Indian Pharmacopoeia (2010) Vol-III

You might also like

You are on page 1of 1112

www.webofpharma.

com
I I
P OPOEI
010

Volume III

lliQfI'lVlllit

Government of India
Ministry of Health & Family Welfare

Published by
THE INDIAN PHARMACOPOEIA COMMISSION
GHAZIABAD

www.webofpharma.com
© 2010, Indian Pharmacopoeia Commission
Application for reproduction should be made to
The Secretary-cum-Scientific Director
INDIAN PHAR1vlACOPOEIA COMMISSION
Sector-23, Raj Nagar,
Ghaziabad-201 002, India
Tel: (91-120)-2783401
Fax: (91-120)-2783311
Website: www.ipc.gov.in
E.mail: ipclab@vsnl.net

ISBN 81-903436-8-8 (Vol. III)


ISBN 81-903436-9-6 (Set)

Sixth Edition (6.0)


Effective from 1st September, 2010

On behalfof Government of India


Ministry ofHealth & Family Welfare

Designed, produced & published by The Indian Pharmacopoeia Commission


Indian Pharmacopoeia Laboratory
Govt. ofIndia, Ministry of Health & Family Welfare
. Sector 23, Raj Nagar, Ghaziabad-201 002

Printed at National Institute of Science Communication And


Information Resources (NISCAIR)
Council ofScientific & Industrial Research
Pusa Gate, Dr. K.S. Krishnan Marg, New Delhi-11O 012

Price per set: Inland Rs 20,000


Foreign $ 900
£ 590

ISBN 81-903436-8-8 ISBN 81-903436-9-6

t88190 343688 08> t88190343695 I


06>

www.webofpharma.com
INDIAN
PHARMACOPOEIA
2010

Volume III

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010

CONTENTS

VOLUME I
Notices v
Preface Vl1

Indian Pharmacopoeia Commission IX

Acknowledgements xv
Introduction XVl1

General Chapters 7

VOLUME II
General Notices 711
General Monographs on Dosage Forms 719
Monographs on Drug substances, Dosage forms and
Pharmaceutical aids (A to M) 755

VOLUME III
General Notices 1729
Monographs on Drug substances, Dosage forms and
Pharmaceutical aids (N to Z) 1737
Monographs on Vaccines and Immunosera for Human Use 2345
Monographs on Herbs and Herbal Products 2463
Monographs on Blood and Blood-related Products 2555
Monographs on Biotechnology Products 2591
Monographs on Veterinary Products 2619
Index 2755

1725

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2007

VolumeIIl
CONTENTS

VOLUME 3

General Notices 1729


Monographs on Drug substances, Dosage fOTITIS and
Phannaceutical aids (N to Z) 1737
Monographs on Vaccines and Immunosera for Human Use 2345
Monographs on Herbs andHerbal Products 2463
Monographs on Blood and Blood-related Products 2555
Monographs on Biotechnology Products 2591
Monographs on Veterinary Products 2619
Index 2755

1727

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 GENERAL NOTICES

GENERAL NOTICES

General Statements 1731


Name 1731
Official and OfficialArticles 1731
Official Standards 1731
Added Substances 1731
Alternative Methods 1731
Meanings ofTerms 1732
Provisions Applicable to Monographs and Test Methods 1732
Expression ofContents 1732
Expression ofConcentrations 1732
Abbreviated Statements ~ ... 1732
Weights and Measures 1733
Monographs 1733
General Monographs 1733
Production 1733
Manufacture ofDrug Products 1733
Excipients 1" ••• 1733
Individual Monographs 1733
Titles 1733
Chemical Formulae 1733
Atomic and Molecular Weights 1733
Definitions 1733
Statement ofContent 1734
Category 1734
Dose 1734
Usual Strength 1734
Description 1734
Solubility 1734
Test Methods 1734
Identification 1734

1729

www.webofpharma.com
GENERAL NOTICES INDIAN PHARMACOPOEIA 2010

Tests and Assays 1734


Tests 1734
Other Tests 1735
Limits 1735
Quantities 1735
Apparatus 1735
Reagents and Solutions 1735
fudicators 1735
Reference Substances 1735
TestsAnimals 1735
Calculation ofResults 1735
Storage 1736
Storage Containers 1736
Labelling 1736

1730

www.webofpharma.com
IP 2010 GENERAL NOTICES

General Notices use but not necessarily to articles that may be sold under the
same name for other purposes.
The active pharmaceutical ingredients (drug substances),
General Statements
excipients (pharmaceutical aids), phannaceutical preparations
The General Notices provide the basic guidelines for the (dosage forms) and other articles described in the monographs
interpretation and application of the standards, tests, assays, are intended for human and veterinary use (unless explicitly
and other specifications of the Indian Phannacopoeia (IP), as restricted to one of these uses).
well as to the statements made in the monographs and other The requirements given in the monographs are not framed to
texts of the Phannacopoeia. provide against all possible impurities, contaminants or
A monograph is to be constructed in accordance with any adulterants; they provide appropriate limitation of potential
general monograph or notice or any appendix, note or other impurities only.
explanatory material that is contained in this Phannacopoeia A preparation must comply throughout the shelf-life assigned
and that is applicable to that monograph. All statements to it by the manufacturer; for opened or broached containers
contained in the monograph, except where a specific general the maximum period of validity for use may sometimes be
notice indicates otherwise and with the exceptions given stated in the individual monograph. Nevertheless, the
hereafter, constitute standards for the official articles. An article responsibility for assigning the period of validity shall be
is not ofpharmacopoeial quality unless it complies with all of with the manufacturer.
the requirements stated.
Added Substances. An official substance, as distinguished
Exceptions to the General Notices do exist, and where they from an official preparation, contains no added substances
do, the wording in the individual monograph or an appendix except when specifically pennitted in the individual monograph.
takes precedence and specifically indicates directions or the Unless otherwise specified in the individual monograph, or
intent. Thus, the specific wording of standards, tests, assays elsewhere in the General Notices, suitable substances may be
and other specifications is binding wherever deviations from added to an official preparation to enhance its stability,
the General Notices exist. Likewise, where there is no specific usefulness or elegance, or to facilitate its preparation. Such
mention to the contrary, the General Notices apply. auxiliary substances shall be hannless in the amounts used,
shall not exceed the minimum quantity required to provide
Name. The full name or title ofthis book, including addenda their intended effect, shall not impair the therapeutic'efficacy
thereto, is Indian Phannacopoeia 2010, abbreviated to IP 2010. or the bioavailability or safety ofthe preparation and shall not
In the texts, the term "Pharmacopoeia" or "IP" without interfere with the tests and assays prescribed for detennining
qualification means the Indian Phannacopoeia 2010 and any compliance with the official standards. Particular care should
addenda thereto. be taken to ensure that such substances are free from hannful
organisms. The freedom to the manufacturers to add auxiliary
Official and Official Articles. The word 'official' wherever
substances imposes on them the responsibility of satisfying
used in this Pharmacopoeia or with reference thereto, is
the licensing authorities on the purpose of the addition and
synonymous with 'pharmacopoeial', with 'IP' and with
the innocuity of such substances.
'compendial'. The designation IP in conjunction with the
official title on the label of an article is an indication that the Alternative Methods. The tests and assays described are the
article purports to comply with IP standards. official methods upon which the standards of the
Pharmacopoeia are based. Alternative methods of analysis
The following tenns are used where the articles for which
may be used for control purposes, provided that the methods
monographs are provided are to be distinguished.
used are shown to give results of equivalent accuracy and
An official substance is a single drug or a drug entity or a enable an unequivocal decision to be made as to whether
pharmaceutical aid for which the monograph title includes no compliance with the standards of the monographs would be
indication of the nature of a dosage fonn. achieved if the official methods were used. Automated
procedures utilising the same basic chemistry as the test
An official preparation is a drug product (dosage form) and is
procedures given in the monograph may also be used to
the finished or partially finished preparation or product ofone
determine compliance. Such alternative or automated
or more official substances fonnulated for use on the patient.
procedures must be validated.
An article is an item for which a monograph is provided,
In the event of doubt or dispute, the methods of analysis of
whether an official substance or an official preparation.
the Phannacopoeia are alone authoritative and only the result
Official Standards. The requirements stated in the obtained by the procedure given in this Phannacopoeia is
monographs apply to articles that are intended for medicinal conclusive.

1731

www.webofpharma.com
GENERAL NOTICES IP 2010

Meanings ofTerms per cent v/v (percentage, volume in volume) expressing


Alcohol. The term "alcohol" without qualification means the number ofmillilitres of substance in 100 millilitres of
ethanol (95 per cent). Other dilutions of ethanol are indicated final product.
by the term "ethanol" or "alcohol" followed by a statement of The expression "parts per million" refers· to the weight in
the percentage by volume of ethanol (C 2H60) required. weight, unless otherwise stated.
Desiccator. A tightly-closed container of suitable size and Where the content of a substance is expressed in terms of the
design that maintains an atmosphere oflow moisture content chemical formula for that substance an upper limit exceeding
by means of silica gel or phosphorus pentoxide or other 100 per cent may be stated. Such an upper limit applies to the
suitable desiccant. result ofthe assay calculated in terms ofthe equivalent content
ofthe specified chemical formula. For example, the statement
Drying and ignition to constant weight. Two consecutive 'contains not less than 99.0 per cent and not more than 101.0
weighings after the drying or igniting operations do not differ per cent of C 7H 60 2 implies that the result of-the assay is not
by more than 0.5 mg, the second weighing following an less than 99.0 per cent and not more than 101.0 per cent,
additional period of drying or of ignition respectively calculated in terms ofthe equivalent content of C 7H 60 2 .
appropriate to the nature and quantity of the residue.
Where the result ofan assay or test is required to be calculated
Ethanol. The term "ethanol" without qualification means with reference to the dried, anhydrous, ignited substance, or
anhydrous ethanol or absolute alcohol. the substance free from solvent, the determination of loss on
drying, water content, loss on ignition, content ofthe specified
Filtration. Unless otherwise stated, filtration is the passing of
a liquid through a suitable filter paper or equivalent device solvent, respectively is carried out by the method prescribed
in the relevant test in the m o n o g r a p h . ·
until the filtrate is clear.
Expression of Concentrations. The following expressions in
Freshly prepared. Made not more than 24 hours before it is
addition to the ones given under Expression of Content are
issued for use..
also used:
Label. Any printed packing material, including package inserts
per cent w/v (percentage, weight in volume) expressing
that provide information on the article.
the number of grams of substance in 100 millilitres of
Negligible. A quantity not exceeding 0.50 mg. product
Solution. Where the name of the solvent is not stated, per cent v/w (percentage, volume in weight) expressing
"solution" implies a solution in water. The water used complies the number of millilitres of substance in 100 grams of
with the requirements of the monograph on Purified Water. product.
The term 'distilled water' indicates Purified Water prepared by Usually, the strength of solutions of solids in liquids is
distillation. expressed as percentage weight in volume, ofliquids in liquids
Temperature. The symbol 0 used without qualification as percentage volume in volume, of solids in semi-solid bases
indicates the use of the Celsius thermometric scale. (e.g. creams) and of gases in liquids as percentage weight in
weight.
Water. Ifthe term is used without qualification it means Purified
Water of the Pharmacopoeia. The term 'distilled water' When the concentration of a solution is expressed as parts of
indicates Purified Water prepared by distillation. dissolved substance in parts of solution, it means parts by
weight (g) ofa solid in parts by volume (m!) ofthe final solution;
Water-bath. A bath of boiling water unless water at another as parts by weight (g) of a gas in parts by weight (g) of the
temperature is indicated. Other methods of heating may be final solution.
used provided the required temperature is approximately
When the concentration of a solution is expressed in molarity
maintained but not exceeded.
designated by the symbol M preceded by a number, it denotes
the number ofmoles ofthe stated solute contained in sufficient
Provisions Applicable to Monographs and Test Methods
Purified Water (unless otherwise stated) to produce 1 litre of
Expression of Contents. Where the content of a substance is solution.
defined, the expression "per cent" is used according to
Abbreviated Statements. Incomplete sentences are employed
circumstances with one of two meanings:
in parts of the monographs for directness and brevity (for
per cent w/w (percentage, weight in weight) expressing example, Iodine Value. Not more than ; Relative Density.
the number of grams of substance in 100 grams of fmal .......to ........) Where the tests are abbreviated, it is to be
product, understood that the test method referred to in brackets

1732

www.webofpharma.com
IP 2010 GENERAL NOTICES

provides the method to be followed and that the values Excipients. Any substance added in preparing an official
specified are the applicable limits. preparation shall be innocuous, shall have no adverse influence
in the therapeutic efficacy of the active ingredients and shall
Weights and Measures. The metric system of weights and
not interfere with the tests and assays of the Pharmacopoeia.
measures is employed in the Pharmacopoeia. All measures are
Care should be taken to ensure that such substances are free
required to be graduated at 25° and all measurements in tests
from halwful organisms.
and assays, unless otherwise stated, are to be made at that
temperature. Graduated glass apparatus used in analytical Individual Monographs
operations shall comply with the requirements stated in
Chapter 2.1.6 Drug products that are the subject ofan individual monograph
are also required to comply with the tests given in the general
monographs.
Monographs Titles. The main title for a drug substance is the International
(;eneralMonographs Non-proprietary Name (INN) approved by the World Health
Organization. Subsidiary names and synonyms have also been
General monographs on dosage fonus include requirements given in some cases; where included, they have the same
ofgeneral application and apply to all preparations within the significance as the main title.
scope of the Introduction section of the general monograph, The main titles of drug products are the ones commonly
except where a preamble limits the application. The recognised in practice. Synonyms drawn from the full non-
requirements are not necessarily comprehensive for a given proprietary name of the active ingredient or ingredients have
specific preparation; additional requirements may sometimes also been given. Where, however, a product contains one or
be given in the individual monograph for it. the other ofdifferent salts ofan active molecule, the main title
Production. Statements given under the heading Production is based on the full name ofthe active ingredient. For example,
relate to particular aspects of the mimufacturing process and Chloroquine Phosphate Tablets and Chloroquine
are not necessarily comprehensive. However, they are SulphateTablets.
mandatory instructions to manufacturers. They may relate, Chemical Formulae. When the chemical structure ofan official
for example, to source materials, to the manufacturing process substance is known or generally accepted, the graphic and
and its validation and control, to any in-process testing that molecular formulae are normally given at the beginning ofthe
is to be carried out by the manufacturer on the final product monograph for. information. This information refers to the
either on selected batches or on each batch prior to release. chemically pure substance and is not to be regarded as an
All this cannot be verified on a sample ofthe final product by indication of the purity of the official material. Elsewhere, in
an independent analyst. It is for the licensing authority to statement of purity and strength and in descriptions of
verify that the instructions have been followed. processes of assay, it will be evident from the context that the
The absence of a section on Production does not imply that formulae denote the chemically pure substances.
attention to features such as those given above is not required. Where the absolute stereochemical configuration is specified,
An article described in a monograph of the Pharmacopoeia is the International Union of Pure and Applied Chemistry
to be manufactured in accordance with the principles of good (IUPAC) RlS and E/Z systems of designation have been used.
manufacturing practice and in accordance with the If the substance is an enantiomer of unlmown absolute
requirements of the Drugs and Cosmetics Rules, 1945. The stereochemistry, the sign ofthe optical rotation, as determined
general principles applicable to the manufacture and quality in the solvent and under the conditions specified in the
assurance of drugs and preparations meant for human use monograph, has been attached to the systematic name. An
apply equally to veterinary products as well. indication ofsign ofrotation has also been given where this is
Manufacture of Drug Products. The opening definitive incorporated in a trivial name that appears on an IUPAC
statement in certain monographs for drug products is given in preferred list.
terms ofthe active ingredient(s) only. Any ingredient(s) other Atomic and Molecular Weights. The atomic weight or
than those included in the statement, must comply with the molecular weight is shown, as and when appropriate at the
general notice on Excipients and the product must conform to top right hand corner of the monograph. The atomic and
the Pharmacopoeial requirements. molecular weights and graphic formulae do not constitute .
Official preparations are prepared only from ingredients that analytical standards for the substances described.
comply with the requirements of the pharmacopoeial Definition. The opening statement of a monograph is one
monographs for those individual ingredients for which that constitutes an official definition of the substance,
monographs are provided. preparation or other article that is the subject of the

1733

www.webofpharma.com
GENERAL NOTICES IP 2010

monograph. In certain monographs for pharmaceutical Solubility. Statements on solubility are given in Chapter 2.4.26
preparations the statement is given in terms of the principal and are intended as infonnation on the approximate solubility
ingredient(s). at a temperature between 15° and 30°, unless otherwise stated,
In monographs on vegetable drugs, the definition indicates and are not to be considered as official requirements. However,
whether the subject of the monograph is, for example, the a test for solubility stated in a monograph constitutes part of
whole drug or the drug in powdered fonn. the standards for the substance that is the subject of that
monograph.
Certain pharmaceutical substances and other articles are
defined by reference to a particular method ofmanufacture. A Test Methods
statement that a substance or article is prepared or obtained
by a certain method constitutes part of the official definition References to general methods of testing are indicated by test
and implies that other methods are not permitted. A statement method numbers in brackets immediately after the heading of
that a substance may be prepared or obtained by a celiain the test or at the end of the text.
method, however, indicates that this is one possible method Identification. The tests given under the heading Identification
and does not imply that other methods are not permissible. are not necessarily sufficient to establish absolute proof of
Statement of content. The limits of content stated are those identity. They provide a means of verifying that the identity
detennined by the method described under Assay. of the material under examination is in accordance with the
label on the container.
Category. The statement of category is provided for
information and is indicative ofthe medical or pharmaceutical In celtain monographs alternative series ofidentification tests
basis for recognition in the Pharmacopoeia. It generally are given; compliance with either one or the other set of tests
represents an application of the best known pharmacological is adequate to verify the identity of the aIticle.
action of the article or of its active ingredient. In the case of When tests for infrared absorption are applied to material
pharmaceutical aids it may indicate the more common usage extracted fi'om fonnulated preparations, strict concordance
ofthe article. The statement is not intended to limit iIi any way with the specified reference spectrum may not always be
the choice or use of the article nor to indicate that it has no possible, but nevertheless a close resemblance between the
other activity or use. spectrum ofthe extracted material and the specified reference
Dose. Doses mentioned in the Pharmacopoeia are intended spectrum should be achieved.
merely for general guidance and represent, unless otherwise
stated, the average range of quantities which are generally Tests andAssays
regarded as suitable for adults when administered by mouth. The tests and assays are the official methods upon which the
They are not to be regarded as binding upon the prescribers. standards of the Pharmacopoeia depend. The requirements
The medical practitioner will exercise his own judgment and are not framed to take into account all possible impurities. It is
act on his own responsibility in respect of the amount of any not to be presumed, for example, that an impurity that is not
therapeutic agent he may prescribe or administer or the detectable by means of the prescribed tests is tolerated.
frequency of its administration. If it is usual to administer a Material found to contain such an impurity is not of
drug by a method other than by mouth, the single dose suitable pharmacopoeial quality ifthe nature or amount ofthe impurity
for that method of administration is mentioned. In the case of found is incompatible with good pharmaceutical practice.
some preparations notes have been given below the statement
of doses to show the approximate quantities of active Pharmacopoeial methods and limits should be used merely as
ingredients contained in the maximal doses as information for compliance requirements and not as requirements to guarantee
the prescriber. total quality assurance. Tests and assays are prescribed for
the minimum sample available on which the attributes ofthe
Usual Strength. The statement on the usual strength(s) of a
alticle should be measured. Assurance of quality must be
preparation given in the individual monograph indicates the
ensured by the manufacturer by the use of statistically valid
strength(s) usually marketed for information ofthe pharmacist
sampling and testing programmes.
and the medical practitioner. It does not imply that a strength
other than the one(s) mentioned in the individual monograph Tests. Unless otherwise stated, the assays and tests are carried
meeting all the prescribed requirements cannot be out at a temperature between 20° and 30°.
manufactured and marketed with the approval of the Where it is directed that an analytical operation is to be carried
appropriate authority. out 'in subdued light', precautions should be taken to avoid
Description. The statements under the heading Description exposure to direct sunlight or other strong light. Where a
are not to be interpreted in a strict sense and are not to be procedure is directed to be performed 'protected from light'
regarded as official requirements. precautions should be taken to exclude actinic light by the

1734

www.webofpharma.com
IP 2010 GENERAL NOTICES

use oflow-actinic glassware, working in a dark room or similar Unless otherwise stated, comparative tests are carried out
procedures. using identical tubes of colourless, transparent, neutral glass
with a flat base, commonly known as Nessler cylinders.
For preparations other than those of fixed strength, the
quantity to be taken for a test or an assay is usually expressed Reagents and Solutions. The reagents required for the tests
in terms of the active ingredient. This means that the quantity and assays of the Pharmacopoeia are defined in the various
ofthe active ingredient expected to be present and the quantity chapters showing their nature, degree of purity and the
of the preparation to be taken are calculated from the strength strengths of the solutions to be made from them. The
stated on the label. requirements set out are not intended to imply that the materials
are suitable for use in medicine; reagents not covered by
Other Tests. In the monographs on dosage forms and certain
monographs in the phannacopoeia shall not be claimed to be
preparations, under the sub-heading 'Other tests' it is stated
ofIP quality.
that the article complies with the tests stated under the general
monograph ofthe relevant dosage form or preparation. Details The term' analytical reagent grade of commerce' implies that
of such tests are provided in the general monographs. the chemical is ofa high degree ofpurity wherein the limits of
various impurities are known. Where it is directed to use a
Limits. The limits given are based on data obtained in normal
'general laboratory reagent grade ofcommerce' it is intended
analytical practice. They take into account nonnal analytical
that a chemically pure grade material, not necessarily required
errors, of acceptable variations in manufacture and of
to be tested for limiting or absence of certain impurities, is to
deterioration to an extent that is acceptable. No further
be used.
tolerances are to be applied to the limits for determining whether
or not the article under examination complies with the Indicators. Where the use ofan indicator solution is mentioned
requirements of the monograph. in an assay or test, approximately 0.1 ml of the solution shall
be added, unless otherwise directed.
Quantities. Unless otherwise stated, the quantities to be taken
for assays, limit tests and other tests are of the substance Reference Substances. Certain monographs require the use
under examination. of a chemical reference substance or a biological reference
preparation or a reference spectrum These are authentic
In tests with numerical limits and assays, the quantity stated
specimens chosen and verified on the basis oftheir suitability
to be taken for testing is approximate. The amount actually
for intended use as prescribed in the Pharmacopoeia and are
used, which may deviate by not more than 10 per cent from
not necessarily suitable in other circumstances.
that stated, is accurately weighed or measured and the result
ofanalysis is calculated from this exact quantity. In tests where IP Reference Substances, abbreviated to IPRS (and referred
the limit is not numerical but usually depends upon to as RS in the individual monographs) are issued by the
comparison with the behaviour of a reference in the same Indian Pharmacopoeia Commission (IPC). They are the official
conditions,. the stated quantity is taken for testing. Reagents standards to be used in cases of arbitration. Secondary
are used in the prescribed amounts. Standards (Working Standards) may be used for routine
analysis, provided they are standardized at regular intervals
Quantities are weighed or measured with an accuracy
against the Reference Substances
commensurate with the indicated degree of precision. For
weighings, the precision is plus or minus 5 units after the last Biological Reference Substances, .also abbreviated to IPRS
figure stated. For example, 0.25 g is to be interpreted as 0.245 and Standard Preparations of antibiotics are issued by
g to 0.255 g. For the measurement of volumes, if the figure agencies authorised by the IPC. They are standardized against
after the decimal point is a zero or ends in a zero, e.g. 10.0 ml or the International Standards and Reference Preparations
0.50 ml, the volume is measured using a pipette, a volumetric established by the World Health Organization (WHO). The
flask or a burette, as appropriate; in other cases, a graduated potency of these preparations is expressed in International
measuring cylinder or a graduated pipette may be used. Units. .
Volumes stated in microlitres are measured using a micropipette Reference spectra are published by the IPC and they are
or microsyringe. accompanied by information concerning the conditions used
The term 'transfer' is used generally to indicate a quantitative for sample preparation and recording of the spectra.
operation. Test Animals. Unless otherwise directed, animals used in a
Apparatus. Measuring and weighing devices and other test or an assay shall be healthy and are drawn from a unifOlID
apparatus are described in the chapter entitled'Apparatus for stock, and have not previously been treated with any material
Tests and Assays'. A specification for a definite size or type that will interfere with the test or the assay.
of container or apparatus in a test or assay is given merely as Calculation of Results. In determining compliance with a
a recommendation. numerical limit in assay or test, the result should be calculated

1735

www.webofpharma.com
GENERAL NOTICES IP 2010

to one decimal place more than the significant figures stated Where no specific storage directions or limitations are given
and then rounded up or down as follows: if the last figure in the monograph or by the manufacturer, it is to be understood
calculated is 5 to 9, the preceding figure is increased by 1; if it that the storage conditions include protection from moisture,
is 4 or less, the preceding figure is left unchanged. freezing and excessive heat (any temperature above 40°).
Storage. Statements under the side-heading Storage constitute Storage Containers. The requirements, guidance and
non-mandatory advice. The articles ofthe Pharmacopoeia are information on containers for pharmaceutical use are given in
to be stored under conditions that prevent contamination and, the chapter entitled Containers (6.1)
as far as possible, deterioration. Precautions that should be In general, an article should be packed in a well-closed
taken in relation to the effects of the atmosphere, moisture, container i.e. one that protects the contents from contamination
heat and light are indicated, where appropriate, in the individual by extraneous solids, liquids or vapours and from loss of the
monograph. article under normal conditions of handling and storage.
Where, additionally, loss or deterioration of the article from
Specific directions are given in some monographs with respect effervescence, deliquescence or evaporation under normal
to the temperatures at which Pharmacopoeial articies should conditions ofstorage is likely, the container must be capable
be stored, where it is considered that usage at a lower or of being tightly closed, and re-closed after use.
higher temperature may produce undesirable results. The
In certain cases, special requirements of pack have been
storage conditions are defined by the following terms:
indicated in some monographs under Storage, using
Store in a dry, well ventilated place at a temperature not expressions that have been defined in chapter 6.1.
exceeding 30°
Labelling. The labelling of drugs and pharmaceuticals is
Store in a refrigerator (20 to 8°). Do not freeze governed by the Drugs and Cosmetics Rules, 1945. The
Store in a freezer (-2° to -18°) statements that are given in the monographs under the side-
heading 'Labelling' are not comprehensive. Only those that
Store in a deep freezer (Below _18°)
are necessary to demonstrate compliance or otherwise with
Storage conditions not related to temperature are indicated in the monograph have been given and they are mandatory. For
the following terms: example, in the monograph on Betamethasone Sodium Tablets
the labelling statement is "The label states the strength in
Store protected from light
terms ofthe equivalent amount ofbetamethasone". Any other
Store protected from light and moisture statements are included as recommendations.

1736

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

DRUG SUBSTANCES, DOSAGE FORMS


AND
PHARMACEUTICAL AIDS

NtoZ .... 1739

1737

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

NalidixicAcid 1743
Nalidixic Acid Tablets 1743
Nalorphine Hydrochloride 1744
Nalorphine Injection 1745
Naloxone Hydrochloride 1745
Naloxone Injection 1747
Naltrexone Hydrochloride 1748
Naltrexone Tablets 1749
Nandrolone Decanoate 1750
Nandrolone Decanoate Injection 1751
Nandrolone Phenylpropionate 1751
Nandrolone Phenylpropionate Injection 1752
Naphazoline Nitrate 1753
Naproxen 1754
Naproxen Oral Suspension 1755
Naproxen Suppositories 1756
Naproxen Sustained-release Tablets 1757
Naproxen Tablets 1758
Nebivolol Hydrochloride 1758
Nebivolol Tablets 1759
NelfinavirMesylate 1760
Nelfinavir Mesylate Oral Powder 1761
Nelfinavir Tablets 1762
Neomycin Sulphate 1763
Neomycin Eye Drops 1764
Neomycin Eye Ointment 1765
Neostigmine Bromide 1766
Neostigmine Tablets 1767
I

Neostigmine Methylsulphate 1767


Neostigmine Injection 1768

1739

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Neotame 1769
Nevirapine 1770
Nevirapine Oral Suspension 1771
Nevirapine Tablets 1772
Niclosamide 1773
Niclosamide Tablets 1774
Nicotinamide 1775
Nicotinamide Tablets 1776
NicotinicAcid 1776
Nicotinic Acid Tablets 1777
Nicoumalone 1777
Nicoumalone Tablets 1778
Nifedipine 1779
Nifedipine Capsules 1780
Nifedipine Sustained-release Tablets 1781
Nifedipine Tablets 1782
Nikethamide 1783
Nikethamide Injection 1784
Nitrazepam 1784
Nitrazepam Tablets 1785
Nitrofurantoin 1786
Nitrofurantoin Tablets 1787
Nitrofurazone 1787
Nitrous Oxide 1788
Noradrenaline Bitartrate 1789
Noradrenaline Bitartrate Injection 1790
Norethisterone 1791
Norethisterone Tablets 1792
Norfloxacin 1793
Norfloxacin Eye Drops 1794
Norfloxacin Tablets 1794
Norgestrel 1795
Norgestrel and Ethinyloestradiol Tablets 1796

1740

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Nortriptyline Hydrochloride 1797


Nortriptyline Tablets 1797
Noscapine 1798
Noscapine Linctus 1800
Novobiocin Sodium 1800
Nystatin 1801
Nystatin Ointment 1802
Nystatin Pessaries 1803
Nystatin Tablets 1803

1741

www.webofpharma.com
www.webofpharma.com
IP 2010 NALIDIXIC ACID TABLETS

Nalidixic Acid Reference solution (a). A 0.002 per cent w/v solution of the
substance under examination in dichloromethane.
Reference solution (b). A 0.0008 per cent w/v solution of the
substance under examination in dichloromethane.
Reference solution (c). A 0.1 per cent w/v solution of nalidixic
acid RS in dichloromethane.
Apply to the plate 10 III ofeach solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with test
Mol. Wt. 232.2 solution (a) is not more intense than the spot in the
chromatogram obtained with reference solution (a) and not
Nalidixic Acid is l-ethyl-l ,4-dihydro-7-methyl-4-oxo-l ,8-
more than one such spot is more intense than the spot in the
naphthyridine-3-carboxylic acid.
chromatogram obtained with reference solution (b).
Nalidixic Acid contains not less than 99.0 per cent and not
Heavy metals (2.3.13).1.0 g complies with the limit test for
more than 101.0 per cent of ClzHI2Nz03, calculated on the
heavy metals, Method B (20 ppm).
dried basis.
Sulphated ash (2.3.18) Not more than 0.1 per cent.
Category. Antibacterial.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Dose. 2 to 4 g daily, in divided doses.
on 1.0 g by drying in an oven at 105 0 •
Description. A white to slightly yellow, crystalline powder.
Assay. Weigh accurately about 0.15 g, dissolve in 10 ml of
Identification dichloromethane, add 30 ml of 2-propanol and 10 mlof carbon
dioxide-free water and titrate with 0.1 M ethanolic sodium
Test A may be omitted iftests B, C andD are carried out. Tests hydroxide, determining the end-point potentiometrically
B, C and D may be omitted if test A is carried out. (2.4.25) and using a glass electrode as the indicator electrode
A. Determine by infrared absorption spectrophotometry (2.4.6). and a silver-silver chloride reference electrode with a sleeve
Compare the spectrum with that obtained with nalidixic acid diaphragm or a capillary tip filled with a saturated solution of
RS or with the reference spectrum ofnalidixic acid. lithium chloride in ethanol. Throughout the titration keep
the temperature ofthe solution at 15 0 to 20 0 and pass a current
B. When examined in the range 230 nm to 360 nm (2.4.7), a of nitrogen through the solution.
0.0005 per cent w/v solution in 0.1 M sodium hydroxide shows
absorption maxima at about 258 nm and 334 nm; ratio of the 1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
absorbance at about 258 nm to that at about 334 nm, 2.2 to 2.4. 0.02322 g ofClzHlzNz03.
C. In the test for Related substances, the principal spot in the Storage. Store protected from light and moisture.
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution (c).
D. Dissolve 0.1 gin 2 ml of hydrochloric acid and add 0.5 ml Nalidixic Acid Tablets
of a 10 per cent w/v solution of 2-naphthol in ethanol (95 per
cent); an orange-red colour develops.
Nalidixic Acid Tablets contain not less than 95.0 per cent and
not more than 105.0 per cent ofthe stated amount ofnalidixic
Tests acid, ClzHlzNz03.

Related substances. Determine by thin-layer chromatography Usual strengths. 250 mg; 500 mg.
(2.4.17), coating the plate with silica gel HF254. ' Identification
Mobile phase. A mixture of 70 volumes of ethanol (95 per
To a quantity ofthe powdered tablets containing 1g ofNalidixic
cent), 20 volumes of dichloromethane and 10 volumes of5 M
Acid add 50 ml of chloroform, shake for 15 minutes, filter and
ammonia.
evaporate the filtrate to dryness. The residue, after drying at
Test solution (a). Dissolve 0.2 g of the substance under 105 0 , complies with the following tests.
examination in 10 ml of dichloromethane.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Test solution (b). A 0.1 per cent w/v solution of the substance Compare the spectrum with that obtained with nalidixic acid
under examination in dichloromethane. RS or with the reference spectrum ofnalidixic acid.

1743

www.webofpharma.com
NALIDIXIC ACID TABLETS IP 2010

B. When examined in the range 230 nm to 360 nm (2.4.7), a Nalorphine Hydrochloride contains not less than 97.0 per cent
0.0005 per cent w/v solution in 0.1 M sodium hydroxide shows and not more than 103.0 per cent ofCI9HzIN03,HCl, calculated
absorption maxima at about 258 nm and 334 nm; ratio of the on the dried basis.
absorbance at about 258 nm to that at about 334 nm, 2.2 to 2.4.
Category. Narcotic antagonist.
Tests Dose. By intravenous injection, 5 mg, repeated twice at three
Related substances. Determine by thin-layer chromatography minute intervals, if necessary.
(2.4.17), coating the plate with silica gel HF254. Description. A white or almost white, crystalline powder;
Mobile phase. A mixture of 70 volumes of ethanol (95 per odourless. It slowly darkens on exposure to air and light.
cent), 20 volumes of dichloromethane and 10 volumes of
5 Mammonia.
Identification
Test solution. Shake a quantity of the powdered tablets Test A may be omitted if tests B, C, D and E are carried out.
containing 0.1 g ofNalidixic Acid with 50 ml of chloroform for Tests C and D may be omitted if tests A, Band E are carried
15 minutes, filter, evaporate the filtrate to dryness and dissolve out.
the residue in 5 ml of chloroform. A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution. Dilute 1 volume of the test solution to Compare the spectrum with that obtained with nalorphine
200 volumes with chloroform. hydrochloride RS.
Apply to the plate 10 III of each solution. After development, B. When examined in the range 230 nm to 360 nm (2.4.7), a
dry the plate in air and examine in ultraviolet light at 254 nm. 0.01 per cent w/v solution in 0.1 M sodium hydroxide shows
Any secondary spot in the chromatogram obtained with the an absorption maximum only at about 298 nm; absorbance at
test solution is not more intense than the spot in the about 298 nm, about 0.6.
chromatogram obtained with the reference solution.
C. To 10 ml of a 2 per cent w/v solution add 0.05 ml of dilute
Other tests. Complies with the tests stated under Tablets. ammonia solution; a white precipitate soluble in sodium
Assay. Weigh and powder 20 tablets. Weigh accurately a hydroxide solution is produced.
quantity of the powder containing about 0.1 g of Nalidixic D. Dissolve 2 mg in 2 ml of water, add 0.15 ml ofpotassium
Acid, add 150 ml of 0.1 M sodium hydroxide, shake for 3 fen-icyanide solution containing, in each ml, 0.05 mlofferric
minutes, dilute to 200.0 ml with 0.1 M sodium hydroxide, mix chloride solution; a deep bluish green colour is produced
and allow to stand for 15 minutes. Dilute 2.0 ml ofthe solution immediately.
to 100.0 ml with water and measure the absorbance of the
resulting solution at the maximum at about 334 nm (2.4.7), E. Gives reaction A ofchlorides (2.3.1).
using 0.1 M sodium hydroxide as the blank Calculate the
content of C 12H 12N z0 3 taking 494 as the specific absorbance Tests
at 334 nm. Melting range (2.4.21). 260 0 to 263 0 •
Storage. Store protected from light and moisture. Acidity. Dissolve 0.2 g in 10 ml offreshly boiled and cooled
water and titrate with O. 02 M sodium hydroxide using methyl
red solution as indicator; not more than 0.2 ml of 0.02 M
Nalorphine Hydrochloride sodium hydroxide is required to change the colour of the
solution.
Specific optical rotation (2.4.22). -122 0 to -125 0 , determined
in a 2.0 per cent w/v solution.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 1000 at a pressure not exceeding
0.7 kPa for 2 hours.
Assay. Weigh accurately about 25 mg and dissolve in sufficient
CI9HzIN03,HCl Mol. Wt. 347.8
water to produce 250 ml. Measure the absorbance of the
Nalorphine Hydrochloride is 17-allyl-7,8-didehydro-4,5a- resulting solution at the maximum at about 285 nm (2.4.7).
epoxymorphinan-3,6a-diol hydrochloride. Calculate the content ofCI9HzIN03,HCl from the absorbance

1744

www.webofpharma.com
IP 2010 NALOXONE HYDROCHLORIDE

obtained by repeating the operation with nalorphine by repeating the operation with nalorphine hydrochloride
hydrochloride RS in place of the substance under RS.
examination.
Storage. Store protected from light.
Storage. Store protected from light and moisture.

Naloxone Hydrochloride
Nalorphine Injection
Naloxone Hydrochloride Dihydrate
Nalorphine Hydrochloride Injection
Nalorphine Injection is a sterile solution of Nalorphine HO
Hydrochloride in Water for Injections containing suitable
buffering agents.
Nalorphine Injection contains not less than 90.0 per cent and N~CH2
not more than 110.0 per cent ofthe stated amount ofnalorphine 1Ilo...00..-'"

,-'
hydrochloride, CI9H21N03,HCl.
Usual strength. 10 mg per ml.
o
CI9H2IN04,HCI,2H20 Mol. Wt. 399.9
Identification Naloxone Hydrochloride is 17-allyl-4,5-a-epoxy-3,14-
dihydroxymorphinan-3-one hydrochloride dihydrate.
A. To a volume containing 50 mg ofNalorphine Hydrochloride
add dilute ammonia solution until the solution is alkaline and Naloxone Hydrochloride contains not less than 98.0 per cent
extract with 25 ml ofa mixture of 1 volume of ethanol (95 per and not more than 102.0 per cent of C19H22ClN04, calculated
cent) and 3 volumes of chloroform and evaporate the extract on the anhydrous basis.
to dryness. Dry the residue at a pressure not exceeding 2 kPa.
Category. Antidote for opioids poisoning.
The residue complies with the following test.
Description. A white to almost white, hygro$copic, crystalline
Determine by infrared absorption spectrophotometry (2.4.6).
powder.
Compare the spectrum with that obtained with nalorphine
hydrochloride RS. Identification
B. To a volume containing 0.1 g ofNalorphine Hydrochloride
Test A may be omitted if tests Band C are carried out. Test B
add 0.05 ml of dilute ammonia solution; a white precipitate
may be omitted if tests A and C are carried out.
soluble in sodium hydroxide solution is produced.
A. Determine by infrared absorption spectrophotometry
C. Gives reaction A ofchlorides (2.3.1).
(2.4.6).Compare the spectruin with that obtained with naloxone
Tests hydrochloride dihydrate RS or with the reference spectrum
of naloxone hydrochloride.
pH (2.4.24).6.0 to 7.5.
B. Detennine by thin-layer chromatography (2.4.17), coating
Other tests. Complies with the tests stated under Parenteral the plate with silica gel G
Preparations (Injections). Mobile phase. Mix 5 volumes of methanol and 95 volumes of
Assay. Transfer an accurately measured volume containing the upper layer from a mixture of60 ml of ammonia and 100 m1
about 10 mg of Nalorphine Hydrochloride to a separating of butan-I-ol.
funnel, add 1 ml of dilute hydrochloric acid and dilute to Test solution. Dissolve 8 mg of the substance under
10 ml with water. Extract with five successive quantities, each examination in 0.5 ml of water and dilute to 1 m1 with methanol.
of 5 ml, of chloroform, allowing the layers to separate before
drawing offeach cWoroform extract and discard the chloroform Reference solution. Dissolve 8 mg of naloxone hydrochloride
extracts. Transfer the aqueous layer to a 100-ml volumetric dihydrate RS in 0.5 ml of water and dilute to 1 ml with methanol.
flask with the aid of small quantities of water and dilute to Apply to the plate 5 I.d of each solution, Allow the mobile
volume with water. Measure'the absorbance of the resulting phase to rise 8 cm. Dry the plate in air and spray with a freshly
solution at the maximum at about 285 nm (2.4.7). Calculate prepared 0.5 per cent w/v solution ofpotassium ferricyanide
the content of C19H21N03,HCI from the absorbance obtained in ferric chloride solution and examine in daylight. The

1745

www.webofpharma.com
NALOXONE HYDROCHLORIDE IP 2010

principal spot in the chromatogram obtained with the test to pH 2.0 with a 50 per cent v/v solution of
solution corresponds to that in the chromatogram obtained orthophosphoric acid,
with the reference solution. - flow rate. 1.5 ml per minute,
- a linear gradient programme using the conditions given
C. Gives reaction A ofchlorides (2.3.1).
_below,
spectrophotometer set at 230 urn,
Tests
- injection volume. 20 /-ll.
Appearance ofsolution. A2.0 per cent w/v solution in carbon Time Mobile phase A Mobile phase B
dioxide-free water (Solution A) is clear (2.4.1) and colourless (in min.) (per cent v/v) (per cent v/v)
(2.4.1).
0~40 100~0 0~100
Acidity or alkalinity. To 10 ml of solution A add 0.05 ml of 40~50 0 100
methyl red solution. Not more than 0.2 ml of 0.02 M sodium
Inject reference solution (a). Adjust the sensitivity of the
hydroxide or 0.02 M hydrochloric acid is required to change
system so that the peak-to-valley ratio is minimum 2.0, where
the colour of the indicator.
Hp is height above the baseline of the peak due to impurity D
Specific optical rotation (2.4.22). -170° to -181°, detennined and H v is the height above the baseline of the lowest point of
in solution A. the curve separating this peak from the peak due to naloxone.
Related substances. Determine by liquid chromatography The relative retention time with reference to naloxone, for
(2.4.14). impurity C, impurity A, impurity F, impurity D, impurity E and
Test solution. Dissolve 0.125 g of the substance under impurity B is about 0.6 minute, 0.8 minute, 0.9 minute, 1.1
examination in 25 m1 of 0.1 M hydrochloric acid. minutes, 3.0 minutes and 3.2 minutes respectively.

Reference solution (a). Dissolve 5 mg of naloxone for peak Inject reference solution (b) and the test solution. In the
identification RS (containing naloxone impurity A (4,5cx -epoxy- chromatogram obtained with the test solution, the area of
3, 14-dihydroxymorphinan-6-one) (noroxymorphone), each secondary peak corresponding to naloxone impurities
naloxone impurity B (4,5cx-epoxy-14-hydroxy-17-(prop-2-enyl)- A, B, C, E, F is not more than the area ofthe principal peak in
3-(prop-2-enyloxy)morphinan-6-one) (3-0-allylnaloxone), the chromatogram obtained with reference solution (b) (0.2
naloxone impurity C (4,5cx -epoxy-3,1Oli,14-trihydroxy-17-(prop- per cent). The area of secondary peak corresponding to
2-enyl)morphinan-6-one) (1 Ocx -hydroxynaloxone), naloxone naloxone impurity D is not more than 1.5 times the area ofthe
impurity D (7,8-didehydro-4,5cx -epoxy-3,14-dihydroxy-17- principal peak in the chromatogram obtained with reference
(prop-2-enyl)morphinan-6-one) (7 ,8-didehydronaloxone), solution (b) (0.3 per cent).The area' of any other impurities is
naloxone impurity E (4,5 cx:4',5' cx-diepoxy-3,3',14,14'- not more than 0.5 times the area of the principal peak in the
tetrahydroxy-17,IT-bis(prop-2-enyl)-2,2'-bimorphinanyl-6,6'- chromatogram obtained with reference solution (b) (0.1 per
dione) (2,2'-bisnaloxone) and naloxone impurity F (4,5cx- cent). The sum ofthe areas of all other secondary peaks is not
epoxy-3,1013,14-trihydroxy-17- (prop-2-enyl)morphinan-6-one) more than 4 times the area of the principal peak in the
(lOcx-hydroxynaloxone) in 1 m1 of 0.1 M hydrochloric acid. chromatogram obtained with reference solution (b) (0.8 per
cent). Ignore any peak with an area less than 0.25 times the
Reference solution (b). Dilute 1.0 m1 ofthe test solution to 20 area of the principal peak in the chromatogram obtained with
m1 with 0.1 Mhydrochloric acid. Dilute 1.0 ml ofthis solution the reference solution (b) (0.05 per cent).
to 25.0 ml with 0.1 M hydrochloric acid.
Water (2.3.43). 7.5 per cent to 11.0 per cent, determined on
Chromatographic system O.2g.
- a stainless steel column 12.5 cm x 4 mm, packed with
octylsilane bonded to porous silica (5 /-lm), Sulphated ash (2.3.18). Not more than 0.2 per cent, determined
- column temperature. 40°, on 0.5 g.
- mobile phase: A. a mixture of20 volumes of acetonitrile, Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of
40 volumes of tetrahydrofuran and 940 volumes of the ethanol (95 per cent) and add 5.0 ml of O. 01 M hydrochloric
solution prepared by dissolving 1.1 g of sodium acid. Titrate with 0.1 M ethanolic sodium hydroxide,
octanesulphonate in 1000 ml of water. Adjust to pH 2.0 determining the end-point potentiometrically (2.4.25). Carry
with a 50 per cent v/v solution of orthophosphoric acid, out a blank titration.
B. a mixture of 40 volumes of
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
tetrahydrofuran , 170 volumes of acetonitrile and 790
0.03638 g ofC19H22ClN04.
volumes of the solution prepared by dissolving 1.1 g of
sodium octanesulphonate in 1000 ml of wate/: Adjust Storage. Store protected from light.

1746

www.webofpharma.com
IP 2010 NALOXONE INJECTION

Naloxone Injection the combined extracts over anhydrous sodium sulphate, filter,
evaporate the filtrate to dryness and dissolve the residue in 1
Naloxone Injection is a sterile solution of Naloxone ml of methanol.
Hydrochloride in Water for Injections.
Reference solution. Dilute 1 ml of the test solution to 200 ml
Naloxone Injection contains not less than 95.0 per cent and with methanol.
not more than 105.0 per cent ofthe stated amount ofnaloxone
hydrochloride, CI9H2IN04,HCI. Apply to the plate 20 Jll of each solution. Allow the mobile
phase to rise 10 cm, protecting the plate from light. After
Usual strength. 400 Jlg per ml. development, dry the plate in a cunent of air, spray with a
freshly prepared 0.5 per cent w/v solution of potassium
Identification
hexacyanoferrate(III) in iron(IIl) chloride solution and
A. In the Assay, the principal peak in the chromatogram examine in daylight. Any secondary spot in the chromatogram
obtained with the test solution conesponds to the peak in the obtained with the test solution is not more intense than the
chromatogram obtained with the reference solution. spot in the chromatogram obtained with the reference solution
(0.5 per cent). Ignore any spot remaining on the line of
B. Detennine by thin-layer chromatography (2.4.17), coating
application.
the plate with silica gel G
Bacterial endotoxins (2.2.3). Not more than 70 ill perml ofthe
Mobile phase. A mixture of 5 volumes of methanol and 95
injection, diluted ifnecessary, with water BETto give a solution
volumes ofthe upper layer from a mixture of60 ml ofMammonia
containing 0.04 per cent w/v of anhydrous Naloxone
and 100 ml of butan-I-ol.
Hydrochloride.
Test solution. Add 1 mlofammoniabufferpH 1O.0toavolume
Other tests. Complies with the tests stated under Parenteral
ofthe injection containing the equivalent of2 mg ofanhydrous
Preparations (Injections).
naloxone hydrochloride, extract with three 20 ml quantities of
a mixture of 1 volume of propan-2-o1 and 3 volumes of Assay. Detennine by liquid chromatography (2.4.14).
chloroform, illy the combined extracts over anhydrous sodium
Solvent mixture. 0.1 volume of orthophosphoric acid, 45
sulphate, filter, evaporate the filtrate to illyness and dissolve
volumes of methanol and 55 volumes of water.
the residue in 1 ml of methanol. Dilute 1 ml ofthis solution to
20 ml with methanol. Test solution. Dilute the injection' equivalent to 0.001 per cent
w/v ofNaloxone Hydrochloride with the solvent mixture.
Reference solution. A 0.01 per cent w/v solution of naloxone
hydrochloride RS in methanol. Reference solution (a). A 0.001 per cent w/v solution of
naloxone hydrochloride RS in the solvent mixture.
Apply to the plate 20 Jll of each solution. Allow the mobile
phase to rise 10 cm, protecting the plate from light. After Reference solution (b). A 0.001 per cent w/v solution of
development, illy the plate in a cunent of air, spray with a naloxone hydrochloride RS and 0.0005 per cent w/v solution
freshly prepared 0.5 per cent w/v solution of potassium of noroxymorphone in the solvent mixture.
hexacyanoferrate (Ill) in iron(III) chloride solution and
Chromatographic system
examine in daylight. The principal spot in the chromatogram
- a stainless steel column 25 cm x 4.6 mm, packed with
obtained with the test solution corresponds to that in the
end-capped octadecylsilane bonded to porous silica (5
chromatogram obtained with the reference solution.
to 10 Jlm)(such as Zorbax C18, 7 to 8 Jlm),
Tests mobile phase: a solution containing 0.068 per cent w/v
of sodium octanesulphonate and 0.1 per cent w/v of
pH (2.4.24).3.0 to 4.5. sodium chloride in the solvent mixture,
Related substances. Detennine by thin-layer chromatography - flow rate. 1 ml per minute,
(2.4.17), coating the plate with silica gel G - spectrophotometer set at 229 nm,
- injection volume. 20 Jll.
Mobile phase. A mixture of 5 volumes of methanol and 95
volumes of the upper layer from a mixture of 60 ml of 2 M Inject reference solution (b). The test is not valid unless the
ammonia and 100 ml of butan-I-of. resolution between the peaks due to naloxone and
noroxymorphone in the chromatogram obtained with reference
Test solution. Transfer a volume of the injection containing solution (b) is not less than 1.3.
about 2 mg of Naloxone Hydrochloride in 1 ml of ammonia
bufferpH I 0.0, extract with three 20 ml quantities ofa mixture Injection reference solution (a) and the test solution.
of 1 volume ofpropan-2-o1 and 3 volumes of chloroform, dry Calculate the content ofCI9H2IN04,HCl in the injection.

1747

www.webofpharma.com
NALTREXONE HYDROCHLORIDE IP 2010

Storage. Store protected from light. Specific optical rotation (2.4.22). - 187° to - 195°, determined
in a 2.0 per cent w/v solution in water.
Labelling. The label states the quantity ofactive ingredient in
terms of the equivalent amount of anhydrous naloxone Related substances. Determine by liquid chromatography
hydrochloride.When naloxone is prescribed for neonatal use, (2.4.14).
Neonatal Naloxone Injection (containing the equivalent of20 Test solution. Dissolve 20 mg of the substance under
micrograms per ml ofanhydrous naloxone hydrochloride) shall examination in 10 ml of 0.1 M hydrochloric acid.
be dispensed.
Reference solution (a). Dissolve 5 mg of 17-but-3-enyl-4,5 a-
epoxy-3,14-dihydroxymorphinan-6-one RS (naltrexone
impurity C RS) in 2.5 ml of 0.1 M hydrochloric acid.
Naltrexone Hydrochloride Reference solution (b). Dilute 1.0 ml of the test solution and
1.0 ml of reference solution (a) to 100 ml with 0.1 M
hydrochloric acid. Dilute 1.0 ml ofthis solution to 10 ml with
0.1 M hydrochloric acid.
Chromatographic system
, Hel - a stainless steel column 15 cm x 4.6 mm, packed with
.........._N~ octadecylsilane bonded to porous silica (5 Ilm),
- column temperature. 40°,
- mobile phase: A. a 0.11 per cent w/v solution ofsodium
octanesulphonate, adjust the pH to 2.3 with
Mol. Wt. 377.9 orthophosphoric acid,
Naltrexone Hydrochloride is 17-(cyclopropylmethyl)-4,5a- B. acetonitrile,
epoxy-3, 14-dihydroxymorphinan-6-one hydrochloride. - a linear gradient programme using the conditions given
below,
Naltrexone Hydrochloride contains not less than 98.0 per cent - flow rate. 1.2 ml per minute,
and not more than 102.0 per cent of CzoHz4ClN04, calculated - spectrophotometer set at 230 nm,
on the anhydrous and ethanol free basis. injection volume. 10 Ill.
Category. Antidote for opioids poisoning. Time Mobile phase A Mobile phase B
Description. A white or almost white powder, very (in min.) (per cent v/v) (per cent v/v)
hygroscopic. 0-45 9H55 1~5

45-47 55~90 45~10


Identification
47-55 ~ 10
A. Dissolve 20 mg ofthe substance under examination in 5 ml Inject reference solution (b). The test is not valid unless the
of water and make allmline with dilute ammonia. Shake with resolution between the peaks due to naltrexone and naltrexone
10 ml of dichloromethane, separate the organic layer and impurity C is not less than 2.0. The relative retention time with
evaporate the solvent. On the residue, determine by infrared reference to naltrexone for 17-formyl-4,5a-epoxy-3,14-
absorption spectrophotometry (2.4.6). Compare the spectrum dihydroxymorphinan-6-one (naltrexone impurity A) is about
with that obtained with naltrexone hydrochloride RS or with 0.4, for 4,5a-epoxy-3,14-dihydroxymorphinan-6-one
the reference spectrum of naltrexone hydrochloride. (noroxymorphone) (naltrexone impurity B) is about 0.7, For
B. Gives reaction A ofchlorides (2.3.1). 17-(cyclopropylmethyl)-4,5a-epoxy-3, lOa, 14-
trihydroxymorphinan-6-one (naltrexone impurity F) is about
Tests 0.8, for 17-(cyclopropylmethyl)-4,5a-epoxy-3,10~,14-
trihydroxymorphinan-6-one (naltrexone impurity G) is about
Appearance of solution. A2.0 per cent w/v solution in carbon-
0.9, for 17-but-3-enyl-4,5a-epoxy-3,14-dihydroxymorphinan-
dioxide free water is clear (2.4.1) and not more intensely
6-one (naltrexone impurity C) is about 1.05,for l7-butyl-4,5a-
coloured than reference solution YS5 or BS5 (2.4.1).
epoxy-3,14-dihydroxymorphinan-6-one (naltrexone impurity H)
Acidity or alkalinity. To 10 ml of2.0 per cent w/v solution in is about 1.1, for 17-(cyclopropylmethyl)-4,5a-:epoxy-3,14-
carbon-dioxidefree water, add 0.05 ml of methyl redsolution. dihydroxymorphinan-6,1O-dione (naltrexone impurity I) is
Not more than 0.2 ml of 0.02 M sodium hydroxide or 0.02. M about 1.2, for 7-(cyclopropylmethyl)-4,5a-epoxy-14-hydroxy-
hydrochloric acid is required to change the colour of the 3-methoxymorphinan-6-one (naltrexone impurity 1) is about
indicator. 1.3, for 17,17'-bis(cyclopropylmethyl)-4,5a:4',5'-a-diepoxy-

1748

www.webofpharma.com
IP 2010 NALTREXONE TABLETS

3,3' ,14,14' -tetrahydroxy-2,2' -bimorphinanyl-6,6' -dione Usual strength. 50 mg.


(pseudonaltrexone) (naltrexone impurity D) is about 1.4, for3-
(cyclopropylmethoxy)-17-(cyclopropylmethyl)-4,5(X-epoxy~ 14- Identification
hydroxymorphinan-6-one (naltrexone impurity E) is about 1.7.
In the Assay, the principal peak in the chromatogram obtained
Inject reference solution (b) and the test solution. In the with the test solution corresponds to the peak in the
chromatogram obtained with the test solution the area any chromatogram obtained with the reference solution.
peak corresponding to naltrexone impurity C, D, E, F and G is
not more than twice the area of the principal peak in the Tests
chromatogram obtained with reference solution (b) (0.2 per
cent); the area ofany peak corresponding to naltrexone impurity Dissolution (2.5.2).
A, B, H, I and J is not more than the area ofthe principal peak Apparatus No.1,
in the chromatogram obtainedwith reference solution (b) (0.1 Medium. 900 ml of water,
per cent); the area of any other secondary peak is not more Speed and time. 50 rpm and 60 minutes.
than the area of the principal peak in the chromatogram
obtained with reference solution (b)(0.1 per cent). The sum of Withdraw a suitable volume of the medium and fllter.
the areas of all secondary peaks is not more than 10 times the Detennine by liquid chromatography (2.4.14).
area ofthe principal peak in the chromatogram obtained with
reference solution (b) (l.0 per cent). Ignore any peak with an Test solution. Use the flltrate.
area less than 0.5 times the area of the principal peak in the Reference solution. A solution of naltrexone hydrochloride
chromatogram obtained with reference solution (b) (0.05 per RS in water to obtain the same concentration as given in the
cent). test solution.
Ethanol (2.3.45). Not more than 3.0 per cent v/v, determined by Chromatographic system
Method I using the following solutions. - a stainless steel column 15 cm x 3.9 mm, packed with
Test solution. Dissolve 0.25 g of the substance under octadecylsilane bonded to porous silica (5 Ilm),
examination in 10 ml of wate/: - column temperature. 45°,
- mobile phase: a mixture of600 volumes of0.05 M buffer
Reference solution. Dilute 0.75 g of anhydrous ethanol to solution prepared by dissolving 7 g of monobasic
1000 ml with water. sodium phosphate in 1000 ml of water, add 1.1 g of
Sulphated ash (2.3.18). Not more than 0.1 per cent. . sodium 1-octane sulphonate monohydrate and 400 ml
of methanol, adjust the pH to 6.7 with dilute sodium
Water (2.3.43). Not more than 10.0 per cent, determined on
hydroxide,
O.2g.
- flow rate. 1 ml per minute,
Assay. Weigh accurately about 0.2 g of the substance under - spectrophotometer set at 280 nm,
examination, dissolve in 60 ml of ethanol (95 per cent) and ~ injection volume. 100 Ill.
add 1.0 ml of 0.1 M hydrochloric acid. Titrate with 0.1 M
Inject the reference solution. The test is not valid unless the
sodium hydroxide, determining the end-point
relative standard deviation for replicate injections is not more
potentiometrically (2.4.25). The curve shows 3 points of
than 2.0.
inflexion. Read the volume added between the flrst 2 points of
inflexion. Inject the reference solution and the test solution.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03779 g of Calculate the content of CzoHz3N04.HCl in the tablets.
CzOHZ4ClN04.
D. Not less than 80 per cent of the stated amount of
Storage. Store protected from light and moisture. CzOHZ3N04.HCl.
Uniformity of content.-Comply with the test stated under
Tablets.
Naltrexone Tablets Determine by liquid chromatography (2.4.14), as described
under Assay, using the following solution as the test solution.
Naltrexone Hydrochloride Tablets
Test solution. Disperse 1 tablet in 100 ml of 0.1 M
Naltrexone Tablets contain not less than 90.0 per cent and not
orthophosphoric acid.
more than 110.0 per cent of the stated amount ofnaltrexone
hydrochloride, C ZO H23N04.HCl. Calculate the content ofCzoHz3N04.HCl in the tablets.

1749

www.webofpharma.com
NALTREXONE TABLETS IP 2010

Other tests. Comply with the tests stated under Tablets. Nandrolone Decanoate
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh and powder 20 tablets. Shake a quantity
o
of powdered tablets containing about 250 mg ofNaltrexone H3 C O)lCH 2 (CH 2 hCH 3
with 80 ml of 0.1 M orthophosphoric acid, sonicate for 30
minutes and dilute to 100 ml with the same solvent, filter.
Reference solution (a). Dissolve 22.5 mg of naltrexone RS in
1.5 ml of methanol and 0.6 ml of 0.1 M hydrochloric acid.
Dilute to 10 ml with 0.1 M orthophosphoric acid. o
Reference solution (b). Dissolve about 3 mg of N-(3-butenyl)- CZ8~03 Mol. Wt. 428.7
noroxymorphone hydrochloride RS (naltrexone impurity A
Nandrolone Decanoate is 3-oxo-4-estren-1713-yl decanoate.
RS) in 3.0 ml of methanol and dilute to 10 ml with 0.1 M
orthophosphoric acid. To 0.5 ml ofthis solution, add 5.0 ml of Nandrolone Decanoate contains not less than 97.0 per cent
reference solution (a) and dilute to 10 ml with 0.1 M and not more than 103.0 per cent of CZ8H4403, calculated on
orthophosphoric acid. the dried basis.
Chromatographic system Category. Anabolic steroid.
- a stainless steel column 15 cm x 3.9 mm packed with Dose. By intramuscular injection, 25 to 50 mg, every 3 weeks.
octadecylsilane bonded to porous silica (5 ).1m),
column temperature. 45°, Description. A white to creamy-offwhite, crystalline powder;
mobile phase: A. dissolve about 1.08 g of sodium 1- odour, faint and characteristic.
octanesulphonate and 23.8 g of sodium acetate in 800 Identification
ml of water. Add 1.0 ml of triethylamine and 200 mlof
methanol, adjust the pH to 6.5 with glacial acetic acid, A. Determine by infrared absorption spectrophotometry (2.4.6).
B. dissolve about 1.08 g sodium 1- Compare the spectrum with that obtained with nandrolone
octanesulphonate and 23.8 g sodium acetate in 400 ml decanoate RS or with the reference spectrum of nandrolone
of water. Add 1.0 ml of triethylamine and 600 ml of decanoate.
methanol, adjust the pH to 6.5 with glacial acetic acid, B. When examined in the range 230 nm to 360 nm (2.4.7), a
a linear gradient programme using the conditions given 0.001 per cent w/v solution in ethanol (95 per cent) shows an
below, absorption maximum only at about 239 nm; absorbance at
flow rate. 1 ml per minute, about 239 nm, about 0.41.
spectrophotometer set at 280 nm,
injection volume. 20 ).11. C. Dissolve 25 mg in 1 ml of methanol, add 2 ml of
semicarbazide acetate solution, heat under a reflux condenser
Time Mobile phase A Mobile phase B
for 30 minutes and cool; the precipitate, after recrystallisation
(min.) (per cent v/v) (per cent v/v)
from ethanol (95 per cent), melts at about 175° (2.4.21).
o 100 o
Tests
1-35 100-70 0-7100
35-36 0-7100 100-70 Specific optical rotation (2.4.22). +32.0 ° to +36.0°, determined
in a 2.0 per cent w/v solution in dioxan.
Inject reference solution (b). The test is not valid unless the
resolution between the peaks due to naltrexone and naltrexone Related substances. Determine by thin-layer chromatography
impurity A is not less than 2.0, the tailing factor is not more (2.4.17), coating the plate with silica gel GF254.
than 1.4, and the relative standard deviation for replicate Mobile phase. A mixture of 70 volumes of heptane and
injections is not more than 2.0 per cent. The relative retention 30 volumes of acetone.
time with reference to naltrexone for noroxymorphone is about Test solution. Dissolve 0.1 g of the substance under
0.55, for 10-hydroxynaltrexone is about 0.7, for naltrexone examination in 10 ml of chloroform.
impurity A is about 1.26, for 2,2a-bisnaltrexone is about 1.80
and for 10-ketonaltrexoneis about 1.99. Reference solution (a). A 0.005 per cent w/v solution of the
substance under examination in chloroform.
Inject reference solution (a) and the test solution.
Reference solution (b). A 0.01 per cent w/v solution of
Calculate the content ofCzoHz3N04.HCl in the Tablets. nandrolone RS in chloroform.

1750

www.webofpharma.com
IP 2010 NANDROLONE PHENYLPROPIONATE

Apply to the plate 5 f.Ll of each solution. After development, Tests


dry the plate in air and examine in ultraviolet light at 254 run. In
Other tests. Complies with the tests stated under Parenteral
the chromatogram obtained with the test solution any spot
Preparations (Injections).
corresponding to nandrolone is not more intense than the
spot in the chromatogram obtained with reference solution Assay. To an accurately measured volume containing about
(b) and any other secondary spot is not more intense than the 0.1 g ofNandrolone Decanoate add sufficient chloroform to
spot in the chromatogram obtained with reference solution (a). produce 100.0 mL Dilute 3.0 ml ofthe solution to 50.0 ml with
chloroform. To 5.0 ml of this solution add 10 ml of isoniazid
Sulphated ash (2.3.18). Not more than 0.1 percent.
solution and sufficient methanol to produce 20.0 ml. Allow to
Loss on drying (2.4.19). Not more than 0.5 per cent, determined stand for 45 minutes and measure the absorbance of the
on 1.0 g by drying over phosphorus pentoxide at a pressure resulting solution at the maximum at about 380 nm (2.4.7),
not exceeding 0.7 kPa for 4 hours. using as the blank 5 ml of chloroform treated in the same
Assay. Weigh accurately about 10 mg and dissolve in sufficient manner. Calculate the content ofC2sH4403 from the absorbance
ethanol (95 per cent) to produce 100.0 mL Dilute 5.0 ml to obtained by repeating the operation using a suitable quantity
50.0 ml with ethanol (95 per cent) and measure the absorbance of nandrolone R8.
ofthe resulting solution at the maximwn at about 239 run (2.4.7). 1 mg ofCIsH2602 is equivalent to 1.562 mg ofC2sH4403.
Calculate the content of C 2s H44 0 3 taking 407 as the specific
Storage. Store protected from light.
absorbance at 239 nm.
Storage. Store protected from light and moisture.

Nandrolone Phenylpropionate
Nandrolone Phenpropionate
Nandrolone Decanoate Injection
Nandrolone Decanoate Injection is a sterile solution of
Nandrolone Decanoate in Ethyl Oleate or other suitable ester,
in a suitable fixed oil or in any mixture ofthese.
Nandrolone Decanoate Injection contains not less than 90.0
per cent and not more than 110.0 per cent ofthe stated amount
ofnandrolone decanoate, C2sH4403.
Usual strength. 25 mg per mL

Identification Mol.Wt. 406.6

Detennine by thin-layer chromatography (2.4.17), coating the Nandrolone Phenylpropionate is 3-oxo-4-estren-17~-yI3­


plate with silica gel GF254. phenylpropionate.

Mobile phase. A mixture of 70 volumes of heptane and Nandrolone Phenylpropionate contains not less than 97.0 per
30 volumes of acetone. cent and not more than 103.0 per cent ofC27H3403, calculated
on the dried basis.
Test solution. Dilute a suitable volume of the injection with
carbon tetrachloride to give a solution containing 0.5 per- Category. Anabolic steroid.
cent w/v solution of Nandrolone Decanoate. Dose. By deep intramuscular injection, 25 to 50 mg weeldy.
Reference solution. A 0.5 per cent w/v solution ofnandrolone Description. A white to creamy-white, crystalline powder;
decanoate RS in carbon tetrachloride. odour, characteristic.
Apply to the plate 5 f.LI of each solution. After development, Identification
dry the plate in air until the odour of solvent is no longer
detectable, spray with a 10 per cent v/v solution of sulphuric A. Detennine by infrared absorption spectrophotometry (2.4.6).
acid in ethanol (95 per cent), heat at 105° for 30 minutes and Compare the spectrum with that obtained with nandrolone
examine in ultraviolet light at 365 run. The principal spot in the phenylpropionate RS or with the reference spectrum of
chromatogram obtained with the test solution corresponds to nandrolone phenylpropionate.
that in the chromatogram obtained with the reference solution. B. When examined in the range 230 run to 360 nm (2.4.7), a
Ignore any subsidiary spots due to the vehicle. 0.001 per cent w/v solution in ethanol (95 per cent) shows an

1751

www.webofpharma.com
NANDROLONE PHENYLPROPIONATE IP 2010

absorption maximum only at about 240 nm; absorbance at Nandrolone Phenylpropionate Injection contains not less than
about 240 nm, about 0.43. 92.5 per cent and not more than 107.5 per cent of the stated
C. Dissolve 25 mg in I ml of methanol, add 2 ml of amount of nandrolone phenylpropionate, C27H3403'
semicarbazide acetate solution, heat under a reflux condenser Usual strengths. 25 mg per ml; 50 m per ml.
for 30 minutes and cool; the precipitate, after recrystallisation
f
from ethanol (95 per cent) melts at about 182° (2.4.21). Identification
Tests Dissolve a volume of the injection containing 50 mg of
Nandrolone Phenylpropionate in 8 ml of light petroleum
Specific optical rotation (2.4.22). +48.0 ° to +51.0°, determined (40° to 60°) and extract with three 8-ml quantities ofa mixture
in a 1.0 per cent w/v solution in dioxan. of 7 volumes of glacial acetic acid and 3 volumes of water.
Related substances. Determine by thin-layer chromatography Wash the combined extracts with 10 ml of light petroleum
(2.4.17), coating the plate with silica gel GF254. (40° to 60°), dilute with water until the solution becomes
turbid, allow to stand for 2 hours in ice and filter. The precipitate,
Mobile phase. A mixture of 70 volumes of heptane and
after washing with water and drying over phosphorus
30 volumes of acetone.
pentoxide at a pressure not exceeding 0.7 kPa, complies with
Test solution. Dissolve 0.1 g of the substance under the following test.
examination in 10 ml ofchloroform.
Determine by thin-layer chromatography (2.4.17), using a
Reference solution (a). A 0.005 per cent w/v solution of the silica gel GF254 precoated plate the surface of which has
substance under examination in chloroform. been modified by chemically-bonded octadecylsilyl groups.
Reference solution (b). A 0.01 per cent w/v solution of Mobile phase. A mixture of20 volumes ofwater, 40 volumes
nandrolone RS in chloroform. of acetonitrile and 60 volumes ofpropan-2-01.
Apply to the plate 5 Jll of each solution. After development, Test solution. A 0.5 per cent w/v solution of the dried
dry the plate in air and examine in ultraviolet light at 254 nm. In precipitate in chloroform.
the chromatogram obtained with the test solution any spot
Reference solution (a). A 0.5 per cent w/v solution of
corresponding to nandrolone is not more intense than the
nandrolone phenylpropionate RS in chloroform.
spot in the chromatogram obtained with reference solution
(b) and any other secondary spot is not more intense than Reference solution (b). A mixture of equal volumes ofthe test
the spot in the chromatogram obtained with reference solution and the reference solution.
solution (a). Apply to the plate 5 Jll of each solution. After development,
Sulphated ash (2.3.18). Not more than 0.1 per cent. dry the plate in air until the solvent has evaporated and heat it
at 100° for 10 minutes. Allow to cool and examine in ultraviolet
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
light at 254 nm. The principal spot in the chromatogram
on 1.0 g by drying over phosphorus pentoxide at a pressure
obtained with the test solution corresponds to that in the
not exceeding 0.7 kPa for 4 hours.
chromatogram obtained with reference solution (a). The
Assay. Weigh accurately about 10 mg, dissolve in sufficient principal spot in the chromatogram obtained with reference
ethanol to produce 100.0 ml, dilute 5.0 ml to 50.0 ml with ethanol solution (b) appears as a single spot.
and measure the absorbance of the resulting solution at the
maximum at about 240 nm (2.4.7). Calculate the content of Tests
C27H3403 taking 430 as the specific absorbance at 240 nm.
Other tests. Complies with the tests stated under Parenteral
Storage. Store protected from light. Preparations (Injections).
Assay. To an accurately measured volume containing about
0.1 g of Nandrolone Phenylpropionate add sufficient
chloroform to produce 100.0 ml. Dilute 3.0 ml ofthis solution
Nandrolone Pbenylpropionate to 50.0 ml with chloroform. To 5.0 ml ofthe resulting solution
Injection add 10 ml of isoniazid solution and sufficient methanol to
produce 20.0 ml. Allow to stand for 45 minutes and measure
Nandrolone Phenylpropionate Injection is a sterile solution the absorbance of the solution at the maximum at about
of Nandrolone Phenylpropionate in Ethyl Oleate or other 380 nm (2.4.7), using as blank 5 ml ofchloroform treated in the
suitable ester, in a suitable fixed oil or in a mixture ofthese. same manner. Calculate the content of C27H3403 from the

1752

www.webofpharma.com
IP 2010 NAPHAZOLINE NITRATE

absorbance obtained from a 0.006 per cent w/v solution of Tests


nandrolone phenylpropionate RS treated in the same manner.
Appearance of solution. A 1.0 per cent w/v solution in carbon
Storage. Store protected from light. dioxide-free water is clear (2.4.1) and colourless (2.4.1).
Labelling. The label states that the preparation is for
pH (2.4.24).5.0 to 6.5, determined in a 1.0 per cent w/v solution.
intramuscular injection only.
Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Dissolve 50 mg of the substance under
N aphazoline Nitrate examination in 100 ml ofthe mobile phase.
Reference solution (a). Dissolve 5 mg of 1-naphthylacetic
acid in the mobile phase, add 5 ml of the test solution and
dilute to 100 ml with the mobile phase.
Reference solution (b). Dissolve 5 mg ofnaphazoline impurity
A RS in 100 ml ofthe mobile phase. Dilute 5.0 ml ofthis solution
to 100.0 ml with the mobile phase.
Reference solution (c). Dilute 2.0 ml of the test solution to
Mol. Wt. 273.3 10.0 ml with the mobile phase. Dilute 1.0 ml ofthis solution to
Naphazoline Nitrate is 2-(l-napthylmethyl)-2-imidazoline 100.0 ml with the mobile phase.
nitrate. Chromatographic system
Naphazoline Nitrate contains not less than 99.0 per cent and - a stainless steel column 25 cm x 4.0 mm, packed with
not more than 101.0 per cent of C4HI4N2,HN03 calculated on octadecylsilane bonded to porous silica (4 Jlm),
the dried basis. mobile phase: dissolve 1.1 g of sodium
octanesulphonate in a mixture of 5 volumes ofglacial
Category. Sympathomimetic.
acetic acid, 300 volumes ofacetonitrile and 700 volumes
Description. A white or almost white crystalline powder. ofwater,
flow rate. 1 ml per minute,
Identification spectrophotometer set at 280 nm,
Test A may be omitted iftests B, C andD are carried out. Tests injection volume. 20 Jll.
Band C may be omitted if tests A and D are carried out. Inject reference solution (a). The test is not valid unless the
A. Determine by infrared absorption spectrophotometry (2.4.6). resolution between the peaks due to naphazoline and
Compare the spectrum with that obtained with naphazoline naphazoline impurity B is not less than 5.0. The relative
nitrate RS. retention time with reference to naphazoline for
naphthylacetylethylenediamine (naphazoline impurity A) is
B. When examined in the range 230 nm to 360 nm (2.4.7), a about 0.76, for I-naphthylacetic acid (naphazoline impurity B)
0.002 per cent w/v solution in 0.01 Mhydrochloricacidshows is about 1.27, for 1-naphthylacetonitrile (naphazoline impurity
absorption maxima at about 270 nm, 280 nm, 287 nm and C) is about 2.8, for B-naphazoline (naphazoline impurity D) is
291 nm; absorbances at these maxima are about 0.43,0.50,0.35 about 1.28.
and 0.34 respectively.
Inject the test solution, reference solution (b) and (c). Run the
C. Dissolve about 0.5 mg in 1 ml ofmethanol, add 0.5 ml ofa
chromatogram 3 times the retention time ofthe principal peak.
freshly prepared 5 per cent w/v solution of sodium
In the chromatogram obtained with the test solution the area
nitroprusside and 0.5 ml ofa 2 per cent w/v solution ofsodium
ofthe peak due to naphazoline impurity A is not more than the
.hydroxide, allow to stand for 10 minutes and add 1 ml of a
area ofthe principal peak in the chromatogram obtained with
8 per cent w/v solution ofsodium bicarbonate; a violet colour
reference solution (b) (0.5 per cent). The area of any other
is produced.
secondary peak is not more than 0.5 times the area of the
D. Dissolve about 10 mg in 5 ml ofwater, add 0.2 g ofmagnesium principal peak in the chromatogram obtained with reference
oxide, shake mechanically for 30 minutes, add 10 ml of solution (c) (0.1 per cent). The sum ofall the secondary peaks
chloroform and shake vigorously. Allow to stand, separate is not more than 5 times the area of the principle peak in the
the chloroform layer, filter and evaporate the aqueous layer to chromatogram obtained with reference solution (c) (1.0 per
dryness. The residue gives reaction A for nitrates (2.3.1). cent), Ignore any peak with an area less than 0.25 times the

1753

www.webofpharma.com
NAPHAZOLINE· NITRATE IP 2010

area ofthe principal peak in the chromatogram obtained with Naproxen contains not less than 99.0 per cent and not more
reference solution (c) (0.05 per cent). than 101.0 per cent OfC14H1403, calculatedon the dried basis.
Naphthylacetylethylenediamine. D.etermine by thin-layer Category. Nonsteroidal antiinflammatory.
chromatography (2.4.17), coating the plate with silica gel G.
Description. A white oralmost white crystalline powder.
Mobile phase. A mixture of 100 volumes of methanol and
1.5 volumes of strong ammonia solution. Identification
Test solution. Dissolve 0.2 g of the substance under Test A may be omitted iftests Band C are carried out. Tests B
examination in 10 ml of methanOL and C may be omitted if test A is carried out.
Reference solution. A solution containing 2 per cent w/vof A. Determine by infrared absorption spectrophotometry (2.4.6).
naphazoline nitrate RS and 0.01 per cent w/v of Compare the spectrum with that obtained with naproxen RS
naphthylacetylethylenediamine hydrochloride RS. or with the reference spectrum of naproxen.
Apply to the plate 10 III of each solution. After development, B. When examined in the range 230 urn to 350 nm (2.4.7), a 0.04
dry the plate at 105° for 5 minutes, spray with a 0.5 percent per cent w/v solution in methanol shows absorption maxima
w/v solution of ninhydrin in methanol and heat at 105° for at about 262 nm, 271 run, 316 urn and 331 nm. The absorbance
10 minutes. Any spot corresponding to naphthylacetyl- at the maxima are216 to 238, 219 to 241, 61 to 69 and 79 to 87
ethylenediamine hydrochloride in the chromatogram obtained respectively.
with the test solution is not more intense than the
corresponding spot in the chromatogram obtained with the C. Melting range (2.4.21). 154° to 158°.
reference solution. The test is not valid unless the
chromatogram obtained with the reference solution shows Tests
two clearly separated spots.
Appearance of solution. A 5.0 per cent w/v solution in
Chlorides (2.3.12). 15.0 ml ofl.O per cent w/v solution in carbon methanol is clear (2.4.1) and not more intensely coloured than
dioxide-free water complies with the limit test for chlorides reference solution BYS6 (2.4.1).
(375 ppm).
Specific optical rotation (2.4.22). + 59° to + 62°, determined in
Sulphated ash (2.3.18). Not more than 0.1 per cent. a 2.0 per cent w/v solution in ethanol (95 per cent).
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined Enantiomeric purity. Determine by liquid chromatography
on 1.0 g by drying in an oven at 105° for 3 hours. (2.4.14).
Assay. Weigh accurately about 0.2 g, dissolve in 30 ml of NOTE-Protect the solutions from light.
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
acid, detenriining the end-point potentiometrically (2.4.25). Test solution. Dissolve 25 mg of the substance under
Carry out a blank titration. examination in 50 ml of tetrahydrojitran.Dilute 2,0 ml ofthis
.solution to 20 ml with the mobile phase.
I ml of 0.1 M perchloric acid is equivalent to 0.02733 g of
C4HI4N2,HN03. Reference solution (a). Dilute 2.5 ml ofthe test solution to 100
ml with the mobile phase.
Storage. Store protected from light.
Reference solution (b). Dissolve 5 mg of racemic naproxen
RS in 10 ml of tetrahydrofitran, dilute to 100 ml with the mobile
phase.
Naproxen Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
silica gel1t-acceptorht-donor for chiral separations (5
JllTI),
- column temperature. 25°,
- mobile phase: a mixture of 5 volumes of glacial acetic
acid, 50 volumes of acetonitrile, 100 volumes of 2-
propanol and 845 volumes of hexane,
CI~1403 Mol. Wt. 230.3
flow rate. 2 ml per minute,
Naproxen is (2S)-2-(6-methoxynaphthalen-2-yl)propionic spectrophotometer set at 263 nm,
acid. - injection volume. 20 Ill.

1754

www.webofpharma.com
IP 2010 NAPROXEN ORAL SUSPENSION

Inject reference solution (b). The test is not valid unless the solution (a) (0.1 per cent). The sum ofthe areas ofall impurities
resolution between the peaks due to (2R)-2-(6- is not more than 3 times the area of the principal peak in the
methoxynaphthalen-2-yl) propanoic acid (naproxen impurity chromatogram obtained with reference solution (a) (0.3 per
G) ((R)-enantiomer) and naproxen is not less than 3.0. cent). Ignore any peak with an area less than 0.5 times the area
of the principal peak in the chromatogram obtained with
Inject reference solution (a) and the test solution. Run the
reference solution (a) (0.05 per cent).
chromatogram 1.5 times the retention time of the principal
peak. The area of the peak due to naproxen impurity G is not Heavy metals (2.3.13). 1.0 g complies with the limit test for
more than the area ofthe principal peak in the chromatogram heavy metals, Method B (20 ppm).
obtained with reference solution (a) (2.5 per cent). Sulphated ash (2.3.18). Not more than 0.1 per cent.
Related substances. Determine by liquid chromatography Loss on drying (2.4.19). Not more than 0.5 per cent, determined
(2.4.14). on 1.0 g by drying in an oven at 105° for 3 hours.
NOTE-Protect the solutions from light. Assay. Weigh accurately about 0.2 g and dissolve in a mixture
Test solution. Dissolve 12 mg of the substance under of25 ml of water and 75 ml of methanol. Titrate with 0.1 M
examination in 20 ml ofthe mobile phase. sodium hydroxide using 1 ml of phenolphthalein solution as
indicator.
Reference solution (a). Dilute 1.0 ml of the test solution to 50
ml with the mobile phase. Dilute 1.0 ml ofthis solution to 20.0 1 ml of 0.1 M sodium hydroxide is equivalent to 0.02303 g of
ml with the mobile phase. C,JII403'
Reference solution (b). Dissolve 6 mg of 2-bromo-6- Storage. Store protected from light.
methoxynaphthalene RS (Naproxen impurity N RS), 6 mg of
1-(6-methoxynaphthalen-2-yl)ethanone RS( Naproxen
impurity L RS) and 6 mg of (1RS)-1-(6- methoxynaphthalen-
2-ylJethanol RS (Naproxen impurity K RS) in 10 ml of Naproxen Oral Suspension
acetonitrile. To 1.0 ml of this solution, add 1.0 ml ofthe test
solution and dilute to 50 ml with the mobile phase. Dilute 1.0 Naproxen Oral Suspension is an aqueous suspension of
ml ofthis solution to 20 ml with the mobile phase. Naproxen in a suitable flavoured vehicle.

Chromatographic system Naproxen Oral Suspension contains not less than 90.0 per
- a stainless steel column 10 cm x 4.0 mm, packed with cent and not more than 110.0 per cent ofthe stated amountof
octadecylsilane bonded to porous silica (3 Ilm), naproxen, C14H1403.
- column temperature. 50°, Usual strength. 25 mg per ml.
- mobile phase: a mixture of 42 volumes of acetonitrile
and 58 volumes of a 0.136 per cent w/v solution of Identification
potassium dihydrogen phosphate adjust the pH to 2.0
Evaporate 50 ml ofsolution A obtained in the Assay, to dryness
with orthophosphoric acid,
using a rotary evaporator. The residue complies with the
- flow rate. 1.5 ml per minute,
following tests.
- spectrophotometer set at 230 nm,
- injection volume. 20 Ill. A. Determine by infrared absorption spectrophotometry
(2.4.6).Compare the spectrum with that obtained with naproxen
Inject reference solution (b). The test is not valid unless the
RS or with the reference spectrum ofnaproxen.
resolution between the peaks due to naproxen impurity K and
naproxen is not less than 2.2. The relative retention time with B. When examined in the range 230 nm to 350 nm (2.4.7), a
reference to naproxen for naproxen impurity K is about 0.9, for 0.004 per cent w/v solution in methanol exhibits maxima at 262
naproxen impurity L is about 1.4 and naproxen impurity N is nm,27l nm,316nmand331 nm.
about 5.3.
Tests
Inject reference solution (a), (b) and the test solution. Run the
chromatogram 1.5 times the retention time ofnaproxen impurity pH (2.4.24).2.1 to 4.0.
N. The area ofthe peak due to naproxen impurity L is not more
Related substances. Determine by thin-layer chromatography
than the area ofthe corresponding peak in the chromatogram
(2.4.17), coating the plate with silica gel GF254.
obtained with reference solution (b) (0.1 per cent); the area of
any other secondary peak is not more than the area of the Mobile phase. A mixture of3 volumes of glacial acetic acid,
principal peak in the chromatogram obtained with reference 9 volumes of tetrahydrojU,:an and 90 volumes of toluene.

1755

www.webofpharma.com
NAPROXEN ORAL SUSPENSION IP 2010

Test solution. Evaporate solution A obtained in the Assay to B. When examined in the range 230 nm to 350 nm (2.4.7), a
dryness on a rotary evaporator and dissolve the residue in 0.004 per cent w/v solution in methanol exhibits maxima at 262
sufficient methanol to produce a solution containing 5.0 per nm,271 nm,316nmand331 nm.
cent w/v ofnaproxen.
Reference solution. Dilute 1 ml of the test solution to 200 ml Tests
with methanol. Related substances. Determine by thin-layer chromatography
Apply to the plate 10 III of each solution. After development, (2.4.17), coating the plate \yith silica gel GF254.
dry the plate in air and examine under ultraviolet light at 254 Mobile phase. A mixture of 3 volumes of glacial acetic acid,
nm. Any secondary spot in the chromatogram obtained with 9 volumes of tetrahydrofuran and 90 volumes of toluene.
the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution (0.5 per Test solution. Weigh 20 suppositories and cut into small
cent). pieces. Dissolve a quantity of the suppositories containing
0.5 g ofNaproxen in 50 ml of 2,2,4-trimethylpentane and extract
Other tests. Complies with the tests stated under Oral Liquids. with four 25-ml quantities of methanol (80 per cent). Combine
Assay. To a quantity of the oral suspension containing 0.5 g the extracts, add 100 ml ofa2 per cent w/v solution of sodium
ofNaproxen add 20 ml of3.5 M hydrochloric acid; mix, extract chloride, extract with four 25-ml quantities of chloroform
with three 50 ml quantities of chloroform, filter each extract filtering each extract through a layer of anhydrous sodium
through anhydrous sodium sulphate, combine the filtrates sulphate on an absorbent cotton plug moistened with
and add sufficient chloroform to produce 200 ml (solution A). chloroform. Evaporate the combined filtrates to dryness using
To 5 ml of solution A add sufficient methanol to produce 250 a rotary evaporator with the aid of gentle heat. Shake the
ml and measure the absorbance of the resulting solution at residue with 10 ml of methanol, centrifuge and use the
the maximum at about 331 nm (2.4.7). supernatant liquid.
Calculate the content of Cl4Hl403 taking 81 as the specific Reference solution. Dilute1 ml of the test solution to 200 ml
absorbance at 331 nm. with methanol.
Apply to the plate 10 III of each solution. After development,
dry the plate in air and examine under ultraviolet light at 254
nm. Any secondary spot in the chromatogram obtained with
N aproxen Suppositories the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Naproxen Suppositories contain Naproxen in a suitable
Other tests. Comply with the tests stated under Suppositories.
suppository base.
Assay. Determine by liquid chromatography (2.4.14).
Naproxen Suppositories contain not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of Test solution. Disperse ten suppositories in 500 ml of methanol
naproxen, C14H1403. on a water-bath for 40 minutes with the aid of ultrasound and
by swirling the flask. Cool at 50 for 1 hour, centrifuge and use
Usual strength. 500 mg.
the clear, supernatant liquid; if the solution is still cloudy
Identification filter through glass-fibre paper (such as Whatrnan OFIe). To 5
ml ofthe filtrate add sufficient ofthe mobile phase to produce
Dissolve a quantity of the suppositories containing 0.5 g of a solution containing 0.01 per cent w/v ofnaproxen.
Naproxen in 50 ml of 2,2,4-trimethylpentane and extract with
Reference solution (a). A 0.01 per cent w/v solution of
four 25 ml quantities of methanol (80 per cent). To the
naproxen RS in the mobile phase.
combined extracts add 100 rnl of a 2 per cent w/v solution of
sodium chloride, extract with four 25 ml quantities of Reference solution (b). A 0.01 per cent w/v each ofnaproxen
chloroform, dry the combined extracts over anhydrous sodium RS and 2-naphthylacetic acid in the mobile phase.
sulphate, filter and add sufficient chloroform to produce 200 Chromatographic system
ml (solution A). Evaporate 100 ml of solution A to dryness a stainless steel column 20 cm x 4 mm, packed with end-
using a rotary evaporator. The residue complies with the capped octadecylsilane bonded to porous silica (5 Ilm)
following tests. (such as Nucleosil C18),
A. Determine by infrared absorption spectrophotometry (2.4.6). mobile phase: a mixture of 1 volume of a 0.52 per cent
Compare the spectrum with that obtained with naproxen RS w/v solution of sodium acetate, adjusted to pH 5.8
9r with the reference spectrum of naproxen. using glacial acetic acid and 1 volume of methanol,

1756

www.webofpharma.com
IP 2010 NAPROXEN SUSTAINED-RELEASE TABLETS

flow rate. 2 ml per minute, the absorbance obtained from a solution of known
spectrophotometer set at 254 nm, concentration of naproxen RS in the same medium.
- injection volume. 20 Ill. D. Not less than 80 per cent ofthe stated amount ofC14H1403.
Inject reference solution (b). The test is not valid unless the Uniformity of content. Comply with the test stated under
resolution between the peaks due to naproxen and 2- Tablets.
naphthylacetic acid in the chromatogram obtained with the
Determine by liquid chromatography (2.4.14), as described
reference solution (b) is more than 3.0.
under Assay using the following solutions.
Inject reference solution (a) and the test solution.
Test solution. Disperse 1 tablet in about 140 ml of solvent
Calculate the content of CI4HI403 in the suppositories. mixture B. Shake for 15 minutes, sonicate for 15 minutes, dilute
Storage. Store protected from light. to 200 ml with solvent mixture B, filter. Dilute 2.0 ml of the
filtrate to 50 ml with the mobile phase.
Reference solution. A 0.025 per cent wIv solution of naproxen
RS in solvent mixture A. Dilute 10 ml ofthis solution to 25 ml
Naproxen Sustained-release Tablets with solvent mixture B.
Calculate the content ofCI4HI403 in the tablet.
Naproxen Sustained-release Tablets contain not less than 90.0
per cent and not more than 110.0 per cent ofthe stated amount Other tests. Comply with the tests stated under Tablets.
ofnaproxen, C14H1403. Assay. Determine by liquid chromatography (2.4.14).
Usual strength. 375 mg. Solvent mixture A. 90 volumes of acetonitrile and 10 volumes
of water.
Identification
Solvent mixture B. 50 volumes of acetonitrile and 50 volumes
A. When examined in the range 200 TIm to 400 TIm (2.4.7), the of water.
solution used in test B of dissolution, exhibits an absorption
Test solution. Weigh and powder 20 tablets. Disperse a quantity
maximum at about 332 TIm.
of powder containing about 250 mg ofNaproxen in 70 ml of
B. In the Assay, the principal peak in the chromatogram solvent mixture B, sonicate for 15 minutes and dilute to 100.0
obtained with the test solution corresponds to that in the ml with solvent mixture B and filter. Dilute 2.0 ml ofthe filtrate
chromatogram obtained with the reference solution (b). to 50 ml with the mobile phase.
Reference solution (a). A 0.05 per cent wlv solution of
Tests
naproxen RS in solvent mixture A.
Dissolution (2.5.2). Reference solution (b). Dilute 10 ml of reference solution (a)
A. Apparatus No.1, to 50 ml with the mobile phase.
Medium. 1000 ml of0.1 M hydrochloric acid, Chromatographic system
Speed and time. 50 rpm and 2 hours. a stainless steel column 25 cm x 4.6 rom, packed with
octadecylsilane bonded to porous silica (5 11m),
Withdraw a suitable volume ofthe medium and filter. Measure
mobile phase: a mixture of 55 volumes of 1.0 per cent
the absorbance of the filtered solution, suitably diluted with
vlv solution of acetic acid and 45 volumes of
the medium if necessary, at the maximum at about 332 TIm
acetonitrile,
(2.4.7). Calculate the content ofC,4H1403in the medium from
- flow rate. 1ml per minute,
the absorbance obtained from a solution of known
spectrophotometer set at 254 nm,
concentration of naproxen RS in the same medium.
injection volume. 50 Ill.
D. Not more than 10 per cent ofthe stated amount ofC,4H,403.
Inject reference solution (b). The test is not valid unless the
B. Apparatus No.1, tailing factor ofthe principal peak is not more than 1.5 per cent
Medium. 1000 ml of 0.2 M phosphate bujferpH 6.8, and the relative standard deviation for replicate injections is
not more than 2.0 per cent.
Speed and time. 50 rpm and 45 minutes.
Inject the test solution and reference solution (b).
Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance of the filtered' solution, suitably diluted with Calculate the content of CI4HI403 in the tablet.
the medium if necessary, at the maximum at about 332 nm Storage. Store protected from moisture, at a temperature not
(2.4.7). Calculate the content ofC,4H1403 in the medium from exceeding 30°.

1757

www.webofpharma.com
NAPROXEN TABLETS IP 2010

Naproxen Tablets Other tests. Comply with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Shake a quantity of the
Naproxen Tablets contain not less than 95.0per cent and not
powder containing about 50 mg ofNaproxen with 70 ml of
more than 105.0 per cent of the stated amount ofnaproxen,
methanol for 30 minutes, add sufficient methanol to produce
C14H1403;
100 ml and filter. Dilute 10 ml of the filtrate to 50 ml with
Usual strengths. 250 mg; 500 mg. methanol and measure the absorbance at the maximum at about
331nm (2.4.7) using a 0.01 per cent wlv solution of naproxen
Identification
RS in methanol.
A. Extract a quantity of the powdered tablets containing 20
Calculate the content of CI4HI403 in the tablets.
mg ofNaproxen with 100 ml of methanol and filter. Evaporate
and dry the residue at 105°. On the residue, determine by Storage. Store protected from light.
infrared absorption spectrophotometry (2.4.6). Compare the
spectrum with that obtained with naproxen RS or with the
reference spectmm of naproxen.
Nebivolol Hydrochloride
B. When examined in the range 230 nm to 350 nm (2.4.7), a . . OH·. OH .
0.002 per cent w Iv solution in methanol exhibits maxima at 262
nm,271 nm,316nmand331 nm.
~O~~~O~ ,Hel
Tests F~ ~F
Dissolution (2.5.2). Mol. Wt. 441.9
Apparatus No.1; Nebivolol Hydrochloride is (IRS, 1'RS)-I, 1'-[(2RS,2'SR)-bis (6-
Medium. 900 ml ofa phosphate buffer prepared by dissolving flurochroman-2-yl)]-2,2'-iminodiethanol hydrochloride.
2.62 g of sodium dihydrogen orthophospate monohydrate Nebivolol Hydrochloride contains not less than 98.0 per cent
and 11.5 g of anhydrous disodium hydrogen orthophosphate and not more than 102.0 per cent of C22H2sF2N04,HCl,
in sufficient water to produce 1000 ml, adjusted to pH 7.4 with calculated on the anhydrous basis.
0.1 M sodium hydroxide or 0.1 M hydrochloric acid,
Speed and time. 50 rpm and 45 minutes. Category. Antihypertensive.

Withdraw a suitable volume ofthe medium and filter. Measure Description. A white to off-white powder.
the absorbance of the filtrate, suitably diluted with the Identification
dissolution medium ifnecessary, at the maximum at about 332
mn (2.4.7). Calculate the content ofC,4H,403, in the mediUlp Detennine by infrared absorption spectrophotometry (2.4.6).
from the absorbance obtained from a solution of known Compare the spectrum with that obtained with nebivolol
concentration of naproxen RS in the same medium. hydrochloride RS or with the reference spectrum ofnebivolol
hydrochloride.
D. Not less than 70 percent ofthe stated amount ofC14H1403.
Related substances. Determine by thin-layer chromatography Tests
(2.4.17), coating the plate with silica gel GF254.
Related substances. Determine by .liquid chromatography
Mobile phase. A mixture on volumes of glacial acetic acid, (2.4.14).
9 volumes of tetrahydrofitran and 90 volumes of toluene.
Test solution. Dissolve 30 mg of the substance under
Test solution. Shake a quantity of the powdered tablets examination in 5 ml of acetonitrile and dilute to 100.0 ml with
containing about 0.5 g ofNaproxen with 10 ml of methanol for the mobile phase.
15 minutes, centrifuge and use the supernatant liquid: Reference solution (a). Dissolve 30 mg of nebivolol
Reference solution. Dilute 1 ml of the test solution to 200 ml hydrochloride RS in 5 ml of acetonitrile and dilute to 100.0 ml
with methanol. with the mobile phase.
Apply to the plate 10 /ll of each solution. After development, Reference solution (b). Dilute 1 ml ofreference solution (a) to
dry the plate in air and examine under ultraviolet light at 254 100.0 ml with the mobile phase.
nm. Any secondary spot in the chromatogram obtained with Chromatographic system
the test solution is not more intense than the spot in the - a stainless steel column 25 cm x 4.6 mm, packed with
chromatogram obtained with the reference solution (0.5 per porous silica (5 /lm) with chemically bonded phenyl
cent). groups,

1758

www.webofpharma.com
IP 2010 NEBIVOLOL TABLETS

- mobile phase: a mixture of28 volumes of acetonitrile, Identification


72 volumes of a buffer solution prepared by dissolving
3.4 g of tetrabutyl ammonium hydrogen sulphate in In the Assay, the principal peak in the chromatogram obtained
1000 ml wate]; and 0.3 volume of diethylamine, with the test solution corresponds to the peak in the
- flow rate. 1 ml per minute, chromatogram obtained with the reference solution.
- spectrophotometer set at 220 nm,
Tests
- injection volume. 20 ,..t.l.
Inject reference solution (a). The test is not valid unless the Dissolution (2.5.2).
column efficiency in not less than 2000 theoretical plates and Apparatus No.1,
the tailing factor is not more than 2.0. Medium. 500 ml 0,1 M hydrochloric acid,
Inject the test solution and reference solution (b). In the Speed and time. 75 rpm and 30 minutes.
chromatogram obtained with the test solution, the area of any Withdraw a suitable volume ofthe medium and filter.
secondary peak is not more than 0.5 times the area ofthe peak
Detennine by liquid chromatography (2.4.14) as described
in the chromatogram obtained with reference solution (b) (0.5
under Assay using the following solutions and a flow rate of
per cent) and the sum of areas of all the secondary peaks is
not more than the area of the peak in the chromatogram 1.5 ml per minute.
obtained with reference solution (b) (1.0 per cent). Test solution. The filtrate obtained as given above.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Reference solution. A solution containing nebivolol
heavy metals, Method B (20 ppm). hydrochloride RS equivalent to 0.001 per cent w/v of
Sulphated ash (2.3 .18). Not more than 0.2 per cent. nebivolol, in the dissolution medium.
Water (2.3.43). Not more than 1.0 per cent, determined on 0.3 g. D. Not less than 70 per cent of the stated amount of
C22H2sF2N04.
Assay. Determine by liquid chromatography (2.4.14).
Uniformity of content. Comply with the test stated under
Test solution. Dissolve 35 mg of the substance under
examination in 5 ml of acetonitrile and dilute to 50.0 ml with Tablets.
the mobile phase. Dilute 10.0 ml of this solution to 100.0 ml Determine by liquid chromatography (2.4.14), using the
with the mobile phase. chromatographic conditions and the reference solution
Reference solution. Dissolve 35 mg of nebivolol described in the Assay.
hydrochloride RS in 5 ml of acetonitrile and dilute to 50.0 ml Test solution. Disperse one tablet in the mobile phase, dilute
with the mobile phase. Dilute 10.0 ml ofthis solution to 100.0 to obtain a solution containing 0.005 per cent w/v ofnebivolol
ml with the mobile phase. in the mobile phase and filter.
Use the chromatographic system described under Related Calculate the content of C22H25F2N04 in the tablets.
substances.
Other tests. Comply with the tests stated under Tablets.
Inject the reference solution. The test is not valid unless the Assay. Determine by liquid chromatography (2.4.14).
relative standard deviation for replicate injections is not more
than 2.0 per cent. Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing about 5 mg ofnebivolol,
Inject the test solution and the reference solution. disperse in 100.0 ml ofthe mobile phase and filter.
Calculate the content ofC22H25F2N04,HCl. Reference solution. A solution containing nebivolol
Storage. Store protected from moisture, at a temperature not hydrochloride RS equivalent to 0.005 per cent w/v of
exceeding 30°. nebivolol, in the mobile phase.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with
Nebivolol Tablets octadecylsilane bonded to porous silica (5 /lm),
Nebivolol Hydrochloride Tablets mobile phase: a mixture of45 volumes ofa buffer solution
prepared by dissolving 6.8 g of potassium dihydrogen
Nebivolol Tablets contain not less than 90.0 per cent and not
orthophosphate in 1000 ml of wate]; adding 2 ml of
more than 110.0 per cent of the stated amount ofnebivolol,
triethylamine and adjusting the pH to 3.0 with 10 per
C22H25F2N04. cent v/v orthophosphoric acid, 25 volumes of
Usual strength. 5 mg acetonitrile and 30 volumes of methanol,

1759

www.webofpharma.com
NELFINAVIR MESYLATE IP 2010

- flow rate. 1 ml per minute, Tests


- spectrophotometer set at 281 nm,
- injection volume. 20 111. Specific optical rotation (2.4.22). -105 0 to -120 0 , determined
in a 1.0 per cent w/v solution in methanol.
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more Related substances. Determine by liquid chromatography
than 2.0 per cent. (2.4.14), using the chromatographic system described in the
Assay.
Inject the test solution and the reference solution.
Test solution. A 0.1 per cent w/v solution of the substance
Calculate the content of C22H2SF2N04 in the tablets, under examination in the mobile phase.
Storage. Store protected from light and moisture. Reference solution (a). A 0.001 per cent w/v solution of the
Labelling. The label states the strength in terms of the substance under examination in the mobile phase.
equivalent amount of nebivolol. Reference solution (b). A 0.01 per cent w/v solution of
methanesulphonic acid in the mobile phase.
Inject reference solution (a). The test is not valid unless the
column efficiency determined from the nelfinavir peak is not
Nelfinavir Mesylate , less than 4000 theoretical plates and the tailing factor is not
more than 2.0.
Separately inject reference solution (b) and record the
chromatograms. Separately inject the test solution and
continue the chromatography for at least three times the
retention time of the principal peak. In the chromatogram
obtained with the test solution, the area of any peak other
than the principal peak is not greater than half of the area of
the principal peak in the chromatogram obtained with reference
solution (a) (0.5 per cent) and the sum of the areas of all such
peaks is not greater than the area of the principal peak in the
chromatogram obtained with reference solution (a) (1.0 per
C32~sN304S,C~03S Mol. Wt. 663.9 cent). Ignore any peak due to methanesulphonic acid
corresponding to the retention time of the principal peak in
Nelfinavir Mesylate is (3S,4aS,8aS)-N-(tert-butyldecahydro- the chromatogram obtained with reference solution (b).
2-[(2R,3R)-3-(3-hydroxy-o-toluamido)-hydroxy-4-
(phenylthio)butyl]isoquinoline-3-carboxamide methyl Methanesulphonic acid. 13.5 per cent to 15.5 per cent w/w,
sulphonate. calculated on the anhydrous basis, determined by the following
method. Weigh accurately about 0.6 g, dissolve in 50 ml of
Nelfinavir Mesylate contains not less than 98.0 per cent and dimethylformamide and titrate with 0.1 M sodium hydroxide,
not more than 101.0 per cent of C32H4SN304S,CH403S, determining the end-point potentiometrically (2.4.25). Carry
calculated on the anhydrous basis. out a blank titration.
Category. Antiretroviral.
1 ml of 0.1 Msodium hydroxide is equivalentto 0.00961 g of
Dose. 1.25 g twice daily. CH3S03H.
Description. A white or almost white powder. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Identification Sulphated ash (2.3 .18). Not more than 0.1 per cent.
A. Determine by infrared absorption spectrophotometry (2.4.6). Water (2.3.43). Not more than 3.0 percent, determined on 0.5 g.
Compare the spectrum with that obtained with neljinavir
Assay. Determine by liquid chromatography (2.4.14).
mesylate RS or with the reference spectrum of nelfinavir
mesylate. Test solution. A 0.01 per cent w/v solution of the substance,
B. In the Assay, the principal peak in the chromatogram under examination in the mobile phase.
obtained with the test solution corresponds to the peak in the Reference solution. A 0.01 per cent w/v solution ofneljinavir
chromatogram obtained with the reference solution. mesylate RS in the mobile phase.

1760

www.webofpharma.com
IP 2010 NELFINAVIR MESYLATE ORAL POWDER

Chromatographic system Use the chromatographic system described under Assay.


a stainless steel column 25 em x 4.6 rom, packed with
Inject the test solution and the reference solution.
octadecylsilane bonded to porous silica (5 /lm),
mobile phase: a filtered and degassed mixture of D. Not less than 75 per cent of the stated amount of
45 volumes of acetonitrile, 20 volumes of methanol C32~5N304S,
and 35 volumes ofa buffer prepared by dissolving 4.0 g
Related substances. Determine by liquid chromatography
of sodium dihydrogen phosphate in 1000 ml of water,
(2.4.14).
to which 1 ml of dimethylamine solution and 1 g of
sodium octanesulphonate are added and mixed to "Test solution. Weigh accurately a quantity of the oral powder
dissolve, containing 50 mg ofNelfinavir Mesylate, disperse in 10 ml of
flow rate. 1 ml per minute, methanol, dilute to 50 ml with the mobile phase and filter.
spectrophotometer set at 215 nm, Reference solution (a). Dissolve 10 mg of nelfinavir mesylate
injection volume. 20 Ill. RS in 2 ml of methanol and dilute to 10 ml with the mobile
Inject the reference solution. The test is not valid unless the phase.
column efficiency determined from the nelfinavir peak is not Reference solution (b). Dilute 1 ml ofreference solution (a) to
less than 5000 theoretical plates, the tailing factor is not more 100 ml with the mobile phase.
than 2.0 and the relative standard deviation for replicate
injections is not more than 2.0 per cent. Chromatographic system
a stainless steel column 15 em x 4.6 mm, packed with
Separately inject the test solution and the reference solution octadecylsilane bonded to porous silica (5 /lm),
and measure the responses for the principal peak. Calculate column temperature. 45°,
the content ofC32H45N304S,CH403S. mobile phase: a mixture of 28 volumes ofa buffer solution
Storage. Storeprotected from light. prepared by dissolving 4.88 g of anhydrous sodium
dihydrogen phosphate in 1000 ml of water, adjusting
the pH to 3.4 with phosphoric acid and filtering,
27 volumes of acetonitrile, 20 volumes of methanol
Nelfinavir Mesylate Oral Powder and 25 volumes of water. Adjust the pH to 4.8 with
0.1 M sodium hydroxide or orthophosphoric acid.
Nelfinavir Mesylate Oral Powder contains not less than
flow rate. 1 ml per minute,
90.0 per cent and not more than 110.0 per cent of the stated
spectrophotometer set at 220 nm,
amount ofnelfinavir, C32~5N304S,
injection volume. 10 Ill.
Usual strength. 50 mg per g.
Inject the reference solution (a). The test is not valid unless
the tailing factor is not more than 2.0 and the column efficiency
Identification
in not less than 4000 theoretical plates.
In the Assay, the principal peak in the chromatogram obtained Inject the test solution and reference solution (b). In the
with the test solution corresponds to the peak in the chromatogram obtained with the test solution, the area of any
chromatogram obtained with the reference solution. secondary peak is not more than the area of the peak in the
Tests chromatogram obtained with the reference solution (b)
(1.0 per cent) and the sum of areas of all the secondary peaks
Dissolution (2.5.2). is not more than twice the area ofthe peak in the chromatogram
Apparatus. No 1 obtained with the reference solution (b) (2.0 per cent).
Medium. 900 ml of 0.1 M hydrochloric acid. Water (2.3.43). Not more than 12.0 per cent, determined on
Speed and time. 75 rpm and 45 minutes. 0.5g.
Withdraw a suitable volume ofthe medium and filter. Assay. De~'rmine by liquid chromatography (2.4.14).
Determine by liquid chromatography (2.4.14). Solvent mixture. 30 volumes of water and 70 volumes of
Test solution. Use the filtrate and, ifnecessary, dilute with the methanol.
dissolution medium. Test solution. Weigh accurately a quantity of the powder
Reference solution. A 0.065 per cent w Iv solution of nelfinavir containing 50 mg ofNelfinavir Mesylate, disperse in 50 ml of
mesylate RS in methanol. Dilute 10 ml ofthe solution to 100 ml 0.1 M hydrochloric acid, dilute to 250.0 ml with the solvent
with the dissolution medium. mixture and filter.

1761

www.webofpharma.com
NELFINAVIR MESYLATE ORAL POWDER IP 2010

Reference solution. Dissolve 10 mg ofneljinavir mesylate RS Tests


in 10 ml ofO.1Mhydrochloric acid and dilute to 50.0 ml with
the solvent mixture. Dissolution (2.5.2).
Apparatus No.1,
Chromatographic system
Medium. 900 mlofO.OI Mhydrochloric acid,
a stainless'steel column 15 cm x 4.6 mm,packed with
octadecylsilane bonded to porous silica (5 /lm), Speed and time. 50 rpm and 30 minutes.
- column temperature 40°, Withdraw a suitable volume ofthe medium and filter promptly
mobile phase: a mixture of 35 volumes ofa buffer solution through a membrane filter disc with an average pore diameter
prepared by dissolving 4 g of sodium dihydrogen not greater than 1.0 /lm. Reject the first few ml ofthefiltrate
phosphate dihydrate and 1g of I-octane sulphonic and dilute a suitable volume of the filtrate with the same
acid sodium salt into 1000 ml of water, adding 1ml of solvent. Measure the absorbance of the resulting solution at
dimethylamine and filtering, 45 volumes acetonitrile the maximum at about 250 nm (2.4.7). Calculate the content of
and 20 volumes of methanol, C32H4SN304S,CH403S from the absorbance of a solution of
flow rate. 2 ml per minute, known concentration of neljinavir mesylate RS.
spectrophotometer set at 220 nm, D. Not less thau 75 per cent of the stated amount of
iIljection volume. 10 Ill. C32~sN304S,C~03S,
Inject the reference solution. The test is not valid unless the Related substances. Determine by liquid chromatography
tailing factor is not more than 2.0, the column efficiency in not (2.4.14).
less than 2000 theoretical plates and the relative standard
Test solution. Weigh accurately a quantity of the powdered
deviation for replicate injections is not more than 2.0 per cent.
tablets containing about 100 mg ofNelfinavir Mesylate, add
Inject the test solution and the reference solution. about 20 ml of methanol, mix with the aid of ultrasound for
Calculate the content of C32H4SN304S in the oral powder. 10 minutes and dilute to 100 ml with.the mobile phase.

Storage. Store protected from moisture, at a temperature not Reference solution. Weigh accurately about 10 mg of
exceeding 30°. neljinavir mesylate RS, add about 10 ml of methanol, shake
for 10 minutes and dilute to 50 ml with the mobile phase.
Labelling. The label states the strength in terms of the
Chromatographic system
equivalent amount ofnelfinavir.
a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica particles or
ceramic microparticles (5 /lm),
- mobile phase: a filtered and degassed mixture of
Nelfinavir Tablets 45 volumes of acetonitrile, 20 volumes of methanol
and 35 volumes ofa buffer prepared by dissolving 4.0 g
Nelfinavir Mesylate Tablets
of sodium dihydrogen phosphate in 1000 ml of water,
Nelfinavir Tablets contain not less than 90.0 per cent and not to which are added 1 ml of dimethylamine solution and
more than 110.0 per cent of the stated amount of nelfinavir 1 g of sodium octanesulphonate and mixing to dissolve,
mesylate, C32H4SN304S,CH403S. flow rate. 1 ml per minute,
Usual strengths. 250 mg; 625 mg. - spectrophotometer set at 215 urn,
injection volume. 20 Ill.
Identification Inject the reference solution. The test is not valid unless the
column efficiency determined from the nelfinavir mesylate peak
A. Shake a quantity ofthe powdered tablets containing about is not less than 4000 theoretical plates and the tailing factor is
0.1 g of Nelfinavir Mesylate with 80 ml of methanol for not more than 2.0.
10 minutes, add sufficient methanol to produce 100 ml, mix
Inject separately the diluent (10 ml of methanol diluted to
and filter. Dilute 5 ml ofthe filtrate to 100 ml with methanol.
50 ml with the mobile phase) and the test solution and continue
When examined in the range 200 nm to 300 urn the resulting the chromatography for 4 times the retention time of the
solution shows an absorption maximum only at about 254 nm principal peak. Examine the diluent chromatogram Jor any
(2.4.7). extraneous peaks and ignore the corresponding peaks
observed in the chromatogram obtained with the test solution.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the Any secondary peak observed in the chromatogram obtained
chromatogram obtained with the reference solution. with the test solution should not be more than 1.0 per cent

1762

www.webofpharma.com
IP 2010 NEOMYCIN SULPHATE

and the sum of the areas of all the secondary peaks should Mobile phase. A freshly prepared 3.85 per cent w/v solution
not be more than 2.0 per cent when calculated by percentage of ammonium acetate.
area normalisation. Inhibit integration of peak due to
Test solution. Dissolve 0.2 g of the substance under
methanesulphonic acid.
examination in 10 ml of water.
Other tests. Complies with the tests stated under Tablets.
Reference solution. A 2.0 per cent w/v solution of neomycin
Assay. Determine by liquid chromatography (2.4.14). sulphate RS in water.
Test solution. Weigh accurately a quantity of the powdered Apply to the plate 1 III of each solution. After development,
tablets containing about 200 mg ofNelfinavir Mesylate, add dry the plate in air for 10 minutes, heat at 100 0 for 1 hour and
about 20 ml of methanol, mix with the aid of ultrasound for spray with a 0.1 per cent w/v solution of ninhydrin in
10 minutes and dilute to 100.0 ml with the mobile phase. Filter I-butanol saturated with water. Heat again at 100 0 for
through a membrane filter disc with an average pore diameter 5 minutes. The principal spot in the chromatogram obtained
not greater than 1.0 11m, rejecting the first few ml ofthe filtrate. with the test solution corresponds to that in the chromatogram
Further dilute 5.0 ml ofthe filtrate to 100.0 ml with the mobile obtained with the reference solution.
phase.
B. Dissolve about 10 mg in 5 ml ofwater, add 0.1 ml ofpyridine
Reference solution. Weigh accurately about 50 mg of and 2 ml of a 0.1 per cent w/v solution of ninhydrin and heat
neifinavir mesylate RS, add about 10 ml ofmethanol, mix with on a water-bath at a temperature of about 70 0 for 10 minutes;
the aid ofultrasound to dissolve and dilute to 50.0 ml with the a deep violet colour is produced.
mobile phase. Dilute 5.0 ml ofthis solution to 50.0 ml with the
C. A 5 per cent w/v solution gives the reactions of sulphates
mobile phase.
(2.3.1).
Use the chromatographic system described in the test for
Related substances. Tests
Inject the reference solution. The test is not valid unless the
pH (2.4.24). 5.0to 7.5, determined in a 1.0 percentw/v solution.
column efficiency determined from the nelfmavir mesylate peak
is not less than 5000 theoretical plates, the tailing factor is not Specific optical rotation (2.4.22).+53.5 0 to +59.0 0 , determined
more than 2.0 and the relative standard deviation for replicate in a 10.0 per cent w/v solution.
injections is not more than 2.0 per cent.
Neamine. Determine by thin-layer chromatography (2.4.17),
Inject separately the test solution and the reference solution coating the plate with silica gel H
and measure the responses for the major peale Calculate the
Mobile phase. A mixture of 30 volumes of methanol, 20
content ofC32H4sN304S,CH403S in the tablets.
volumes of strong ammonia solution and 10 volumes of
Storage. Store protected from light. dichloromethane.
Test solution. Dissolve 0.25 g of the substance under
examination in 10 ml of water.
Neomycin Sulphate Reference solution. A 0.05 per cent w/v solution of neamine
Neomycin Sulphate is a mixture ofthe sulphates ofsubstances RSin water.
obtained by the growth of certain selected strains of Apply to the plate as 5-mm bands 5 III of each solution. Dry
Streptomyces fradiae. the bands; allow the mobile phase to rise at least 8 cm. Dry the
Neomycin Sulphate has a potency of not less than 600 Units plate in a current ofwann air, heat at 1100 for 10 minutes, spray
per mg, calculated on the dried basis. the plate with ninhydrin and stannous chloride reagent and
heat at 1100 for 15 minutes. Spray the plate again with the
Category. Antibacterial (topical and systemic). same reagent and heat at 110 0 for 15 minutes. Any band
Dose. 1 g every 4 hours. corresponding to neamine in the chromatogram obtained with
the test solution is not more intense than the spot in the
Description. A white or yellowish-white powder; odourless
chromatogram obtained with the reference solution.
or almost odourless; hygroscopic.
Neomycin C. Determine by thin-layer chromatography (2.4.17),
Identification coating the plate with silica gel of a suitable grade.
A. Determine by thin-layer chromatography (2.4.17), coating Mobile phase. A mixture of 80 volumes of a 20 per cent w/v
the plate with silica gel H solution of sodium chloride and 20 volumes of methanol.

1763

www.webofpharma.com
NEOMYCIN SULPHATE IP 2010

Test solution. Dissolve 40 mg of the substance under Mobile phase. A mixture of 60 volumes of methanol,
examination in water and dilute to 5 ml with the same solvent. 40 volumes of strong ammonia solution and 20 volumes of
Reference solution (a). Dissolve 30 mg ofjramycetin sulphate chloroform.
RS in water and dilute to 25 ml with the same solvent. Test solution. Dilute ifnecessary a volume ofthe eye drops to
produce a solution containing 0.5 per cent w/v of Neomycin
Reference solution (b). Dilute 5 ml ofreference solution (a) to
Sulphate in water.
25 ml with water.
Reference solution (a). A 0.5 per cent w/v solution of
Reference solution (c). Dissolve 40 mg of neomycin sulphate
neomycin sulphate RS in water.
RS in water and dilute to 5 ml with the same solvent.
Reference solution (b). A mixture of equal volumes ofthe eye
Apply to the plate as 5 mm bands 5 J..Ll of each solution. Dry
drops and reference solution (a).
the bands; allow the mobile phase to rise at least 12 cm. Dry
the plate at 100° to 105° for 10 minutes. Spray the plate with Apply to the plate 1 J..Ll of each solution. After development,
ethanolic ninhydrin solution and heat at 100° to 105° for dry the plate in air, spray with a 1 per cent w/v solution of
10 minutes. In the chromatogram obtained with the test ninhydrin in I-butanol and heat at 105° for 2 minutes. The
solution the principal band corresponds to the principal band principal red spot in the chromatogram obtained with the test
in the chromatogram obtained with reference solution (c) and solution corresponds to that in the chromatogram obtained
the band due to neomycin C with Rf value slightly less than with reference solution (a) and the principal red spot in the
that of the principal band is not more intense than the band chromatogram obtained with reference solution (b) appears
obtained with reference solution (a) (15 per cent) but is more as a single spot.
intense than the band in the chromatogram obtained with
reference solution (b) (3 per cent). The test is not valid unless Tests
in the chromatogram obtained with reference solution (c) a Neamine. Determine by thin-layer chromatography (2.4.17),
band appears with R f value slightly less than that of the coating the plate with silica gel H
principal band.
.Mobile phase. A mixture of 30 volumes of methanol,
Sulphated ash (2.3.18). Not more than 1.0 per cent. 20 volumes of strong ammonia solution and 10 volumes of
Loss on drying (2.4.19). Not more than 8.0 per cent, determined dichloromethane.
on 0.5 g by drying in an oven at 60° over phosphoruspentoxide Test solution. A volume of the eye drops containing 5 J..Lg
at a pressure not exceeding 0.7 kPa for 3 hours. (3.5 Units).
Assay. Determine by the microbiological assay ofantibiotics, Reference solution. The same volume of water containing
Method A (2.2.1 0). 0.1 J..Lg ofneamine RS.
Storage. Store protected from light and moisture. Apply to the plate each solution. After development, dry the
Labelling. The label states the strength in terms of Units of plate in a current ofwarm air, heat at 110° for 10 minutes, spray
neomycin per mg. the plate with ninhydrin and stannous chloride reagent and
heat at 11 0° for 15 minutes. Spray the plate again with the
same reagent and heat at 110° for 15 minutes. Any spot
corresponding to neamine in the chromatogram obtained with
Neomycin Eye Drops the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Neomycin Sulphate Eye Drops
Neomycin C. Determine by liquid chromatography (2.4.14).
Neomycin Sulphate Eye Drops are a sterile solution of
Neomycin Sulphate in Purified Water. Test solution. Dilute the eye drops with 0.02 M borax to
contain 1 mg (700 Units) per mI. To 0.5 mI ofthe diluted solution
Neomycin Sulphate Eye Drops contain not less than 90.0 per add 1.5 ml of a freshly prepared 2 per cent w/v solution of
cent and not more than 115.0 per cent w/v ofthe stated amount I-jluoro-2,4-dinitrobenzene in methanol, dilute to 25 ml with
of neomycin sulphate. the mobile phase, allow to stand and use the clear lower layer.
Usual strength. 0.5 per cent w/v (3500 Units per ml). Reference solution. Add 1.5 ml of the 1-fluoro-2,4-
dinitrobenzene solution to 0.5 ml ofa 0.1 per cent w/v solution
Identification
of neomycin sulphate RS in 0.02 M borax, heat in a water-
Determine by thin-layer chromatography (2.4.17), coating the bath at 60° for 1 hour and cool; dilute the solution to 25 ml with
plate with silica gel. the mobile phase, allow to stand and use the clear lower layer.

1764

www.webofpharma.com
IP 2010 NEOMYCIN EYE OINTMENT

Chromatographic system Usual strength. 0.5 per cent w/w (3500 Units per g).
a stainless steel column 20 em x 4.6 mm, packed with
porous silica particles (5 /-lm) (such as NucleosiI100-5), Identification
- mobile phase: a mixture of 97 ml of tetrahydrofuran, Determine by thin-layer chromatography (2.4.17), coating the
1.0 ml of water and 0.5 ml of glacial acetic acid diluted plate with silica gel.
with sufficient of a 2.0 per cent v/v solution of ethanol
Mobilephase. A mixture of60 volumes of methanol, 40 volumes
in ethanol-free chloroform to produce 250 ml,
of strong ammonia solution and 20 volumes of chloroform.
flow rate. 1.6 m1 per minute,
- spectrophotometer set at 350 nm, Test solution. Disperse a quantity of the eye ointment
- injection volume. 10 Ill. containing 20 mg ofNeoinycin Sulphate in 20 ml of chloroform,
extract with 5 ml of water and use the aqueous extract.
If necessary the tetrahydrofuran and water content of the
mobile phase may be adjusted so that the chromatogram Reference solution (a). A 0.4 per cent w/v solution of
obtained with the reference solution shows resolution similar neomycin sulphate RS in water.
to that in the specimen chromatogram supplied withframycetin Reference solution (b). A mixture of equal volumes of test
sulphate RS. The mobile phase should be passed through the solution and reference solution (a).
column for several hours before the solutions are injected.
Continue the chromatography for 1.4 times the retention time Apply to the plate 1 /-ll of each solution. After development,
dry the plate in air, spray with a 1 per cent w/v solution of
ofthe peak due to neomycin B.
ninhydrin in i-butanol and heat at 105° for 2 minutes. The
The column efficiency, determined using the peak due to principal red spot in the chromatogram obtained with the test
Neomycin B in the chromatogram obtained with the test solution corresponds to that in the chromatogram obtained
solution, should be not less than 13,000 theoretical plates. with reference solution (a) and the principal red spot in the
In the chromatogram obtained with the test solution the area chromatogram obtained with reference solution (b) app~ars
of the peak corresponding to neomycin C is not less than as a single spot.
3.0 per cent and not more than 15.0 per cent ofsum ofthe areas Tests
ofthe peaks corresponding to Neomycin B and Neomycin C.
Neamine. Determine by thin-layer chromatography (2.4.17),
Other tests. Complies with the tests stated under Eye Drops.
coating the plate with silica gel H.
Assay. Measure accurately a quantity containing 5 mg of Mobile phase. A mixture of 30 volumes of methanol,
Neomycin Sulphate and dilute to 50.0 ml with sterile phosphate 20 volumes of strong ammonia solution and 10 volumes of
bufferpH 8.0 and mix. Dilute 10.0 ml ofthe resulting solution dichloromethane.
to 100.0 ml with the same solvent.
Test solution. Disperse a quantity of the eye ointment
Determine by the microbiological assay ofantibiotics, Method containing 20 mg ofNeomycin Sulphate in 20 m1 of chloroform,
A (2.2.10) shake gently with 8 ml of water, allow the layers to separate
The upper fiducial limit of error is not less than 90.0 per cent and use the aqueous layer.
and the lower fiducial limit oferror is not more than 115.0 per Reference solution. A 0.005 per cent w/v solution of neamine
cent of the stated number of Units per ml. RSin water.
Storage. Store protected from light. Apply to the plate 2 /-l1 of each solution. After development,
Labelling. The strength is stated in terms of percentage w/v dry the plate in a current ofwarm air, heat at 110° for 10 minutes,
as well as the number ofUnits per ml. spray with ninhydrin and stannous chloride reagent and
heat at 110° for 15 minutes. Spray the plate again with the
same reagent and heat at 1l0° for 15 minutes. Any spot
corresponding to neamine in the chromatogram obtained with
Neomycin Eye Ointment the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Neomycin Sulphate Eye Ointment
Neomycin C. Determine by liquid chromatography (2.4.17)
Neomycin Sulphate Eye Ointment is a sterile preparation
Test solution. Disperse a quantity of the eye ointment
containing Neomycin Sulphate in a suitable basis.
containing 5 mg of Neomycin Sulphate in 20 ml of light
Neomycin Sulphate Eye Ointment contains not less than petroleum (120° to 160°), add 5 m1 of 0.02 Mborax, shake,
90.0 per cent and not more than 115.0 per cent of the stated separate the aqueous layer and centrifuge. To 0.5 m1 of the
amount of neomycin sulphate. separated aqueous layer add 1.5 ml ofa freshly prepared 2 per

1765

www.webofpharma.com
NEOMYCIN EYE OINTMENT IP 2010

cent w/v solution of I-jluoro-2,4-dinitrobenzenein methanol, Neostigmine Bromide


heat on a water-bath at 60° for 1 hour and cool. Dilute the
resulting solution to 25 ml with the mobile phase, allow to
stand and use the clear lower layer.
Refaence solution. Add 1.5 ml of the I-fluoro-2,4-
dinitr,obenzene solution to 0.5 ml ofa 0.1 per cent w/v solution
of neomycin sulphate RS in 0.02 M borax and proceed as for
the test solution.
Chromatographic system
a stainless steel column 20 cm x 4.6 mm, packed with
porous silica particles (5 Ilm), Mol. Wt. 303.2
mobile phase: 97 ml of tetrahydrojitran, 1.0 ml of water
Neostigmine Bromide is 3-(dimethylcarbamoyloxy)
and 0.5 ml of glacial acetic acid with sufficient of a
trimethylanilinium bromide.
2.0 per cent v/v solution of ethanol in ethanol-free
chloroform to produce 250 ml, Neostigmine Bromide contains not less than 98:0 per cent and
- flow rate. 1.6 ml per minute, not more than 101.0 per cent ofClzH19BrNzOz, calculated on
- spectrophotometer set at 350 run, the dried basis.
- injection volume. 10 Ill. Category. Anticholinesterase.
If necessary the tetrahydrofuran and water content of the
Dose. 15 to 30 mg, repeated at suitable intervals; total daily
mobile phase may be adjusted so that the chromatogram
dose, 75 to 300 mg.
obtained with reference solution shows resolution similar to
that in the specimen chromatogram supplied withji-amycetin Description. Colourless crystals or a white, clystalline powder;
sulphate RS. The mobile phase should be passed through the odourless; hygroscopic.
column for several hours before the solutions are injected.
Continue the chromatography for 1.4 times the retention time
Identification
of the peakdue to neomycin B. Test A may be omitted if tests B, C, D and E are carried out.
The column efficiency, determined using the peak due to Tests B, C and D may be omitted if tests A and E are carried
Neomycin B in the chromatogram obtained with the test out.
solution, should be not less than 13,000 theoretical plates. A. Detennine by infrared absorption spectrophotometly (2.4.6).
In the chromatogram obtained with the test solution the area Compare the spectrum with that obtained with neostigmine
of the peak corresponding to neomycin C is not less than bromide RS.
3.0 per cent and not more than 15.0 per cent ofthe sum ofthe B. When examined in the range 230 nm to 360 nm (2.4.7), a
areas ofthe peaks corresponding to Neomycin B and Neomycin 0.02 per cent w/v solution in 0.5 M sulphuric acid shows
C. absorption maxima at about 260 nm and 266 nm.
Other tests. Complies with the tests stated under Eye C. Warm about 50 mg with 1 ml of dilute sodium hydroxide
Ointments. solution; an odour of dimethylamine develops slowly.
Assay. Weigh accurately a quantity containing 5 mg of D. Warm about 50 mg with 0.4 g ofpotassium hydroxide and
Neomycin Sulphate, dissolve in 25 ml of chloroform, extract 2 ml of ethanol (95 per cent) on a water-bath for 3 minutes,
with four quantities, each of20 ml, of sterile phosphate buffer replacing the evaporated ethanol. Cool, add 2 ml of dilute
pH 8.0, combine the extracts and add sufficient of the buffer diazobenzenesulphonic acid solution; an orange-red colour
solution to produce 100.0 ml. is produced.
Cany out the microbiological assay of antibiotics, Method A E. Gives the reactions ofbromides(2.3.1).
(2.2.10).
The upper fiducial limit of error is not less than 90.0 per cent Tests
and the lower fiducial limit ofelTor is not more than 115.0 per Appearance of solution. A 0.5 per cent w/v solution is clear
cent of the stated number of Units per g. (2.4.1), and colourless (2.4.1).
Storage. Store protected from light. Acidity. Dissolve 0.2 g in20 ml of carbondioxide-free water
Labelling. The strength is stated in terms of percentage w/v and titrate to pH 7.0 with 0.02 Msodium hydroxide (carbonate-
as well as the number of Units per ml. free); not more than 0.1 ml is required.

1766

www.webofpharma.com
IP 2010 NEOSTIGMINE METHYLSULPHATE

3-Hydroxytrimethylanilinium bromide. Dissolve 50 mg in a Bromide, transfer to a semi-micro ammonia-distillation


mixture of 1 ml of sodium carbonate solution and 9 ml of apparatus, add 20 ml of a 50 per cent w/v solution of sodium
water. Absorbance of the resulting solution at about 294 nm, hydroxide and 0.5 ml ofa 2 per cent w/v solution of 2-octanol
measured immediately after preparation, not more than in liqUidparaffin. Pass a current ofsteam through the mixture,
0.25 (2.4.7). . collect the distillate in 50 ml of 0.01 Msulphuric acid until the
volume is about 200 ml and titrate the excess of acid with
Sulphates (2.3.17). 0.75 g complies with the limit test for
sulphates (200 ppm). 0.02 M sodium hydroxide using methyl red solution as
indicator. Repeat the operation without the substance unde~'
Sulphated ash (2.3.18). Not more than 0.1 percent. examination. The difference between the titrations represents
Loss on drying (2.4.19). Not more than 1.0 per cent, detennined the amount of sulphuric acid required to neutralise the
on 1.0 g by drying in an oven at 105°. dimethylamine produced.

Assay. Weigh accurately about 0.5 g, dissolve in 20 ml of 1 ml of 0.01 M sulphuric acid is equivalent to 0.006064 g of
anhydrous glacial acetic acid, add 5 ml of acetic anhydride. C12H19BrN202'
Titrate with 0.1 M perchloric acid, using crystal violet Storage. Store protected from light and moisture.
solution as indicator. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.03032 g of
C,2HI9BrN202. Neostigmine Methylsnlphate
Storage. Store protected from light and moisture.
Mol. Wt. 334.4
Neostigmine Methylsulphate is 3-(dimethylcarbamoyloxy)-
trimethylanilinium methyl sulphate.
Neostigmine Tablets
Neostigmine Methylsulphate contains not less than 98.5 per
Neostigmine Bromide Tablets cent and not more than 101.0 per cent of CI3H22N206S,
calculated on the dried basis.
Neostigmine Bromide Tablets contain not less than 92.5 per
cent and not more than 107.5 per cent of the stated amount of Category. Anticholinesterase.
neostigmine bromide, CI2H,9BrN202.
Dose. By subcutaneous or intramuscular injection, 1 to 2.5
Usualstrength. 15 mg. mg, repeated at suitable intervals; total daily dose,S to 20 mg.

Identification Description. Colourless crystals or a white, crystalline powder;


hygroscopic.
Triturate a quantity of the powdered tablets containing about
0.3 g of Neostigmine Bromide with three quantities, each of Identification
5 ml of ether and discard the ether. Macerate the residue with Test A may be omitted iftests B, C andD are carried out. Tests
several quantities, each of 10 ml of ethanol (95 per cent), B, C and D may be omitted if test A is carried out.
filtering after each maceration. Evaporate the combined filtrates
on a water-bath and dry the residue at 105° for 1 hour. The A. Detennine by infrared absorption spectrophotometry (2.4.6).
residue melts at about 167°, with decomposition. The residue Compare the spectrum with that obtained with neostigmine
complies with the following tests. methylsulphate RS or with the reference spectrum of
neostigmine methylsulphate.
A. Wann about 50 mg with 0.4 g ofpotassium hydroxide and
2 ml of ethanol (95 per cent) on a water-bath for 3 minutes, B. When examined in the range 230 nm to 360 nm (2.4.7), a
replacing the evaporated ethanol. Cool, add 2 ml of dilute 0.05 per cent w/v solution in 0.5 M sulphuric acid shows
diazobenzenesulphonic acid solution; an orange-red colour absorption maxima, at about 261 nm and 267 nm. The ratio of
is produced. the absorbance at the maximum at about 267 run to that at the
maximum at261nm is 0.84 to 0.87.
B. Gives the reactions ofbromides (2.3.1).
C. Dissolve 0.1 g in 5 ml of distilled water and add 1 ml of a
Tests 6 per cent w/v solution of barium chloride; no precipitate is
produced. Add 2 ml of hydrochloric acid and heat in a water-
Other tests. Complies with the tests stated under Tablets. bath for 10 minutes; a white precipitate is produced.
Assay. Weigh and powder 20 tablets. Weigh accurately a D. Wann about 50 mg with 0.4 g ofpotassium hydrOXide and
quantity ofthe powder containing about 0.15 g ofNeostigmine 2 ml of ethanol (95 per cent) on a water-bath for 3 minutes,

1767

www.webofpharma.com
NEOSTIGMINE METHYLSULPHATE IP 2010

replacing the evaporated ethai1Ol. Cool, add 2 ml of dilute Usualstrengths. 0.5 mgperml; 2.5 mg per ml.
diazobenzenesulphonic acid solution; an orange-red colour
is produced. Identification
Tests A. Dilute, if necessary, a volume of the injection containing
2.5 mg of Neostigmine Methylsulphate to 5 ml with water,
Appearance ofsolution. A 5.0 per cent w/v solution in distilled shake with three quantities, each of 10 ml, of ether and discard
water is clear (2.4.1), and colourless (2.4.1). the ether extracts.
Acidity or alkalinity. To 4.0ml ofa 5.0 per centw/v solution in When examined in the range 230 urn to 360 urn (2.4.7), a 2 cm
distilled water add 6.0 ml of water and 0.1 ml of phenol- layer of the resulting solution shows absorption maxima at
phthalein solution; the solution is colourless. Add about 260 urn and 267 urn.
0.3 ml of O. 01 M sodium hydroxide; the solution becomes red.
B. Determine by thin-layer chromatography (2.4.17), coating
Add 0.4 ml of 0.01 M hydrochloric acid; the solution becomes
the plate with silica gel G
colourless. Add 0.1 ml of methyl red solution; the solution
becomes red or yellowish-red. Mobile phase. A mixture of 50 volumes of chloroform,
35 volumes of methanol, 10 volumes of formic acid and
3-Hydroxytrimethylanilinium methyl sulphate. Dissolve
5 volumes of water.
50 mg in a mixture of 1 ml of sodium carbonate solution and
9 ml of water. Absorbance of the resulting solution at about Test solution. Dilute the injection under examination, if
294 urn, measured immediately after preparation, not more than necessary, with water to produce a solution containing
0.20(2.4.7). 0.05 per cent w/v ofNeostigmine Methylsulphate.
Chlorides (2.3.12). 1.0 g complies with the limit test for Reference solution (a). A 0.05 per cent w/v solution of
chlorides (250 ppm). neostigmine methylsulphate RS in water.
Sulphates (2.3.17). 0.75 g complies with the limit test for Reference solution (b). A mixture of equal volumes ofthe test
sulphates (200 ppm). solution and reference solution (a).

Loss on drying (2.4.19). Not more than 1.0 per cent, determined Apply to the plate 10 /!l ofeach solution. After development,
on 1.0 g by drying in an oven at 105°. dry the plate in air, spray with dilute potassium
iodobismuthate solution. The principal spot in the
Sulphated ash (2.3.18). Not more than 0.1 per cent. chromatogram obtained with the test solution corresponds to
Assay. Weigh accurately about 0.3 g and dissolve in 150 m1 of that in the chromatogram obtained with reference solution (a).
water. Add 100 ml of2 M sodium hydroxide, distill and collect The principal spot in the chromatogram obtained with
the distillate in 50 ml ofa 4 per cent w/v solution of boric acid reference solution (b) appears as a single, compact spot.
until a total volume of250 ml is reached. Titrate the distillate C. To 1 ml add 0.5 ml of sodium hydroxide solution and
with 0.1 M hydrochloric acid using 0.25 ml of methyl red- evaporate to dryness on a water-bath. Heat quickly in an oil-
methylene blue solution as indicator. Repeat the operation bath to about 250° and maintain at this temperature for about
without the substance under examination. The difference 30 seconds. Cool, dissolve the residue in 1 ml of water, cool in
between the titrations represents the amount of hydrochloric ice water and add 1 ml of diazobenzenesulphonic acid
acid required. solution; an orange-red colour is produced.
1 ml of 0.1 M hydrochloric acid is equivalentto 0.03344 g of
Tests
C13H22N206S.
Storage. Store protected from light and moisture. pH. (2.4.24) 4.5 to 6.5.
3-Hydroxy trimethylanilinium methyl sulphate. Determine
by liquid chromatography (2.4.14).
Test solution. Dilute the injection if necessary, with water to
Neostigmine Injection contain a 0.05 per cent w/v solution of Neostigmine
Neostigmine Methylsulphate Injection Methylsulphate.
Neostigmine Injection is a sterile solution of Neostigmine Reference solution (a). Dilute 1 volume ofthe test solution to
Methylsulphate in Water for Injections. 100 volumes with water.
Neostigmine Injection contains not less than 90.0 per cent Reference solution (b). Add 0.05 ml of5 M sodium hydroxide
and not more than 110.0 per cent of the stated amount of to 1 ml of the test solution and allow to stand for 5 minutes.
neostigmine methylsulphate, C13H22N206S. Add 0.1 ml of 5 M hydrochloric acid and use immediately.

1768

www.webofpharma.com
IP 2010 NEOTAME

Chromatographic system Category. Non-nutritive sweetener.


a stainless steel column 25 em x 4.6 rom packed with
Description. A white crystalline powder.
octadecylsilane chemically bonded to porous silica
particles (5 f.!m)(such as Lichrosphere 60 RP-selectB), Identification
mobile phase: 0.0015 M solution of sodium
heptanesulphonate in a mixture of 15 volumes of A. Determine by infrared absorption spectrophotometry
acetonitrile and 85 volumes of 0.05 M potassium (2.4.6). Compare the spectrum with that obtained with neotame
dihydrogen orthophosphate adjusted to pH 3.0 with RS or with the reference spectrum ofneotame.
orthophosphoric acid, B. In the Assay, the principal peak in the chromatogram
flow rate. of 1.1 m1 per minute, obtained with the test solution corresponds to the peak in the
spectrophotometer set at 215 nm, chromatogram obtained with the reference solution.
injection volume. 10 f.!l.
In the chromatogram obtained with reference solution (b) the Tests
principal peak has a retention time of about 6.8 minutes Specific optical rotation (2.4.22). -40° to-43.4°, determined in
(neostigmine methylsulphate) and there is a peak with a a 0.5 per cent w/v solution.
relative retention time of about 0.5 (3-hydroxy)
trimethylani1inium methylsulphate. In the chromatogram Related substances. Determine by liquid chromatography
obtained with the test solution, the area of any secondary (2.4.14).
peak with a retention time corresponding to that ofthe peak Test solution. Dissolve 100 mg ofsubstance under examination
due to (3-hydroxy)trimethylanilinium methylsulphate in the in 50.0 ml ofthe mobile phase.
chromatogram obtained with reference solution (b) is not
Reference solution (a). A 0.002 per cent w/v solution of
greater than the area ofthe principal peak in the chromatogram
neotame RS in the mobile phase.
obtained with reference solution (a) (1 per cent).
Reference solution (b). A 0.003 per cent w/v solution of
Other tests. Complies with the tests stated under Parenteral
neotame impurity A RS in the mobile phase.
Preparations (Injections).
Use the chromatographic system as described under Assay.
Assay. Dilute an accurately measured volume containing about
25 mg ofNeostigmine Methy1sulphate to 50.0 ml with water. Inject reference solution (b). The test is not valid unless the
Measure the absorbance of the resulting solution at the relative standard deviation for replicate injections is not more
maximum at about 260 nm (2.4.7). Calculate the content of than 5.0 per cent.
C 13 H zzNz0 6 S taking 14.35 as the specific absorbance at Inject the test solution and reference solution (a). In the
260nm. chromatogram' obtained with the test solution, the area of
Storage. Store protected from light. peak corresponding to neotame impurity A is not more than
1.5 times the area of the principal peak in the chromatogram
obtained with reference solution (a) (1.5 percent) and the sum
of the areas of all other secondary peaks is not more than
Neotame twice the area of the principal peak in the chromatogram
obtained with reference solution (a) (2.0 per cent).
Loss on ignition (2.4.20). Not more than 0.2 per cent.
Water (2.3.43). NotmorethaiJ. 5.0 per cent, determined on 1.0 g.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve 50 mg ofsubstance under examination
in 25.0 ml ofmobile phase. Dilute 5.0 ml ofthis solution to 10.0
ml with the mobile phase.
Mol.Wt. 378.4
Reference solution. A 0.1 per cent w/v solution ofneotame RS
Neotame isN-(3,3-dimethylbuty1)-L-a-aspartyl-L- in the mobile phase.
phenylalanine 2-methy1 ester.
Chromatographic system
Neotame contains not less than 97.0 per cent and not more - a stainless steel column 10 em x 4.6 mm, packed with
than 102.0 per cent of CzoH30NzOs, calculated on the dried endcapped octadecylsilane bonded to porous silica (5
basis. f.!ID),

1769

www.webofpharma.com
NEVlRAPINE IP2010

column temperature. 45°, Test solution. Dissolve 0.1 g of the substance under
mobile phase: dissolve 3 gm of sodium 1- examination in· 100 ml of methanol.
heptanesulphonate and 3.8 ml of tri~thylamine in 750 Reference solution. Dilute 1 ml of the test solution to 100 ml
ml of water, adjust the pH to 3.5 with orthophosphoric
with methanol.
acid. Add 250 ml of acetonitrile, adjust the pH to 3.7
with orthophosphoric acid, Chromatographic system
flow rate. 1.5 ml per minute, a stainless steel column 25 cm x 4.6 mm, packed with
spectrophotometer set at 210 urn, octadecylsilane bonded to porous silica or ceramic
injection volume. 20 Ill. microparticles (5 Ilm),
mobile phase: a filtered and degassed mixture of
Inject the reference solution; The test is not valid unless the 20 volumes of methanol, 20 volumes of acetonitrile
tailing factor is not more than 2.0 and the relative standard and 60 volumes of a buffer prepared by dissolving
deviation for replicate injections is not more than 2.0 per cent. 12.0 g of sodium dihydrogen phosphate in about 800 ml
Inject the test solution and the reference solution. of water, adjusting the pH to 3.0 with phosphoric acid
and diluting to 1000.0 ml with water;
Calculate the content of C2oH30N20s.
- flow rate. 1.2 ml per minute,
Storage. Store protected from moisture. spectrophotometer set at 230 urn,
- injection volume. 20 Ill.
Inject the reference solution. The test is not valid unless the
Nevirapine column efficiency determined from the nevirapine peak is not
less than 5000 theoretical plates and the tailing factor is not
more than 2.0.
Separately inject the test solution and the reference solution.
In the chromatogram obtained with the test solution, the area
of any peak other than the principal peak is not greater than
half of the area of the principal peak in the chromatogram
obtained with the reference solution (0.5 per cent) and the
sum of the areas of all such peaks is not greater than the area
of the principal peak in the chromatogram obtained with the
Mol. Wt. 266.3 reference solution (1.0 per cent).
Nevirapine is ll-cyclopropyl-5,11-dihydro-4-methy-6H- Heavy metals (2.3.13). 1.0 g complies with the limit test for
dipyrido[3,2-b:2' ,3' -e][l,4]diazepin-6-one. heavy metals, Method B (20 ppm).
Nevirapine contains not less than 98.0 per cent and not more Sulphated ash (2.3.18). Not more than 0.2 per cent.
than 102.0 per cent ofClsHl4N40, calculated on the dried basis. Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
Category. Antiretroviral. on 1.0 g by drying at 105° for 3 hours.

Dose. 200 mg once or twice daily. Assay: Determine by liquid chromatography (2.4.14).

Description. A white or almost white crystalline powder. Test solution. A 0.005 per cent w/v solution of the substance
under examination in methanol.
Identification Reference solution. A 0.005 per cent w/v solution of
nevirapine RS in methanol.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with nevirapine RS Chromatographic system
or with the reference spectrum of nevirapine. - a stainless steel column 25- cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica or ceramic
B. In the Assay, the principal peak in the chromatogram
microparticles (5Ilm),
obtained with the test solution corresponds to that in the
mobile phase: a filtered and degassed mixture of
chromatogram obtained with the reference solution.
20 volumes of methanol, 20 volumes of acetonitrile
Tests and 60 volumes of a buffer prepared by dissolving
12.0 g of sodium dihydrogen phosphate in about 800 ml
Related substances. Determine by liquid chromatography of water, adjusting the pH to 3.0 with orthophosphoric
(2.4.14). acid and diluting to 1000.0 ml with wate/;

1770

www.webofpharma.com
IP 2010 NEVIRAPINE ORAL SUSPENSION

flow rate. 1.2 ml per minute, Test solution. To an accurately measured volume of the
spectrophotometer set at 230 urn, preparation under examination containing about 25 mg of
- injection volume. 20 ,.ti. Nevirapine add about 10 ml of methanol, mix with the aid of
Inject the reference solution. The test is not valid unless the ultrasound for 10 minutes, dilute to 50 ml with water, mix and
column efficiency determined from the nevirapine peak is not filter.
less than 3000 theoretical plates, the tailing factor is not more Reference solution. Weigh accurately about 25 mg of
than 2.0 and the relative standard deviation for the replicate nevirapine RS, add about 10 ml of methanol, mix with the aid
injections is not more than 2.0 per cent. of ultrasound to dissolve and dilute to 50 ml with water.
Separately inject the test solution and the reference solution Chromatographic system
and measure the responses for the principal peak. Calculate a stainless steel column 25 cm x 4.6 mm, packed with
the content ofClsHl4N40. octylsilyl silica gel for chromatography (5Ilm) (Such as
Storage. Store protected from moisture. Hypersil C8),
mobile phase: A. 0.1 M ammonium acetate,
B. methanol,
- flow rate. 1 ml per minute,
Nevirapine Oral Suspension a linear gradient programme using the conditions given
below,
Nevirapine Oral Suspension is a suspension ofNevirapine in spectrophotometer set at 270 nm,
a suitable flavoured vehicle. injection volume. 20 ,.ti.
Nevirapine Oral Suspension contains not less than 90.0 per Time Mobile phase A Mobile phase B
cent and not more than 110.0 per cent ofthe stated amount of (in min.) (per cent v/v) (per cent v/v)
nevirapine, ClsH14N40.
o 95 5
Usual strength. 50 mg in 5 ml. 5 95 5
25 20 80
Identification
30 20 80
A. Detennine by thin-layer chromatography (2.4.17), coating 31 95 5
the plate with silica gel GF254. 40 95 5
Mobile phase. A mixture of 40 volumes of 1-butanol, Inject the reference solution. The test is not valid unless the
30 volumes of heptane, 30 volumes of acetone and 10 volumes column efficiency determined from the nevirapine peak is not
of strong ammonia solution. less than 3000 theoretical plates and the tailing factor is not
Test solution. Dilute the preparation under examination with more than 2.0.
methanol to obtain a solution containing 1 mg ofNevirapine Inject separately the diluent (dilute 10 ml of methanol to 50 ml
perml. with water) and the test solution. Examine the diluent
Reference solution. A 0.1 per cent w/v solution of nevirapine chromatogram for any extraneous peaks and ignore the
RS in a mixture of75 volumes of methanol and 25 volumes of corresponding peaks observed in the chromatogram obtained
water. with the test solution. Ignore any peaks due to preservatives
Apply to the plate 10 III of each solution. After development, also.
dry the plate in air and examine in ultraviolet light at 254 urn. Any secondary peak observed in the chromatogram obtained
The principal spot in the chromatogram obtained with the test with the test solution should not be more than 1.0 per cent
solution corresponds to that in the -chromatogram obtained and the sum of the areas of all the secondary peaks should
with the reference solution. not be more than 2.0 per cent when calculated by percentage
B. In the Assay, the principal peak in the chromatogram area normalisation.
obtained with the test solution corresponds to the peak in the Other tests. Comply with the tests stated under Oral Liquids.
chromatogram obtained with the reference solution. Assay. Detennine by liquid chromatography (2.4.14).
Tests Test solution. Weigh accurately a quantity of the preparation
under examination containing 25 mg ofNevirapine, add abol1t
pH (2.4.24). 5.0to 7.0.
10 ml ofmethanol, mix with the aid ofultrasound for 10 minutes,
Related substances. Determine by liquid chromatography dilute to 50.0 ml with water, mix and filter. Further dilute 10.0 ml
(2.4.14). ofthe :filtrate to 25.0 ml with water.

1771

www.webofpharma.com
NEVIRAPINE TABLETS IP 2010

Reference solution. Weigh accurately about 50 mg of containing 0.05 per cent w/v ofNevirapine and filter through
nevirapine RS, add about 20 ml ofmethanol, mix with the aid a membrane filter disc with an average diameter not exceeding
of ultrasound to dissolve and dilute to 100.0 ml with water. 1.0 /lm, rejecting the first few ml ofthe filtrate.
Dilute 10.0 ml ofthis solution to 25.0 ml with water.
Reference solution. A 0.05 per cent w/v solution ofnevirapine
Use the chromatographic system described under the test for RS in the mobile phase.
Related substances.
Chromatographic system
Inject the reference solution. The test is not valid unless the a stainless steel column 25 cm x 4.6 mm, packed with
relative standard deviation for replicate injections is not more octadecylsi1ane bonded to porous silica or ceramic
than 2.0 per cent. microparticles (5/lm),
mobile phase: a filtered and degassed mixture of
Inject separately the test solution and the reference solution
20 volumes of methanol, 20 volumes of acetonitrile
and measure the responses for the princip~l peak.
and 60 volumes of a buffer prepared by dissolving
Determine the weight per ml of the oral suspension (2.4.29) 12.0 g ofsodium dihydrogen phosphate in about 800 ml
and calculate the content of CIsHI4N40 weight in volume. of water, adjusting the pH to 3.0 with orthophosphoric
Storage. Store protected from light. acid and diluting to 1000.0 ml with water;
flow rate. 1.2 ml per minute,
spectrophotometer set at 230 nm,
injection volume. 20 Ill.
Nevirapine Tablets Inject the reference solution. The test is not valid unless the
Nevirapine Tablets contain not less than 90.0 per cent and not column efficiency determined from the nevirapine peak is not
more than 110.0 per cent of the stated amount ofnevirapine, less than 7500 theoretical plates and the tailing factor is not
ClsH14N40. more than 1.5 and the relative standard deviation for replicate
injections is not more than 2 per cent.
Usual strength. 200 mg.
Inject the test solution and continue the chromatography for
Identification at least five times the retention time of the principal peak.
A. When examined in the range 200 urn to 400 nm (2.4.7) a Determine the amount of related substances by the area
0.001 per cent w/v solution in the mobile phase described normalisation method. Any individual impurity is not more
under Assay, shows an absorption maximum at about 230 urn. than 1.0 per cent and the sum ofall impurities is not more than
2.0 per cent.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the Other tests. Comply with the tests stated under Tablets.
chromatogram obtained with the reference solution. Assay. Determine by liquid chromatography (2.4.14).
Dissolution (2.5.2). Test solution. Weigh and powder 20 tablets. Shake a quantity
Apparatus No.1, of powder containing about 100 mg of Nevirapine with
Medium. 900 ml of 0.1 M hydrochloric acid, . sufficient of the mobile phase to obtain a mixture containing
Speed and time. 50 rpm and 45 minutes. 0.05 per cent w/v of Nevirapine. Mix and filter through a
membrane filter disc with an average pore diameter not greater
Withdraw a suitable volume of the medium and filter through
than 1.0 /lm, rejecting the first few ml ofthe filtrate.
a membrane filter disc with an average pore diameter not greater
than 1.0 /lm, rejecting the first few ml of the filtrate. Dilute Reference solution. A 0.05 per cent w/v solution ofnevirapine
suitably, ifnecessary. Measure the absorbance ofthe resulting RS in the mobile phase.
solution, at the maximum at about 313 urn (2.4.7). Chromatographic system
Calculate the content of CIsHI4N40 from the absorbance a stainless steel column 25 cm x 4.6 mm, packed with
obtained from a solution ofknown concentration ofnevirapine octadecylsilane bonded to porous silica or ceramic
RS in 0.1 M hydrochloric acid. microparticles (5 /lm),
D. Not less than 70 per cent ofthe stated amount ofC,sH'4N40. mobile phase: a filtered and degassed mixture of
20 volumes of methanol, 20 volumes of acetonitrile
Related substances. Determine by liquid chromatography and 60 volumes of a buffer prepared by dissolving
(2.4.14). 12.0 g ofsodium dihydrogen phosphate in about 800 ml
Test solution. Shake a quantity ofthe powdered tablets with a of water, adjusting the pH to 3.0 with phosphoric acid
suitable quantity of the mobile phase to obtain a mixture and diluting to 1000.0 ml with water;

1772

www.webofpharma.com
IP 2010 NICLOSAMIDE

flow rate. 1.2 ml per minute, cent w/v solution of ammonium sulphamate, shake, allow to
- spectrophotometer set at 230 nm, stand for 3 minutes and add 2 ml ofa 0.5 per cent w/v solution
injection volume. 20 Ill. ofN-(l-naphthyl) ethylenediamine dihydrochloride; a violet
colour is produced.
Inject the reference solution. The test is not valid unless the
column efficiency determined from the nevirapine peak is not C. Heat the substance under examination on a copper wire in
less than 7500 theoretical plates, the tailing factor is not more a non-luminous flame; a green colour is imparted to the flame.
than 1.5 and the relative standard deviation for replicate
injections is not more than 2.0 per cent. Tests
Separately inject the test solution and the reference solution Related substances. Determine by liquid chromatography
and measure the responses for the principal peale Calculate (2.4.14).
the content ofCIsHI4N40 in the tablets. .
Test solution. Dissolve 50 mg of the substance under
Storage. Store protected from moisture at a temperature not examination in 50 ml ofthe methanol.
exceeding 30°.
Reference solution. Dilute 1.0 ml ofthe test solution to 100.0
ml with acetonitrile. Dilute 1.0 ml of this solution to 20.0 ml
with acetonitrile.
Niclosamide Chromatographic system
Anhydrous Niclosamide - a stainless steel column 12.5 cm x 4.0 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm),
mobile phase: a mixture of 50 volumes of acetonitrile,

~~xrN02
50 volumes ofa solution containing 0.2 per cent w/v of
potassium dihydrogen orthophosphate, 0.1 per cent

yH CI
w/v of disodium hydrogen phosphate and 0.2 per cent
w/v of tetrabutylammonium hydrogen sulphate,
flow rate. 1 ml per minute,
spectrophotometer set at 230 nm,
- injection volume. 20 Ill.
Mol. Wt. 327.1 Inject the reference solution. Adjust the sensitivity so that
Niclosamide is 2' ,5-dichloro-4' -nitrosalicylanilide. the height of the principal peak is not less than 20 per cent of
full scale ofthe recorder.
Niclosamide contains not less than 98.0 per cent and not more
than 101.0 per cent ofC 13H gChNz0 4, calculated on the dried Inject the test solution and the reference solution. Run the
basis. chromatogram twice the retention time of the principal peak.
In the chromatogram obtained with the test solution, the sum
Category. Anthelmintic. ofthe areas ofall the secondary peaks is not more than 4 times
Dose. 2 g as a single dose after a light breakfast followed by a the area of the principle peak in the chromatogram obtained
purgative 2 hours later. with the reference solution. Ignore any peak with an area less
than 10 per cent of the area of the principal peak in the
Description. A yellowish white to yellowish, fine crystals or
chromatogram obtained with the reference solution.
powder; odourless.
Chlorides (2.3.12). T02.0 g add a mixture of40 ml of water and
Identification 1.2 ml of 5 M acetic acid, boil for 2 minutes, cool and filter;
10 ml ofthe filtrate diluted to 15 ml with water complies with
Test A may be omitted if tests Band C are carried out. Tests B
the limit test for chlorides (500 ppm).
and C may be omitted if test A is carried out.
2-Chloro-4-nitroaniline. Not more than 100 ppm, determined
A. Determine by infrared absorption spectrophotometry (2.4.6). by the following method. Boil 0.25 g with 5 ml of methanol,
Compare the spectrum with that obtained with niclosamide cool, add 45 ml of 1 M hydrochloric acid, heat to boiling,
RS or with the reference spectrum of nic1osamide.
coot filter and dilute the filtrate to 50.0 ml with 1 M
B. Heat 50 mg with 5 ml of 1 Mhydrochloric acid and 0.1 g of hydrochloric acid. To 10.0 ml ofthis solution add 0.5 ml of a
zinc powder in a water-bath for 10 minutes, cool and filter. To 0.5 per cent w/v solution of sodium nitrite and allow to stand
the filtrate add 1 ml of a 0.5 per cent w/v solution of sodium for 3 minutes. Add 1.0 ml of a 2 per cent w/v solution of
nitrite and allow to stand for 3 minutes. Add 2 ml of a 2 per ammonium sulphamate, shake, allow to stand for 3 minutes

1773

www.webofpharma.com
NICLOSAMIDE IP 2010

and add 1.0 ml ofa 0.5 per cent w/v solution ofN-(I-naphthyij B. Heat 50 mg with 5 ml of 1 Mhydrochlodc acid and 0.1 g of
ethylenediamine dihydrochloride. Any.pinldsh violet colour zinc powder in a water-bath for 10 minutes, cool and filter. To
produced is not more intense than that obtained in a solution the filtrate add 1 ml of a 0.5 per cent w/v solution of sodium
prepared at the same time and in the same manner using nitrite and. allow to stand for 3 minutes. Add 2 ml of a 2 per
10.0 ml of a solution prepared by diluting 2.0 ml of a cent w/v solution of ammonium sulphamate, shake, allow to
0.00050 per cent w/v solution of 2-chloro-4-nitroaniline in stand for 3 minutes apd add 2 ml of a 0.5 per cent w/v solution
methanol to 20 ml with 1 M hydrochloric acid and beginning ofN- (I-naphthyl) ethylenediamine dihydrochloride; a violet
at the words "add 0.5 ml of a 0.5 per cent w/v solution of colour is produced.
sodium nitrite.....".
Tests
5-Chlorosalicylic acid. Not more than 60 ppm, detenniiled by
the following method. Boil 1.0 g with 15 ml of water for Related substances. Detennine by liquid chromatography
2 minutes, cool, filter through a membrane filter (pore size (2.4.14).
0.45 !lm), wash the filter and dilute the combined filtrate and
Test solution. Shake a quantity of the powdered tablets
washings to 20 ml with water: (solution A). Dissolve 30 mg of
containing about 100 mg ofanhydrous nicolsamide with 80 ml
5-chlorosalicylic acid in20 ml of methanol and add sufficient
of methanol for 15 minutes and dilute to 100.0 ml with the
water to produce 100.0 ml. Dilute 1.0 ml of this solution to
same solvent.
100.0 ml with water (solution B). To 10.0 ml ofeach ofsolutions
A and B add separately 0.1 ml offerrie chloride solution; any Reference solution. Dilute 1.0 ml ofthe test solution to 100.0
violet colour produced in solution A is not more intense than ml with acetonitrile and further dilute 1.0 ml ofthis solution to
that obtained in solution B. 20.0 ml with acetonitrile.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Chromatographic system
a stainless steel column 10 cm x 4.6 mm, packed with
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
octadecylsilane bonded to porous silica (5 !lm) ,
on 1.0 g by drying in an oven at 105° for 4 hours.
- mobile phase: a mixture of 50 volumes of acetonitrile
Assay. Weigh accurately about 0.3 g, dissolve in 80 ml of a and 50 volumes ofa solution containing about 0.2 per
mixture of equal volumes of acetone and methanol. Titrate cent w/v ofpotassium dihydrogen orthophosphate, 0.2
with 0.1 M tetrabutylammonium hydroxide, determining the per cent w/v of tetrabutylammonium hydrogen sulphate
end-point potentiometrically (2.4.25). Carry out a blank titration. and 0.1 per cent w/v of disodium hydrogen
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to orthophosphate,
flow rate. I ml per minute,
0.03271 gofCl3HsChN204'
spectrophotometer set at 230 nm,
Storage. Store protected from light and moisture. injection volume. 20 Ill.
Inject the reference solution. Adjust the sensitivity so that
the height of the principal peak is not less than 20 per cent of
full scale of the recorder.
Niclosainide Tablets
Inject the test solution and the reference solution. Run the
Niclosamide Tablets contain not less than 95.0 per cent and chromatogram twice the retention time of the principal peak.
not more than 105.0 per cent of the stated amount of In the chromatogram obtained with the test solution, the sum
niclosamide, C13HsC12N204' The tablets may contain ofthe areas of all the secondary peaks is not more than 4 times
sweetening and flavouring agents. the area of the principle peak in the chromatogram obtained
Usual strength. 500 mg. with the reference solution. Ignore any peak with an area less
than 10 per cent of the area of the principal peak in the
Identification chromatogram obtained with the reference solution.
2-Chloro-4-nitroaniline. Not more than 100 ppm, Boil a
Heat a quantity of the powdered tablets containing 0.5 g of
quantity of the powdered tablets containing 0.1 g of
Niclosamide with 25 ml of hot ethanol (95 per cent), filter
Niclosamide with 20 ml of methanol and 20 ml ofa solution in
while hot and evaporate the filtrate to dryness on a water-
methanol containing 10 !lg of 2-chloro-4-nitroaniline, cool,
bath. The residue complies with the following tests.
add 45 ml of 1 M hydrochloric acid, heat to boiling, cool, filter
A. Detennine by infrared absorption spectrophotomeuy (2.4.6). and dilute the filtrate to 50.0 ml with 1 M hydrochloric acid.
Compare the spectrum with that obtained with niclosamide To 10.0 ml of this solution add 0.5 ml of a 0.5 per cent w/v
RS or with the reference spectrum of niclosamide. solution of sodium nitrite and allow to stand for 3 minutes.

1774

www.webofpharma.com
IP 2010 NICOTINAMIDE

Add 1.0 ml of a 2 per cent w/v solution of ammonium Identification


sulphamate, shake, allow to stand for 3 minutes and add
Test A may be omitted iftests B, C andD are carried out. Tests
1.0 ml of a' 0.5 per cent w/v solution of N-(l-naphthyl)
ethylenediamine dihydrochloriqe. Any pinkish violet colour B, C and D may be omitted if test A is carried out.
produced is not more intense than that obtained in a solution A. Determine by infrared absorption spectrophotometry (2.4.6).
prepared at the same time and in the same manner using Compare the spectrum with that obtained with nicotinamide
10.0 ml ofa solution prepared by diluting 2.0 ml ofa 0.0005 per RS or with the reference spectnllTI of nicotinamide.
cent w/v solution of 2-chloro-4-nitroaniline in methanol to
B. Heat about 5 mg in a dry tube; pyridine is evolved.
20.0 ml with 1 M hydrochloric acid and beginning at the
words "add 0.5 ml of a 0.5 per cent w/v solution of sodium C. Boil 0.1 g with 1 ml of dilute sodium hydroxide solution;
nitrite.....". ammonia is evolved.
5-ChlorosaUcyUc acid. Boil a quantity ofthe powdered tablets D. To 2 ml ofa 0.1 per cent w/v solution add 6 ml of cyanogen
containing 0.5 g of Niclosamide with 10 ml of water for bromide solution and I ml of a 2.5 per cent w/v solution of
2 minutes, cool, filter and to the filtrate add 0.2 ml offerric aniline; a golden yellow colour develops.
chloride solution; no red or violet colour is produced.
Tests
Other tests. Comply with the tests stated under Tablets.
Appearance of solution. A 5.0 per cent w/v solution in carbon
Assay. Weigh and powder 20 tablets. Weigh accurately a dioxide-fi-ee water is clear (2.4.1), and not more intensely
quantity of the powdered tablets containing about 0.3 g of coloured than reference solution BYS7 (2.4.1).
Niclosamide dissolved in 60 ml of dimethylformamide. Titrate
pH(2.4.24). 6.0 to 7.5, determined in a 5.0 per centw/v solution.
with 0.1 M teirabutylammonium hydroxide, determining the
end-point potentiometrically (2.4.25). Carry out a blank titration. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
0.03271 gofC13HsC12N204' Mobile phase. A mixture of 48 volumes of chloroform,
45 volumes of ethanol and 4 volumes of water.
Storage. Store protected from light and moisture.
Test solution. Dissolve 0.8 g of the substance under
Labelling. The label states that the tablets should be chewed
examination in 10 ml of ethanol (50 per cent).
thoroughly before swallowing.
Reference solution. A 0.02 per cent w/v solution of the
substance under examination in ethanol (50 per cent).
Apply to the plate 5 f..ll of each solution. Allow the mobile
Nicotinamide phase to rise 10 cm. Dry the plate in air and examine in ultraviolet
Niacinamide light at 254 mTI. Any secondary spot in the chromatogram
obtained with the test solution is not more intense than the
N spot in the chromatogram obtained with the reference solution.

OyNH 2
Heavy metals (2.3.13). Dissolve 0.67 gin 10 mlofwater, 7.5 ml
of 1 M hydrochloric acid and sufficient water to produce
o 25 ml. The solution complies with the limit test for heavy metals,
Method A (30 ppm).
C6H6N 20 Mol. Wt. 122.1
Chlorides (2.3.12). 1.0 g complies with the limit test for
Nicotinamide ispyridine-3-carboxamide. chlorides (250 ppm).

Nicotinamide contains not less than 99.0 per cent and not Sulphates (2.3.17). 1.2 g complies with the limit test for
more than 101.0 per cent ofC6H6N 20, calculated on the dried sulphates (125 ppm).
basis. Sulphated ash (2.3.18). Not more than 0.1 per cent.

Category. B-group vitamin. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying over phosphorus pentoxide at a pressure
Dose. Orally, prophylactic, 15 to 30 mg daily; therapeutic, 50 of 1.5 to 2.7 kPa for 18 hours.
to 250 mg daily. By intravenous injection, .50 to 250 mg daily.
Assay. Weigh accurately about 0.25 g, dissolve in 20 ml of
DescriptioR Colourless crystals or a white, crystalline powder; anhydrous glacial acetic acid, heating slightly if necessary.
odour, faint and characteristic. Add 5 ml of acetic anhydride. Titrate with 0.1 M perchloric

1775

www.webofpharma.com
NICOTINAMIDE TABLETS IP 2010

acid, using crystal violet solution as indicator. Carry out a light at 254 urn. Any secondary spot in the chromatogram
blank titration. obtained with the test solution is not more intense than the
spot in the chromatogram obtained with the reference solution.
I ml of 0.1 M perchloric acid is equivalent to 0.01221 g of
C6H6NzO. Other tests. Comply with the tests stated under Tablets.
Storage. Store protected from moisture. Assay. Weigh and powder 20 tablets. Weigh. accurately a
quantity of the powder containing about 50 mg of
Nicotinamide, shake with 50 ml of ethanol (95 per cent) for 15
minutes and dilute to 100.0 ml with ethanol (95 per cent). Mix,
Nicotinamide Tablets filter, dilute 5.0 ml ofthe filtrate to 100.0 ml with ethanol (95
Niacinamide Tablets per cent) and measure the absorbance ofthe resulting solution
at the maximum at about 262 nm (2.4.7). Calculate the content
Nicotinamide Tablets contain not less than 92.5 per cent and of C6H 6N zO taking 241 as the specific absorbance at 262 nm.
not more than 107.5 per cent of the stated amount of
nicotinamide, C6H 6N zO. Storage. Store protected from light and moisture.

Usual strength. 50 mg.

Identification Nicotinic Acid


Shake a quantity of the powdered tablets containing 0.2 g of Niacin
Nicotinamide with 50 ml of ethanol for 15 minutes, filter and
evaporate the filtrate to dryness on a water-bath. The residue N
complies with the following tests. ~")
A. Determine by infrared absorption spectrophotometry (2.4.6). ~COOH
Compare the spectrum with that obtained with nicotinamide
RS or with the reference spectrum of nicotinamide. C6HsNOz Mol. Wt. 123.1

B. Boil 0.1 g with I ml of dilute sodium hydroxide solution; Nicotinic Acid is pyridine-3-carboxylic acid.
ammonia is evolved. Nicotinic Acid contains not less than 99.5 per cent and not
C. To 2 ml ofa 0.1 per cent w/v solution add 6 ml of cyanogen more than 100.5 per cent ofC 6H sNOz, calculated on the dried
bromide solution and 1 ml of a 2.5 per cent w/v solution of basis.
aniline; a golden yellow colour develops. Category. B-group vitamin.
D. When examined in the range 230 nm to 360 nm (2.4.7), the Dose. Prophylactic, 15 to 30 mg daily; therapeutic, 50 to 250
solution obtained in the Assay shows an absorption maximum mgdaily.
only at about 262 nm and two shoulders at about 258 urn and
Description. A white or creamy-white, crystalline powder.
269nm.

Tests Identification
Related substances. Determine by thin-layer chromatography Test A may be omitted iftests B, C andD are carried out. Tests
(2.4.17), coating the plate with silica gel GF254. B, C and D may be omitted if test A is carried out.
Mobile phase. A mixture of 48 volumes of chloroform, A. Determine by infrared absorption spectrophotometry (2.4.6).
45 volumes of ethanol and 4 volumes of water. Compare the spectrum with that obtained with nicotinic acid
RS or with the reference spectrum of nicotinic acid.
Test solution. Shake a quantity of the powdered tablets
containing 0.1 g of Nicotinamide with 15 ml of ethanol for B. Heat a small quantity with twice its weight of soda lime;
15 minutes, filter, evaporate to dryness on a water-bath and pyridine is evolved.
dissolve the residue as completely as possible in 1 ml of C. Dissolve about 50 mg in 20 ml of water, neutralise to litmus
ethanol. paper with 0.1 M sodium hydroxide, add 3 ml of copper
Reference solution. Dilute 1 volume of the test solution to sulphate solution; a blue precipitate is gradually produced.
400 volumes with ethanol D. To 2 ml ofa 0.1 per cent w/v solution add 6 ml of cyanogen
Apply to the plate 5 III of each solution. Allow the mobile bromide solution and I ml of a 2.5 per cent w/v solution of
phase to rise 10 cm. Dry the plate in air and examine in ultraviolet aniline; a golden yellow colour is produced.

1776

www.webofpharma.com
IP 2010 NICOUMALONE

Tests Test solution. Shake a quantity of the powdered tablets


containing 50 mg ofNicotinic Acid with 50 ml of hot ethanol
Related substances. Determine by thin-layer chromatography
(95 per cent), filter and allow the filtrate to cool.
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 85 volumes of I-propanol, Reference solution. A 0.1 per cent w/v solution of nicotinic
10 volumes of anhydrousformic acid and 5 volumes of water.
acid RS in ethanol (95 per cent).

Test solution. Dissolve 0.2 g of the substance under Apply to the plate 5 !!l of each solution. After development,
examination in 10 ml of water. Warm slightly, ifnecessary. dry the plate in air and examine in ultraviolet light at 254 om.
The principal spot in the chromatogram obtained with the test
Reference solution. A 0.01 per cent w/v solution of the
solution corresponds to that in the chromatogram obtained
substance under examination in water.
with the reference solution.
Apply to the plate 5 !!l of each solution. After development,
B. Triturate a quantity of the powdered tablets containing
dry the plate at 105° for 10 minutes and examine in ultraviolet
50 mg ofNicotinic Acid with 10 ml of water and filter. To 2 ml
light at 254 nm. Any secondary spot in the chromatogram
ofthe filtrate add 6 ml of cyanogen bromide solution and 1 ml
obtained with the test solution is not more intense than the
of a 2.5 per cent w/v solution of aniline; a golden yellow
spot in the chromatogram obtained with the reference solution.
precipitate is produced.
Heavy metals (2.3.13). Mix 1.0 g with 1.5 ml of dilute
hydrochloric acid and sufficient water to produce 25 ml, heat C. Shake a quantity of the powdered tablets containing 0.1 g
gently and cool to room temperature. The resulting solution of Nicotinic Acid with ethanol (95 per cent), filter and
complies with the limit test for heavy metals, Method B evaporate the filtrate to dryness. Add to the residue 10 mg of
(20 ppm). citric acid and 0.15 ml of acetic anhydride and heat on a
water-bath; a reddish-violet colour is produced.
Chlorides (2.3.12). 1.0 g complies with the limit test for
chlorides (250 ppm). Tests
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Other tests. Comply with the tests stated under Tablets.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105° for 1 hour. Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.25 g of Nicotinic
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of Acid, add 40 ml of hot ethanol (95 per cent), previously
carbon dioxide-free water and titrate with 0.1 M sodium neutralised to phenolphthalein solution, and shake. Allow to
hydroxide using phenol red solution as indicator. Repeat the stand for 15 minutes, swirling occasionally, and then shake
operation without the substance under examination. The for 10 minutes. Filter through a plug of cotton and wash the
difference between the titrations represents the amount of filter with ethanol (95 per cent). Add 50 ml of carbon dioxide-
sodium hydroxide required. free water and titrate with 0.1 M sodium hydroxide using
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01231 g of phenol red solittion as indicator.
C6H sNOz.
1 ml of 0.1 M sodiilm hydroxide is equivalent to 0.01231 g of
Storage. Store protected from light and moisture. C6H sNO z. '
Storage. Store protected from light and moisture.

Nicotinic Acid Tablets


Niacin Tablets Nicoumalone
Nicotinic Acid Tablets contain not less than 92.5 per cent and
Acenocoumarol
not more than 107.5 per cent ofthe stated amount ofnicotinic
acid, C6H sNO z.
NOz
Usual strengths. 25 mg; 50 mg; 100mg.
Identification
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Mobile phase. A mixture of 48 volumes of chloroform,
45 volumes of ethanol (95 per cent) and 8 volumes of water. CI9HISN06
° Mol. Wt. 353.3

1777

www.webofpharma.com
NICOUMALONE IP 2010

Nicoumalone is (RS)-4-hydroxy-3-[ 1-(4-nitrophenyl)-3- Sulphated ash (2.3.18). Not more than 0.1 per cent.
oxobutyl]coumarin.
Loss on drying (2.4.19). Not more than 0.5 per cent; determined
Nicoumalone contains not less than 98.5 per cent and not on 1.0 g by dIying in an oven at 105°.
more than 100.5 percent ofC19HlsN06, calculated on the dried
Assay. Weigh accurately about 0.75 g, dissolve in 50 ml of
basis.
acetone and titrate with 0.1 M sodium hydroxide using
Category. Anticoagulant. bromothymol blue solution as indicator.. Repeat the operation
without the substance under examination. The difference
Dose. Initial dose, first day, 8 to 12 mg; second day, 4 to 8 mg;
between the titrations represents the amount of sodium
maintenance dose, 1 to 8 mg daily.
hydroxide required.
Description. A white to brownish-white powder; odourless or
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03533 g of
almost odourless.
C19HlsN06.

Identification Storage. Store protected from light.

A. Determine by infrared absorption spectrophotometry (2.4.6).


Compare the spectrum with that obtained with nicoumalone
RS or with the reference spectrum of nicoumalone.

B. When examined in the range 230 nm to 360 nm (2.4.7), a


Nicoumalone Tablets
0.001 per cent w/v solution in a mixture of 9 volumes of Acenocoumarol Tablets
methanol and 1 volume of 1 M hydrochloric acid shows
absorption maxima at about 283 mn and 306 nm; absorbances Nicoumalone Tablets contain not less than 90.0 per cent and
at the maxima, about 0.65 and about 0.52, respectively. not more than 110.0 per cent of the stated amount of
nicoumalone, C19HlsN06.
C. Heat 50 mg with 2.5 ml of glacial acetic acid, 0.5 ml of
Usual strengths. 1 mg; 2 mg; 3 mg; 4 mg.
hydrochloric acid and 0.2 g of zinc powder on a water-bath
for 5 minutes, cool and filter. To the filtrate add 0.05 ml of
sodium nitrite solution and add the mixture to 10 ml ofa·l per Identification
cent wlvsolution of 2-naphthol containing 3 ml of5 M sodium
A. Heat a quantity of the powdered tablets containing 50 mg
hydroxide; a bright red precipitate is produced.
ofNicoumalone with 30 ml of acetone under a reflux condenser
Tests for 5 minutes, filter and wash the residue with two quantities,
each of 10 ml, of acetone. Evaporate the combined filtrate and
Appearance of solution. A 2.0 per centw/v solution in acetone washings to 5 ml, add water dropwise until the solution
is clear (2.4.1). » becomes turbid, heat on a water-bath until the solution is
clear and allow to stand. Filter, wash the crystals with a mixture
B. A 2.0 per cent w/v solution in 0.1 M sodium hydroxide is
of equal volumes of acetone and water and dry at 100° at a
clear (2.4.1), and yellow.
pressure of2 kPa for 30 minutes.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. On the residue determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that
Mobile phase. A mixture of 50 volumes of chloroform, obtained with nicoumalone RS or with the reference spectrum
50 volumes of cyclohexane and 20 volumes of glacial acetic ofnicoumalone.
acid.
B. When examined in the range 230 nm to 360 nm (2.4.7), the
Test solution. Dissolve 0.2 g of the substance under final solution obtained in the Assay shows absorption maxima
examination in 10 ml of acetone. at about 283 nm and 306 nm.
Reference solution. A 0.002 per cent w/v solution of the C. Heat 50 mg ofthe residue obtained in testA, with 2.5 ml of
substance under examination in acetone.
glacial acetic acid, 0.5 ml of hydrochloric acid and 0.2 g of
Apply to the plate 20 J.ll of each solution. After development, zinc powder on a water-bath for 5 minutes, cool and filter. To
illy the plate in air and irmnediately examine in ultraviolet light the filtrate add 0.05 ml of sodium nitrite solution and add the
at 254 nm. Any secondary spot in the chromatogram obtained mixture to 10 ml of a 1 per cent w/v solution of 2-naphthol
with the test solution is not more intense than the spot in the containing 3 ml of 5 M sodium hydroxide; a bright red
chromatogram obtained with the reference solution. precipitate is produced.

1778

www.webofpharma.com
IP 2010 NIFEDIPINE

Tests Nifedipine
Related substances. Detennine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.

Mobile phase. A mixture of 50 volumes of chloroform,


50 volumes of cyclohexane and 20 volumes of glacial acetic OCH 3
acid.
o
Test solution. Shake a quantity of the powdered tablets
containing 20 mg of Nicoumalone with 5 ml of acetone,
centrifuge and use the supernatant liquid.

Reference solution. Dilute 1 volume of the test solution to C17HISN206 Mol. Wt. 346.3
200 volumes with acetone.
Nifedipine is dimethyll,4-dihydro-2,6-dimethyl-4-(2-
Apply to the plate 20 J1.1 of each solution. After development, nitrophenyl)pyridine-3,5-dicarboxylate. .
dry the plate in air and immediately examine in ultraviolet light Nifedipine contains not less than 98.0 per cent and not more
at 254 nm. Any secondary spot in the chromatogram obtained than 102.0 per cent of CI7HISN206, calculated on the dried
with the test solution is not more intense than the spot in the basis.
chromatogram obtained with the reference solution.
Category. Antianginal (calcium channel blocker).
Uniformity of content. Comply with the test stated under
Dose. Initial dose, 5-20 mg daily, in divided doses; subsequent
Tablets.
doses, in accordance with the needs of the patient but total
Finely crush one tablet, add 30 ml of methanol, stir the mixture daily dose not to exceed 100 mg.
for 30 minutes and filter through sintered glass, washing the Description. A yellow, crystalline powder; readily affected by
residue with three quantities, each of 15 ml, of methanol. To exposure to light.
the combined. filtrate and washings add 10 ml of 1 M
hydrochloric acid and sufficient methanol to produce NOTE- Nifedipine, when exposed to daylight and certain
100.0 ml. Ifnecessary, dilute further with a solvent prepared wavelengths of artificial light, readily converts to a
by diluting 1 volume of 1 M hydrochloric acid to 10 volumes nitrosophenyl derivative. Exposure to ultraviolet light leads
with methanol to produce a solution containing about to the formation of a nitrophenyl derivative. Pelform the
0.001 per cent w/v solution of Nicoumalone. Measure the tests and assay in the dark or under long-wavelength light
absorbance ofthe resulting solution at the maximum at about (greater than 420 nm). Use low-actinic glassware.
306 nm (2.4.7). Calculate the content OfC19HlSN06 taking 521
as the specific absorbance at 306 run. Identification
Test A may be omitted iftests B, C and D are carried out. Tests
Other tests. Comply with the tests stated under Tablets.
B, C and D may be omitted iftest A is carried out.
Assay. Weigh and powder 20 tablets. Weigh accurately a A. Detennine by infrared absorption spectrophotometry (2.4.6).
quantity of the powder containing about 10 mg of Compare the spectrum with that obtained with nifedipine RS
Nicoumalone, add 30 ml of methanol, stir the mixture for or with the reference spectrum of nifedipine.
30 minutes and filter through sintered-glass, washIng the .. .
'd .h h .. 1 f 15 1 f h I 'T' B. In the test for Related substances, the pnnclpal peak m the
resl ue WIt tree quantttles, eac 1 0 m , 0 met ana. 10 ." .
· d f'l d l' dd 10 1 f 1 M . chromatogmm obtamed WIth the test solutIon corresponds to
th e com b me I trate an was Hngs a mo.... . .
' 'd d ff" f I d the peak due to mfedlpme 111 the chromatogram obtamed WIth
hy droc hionc aCI an su IClent met tano to pro uce tl fi 1 t'
100.0 ml. Dilute 5.0 ml ofthis solution to 50.0 ml with a solvent le re erence so u Ion (a).
prepared by diluting 1 volume of 1 M hydrochloric acid to C. Detennineby thin-layer chromatography (2.4.17), coating
10 volumes with methanol and measure the absorbance ofthe the plate with silica gel GF254.
resulting solution at the maximum at about 306 nm (2.4.7). Mobile phase. A mixture of 60 volumes of cyclohexane and
Calculate the content ofC19HIsN06 taking 521 as the specific 40 volumes of ethyl acetate.
absorbance at 306 nm.
Test solution. Dissolve 0.1 g of the substance under
Storage. Store protected from light and moishlre. examination in 100 ml of methanol.

1779

www.webofpharma.com
NIFEDIPINE IP 2010

Reference solution. A 0.1 per cent w/v solution of nifedipine analogue and nifedipine (retention time about 15.5 minutes).
RS in methanol. The test is not valid unless, in the chromatogram obtained
Apply to the plate 5 !-t1 of each solution. After development, with reference solution (d), (a) the resolution factor between
dry the plate in air and examine in ultraviolet light at 254 urn. the peaks due to the nitrophenylpyridine analogue and the
The principal spot in the chromatogram obtained with the test nitrosophenylpyridine analogue is greater than 1.5, (b) the
solution corresponds to that in the chromatogram obtained resolution between the peaks due to the nitrosophenylpyridine
with the reference solution. analogue and nifedipine is greater than 1.5, and (c) the height
of the peak due to the nitrophenyl-pyridine analogue is at
D. To 25 mg add 10 ml of a mixture of 5 volumes of ethanol least 20 per cent ofthe full-scale deflection.
(95 per cent), 3.5 volumes of water and 1.5 volumes of
hydrochloric acid and dissolve with gentle heating. Add Inject the test solution and reference solutions (a) and (d) and
0.5 g of granulated zinc and allow to stand for 5 minutes, record the chromatograms for twice the retention time of
swirling occasionally. Filter, add 5 ml ofa 1per cent w/v solution nifedipine. In the chromatogram obtained with the test solution
ofsodium nitrite to the filtrate and allow to stand for 2 minutes. no secondary peak other than any peaks corresponding to
Add 2 m1 ofa 5 per cent w/v solution of ammonium sulphamate, the nitrophenylpyridine analogue and the nitrosophenylpyridine
shake vigorously with care and add 2 ml of a 0.5 per cent w/v analogue has an area greater than that of the peak due to
solution ofN-(I- naphthyl) ethylenediamine dihydrochloride; nifedipine in the chromatogram obtained with reference solution
an intense red colour develops which persists for more than (d) and the areas of any peaks corresponding to the
5 minutes. nitrophenylpyridine analogue and the nitrosophenylpyridine
analogue are not greater than the areas of the corresponding
Tests peaks in the chromatogram obtained with reference solution
Related substances. Determine by liquid chromatography (d). The total amount of related substances is not greater than
(2.4.14). 0.3 per cent. Ignore any peak with an area less than 10 per cent
ofthe area ofthe peak due to nifedipine in the chromatogram
Test solution. Dissolve 0.2 g of the substance under obtained with reference solution (d).
examination in 20 ml of methanol and dilute to 50 ml with the
mobile phase. Heavy metals (2.3.13). 2.0 g complies with limittest for heavy
Reference solution (a). Dissolve an accurately weighed metals, Method B (l0 ppm).
quantity of nifedipine RS in sufficient methanol to produce a Sulphated ash (2.3.18). Not more than 0.1 per cent.
1.0 per cent w/v solution and dilute quantitatively with the
mobile phase to obtain a 0.4 per cent w/v solution. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105° for 2 hours.
Reference solution (b). A 0.04 per cent w/v solution of
dimethyl-2,6-dimethyl-4-(2-nitrophenyl)pyridine-3, 5- Assay. Weigh accurately about 0.13 g, dissolve in a mixture of
dicaboxylate RS (nitrophenylpyridine analogue) in methanol. 25 ml of 2-methyl-2-propanol and 25 ml of 1 M perchloric
Reference solution (c). A 0.04 per cent w/v solution of acid and titrate with 0.1 M ceric ammonium sulphate, using
dimethyl- 2, 6-dimethyl-4-(2-nitrosophenyl)pyridine-3, 5- 0.1 ml offerroin solution as indicator until the pink colour is
dicarboxylate RS (nitroso- phenylpyridine analogue) in discharged, titrating slowly towards the end-point. Carry out
methanol. a blank titration.

Reference solution (d). Mix 1 volume of each of reference 1 ml of 0.1 M ceric ammonium sulphate is equivalent to
solutions (b) and (c) and 0.1 volume ofthe test solution, dilute 0.01732 g ofCI7HlsN206.
to 10 volumes with the mobile phase and then dilute 2 volumes
Storage. Store protected from light.
ofthe resulting solution to 10 volumes with the mobile phase.
Chromatographic system
- a stainless steel column 15 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 !-tm), Nifedipine Capsules
- mobile phase: 55 volumes of water, 36 volumes of
Nifedipine Capsules contain not less than 90.0 per cent and
methanol and 9 volumes of acetonitrile,
not more than 110.0 per cent ofthe stated amount ofnifedipine,
- flow rate. 1 ml per minute,
CI7HISN206.
- spectrophotometer set at 235 urn,
injection volume. 20 !-tl. Usualstrengths. 5 mg; 10 mg.
Inject reference solution (d). The peaks appear in the order NOTE - Nifedipine, when exposed to daylight and certain
nitrophenylpyridine analogue, nitrosophenylpyridine wavelengths of artificial light, readily converts to a

1780

www.webofpharma.com
IP 2010 NIFEDIPINE SUSTAINED-RELEASE TABLETS

nitrosophenyl derivative. Exposure to ultraviolet light leads with the aid ofsmall quantities of methanol. Dilute to volume
to the formation of a nitrophenyl derivative. Perform the with methanol and mix. To 20.0 ml add sufficient methanol to
tests and assay in the dark or under long-wavelength light produce 100.0 ml and mix. Measure the absorbance of the
(greater than 420 nm). Use low-actinic glassware. resulting solution at the maximum at about 350 nm (2.4.7).
Calculate the content ofC,7H,sN206 in the capsules from the
Identification absorbance obtained by repeating the operation with a
0.005 per cent w/v solution of nifedipine RS in methanol.
Determine by thin-layer chromatography (2.4.17), coating the
plate with silica gel GF254. Storage. Store protected from light.
Mobile phase. A mixture of equal volumes of ethyl acetate
and cyclohexane.
Test solution. Transfer a quantity ofthe contents ofthe capsules Nifedipine Sustained-release Tablets
containing 30 mg of Nifedipine into a centrifuge tube containing
Nifedipine Sustained-release Tablets contain not less than
0.1 M sodium hydroxide, add 25 ml of dichloromethane,
90.0 per cent and not more than 110.0 per cent of the stated
stopper the tube and shake gently for 1 hour. Centrifuge for
amountofnifedipine, CI7HISN206.
10 minutes' at 2000 to 2500 rpm. Remove the supernatant
aqueous layer by aspiration with a syringe and transfer 5 mlof NOTE - Nifedipine, when exposed to daylight and certain
the clarified lower layer to a suitable vial. wavelengths of artifiCial light, readily converts to a
nitrosophenyl derivative. Exposure to ultraviolet light leads
Reference solution (a). A 0.12 per cent w/v solution of
to the formation of a nitrophenyl derivative. Perform the
nifedipine RS in dichloromethane.
tests and the assay in the dark or under long-wavelength
Reference solution (b). A mixture of equal volumes of test light (greater than 420 nm). Use low-actinic glassware.
solution and reference solution (a).
Usualstrengths. 10 mg; 20 mg.
Apply to the plate 500 III ofeach solution as bands 20 mm by
3 mm. After development, dry the plate in air until the solvent Identification
is not detectable and immediately examine in ultraviolet light
at 254 Dm. The principal band, appearing as a dark blue band, Determine by thin-layer chromatography (2.4.17), coating the
in the chromatogram obtained with the test solution plate with silica gel GF254.
corresponds to that in the chromatogram obtained with the Mobile phase. A mixture of equal volumes of ethyl acetate
reference solution (a). Spray with a solution prepared in the and cyclohexane.
following manner. Dissolve 3 g of bismuth subnitrate and 30 g
Test solution. Transfer a quantity of the powdered tablets
of potassium iodide in 10 ml of 3 M hydrochloric acid and
containing 30 mg ofNifedipine into a centrifuge tube containing
dilute with water to 100 m1; dilute 10 ml to 100 ml with 0.3 M
0.1 M sodium hydroxide, add 25 ml of dichloromethane,
hydrochloric acid. In the chromatogram obtained with test
stopper the tube and shake gently for 1 hour. Centrifuge for 10
solution the principal band, appearing as a compact light
minutes at 2000 rpm to 2500 rpm. Remove the supernatant
orange band against a yellow background, corresponds to
aqueous layer by aspiration with a syringe and use 5 ml ofthe
that in the chromatogram obtained with reference solution (a).
clarified lower layer.
The band obtained with reference solution (b) appears as a
single band under both visualisation procedures. Reference solution. A 0.12 per cent w/v solution of nifedipine
RS in dichloromethane.
Tests
Apply to the plate 500 III of each solution as bands 20 mm by
Uniformity of content. Comply with the test stated under 3 mm. After development, dry the plate in air until the odour of
Capsules. the solvent is not detectable and immediately examine in
Transfer the contents of a capsule quantitatively to a 200-ml ultraviolet light at 254 Dm. The principal band, appearing as a
volumetric flask with the aid of methanol, dilute to volume dark blue band, in the chromatogram obtained with the test
with methanol and mix. Complete the Assay beginning at the solution corresponds to that in the chromatogram obtained
words "Measure the absorbance...." and calculate the content with the reference solution. Spray with a solution prepared in
ofC17HlsN206 in the capsule. the following manner. Dissolve 3 g of bismuth subnitrate and
30 g of potassium iodide in 10 ml of 3 M hydrochloric acid
Other tests. Comply with the tests stated under Capsules.
and dilute to 100 ml with water; dilute 10 ml ofthis solution to
Assay. Transfer the contents of 5 capsules containing about 100 ml with 0.3 M hydrochloric acid. In the chromatogram
50 mg ofNifedipine quantitatively to a 200-ml volumetric flask obtained with the test solution the principal band, appearing

1781

www.webofpharma.com
NIFEDIPINE SUSTAINED-RELEASE TABLETS IP 2010 .

as a compact light orange band againsta yellow background, Nifedipine Tablets


corresponds to that in the chromatogram obtained with
reference solution. Nifedipine Tablets contain not less than 90.0 per cent anduot
more than 110.0 per cent of the stated amount ofnifedipine,
Tests C17HISNz06' The tablets may be coated.
Usual strengths. 5 mg; 10 mg.
Dissolution (2.5.2).
A. Apparatus No.1,
NOTE - Nifedipine, when exposed to daylight and certain
wavelengths of artificial light, readily converts to a
Medium. 900 ml of 0.1 M hydrochloric acid,
nitrosophenyl derivative. Exposure to ultraviolet light leads
Speed and time. 150 rpm and 120 minutes.
to the formation of a nitrophenyl derivative. Pelform the
Withdraw a suitable volume ofthe medium and filter. Measure tests and assay in the dark or under long-wavelength light
the absorbance of the filtrate, suitably diluted with the (greater than 420 nm). Use low-actinic glassware.
dissolution medium, ifnecessary, at the maximum at about 340
run (2.4.7). Identification

Calculate the content ofC17HlsNz06 in the medium from the Detennine by thin-layer chromatography (2.4.17), coating the
absorbance obtained from a solution oflmown concentration plate with silica gel GF254.
of nifedipine RS prepared by dissolving in minimum volume Mobile phase. A mixture of equal volumes of ethyl acetate
of methanol and then diluting with the dissolution medium. and cyclohexane.
D. Not less than 25 per cent and not more than 45 per cent of Test solution. Transfer a quantity of the powdered tablets
the stated amount of C17HISNz06. containing 30 mg ofNifedipine to a centrifuge tube containing
20 ml of 0.1 M sodium hydroxide, add 25 mlof dichloromethane,
B. Apparatus No.1,
stopper the tube and shake gently for 1 hour. Centrifuge for 10
Medium. 900 ml of phosphate buffer pH 6.8 prepared by minutes at 2000 to 2500 rpm. Remove the supernatant aqueous
dissolving 30 g of sodium lawyl sulphate, 24.8 g of disodium layer by aspiration with a syringe and transfer 5.0 ml of the
hydrogen phosphate anhydrous, 2.85 g of citric acid clarified lower layer to a suitable vial.
monohydrate and 0.75 ml of orthophosphoric acid in sufficient Reference solution (a). A 0.12 per cent w/v solution of
water to produce 6000 ml. Adjust the pH to 6.8, if necessary, nifedipine RS in dichloromethane.
Replace 0.1 M hydrochloric acid with phosphate buffer pH Reference solution (b). A mixture of equal volumes ofthe test
6.8 and run the apparatus at 150 rpm for 6 hours. Withdraw a solution and reference solution (a).
suitable volume of the medium and filter. Measure the
absorbance ofthe filtrate, suitably diluted with the dissolution Apply to the plate 500 f..Ll of each solution as bands 20 mm by
medium, ifneceSSaIy, at the maximum at about 340 nm (2.4.7). 3 mrn. After development, dry the plate in air until the solvent
is not detectable and immediately examine in ultraviolet light
Calculate the content ofC17HlsNz06 in the medium from the at 254 run. The principal band, appearing as a dark blue band,
absorbance obtained from a solution oflmown concentration in the chromatogram obtained with the test solution
of nifedipine RS prepared by dissolving in minimum volume corresponds to that in the chromatogram obtained with
of methanol and then diluting with the dissolution medium. reference solution (a). Spray with a solution prepared in the
D. Not less than 60 per cent of the stated amount of following manner. Dissolve 3 g of bismuth subnitrate and 30 g
C17HISNz06. of potassium iodide in 10 ml of 3 M hydrochloric acid and
dilute with water to 100 ml; dilute 10 ml to 100 ml with 0.3 M
Other tests. Comply with the tests stated under Tablets. hydrochloric acid. In the chromatogram obtained with the
Assay. Weigh and powder 20 tablets. Weigh accurately a test solution the principal band, appearing as a compact light
quantity ofthe powder containing about 25 mg ofNifedipine, orange band against a yellow background, corresponds to
disperse in methanol, shake and dilute to 100.0 ml with that in the chromatogram obtained with reference solution (a).
methanol, filter. Dilute 20.0 ml ofthe filtrate to 100.0 ml with The band obtained with reference solution (b) appears as a
methanol. Measure the absorbance of the resulting solution single band under both visualisation procedures.
at the maximum at about 350 nm (2.4.7). Calculate the content
OfC17HISNz06 from the absorbance obtained with a 0.005 per Tests
cent w/v solution of nifedipine RS in methanol.
Uniformity of content. Comply with the test stated under
Storage. Store protected from light and moisture. Tablets

1782

www.webofpharma.com
IP 2010 NIKETHAMIDE

Shake one tablet with methanol in a 200-ml volumetric flask, C. Heat 0.1 g with I ml of 2 M sodium hydroxide; diethylamine,
dilute to volume with methanol, mix and filter. Complete the recognisable by its odour, is evolved progressively; the fumes
Assay beginning at the words "Measure the absorbance...." tum red litmus paper blue.
and calculate the content ofC'7H,sNz06 in the tablet.
D. To 2 ml ofa 0.1 percent w/v solution add 2 ml of cyanogen
Other tests. Comply with the tests stated under Tablets. bromide solution and 3 ml of a 2.5 per cent w/v solution of
Assay. Weigh and powder 20 tablets. Weigh accurately a aniline and mix; a yellow colour is produced.
quantity of the powder containing 50 mg ofNifedipine into a
200 ml volumetric flask. Dissolve with the aid of 50 mt of Tests
methanol. Dilute to volume with methanol, mix and filter. Dilute Appearance of solution. The ;ubstance, in liquid form or
20 ml ofthe filtrate to 100 ml with methanol and mix. Measure liquefied by gentle heating, is clear (2.4.1), and not more
the absorbance of the resulting solution at the maximum at intensely coloured than reference solution YS5 (2.4.1).
about 350 nm (2.4.7). Calculate the content ofC17HlsNz06 £i'om
the absorbance obtained by repeating the operation with a pH (2.4.24). 6.0 to 7.8, detelmined in a 25.0 per cent w/v solution.
0.005 per cent w/v solution of nifedipine RS in methanol.
Congealing temperature (2.4.10).23° to 24.so.
Storage. Store protected from light and moisture.
Refractive index (2.4.27). 1.522 to 1.526.
Related substances. Detennine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Nikethamide Mobile phase. A mixture of 75 volumes of chloroform and
25 volumes of I-propanol.
Test solution. Dissolve 0.4 g of tile su'bstance under
examination in 10 ml of methanol.
Reference solution (a). A 0.04 per cent w/v solution of
ethylnicotinamide RS in methanol.
Reference solution (b). A 0.004 per cent w/v solution of
ethylnicotinamide RS in methanol. .
Mol. Wt. 178.2
Apply to the plate 10 J.lI. of each solution. After development,
Nikethamide isN,N-diethylpyridine-3-carboxamide. dry the plate in air and examine in ultraviolet light at 254 nm.
Nikethamide contains not less than 99.0 per cent and not more Any spot corresponding to ethylnicotinamide in the
than 101.0 per cent ofC IDH I4N zO, calculated on the anhydrous chromatogram obtained with the test solution is not more
basis. intense than the spot in the chromatogram obta~ned with
reference solution (a) and any other secondary spot is not
Category. CNS and Respiratory stimulant.
more intense than the spot in the chromatogram obtained
Dose. By slow intravenous injection, 500 mg to 2 g repeated at with reference solution (b).
intervals of 15 to 30 minutes as necessary.
Heavy metals (2.3.13).2.0 g complies with limit test for heavy
Description. A colourless or slightly yellowish, oily liquid or metals, Method B (10 ppm):
crystalline mass; odour, slight and characteristic.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Identification Water (2.3.43). Not more than 0.3 per cent, detennined on
2.0g.
Test A may be omitted iftests B, C andD are carried out. Tests
C and D may be omitted iftests A and B are carried out. Assay. Weigh accurately about 0.15 g, dissolve in 20 ml of
anhydrous glacial acetic acid and 5 ml of acetic anhydride.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Titrate with 0.1 M perchloric acid, determining the end-point
Compare the spectrum with that obtained with nikethamide
potentiometrically (2.4.25). Carry out a blank titration.
RS or with the reference spectrum ofnikethamide.
1 ml of 0.1 M perchloric acid is equivalent to 0.01782 g of
B. When examined in the range 230 nm to 360 nm (2.4.7), a
CIDHI~ZO.
0.002 per cent w/v solution shows an absorption maximum
only at about 263 nm; absorbance at about 263 nm, about 0.57. Storge. Storeprotected from light.

1783

www.webofpharma.com
NIKETHAMIDE INJECTION IP 2010

Nikethamide Injection resulting solution at the maximum at about 263 nm (2.4.7).


Calculate the content ofC IOH l4N zO taking 282 as the specific
Nikethamide Injection is a sterile solution containing 25 per absorbance at 263 nm.
cent w/v solution ofNikethamide in Water for Injections.
Storage. Store protected from light, in single dose containers.
Nikethamide Injection contains not less than 24.0 per cent
and not more than 26.0 per cent w/v solution of nikethamide,
CIOHI~zO.
Nitrazepam
Identification
A. Make I ml alkaline with 5 M sodium hydroxide, extract with
5 ml of dichloromethane and evaporate the solvent.
On the residue determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with nikethamide RS or with the reference spectrum
ofnikethamide.
B. Gives a voluminous precipitate with alkaline potassium
mercuri-iodide solution and a greyish-brown flocculent
precipitate with tannic acid solution. Gives no precipitate
Mol. Wt. 281.3
with iodine solution or with potassium mercuri-iodide
solution. Nitrazepam is 1,3-dibydro-7-nitro-5-phenyl-2H-1 ,4-
benzodiazepin-2-one.
C. Heat I ml with 0.2 g of sodium hydroxide; diethylamine,
recognisable by its odour, is evolved. Nitrazepam contains not less than 99.0 per cent and not more
than 101.0 per cent ClsHIIN303, calculated on the dried basis.
Tests
Category. Hypnotic; sedative.
pH (2.4.24).6.0 to 8.0.
Dose. 5 to 10 mg daily, at bed time.
Related substances. Determine by thin-layer chromatography Description. A yellow, crystalline powder; odourless or almost
(2.4.17), coating the plate with silica gel GF254.
odourless.
Mobile phase. A mixture of 75 volumes of chloroform and
25 volumes of I-propanol. Identification
Test solution. Dilute 1 ml ofthe injection to 5 ml with methanol. Test A may be omitted iftests B, C andD are carried out. Tests
B, C and D may be omitted if test A is carried out.
Reference solution (a). A 0.05 per cent w/v solution of
ethylnicotinamide RS in methanol. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with nitrazepam RS
Reference solution (b). A 0.005 per cent w/v solution of
or with the reference spectrum of nitrazepam.
ethylnicotinamide RS in methanol.
B. Carry out the following procedure in subdued light.
Apply to the plate 10 III of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. When examined in the range 230 nm to 360 nm (2.4.7), a freshly
Any spot corresponding to ethylnicotinamide in the prepared 0.0005 per cent w/v solution in a 0.5 per cent w/v
chromatogram obtained with the test solution is not more solution of sulphuric acid in methanol shows an absorption
intense than the spot in the chromatogram obtained with maximum only at about 280 nm; absorbance at about 280 nm,
reference solution (a) and any other secondary spot is not about 0.45.
more intense than the spot in the chromatogram obtained
C. Dissolve 10 mg in 1 ml of methanol, warming ifnecessary,
with reference solution (b).
and add 0.05 ml of 2 M sodium hydroxide; an intense yellow
Other tests. Complies with the tests stated under Parenteral colour is produced.
Preparations (Injections).
D. Dissolve 20 mg in a mixture of 5 ml of hydrochloric acid
Assay. Dilute 5.0 ml to 500.0 m1 with water. To 5.0 ml ofthe and 10 ml of water, boil for 5 minutes, cool and add 2 ml of a
solution add 5 ml of I M hydrochloric acid and sufficient 0.1 per cent w/v solution of sodium nitrite. Allow to stand for
water to produce 500.0 ml. Measure the absorbance of the 1 minute, add 1 ml ofa 0.5 percent w/v solution of sulphamic

1784

www.webofpharma.com
IP 2010 NITRAZEPAM TABLETS

acid, mix, allow to stand for 1minute, add 1ml ofa 0.1 per cent Mobile phase. A mixture of 100 volumes of chloroform and
w/v solution of N-(l-naphthyl)ethylenediamine 10 volumes of methanol.
dihydrochloride; a red colour is produced.
Test solution. Shake a quantity of the powdered tablets with
Tests sufficient methanol to produce a solution containing 5 mg of
Nitrazepam per ml, allow to settle and decant the supernatant
Related substances. Determine by thin-layer chromatography liquid.
(2.4.17), coating the plate with silica gel GF254.
Reference solution. A 0.5 per cent w/v solution of nitrazepam
Mobile phase. A mixture of 85 volumes of nitromethane and RS in methanol.
15 volumes of ethyl acetate.
Apply to the plate 2 III of each solution. After development,
Test solution. Dissolve 0.2 g of the substance under dry the plate in air, spray it with ethanolic sulphuric acid
examination in 10 ml of acetone. (10 per cent v/v), heat at 105° for 10 minutes and examine in
Reference solution. A 0.002 per cent w/v solution of the ultraviolet light at 365 nm. The principal spot in the
substance under examination in acetone. chromatogram obtained with the test solution corresponds to
that in the chromatogram obtained with the reference solution.
Apply to the plate 10 III of each solution. Allow the mobile
phase to rise 12 cm. Dry the plate in air and examine in ultraviolet C. To a quantity of the powdered tablets containing 5 mg of
light at 254 nm. Any secondary spot in the chromatogram Nitrazepam add 5 ml of hydrochloric acid and 10 ml of water,
obtained with the test solution is not more intense than the heat on a water-bath for 15 minutes, filter and cool. To the
spot in the chromatogram obtained with the reference solution. clear filtrate add 1 ml ofa 0.1 per cent w/v solution of sodium
nitrite, allow to stand for 3 minutes and add 1 ml of a 0.5 per
Heavy metals (2.3.13). 1.0 g complies with the limit test for
cent w/v solution of sulphamic acid. Allow to stand for
heavy metals, Method B (20 ppm).
3 minutes and add 1 ml ofa 0.1 per cent w/v solution ofN-(1-
Sulphated ash (2.3.18). Not more than 0.1 per cent. naphthyl) ethylenediamine dihydrochloride; a red colour is
Loss on drying (2.4.19). Not more than 0.5 per cent, determined produced.
on 1.0 g by drying in an oven at 105° for 4 hours.
Tests
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of
acetic anhydride. Titrate with 0.1 M perchloric acid, Dissolution (2.5.2).
determining the end-point potentiometrically (2.4.25). Carry Apparatus No.1,
out a blank titration. Medium. 900 ml of 0.1 M hydrochloric acid,
1 ml of 0.1 M perchloric acid is equivalent to 0.02813 g of Speed and time. 50 rpm and 45 minutes.
ClsHIlN303. Withdraw a suitable volume ofthe medium and filter. Measure
Storage. Store protectedfrom light and moisture. the absorbance of the filtrate, suitably diluted ifnecessary, at
the maximum at about 280 nm (2.4.7). Calculate the content of
ClsHIIN303, in the medium from the absorbance obtained from
a solution of known concentration of nitrazapam RS in. the
Nitrazepam Tablets same medium.

Nitrazepam Tablets contain not less than 90.0 per cent and not D. Not less than 70 per cent of the stated amount of
more than 110.0 per cent of the stated amount ofnitrazepam, ClsH11N303.
ClsHIIN303. Related substances. Determine by thin-layer chromatography
Usual strengths. 5 mg; 10 mg. (2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 40 volumes of nitromethane,
Identification
40 volumes of toluene and 20 volumes of chloroform.
Carry out the following procedure in subdued light. Test solution. Shake a quantity of the powdered tablets
A. When examined in the range 230 nm to 360 nm (2.4.7), the containing 40 mg ofNitra'zepam with 25 ml of chloroform,
final solution obtained in the Assay shows an absorption filter, carefully evaporate the filtrate to dryness and dissolve
maximum only at about 280 nm. the residue in 2 ml of chloroform.
B. Determine by thin-layer chromatography (2.4.17), coating Reference solution. Dilute 1 volume of the test solution to
the plate with silica gel G 200 volumes with chloroform.

1785

www.webofpharma.com
NITRAZEPAM TABLETS IP 2010

Apply to the plate 5 Jll of each solution. Allow the mobile Nitrofurantoin contains not less than 98.0 per cent-and not
phase to rise 12 cm. Dry the plate in air and examine in ultraviolet morethan 102.0 per cent ofCgH6N40 s, calculated on the dried
light at 254 nm. Any secondary spot in the chromatogram basis.
obtained with the test solution is not more intense than the
Category. Antibacterial.
spot in the chromatogram obtained with the reference solution.
Dose. 50 to 100 mg four times daily.
Uniformity of content. Comply with the test stated under
Tablets. Description. Lemon yellow crystals or a crystalline powder;
odourless or almost odourless.
NOTE - CarlY out thefollowing procedure in subdued light.
Powder 1 tablet, add 5 ml of water, mix and allow to stand for Identification
15 minutes protected fi'om light. Add 70 ml of a 0.5 per cent Carry out the following test in subdued light.
v/v solution of hydrochloric acid in methanol, shake for
15 minutes protected from light, add sufficient of the A. When examined in the range 230 nm to 400 nm (2.4.7), the
hydrochloric acid solution to produce 100.0 ml and filter. final solution obtained in the Assay shows absorption maxima
Dilute 10.0 ml ofthe filtrate with sufficient ofthe hydrochloric at about 266 nm and 367 nm; the ratio ofthe absorbance at the
acid solution to produce a solution containing 0.0005 per maximum at about 367 nm to that at the maximum at about
cent w/v solution of Nitrazepam. Measure the absorbance of 266nmis 1.36 to 1.42.
the resulting solution immediately at the maximum at about B. To 1 ml ofa 0.1 per cent w/v solution in dimethylformamide
280 urn (2.4.7). Calculate the content ofC,sHIIN303 in the tablet add 0.1 ml of 0.5 M ethanolic potassium hydroxide; a brown
taking 910 as the specific absorbance at 280 nm. colour develops.
Other tests. Comply with the tests stated under Tablets.
Tests
Assay. CarlY out the following procedure in subdued light.
Related substances. Detennine by thin-layer chromatography
Weigh and powder 20 tablets. Weigh accurately a quantity of (2.4.17), coating the plate with silica gel HF254.
the powder containing about 5 mg ofNitrazepam, add 5 ml of
water, mix and allow to stand for 15 minutes protected from Mobile phase. A mixture of 90 volumes of nitromethane and
light. Add 70 ml of a 0.5 per cent v/v solution of hydrochloric 10 volumes of methanol.
acid in methanol, shake for 15 minutes protected from light, Test solution. Dissolve 0.25 g of the substance under
add sufficient of the hydrochloric acid solution to produce examination in minimum volume of dimethylformamide and
100.0 ml and filter. Dilute 10.0 ml ofthe filtrate to 100.0 rnl with dilute to 10 ml with acetone.
the same solvent arid measure the absorbance of the resulting
Reference solution. Dilute 1 volume of the test solution to
solution immediately at the maximum at about 280 nm (2.4.7).
100 volumes with acetone.
Calculate the content ofCIsHIIN303 taking 910 as the specific
absorbance at 280 nm. Apply to the plate 10 Jll of each solution. After development,
dry the plate in air, heat at 105° for 5 minutes and examine in
Storage. Store protected from light and moisture.
ultraviolet light at 254 nm. Spray with phenylhydrazine
hydrochloride solution and heat the plate at 105° for further
10 minutes. Any secondaty spot in the chromatogram obtained
with the test solution is not more intense than the spot in the
Nitrofurantoin chromatogram obtained with the reference solution by both
methods of visualisation.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). 5.0 to 7.1 per cent (hydrous fonn) and
not more than 1.0 per cent (anhydrous fonn), determined on
1.0 g by drying in an oven at 105°.
Assay. CarlY out the following procedure in subdued light.
CgH6N40 S Mol. Wt. 238.2 (anhydrous)
Weigh accurately about 75 mg and dissolve in 25.0 ml of
CgH6N40S,HzO Mol. Wt. 256.2 (hydrous)
dimethylformamide, add sufficient water to produce 500.0 ml
Nitrofurantoin is 1-(5-nitrofurfurylideneamino)imidazolidine- and mix. Dilute 5.0 ml to 100.0 ml with a solution containing
2,4-dione. It is anhydrous or contains one molecule of water 1.8 per cent w/v solution of sodium acetate and 0.14 per cent
of hydration. v/v of glacial acetic acid. Measure the absorbance of the

1786

www.webofpharma.com
IP 2010 NITROFURAZONE

resulting solution at the maximum at about 367 nm (2.4.7), acid. Measure the absorbance of the resulting solution at the
using as the blank the sodium acetate-acetic acid solution. maximum at about 367 urn (2.4.7), using as the blank the sodium
Calculate the content ofCsH 6N40 s taking 765 as the specific acetate-acetic acid solution. Calculate the content ofCsH6N 40 s
absorbance at 367 nm. taking 765 as the specific absorbance at 367 run.
Storage. Store protected from light and moisture. Storage. Store protected from light and moisture.
Labelling. The label states whether the material is anhydrous
or hydrous. .

Nitrofurazone
Nitrofurantoin Tablets
Nitofural
Nitrofurantoin Tablets contain not less than 90.0 per cent and
not more than I 10.0 per cent of the stated amount of
nitrofurantoin, CSH 6N4 0 S '
Usual strengths. 50 mg; 100 mg.

Identification
C6H6N40 4 Mol. Wt. 198.1
CarlY out the following proceduJ'e in subdued light.
Nitrofurazone is 5-nitro-2-furaldehyde semicarbazone.
A. When examined in the range 230 nm to 400 nm (2.4.7), the
final ~olution obtained in the Assay shows absorption maxima Nitrofurazone contains not less than 97.0 per cent and not
at about 266 nm and 367 nm. more than 103.0 per c.ent ofC 6H 6N 40 4 , calculated on the dried
basis.
Tests Category. Antibacterial.
Related substances. Detelmine by thin-layer chromatography
Description. A yellow to brownish-yellow, crystalline powder;
(2.4.17), coating the plate with silica gel HF254.
odourless or almost odourless.
Mobile phase. A mixture of 90 volumes of nitromethane and
10 volumes of methanol. Identification
Test solution. Shake a quantity of the powdered tablets A. Detennineby infrared absorption spectrophotometry (2.4.6).
containing 0.1 g ofNitrofurantoin with 10 ml of a mixture of Compare the spectrum with that obtained with nitrojitrazone
9 volumes of acetone and 1 volume of dimethylformamide RS or with the reference spectrum ofnitrofurazone.
and filter.
B. Dissolve 1 mg in 1 ml of dimetliylfohnamide and add 0.05 ml
Reference solution. Dilute 1 volume of the test solution to 'of 1 M ethanoUc potassium hydroxide; a ruby red colour is
100 volumes with acetone. produced.
Apply to the plate 10 III of each solution. After development,
dry the plate in air, heat at 105° for 5 minutes and examine in Tests
ultraviolet light at 254 nm. Spray with phenylhydrazine
pH (2.4.24).5.0 to 7.0, detennined in the filtrate obtained by
hydrochloride solution and heat the plate at 105° for further
shaking 1.0 g with 100 ml of carbon dioxide-free water and
10 minutes. Any secondary spot in the chromatogram obtained
filtering.
with the test solution, is not more intense than the spot in the
chromatogram obtained with the reference solution by both Related substances. Detennine by liquid chromatography
methods of visualisation. (2.4.14).
Other tests. Comply with the tests stated under Tablets. Test solution. Dissolve 100 mg of the substance under
examination in 100.0 ml ofthe mobile phase.
Assay. Carry out the following procedure in sub.dued light.
Reference solution (a). A 0.0005 per cent w/v solution of(5-
Weigh and powder 20 tablets. Weigh accurately a quantity of
nitro-2-fil1yl)methylene diacetate (Nitrofurazone impurity
the powder containing about 0.15 g of Nitrofurantoin, add
B) in the mobile phase.
50.0 ml of dimethylformamide, shake for 5 minutes, add
sufficient water to produce 1000.0 ml and mix. Dilute 5.0 rnI to Reference solution (b). Dissolve 10 mg each ofnitrojitral RS
100.0 ml with a solution containing 1.8 per cent w/v solution and nitrojitrantoin in 100 ml of the mobile phase. Dilute 5 ml
of sodium acetate and 0.14 per cent v/v of glacial acetic of this solution to 100 ml with the mobile phase.

1787

www.webofpharma.com
NITROFURAZONE IP 2010

Chromatographic system Category. General anaesthetic.


a stainless steel column 25 em x 4.6 mm, packed with Dose. By inhalation, 60 to 80 per cent, with oxygen 20 to 40 per
octadecylsilane bonded to porous silica (5 /!m), cent, as required.
- mobile phase: a mixture of 40 volumes of acetonitrile
and 60 volumes of water, Description. A colourless gas; odourless.
- flow rate. 1 ml per minute,
- spectrophotometer set at 310 nm, Identification
- injection volume. 20 Ill. A. A glowing splinter of wood bursts into flame on contact
Inject reference solution (b). The test is not valid unless the with the gas.
resolution between the peaks due to nitrofural and B. Shake with alkaline pyrogallol solution; the gas being
nitrofurantoin is not less than 2.0. examined is not absorbed and the solution does not become
Inject the test solution and reference solution (a). Run the brown.
chromatogram 10 times the retention time ofnitrofural. In the
chromatogram obtained with the test solution, the area of any Tests
secondary peak is not more than the area ofthe principal peak
Acidity or alkalinity. Use hermetically-closed, flat-bottomed,
in the chromatogram obtained with reference solution (a) (0.5
glass cylinders with dimensions such that 50 ml of liquid
per cent) and the sum of the areas of all the secondary peaks
reaches a height of 12 to 14 em, fitted with an outlet tube and
is not more than twice the area of the principal peak in the
with an inlet tube with an orifice of 1 mm in internal diameter
chromatogram obtained with reference solution (a) (1.0 per
reaching to within 2 mm of the bottom of the cylinder. For
cent). Ignore any peaks with the area less than 0.05 times the
solution (1) pass 2.0 litres ofthe gas under examination through
area ofthe principal peak in the chromatogram obtained with
a mixture of 0.1 ml of O. 01 M hydrochloric acid and 50 ml of
reference solution (a) (0.025 per cent).
carbon dioxide-free water. For solution (2) use 50 ml of carbon
Sulphated ash (2.3.18). Not more than 0.1 per cent. dioxide-free water. For solution (3) add 0.2 ml of 0.01 M
Loss on drying (2.4.19). Not more than 0.5 per cent, determined hydrochloric acid to 50 ml of carbon dioxide-free water. To
on 1.0 g by drying in an oven at 105 0 • each solution add 0.1 ml of a 0.02 per cent w/v solution of
methyl red in ethanol (70 per cent). The intensity ofthe colour
Assay. Carry out the following procedure in subdued light.
of solution (1) is between those of solutions (2) and (3).
Weigh accurately about 60 mg, add 20.0 ml of
dimethylformamide, swirl to dissolve and add sufficient water Arsine and phosphine. Through a mercuric chloride paper
to produce 500.0 m!. Dilute 5,0 ml ofthe solution to 100.0 ml attached to a glass tube as in the limit test for arsenic (2.3.10),
with water and mix. Measure the absorbance of the resulting pass 2.0 litres of the gas; no visible stain is produced.
solution at the maximum at about 375 nm (2.4.7). Calculate the Carbon dioxide. Not more than 300 ppm v/v determined by the
content ofC 6H 6N 40 4 taking 822 as the specific absorbance at following method. Use the apparatus described in the test for
375nm. Acidity or alkalinity. Pass 1.0 litre through 50 ml ofclear barium
Storage. Store protected from light and moisture. hydroxide solution. Any turbidity produced in the resulting
solution is not more. than that obtained in a reference solution
prepared at the same time by adding 1 ml of a 0.11 per cent
Nitrous Oxide w/v solution of sodium bicarbonate in carbon dioxide-free
water to 50 ml of barium hydroxide solution.
Mol. Wt. 44.0
Carbon monoxide. Notmore than 10 ppm v/v, determined by
Nitrous Oxide contains not less than 98.0 per cent v/v ofNzO. the following method. Connect in series a U-tube containing
NOTE - Carry out thefollowing tests on afull cylinderfrom silica gel impregnated with chromium trioxide, a drechsel
which no gas has been withdrawn. The cylinder from which bottle containing 100 ml of a 40 per cent w/v solution of
the gas is taken should be kept at room temperature for not potassium hydroxide, a U-tube containing pellets of
less than 6 hours before carrying out the tests. Keep the potassium hydroxide, a U-tube containing phosphorus
cylinder in the vertical position with the outlet valve pentoxide dispersed on previously granulated, fused pumice,
uppermost and deliver the gas at a rate of 4 litres per hour, a tube containing iodine pentoxide in granules, previously
unless otherwise directed. The test for Carbon monoxide dried at 200 0 and kept at a temperature of 1200 , packed in I-em
should be carried out on the first portion ofgas drawn from columns separated by I-em columns of glass wool giving an
the cylinder and that for Nitric oxide and nitrogen dioxide effective length of 5 em, and a flask containing 2.0 ml of 1 M
immediately thereafter. potassium iodide and 0.15 ml of starch solution.

1788

www.webofpharma.com
IP 2010 NORADRENALINE BITARTRATE

Flush the apparatus with 5.01itres of carbon dioxide-free air Water. Pass a measured quantity at a rate of6litres per hour
and, if necessary, discharge the blue colour in the iodide through an absorption tube containing magnesium
solution by adding a small quantity of freshly prepared perchlorate; the increase in weight ofthe tube does not exceed
0.002 M sodium thiosulphate. Continue flushing until not 2 mg per litre ofgas, the initial and final weighings ofthe tube
more than 0.045 ml of0.002 M sodium thiosulphate is required being made when the air in it has been displaced by the nitrous
after passing 5.0 litres ofcarbon dioxide-free air. Pass 5.0 litres oxide.
ofthe gas under examination through the apparatus and flush Assay. Carry out the assay of nitrous oxide (2.3.32), using
the last traces of liberated iodine into the reaction flask by 100 ml ofthe gas under examination. Use a cylinder ofthe gas
passing through the apparatus 1.0 litre of carbon monoxide- under examination from which at least 1 per cent w/w of the
free air. Titrate the liberated iodine with 0.002 M sodium contents have been removed.
thiosulphate. Carry out a blank titration under the same
Storage. Store under pressure in metal cylinders of the type
conditions, using 5.0 litres of carbon dioxide-free air. The
conforming to the appropriate safety regulations and at a
difference between the volumes of 0.002 M sodium
temperature not exceeding 37°.
thiosulphate used in the two titrations is not greater than
1.0rnl. Labelling. The cylinder is painted blue and carries a label
stating "Nitrous Oxide". In addition, ''Nitrous Oxide" or the
Halogens and hydrogen sulphide. Pass a volume containing symbol ''N20'' should be stencilled in paint on the shoulder of
1.0 litre measured at 25° and at 101.3 kPa through a mixture of the cylinder. .
100 ml of water and 1 ml of silver nitrate solution; neither
opalescence nor darkening is produced.
Nitric oxide and nitrogen dioxide. Not more than 2 ppm v/v in Noradrenaline Bitartrate
both the liquid and gaseous phases, determined by the
following method. Use two of the cylinders described in the Noradrenaline Acid Tartrate; Levarterenol Bitartrate;
test for Acidity or alkalinity connected in series. Examine Norepinephrine Bitartrate
separately both the liquid and gaseous phases of the gas
OH
under examination. To obtain the liquid phase invert the
cylinder. The liquid vaporises on leaving the valve.
For solution A dissolve 1 g ofsulphanilic acid in a mixture of
10 ml ofglacial acetic acid and 180 ml ofwater. For solution
0
HO~ .
NH2 H OH
HOOe)( eOOH ,H 20
. HXOH
B dissolve 0.2 g of N-(l-naphthyl)ethylenediamine OH
dihydrochloride in 10 ml of a 50 per cent v/v solution of CSHIIN03,C4Ht;06,H20 Mol. Wt. 337.3
glacial acetic acid, heating gently, and dilute to 200 ml with
water. Mix 9 volumes ofsolution A with 1 volume ofsolution Noradrenaline Bitartrate is (R)-2-amino-l-(3,4-
B (reagent A). dihydroxyphenyl)ethanol tartrate monohydrate.
Noradrenaline Bitartrate contains not less than 98.5 per cent
In the first cylinder place 15 ml ofa solution containing 2.5 per
and not more than 101.0 per cent of CSHllN03,C4H606,
cent w/v solution of potassium permanganate and 1.2 per
calculated on the anhydrous basis.
cent v/v ofsulphuric acid (96 per cent). Place 20 ml ofreagent
A in the second cylinder and connect the outlet tube of the Category. Sympathomimetic.
first cylinder to the inlet tube of the second cylinder. Pass
Dose. By intravenous infusion, 2 to 20 /Lg per minute, according
2.5 litres ofthe gas under examination through the reagents at
to the blood pressure of the patient.
a rate of 15 litres per hour. Prepare a reference solution by
adding 0.25 ml of a 0.00616 per cent w/v solution ofsodium Description. A white or almost white, crystalline powder;
nitrite to 20 ml of reagent A. Allow both the sample and odourless. It gradually darkens on exposure to air and light.
reference solutions to stand for 10 minutes. For both liquid
and gaseous phases, any red colour in the sample solution is Identification
not more intense than that in the reference solution. Test A may be omitted iftests B, C, D, E andF are carried out.
Oxidising substances. Pass a volume containing 2.0 litres Tests C, D, E may be omitted iftests A, BandF are carried out.
measured at 25° and at 101.3 kPa through a freshly prepared A. Dissolve 0.2 g in 2 ml ofwater containing about 10 mg of
solution of 0.5 g of soluble starch and 0.5 g of potassium sodium sulphite and add sufficient dilute ammonia solution
iodide in 100 ml ofwater containing 0.05 ml ofglacial acetic to give an alkaline reaction. Keep the mixture at about 4° for
acid; the colour of the liquid is not changed. 1 hour and filter.

1789

www.webofpharma.com
NORADRENALINE BITARTRATE IP 2010

On the residue (residue R) determine by infrared absorption bands 20mm by 2 mm. Allowthe applied bands to dry in air,
spectrophotometry (2,4,6). Compare the spectrum with that spray them with a saturated solution of sodium bicarbonate,
obtained with noradrenaline acid tartrate RS treated in the allow to dry in air and spray the' bands twice with acetic
same manner. anhydride, drying between the two sprayings. Heat the plate
B. When examined in the range 230 nm to 360 nm (2.4.7), a at. 50° for 90 minutes and develop the chromatograms. After
0.005 per cent w/v solution in 0.01 M hydrochloric acid shows removal of the plate, allow it to dry in air and spray with a
an absorption maximum at about 279 nm; absorbance at about freshly prepared mixture of8 volumes of methanol, 2 volumes
279 nin, about 0.40. of ethylenediamine and 2 volumes of a 0.5 per cent w/v
solution of potassium ferricyanide. Dry the plate at 60° for
C. Wash residue R obtained in test A with three quantities,
10 minutes and examine in ultraviolet light at 254 and 365 nm.
each of2 ml, of water, followed by 5 ml of ethanol (95 per
, In the chromatogram obtained with the test solution any band
cent) and 5 ml of ether and dry the precipitate under preSSUTe
with a slightly higher R r value than the principal band is not
of 1.5 to 2.5 kPa for 3 hours. The specific optical rotation
more intense than the corresponding band in the chromatogram
(2.4.22), determined in a 2.0 per cent w/v solution,ofthe dried
obtained with reference solution (b). The chromatogram
precipitate in 0.5 M hydrochloric acid is -44° ,to -48°.
obtained with reference solution (c) shows a clearly separated
D. To 1 ml of a 1 per cent w/v solution,add 0,05 ml offerric band corresponding to the most intense band in the
chloride solution; an intense green colour is produced. Add, chromatogram obtained with reference solution (a) at a higher
drop by drop, sodiwn bicarbonate solution; ,the colour R r value than the most intense band.
changes to blue and then red.
Noradrenalone. Absorbance of a 0.2 per cent w/v solution in
E. To 1 ml ofa 0.1 per cent w/v solution add 10 ml ofphthalate
0.01 M hydrochloric acid at 310 nm, not more than 0.40 (2.4.7).
btifferpH 3.6, add 1 ml of O. 05 M iodine, setaside for 5 minutes , . . ' :

and add 2 ml of 0.1 M sodium thiosulphate; not more than a Sulph~ted ash (2.3.18). Not more than 0.1 percent.
faint red colour is produced; Repeat the test using buffei· Water (2.3.43). 4.5 to 5.8 per cent, determined on 0.5 g.
solution pH 6.6 instead of phthalate buffer pH 3.6; a strong
reddish violet colour is produced (distinction from adrenaline Assay. Weigh accurately about 0.6 g, dissolve in 50 ml of
and isoprenaline). anhydrous glacial acetic acid, warming if necessary.Titrate
with 0.1 M perchloric acid, using Clystal violet solutiOn as
F. The filtrate obtained in test A gives the reactions oftartrates indicator, until a bluish green colour is obtained. Carry out a
(2.3.1).
blank titration.
Tests 1 ml of 0.1 M perchloric acid is equivalent to 0.03193 g of
Appearance of solution. A freshly prepared 2.0 per cent w/v CgHIIN03,CJf606.
solution is clear (2.4.1), and not more intensely coloured than Storage. Store protected from moisture.
reference solution BYS5 (2.4.1).
pH (2.4.24). 3.5 to 5.0, detennined in a 1.0 per centw/v solution.
Melting range (2.4.21). 100° to 106°, with decomposition. Noradrenaline Bitartrate Injection
Adrenaline. Determine by thin-layer chromatography (2.4.17),
Noradrenaline Acid Tartrate Injection; Noradrenaline
coating the plate with silica gel G
Injection; Levarterenol Bitartrate Injection;
Mobile phase. A mixture of50 volumes of acetone, 50 volumes Norepinephrine Bitartrate Injection
of dichloromethane and 0.5 volume ofanhydrousformic acid.
Noradrenaline Bitartrate Injection is a sterile solution of
Prepare the following solutions immediately before use. Noradrenaline Bitartrate. It is prepared by diluting Sterile
Test solution. Dissolve 0.25g of the substance under Noradrenaline Concentrate to 250 times its volume with Sodium
examination in 10 ml of water. Chloride and Dextrose Injection or with Dextr'ose Injection
Reference solution (a). A 0.125 per cent w/v solution of (5 per cent w/v) immediately before use.
adrenaline tartrate RS in water. Noradrenaline Bitmtrate Injection contains in 1 ml 8 Ilg of
Reference solution (b). A 0.025 per cent w/v solution of Noradrenaline Bitmtrate equivalent to approximately 4 Ilg of
adrenaline tartrate RS in water. noradrenaline.
Reference solution (c). A mixture of equal volumes ofthe test
solution and reference solution (b).
Tests
Apply to the plate 6 III of each of test solution, reference Other tests. Complies with the tests stated under Parenteral
solutions (a) and (b) and 12 III of reference solution (c) as Preparations (Injections).

1790

www.webofpharma.com
IP 2010 NORETHISTERONE

Sterile Noradrenaline Concentrate Norethisterone contains not less than 98.0 per cent and not
more than 102.0 per cent of CZOHZ60Z, calculated on the dried
Sterile Noradrenaline Concentrate is asterile, isotonic solution
basis.
containing 0.2 per cent w/v of Noradrenaline Bitartrate in
Water for Injections. Category. Progestogen.
.. ,.

Sterile Noradrenaline C,onceiltr.ate contains not less than Dose. 5 to 20 mg daily, in single or divided doses.
0.18 per cent and not more than 0.23 per cent w/v of
Description. A white or yellowish-white, crystalline powder;
noradrenaline bitartrate, CgHIIN03,C4H606,HzO.
odourless.
Identification
Identification
Mix 0.5 ml with 10 ml ofphthalate bufferpH 3.6, add l'ml of
A. Detennine by inli"ared absorption spectrophotometry (2.4.6).
0.05 M iodine, allow to stand for 5 minutes and add 2 ml of
Compare the spectrum with that obtained with norethisterone
0.1 M sodium thiosulphate; not more than a velY faint red
RS or with the reference spectrum of norethisterone.
colour is produced. Repeat the test using phosphate buffer
pH 6.6 instead of phthalate buffer pH 3.6; a strong reddish B. Determine by thin~layer chromatography (2.4.17), coating
violet colour is produced; the plate with silica gel G: '

Tests Solvent n1ixture. A mixture of 90 volumes of acetone and


10 volliines offormamide.
pH (2.4.24). 3.0 to 4.6.
Mobile phase. A mixture of 40 volumes of hexane and
Other tests. Complies with the tests stated under Parenteral 10 volumes of dioxan.
Preparations (Concentrated Solutions for Injection).
Test solution. Dissolve 10 mg of the substance under
Assay. Dilute 5.0 ml to 200.0 ml with water and measure the examination in 10 ml of chloroform.
absorbance ofthe resulting solution at the maximum at about
Reference solution. Dissolve 10 mg of norethisterone RS in
279 nm (2.4.7). Calculate the content ofCgH"N03,C4H606,HzO
taking 80 as the specific absorbance at 279 nm.. 10 nil of chloroform.

Storage. Store protected from light, in single dose containers. Place the dry plate in a tank containing a shallow layer of the
solvent mixture, allow the solvent mixture to ascend to the
Labelling. The label states (1) "Sterile Noradrenaline top, remove the plate from the tank and allow the solvent to
Concentrate"; (2) that 1 voillme of the solution diluted to evaporate. Use within 2 hours, with the flow of the mobile
250 volumes with Sodium Chloride and Dextrose Injectionor phase in the direction in which the aforementioned treatment
with Dextrose Injection (5 per cent w/v) produces was done.
Noradrenaline Bitartrate Injection, which must be used
immediately after preparation; (3) that ifthe solution isbrbwn Apply to the plate 2 J..lI of each solution. Allow the mobile
it should not be used. phase to l~ise 12 cm. DIy the plate in a current ofwann air, allow
the solvent to evaporate, heat at 1200 for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at l20~ for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
Norethisterone spot in the chromatogram obtained with the test solution
Norethindrone corresponds to and exhibits fluorescence similar to that in tIie
chromatogram obtained with the reference solution.
OH C. Dissolve about 2 mg in 2 ml of ethanol (95 per cent) and
-C=CH add 1 ml of a 1 per cent w/v solution of butylated
hydroxytoluene in ethanol (95 per cent) and 2 ml of I M
sodium hydroxir;fe. Heat in a water~bath for 30 minutes and
cool; a yellowi,sh pink colour is produced. '
o Tests
Mol. Wt. 298.4 Appearance of solution. Dissolve 0.2 g in sufficient dioxan to
Norethisterone is 17~-hydroxy-19-nor-17 a-pregn-4-en-20-yn- produce 10 ml (solution A). The solution is clear (2.4.1), and
3-one. not more intensely coloured than reference solution YS6 (2.4.1).

1791

www.webofpharma.com
NORETHISTERONE IP 2010

Specific optical rotation (2.4.22). -33.0° to -37.0°, determined Assay. Weigh accurately about 0.4 g," dissolve in 40 ml of
in a solution prepared by diluting 5.0 ml of solution A to tetrahydrofuran, add 10 ml of a 10 per cent w/v solution of
10.0 ml with dioxan. silver nitrate and titrate with 0.1 M sodium hydroxide using
Light absorption. Dissolve 10 mg in sufficient ethanol 2 ml of bromocresol green solution as indicator. Repeat the
(95 per cent) to produce 100 ml, dilute 10 ml ofthe solution to operation without the substance under examination. The
100 ml with methanol (98 per cent). Absorbance of the difference between the titrations represents the amount of
resulting solution at the maximum at about 240 nm, 0.55 to 0.59 sodium hydroxide required.
(2.4.7). 1 ml of 0.1 M sodium hydroxide is equivalent to 0.02984 g of
Related substances. Determine by liquid chromatography CZOHZ60Z'

(2.4.14). Storage. Store protected from light and moisture.


Solvent mixture. A mixture of 40 volumes of water and 60
volumes of acetonitrile.
Test solution. Dissolve 25 mg of the substance under
examination in 10 ml ofthe solvent mixture. Norethisterone Tablets
Reference solution. Dilute 1.0 ml ofthe test solution to 100.0
Norethindrone Tablets
ml with the solvent mixture. Dilute 1.0 ml ofthis solution to
10.0 ml with the solvent mixture. Norethisterone Tablets contain not less than 90.0 per cent
and not more than 110.0 per cent of the stated amount of
Chromatographic system
norethisterone, CZOHZ60Z'
- a stainless steel column 15 cm x 4.6 mm, packed with
end-capped octylsilane bonded to porous silica (5 J!m), Usual strength. 5 mg.
mobile phase: A. water,
B. acetonitrile, Identification
a linear gradient programme using the condition given
below, Place a quantity of the powdered tablets containing 25 mg of
flow rate. 1 ml per minute, Norethisterone on a small filter, wash with three quantities,
spectrophotometer set at 254 nrn, each of 5 ml, of light petroleum (60° to 80°) and discard the
injection volume. 20 J!l. washings. Extract the residue with 15 ml of chloroform,
evaporate the extract to dryness and recrystallise from aqueous
Time Mobile phase A Mobile phase B
methanol. The residue complies withthe following test.
(in min.) (per cent w/v) (per cent w/v)
0-20 63 37 Determine by thin-layer chromatography (2.4.17), coating the
plate with silica gel G.
20-25 63-20 37-80
Solvent mixture. A mixture of 90 volumes of acetone and
25-35 20 80
10 volumes of 1,2-propanediol.
35-36 20-63 80-37
Mobile phase. A mixture of40 volumes of cyclohexane and 10
36-50 63 37
volumes of toluene.
Inject the test solution and the reference solution. In the
Test solution. Dissolve 25 mg of the substance under
chromatogram obtained with the test solution, the area of any
examination in 10 ml ofthe solvent mixture.
secondary peak is not more than twice the area ofthe principal
peak in the chromatogram obtained with the reference solution Reference solution (a). Dissolve 25 mg of norethisterone RS
(0.2 per cent) and the sum of the areas of all the secondary in 10 ml ofthe solvent mixture.
peaks is not more than 3 times the area ofthe principal peak in
Reference solution (b). Mix equal volumes ofthe test solution
the chromatogram obtained with reference solution (0.3 per
and reference solution (a).
cent). Ignore any peaks with the area less than 0.5 times the
area of the principal peak in the chromatogram obtained with Place the dry plate in a tank containing a shallow layer of the
the reference solution (0.05 per cent). solvent mixture, allow the solvent mixture to ascend to the
top, remove the plate from the tank and allow the solvent to
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
evaporate. Use within 2 hours, with the flow of the mobile
Loss on drying (2.4.19). Not more than 0.5 per cent, determined phase in the direction in which the aforementioned treatment
on 1.0 g by drying in an oven at 105° for 3 hours. was done.

1792

www.webofpharma.com
IP 2010 NORFLOXACIN

Apply to the plate 2 J.ll of each solution. Allow the mobile N orfloxacin
phase to rise 12 cm. Dry the plate in a current ofwarm air, allow
the solvent to evaporate, heat at 120 0 for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 1200 for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with
reference solution (a). The principal spot in the chromatogram
obtained with reference solution (b) appears as a single,
compact spot.
Mol. WUI9.3
Tests
Norfloxacin is l-ethyl-6-fluoro-4-oxo-7-(piperazin-l-yl)-l ,4-
Uniformity ofcontent. (For tablets containing 10 mg or less)- dihydroquinoline-3-carboxylic acid.
Comply with the test stated under Tablets. Norfloxacin contains not less than 99.0 per cent and not more
Carry out the procedure described under Assay but using the than 101.0 per cent ofC16H18FN303, calculated on the dried
following test solution. basis.
Category. Antibacterial.
Test solution. Powder one tablet and dissolve as completely
as possible in 2 ml of water with the aid of ultrasound for 15 Dose. 400 mg to 800 g daily, in divided doses.
minutes and dilute to 5.0 ml with methanol. Centrifuge for 15 Description. A white to pale yellow, crystalline powder.
minutes and use the clear supernatant liquid.
Calculate the content of C2oH2602 in the tablet. Identification
Other tests. Comply with the tests stated under Tablets. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with norfloxacin
Assay. Determine by liquid chromatography (2.4.14). RS or with the reference spectrum ofnorfloxacin.
Test solution. Weigh and powder 20 tablets. Disperse a B. When examined in the range 230 nm to 360 urn (2.4.7), a
quantity ofpowder containing about 5 mg of Norethisterone 0.0005 per cent w/v solution in 0.1 M sodium hydroxide shows
in 10 ml of water, sonicate for 15 minutes and dilute to 25.0 ml an absorption maximum at about 273 urn.
with methanol, filter.
Tests
Reference solution. A 0.02 per cent w/v solution of
norethisterone RS in methanol. Related substances. Determine by thin-layer chromatography
Chromatographic system (2.4.17), coating the plate with silica gel G, previously washed
- a stainless steel column 25 cm x 4.6 mm, packed with with methanol and dried.
octadecylsilane bonded to porous silica (10 J.lm) (Such Mobile phase. A mixture of 40 volumes of dichloromethane,
as Spherisorb ODS 1), 40 volumes of methanol, 20 volumes of toluene, 14 volumes
mobile phase: a mixture of28 volumes of water and 72 of diethylamine and 8 volumes of water.
. volumes of methanol, Test solution. Dissolve 0.8 g of the substance under
- flow rate. 1 ml per minute, examination in 100 rnl ofa mixture ofequal volumes ofmethanol
- spectrophotometer set at 254 urn, and dichloromethane.
- injection volume. 20 J.ll.
Reference solution. Dissolve 4.0 mg ofnorfloxacin RS in 1 ml
Inject the reference solution. The test is not valid unless the of glacial acetic acid, add 4 ml of methanol and mix; dilute
relative standard deviation for replicate injections is not more 1 mlofthe solution with 9 ml ofamixture ofequal volumes of
than 2.0 per cent. methanol and dichloromethane (reference solution A). Dilute
Inject the reference solution and the test solution. a portion ofreference solution A with an equal volume ofthe
methanol-dichloromethane mixture (reference solution B).
Calculate the content of C2oH2602 in the tablets.
Apply separately to the plate spots of the three solutions in
Storage. Store protected from light and moisture. quantities indicated below. For spot 1 use 5 J.lI of the test

1793

www.webofpharma.com
NORFLOXACIN IPZ010

solution; for spots 2, 3 and 4 use 1 J.ll, 1.5 J.ll and2 J.ll respectively Other tests. Comply with the tests stated under Eye Drops.
ofreference solution A; for spot 5 use 5 J.ll ofreference solution
Assay. Detennine by liquid chromatography (2.4.14).
B. Place the plate in a paper-lined chamber previously
equilibrated with the mobile phase and allow the solvent front Test solution. Dilute a suitable volume ofthe eye drops with a
to move about nine-tenths of the length of the plate. After 0.1 per cent v/v solution of orthophosphoric acid to produce
development, dry the plate in air and examine in ultraviolet a solution containing 0.Q05 per cent w/v ofNorfloxacin.
light at 254 nm and 365 run. Compare the intensities of any Reference solution. A 0.005 per cent w/v solution of
secondary spots in the chromatogram obtained with the test nOifloxacin RS inO.1per cent v/v solution of orthophosphoric
solution with those ofthe principal spots (2), (3), (4) and (5). acid.
The sum of the intensities of secondary spots obtained with
the test solution is not more than 0.5 per cent of impurities. Chromatographic system
(The spots (2) (3) (4) and (5) represent 0.2 per cent, 0.3 per a stainless steel column 30 cm x 3.9 mm, packed with
cent, 0.4 per cent and 0.5 per cent respectively of impurities). octadecylsilane bonded to porous silica (10 J.lm) (Such
as Bondapack C 18),
Heavy metals (2.3.13).1.33 g complies with the limit test for column temerature. 50°,
heavy metals, Method B (15 ppm). mobile phase: a mixture of 300 volumes of methanol
Sulphated ash (2.3.18). Not more than 0.1 per cent, detennined and 700 volumes of 0.1 per cent v/v orthophosphoric
on 1.0 g in a platinum crucible. acid, .
flow rate. 2 ml per minute,
Loss on drying (2.4.19). Not more than 1.0 per cent, detennined
spectrophotometer set at 280 nm,
on 0.5 g by drying in an oven at 100° at a pressure not exceeding
- injection volume. 20 J.ll.
0.7kPa.
Precondition the column using 0.01 M anhydrous sodium
Assay. Weigh accurately about 0.3 g and dissolve in 100 ml of
dihydrogen orthophosphate, adjusted to pH 4.0 with
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric orthophosphoric acid, at a flow rate of0.5 ml per minute for 8
acid, detennining the end-point potentiometrically (2.4.25) and hours. Equilibrate the column with the mobile phase for about
using a suitable anhydrous electrode system. (The electrode
30 minutes before starting the chromatography.
system may be rendered anhydrous by filling the electrode
Inject the reference solution. The test is not valid unless the
with 0.1 M lithium perchlorate in acetic anhydride after
tailing factor is not more than 2.0. The relative standard
removing any aqueous solution contained in it). Carry out a
deviation for replicate injections is not more than 2.0 per cent.
blank titration.
Inject altemately the test solution and the reference solution.
1 ml of 0.1 Mperchloric acid is equivalent to 0.03193 g of
C16HISFN303' Calculate the content of C16H.ISFN303 in the eye drops.
Storage. Store protected from light and moisture. Storage. Store protected from light.

Norfloxacin Eye Drops Norfloxacin Tablets


Norfloxacin Eye Drops are a sterile solution ofNorfloxacin in Norfloxacin Tablets contain not less than 90.0 per cent and
PUlified water. not more than 110.0 per cent of the stated amount of
Norfloxacin Eye Drops contain not less than 90.0 percent and norfloxacin, C16HISFN303'
not more than 110;0 per cent of the stated amount of Usual strengths. 200 mg; 400 mg; 800 mg.
norfloxacin, C16HlsFN303'
Identification
Usual strength. 0.3 per cent w/v.
A. Determine by thin-layer chromatography (2.4.17), coating
Identification the plate with silica gel GF254.

In the Assay, the principal peak in the chromatogram obtained Mobile phase. A mixture of 40 volumes of chloroform,
with the test solution corresponds to the peak in the 40 volumes of methanol, 20 volumes of toluene, 14 volumes
chromatogram obtained with the reference solution. of diethylamine and 8 volumes of water.
Test solution. Shake a quantity ofthe fine~y powdered tablets
Tests
containing 75 mg ofNorfloxacin with 50 ml of a mixture of
pH (2.4.24).4.6 to 5.5. equal volumes of acidified methanol (containing 0.9 per cent

1794

www.webofpharma.com
IP 2010 NORGESTREL

v/v of hydrochloric acid) and dichloromethane, centrifuge flow rate. 2 ml per minute,
and use the clear supernatant solution. spectrophotometer set at 275 nm,
Reference solution. A 0.15 per cent w/v solution of nOlj!oxacin
- injection volume. 20 fll.
RS in the same solvent mixture. lriject the test solution and the reference solution. The assay
is not valid unless the capacity factor is not less than 2, the
Apply to the plate 50 fll of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. column efficiency is not less than 1500 theoretical plates, the
tailing factor for the norfloxacin peak is not more than 2.0 and
Theprincipal spot in the chromatogram obtained with the test
the relative standard deviation for replicate injections is not
solution corresponds to that in the chromatogram obtained
with the reference solution. more than 2.0 per cent.

B. In the Assay, the principal peak in the chromatogram Calculate the content ofCI6HIsFN303 in the tablets.
obtained with the test solution cOlTesponds to the peak due Storage. Store protected from light and moisture.
to norfloxacin in the chromatogram obtained with the reference
solution.

Tests Norgestrel
Dissolution (2.5.2).
Apparatus No.1,
Medium. 750 ml of acetate bufferpH4.0,
Speed and time. 50 rpm and 30 minutes.
Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance of the filtrate, suitably diluted with acetate
buffer pH 4.0, if necessary, at the maximum at about 278 nm
(2.4.7). Concomitantly measure the abs.orbance of a solution Mol. Wt. 312.5
oflmown concentration of nOlj!oxacin RS in the same medium. Norgestrel is rac-13-ethyl-17-hydroxy-18,19-dinor-17Cf.,-
Calculate the total content ofCI6HIsFN303 in the medium. pregn-4-en-20-yn-3-one.
D. Not less than 70 per cent of the stated amount of Category. Progestogen.
C16HlsFN303. Dose. As a contraceptive, 150 to 300 flg in combination with
Other tests. Comply with the tests stated under Tablets. 20 to 50 flg ofethinyloestradiol daily.
Assay. Determine by liquid chromatography (2.4.14). Description. A white or almost white, clystalline powder;
Test solution. Weigh and powder 20 tablets. Add 80 ml ofthe practically odourless.
mobile phase to an accurately weighed quantity of the
Identification
powdered tablets containing about 100 mg ofNorfloxacin, mix
with the aid ofultrasound for 10 minutes, dilute with a 0.1 per A. Determine by infi'ared absorption spectrophotometry (2.4.6).
cent v/v solution of phosphoric acid to 200.0 ml and mix. Compare the spectTUm with that obtained with norgestrel RS.
Dilute 10.0 ml ofthis solution to 25.0 ml with the mobile phase, B. When examined in the range 220 nm to 360 111TI (2.4.7), a
mix and use the resulting solution after filtration through a 0.001 per cent w/v solution in methanol shows an absorption
filter with porosity ofnot more than 0.1 fllU. maximum only at about 240 nm.
Reference solution. A 0.02 per cent w/vsolution ofnOlj!oxacin
C. Melting range (2.4.21). 205° to 212°, but the range between
RS in the mobile phase. beginning and end of melting does not exceed 4°.
Chromatographic system
- a stainless steel colulUn 30 cm x 3.9 mm, packed with Tests
octadecylsilane bonded to porous silica (5 flm), Specific optical rotation (2.4.22). -0.1 ° to +0.1 0, determined in
mobile phase: 85 volumes ofa 0.1 per cent v/v solution a 5.0 per centw/v solution in chloroform.
of phosphoric acid and 15 volumes of acetonitrile,
- temperature. COlutm140° ± 1°, after preconditioning with Related substances. Determine by thin-layer chromatography
degassed 0.01 M sodium dihydrogen phosphate (2.4.17), coating the plate with silica gel G
adjusted to pH 4.0 with phosphoric acid flowing at a Mobile phase. A mixture of 80 volumes of dichloromethane
rate of0.5 ml per minute for 8 hours, and 20 volumes of ethyl acetate.

1795

www.webofpharma.com
NORGESTREL IP 2010

Test solution. Dissolve 0.2 g of the substance under Reference solution. A solution containing 0.06 per cent w/v
examination in 10 ml of chloroform. solution of norgestrel RS and 0.006 per cent w/v solution of
Reference solution (a). A 0.01 per cent w/v solution of the ethinyloestradiol RS.
substance under examination in chloroform. Apply to the plate 40 III of each solution. After development,
Reference solution (b). A 0.004 per cent w/v solution of the dry the plate in air, spray with ethanolic sulphuric acid
substance under examination in chloroform. (80 per cent v/v) ,heat at 110° for 10 minutes and examine in
ultraviolet light at 365 nm. The principal spots in the
Apply to the plate 10 III of each.solution. After development, chromatogram obtained with the test solution correspond to
dry the plate in air and spray with phosphomolybdic acid the spots for norgestrel (red fluorescence) and
solution. Any secondary spot in the chromatogram obtained ethinyloestradiol (orange-yellow fluorescence) in the
with the test solution is not more intense than the spot in the chromatogram obtained with the reference solution.
chromatogram obtained with reference solution (a) and not
more than two such spots are more intense than the spot in Tests
the chromatogram obtained with reference solution (b).
Uniformity of content. Comply with the test stated under
Sulphated ash. (2.3.18) Not more than 0.3 per cent. Tablets.
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined Carry out the procedure described under Assay but using the
on 1.0 g by drying in an oven at 105° for 5 hours. following test solution.
Assay. Weigh accurately about 0.1 g, dissolve in sufficient Test solution. Add 2.0 ml of methanol (70 per cent) and 2.0 ml
ethanol (95 per cent) to produce 100.0 ml, dilute stepwise of a 0.00002 per cent w/v solution of diphenyl in methanol
with ethanol (95 per cent) to obtain a solution containing (70 per cent) (internal standard solution) to one tablet, shake
0.001 per cent w/v of levonorgestre1 and measure the for 20 minutes, centrifuge, filter the supernatant liquid through
absorbance of the resulting solution at the maximum at about a membrane filter with a pore size ofnot more than 0.2 mm and
241 nm, (2.4.7). Calculate the content of CZIHzsOz from the use the filtrate.
absorbance obtained with a 0.001 per cent w/v solution of
norgestrel RS in ethanol (95 per cent). Calculate the contents of norgestrel CZIHzsOz, and
ethinyloestradiol, CZOHZ40Z' in the tablet.
Storage. Store protected from moisture.
Other tests. Comply with the tests stated under Tablets.
Assay. Detennine by liquid chromatography (2.4.14) using the
Norgestrel and Etbinyloestradiol chromatographic system described under Uniformity of
content.
Tablets
Test solution. Weigh and powder 20 tablets. To a quantity of
Norgestrel and Ethinyloestradiol Tablets contain not less than the powder equivalent to one tablet add 2.0 ml of methanol
90.0 per cent and not more than 110.0 per cent of the stated (70 per cent) and 2.0 ml of a 0.00002 per cent w/v solution of
amounts of norgestrel, CZIHzsOz and ethinyloestradiol, diphenyl in methanol (70 per cent) (internal standard
CzOHZ40 Z. solution), shake for 20 minutes, centrifuge, filter the
Category. Oral contraceptive. supernatant liquid through a membrane filter with a pore size
afnot more than 0.2 mm and use the filtrate.
Dose. One tablet daily for 21 days starting from the fifth day of
menstrual cycle. Reference solution. A solution in methanol (70 per cent)
containing 0.15 mg per ml of norgestrel RS and 0.015 mg per
Usual strength. Norgestrel, 300 Ilg and Ethinyloestradiol, ml of ethinyloestradiol RS. Take 2.0 ml of this solution and
30 Ilg. add 2.0 ml of a 0.00002 per cent w/v solution of diphenyl in
Identification methanol (70 per cent) and use the resulting solution.

Detennine by thin-layer chromatography (2.4.17), coating the Chromatographic system


plate with silica gel G - a stainless steel column 15 cm x 4.6 rntD, packed with
octadecylsilane bonded to porous silica (5 to 7 Ilm),
Mobile phase. A mixture of 96 volumes of dichloromethane - mobile phase: a mixture of35 volumes of acetonitrile,
and 4 volumes of ethanol (95 per cent). 15 volumes of methanol and 45 volumes of water,
Test solution. Powder 20 tablets finely, triturate with 20 ml of - flow rate. 1 to1.5 ml per minute,
dichloromethane, allow the solids to sediment and use the - spectrophotometer set at 215 urn,
clear supernatant liquid. - injection volume. 20 Ill.

1796

www.webofpharma.com
IP 2010 NORTRIPTYLINE TABLETS

Inject the reference solution. The resolution between the two C. To about 50 mg dissolved in 3 ml ofwarm water, add 1drop
major peaks is not less 2.5 and the relative standard deviation of a 2.5 per cent w/v solution of quinhydrone in methanol; a
for replicate injections is not more than 2.0 per cent. red colour is produced after a few minutes (distinction from
Inject the test solution and the reference solution. The relative amitriptyline).
retention times are about 0.7 for ethinyloestradiol and about D. Gives the reactions ofchlorides (2.3.1).
1.0 for norgestrel.
Tests
Calculate the contents of norgestrel, C21H2S02, and
ethinyloestradiol, C2oH2402 in the tablets. Related substances. Determine by thin-layer chromatography
Storage. Store protected from light. (2.4.17), coating the plate with silica gel G
Mobile phase. A mixture of 85 volumes of cyclohexane,
15 volumes of ethyl acetate and 3 volumes of diethylamine.
Test solution. Dissolve 0.2 g of the substance under
Nortriptyline Hydrochloride examination in 10 ml of ethanol (95 per cent) prepared in
subdued light.
Reference solution. A 0.001 per cent w/v solution of
dibenzosuberone RS in ethanol (95 per cent) prepared in
, Hel subdued light.
Apply to the plate 5 III of each solution. Allow the mobile
phase to rise 14 cm in an unsaturated tank protected from
light. Dry the plate in air, spray with a freshly prepared solution
Mol. Wt. 299.8 of sulphuric acid containing 4 per cent v/v offormaldehyde
solution and examine immediately in ultraviolet light at
Nortriptyline Hydrochloride is 3-(10, II-dihydro-5H-dibenzo 365 nm. Any secondary spot in the chromatogram obtained
[a,dJcyclohept-5-ylidene)propyl(methyl)amine hydrochloride. with the test solution is not more intense than the spot in the
Nortriptyline Hydrochloride contains not less than 98.0 per chromatogram obtained with the reference solution.
cent and not more than 101.5 percent ofC ,9H2I N,HC1, calculated Heavy metals (2.3.13). 2.0 g complies with the limit test for
on the dried basis. heavy metals, Method B (10 ppm).
Category. Antidepressant.
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
Dose. Initially, 50 to 150 mg daily; maintenance dose, 30 to 75
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
mgdaily.
on 1.0 g by drying in an oven at 105° for 3 hours.
Description. A white to off-white powder; odour slight and
Assay. Weigh accurately about 0.25 g and dissolve in 25 ml of
characteristic.
anhydrous glacial acetic acid, warm slightly, if necessary, to
effect solution. Cool, add 5 ml of mercuric acetate solution.
Identification
Titrate with 0.1 M perchloric acid, determining the end-point
Test A may be omitted iftests B, C andD are carried out. Tests potentiometrically (2.4.25). Carry out a blank titration.
Band C may be omitted if tests A and, D are carried out. 1 ml of 0.1 M perchloric acid is equivalent to 0.02998 g of
A. Dissolve 0.1 gin 10 ml of water, make allmline with 1 M CI9H2I N,HCl.
sodium hydroxide, extract with 5 ml of chloroform and
Storage. Store protected from light and moisture.
evaporate to dryness using a current of nitrogen.
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with nortriptyline
hyrdochloride RS treated in the same manner or with the Nortriptyline Tablets
reference spectrum ofnortriptyline.
Nortriptyline Hydrochloride Tablets
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.001 per cent w/v solution in methanol shows an absorption Nortriptyline Tablets contain less than 90.0 per cent and not
maximum only at about 239 nm; absorbance at about 239 nm, more than 110.0 per cent ofthe stated amount ofnortriptyline,
about 0.48. C I9H2I N. The tablets are coated.

1797

www.webofpharma.com
NORTRIPTYLINE TABLETS IP 2010

Usual strengths. The equivalent of 10 mgand 25mg of methanol and shake for 30 minutes. Add sufficient water to
nortriptyline. produce 10 ml, centrifuge and use the clear supernatant liquid.

Identification Reference solution. A 0.01 per cent w/v solution of


nortriptyline hydrochloride RS in methanol (50 per cent).
A. Shake a quantity ofthe powdered tablets containing about
Follow the procedure given in the Assay. Calculatethe content
5 mg ofnortriptyline with 20 ml of methanol and filter. To 1 ml
ofCzo Hz3 N in the tablet.
ofthe filtrate add 1 ml of a 2.5 per cent w/v solution of sodiurn
bicarbonate, 1 ml of a 2 per cent w/v solution of sodium Other tests. Comply with the tests stated under Tablets,
periodate and 1 ml ofa 0.3 per cent w/v solution ofpotassium Assay. Detennine by liquid chromatography (2.4.14).
permanganate. Allow to stand for 15 minutes, acidifY with
1 M sulphuric acid and extract with 10 ml of 2i2,4~ Test solution. Shake vigorously 20 tablets with 50 ml of water
trimethylpentane. until the tablets disintegrate completely, add 100.0 ml of
methanol and shake for 30 minutes. Add sufficient water to
When examined in the range 230 nm to 360 nm (2.4.7), the produce 200.0 ml, filter and dilute a volume of the filtrate
resulting trimethylpentane solution shows an absorption containing about 25 mg of nOltriptyline to 100.0 ml with
maximum only at about 265 nm. methanol (50 per cent).
B. Triturate a quantity of the powdered tablets containing Reference solution. A 0.025 per cent w/v solution of
0.1 g of nortriptyline with 10 ml of chloroform, filter and nortriptyline hydrochloride RS in methanol (50 per cent).
evaporate the filtrate to a low volume. Add ether until a
turbidity is produced and allow to stand. Dissolve 50 mg of Chromatographic system
the precipitate in 3 ml ofwann water, cool and add 1 drop ofa a stainless steel column 20 cm x 4.6 mm, packed with
2.5 per cent w/v solution of quinhydrone in methanol; a red octadecylsilane bonded to porous silica (l0 Ilm),
colour is produced after a few minutes (distinction from mobile phase: a 0.56 per cent w/v solution of sodium
amitriptyline): hexanesulphonate in a mixture of equal volumes of
water and acetonitrile adjusted to pH 4.5 with glacial
Tests acetic acid;
flow rate. 2 ml per minute,
Related substances. Detel1lline by thin71ayer chromatography - spectrophotometer set at 239 nm,
(2.4.17), coating the plate with silica gel G injection volume. 20 Ill.
Mobile phase. A mixture of 85 volumes of cyclohexane, Inject the test solution and the reference solution.
15 volumes of ethyl acetate and 3 volunies of diethylamine.
Calculate the content of C I9Hz1 N in the tablets.
Test solution. Extract a quantity of the powdered tablets
containing 20 mg of nortriptyline with 5 ml of a mixture of Storage. Store protected from light and moisture.
9 volumes of ethanol (95 per cent) and 1 volume of 2 M
hydrochloric acid, centrifuge and use the supernatant liquid.
Reference solution. A 0.001 per cent w/v solution of
dibenzosuberone RS in ethanol (95 per cent) prepared in Noscapine
subdued light. Narcotine
Apply to the plate 5 III of each solution. Allow the mobile
phase to rise 14 cm in an unsaturated tank protected from
light. Dry the plate in air, spray with a freshly prepared solution
of sulphuric acid containing 4 per cent v/v offormaldehyde
solution and examine immediately in ultraviolet light at
365 nm. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Uniformity of content (For tablets ~ontaining 10 mg or less).
Comply with the test stated under Tablets.
Detel1lline by liquid chromatography (2.4.14).
Test solution. Powder one tablet, add 2.5 ml of water, shake
vigorously to completely disperse the tablet, add 5 ml of Mol. Wt. 413.4

1798

www.webofpharma.com
IP 2010 NOSCAPINE

Noscapine is (38)-6,7-dimethoxy-3-[(5R)-5,6, 7,8-tetrahydro- Reference solution (c). Dissolve 1.5 mg of papaverine


4-methoxy-6-methyl-1 ,3-dioxolo[4,5-g]isoquinolin-5- hydrochloride in 10 ml ofthe test solution and dilute to 50 ml
yl]phthalide, an alkaloid obtained from opium. with the mobile phase.
Noscapine contains not less than 98.5 per cent and not more Chromatographic system
than 100.5 per cent ofC22H23N07, calculated on the dried basis. - a stainless steel column 12.5 cm x 4.6 mm, packed with
Category. Cough suppressant. nitrile groups chemically bonded to porous silica (5 11m),
- mobile phase: a mixture of35 volumes of methanol and
Dose. 15 to 30 mg three to four times daily. 65 volumes of phosphate buffer pH 6.0,
Description. Colourless crystals or a white crystalline powder. flow rate. I ml per minute,
- spectrophotometer set at 240 nm,
Identification - injection volume. 20 Ill.
Inject reference solution (c). The test is not valid unless the
Test A may be omitted iftests B, C andD are carried out. Tests
resolution between the peaks due to noscapine and papaverine
B, C and D may be omitted if test A is carried out.
(noscapine impurity A) is not less than 2.0. The relative
A. Determine by infrared absorption spectrophotometry (2.4.6). retention· time with reference to noscapine for noscapine
Compare the spectrum with that obtained with noscapine RS Impurity Ais about 1.3.
or with the reference spectrum of noscapine.
Inject the test solution, reference solution (a) and (b). Run the
B. When examined in the range 230 nm to 360 nm (2.4.7), a
chromatogram 2.5 times the retention time of the principal
0.005 per cent w/v solution in methanol shows absorption
peak. In the chromatogram obtained with the test solution the
maxima at about 291 nm and 310 nm; ratio ofabsorbance at the
area ofthe peak due to noscapine impurity A is not more than
maximum at about 310 nm to that at the maximum at about
the area ofthe principal peak in the chromatogram obtained
291 nm, 1.2 to 1.3.
with reference solution (b) (0.5 per cent), the area ofthe any
C. To 0.1 g in a porcelain dish add a few drops of sulphuric oth~r secondary peak is not more than 0.4 times the area of
acid and stir; a greenish-yellow solution is formed which on the principal peak in the chromatogram obtained with reference
warming becomes red and finally violet. solution (a) (0.2 per cent), and the sum of the areas of
D. Dissolve 50mg inS mlof5 Mhydrochloricacid, add 10ml secondary peaks other than noscapine impurity A is not more
of a mixture of equal volumes of ethanol (95 per cent) and a than the area of the principal peak in the chromatogram
saturated solution of sodium acetate, mix and allow to stand obtained with reference solution (a) (0.5 per cent). Ignore any
for about 3 minutes; shining crystals separate. peak with an area less than 0.1 times the area of the principal
peak in the chromatogram obtained with reference solution
Tests (a) (0.05 per cent).

Appearance ofsolution. A 2.0 per cent w/v solution in acetone Morphine. Dissolve 0.1 gin 10 m1 of 0.1 M hydrochloric acid.
examined immediately after preparation is clear (2.4.1), and not To 1 m1 of the resulting solution add a mixture of 1 ml of
more intensely coloured than reference solution YS6 (2.4.1). potassium ferricyanide solution, 0.05 m1 of ferric chloride
test solution and 4 m1 of water; no blue or dark green colour
Specific optical rotation (2.4.22). +42.0° to +48.0°, determined
develops within 1 minute.
at 20° in a solution prepared by dissolving 0.5 g in sufficient
0.1 M hydrochloric acid to produce 25.0 ml. Sulphated ash (2.3.1 8). Not more than 0.1 per cent.
Related substances: Determine by liquid chromatography
(2.4.14). Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105°.
Test solution. Dissolve 20 mg of the substance under
examination with gentle heating in 14 ml of methanol, cool, Assay. Weigh accurately about 0.35 g, dissolve in 40 m1 of
and dilute to 20 ml with phosphate buffer solution pH 6.,0. anhydrous glacial acetic acid, warming gently. Titrate with
Reference solution (a). Dilute 1.0 ml of the test solution to 0.1 M perchloric acid, determining the end-point
20.0 ml with the mobile phase. Dilute 1.0 ml ofthe solution to potentiOluetrically (2.4.25). Carry out a blank titration.
10.0 ml with the mobile phase.
1 m1 of0.1 M perchloric acid is equivalentto 0.04134 g of
Reference solution (b). Dissolve 5 mg of papaverine C22H23N07•
hydrochloride in 50 ml of the test solution. Dilute 1.0 ml of
this solution to 20 ml with the mobile phase. Storage. Store protected from light and moisture.

1799

www.webofpharma.com
NOSCAPINE LINCTUS IF 2010

Noscapine Linctus Novobiocin Sodium


Narcotine Linctus
Noscapine Linctus is a solution of Noscapine in a suitable
flavoured vehicle. It may contain up to 1per cent w/v solution
ofCitric Acid.
Noscapine Linctus contains not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
noscapine, C22H23N07'
Usual strength. 7 mg per 5 ml.
Mol. Wt. 634.6
Identification Novobiocin Sodium is the monosodium salt of novobiocin,
N-[7- {3-0-( aminocarbonyl)-6-deoxy-5-C-methyl-4-0-methyl-
To a quantity containing 60 mg ofNoscapine add 20 ml of
~-L-Iy.xo-hexopyranosyl}-oxy-4-hydroxy-8-methyl-2-oxo-2H­
water, 2 g of sodium chloride and 2 ml of 5 M sodium
I-benzopyran-3-yl]-4-hydroxy-3-(3-methyl-2-
hydroxide. Extract with successive quantities of50, 50, 25 and
butenyl)benzamide, an antimicrobial substance produced by
25 ml ofether. Combine the extracts, wash with three quantities,
the growth of certain strains of Streptomyces niveus or
each of5 ml, of water and evaporate to dryness. Dissolve the
related organisms or by other means.
residue in 20 ml of chloroform. Wash with three quantities,
each of20 ml, of water, dry the chloroform layer with anhydrous Novobiocin Sodium contains the equivalent of not less than
sodium sulphate, filter and evaporate the solvent. Ifnecessary, 850 Ilg of novobiocin per mg, calculated on the dried basis.
induce crystallisation by scratching with a glass rod. The Category. Antibacterial.
crystals comply with the following tests.
Dose. The equivalent of 1 to 2 g ofnovobiocin daily, in divided
A. Determine by infrared absorption spectrophotometry (2.4.6). doses.
Compare the spectrum with that obtained with noscapine RS
or with the reference spectrum of noscapine. Description. A white or yellowish white, crystalline powder.
B. When examined in the range 230 nm to 360 nm (2.4.7), a Identification
0.005 per cent w/v solution in methanol shows absorption
maxima at about 291 nmand 310 nm; ratio ofabsorbance at the A. Determine by thin-layer chromatography (2.4.17), coating
maximum at about 310 nm to that at the maximum at about the plate with silica gel G. .
291 nm, 1.2 to 1.3. Mobile phase. A mixture of 75 volumes of chloroform,
25 volumes of methanol and 1 volume of strong ammonia
Tests solution.
Other tests. Complies with the tests stated under Oral Liquids. Test solution. Dissolve a quantity of the substance under
examination in methanol so as to obtain a solution containing
Assay. Weigh accurately a quantity containing 60 mg of
0.1 per cent w/v solution of novobiocin.
Noscapine, add 20 ml of water, 2 g of sodium chloride and
2 ml of 5 M sodium hydroxide and extract with successive Reference solution. A 0.1 per cent w/v solution of novobiocin
quantities of50, 50,25 and 25 ml ofether. Combine the extracts, RS in methanol.
wash with three quantities, each of5 ml, of water and evaporate Apply to the plate 1 III of each solution. After development,
to dryness. To the residue add 50.0 ml of 0.1 M hydrochloric dry the plate in air and examine in ultraviolet light at 254 nm.
acid, warm on a water-bath to dissolve and to remove any The principal spot in the chromatogram obtained with the test
traces ofether and dilute to 100.0 ml with water. Dilute 3.0 ml solution corresponds to that in the chromatogram obtained
to 50.0 ml with water and measure the absorbance of the with the reference solution.
resulting solution at the maximum at about 310 nm (2..4.7).
Calculate the content ofC22H23N07 taking 90.7 as the specific B. When examined in the range 230 nm to 360 nm (2.4.7), a
absorbance at 310 nm. 0.001 per cent w/v solution in a 0.4 per cent w/v solution of
potassium hydroxide shows an absorption maximum only at
Determine the weight per ml ofthe linctus (2.4.29), and calculate about 307 nm.
the content ofC22H23N07, weight in volume.
C. The residue obtained by igniting it gives the tests for sodium
Storage. Store protected from light and moisture. salts (2.3~ 1).

1800

www.webofpharma.com
IP 2010 NYSTATIN

Tests Dose. In the treatment of alimentary candidiasis, 500,000 to


1,000,000 Units daily, in divided doses.
pH (2.4.24).6.6 to 8.5, determined in a 2.5 percentw/v solution.
Description. A yellow to slightly brown powder; odour,
Specific optical rotation (2.4.22). -50.0° to-58.0°, determined characteristic; hygroscopic.
in a 5.0 per cent w/v solution in methanol containing I per
cent v/v of hydrochloric acid. Identification
Loss on drying (2.4.19). Not more than 6 per cent, determined A. When examined in the range 220 nm to 360 nm (2.4.7), the
on 0.2 g by drying in an oven at 60° over phosphoruspentoxide final solution obtained in the test for Light absorption shows
at a pressure not exceeding 0.7 kPa for 3 hours. absorption maxima at about 230 nm, 291 nm, 305 nm and
Assay. Determine by the microbiological assay ofantibiotics, 319 nm. The ratios of the absorbances at the maxima at about
Method A (2.2.1 0), and express the results in J.lg ofnovobiocin 291 nm and about 319 nm to the absorbance at the maximum at
per mg. about 305 nm are 0.61 to 0.73 and 0.83 to 0.96, respectively.
Use as the blank a solution prepared in the same manner
Novobiocin Sodium intended for use in the manufacture of
without the substance under examination.
Parenteral Preparations complies with the following
additional requirements. B. Shake 30 mg with 5 ml of water for 2 minutes, add 2 ml of
sodium molybdotungstophosphate solution, and allow to
Bacterial endotoxins (2.2.3). Not more than 0.7 Endotoxin units
stand for I hour; the green colour produced is darker than
per mg.
that produced by repeating the test without the substance
Sterility. Complies with the test for sterility (2.2.11). under examination.
Storage. Store protected from light and moisture at a C. Shake 30 mg with 5 ml of water for 2 minutes, add 2 ml ofa
temperature not exceeding 30°. If it is intended for use in the solution prepared by dissolving 0.1 g ofpyrogallol in 100 ml
manufacture of parenteral preparations, the container should of decolorised magenta solution, heat on a water-bath until a
be sterile and sealed so as to exclude micro-organisms. dark pink colour is produced, cool and allow to stand for
Labelling. The label states whether or not the contents are 1 hour; the pink colour is retained.
intended for use in the manufacture ofparenteral preparations. D. To 2 mg add 0.1 ml of hydrochloric acid; a brown colour is
produced.
E. To 2 mg add 0.1 ml of sulphuric acid; a brown colour is
Nystatin produced which becomes violet on standing.

Tests
pH (2.4.24). 6.5 to 8.0, determined in a 3.0 per cent w/v
OH suspension in water.
Light absorption (2.4.7). Dissolve 0.1 g in a mixture of5.0 ml of
COOH
glacial acetic acid and 50 ml of methanol, add sufficient
methanol to produce 1 00.0 ml and dilute 1.0 ml ofthe resulting
o solution to 100.0 ml with methanol. Absorbance ofthe resulting
\--o-\CH 3 solution, measured within 30 minutes of preparation, at the
~OH maximum at about 305 nm, not less than 0.60. Use as the blank
OH NH 2 a solution prepared in the same manner without the substance
under examination.
Mol. Wt. 926.1 Composition. Determine by liquid chromatography (2.4.14).
Nystatin is an antifungal substance produced by the growth Note-Carry out the test protectedfrom light.
of certain strains of Streptomyces noursei or by any other Test solution. Dissolve 20 mg of the substance under
means. It consists mainly ofpolyenes, the principal component examination in 50 ml of dimethyl sulphoxide.
being nystatin AI.
Reference solution (a). A 0.04 per cent w Iv solution of nystatin
Nystatin has a potency of not less than 4400 Units per mg, RS in dimethyl sulphoxide.
calculated on the dried basis.
Reference solution (b). Dissolve 20 mg ofthe substance under
Category. Antifungal. examination in 25 ml ofthe methanol and dilute to 50 ml with

1801

www.webofpharma.com
NYSTATIN IP 2010

water. To 10.0 ml of this solution add 2.0 ml of dilute Determine by the microbiological assay ofantibiotics (2.2.10).
hydrochloric acid. Allow to stand at room temperature for 1
Nystatin intendedfor oral administration complies with the
hour.
following additional requirement.
Reference solution (c). Dilute 1.0 ml of reference solution (a)
Abnormal toxicity (2.2.1). Complies with the test for abnormal
in 100.0 rnl of dimethyl sulphoxide. Dilute 1.0 rnl ofthis solution
toxicity, using a quantity containing not less than 600 Units
to 10.0 ml with dimethyl sulphoxide.
suspended in not more than 0.5 ml ofa 0.5 per centw/v solution
Chromatographic system of acacia and injecting the suspension intraperitoneally.
- a stainless steel column 15 cm x 4.6 mm, packed with
Storage. Store protected from light and moisture.
base-deactivated end-capped octadecylsilane bonded
to porous silica (5 /lm), Labelling. The label states the strength in terms ofthe number
- mobile phase: A. a mixture of29 volumes ofacetonitrile of Units ofNystatin per mg.
and 71 volumes of 0.385 per cent w/v solution of
ammonium acetate,
B. a mixture of 40 volumes of 0.385 per
cent w/v solution ofammonium acetate and 60 volumes
Nystatin Ointment
of acetonitrile. Nystatin Ointment is a dispersion of Nystatin in microfine
- a linear gradient programme using the conditions given powder in a suitable ointment basis.
below,
Nystatin Ointment contains not less than 90.0 per cent and
- flow rate. 1 ml per minute,
not more than 130.0 per cent ofthe stated number ofUnits of
- spectrophotometer set at 305 nm,
nystatin.
- injection volume. 20 /ll.
Time Mobile phase A Mobile phase B Usual strength. 100,000 Units per g.
(in min.) (per cent w/v) (per cent w/v)
Identification
0-25 100 o
25-35 100-0 0-100 Disperse a quantity containing 25,000 Units in 10 ml of
35-45 0 100 chloroform, add 40 ml ofmethanol and shake. Filter and dilute
1 ml ofthe filtrate to 25 ml with methanol.
45-50 0-100 100-0
50-55 100 o When examined in the range 230 nm to 360 nm (2.4.7), the
Inject reference solution (b). The test is not valid unless the resulting solution shows absorption maxima at about 291 nm,
resolution between the two principal peaks is not less than 305 nm and 319 nm. The ratios ofthe absorbances at the maxima
3.5. at about 291 nm and about 319 nm to the absorbance at the
maximum at about 305 nm are 0.61 to 0.73 and 0.83 to 0.96,
Inject the test solution, reference solution (a) and (c). In the
respectively. Use as the blank a solution prepared exactly in
chromatogram obtained with the test solution the area of the
the same manner without the substance under examination.
peak due to nystatin A 1 is not less than 85 per cent the area of
the plincipal peak in the chromatogram obtained with reference Tests
solution (a), the area of the any other compound is not more
than 4.0 per cent the area of the principal peak in the Other tests. Complies with the tests stated under Ointments.
chromatogram obtained with reference solution (a). Ignore Assay. Protect the solution from light throughout the assay.
any peak with a retention time of less than 2 minutes.
Weigh accurately a quantity containing 400,000 Units, disperse
Heavy metals (2.3.13). 1.0 g complies with the limit test for in 20 ml of ether in a stoppered flask, add 70 ml of
heavy metals, Method B (20 ppm). dimethylformamide, shake for a few minutes, add 10 ml of
Sulphated ash (2.3.18). Not more than 3.5 per cent. water, shake vigorously for a few minutes and add sufficient
dimethylformamide to produce 100.0 ml. Mix well, filter and
Loss on drying (2.4.19). Not more than 5.0 per cent, determined
dilute 10.0 ml of the filtrate to 100.0 ml with buffer solution
on 1.0 g by drying in an oven at 60° at a pressure not exceeding
No 4 (2.2.10).
0.1 kPa for 3 hours.
Determine by the microbiological assay ofantibiotics (2.2.10).
Assay. Protect the solution from light throughout the assay.
Storage. Store protected from moisture.
Weigh accurately about 75 mg and dissolve in sufficient
dimethylformamide to produce 50.0 ml; dilute 10.0 ml of the Labelling. The label states the strength in terms ofthe number
resulting solution to 200.0 rnl with buffersolutionNo4 (2.2.10). of Units of Nystatin per g.

1802

www.webofpharma.com
IP 2010 NYSTATIN TABLETS

Nystatin Pessaries Nystatin Tablets


Nystatin Vaginal Tablets Nystatin Tablets contain not less than 90.0 per cent and not
Nystatin Pessaries contain not less than 90.0 per cent and not more than 130.0 per cent of the stated number of Units of
more than 130.0 per cent of the stated number of Units of nystatin. The tablets are coated.
nystatin. Usual strength. 500,000 Units.
Usual strength. 100,000 Units.
Identification
Identification Extract a quantity of the powdered tablets containing
Extract a quantity of the powdered pessaries containing 300,000 Units with a mixture of50 ml of methanol and 5 ml of
300,000 Units with a mixture of50 ml of methanol and 5 ml of glacial acetic acid, add sufficient methanol to produce
glacial acetic acid, add sufficient methanol to produce 100 ml and filter. Dilute 1 ml of the filtrate to 100 ml with
100 ml and filter. Dilute 1 ml of the filtrate to 100 ml with methanol. The resulting solution complies with the follwing
methanol. The resulting solution complies with the follwing test.
test. When examined in the range 230 nm to 360 nm (2.4.7), the
Disperse a quantity containing 25,000 Units in 10 ml of solution shows absorption maxima at about 291 nm, 305 nm
chlor%nn, add 40 ml of methanol and shake. Filter and dilute and 319 urn. The ratios of the absorbances at the maxima at
1 ml ofthe filtrate to 25 ml with methanol. about 291 nm and about 319 nm to the absorbance at the
maximum at about 305 nm are 0.61 to 0.73 and 0.83 to 0.96,
When examined in the range 230 urn to 360 urn (2.4.7), the respectively. Use as the blank a solution prepared exactly in
resulting solution shows absorption maxima at about 291 urn, the same manner without the substance under examination.
305 urn and 319 urn. The ratios ofthe absorbances at the maxima
at about 291 urn and about 319 nm to the absorbance at the Tests
maximum at about 305 urn are 0.61 to 0.73 and 0.83 to 0.96,
respectively. Use as the blank a solution prepared exactly in Disintegration (2.5.1). 30 minutes, but using a 0.6 per cent
the same manner without the substance under examination. v/v solution of hydrochloric acid in place of water. If the
tablets fail to disintegrate, wash them rapidly by immersion in
Tests water and continue the test using phosphate buffer pH 6.8;
the tablets then disintegrate within a further 30 minutes.
Other tests. Comply with the tests stated under Pessaries.
Loss on drying (2.4.19). Not more than 5 per cent, determined Loss on drying (2.4.19). Not more than 5.0 per cent, determined
on 1.0 g ofthe powdered pessaries by drying in an oven at 60° on 1.0 g ofthe powdered tablets by drying in an oven at 60° at
at a pressure not exceeding 0.7 kPa for 3 hours. a pressure not exceeding 0.7 kPa for 3 hours.

Assay. Protect the solution from light throughout the assay. Other tests. Comply with the tests stated under Tablets.

Weigh and powder 20 pessaries. Weigh accurately a quantity Assay. Weigh and powder 20 tablets. Weigh accurately a
ofthe powder containing 200,000 Units and shake with 50.0 ml quantity of the powder containing 200,000 Units and shake
of dimethylfonnamide for 1 hour. Centrifuge, dilute 10.0 ml of with 50.0 ml of dimethylfonnamide for 1 hour. Centrifuge, dilute
the clear, supernatant liquid to 200.0 ml with buffer solution 10.0 ml ofthe clear, supernatant liquid to 200.0 ml with buffer
No 4 (2.2.10). solution No 4 (2.2.10).
Determine by the microbiological assay ofantibiotics (2.2.10). Determine by the microbiological assay ofantibiotics (2.2.10).
Storage. Store protected from light and moisture. Storage. Store protected from moisture.
Labelling. The label states the strength in terms ofthe number Labelling. The label states the strength in tenns ofthe number
of Units ofNystatin. of Units of Nystatin.

1803

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

0
Oestradiol Benzoate 1807
Oestradiol Injection 1807
Ofloxacin 1808
Ofloxacin Infusion 1809
Ofloxacin Opthalmic Solution 1809
Ofloxacin Tablets 1810
Olanzapine 1811
Olanzapine Tablets 1812
Oleic Acid 1812
Omeprazole 1813
Omeprazole Capsules 1814
Ondansetron Hydrochloride 1815
Ondansetron Injection 1816
Ondansetron Orally Disintegrating Tablets 1816
Ondansetron Oral Solution 1818
Ondansetron Tablets 1819
Oral Rehydration Salts 1820
Ormeloxifene Hydrochloride 1821
Ormeloxifene Hydrochloride Tablets 1822
Ornidazole 1823
Ornidazole Tablets 1824
Orphenadrine Citrate 1825
Orphenadrine Hydrochloride 1826
Orphenadrine Tablets 1827
OseltamivirPhosphate 1827
Oseltamivir Capsules 1828
Oseltarnivir Oral Suspension 1829
Oxazepam 1830
Oxazepam Tablets 1831
Oxcarbazepine 1832

1805

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Oxcarbazepine Tablets 1833


Oxprenolol Hydrochloride 1834
Oxprenolol Tablets 1834
Oxygen 1835
Oxygen 93 Per cent 1835
Oxytetracycline Dihydrate 1836
Oxytetracycline Injection 1838
Oxytetracycline Hydrochloride 1838
Oxytetracycline Capsules 1840
Oxytetracycline Eye Ointment 1841
Oxytetracycline Hydrochloride Injection 1842
Oxytocin 1842
Oxytocin Injection 1845
Oxytocin Nasal Solution 1846

1806

www.webofpharma.com
IP 2010 OESTRADIOL INJECTION

Oestradiol Benzoate Test solution (a). Dissolve 0.2 g of the substance under
examination in 10 ml ofa mixture of90 volumes of chloroform
and 10 volumes of methanol.
Test solution (b). Dissolve 0.1 g of the substance under
examination in 100 ml ofthe same solvent mixture.

o Reference solution (a). A 0.02 per cent w/v solution of the

~O
substance under examination in the same solvent mixture.
Reference solution (b). A 0.1 per cent w/v solution of
oestradiol benzoate RS in the same solvent mixture.
Apply to the plate 5 III of each solution. After development,
Mol. Wt. 376.5 dry the plate in air until odour of the solvent is no longer
Oestradiol Benzoate is 17~-hydroxyestra-1 ,3,5(1 0)-trien-3-yl detectable, heat at 1100 for 10 minutes, spray the plate while
benzoate. hot with ethanolic sulphuric acid (20 per cent), heat again at
1100 for 10 minutes and examine in ultraviolet light at 365 nm.
Oestradiol Benzoate contains not less than 97.0 per cent and Any secondary spot in the chromatogram obtained with test
not more than 103.0 per cent ofC2sH2s03, calculated on the solution (a) is not more intense than the spot in the
dried basis. chromatogram obtained with reference solution (a).
Category. Oestrogen. Sulphated ash (2.3.18). Not more than 0.2 per cent, determined
Dose. By intramuscular injection, I to 2 mg daily. on 0.5 g.
Description. Colourless crystals or a white, crystalline powder; Loss on drying (2.4.19). Not more than 0.5 per cent, determined
odourless. on 0.5 g by drying in an oven at 105 0 for 3 hours.
Assay. Weigh accurately about 25 mg, dissolve in sufficient
Identification
ethanol (95 per cent) to produce 250.0 ml. Dilute 10.0 ml to
Test A may be omitted if tests Band C are carried out. Test C 100.0 ml with ethanol (95 per cent) and measure the
may be omitted if tests A and B are carried out. absorbance ofthe resulting solution at the maximum at about
231 nm (2.4.7). Calculate the content ofC2sH2s03 taking 500 as
A. Determine by infrared absorption spectrophotometry (2.4.6).
the specific absorbance at 231 nm.
Compare the spectrum with that obtained with oestradiol
benzoate RS or with the reference spectrum of oestradiol Storage. Store protected from light and moisture.
benzoate.
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram.obtained with reference solution (b) Oestradiol Injection
when examined in daylight and in ultraviolet light at 365 nm.
Oestradiol Benzoate Injection
C. To about 1 mg add 0.5 ml of a 5 per cent w/v solution of
Oestradiol Injection is a sterile solution ofOestradiol Benzoate
ammonium molybdate in sulphuric acid; a yellowish green
in Ethyl Oleate or other suitable ester, in a suitable fixed oil or
colour develops which exhibits an intense green fluorescence
in any mixture of these. It may contain suitable alcohols.
when examined in ultraviolet light at 365 nm. Add 1 illl of
sulphuric acid and 9 ml of water; the solution becomes pink Oestradiol Benzoate Injection contains not less than 90.0 per
with a yellowish fluorescence. cent and not more than 110.0 per cent ofthe stated amount of
oestradiol benzoate, C2sH2S03'
Tests Usual strength. 1 mg per ml.
0 0
Specific optical rotation (2.4.22). +57.0 to +63.0 , determined
in a 1.0 per cent wN solution in dioxan. Identification
Related substances. Determine by thin-layer chromatography Determine by thin-layer chromatography (2.4.17), coating the
(2.4.17), coating the plate with silica gel G plate with silica gel G.
Mobile phase. A mixture of 90 volumes of toluene and Mobile phase. A mixture of 80 volumes of toluene and
10 volumes of ethanol (95 per cent). 20 volumes of ethyl acetate.

1807

www.webofpharma.com
OESTRADIOL INJECTION IP 2010

Test solution. Add 10 ml of2, 2, 4-trimethylpentane to a volume Labelling. The label states (1) the nature and composition of
of the injection containing 2 mg of Oestradiol Benzoate and the solvent; (2) that it is meant for intramuscular injection
extract with three quantities, each of 10 ml, of ethanol (70 per only; (3) that any solid matter that may have separated on
cent). Wash the combined extracts with 15 ml of 2,2,4- standing should be redissolved by warming before use.
trimethylpentane, evaporate the ethanolic extract to dryness
using a rotary evaporator and dissolve the residue in 2 ml of
chloroform. Ofloxacin
Reference solution. A 0.1 per cent w/v solution of oestradiol
benzoate RS in chloroform.
Apply to the plate 5 f.!l of each solution. After development,
dry the plate in air, spray with ethanolic sulphuric acid
(20 per cent), heat at 105° for 10 minutes and examine in
ultraviolet light at 365 nm. The principal spot in the
chromatogram obtained with the test solution corresponds to
that in the chromatogram obtained with the reference solution. Mol. Wt. 361.4

Tests Ofloxacin is (RS)-9- fluoro-3-methyl-l0-(4-methylpiperazin-l-


yl)-7-oxo-2,3-dihydro-7H-pyrido[1,2,3,-de]-1 ,4-benzoxazine-
Other tests. Complies with the tests stated under Parenteral 6-carboxylic acid
Preparations (Injections).
Ofloxacin contains not less than 98.5 per cent and not more
Assay. Determine by liquid chromatography (2.4.14). than 101.5 per cent of ClsH2oFN304. calculated on the dried
Test solution (a). Dilute an accurately measured quantity of basis.
the injection containing about 1 mg of Oestradiol Benzoate to Category. Antibacterial.
10.0 ml with a mixture of 90 volumes of cyclohexane and
10 volumes of dioxan. Dose. 200 to 400 mg daily.

Test solution (b). Add 1 ml ofa solut~on prepared by dissolving Description. A pale yellow or bright yellow, cryst~lline powder.
15 mg of 4-hydroxybenzaldehyde (internal standard) in Identification
10.0 ml of dioxan, adding sufficient cyclohexane to produce
100.0 ml (solution A), to an accurately measured quantity of Determine by infrared absorption spectrophotometry (2.4.6).
the injection containing about 1 mg of Oestradiol Benzoate Compare the spectrum with that obtained with ofloxacin RS
and dilute to 10.0 ml with sufficient ofa mixture of90 volumes or the spectrum obtained with the reference solution.
of cyclohexane and 10 volumes of dioxan.
Tests
Reference solution. Add 10 ml of solution A to 10 mg of
oestradiol benzoate RS, accurately weighed, and dilute to Light absorption. Absorbance at 440 urn (2.4.7), of0.5 per cent
100.0 ml with the same solvent mixture. w/v solution in 0.1 M hydrochloric acid is not more than 0.25.

Chromatographic system Related substances. Determine by liquid chromatography


- a stainless steel column 30 cm x 4 mm, packed with porous (2.4.14).
silica particles{1O f.!m), Test solution. Dissolve 10 mg of the substance under
- mobile phase: 90 volumes of cyclohexane and examination in 10 ml of methanol.
10 volumes of dioxan,
Reference solution (a). A 0.1 per cent w/v solution of
- flow rate. 2 ml per minute,
ofloxacin RS in methanol.
- spectrophotometer set at 254 nm,
- injection volume. 20 f.!l. Reference solution (b). Dilute I ml ofreference solution (a) to
100 ml with methanol.
Inject test solutions (a), (b) and the reference solution. The
assay is not valid unless the resolution between the peaks Chromatographic system
due to benzyl alcohol (ifpresent) and oestradiol benzoate and - a stainless steel column 10 cm x 4.6 mm packed with
between the peaks due to oestradiol benzoate and the internal octadecylsilane bonded to porous silica (5 f.!m),
standard is more than 1.5. mobile phase: a mixture of 10 volumes of acetonitrile
and 90 volumes ofphosphate bufferpH 2.4 prepared by
Calculate the content of C2sH2s03 in the injection.
dissolving 27.2 g of monobasic potassium phosphate
Storage. Store protected from light. in 1000 ml of water, adjust the pH to 2.4 with

1808

www.webofpharma.com
IP 2010 OFLOXACIN OPHTHALMIC SOLUTION

orthophosphoric acid, NOTE -Protect the solutions from the light.


- flow rate. 2 ml per minute,
Test solution. Measure accurately a volume containing 50 mg
- spectrophotometer set at 294 nm,
ofOfloxacin in 100.0 ml with mobilephase. Dilute 5.0 ml ofthe
- injection volume. 10 fll.
solution to 50.0 ml with mobile phase.
Inject reference solution (b). The test is not valid unless the
Reference solution. A 0.005 per cent w/v solution of ojloxacin
column efficiency in not less than 1400 theoretical plates.
RS in mobile phase.
Inject the test solution and reference solution (b). Run the
Chromatographic system
chromatogram three times of the principal peak. In the
- a stainless steel column 25 cm x 4.6 mm packed with
chromatogram obtained with the test solution, the area of any
octadecylsilane bonded to porous silica (5 flm),
secondary peak is not more than 0.5 times the area ofthe peak
- mobile phase: a mixture of80 volumes ofbuffer solution
in the chromatogram obtained with reference solution (b) (0.5
prepared by dissolving 6.8 g of potassium dihydrogen
per cent) and the sum of areas of all the secondary peaks is
orthophosphate and 0.47 g sodium 1-hexane
not more than the area of the peak in the chromatogram
sulphonate in 1000 ml of water, add 1 ml of triethylamine
obtained with the reference solution (b) (1.0 per cent).
and adjust the pH to 3.0 with orthophosphoric acid
Heavy Metals (2.3.13). 2.0 g complies with the limit test for and 20 volumes of acetonitrile,
heavy metals, Method B (l0 ppm). - flow rate. 1 ml per minute,
Sulphated ash (2.3.18). Not more than 0.1 per cent. - spectrophotometer set at 294 nm,
Loss on drying (2.4.19). Not more than 0.2 per cent, determined - injection volume. 20 fll.
on 1.0 g by drying in an oven at 105° for 4 hours. Inject the reference solution. The test is not valid unless the
Assay. Weigh accurately about 0.3 g, dissolve in 100 ml of tailing factor is not more than 2.0 and the relative standard
anhydrous glacial acetic acid. l)trate with 0.1 M perchloric deviation for replicate injections is not more than 2.0 per cent.
acid, determining the end-point potentiometrically (2.4.25). Inject the test solution and the reference solution.
Carry out a blank titration.
Calculate the content ofClsH2oFN304 in infusion.
1 ml of 0.1 M perchloric acid is equivalent to 0.03614 g of
ClsH20FN304.
Storage. Store protected from light and moisture. Ofloxacin Ophthalmic Solution
Ofloxacin Ophthalmic Solution is a sterile aqueous solution of
Ofloxacin.
Ofloxacin Infusion
Ofloxacin Ophthalmic Solution contains not less than 90.0 per
Ofloxacin Infusion is a sterile solution of Ofloxacin in 5 per cent and not more than 110.0 per cent of the stated amount of
cent Dextrose Injection or in Sodium Chloride Injection. ofloxacin, ClsH20FN304.
Ofloxacin Infusion contain not less than 90.0 per cent and not Usual strength. 0.3 per cent w/w.
more than 120.0 per cent of the stated amount of ofloxacin,
ClsH20FN304' Identification
Usual strengths. 25 mg; 50 mg; 100 mg; 200 mg; 400mg. In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the peak in the
Identification chromatogram obtained with the reference solution.
In the Assay, the principal peak in the chromatogram obtained Tests
with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. pH (2.4.24).6.0 to 7.2.
Sterility (2.2.11). Comply with the test for sterility.
Tests
Assay. Determine by liquid chromatography (2.4.14).
pH (2.4.24). 3.8to 7.5.
Solvent mixture. Phosphate buffer pH 7.25 prepared by
Other tests. Complies with the tests stated under Parenteral
dissolving 3.56 g of disodium hydrogen phosphate in 1000 ml
Preparation (Infusions).
water, adjusted pH to 7.25 using orthophosphoric acid or
Assay. Determine by liquid chromatography (2.4.14). sodium hydroxide solution.

1809

www.webofpharma.com
OFLOXACIN TABLETS IP 2010

Test solution. Measure accurately a volume of Opthalmic obtained from a solution of known concentration of
Solution, containing 30 mg ofOfloxacin in 100.0 ml ofsolvent ojloxacin RS in the same medium.
mixture. Dilute 1.0 ml of the solution to 10.0 with solvent
D. Not less than 75 per cent of the stated amount of
mixture.
ClsH20FN304.
Reference solution. A 0.003 per cent w/v solution of ojloxacin Related substances. Determine by liquid chromatography
RS in solvent mixture. (2.4.14).
Chromatographic system Test solution. Weigh and powder 20 tablets. Weigh accurately
- a stainless steel column 15 cm x 4.6 mm packed with a quantity ofpowdered tablet containing 100 mg ofOfloxacin,
octadecylsilane bonded to porous silica (5 !Jm) (Such disperse in 60 ml of methanol and dilute to 100 ml with methanol
as TSK GEL), . and filter.
- mobile phase: a mixture of 20 volumes of acetonitrile,
80 volumes of phosphate buffer pH 7.25 prepared by
Reference solution (a). A 0.1 per cent w/v solution of
dissolving 2.54 g of tetrabutyl ammonium hydrogen
ojloxacin RS in methanol.
sulphate and 3.56 g of disodium hydrogen phosphate Reference solution (b). Dilute 1 ml ofreference solution (a) to
in 1000 ml water, 100 ml with methanol.
- flow rate. 1.5 ml per minute, Chromatographic system
- spectrophotometer set at 294 nm, a stainless steel column 15 cm x 4.6 mmpacked with
- injection volume. 20 !Jl. octadecylsilane bonded to porous silica (5 !Jm),
Inject the reference solution. The test is not valid unless the - mobile phase: a mixture of 8 volumes of acetonitrile
relative standard deviation for replicate injections is not more and 92 volumes ofphosphate bufferpH 2. 4 prepared by
than 2.0 per cent. dissolving 27.2 g of monobasic potassium phosphate
in 1000 ml of water, adjust the pH to 2.4 with
Inject the test solution and the reference solution.
orthophosphoric acid,
Calculate the content of CJsH2oFN304. - flow rate. 2 ml per minute,
spectrophotometer set at 294 nm,
Storage. Store protected from light.
- injection volume. 10 !Jl.
Inject reference solution (a). The test is not valid unless the
column efficiency in not less than 1400 theoretical plates.
Ofloxacin Tablets Inject the test solution and reference solution (b). Run the
chromatogram three times of the principal peak. In the
Ofloxacin Tablets contain not less than 90.0 per cent and not
chromatogram obtained with the test solution, the area of any
more than 110.0 per cent of the stated amount ofofloxacin,
secondary peak is not more than the area of the peak in the
CJSH20FN304' chromatogram obtained with reference solution (b) (1.0 per
Usual strengths. 200 mg; 400 mg. cent) and the sum of areas of all the secondary peaks is not
more than twice the area of the peak in the chromatogram
Identification obtained with the reference solution (b) (2.0 per cent).

In the Assay, the principal peak in the chromatogram obtained Other tests. Comply with the tests stated under the Tablets.
with the test solution corresponds to the peak in the Assay. Determine by liquid chromatography (2.4.14).
chromatogram obtained with the reference solution.
Test solution. Weigh and powder 20 tablets. Weigh accurately
Tests a quantity ofpowdered tablet containing 25 mg ofOfloxacin,
disperse in 60 m1 of methanol and dilute to 100 ml with methanol
Dissolution (2.5.2). and filter.
Apparatus No.1, Reference solution. A 0.025 per cent w/v solution of ojloxacin
Medium. 900 ml of 0.1 M hydrochloride acid, RS in methanol.
Speed and time. 50 rpm and 30 minutes.
Chromatographic system
Withdraw a suitable volume ofthe medium and filter. Measure - a stainless steel column 15 cm x 4.6 mm packed with
the absorbance ofthe filtrate, suitably diluted with the medium octadecylsilane bonded to porous silica (5 !Jm),
ifnecessary, at the maximum at about 294 nm (2.4.7). Calculate - mobile phase: a mixture of92 volumes ofbuffer solution
the content ofCJsH2oFN304 in the medium from the absorbance prepared by dissolving 27.2 g ofpotassium dihydrogen

1810

www.webofpharma.com
IP 2010 OLANZAPINE

phosphate in 1000 ml of water and adjust the pH to 2.4 Reference solution (aj. A 0.2 per cent w/v solution of
with orthophosphoric acid and 8 volumes of olanzapine RS in solvent mixture.
acetonitrile, Reference solution (bj. Dilute 1 ml ofreference solution (a) to
- flow rate. 2 ml per minute,
100 ml with solvent mixture.
- spectrophotometer set at 294 nm,
- injection volume. 10 Ill. Chromatographic system

Inject the reference solution. The test is not valid unless the - a stainless steel column 25 cm x 4.6 mm packed with
relative standard deviation for replicate injections is not more octadecylsilane bonded to porous silica (5 Ilm),
than 2.0 per cent. column temperature. 40°,
mobile phase: A. a mixture of 80 volumes of buffer
Inject the test solution and the reference solution. solution pH 6.8 prepared by dissolving 4.825 g of
Calculate the content ofClsHzoFN304. sodium dihydrogen orthophosphate monohydrate in
1000 ml of water, adjust pH to 6.8 with 10 per cent w/v of
Storage. Store protected from light and moisture.
sodium hydroxide and 20 volumes of acetonitrile,
B. acetonitrile,
flow rate. 1.2 ml per minute,
Olanzapine spectrophotometer set at 230 nm,
injection volume. 10 Ill.
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent v/v)
o 100 o
20 63 37
30 45 55
32 100 o
38 100 o
Inject reference solution (b). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
Mol. Wt. 312.4
not less than 2000 theoretical plates.
Olanzapine is 2-methyl-4-(4-methyl-l-piperazinyl)-10H-
Inject the test solution and reference solution (b). In the
thieno[2,3-b][l ,5]benzodiazepine.
chromatogram obtained with the test solution, the area ofany
Olanzapine contains not less than 98.0 per cent and not more secondary peak is not more than 0.5 times the area ofthe peak
than 102.0 per cent ofC 17H zoN 4S, calculated on the anhydrous in the chromatogram obtained with reference solution (b) (0.5
basis. per cent) and the sum of areas of all the secondary peaks is
Category. Antipsychotic. not more than twice the area of the peak in the chromatogram
obtained with the reference solution (b) (2.0 per cent).
Dose. 10 mg daily.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Description. A yellow crystalline powder.
heavy metals, Method B (20 ppm).
Identification Sulphated ash (2.3.18). Not more than 0.1 per cent.
Determine by infrared absorption spectrophotometry (2.4.6). Water (2.3.43). Not more than 1.0 per cent, determined on
Compare the spectrum with that obtained with olanzapine RS LOg.
or with the reference spectrum of olanzapine.
Assay. Weigh accurately about 0.2 g, dissolve in 40 ml of
Tests glacial acetic acid. Titrate with 0.1 M perchloric acid,
Related substances. Determine by liquid chromatography determining the end-point potentiometrically (2.4.25). Carry
(2.4.14). out a blank titration.

Solvent mixture. 40 volumes of water and 60 volumes of 1 ml of 0.1 M perchloric acid is equivalent to 0.01562 g of
acetonitrile. C 17HzoN 4S.
Test solution. Dissolve 50 mg of the substance under Storage. Store protected from light and moisture, at a
examination in 25 ml ofsolvent mixture. temperature not exceeding 30°.

1811

www.webofpharma.com
OLANZAPINE TABLETS IP 2010

Olanzapine Tablets orthophosphate in 900 ml water, add 2 ml of


triethylamine and dilute to 1000 ml with wate/: Adjust
Olanzapine Tablets contain not less than 90.0 per cent and not the pH to 2.5 with orthophosphoric acid and 30 volumes
more than 110.0 per cent ofthe stated amount of olanzapine, of methanol,
C 17HzoN 4S. - flow rate. 1 ml per minute,
Usual strengths. 2.5 rng; 5 mg; 7.5 mg; 10 mg. - spectrophotometer set at 220 urn,
- injection volume. 10 Ill.
Identification Inject the reference solution. The test is not valid unless the
In the Assay, the principal peak in the chromatogram obtained tailing factor is not more than 2.0, the column efficiency is not
with the test solution corresponds to the peak in the less than 2500 theoretical plates. The relative standard
chromatogram obtained with the reference solution. deviation for replicate injections is not more than 2 per cent.
Inject the test solution and the reference solution.
Tests
Calculate the content ofC 17H zoN 4S.
Dissolution (2.5.2).
Apparatus No.1, Storage. Store protected from light and moisture, at a
temperature not exceeding 25°.
Medium. 900 ml of O. 01 M hydrochloric acid,
Speed and time. 50 rpm for 45 minutes.
Withdraw a suitable volume ofthe medium and filter. Determine
by liquid chromatography (2.4.14). Oleic Acid
NOTE - Protect all the solutions ji-om light.
H3C(H2C)6V=V(CH2)6COOH
Test solution. Use the filtrate, if necessary dilute with
dissolution medium. Mol. Wt. 282.5
Reference solution. Weigh 16 mg of olanzapine RS, dissolve Oleic Acid consists mainly of (Z)-octadec-9-enoic acid,
in about 2.5 ml of acetonitrile and dilute to 25 ml with 0.01 M ClsH340Z, together with varying amounts of saturated and
hydrochloroc acid.Dilute suitabely to get 0.00016 per cent other unsaturated fatty acids and is obtained by the hydrolysis
w/v in dissolution medium. of fats or fixed oils and separation of the liquid acids by
Chromatographic system as described under Assay, using expression or other suitable means. It may contain a suitable
Injection volume 50 ).1.1. antioxidant.
Inject the reference solution. The test is not valid unless the Category. Pharmaceutical aid (emulsion adjuvant).
tailing factor is not more than 2.0. The column efficiency is not Description. A clear, yellowish to pale brown, oily liquid; odour,
less than 2500 theoretical plates. characteristic. On exposure to air it darkens in colour and the
Inject the test solution and the reference solution. odour becomes more pronounced.
Calculate the content ofC 17H zoN 4S. Identification
D. Not less than 70 per cent ofthe stated amount ofC 17H zoN 4S. A. To 1 ml add 1 ml of ethanol (95 per cent,),' the solution is
Other tests. Comply with the tests stated under Tablets. clear. It turns orange or red on addition of 0.1 ml of methyl
Assay. Determine by liquid chromatography (2.4.14). orange solution.
Test solution. Take 10 intact tablets to a suitable volumetric B.Take a mixture of 1 ml of nitric acid and 1 ml of water in a
flask, disperse in acetonitrile and dilute with 0.01 M test-tube with an internal diameter of about 12.5 mm and add
hydrochloric acid to get a final concentration of 0.01 per cent 1 ml ofthe substance under examination on the surface of the
w/v ofOlanzapine. mixture. Introduce 0.5 g of copper turnings into the lower
layer and allow to stand under a hood for 4 hours; the upper
Reference solution. Weigh 10 mg of olanzapine RS, dissolve
layer solidifies.
in about 25 rnl of acetonitrile and dilute to 100 ml with 0.01 M
hydrochloric acid. C.Complies with the test for Iodine value (2.3.28).
Chromatographic system Tests
- a stainless steel column 25 em x 4.6 mm packed with
Weight per ml (2.4.29).0.889 g to 0.895 g.
octadecylsilane bonded to porous silica (5 Ilm),
- mobile phase: a mixture of70 volumes of buffersolution Peroxide value (2.3.35). Not more than 10.0.
prepared by dissolving 3 g of ammonium dihydrogen Acid value (2.3.23). 195 to 202, determined on 0.5 g..

1812

www.webofpharma.com
IP 2010 OMEPRAZOLE

Iodine value (2.3.28). 85 to 95, determined by MethodA.. Identification


Water-solubie acids. Shake 5 ml with 5 ml of water for Test A may be omitted if tests Band C are carried out. Tests B
2 minutes, allow the liquids to separate and filter the aqueous and C may be omitted if test A is carried out.
layer through paper moistened with water. To the filtrate add
A. Determine by infrared absorption spectrophotometry (2.4.6).
0.05 ml ofmethyl orange solution; the liquid does not become
Compare the spectrum with that obtained with omeprazole
red.
RS or with the reference spectrum of omeprazole.
Neutral fats and mineral oils. Boil I ml with 5 ml of 0.5 M
B. When examined in the range 230 nm to 360 nm (204.7), a
sodium carbonate and 25 ml of water in a large flask. The
0.002 per cent w/v solution in 0.1 M sodium hydroxide shows
solution, while still hot, is not more opalescent than
absorption maxima at about 276 nm and 305 nm; the ratio of
opalescence standard OS2 (204.1).
the absorbance at about 305 nm to that at about 276 nm, 1.6
Congealing point. Dry about 10 ml by heating to 110° with to 1.8.
frequent stirring, transfer to a test-tube about 20 rom in diameter, C. In the test for Related substances, the principal spot in the
cool and when at 15° immerse the tube in a suitable water-bath chromatogram obtained with the test solution corresponds to
so that the cooling takes place at the rate of2° per minute. Stir that in the chromatogram obtained with reference solution (a).
the sample with a thermometer; it does not become cloudy
until the temperature has fallen to 10°. On further cooling it Tests
congeals to a white solid or semi-solid mass at about 4°.
Appearance of solution. A 2.0 per cent w/v solution in
Sulphated ash (2.3.18). Not more than 0.1 per cent. dichloromethane is clear (204.1). .
Storage. Store protected from light and moisture in a refrigerator Light absorption (204.7). Absorbances of a freshly prepared
esc to 15°). 2.0 per cent w/v solution in dichloromethane at 400 nm,
Labelling. The label states (1) where applicable, that it is used 500 nm and 600 nm are not more than 0.25, 0.10 and 0:10
for external use only; (2) the name and concentration of any respectively.
added antioxidant. Related substances. A. Detennine by thin-layer chromato-
graphy (204.17), coating the plate with silica gel OF254.
Mobile phase. Arnixture of50 volumes ofbenzene, 30 volumes
of ethyl acetate and 10 volumes of methanol.
Omeprazole Test solution. Dissolve 0.4 g of the substance under
examination in 100 ml ofethanol.

O~N:/'
H
I CH3
Reference solution (a). A 004 per cent w/v solution of

~
:;/ II N omeprazole RS in ethanol.
rr,)-8
HCO~N
OCH Reference solution (b). A 0.004 per cent w/v solution of
3
3 CH 3 omeprazole RS in ethanol.
Reference solution (c). A 0.002 per cent w/v solution of
omeprazole RS in ethanol.
C17H19N303S Mol. Wt. 34504
Apply to the plate 10 III of each solution. After development,
Omeprazole is 5-methoxy-2-[[(4-methoxy-3,5-
dry the plate in air and examine in ultraviolet light at 254 nm
dimethylpyridin-2-yl)methyl]sulfinyl]-IH-benzirnidazole.
immediately. Any secondary spot in the chromatogram
Omeprazole contains not less than 99.0 per cent and not mbre obtained with the test solution is not more intense than the
than 101.0 per cent of C17HI9N303S, calculated on the dried spot in the chromatogram obtained with reference solution (c)
basis. and the total intensity of all such spots in the chromatogram
NOTE - Peiform the· tests and assay in subdued light and obtained with the test solution is not more than the intensity
use low-actinic glassware. of the spot obtained with reference solution (b).
B. In the Assay, the sum of the areas of all the secondary
Category. Antiulcerative (proton pump inhibitor).
peaks is not greater than 1.5 rer cent of the total area of all
Dose. For duodenal and gastric ulcers and for reflux peaks.
oesophagitis, 20 to 40 mg daily; for Zollinger-Ellison syndrome,
Heavy metals (2.3.13). The residue obtained in the test for
initially 60 mg once daily; usual range, 20 to 120 mg daily.
Sulphated ash, complies with the limit test fo~ heavy metals,
Description. A white or almost white powder. Method B (20 ppm).

1813

www.webofpharma.com
OMEPRAZOLE IP 2010

Sulphated ash (2.3.18). Not more than 0.2 per cent. filter. Dilute 2 ml of the filtrate to 1QO mI with O.lM sodium
hydroxide.
Loss on drying (2.4.19). Not more than 0.2 per cent, determined
on 1.0 g by drying in an oven at 60° at a pressure not exceeding When exarninedin the.range 230 nm to 360 nm (2.4.7); the
0.7 kPa for 4 hours. resulting solution shows absorption maxima at about 276 nm
and305nm.
Assay. Determine by liquid chromatography (2.4.14).
B. In the Assay, the principal peak in the chromatogram
Test solution. A 0.005 per cent w/v solution of the substance
obtained with the test solution corresponds to the peak due
under examination in the mobile phase.
to omeprazole in the chromatogram obtained with the reference
Reference solution. A 0.005 per cent w/v solution of solution.
omeprazole RS in the mobile phase.
Chromatographic system Tests
- a stainless steel column 30 cm x 3.9 mm, packed with Dissolution (2.5.2).
octadecylsilane bonded to porous silica (5 ILm),
- mobile phase: a mixture of 65 volumes of phosphate A. Apparatus No.1,
buffer pH 7.4 and 35 volumes of acetonitrile, Medium. 900 ml of 0.1 M hydrochloric acid, .
- flow rate. 1 ml per minute, Speed and time. 100 rpm and 2 hours.
- spectrophotometer set at 302 nm, Tap the granules from a capsule slightly with a glass rod to
- injection volume. 20 ILL make them settle to the bottom. Rotate the paddle at 100 rpm
Inject the reference solution. Repeat the procedure at least for 2 hours, drain the solution slowly without losing any
five times and measure the peak responses ofthe peak due to granules. Transfer them quantitatively to a 1OO-ml volumetric
omeprazole. The relative standard deviation of the replicate flask, add 20 ml of 0.1 M sodium hydroxide and mix with the
injections is not more than 2.0 per cent. aid of ultrasound. Dilute to volume with 0.1 M sodium
hydroxide, centrifuge about 15 ml for 5 minutes and dilute
Inject the test solution and the reference solution. 5.0 mI ofthe clear supernatant liquid to 50.0 ml with the mobile
Calculate the content ofC17HJ9N303S, phase. Using the resulting solution as the test solution, carry
out the determination as described in the Assay. Calculate the
Storage. Store protected from light and moisture in a refrigerator
content of C17H19N303S in the supernatant liquid. Calculate
(2° to 8°).
the percentage of omeprazole released in the acid medium by
NOTE - A combination of elevated temperatures (3JO-500) subtracting the content of C17H19N303S in the test solution
and high humidity degrades Omeprdzole. It rapidly degrades from the total content ofomeprazole determined in the Assay.
under acidic conditions.
Not more than 15 per cent ofthe stated amount ofC 17HJ9N303S
is dissolved in 2 hours.

Omeprazole Capsules B. Apparatus No.1,


Medium. 900 ml ofphosphate bufferpH 6.8,
Omeprazole Capsules contain enteric-coated granules of Speed and time. 100 rpm and 45 minutes.
Omeprazole.
Tap the granules from a capsule slightly with a glass rod to
Omeprazole Capsules contain not less than 90.0 per cent and make them settle to the bottom. Rotate the paddle at 100 rpm
not more than 110.0 per cent of the stated amount of for 45 minutes and filter the solution. Using the filtered medium
omeprazole, C17HI9N303S, as the test solution, carry out the determination as described
NOTE - Perform the tests and assay in subdued light and in the Assay. Calculate the content of C17H19N303S in the
use low-actinic glassware. medium.

Usual strength. 20 mg. D. Not less than 70 per cent of the stated amount of
C17HI9N303S,
Identification
Other tests. Comply with the tests stated under Capsules.
A.To a quantity of the contents of the capsules containing
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
50 mg of Omeprazole in a 1OO-ml volumetric flask add about
on 0.5 g of the contents of the capsules by drying in an oven
70 m1 of O. I M sodium hydrOXide. Mixin an ultrasonic bath for
at 60° at a pressure not exceeding 0.7 kPa for 4 hours.
about 5 minutes and heat on a water-bath for 10 minutes.
Cool, make up to volume with 0.1 M sodium hydroxide and Assay. Determine by liquid chromatography (2.4.14).

1814

www.webofpharma.com
IP 2010 ONDANSETRON HYDROCHLORIDE

Test solution. Mix the contents of 20 capsules. Weigh and ondansetron hydrochloride RS or with the reference spectrum
transfer the granules containing·about 20 mg of Omeprazole of ondansetron hydrochloride.
to a 100-ml volumetric flask, add 20 ml of 0.1 M sodium
B. It gives the reactions ofchlorides"(2.3.12).
hydroxide, mix with the aid ofultrasound and dilute to volume
with 0.1 M sodium hydroxide. Centrifuge for 5 minutes and
Tests
dilute 5.0 ml ofthe clear supernatant liquid to 50.0 ml with the
mobile phase. Related substances. Determine by liquid chromatography
Reference solution. Take 20 mg of omeprazole RS in a dry, (2.4.14).
stoppered test-tube, add 20.0 rnl of 0.1 M sodium hydroxide, Use the chromatographic system,the test solution and
shake vigorously for 5 minutes and dilute 1.0 ml ofthe solution reference solution (b) described under Assay.
with the mobile phase to produce 50.0 ml.
Inject the test solution and reference solution (b). In the
Chromatographic system chromatogram obtained with the test solution, the area of any
- a stainless steel column 30 cm x 3.9 mm, packed with secondary peak is not more than 0.2 times the area ofthe peak
octadecylsilane bonded to porous silica (5 11m), in the chromatogram obtained with reference solution (b)
- mobile phase: a mixture of 65 volumes of phosphate (0.2 per cent) and the sum of the areas of all the secondary
buffer pH 7.4 and 35 volumes of acetonitrile, peaks is not more than 0.5 times the area of the peak in the
- flow rate. 1 ml per minute, chromatogram obtained with reference solution (b) (0.5 per
- spectrophotometer set at 302 nm, cent).
- injection volume. 20 Ill.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Inject the reference solution. Repeat the procedure at least
five times and measure the peak responses of the peak due to Water (2.3.43).9.0 to 10.5 per cent, determined on 0.2 g.
omeprazole. The relative standard deviation of the replicate Assay. Determine by liquid chromatography (2.4.14).
injections is not more than 2.0 per cent.
Test solution. Dissolve 50 mg of the substance under
Calculate the content ofCi7H 19N 30 3S in the capsules. examination in 50 ml of the mobile phase and filter. Dilute
Storage. Store protected from light and moisture. 5.0 ml ofthe solution to 50.0 ml with the mobile phase.
Reference solution (a). A 0.01 per cent w/v solution of
ondansetron hydrochloride RS in the mobile phase.
Ondansetron Hydrochloride Reference solution (b). Dilute 1 ml ofreference solution (a) to
100.0 ml with the mobile phase.
Reference solution (c). A solution containing 0.01 per cent
w/v of ondansetron hydrochloride RS and 0.002 per cent w/v
of 3[Dimethylaminomethylj-1, 2, 3, 9-tetrahydro-9-methyl-
4H-carbozol-4-one RS (ondansetron impurity A RS) "in the
mobile phase.
Chromatographic system
ClsH19N30,HCl,2HzO Mol. Wt. 365.9 - a stainless steel column 25 cm x 4.6 mm, packed with
Ondansetron HydrocWoride is (RS)-9-methyl-3-[(2-methyl-lH- porous silica (5 11m) with chemically bonded nitrile
imidazol-l-yl)methyl]-1 ,2,3,9-tetrahydro-4H-carbazol-4-one groups (Such as Supelco LC-CN),
hydrochloride dihydrate. - mobile phase: a mixture of 50 volumes of 0.02 M
monobasic potassium phosphate with the pH
Ondansetron Hydrochloride contains not less than 98.0 per
previously adjusted to 5.4 with 1 M sodium hydroxide
cent and not more than 102.0 per cent of ClsH19N30,HCl,
and 50 volumes of acetonitrile,
calculated on the anhydrous basis.
- flow rate. 1.5 ml per minute,
Category. Antiemetic. (5-HT3 receptor antagonist) - spectrophotometer set at 216 nm,
Description. A white or almost white powder. - injection volume. 10 Ill.

Identification Inject reference solution (c). The test is not valid unless the
relative retention time for ondansetron is about 1.0 and for
A. Determine by infrared absorption spectrophotometry ondimsetron impurity A is about 1.1. The resolution between
(2.4.6).Compare the spectrum with that obtained with ondansetron and ondansetron impurity A is not less than 1.5.

1815

www.webofpharma.com
ONDANSETRON INJECTION IP 2010

Inject reference solution (a). The tailing factor is not more Reference solution (b). Dilute 1 ml ofreference solution (a) to
than 2.0 and the relative standard deviation for replicate 100 ml with the mobile phase.
injections is not more than 2.0 per cent. Reference solution (c). A solution containing 0.01 per cent
Inject the test solution and reference solution (a). w/v of ondansetron hydrochloride RS and 0.005 per cent w/v
of 3[Dimethylaminomethyl}-1, 2, 3, 9-tetrahydro-9-methyl-
Calculate the content ofClsHI9N30,HCI.
4H-carbozol-4-one RS (ondansetron impurity A RS) in the
Storage. Store protected from light and moisture, at a mobile phase.
temperature not exceeding 30°.
Chromatographic system
- a stainless steel column 20 cm x 4.6 lTIlll, packed with
porous silica (5 11m) bonded to nitrile groups (such as
Ondansetron Injection Supelco LC-CN),
- mobile phase: a mixture of 50 volumes of 0.02 M
Ondansetron Hydrochloride Injection monobasic potassium phosphate with the pH
previously adjusted to 5.4 with 1 M sodium hydroxide
Ondansetron Injection contains not less than 95.0 per cent
and 50 volumes of acetonitrile,
and not more than 105.0 per cent of the stated amount of
- flow rate. 1.5 rn1 per minute,
ondansetron, ClsH19N30.
- spectrophotometer set at 216 nm,
Usual strengths. 2 mg; 4 mg; 8 mg. - injection volume. 10 Ill.
Inject reference solution (c). The test is not valid unless the
Identification
relative retention time for ondansetron is about 1.0 and for
In the Assay, the principal peak in the chromatogram obtained ondansetron impurity A is about 1..1. The resolution between
with the test solution corresponds to the peak in the ondansetron and ondansetron impurity A is not less than 1.5.
chromatogram obtained with the reference solution (a). Inject reference solution (a). The tailing factor is not more
than 2.0 and the relative standard deviation for replicate
Tests injections is not more than 2.0 per cent.
pH (2.4.24).3.3 to 4.0. Inject the test solution and reference solution (a).
Related substances. Determine by liquid chromatography Calculate the content ofCIsHI9N30 in the injection.
(2.4.14). Storage. Store protected from light, at a temperature not
Use the chromatographic system, test solution and reference exceeding 30°.
solution (b) described under Assay. Labelling. The label states the strength in terms of the
Inject the test solution and reference solution (b). In the equivalent amount of ondansetron.
chromatogram obtained with the test solution, the area of any
secondary peale is not more than 0.2 times the area ofthe peak
in the chromatogram obtained with reference solution (b) (0.2
per cent) and the sum ofthe areas of all the secondary peaks
Ondansetron Orally Disintegrating
is not more than 1.5 times the area of the peak in the Tablets
chromatogram obtained with reference solution (b) (1.5 per
Ondansetron Orally Disintegrating Tablets contain not less
cent).
than 90.0 per cent and not more than 110.0 per cent of the
Bacterial endotoxins (2.2.3). Not more than 9.9 Endotoxin stated amount of ondansetron, ClsH19N30.
Units per mg of ondansetron.
Usual strength. 4 mg; 8 mg.
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections). Identification
Assay. Determine by liquid chromatography (2.4.14).• In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the peak in the
Test solution.Transfer an accurately measured volume of the chromatogram obtained with the reference solution.
injection containing about 2 mg of ondansetron to a 25-ml
volumetric flask, dilute to volume with the mobile phase and mix. Tests
Reference solution (a). A 0.01 per cent w/v solution of Dissolution (2.5.2).
ondansetron hydrochloride RS in the mobile phase. Apparatus No.1,

1816

www.webofpharma.com
IP 2010 ONDANSETRON ORALLY DISINTEGRATINGTABLETS

Medium. 500 ml of 0.1 M hydrochloric acid, Inject reference solution (d) and the test solution. In the
Speed and time. 50 rpm and 10 minutes. chromatogram obtained with the test solution, the area of the
peak due to 2-methylimidazole is not more than 0.1 5 per cent,
Withdraw a suitable volume ofthe medium and filter. Measure
multiply with response factor 0.5, the area of peak due to
the absorbance of the filtrate, suitably diluted with the
ondansetron impurity D is not more than 0.12 per cent, multiply
dissolution medium ifnecessary, at the maximum at about 31 0
with response factor 1.2. The area of any other secondary
nm (2.4.7). Calculate the content ofClsH19N30 in the medium
peak is not more than 0.1 per cent and the sum of all the
from the absorbance obtained by using a solution of known
secondary peaks is not more than 0.5 per cent.
concentration of ondansetron RS in same medium.
Water (2.3.43). Not more than 4.0 per cent, determined on
D. Not less than 80 per cent ofthe stated amount ofC ls H19N30.
LOg.
Related substances. Determine by liquid chromatography
(2.4.14). Other tests. Comply with the tests stated under tablets.

Test solution. Disperse a quantity of powdered tablets Assay. Determine by liquid chromatography (2.4.14).
containing about 40 mg of ondansetron with 100.0 ml of the Test solution. Weigh and powder 20 tablets. Disperse a quantity
mobile phase. Centrifuge a portion ofthis solution at 3000 rpm of powder containing about 40 mg of ondansetron with
for 10 minutes. Use the supernatant. 100.0 mlO.01 Mhydrochloricacid.Dilute 1.0 ml ofthe solution
Reference solution (a). A 0.004 per cent w/v solution of to 10.0 ml with 0.01 Mhydrochloric acid.
1,2,3,9-tetrahydro-9-methyl-3-methylene-4H-carbazol-4-one Reference solution (a). A 0.004 per cent w/v solution of
RS (ondansetron impuirty DRS) in acetonitrile. ondansetron RS in 0.01 M hydrochloric acid.
Reference solution (b). A 0.004 per cent w/v solution of Reference solution (b). A 0.014 per cent w/v solution of
2-methylimidazole in acetonitrile. 3[Dimethylamino methylj-1,2,3,9-tetrahydro-9-methyl-H-
Reference solution (c). A 0.004 per cent w/v solution of carbozol-4-one-RS (ondansetron impurity A RS) in 0.01 M
ondansetron RS in acetonitriile. hydrochloric acid.
Reference solution (d). Dilute 5.0 ml ofreference solution (c) Reference solution (c). Transfer 8.0 ml each of reference
to 100.0 ml with the mobile phase. solution (a) and (b) to 50-ml volumetric flask and dilute to
Reference solution (e). Dilute 10.0 ml ofreference solution (c) volume with 0.01 M hydrochloric acid.
to 100.0 ml with the mobile phase. Chromatographic system
Reference solution (j). Transfer 5.0 ml of each reference - a stainless steel column 25 cm x 4.6 rom, packed with
solution (a), (b) and (c) to a 100-ml volumetric flask, dilute to porOl.jS silica bonded to nitrile groups (5 /-lm),
volume with the mobile phase. - mobile phase: a mixture of52 volumes of0.272 per cent
w/v solution of monobasic potassium phosphate,
Chromatographic system
adjusted to pH 5.4 with 1 M sodium hydroxide and 48
- a stainless steel column 25 cm x 4.6 rom, packed with
volumes of acetonitrile,
porous silica chemically bonded to nitrile groups (5 /-lm),
- flow rate. 1.5 ml per minute,
- mobile phase: a mixture of 80 volumes of phosphate
- spectrophotometer set at 216 urn,
buffer prepared by dissolving 2.72 g of monobasic
- injection volume. 10 /-ll.
potassium phosphate in 1000 ml of water, adjusted to
pH 5.4 with 1 M sodium hydroxide and 20 volumes of Inject reference solution (c). The test is not valid unless the
acetonitrile, resolution between the peaks due to ondansetron and
- flow rate. 1.5 ml per minute, ondansetron impurity A is not less than 1.5. The relative
- spectrophotometer set at 216 nm, retention time with reference to ondansetron for ondansetron
- injection volume. 20 /-ll. impurity A is about 1.1. The tailing factoris not more than 2.0
for ondansetron peak.
Inject reference solution (t). The test is not valid unless the
resolution between ondansetron and adjacent peak is not less Inject reference solution (a) and the test solution.
than 1.5, the theoretical plates is not less than 8000 for
Calculate the content ofCIsH19N30 in the tablets.
ondansetron and tailing factor is not more than 2.0.
Storage. Store protected form light and moisture, at a
Inject reference solution (e). The signal to noise ratio for the
temperature not exceeding 30°.
ondansetron peak is not less than 15. The relative retention
time with reference to ondansetron for 2-methylimidazole is Labelling. The label states the active ingredient in terms of
about 0.16 and for ondansetron impurity D is about 0.45. equivalent amount of ondansetron.

1817

www.webofpharma.com
ONDANSETRON ORAL SOLUTION IP 2010

Ondansetron Oral Solution Chromatographic system


- a stainless steel column 25 cm x 4.6 mm, packed with
Ondansetron Hydrochloride Oral Solution porous silica bonded to nitrile groups (5 /J.m),
- mobile phase: a mixture of 80 volumes of 0.02 M
Ondansetron Oral Solution is a solution of Ondansetron
monobasic potassium phosphate, previously adjusted
Hydrochloride in a suitable vehicle..
to pH 5.4 and 20 volumes of acetonitrile,
Ondansetron Oral Solution contains not less than 95.0 per - flow rate. 1.5ml per minute,
cent and not more than 105.0 per cent ofthe stated amount of - spectrophotometer set at 328 nm,
ondansetron, ClsH'9N30. injection volume. 20/J.1.
Usual strength. 2 mg per 5 ml; 4 mg per 5 ml. Inject reference solution (b). The test is not valid unless the
resolution between ondansetron impurity D and ondansetron
Identification impUlity C is not less than 2.0, the tailing factor for ondansetron
impurity D is not more than 2.0 and the relative standard
A. Detennine by thin-layer chromatography (2.4.17), coating deviation forreplicate injections is notmore than 4.0 per cent.
the plate with silica gel G.
Inject reference solution (a) and the test solution.
Mobile phase. A mixture of 90 volumes of chloroform, 50 Calculate the content of ondansetron impurity D in the oral
volumes of ethyl acetate, 40 volumes of methanol and 1volume solution.
of ammonium hydroxide.
Related substances. Detennine by liquid chromatography
Test solution. Dilute an accurately measured volume of the (2.4.14), as described under Assay with the following
oral solution containing about 20 mg of ondansetron to 100.0 modifications.
ml in a mixture of 50 volumes of methanol and 50 volumes of
Test solution. Weigh accurately a quantity of oral solution
water.
containing about 45 mg of Ondansetron Hydrochloride in
Reference solution. A 0.025 per cent w/v solution of 50-ml volumetric flask, dissolve and dilute with the mobile
ondansetron RS in methanol. phase and mix. Dilute 5.0 ml ofthis solution to 50 ml with the
mobile phase.
Apply 10 /J.I of each solution. Allow the solvent front to rise
15 cm. Dry the plate in air and examine in ultraviolet light at Inject reference solution (c). The test is not valid unless the
254 nm. The chro\natogram obtained with the test solution resolution between ondansetron impuirty A and ondansetron
corresponds to that obtained with the reference solution. is not less than 1.5, the tailing factor is not less than 2.0 and
the relative standard deviation for replicate injections is not
B. In the Assay, the principal peak in the chromatogram more than 1.5 per cent. The relative retention time with
obtained with the test solution corresponds to the peak in the reference to ondansetron for ondansetron impurity A is
chromatogram obtained with the reference solution. about 1.1.
Inject reference solution (b) and the test solution. In the
Tests chromatogram obtained with the test solution, the area ofthe
peak due to ondansetron impurity D is not more than
pH (2.4.24).3.3 to 4.0.
0.1 per cent, the area of each peak due to imidazole, 2-methyl
Ondansetron impurity D. Not more than 0.1 per cent. imidazole, des-(-methy1)ondansetron hydrochloride,
Detennine by liquid chromatography (2.4.14). N-Desmethyl ondansetron maleate, ondansetron impurity A
is not more than 0.2 per cent. The area of any other secondary
Test solution. Dilute an accurately measured volume of oral peak is not more than 0.2 per cent and the sum of all the
solution to obtain a solution having a concentration of 0.08 secondary peaks is not more than 0.5 per cent.
per cent w/v of ondansetron in the mobile phase. Other tests. Complies with the tests stated under Oral Liquids.
Reference solution (a). A 0.00005 per cent w/v solution of Assay. Detennine by liquid chromatography (2.4.14).
1,2,3,9-tetrahydro-9-methyl-3-methylene-4H-carbazol~4-on
e
RS (ondansetron impurity D RS) in the mobile phase. Test solution. Dilute a volume of the oral solution containing
about 9 mg ofondansetron to 100.0 ml with themobi1e phase.
Reference solution (b). A solution containing 0.00005 per cent
w/v of ondansetron impurity D RS and 0.0002 per cent w/v of Reference solution (a). A 0.01 per cent w/v solution of
N-methyl [2 -(2 -methylbenzhydryloxy) ethyl} am ine ondansetron hydrochloride RS in the mobile phase.
hydrochloride RS (ondansetron impurity C RS) in the mobile Reference solution (b). Dilute 1 ml ofreference solution (a) to
phase. 100.0 ml with the mobile phase.

1818

www.webofpharma.com
IP 2010 ONDANSETRON TABLETS

Reference solution (c). A solution containing 0.01 per cent Medium. 500 ml ofwatel;
w/v of ondansetron hydrochloride RS and 0.002 per cent w/v Speed and time. 50 rpm and 45 minutes.
of 3[DimethylaminomethylJ-l, 2, 3, 9-tetrahydro-9-methyl-
Withdraw a suitable volume ofthe medium and filter. Measure
4H-carbozol-4-one RS (ondansetron impurity A RS) in the
the absorbance of the filtered solution, suitably diluted with
mobile phase.
the medium if necessary, at the maximum at about 310 nm
Chromatographic system (2.4.7). Calculate the content ofCIsHI9N30 in the medium from
- a stainless steel column 25 cm x 4.6 mrn, packed with the absorbance obtained from a solution of known
porous silica (5 11m) with chemically bonded nitrile concentration of ondansetron hydrochloride dihydrate RS
groups (such as Supelco LC-CN), in the same medium.
- mobile phase: a mixture of 50 volumes of 0.02 M
1 mg of CIsHI9N30 is equivalent to 1.25 mg of
monobasic potassium phosphate with the pH
C,sHI9N30,HCl,2HzO.
previously adjusted to 5.4 with 1 M sodium hydroxide
and 50 volumes of acetonitrile, D. Not less than 70 per cent ofthe stated amount ofClsHl9N30.
flow rate. 1.5 ml per minute, Related substances. Determine by liquid chromatography
- spectrophotometer set at 216 nm, (2.4.14).
- injection volume. 10 Ill. Use the chromatographic system, test solution and reference
Inject reference solution (c). The test is not valid unless the solution (b) described under Assay.
relative retention time for ondansetron is about 1.0 and for Inject the test solution and reference solution (b). In the
ondansetron impurity A is about 1.1. The resolution between chromatogram obtained with the test solution, the area ofany
ondansetron and ondansetron impurity A is not less than 1.5. secondary peak is not more than 0.2 times the area ofthe peak
Inject reference solution (a) The tailing factor is not more in the chromatogram obtained with reference solution (b) (0.2
than 2.0 and the relative standard deviation for replicate per cent) and the sum of the areas of all the secondary peaks
injections is not more than 2.0 per cent. is not more than 0.5 times the area of the peak in the
Inject tbe test solution and reference solution (a). chromatogram obtained with reference solution (b) (0.5 per
cent).
Calculate the content ofClsHl9N30, HCl in the oral solution.
Uniformity of content. Comply with the test stated under
Microbial contamination (2.2.9). Complies with the microbial Tablets.
contamination test. The total aerobic count is not more than
Determine by liquid chromatography (2.4.14), using the
100 cfu per g, Enterobacteriaceae count is not more than 10
chromatographic conditions and reference solution (a)
cfu per g and the total combined molds and yeasts count is
described in the Assay.
not more than 50 cfu per g.
Test solution. Disperse one tablet in' the mobile phase and
Storage. Store protected from light and moisture.
dilute to obtain a solution containing 0.01 per cent w/v of
Ondanseteron Hydrochloride and filter.
Calculate the content ofCIsHI9N30 in the tablet.
Ondansetron Tablets Other tests. Comply with the tests stated under Tablets.
Ondansetron Hydrochloride Tablets Assay. Determine by liquid chromatography (2.4.14).
Ondansetron Tablets contain not less than 95.0 per cent and Test solution. Weigh and powder 20 tablets. Weigh accurately
not more than 105.0 per cent of the stated amount of a quantity ofpowder containing about 50 mg of ondansetron,
ondansetron, ClsH19N30. disperse in 50 ml ofthe mobile phase and filter. Dilute 5.0 ml of
Usual strengths. 2 mg; 4 mg; 8 mg the solution to 50.0 ml with the mobile phase.
Reference solution (a). A 0.01 per cent w/v solution of
Identification ondansetron hydrochloride RS in the mobile phase.
In the Assay, the principal peak in the chromatogram obtained Reference solution (b). Dilute 1 ml ofreference solution (a) to
with the test solution corresponds to the peak in the 100 ml with the mobile phase.
. chromatogram obtained withthe reference solution (a). Reference solution (c). A solution containing 0.01 per cent
w/v of ondansetron hydrochloride RS and 0.002 per cent w/v
Tests
of 3[DimethylaminomethylJ-l, 2, 3, 9-tetrahydro-9-methyl-
Dissolution (2.5.2) 4H-carbozol-4-one RS (ondansetron impurity A RS) in the
Apparatus No.1, mobile phase.

1819

www.webofpharma.com
ORAL REHYDRATION SALTS IF 2010

Chromatographic system Composition ofthe formulation in terms ofthe amount, in g, to


- a stainless steel column 25 cm x 4.6 mm, packed with be dissolved in sufficient water to produce 1000 ml.
porous silica (5 /lm) bonded to nitrile groups (such as
Sodium Chloride 2.6
Supelco LC-CN),
Dextrose (anhydrous) 13.5
mobile phase: a mixture of 50 volumes of 0.02 M
or
monobasic potassium phosphate with the pH
Dextrose Monohydrate 14.85
previously adjusted to 5.4 with 1 M sodium hydroxide
and 50 volumes of acetonitrile, Potassium Chloride 1.5
flow rate. 1.5 ml per minute, Sodium Citrate 2.9
spectrophotometer set at 216 nm, The molar concentrations of sodium, potassium, chloride and
- injection volume. 10 Ill. citrate ions in terms ofmillimoles per litre are given below:
Inject reference solution (c). The test is not valid unless the mmol/l
relative retention time for ondansetron is about 1.0 and for Sodium 75
ondansetron impurity A is about 1.1. The resolution between Potassium 20
ondansetron and ondansetron impurity A is not less than 1.5.
Chloride 65
Inject reference solution (a). The tailing factor is not more Citrate 10
than 2.0 and the relative standard deviation for replicate
Dextrose 75
injections is not more than 2.0 per cent.
The total osmolar concentration of the solution in terms of
Inject the test solution and reference solution (a).
mOsmol per litre is 245.
Calculate the content ofC,sH,9N30 in the tablets.
Oral Rehydration Salts contain not less than 90.0 per cent and
Storage. Store protected from light and moisture, at a not more than 110.0 per cent ofthe stated amount ofDextrose
temperature not exceeding 30°. (anhydrous) or Dextrose Monohydrate (as appropriate) and
Labelling. The label states the strength in terms of the ofthe requisite amQunts ofsodium, Na, potassium, K, chloride,
equivalent amount of ondansetron. Cl, and citrate, C6H s0 7, calculated from the stated amounts of
the relevant constituents.
Description. A white to creamy-white, amorphous or
crystalline powder; odourless.
Oral Rehydration Salts
ORS Powder Identification
Oral Rehydration Salts are dry, homogeneously mixed powders A. When heated, melting and charring occurs and an odour of
containing Dextrose, Sodium Chloride, Potassium Chloride burnt sugar is produced.
and either Sodium Bicarbonate or Sodium Citrate for use in B. Add a few drops ofthe solution prepared as directed in the
oral rehydration therapy after being dissolved in the requisite label to 5 ml of potassium cupri-tartrate solution; a copious
amount ofwater. red precipitate is produced on boiling.
They may contain suitable flavouring agents and, where C. Gives reaction A of sodium salts, reaction A of potassium
necessary, suitable flow agents in the minimum quantity salts and reaction A of chlorides (2.3.1).
required to achieve a satisfactory product but may not contain
artificial sweetening agents like mono- and/or poly- D. A quantity containing about 50 mg of citric acid gives the
saccharides. If saccharin/saccharin sodium or aspartame is reactions ofcitrates (2.3.1).
used in preparations meant for paediatric use, the concentration
of saccharin should be such that its daily intake is not more
Tests
than 5 mg/kg ofbody weight and that of aspartame should be Uniformity of weight. Comply with the test for contents of
such that its daily intake is not more than 40 mg/kg of body packaged dosage forms (2.5.6).
weight.
Seal test (onlyfor sachets). Loosely bundle 10 sachets with a
Category: Replacement solution for diarrhoeal rehydration. rubber band and submerge the bundle under water in a vacuum
Usual strength. A formulation of reduced osmolarity (given desiccator maintained at a pressure not exceeding 18 kPa for
below) recommended by the World Health Organization one minute. Examine the bundle for any fine stream ofbubbles.
(WHO) for the Diarrhoeal Diseases Control Programme, and Re-establish normal pressure and open the bundle. No
ofthe United Nations Children's Emergency Fund (UNICEF). penetration of water is observed in any sachet.

1820

www.webofpharma.com
IP 2010 ORMELOXIFEN HYDROCHLORIDE

Other tests. Comply with the tests stated under Oral Powders. in g ofC6H 120 6and C6H1206,H20 respectively, as appropriate,
in the weight taken for the Assay.
Assay. Carry out the following assays on the well-mixed
contents of an individual sachet or on a suitable sample from Storage. Store protected from moisture in sachets, preferably
the well-mixed contents ofa bulle container. Where the amount made ofaluminum foil, containing sufficient powder for a single
in an individual sachet is insufficient to carry out all the assays, dose or for a day's treatment or for use in hospitals, in bulle
take a separate sachet for the Assay for citrate and for the containers containing sufficient quantity to produce a volume
Assay for dextrose. For the Assays for total sodium, for of solution appropriate to the daily requirements of the
potassium and for total ch.loride weigh accurately about hospital concerned.
8.0 g and dissolve in sufficient water to produce 500.0 ml
Labelling. The label states (l) for sachets, the total weights,
(solution A).
in g, of each constituent; (2) for bulle containers, the weights,
For total sodium - Dilute a suitable volume of solution A in g, of each constituent in a stated quantity, in g, of the oral
with a s1,1fficient volume of a solution of strontium chloride powder; (3) the molar concentration in millimoles per litre of
such thatthe final solution contains a 1500 to 2000-fold excess sodium, potassium, chloride and citrate ions, and of dextrose
of strontium ions and determine by Method A for atomic as well as the total osmolar concentration in mOsmol per litre
absorption spectrophotometry (2.4.2), measuring at 589 nm of the solution prepared from the oral powder; (4) the total
and using sodium solution AAS, suitably diluted with the weight ofthe contents of the container; (5) the directions for
strontium chloride solution, for the standard solutions or, use; (6) that any portion ofthe solution prepared from the oral
alternately by Method A for flame photometry (2.4.4). powder that remains unused for 24 hours after preparation
should be discarded; (7) the storage conditions. g, of the oral
1 g ofSodium Chloride, and ofSodium Citrate is equivalent to
powder.
0.3934 and 0.2345 g ofNa respectively.
For potassium - Dilute a suitable volume of solution A with
a sufficient volume of solution of strontium chloride such
that the final solution contains a 1500 to 2000-fold excess of Ormeloxifene Hydrochloride
strontium ions and determine by Method A for atomic
Centchroman Hydrochloride; Centchroman
absorption spectrophotometry (2.4.2), measuring at 767 nm
and using potassium solution AAS, suitably diluted with the
strontium chloride solution, for the standard solutions or,
H3 CO 0 ,CH 3
alternatively by Method A for flame photometry (2.4.4).
CH 3
1 g ofPotassium Chloride is equivalent to 0.5245 g ofK.
For total chloride - Titrate 50 ml of solution A with 0.1 M I I":

Y
~
silver nitrate using potassium chromate solution as indicator. • HCI
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g ofCI.
1 g of Sodium Chloride, and of Potassium Chloride is
equivalent to 0.6066 and 0.4756 g ofCl respectively. O~O
For citrate - Weigh accurately about 2.0 g and dissolve in
50 ml of anhydrous glacial acetic acid by heating at about
50°. Cool, allow to stand for 10 minutes. Titrate with 0.1 M Mol. Wt. 493.5
perchloric acid, using 1-naphtholbenzein solution as
Ormeloxifene Hydrochloride is trans-7-methoxy-2,2-
indicator. Carry OJ.lt a blank titration.
dimethyl-3-phenyl-4[4-(2-
1 ml of 0.1 M perchloric acid is equivalent to 0.006303 g of pyrrolidinoethoxy)phenyl]chroman hydrochloride.
CJfS07' Ormeloxifene Hydrochloride contains not less than 98.5 per
1 g ofSodium Citrate is equivalentto 0.6430 g ofC6Hs0 7• cent and not more than 101.5 per cent of C30H3sN03,HCl,
For dextrose - Weigh accurately about 7.5 g, dissolve in calculated on the dried basis.
40 ml of water, add 0.5 ml of dilute ammonia solution, and Category. Antifertility agent (female oral contraceptive).
dilute to 50 ml with water. Mix well, allow to stand for
Dose. 120 mg twice a week, 3 days apart, for three months and
30 minutes, filter the solution, ifturbid, and measure the optical
thereafter once a week.
rotation in a 2-dm tube (2.4.22). The observed rotation, in
degrees, multiplied by 0.9477 and 1.0424 represents the weight Description. A white to off-white powder.

1821

www.webofpharma.com
ORMELOXIFEN HYDROCHLORIDE IP 2010

Identification . Chromatographic system


a stainless steel column 25 cm x 4.6 mm,packed with
A. Determine by infrared absorption spectrophotometry (2.4.6).
octadecylsilane bonded to porous silica (5 Ilm),
Compare the spectrum with that obtained with ormeloxifene
mobile phase: 80 volumes of acetonitrile and 20 volumes
hydrochloride RS orwith the reference spectrum oformeloxifene.
of water containing 0.04 per cent w/v solution of
B, When examined in the range 230 urn to 360 run (2.4.7), a tetramethylammonium hydroxide adjusted to pH 7.6
0.01 per cent w/v solution in methanol shows absorption with phosphoric acid, . .
maxima at about 278 and 282 urn. flow rate. 1.5 ml per minute,
C. Determine by thin-layer chromatography (2.4.17), coating spectrophotonieter set at 280 run,
the plate with silica gel G injection volume. 20 Ill.

Mobile phase. A mixture of 90 volumes of chloroform and Inject the reference solution and use an attenuation such that
10 volumes of methanol. the response for theprincipal peak due to trans-ormeloxifene
hydrochloride is more than 50 per cent of the full-scale
Test solution. Dissolve 0.25 g of the substance under
deflection ofthe recorder. Make a total ofsix injections. When
examination in 100 ml of chloroform.
the chromatograms are recorded under the conditions
Reference solution. A 0.25 per cent w/v solution of described above, trans-ormeloxifene hydrochloride is eluted
ormeloxifene hydrochloride RS in chloroform. before cis-ormeloxifene hydrochloride.The test is not valid
Apply to the plate 2 III of each solution. After development, unless the relative standard deviation ofsix replicate injections
dry the plate in air, until the odour ofthe solvents is no longer is not greater than 6 per cent and the resolution between the
detectable and spray with a 0.3 per cent w/v solution of peaks due to cis-ormeloxifene hydrochloride and trans-
potassium permanganate. The principal spot in the ormeloxifene hydrochloride is greater than 1. If necessary,
chromatogram obtained with the test solution corresponds to adjust the proportions and the flow rate of the mobile phase
that in the chromatogram obtained with the reference solution. to obtain proper resolution. Inject a suitable volume of test
solutiop and record the response. From the average peak areas
D. To 1 ml ofa 2 per centw/v solution in ethanol (95 percent)
of the six replicate analyses calculate the content of trans-
add 1 ml of a saturated solution of picric acid in water, stir
ormeloxifene hydrochloride and cis-ormeloxifene
well and set aside for 5 minutes. The yellow precipitate obtained
hydrochloride in the substance under examination by
after washing with water and drying at 60° for 4 hours melts at
comparing with the peak responses for trans-ormeloxifene
212°to 218°(2.4.21).
hydrochloride and cis-ormeloxifene hydrochloride respectively
Tests obtained with reference solution.
Total basic substances. Weigh accurately 0.5 g, dissolve in Storage. Store protected from moisture.
25 ml of anhydrous glacial acetic acid, add 10 ml of a 5 per
cent w/v solution of mercuric acetate in anhydrous glacial
acetic acid. Titrate with 0.1 M perchloric acid, using clystal
violet solution as indicator to a bluish green end-point. Carry Ormeloxifene Hydrochloride Tablets
out a blank titration.
Centchroman Hydrochloride Tablets; Centchroman
1 ml of 0.1 M perchloric acid is equivalent to 0.04935 g of Tablets
C30H3sN03,HCl.
Ormeloxifene Hydrochloride Tablets contain not less than
cis~Isomer.Not more than 1.5 per cent ofthe total content of 90.0 per cent and not more than 110.0 per cent of the stated
hydrochlorides of trans-and cis-isomers determined in the amount ofOrmeloxifene hydrochloride, C30H3sN03,HCI.
Assay.
Usual strength. 30 mg.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 3.5 per cent, determined Identification
on 1.0 g by drying in an oven at 105° for 4 hours.
Shake a quantity of the powdered tablets containing 0.1 g of
Assay. Determine by liquid chromatography (2.4.14). Ormeloxifene Hydrochloride with! 0 ml of chloroform, filter
Test solution.. A 0.004 per cent w/v solution of the substance and evaporate the filtrate to dryness at a pressure not
under examination in the mobile phase. exceeding 0.7 kPa. The residue complies with the following
Reference solution. A solution containing 0.004 per cent w/v tests.
of trans-ormeloxifene hydrochloride RS and 0.0006 per cent A. Determine by infrared absorption spectrophotometry (2.4.6).
w/v of cis-ormeloxifene hydrochloride RS in the mobile phase. Compare the spectrum with that obtained with ormeloxifene

1822

www.webofpharma.com
IP 2010 ORNlDAZOLE

hydrochloride RS or with the reference spectrum of Chromatographic system


ormeloxifene. a stainless steel column 25 cm x 4.6 mm, packed with
B. When examined in the range 230 nm to 360 nm (2.4.7), a octadecylsilane bonded to porous silica (5 Ilm),
mobile phase: 80 volumes of acetonitrile and 20 volumes
0.01 per cent w/v solution in methanol shows absorption
maxima at about 278 and 282 nm. of water containing 0.04 per cent w/v solution of
tetramethylammonium hydroxide adjusted to pH 7.6
C. Determine by thin-layer chromatography (2.4.17), coating with phosphoric acid,
the plate with silica gel G - flow rate. 1.5 ml per minute,
Mobile phase. A mixture of 90 volumes of chloroform and - spectrophotometer set at 280 nm,
10 volumes of methanol. injection volume. 20 Ill.
Inject the reference solution and use an attenuation such that
Test solution. Dissolve 0.25 g of the substance under
the response for the principal peak due to trans-ormeloxifene
examination in 100 ml of chloroform.
hydrochloride is more than 50 per cent offull-scale deflection
Reference solution. A 0.25 per cent w/v solution of of the recorder. Make a total of six injections. When the
ormeloxifene hydrochloride RS in chloroform. chromatograms are recorded under the conditions described
Apply to the plate 2 III of each solution. After development, above, trans-ormeloxifene hydrochloride is eluted before cis-
dry the plate in air, until the odour of the solvents is no longer ormeloxifene hydrochloride.The test is not valid unless the
detectable and spray with ~ 0.3 per cent w/v solution of relative standard deviation of six replicate injections is not
potassium permanganate. The principal spot in the greater than 6 per cent and the resolution between the peaks
chromatogram obtained with the test solution corresponds to due to cis-ormeloxifene hydrochloride and trans-ormeloxifene
that in the chromatogram obtained with the reference solution. hydrochloride is greater than 1. If necessary, adjust the
proportions and the flow rate of the mobile phase to obtain
Tests proper resolution. Inject a suitable volume of test solution
and record the response. From the average peak areas of the
Dissolution (2.5.2).
six replicate analyses calculate the content of trans-
Apparatus No.2, ormeloxifene hydrochloride and cis-ormeloxifene
Medium. 900 ml of water, hydrochloride in the substance under examination.
Speed and time. 100 rpm and 45 minutes.
Storage. Store protected from moisture.
Withdraw a suitable volume ofthe medium , and filter. Measure
the absorbance of the filtrate, suitably diluted if necessary, at
the maximum at about 278 nm (2.4.7). Calculate the content of Ornidazole
C30H3sN03,HCl in the medium from the absorbance obtained
OH
from a solution of known concentration of Ormeloxifene
hydrochloride RS in water. ~CI
D. Not less than 70 per cent of the stated amount of
C30H3sN03,HCl. 02N~rCH3
cis-Isomer. Not more than 1.5 per cent ofthe total content of
ormeloxifene hydrochlorides of trans- and cis-isomers C7HIQClN30 3 Mol. Wt. 219.6
determined in the Assay. Omidazole is (RS)-I-chloro-3-(2-methyl-5-nitroimidazol-l-
Other tests. Comply with the tests stated under Tablets. yl)propan-2-ol.
Omidazole contains not less than 98.0 per cent and not more
Assay. Determine by liquid chromatography (2.4.17).
than 101.0 p~r cent ofC7H IQClN30 3,calculated on the anhydrous
Test solution. Weigh and powder 20 tablets. Shake a quantity basis.
ofthe powder containing 0.1 g ofOrmeloxifene Hydrochloride Category. Antiamoebic.
with three quantities, each of 5 ml, of methanol, centrifuge
Dose. 500 mg twice a day orally for five days.
each extract, dilute the combined extracts to 25 ml with
methanol and then dilute 250 III ofthe resulting solution to 25 Description. A white to yellowish white crystalline powder.
ml with the mobile phase. Identification
Reference solution. A solution containing 0.004 per cent w/v A. Determine by infrared absorption spectrophotometry (2.4.6).
of trans-ormeloxifene hydrochloride RS and 0.0006 per cent Compare the spectrum with that obtained with ornidazole RS
w/v of cis-ormeloxifene hydrochloride RS in the mobile phase. or with the reference spectrum of omidazole.

1823

www.webofpharma.com
ORNIDAZOLE IP 2010

B. When examined in the range 230 nm to 360 nm (2.4.7), a Storage. Store protected from light and moisture, at a
0.002 per cent w/v solution in 0.1 M hydrochloric acid shows temperature not exceeding 30°.
an absorption maximum at about 277 nm; absorbance at
277 nm, 0.580 to 0.630.

Tests Ornidazole Tablets


Ornidazole Tablets contain not less than 95.0 per cent and not
Related substances. Determine by liquid chromatography
more than 105.0 per cent of the stated amount of ornidazole,
(2.4.14).
C7H IOCIN30 3•
Test solution. Dissolve 25 mg of the substance under
Usual strength. 500 mg.
examination in 25.0 ml ofthe mobile phase.
Reference solution (a). A 0.001 per cent w/v solution of Identification
2-methyl-5-nitroimidazole RS in the mobile phase. Extract a quantity ofthe powdered tablets containing 0.1 g of
Reference solution (b). A 0.001 per cent w/v solution of Ornidazole with 40 ml of chloroform, filter and evaporate the
ornidazole RS in the mobile phase. filtrate to dryness. The residue complies with the following
test.
Reference solution (c). Dilute 20.0 ml ofreference solution (a)
and 1.0 ml ofreference solution (b) to 200.0 ml with the mobile A. Determine by infrared absorption spectrophotometry (2.4.6).
phase. Compare the spectrum with that obtained with ornidazole RS
or with the reference spectrum of ornidazole.
Chromatographic system .
- a stainless steel column 25 cm x 4.6 mID, packed with B. Heat a quantity ofthe powdered tablets containing 0.1 g of
octadecylsilane bonded to porous silica (5 /lm), Ornidazole with 10 mg of zinc powder, 1 ml of water and
- mobile phase: a mixture of 70 volumes of O.OlM 0.25 m1 hydrochloric acid for 5 minutes, cool in ice, add 0.5 ml
potassium dihydrogen orthophosphate and 30 of sodium nitrite solution and remove the excess of nitrite
volumes of methanol, with sulphuric acid. Add 0.5 ml of 2-naphthol solution and 2
- flow rate. 1 ml per minute, ml of5 M sodium hydroxide solution. An orange-red colour is
- spectrophotometer set at 318 nm, produced.
- injection volume. 20 Ill. Tests
Inject reference solution (c). The test is not valid unless the
Dissolution (2.52).
resolution between the peaks corresponding to 2-methyl-5-
Apparatus No. 1,
nitroimidazole and ornidazole is at least 1.5.
Medium. 900 ml of 0.1 M hydrochloric acid,
Inject the test solution, reference solutions (a) and (b). In the Speed and time. 75 rpm and 30 minutes.
chromatogram obtained with the test solution, the area of any
Withdraw a suitable volume ofthe medium and filter promptly,
peak corresponding to 2-methyl-5-nitroimidazole is not more
rejecting the first few ml of the filtrate and dilute a suitable
than 0.2 times the area of the peak obtained with reference
volume ofthe filtrate with 0.1 M hydrochloric acid. Measure
solution (a) (0.2 per cent) and the sum of areas of all the
the absorbance of the resulting solution at the maximum at
secondary peaks is not more than the area of the peak in the
about 277 nm (2.4.7). Calculate the content of ornidazole,
chromatogram obtained with reference solution (b)
C7H IOCIN30 3 in the medium from the absorbance obtained from
(1.0 per cent).
a solution of known concentration of ornidazole RS
Heavy metals (2.3.13). 1.0 g complies with the limit test for
D. Not less than 80 per cent of the stated amount of
heavy metals, Method B (20 ppm).
C7H IOCIN30 3•
Sulphated ash (2.3.18). Not more than 0.1 per cent. Related substances. Determine by liquid chromatography
Water (2.3.43). Not more than 1.0 per cent, determined on 0.5 g. (2.4.14).
Assay. Weigh accurately about 0.15 g, dissolve in 50 ml of Test solution. Disperse a quantity of the powdered tablets
glacial acetic acid. Titrate with 0.1 M perchloric acid, containing 25 mg ofornidazole in 25.0 m1 of the mobile phase.
determining the end-point potentiometrically (2.4.25). Carry Reference solution (a). A 0.001 per cent w/v solution of
out a blank titration. 2-methyl-5-nitroimidazole RS in the mobile phase.
1 ml of 0.1 M perchloric acid is equivalent of 0.02196 g of Reference solution (b). A 0.001 per cent w/v solution of
C7H IOCIN30 3. ornidazole RS in the mobile phase.

1824

www.webofpharma.com
IP 2010 ORPHENADRINE CITRATE

Reference solution (c). Dilute 20.0 ml ofreference solution (a) Orphenadrine Citrate contains not less than 98.5 per cent and
and 1.0 ml ofreference solution (b) to 200.0 ml with the mobile not more than 101.0 per cent ofC 1sH 23NO, C6H s0 7, calculated
phase. on the dried basis.
Chromatographic system Category. Skeletal muscle relaxant.
- a stainless steel column 25 cm x 4.6 mm, packed with
Dose. By intramuscular or by slow intravenous injection (over
octadecylsilane bonded to porous silica (5 11m),
5 minutes), 60 mg repeated after 12 hours if necessary.
- mobile phase: a mixture of 70 volumes of 0.01 M
potassium dihydrogen orthophosphate and 30 volumes Description. A white or almost white, crystalline powder;
of methanol, odourless or almost odouriess.
- flow rate. 1 ml per minute,
spectrophotometer set at 318 nm, Identification
- injection volume. 20 Ill. Test A may be omitted if tests B, C, D and E are carried out.
Inject reference solution (c). The test is not valid unless the Tests B, C and D may be omitted if tests A and E are carried
resolution between the peaks corresponding to 2-methyl-5- out.
nitroimidazole and omidazole is at least 2.0. A. Determine by infrared absorption spectrophotometry,(2.4.6).
Inject the test solution, reference solution (a) and (b). In the Compare the spectrum with that obtained with orphenadrine
chromatogram obtained with the test solution, the area ofany citrate RS.
peak corresponding to 2-methyl-5-nitroimidazole is not more B. When examined in the range 230 nm to 360 nm (2.4.7), a
than 0.2 times the area of the peak obtained with reference 0.06 per cent w/v solution in ethanol (95 per cent) shows
solution (a) (0.2 per cent) and the sum of areas of all the
secondary peaks is not more than the area of the peak in the
absorption maxima at 258 nm, 264 mnand 271 nm; absorbances
at the maxima are about 0.68, 0.72 and 0.47 respectively.
chromatogram obtained with the reference solution (b) (1.0
per cent). C. Dissolve 50 mg in 10 ml of ethanol (50 per cent), add 10 ml
of picric acid solution and allow to stand. The precipitate
Other tests. Comply with the tests stated under Tablets. after recrystallisation from ethanol (95 per cent), melts at
Assay. Weigh and powder 20 tablets. Weigh accurately a about 89° or at about 107° (2.4.21).
quantity ofthe powder containing 0.2 g ofOmidazole, transfer D. Dissolve 5 mg in 2 ml of sulphuric acid; an orange-red
to a sintered-glass crucible and extract with 10 ml of hot colour is produced.
acetone. Repeat the extraction 6 times with hot acetone. Cool,
add to the combined extracts 50 ml of acetic anhydride. Titrate E. To 1 g add 10 ml water and 2 ml of 5 M sodium hydroxide,
with 0.1 M perchloric acid, determining the end point shake with two quantities, each of 10 ml, of chloroform and
potentiometrically (2.4.25). Carry out a blank titration. discard the chloroform. Heat the aqueous solution to boiling
with an excess of mercuric sulphate solution, filter ifnecessary
1 ml of 0.1 M perchloric acid is equivalent to 0.02196 g of and boil the resulting solution with 0.2 ml of dilute potassium
C7H IOClN30 3• permanganate solution; the solution is decolorised and a
Storage. Store protected from ligh! and moisture, at a white precipitate is produced.
temperature not exceeding 30°.
Tests
Quaternary ammonium salt. Determine by thin-layer
Orphenadrine Citrate chromatography (2.4.17), coating the plate with silica gel G.
Mobile phase. A mixture of 50 volumes of 2-propanol,
30 volumes of butyl acetate. 15 volumes of water and
5 volumes of strong ammonia solution.
HO eOOH Test solution. Dissolve 0.1 g of the substance under
, Hooe~eooH examination in 10 ml methanol.
Reference solution. A 0.005 per cent wlv solution of
ethyldimethyl [2-(2-methylbenzhydryloxy)ethyl ]ammonium
Mol. Wt. 461.5 chloride RS.
Orphenadrine Citrate is (RS)-dimethyl[2-(2- Apply to the plate 5 III of each solution. After development,
methylbenzhydryloxy)ethyl]amine dihydrogencitrate. dry the plate in air and spray with dilute potossium

1825

www.webofpharma.com
ORPHENADINE CITRATE IP 2010

iodobismuthate solution. Any spot corresponding to Description. A white or almost white, crystalline powder;
ethyldimethyl [2-(2-methylbenzhydryloxy) ethyl] ammonium odourless or almost odourless.
chloride in the chromatogram obtained with the test solution
is not more intense than the spot in the chromatogram obtained Identification
with the reference solution. Tests A andF may be omitted iftestsB, C, D andE are carried
Secondary amine. Determine by thin-layer chromatography out. Tests B, C and D may be omitted iftests A, E and Fare
(2.4.17) coating the plate with silica gel GF254. carried out.
Mobile phase. A mixture of 96 volumes of I-butanol and A. Determine by infrared absorption spectrophotometry,(2.4.6).
4 volumes of strong ammonia solution. Compare the spectrum with that obtained with orphenadrine
hydrochloride RS.
Test solution. Dissolve 0.4 g of the substance under
examination in'l 0 ml methanol. B. When examined in the range 230 nm to 360 nmJ2.4.7), a
0.06 per cent wlv solution in ethanol (95 per cent) shows
Reference solution. A 0.02 per cent wlv solution of methyl [2-
three absorption maxima at about 258 nm,264nm and 271 nm;
(2-methylbenzhydlyloxy) ethyl]amine hydrochloride R8.
absorbances at the maxima, about 1.07, 1.13 and 0.73
Apply to the plate 10 /!l of each solution. After development, respectively.
dry in air and examine in ultra-violet light at 254 nm. Spray the
C. Dissolve about 5 mg in 2 mlof sulphuric acid; an orange-
plate with dilute potassium iodobismuthate solution and
red colour is produced.
examine again. By each method of visualisation any spot
corresponding to methyl [2-(2-methylbenzhydryloxy) D. Dissolve about 50 mg inl0 ml of ethanol (95percent), add
ethyl]amine hydrochloride in the chromatogram obtained with 10 ml of picric acid solution and allow to stand. The
the test solution is not more intense than the spot in the precipitate, after recrystallisation from ethanol (95 per cent)
chromatogram obtained with the reference solution. melts at about 89° or at about 107° (2.4.21).
Heavy metals (2.3.13). 2.0 g complies with the limit test for Tests
heavy metals, Method B (10 ppm). Use 2 ml oflead standard
solution (l0 ppm Pb) to prepare the standard. Quaternary ammonium salt. Determine by thin-layer
chromatography (2.4.17), coating the plate with silica gel G.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Mobile phase. A mixture of 50 volumes of 2-propanol,
Loss on drying (2.3.19). Not more than 0.5 per cent, determined
30 volumes of butyl acetate, 15 volumes of water and
on 1.0 g by drying in an oven at 105° for 3 hours.
5 volumes of strong ammonia solution.
Assay. Weigh accurately about 1.0 g, dissolve in 30 ml of
Test solution. Dissolve 0.1 g of the substance under
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
examination in 10 ml of methanol.
acid, determining the end-point potentiometrically (2.4.25).
Carry out a blank titration. Reference solution. A 0.005 per cent wlv solution of
ethyldimethyl [2-(2-methylbenzhydryloxy) ethyl] ammonium
1 ml of 0.1 M perchloric acid is equivalent to 0.04615 g of
chloride R8.
C 1sH23NO, C6Hs07.
Apply to the plate 5 /!l of each solution. After development,
Storage. Store protected from light.
dry the plate in air and spray with dilute potassium
iodobismuthate solution. Any spot corresponding to
ethyldimethyl [2-(2-methylbenzhydryloxy) ethyl] ammonium
Orphenadrine Hydrochloride chloride in the chromatogram obtained with the test solution
is not more intense than the spot in the chromatogram obtained
C 1sH23NO,HCl Mol. Wt.305.8 with the reference solution.
Orphenadrine Hydrochloride is (RS)-dimethyl[2- Secondary amine. Determine by thin-layer chromatography
(2methylbenzhydryloxy) ethyl]amine hydrochloride. (2.4.17) coating the plate with silica gel GF254.
Orphenadrine Hydrochloride contains not less than 98.5 per Mobile phase. A mixture of 96 volumes of I-butanol and
cent and not more than 101.0 percent of C 1sH 23NO,HCl. 4 volumes of strong ammonia solution.
calculated on the dried basis.
Test solution. Dissolve 0.4 g of the substance under
Category. Antiparkinsonian. examination in 10 ml of methanol.
Dose. 150 mg daily, in divided doses, gradually increased; Reference solution. A 0.02 per cent wlv solution of methyl [2-
maximum 400 mg daily. (2-methylbenzhydryloxy) ethyl]amine hydrochloride R8.

1826

www.webofpharma.com
IP 2010 OSELTAMIVIR PHOSPHATE

Apply to the plate 10 III of each solution. After development, Assay. Weigh and powder 20 tablets. Weigh accurately a
dry in air and examine in ultra-violet light at 254 nm. Spray the quantity ofthe powder containing about 70 mg Orphenadrine
plate with dilute potassium iodobismuthate solution and Hydrochloride and dissolve as completely as possible in a
examine again. By each method of visualisation any spot mixture of 5 ml of water and 5 ml of 2 M hydrochloric acid
corresponding to methyl [2-(2-methylbenzhydryloxy) Without delay extract with four quantities, each 15 ml, of
ethyl]amine hydrochloride in the chromatogram obtained with chloroform, filter the combined extracts and evaporate to about
the test solution is not more intense than the spot in the 20 m1. Add 30 ml of anhydrous glacial acetic and 2 ml of
chromatogram obtained with the reference solution. mercuric acetate solution and titrate with 0.02 M perchloric
Heavy metals (2.3.13). 2.0 g complies with the limit test for acid determining the end point potentiometrically (2.4.25).
heavy metals, Method B (10 ppm). Use 2 ml oflead standard Carry out a blank titration.
solution (10 ppm Pb) to prepare the standard. I ml of 0.02 Mperchloric acid is equivalent to 0.006117 g of
Sulphated ash (2.3.18). Not more than 0.1 percent. C 1SH23NO,HCI.

Loss on drying (2.3.19). Not more than 0.5 per cent, determined Storage. Store protected from light and moisture.
on 1.0 g by drying in an oven at 105° for 3 hours.
Assay. Weigh accurately about 1.0 g, dissolve in 20 ml of
anhydrous glacial acetic acid, add 20 ml of mercuric acetate Oseltamivir Phosphate
solution and titrate with 0.1 M perchloric acid using
l-naphtholbenzein solution as indicator. Carry out a blank
titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.03058 g of
C 1sH23NO, HCI.
Storage. Store protected from light.

Orphenadrine Tablets CI6H2SN204, H3P04 Mol. Wt. 410.4

Orphenadrine Hydrochloride Tablets Oseltamivir Phosphate is phosphoric acid salt of ethyl


(3R,4R,5S)-4-(acetylamino)-5-amino-3-(1-ethylpropoxy)-I-
Orphenadrine Tablets contain not less than 92.5 per cent and cyclohexene-l-carboxylate.
not more than 107.5 per cent of the stated amount of
orphenadrine hydrochloride, C 1SH23NO,HCI. Oseltamivir Phosphate contain not less than 98.0 per cent and
not more than 102.0 per cent ofCI6H2sN204 .H3P0 4,calCulated
Usual strength. 50 mg. on the dried basis.
Identification Category. Antiviral.
Extract a quantity of the powdered tablets containing about Description. A white or off-white powder.
0.15 g ofOrphenadrine Hydrochloride with chloroform, filter
and evaporate the filtrate to dryness. The residue complies Identification
with the following tests. A. Determine by infrared absorption spectrophotometry (2.4.6).
A. Dissolve· 5 mg in 2 ml of sulphuric acid; an orange-red Compare the spectrum with that obtained with oseltamivir
colour is produced. phosphate RS.
B. Dissolve 50 mg in 10 ml of ethanol (50 per cent), add 10 ml B. In the Assay, the principal peak in the chromatogram
of picric acid solution and allow to stand. The precipitate obtained with the test solution corresponds to the peak in the
after recrystallisation from ethanol (95 per cent), melts at chromatogram obtained with the reference solution.
about 89° or at about 107°(2.4.21).
Tests
C. A 5 per cent w/v solution gives reaction A of chlorides
(2.3.1). Specific optical rotation (2.4.22). - 42.0° to - 48.0°, determined
in a 1.0 per cent w/v solution in methanol.
Tests
Related substances. Determine by liquid chromatography
Other tests. Comply with the tests stated under Tablets. (2.4.14).

1827

www.webofpharma.com
OSELTAMIVIR PHOSPHATE IP 2010

NOTE - Prepare the solutions immediately before use. Chromatographic system


a stainless steel column 15 cm x 4.6 mm, packed with
Test solution. Dissolve 25 mg of the substance under
octylsilanebonded to porous silica (5 flm) (Such as
examination in 25 ml ofthe mobile phase.
YMC pack PRO C8),
Reference solution (a). A 0.1 per cent w/v solution of - column temperature. 50°,
oseltamivir phosphate RS in the mobile phase. - mobile phase: a mixture of66 volumes of buffer solution
Reference solution (b). Dilute 1 ml ofreference solution (a) to prepared by dissolving 6.8 g of anhydrous monobasic
100 ml with the mobile phase. potassium phosphate in 1000 ml of water, adjusting the
pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
Chromatographic system of methanol and 23.5 volumes of acetonitrile,
- a stainless steel column 15 cm x 4.6 mm, packed with flow rate. 1 ml per minute,
octylsilane bonded to porous silica (5 flm) (such as YMC spectrophotometer set at 207 nm,
pack PRO C8), - injection volume. 20 fll.
- column te~perature 50°,
- mobile phase: a mixture of66 volumes ofa buffer solution Inject the reference solution. The test is not valid unless the
prepared by dissolving 6.8 g of anhydrous monobasic tailing factor is not more than 2.0, the column efficiency in not
potassium phosphate in 1000 ml of water and adjusting less than 2000 theoretical plates and the relative standard
the pH to 6.0 with dilute sodium hydroxide, 24.5 volumes deviation for replicate injections is not more than 2.0 per cent.
of methanol and 23.5 volumes of acetonitrile, Inject the test solution and the reference solution.
- flow rate. 1 ml per minute,
- spectrophotometer set at 207 nm, Calculate the content ofC16H28N204,H3P04'
injection volume. 20 fll. Storage. Store protected from light and moisture.
Inject reference solution (a). The test is not valid unless the
column efficiency is not less than 2000 theoretical plates and
the tailing factor is not more than 2.0. Oseltamivir Capsules
Inject the test solution and reference solution (b). In the Oseltamivir Phosphate Capsules
chromatogram obtained with the test solution, the area of any Oseltamivir Capsules contain not less than 90.0 per cent and
secondary peak is not more than 0.5 times the area ofthe peak not more than 110.0 per cent of the stated amount of
in the chromatogram obtained with the reference solution (b) oseltamivir, C16H28N204.
(0.5 per cent) and the sum of all the secondary peaks is not
more than the area of the peak in the chromatogram obtained Usual strength. 75 mg.
with the reference solution (b) (1.0 per cent).
Identification
Phosphoric acid. 23.4 to 24.4 per cent, calculated on a dried
basis. In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the peak in the
Weigh accurately about 0.2 g and dissolve in 40 ml ofdistilled chromatogram obtained with the reference solution.
water. Titrate with 0.1 M sodium hydroxide, determining the
end-point potentiometrically (2.4.25). Carry out a blank titration. Tests
1 ml of 0.1 M sodium hydroxide is equivalent to 0.0049 g of Dissolution (2.5.2).
phosphoric acid. Apparatus No.1,
Heavy metals (2.3.13). 1 g complies with the limit test for heavy Medium. 900 ml of 0.1 M hydrochloric acid,
metals, Method A (20 ppm). Speed and time. 50 rpm and 45 minutes
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Withdraw a suitable volume ofthe medium and filter. Determine
on 1 g by drying in an oven at 105°. by liquid chromatography (2.4.14).
Assay. Determine by liquid chromatography (2.4.14). Test solution. Use the filtrate.
Test solution. Dissolve 50 mg of the substance under Reference solution. Dissolve 50 mg of oseltamivirphosphate
examination in 50.0 ml ofthe mobile phase. Dilute 10.0 ml of RS in 20 ml ofthe mobile phase and dilute to 100 ml with the
the solution to 50.0 ml with the mobile phase. dissolution medium. Dilute 5 ml ofthe solution to 25 ml with
Reference solution. A 0.02 per cent w/v solution of oseltamivir the dissolution medium.
phosphate RS in the mobile phase. Use the chromatographic system described under Assay.

1828

www.webofpharma.com
IP 2010 OSELTAMIVIR ORAL SUSPENSION

D. Not less than 80 per cent of the stated amount of mobile phase: a mixture of66 volumes ofa buffer solution
C16H2sN204' prepared by dissolving 6.8 g of anhydrous monobasic
potassium phosphate in 1000 ml of water and adjusting
Related substances. Determine by liquid chromatography
the pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
(2.4.14).
of methanol and 23.5 volumes of acetonitrile,
NOTE - Prepare the solutions immediately before use. - flow rate. 1.2 ml per minute,
Test solution. Weigh accurately a quantity of the mixed spectrophotometer set at 207 nm,
contents of20 capsules containing about 50 mg ofoseltamivir, - injection volume. 20 /-ll.
disperse in 50 ml ofmobile phase and filter. Inject the reference solution. The test is not valid unless the
Reference solution (a). A 0.1 per cent w/v solution of tailing factor is not more than 2.0, the column efficiency is not
oseltamivir phosphate RS in the mobile phase. less than 2000 theoretical plates and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
Reference solution (b). Dilute 1 ml of reference solution to
100 ml with the mobile phase. Inject the test solution and the reference solution.

Chromatographic system Calculate the content ofC16H2sN204.


- a stainless steel column 25 cm x 4.6 rom, packed with Storage. Store protected from moisture, at a temperature not
octylsilane bonded to porous silica (5 /-lm) (such as YMC exceeding 30°.
pack PRO C8),
mobile phase: a mixture of66 volumes ofa buffer solution Labelling. The label states the strength in terms of the
prepared by dissolving 6.8 g of anhydrous monobasic equivalent amount of oseltamivir.
potassium phosphate in 1000 ml of water, adjusting the
pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
of methanol and 23.5 volumes of acetonitrile, Oseltamivir Oral Suspension
- flow rate. 1.5 ml per minute,
spectrophotometer set at 207 nm, Oseltamivir Phosphate Oral Suspension
injection volume. 20 /-ll.
Oseltamivir Oral Suspension is a mixture consisting of
Inject reference solution (a). The test is not valid unless the Oseltamivir phosphate with buffering agents and other
column efficiency is not less than 2000 theoretical plates and excipients. It contains a suitable flavouring agent. It is filled in
the tailing factor is not more than 2.0. a sealed container.
Inject the test solution and reference solution (b). In the The suspension is constituted by dispersing the contents of
chromatogram obtained with the test solution, the area of any the sealed container in the specified volume of water just
secondary peak is not more than the area of the peak in the before issue.
chromatogram obtained with the reference solution (b) (1.0
Oseltamivir Oral Suspension contains not less than 90.0 per
per cent) and the sum of the areas of all the secondary peaks
cent and not more than 110.0 percent ofthe stated amount of
is not more than twice the area ofthe peak in the chromatogram
oseltamivir C16H2sN204.
obtained with the reference solution (b) (2.0 per cent).
The contents of the sealed container comply with the
Other tests. Comply with the tests stated under Capsules.
following requirement.
Water (2.3.43). Not more than 5.0 per cent, determined on 0.5 g.
Usual strength. 15 mg per mI.
Assay. Determine by liquid chromatography (2.4.14)..
Water (2.3.43). Not more than 2.5 per cent, determined on 1.0 g.
NOTE - Prepare the solutions immediately before use.
The constituted suspension complies with the requirements
Test solution. Weigh accurately a quantity of the mixed stated under Oral liquids and with the following
contents of20 capsules containing about 200 mg ofoseltamivir, requirements.
disperse in 100.0 mI ofthe mobile phase and filter. Dilute 5.0 ml
ofthe solution to 50.0 ml with the mobile phase. Identification
Reference solution. A 0.02 per cent w/v solution of oseltamivir A. In the Assay, the principal peak in the chromatogram
phosphate RS in the mobile phase. obtained with the test solution corresponds to the peak in the
Chromatographic system chromatogram obtained with reference solution (b).
- a stainless steel column 15 cm x 4.6 rom, packed with B. To 2 ml of the reconstituted suspension, add 2 ml of dilute
octylsilane bonded to porous silica (5 /-lm), nitric acid and 4ml ofa 10 per cent w/v solution of ammonium

1829

www.webofpharma.com
OSELTAMIVIR ORAL SUSPENSION IP 2010

molybdate and wann the solution. A bright yellow precipitate Chromatographic system
isfonned. a stainless steel column 15 cm x 4.6 mm, packed with
octy1silane bonded to porous silica (5 Ilm),
Tests mobile phase: a mixture of 60 volumes ofa buffer solution
prepared by dissolving 6.8 g of potassium dihydrogen
pH (2.4.24). 3.0 to 5.0.
phosphate in 1000 m1 of water and adjusting the pH to
Related substances. Detennine by liquid chromatography 6.0 with dilute sodium hydroxide, 24.5 volumes of
(2.4.14). methanol and 23.5 volumes of acetonitrile,
NOTE - Prepare the solutions immediately before use. flow rate. 1.2 ml per minute,
spectrophotometer set at 207 urn,
Test solution. Weigh accurately a quantity of the suspension injection volume. 20 Ill.
containing 25 mg ofoseltamivir, dissolve in 25 ml ofthe mobile
Inject the reference solution. The test is not valid unless the
phase and filter.
tailing factor is not more than 2.0, the column efficiency in not
Reference solution (a). A 0.1 per cent w/v solution of less than 2000 theoretical plates and the relative standard
oseltamivir phosphate RS in the mobile phase. deviation for replicate injections is not more than 2.0 per cent.
Reference solution (b). Dilute 1 ml ofreference solution (a) to Inject the test solution and the reference solution.
100 ml with the mobile phase.
Calculate the content of CI6HzsNz04'
Chromatographic system Storage. Store protected from moisture, at a temperature not
- a stainless steel column 25 cm x 4.6 mm, packed with exceeding 30°.
octylsilane bonded to porous silica (5 Ilm),
- column temperature 35°, Labelling. The label states (1) the quantity ofactive ingredient
- mobile phase: 70 volumes of a buffer solution prepared in tenns of the equivalent amount of oseltamivir; (b) the
by dissolving 6.8 g ofpotassium dihydrogen phosphate temperature of storage and the period during which the
in 1000 ml of water and adjusting the pH to 6.0 with constituted suspension may be expected to be satisfactory
dilute sodium hydroxide, 15 volumes of methanol and for use.
15 volumes of acetonitrile,
flow rate. 1.5 ml per minute,
spectrophotometer set at 207 urn, Oxazepam
injection volume. 20 Ill.
Inject reference solution (a). The test is not valid unless the
column efficiency is not less than 2000 theoretical plates and
the tailing factor is not more than 2.0.
Inject the test solution and reference solution (b). In the CI
chromatogram obtained with the test solution, the area of any
secondary peak is not more than the area of the peak in the
chromatogram obtained with the reference solution (b)
(1.0 per cent) and the sum of areas of all the secondary peaks
Mol. Wt. 286.7
is not more than 3 times the area of the peak in the
chromatogram obtained with 'the reference solution (b) Oxazepam is 7-chloro-3-hydroxy-5-phenyl-1,3-dihydro-2H-
(3.0 per cent). 1,4-benzodiazepin-2-one.

Other tests. Complies with the tests stated under Oral liquids. Oxazepam contains not less than 98.5 per cent and not more
than 101.0 per cent ofClsHIICINzOz, calculated on the dried
Assay. Detennine by liquid chromatography{2.4. 14). basis.
NOTE - Prepare the solutions immediately before use. Category. Anxiolytic.
Test solution. Weigh accurately a quantity of the suspension Dose. 15 to 30 mg, 2 to 3 times daily.
containing 60 mg of oseltamivir, dissolve in 250.0 ml of the
Description. A white or alrnostwhite, crystalline powder.
mobile phase and filter. Dilute 10.0 ml ofthe solution to 25.0 ml
with the mobile phase and filter. Identification
Reference solution. A 0.0125 per cent w/v solution of Test A may be omitted iftests B, C and D are carried out. Tests
oseltamivir phosphate RS in the mobile phase. Band D may be omitted if tests A and C are carried out.

1830

www.webofpharma.com
IP 2010 OXAZEPAM TABLETS

A. Detennine by infrared absorption spectrophotometry (2.4.6). Any secondary spot in the chromatogram obtained with test
Compare the spectrum with that obtained with oxazepam RS. solution (a) is not more intense than the spot in the-
B. Prepare the solutions immediately before use, protected chromatogram obtained with reference solution (c) and at most
./i·om light. one such spot is more intense than the spot in the
chromatogram obtained with reference solution (d). The test
Dissolve about 20 mg in sufficient ethanol (95 per cent) to is not valid unless the chromatogram obtained with reference
produce 100 ml. Dilute 10 ml of the solution to 50 ml with solution (b) shows two clearly separated spots.
ethanol (95 per cent) (solution A). Dilute 10 ml of solution A
to 100 ml with ethanol (95 per cent) (solution B). Sulphated ash (2.3.18). Not more than 0.1 per cent.

When examined in the range 230 nm to 360 urn (2.4.7), solution Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
A shows an absorption maxjmum at about 316 nm. When on 1.0 g by drying in an oven at 105°at a pressure not exceeding
examined in the range 220 urn to 250 urn, solution B shows an 0.7kPa.
absorption maximum at about 229 nm; absorbance at about Assay. Weigh accurately about 0.25 g and dissolve in a mixture
229 nm, 1.220 to 1.300. of 10 m1 of anhydrous glacial acetic acid and 90 ml of acetic
C. In the test for Related substances, the principal spot in the anhydride and titrate with 0.1 M perchloric acid detennining
chromatogram obtained with test solution (b) corresponds to the end point potentiometrically (2.4.25). Carry out a blank
that in the chromatogram obtained with reference solution (b). titration.
D. Dissolve about 20 mg in a mixture of5 ml of hydrochloric 1 ml of 0.1 M perchloric acid is eqUivalent to 0.02867 g of
acid and 10 ml of wate/: Heat to boiling for 5 minutes and cool. ClsHIICIN202.
Add 2 ml of a 0.1 per cent w/v solution of sodium nitrite and Storage. Store protected from light and moisture.
allow to stand for 1 minute. Add 1 ml of a 0.5 per cent w/v
solution ofsulphamic acid, mix and allow to stand for 1minute.
Add 1 ml of 0.1 per cent w/v solution of a N- (i-naphthyl)
ethylenediamine dihydrochloride; a red colour is produced. Oxazepam Tablets
Tests Oxazepam Tablets contain not less than 90.0 per cent and not
more than 110.0 per cent of the stated. amount of oxazepam,
Related substances. Carry out the test protectedfrom light. ClsH11ClN202.
Detennine by thin-layer chromatography (2.4.17), coating the Usual strengths. 10 mg; 15 mg; 30 mg.
plate with silica gel GF254.
Identification
Wash the plate with methanol before use.
A. Extract a quantity of the powdered tablets containing
Mobile phase. A mixture of 100 volumes of dichloromethane
20 mg ofOxazepam with 25 ml of chloroform, filter, evaporate
and 10 volumes of methanol.
to dryness and dry the residue at 60° at a pressure not exceeding
Test solution (a). Dissolve 50 mg of the substance under 0.7kPa.
examination in sufficient acetone to produce 10m!.
On the residue, determine by infrared absorption
Test solution (b). Dilute 2 ml oftest solution (a) to 10 ml with spectrophotometry (2.4.6). Compare the spectrum with that
acetone. obtained with oxazepam RS or with the reference spectnnn of
Reference solution (a). Dissolve 10 mg of oxazepam RS in oxazepam.
sufficient acetone to produce 10 ml. B. When examined in the range 210 nm to 360 urn (2.4.7), the
Reference solution (b). Dissolve 10 mg each of oxazepam RS solution obtained in the Assay shows absorption maxima at
and bromazepam RS in sufficient acetone to produce about 230 nm and 316 nm.
lOml.
Tests
Reference solution (c). Dilute 1 ml ofreference solution (a) to
100 ml with acetone. Related substances. CarlY out the test protectedfrom light.
Reference solution (d). Dilute 5 ml ofsolution (d) to 10 m1 with Detennine by thin-layer chromatography (2.4.17) coating the
acetone. plate with silica gel GF 254. .
Apply to the plate 20 !J.I of each solution. Allow the mobile Wash the plate with methanol before use.
phase to rise 17 cm in the same direction as the washing with Mobile phase. A mixture of 100 volumes of dichloromethane
methanol. Dry in air and examine in ultraviolet light at 254 urn. and 10 volumes of methanol.

1831

www.webofpharma.com
OXAZEPAM TABLETS IP 2010

Test solution. Shake a quantity ofpowdered tablets containing Identification


30 mg ofOxazepam with 6 rnl ofacetone and centrifuge.
Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution (a). Dilute 1 volume ofthe test solution to
Compare the spectrum with that obtained with oxcarbazepine
100 volumes with acetone.
RS or with the reference spectrum of oxcarbazepine.
Reference solution (b). Dilute 1 volume ofthe test solution to
500 volumes with acetone. Tests
Reference solution (c). A solution containing 0.1 per cent Related substances. Determine by liquid chromatography
w/v each of oxazepam RS and bromazepam RS. (2.4.14).
Apply to the plate 20 fl1 of each of the solutions. Allow the
Note - Use freshly prepared solutions.
mobile phase to rise 17 em in the same direction as in the
washing with methanol. Dry in air and examine in ultraviolet Test solution. Dissolve 50 mg of the substance under
light at 254 nm. Any secondary spot in the chromatogram examination in 100.0 ml ofthe mobile phase.
obtained with the test solution is not more intense than the Reference solution (a). A 0.05 per cent w/v solution of
spot in the chromatogram obtained with reference solution (a) oxcarbazepine RS in the mobile phase.
(1 per cent) and not more than one such spot is more intense
Reference solution (b). Dilute 1.0 ml ofreference solution (a)
than the spot in the chromatogram obtained with reference
to 100.0 ml with the mobile phase.
solution (b) (0.2 per cent). The test is not valid unless the
chromatogram obtained with reference solution (c) shows two Chromatographic system
clearly separated spots. a stainless steel column 25 em x 4.6 rom, packed with
octadecylsilane bonded to porous silica (5 flm) (such as
Otber tests. Comply with the tests stated under Tablets.
Inertsil C18),
Assay. Weigh and powder 20 tablets. Weigh accurately a - column temperature 50 0 ,
quantity of the powder containing about 25 mg of Oxazepam - mobile phase: a mixture of22 volumes of methanol, 16
and shake with 150 ml ofethanol (95 per cent) for 30 minutes. volumes of acetonitrile and 62 volumes of a buffer
Add sufficient ethanol (95 per cent) to produce 250.0 rnl and solution prepared by dissolving 6.8 g of potassium
centrifuge. Dilute 5.0 ml ofthe supernatant liquid to 100.0 ml dihydrogen orthophosphate in 1000 ml ofwater, adding
with the same solvent and measure the absorbance of the 2 ml of triethylamine and adjusting the pH to 6.0,
resulting solution at the maximum at about 230 nm (2.4.7). flow rate. 1.5 ml per minute,
Calculate the content of ClsHIIClNzOz taking 1250 as the - spectrophotometer set at 215 TIm,
specific absorbance at 230 TIm. injection volume. 10 fll.
Storage. Store protected from light at a temperature not
Inject reference solution (a). The test is not valid unless the
exceeding 30°.
column efficiency is not less than 2000 theoretical plates and
the tailing factor is not more than 2.0.
Oxcarbazepine Inject the test solution and reference solution (b). Run the
chromatograms for about 60 minutes~ The relative retention
times with reference to oxcarbazepine are· 1.7 for
dibenz[b,fJazepine-5-carboxylic acid amide (oxcarbazepine
impurity A), 7.9 for 10-methoxy-5H-dibenz[b,fJazepine
(oxcarbazepine impurity B), and 2.5 for 10-methoxy-5H-
dibenz[b,fJazepine-5-carboxylic acid amide (oxcarbazepine
impurity C ).
Calculate the contents of the impurities in the test solution
ClsHIZNzOz, Mol. Wt. 252.3 using the correction factors, 0.68 for oxcarbazepine impurity
Oxcarbazepine is 10,11-dihydro-1O-oxo-5H-dibenz[b,jJazepine- A, 0.76 for oxcarbazepine impurity Band 0.80 for oxcarbazepine
5-carboxarnide. impurity C. Not more than 1.0 per cent of oxcarbazepine
Oxcarbazepine contains not less than 98.0 per cent and not impurity A and not more than 0.1 per cent each of
more than 102.0 per cent ofClsHlzNzOz, calculated on the dried oxcatbazepine impurity Band oxcarbazepine impurity C and
basis. not more than 0.1 per cent ofany other impurity is found. The
Category. Non-opioid analgesics. sum of all the impurities is not more than 1.5 per cent.
Dose. 300 mg twice a day. Heavy metals (2.3.13). 1.0 g complies with the limit test for
Description. An off-white to yellow crystalline powder. heavy metals, Method B (20 ppm).

1832

www.webofpharma.com
IP 2010 OXCARBAZEPINE TABLETS

Sulphated ash (2.3.18). Not more than 0.1 per cent. D. Not less than 75 per cent of the stated amount of
Loss on drying (2.4.19). Not more than 0.5 per cent, determined ClsH12N202.
on 1.0 g by drying in an oven at 105 0 • Related substances. Determine by liquid chromatography
Assay. Determine by liquid chromatography (2.4.14) as (2.4.14).
described under related substances using the following Note - Useji-eshly prepared solutions.
solution.
Test solution (a). Weigh accurately a quantity ofthe powdered
Test solution. Dissolve 10 mg of the substance under tablets containing 50 mg of Oxcarbazepine add 15 ml methanol,
examination in 100.0 ml ofthe mobile phase.
mix with the aid of ultrasound for 15 minutes and dilute to
Reference solution. A 0.01 per cent w/v solution of 100.0 ml with methanol. Mix and centrifuge.
oxcarbazepine RS in the mobile phase.
Test solution (b). Dilute 5.0 ml of test solution (a) to 25.0 ml
Inject the reference solution. The test is not valid unless the with the mobile phase.
tailing factor is not more than 2.0, the column efficiency is not
less than 2000 theoretical plates and the relative standard Reference solution (a). Dissolve 25.0 mg of oxcarbazepine
deviation for replicate injections is not more than 2.0 per cent. RS in methanol with the aid ofultrasound and dilute to 50.0 ml
with methanol.
Inject the test solution and the reference solution.
Reference solution (b). Dilute 5.0 ml ofreference solution (a)
Calculate the content ofC,sH,2N202'
to 25.0 ml with the mobile phase.
Storage. Store protected from moisture.
Reference solution (c). Dilute 1.0 ml of reference solution (a)
to 100.0 ml with the mobile phase.

Oxcarbazepine Tablets Chromatographic system


a stainless steel column 25 cm x 4.6 mm, packed with
Oxcarbazepine Tablets contain not less than 90.0 per cent and octadecylsilane bonded to porous silica (5 Ilm) (Such
not more than 110.0 per cent of the stated amount of as Hypersil BDS CI8),
oxcarbazepine, ClsH12N202. column temperature 500 ,
Usual strengths. 150 mg; 300 mg; 450 mg; 600 mg. mobile phase: a mixture of 66 volumes of O.05M
potassium dihydrogen phosphate, 14 volumes of
Identification acetonitrile and 20 volumes of methanol, add 0.1 mlof
A. In the Assay, the principal peak in the chromatogram triethylamine and adjust the pH to 6.0 with.
obtained with the test solution (b) corresponds to the peak in orthophosphoric acid,
the chromatogram obtained with reference solution (b). flow rate. 2 ml per minute,
- spectrophotometer set at 215 hm,
B. Extract a quantity ofthe powdered tablets containing about
injection volume. 20 Ill.
50 mg ofoxcarbazepine with 100 ml with methanol and filter.
Diluted 2.0 ml ofthis solution to 100.0 ml with methanol. When Inject reference solution (a). The test is not valid unless the
examined in the range 200 urn to 350 nm (2.4.7), the solution column efficiency is not less than 2000 theoretical plates, the
shows an absorption maximum at about 254 nm. tailing factor is not more than 2.0 and relative standard deviation
for replicate injections is not more than 2.0 per cent.
Tests
Inject test solution (a) and reference solution (c). Run the
Dissolution (2.5.2). chromatograms for three times the retention time of the
Apparatus No. 1 principal peak. In the chromatogram obtained with the test
Medium. 900 ml of 0.75 per cent w/v solution ofsodium lauryl solution, the area of any secondary peak is not more than the
sulphate in water, area ofthe peak in the chromatogram obtained with reference
Speed and time. 75 rpm and 45 minutes. solution (c) (1.0 per cent) and the sum of areas of all the
Withdraw a suitable volume ofthe medium and filter. Measure secondary peaks is not more than twice the area ofthe peak in
the absorbance of the filtered solution, suitably diluted with the chromatogram obtained with reference solution (c) (2.0
the medium if necessary, at the maximum at about 305 urn per cent).
(2.4.7). Calculate the content of ClsH12N202 in the medium Other tests. Comply with the tests stated under Tablets.
from the absorbance obtained from a solution of known
Assay. Determine by liquid chromatography (2.4.14) as
concentration of oxcarbazepine RS prepared by dissolving
described under Related substances.
in minimum volume of methanol and then diluting with the
dissolution medium. Inject test solution (b) and reference solution (b).

1833

www.webofpharma.com
OXPRENOLOL HYDROCHLORIDE IP 2010

Calculate the contentofCIsHI2N202 in the tablets. Test solution (a). Dissolve 0.5 g of the substance under
examination in 10 rnl ofa mixture of90 volumes of chloroform
Storage. Store protected from moisture, at a temperature not
and 10 volumes of methanol.
exceeding 30°.
Test solution (b). Dissolve 0.5 g of the substance under
examination in 100 ml ofa mixture of90 volumes of chloroform
and 10 volumes of methanol.
Oxprenolol Hydrochloride
Reference solution (a). A 0.02 per cent w/v solution of the
OH H substance under examination in a mixture of 90 volumes of

o:
O~NyCH3 chloroform and 10 volumes of methanol.
I ,HCI Reference solution (b). A 0.01 per cent w/v solution of the
~ O~CH2 CH 3 substance under examination in a mixture of 90 volumes of
chloroform and 10 volumes of methanol.
C 1sH23N03,HCl Mol. Wt. 301.8
Reference solution (c). A 0.5 per cent w/v solution of
Oxprenolol Hydrochloride is (RS)-I-(2-allyloxyphenoxy)-3- oxprenolol hydrochloride RS in a mixture of 90 volumes of
isopropylaminopropan-2-01 hydrochloride. chloroform and 10 volumes of methanol.
Oxprenolol Hydrochloride contains not less than 98.5 per cent Apply to the plate 2 f.l.1 of each solution. Allow the mobile
and not more than 101.5 per cent ofClsH23N03,HCl, calculated phase to rise 13 em. Dry the plate in warm air for 10 minutes,
on the dried basis. allow to cool, spray with anisaldehyde solution, heat at 105°
Category. Beta-adrenoceptor antagonist (antihypertensive; for 10 minutes and examine in daylight. Any secondary spot
antianginal; antiarrhythmic). in the chromatogram obtained with test solution (a) is not
more intense than the spot in the chromatogram obtained
Dose. As antihypertensive, initially 80 mg twice daily, increased
with reference solution (a) and not more than one such spot is
as required at weekly intervals; maximum 480 mg daily. As
more intense than the spot in the chromatogram obtained
antianginal, 40 to 160 mg thrice daily. As antiarrhythmic, initially
with reference solution (b).
20 to 40 mg thrice daily, increased as necessary.
Heavy metals (2.3.13). 1.0 g complies with the iimit test for
Description. A white Of almost white, crystalline powder.
heavy metals, Method B (20 ppm).
Identification Sulphated ash (2.3 .18). Not more than 0.1 per cent.
Test A may be omitted if tests Band C are carried out. Test B Loss on drying (2.4.19). Not more than 0.5 per cent, determined
may be omitted if tests A and C are carried out. on 1.0 g by drying in an oven at 60° over phosphoruspentoxide
A. Determine by infrared absorption spectrophotometry (2.4.6). at a pressure of 1.5 to 2.5 kPa for 6 hours.
Compare the spectrum with that obtained with oxprenolol RS Assay. Weigh accurately about 0.25 g, dissolve in 60 ml of
or with the reference spectrum of oxprenolol. anhydrous glacial acetic acid, add 5 ml of mercuric acetate
B. In the test for Related substances, the principal spot in the solution. Titrate with 0.1 Mperchloric acid, determining the
chromatogram obtained with test solution (b) corresponds to end-point potentiometrically (2.4.25).Carry out a blank
that in the chromatogram obtained with reference solution (c). titration.

C. Gives reaction A ofchlorides (2.3.1). 1 ml of 0.1 M perchloric acid is equivalent to 0.03018 g of


C 1SH23N03,HCl.
Tests
Storage. Store protected from light and moisture.
Appearance ofsolution. A 10.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution GYS6 (2.4.1).
Oxprenolol Tablets
pH (2.4.24). 4.5 to 6.0, determined in a freshly prepared
10.0 per cent wIv solution. Oxprenolol Hydrochloride Tablets
Related substances. Determine by thin-layer chromatography Oxprenolol Hydrochloride Tablets contain not less than
(2.4.17), coating the plate with silica gel G 95.0 per cent and not more than 105.0 per cent of the stated
Mobile phase. A mixture of 88 volumes of chloroform, amount of oxprenolol hydrochloride, CISH23N03,HCl. The
12 volumes of methanol and 2 volumes of strong ammonia tablets are coated.
solution. Usual strengths. 40 mg; 80 mg.

1834

www.webofpharma.com
IP 2010 OXYGEN 93 PER CENT

Identification Oxygen
A.To a quantity of the powdered tablets containing 50 mg of Mol. Wt. 32.0
Oxprenolol Hydrochloride add 10 ml of water and 2 ml of
Oxygen contains not less than 99.0 per cent vlv of Oz.
dilute sodium hydroxide solution, extract with 10 ml of
chloroform and reserve the aqueous layer for test C. Dry the This monograph applies to oxygen for medicinal use only.
chloroform extract over anhydrous sodium sulphate, filter and Description. A colourless gas; odourless.
evaporate the filtrate to dryness.
On the residue, determine by infrared absorption Identification
spectrophotometry (2.4.6). Compare the spectrum with that A. A glowing splinter of wood bursts into flame on contact
obtained with oxprenolol hydrochloride RS or with the with the gas.
reference spectrum of oxprenolol hydrochloride.
B. Shake with alkaline pyrogallol solution; the gas under
B. When examined in the range 230 nm to 360 nm (2.4.7), the examination is absorbed and the solution becoming dark
solution obtained in the Assay shows an absorption maximum brown.
only at about 273 nm.
C. When tested as described under Assay, not more than
C. The aqueous layer obtained in test A gives reaction A of 1.0 ml ofgas remains.
chlorides (2.3.1).
Tests
D. The residue obtained in test A melts at about 76° (2.4.21).
Carbon dioxide. Not more than 300 ppm vlv, determined by
Tests using a carbon dioxide detector tube (2.1.1).

Related substances. Determine by thin-layer chromatography Carbon monoxide. Not more than 5 ppm vlv, determined by
(2.4.17), coating the plate with silica gel G using a carbon monoxide detector tube (2.1.1).

Mobile phase. A mixture of 88 volumes of chloroform, Water vapour. Not more than 67 ppm vlv, determined by using
12 volumes of methanol and 2 volumes of strong ammonia a water vapour detector tube (2.1.1).
solution. Assay (2.3.33). Use 100 ml ofthe gas under examination and
Test solution. Extract a quantity of the powdered tablets place spirals of freshly cleaned copper wire and 125 ml of
containing 0.25 g of Oxpreno101 Hydrochloride with 5 ml of ammonia buffer pH 10.9 in the pipette. The volume of the
water, centrifuge and use the supernatant liquid. residual gas in the burette is not more than 1.0 m!.

Reference solution. Dilute 1 volume of the test solution to Storage. Store under pressure in metal cylinders of the type
200 volumes with water. conforming to the appropriate safety regulations. Valves and
taps should not be lubricated with oil or grease.
Apply to the plate 2 III of each solution. Allow the mobile
Labelling. The shoulder of the metal cylinder should be
phase to rise 13 cm. Dry the plate in warm air for 10 minutes,
painted white and the remainder should be painted black. The
allow to cool, spray with anisaldehyde solution, heat at 105°
cylinder should carry a label stating "Oxygen". In addition,
for 10 minutes and examine in daylight. Any secondary spot
"Oxygen" or the symbol "Oz" should be stencilled in paint on
in the chromatogram obtained with the test solution is not
the shoulder of the cylinder.
more intense than the spot in the chromatogram obtained
with the reference solution. .
Other tests. Comply with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Oxygen 93 Per Cent
quantity ofthe powder containing about 20 mg ofOxprenolol Oxygen 93 per Cent contains not less than 90.0 per cent and
Hydrochloride, add 25 ml of 0.1 M hydrochloric acid and not more than 96.0 per cent, vlv ofOz, the remainder consisting
sufficient water to produce 250.0 m!. Mix with the aid of mostly of argon and nitrogen. It is produced from air by the
ultrasound for 5 minutes, shake for 15 minutes and filter. molecular sieve process.
Measure the absorbance of the resulting solution at the
Description. A colourless gas; odourless.
maximum at about 273 nm (2.4.7). Calculate the content of
C,sHz3N03,HCl taking 74.5 as the specific absorbance at Identification
273nm.
A. A glowing splinter of wood bursts into flame on contact
Storage. Store protected from moisture. with the gas.

1835

www.webofpharma.com
OXYGEN 93 PER CENT IP 2010

B. Shake with alkaline pyrogallol solution; the gas under Oxytetracycline has a potency not less than 900 flg of
examination is absorbed and the solution becomes dark brown. CzzHz4Nz09, per mg, calculated on the anhydrous basis.
C. When tested as described under Assay, not more than Category. Antibacterial.
10.0 ml and not less than 4.0 ml ofgas remain.
Dose. Orally, 250 to 500 mg every 6 hours; by intramuscular
Tests injection, 1 to 2 g daily.
Description. A tan yellow or light yellow (with or without a
Carbon dioxide. Not more than 300 ppm v/v, determined by
greenish tinge), crystalline powder; odourless.
using a carbon dioxide detector tube (2.1.1).
Carbon monoxide. Not more than 5 ppm v/v, determined by Identification
using a carbon monoxide detector tube (2.1.1).
A. Determine by thin-layer chromatography (2.4.17), coating
Assay (2.3.33). Use 100 ml of the gas under examination and
the plate with the substance prepared by mixing 25 g of silica
place spirals of freshly cleaned copper wire and 125 ml of
gel Gwith 50 ml ofa mixture of2.5 ml ofglycerin and 47.5 ml of
ammonia buffer pH 10.9 in the pipette. The volume of the
0.1 M disodium edetate previously adjusted to pH 7.0 with
residual gas in the burette is not more than 10.0 ml and not
dilute ammonia solution. After spreading the plate, allow it to
less than 4.0 ml.
stand at room temperature till it is dry (70 to 90 minutes).
Storage. Store in cylinders or in a low pressure collecting
tank. Containers used for Oxygen 93 Per Cent must not be Mobile phase. The lower layer formed after shaking 200 ml of
treated with any toxic, sleep-inducing or narcosis-producing a mixture of 2 volumes of ethyl acetate, 2 volumes of
compounds and must not be treated with any compound that chloroform and 1 volume of acetone with 25 ml of 0.1 M
will be irritating to the respiratory tract when the Oxygen disodium edetate previously adjusted to pH 7.0 with dilute
93 Per Cent is used. ammonia solution.

Labelling. Label each outlet "Oxygen 93 Per Cent", when it is Test solution. Dissolve 0.05 g of the substance under
piped directly from the collecting tanle to the point ofuse. Ifit examination in 100 ml of methanol.
is stored in cylinders, reduce the pressure by means ,of a Reference solution (a). A 0.05 per cent w/v solution of
regulator. Measure the gases with a gas volume meter oxytetracycline RS in methanol.
downstream from the detector tube in order to minimise
contamination or change of the specimens. The shOlilder of Reference solution (b). A solution containing 0.05 per cent
the cylinder should be painted white and the remainder should w/v each of demethylchlortetracycline hydrochloride RS,
be painted black. The cylinder should carry a label stating oxytetracycline hydrochloride RS and tetracycline
"Oxygen 93 Per Cent" and "For medicinal use". hydrochloride RS in methanol.
Apply to the plate 1 III of each solution, freshly prepared.
After development, dry the plate in air, expose to the vapours
Oxytetracycline Dihydrate of ammonia and examine in ultraviolet light at 365 urn. The
principal spot in the chromatogram obtained with the test
Oxytetracycline solution corresponds to that in the chromatogram obtained
with reference solution (a). The test is not valid unless the
OH 0 o chromatogram obtained with reference solution (b) shows
three clearly separated spots.
NH z
,2H zG B. To about 0.5 mg add 2 ml of sulphuric acid; a red colour is
produced. Add the solution to 1 ml of water; the colour
changes to yellow.
C. Dissolve about 2 mg in 5 ml of a 1 per cent w/v solution of
sodium carbonate and add 2 ml of diazotised sulphanilic
C:1zHz<tNZC?9,2HzO Mol. Wt. 496.5
acid solution; an orange-red to brownish-red colour is
Oxytetracycline is (4S,4aR,5S,5aR,6S, 12aS)-4-dimethyl produced.
amino-1 ,4,4a,5,5a,6,11,12a-octahydro-3,5,6,10, 12,12a-
hexahydroxy-6-methyl-1,II-dioxonaphthacene-2- Tests
carboxamide dihydrate, a substance produced by the
growth of certain strains of Streptomyces rimosus or obtained pH (2.4.24). 4.5 to 7.5, determined in a 1.0 per cent w/v
by any other means. It contains a variable quantity of water. suspension in freshly boiled and cooled water.

1836

www.webofpharma.com
IP 2010 OXYTETRACYCLINE DIHYDRATE

Specific optical rotation (204.22). -203° to -216°, detennined spectrophotometer set at 254 run,
at 20° in a 1.0 per cent w/v solution in 0.1 M hydrochloric injection volume. 20 /ll.
acid, after allowing the solution to stand protected from light
Adjust the sensitivity so that the heights of the peaks in the
for 30 minutes before measurement.
chromatogram obtained with reference solution (d) are at least
Light absorption. Absorbance ofa 0.002 per cent w/v solution 50 per cent of the full-scale deflection of the recorder.
in buffersolution pH2.0 at the maximum at about 353 run, 0.58
The test is not valid unless (a) the resolution between the first
to 0.62 (204.7).
peak (4-epioxytetracycline) and the second peak
Light-absorbing impurities. A. Dissolve 20 mg in sufficient (oxytetracycline) is at least 4.0 (b) the resolution between the
of a mixture of I volume of 1 M hydrochloric acid and second peak and the third peak (tetracycline) is at least 5.0
99 volumes of methanol to produce 10 ml. Absorbance ofthe (the content of 2-methyl-2-propanol in the mobile phase may
resulting solution at about 430 nm, when measured within be adjusted if necessary) and (c) the symmetry factor for the
1 hour ofpreparing the solution, not more than 0.25, calculated second peak is at most 1.25.
on the anhydrous basis (204.7).
Inject reference solution (a) six times. The test is not valid
B. Dissolve 0.1 g in sufficient ofa mixture of 1 volume of 1 M unless the relative standard deviation of the area of the peak
hydrochloric acid and 99 volumes of methanol to produce due to oxytetracycline is not greater than 1.0 per cent.
10 ml. Absorbance ofthe resulting solution at about 490 nm,
Inject the test solution and reference solution (e). In the
when measured within 1 hour of preparing the solution, not
chromatogram obtained with the test solution the area of any
more than 0.20, calculated on the anhydrous basis (204.7).
peak corresponding to 4-epioxytetracycline or tetracycline is
Related substances. Detennine by liquid chromatography not greater than the area of the corresponding peak in the
(204.14). chromatogram obtained with reference solution (e). In the
Test solution. Dissolve 20 mg of the substance under chromatogram obtained with the test solution the area of any
examination in 25 ml of O. DIMhydrochloric acid. peak appearing on the tail of the principal peak is not greater
than 4.0 times that ofthe peak due to 4-epioxytetracycline in
Reference solution (a). Dissolve 20 mg of oxytetracycline RS
the chromatogram obtained with reference solution (e).
in 25 ml of O. DIM hydrochloric acid.
Heavy metals (2.3.13).004 g complies with limit the test for
Reference solution (b). Dissolve 20 mg of 4-epioxytetra-
heavy metals, Method B (50 ppm).
cycline RS in 25 ml of 0.01 M hydrochloric acid.
Sulphated ash (2.3.18). Not more than 0.5 per cent.
Reference solution (c). Dissolve 20 mg of tetracycline
hydrochloride RS in 25 ml of 0.01 M hydrochloric acid. Water (2.3043). 4.0 to 9.0 per cent, detennined on 0.5 g.
Reference solution (d). Dilute a mixture of 1.5 ml ofreference Assay. Detennine by the microbiological assay ofantibiotics,
solution (a), 1.0 ml of reference solution (b) and 3.0 ml of Method A or B (2.2.10), and express the results in /lg of
reference solution (c) to 25 ml with 0.01 M hydrochloric oxytetracycline, CzzHz4Nz09' per mg.
acid. Oxytetracycline intended for use in the manufacture of
Reference solution (e). Dilute a mixture of 1.0 ml ofreference parenteral preparations without a fUrther procedure for the
solution (b) and 4.0 ml.ofreference solution (c) to 200 rn1 with removal ofbacterial endotoxins complies with the jrjl!owing
0.01 M hydrochloric acid. additional requirement.
Chromatographic system Bacterial endotoxins (2.2.3). Not more than 004 Endotoxin Unit
- a stainless steel column 25 cm x 4.6 mm, packed with . per mg ofOxytetracycline.
styrene- divinylbenzene co-polymer (8 to 10 /lm), Oxytetracycline intended for use in the manufacture of
- column temperature. 60°, parenteral preparations without a further sterilisation
- 'mobile phase: add 60 g of 2-methyl-2-propanol to a procedure complies with the following additional
volumetric flask with the aid of 200 ml of water, add requirement.
60 ml of 0.33 M phosphate buffer pH 7.5, 50 ml of a
Sterility. Complies with the test for sterility (2.2.11).
1.0 per cent w/v solution of tetrabutylammonium
hydrogen sulphate previously adjusted to pH 7.5 with Storage. Store protected from light and moisture. If it is
2 M sodium hydroxide and 10 ml of a 0.04 per cent w/v intended for use in the manufacture ofparenteral preparations,
solution of disodium edetate previously adjusted to the container should be sterile and sealed so as to exclude
pH 7.5 with 2 M sodium hydroxide and dilute to micro-organisms.
1000.0 ml with water, .Labelling. The label states whether or not the contents are
- flowrate. 1 ml per minute, intended for use in the manufacture ofparenteral preparations.

1837

www.webofpharma.com
OXYTETRACYCLINE INJECTION IP 2010

Oxytetracycline Injection Bacterial endotoxins (2.2.3). Not more than 004 Endotoxin Unit
per mg ofOxytetracycline.
Oxytetracycline Injection is a sterile solution ofoxytetracycline
Other tests. Complies with the tests stated under Parenteral
with or without one or more suitable buffering agents,
Preparations (Injections).
anaesthetics, preservatives, antioxidants, complexing agents
and solvents. Assay. Determine by the microbiological assay ofantibiotics,
Method A or B (2.2.10), and express the result in mg of
Oxytetracycline Injection contains not less than 90.0 per cent
and not more than 120.0 per cent of the stated amount of oxytetracycline, CzzHz4Nz09' per ml.
anhydrous oxytetracycline, CzzHz4Nz09' Storage. Store protected from light.
Usual strengths. 50 mg per ml; 125 mg per ml. Labelling. The label states (1) the strength in mg ofanhydrous
oxytetra,cycline per ml; (2) that the contents are to be used for
Description. A clear, yellow to tan yellow solution. It may
intramuscular use only; (3) the names of any preservatives
have a greenish tinge.
used.
Identification
A. Determine by thin-layer chromatography (204.17), coating Oxytetracycline Hydrochloride
the plate with the substance prepared by mixing 25 g of silica
gel G with 50 ml ofa mixture of2.5 ml ofglycerin and 47.5 ml of Mol. Wt. 496.9
0.1 M disodium edetate previously adjusted to pH 7.0 with Oxytetracycline Hydrochloride is (4S,4aR,5S,5aR,6S, l2a8)-
dilute ammonia solution. After spreading the plate, allow it to 4-dimethylaIDino-l,4,4a,5,5a,6,11,12a-octahydro-3,5,
stand at room temperature till it is dry (70 to 90 minutes). 6,10,12, 12a-hexahydroxy-6-methyl-l ,11-dioxonaphthacene-2-
Mobile phase. The lower layer fonned after shaking 200 ml of carboxamide hydrochloride, a substance produced by
a mixture of 2 volumes of ethyl acetate, 2 volumes of the growth of certain strains of Streptomyces rimosus or
chloroform and 1 volume of acetone with 25 ml of 0.1 M obtainedby any other means.
disodium edetate previously adjusted to pH 7.0 with dilute Category. Antibacterial.
ammonia solution.
Dose. Orally, 250 to 500 mg every 6 hours; by intravenous
Test solution. Shake a quantity containing 10 mg ofanhydrous infusion, in a concentration of 0.1 per cent w/v, 1 to 2 g daily.
oxytetracycline with 20 ml of methanol, centrifuge if necessary
and use the clear supernatant liquid. Description. A pale yellow, crystalline powder; odour1ess;
hygroscopic.
Reference solution (a). A 0.05 per cent w/v solution of
oxytetracycline hydrochloride RS in methanol. Oxytetracycline Hydrochloride has a potency not less than
835 flg of CzzHz4Nz09, per mg, calculated on the anhydrous
Reference solution (b). A solution containing 0.05 per cent basis.
w/v each of demethylchlortetracycline hydrochloride RS,
oxytetracycline hydrochloride RS and tetracycline Identification
hydrochloride RS in methanol.
A. Determine by thin-layer chromatography (204.17), coating
Apply to the plate 1 fll of each solution, freshly prepared. the plate with the substance prepared by mixing 25 g of silica
After development, dry the plate in air, expose to the vapours gel G with 50 ml ofa mixture of2.5 mlofglycerin and 47.5 ml of
of ammonia and examine in ultraviolet light at 365 nm. The 0.1 M disodium edetate previously adjusted to pH 7.0 with
principal spot in the chromatogram obtained with the test dilute ammonia solution. After spreading the plate, allow it to
solution corresponds to that in the chromatogram obtained stand at room temperature till it is dry (70 to 90 minutes).
with reference solution (a). The test is not valid unless the Mobile phase. The lower layer formed after shaking 200 m1 of
chromatogram obtained with reference solution (b) shows a mixture of 2 volumes of ethyl acetate, 2 volumes of
three clearly separated spots.
chloroform and 1 volume of acetone with 25 m1 of 0.1 M
B. Add 0.1 ml to 2 ml ofsulphuric acid; a red colour is produced. disodium edetate previously adjusted to pH 7.0 with dilute
Add the solution to 1 ml of water; the colour changes to ammonia solution.
yellow. Test solution. Dissolve 0.05 g of the substance under
examination in 100 ml of methanol.
Tests
Reference solution (a). A 0.05 per cent w/v solution of
pH (204.24). 8.0 to 9.0. oxytetracycline hydrochloride RS in methanol.

1838

www.webofpharma.com
IP 2010 OXYTETRACYCLINE HYDROCHLORlDE

Reference solution (b). A solution containing 0.05 per cent Reference solution (c). Dissolve 20 mg of tetracycline
w/v each of demethylchlortetracycline hydrochloride RS, hydrochloride RS in 25 ml of 0.01 M hydrochloric acid.
oxytetracycline hydrochloride RS and tetracycline
Reference solution (d). Dissolve 20 mg of a..-apo-
hydrochloride RS in methanol.
oxytetracycline RS in 20 ml of 0.01 M sodium hydroxide and
Apply to the plate 1 J!l of each solution, freshly prepared. dilute to 250 ml with 0.01 M hydrochloric acid.
After development, dry the plate in air, expose to the vapours
Reference solution (e). Dissolve 20 mg of ~ -apo-
of ammonia and examine in ultraviolet light at 365 nm. The
oxytetracycline RS in 20 ml of 0.01 M sodium hydroxide and
principal spot in the chromatogram obtained with the test
dilute to 250 ml with 0.01 M hydrochloric acid.
solution corresponds to that in the chromatogram obtained
with reference solution (a). The test is not valid unless the Reference solution (f), Dilute a mixture of 1.5 ml ofreference
chromatogram obtained with reference solution (b) shows solution (a), 1.0 ml of reference solution (b), 3.0 ml each of
three clearly separated spots. reference solutions (c) (d) and (e) to 25 ml with 0.01 M
hydrochloric acid.
B. To about 0.5 mg add 2 ml of sulphuric acid; a red colour is
produced. Add the solution to 1 ml of water; the colour Reference solution (g). Dilute a mixture of 1.0 ml ofreference
changes to yellow. solution (b), 4.0 ml of reference solution (c) and 40.0 ml of
reference solution (e) to 200 ml with 0.01 M hydrochloric
C. Dissolve about 2 mg in 5 ml of a 1 per cent w/v solution of
acid.
sodium carbonate and add 2 ml of diazotised sulphanilic
acid solution; an orange-red to brownish-red colour is Chromatographic system
produced. - a stainless steel column 25 cm x 4.6 mm, packed with
styrene-divinylbenzene co-polymer (8 to 10 J!m),
D. A 5 per cent w/v solution gives the reactions of chlorides
- column temperature. 60°,
(2.3.1).
- mobile phase: transfer separately 30 g (for mobile phase
Tests A) orl 00 g (for mobile phase B) of 2-methyl-2-propanol
to volumetric flasks with the aid of200 ml of water; to
pH (2.4.24).2.0 to 3.0, determined in a 1.0 per cent w/v solution. each flask add 60 ml of 0.33 M phosphate buffei'pH 7.5,
Specific optical rotation (2.4.22). -188° to -200°, determined 50 ml of a 1.0 per cent w/v solution of tetrabutyl-
at 20° in a 1 per cent w/v solution in 0.1 M hydrochloric acid. ammonium hydrogen sulphate previously adjusted to
pH 7.5 with 2 M sodium hydroxide and 10 ml of a 0.04
Light absorption (2.4.7). Absorbance ofa 0.002 per cent w/v
per cent w/v solution of disodium edetate previously
solution in chloride buffer solution pH 2.0 at the maximum at
adjusted to pH 7.5 with 2 M sodium hydroxide and
about 353 nm, 0.54 to 0.58.
dilute each solution to 1000.0 ml with water.,
Light-absorbing impurities. A. Dissolve 20 mg in sufficient flow rate. 1 ml per minute,
of a mixture of 1 volume of 1 M hydrochloric acid and - spectrophotometer set at 254 nm,
99 volumes of methanol to produce 10 ml. Absorbance ofthe - injection volume. 20 J!l.
resulting solution at about 430 urn, when measured within
Carry out a one-step gradient elution in the following manner.
1 hour ofpreparing the solution, not more than 0.50, calculated
Pump a mixture containing 30 volumes ofmobile phase Band
on the anhydrous basis (2.4.7).
70 volumes of mobile phase A for 15 minutes, then pump a
B. Dissolve 0.1 g in sufficient ofa mixture of 1 volume of 1 M mixture containing 30 volumes of mobile phase A and
hydrochloric acid and 99 volumes of methanol to produce 70 volumes of mobile phase B for 15 minutes and finally
10 ml. Absorbance of the resulting solution at about 490 nm, equilibrate with the first mixture. Adjust the sensitivity so that
when measured within 1 hour of preparing the solution, not the heights of the peaks in the chromatogram obtained with
more than 0.20, calculated on the anhydrous basis (2.4.7). reference solution (f) are at least 50 per cent of full-scale
deflection ofthe recorder.
Related substances. Determine by liquid chromatography
(2.4.14). The test is not valid unless, in the chromi;ltogram obtained
with reference solution (f), (a) the resolution between the first
Test solution. Dissolve 20 mg of the substance under
peak (4-epioxytetracycline) and the second peak
examination in 25 ml of O. 01 M hydrochloric acid.
(oxytetracycline) is at least 4.0, (b) the resolution between the
Reference solution (a). Dissolve 20 mg of oxytetracycline RS second peak and the third peak (tetracycline) is at least 5.0, (c)
in 25 ml of O. 01 M hydrochloric acid. the resolution between the fourth peak (a..-apo-
Reference solution (b). Dissolve 20 mg of 4-epioxytetra- oxytetracycline) and the fifth peak (~-apo-oxytetracycline)
cycline RS in 25 ml of 0.01 M hydrochloric acid. is at least 3.5, and (d) the symmetry factor ofthe second peak

1839

www.webofpharma.com
OXYTETRACYCLINE HYDROCHLORIDE IP 2010

is at most 1.25. If necessary adjust the proportions of the Oxytetracycline Capsules


mobile phases used to produce the one-step gradient elution.
Adjust the time-programme for the one-step gradient elution Oxytetracycline Hydrochloride Capsules
if necessary.
Oxytetracycline Capsules contain not less than 95.0 per cent
Inject reference solution (a) six times. The test is not valid if and not more than 110.0 per cent of the stated amount of
the relative standard deviation of the area of the peak due to oxytetracycline hydrochloride, CzzHz4Nz09,HCl.
oxytetracycline is greater than 1.0 per cent. Ifnecessary, adjust
Usual strengths. 250 mg; 500 mg.
the integrator parameters.
Inject the test solution and reference solution (g). In the Identification
chromatogram obtained with the test solution the area of any
peak corresponding to 4-epioxytetracycline or tetracycline A. Determine by thin-layer chromatography (204.17), coating
is not greater than the area of the corresponding peak in the the plate with the substance prepared by mixing 25 g of
chromatogram obtained with reference solution (g) and the silica gel G with 50 ml of a mixture of 2.5 ml of glycerin and
total area ofthe peaks corresponding to a-apo-oxytetracycline 47.5 ml of 0.1 M disodium edetate previously adjusted to pH
and to ~ -apo-oxytetracycline and any peak between the latter 7.0 with dilute ammonia solution. After spreading the plate,
two is not greater than the area of the peak due to ~ -apo- allow it to stand at room temperature till it is dry (70 to 90
oxytetracycline in the chromatogram obtained with reference minutes).
solution (g). In the chromatogram obtained with the test
Mobile phase. The lower layer formed after shaking 200 ml of
solution the area of any peak appearing on the tail of the
a mixture of 2 volumes of ethyl acetate, 2 volumes of
principal peak is not greater than 4.0 times that ofthe peak due
chloroform and 1 volume of acetone with 25 ml of 0.1 M
to 4-epioxytetracycline in the chromatogram obtained with
disodium edetate previously adjusted to pH 7.0 with dilute
reference solution (g).
ammonia solution.
Heavy metals (2.3.13).004 g complies with the limit test for
Test solution. Extract a quantity ofthe contents ofthe capsules
heavy metals, Method B (50 ppm).
containing 10 mg ofOxytetracycline Hydrochloride with 20 ml
Sulphated ash (2.3.18). Not more than 0.5 per cent. methanol, centrifuge and use the supernatant liquid.
Water (2.3043). Not more than 2.0 per cent w/w, determined on Reference solution (a). A 0.05 per cent w/v solution of
0.5 g. oxytetracycline hydrochloride RS in methanol.
Assay. Determine by the microbiological assay ofantibiotics, Reference solution (b). A solution containing 0.05 per cent
Method A or B (2.2.10), and express the result in Ilg of w/v each of demethylchlortetracycline hydrochloride RS,
oxytetracycline, CzzHz4Nz09, per mg. oxytetracycline hydrochloride RS and tetracycline
hydrochloride RS in methanol.
Oxytetracycline Hydrochloride intended for use in the
manufacture ofparenteral preparations without a fitrther Apply to the plate 1 III of each solution, freshly prepared.
procedure for the removal ofbacterial endotoxins complies After development, dry the plate in air, expose to the vapours
with the following additional requirement. of ammonia and examine in ultraviolet light at 365 nm. The
principal spot in the chromatogram obtained with the test
Bacterial endotoxins (2.2.3). Not more than 004 Endotoxin Unit
solution corresponds to that in the chromatogram obtained
per mg.
with reference solution (a). The test is not valid unless the
Oxytetracycline Hydrochloride intended for use in the chromatogram obtained with reference solution (b) shows
.manufacture ofparenteral preparations without a jitrther three clearly separated spots.
sterilisation procedure complies with the following
B. To 0.5 mg of the contents of the capsules add 2 ml of
additional requirement.
sulphuric acid; a red colour is produced. Add the solution to
Sterility. Complies with test for sterility (2.2.11). 1 ml of water; the colour changes to yellow.
Storage. Store protected from light and moisture. If it is C. Dissolve about 2 mg ofthe contents ofthe capsules in 5 ml
intended for use in the manufacture ofparenteral preparations, ofa 1 per cent w/v solution of sodium carbonate and add 2 ml
the container should be sterile and sealed so as to exclude of diazotised sulphanilic acid solution; a light brown colour
micro-organisms. is produced.
Labelling. The label states whether or not the contents are D. The contents ofthe capsules give the reactions ofchlorides
intended for use in the manufacture ofparenteral preparations. (2.3.1).

1840

www.webofpharma.com
IP 2010 OXYTETRACYLINE EYE OINTMENT

Tests Identification
Light-absorbing impurities. Dissolve a portion ofthe mixed Determine by thin-layer chromatography (2.4.17), coating the
contents of five capsules as completely as possible in plate with the substance prepared by mixing 25 g of silica gel
sufficient of a mixture of 1 volume of 1 M hydrochloric acid G with 50 ml of a mixttLre of2.5 ml ofglycerin and 47.5 ml of
and 99 volumes of methanol to produce two solutions of 0.1 M disodium edetate previously adjusted to pH 7.0 with
Oxytetracycline Hydrochloride containing (1) 0.2 per cent w/v dilute ammonia solution. After spreading the plate, allow it to
and (2) 1.0 per cent w/v and filter each solution. Absorbance stand at room temperature till it is dry (70 to 90 minutes).
ofthe filtrate obtained from solution (1) at about 430 om, when Mobile phase. The lower layer formed after shaldng 200 ml of
measured within 1 hour of preparing the solution, not greater a mixture of 2 volumes of ethyl acetate, 2 volumes of
than 0.75 and ofthe filtrate obtained from solution (2) at about chloroform and 1 volume of acetone with 25 ml of 0.1 M
490 om, not more than 0.40 (2.4.7). disodium edetate previously adjusted to pH 7.0 with dilute
ammonia solution.
Dissolution (2.5.2).
Test solution. A solution prepared by heating a quantity
Apparatus No.1,
containing 20 mg ofOxytetracycline Hydrochloride with 20 ml
Medium. 900 ml of 0.1 M hydrochloric acid,
of methanol for 20 minutes, cooling in ice, filtering, carefully
Speed and time. 100 rpm and 45 minutes. evaporating the filtrate to dryness and dissolving the residue
Withdraw a suitable volume ofthe medium and filter through in 20 ml of methanol.
a membrane filter disc with an average pore diameter not greater Reference solution (a). A 0.05 per cent w/v solution of
than 1.0 j.Lm. Measure the absorbance of the filtrate, suitably oxytetracycline hydrochloride RS in methanol.
diluted ifnecessary, at the maximum at about 353 nm (2.4.7).
Reference solution (b). A solution containing 0.05 per cent
Calculate the content ofCzzH24N209,HCI in the medium taldng
w/v each of demethylchlortetracycline hydrochloride RS,
282 as the specific absorbance at 353 om.
oxytetracycline hydrochloride RS and tetracycline
D. Not less than 75 per cent of the stated amount of hydrochloride RS in methanol.
CnH2~209,HCl.
Apply to the plate 1 j.LI of each solution, freshly prepared.
Loss on drying (2.4.19). Not more than 5.0 percent, determined After development, dry the plate in air, expose to the vapours
on 1.0 g ofthe mixed contents ofthe capsules by drying in an of ammonia and examine in ultraviolet light at 365 nm. The
oven at 60° at a pressure not exceeding 0.7 kPa for 3 hours. principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Other tests. Comply with the tests stated under Capsules.
with reference solution (a). The test is not valid unless the
Assay. Weigh accurately a quantity of the mixed contents of chromatogram obtained with reference solution (b) shows
20 capsules containing about 0.25 g of Oxytetracycline three clearly separated spots.
Hydrochloride, add 250.0 ml of water, shake, filter.
Tests
Determine by the microbiological assay ofantibiotics, Method
A or B (2.2.10), and express the results in mg ofoxytetracycline Water (2.3.43). Not more than 1.0 percent, determined on 0.5 g.
hydrochloride per capsule taking each mg of oxytetracycline Other tests. Complies with the tests stated under Eye
to be equivalent to 1.079 mg ofoxytetracycline hydrochloride, Ointments.
CnH2~209,HCl. Assay. Weigh accurately about 1.0 g and transfer to a
Storage. Store protected from light and moisture. separating funnel. Add 25 ml of peroxide-free ether, shake
well and extract with five quantities, each of20 ml, of 0.1 M
hydrochloric acid. Combine the extracts and dilute to
200.0 ml with 0.1 Mhydrochloric acid. Dilute a suitable volume
of the resulting solution with buffer solution No 3 (2.2.10), to
Oxytetracycline Eye Ointment produce a solution containing 1 j.Lg of oxytetracycline per mI.

Oxytetracycline Hydrochloride Eye Ointment Determine by the microbiological assay ofantibiotics, Method
B (2.2.10), and express the results as a percentage of
Oxytetracycline Eye Ointment contains not less than 90.0 per oxytetracycline hydrochloride taking each mg of
cent and not more than 115.0 per cent of the stated amount of oxytetracycline to be equivalent to 1.079 mg ofoxytetracycline
oxytetracycline hydrochloride, CZ2H24N209,HCI. hydrochloride, C22H24Nz09,HCl.
Usual strength. 1 per cent w/w. Storage. Store protected from light and moisture.

1841

www.webofpharma.com
OXYTETRACYCLINE HYDROCHLORIDE INJECTION IP 2010

Oxytetracycline Hydrochloride principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Injection with reference solution (a). The test is not valid unless the
Oxytetracycline Hydrochloride Injection is a sterile material chromatogram obtained with reference solution (b) shows
consisting of Oxytetracycline Hydrochloride with or without three clearly separated spots.
buffering agents and other excipients. It is filled in a sealed B. To about 0.5 mg add 2 ml of sulphuric acid; a red colour is
container. produced. Add the solution to I ml of water; the colour
The injection is constituted by dissolving the contents ofthe changes to yellow.
sealed container in the requisite amount of sterile Water for C. Dissolve about 2 mg in 5 ml of a I per cent w/v solution of
Injections, immediately before use. sodium carbonate and add 2 ml of diazotised sulphanilic
The constituted solution complies with the requirements for acid solution; an orange-red to brownish-red colour is
Clarity of solution and Particulate matter stated under produced.
Parenteral Preparations (Injections).
Tests
Storage. The constituted solution may be used within three
days of preparation when stored in a refrigerator (20 to 8°). Appearance of solution. A 10.0 per centw/v solution is clear
(2.4.1) and yellow.
Oxytetracycline Hydrochloride Injection contains not less than
pH (2.4.24). 2.0 to 3.0, determined in a 1.0 per cent w/v solution.
90.0 per cent and not more than 115.0 per cent of the stated
amount ofoxytetracycline, CzzHz4NzOg. Light-absorbing impurities. A. Dissolve 20 mg in sufficient
of a mixture of I volume of 1 M hydrochloric acid and
The contents of the sealed container comply with the
99 volumes of methanol to produce 10 ml. Absorbance ofthe
requirements stated under Parenteral Preparations
resulting solution at about 430 nm, when measured within
(Powdersfor Injection) and with thefollowing requirements.
I hour ofpreparing the solution, not more than 0.50, calculated
Usual strengths. The equivalent of 250 mg and 500 mg of on the anhydrous basis (2.4.7).
oxytetracycline.
B. Dissolve 0.1 g in sufficient ofa mixture of I volume of 1 M
Description. A pale yellow, crystalline powder. hydrochloric acid and 99 volumes of methanol to produce
10 ml. Absorbance of the resulting solution at about 490 nm,
Identification when measured within 1 hour of preparing the solution, not
A. Determine by thin-layer chromatography (2.4.17), coating more than 0.20, calculated on the anhydrous basis (2.4.7).
the plate with the substance prepared by mixing 25 g of silica Assay. Determine by the microbiological assay ofantibiotics,
gel G with 50 ml ofa mixture of2.5 ml ofglycerin and 47.5 m1 of method A or B (2.2.10), and express the result in )..I.g of
0.1 M disodium edetate previously adjusted to pH 7.0 with oxytetracycline, CzzHz4NzOg, per mg.
dilute ammonia solution. After spreading the plate, allow it to
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit
stand at room temperature till it is dry (70 to 90 minutes).
per mg.
Mobile phase. The lower layer formed after shaking 200 ml of
Storage. Store protected from light and moisture.
a mixture of 2 volumes of ethyl acetate, 2 volumes of
chloroform and I volume of acetone with 25 ml of 0.1 M Labelling. The label states (I) the quantity ofOxytetracycline
disodium edetate previously adjusted to pH 7.0 with dilute Hydrochloride contained in it in terms of the equivalent
ammonia solution. amount of oxytetracycline; (2) that the contents are to be
used for intravenous injection only; (3) the names of the
Test solution. Dissolve 0.05 g of the substance under buffering agents used.
examination in 100 m1 of methanol.
Reference solution (a). A 0.05 per cent w/v solution of
oxytetracycline hydrochloride RS in methanol.
Oxytocin
Reference solution (b). A solution containing 0.05 per cent
w/v each of demethylchlortetracycline hydrochloride RS, Cys-Tyr-lle-Gln-Asn-Cys-Pro-Leu-GlyNH z
oxytetracycline hydrochloride RS and tetracycline
hydrochloride RS in methanol.
I I
Apply to the plate I )..1.1 of each solution, freshly prepared. Mol. Wt.1007.2
After development, dry the plate in air, expose to the vapours Oxytocin is a cyclic nonapeptide hormone obtained by a
of ammonia and examine in ultraviolet light at 365 nm. The process offractionation from the posterior lobe ofthe pituitary

1842

www.webofpharma.com
IP 2010 OXYTOCIN

gland of healthy oxen or other mammals or by synthesis that containing the dried samples in a vessel that contains an
has the property of stimulating contraction of the uterus and appropriate amount of hydrolysis solution. The hydrolysis
mille ejection in receptive animals. It may be presented as a solution does not come in contact with the test sample. Apply
solid or as a solution in a solvent containing an appropriate an inert atmosphere or vacuum (less than 200 fl.m ofmercury
antimicrobial preservative such as 0.2 per cent w/v solution of or 26.7 Pa) to the headspace of the vessel, and heat to about
chlorbutol. 110° for a 24 hours hydrolysis time. Acid vapour hydrolysis
the dried sample. Any condensation ofthe acid in the sample
Ifit is derived from animal species, Oxytocin contains not less
vials is to be minimised. After hydrolysis, dry the test sample
than 95.0 percent and not more than 105.0 per cent of the
in vacuum to remove any residual acid.
stated number of Units of oxytocic activity. If it is a synthetic
product presented as a solid, it contains not less than 560 For analysis-Past-column ninhydrin derivatisation. Ion-
Units per mg, calculated with reference to the peptide content exchange chromatography with post-column ninhydrin
and when presented as a liquid, it contains not less than 150 derivatisation is one ofthe most common methods employed
Units per ml. for quantitative amino acid analysis. As a rule, a lithium-based
Category. Oxytocic. cation-exchange system is employed for the analysis of the
more complex physiological samples, and the faster sodium-
Description. When presented as a solid, a white or almost based cation-exchange system is used for the more simplistic
white powder. When presented as a liquid, a clear colourless amino acid mixtures obtained with protein hydrolysates
liquid. (typically containing 17 amino acid components). Separation
ofthe amino acids on an ion-exchange column is accomplished
Identification
through a combination of changes in pH and cation strength.
Test B may be omitted if tests A and C are carried out. Test C A temperature gradient is often employed to enhance
may be omitted if tests A and B are carried out. separation.

A. In the Assay, the principal peak in the chromatogram When the amino acid reacts with ninhydrin, the reactant has a
obtained with the test solution corresponds to the peak in the characteristic purple or yellow colour. Amino acids, except
chromatogram obtained with the reference solution. imino acid, give a purple colour, and show an absorption
maximum at 570 nm. The imino acids such as proline give a
B. Amino acid analysis.
yellow colour, and show an absorption maximum at 440 urn.
For hydrolysis-Acid hydrolysis using hydrochloric acid The post-column reaction between ninhydrin and amino acids
containing phenol is the most common procedure used for eluted from the column is monitored at 440 nm and 570 urn,
protein/peptide hydrolysis preceding amino acid analysis. The and the chromatogram obtained is used for the determination
addition of phenol to the reaction prevents the halogenation of amino acid composition.
of tyrosine.
The detection limit is considered to be 10 pmol for most ofthe
Hydrolysis solution. 6 M hydrochloric acid containing 0.1 amino acid derivatives, but 50 pmol for the proline derivative.
per cent to 1.0 per cent of phenol. Response linearity is obtained in the range of 20-500 pmol
with con-elation coefficients exceeding 0.999. To obtain good
Procedure
composition data, samples larger than 1 fl.g before hydrolysis
Liquidphase hydrolysis. Place the protein or peptide sample are best suited for this amino acid analysis ofprotein/peptide.
in a hydrolysis tube, and dry (the sample is dried so that water
Express the content ofeach amino acid in moles. Calculate the
in the sample will not dilute the acid used for the hydrolysis).
relative proportions of the amino acids, taking 1/6 of the sum
Add 200 fl.l of hydrolysis solution per 500fl.g oflyophilised
ofthe number ofmoles ofaspartic acid, glutamic acid, proline,
protein. Freeze the sample tube in a dry ice-acetone bath, and
glycine, isoleucine and leucine as equal to 1.
flame seal in vacuum. Samples are typically hydrolysed at
110° for 24 hours in vacuum or in an inert atmosphere to The values fall within the following limits: aspartic acid: 0.90
prevent oxidation. Longer hydrolysis times (e.g. 48 hours and to 1.10; glutamic acid: 0.90 to 1.10; proline: 0.90 to 1.10; glycine:
72 hours) are investigated ifthere is a concern that the protein 0.90 to I.I 0; leucine: 0.90 to I.I 0; isoleucine: 0.90 to I.I 0;
is not completely hydrolysed. tyrosine: 0.7 to 1.05; half-cystine: 1.4 to 2.1. Not more than
traces of other amino acids are present.
Vapour phase hydrolysis. This is one of the most common
acid hydrolysis procedures, and it is preferred for microanalysis C. For biological response, dissect out the uterus from a virgin
when only small amounts of the sample are available. female rat weighing between 120 to 200 g and in diestrus stage.
Contamination of the sample from, the acid reagent is also Immediately before injection confirm the uterine stage ofthe
minimised by using vapour phase hydrolysis. Place vials rat by vaginal smear. Suspend one hom of the uterus in a

1843

www.webofpharma.com
OXYTOXIN IP 2010

organ bath containing 9.0 g of sodium chloride, 0.42 g of mobile phase: appropriate proportions ofa 1.56 per cent
potassium chloride, 0.16 g of calcium chloride, 0.50 g of w/v solution of sodium dihydrogen phosphate (mobile
sodium bicarbonate, 0.25 g of dextrose, and 0.0053 g of phase A) and a mixture ofequal volumes of acetonitrile
magnesium chloride per litre of the solution. Maintain the and water (mobile phase B),
bath temperature at 32° or any other suitable temperature at flow rate. 1 ml per minute,
which spontaneous contractions of the uterus are abolished spectrophotometer set at 220 urn,
and the preparations maintain its sensitivity. Oxygenate the injection volume: 20 /ll
bath solution with a mixture of 95 per cent oxygen and 5 per Equilibrate the column with a mixture of 70 volumes ofmobile
cent carbon dioxide. Record the contractions of the uterine phase A and 30 volumes of mobile phase B and record the
muscle on a recorder, using a suitable instrument producing chromatograms as follows. Operate by gradient elution
linear response (for example an isotonic liver with a load of increasing continuously and linearly the proportion ofmobile
not exceeding 2 g or isotonic and linear transducer). Add to phase B by 1.0 per cent v/v per minute for 30 minutes. Finally
the bath two appropriate dilutions of the oxytocin reference elute using the same mixture for 15 minutes to re-equilibrate
solution, and record the contraction of the muscle following the column.
each dilution. The appropriate dilutions (doses) are those
dilutions of the reference solution which produce clearly Calculate the content ofthe peptide, C43H66NIZ0IZSZ.
distinctive submaximal contractions. The required dilution Assay. Determine by liquid chromatography (2.4.14).
normally lies between 0.1 to 5 micro units per ml of the bath Test solution. Dissolve about 25 mg of the substance under
solution. When maximal contraction is reached, replace the examination in 100 ml ofmobile phase A.
bath solution by a fresh solution and wait until the muscle is
relaxed completely and the pointer of the recorder returns to Reference solution. A 0.025 per cent w/v solution of oxytocin
the base line. The doses of the different reference solutions RS in mobile phase A.
should be added at regular intervals depending upon the rate Chromatographic system
of the recovery of the uterine muscle. Dissolve or dilute the a stainless steel column 12.5 cm x 4.6 mm, packed with
preparation to be tested in a suitable diluent (preferably using octadecylsilane bonded to porous silica (5 /lm),
bath solution) to obtain responses on the addition of two mobile phase: A. a 1.6 per cent w/v solution of sodium
dilutions similar to the one used with the oxytocin reference dihydrogen phosphate,
solution. The two selected dilutions ofthe reference solution B. a mixture of 50 volumes of acetonitrile
and preparation under examination should be applied and 50 volumes of water,
according to a randomised block or Latin square design and a linear gradient programme using the conditions given
at least three responses to each dilution should be recorded. below,
The magnitude of contractions obtained with the reference flow rate. 1 ml per minute,
solution is comparable to the contractions obtained with the spectrophotometer set at 220 urn,
test solution. injection volume. 25 Ill.
Time Mobile phase A Mobile phase B
Tests (in min) (per cent v/v) (per cent v/v)
0-30 70-740 30-760
Peptide. 90.0 to 11 0.0 per cent ofthe stated amount ofoxytocin,
C43H66NIZ0IZSZ expressed per mg for the solid, and in mg per 30-30.1 40-770 60-730
ml for the liquid, 30.1- 45 70 30
Determine by liquid chromatography (2.4.14). Inject the test solution and the reference solution.

Test solution. Dissolve 3.5 mg of the substance under Calculate the content OfC43H66N120IzSz.
examination in sufficient of a 1.56 per cent w/v solution of Oxytocin intended for use in the manufacture ofparenteral
sodium dihydrogen phosphate to produce 10.0 ml or use the preparations without a fUrther procedure for the removal of
liquid preparation as appropriate. bacterial endotoxins complies with the following additional
Reference solution. Dissolve 3.5 mg ofoxytocin RS in sufficient requirement.
of a 1.56 per cent w/v solution of sodium dihydrogen Bacterial endotoxins (2.2.3). Less than 0.5 Endotoxin Units
phosphate to produce 10.0 ml. per Unit ofoxytocin.
Chromatographic system Oxytocin intended for use in the manufacture ofparenteral
a stainless steel column 12 cm x 4.6 mm, packed with preparations without a further sterilisation procedure
octadecylsilane bonded to porous silica (5 /lm), complies with the following additional requirement.

1844

www.webofpharma.com
IP 20ID OXYTOCIN INJECTION

Sterility (2.2.11). Complies with the test for sterility. - mobile phase: a mixture of85 volumes ofa 0.2 per cent
Storage. Store protected from moisture. If the substance is v/v solution of orthophosphoric acid and 15 volumes
intended for use in the manufacture ofparenteral preparations, of acetonitrile,
the container should be sterile, tamper-evident and sealed so flow rate. 1 ml per minute,
as to exclude micro-organisms. spectrophotometer set at 220 nm,
- injection volume. 200 J.Ll.
Labelling. The label states (1) the number ofUnits ofoxytocic
activity per mg (for solid) or per ml (for liquid); (2) either the Inject the reference solution. The test is not valid unless the
animal species from which it is obtained or whether it is theoretical plates are not less than 50,000.
synthetic, as appropriate; (3) whether or not the contents are Inject the test solution and the reference solution.
intended for use in the manufacture ofparenteral preparations.
Calculate the content ofC43H66NlzOlzSz in the injection.
Oxytocin Injection containing Oxytocin of natural origin
obtained by extraction and purification complies with the
follOWing additional requirement.
Oxytocin Injection
Vasopressin impurity. Not more than 0.5 Unit perml.
Oxytocin Injection is a sterile solution of Oxytocin in Water
for Injections. Determine by liquid chromatography (2.4.14).

Oxytocin Injection contains not less than 95.0 per cent and Solvent mixture. Dissolve 5.0 g of chlorobutanol in 5.0 ml of
not more than 105.0 per cent ofthe stated number ofUnits of glacial acetic acid, add 1.1 g of sodium acetate, 5.0 g of
oxytocin activity. ethanol, and dilute to 1000 ml with water and mix.

Usual strengths. 5 Units per ml; 10 Units per ml. Test solution. Dilute 2.0 ml of injection under examination to
25 ml with 0.25 per cent w/v of glacial acetic acid and mix.
Description. A clear colourless liquid.
Reference solution. Dissolve the contents of one vial of
Identification vasopressin RS in a lmown volume of solvent mixture. If
necessary dilute the prepared solution to a working
In the Assay, the principal peak in the chromatogram obtained concentration range.
with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with
Tests endcapped octadecylsilane bonded to porous silica
(5 J.Lm),
pH(2.4.24).3.0t05.0 column temperature. 30°,
mobile phase: a mixture of 87 volumes of 6.6 per cent
Bacterial endotoxins (2.2.3). Less than 0.5 Endotoxin Units
v/v solution of dibasic ammonium phosphate, adjusted
per Unit ofoxytocin.
to pH 3.0 with orthophosphoric acid and 13 volumes of
Other tests. Complies with the tests stated under Parenteral acetonitrile, filter through 0.45 J.Lm nylon membrane,
Preparations (Injections). flow rate. 1 ml per minute,
spectrophometer set at 220 nm,
Assay. Determine by liquid chromatography (2.4.14).
- injection volume. 20 J.L1.
Test solution. Use the injection under examination.
Equilibrate the column at least for one hour. Run the
Reference solution. Dissolve the contents of one vial of chromatogram minimum of60 minutes.
oxytocin RS in a 1.65 per cent w/v solution of sodium
Inject the reference solution. The test is not valid unless the
dihydrogen orthophosphate to produce a solution containing
resolution between vasopressin and the adjacent peak is not
the same concentration in J.Lg of oxytocin as that stated on the
less than 1.5 and relative standard deviation for replicate
label ofthe injection.
injections is not more than 2.0 per cent.
Chromatographic system
Inject the reference solution and the test solution.
a stainless steel column 10 cm x 4.6 mm, packed with
endcapped octadecylsilane bonded to porous silica (5 Labelling. The label states (1) the number ofUnits ofoxytocin
J.Lm) (such as Nucleosil C18), activity per ml; (2) either the animal spieces from which it is
column temperature. 40°, obtained or whether it is synthetic, as appropriate.

1845

www.webofpharma.com
OXYTOXIN NASAL SOLUTION IF 2010

Oxytocin Nasal Solution Calculate the content ofC43H66NlzOlzSzin the nasal solution.
Oxytocin Nasal Solution containing Oxytocin of natured
Oxytocin Nasal Solution is a solution ofOxytocin in a suitable
origin obtained by extraction and purification complies with
solvent containing an appropriate antimicrobial preservative.
the follOWing additional requirement.
Oxytocin Nasal Solution contains not less than 85.0 per cent
Vasopressin impurity. Not more than 0.5 Unit perml.
and not more than 120.0 per cent ofthe stated number ofUnits
ofoxytocic activity. Detennine by liquid chromatography (2.4.14).
Usual streugth. 40 Units perml. Solvent mixture. Dissolve 5.0 g of chlorobutanol in 5.0 ml of
Description. A clear, colourless solution. glacial acetic acid, add 1.1 g of sodium acetate, 5.0 g of
ethanol, and dilute to 1000 ml with water and mix.
Identification Test solution. Dilute 2.0 ml of injection under examination to
25 ml with 0.25 per cent w/v of glacial acetic acid and mix.
In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the peak in the Reference solution. Dissolve the contents of one vial of
chromatogram obtained with the reference solution. vasopressin RS in a known volume of solvent mixture. If
necessary dilute the prepared solution to a working
Tests concentration range.
pH (2.4.24). 3.0 to 5.0 Chromatographic system
Other tests. Complies with the tests stated under Nasal a stainless steel column 25 cm x 4.6 mm, packed with
Preparations. endcapped octadecylsilane bonded to porous silica (5
/lIn),
Assay. Detennine by liquid chromatography (2.4.14). column temperature. 30°,
Test solution. Use the nasal solution under examination. mobile phase: a mixture of87 volumes of6.6per cent vi
v solution of dibasic ammonium phosphate, adjusted
Reference solution. Dissolve the contents of one vial of
to pH 3.0 with orthophosphoric acid and 13 volumes of
oxytocin RS in a 1.65 per cent w/v solution of sodium
acetonitrile, filter through 0.45 f-lm nylon membrane,
dihydrogen orthophosphate to produce a solution containing
flow rate. 1 ml per minute,
the same concentration in f-lg ofoxytocin as that stated on the
spectrophometer set at 220 urn,
label of the nasal solution.
injection volume. 20 f-ll.
Chromatographic system
a stainless steel column 10 cm x 4.6 mm, packed with Equilibrate the column at least for one hour. Run the
endcapped octadecylsilane bonded to porous silica (5 chromatogram minimum of60 minutes.
f-lm) (such as Nucleosil CI8), Inject the reference solution. The test is not valid unless the
column temperature. 40°, resolution between vasopressin and the adjacent peak is not
mobile phase: a mixture of85 volumes ofa 0.2 per cent less than 1.5 and relative standard deviation for replicate
v/v solution of orthophosphoric acid and 15 volumes injections is not more than 2.0 per cent.
of acetonitrile,
- flow rate. 1 ml per minute, Inject the reference solution and the test solution.
- spectrophotometer set at 220 nm, Storage. Store at a temperature not exceeding 30°.
- injection volume. 200 f-ll.
Labelling. The label states (1) the number ofUnits ofoxytocic
Inject the reference solution. The test is not valid unless the activity per ml; (2) either the animal species from which it is
theoretical plates is not less than 50,000. obtained or whether it is synthetic, as appropriate; (3) that the
Inject the test solution and the reference solution. preparation is intended for intranasal administration only.

1846

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Paclitaxel 1853
Paclitaxel fujection 1854
Pancreatin 1855
D-Panthenol 1856
Pantoprazole Sodium 1856
Pantoprazole Sustained-release Tablets 1857
Paracetamol 1859
Paracetamol Syrup 1860
Paracetamol Tablets 1861
Hard Paraffin 1862
Liquid Paraffin . 1862
Light Liquid Paraffin 1863
Liquid ParaffinEmulsion 1863
White SoftParaffin 1863
Yellow SoftParaffin 1864
Paraffin Ointment 1865
Paraldehyde 1865
Penicillamine 1866
Penicillamine Tablets 1868
Diluted Pentaerythritol Tetranitrate 18~68

Pentaerythritol Tetranitrate Tablets 1870


Pentamidine Isethionate 1871
Pentamidine fujection 1872
Pentazocine 1873
Pentazocine Hydrochloride 1873
Pentazocine Tablets 1874
Pentazocine Lactate 1875
Pentazocine fujection 1876
Pentobarbitone Sodium 1877
Pentobarbitone Tablets 1877
Pepsin 1878

1847

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Peritoneal Dialysis Solutions 1879


Perphenazine 1881
Perphenazine Tablets 1882
Pethidine Hydrochloride 1883
Pethidine Injection 1884
Pethidine Tablets 1885
Phenindione 1886
Phenindione Tablets 1887
Pheniramine Maleate 1888
Pheniramine Injection 1889
Pheniramine Tablets 1889
Phenobarbitone 1890
Phenobarbitone Tablets 1891
Phenobarbitone Sodium 1891
Phenobarbitone Injection 1892
Phenobarbitone Sodium Tablets 1893
Phenol 1893
Phenolphthalein 1894
Phenoxyethanol 1895
PhenoxymethylpenicillinPotassium 1896
PhenoxymethylpenicillinPotassiumTablets 1897
Phentolamine Mesylate 1898
Phentolamine Injection 1899
Phenylephrine Hydrochloride 1899
Phenylephrine Injection 1900
PhenylmercuricAcetate 1901
Phenylmercuric Nitrate 1901
Phenylpropanolamine Hydrochloride 1902
Phenytoin 1903
Phenytoin Sodium 1904
PhenytoinInjection 1905
Phenytoin Capsules 1906
Phenytoin Tablets 1906

1848

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Phenytoin Oral Suspension 1907


Pholcodine 1908
Pholcodine Linctus 1908·
Phosphoric Acid 1909
Physostigmine Salicylate 1910
Physostigmine fujection 1911
Pilocarpine Nitrate 1911
Pimozide 1912
Pimozide Tablets 1913
Pindolol 1914
Pindolol Tablets 1915
Pioglitazone HydrocWoride 1916
Pioglitazone Tablets 1917
Piperacillin 1918
Piperacillin futravenous Infusion 1919
PiperazineAdipate 1920
PiperazineAdipate Tablets 1921
Piperazine Citrate 1921
Piperazine Citrate Syrup 1922
Piperazine Hydrate 1923
Piperazine Phosphate 1924
Piperazine Phosphate Tablets 1924
Piracetam 1925
Piroxicam 1926
Piroxicam Capsules 1927
Plaster of Paris 1928
Poloxamer 1928 .
Polyethylene Glycol 1500 1929
Polyethylene Glycol 4000 1930
Polyethylene Glycol 6000 1930
Polyoxyl35 Castor Oil 1931
Polyoxyl40 Hydrogenated Castor Oil 1931
Polysorbate 20 1932
:1'~
1849

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Polysorbate 80 ~ ... 1932


Potassium CWoride 1933
Potassium Chloride and Dextrose Injection 1934
Potassium CWoride, Sodium Chloride and Dextrose Injection 1935
PotassiumCitrate 1936
Potassium Clavulanate 1937
Potassium Clavulanate Diluted 1938
Potassium Iodide 1939
Potassium·Permanganate 1940
Povidone 1941
Povidone..Iodine 1943
Povidone-Iodine Solution 1943
PralidoximeCWoride 1944
Pralidoxime CWoride Injection 1944
Pravastatin Sodium 1945
Pravastatin Tablets 1946
Praziquantel 1947
Praziquantel Tablets 1948
Prazosin HydrocWoride 1949
Prazosin Tablets 1950
Prednisolone 1951
Prednisolone Tablets 1952
Prednisolone Acetate 1954
Prednisolone Sodium Phosphate 1955
Prednisolone Sodium Phosphate Injection 1956
Prednisone 1957
Prednisone Tablets 1958
Pregabalin 1960
Pregabalin Capsules 1961
Pregelatinised Starch .... 1963
Primaquine Phosphate 1963
PrimaquineTablets 1964
Probenecid 1965

1850

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Probenecid Tablets 1966


Procainamide Hydrochloride 1966
Procainamide fujection 1967
Procainamide Tablets 1968
Procaine Hydrochloride 1968
Procaine andAdrenaline fujection 1969
Procaine Penicillin 1970
Fortified Procaine Penicillin fujection 1971
ProchlOlperazine Maleate 1972
Prochlorperazine Tablets 1972
Prochlorperazine Mesylate 1973
Prochlorperazine fujection 1974
Procyclidine Hydrochloride 1974
Procyclidine Tablets 1975
Progesterone fujectable Suspension 1976
Proguanil Hydrochloride 1977
Proguanil Tablets 1977
Promazine Tablets 1978
Promethazine Hydrochloride 1979
Promethazine fujection 1979
Promethazine Syrup 1980
Promethaiine Tablets 1981
Promethazine Theoclate 1982
Promethazine Theoclate Tablets 1982
Propane 1983
Propionic Acid 1984
Propofol 1985
Propofol fujection 1986
Propranolol Hydrochloride 1987
Propranolol fujection 1988
Propranolol Tablets 1989
Propyl Gallate 1990
Propylene Glycol 1990

1851

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Propylparaben 1991
Propylthiouracil 1992.
Propylthiouracil Tablets . 1993.
Propyphenazone 1994
Protamine Sulphate 1995
Protamine Sulphate Injection 1996
Prothionamide 1996
Prothionamide Tablets 1997
Protriptylihe HydrocWoride 1998
Protriptyline Tablets 1999
Pseudoephedrine HydrocWoride 1999
Pseudoephedrine Syrup 2000
Pseudoephedrine Tablets 2001
Psoralen 2001
Pyrantel Pamoate .... 2002
Pyrante1Pamoate Oral Suspension 2003
Pyrazinamide 2004
Pyrazinamide Tablets 2005
Pyridoxine HydrocWoride ·2005
Pyridoxine Tablets 2006
Pyrimethamine 2007
Pyrimethamine Tablets ..... 2008
Pyrimethamine and Sulphadoxine Tablets .... 2009

1852

www.webofpharma.com
IP 2010 PACLITAXEL

Paclitaxel Chromatographic system


- a stainless steel column 15 em x 4.6 mm, packed with
Taxo! octadecylsilane bonded to porous silica(3/lm),
column temperature. 35°,
mobile phase: A. a mixture of 60 volumes of water and
40 volumes of acetonitrile,
B. acetonitrile
flow rate. 1.2 ml per minute,
a linear gradient programme using the conditions given
below,
spectrophotometer set at 227nm,
injection volume. 10 Ill.
Time Mobile phase A Mobile phase B
(in min) (per cent v/v) (per cent v/v)
o 100 O.
C47HsINOl4 Mol. Wt. 853.9
20 100 o
Paclitaxel is 5~,20-epoxy-l,2a;4,7 f3,lOf3,13a-hexahydroxytax-
60 10 90
ll-en-9-one 4,1 O-diacetate 2-benzoate 13-ester with(2R,3S)-
N-benzoyl-3-phenylisoserine. 62 100 o
A taxane derivative first isolated from the bark ofthe Pacific 70 100 o
yew tree, Taxus brevifolia. Inject reference solution (b). The test is not valid unless the
Paclitaxel contains not less than 97.0 per cent and not more resolution between the peak due to paclitaxel and 10-deacetyl-
than 102.0 per cent ofC47Hs1N014, calculated on the anhydrous 7-epipaclitaxel is not less than 1.2. The relative retention time
basis. for 10-deacetyl-7-epipaclitaxel is about 0.94 in respect to
paclitaxel.
Category. Anticancer.
Inject the test solution and reference solution (a). In the
Description. A white or almost white powder.
chromatogram obtained with the test solution, the area ofany
CAUTION - Paclitaxel is potentially cytotoxic. Great care secondary peak is not more than 0.5 times the area ofthe peak
should be taken in handling the powder and preparing in the chromatogram obtained with the reference solution (a)
solutions. (0.5 per cent) and the sum of areas of all the secondary peaks
is not more thantwi<;:e the area ofthe peak in the chromatogram
Identification
obtained with the reference solution (a) (2.0 per cent).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Heavy metals (2.3.13). 1 g complies with the limittest for heavy
Compare the spectrum with that obtained with paciltaxel RS. metals, Method B (20ppm ). '
B. In the Assay, the principal peak in the chromatogram
Sulphated ash (2.3.18). Not more than 0.2 per cent. '
obtained withthe test solution corresponds to the peak inthe
chromatogram obtained with the reference solution. Water (2.3.43). Not more than 4.0 percent, determined on 0.1
g by coloumetry method.
Tests
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit
Specific optical rotation (2.4.22). -49.00 to - 55.00 , determined per mg ofpaclitaxel.
in 1.0 per cent w/v solution in methanol.
Microbial contamination (2.2.9). The total viable aerobic count
Related substances. Determine by liquid chromatography
does not exceed 100 cfu per g. It meets the requirements ofthe
(2.4.14).
tests for the absence of Staphylococcus aureus, Pseudomonas
Test solution. Dissolve 10 mg ofsubstance under examination aeruginosa, Salmonella species, and Escherichia coli.
in 10 ml of acetonitrile.
Assay. Determine by liquid chromatography (2.4.14).
Reference solution (a). A 0.001 per cent w/v solution of
paclitaxel RS in acetonitrile. Solvent mixture. Dissolve 200 III of glacial acetic acid in
Reference solution (b). A solution containing 0.008 per cent 1000 ml of methanol.
w/vof 10 deacetyl-7-epipaclitaxel and 0.1 per cent w/v of Test solution. Dissolve 0.1 g of the substance under
paclitaxel RS in acetonitrile. examination in 100.0 ml in solvent mixture.

1853

www.webofpharma.com
PACLITAXEL INJECTION IP 2010

Reference solution. A 0.1 per cent w/v solution ofpaclitaxel Reference solution (b). A solution containing 0.006 per cent
RS in solvent mixture. w/v of 10 deacetyl-7-epipaclitaxel RS and 0.12 per cent w/v
of paclitaxel RS in acetonitrile.
Chromatographic system
a stainless steel column 25 ern x.4;6 mm, packed with Chromatographic system
pentafltioro phenyl groups bonded to porous silica (5 a stainless steel column 15 em x 4.6 mm, packed with
J.Ull), octadecylsilane bonded to porous silica (311m),
column temperature 35°, column temperature. 35°,
mobile phase: a mixture of 11 volumes of water and mobile phase: A. a mixture of 00 volumes of water and
9 volumes of acetonitrile, 40 volumes of acetonitrile,
flow rate. 1.5 rn1 per minute, B. acetonitrile,
spectrophotometer set at 227nm, flow rate. L2 rn1 per minute,
injection volume. 10 Ill. a linear gradient programme using the conditions given
below,
Inject the reference solution. The test is not valid unless the
- spectrophotometer set at 227 nm,
tailing factor is not more than L5. The relativestandard
deviation for replicate injections is not more than 2.0 per cent. injection volume. lO Ill.
Time Mobile phase A Mobile phase B
Inject the test solution and reference solution.
(in min) (per cent v/v) (percent v/v)
Calculate the content OfC47HsIN014.
o 100 o
Storage. Store protected from light, at a temperature not
exceeding 25°.
26 100 o
66 17 83
67 100 o
Paclitaxel Injection Inject reference solution (b). The test is not valid uniess the
resolution between the peak due to paclitaxel and 10-deacetyl-
Paclitaxel Injection is a sterile solution of Paclitaxel suitable 7-epipaclitaxel is not less than 1.2. The relative retention time
for dilution,for intravenous use. for 1O-deacetyl-7-epipaclitaxe1 is about 0.94 in respect to
Paclitaxel Injection contains not less than 90.0 per cent and paclitaxel.
not more than 110.0 per centofthestated amount of paclitaxel, Inject the test solution and reference solution (a). In the
C47HsIN014. . chromatogram obtained with the test solution, the area of any
Description. A clear colourless to slight yellow viscous secondary peak is not more than 0.8 times the area ofthe peak
solution. in the chromatogram obtained with the reference solution (a)
(0.8 per cent) and the sum of areas of all the secondary peaks
Identification is not more than twice the area ofthe peak in the chromatogram
obtained with the reference solution (a) (2.0 per cent).
In the Assay, the principal peak in the chromatogram obtained
with test solution corresponds to the peak in the chromatogram Other tests. Complies with the tests stated under Parenteral
obtained with reference solution. Preparations (Injections).

Tests Bacterial endotoxins (2.2.3). Not more than 0.67 Endotoxin


Unit per mg ofpaclitaxel.
pH (2.4.24). 3.0to 7.0, determined in a 10percentv/v solution
in water. Sterility (2.2.11). Complies with the test for sterility.

Light absorption. Absorbance of the injection at about 425 Assay. Determine by liquid chromatography (2.4.14).
nm, not more than 0.1. Solvent mixture. Dissolve 200 III of glacial acetic acid in
Related substances. Determine by liquid chromatography 1000 ml of methanol.
(2.4.14).
Test solution. Accurately measure the volume of injection
Test. solution. Accurately measure the volume of injection containing 6 mg ofPaclitaxel and dissolve in 10 ml ofsolvent
containing 12 mg of Paclitaxel, dilute to 10 rn1 with acetonitrile. mixture.
Reference solution (a). A 0.0012 per cent w/v solution of Reference solution. A 0.06 per cent w/v solution ofpaclitaxel
paclitaxel RS in acetonitrile. RS in solvent mixture.

1854

www.webofpharma.com
lP 2010 PANCREATIN

Chromatographic system and solution (2) to the remainder and maintain the mixtures at
a stainless steel column 30 cm x 3.9 mm, packed with 39° ± 1° for 5 minutes. To I ml of each mixture add 10 mlof
penta fluro phenyl group chemically bonded to porous iodinated potassium iodide solution. The liquid containing
silica'(5 jlm), solution (2) retains the colour of the solution of iodine and
column temperature 35°, the liquid containing solution (1) acquires an intense blue
mobile phase: a mixture of 11 volumes of water and 9 colour.
volumes of acetonitrile,
- flow rate. 1.5 ml per minute, Tests
- spectrophotometer set at 227nm, Fat. Not more than 5.0 per cent, determined by the following
- injection volume. 10 jll. method. Extract I g with light petroleum (40° to 60°) for
Inject the reference solution. The test is not valid unless the 3 hours in an apparatus for the continuous extraction ofdrugs
tailing factor is not more than 1.5. The relative standard (2.1.8), evaporate the extract and dry the residue at 105° for
deviation for replicate injections is not more than 2.0 per cent. 2 hours.
Inject the test solution and reference solution. Microbial contamination (2.2.9). 1g is free from Escherichia
coli; 10 g is free from Salmonellae.
Calculate the content ofC47HsINOI4.
Loss on drying (2.4.19). Not more than 5.0 per cent, determined
Storage. Store protected from light, at a temperature not
on 0.5 g by dying in an oven at 60° at a pressure not exceeding
exceeding 25°.
0.7 kPa for 4 hours.
Assay. For protease activity - Weigh accurately 4.0 g of
purified casein and dissolve in about 90 ml of water containing
Pancreatin 3 ml of1 M sodium hydroxide, adjust the pH ofthe solution to
8.7 and add sufficient water to produce 100.0 ml. Weigh
Pancreatin is a preparation ofmammalian pancreas containing accurately about 0.5 g of the substance under examination,
protease, lipase and amylase activity. It may contain Sodium triturate with water and add sufficient water to produce
Chloride. 300.0 ml to give the test solution. Dilute 15.0 ml ofthe casein
Pancreatin contains not less than the minimum protease solution with 30 ml of water, warm to 55° and add 10.0 ml ofthe
activity, amylase activity and lipase activity determined under unfiltered test solution. Heat rapidly to 55° and keep at this
the conditions of the Assay. temperature for 20 minutes. Cool rapidly to room temperature.
Dilute a further portion of 15.0 ml ofthe casein solution with
Category. Digestive enzyme. 30 ml of water, add 10.0 ml of the unfiltered test solution,
Dose. 500 mg to 1g. previously boiled and cooled, heat rapidly to 55° and keep at
this temperature for 20 minutes. Cool to room temperature. To
Description. A white or buff-coloured, amorphous powder;'
each solution add 0.75 ml ofphenolphthalein solution and 10
odour, meaty and not unpleasant. ml ofJormaldehyde solution. Titrate each solution with 0.1 M
sodium hydroxide until the colour of the solution matches
Identification
that produced by mixing 10 ml of buffer solution pH 8.7 and
A. Triturate 0.5 g with 10 ml of water and adjust to pH 8.0 by 0.15 ml of phenolphthalein solution. The difference between
the addition of 1 M sodium hydroxide using cresol red the two titrations is not less than 4.5 ml.
solution as indicator. Divide the resulting solution into two
For lipase activity- To 95 ml of water, add 6.5 mloftriacetin
equal portions. Boil one portion [solution (1)] and leave the and 0.2 ml ofa 0.1 per centw/v solution of bromocresolpurple,
other untreated [solution (2)]. To each add a few shreds of neutralise with 0.5 M sodium hydroxide and add sufficient
congo red fibrin, warm to 39° ± 1° and maintain at this
water to produce 110 mI. Place 50 ml ofthis solution in each of
temperature for 1 hour. Solution (2) is stained red and solution two large tubes 3 cm x 20 cm A and B contained in a thermostat
(1) is colourless or not more than slightly pink.
at 30°. Insert in each tube a rubber stopper having two holes,
B. Triturate 0.25 g with 10 ml of water and adjustto pH 8.0 by onefor the tip ofa burette and the other fora short glass tube
the addition of 1 M sodium hydroxide using cresol red through which passes a thread operating a glass stirring coil.
solution as' indicator. Divide the resulting solution into two Stir the contents of the tube until they attain the temperature
equal portions. Boil one portion [solution (1)] and leave the of the thermostat. Prepare a solution of 0.1 g ofthe substance
other untreated [solution (2)]. Dissolve 0.1 g ofsoluble starch under examination in 10.0 ml of water. To tube A add 1.0 ml of
in 100 ml ofboiling water, boil for 2 minutes, cool and dilute to the solution, to tube B add 1.0 ml of the solution previously
150 ml with water. Add solution (1) to halfthe starch mucilage boiled. Adjust and maintain the pH ofthe solutions in the two

1855

www.webofpharma.com
PANCREATIN IP 2010

tubes to 6.2 to 6.4 by the addition of 0.05 M sodium hydroxide Identification


dropwise, stirring frequently. After 30 minutes, the difference
Boil 50 mg with 5 ml of 0.1 M sodium hydroxide for 1 minute,
between the volumes of 0.05 M sodium hydroxide added to
cool and add 5 ml of1 M hydrochloric acid and 0.1 mlofjerric
the two tubes is not less than 1.0 ml.
chloride test solution; a deep yellow colour is produced.
For amylase activity- Not less than I00 Units per g. Dissolve
0.1 g or a quantity containing 10 Units, accurately weighed, in Tests
sufficient buffer solution pH 6.8 to produce 1000.0 ml. Filter if pH (2.4.24). Not more than 10.5, determined in a 5.0 percent
necessary (1 ml of the test solution should be capable of w/v solution in carbon dioxide-free water.
digesting about 10 mg of dry soluble maize or corn starch).
Into each of six stoppered test~tubes add 5.0 ml of starch Specific optical rotation (2.4.22). +28.2° to +30.2°, determined
substrate without touching the sides of the test-tube. Place at 20° in a 5.0 per cent w/v solution.
the test-tubes in a water-bath maintained at 40° ± 0.1°. When Refractive index (2.4.27). 1.490 to 1.498, determined at 20°.
the temperature of the solution in the tubes has reached 40°, Heavy metals (2.3.13). 1.0 g dissolved in 25 rnl ofwater complies
add 0.35 ml, 0.4 rnl, 0.45 rnl, 0.5 ml, 0.55 ml and 0.6 rnl ofthe test with the limit test for heavy metals, Method A (20 ppm).
solution to each of the test-tubes marked 1 to 6 respectively
and record the time of addition. Mix thoroughly and replace Assay. Weigh accurately about 0.5 g and carry out Method A
the tubes in the water-bath. After exactly 60 minutes remove for the determination ofnitrogen (2.3 .30).
the tubes and cool rapidly in cold water. Add to each tube 1 ml of 0.05 Msulphuricacidis equivalentto 0.001401 g ofN.
0.05 ml of 0.02 M iodine and mix well. Note the tube containing Storage. Store protected from moisture.
the lowest volume of test solution, which does not show a
bluish or violet tinge (if there is doubt, warm the solution
slightly, when the colour distinction is prominent). From this
volume calculate the number ofgrams ofdry soluble maize or Pantoprazole Sodium
corn starch digested by 1.0 g of the substance under
examination. This represents the number ofUnits of amylase
activity per g.
Storage. Store protected from moisture.
Labelling. The label states the name of any added substance.

Mol. Wt. 432.4


Pantoprazole Sodium is sodium 5-(difluromethoxy)-2[[(3,4
D-Panthenol
uimethoxy-pyridin-2-yl)methyl]sulphinyl]benzimidazol-1-ide,
Pantothenol; Dextro-pantothenyl Alcohol sesquihydrate.
Pantoprazole Sodium contains not less than 98.0 per cent and
not more than 102.0 per cent ofCI6HI4F2N3Na04S, calculated
on the anhydrous basis.
Category. Antiulcer.
Description. A white to off-white powder.
C9H I9N04 Mol. Wt. 205.3 Identification
D-Panthenol is (R)-2,4-dihydroxy-N-(3-hydroxypropyl)-3, 3- Test A may be omitted if tests Band C are carried out. Test B
dimethylbutanamide. may be omitted if tests A and C are carried out.
D-Panthenol contains not less than 6.60 per cent and not more A. Determine by infrared absorption spectrophotometry (2.4.6).
than 6.95 per cent of nitrogen, N. Compare the spectrum with that obtained with pantoprazole
Category. Vitamin B (enzyme co-factor). sodium RS or with the reference spectrum of pantoprazole
sodium.
Dose. 250 to 500 mg.
B. In the Assay, the principal peak in the chromatogram
Description. A clear, colourless or slightly yellow, viscous obtained with the test solution· corresponds to the principal
liquid; odourless. peak in the chromatogram obtained with the reference solution.

1856

www.webofpharma.com
IP 2010 PANTOPRAZOLE SUSTAINED-RELEASE TABLETS

C. Gives the reaction (a) ofsodium (2.3.1). Assay. Determine by liquid chromatography (2.4.14).

Tests Solvent mixture (a). Dilute 25.0 ml of ammonium hydroxide


to 500.0 ml with water.
Optical rotation (2.4.22). -0.40° to +0.40°, determined on 1.0
Solvent mixture (b). Equal volumes of acetonitrile and water.
per cent w/v solution.
Test solution. Dissolve about 20 mg of the substance under
Related substances. Determine by liquid chromatography
examination 10.0 ml ofsolvent mixture (b) and dilute to 50.0 ml
(2.4.14).
with solvent mixture (a). Dilute 3.0 ml ofthis solution to 20.0 ml
Solvent mixture. Equal volumes of acetonitrile and 0.001 M with solvent mixture (a).
sodium hydroxide.
Reference solution. Dissolve 20 mg of pantoprazole sodium
Test solution. Dissolve about 23 mg of the substance under RSin 10.0 ml of solvent mixture (b) and dilute to 50.0 ml with
examination in 50.0 ml ofthe solvent mixture. solvent mixture (a). Dilute 3.0 ml ofthis solution to 20.0 ml with
Reference solution. A 0.06 per cent w/v solution of solvent mixture (a).
pantoprazole sodium RS in the solvent mixture. Dilute 2.5 ml
Chromatographic system
ofthe solution to 50.0 ml with the solvent mixture.
- a stainless steel column 15 cm x 3.9 mm packed with
Chromatographic system octadecylsilane bonded to porous silica (4 Ilm) (Such
- a stainless steel column 12.5 cm x 4.0 mm packed with as Inertsil ODS-3),
octadecylsilane bonded to porous silica (5 Ilm) (Such - column temperature. 30°,
as Inertsil ODS-3), - sample temperature.4°,
- column temperature. 40°, mobile phase: A. a mixture of 85 volumes of buffer
- mobile phase: A. dissolve 1.74 g of dibasic potassium solution prepared by dissolving 1.32 g of dibasic
phosphate to 1000 ml with water; adjusted to pH ammonium phosphate to 1000 ml with water, adjusted
7.0 with orthophosphoric acid, to pH 7.5 with orthophosphoric acid and 15 volumes of
B. acetonitrile, acetonitrile,
- flow rate. 1 ml per minute, B. a mixture of7 volumes of acetonitrile
- spectrophotometer set at 290 run, and3 volumes of methanol,
- injection volume. 20 Ill. flow rate. 1 ml per minute,
Time Mobile phase A Mobile phase B - spectrophotometer set at 285 run,
(in min) (per cent v/v) (per cent v/v) - injection volume. 20 Ill.
040 80-720 20-780 Time Mobile phase A Mobile phase B
40-45 20 80 (in min) (per cent v/v) (per cent v/v)
45-55 20-780 80-720 0-10 86 14
Inject the reference solution. The test is not valid unless the 35 42 58
relative standard deviation for replicate injections is not more 36 86 14
than 2.0 per cent. 46 86 14
Inject the test solution and the reference solution. In the Inject the reference solution. The test is not valid unless the
chromatogram obtained with the test solution the area of the relative standard deviation for replicate injections is not more
peak due to impurity C determined at wavelength 305 nm at than 2.0 per cent.
relative retention time about 0.6 is not more than 0.77 times the
area of the principal peak at 290 nm in the chromatogram Inject the test solution and the reference solution.
obtained with reference solution (b) (0.5 per cent), the area of Calculate the content ofC16H14F2N3Na04S,
any secondary peak is not more than 0.077 times the area of
Storage. Store protected from light and moisture, between 2°
the principal peak in the chromatogram obtained with reference
to 8°.
solution (b) (0.5 per cent) and the sum of the areas of all the
secondary peaks is not more than 0.15 times the area of the
principal peak in the chromatogram obtained with reference
solution (b) (1.0 per cent).
Pantoprazole Sustained-release Tablets
Heavy metals (2.3.13). 1.0 g complies with the limit test for Pantoprazole Sodium Sustained-release Tablets
heavy metals, Method B (20 ppm). Pantoprazole Sustained-release Tablets contain not less than
Water (2.3.43). Not less than 5.0 per cent and not more than 90.0 per cent and not more than 110.0 per cent of the stated
8.0 per cent, determined on 0.15 g. amount ofpantoprazole, C16HlSF2N304S.

1857

www.webofpharma.com
PANTOPRAZOLE SUSTAINED-RELEASE TABLETS IP 2010

Usual strenth. 40 mg. Uniformity of content. Comply with the test stated under
Tablets.
Identification
Determine by liquid chromatography (2.4.14), as described in
A. In the Assay, the principal peak in the chromatogram the Assay using the following solutions.
obtained with the test solution corresponds to the principal
peak: in the chromatogram obtained with the reference solution. Test solution. Disperse 1 intact tablet in 100.0 ml ofthe mobile
phase and filter.
B. When examined in the range 230 urn to 350 nm (2.4.7), a
0.001 per cent w/v solution in methanol shows absorption Reference solution. Dissolve an accurately weighed quantity
maxima at about 289 urn. of pantoprazole sodium RS in the mobile phase and dilute
with the mobile phase to obtain a solution having a known
Tests concentration similar to the test solution.
Dissolution (2.5.2). Inject the reference solution and the test solution.
NOTE-Prepare the solutions immediately before use. Calculate the content ofCI6HIsF2N304S in the tablet.
Protect the solutions from light.
Other tests. Comply with the tests stated under Tablets.
A. Apparatus No.1,
Medium. 1000 ml of 0.1 M hydrochloric acid, Assay. Determine by liquid chromatography (2.4.14).
Speed and time. 100 rpm and 120 minutes. NOTE-Prepare the solutions immediately before use.
Determine by liquid chromatography (2.4.14). Protect the solutions from light.
Test solution. Withdraw the medium completely and disperse Test solution. Weigh and powder 20 tablets. Weigh accurately
the intact tablet in 100 ml of the mobile phase and filter. a quantity ofpowder containing about 20 mg ofPantoprazole
Sodium, disperse in 100 ml ofthe mobile phase and filter.
Reference solution. Dissolve an accurately weighed quantity
of pantoprazole sodium RS in the mobile phase and dilute Reference solution. A 0.02 per cent w/v solution of
with the mobile phase to obtain a solution having a known pantoprazole sodium RS in the mobile phase.
concentration similar to the test solution.
Chromatographic system
Use chromatographic system as described under Assay. stainless steel column 25 cm x 4.6 mm, packed with
Inject the reference solution and the test solution. octadecylsilane bonded to porous silica (5 Ilm) (such as
Inertsil ODS-3),
Calculate the content of CI6HISF2N304S released in the acid mobile phase: a mixture of50 volumes ofbuffer solution
medium by subtracting the content of CI6HISF2N304S in the prepared by dissolving 6.8 g of potassium dihydrogen
test solution from the total content of Pantoprazole, orthophosphate and. 1 g of hexane sulphonic acid
CI6HISF2N304S determined in the Assay. sodium salt in 1000 ml of water, adjusted to pH 7.3 with
D. Not more than 10 per cent of the stated amount of 1 M sodium hydroxide and 50 volumes of acetonitrile,
CI6HISF2N304S. flow rate. 1.5 m1 per minute,
spectrophotometer set at 290 urn,
B. Apparatus No.1,
injection volume. 10 Ill.
Medium. 1000 ml of tris-acetate buffer solution pH 8.5,
Inject the reference solution. The test is not valid unless the
Speed and time. 75 rpm and 60 minutes.
theoretical plates are not less than 2000, the tailing factor is
Run method A on another 6 tablets and discard the medium not more than 2.0 and the relative standard deviation for
completely and fill the empty vessel with the dissolution replicate injections is not more than 2.0 per cent.
medium. Withdraw a suitable volume ofthe medium and filter.
Dilute the filtrate, if necessary, with the dissolution medium. Inject the test solution and the reference solution.
Measure. the absorbance at the maximum at about 290 nm Calculate the content OfC16HISF2N304S in the tablets.
(2.4.7). Calculate the content ofC,6H,sF2N304S in the medium
from the absorbance obtained from a solution of known Storage. Store protected from moisture, at a temperature not
concentration of pantoprazole sodium RS. exceeding 30°.
D. Not less than 75 per cent of the stated amount of Labelling. The label states the strength in terms ofthe amount
CI6HISF2N304S. ofPantoprazole.

1858

www.webofpharma.com
IP 2010 PARACETAMOL

Paracetamol Related substances. Determine by liquid chromatography


(2.4.14).
Acetaminophen
Note-Prepare the solutions immediately before use.
Test solution. Dissolve 0.2 g of the substance under
examination in 2.5 ml of methanol containing 0.46 per cent
w/v of tetrabutylammonium hydroxide solution (40 per cent
w/v) and dilute to 10.0 ml with the solution containing equal
volumes of 1.79 per cent w/v ofdisodium hydrogenphosphate
and 0.78 per cent w/v of sodium dihydrogen phosphate.
Reference solution (a). Dilute 1.0 ml of the test solution to
50.0 ml with the mobile phase. Dilute 5.0 ml ofthis solution to
Mol. Wt. 151.2 100.0 ml with the mobile phase.

Paracetamol is 4-hydroxyacetanilide. Reference solution (b). Dilute 1.0 ml of reference solution (a)
to 10.0 ml with the mobile phase.
Paracetamol contains not less than 99.0 per cent and not more
than 101.0 per cent ofCsHgN02, calculated on the dried basis. Reference solution (c). Asolution containing 0.025 per cent
w/v each of 4-aminophenol, paracetamol RS and
Category. Analgesic; antipyretic. chloroacetanilide in methanol. Dilute 1.0 ml of this solution
Dose. 500 mg to I g every 4 to 6 hours, upto 4 g daily, in to 250.0 ml with the mobile phase.
divided doses
Reference solution (d). Dissolve 20 mg of 4-nitrophenol in
Description. White crystals or a white, crystalline powder. 50.0 ml of methanol. Dilute 1.0 ml ofthis solution to.20.0 ml
with the mobile phase.
Identification
Chromatographic system
TestA may be omitted if tests B C andD are carried out. Tests - a stainless steel column 25 cm x 4.0 rom, packed with
B, C and D may be omitted if test A is carried out. . octylsilane bonded to porous silica (5 /lm),
A. Determine by infrared absorption spectrophotometry (2.4.6). - column temperature. 35°,
Compare the spectrum with that obtained with paracetamol - mobile phase: a mixture of 37.5 volumes of a 1.79 per
RS or with the reference spectrum ofparacetamol. cent w/v solution of disodium hydrogen phosphate,
37.5 volumes of a 0.78 per cent w/v solution ofsodium
B. Dissolve 50 tng in sufficient methanol to produce 100 ml. dihydrogen phosphate and 25 volumes of methanol
To 1 ml ofthis solution add 0.5 m1 of0.1 M hydrochloric acid containing 0.46 per cent v/v of tetrabutylammonium
and dilute to 100 ml with methanol. Protect the resulting hydroxide solution (40 per cent w/v),
solution from bright light and immediately measure the - flow rate. 1.5 ml per minute,
absorbance at the maximum at about 249 nm; absorbance at - spectrophotometer set at 245 nm,
249 nm, about 0.44 (2.4.7). - injection volume. 20 Ill.
C. Boil 0.1 g in 1 ml of hydrochloric acid for 3 minutes, add
Inject reference solution (c). The test is not valid unless the
10 ml of water and cool; no precipitate is produced. Add
resolution between the peaks due to 4-aminophenol
0.05 ml of 0.0167 M potassium dichromate; a violet colour
(paracetamol impurity K) and paracetamol is not less than 4.0
develops which does not tum red.
and the signal-to-noise ratio of the peak due to
D. Gives the reaction of acetyl groups (2.3.1). chloroacetanilide (paracetamol imputity J) is not less than 50.
The relative retention time with reference to paracetamol for
Tests paracetamol impurity K is about 0.8, for 4-nitrophenol
(paracetamol impurity F) is about 3.0 and for paracetamol
4-Aminophenol. Dissolve 0.5 g in sufficient methanol (50 per
impurity J is about 7.0.
cent) to produce 10 ml. Add 0.2 ml offreshly prepared alkaline
sodium nitroprusside solution, mix and allow to stand for Inject the test solution, reference solution (a), (b) and (c). Run
30 minutes. Any blue colour in the solution is not more intense the chromatogram 12 times the retention time ofthe principal
than that in 10 ml of a solution prepared at the same time and peale In the chromatogram obtained with the test solution the
in the same manner containing 0.5 g of 4-aminophenol-free area ofthe peak due to paracetamol impurity J is not more than
paracetamol and 0.5 ml of a 0.005 per cent w/v solution of 0.2 times the area of the corresponding peak in the
4-aminophenol in methanol (50 per cent) (50 ppm). chromatogram obtained with reference solution (c) (10 ppm)

1859

www.webofpharma.com
PARACETAMOL SYRUP IP 2010

and the area ofthe peak due to paracetamol impurity K is not Reference solution. A 0.25 per cent w/v solution of
more than the area of the corresponding peak in the paracetamol RS in methanol
chromatogram obtained with reference solution (c) (50 ppm). Apply to the plate 10 III of each solution. After development,
The area of any other secondary peak is not more than 0.5 dry the plate in a current of warm air, examine in ultraviolet
times the area of the principal peak in the chromatogram light at 254 nm. The principal spot in the chromatogram
obtained with reference solution (a) (0.05 per cent). The sum obtained with the test solution corresponds to that in the
ofareas ofother secondary peaks is not more than the area of chromatogram obtained with the reference solution.
the principal peak in the chromatogram obtained with reference
solution (a) (0.1 per cent). Ignore any peak with an area less B. In the Assay, the principal peak in the chromatogram
than 0.5times the area ofthe principal peak in the chromatogram obtained with the test solution corresponds to the peak in the
obtained with reference solution (b) (0.01 per cent). chromatogram obtained with reference solution.
Heavy metals (2.3.13).2.0 g complies with the limit test for Tests
heavy metals, Method B (10 ppm).
4-Aminophenol. Determine by liquid chromatography (2.4.14).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Test solution. Shake 5 ml ofthe preparation under examination
Loss on drying (2.4.19). Not more thanO.5 per cent, determined
with 15 ml ofthe mobile phase, dilute to 25 ml with the mobile
on 1.0 g by drying in an oven at 105°.
phase and filter if necessary.
Assay. Weigh accurately about 0.5 g, dissolve in a mixture of
Reference solution. A 0.0025 per cent w/v solution of
10 ml of water and 50 ml of 1 M sulphuric acid. Boil under a
4-aminophenol in the mobile phase.
reflux condenser for 1 hour, cool and dilute to 100.0 ml with
water. To 20.0 ml of the solution add 40 mlofwater, 40 g of Chromatographic system
water in the form of ice, 15 ml of 2 M hydrochloric acid and a stainless steel column 20 cm x 4.6 mm, packed with
0.1 ml of ferro in solution and titrate with 0.1 M eerie octadecylsilane bonded to porous silica (10 11m),
ammonium sulphate until a yellow colour is produced. Carry - mobile phase: 0.01 M sodium butanesulphonate in a
out a blank titration. mixture of85 volumes of water, 15 volumes of methanol
and 0.4 volume offormic acid,
1 ml of 0.1 M eerie ammonium sulphate is equivalent to
- flow rate. 2 ml per minute,
0.00756 gofCsHgN02 •
- spectrophotometer set at 272 urn,
Storage. Store protected from light and moisture. - injection volume. 20 Ill.
In the chromatogram obtained with the test solution the area
of any peak corresponding to 4-aminophenol is not greater
than the area of the peak in the chromatogram obtained with
Paracetamol Syrup the reference solution. In the chromatogram obtained with the
test solution peaks with a long retention time may occur due
Paracetamol Oral Solution; Acetaminophen Syrup
to preservatives in the preparations.
Paracetamol Syrup is a solution ofParacetamol in a suitable
Other tests. Complies with the tests stated under Oral Liquids.
flavoured vehicle. ,
Assay. Determine by liquid chromatography (2.4.14).
Paracetamol Syrup contains not less than 95.0 per cent and
not more than 105.0 per cent w/v solution ofthe stated amount Test solution. Mix an accurately weighed quantity of the
ofparacetamol, CsHgN0 2 • preparation under examination containing 25 mg of
Paracetamol in 100 ml ofthe mobile phase, dilute to 200.0 ml
Usual strength. 125 mgper 5 ml.
with the mobile phase and filter if necessary.
Identification Reference Solution. A 0.0125 per cent w/v solution of
paracetamol RS in the mobile phase.
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254. Chromatographic system
- a stainless steel column 20 cm x 4.6 mm, packed with
Mobile phase. A mixture of 65 volumes of chloroform,
octadecylsilane bonded to porous silica (10 11m),
25 volumes of acetone, 10 volumes of toluene and 0.5 volumes
- mobile phase: 0.01 M sodium butanesulphonate in a
of glacial acetic acid.
mixture of85 volumes of water, 15 volumes of methanol
Test solution. Dilute a volume containing 25 mg ofParacetamol and 0.4 volume offormic acid,
to 10 ml with methanol and filter ifnecessary. - flow rate. 2 ml per minute,

1860

www.webofpharma.com
IF 2010 PARACETAMOL TABLETS

- spectrophotometer set at 243 nm, test solution peaks with a long retention times may occur due
- injection volume. 20 !-d. to excipients.
Determine the weight per ml (2.4.29) ofthe syrup and calculate Related substances. Determine by liquid chromatography
the percentage content ofCsH 9N02, weight in volume. (2.4.14).
Storage. Store protected from light and moisture. Test solution. Disperse a quantity of powdered tablets
containing about 0.2 g ofParacetamol in 10.0 ml ofthe mobile
phase, filter.

Paracetamol Tablets Reference solution (a). Dilute 1 ml ofthe test solution to 20 ml


with the mobile phase. Dilute lml ofthis solution to 20 ml with
Acetaminophen Tablets the mobile phase.
Paracetamol Tablets contain not less than 95.0 per cent and Reference solution (b). A solution containing 0.002 per cent
not more than 105.0 per cent of the stated amount of w/v each of 4-aminophenol and paracetamol RS in the
paracetamol, CsH9N02 • mobile phase.
Usual strengths. 300 mg; 500 mg. Reference solution (c). ) A 0.00002 per cent w/v solution of 4-
chloroacetanilide in the mobile phase.
Identification
Chromatographic system
Extract a quantity ofthe powdered tablets containing 0.5 g of . - a stainless steel column 25 cm x 4.0 mm, packed with
Paracetamol with 20 ml of acetone, filter, evaporate the filtrate octylsilane bonded to porous silica (5 J.1m) (Such as
to dryness and dry at 105°. The residue complies with the Zorbax Rx C8),
following tests. - column temperature. 35 0,
A. Determine by infrared absorption spectrophotometry (2.4.6). - mobile phase: a mixture of 25 volumes of methanol
Compare the spectrum with that obtained with paracetamol containing 1.15 g of tetrabutylammonium hydroxide
RSor with the reference spectrum ofparacetamol. solution (40 per centw/v), with 37.5 volumes of 0.05 M
disodium hydrogen orthophosphate and 37.5 volumes
B. Boil 0.1 gin 1 ml of hydrochloric acid for 3 minutes, add
of 0.05 M sodium dihydrogen orthophosphate,
10 ml of water and cool; no precipitate is produced. Add
- flow rate. 1.5 ml per minute,
0.05 ml of 0.0167 M potassium dichromate; a violet colour
- spectrophotometer set at 245 nm,
develops which does not tum red.
- injection volume. 20 j..1l.
Tests Inject reference solution (b). The test is not valid unless the
4-Aminophenol. Determine by liquid chromatography (2.4.14). resolution between the two principal peaks is not less than
4.0.
Test solution. Shake a quantity of the powdered tablets
containing 1 g ofParacetamol with 15 ml of methanol, dilute to Inject the test solution and reference solution (a), (b) and (c).
100 ml with water and filter. Run the chromatogram 12 times the retention time of the
principal peak. In the chromatogram obtained with the test
Reference solution. A 0.001 per cent w/v solution of solution the area of peak corresponding to 4-aminophenol is
4-aminophenol in methanol (15 per cent). not more than the area ofthe corresponding peak in reference
Chromatographic system solution (b) (0.1 per cent), the area of peak corresponding to
- a stainless steel column 20 cm x 4.6 mm, packed with 4- chloroacetanilide is not more than the area of the principal
octadecylsilane bonded to porous silica (10 j..1m), peak in reference solution (c) (10 ppm) and the area of any
- mobile phase: 0.01 M sodium butanesulphonate in a other secondary peak is not more than the area ofthe principal
mixture of85 volumes of water, 15 volumes of methanol peak obtained with reference solution (a) (0.25 per cent).
and 0.4 volume offormic acid, Dissolution (2.5.2).
- flow rate. 2 ml per minute,
- spectrophotometer set at 272 nm, Apparatus. No 1
- injection volume. 20 J.1l. Medium. 900 ml ofphosphate bufferpH 5.8
Speed and time. 50 rpm and 30 minutes.
In the chromatogram obtained with the test solution the area
of any peak corresponding to 4-aminophenol is not greater Withdraw a suitable volume ofthe medium and filter and dilute
than the area of the peak in the chromatogram obtained with a suitable volume ofthe filtrate with the same solvent. Measure
the reference solution. In the chromatogram obtained with the the absorbance of the resulting solution at the maximum at

1861

www.webofpharma.com
HARD PARAFFIN IP 2010

about 243 nm (2.4.7). Similarly measure the absorbance of a Dose. 10 to 30 ml.


solution oflmown concentration ofparacetamol RS. Calculate
Description. A transparent, colourless, oily liquid~ free from
the content of CsHgNO z. fluorescence by daylight; odourless or almost odouiless.
D. Not less than 80 per cent ofthe stated amount ofCsHgNO z.
Tests
Other tests. Complies with the tests stated under Tablets.
Weight per ml (2.4.29). 0.860 g to 0.904 g.
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity ofthe powder containing about 0.15 g ofParacetamol, Dynamic viscosity (2.4.28). 110 mPas to 230 mPas, determined
at200±10 bymethodB or Viscosity (2.4.28). 10 cps to 40 cps,
add 50 mloiO.l M sodium hydroxide, dilute with 100 ml of
water, shake for 15 minutes and add sufficient water to produce determined at 30° ±l ° by method C using LVI spindle at 30
200.0 ml. Mix, filter and dilute 10.0 ml ofthe filtrate to 100.0 ml rpm.
with water. To 10.0 ml of the resulting solution add 10 ml of Acidity or alkalinity. To 10.0 g add 20 ml ofboiling water, heat
0.1 M sodium hydroxide, dilute to 100.0 ml with water and in a water-bath for 5 minutes, shake vigorously for 1 minute,
mix. Measure the absorbance of the resulting solution at the cool, allow to separate and filter the aqueous layer. To 10 ml of
maximum at about 257 nm (2.4.7). Calculate the content of the filtrate add 0.1 ml of phenolphthalein solution. The"
CsHgNO z taking 715 as the specific absorbance at 257 nm. solution is colourless and not more than 0.1 ml of 0.1 M sodium
hydroxide is required to change the colour of the solution to
Storage. Store protected from light and moisture.
pink.
Light absorption. When examined in the range 240 nrn to
280 nm (2.4.7), a 2.0 per cent w/v solution in 2,2,4-trimethyl-
Hard Paraffin pentane shows an absorption of not more than 0.1.
HardParaffin is a purified mixture of solid hydrocarbons Readily carbonisable substances. Place 5 ml in a dry, heat-
obtained from petroleum or from shale oil. resistant glass-stoppered test-tube (125 mm x 18 mm)
Category. Pharmaceutical aid(stiffening agent). previously rinsed with chromic acidsolution, then with water
and dried. Add 5 ml of nitrogen-free sulphuric acid
Description. A white or colourless, translucent mass, (containing 94.5 per cent to 95.5 per cent w/w ofHzS04), insert
frequently showing a crystalline structure; odourless even the stopper and shake as vigorously as possible in the
when freshly cut; slightly greasy to the touch. Bums with a longitudinal direction of the tube for 5 seconds. Loosen the
luminous flame. When melted, the liquid is free from stopper, immediately place the tube in a bath ofboiling water,
fluorescence by daylight. supporting it so as to prevent contact of the tube with the
bottom or side ofthe bath and heat for 10 minutes. At the end
Tests
of the second, fourth, sixth, and eighth minutes, remove the
Acidity or alkalinity. To 10.0 g add 20 ml ofboiling water, heat tube from the bath and shake as vigorously as possible in the
in a water-bath for 5 minutes, shake vigorously for I minute, longitudinal direction ofthe tube for 5 seconds. At the end of
cool, allow to separate and filter the aqueous layer. To 10 ml of 10 minutes from the time the tube was placed in the bath
the filtrate add 0.1 rnl of phenolphthalein solution. The remove the tube and allow to stand for 10 minutes. The lower
solution is colourless and not more than 0.1 ml of 0:1 M sodium acid layer is notmore intensely coloured than a mixture oD ml
hydroxide is required to change the colour of the solution to ofFCS, 1.5 ml ofCCS and 0.5 ml ofCSS (2.4.1), overlaid with
pink 5 ml ofliquid paraffin. lithe sulphuric acid remains dispersed
in the molten paraffin, the colour ofthe emulsion is not darker
Congealing range (2.4.10). 50° to 65°.
than that of the standard mixture when shaken vigorously.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Solid paraffins. Place a suitable quantity, previously dried by
heating at 100° for 2 hours and cooled in a desiccator over
sulphuric acid, in a glass cylindrical vessel having an internal
Liquid Paraffin diameter ofapproximately 25 mm. Close the vessel and immerse
in a mixture of ice and water; after 4 hours the liquid is
White Mineral Oil; Liquid Petrolatum sufficiently clear that a black line, 0.5 mm in width, held vertically
Liquid Paraffin is a pmified mixture of liquid hydrocarbons behind the vessel is easily seen.
obtained from petroleum to which not more than 10 ppm of Sulphur compounds. Mix 4 ml with 2 ml of ethanol (95 per
. tocopherol or of butylated hydroxytoluene may be added. cent), and 2 drops of a clear, saturated solution of lead
Category. Laxative; faecal softener. monoxide iii sodium hydroxide solution and heat at 70° for

1862

www.webofpharma.com
IP 2010 WHITE SOFT PARAFFIN

10 minutes with frequent shaking; the mixture remains dispersed in the molten paraffin, the colour ofthe emulsion is
colourless. not darker than that of the standard mixture when shaken
Storage. Store protected from light. vigorously.
Solid paraffins. Place a suitable quantity, previously dried by
heating at 100° for 2 hours and cooled in a desiccator over
Light Liquid Paraffin sulphuric acid, in a glass cylindrical vessel having an internal
diameter ofapproximately 25 mm. Close the vessel and immerse
Light Mineral Oil; Light Liquid Petrolatum in a mixture of ice and water; after 4 hours the liquid is
sufficiently clear that a black line, 0.5 mm in width, held vertically
Light Liquid Paraffin is a purified mixture ofliquid saturated
behind the vessel is easily seen.
hydrocarbons obtained from petroleum. It may contain a
suitable stabiliser. Sulphur compounds. Mix 4 ml with 2 ml of ethanol (95 per
cent), and 2 drops of a clear, saturated solution of lead
Category. Pharmaceutical aid (vehicle).
monoxide in sodium hydroxide solution and heat at 70° for
Description. A transparent, colourless, oily liquid, free from 10 minutes with frequent shaking; the mixture remains
fluorescence by daylight; almost odourless when cold. colourless.

Tests Storage. Store protected from light.

Weight per ml (2.4.29).0.820 g to 0.880 g.


Dynamic viscosity (2.4.28). 25 mPas to 80 mPas, determined at Liquid Paraffin Emulsion
20° ±l ° by method B or Viscosity (2.4.28). 10 cps to 40 cps,
determined at 30° ±l ° by method C using LVI spindle at 30 Liquid Paraffm Oral Emulsion
rpm. Liquid Paraffin Emulsion is an oral emulsion ofLiquid Paraffin
Acidity or alkalinity. To 10.0 g add 20 ml ofboiling water, heat in Purified Water.
in a water-bath for 5 minutes, shake vigorously for 1 minute, Liquid Paraffin Emulsion contains not less than 44.0 per cent
cool, allow to separate and filter the aqueous layer. To 10 ml of and not more than 49.0 per cent w/w ofliquid paraffin.
the filtrate add 0.1 ml of phenolphthalein solution. The
Category. Laxative; faecal softener.
solution is colourless and not more than 0.1 ml of 0.1 M sodium
hydroxide is required to change the colour of the solution to Dose. 10 to 30 m!.
pink.
Tests
Light absorption. When examined in the range 240 nm to 280
nm (2.4.7), a 2.0 per cent w/v solution in 2,2,4-trimethylpentane Other tests. Complies with the tests stated under Oral Liquids.
shows an absorption of not more than 0.1. Assay. Weigh accurately about 5.0 g, add 10 ml ofwater, extract
Readily carbonisable substances. Place 5 ml in a dry, heat- with two quantities, each of40 ml, ofa mixture of2 volumes of
resistant glass-stoppered test-tube (125 mm x 18 mm) ethanol (95 per cent), 3 volumes of light petroleum (40° to
previously rinsed with chromic acid solution, then with water 60°) and 3 volumes of ether and then with 30 ml ofa mixture of
and dried. Add 5 ml of nitrogen-free sulphuric acid equal volumes of lightpetroleum (40° to 60°) and ether. Wash
(containing 94.5 per cent to 95.5 per cent w/w ofH 2S04), insert the combined extracts with 15 ml of 0.5 M sodium hydroxide
the stopper and shake as vigorously as possible in the and then with 15 ml of water, evaporate the solvent, add 5 ml
longitudinal direction of the tube for 5 seconds. Loosen the of acetone and evaporate again. Repeat the addition and
stopper, immediately place the tube in a bath ofboiling water, evaporation of acetone until the residue is free from water,
supporting it so as to prevent contact of the tube with the dry at 105° for 15 minutes and weigh.
bottom or side ofthe bath and heat for 10 minutes. At the end Storage. Store protected from moisture.
of the second, fourth, sixth, and eighth minutes, remove the
tube from the bath and shake as vigorously ssas possible in
the longitudinal direction of the tube for 5 seconds. At the
end of 10 minutes from the time the tube was placed in the White Soft Paraffin
bath remove the tube and allow to stand for 10 minutes. The
lower acid layer is not more intensely coloured than a mixture
White Petroleum Jelly
of3 ml ofFCS, 1.5 ml ofCCS and 0.5 ml ofCSS (2.4.1), overlaid White Soft Paraffin is a purified, semi-solid mixture of
with 5 ml of liquid paraffin. If the sulphuric acid remains hydrocarbons obtained from petroleum and bleached.

1863

www.webofpharma.com
WHITE SOFT PARAFFIN IP 2010

Category. Pharmaceutical aid (ointment base). the test substance at a spot 25 mID to 38 mm from the edge of
the container. Adjust the zero setting and quickly release the
Description. A white, translucent, soft unctuous mass,
plunger, then hold it free for 5 seconds. Secure the plunger
retaining these characteristics on storage and when melted
and read the total penetration from the scale. Make three or
. and allowed to cool without stirring; not more than slightly
more trials, each so spaced that there is no overlapping ofthe
fluorescent by daylight, even melted; odour1ess when rubbed
areas of penetration. Where the penetration exceeds 20 mID,
on the skin.
use a separate container of the test substance for each trial.
Tests Read the penetration to the nearest 0.1 mm. Calculate the
average of the three or more readings and conduct further
Melting range (2.4.21).38° to 56°, determined by Method IV trials to a total of 10 if the individual results differ from the
Acidity or alkalinity. To 10.0 g add 20 ml ofboiling water, heat average by more than ± 3 per cent. The final average of the
in a water-bath for 5 minutes, shake vigorously for 1 minute, trials is not less than 10.0 mID and not more than 30.0 mID
cool, allow to separate and filter the aqueous layer. To 10 ml of indicating a consistency value between 100 and 300.
the filtrate add 0.1 ml of phenolphthalein solution. The Sulphated ash (2.3.18). Not more than 0.1 percent.
solution is colourless and not more than 0.1 m1 of 0.1 M sodium
hydroxide is required to change the colour of the solution to Storage. Store protected from light and moisture.
pink.
Light absorption (2.4.7). Absorbance o(a 0.05 per cent w/v
solution in 2,2,4-trimethylpentane at about 290 nm, not more Yellow Soft Paraffin
than 0.5. Yellow Petroleum Jelly
Fixed oils, fats and resin. Digest 109 with 50 ml of sodium Yellow Soft Paraffin is a purified, semi-solid mixture of
-hydroxide solution at 100° for 30 minutes and allow the hydrocarbons obtained from petroleum.
aqueous layer to separate. On acidifYing the aqueous layer
with dilute sulphuric acid, no precipitate or oily matter is Category. Pharmaceutical aid (ointment base).
produced. Description. A pale yellow to yellow, translucent, soft
Foreign organic matter. Volatilises when heated, without unctuous mass, retaining these characteristics on storage and
emitting an acrid odour. when melted and allowed to cool without stirring; not more
than slightly fluorescent by daylight, even melted; odourless
Consistency. 100 to 300, determined by the following method. when rubbed on the skin.
Apparatus. The apparatus is essentially in agreement with IS
4887: 1980 and comprises a penetrometer fitted with a polished
Tests
cone-shaped metal plunger weighing 150 g having a detachable Melting range (2.4.21).38° to 56°, determined by Method IV.
steel tip of the following dimensions. The tip of the cone has
Light absorption (2.4.7). Absorbance of a 0.05 per cent w/v
an angle of 30°, the point being truncated to a diameter of
solution in 2,2,4-trimethylpentane at about 290 nm, not more
0.38 ± 0.08 mID, the base ofthe tip is 8.38 ± 0.13 mID in diameter
than 0.75.
and the length ofthe tip is 15 ± 0.25 mID. The remaining portion
ofthe cone has an angle of 90°, is 28 to 29 mm in height, and Acidity or alkalinity. To 10.0 g add 20 ml ofboiling water, heat
has a maximum diameter of65.1 mID at the base. The containers in a water-bath for 5 minutes, shake vigorously for 1 minute,
of the test are flat-bottomed metal or glass cylinders that are cool, allow to separate and filter the aqueous layer. To 10 ml of
102 ± 6 mID in diameter and not less than 60 mm in height. the filtrate add 0.1 ml of phenolphthalein solution. The
solution is colourless and not more than 0.1 ml of 0.1 M sodium
Procedure. Melt a sufficient quantity at a temperature below hydroxide is required to change the colour of the solution to
85° and pour into one or more ofthe containers filling to within pink.
6 mID ofthe rim. Cool to 25°± 2.so over a period ofnot less than
16 hours, protected from drafts. Two hours before the test, Fixed oils, fats and resin. Digest 109 with 50 ml of sodium
place the containers in a water-bath at 25° ± 0.5°. If the room hydroxide solution at 100° for 30 minutes and allow the
temperature is below 23.5° or above 26.5°, adjust the aqueous layer to separate. On acidifYing the aqueous layer
temperature of the cone to 25° ± 0.5° by placing it in a water- with dilute sulphuric acid, no precipitate or oily matter is
bath. produced.
Foreign organic matter. Volatilises when heated, without
Without disturbing the surface of the substance under
emitting an acrid odour.
examination, place the container on the penetrometer table,
and lower the cone until the tip just touches the top surface of Consistency. 100 to 300, determined by the following method.

1864

www.webofpharma.com
IP 2010 PARALDEHYDE

Apparatus. The apparatus is essentially in agreement with IS Tests


4887.1980 and comprises a penetrometer fitted with a polished
Paraffin Ointment complies with the tests stated under
cone-shaped metal plunger weighing 150 g having a detachable
Ointments.
steel tip of the following dimensions. The tip of the cone has
an angle of 30°, the point being truncated to a diameter of
0.38 ± 0.08 rom, the base ofthe tip is 8.38 ± 0.13 rom in diameter
and the length ofthe tip is 15 ±0.25 rom. The remaining portion Paraldehyde
of the cone has an angle of 90°, is 28 to 29 mm in height, and
has a maximum diameter of65.1 rom at the base. The containers
of the test are flat-bottomed metal or glass cylinders that are
102 ± 6 mm in diameter and not less than ,60 mm in height.
Procedure. Melt a sufficient quantity at a temperature below
85° and pour into one or more ofthe containers filling to within
Mol. Wt. 132.7
6 rom ofthe rim. Cool to 25°± 2S over a period ofnot less than
16 hours, protected from drafts. Two hours before the test, Paraldehyde is 2,4,6-trimethyl-1 ,3,5-trioxane, the cyclic trimer
place the containers in a water-bath at 25° ± 0.5°. If the room of acetaldehyde. It may contain a suitable amount of
temperature is below 23.5° or above 26.5°, adjust the antioxidant.
temperature of the cone to 25° ± 0.5° by placing it in a water- Category. Anticonvulsant in status epilepticus; hypnotic;
bath. sedative.
Without disturbing the surface of the substance under
Dose. By deep intramuscular injection, as a single dose, 5 to
examination, place the container on the penetrometer table, 10 ml; by rectal injection, 5 to 10 ml, suitably diluted with
and lower the cone until the tip just touches the top surface of physiological saline.
the test substance at a spot 25 mm to 38 mm from the edge of
the container. Adjust the zero setting and quickly release the Description. A colourless or slightly yellow, transparent liquid;
plunger, then hold it free for 5 seconds. Secure the plunger odour, strong and characteristic. Solidifies at low temperature
and read the total penetration from the scale. Make three or to form a crystalline mass.
more trials, each so spaced that there is no overlapping of the Identification'
areas of penetration. Where the penetration exceeds 20 rom,
use a separate container of the test substance for each trial. A. Heat 5 ml with 0.1 ml of 1 M sulphuric acid; acetaldehyde,
Read the penetration to the nearest 0.1 mm. Calculate the recognisable by its odour, is evolved.
average of the three or more readings and conduct further B. To 5 ml ofa 10 per cent v/v solution add 5 ml ofammoniacal
trials to a total of 10 if the individual results differ from the silver nitrate solution in a test-tube and heat on a water-bath;
average by more than ± 3 per cent. The final average of the metallic silver is deposited as a mirror on the sidesofthe tube.
trials is not less than 10.0 rom and not more than 30.0 mm
C. A 10 per cent w/v solution in carbon dioxide-free water is
indicating a consistency value between 100 and 300.
clear (2.4.1), but becomes turbid on warming.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Tests
Storage. Store protected from light and moisture.
Congealing range (2.4.1 0). 10° to 13°.
Distillation range (2.4.8). Not more than 10 per cent distils
Paraffin Ointment below 123° and not less than 95 per cent distils below 126°.
Category. Pharmaceutical aid (ointment basis). Refractive index (2.4.27). 1.403 to 1.406.

White Beeswax 20 g Relative density (2.4.29).0.991 to 0.996.


Hard Paraffin 30 g Acetaldehyde. Shake 5 ml with a mixture of5 ml of ethanol
(60 per cent), 5 ml of hydroxylamine hydrochloride reagent
Cetostearyl Alcohol 50 g
in ethanol (60 per cent) and 2 drops of methyl orange solution
White Soft Paraffin· 900 g and titrate with 0.5 M sodium hydroxide to full yellow colour;
*May be replaced by Yellow Soft Paraffin ifother medicaments not more than 0.8 ml of 0.5 M sodium hydroxide is required.
to be incorporated are coloUred. Acidity. Mix 5 ml with 45 ml of carbon dioxide-free water and
Mix the ingredients, heat gently with stirring until titrate with 0.1 M sodium hydroxide using phenolphthalein
homogeneous and stir until cold. solution as indicator; not more than 1.5 ml is required.

1865

www.webofpharma.com
PARALDEHYDE IP 2010

Chlorides. To 5 ml ofa 1 per cent v/v solution add one drop of Identification
nitric acid and three drops of silver nitrate solution; no
opalescence is produced immediately. Test A may be omitted if test B, C and D carried out. Test D
may be omitted if test A, Band C are carried out.
Sulphates. To 5 ml ofa 1 per cent v/v solution add one drop of
hydrochloric acid and three drops of barium chloride A. Determine by thin layer chromatography (2.4.17), coating
solution; no turbidity is produced. the plate with silica gel G.
Peroxides. In a stoppered vessel, dissolve 5 ml in sufficient of Mobile phase. A mixture of 40 volumes of 1-butanol,
recently boiled and cooled water to produce 50 ml, add 5 ml of 10 volumes of glacial acetic acid and 10 volumes of water.
dilute sulphuric acid and 10 ml ofpotassium iodide solution. Test solution.. Dissolve 0.25 g of the substance under
Close the flask and set aside in the dark for 15 minutes. Titrate examination in 100 ml of wate]:
with 0.1 M sodium thiosulphate using starch solution as
indicator; set aside for 5 minutes and, if necessary, complete Reference solution. A 0.25 per cent w/v solution of
the titration. Not more than 2.0 ml of 0.1 M sodium penicillamine RS in water.
thiosulphate is required. Apply to the plate 2 III of each solution. Allow the mobile.
Non-volatile matter. Heat 5 ml in a small dish on a water-bath phase to rise 10 cm. Dry the plate at 105° for 5 to 10 minutes
and dry at 105° for 1 hour; the residue weighs not more than 3 and expose to iodine vapour for 5 to 10 minutes. The principal
mg (0.06 per cent w/v). spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with the
Storage. Store protected from moisture, in complete darkness
reference solution.
and at a temperature of8° to 15°. Ifsolidified, the whole ofthe
contents of the container should be liquified by warming B. Dissolve 0.5 g in a mixture of 0.5 ml of hydrochloric acid
before use. and 4 ml ofwarm acetone, cool in ice and scratch the inside of
the tube with a glass rod to initiate crystallisation; a white
NOTE - Do not use Paraldehyde if it has a brownish colour
precipitate is produced. Filter under vacuum, wash the
or an odour of acetic acid. Avoid contact with rubber and
precipitate with acetone and dry with suction. A 1 per cent
plastics.
w/v solution of the dried material is dextrorotatory.
Labelling. The label states (1) the nature and the proportion
of any antioxidant added; (2) that it may decompose on C. To 4 ml of a 1 per cent w/v solution add 2 ml of
standing to form potentially harmful substances. phosphotungstic acid solution and heat nearly to boiling; a
blue colour is produced.
D. In the testfor Penicillamine disulphide, the principal peak
Penicillamine in the chromatogram obtained with test solution (b)
corresponds to the peak due to penicillamine in the
D-Penicillamine chromatogram obtained with reference solution (a).
CH s 0 Tests
HsC--L - Jl
HS/ ~N~~H Appearance ofsolution. A 10.0 per cent w/v solution in carbon
dioxide-free water (solution A) is clear (2.4.1), and not more
CsHIlNOzS Mol. Wt. 149.2 intensely coloured than degree 6 of the appropriate range of
reference solutions (2.4.1).
Penicillamine is 3-mercapto-o-valine.
Penicillamine contains not less than 98.0 per cent and not pH (2.4.24). 4.5 to 5.5, determined ina 1.0percentw/v solution.
more than 101.0 per cent ofCsH II NOzS, calculated on the dried Specific optical rotation (2.4.22). -61.0° to-65.0°, determined
basis. in a 5.0 per cent w/v solution in 1 M sodium hydroxide.
Category. Chelating agent in copper and lead poisoning; Heavy metals (2.3.13).10 ml ofsolution A, complies with the
antirheumatoid arthritic. limit test for heavy metals, Method D (20 ppm). Use lead
Dose. In poisoning, 500 mg to 2 g daily, in divided doses or in standard solution (2 ppm Pb) to prepare the standard.
accordance with the needs of the patient. In rheumatoid Mercuric salts. Determine by atomic absorption
arthritis, initial dose, 125 to 250 mg daily before food, increased spectrophotometry (2.4.2), using a solution prepared in the
gradually every 4 to 12 weeks until remission occurs; usual following manner. To 1.0 g ofthe substance under examination
maintenance dose, 500 to 750 mg daily. add 10 ml of water and 0.15 ml of perchloric acid and swirl
Description. A white or almost white, crystalline powder. until dissolution is complete. Add 1 ml of ammonium

1866

www.webofpharma.com
IP 2010 PENICILLAMINE

pyrrolidinedithiocarbamate solution that has been washed sensitivity and formation of clearly defined inhibition zones
three times immediately before use, each time with an equal ofsuitable diameter. Immediately pour the inoculated medium
volume of 4-methyl-2-pentanone. Mix, add 2 ml of 4-methyl- into five Petri dishes (10 cm in diameter) to give uniform layers
2-pentanone, shake for 1 minute, dilute to 25 mlwith water, 2 to 5 mm in depth. Alternatively, the medium may consist of
allow the layers to separate and use the 4-methyl-2-pentanone two layers, only the upper layer being inoculated. Store the
layer. Measure the absorbance at 254 nm using a mercury dishes so that no appreciable growth or death of micro-
hollow-cathode lamp and an air-acetylene flame and setting organisms occurs before use and so that the surface of the
the zero using a 4-methyl-2-pentanone layer obtained by medium is dry at the time of use. In each dish, place five
repeating the procedure described above but omitting the stainless steel hollow cylinders (6 mm in diameter) on the
substance under examination. For the standard solution surface ofthe medium evenly spaced on a circle with a radius
dissolve 0.108 g of yellow mercuric oxide in the minimum of about 25 mm and concentric with the dish. For each dish,
volume of 2 M hydrochloric acid, add sufficient water to place in separate cylinders 0.15 ml of each of the following
produce 1000.0 ml and treat suitable volumes in the same manner five solutions.
as the solution ofthe substance under examination (10 ppm). For solution (1) dissolve LO g of the substance under
Penicillamine disolphide. Determine by liquid examination in 8 ml ofphosphate buffer pH 2.5, add 8 ml of
chromatography (2.4.14). ether and shake vigorously for 1minute. Repeat the extraction
Test solution (a). Dissolve 40 mg of the substance under and combine the ether layers. Add 8 ml ofphosphate buffer
examination in 5 ml ofthe mobile phase, add 1 ml ofa 0.0025 pH 2.5, shake for 1 minute, allow to settle and separate the
per cent w/v solution of sulphanilamide (internal standard) ether layer quantitatively, taking care to eliminate the aqueous
in the mobile phase and dilute to 10 ml with the mobile phase. phase completely. (Penicillin is unstable at pH 2.5; carry out
the operations at this pH within 6 to 7 minutes). Add 8 mlof
Test solution (b). Dissolve 40 mg of the substance under phosphate bufferpH 6.0, shake for 5 minutes, allow to settle,
examination in the mobile" phase and dilute to 10 ml with the separate the aqueous layer and check that the pH is 6.0. For
same solvent. solution (2) add 20 III of pr:micillinase solution to 2 ml of
Reference solution (a). A 0.4 per cent w/v solution of solution (1) and incubate at 37° for 1 hour. For solution (3)
penicillamine RS in the mobile phase. dissolve 5 mg of benzylpenicillin sodium in 500 ml of
Reference solution (b). Add 1 m1 oftest solution (a) to 1 m10f phosphate buffer pH 6.0 and dilute 0.25 mlofthis solution to
a 0.04 per cent w/v solution ofpenicillamine disulphide RS in 200 ml with phosphate bufferpH 2.5. Carry outthe extraction
the mobile phase and dilute to 10 ml with the mobile phase. procedure described under solution (1) using 8 ml of this
solution and beginning at the words "add 8 ml ofether...". For
Chromatographic system solution (4) add 20 ml of penicillinase solution to 2 ml of
- a stainless steel column 25 cm x 5 mm, packed with solution (3) and incubate at 37° for 1 hour. Prepare solution (5)
octylsilane bonded to porous silica (5 to 10 Ilm), in the same manner as solution (1) but omitting the substance
- mobile phase: an equal volume of 0.2 per cent w/v under examination.
solution of methanesulphonic acid and 0.01 per cent
w/v solution of disodium edetate, Maintain the dishes at 30° for at least 24 hours. Measure the
- flow rate. 2 ml per minute, diameters ofthe zones ofinhibition to within 0.1 mm. The test
- spectrophotometer set at 220 nm, is not valid unless solution (3) gives a clear zone of inhibition
injection volume. 20 Ill. and solutions (4) and (5) give no zones ofinhibition. Ifsolution
(1) gives a zone ofinhibition it is caused by penicillin provided
In the chromatogram obtained with test solution (a) the ratio solution (2) gives no zone of inhibition. If this is the case, the
of the area of any peak corresponding to penicillamine average diameter of the zones of inhibition given by solution
disulphide to the area of the peak due to the internal standard (1) for the five Petri dishes is less than that given by solution
is not greater than the corresponding ratio in the chromatogram (3)(0.1 ppm).
obtained with reference solution (b).
Nutrient medium
Penicillin. Carry out the following procedure in a penicillin-
Peptone 5 g
free atmosphere and with equipment reserved for the test.
Yeast extract L5g
Sterilise the equipment at 180° for 3 hours and the buffer
solutions at 121° for 20 minutes before use. Meat extract L5g
Sodium chloride 3.5g
LiquefY a suitable nutrient medium such as that described
below and inoculate at a suitable temperature with a culture of Agar 15 g
Micrococcus flavus (ATCC 9341) to give 5 x 104 micro- Distilled water 1000 ml
organisms per ml or a quantity necessary to obtain the required Adjust the pH to 6.0

1867

www.webofpharma.com
PENICILLAMINE TABLETS IP 2010

Penillic acid. Absorbance of a 0.2 per cent w/v solution at Penicillamine disulphide. Determine by liquid
about 268 run, not more than 0.07 (2.4.7)(about0.5 per cent). chromatography (2.4.14).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Test solution. Shake quantity of the powdered tablets
containing 40 mg of Penicillamine with 10 ml of the mobile
Loss on drying (2.4.19). Not morethan 0.5 per cent, determined
phase, filter and use the filtrate.
on 1.0 g by drying in an oven at 60° over phosphoruspentoxide
at a pressure not exceeding 0.7 kPa. Reference solution. A 0.004 per cent w/v solution of
penicillamine disulphide RS in the mobile phase.
Assay. Dissolve 0.1 gin 30 ml of anhydrous glacial acetic
acid Titrate with 0.1 M perchloric acid, determining the end- Chromatographic system
point potentiometrically (2.4.25). Carry out a blank titration. a stainless steel column 25 cm x 5 mm, packed with
octylsilane bonded to porous silica (5 to 10 /lm),
1 ml of 0.1 M perchloric acid is equivalent to 0.01492 g of
mobile phase: an equal volume of 0.2 per cent w/v
CsH11NOzS.
solution of methanesulphonic acid and 0.01 per cent
Storage. Store protected from moisture. w/v solution of disodium edetate,
flow rate. 2 ml per minute,
- spectrophotometer set at 220 run,
- injection volume. 20 /ll.
Penicillamine Tablets
In the chromatogram obtained with the test solution the area
D-Penicillamine Tablets of any peak corresponding to penicillamine disulphide is not
Penicillamine Tablets contain not less than 95.0 per cent and greater than the area ofthe principal peak in the chromatogram
not more than 105.0 per cent of the stated amount of obtained with the reference solution (1.0 per cent).
penicillamine, CsH11NOzS. The tablets are coated. Other tests. Complies with the tests stated under Tablets.
Usual strengths. 50 mg; 125 mg; 250 mg. Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing 0.1 g of Penicillamine,
Identification
dissolve as completely as possible in 50 ml of water and filter.
A. Shake a quantity ofthe powdered tablets containing 20 mg Add to the filtrate 5 ml of1 M sodium hydroxide and 0.2 ml of
ofPenicillamine with 4 ml of water and filter. Add to the filtrate a 0.1 per cent w/v solution of dithizone in ethanol (95 per
2 ml of phosphotungstic acid solution and allow to stand for cent) and titrate with 0.02 M mercuric nitrate.
5 minutes; a blue colour is produced.
1 ml of 0.02 M mercuric nitrate is equivalentto 0.005968 gof
B. Dissolve a quantity of the powdered tablets containing CsH11NOzS.
10 mg ofPenicillamine in 5 ml of water and add 0.3 ml of5 M Storage. Store protected from moisture.
sodium hydroxide and 20 mg of ninhydrin; an intense blue or
violet-blue colour is produced immediately.

Tests Diluted Pentaerythritol Tetranitrate


Mercuric salts. Disperse a quantity of the powdered tablets
containing 1 g ofPenicillamine in 10 ml of water in a stoppered
flask, add 0.2 ml of9 M perchloric acid and swirl to dissolve.
Add 1 ml of ammonium pyrrolidinedithiocarbamate solution,
mix, add 2 ml of 4-methyl-2-pentanone, shake for 1 minute and
add sufficient water to produce 25 ml. Detennine by atomic
absorption spectrophotometry (2.4.2), using a mercury hollow-
cathode lamp and an air-acetylene flame and setting the zero
using a 4-methyl-2-pentanone layer obtained by repeating Diluted Pentaerythritol Tetranitrate is a dry mixture of2,2-
the procedure described above but omitting the substance bis(hydroxymethyl)propane-l ,3-diol tetranitrate with
under examination, measuring at 254 run. Use mercury solution Lactose or Mannitol or a mixture of Lactose and Starch or
AAS, suitably diluted with water, for the standard solutions, any other suitable inert excipients which permit safe
adjusted to contain the same concentrations of9 M perchloric handling.
acid, ammonium pyrrolidinedithiocarbamate solution and Diluted Pentaerythritol Tetranitrate contains not less than
4-methyl-2-pentanone as the solution under examination 95.0 per cent and not more than 105.0 per cent of the stated
(40 ppm). amount ofpentaerythritol tetranitrate, CsHgN40IZ'

1868

www.webofpharma.com
IP 2010 DILUTED PENTAERYTHRlTOL TETRANITRATE

Category. Antianginal. prepared potassium iodide and starch solution and examine
at 254 urn. The spot due to nitrate in the chromatogram obtained
Dose. The equivalent of 20 to 60 mg of pentaerythritol
with the test solution is not more intense than the spot in the
tetranitrate, three to four times daily.
chromatogram obtained with the reference solution (0.5 per
Description. A white or almost white, powder; odour, faint cent, calculated as potassium nitrate).
and mild.
Related substances. Determine by liquid chromatography
(2.4.14).
Identification
Test solution (a). Shake about 25 mg of the substance under
A. Transfer a quantity of powder containing 10 mg of examination in 20 ml of the mobile phase for 15 rp.inutes and
pentaerythritol tetranitrate to a medium porosity sintered-glass dilute to 25.0 ml with the mobile phase, filter.
filter, add 5 ml of dry acetone and collect the filtrate. Repeat
with two further quantities, each of 5 ml, of dry acetone and Test solution (b). Dilute 1.0 ml of test solution (a) to 10.0 ml
evaporate the combined filtrate at a temperature not exceeding with the mobile phase.
60°, with the aid of a gentle current of air, and dry the residue Reference solution (a). Dissolve a quantity of diluted
at 60° for 4 hours; the residue melts at 138° to 142°(2.4.21). pentaerythritol tetranitrate RS containing about 25 mg of
B. Suspend 10 mg ofthe residue obtained in test A in a mixture Pentaerythritol Tetranitrate in 20 ml of the mobile phase,
of2 ml of sulphuric acid and 1 ml of water; cool and carefully sonicate for 15 minutes and dilute to 25.0 ml with the mobile
overlay with 3 ml offerrous sulphate solution; a reddish brown phase.
colour is produced at the interface of the two liquids. Reference solution (b). Dilute 1.0 ml ofreference solution (a)
C. Determine by thin-layer chromatography (2.4.17), coating to 10.0 ml with the mobile phase.
the plate with silica gel GF254. Reference solution (c). Dilute 0.3 ml ofreference solution (b)
Mobile phase. A mixture of20 volumes of ethyl acetate and 80 to 10.0 ml with the mobile phase.
volumes of toluene. Reference solution (d). Dilute 200 III of glyceryl trinitrate
Test solution. Dissolve a quantity of the substance under solution RS to 25.0 ml with the mobile phase.
examination containing about 10 mg of Pentaerythritol Reference solution (e). To 1 ml of reference solution (b), add
Tetranitrate in 10 ml of ethanol (95 per cent), filter. 1 ml of reference solution (d) and dilute to 10.0 ml with the
Reference solution. Shake a quantity of diluted mobile phase.
pentaerythritol tetranitrate RS containing about 10 mg of Reference solution (f). Dilute 1.0 ml ofreference solution (a)
Pentaerythritol Tetranitrate with 10 ml of ethanol (95 per to 20.0 ml with the mobile phase. Dilute 0.5 ml ofthis solution
cent). to 50.0 ml with the mobile phase.
Apply to the plate 10 III of each solution. Allow the mobile Chromatographic system
phase to rise 8 cm. Dry the plate in air and examine at 254 urn, - a stainless steel column 15 cm x 3.9 mm, packed with
The principal spot in the chromatogram obtained with the test octylsilane bonded to porous silica (5 11m),
solution corresponds to that in the chromatogram obtained - mobile phase: a mixture of 35 volumes of water and 65
with the reference solution. volumes of acetonitrile,
- flow rate. 1.4 ml per minute,
Tests - spectrophotometer set at 220 urn,
Impurity A. Determine by thin-layer chromatography (2.4.17), - injection volume. 20 Ill.
coating the plate with silica gel G. Inject reference solution (e). The test is not valid unless the
Mobile phase. A mixture of 15 volumes of glacial acetic acid, resolution between the peaks due to glyceryl trinitrate and
30 volumes of acetone and 60 volumes of toluene. pentaerythritol tetranitrate is not less than 3.0. The relative
retention time with reference to pentaerythritol tetranitrate for
Test solution. Dissolve about 0.1 g of the substance under
pentaerythritol tetranitrate impurity B is about 0.7 and for
examination in 5.0 ml of ethanol (95 per cent), filter.
pentaerythritol tetranitrate impurity C is about 0.3.
Reference solution. Dissolve about 10 mg ofpotassium nitrate
Inject test solution (a), reference solution (c) and (t). Run the
in I ml of water and dilute to 100 ml with ethanol (95 per
chromatogram 5 times the retention time ofthe principal peak.
cent).
In the chromatogram obtained with test solution (a) the area
Apply to the plate 10 III of each solution. Allow the mobile ofthe peak due to pentaerythritol tetranitrate impurity C is not
phase to rise 8 cm. Dry the plate in air and spray with freshly more than the area ofthe principal peak in the chromatogram

1869

www.webofpharma.com
PENTAERYTHRITOL TETRANlTRATE TABLETS IP 2010

obtained with reference solution (c) (0.3 per cent), the area of Transfer a portion of the mixture to a glass-stoppered
any other secondary peak is not more than not more than centrifuge tube and centrifuge at 1500 rpm for 5 minutes.
twice the area of the principal peak in the chromatogram Transfer 2.5 ml of the supernatant solution to a 100-ml
obtained with reference solution (t) (0.1 per cent). The sum of volumetric flask and evaporate at 35° with the aid ofa current
all the secondary peaks is not more than twice the area of the of air to dryness. To the residue add 1.0 m1 of glacial acetic
principal peak in the chromatogram obtained with reference acid and swirl to dissolve. Add 2 ml of phenoldisulphonic
solution (c) (0.6 per cent). Ignore any peak with an area less acidsolution, mix and allow to stand for 5 minutes. Add 25 ml
than the area of the principal peak in the chromatogram ofwater and 10 ml ofstrong ammonia solution, cool, dilute to
obtained with reference solution (t) (0.05 per cent). volume with water and mix. Measure the absorbance of the
Heavy metals (2.3.13). 2.0 g complies with limit test for heavy resulting solution at the maximum at about 409 nm (2.4.7),
metals, Method B (10 ppm). using water as the blank.
Weigh accurately 0.130 g of potassium nitrate, previously
Assay. Determine by liquid chromatography (2.4.14), as
dried at 105° for 4 hours, dissolve in 3 ml ofwater, dilute with
described in the test for Related substances.
sufficient glacial acetic acid to produce 200.0 ml and mix
Inject test solution (b) and reference solution (b). well. Using 1.0 ml of this solution repeat the procedure
Calculate the content ofCsHsN4012. beginning at the words "Add 2 ml ofphenoldisulphonic acid
solution,.......".Calculate the content ofCsHsN4012 in the tablet.
Storage. Store protected from light and moisture.
1 ml ofthe potassium nitrate solution is equivalent to 0.000503
NOTE - Undilutedpentaelythritol tetranitrate is a powerfUl g ofCsHsN40'2'
explosive. It can be exploded with percussion or excessive Other tests. Comply with the tests stated under Tablets.
heat. Great care and appropriate precautions should be
Assay. Weigh and powder 20 tablets. Weigh accurately a
taken in handling and only exceedingly small amounts should
quantity of the powder containing about 50 mg of
be isolated.
pentaerythritol tetranitrate and transfer to a 100-m1 volumetric
Labelling. The label states the percentage content of flask with the aid of about 30 ml of acetone. Add sufficient
pentaerythritol tetranitrate. acetone to produce 50 ml and warm on a water-bath at a
temperature not exceeding 60° and boil gently, with occasional
swirling, for 5 minutes. Cool, dilute to volume with acetone
and mix. Transfer a portion ofthe mixture to a glass-stoppered
Pentaerythritol Tetranitrate Tablets centrifuge tube and centrifuge at 1500 rpm for 5 minutes.
Pentaerythritol Tetranitrate Tablets contain not less than Transfer 1.0 ml of the supernatant solution to a 100-ml
90.0 per cent and not more than 110.0 per cent of the stated volumetric flask and evaporate at 35° with the aid ofa current
amount ofpentaerythritol tetranitrate, CsHsN 40 12 • of air to dryness. To the residue add 1.0 ml of glacial acetic
acid and swirl to dissolve. Add 2 ml of phenoldisulphonic
Usual strengths. 10 mg; 30 mg.
acidsolution, mix and allow to stand for 5 minutes. Add 25 ml
Identification ofwater and 10 ml ofstrong ammonia solution, cool, dilute to
volume with water and mix. Measure the absorbance of the
A. Transfer a quantity of powder containing 10 mg of
resulting solution at the maximum at about 409 urn (2.4.7),
pentaerythritol tetranitrate to a medium porosity sintered-glass
using water as the blank.
filter, add 5 ml of dry acetone and collect the filtrate. Repeat
Weigh accurately 0.13 g ofpotassium nitrate, previously dried
with two further quantities, each of 5 ml, of dry acetone and
evaporate the combined filtrate at a temperature not exceeding at 105° for 4 hours, dissolve in 3 ml of water, dilute. with
60°, with the aid of a gentle current of air, and dry the residue sufficient glacial acetic acid to produce 200.0 mland mix
at 60° for 4 hours; the residue melts at 138° to 142° (2.4.21). well. Using 1.0 ml of this solution repeat the procedure
beginning at the words "Add 2 ml ofphenoldisulphonic acid
Tests solution,.......".
Uniformity of content. (For tablets containing 10 mg or less) Calculate the content of CsHsN40 12 from the values of the
- Comply with the test stated under Tablets. absorbances so obtained.
1 ml of the potassium nitrate solution is equivalent to
Crush one tablet and transfer to a 50-ml volumetric flask with
0.000503 g ofCsHsN40 12 •
the aid of 15 m1 ofacetone. Add sufficient acetone to produce
25 ml, heat the mixture on a water-bath at a temperature not Storage. Store protected from light and moisture.
exceeding 60° and boil gently, with occasional swirling, for Labelling. The label states the strength in terms of the
5 minutes. Cool, dilute to volume with acetone and mix. equivalent amount of pentaerythritol tetranitrate.

1870

www.webofpharma.com
IP 20ra PENTAMIDINE ISETHIONATE

Pentamidine Isethionate and dilute to 50 ml with water. Dilute 1.0 ml ofthe solution to
50.0 ml with the mobile phase.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 ilm),
- mobile phase: a mixture of65 volumes of methanol and
Mol. Wt. 592.7 35 volumes of 3 per cent w/v solution of ammonium
acetate, adjusted to pH 7.5 with triethylamine,
Pentamidine Isethionate is 4,4' -[pentane-l ,5-diylbis(oxy)] - flow rate. 1 ml per minute,
bisbenzenecarboximidamide di(2-hydroxyethanesulphonate). - spectrophotometer set at 265 nm,
Pentamidine Isethionate contains not less than 98.5 per cent - injection volume. 10 ill.
and not more than 101.0 per cent ofC19H24N402, 2C2H 60 4S, Inject reference solution (b). The test is not valid unless the
calculated on the dried basis. resolution between the 2 principal peaks is not less than 2.0.
Category. Antiprotozoal. Inject the test solution and reference solution (a). Run the
Dose. By intramuscular injection, 3 to 4 mg per kg body weight chromatogram 3.5 times the retention time of the principal
daily for 10 days, maximum 200 mg. peale In the chromatogram obtained with the test solution,
the area of any secondary peak is not more than the area of
Description. A white or almost white powder or crystals;
the principal peak in the chromatogram obtained with reference
odourless or almost odourless; hygroscopic.
solution (a) (0.2 per cent), the sum of all the secondary peaks
is not more than twice the area of the principal peak in the
Identification
chromatogram obtained with reference solution (a) (0.4 per
A. Determine by infrared absorption spectrophotometry (2.4.6). cent). Ignore any peak with an area less than 0.1 times the area
Compare the spectrum with that obtained with pentamidine of the principal peak in the chromatogram obtained with
isethionate RS or with the reference spectrum ofpentamidine reference solution (a) (0.02 per cent).
isethionate. Ammonium isethionate. To 1.0 g in a test-tube (about 4 cm in
B. When examined in the range 230 om to 360 nm (2.4.7), a diameter) add 10 ml of water and 20 ml of 1 M sodium
0.001 per cent w/v solution in 0.01 Mhydrochloric acid shows hydroxide. Immediately attach a bung carrying a splash head
an absorption maximum only at about 262 om; absorbance at and an aspirator tube (about 5 mm in diameter).. Connect the
about 262 om, about 0.46). splash head to two test-tubes in series, each containing 20 ml
of 0.01 M sulphuric acid. Heat the tube containing the
C. To 10 ml ofa 0.05 per cent w/v solution add 1 ml ofa 0.1 per substance under examination in a water-bath at 45° to 50° and,
cent w/v solution of glyoxal sodium bisulphite and 1 ml of a maintaining this temperature, draw a current ofair, previously
solution prepared by dissolving 4 g of boric acid in a mixture passed through 1 M sulphuric acid, through the liquids in a
of 27 ml of 1 M sodium hydroxide and sufficient water to series of tubes for 3 hours at such a rate thatthe bubbles are
produce 100 ml. Heat on a water-bath for 10 minutes; a magenta just too rapid to count. Titrate the combined solutions from
colour is produced. the two absorption tubes with 0.02 M sodium hydroxide using
methyl red-methylene blue solution as indicator; not less than
Tests 36.5 ml of 0.02 M sodium hydroxide is required.
pH (2.4.24). 4.5 to 6.5, determined in a 5.0 percentw/v solution. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Related substances. Determine by liquid chromatography Loss on drying (2.4.19). Not more than 4.0 per cent, determined
(2.4.14). on 1.0 g by drying in an oven at 105°.
Test solution. Dissolve about 0.1 g of the substance under Assay. Dissolve 0.250 g in 50 ml of dimethylformamide. Add
examination in 100.0 ml ofthe mobile phase. 0.25 ml of thymol blue solution and titrate with 0.1 M
tetrabutylammonium hydroxide, under a current of nitrogen,
Reference solution (a). Dilute 2.0 ml of the test solution to
until the colour of the indicator changes to blue. Carry out a
100.0 ml with the mobile phase. Dilute 1.0 ml ofthis solution to
blank titration.
10.0 ml with the mobile phase.
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
Reference solution (b). To 0.1 g in a conical flask, add 40 ml of
water and glass beads, adjusted to pH 10.5 with dilute sodium 0.02963 g ofC23H36N401OS2.
hydroxide solution and boil under reflux for 20 minutes. Cool Storage. Store protected from moisture.

1871

www.webofpharma.com
PENTAMIDINE INJECTION IP 2010

Pentamidine Injection Reference solution (b). Dissolve 0.1 g of substance under


examination in 40 ml of water, adjusted to pH 10.5 with 2 M
Pentamidine Isethionate Injection sodium hydroxide, heat under a reflux condenser for 20
Pentamidine Injection is a sterile material consisting of minutes, cool and dilute to 50 ml with water. Dilute 1.0 m1 of
Pentamidine Isethionate with or without buffering agents and this solution to 50.0 ml with the mobile phase.
other excipients. It is filled in a sealed container. Chromatographic system
The injection is constituted by dissolving the contents of the a stainless steel column 25 cm x 4.6 mm packed with
sealed container in the requisite amount of sterile Water for octadecylsilane bonded to porous silica (5 f!m) (Such
Injections, immediately before use. as Spherisorb ODS 1), .
mobile phase: a mixture of 130 volumes of methanol and
The constituted solution complies with the requirements for
70 volumes of3 per cent w/v solution of ammonium
Clarity of solution and Particulate matter stated under
acetate, adjusted to pH 7.5 with triethylamine,
Parenteral Preparations (Injections).
flow rate. 1 ml per minute,
Storage. The constituted solution should be used immediately spectrophotometer set at 265 nm,
after preparation but, in any case, within the period
injection volume. 10 f!l.
recommended by the manufacturer.
Inject reference solution (b). The test is not valid unless the
Pentamidine Injection contains not less than 95.0 per cent and
resolution between the two principal peaks is not less than
not more than 105.0 per cent of the stated amount of
2.0.
pentamidine isethionate, C19Hz4N40z, 2CzH 60 4S.
Usual strength. 200 mg. Inject the test solution and reference solution (a). Run the
chromatogram 3.5 times the retention time of the principal
The contents of the sealed container comply with the
peak. In the chromatogram obtained with the test solution the
requirements stated under Parenteral Preparations
area of any secondary peak is not more 0.2 times the area of
(Powders for Injection) and with thefollowing requirements.
the principal peak in the chromatogram obtained with reference
solution (a) (0.2 per cent) and the sum of the areas of all the
Identification
secondary peaks is not more than 0.4 times the area of the
A. Determine by infrared absorption spectrophotometry (2.4.6). principal peak in the chrotnatogram obtained with reference
Compare the spectrum with that obtained with pentamidine solution (a) (0.4 per cent).
isethionate RS or with the reference spectrum ofpentamidine
isethionate. Ammonium isethionate. To 1.0 g in a test-tube (about 4 cm in
diameter) add 10 ml of water and 20 ml of 1 M sodium
B. When examined in the range 230 nm to 360 nm (2.4.7), a hydroxide. Immediately attach a bung carrying a splash head
0.001 per cent w/v solution in 0.01 M hydrochloric acid shows and an aspirator tube (about 5 mm in diameter). Connect the
an absorption maximum only at about 262 nm; absorbance at splash head to two test-tubes in series, each containing 20 ml
about 262 nm, about 0.46. of 0.01 M sulphuric acid. Heat the tube containing the
C. To 10ml ofaO.05 per cent w/v solution add 1 m1 ofaO.1 per substance under examination in a water-bath at 45° to 50° and,
cent w/v solution of glyoxal sodium bisulphite and 1 ml of a maintaining this temperature, draw a current ofair, previously
solution prepared by dissolving 4 g of boric acid in a mixture passed through 1 M sulphuric acid, through the liquids in a
of 27 ml of 1 M sodium hydroxide and sufficient water to series of tubes for 3 hours at such a rate that the bubbles are
produce 100 ml. Heat on a water-bath for 10 minutes; a magenta· just too rapid to count. Titrate the combined solutions from
colour is produced. the two absorption tubes with 0.02 M sodium hydroxide using
methyl red-methylene blue solution as indicator; not less than
Tests 36.5 ml of 0.02 M sodium hydroxide is required.

pH (2.4.24). 4.5 to 6.5, determined in a5.0 per centw/v solution. Assay. Dissolve 0.25 g ofthe mixed contents of 10 containers
in 50 ml of dimethylformamide. Titrate with 0.1 M
Related substances. Determine by liquid chromatography
tetrabutylammonium hydroxide, determining the end-point
(2.4.14).
potentiometrically (2.4.25). Carry out a blank titration.
Test solution. Dissolve 0.1 g of substance under examination
in 100.0 ml ofthe mobile phase. 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
0.02963 g ofC,9Hz4N40z, 2CzH 60 4S.
Reference solution (a). Dilute 1 ml ofthe test solution to 100
ml with the mobile phase. Storage. Store in single dose containers.

1872

www.webofpharma.com
IP 2010 PENTAZOCINE HYDROCHLORIDE

Pentazocine Reference solution (b). A 0.01 per cent w/v solution of the
substance 'under examination in chloroform.
Reference solution (c). A 0.005 per cent w/v solution of the
substance under examination in chloroform.
Apply to the plate 10 III of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Heat the plate at 105° for 15 minutes, allow to cool, expose to
iodine vapour and re-examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the
Mol. Wt. 285.4
chromatogram obtained with reference solution (a); not more
Pentazocine is (2RS, 6RS, llRS)-6, II-dimethyl-3-(3-methylbut- than one such spot is more intense than the spot in the
2-enyl)-1,2,3,4,5,6-hexahydro-2,6-methano-3-benzazocin-8-01. chromatogram obtained with reference solution (b) and not
Pentazocine contains not less than 98.0 per cent and not more more than four such spots are more intense than the spot in
than 101.0 per cent ofC I9H27NO, calculated on the dried basis. the chromatogram obtained with reference solution (c).

Category. Narcotic analgesic. Sulphated ash (2.3.18). Not more than 0.1 percent.

Dose. By subcutaneous, intramuscular or intravenous Loss on drying (2.4.19). Not more than 1.0 per cent, determined
injection, 30 to 60 mg every 3 to 4 hours. on 1.0 g by drying in an oven at 60° at a pressure not exceeding
0.7 kPa for 4 hours.
Description. A white or pale cream powder.
Assay. Weigh accurately about 0.6 g, dissolve in 50 ml of
Identification anhydrous glacial acetic acid and titrate with 0.1 M perchlpric
acid, using crystal violet solution as indicator. Carry out a
A Determine by infrared absorption spectrophotometry (2.4.6).
blank titration.
Compare the spectrum with that obtained with pentazocine
RS or with the reference spectrum ofpentazocine. 1 ml of 0.1 Mperchloric acid is equivalent to 0.02854 g of
C I9H27NO.
B.To 1 mg in a porcelain crucible add 0.5 ml of a solution of
sulphuric acid containing 1 per cent w/v solution of Storage. Store protected from light and moisture.
ammonium molybdate; an intense blue colour is produced
which changes to bluish green, green and finally, on standing,
yellow.
Pentazocine Hydrochloride
C. Dissolve 5 mg in 5 ml ofsulphuric acid, add 0.05 mlofferric
chloride solution and mix; a yellow colour is produced which
deepens slightly in intensity on warming. On the addition of
0.05 ml ofnitric acid the yellow colour is unchanged.

Tests
Light absorption (2.4.7). To 0.1 g add 20 ml ofwater and 10 ml
of 1 M hydrochloric acid, shake to dissolve and add sufficient
water to produce 100 ml. Dilute 10 ml to 100 ml with water. The
absorbance of the resulting solution at the maximum at about
278 nm, 0.67 to 0.71. C I9H27NO,HCl Mol. Wt. 321.9

Related substances. Determine by thin-layer chromatography Pentazocine Hydrochloride is (2RS,6RS, llRS)-6, 11-
(2.4.17), coating the plate with silica gel HF254. dimethyl-3-(3-methylbut-2-enyl)-1,2,3,4,5,6-hexahydro-2,6-
methano-3-benzazocin-8-01 hydrochloride.
Mobile phase. A mixture of 94 volumes of chloroform,
3 volumes of 2-propylamine and 3 volumes of methanol. Pentazocine Hydrochloride contains not less than 98.0 per
cent and not more than 101.0 per cent of C I9 H27NO,HCl,
Test solution. Dissolve 0.2 g of the substance under
calculated on the dried basis.
examination in 10 ml ofchloroform.
Category. Narcotic analgesic.
Referencesolution (a). A 0.02 per cent w/v solution of the
substance under examination in chloroform. Dose. 25 to 100 mg, every 3 to 4 hours after food.

1873

www.webofpharma.com
PENTAZOCINE HYDROCHLORIDE IP 2010

Description. A white or pale cream-coloured, crystalline Loss on drying (2.4.19). Not more than 1.0 per cent, determined
powder; odourless. The material exhibits polymorphism. on 1.0 g by drying in an oven at 100° at a pressure not exceeding
0.7kPa.
Identification Assay. Weigh accurately about 0.5 g, dissolve in 50 ml of
A. Determine by infrared absorption spectrophotometry (2.4.6). anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
Compare the spectrum with that obtained with pentazocine acid, using crystal violet solution as indicator.· Carry out a
hydrochloride RS or with the reference spectrum of blank titration.
pentflzocine hydrochloride. 1 ml of 0.1 M perchloric acid is equivalent to 0.03219 g of
B. To 1 mg in a porcelain crucible add 0.5 ml ofa solution of C I9H27NO,HCl.
sulphuric acid containing 1 per cent w/v solution of Storage. Store protected from light and moisture.
ammonium molybdate; an intense blue colour is produced
which changes to bluish green, green and finally, on standing,
yellow.
C. Gives reaction A ofchlorides (2.3.1).
Pentazocine Tablets
Pentazocine Hydrochloride Tablets
Tests
Pentazocine Tablets contain not less than 92.5 per cent and
pH (2.4.24). 4.0 to 6.0, determined ina 1.0 per centw/v solution. not more than 107.5 per cent of the stated amount of
pentazocine hydrochloride, C I9H 27NO,HCl.
Light absorption (2.4.7). Dissolve 0.1 g in 10 ml of 1 M
hydrochloric acid and add sufficient water to produce Usual strength. 25 mg.
100 ml; dilute 10 ml to 100 ml with water. Absorbance ofthe
resulting solution at the maximum at about 278 urn, 0.59 to Identification
0.63. A. Shake a quantity of the powdered tablets containing 0.1 g
Related substances. Determine by thin-layer chromatography of Pentazocine Hydrochloride with 10 ml of water, filter, add
(2.4.17), coating the plate with silica gel HF254. 1 ml of1 M sodium hydroxide and shake the resulting solution
with 20 ml of chloroform. Wash the chloroform extract with
Mobile phase. A mixture of 94 volumes of chlorofor.m, 5 ml of water, dry over anhydrous sodium sulphate and filter.
3 volumes of 2-propylamine and 3 volumes of methanol. Evaporate the chloroform using a rotary evaporator and dry
Test solution. Dissolve 0.2 g o·f the substance under the oily residue at a temperature not exceeding 25° at a pressure
examination in 10 ml of chloroform. of2 kPa for 1 hour.

Reference solution (a). A 0.02 per cent w/v solution of the On the residue, determine by infrared absorption
substance under examination in chloroform. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with pentazocine RS or with the reference spectrum
Reference solution (b). A 0.01 per cent w/v solution of the of pentazocine.
substance under examination in chloroform.
B. To a quantity of the powdered tablets containing 50 mg of
Reference solution (c). A 0.005 per cent w/v solution of the Pentazocine Hydrochloride add 70 ml of water, shake for
substance under examination in chloroform. 15 minutes, add sufficient water to produce 100 ml and filter.
To 10 ml ofthe filtrate add 10 ml of1 M sodium hydroxide and
Apply to the plate 10 III of each solution. After development,
sufficient water to produce 100 ml. When examined in the
dry the plate in air and examine in ultraviolet light at 254 urn.
range 230 nm to 360 urn (2.4.7), the resulting solution shows
Heat the plate at 105° for 15 minutes, allow to cool, expose to
absorption maxima at about 238 nm and 298 nm.
iodine vapour and re-examine in ultraviolet light at 254 urn.
Any secondary spot in the chromatogram obtained with the C. Shake a quantity ofthe powdered tablets containing 25 mg
test solution is not more intense than the spot in the ofPentazocine Hydrochloride with 5 ml of water and 0.5 ml of
chromatogram obtained with reference solution (a); not more 2 M nitric acid for 1 minute and filter. The filtrate gives reaction
than one such spot is more intense than the spot in the A ofchlorides (2.3.1).
chromatogram obtained with reference solution (b) and not
more than four such spots are more intense than the spot in Tests
the chromatogram obtained with reference solution (c).
Related substances. Determine by thin-layer chromatography
Sulphated ash (2.3.18). Not more than 0.1 per cent. (2.4.17), coating the plate with silica gel HF254.

1874

www.webofpharma.com
IP 2010 PENTAZOCINE LACTATE

Mobile phase. A mixture of 94 volumes of chloroform, Pentazocine Lactate contains not less than 98.5 per cent and
3 volumes of 2-propylamine and 3 volumes of methanol. not more than 101.0 per cent ofCI9H27NO,C3H603, calculated
on the dried basis.
Test solution. Shake a quantity of the powdered tablets
containing 0.2 g ofPentazocine Hydrochloride with 10 ml of Category. Narcotic analgesic.
0;1 M methanolic ammonia for 10 minutes, centrifuge and
Dose. By subcutaneous, intramuscular or intravenous
use the supernatant liquid.
injection, the equivalent ono to 60 mg ofpentazocine every 3
Reference solution (a). Dilute 1 volume of test solution to to 4 hours.
100 volumes with the same solvent.
Description. A white or pale cream-coloured, crystalline
Reference solution (b). Dilute 1 volume of test solution to powder; odourless.
200 volumes with the same solvent.
Identification
Reference solution (c). Dilute 1 volume of test solution to
400 volumes with the same solvent. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with pentazocine
Apply to the plate 10 fll of each solution. After development,
lactate RS or with the reference spectrum of pentazocine
dry the plate in air and examine in ultraviolet light at 254 nm.
lactate.
Heat the plate at 105° for 15 minutes, allow to cool, expose to
iodine vapour and re-examine in ultraviolet light at 254 nm. B. To 1 mg in a porcelain crucible add 0.5 ml of a solution of
Any secondary spot in the chromatogram obtained with the sulphuric acid containing 1 per cent w/v solution of
test solution is not more intense than the spot in the ammonium molybdate; an intense blue colour is produced
chromatogram obtained with reference solution (a), not more which changes to bluish green, green and finally, on standing,
than one such spot is more intense than the spot in the yellow.
chromatogram obtained with reference solution (b) and not C. Gives reaction A oflactates (2.3.1).
more than four such spots are mo~e intense than the spot in
the chromatogram obtained with reference solution (c). Tests
Other tests. Complies with the tests stated under Tablets. pH (2.4.24). 5.5 to 6.5, determined in a 1.0percentw/v solution.
Assay. Weigh and powder 20 tablets. Weigh accurately a Light absorption (2.4.7). Dissolve 0.1 g in 10 ml of 1 M
quantity ofthe powder containing about 25 mg ofPentazocine hydrochloric acid and add sufficient water to produce
Hydrochloride, shake with 100 ml ofwater for 15 minutes, add 100 ml; dilute 10 ml to 100 ml with water. Absorbance ofthe
2.5 ml of 1 M hydrochloric acid and sufficient water to resulting solution at the maximum at about 278 nm, 0.50 to
produce 250.0 ml and filter. Measure the absorbance of the 0.54.
filtrate at the maximum at about 278 nm (2.4.7). Calculate the
content ofC I9H27NO,HCl taking 61.2 as the specific absorbance Related substances. Determine by thin-layer chromatography
at278nm. (2.4.17), coating the plate with silica gel HF254.

Storage. Store protected from light and moisture. Mobile phase. A mixture of 94 volumes of chloroform,
3 volumes of 2-propylamine and 3 volumes of methanol.
Test solution. Dissolve 0.2 g of the substance under
Pentazocine Lactate examination in 10 ml ofchloroform.

HO» . I

H,C"'. CH N-"'=t,
Reference solution (a). A 0.02 per cent w/v solution of the
substance under examination in chloroform.
Reference solution (b). A 0.01 per cent w/v solution of the
substance under examination in chloroform.
Reference solution (c). A 0.005 per cent w/v solution of the
substance under examination in chloroform.
3 CH 3
Apply to the plate 25 fll of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Mol. Wt. 375.4
Heat the plate at 105° for 15 minutes, allow to cool, expose to
Pentazocine Lactate is (2RS, 6RS, llRS)-6, ll-dimethyl-3-(3- iodine vapour and re-examine in ultraviolet light at 254 nm.
methylbut-2-enyl)-1,2,3,4,5,6-hexahydro-2,6-methano-3- Any secondary spot in the chromatogram· obtained with the
benzazocin-8-ol1actate. test solution is not more intense than the spot in the

1875

www.webofpharma.com
PENTAZOCINE INJECTION IP 2010

chromatogram obtained with reference solution (a); not more C. To a volume containing 30 mg ofpentazocine add 2 ml of
than one such spot is more intense than the spot in the 0.1 M sodium hydroxide, extract with 2 ml of chloroform and
chromatogram obtained with reference solution (b) and not evaporate 0.1 ml of the chloroform extract to dryness in a
more than four such spots are more intense than the spot in porcelain crucible. Add to the residue 0.5 ml of a 1 per cent
the chromatogram obtained with reference solution (c). w/v solution of ammonium molybdate in sulphuric acid; an
intense blue colour is produced which changes to bluish green,
Su.phated ash (2.3.18). Not more than 0.1 per cent.
green and finally, on standing, yellow.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 60° at a pressure not exceeding Tests
0.7 kPa for 16 hours.
pH (2.4.24). 4.0 to 5.0.
Assay. Weigh accurately about 0.75 g of the substance under
examination, dissolve in 50 ml of anhydrous glacial acetic Related substances. Determine by thin-layer chromatography
acid. Titrate with 0.1 M perchloric acid, using crystal violet (2.4.17), coating the plate with silica gel HF254.
solution as indicator. Carry out a blank titration.
Mobile phase. A mixture of 94 volumes of chloroform,
1 ml of 0.1 M perchloric acid is equivalent to 0.03754 g of 3 volumes of 2-propylamine and 3 volumes of methanol.
CI9H27NO,C3H603.
Test solution. Dilute a volume ofthe injection with sufficient
Storage. Store protected from light and moisture. ethanol (95 per cent) to produce a solution containing
2.0 per cent w/v solution ofpentazocine.
Reference solution (a). Dilute 1 volume ofthe test solution to
Pentazocine Injection 100 volumes with ethanol (95 per cent).
Pentazocine Lactate Injection Reference solution (b). Dilute 1 volume ofthe test solution to
Pentazocine Injection is a sterile solution in Water for 200 volumes with ethanol (95 per cent). .
Injections ofeither Pentazocine Lactate or pentazocine lactate
Reference solution (c). Dilute 1 volume ofthe test solution to
prepared by the interaction of Pentazocine and Lactic Acid.
400 volumes with ethanol (95 per cent).
Pentazocine Injection contains not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of Apply to the plate 10 III of each solution. After development,
pentazocine, C I9 H27NO. dry the plate in air and examine in ultraviolet light at 254 nm.
Heat the plate at 105° for 15 minutes, allow to cool, expose to
Usual strength. The equivalent of30 mg of pentazocine per iodine vapour and re-examine in ultraviolet light at 254 urn.
ml; the equivalent of60 mg ofpentazocine per ml. Any secondary spot in the chromatogram obtained with the
Description. A clear, colourless or almost colourless liquid. test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a), not more
Identification than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b) and not
A. To a volume containing 90 mg ofpentazocine add 5 ml of
more than four such spots are more intense than the spot in
0.1 M sodium hydroxide and shake the resulting solution
the chromatogram obtained with reference solution (c).
with 5 ml of chloroform. Wash the chloroform extract with 2 ml
of water, dry over anhydrous sodium sulphate and filter. Other tests. Complies with the tests stated under Parenteral
Evaporate the chloroform without applying heat and dry the Preparations (Injections).
oily residue at a temperature not exceeding 25° and at a pressure
of2 kPa for 1 hour. Assay. To an accurately measured volume containing about
0.15 g of pentazocine add sufficient water to produce
On the residue, determine by infrared absorption 100.0 ml. To 5.0 ml add 1 ml of 1 M hydrochloric acid and
spectrophotometry (2.4.6). Compare the spectrum with that sufficient water to produce 100.0 ml. Measure the absorbance
obtained with pentazocine RS or with the reference spectrum ofthe resulting solution atthe maximum at about 278 urn (2.4.7).
ofpentazocine (form B). Calculate the content of C I9H27NO, taking 69 as the specific_
B. Dilute a volume containing 30 mg ofpentazocine to 100 ml absorbance at 278 urn.
with water. To 10 ml add 10 ml of 1 M sodium hydroxide and Storage. Store protected from light.
sufficient water to produce 100 ml. When examined in the
range 230 urn to 360 urn (2.4.7), the solution shows absorption. Labelling. The label states the strength in terms of the
maxima, at about 238 urn and 298 nm. equivalent amount ofpentazocine in a suitable dose-volume.

1876

www.webofpharma.com
IP 2010 PENTOBARBITONE TABLETS

Pentobarbitone Sodium Test solution. A 2.0 per cent w/v solution of the substance
under examination.
Soluble Pentobarbitone; Pentobarbital Sodium
Reference solution. A 0.01 per cent w/v solution of the
substance under examination.
Do not ignore any spot remaining on the line of application.
Free pentobarbitone. Not more than 3.5 per cent, determined
by the following method. Weigh accurately about 2.0 g and
dissolve in 75 ml of dimethylformamide, heating gently if
necessary. Add 0.25 ml of a 1 per cent w/v solution of thymol
blue in dimethylformamide and titrate with 0.1 M sodium
Mol. Wt. 248.3 methoxide to a blue end-point. Carry out a blank titration.
Pentobarbitone Sodium is sodium 5-ethyl-5-[(IRS)-I- 1 ml of 0.1 M sodium methoxide is equivalent to 0.02263 g of
methylbutyl]barbiturate. pentobarbitone.
Pentobarbitone Sodium contains not less than 99.0 per cent Isomer. Dissolve 0.3 gin 5 ml of a 5 per cent w/v solution of
and not more than 101.5 per cent ofC\\HI7N2Na03, calculated anhydrous sodium carbonate and add 0.3 g of 4-nitrobenzyl
on the dried basis. bromide dissolved in 10 ml of ethanol (95 per cent). Heat
Category. Hypnotic., under a reflux condenser for 30 minutes, cool to 25° scratch
the side of the vessel with a glass rod if necessary to induce
Dose. 100 to 200 mg.
crystallisation and filter. Wash the residue with five quantities,
Description. A white, crystalline powder or granules; each of 5 ml, of water. Transfer the residue as completely as
hygroscopic. possible to a small flask, add 25 ml of ethanol (95 per cent)
and heat under a reflux condenser for 10 minutes; the solid
Identification dissolves completely. Cool to 25° and scratch the side of the
flask with a glass rod to induce crystallisation. Filter, wash the
A. To 10 mlofa 10 per cent w/v solution add 5 mlof2 M acetic
residue with two quantities, each of 5 ml, of water and dry at
acid; a white, crystalline precipitate is produced. Filter, wash
105° for 30 minutes. The dried residue melts at 136° to 148°
the precipitate with water and dry at 105°. Determine the
(2.4.21).
melting point ofthe dried precipitate (2.4.21). Mix equal parts
ofthe dried precipitate and phenobarbitone RS and determine Heavy metals (2.3.13). 0.67 g complies with the limit test for
the melting point (2.4.21). The difference between the melting heavy metals, Method B (30 ppm).
points (which are about 131°) is not greater than 2°.
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
B. Complies with the test for identification of barbiturates on 1.0 g by drying in an oven at 105°.
(2.3.1), using the dried precipitate obtained in test A for
Assay. Weigh accurately about 0.4 g, dissolve in 25 ml of a
preparing the test solution.
12.75 per cent w/v solution of silver nitrate in pyridine and
.C. To 10 mg add 10 mg of vanillin and 2 ml of sulphuric acid, titrate with 0.1 M ethanolic sodium hydroxide using 0.5 mlof
mix and heat on a water-bath for 2 minutes; a reddish-brown thymolphthalein solution as indicator, until a pure blue colour
colour is produced. Cool and add 5 ml of ethanol; the colour is obtained. Carry out a blank titration.
changes to violet and then blue.
1 ml of 0.1 Methanolic sodium hydrOXide is equivalent to
D. Ignite 1 g; the residue gives reaction A of sodium salts 0.02483 g ofC\\HI7N2Na03'
(2.3.1).
Storage. Store protected from moisture.
Tests
Appearance of solution. Prepare freshly a 10.0 per cent w/v
solution in carbon dioxide-free water (solution A). Solution Pentobarbitone Tablets
A is clear (2.4.1). Pentobarbitone Sodium Tablets; Pentobarbital Sodium
pH (2.4.24). 9.6 to 11.0, determined in solution A. Tablets
Related substances. Complies with the test for related Pentobarbitone Tablets contain not less than 92.5 per cent
substances in barbiturates (2.3.4), but applying 10 III of each and not more than 107.5 per cent of the stated amount of
of the following solutions. pentobarbitone sodium, C\\HI7N2Na03.

1877

www.webofpharma.com
PENTOBARBITONE TABLETS IP 2010

Usual strength. 100 mg. Pepsin


Identification Pepsin is obtained from the gastric mucosa of pigs, cattle or
sheep. It contains gastric proteinases that are active in an
A. Shake a quantity of the powdered tablets containing 0.1 g acid medium, pH 1 to 5. It may contain a suitable diluent such
of Pentobarbitone Sodium with 10 ml of a 10 per cent w/v
as Lactose.
solution ofpyridine and filter. Add to the filtrate 1 ml ofcupric
sulphate with pyridine solution and set aside for 10 minutes; Pepsin has an activity equivalent to its ability to digest not
a reddish violet precipitate is produced. less than 3000 times its weight of coagulated egg albumin
when determined by the method given under Assay.
B. Shake a quantity of the powdered tablets containing 0.1 g
of Pentobarbitone Sodium with 10 ml of water and filter. To Category. Proteolytic enzyme.
the filtrate add 2 ml of hydrochloric acid; a white precipitate Dose. 300 mg to 1 g.
is produced (distinction from pentobarbitone).
Description. A white or light buff-coloured, crystalline or
C. The residue obtained in the Assay melts at 127° to amorphous powder or translucent scales; odour, faint and
130°(2.4.21). meaty but not rancid; hygroscopic.
D. The powdered tablets, when moistened with hydrochloric
acid and introduced on a platinum wire into a flame, impart a Identification
yellow colour to the flame. A. Place 1 ml of congo redfibrin on a filter paper and wash
until a colourless filtrate is obtained with a solution prepared
Tests by diluting 30 ml of 1 M hydrochloric acid to 1000 ml with
Isomer. Dissolve a quantity ofthe powdered tablets containing water and adjusting the pH 1.5 to 1.7. Perforate the filter paper
0.3 g of Pentobarbitone Sodium in 5 ml of a 5 per cent w/v and wash the congo red fibrin through it with 20ml of the
solution of anhydrous sodium carbonate and add 0.3 g of same hydrochloric acid solution. Shake this suspension before
4-nitrobenzyl bromide dissolved in 10 ml of ethanol (95 per use. Dissolve about 10 mg ofthe substance under examination
cent). Heat under a reflux condenser for 30 minutes, cool to in 2 ml ofthe hydrochloric acid solution and adjust the pH 1.5
25° scratch the side of the vessel with a glass rod ifnecessary to 1.7. Place 4 ml ofthe congo red fibrin suspension in each of
to induce crystallisation and filter. Wash the residue with five two tubes. To one of the tubes add 1 m1 ofthe solution of the
quantities, each of 5 ml, of water. Transfer the residue as substance under examination and to the other tube add 1 mlof
completely as possible to a small flask, add 25 ml of ethanol water (control solution). Mix the contents of each tube and
(95 per cent) and heat under areflux condenser for 10 minutes; place ina water~bathat 25° with gentle shaking for 15 minutes;
filter the hot solution. Cool to 25° and scratch the side of the the control solution is eoi()urless and the solution of the
flask with a glass rod to induce crystallisation. Filter, wash the substance under examination is violet blue.
residue with two quantities, each of 5 ml, of water and dry at B. The proteolytic activity of a solution in water is destroyed
105° for 30 minutes. The dried residue melts at 136° to 148° at once by boiling. It is destroyed by warming for 10 minutes
(2.4.21). at 40° at a pH of8.0.
Other tests. Complies with the tests stated under Tablets.
Tests
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.3 g of Microbial contamination (2.2.9). 1 g is free from Escherichia
Pentobarbitone Sodium, dissolve as completely as possible coli and 109 is free from Salmonellae.
in 10 ml of a 2 per cent w/v solution of sodium hydroxide, Sulphated ash (2.3.18). Not more than 5.0 per cent.
saturate with sodium chloride, acidify with hydrochloric acid
Loss on drying (2.4.19). Not more than 5.0 per cent, determined
and extract with successive quantities, each of 15 ml, of ether
on 1.0 g by drying in an oven at 60° at a pressure not exceeding
until complete extraction is effected. Wash the combined
0.7 kPa for 4 hours.
extracts with two quantities, each of2 ml, ofwater and extract
the combined washings with 10 ml of ether. Add the ether to Assay. Weigh accurately 0.25 g, triturate with 1.0 g ofsodium
the main ether layer, filter and wash the filter with ether. chloride, add slowly acidified water prepared by diluting
Evaporate the solvent and dry the residue to constant weight 65 ml of1 M hydrochloric acidto 1000 ml with water, continue
atl05°. the trituration, dilute to 1000.0 ml with the acidified water and
shake for 15 minutes. Prepare coagulated egg albumin by
1 g ofresidue is equivalent to 1.097 g ofCIIHI7N2Na03'
boiling fresh hen-eggs in water for 15minutes, cooling rapidly
Storage. Store protected from moisture. to room temperature by immersion in cold water, separating

1878

www.webofpharma.com
IP 2010 PERlTONEAL DIALYSIS SOLUTIONS

the whites and rubbing through a no. 44 sieve. Reject the first Unless otherwise justified and authorised, antioxidants such
portion that passes through the sieve and triturate 15.0 g of as metabisulphite salts are not added to the solutions.
freshly prepared coagulated egg albumin with 50 ml of the Description. Clear, colourless or faintly straw-coloured
acidified water ensuring that the particles of egg albumin are solutions.
thoroughly disintegrated, add a further 50 ml of the acidified
water and keep in a water-bath at 51 0± 10 for 15 minutes. Add Identification
20.0 ml of the prepared solution of the substance under
A. To 5 ml of the solution under examination, add 2 ml of
examination and digest at 51 0 ± 10 for 4 hours, shaking at
dilute sodium hydroxide solution and 0.05 ml of potassium
intervals of 15 minutes. Centrifuge and decant offmost ofthe
cupri-tartrate solution; the solution remains blue and clear.
clear supernatant liquid, wash the remainder into a 10-ml
Heat to boiling; a copious red precipitate is formed.
graduated cylinder and allow to stand for 30 minutes. The
volume of the undissolved albumin is not more than 2 ml. B. 20 ml gives reactions of chlorides, sodium salts, potassium
salts and calcium salts (2.3.1).
Storage. Store protected from moisture.
C. To 5 ml add 1 ml of hydrochloric acid in a test-tube fitted
with a stopper and a bent tube, heat and collect a few ml ofthe
distillate. The distillate gives reaction C of acetates (2.3.1).
Peritoneal Dialysis Solutions D. ToO.l ml of titan yellow solution add lOmlofwater,2mlof
Intraperitoneal Dialysis Fluids the solution under examination and 1 mt of 1 M sodium
hydroxide; a pink colour is produced if magnesium salts are
Peritoneal Dialysis Solutions are sterile preparations for present.
intraperitoneal use containing electrolytes with a concentration
close to the electrolytic composition ofplasma. They contain E. Lactates and bicarbonates are identified together with ,the
dextrose in varying concentrations and/or other suitable Assay for lactate and bicarbonate.
osmotic agents. They do not contain antioxidants such as Tests
metabisulphite salts.
Appearance of solution. The solution under examination is
Peritoneal Dialysis Solutions contain not less than 97.5 per clear (2.4.1), and not more intensely coloured than reference
cent and not more than 102.5 per cent ofthe stated amount of solution YS4 (2.4.1).
sodium, Na, not less than 95.0 per cent and not more than
105.0 per cent ofthe stated amounts ofpotassium, K, calcium, pH (2.4.24). 4.5 to 6.5. Ifthe solution contains bicarbonate, 6.5
Ca, magnesium, Mg, chloride, Cl, acetate, C2H3 0 2 , lactate, to 8.0.
C3H;03, sodium bicarbonate, NaHC03, bicarbonate, HC0 3and 5-Hydroxymethylfurfural and Related substances. Dilute a
dextrose,·C6H 120 6• volume containing 1.0 g of Dextrose to 250.0 ml with water
and measure the absorbance of the resulting solution (2.4.7)
Usual strengths: Several formulations are used. The
at the maximum at about 284 urn; absorbance at'about 284 urn,
concentrations of the components per litre of solution are
not more than 0.25.
usually in the following range.
Aluminium. Adjust the pH of 400 ml of the solution under
Concentration in examination to pH 6.0 and add 10 ml of acetate bufferpH 6.0.
mmolllitre Extract the resulting solution with successive quantities of
Sodium 125-150 20, 20 and 10 ml of a 0.5 per cent w/v solution of
Potassium 0-4.5 8-hydroxyquinoline in chloroform and dilute the combined
Calcium 0-2.5 extracts to 50.0 ml with chloroform. Use as the blank a mixture
Magnesium 0.25- 1.5 of lO ml of acetate bufferpH 6. 0 and 100 ml of water treated in
Acetate and/or the same manner and as the standard solution a mixture of
Lactate and/or 2.0 ml of aluminium standard solution (2 ppm AI), 10 ml of
Bicarbonate 30-60 acetate buffer pH 6.0 and 90 ml of water treated in the same
CWoride manner. Measure the fluorescence of the test solution (II), of
90-120
the standard solution (12) and ofthe blank (13), (2.4.5), using an
Dextrose 25-250
excitation wavelength of392 nm and a secondary ftlterwith a
When bicarbonate is present, the solution of sodium transmission band centred at 518 urn, or a monochromator set
bicarbonate is supplied in a separate container or a separate to transmit at this wavelength. The fluorescence of the teSt
compartment and is added to the electrolyte solution solution (I, - 13) is not greater than that ofthe standard solution
immediately before use. (I2 - 13),

1879

www.webofpharma.com
PERITONEAL DIALYSIS SOLUTIONS IP 2010

Particulate contamination (2.5.9). Carry out the test using solution as indicator until a reddish yellow colour is produced.
50 ml ofthe solution under examination. Carry out a blank titration.
The preparation meets the requirements ofthe test ifit contains 1 ml of 0.1 M silver nitrateisequivalent to 0.003545 g oftota1
particles within the maximum limits shown below. chloride, calculated as Cl.
Particle size in /lm Maxirnumnumber For acetate (if present) - Determine by liquid
(Equal to or larger than) ofparticles per ml chromatography (2.4.14).
10 25 Test solution. Dilute an accurately measured volume of the
preparation under examination quantitatively with water to
25 3
obtain a solution containing about 1.0 mg of acetate per ml.
Other tests. Complies with the tests stated under Parenteral Reference solution. Dissolve an accurately weighed quantity
Preparations (Injections). of sodium acetate in water to obtain a solution having a known
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin concentration of about 0.12 per cent w/v of sodium acetate.
Unitperml. Chromatographic system
Pyrogens (2.2.8). Solutions for which a validated test for a stainless steel column 30 cm x 7.8 mm, packed with a
bacterial endotoxins cannot be carried out, comply with the strong cation-exchange resin consisting of sulphonated
test for pyrogens, injecting 10 ml of the solution per kg ofthe cross-linked styrene-divinylbenzene copolymer in the
rabbit's body weight. hydrogen form (7 /lm),
mobile phase: filtered and degassed 0.1 M sulphuric
Sterility (2.2.11). Complies with the test for sterility.
acid,
Assay. For sodium - Dilute suitably with water and determine flow rate. 0.8 ml per minute,
by Method A for flame photometry (2.4.4), or by Method A for column temperature. 60°,
atomic absorption spectrophotometry (2.4.2), measuring at spectrophotometer set at 210 Dill,
589 Dill and using sodium solution FP, or sodium solution injection volume. 20 Ill.
AAS respectively, suitably diluted with water for the standard
solutions. Inject the reference solution and record the chromatograms.
The test is not valid unless the tailing factor is not more than
For potassium - Dilute suitably with water and determine 2.0 and the relative standard deviation for replicate injections
by MethodA for flame photometry (2.4.4), or by Method A for is not more than 2.0 per cent.
atomic absorption spectrophotometry (2.4.2), measuring at
767 Dill and using potassium solution FP orpotassimn solution Iriject separately the test and standard solutions and record
AAS respectively, suitably diluted with water for the standard the chromatograms. Measure the responses forthe major peak
solutions. and calculate the content of acetate in the preparation under
examination.
For calcium - Dilute suitably with water and determine by
Method A for flame photometry (2.4.4), or by Method A for For lactate (ifpresent) - Determine by liquid chromatography
atomic absorption spectrophotometry (2.4.2), measuring at (2.4.14).
422.7 Dill and using calcium solution FP or calcium solution Test solution. Use the preparation under examination.
AAS respectively, suitably diluted with water for the standard
Reference solution(a). Dissolve an accurately weighed
solutions.
quantity of sodium lactate RS in water to obtain a solution
For magnesium - To 50.0 ml add 50 ml of water and 5 ml of having a lmown concentration of about 2 mg per ml.
strong ammonia"ammonium chloride solution and titrate with
Reference solution (b). Prepare a solution in water containing
0.005 M disodium edetate using 50 mg of eriochrome black
about 3 mg of anhydrous sodium acetate and 3 mg of sodium
T mixture as indicator.
lactate RS per ml.
1 ml of O. 005 M disodium edetate is equivalent to 0.1215 mg
Chromatographic system
of Mg.
a stainless steel column 10 cm x 4.6 mm, packed with
For total chloride - Dilute an accurately measured volume octadecylsi1ane chemically bonded to porous silica or
containing about 60 mg ofchloride to 50.0 ml with water. Add ceramic microparticles 3 to 10 /lm,
25.0 ml of 0.1 M silver nitrate and 2 ml of nitric acid. Filter, mobile phase: a filtered and degassed solution in water
wash the precipitate with water slightly acidified with nitric containing about I ml of formic acid and 1 ml of
acid and titrate the excess of silver nitrate with 0.1 M dicyclohexylamine per litre,
ammonium thiocyanate using ferric ammonium sulphate flow rate. 1 ml per minute,

1880

www.webofpharma.com
IP 2010 PERPHENAZINE

- spectrophotometer set at 210 nm, Titrate with 0.1 M sodium thiosulphate using starch solution
- injection volume. 20 /ll. as indicator. Carry out a blank titration using 25 ml of water.
Inject separately reference solutions (a) and (b), and record Calculate the content of anhydrous dextrose, C 6H 120 6, from
the chromatograms. The test is not valid unless the resolution the following Table.
between the peaks due to acetate and lactate is not less than Volume of Anhydrous dextrose
2.0, the tailing factor for the analyte peak is not more than 2.0 0.1 M sodium thiosulphate
and the, relative standard deviation for replicate injections is consumed (ml) (mg)
not more than 2.0 per cent.
8 19.8
Inject separately the test solution and reference solution (a), 9 22.4
and record the chromatograms. Measure the responses for 10 25.0
the major peak and calculate the content of lactate in the
11 27.6
preparation under examination.
12 30.3
For sodium bicarbonate - Titrate with 0.1 M hydrochloric 13 33.0
acid a volume ofthe preparation under examination containing 14 35.7
about 0.1 g of sodium bicarbonate, determining the end-point 15 38.5
potentiometrically (2.4.25). 16 41.3
1 ml of 0.1 M hydrochloric acid is equivalent to 8.40 mg of Storage. Peritoneal Dialysis Solutions are supplied in rigid or
NaHC03• semi-rigid plastic containers, in flexible plastic containers fitted
For lactate and bicarbonate - Determine by liquid with a special connecting device (these are generally pIled to
chromatography (2.4.14). a volume below their nominal capacity and presented in clo:>ed
Test solution. Use the preparation under examination. protective envelopes) or in glass containers. Store at a
temperature not exceeding 30°.
Reference solutions. Dissolve accurately weighed quantities
oflactates and bicarbonates in order to obtain solutions having CAUTION - Exposure to temperatures below 4° may cause
concentrations of about 90 per cent, 100 per cent and 110 per crystallisation and separation of solid particles rendering
cent ofthe claim in 100 ml of water. the preparation unsuitable for use.
Chromatographic system Labelling. The label states (1) the formula ofthe solution for
a stainless steel column 30 cm x 7.8 mm, packed with a peritoneal dialysis, expressed in grams per litre and in millimoles
cation-exchange resin (9 /lm), per litre; (2) the total osmolar concentration in mOsmol per
column temperature. 85°, litre; (3) the nominal volume ofthe solution in the container;
mobile phase: filtered and degassed 0.005 M sulphuric (4) that the solution is free from bacterial endotoxins, or where
acid, applicable, that it is apyrogenic; (5) that the solution is not to
- ,flow rate. 0.6 ml per minute, be used for intravenous infusion; (6) that any unused portion
differential refractometer detector, of the solution is to be discarded; (7) that the solution
injection volume. 20 /ll. containing visible particles should not be used; (8) the storage
Inject separately the test solution and the reference solutions conditions.
in duplicate, and record the chromatograms in the prescribed
conditions. The peaks elute in the following order, lactates,
then bicarbonates. Determine the concentration of lactates Perphenazine
and bicarbonates in the test solution by interpolating the peak
area for lactate and the peak height for bicarbonate from the
linear regression curve obtained with the solutions prepared
as reference solutions.
For dextrose - Transfer a volume of the preparation under
examination containing about 25 mg ofDextrose to a 250-ml
conical flask with a ground-glass neck and add 25.0 ml of
cupri-citric solution. Add a few grains of pumice, fit a reflux
condenser, heat so that boiling occurs within 2 minutes and
C:z 1H26ClN30S Mol. Wt. 404.0
boil for exactly 10 minutes. Cool and add 3 g of potassium
iodide dissolved in 3 ml of water. Carefully add, in small Perphenazine is 2-[4-[3-(2-chloro-l OH-phenothiazin-l0-
amounts, 25 rnl ofa 25 per cent w/w solution ofsulphuric acid. yl)propyl]piperazin-l-yl]ethanol.

1881

www.webofpharma.com
PERPHENAZINE IP 2010

Perphenazine contains not less than 99.0 per cent and not D. Melting range (2.4.21). 96° to 100°.
more than 101.0 per cent OfCZIHz6ClN30S, calculated on the
dried basis. Tests
Category. Dopamine receptor antagonist; neuroleptic. Appearance of solution. A 2.0 per cent w/v solution in
methanol is clear (2.4.1).
Description. A white or yellowish-white, crystalline powder.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Identification
NOTE-Prepare the solutions immediately before use.
Test A may be omitted iftests B, C and D are carried out. Tests
Band C may be omitted if tests A and D are carried out. Mobile phase. A mixture of 1 volume of concentrated
ammonia, 14 volumes of water and 85 volumes of butanol.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with perphenazine Test solution. Dissolve 0.1 g of the substance under
RS or with the reference spectrum of perphenazine. examination in 10 ml of methanol.

B. Determine by thin-layer chromatography (2.4.17), coating Reference solution. Dilute 0.5 ml ofthe test solution to 100 m1
the plate with kieselguhr G. with methanol.

Mobile phase. A mixture of 2 volumes of diethylamine and Apply to the plate 10 III of each solution. Allow the mobile
100 volumes of light petroleum, saturated with phase to rise 15 cm. Dry the plate in air and examine under
phenoxyethanol (add 6 volumes phenoxyethanol to 8 volumes ultraviolet light at 254 nm. Any secondary spot in the
ofthe above mixture until there is a persistent cloudiness after chromatogram obtained with the test solution is not more
shaking, decant, and use the supernatant liquid, even if it is intense than the spot in the chromatogram obtained with the
cloudy). reference solution.

Test solution. Dissolve 20 mg of the substance under Sulphated ash (2.3.18). Not more than 0.1 per cent.
examination in 10 ml of chloroform. Loss on drying (2.4.19). Not more than 0.5 percent, determined
Reference solution. A 0.2 per cent w/v solution of on 1.0 g by drying in vacuum at 65° for 4 hours.
perphenazine RS in chloroform. Assay. Weigh accurately about 0.15 g and dissolve in25 mlof
Impregnate the plate by placing it in a closed tank containing anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
the necessary quantity ofthe impregnation mixture containing acid, determining the end-point potentiometrically (2.4.25).
2.5 per cent v/v of phenoxyethanol and 7.5 per cent v/v of Carry out a blank titration.
formamide in acetone so that the plate dips about 5 mm beneath 1 ml of 0,1 M perchloric acid is equivalent to 0.0202 g of
the surface of the liquid. When the impregnation mixture has CZIHz6CIN30S.
risen at least 17 cm from the lower edge of the plate, remove
the plate and use immediately. Carry out the chromatography Calculate the content ofCzlHz6ClN30S.
in the same direction as the impregnation. Storage. Store protected from light.
Apply to the plate 2 III of each solution. Develop in the dark
and allow the mobile phase to rise 15 cm. Expose the plate to
ultraviolet light at 365 nm and examine after a few minutes. Perphenazine Tablets
The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained Perphenazine Tablets contain not less than 92.5 per cent and
with the reference solution. Dry the plate at 120° for 20 minutes, not more tha.n 107.5 per cent of the stated amount of
allow to cool and spray with a 10 per cent v/v solution of perphenazine, CZIHz6CIN30S.
sulphuric acid in ethanol (95 per cent). The principal spot in Usual strengths. 2 mg; 4 mg.
the chromatogram obtained with the test solution corresponds
to that in the chromatogram obtained with the reference Identification
solution. A. To a quantity of the powdered tablets containing 40 mg of
C. When examined in the range 230 Dill to 350 Dill (2.4.7), a Perphenazine, add 10 ml of water and 2 .ml of 1 M sodium
0.001 per cent w/v solution in methanol shows absorption hydroXide, shake and extract with 15 ml of ether. Wash the
maxima at 257 Dill and 313 Dill. The ratio of the absorbance ether layer with 5 ml of water, dry with anhydrous sodium
measured at the· maximum at 313 Dill to that measured at the sulphate and evaporate the ether to dryness. On the residue,
maximum at 257 nmis 0.120 to 0.128. determine by infrared absorption spectrophotometry (2.4:6).

1882

www.webofpharma.com
IP 2010 PETHIDINE HYDROCHLORIDE

Compare the spectrum with that obtained with pelphenazine disintegrated, heat on a water-bath for 3 minutes, swirling
RS or with the reference spectrum of perphenazine. continuously, cool, add 400 ml of ethanol (95 per cent), mix
with the aid ofultrasound for 2 minutes, shake for 5 minutes,
B. Extract a quantity ofthe powdered tablets containing 20 mg
dilute to 500 ml with ethanol (95 per cent) and filter through a
ofPerphenazine with 10 ml of chloroform, filter and evaporate
glass microfibre filter paper. Dilute the filtrate with ethanol
the filtrate to dryness. Dissolve the residue in 2 ml of methanol,
(95 per cent) to produce a solution containing 0.001 per cent
pour into a 4.0 per cent w/v solution ofpicric acid in methanol
w/v of Perphenazine. Record second-derivative ultraviolet
at 50 0 , cool and allow to stand for 4 hours. After recrystallisation
absorption spectra ofthe solutions in the range 210 to 290 nm
from methanol, the precipitate melts at about 248 0 (2.4.21)
(2.4.7). Measure the amplitude from the peak at 265 nm to the
with decomposition.
trough at 255 nm.
C. To a quantity of the powdered tablets containing 5 mg of
Calculate the content of C21H26N30S in the tablet from the
Perphenazine, add 5 ml of sulphuric acid and allow to stand
absorbance obtained by repeating the operation using
for 5 minutes. A red colour is produced.
pelphenazine RS in place ofthe substance under examination.
Tests For tablets containing 10 mg or less.
Related substances. Comply with the test for Related Use the average of the 10 individual results obtained in the
substances in Phenothiazines (2.3.5) with the following test for Uniformity of content.
modifications. .
Use mobile phase (c) and applying to the plate 50 III ofeach of
the following freshly prepared solutions. Pethidine Hydrochloride
Test solution. Extract a quantity of the powdered tablets
Meperidine Hydrochloride
containing 20 mg ofPerphenazine with 10 ml of ethanol (95
per cent) and filter.
Reference solution. Dilute 1 ml of the test solution to 200 ml CH 3
I
with the ethanol (95 per cent). N
Uniformity of content. (For tablets containing 10 mg or
less). Comply with the test stated under Tablets.
NOTE-Carry out the test protectedfrom light. o
Transfer one tablet to a 1OO-rnl volumetric flask and add 5 ml of
water, mix with the aid ofultrasound for 15 minutes or until the
tablet has completely disintegrated, heat on a water-bath for 3 ClsH2IN02,HCI Mol. Wt. 283.8
minutes, swirling continuously and cool. Add 50 ml of ethanol
(95 per cent), mix with the aid of ultrasound for 2 minutes, Pethidine Hydrochloride is ethyll-methyl-4-
shake for 5 minutes, dilute to 100 ml with ethanol (95 per cent) phenylpiperidine-4-carboxylate hydrochloride.
and filter through a glass microfibre filter paper. Dilute the Pethidine Hydrochloride contains not less than 99.0 per cent
filtrate with ethanol (95 per cent) to produce a solution and not more than 10 1.0 per cent ofCIsH21N0 2,HCl, calculated
containing 0.001 per cent w/v of Perphenazine. Record the on the dried basis.
second-derivative ultraviolet absorption spectra of the
Category. Narcotic analgesic.
resulting solution inthe range 210 to 290nm (2.4.7). Measure
the amplitude from the peak at 265 nm to the trough at 255 nm. Dose. Orally, 50 to 150 mg every 4 hours; by subcutaneous or
intramuscular injection, 25 to 100 mg every 4 hours; by slow
Calculate the content of C21H26N30S in the tablet from the
intravenous injection, 25 to 50 mg every 4 hours.
absorbance obtained by repeating the operation using
perphenazine RS in place ofthe substance under examination. Description. Colourless crystals or a white, crystalline powder.
Other tests. Comply with the tests stated under Tablets. Identification
Assay. For tablets containing more than 10 mg.
Test A may be omitted iftests B, C andD are carried out. Tests
NOTE-Carry out the test protectedfrom light. Band C may be omitted if tests A and D are carried out.
Shake 10 whole tablets with 50 ml of water and mix with the aid A. Determine by infrared absorption spectrophotometry (2.4.6).
ofultrasound for 15 minutes or until the tablets have completely Compare the spectrum with that obtained with pethidine

1883

www.webofpharma.com
PETHIDINE HYDROCHLORIDE IP 2010

hydrochloride RS or with the reference spectrum ofpethidine Time Mobile phase A Mobile phase B
hydrochloride. (min.) (per cent v/v) (per cent v/v)
B. To 5 ml of a 1 per cent w/v solution add a few drops of 0-15 80 -'775 20 -'725
potassium men::uri":iodide solution; a cream-coloured 15-31 75 -'755 25 -745
precipitate is produced. 31-40 55 45
40-41 55 -'780 45 -'720
C. Dissolve 5 mg in 0.5 ml of water and add 0.1 ml of
41-50 80 20
formaldehyde solution and 2 ml of sulphuric acid; an orange-
red colour is produced. Inject reference solution (c). The test is not valid unless the
signal-to-noise ratio for the first peak is not less than 10 and
D. Gives reaction A ofchlorides (2.3.1).
peak-to-valley ratio where H p is height above the baseline,
Tests and H v is height above the baseline ofthe lowest point of the
curve separating this peak from the peak due to impurity A is
Appearance ofsolution. A2.0 per centw/v solution in carbon not less than 4.0. The relative retention time with reference to
dioxide-free water is clear (2.4.1), and colourless (2.4.1). pethidine for pethidine impurity B is about 0.66 and for
Acidity or alkalinity. To 10 ml ofa 2.0 per cent wIv solution in pethidine impurity A is about 0.68.
carbon dioxide-free water add 0.2 ml of methyl red solution Inject test solution (a), (b), reference solution (a) and (c). In
and 0.2 ml of 0.01 Msodium hydroxide; the solution is yellow. the chromatogram obtained with test solution (b), the area of
Add 0.3 ml of 0.01 M hydrochloric acid; the solution is red. the peale due to I-methyl-4-phenyl-l ,2,3,6-tetrahydropyridine
(pethidine impurity B) is not more than the area of the
Related substances. Determine by liquid chromatography
corresponding peak in the chromatogram obtained with
(2.4.14).
reference solution (c) (10 ppm). In the chromatogram obtained
Solvent mixture. 20 volumes of acetonitrile and 80 volumes with test solution (a), the area of any secondary peak is not
of water. more than the area ofthe principal peak in the chromatogram
Test solution (a). Dissolve about 0.1 g ofthe substance under obtained with reference solution (a) (0.5 per cent) and the sum
examination in 25.0 ml ofthe solvent mixture. of all the secondary peaks is not more than twice the area of
the principal peak in the chromatogram obtained with reference
Test solution (b). Dissolve about 0.125 g of the substance solution (a) (1.0 per cent). Ignore any peak with an area less
under examination in 10.0 ml ofthe solvent mixture. than 0.1 times the area ofthe principal peak in the chromatogram
Reference solution (a). Dilute 0.5 ml of test solution (a) to obtained with reference solution (a) (0.05 per cent).
100.0 ml with the solvent mixture. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Reference solution (b). Dissolve 12.5 mg of 1-methyl-4- Loss on drying (2.4.19). Not more than 0.5 per cent, determined
phenyl-1,2,3,6- tetrahydropyridine in 10.0 ml of the solvent on 1.0 g by drying in an oven at 105°.
mixture. Dilute 1.0 ml of this solution to 100.0 ml with the Assay. Weigh accurately about 0.5 g, dissolve in 30 ml of
solvent mixture: anhydrous glacial acetic acid, add 12 ml of mercuric acetate
Reference solution (c). Dilute 5.0 ml ofreference solution (b) solution. Titrate with 0.1 M perchloric acid, using crystal
and 1.0 ml ofreference solution (c) to 100.0 ml with the solvent violet solution as indicator. Carry out a blank titration.
mixture. 1 ml of 0.1 M perchloric acid is equivalent to 0.02838 g of
Chromatographic system ClsH21N02,HCI.
- a stainless steel column 25 cm x 4.0 mm packed with Storage. Store protected from light and moisture.
endcapped octadecylsilane bonded to porous silica (5
/lm) (Such as Inertsil ODS2),
mobile phase: A. a mixture of equal volumes of 4.2 per Pethidine Injection
cent w/v solution of sodium perchlorate and 1.2 per
cent v/v solution of orthophosphoric acid, adjusted to Pethidine Hydrochloride Injection; Meperidine
pH 2.0 with triethylamine, Hydrochloride Injection
B. acetonitrile,
Pethidine Injection is a sterile solution of Pethidine
a linear gradient programme using the conditions given
Hydrochloride in Water for Injections.
below,
flow rate. 1 ml per minute, Pethidine Injection contains not less than 95.0 per cent and
spectrophotometer set at 210 nm, not more than 105.0 per cent of pethidine hydrochloride,
injectiOn volume. 50 Ill. ClsH21N02,HCI.

1884

www.webofpharma.com
IP 2010 PETHIDINE TA-\3LETS

Usual strength. 50 mg per ml. Inject the test solution and the reference solution. In the
chromatogram obtained with the test solution, the area of any
Identification secondary peak is not more than the area ofthe principal peak
A. To a volume containing 50 mg ofPethidine Hydrochloride in the chromatogram obtained with the reference solution (0.5
add sufficient 1. M sodium hydroxide to make strongly alkaline per cent) and the sum ofthe areas of all the secondary peaks
to litmus paper and extract with two quantities, each of 10 ml, is not more than twice the area of the principal peak in the
of chloroform. Wash the combined extracts with 5 ml of water, chromatogram obtained with the reference solution (1.0 per
dry over anhydrous sodium sulphate, filter and evaporate the cent). Ignore any peak with an area less than 0.1 times the area
filtrate to dryness. Remove the last traces of chloroform by of the principal peak in the chromatogram obtained with the
drying the residual oil at 60° ata pressure not exceeding reference solution (0.05 per cent).
0.7kPa. Other tests. Complies with the tests stated under Parenteral
On the oily residue, determine by infrared absorption Preparations (Injections).
spectrophotometry (2.4.6). Compare the spectrum with that Assay. Determine by liquid chromatography (2.4.14).
obtained with pethidine hydrochloride RS or with the
Test solution. Dilute a volume ofthe injection containing about
reference spectrum of pethidine.
0.1 g of Pethidine Hydrochloride with 100.0 ml of the water.
B. To 0.5 ml add 0.1 ml offormaldehyde solution and 2 ml of Dilute 3.0 ml ofthis solution to 25.0 ml with the mobile phase.
sulphuric acid; an orange-red colour is produced.
Reference solution. A 0.1 per cent w/v solution of pethidine
C. Gives the reactions ofchlorides (2.3.1). hydrochlorideRS. Dilute 3.0 ml ofthis solution to 25.0 ml with
the mobile phase.
Tests
Chromatographic system
Related substances. Determine by liquid chromatography - a stainless steel column 25 cm x 4.0 rom packed with
(2.4.14). endcapped octadecylsilane bonded to porous silica
Solvent mixture. 20 volumes of acetonitrile and 80 volumes (5Ilm) (Such as Spherisorb ODSl),
of water. - column temperature. 40°,
Test solution. Dilute a volume ofthe injection containing 0.1 g - mobile phase: a mixture of 110 volumes of acetonitrile
ofPethidine HydrocWoride with 25.0 ml ofthe solvent mixture. and 90 volumes of a solution prepared by dissolving
6.8 g ofpotassium dihydrogen orthophosphate in 1000
Reference solution. Dilute 0.5 ml of the test solution to 100.0 ml of water, add 10 ml triethylamine, adjusted to pH 7.0
ml with the solvent mixture. with orthophosphoric acid,
Chromatographic system - flow rate. 2 ml per minute,
a stainless steel column 25 cm x 4.0 rom packed with spectrophotometer set at 230 urn,
endcapped octadecylsilane bonded to porous silica - injection volume. 20 ,.d.
(5 Ilm) (Such as Kromasil C18),
Inject the reference solution. The test is not valid unless the
- mobile phase: A. a mixture of equal volumes of 4.2 per
theoritical plates is not less than 8000.
cent w/v solution of sodium perchlorate and 1.2 per
cent v/v solution of orthophosphoric acid, adjusted to Injet the test solution and the reference solution.
pH 2.0 with triethylamine, Calculate the content ofClsHzlNOz,HCl in the injection.
B. acetonitrile,
a linear gradient programme using the conditions given Storage. Store protected from light.
below,
flow rate. 1 ml per minute,
spectrophotometer set at 210 urn,
injection volume. 20 Ill.
Pethidine Tablets
Time Mobile phase A Mobile phase B
(min.) (per cent v/v) (per cent v/v) Pethidine Hydrochloride Tablets; Meperidine
0-15 80 -775 20 -725 Hydrochloride Tablets
15-31 75 -755 25 -745 Pethidine Tablets contain not less than 92.5 per cent and not
31-40 55 45 more than 107.5 per cent of the stated amount of pethidine
40-41 55 -780 45 -720 hydrochloride, ClsHzlNOz,HCI.
41-50 80 20 Usual strengths. 25 mg; 50 mg.

1885

www.webofpharma.com
PETHIDINE TABLETS IP 2010

Identification intense than the spot in the chromatogram obtained with the
" reference solution.
A. Shake a quantity ofthe powdered tablets containing 50 mg
of Pethidine Hydrochloride with 20 ml of chloroform, filter, Other tests. Comply with the tests stated under Tablets.
evaporate the filtrate to dryness and dry the residue at a Assay. Weigh and powder 20 tablets. Weigh accurately a
pressure of 2 kPa. quantity of the powder containing about 0.3 g of Pethidine
On the residue, determine by infrared absorption Hydrochloride in 40 ml of water,add 2 rnl of 5 M sodium
spectrophotometry (2.4.6). Compare the spectrum with that hydroxide and extract immediately with successive quantities
obtained with pethidine hydrochloride RS or with the of25, 10 and 10 ml of chloroform. Wash each extract with the
reference spectrum of pethidine hydrochloride. same 15 ml of water and filter into a dry flask. Combine the
extracts (which should be clear and free from droplets of water).
B. Shake a quantity ofthe powdered tablets containing 0.2 g Titrate with 0.05 M perchloric acid, using 0.15 ml of
of Pethidine Hydrochloride with 20 ml of water and filter. To 1-naphtholbenzein solution as indicator. Carry out a blank
5 ml ofthe filtrate add 10 ml ofpicric acid solution. The crystals titration.
so obtained, after washing with water and drying, melt at
about 190°(2.4.21). 1 ml of 0.05 Mperchloric acid is equivalent to 0.01419 g of
ClsH21N02,HCl.
Tests Storage. Store protected from light and moisture.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with laeselguhr G.
Mobile phase. The upper layer obtained by shaking together
100 volumes of light petroleum (50° to 70°), 8 volumes of Phenindione
2-phenoxyethanol and 1 volume of diethylamine.
Test solution. The upper layer obtained by shaking a quantity o
of the powdered tablets containing 0.1 g of Pethidine
Hydrochloride with 5 ml of water, filtering, shaking the filtrate
with 0.5 ml of 5 M sodium hydroxide and 2 ml of ether and
allowing the layers to separate.
o}o o
Reference solution. Dilute 0.5 mlofthe test solution to 50 ml Mol. Wt. 222.2
with ether.
Phenil1dione is 2-phenylindane-l ,3-dione.
Impregnate the dry plate by placing it in a closed tank
Phenindione contains not less than 98.0 per cent and not
containing a mixture of90 volumes of acetone and 10 volumes
more than 100.5 per cent ofC 1sHIQ02, calculated on the dried
of 2-phenoxyethanol so that the plate dips about 5 mm beneath
basis.
the surface ofthe liquid, allowing the impregnating solvent to
ascend at least 15 cm, removing the plate from the tank and Category. Anticoagulant.
drying in a current ofair. Use immediately, with the flow ofthe Dose. Initially, 200 to 300 mg; subsequently 25 to 100 mg daily,
mobile phase in the same direction as the impregnation. in accordance with the needs of the patient.
Apply to the plate 5 )11 of each solution. Allow the mobile Description. Soft, white or creamy-white crystals; almost
phase to rise 12 cm. Dry the plate in air for 10 minutes, return odourless.
the plate to the tanle and repeat the development. Remove the
plate, allow it to dry in air for 10 minutes and spray with a Identification
0.2 per cent w/v solution of 2,7-dichlorofluorescein in
methanol. Allow to stand for 5 minutes and spray with water A. Determine by infrared absorption spectrophotometry (2.4.6).
until the background is white to pale yellow. Examine in Compare the spectrum with that obtained with phenindione
daylight. The chromatograms show red or orange spots. Any RS.
secondary spot in the chromatogram obtained with the test B. Dissolve 0.1 gin 30 ml of ethanol (95 per cent) with the aid
solution is not more intense than the spot in the chromatogram of heat, cool and add sufficient ethanol (95 per cent) to
obtained with the reference solution. Examine without delay produce 50 ml. Dilute 10 ml of this solution to 250 ml with
in ultraviolet light at 365 nm. The chromatograms show spots 0.1 M sodium hydroxide and ftuther dilute 5 ml to 100 ml with
with intense yellow fluorescence. Any secondary spot in the 0.1 M sodium hydroxide. When examined in the range 230 nm
chromatogram obtained with the test solution is not more and 360 nm (2.4.7), the solution shows absorption maxima at

1886

www.webofpharma.com
IP 2010 PHENINDIONE TABLETS

about 278 nm and at about 330 nm; absorbance at about Phenindione Tablets
278 nm, about 0.55 and at about 330 nm, about 0.16.
Phenindione Tablets contain not less than 92.5 per cent and
C. To 1 g add 50 ml of ethanol (95 per cent) and 0.5 mlof not more than 107.5 per cent of the stated amount of
aniline, heat gently under a reflux condenser for 3 hours, cool phenindione, ClsHIOO Z•
in ice and filter. The residue, after washing with 2 ml ofethanol
(95 per cent) and recrystallisation from chloroform, melts at Usual strength. 50 mg.
about 225°(2.4.21).
Identification
Tests Shake a quantity of the powdered tablets containing 0.2 g of
Phenindione with 50 ml ofchloroform, filter and evaporate the
Related substances. Determme by thin-layer chromatography
filtrate to dryness. Recrystallise the residue from ethanol
(2.4.17), coating the plate with silica gel GF254.
(95 per cent). The crystals complies with the following tests.
Mobile phase. A 0.02 per cent w/v solution of butylated A. Determine by infrared absorption spectrophotometry (2.4.6)..
hydroxytoluene in a mixture of 80 volumes of toluene, Compare the spectrum with that obtained with phenindione
20 volumes of ethyl acetate and 4 volumes of glacial acetic RS.
acid.
B. Dissolve 0.1 gin 30 ml ofethanol (95 per cent) with the aid
Test solution. Dissolve 0.1 g of the substance under of heat, cool and add sufficient. ethanol (95 per cent) to
examination in 10 m1 ofdichloromethane. produce 50 ml. Dilute 10 ml of this solution to 250 ml with
0.1 M sodium hydroxide and further dilute 5 ml to 100 ml with
Reference solution (a). A 0.02 per cent w/v solution of the 0.1 M sodium hydroxide. When examined in the range 230nm
substance under examination in dichloromethane. to 360 nm(2.4.7), the solution shows absorption maxima at
Reference solution (b). A 0.005 per cent w/v solution of the about 278 nm and 330 nm; absorbance at about 278. about
substance under examination in dichloromethane. 0.55 and at about 330 nm, about 0.16.
C. To 50 mg add 1 ml ofsulphuric acid; a deep blue to violet
Apply to the plate 10 III of each solution. Allow the mobile
solution is produced. On dilution with water the solution
phase to rise 4 cm. Dry the plate in warm air and examine in
becomes colourless and a white precipitate is produced.
ultraviolet light at 254 nm. Any secondary spot in the
chromatogram obtained with the test solution is not more
intense than the spot in the chromatogram obtained with
Tests
reference solution (a) and not more than one such spot is Related substances. Determine by thin-layer chromatography
more intense than the spot in the chromatogram 9btained (2.4.17), coating the plate with silica gel GF254.
with reference solution (b).
Mobile phase. A 0.02 per cent w/v solution of butylated
Sulphated ash (2.3 .18). Not more than 0.1 per cent. hydroxytoluene in a mixture of 80 volumes of toluene,
20 volumes of ethyl acetate and 4 volumes of glacial acetic
Loss on drying (2.4.19). Not more than 1.0 per cent, determined acid.
on 1.0 g by drying in oven at 105° for 2 hours.
Test solution. Shake a quantity of the powdered tablets
Assay. Weigh accurately about 0.3 g, add 50 ml of ethanol containing 50 mg of P4enindione with 15 m1 of
(95 per cent) and warm until solution is effected. Cool to dichloromethane, filter, evaporate the filtrate to dryness and
room temperature, add 10 ml ofa 10 per cent v/v solution of dissolve the residue in 5 ml of dichloromethane.
bromine in ethanol (95 per cent) and allow to stand for
Reference solution (a). Dilute 1 volume ofthe test solution to
10 minutes, shaking occasionally. Add 1 g of 2-naphthol and
50 volumes with dichloromethane.
shake until the colour ofthe bromine is discharged. Remove
any vapour of bromine in the flask with a current of air, add Reference solution (b). Dilute 1 volume of the test solution to
50 m1 of water and 10 ml of dilute potassium iodide solution 200 volumes with dichloromethane.
and titrate the liberated iodine with 0.1 M sodium thiosulphate Apply to the plate 10 III of each solution. Allow the mobile
using starch solution as indicator. phase to rise 4 cm. Dry the plate in warm air and examine in
1 ml of 0.1 M sodium thiosulphate is equivalent to 0.01111 g ultraviolet light at 254 nm. Any secondary spot in the
ofC1SHIOOz. chromatogram obtained with the test solution is not more
intense than the spot in the chromatogram obtained with
Storage. Store protected from moisture. reference solution (a) and not more than one such spot is

1887

www.webofpharma.com
PHENlRAMINE MALEATE IP 2010

more intense than the spot in the chromatogram obtained Identification


with reference solution (b).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Uniformity ofcontent. (For tablets containing 50 mg or less) Compare the spectrum with that obtained with pheniramine
- Comply with the test stated under Tablets. maleate RS or with the reference spectrum of pheniramine
Place one tablet in 50 ml of0.1 M sodium hydroxide, dissolve maleate.
completely by shaldng gently, add a further 100 ml of 0.1 M B. When examined in the range 230 nm to 360 nm (2.4.7), a
sodium hydroxide and shake for 1 hour. Dilute to 250.0 ml with 0.002 per cent w/v solution in 0.1 Mhydrochloric acid shows
0.1 M sodium hydroxide, filter and dilute a portion of the an inflection at about 262 nm; absorbance at about 265 nm,
filtrate with sufficient 0.1 M sodium hydroxide to produce a about 0.42.
solution containing 4 Ilg ofPhenindione per ml. Measure the
C. Dissolve 0.25 gin 5 ml ofwater, add 2 ml ofstrong ammonia
absorbance ofthe resulting solution at the maximum at about
solution and extract with three quantities, each of 5 ml, of
278 nm (2.4.7). Calculate the content ofC 1SH lO0 2taking 1310
chloroform. Evaporate the aqueous extract to dryness, add
as the specific absorbance at 278 nm.
0.2 ml of 1 M sulphuric acid and 5 ml of water, extract with
Other tests. Complies with the tests stated under Tablets. four quantities, each of 25 ml, of ether and evaporate the
Assay. Weigh and powder 20 Tablets. Weigh accurately a combined ether extracts to dryness in a current ofwarm air. To
quantity ofthe powder containing about 50 mg ofPhenindione the residue add 50 mg of resorcinol and 1 ml of sulphuric
and shake with 150 ml of 0.1 M sodium hydroxide for 1 hour, acid, heat in a water-bath.for 2 minutes, shake well, heat in a
add sufficient 0.1 M sodium hydroxide to produce 250.0 ml, water-bath for a further 30 minutes and cool in ice. Carefully
filter and dilute 5.0 ml of the filtrate to 250.0 ml with 0.1 M add 5 ml ofwater; a yellow colour is produced. To 2 ml ofthe
sodium hydroxide. Measure the absorbance of the resulting solution add 3 ml of a 50 per cent w/v solution of ammonium
solution at the maximum at about 278 nm (2.4.7). Calculate the acetate, previously cooled in ice; a pink colour is produced
content ofC 1SH lO0 2taking 1310 as the specific absorbance at which persists for at least 10 minutes in the cooled solution.
278nm. Tests
Storage. Store protected from moisture.
pH (2.4.24). 4.5 to 5.5, determined in a 1.0 per cent w/v solution.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G
Pheniramine Maleate
Mobile phase. A mixture of 50 volumes of cyclohexane,
40 volumes of chloroform and 10 volumes of diethylamine.
Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml ofmethanol.

N (COOH Reference solution (a). A 0.02 per cent w/v solution of the
" substance under examination in methanol.
COOH
Reference solution (b). A 0.004 per cent w/v solution of the
substance under examination in methanol.
Mol. Wt. 356.4 Apply to the plate 10 III of each solution. After development,
Pheniramine Maleate is (3RS)-N,N-dimethyl-3-phenyl-3- dry the plate in air, spray with dilute potassium
(pyridin-2-yl)propan-l-amine hydrogen maleate. iodobismuthate solution. Any secondary spot in the
chromatogram obtained with the test solution is not more
Pheniramine Maleate contains not less than 99.0 per cent and
intense than the spot in the chromatogram obtained with
not more than 101.0 per cent ofC16H2oN2,C4H404, calculated
reference solution (a) and not more than one such spot is
on the dried basis.
more intense than the spot in the chromatogram obtained
Category. Antihistaminic. with reference solution (b).
Dose. Orally, 25 to 50 mg daily, in divided doses; by Heavy metals (2.3.13). Dissolve 1.0 g in 10 ml ofwater and add
intramuscular or slow intravenous injection, 22.5 to 45 mg 2 ml ofacetic acid and sufficient water to produce 25 ml. The
daily, in divided doses. resulting solution complies with the limit test for heavy metals,
Description. A white or almost white, crystalline powder; Method A (20 ppm).
odourless or almost odourless. Sulphated ash (2.3.18). Not more than 0.1 per cent.

1888

www.webofpharma.com
IP 2010 PHENIRAMINE TABLETS

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Related substances. Determine by the method described under
on 1.0 g by drying in an oven at 60° at a pressure not exceeding th'e Identification test using as the reference solution a
0.7kPa. solution prepared by diluting 1 volume of the test solution to
500 volumes with chloroform. Any secondary spot in the
Assay. Weigh accurately about 0.4 g, dissolve in 20 ml of
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric chromatogram obtained with the test solution is not more
acid, using 1- naphtholbenzein solution as indicator. Carry intense than the spot in the chromatogram obtained with the
out a blank titration. reference solution.

1 ml of 0.1 M perchloric acid is equivalent to 0.01782 g of Other tests. Complies with the tests stated under Parenteral
CI6H20N2,C4~04.
Preparations (Injections).

Storage. Store protected from light and moisture. Assay. To an accurately measured volume containing about
0.11 g ofPheniramine Maleate add sufficient water to produce
50.0 ml and mix well. To 20.0 ml add sufficient 1 M sodium
hydroxide to make the solution just alkaline to litmus paper,
add 2 ml in excess and extract with two quantities, each of
Pheniramine Injection 50 ml, ofether. Wash each ether extract in succession with 20,
Pheniramine Maleate Injection 20 and 5 ml of 0.1 M hydrochloric acid, dilute the combined
extracts to 100.0 ml with 0.1 M hydrochloric acid and mix.
Pheniramine Injection is a sterile solution of Pheniramine Dilute 5.0 ml to 100.0 ml with 0.1 M hydrochloric acid and
Maleate in Water for Injections. measure the absorbance of the resulting solution at the
Pheniramine Injection contains not less than 90.0 per cent and maximum at about 265 nrn (2.4.7), using 0.1 M hydrochloric
not more than 110.0 per cent of the stated amount of acid as the blank. Calculate the content OfCI6H2oN2, C4H40 4
pheniramine maleate, C16H20N2, C4H40 4. taking 210 as the specific absorbance at 265 nm.
Usual strength. 22.75 mg per mI. Storage. Store protected from light.

Identification
Determine by thin-layer chromatography (2.4.17), coating the
plate with silica gel GF254. Pheniramine Tablets
Mobile phase. A mixture of 50 volumes of cyclohexane, Pheniramine Maleate Tablets
40 volumes of chloroform and 10 volumes of diethylamine.
Pheniramine Tablets contain not less than 92.5 per cent and
Test solution. Evaporate an appropriate volume ofthe injection not more than 107.5 per cent of the stated amount of
to dryness in a current ofnitrogen using the minimum amount pheniramine maleate, C16H20N2, C4~04'
of heat, dissolve· the residue in sufficient chloroform to
produce a solution containing 2.0 per cent w/v solution of Usual strengths. 12.5 mg; 25 mg; 50 mg.
Pheniramine Maleate and centrifuge.
Reference solution. A2.0 per cent w/v solution ofpheniramine
Identification
maleate RS in chloroform. Boil a quantity ofthe powdered tablets containing about 0.5 g
Apply to the plate 10 ).11 of each solution. After development, ofPheniramine Maleate with 150 ml ofacetone under a reflux
dry the plate in air and examine in ultraviolet light at 254 nrn. condenser for about 45 minutes. Filter and evaporate the filtrate
The two principal spots in the chromatogram obtained with to dryness on a water-bath. The residue complies with the
the test solution correspond to those in the chromatogram following tests.
obtained with the reference solution. Spray the plate with A. Determine by infrared absorption spectrophotometry (2.4.6).
dilute potassium iodobismuthate solution. The principal spot Compare the spectrum with that obtained with pheniramine
in the chromatogram obtained with the test solution maleate RS or with the reference spectrum of pheniramine
corresponds to that in the chromatogram obtained with the maleate.
reference solution.
B. When examined in the range 230 nrn to 360 nrn (2.4.7), a
Tests 0.002 per cent w/v solution in 0.1 M hydrochloric acid shows
an inflection at about 262 nm; absorbance at about 265 nrn,
pH (2.4.24). 4.5 to 5.5. about 0.42.

1889

www.webofpharma.com
PHENIRAMINE TABLETS IP 2010

C. Dissolve 0.25 gin 5 ml of water, add 2 ml ofstrong ammonia Phenobarbitone


solution and extract with three quantities, each of 5 ml, of
chloroform. Evaporate the aqueous extract to dryness, add Phenobarbital
0.2 ml of 1 M sulphuric acid and 5 ml of water, extract with
four quantities, each of 25 ml, of ether and evaporate the
combined ether extracts to dryness in a current ofwarm air. To
the residue add 50 mg of resorcinol and 1 ml of sulphuric
acid, heat in a water-bath for 2 minutes, shake well, heat in a
water-bath for a further 30 minutes and cool in ice. Carefully
add 5 ml of water; a yellow colour is produced. To 2 ml ofthe
solution add 3 ml of a 50 per cent w/v solution of ammonium
acetate, previously cooled in ice; a pink colour is produced Mol. Wt. 232.2
which persists for at least 10 minutes in the cooled solution. Phenobarbitone is 5-ethyl-5-phenylbarbituric acid.
Phenobarbitone contains not less than 99.0 per cent and not
Tests
more than 101.0 per cent of ClzHlZNz03, calculated on the
Related substances. Determine by thin-layer chromatography dried basis.
(2.4.17), coating the plate with silica gel G Category. Sedative; anticonvulsant.
Mobile phase. A mixture of 50 volumes of cyclohexane, Dose. 60 to 300 mg at night.
40 volumes of chloroform and 10 volumes of diethylamine.
Description. Colourless crystals or a white, crystalline powder;
Test solution. Shake a quantity of the powdered tablets odourless.
containing 20 mg of Pheniramine Maleate with 10 ml of
methanol, centrifuge and use the supernatant liquid. Identification
Reference solution (a). Dilute 1 volume ofthe test solution to Test A may be omitted if tests B, C, D and E are carried out.
100 volumes with methanol. Tests C, D and E may be omitted if tests A and B are carried
out.
Reference solution (b). Dilute 1 volume of reference solution
(a) to 20 volumes with methanol. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with phenobarbitone
Apply to the plate 10 J.Ll of each solution. After development, RS or with the reference spectrum of phenobarbitone.
dry the plate in air, spray with . dilute potassium
iodobismuthate solution. Any secondary spot in the B. Determine the melting point (2.4.21) ofthe substance under
chromatogram obtained with the test solution is not more examination and of a mixture of equal quantities of the
intense than the spot in the chromatogram obtained with substance under examination and phenobarbitone RS. The
reference solution (a) and not more than one such spot is difference between the melting points, which are about 175°,
more intense than the spot in the chromatogram obtained is not greater than 2°.
with reference solution (b). C. Complies with the test for identification of barbiturates
Other tests. Comply with the tests stated under Tablets. (2.3.2).
D. Dissolve about 20 mg in 5 ml of eth.anol, add a drop of
Assay. Weigh and powder 20 tablets. Weigh accurately a
cobalt chloride solution and a drop of dilute ammonia
quantity ofthe powder containing about 45 mg ofPheniramine
solution; a violet colour is produced.
Maleate, shake with 20 l11l of 0.1 Mhydrochloric acid, centrifuge
and transfer the supernatant liquid to a 1OO-ml volumetric flask. E. Gives the reaction ofnon-nitrogen substituted barbiturates
Repeat the extraction with three further quantities, each of20 (2.3.1).
ml, of 0.1 M hydrochloric acid. Combine the extracts and add
sufficient 0.1 M hydrochloric acid to produce 100.0 ml. Mix Tests
and dilute 5.0 ml to 100.0 ml with 0.1 M hydrochloric acid; Appearance of solution. A 10.0 per cent w/v solution in a
measure the absorbance of the resulting solution at the mixture of20 volumes of2 M sodium hydroxide and 30 volumes
maximum at about 265 nm (2.4.7), using 0.1 M hydrochloric of water is clear (2.4.1), and not more intensely coloured than
acid as the blank. Calculate the content of C16HzoNz,C4H404 reference solution YS6 (2.4.1).
taking 210 as the specific absorbance at 265 urn.
Acidity. Mix 1.0 g with 50 ml ofwater, boil for 2 minutes, allow
Storage. Store protected from light and moisture. to cool, filter and adjust the volume to 50 ml. To 10 m1 of the

1890

www.webofpharma.com
IP 2010 PHENOBARBITONE SODIUM

filtrate add 0.1 5 ml of methyl redsolution; not more than 0.1 ml anhydrous sodium sulphate and evaporate the ether to
of 0.1 M sodium hydroxide is required to change the colour of dryness on a water-bath. The residue complies with the
the solution from orange-yellow to pure yellow. following tests.
Related substances. Determine by thin-layer chromatography A. Determine by infrared absorption spectrophotometry (2.4.6).
(2.4.17), coating the plate with silica gel GF254. Compare the spectrum with that obtained with phenobarbitone
RS or with the reference spectrum of phenobarbitone.
Mobile phase. A mixture of 5 volumes of concentrated
ammonia, 15 volumes of ethanol (95 per cent) and 80 volumes B. Dissolve 50 mg in 2 ml of a 0.2 per cent w/v solution of
of chloroform. cobaltous acetate in methanol, warm, add 50 mg ofpowdered
borax and heat to boiling; a bluish violet colour is produced.
Test solution. Dissolve about 1.0 g of the substance under
examination in 100 ml of ethanol (95 per cent).
Tests
Reference solution. Dilute 0.5 ml ofthe test solution to 100 ml
with ethanol (95 per cent). Disintegration (2.5.1). 30 minutes

Apply 20 JlI of each solution. Allow the mobile phase to rise Other tests. Complies with the tests stated under Tablets.
15 em. Dry the plate in air and examine in ultraviolet light at 254 Assay. Weigh and powder 20 tablets. Weigh accurately a
om. Spray with diphenylcarbazone mercuric reagent. Allow quantity of the powder containing about 0.3 g of
the plate to dry in air and spray with freshly prepared Phenobarbitone and extract in a continuous extraction
alcoholic potassium hydroxide solution ( 20 per cent w/v). apparatus (2.1.8) with ether until complete extraction is
Heat at 105° for 5 minutes. Any secondary spot in the effected. Remove the ether and dry the residue, which is
chromatogram obtained with the test solution, is not more C12H12N203, to constant weight at 105°,
intense than the spot in the chromatogram obtained with the
reference solution (0.5 per cent). Storage. Store protected from moisture.

Sulphated ash (2.3.18). Not more than 0.1 per cent.


Loss on drying (2.4.19). Not more than 1.0 per cent w/w,
determined on 1.0 g by drying in an oven at 105° for 2 hours.
Phenobarbitone Sodium
Assay. Weigh accurately about 0.1 g, dissolve in 5 ml of
pyridine, add 0.25 ml of thymolphthalein solution and 10 ml Phenobarbital Sodium; Soluble Phenobarbitone; Soluble
of silver nitrate-pyridine reagent and titrate with 0.1 M Phenobarbital
ethanolic sodium hydroxide until a pure blue colour is
obtained. Repeat the operation without the substance under H
examination. The difference between the titrations represents 0 NyONa

&
the amount of sodium hydroxide required. H3C I
N
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
0.01161 g OfC12H12N203.
~ ~ 0
Storage. Store protected from moisture.

C l2H ll N 2Na03 Mol. Wt. 254.2


Phenobarbitone Tablets Phenobarbitone Sodium is sodium 5-ethyl-5-phenylbarbiturate.

Phenobarbital Tablets Phenobarbitone Sodium contains not less than 99.0 per cent
and not more than 101.0 per cent OfC12HllN2Na03, calculated
Phenobarbitone Tablets contain not less than 92.5 per cent
on the dried basis.
and not more than 107.5 per cent of the stated amount of
phenobarbitone, CI2H12N203. Category. Sedative; anticonvulsant.
Usual strengths. 15 mg; 30 mg; 60 mg; 100 mg. Dose. Orally, 60 to 300 mg at night; by intramuscular or
intravenous injection, 50 to 200 mg, repeated after 6 hours if
Identification necessary; maximum 600 mg daily.
Extract a quantity of the powdered tablets containing about Description. A white powder or crystalline granules or flaky
0.5 g of Phenobarbitone with 50 rn1 of ether, filter through crystals; hygroscopic.

1891

www.webofpharma.com
PHENOBARBITONE SODIUM IP 2010

Identification Apply 20 I!l ofeach solution. Allow the mobilephase to rise 15


em. Dry the plate in air and examine in ultraviolet light at 254
Test A may be omitted iftests B, C, D, E andF are carried out. nm. Spray with diphenylcarbazone mercuric reagent. Allow
Tests C, D andE may be omitted iftests A, BandF are carried the plate to dry in air and spray, with freshly prepared
out. alcoholic potassium hydroxide solution ( ,20 per ,cent w/v).
A. Dissolve 0.2 gin 20 ml of ethanol (50 per cent), acidify Heat at 105 0 for 5 minutes. Any secondary spot in the
with dilute hydrochloric acid and extract with 50 ml of ether. chromatogram' obtained with the test solution, is not more
Wash the ether layer with 10 ml of water, dry over anhydrous intense than the spot in the chromatogram obtained with the
sodium sulphate and filter. Evaporate the filtrate to dryness reference solution (0.5 per cent).
and dry the residue at 105°. Loss on drying (2.4.19). Not more than 7.0 per cent, determined
On the residue, determine by infrared absorption on 0.5 g by drying in an oven at 150° for 4 hours.
spectrophotometry (2.4.6). Compare the spectrum with that Assay. Weigh accurately about 0.15 g, dissolve in 2 ml of
obtained with phenobarbitone RS or with the reference water and add 8 ml of 0.05 M sulphuric acid. Heat to boiling
spectrum of phenobarbitone. and cool. Add 30 ml of methanol and shake until dissolution
B. Determine the melting point (2.4.21), ofthe residue obtained is complete. Titrate with 0.1 M sodium hydroxide, determining
in test A and ofa mixture ofequal quantities ofthe residue and the end-point potentiometrically (2.4.25). After the first
phenobarbitone RS. The difference between the melting inflection, stop the addition of the sodium hydroxide, add
points, which are about 175°, is not greater than 2°. 10 ml of pyridine, mix and continue the titration until the
second inflection is reached. The difference between the
C. Complies with the test for identification of barbiturates volumes represents the amount of sodium hydroxide
(2.3.2), but using the following solutions. required.
Test solution. A 0.1 per cent w/v solution of the substance 1 ml of 0.1 M sodium hydroxide is equivalent to 0.02542 g of
under examination in ethanol (50 per cent). C12HIlNzNa03'
Reference solution. A 0.09 per cent w/v solution of Storage. Store protected from moisture.
phenobarbitone RS in ethanol (50 per cent).
D. 1 g dissolves completely in 20 ml of ethanol (90 per cent)
(distinction from barbitone sodium). Phenobarbitone Injection
E. Gives the reaction of non-nitrogen substituted barbiturates Phenobarbital Sodium Injection; Phenobarbitone Sodium
(2.3.1). InjeCtion; Soluble PhenobarbitoneInjeetion
F. Ignite about 0.1 g; the residue gives the reactions ofsodium Phenobarbitone Injection is a sterile solution of
salts (2.3.1). Phenobarbitone Sodium in a mixture of nine volumes of
Propylene Glycol and one volume of Water for Injections.
Tests
Phenobarbitone Injection contains not less than 95.0 per cent
Appearance ofsolution. Al 0.0 per cent w/v solution in ethanol and not more than 105.0 per cent of the stated amount of
(50 per cent) is clear (2.4.1), and not more intensely coloured phenobarbitone sodium, ClzHIINzNa03'
than reference solution YS7 (2.4.1).
Usual strengths. 30 mg, 60 mg, 65 mg and 130 mg per ml.
pH (2.4.24). Not more than 10.2, determined in a 10.0 per cent
Identification
w/v solution.
To a volume containing 1 g of Phenobarbitone Sodium add
Related substances. Deterrriine by thin-layer chromatography
15 ml of water if necessary, make slightly acidic with 1 M
(2.4.17), coating the plate with silica gel GF254.
sulphuric acid and filter. The residue, after washing with water
Mobile phase. A mixture of 5 volumes of concentrated and drying at 105°, complies with the following tests.
ammonia, 15 volumes of ethanol (95 per cent) and 80 volumes
A. Determine by infrared absorption spectrophotometry (2.4.6).
of chloroform.
Compare the spectrum with that obtained with phenobarbitone
Test solution Dissolve about 1.0 g of the substance under RS or with the reference spectrum of phenobarbitone.
examination in 100 ml of ethanol (95 per cent).
B. Dissolve 50 mg in 2 ml of a 0.2 per cent w/v solution of
Reference solution. Dilute 0.5 ml ofthe test solution to 100 ml cobalt acetate in methanol, warm, add 50 mg of powdered
with ethanol (95 per cent). borax and heat to boiling; a bluish violet colour is produced.

1892

www.webofpharma.com
PHENOL

Tests the filtrate is alkaline to litmus solution and yields a white


precipitate on the addition of dilute hydrochloric acid.
pH (2.4.24).10.0 to 11.0.
E. The powdered tablets, when moistened with hydrochloric
Other tests. Complies with the tests stated under Parenteral acid and introduced on a platinum wire into a flame, impart a
Preparations (Injections). yellow colour to the flame.
Assay. Weigh accurately about 2.0 g, add 30 ml of water and
3 g of sodium carbonate, stir to dissolve and titrate with Tests
0.1 M silver nitrate until a distinct turbidity is observed when Other tests. Complies with the tests stated under Tablets.
viewed against a black background, the solution being stirred
vigorously throughout the titration. Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.3 g of
1 ml of 0.1 M silver nitrate is equivalent to 0.02542 g of Phenobarbitone Sodium, dissolve as completely as possible
Ct2HIIN2Na03' in 10 ml of a 2 per cent w/v solution of sodium hydroxide,
Determine the weight per ml of the injection (2.4.29) and saturate with sodium chloride, acidify with hydrochloric acid
calculate the percentage weight in volume ofC12H11N2Na03. and extract with successive quantities, each of 15 ml, of ether
until complete extraction is effected. Wash the combined
Storage. Store in single dose containers. extracts with two quantities, each of2 ml, of water and extract
Labelling. The label states that the injection should not be the combined washings with 10 ml of ether. Add the ether to
used ifthe solution is discoloured or ifit contains a precipitate. the main ether layer and dry the residue to constant weight at
105°.
1 g ofthe residue is equivalent to 1.095 g ofC12HlIN2Na03.
Storage. Store protected from moisture.
Phenobarbitone Sodium Tablets
Phenobarbital Sodium Tablets; Soluble Phenobarbitone
Tablets; Soluble Phenobarbital Tablets Phenol
Phenobarbitone Sodium Tablets contain not less than Carbolic acid
92.5 per cent and not more than 107.5 per cent of the stated
OH
amount ofphenobarbitone sodium, C12HllN2Na03'
Usual strengths. 30 mg; 60 mg.

Identification
6 Mol. Wt. 94.1
A. Heat 0.1 g ofthe residue obtained in Assay on a water-bath
Phenol contains not less than 99.0 per cent and not more than
with 15 ml of ethanol (25 per cent) until dissolved, filter while
100.5 per cent ofC 6H 60.
hot and allow to cool. Filter through a sintered-glass crucible,
wash with a small quantity of ethanol (25 per cent) and dry at Category. Antiseptic; antipruritic; pharmaceutical aid
105°. Heat in a sealed tube at 105° for 1 hour. (antimicrobial preservative).
On the residue, determine by infrared absorption Description. Colourless or faintly pink or faintly yellowish
spectrophotometry (2.4.6). Compare the spectrum with that crystals or crystalline masses; odour, characteristic;
obtained with phenobarbitone RS or with the reference deliquescent. .
spectrum of phenobarbitone.
Identification
B. The residue obtained in testA melts at about 175° (2.4.21).
A. Dissolve 0.5 gin 2 ml of strong ammonia solution. Dilute
C. Dissolve 50 mg ofthe residue obtained in Assay in 2 ml of this solution to about 100 ml with water and to 2 ml of the
a 0.2 per cent w/v solution of cobaltous acetate in methanol, resulting solution, add 0.05 ml of 3 per cent w/v solution of
warm, add 50 mg of powdered borax and heat to boiling; a sodium hypochlorite; a blue colour develops which becomes
bluish violet colour is produced. progressively more intense.
D. Triturate a quantity of the powdered tablets containing B. Dissolve 1.0 g in sufficient water to produce 15 ml (solution
0.2 g ofPhenobarbitone Sodium with 5 ml of water and filter; A) and to 1 ml,add 10 ml of water and 0.1 mlofJerricchloride

1893

www.webofpharma.com
PHENOL IP 2010

solution; a violet colour is produced which disappears on the Dose. 30 to 200 mg.
addition of 5 ml of 2-propanol.
Description. A white or yellowish white, crystallimi or
C. To 1 ml ofsolutionA add 10 ml of water and 1 m1 of bromine amorphous powder; odourless or almost odourless.
solution; a pale yellow precipitate is produced.
Identification
Tests
A. Dissolves in dilute solutions of alkali hydroxides and in
Appearance of solution. Solution A is clear (2.4.1), and not hot solutions ofalkali carbonates forming a red solution which
more intensely coloured than reference solution BS6 (2.4.1). is decolorised by dilute acids.
Acidity. To 2 ml of solution A add 0.05 ml of methyl orange B. Dissolve 25 mg in 100 ml of ethanol (95 per cent) (solution
solution; the solution is yellow. A). To 2.0 ml ofsolutionAadd 5.0 ml of 1 Mhydrochloric acid
and dilute to 50.0 ml with ethanol (95 per cent) (solution AI)'
Freezing point (2.4.11). Not less than 39.so.
To 10.0 ml of solution A add 5.0 ml ofl M hydrochloric acid
Non-volatile matter. Not more than 0.05 per cent, when 5.0 g is and dilute to 50.0 ml with ethanol (95 per cent) (solution A 2).
volatilised on a water-bath and dried to constant weight at To 2.0 ml of solution A add 5.0 ml of 1 M sodium hydroxide
105°. and dilute to 50.0 ml with ethanol (95 per cent) (solution B).
Assay. Weigh accurately about 0.5 g and dissolve in sufficient Examined between 220 urn and 250 urn (2.4.7), solution AI
water to produce 250.0 ml. Transfer 25.0 ml to a ground-glass- shows an absorption maximum at 229 nm. The specific
stoppered flask, add 50.0 ml of 0.05 M bromine and 5 ml of absorbance at the maximum at 229 urn is 922 to 1018. Examined
hydrochloric acid, stopper, allow to stand for 30 minutes, between 250 urn and 300 urn, solutionA2 shows an absorption
swirling occasionally, and allow to stand for a further maximum at 276 urn. The specific absorbance at the maximum
15 minutes. Add 5 ml ofa 20 per cent w/v solution ofpotassium at 276 urn is 142 to 158. Examined between 230 urn and 270 urn,
iodide, shake and titrate with 0.1 M sodium thiosulphate until solution B shows an absorption maximum at 249 urn. The
a faint yellow colour remains. Add 0.5 ml of starch solution specific absorbance at the maximum at 249 urn is 744 to 822.
and 10 ml of chloroform and continue the titration with
Tests
vigorous shaking. Repeat the operation without the substance
under examination. The difference between the titrations Solution A. To 2.0 g add 40 ml of distilled water, boil, cool
represents the amount of bromine required. and filter.
1 ml of 0.05 Mbromine is equivalentto 0.001569 g ofC6H 60. Appearance of solution. A 4 per cent w/v solution in ethanol
(95 per cent) is clear (2.4.1) and not more intensely coloured
Storage. Store protected from light and moisture.
than reference solution YS7 (2.4.1).
Acidity or alkalinity. To 10 ml of solution A, add 0.15 ml of
bromothymol blue solution. Add 0.05 ml of 0.01 M
Phenolphthalein hydrochloric acid, the solution is yellow. Add 0.1 ml of 0.01
M sodium hydroxide, the solution is blue.
o Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel F254.
Mobile phase. A mixture of 50 volumes of acetone and 50
volumes of dichloromethane.

OH Test solution. Dissolve 0.5 g of the substance under


examination in 10 ml ethanol (95 per cent).
HO Reference solution (a). Dilute 1 ml ofthe test solution to 10 ml
with ethanol (95 percent). Dilute 5 ml ofthis solution to 100
Mol. Wt. 318.3 ml with ethanol (95 per cent).
Phenolphthalein is 3,3-bis(4-hydroxyphenyl)phthalide. Reference solution (b). Dissolve 25 mg ofjluorene in ethanol
Phenolphthalein contains not less than 98.0 per cent and not (95 per cent), add 0.5 ml ofthe test solution and dilute to 10ml
more than 102.0 per cent of C2oH1404, calculated on the dried with ethanol (95 per cent).
basis. Apply to the plate 5 III of each solution. Allow the mobile
Category. Laxative. phase to rise 15 em. Dry the plate in air and examine at 254 urn

1894

www.webofpharma.com
IP 2010 PHENOXYETHANOL

and re-examine after exposure to ammonia vapour. Any Identification


secondary spot in the chromatogram obtained with the test
solution is not more intense than the spot in the chromatogram Shake 2 ml of Phenoxyethanol with a mixture of 4.0 g of
obtained with reference solution (a) (0.5 per cent). The test is potassium permanganate, 5.4 g of sodium carbonate and 75
not valid unless the chromatogram obtained with reference ml of water for 30 minutes. Add 25 g of sodium chloride and
solution (b) shows 2 clearly separated spots. stir continuously for 60 minutes, filter and adjusted to pH 1.7
with hydrochloric acid. The precipitate, after recrystallisation
Chlorides (2.3.12). Dilute 10 ml of solution A to15 ml with from water complies with the following tests.
water. The solution complies with the limit test for chlorides
(lOOppm). A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with phenoxyethanol
Heavy metals (2.3.13). Heat 5 g with 50 ml of dilute RS or with the reference spectrum of phenoxyethanol.
hydrochloric acid on a water-batl). for 5 min and filter.
Evaporate the filtrate almost to dryness and dissolve the B. When examined in the range 240 nm to 350 nm (2.4.7), a
residue in 50 ml of water. 12 ml ofthis solution complies with 0.008 per cent w/v solution in water, shows two absorption
the limit test for heavy metals, Method D (20 ppm). Use 10 ml maxima at 269 nm and 275 nm and specific absorbance at 269
of lead standard solution (2 ppm Pb) to prepare the standard. nm is 95 to 105 and at 275 nm is 75 to 85.

Sulphates (2.3.17). 15 ml ofsolution A complies with the limit Tests


test for sulphates (200 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Refractive index (2.4.27). 1.537 to 1.539

Loss on drying (2.4.19). Not more than 1.0 per cent, determined Relative density (2.4.29). 1.105 to 1.110
on 1.0 g by drying in an oven at 105°. Related substances. Determine by Gas chromatography
Assay. Dissolve 0.1 gin 5 ml of dimethylformamide. Add 5 ml (2.4.13).
of sodium carbonate solution, 10 ml of sodium hydrogen Internal standard solution. Dissolve 1.25 g of methyllaurate
carbonate solution, 35 ml of water and 50.0 ml of 0.05 M in 25.0 ml of dichloromethane.
iodine. Add 10 ml of dichloromethane and 20 ml of dilute
sulphuric acid. Titrate the excess ofiodine with 0.1 M sodium Test solution. Dissolve 5 g ofthe substance under examination
thiosulphate; using 0.3 ml of starch solution added towards in 10.0 ml of dichloromethane and add 1.0 ml ofthe internal
the end ofthe titration, as indicator. Carry out a blank titration. standard solution.

1 mlof 0.05 M iodine is equivalent to 0.003979 g ofCzoHl404. Reference solution. Add 10.0 ml of the internal standard
solution to 1.0 ml of the test solution and dilute to 100.0 ml
Storage. Store protected from moisture. with dichloromethane.
Chromatographic system
- a glass column 1.5 m x 4 mm, packed with silanised
Phenoxyethanol diatomaceous earth support (150 to 180 mesh) coated
with 3 per cent w/w solution of polymethylphenyl-
2-Phenoxyethanol siloxane,
temperature:
column. 130°,
inlet port and detector at 200°,
flame ionization detector,
flow rate. 30 ml per minute using nitrogen as a carrier
gas.
Mol. Wt.138.2 Inject 1 f!1 ofthe reference solution. The test is not valid unless
Phenoxyethanol is l-hydroxy-2-phenoxyethane. the resolution between the peaks due to phenoxyethanol and
methyllaurate is not less than 12.
Phenoxyethanol contains not less than 99.0 per cent and not
more than 100.5 per cent ofC~HIOOz, calculated on the dried Inject 1 f!1 of the test solution and the reference solution. In
basis. the chromatogram obtained with the test solution, the ratio of
the area due to peak corresponding tophenoxyethanol to the
Category. Pharmaceutical aid.
area of the peak due to the internal standard from the
Description. A colourless, slightly viscous liquid. chromatogram obtained with the reference solution. from the

1895

www.webofpharma.com
PHENOXYMETHYLPENICILLIN POTASSIUM IP 2010

chromatogram obtained with test solution, the ratio of the Identification


area ofthe sum ofany secondary peale and the peak due to the
internal standard, 'is not more than 1.0 per cent. Test A may be omitted if tests Band C are carried out. Test B
may be omitted if tests A and C are carried out.
Phenol. Not more than 0.1 per cent.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Shake about 1 g ofpropoxyphenol in 50 ml of dichloromethane Compare the spectrum with that obtained with
with 1 ml of dilute sodium hydroxide solution and 10 ml of phenoxymethylpenicillin potassium RS.
water. Wash the upper layer with 2 quantities, each of20 ml of
dichloromethane and dilute to 100.0 ml with water. The B. Gives reaction B ofpenicillins and cephalosporins (2.3.1).
absorbance at the maxima at 287 urn (2.4.7) is not more than C. Gives reaction A ofpotassium salts (2.3.1).
0.27.
Assay. Weigh accurately 2 g, dissolve in 10.0 ml of freshly Tests
prepared acetic anhydride solution, heat in a water bath for pH (2.4.24). 5.5 to 7.5, determined in a 0.5 per centw/v solution.
45 minutes. Cool, add 10.0 ml of water. further heat for 2
minutes, cool and add 10.0 ml of butanol, shake vigorously Specific optical rotation (2.4.22). +215.00 to +230.0°, determined
and titrate excess of acetic acid with 1 M sodium hydroxide in a 1.0 per cent w/v solution in carbon dioxide-free water.
using 0.2 ml of phenolphthalein solution as indicator. Carry Light absorption (2.4.7). Absorbance of a 0.1 per cent w/v
out a blank titration. solution in 0.1 M sodium hydroxide at the maximum at about
1 ml of 1 M sodium hydroxide is equivalent to 0.1382 g of 306 nm, not more than 0.33. Absorbance of a 0.02 per cent
C8H IOOZ• w/v solution in O.lM sodium hydroxide at the maximum at
about 274 nm, not less than 0.50.
Phenoxyacetic acid. Not more than 0.5 per cent.
Determine by liquid chromatography (2.4.14).
Phenoxymethylpenicillin Potassium
Test solution. Weigh accurately a suitable quantity of the
Penicillin V Potassium substance under examination, dissolve in phosphate buffer
pH 6.6 and dilute to obtain a solution having a known
concentration of about 20 mg per ml.
Reference solution. Weigh accurately a suitable quantity of·
phelloxyacetlc acid, dissolve in the phosphate buffer pH 6.6
and dilute to obtain a solution having a known concentration
of about 0.1 mg per ml.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
Mol. Wt. 388.5 octadecylsilane chemically bonded to porous silica
Phenoxymethylpenicillin Potassium is potassium (6R)-6-(2- (5Ilm),
phenoxyacetamido)penicillinate, produced by the growth of - mobile phase: a mixture of 65 volumes of water,
certain strains of Penicillium notatum or related organisms 35 volumes of acetonitrile and 1 volume ofglacial acetic
on a culture medium containing an appropriate precursor, or acid,
obtained by any other means. - flow rate. 1 ml per minute,
- spectrophotometer set at 254 nm,
Phenoxymethylpenicillin Potassium· contains not less than
injection volume. 20 Ill.
86.0 per cent of total penicillins CI6HI7NzOsS, calculated on
the anhydrous basis. Inject the reference solution. The tailing factor is not more
than 1.5 and the relative standard deviation for replicate
Category. Antibacterial.
injections is not more than 2.0 per cent.
Dose. For an adult, the equivalent of 250 to 500 mg of
Inject alternately the test solution and the reference solution.
phenoxymethylpenicillin every 6 hours, at least 30 minutes
before food. For a child every 6 hours, upto 1 year, 62.5 mg; 1 Calculate the content of phenoxyacetic acid.
to 5 years, 125 mg; and 6 to 12 years, 250 mg.
Limit ofp-hydroxyphenoxymethylpenicillin. Not more than
Description. A white, crystalline powder. 5.0 per cent.

1896

www.webofpharma.com
IP 2010 PHENOXYMETHYLPENICILLIN POTASSIUM TABLETS

Using the chromatograms obtained with the test solution in Phenoxymethylpenicillin Potassium
the Assay, calculate the content ofp-hydroxyphenoxymethyl-
penicillin from the peak response of p-hydroxyphenoxy~ Tablets
methylpenicillin and the sum of the peak responses of Phenoxymethylpenicillin Tablets; Penicillin V Potassium
p-hydroxyphenoxymethylpenicillin and phenoxymethyl-
Tablets; Penicillin V Tablets
penicillin.
Phenoxymethylpenicillin Potassium Tablets contain not less
Water (2.3.43). Not more than 1.0 per cent, determined on than 92.5 per cent and not more than 107.5 per cent of the
1.0 g. stated amount ofphenoxymethylpenicillin, CI6HISNzOsS.
Assay. Determine by liquid chromatography (2.4.14). Usual strengths. The equivalent of62.5 mg, 125 mg, 250 mg
Test solution. Dissolve about 125 mg of the substance under and 500 mg ofphenoxymethylpenicillin.
examination in the mobile phase and dilute to 50.0 ml with the
Identification
mobile phase.
A. Shake a quantity ofthe powdered tablets containing 80 mg
Reference solution (a). Dissolve an accurately weighed
ofphenoxymethylpenicillin with water, dilute to 250 ml with
quantity of phenoxymethylpenicillin potassium RS in the
water and filter. When examined between 230 and· 360 nm
mobile phase and dilute to obtain a solution having a known
(2.4.7), the filtrate shows absorption maxima at about 268 nm
concentration of about 2.5 mg per ml.
and 274 nm and a minimum at about 272 urn.
Reference solution (b). A solution in the mobile phase
B. Shake a quantity ofthe powdered tablets containing 10 mg
containing 0.25 per cent w/v each ofbenzylpenicillin potassium
ofphenoxymethylpenicillin with I 0 ml of water, filter and add
and phenoxymethylpenicillin potassium.
0.5 ml of neutral red solution. Add sufficient 0;01 M sodium
Chromatographic system hydroxide to produce a permanent orange colour and then
- a stainless steel column 30 cm x 4 rom, packed with add 1.0 ml of penicillinase solution; the solution changes
octadecylsilane chemically bonded to porous silica rapidly to red.
(3 to 10 Jlm), C. Ignite 0.5 g of the powdered tablets, add 5 ml of 2 M
- mobile phase: a mixture of 65 volumes of water, hydrochloric acid, boil, cool and filter. The filtrate gives
35 volumes of acetonitrile and 5.75 volumes of glacial reaction B ofpotassium salts (2.3.1).
acetic acid,
- flow rate. 1 ml per minute, Tests
- spectrophotometer set at 254 nm,
Dissolution (2.5.2).
- injection volume. 10 Jll.
Apparatus No.1,
Inject reference solution (b).The relative retention times are
Medium. 900 m1 of watel;
about 0.8 for benzylpenicillin and 1.0 for phenoxymethyl-
Speed and time. 50 rpm and 45 minutes.
penicillin. The column efficiency determined from the
phenoxymethylpenicillin peak is not less than Withdraw a suitable volume ofthe medium and filter. Measure
1800 theoretical plates and the resolution between benzyl- the absorbance ofthe filtrate, suitably diluted if necessary, at
penicillin and phenoxymethylpenicillin is not less than 3.0. the maximum at about 268 urn (2.4.7). At the same time measure
the absorbance of a solution of known concentration of
Inject reference solution (a). The relative standard deviation
phenoxymethylpenicillin potassium RS at the maximum at
for replicate injections is not more than 1.0 per cent.
about 268 urn. Calculate the content of CI6HlsNzOsS, in the
Inject the test solution and reference solution (a). Record the medium.
chromatograms and measure the responses for the D. Not less than 75 per cent of the stated amount of
phenoxymethylpenicillin peak and any p-hydroxyphenoxy- CI6HlsNzOsS.
methylpenicillin peak with a retention time ofabout 0.4 relative
Other tests. Complies with the tests stated under Tablets.
to that ofthe main phenoxymethylpenicillin peak.
Assay. Determine by liquid chromatography (2.4.14).
Calculate the content of CI6HI7NzOsS, from the sum of the
peak responses ofthep-hydroxyphenoxymethylpenicillin and Test solution. Weigh and finely powder 20 tablets. Dissolve
phenoxymethylpenicillin peaks in the chromatograms obtained an accurately weighed quantity ofthe powder containing about
with the test solution and reference solution (a). 0.25 g of phenoxymethy1penicillin in the mobile phase by
shaking for 5 minutes and dilute to 100.0 ml with the mobile
Storage. Store protected from moisture. phase. Filter through a 0.5 Jlm or finer filter and use the filtrate.

1897

www.webofpharma.com
PHENOXYMETHYLPENICILLIN POTASSIUM TABLETS IP 2010

Reference solution (a). Dissolve an accurately weighed Phentolamine Mesylate is 3-[[4,5-dihydro-1H-imidazol-2-


quantity of phenoxymethylpenicillin potassium RS in the yl)methy1](4-methylphenyl)aminophenol
mobile phase and dilute to obtain a solution having a known methanesulphonate.
concentration ofabout 2.5 mg per ml.
Phentolamine Mesylate contains not less than 99.0 per cent
Reference solution (b). A solution in the. mobile phase and not more than 100.5 per cent of C 17H I9N 30, CH4 0 3 S,
containing 0.25 per cent w/v each ofbenzylpenicillin potassium calculated on the dried basis.
and phenoxyinethylpenicillin potassium.
Category. Alpha-adrenoceptor antagonist.
Chromatographic system
Dose. By intravenous injection, 5 to 10 mg.
- a stainless steel column 30 cm x 4 mm, packed with
octadecylsilane chemically bonded to porous silica,
- mobile phase: a mixture of 650 volumes of water, Identification
350 volumes of acetonitrile and 5.75 volumes ofglacial Test A may be omitted iftests B, C andD are carried out. Tests
acetic acid, B, C and D may be omitted if test A is carried out.
- flow rate. 1 ml per minute,
- spectrophotometer set at 254 nm, A. Determine by infrared absorption spectrophotometry(2A.6).
- injection volume. 10 Jll. Compare the spectrum with that obtained with phentolamine
mesylate RS or with the reference spectrum of phentolamine
Inject reference solution (b). The relative retention times are
mesylate.
about 0.8 for benzylpenicillin and 1.0 for phenoxymethyl-
penicillin. The column efficiency determined from the B. When examined in the range 230 nm to 360 nm (204.7), a
phenoxymethylpenicillin peak is not less than 1800 theoretical 0.002 per cent w/v solution shows an absorption maximum
plates and the resolution between the benzylpenicillin and only at about 278 nm; absorbance at about 278 nm, about 0.5.
phenoxymethylpenicillin peaks is not less than 3.0.
C. Dissolve 0.5 gin 5 ml of ethanol (95 per cent) and 5 mlof
Inject reference solution (b). The relative standard deviation 0.1 M hydrochloric acid and add 2 ml of a 0.5 per cent w/v
for replicate injections is not more than 1.0 per cent. solution of ammonium metavanadate; a light green precipitate
is produced.
Inject the test solution and reference solution (a). Record the
chromatograms and measure the responses for the D. Mix 50 mg with 0.2 g ofpowdered sodium hydroxide, heat
phenoxymethylpenicillin peak and any p-hydroxyphenoxy- to fusion and continue the heating for a few seconds longer.
methylpenicillin peak with a retention time ofabout 004 relative Cool, add 0.5 ml of water and a slight excess of 2 M
to that ofthe main phenoxymethylpenicillin peak. hydrochloric acid amI warm; szdphur dioxide is evolved,
which turns moistened starch iodate paper blue.
Calculate the content of C16HI7NzOsS, from the sum of the
peak responses ofthe p-hydroxyphenoxymethylpenicillin and
phenoxymethylpenicillin peaks in the chromatograms obtained Tests
with the test solution and reference solution (a)
Acidity or alkalinity. Dissolve 0.1 gin 10 m1 of carbon dioxide-
Storage. Store protected from moisture. free water and add 0.1 ml of methyl red solution. The solution
is not red and not more than 0.05 ml of 0.1 Msodium hydroxide
Labelling. The label states the strength in terms of the
is required to change the colour of the solution.
equivalent amount ofphenoxymethylpenicillin.
Related substances. Determine by thin-layer chromatography
(204.17), coating the plate with silica gel G.
Phentolamine Mesylate Mobile phase. A mixture of 85 volumes of 2-butanone,
15 volumes of acetone and 5 volumes of strong ammonia
solution.
Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml of ethanol (95 per cent).
Reference solution. A 0.01 per cent w/v solution of the
substance under examination in ethanol (95 per cent).
Apply to the plate 10 Jll of each solution. After development,
dry the plate in air, spray with dilute potassium
Mol. Wt. 377.5 iodobismuthate solution. Any secondary spot in the

1898

www.webofpharma.com
IP 2010 PHENYLEPHRINE HYDROCHLORlDE

chromatogram obtained with the test solution is not more Phenylephrine Hydrochloride
intense than the spot in the chromatogram obtained with the
reference solution.
H,,,OH H
Sulphated ash (2.3.18). Not more than 0.1 per cent.
HO~N'CH3 ,HCI
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105°.
Assay. Weigh accurately about 0.3 g and dissolve in 100 ml of ~H13N02,HCI Mol. Wt. 203.7
anhydrous 2-propanol with the aid ofultrasound ifnecessary. Phenylephrine Hydrochloride is (R)-I-(3-hydroxyphenyl)-2-
Titrate with 0.1 M tetrabutylammonium hydroxide in methylaminoethanol hydrochloride.
2-propanol. Determine the end-point potentiometrically
Phenylephrine Hydrochloride contains not less than 98.5 per
(2.4.25), using a glass electrode and a calomel electrode
cent and not more than 101.0 per cent of C9H 13N02, HCI,
containing a saturated solution of tetramethylammonium
calculated on the dried basis.
chloride in 2-propanol. Carry out a blank titration.
Category. Sympathomimetic.
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
0.03775 g ofC 17H 19N 30, CH40 3S. Dose. By subcutaneous or intramuscular injection, 2 to 5 mg;
by slow intravenous injection, 100 to 500 IJ.g; by intravenous
Storage. Store protected from light and moisture. infusion 5 to 20 mg in 500 ml, adjusted according to response.
Description. A white or almost white, crystalline powder.

Identification
Phentolamine Injection Test A may be omitted iftests B, C andD are carried out. Tests
Band Cmay be omitted if tests A and D are carried out.
Phentolamine Mesylate Injection
A. Determine by infrared absorption spectrophotometry (2.4.6).
Phentolamine Injection is a sterile solution of Phentolamine Compare the spectrum with that obtained with phenylephrine
Mesylate in Water for Injections containing Dextrose. hydrochloride RS or with the reference spectrum of
Phentolamine Injection contains not less than 95.0 per cent phenylephrine hydrochloride.
and not more than 105.0per cent of the stated amount of B. Dissolve about 10 mgin I ml of water and add 0.05 mlof
phentolamine mesylate, C 17H 19N 30, CH40 3S. cupric sulphate solution and I ml of 5 M sodium hydroxide;
Usual strength. 10 mg per ml. a violet colour is produced. Add I ml of ether and shake; the
etherlayer remains colourless.
Identification C. Dissolve 0.3 gin 3 ml of water, add I ml of 6 M ammonia
and initiate crystallisation by scratching the side of the tube
The residue obtained in the Assay melts at about 138° (2.4.21 ). with a glass rod. The melting range of the crystals, after
washing with iced water and drying at 105° for 2 hours, is 171°
Tests to 176°(2.4.21).
D. Gives reaction A ofchlorides (2.3.1).
pH (2.4.24). 3.5 to 5.0.

Other tests. Complies with the tests stated under Parenteral Tests
Preparations (Injections). Appearance of solution. Dissolve 2.0 g in 100 ml of carbon
Assay. Dilute an accurately measured volume containing about
dioxide-free water prepared from distilled water (solution
A). Solution A is clear (2.4.1), and colourless (2.4.1).
0.1 g ofPhentolamine Mesylate to 40 ml with water, add 20 ml
of a 20 per cent w/v solution of trichloroacetic acid, allow to Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of
stand for 3 hours, filter, wash the residue with two quantities, methyl red solution and 0.2 ml of 0.01 M sodium hydroxide.
each of 5 ml, of water and dry to constant weight at 105°. The solution is yellow and not more than 0.4 ml of 0.01 M
hydrochloric acid is required to change the colour of the
I g of the residue is equivalent to 0.8487 g of C 17H 19N 30,
solution to red.
C~03S,
Specific optical rotation (2.4.22). -43.0° to -47.0°. determined
Storage. Store protected from light. in solution A.

1899

www.webofpharma.com
PHENYLEPHRINE HYDROCHLORIDE IP 2010

Related substances. Determine by thin-layer chromatography Usual strength. 10 mg per ml.


(2.4.17), coating the plate with silica gel H.
Identification
Mobile phase. A mixture of 80 volumes of 2-propanol,
15 volumes of 10Mammonia and 5 volumes of chloroform. A. In the test for Related substances, the principal spot in the
Test solution. Dissolve 0.2 g of the substance under chromatogram obtained with test solution (b) c9rresponds to
examination in 10 ml of methanol. that in the chromatogram obtained with reference solution (b).

Reference solution (a). A 0.01 per cent w/v solution of the B. To a volume containing 10 mg of Phenylephrine
substance under examination in methanol. Hydrochloride add, if necessary, sufficient water to produce
1 ml and then add 0.05 ml of cupric sulphate solution and I ml
Reference solution (b). A 0.004 per cent w/v solution of the of 5 M sodium hydroxide; a violet colour is produced. Add
substance under examination in methanol. 1 ml of ether and shake; the ether layer remains colourless.
Apply to the plate 5 !J.I of each solution. After development, C. Gives reaction A ofchlorides (2.3.1).
dry the plate in cold air, spray with ninhydrin solution, heat at
105° for 10 minutes and examine in daylight. Any secondary Tests
spot in the chromatogram obtained with the test solution is
not moreintense than the spot in the chromatogram obtained pH (2.4.24). 4.5 to 6.5.
with reference solution (a) and not more than two such spots Related substances. Determine by thin-layer chromatography
are more intense than the spot in the chromatogram obtained (2.4.17), coating the plate with silica gel H.
with reference solution (b).
Mobile phase. A mixture of 80 volumes of 2-propanol,
Sulphates (2.3.17). 15 ml ofsolution A complies with the limit 15 volumes of 10Mammonia and 5 volumes of chloroform.
test for sulphates (500 ppm).
Test solution (a) Evaporate a volume ofthe injection containing
Phenones. To 10 ml of solution A add sufficient 0.01 M 20 mg ofPhenylephrine Hydrochloride to dryness and dissolve
hydrochloric acid to produce 50 ml. Absorbance of the the residue in 1 rol of methanol.
resulting solution at the maximum at about 310 om, not more
than 0.20 (2.4.7). Test solution (b). Dilute 1 volume of test solution (a) to
200 volumes with methanol.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Reference solution (a). Dilute 1 volume of test solution (b) to
Losson drying{2.4.19). Not more than 1.0 per cent,deterrnined 2.5 volumes with methanol.
on 1.0 g by drying in an oven at 105°.
Reference solution (b). A 0.01 per cent w/v solution of
Assay. Weigh accurately about 0.15 g, dissolve in a mixture of phenylephrine hydrochloride RS in methanol.
0.5 ml of 0.1 M hydrochloric acid and 80 ml of ethanol
(95 per cent) and titrate with 0.1 M ethanolic sodium Apply to the plate 5 !J.I of each solution. After development,
hydroxide determining the end-point potentiometrically dry the plate in cold air, spray with ninhydrin solution, heat at
(2.4.25). Record the volume added between the two inflections. 105° for 10 minutes and examine in daylight. Any secondary
spot in the chromatogram obtained with test solution (a) is
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to not more intense than the spot in the chromatogram obtained
0.02037 gofCgH 13NOz,HCl. with test solution (b) and not more than two such spots are
Storage. Store protected from light and moisture. more intense than the spot in the chromatogram obtained
with reference solution (a).
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Phenylephrine Injection Assay. To an accurately measured volume containing about
50 mg of Phenylephrine Hydrochloride add sufficient 0.5 M
Phenylephrine Hydrochloride Injection
sulphuric acid to produce 100.0 ml. Dilute 10.0 ml of this
Phenylephrine Injection is a sterile solution of Phenylephrine solution to 100.0 ml with 0.5 M sulphuric acid and measure
Hydrochloride in Water for Injections. the absorbance of the resulting solution at the maximum at
about 273 om (2.4.7). Calculate the content ofC gH I3NO z,HCl
Phenylephrine Injection contains not less than 95.0 per cent
taking 90 as the specific absorbance at 273 nm.
and not more than 105.0 per cent of the stated amount.of
phenylephrine hydrochloride, CgH 13NOz,HCl. Storage. Store protected from light.

1900

www.webofpharma.com
IP 2010 PHENYLMERCURIC NITRATE

Phenylmercuric Acetate thiocyanate. Repeat the operation without the substance under
examination. The difference between the titrations represents
the amount of ammonium thiocyanate required.
1 ml of 0.1 M ammonium thiocyanate is equivalent to
0.01684 g ofC gHgHg02 •
Storage. Store protected from light and moisture.
Mol. Wt. 336.7
Phenylmercuric Acetate is (acetato)phenylmercury.
Phenylmercuric Acetate contains not less than 98.0 per cent
Phenylmercuric Nitrate
and not more than 100.5 per cent ofC gH gHg02 • Phenylmercuric Nitrate is a mixture ofphenylmercuric nitrate,
C6H sHgN03 and phenylmercuric hydroxide, C 6H sHgOH.
Category. Pharmaceutical aid (antimicrobial preservative).
Phenylmercuric Nitrate contains not less than 62.5 per cent
Description. A white to creamy white, crystalline powder, or
and not more than 63.5 per cent ofmercury, Hg, calculated on
small white prisms or leaflets.
the dried basis.
Identification Category. Antiseptic; pharmaceutical aid (antimicrobial
preservative).
A. To 100 mg add 0.5 mlofnitricacid, warm gently until a dark
brown colour is produced and dilute with water to 10 ml; the Description. A white or pale yellow powder.
characteristic odour of nitrobenzene is produced.
Identification
B. To 100 mg add 0.5 ml of sulphuric acid and I ml of ethanol
(95 per cent) and warm; the characteristic odour of ethyl A. To 0.1 g add 3 ml of sulphuric acid; the mixture becomes
acetate is produced. yellow and the characteristic odour of nitrobenzene is
C. To 5 ml of a saturated solution in water, add a few drops of produced.
a freshly prepared 10 per cent w/v solution ofsodium sulphide; B. To 0.1 g add 45 ml of water and heat to boiling with shaking.
a white precipitate is formed which darkens slowly on boiling Cool, filter and add sufficient water to produce 50 ml (solution
and allowing to stand. A). To 1 ml ofsolution A add 1 mlof2 Mhydrochloricacid; a
white, flocculent precipitate is produced.
Tests
C. To 5 ml of solution A add 8 ml of water and 0.1 ml of a
Mercuric salts and heavy metals. Heat about 100 mg with freshly prepared 10 per cent w/v solution of sodium sulphide;
15 rn1 of water, cool and filter. To the filtrate add a few drops of a white precipitate is formed which darkens slowly on boiling
a freshly prepared 10 per cent w/v solution ofsodium sulphide; and allowing to stand.
the precipitate formed shows no immediate colour.
D. To 5 ml of solution A add 1 ml of 2 M hydrochloric acid,
Polymercurated benzene compounds. Shake 2.0 g with 100 rn1 2 m1 of dichloromethane and 0.2 ml of dithizone solution and
of acetone. Filter, wash the residue with successive portions shake; the lower layer is orange-yellow.
of acetone until a total of 50 ml is used, dry the residue at 105°
E. Solution A gives reaction ofnitrates (2.3.1).
for 1 hour and weigh. The weight of the residue does not
exceed 30 mg (1.5 per cent). Tests
Sulphated ash (2.3 .18). Not more than 0.2 per cent.
Appearance ofsolution. Solution A is colourless (2.4.1).
Assay. Weigh accurately about 0.4 g, transfer to a 100-ml flask,
Inorganic mercuric compounds. To a 10 ml ofsolution A add
add 15 ml of water, 5 ml ofjormic acid and 1 g ofzinc dust and
2 ml ofpotassium iodide solution and 3 ml of2 M hydrochloric
reflux for 30 minutes. Cool, filter and wash the filter paper and
acid and filter; the filtrate is colourless. Wash the precipitate
the amalgam with water until the washings are no longer acidic
with 2 ml of water, combine the filtrate and washings and add
to litmus. Dissolve the amalgam in 40 ml of 8 M nitric acid.
2 ml of2 M sodium hydroxide and sufficient water to produce
Heat on a water-bath for 3 minutes and add 0.5 g urea and
20 m!. 12 ml of the solution complies with the limit test for
sufficient 0.02 M potassium permanganate to produce a
heavy metals, Method A (2.3.13). Use lead standard solution
permanent pink colour. Cool and add hydrogen peroxide
(1 ppm Pb) to prepare the standard (0.1 per cent).
. solution to decolorise the solution. Add 1 ml of jerrie
ammonium sulphate solution and titrate with 0.1 M ammonium Sulphated ash (2.3 .18). Not more than 0.1 per cent.

1901

www.webofpharma.com
PHENYLPROPANOLAMINE HYDROCHLORIDE IP 2010

Loss on drying (2.4.19). Not more than 1.0 per cent, detennined D. Melting range (2.4.21). 194° to 197°
on 1.0 g by drying overphosphoruspentoxide at a pressure
E. Gives reaction A ofchlorides (2.3.1).
of 1.5 to 2.5 kPa for 24 hours.
Assay. Weigh accurately about 0.2 g and dissolve in a mixture Tests
of 90 ml of water and 10 ml of nitric acid. Add 2 ml offerric
ammonium sulphate solution and titrate with 0.1 M ammonium Appearance ofsolution. AS.0 per cent w/v solution in water is
thiocyanate until a persistent reddish yellow colour is clear (2.4.1) and colourless (2.4.1).
obtained. Repeat the operation without the substance under Acidity or alkalinity. To 10 ml of 5.0 per cent w/v solution in
examination. The difference between the titrations represents water, add 0.1 ml of methyl red solution and 0.2 ml of 0.01 M
the amount of ammonium thiocyanate required. sodium hydroxide. The solution turns yellow. Add 0.4 ml of
1 ml of 0.1 A1ammonium thiocyanate is equivalent to 0.02006 0.01 M hydrochloric acid. The solution turns red.
gofHg.
Related substances. Determine by thin-layer chromatography
Storage. Store protected from light and moi~ture. (2.4.17), coating the plate with silica gel H.
Mobile phase. Amixture of6 volumes of 13.5 M ammonia, 24
volumes of ethanol (95 per cent) and 70 volumes of butanol.

Phenylpropanolamine Hydrochloride Test solution (a). Dissolve 0.2 g of the substance under
examination in 10 ml of ethanol (95 per cent).
Test solution (b). Dilute 1.0 ml of test solution (a) to 10.0 ml
, Hel with ethanol (95 per cent).
Reference solution (a). Dissolve 20. mg of
phenylpropanolamine hydrochloride RS in 10 ml of ethanol
~HI3NO,HCl Mol. Wt.187.7 (95 per cent).
Phenylpropanolamine Hydrochloride is (IRS,2SR)-2-amino- Reference solution (b). Dilute 1.0 ml ofreference solution (a)
I-phenylpropan-l-01 hydrochloride. to 10.0 ml with ethanol (95 per cent).
Phenylpropanolamine Hydrochloride contains not less than Reference solution (c). Dissolve 20 mg of
99.0 percent and not more than 101.5 per cent ofCcjH I3NO,HCl, norpseudoephedrine hydrochloride RS in ethanol (95 per
calculated on the dried basis. cent), add 1 ml of test solution (a) and dilute to 10 mlwith
Category. Adrenoreceptor antagonist. ethanol (95 per cent).

Description. A white or almost white crystalline powder. Reference solution (d). Dissolve 60 mg of ammonium chloride
in 10 ml ofthe methanol.
Identification
Before use, spray the plate with a 2 per cent w/v solution of
Tests A and E may be omitted if tests B, C and D are carried disodium tetraborate, using 8 ml for a plate 100 mm by 200 mm
out. Tests B, C and D may be omitted if tests A and E are and dry in a stream of cold· air for 30 minutes. Apply to the
carried out. plate 10 f.tl of each solution as bands about 10 mm by 3 mm.
Allow the mobile phase to rise 10 cm. Dry the plate in a current
A. Determine by infrared absorption spectrophotometry (2.4.6).
ofwarm air, allow to cool, spray with a 0.2 per cent w/v solution
Compare the spectrum with that obtained with
of ninhydrin in ethanol (95 per cent) and heat at 110° for 15
phenylpropanolamine hydrochloride RSor with the reference
minutes. Any spot in the chromatogram obtained with test
spectrum of phenylpropanolamine hydrochloride.
solution (a) other than the principal spot and the spot
B. In the test for Related substances, the principal spot in the corresponding to ammonium chloride is not more intense than
chromatogram obtained with test solution (b) corresponds to the spot in the chromatogram obtained with reference solution
that in the chromatogram obtained with reference solution (a). (b) (1.0 per cent). The test is not validunless the chromatogram
C. To a 1.0 per cent w/v solution of the substance under obtained with reference solution (c) shows two clearly
examination in the water, add 0.2 ml of copper sulphate separated spots.
solution and 0.3 ml of dilute sodium hydroxide solution. A Phenylpropanonamine. Absorbance of a 2.0 per cent w/v
violet colour develops. Add 2 ml of ether and shake. A violet solution in 0.01 M hydrochloric acid at the maximum at about
precipitate is formed between the two layers. 283 urn (2.4.7) is not more than 0.1.

1902

www.webofpharma.com
IP 2010 PHENYTOIN

Heavy metals (2.3.13).12 ml of5.0 per cent w/v solution in Tests


water complies with the limit test for heavy metals, Method D
(20 ppm). Appearance of solution. Dissolve 1.0 g in a mixture of5 m1 of
1 M sodium hydroxide and 20 ml of water. The solution is
Sulphated ash (2.3.18). Not more than 0.1 per cent. clear (2.4.1) and not more intensely coloured than reference
Loss on drying (2.4.19). Not more than 0.5 per cent, determined solution BYS5 (2.4.1).
on 1.0 g by drying in an oven at 105°. Acidity or alkalinity. To 1.0 g, add 45 ml of water and boil for
Assay. Weigh accurately about 0.15 g and dissolve in a mixture 2 minutes. Allow to cool and filter. Wash the filter with carbon
of5 ml of 0.01 Mhydrochloric acid and 50 m1 of ethanol (95 dioxide-free water and dilute the combined filtrate and
per cent). Titrate with 0.1 M sodium hydroxide. Determine washings to 50 ml with the same solvent. To 10 ml of the
the end-point potentiometrically (2.4.25). Carry out a blank solution add 0.15 ml of methyl red solution; not more than 0.5
titration. ml of 0.01 M hydrochloric acid is required to change the
colour ofthe solution to red. To 10 ml ofthe solution add 0.15
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01877 g of
ml of bromothymol blue solution; not more than 0.5 ml of O. 01
~HI4CINO.
M sodium hydroxide is required to change the colour of the
solution to blue.
Related substances. Determine by thin-layer chromatography
Phenytoin (2.4.17), coating the plate with silica gel GF254.
Diphenylhydantoin Solvent mixture. Equal volumes of acetone and meth,anol.
Mobile phase. A mixture of 30 volumes of dioxan and 75
volumes of hexane.
Test solution (a). Dissolve 0.4 g of the substance under
examination in 10 ml ofthe solvent mixture.
Test solution (b). Dilute 1 ml oftest solution (a) to 20 ml with
the solvent mixture.
Reference solution (a). Dissolve 20 mg ofphenytoin RSin 10
Mol.Wt. 252.3 ml ofthe solvent mixture.
Phenytoin is 5,5-diphenylimidazolidine-2,4-dione. Reference solution (b). Dissolve 8 mg of benzophenone in
Phenytoin contains not less than 99.0 per cent and not more 100 ml ofthe solvent mixture.
than 101.0 per cent of ClsHI2NzOz, calculated on the dried Reference solution (c). Dissolve 8 mg of benzil in 100 ml of
basis. the solvent mixture.
Category. Anticonvulsant. Reference solution (d). Dilute 1 ml oftest solution (a) to 100
Description. A white or almost white, crystalline powder. ml with the solvent mixture.
Reference solution (e). Mix 1 ml ofreference solution (b) and
Identification 1 ml ofreference solution (c).
Test A may be omitted if tests Band C are carried out. Tests B Before use, wash the plate with a mixture of 30 volumes of
and C may be omitted if test A is carried out. dioxan and 75 volumes of hexane. Allow the plate to dry in air.
A. Determine by infrared absorption spectrophotometry (2.4.6). Apply to the plate 10 ,...1 of each solution. Dry the plate in a
Compare the spectrum with that obtained with phenytoin RS current ofcold air for 2 minutes. Allow the mobile phase to rise
or with the reference spectrum of phenytoin. 15 cm. Dry the plate in air and examine under ultraviolet light
at 254 nm. In the chromatogram obtained with test solution (a)
B. In the test for Related substances, the principal spot in the
any spot corresponding to benzophenone is not more intense
chromatogram obtained with test solution (b) corresponds to
than the spot in the chromatogram obtained with the reference
that in the chromatogram obtained with reference solution
solution (b) (0.2 per cent); any spot corresponding to benzil
(a).
is not more intense than the spot in the chromatogram obtained
C. To about 10mg, add 1 mlofwaterand 0.05 mlofammonia. with reference solution (c) (0.2 per cent) and any spot, apart
Heat until boiling begins. Add 0.05 ml of a 5 per cent w/v from the principal spot and any spot corresponding to benzo-
solution of copper sulphate in dilute ammonia and shake, a phenone and benzil, is not more intense than the spot in the
pink crystalline precipitate is formed. chromatogram obtained with reference solution (d) (1 per

1903

www.webofpharma.com
. PHENYTOIN SODIUM IP 2010

cent). The test is not valid unless the chromatogram obtained B. Dissolve 0.25 gin 5 ml of water and acidifY with dilute
with reference solution (e) shows two clearly separated spots. hydrochloric acid; a white precipitate is produced.
Heavy metals (2.3.13). 2.0 g complies with the limit test for C. Dissolve 0.1 g in 10 ml of a 10 per cent w/v solution of
heavy metals, Method D (10 ppm). pyridine, add 1 ml of cupric sulphate with pyridine solution
Sulphated ash (2.3.18); Not more than 0.1 per cent. and allow to stand. for 10 minutes; a blue precipitate is
produced. .
Loss ondrying (2.4.19). Not more than 0.5 per cent, detenhined
on LOg by drying in an oven at 105°. D. Incinerate 0.1 g; the residue after neutralisation with
hydrochloric acid and addition of 2 ml of water gives the
Assay. Weigh accurately about 0.2 g, dissolve in 50 ml of reactions ofsodium salts (2.3.1).
dimethylformamide. Titrate with 0.1 M sodium methoxide,
determining the end-point potentiometrically (2.4.25). Carry Tests
out a blank titration.
Appearance of solution. Suspend 1.0 g in 5 ml of water and
1 ml of 0.1 M sodium methoxide is equivalent to 0.02523 gof dilute to 20 ml with 0.1 M sodium hydroxide; the solution is
ClsHlzNzOz. clear (2.4.1), and not more intensely coloured than reference
Storage. Store protected from light. solution BYS6 (2.4.1).
Free phenytoin. Dissolve 0.3 gin 10 ml of a mixture of equal
volumes of pyridine and water and add 0.5 ml of dilute
Phenytoin Sodium phenolphthalein solution and 3 ml of silver nitrate-pyridine
reagent. Not more than 1.0 ml of 0.1 M sodium hydroxide is
Diphenylhydantoin Sodium required to change the colour of the solution to pink.
Related substances. Detenhine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 45 volumes of chloroform,
45 volumes of 2-propanol and 10 volumes ofstrong ammonia
solution.
Mol. Wt. 274.3 Test solution. Dissolve 0.4 g of the substance under
Phenytoin Sodium is 4-oxo-5,5-diphenyl-2-imidazolidin-2-01il.te examination in 10 ml of methanol.
Phenytoin Sodium contains not less than 98.0 per cent and Reference solution (a). A 0.04 per cent w/v solution of the
not more than 101.0 per cent ofClsHIINzNaOz, calculated on substance under examination in methanol.
the anhydrous basis. . Reference solution (b). A 0.02 per cent w/v solution of
Category. Anticonvulsant; antiarrhythmic. benzophenone in methanol.

Dose. As anticonvulsant, orally, 150 to 300 mg daily, increasing Apply to the plate 10 !J.l of each solution. After development,
gradually to 600 mg in accordance with the needs of patient. dry the plate at 80° for 5 minutes and examine in ultraviolet
In status epileptieus, by slow intravenous injection, 10 to 15 light at 254 nm. Any secondary spot in the chromatogram
mg per kg at a rate not exceeding 50 mg per minute as a loading obtained with the test solution is not more intense than the
dose; maintenance doses of 100 mg thereafter every 6 hours. spot in the chromatogram obtained with reference solution (a)
In arrhythmias, by intravenous injection, 3.5 to 5 mg per kg at and any spot corresponding to benzophenone is not more
a rate not exceeding 50 mg per minute and repeated once if intense than the spotin the chromatogram obtailled with
necessary. reference solution (b).

Description. A white powder; odourless; somewhat Heavy metals (2.3.13). 1.0 g complies with the limit test for
hygroscopic. heavy metals, Method B (20 ppm).
Water (2.3.43). Not more than 3.0 per cent, determined on 1.0 g.
Identification
Assay. Weigh accurately about 0.18 g, suspend in 2 ml of
Test A may be omitted iftests B, C andD are carried out. Tests water, add 8 ml of 0.05 M sulphuric acid and heat gently for
Band C may be omitted if tests A and D are carried out. 1 minute. Add 30 ml of methanol, cool. Titrate with 0.1 M
A. Detenhine by infrared absorption spectrophotometry (2.4.6). sodium hydroxide, determining the end-point
Compare the spectrum with that obtained with phenytoin potentiometrically (2.4.25). After the fIrst inflection, stop the
sodium RS or with the reference spectrum ofphenytoin sodium. addition of sodium hydroxide, add 5 ml of silver nitrate

1904

www.webofpharma.com
IP 2010 PHENYTOIN INJECTION

solution in pyridine, mix and continue the titration until a D. Incinerate 0.1 g; the residue after neutralisation with
second inflection is reached. Record the volume of 0.1 M hydrochloric acid and addition of 2 ml of water gives the
sodium hydroxide added between the two inflections. reactions of sodium salts (2.3.1).
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02743 g of
CjsHjjNzNaOz.
Tests
Storage. Store protected from moisture. Appearance of solution. Suspend 1.0 g in 5 ml of water and
dilute to 20 ml with 0.1 M sodium hydroxide; the solution is
clear (2.4.1), and not more intensely coloured than reference
solution BYS6 (2.4.1).
Phenytoin Injection Completeness of solution. The contents dissolve in the
quantity of the solvent recommended on the label and give a
Phenytoin Sodium Injection; Diphenylhydantoin Sodium clear solution.
injection
pH (2.4.24). 10.0 to 12.0, determined in a 5.0 per cent w/v
Phenytoin Injection is a sterile material consisting ofPhenytoin solution in the stated solvent.
Sodium with or without buffering agents and other excipients.
It is filled in a sealed container. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
The injection is constituted by dissolving the contents of the
sealed container in the requisite amount of sterile Water for Mobile phase. A mixture of 45 volumes of chloroform, .
Injection or other suitable solvent, immediately before use. 45 volumes of 2-propanol and 10 volumes of strong ammonia
solution.
The constituted solution complies with the requirements for
Clarity of solution and Particulate matter stated under Test solution. Dissolve 0.4 g of the substance under
Parenteral Preparations (Injections). examination in 10 ml of methanol.
Storage. The constituted solution should be used immediately Reference solution (a). A 0.04 per cent w/v solution of the
after preparation but, in any case, within the period substance under examination in methanol.
recommended by the manufacturer.
Reference solution (b). A 0.02 per cent w/v solution of
Phenytoin Injection contains not less than 90.0 per cent and benzophenone in methanol.
not more than 110.0 per centofthe stated amount ofphenytoin
sodium, ClsHjjNzNaOz. Apply to the plate 10 p.l of each solution. After development,
dry the plate at 800 for 5 minutes and examine in ultraviolet
Usual strengths. 100 mg; 250 mg. light at 254 nm. Any secondary spot in the chromatogram
Description. A white powder; odourless; somewhat obtained with the test solution is not more intense than the
hygroscopic. spot in the chromatogram obtained with reference solution (a)
and any spot corresponding to benzophenone is not more
The contents of the sealed container comply with the
intense than the spot in the chromatogram obtained with
requir~ments stated under Parenteral Preparations
reference solution (b).
(Powders for Injection) and with thefollowing requirements.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Identification heavy metals, Method B (20 ppm).

Test A may be omitted iftests B, C andD are carried out. Tests Water (2.3.43). Not more than 3.0 per cent, determined on
Band C may be omitted if tests A and D are carried out. l.Og.

A. Determine by infrared absorption spectrophotometry (2.4.6). Assay. Weigh accurately about 0.18 g ofthe mixed contents of
Compare the spectrum with that obtained with phenytoin 10 containers, suspend in 2 ml of water, add 8 ml of 0.05 M
sodium RS or with the reference spectrum ofphenytoin sodium. sulphuric acid and heat gently for 1 minute. Add 30 ml of
methanol, cool. Titrate with 0.1 M sodium hydroxide,
B. Dissolve 0.25 g in 5 ml of water and acidify with dilute
determining the end-point potentiometrically (2.425). After
hydrochloric acid; a white precipitate is produced.
the first inflection, stop the addition of sodium hydroxide,
C. Dissolve 0.1 g in 10 ml of a 10 per cent w/v solution of add 5 ml ofsilver nitrate solution inpyridine, mix and continue
pyridine, add 1 ml of cupric sulphate with pyridine solution the titration until a second inflection is reached. Record the
and allow to stand for 10 minutes; a blue precipitate is volume of 0.1 M sodium hydroxide added between the two
produced. inflections.

1905

www.webofpharma.com
PHENYTOIN CAPSULES IP 2010

1 m1 of 0.1 M sodium hydroxide is equivalent to 0.02743 g of Apply to the plate 10 l!1 of each solution. After development,
ClsHIINzNaOz. dry at 80° for 5 minutes and examine in ultraviolet light at 254
nm. Any spot in the chromatogram obtained with the test
Storage. Store protected from moisture at a temperature not
solution corresponding to benzophenone is not more intense
exceeding 30°.
than the spot in the chromatogram obtained with reference
Labelling. The label states (1) the quantity of Phenytoin solution (b) (0.5 per cent) and any other secondary spot is not
Sodium contained in it; (2) the directions for preparing the more intense than the spot in the chromatogram obtained
Injection. with reference solution (a) (0.5 per cent).
Other tests. Comply with the tests stated under Capsules.
Assay. Shake a quantity ofthe mixed contents of20 capsules
containing about 0.25 g of Phenytoin Sodium with 50 ml of
Phenytoin Capsules 0.01 M sodium hydroxide. Centrifuge, acidify 25 ml of the
Phenytoin Sodium Capsules; Diphenylhydantoin Sodium clear liquid with 10 ml of 0.1 M hydrochloric acid and extract
Capsules· with successive quantities of50, 40 and 25 ml of ether. Wash
the combined extracts with 10 ml of water, evaporate to
Phenytoin Capsules contain not less than 92.7 per cent and
dryness and dry the residue at 105°. Dissolve in 50 ml of
not more than 107.5 per cent ofthe stated amount ofphenytoin
anhydrous pyridine. Titrate with 0.1 M tetrabutylammonium
sodium, CISHIINzNaOz. hydroxide using 0.3 per cent w/v solution of thymol blue in
Usual strength. 10 mg. anhydrous pyridine as indicator.
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
Identification 0.02743 g ofClsHIINzNaOz.
A. Centrifuge the precipitated mixture obtained in test B,
dissolve the residue in methanol, evaporate and dry at 105°
for 30 minutes. The residue complies with the following test.
Phenytoin Tablets
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with phenytoin Phenytoin Sodium Tablets; Diphenylhydantoin Sodium
sodium RS or with the reference spectrum ofphenytoin sodium. Tablets
B. Shake a quantity of the content of capsules containing Phenytoin Tablets contain not less than 90.0 per cent and not
about 0.5 g ofPhenytoin Sodium with 10 ml ofwater and filter. more than 110.0 per cent of the stated amount of phenytoin
The filtrate yields a white precipitate on the addition of 2 M sodium, ClsHIINzNaOz. The tablets are coated.
hydrochloric acid. Usual strengths. 50 mg; 100 mg.
C. To 0.1 g ofthe residue obtained in testA, add 0.5 ml of 1 M
Identification
sodium hydroxide, 10 ml of a 10per cent w/vsolution of
pyridine and 1 ml of copper sulphate-pyridine reagent and A. Shake a quantity of the powdered tablets containing 0.1 g
allow to stand for 10 minutes. A blue precipitate is produced. of Phenytoin Sodium with 20 ml of water, filter, acidify with
dilute hydrochloric acid and extract with 10 ml of chloroform.
Tests Wash the chlorofonn extract with water, dry with anhydrous
sodium sulphate and evaporate to dryness and dry the residue
Related substances. Determine by thin-layer chromatography
at 105°. The residue complies with the following test.
(2.4.17), coating the plate with silica gel GF254.
DetermIne by infrared absorption spectrophotometry (2.4.6).
Mobile phase. A mixture of 10 volumes of 13.5 M ammonia,
Compare the spectrum with that obtained with phenytoin
45 volumes of chloroform and 45 volumes ofpropan-2-ol.
sodium sodium RS treated in the same manner or with the
Test solution. Shake a quantity of the content of capsules reference spectrum of phenytoin sodium.
containing about 0.2 g of Phenytoin Sodium with 5 m1 of
B. Triturate a quantity ofthepowdered tablets containing 0.5
methanol, warm on a water-bath with shaking and filter.
g ofPhenytoin Sodium with 10 ml of water and filter. Acidify
Reference solution (a). Dilute 1 ml ofthe test solution to 200 with dilute hydrochloric acid; a white precipitate is produced.
ml with methanol.
C. The powdered tablets, when moistened with hydrochloric
Reference solution (b). A 0.02 per cent w/v solution of acid and introduced on a platinum wire into a flame, impart a
benzophenone in methanol. yellow colour to the flame.

1906

www.webofpharma.com
IP 2010 PHENYTOIN ORAL SUSPENSION

Tests When stored at the temperature and for the period stated on
the label during which the constituted suspension may be
Related substances. Detennine bythin-layer chromatography expected to be satisfactory for use, it contains not less than
(2.4.17), coating the plate with silica gel GF254. 80.0 per cent of the stated amount of phenytoin, ClsH12N202.
Mobile phase. A mixture of 75 volumes of hexane and Usual strength. 25 mg per m!.
30 volumes of dioxan.
Storage. Store protected from moisture at a temperature not
Test solution. Shake a quantity of the powdered tablets exceeding 30°.
containing 0.1 g ofPhenytoin Sodium with 5 ml of methanol,
wann on a water-bath with shaking and filter. The constituted suspension complies with the tests stated
under Oral Liquids and with the following tests.
Reference solution. A 0.01 per cent w/v solution of
benzophenone in methanol. Identificatio~
Apply to the plate 5 III of each solution. Allow the mobile
Detennine by infrared absorption spectrophotometry (2.4.6).
phase to rise 12 cm. Dry the plate in air and examine in ultraviolet
Compare the spectrum with that obtain with phenytoin RS or
light at 254 nm. In the chromatogram obtained with the test
with the reference spectrum of phenytoin.
solution any spot corresponding tobenzophenone is not more
intense than the spot in the chromatogram obtained with the Tests
reference solution.
pH (2.4.24).4.5 to 5.5 determined on 1.0 g.
Other tests. Comply with the tests stated under Tablets.
Benzil and benzophenone. Determine by thin-layer
Assay. Weigh and powder 20 tablets. Weigh accurately a
chromatography (2.4.17), coating the plate with silica gel
quantity of the powder containing about 0.25 g of Phenytoin
GF254.
Sodium, shake with 40 ml of 0.01 M sodium hydroxide for
5 minutes and add sufficient 0.01 M sodium hydroxide to Mobile phase. A mixture 000 volumes of 1, 4-dioxan and 75
produce 50.0 m!. Centrifuge, aciditY 25.0 rnl ofthe clear liquid volumes of hexane.
with 10 ml of 0.1 M hydrochloric acid and extract successively Test solution. Disperse a quantity of suspension containing
with 50, 40, 25 and 25 ml of ether. Wash the combined extracts about 30 mg of Phenytoin in 5 ml of water add 2 ml of 2 M
with 10 ml of water, evaporate to dryness and dry the residue hydrochloric acid, mix well and extract with five 20 ml quantities
at 105°. Dissolve in 50 m1 of anhydrous pyridine and titrate of ether. Combine the ether extracts, wash with three 10 ml
with 0.1 Mtetrabutylammonium hydroxide, using 0.3 per cent quantities of water, evaporate to dryness and dissolve the
w/v solution of thymol blue in pyridine as indicator and taking residue in 1.5 ml ofa mixture on volume ofglacial acetic acid
care to prevent absorption of carbon dioxide from the and 9 volumes of acetone.
atmosphere. Carry out a blank titration.
Reference solution (a). A 0.004 per cent w/v solution of
1 m1 of 0.1 M tetrabutylammonium hydroxide is equivalent to benzophenone in ethanol (95 per cent).
0.02743 g ofClsHIIN2Na02.
Reference solution (b). A 0.004 per cent w/v solution of benzil
Storage. Store protected from moisture.
in ethanol (95 per cent).
Apply to the plate 5 III of each solution. Allow the mobile
phase to.rise 12 cm. Dry the plate in air and examine in ultraviolet
Phenytoin Oral Suspension light at 254 nm. Any secondary spot in the chromatogram
Diphenylhydantoin Oral Suspension obtained with the test solution is not more intense than the
spot in the chromatogram obtain with reference solution (a)
Phenytoin Oral Suspension is a mixture of consisting of (0.2 per cent) and not more than one such spot is more intense
Phenytoin with buffering agents and other excipients. It than the spot in the chromatogram obtained with reference
contains a suitable flavouring agent. It is filled in a sealed solution (b) (0.2 per cent).
container.
Assay. Disperse a quantity ofthe suspension containing about
The suspension is constituted by dispersing the contents of
0.2 g of Phenytoin, add 10 ml of 2 M hydrochloric acid and
the sealedcontainer in the specific volume ofWaterjust before
15 ml of water, extract with three 50 ml quantities ofa mixture
use.
00 volumes of chloroform and 1volume ofpropan-2-01 and
Phenytoin Oral Suspension contains not less than 90.0 per evaporate the combined extract to dryness. Dry the residue at
cent and not more than 110.0 per cent of the stated amount of 105° for 1 hour, cool and dissolve in a mixture 00 ml of· 1 M
phenytoin, ClsHI2N202. sodium hydroxide and 50 ml of water with the aid of gentle

1907

www.webofpharma.com
PHOLCODINE IP 2010

heat. Cool and pass a current of carbon dioxide through the sodium nitrite, allow to stand for 15 minutes and add 3 ml of
solution until precipitation of phenytoin is complete, filter 6 M ammonia. The solution is not more intensely coloured
and wash the residue with two 10 ml quantities of water and than reference solution BS4 (2.4.1).
dry at 105° for 2 hours.
Related substances. Determine by thin-layer chromatography
Determine the weight per ml (2.4.29) ofthe oral suspension. (2.4.17), coating the plate with silica gel G
Calculate the content OfClSHI2N202, weight in volume. Mobile phase. A mixture of 70 volumes of ethanol (95 per
cent), 70 volumes of toluene, 65 volumes of acetone and
5 volumes of strong ammonia solution.
Pbolcodine Test solution. Dissolve 0.25 g of the substance under
examination in 10 ml of chloroform.
Reference solution (a). A 0.025 per cent w/v solution of the
substance under examination in chloroform.
Referencesolution (b). A 0.0125 per cent w/v solutioriofthe
substance under examination in chloroform.
Apply to the plate 10 Jll of each solution. After development,
dry the plate in air, spray with dilute potassium
C23H30N204,H20 Mol. Wt. 416.5 iodobismuthate solution. Any secondary spot in the
chromatogram obtained with the test solution is not more
Pholcodine is 7,8-didehydro-4,5a-epoxy-17-methyl-3-[2-
intense than the spot in the chromatogram obtained with
(morpholin-4-yl)ethoxy]morphinan-6a-ol monohydrate.
reference solution (a) and not more than one such spot of
Pholcodine contains not less than 98.0 per cent and not more higher Rf value than the principal spot is more intense than
than 101.0 per cent of C23H30N204, calculated on the dried the spot in the chromatogram obtained with reference solution
basis. (b).
Category. Cough suppressant. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Dos~. Upto 40 mg daily, in divided doses. Loss on drying (2.4.19). 3.9 to 4.5 per cent, determined on 0.5
Description. A white or almost white, crystalline powder. g by drying in an oven at 105°.

Identification Assay. Weigh accurately about 0.18 g, dissolve in 50 ml of


anhydrous glacial acetic acid, warming gently. Titrate with
A. Determine by infrared absorption spectrophotometry (2.4.6). 0.1 M perchloric acid, determining the end-point
Compare the spectrum with that obtained with pholcodine RS potentiometrically at the second inflection (2.4.25). Carry out
or with the reference spectrum of pholcodine. a blank titration.
B. To 10 ml ofa 0.1 per cent w/v solution add 75 ml of water 1 ml of 0.1 M perchloric acid is equivalent to 0.01993 g of
and 10 ml of 1 M sodium hydroxide and dilute to 100 ml with C23H30N204.
water. When examined in the range 230 nm and 360 nm (2.4.7),
the resulting solution shows an absorption maximum at about Storage. Store protected from moisture.
284 nm; absorbance at about 284 nm, 0.36 to 0.39.
C. Dissolve 50 mgin 1 ml of sulphuric acid and add 0.05 ml of
a 10 per cent w/v solution of ammonium molybdate; a pale
blue colour is produced. Warm gently; the colour changes to Pbolcodine Linctus
deep blue. Add 0.05 ml of2 M nitric acid; the colour changes
to brownish red. Pholcodine Linctus is a solution containing 0.1 per cent w/v
solution of Pholcodine and 1 per cent w/v solution of Citric
Tests Acid Monohydrate in a suitable flavoured vehicle.

Specific optical rotation (2.4.22). -94.0° to -98.0°, determined Pholcodine Linctus contains not less than 0.090 per cent and
at 20° in a 2.0 per cent w/v solution in ethanol (95 per cent). not more than 0.110 per cent w/v of pholcodine,
C23H30N204.H20.
Morphine. Dissolve 0.1 g in sufficient of 0.1 M hydrochloric
acid to produce 5 ml, add 2 ml of a 1 per cent w/v solution of Category. Opioid cough suppressant.

1908

www.webofpharma.com
IP 2010 PHOSPHORIC ACID

Identification Category. Pharmaceutical aid (acidifYing agent).

To 20 ml add 20 ml of water, make alkaline to litmus paper with Description. A clear, colourless, syrupy liquid; corrosive.
5 M ammonia, extract with two quantities, each of 20 ml, of When kept at a low temperature it may solidifY, producing a
chloroform, washing each extract with 5 ml of water, dry the mass of colourless crystals which do not melt until the
combined extracts with anhydrous sodium sulphate, filter and temperature reaches 28°.
evaporate to dryness. If necessary, add 0.1 ml of ether and
scratch the sides of the vessel with a glass rod to induce Identification
crystallisation. The crystals, dried at a pressure not exceeding
A. Dilute with water; the solution is strongly acidic.
2 kPa, comply with the following tests.
B. Dilute 10.0 g to 150 ml with water (solution A). Neutralise
A. Determine by infrared absorption spectrophotometry (2.4.6).
with 2 M sodium hydroxide; the resulting solution gives the
Compare the spectrum with that obtained with pholcodine RS
reactions of phosphates (2.3.1).
or with the reference spectrum of pholcodine.
B. When examined in the range 230 urn and 360 nm (2.4.7), a Tests
0.01 per cent w/v solution in 0.01 Msodium hydroxide shows
an absorption maximum only at about 284 nm. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
C. To a portion ofthe crystals add 0.05 ml of nitric acid and
mix; a yellow colour is produced. Arsenic (2.3.1 0). Dissolve 5.0 g in 50 ml of water and add 10 ml
of stannated hydrochloric acid; the resulting solution
D. Dissolve the remainder ofthe crystals in 1 ml of sulphuric
complies with the limit test for arsenic (2 ppm).
acid and add 0.05 ml of ammonium molybdate-sulphuric acid
solution; a pale blue colour is produced. Warm gently; the Heavy metals (2.3.13). Dilute 1.2 ml with 10 ml of water,
colour changes to deep blue. Add 0.05 ml of 2 M nitric acid; neutralise with dilute ammonia solution, add sufficient dilute
the colour changes to brownish red. acetic acid to render the solution acidic and then.dilute to
25 ml with water. The resulting solution· complies with the
Tests limit test for heavy metals, Method A (I 0 ppm).
Other tests. Complies with the tests stated under Oral Liquids.
Iron (2.3.14). 10 ml ofsolutionA complies with the limit testfor
Assay. Weigh accurately about 50 g and add sufficient 5 M iron (60 ppm).
ammonia to make the solution alkaline to litmus paper, extract
with four quantities, each of 25 ml, of chloroform, washing Chlorides (2.3.12). 3 ml ofsolution A complies with the limit
each extract with the same 5 ml of water. Combine the extracts test for chlorides (50 ppm).
and evaporate until the volume is reduced to 15 m!. Titrate Sulphates (2.3.17). 20 ml ofsolution A complies with the limit
with 0.02 M perchloric acid, using quinaldine red solution test for sulphates (100 ppm).
as indicator. Carry out a blank titration.
Alkali phosphates. To 1.7 g in a graduated cylinder add 6 ml of
1 ml of 0.02 Mperchloric acid is equivalent to 0.004165 g of
ether and 2 ml of ethanol (95 per cent); no turbidity is
~3H30Nz04,HzO.
produced.
Determine the weight perm! ofthe linctus (2.4.29), and calculate
the content ofCz3H30Nz04,HzO, weight in volume. Aluminium and calcium. To 1.7 g add 10 ml of water and 8 ml
of dilute ammonia solution; the solution remains clear.
Storage. Store protected from light.
Hypophosphorus acid and phosphorous acid. To 5 m! ofsolution
Labelling. The label states the strength in terms of the
A add 2 ml of a 1.7 per cent w/v solution of silver nitrate and
equivalent amount of pholcodine.
heat on a water-bath for 5 minutes; the appearance of the
solution does not change.

Assay. Weigh accurately about 1.0 g, add a solution oflO g of


Phosphoric Acid sodium chloride in 30 ml of water and titrate with1 Msodium
Orthophosphoric Acid; Concentrated Phosphoric Acid hydroxide using dilute phenolphthalein solution as indicator.

H3P04 Mol. Wt. 98.0 1 ml of 1 M sodium hydroxide is equivalent to 0.04900 g of


H 3P04•
Phosphoric acid contains not less than 84.0 per cent w/w
and not more than 90.0 per cent w/w ofH3P04. Storage. Store protected from moisture, in glass containers.

1909

www.webofpharma.com
PHYSOSTIGMINE SALICYLATE IP 2010

Physostigmine Salicylate water, and dilute to 100 ml with the same solvent (solutionA).
The solution, examined immediately after preparation, is clear
Eserine Salicylate (2.4.1), and colourless (2.4.1).
pH (2.4.24). 5.1 to 5.9, determined in solution A immediately
after preparation.

&
COO~H Specific opticalrotation (2.4.22). -90.0° to -94.0°, determined
in solution A immediately after preparation.
I"::
o Eseridine. To 5 ml of solution A, examined immediately after
preparation, add a few crystals of potassium iodate and a
drop of 2 M hydrochloric acid and 2 ml of chloroform and
shake; the cWoroform layer does not tum violet within 1·minute.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G

Mol. Wt. 413.5


Mobile phase. A mixture of 100 volumes of cyclohexane,
23 volumes of 2-propanol and 2 volumes of strong ammonia
Physostigmine Salicylate is (3aS,8aR)-1 ,2,3,3a,8,8a- solution.
hexahydro-l ,3a,8-trimethylpyrrolo[2,3-b]indol-5-yl
methylcarbamate salicylate. Test solution (a). Dissolve 0.2 g of the substance under
examination in 10 ml of ethanol (95 per cent).
Physostigmine Salicylate contains not less than 98.5 per cent
and not more than 101.0 per cent of ClsH21N302, C 7H 60 3, Test solution (b). Dissolve 0.1 g of the substance under
calculated on the dried basis. examination in 100 ml of ethanol (95 per cent).

Category. Anticholinesterase. Reference solution (a). A 0.1 per cent w/v solution of
physostigmine salicylate RS in ethanol (95 per cent).
Dose. By subcutaneous or intramuscular injection, 600 flg to
1.2 mg. Reference solution (b). A 0.01 per cent w/v solution of
physostigmine salicylate RS in ethanol (95 per cent).
Description. A colourless or faintly yellow crystals, turning
red gradually under the action of air and light and rapidly in Apply to the plate 20 fll of each solution. After development,
presence of moisture; odourless. dry the plate in cold air, carry out a second chromatographic
development in same direCtion, dry the plate in air and spray
Identification with freshly prepared acetic potassium iodobismuthate
solution and then with hydrogen peroxide solution (IO vol).
Test A may be omitted if tests B, C and D are carried out. Test Examine the plate within 2 minutes. Any secondary spot in the
C may be omitted if tests A, Band D are carried out. chromatogram obtained with test solution (a) is not more
intense than the spot in the chromatogram obtained with
A. Determine by infrared absorption spectrophotometry (2.4.6).
reference solution (b).
Compare the spectrum with that obtained with physostigmine
salicylate RS. Sulphates (2.3.17). 15 ml ofsolution A complies with the limit
test for sulphates (0.1 per cent).
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Sulphated ash (2.3.18). Not more than 0.1 per cent, determined
that in the chromatogram obtained with reference solution (a). on the residue obtained in the test for Loss on drying.
C. To a 1 per cent w/v solution add 1 M sodium hydroxide; a Loss on drying (2.4.19). Not more than 1.0 per cent, determined
white precipitate which turns pink is produced. The precipitate on 1.0 g by drying in an oven at 105°.
dissolves in an excess ofthe reagent, producing a red solution.
Assay. Weigh accurately about 0.35 g, dissolve in 50 ml of a
D. A 0.9 per cent w/v solution in carbon dioxide-free water mixture ofequal volumes of chloroform and anhydrous glacial
gives reaction A of salicylates (2.3.1). acetic acid. Titrate with 0.1 M perchloric acid, determining
the end-point potentiometrically (2.4.25). Carry out a blank
Tests titration.
Appearance of solution. Dissolve 0.9 g without heating in 1 ml of 0.1 M perchloric acid is equivalent to 0.04135 g of
95 ml of carbon dioxide-free water prepared from distilled ClsH2IN302,C7~03'

1910

www.webofpharma.com
IP 2010 PILOCARPINE NITRATE

Storage. Store protected from light and moisture. Pilocarpine Nitrate

Physostigmine Injection
~ NlJ+JO
f'l
I
\. I
CH 3
• HN0 3
H3 C HH
Physostigmine Salicylate Injection; Eserine Salicylate
Injection CIIHI6N202, RN03 Mol. Wt. 271.3
Pilocarpine Nitrate is (3S,4R)-3-ethyl-4-[(I-methyl-lH-
Physostigmine Injection is a sterile solution ofPhysostigmine
imidazol-5-yl)methyl]dihydrofuran-2(3H)-one nitrate..
Salicylate in Water for Injections.
Pilocarpine Nitrate contains not less than 98.5 per cent and
Physostigmine Injection contains not less than 90.0 per cent not more than 101.0 per cent ofCIIHI6N202, RN0 3, calculated
and not more than 110.0 per cent of the stated amount of on the dried basis.
physostigmine salicylate, ClsH21N302, C7H 60 3.
Category. Cholinergic (ophthalmic) in glaucoma.
Usual strength. 600 flg per ml.
Description. Colourless crystals or a white, crystalline powder;
odourless; sensitive to light.
Identification
Identification
A. Warm a volume containing 3 mg of Physostigmine
Salicylate with 0.3 ml of5 M ammonia; a yellowish red solution Test A may be omitted iftests B, C andD are carried out. Tests
is produced which on evaporation gives a bluish residue. Band C may be omitted if tests A andD are carried out.
A. Determine by infrared absorption spectrophotometry (2.4.6).
B. The residue obtained in testA dissolves in ethanol (95 per
Compare the spectrum with that obtained with pilocarpine
cent) producing a blue solution which, on the addition of
nitrate RS.
6 M acetic acid, appears blue by transmitted light and exhibits
a red fluorescence which intensifies on dilution with water. B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to
C. The residue obtained in test A dissolves in sulphuric acid that in the chromatogram obtained with reference solution (a).
producing a green solution which, on the gradual addition of
C. Dissolve about 10 mg in 2 ml of water, add 2 drops ofa 5 per
ethanol (95 per cent), changes to red but reverts to green
cent w/v solution ofpotassiumdichromate, 1 ml of hydrogen
when the ethanol is evaporated.
peroxide solution (10 vol) and 2 ml of chloroform and shake;
the chlorofonn layer turns violet.
Tests
D. Gives reaction A ofnitrates (2.3.1).
pH (2.4.24).4.0 to 6.0. Tests
Other tests. Complies with the tests stated under Parenteral Appearance ofsolution. A 5.0 per cent w/v solution in carbon
Preparations (Injections). dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution YS6 (2.4.1).
Assay. Transfer an accurately measured volume containing
30 mg of Physostigmine Salicylate to a separator, add about pH (2.4.24).3.5 to 4.5, determined in a 5.0 per cent w/v solution
0.25 g of sodium bicarbonate and extract with six quantities, prepared immediately before use in carbon dioxide-free water.
each of 15 ml, of chloroform. Filter the combined chloroform Specific optical rotation (2.4.22). +79.5° to +83.0°, determined
extracts through about 109 of anhydrous sodium sulphate. in a 5.0 per cent w/v solution in carbon dioxide-free water,
Add 25 ml of anhydrous glacial acetic acid to the filtrate. prepared immediately before use.
Titrate with 0.1 M perchloric acid, determining the end-point
Related substances. Determine by thin-layer chromatography
potentiometrically (2.4.25). Carry out a blank titration.
(2.4.17), coating the plate with silica gel G
1 ml of 0.01 Mperchloric acid is equivalent to 0.004135 g of Mobile phase. A mixture of 85 volumes of chloroform,
ClsH21N302, C7H 60 3· 14 volumes of methanol and 1 volume of strong ammonia
solution.
Storage. Store protected from light, in single dose containers.
The injection should not be used if it is more than slightly Test solution (a). Dissolve 0.3 g of the substance under
discoloured. examination in 10 ml of water.

1911

www.webofpharma.com
PILOCARPINE NITRATE IP 2010

Test solution (b). Dissolve 0.1 g of the substance under Pimozide contains not less than 99.0 per cent and not more
examination in 100 m1 ofwater. than 101.0 per cent ofCzsHz9FzN30, calculated on the dried
basis.
Reference solution (a). A 0.1 per cent w/v solution of
pilocarpine nitrate RS in water. Category. Antipsychotic.
Reference solution (b). A 0.03 per cent w/v solution of Description. A white to almost white powder.
pilocarpine nitrate RS in water.
Identification
Apply to the plate 10 III of each solution. After development,
dry the plate at 105° for 10 minutes, cool and spray with Test A may be omitted iftests B, C andD are carried out. Tests
potassium iodobismuthate solution. Any secondary spot in B, C and D may be omitted if test A is carried out.
the chromatogram obtained with test solution (a) is not more A. Determine by infrared absorption spectrophotometry (2.4.6).
intense than the spot in the chromatogram obtained with Compare the spectrum with that obtained with pimozide RS or
reference solution (b). with the reference spectrum ofpimozide.
Other alkaloids. To a 1 per cent wlv solution add dilute B. Determine by thin-layer chromatography (2.4.17), coating
ammonia solution; no turbidity is produced. To a 1 per cent the plate with silica gel G
w/v solution add a few drops of potassium dichromate
Mobile phase. A mixture of 10 volumes of acetone and 90
solution; no turbidity is produced.
volumes of methanol.
Chlorides (2.3.12). 25 m1 ofa 10.0 per cent w/v solution complies
Test solution. Dissolve 30 mg of the substance under
with the limit test for chlorides (100 ppm).
examination in 10 ml ofthe mobile phase.
Iron (2.3.14). 20 ml ofa 10.0 per cent w/v solution complies Reference solution (a). A 0.3 per cent w/v solution ofpimozide
with the limit test for iron (20 ppm). RS in the mobile phase.
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined Reference solution (b). A solution containing 0.3 per cent wi
on 0.5 g. veach ofpimozide RS and benperidol RS in the mobile phase.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Apply to the plate 10 III of each solution. Allow the mobile
on 1.0 g by drying in an oven at 105°. phase to rise 15 cm. Dry the plate in a current ofwarm air for 15
Assay. Weigh accurately about 0.25 g, dissolve in 30 ml of minutes and expose it to iodine vapour until the spots appear.
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric The principal spot in the chromatogram obtained with the test
acid, determining the end-point potentiometrically (2.4.25). solution corresponds to that in the chromatogram obtained
Carry out a blank titration. with reference solution (a). The test is not valid unless the
chromatogram obtained with reference solution (b) shows two
1 ml of 0.1 M perchloric acid is equivalent to 0.02713 g of
clearly separated spots.
CIIHI6NzOz, RN03.
C. Mix about 5 mg ofthe substance under examination with 45
Storage. Store protected from light and moisture.
mg ofheavy magnesium oxide and ignite in a crucible until an
almost white residue is obtained. Allow to cool, add 1 ml of
water, 0.05 ml ofphenolphthalein solution and about 1 mlof
Pimozide dilute hydrochloric acid to render the solution colourless.
Filter. To a freshly prepared mixture of 0.1 ml of alizarin S
solution and 0.1 ml ofzirconyl nitrate solution, add 1.0 ml of
F the filtrate. Mix allow to stand for 5 minutes and compare the
colour of the solution with that of a blank prepared in the
same manner. The test solution is yellow and the blank is red.
D. Melting point (2.4.21).216° to 220°.

Tests
F Appearance of solution. A 1.0 per cent w/v solution in
methanol is clear (2.4.1) and not more intensely coloured than
Mo1.Wt. 461.6 reference solution YS7 (2.4.1).
Pimozide is 1-[I-[4,4-bis(4-fluorophenyl)butyl]piperidin-4- Related substances. Determine by liquid chromatography
yl]-l ,3-dihydro-2H-benzimidazol-2-one. (2.4.14).

1912

www.webofpharma.com
IP 2010 PIMOZIDE TABLETS

Test solution. Dissolve 100 mg of the substance under Assay. Dissolve 0.3 g in 50 ml of a mixture of 1 volume of
examination in 10 ml of methanol. anhydrous glacial acetic acid and 7 volumes of methyl ethy(
Reference solution (a). Dissolve 5 mg of pimozide RS and 2 ketone and titrate with 0.1 M perchloric acid, using 0.2 ml of
mg of mebendazole RS in 100 ml of methanol. naphtholbenzein solution as indicator. Carry out a blank
titration.
Reference solution (b). Dilute 5.0 ml ofthe test solution to 100
ml with methanol. Dilute 1.0 ml ofthis solution to 10 ml with I ml 0.1 M perchloric acid is equivalent to 0.04616 g of
methanol. C2sH29F2N30.
Chromatographic system Storage. Store protected from light.
- a stainless steel column 10 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (3 /lm),
mobile phase: A. a solution containing 0.25 per cent
w/v of ammonium acetate and 0.85 per cent w/v of
Pimozide Tablets
tetrabutylammonium hydrogen sulphate, Pimozide Tablets contain not less than 95.0 per cent and not
B. acetonitrile, more than 105.0 per cent of the stated amount ofpimozide,
flow rate. 2ml per minute, C2sH29F2N30.
a linear gradient programme using the conditions given
Usual strengths. 1 mg; 2 mg; 4 mg.
below,
spectrophotometer set at ·280 run, Identification
injection volume. 10 /ll.
A. Shake a quantity ofthe powdered tablets containing about
Time Mobile phase A Mobile phase B
20 mg ofPimozide with 20 rnl of dichloromethane for 5 minutes,
(in min.) (per cent v/v) (per cent v/v)
filter and evaporate the filtrate to dryness under reduced
0-10 80~70 20~30
pressure. On the residue, determine by infrared absorption
10-15 70 30 spectrophotometry (2.4.6). Compare the spectrum with that
15-20 80 20 obtained with pimozide RS or with the reference spectrum of
Inject reference solution (a). The test is not valid unless the pimozide.
resolution between the peaks due to mebendazole and pimozide B. In the Assay, the principal peak in the chromatogram
is not less than 5.0. . obtained with the test solution corresponds to the peak in the
Inject the test solution and reference solution (b). In the chromatogram obtained with the reference solution.
chromatogram obtained with the test solution, the area ofany
Tests
peak due to (1-(piperidin-4-yl)-1 ,3-dihydro-2H-benzimidazol-
2-one) (pimozide impurityA) , (1-[1-[(4RS)-4-(4-fluorophenyl)- Dissolution (2.5.2).
4-phenylbutyl]piperidin-4-yl]-1,3-dihydro-2H-benzimidazol-2- Apparatus No.1,
one) (pimozide impurity B), (1-[1-[(4RS)-4-(2-fluorophenyl)-4- Medium. 900 ml of 0.01 M hydrochlorIc acid,
(4-fluorophenyl)butyl]piperidin-4-yl]-1 ,3-dihydro-2H- Speed and time. 100 rpm and 45 minutes.
benzimidazol-2-one) (pimozide impurity C), (1-[ 1-[4,4-bis(4-
fluorophenyl)butyl]-1 ,2,3,6-tetrahydropyridin-4-yl]-1 ,3- Withdraw a suitable volume ofthe medium and filter.
dihydro-2H-benzimidazol-2-one) (pimozide impurity D), (1-[1- Determine by liquid chromatography (2.4.14).
[4,4-bis(4-fluorophenyl)butyl]piperidin-4-yl I-oxide]-1,3-
Test solution. Dilute the filtrate, if necessary with the
dihydro-2H-benzimidazol-2-one) (pimozide impurity E), is not
dissolution medium.
more than the area ofthe principal peak in the chromatogram
obtained with reference solution (b) (0.5 per cent). The sum of Reference solution. A 0.0002 per cent w/v solution ofpimozide
the areas of all the secondary peaks is not more than 1.5 times RS in dissolution medium.
the area of the principal peak in the chromatogram obtained
Use chromatographic system as described under Related
with reference solution (b) (0.75 per cent). Ignore any peak substances.
with an area less than 0.1 times the area ofthe principal peale in
the chromatogram obtained with reference solution (b) (0.05 Calculate the content ofpimozide, C2sH29F2N30.
per cent). D. Not less than 70 per cent of the stated amount of
Sulphated ash (2.3.18). Not more than 0.1 per cent. C2sH29F2N30.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Related substances. Determine by liquid chromatography
on 1.0 g by drying in an oven at 105°. (2.4.14).

1913

www.webofpharma.com
PIMOZIDE TABLETS IP 2010

Test solution. Shake a quantity ofpowdered tablets containing Reference solution (b). A solution containing 0.005 per cent
about 40 mg ofPimozide with 20 ml ofmethanol for 30 minutes, w/v of pimozide RS and 0.002 per cent w/v of mebendazole
mix with the aid ofultrasound for 10 minutes, centrifuge and RS in methanol.
filter the supernatant liquid.
Use chromatographic system as described under Related
Reference solution (a). Dilute 1 ml of the test solution to 200 substances.
ml with methanol.
Inject reference solution (b). The test is not valid unless the
Reference solution (b). A solution containing 0.005 per cent resolution between the peaks due to pimozide and mebendazole
w/v of pimozide RS and 0.002 per cent w/v of mebendazole is not less than 5.0.
RS in methanol.
Inject the test solution and reference solution (a).
Chromatographic system
Calculate the content ofCzsHz9FzN30 in each tablet.
- a stainless steel column 10 cm x 4.6 mm packed with
endcapped octadecylsilane bonded to porous silica (3 Other tests. Comply with the tests stated under Tablets.
/lm) (Such as Hypersil ODS), Assay. Determine by liquid chromatography (2.4.14).
mobile phase: A. a solution containing 0.25 per cent
w/v of ammonium acetate and 0.85 per cent w/vof Test solution. Shake a quantity ofpowdered tablets containing
tetrabutylammonium hydrogen sulphate, about 20 mg ofPimozide with 35 ml of methanol for 30 minutes,
B. acetonitrile, dilute to 50 ml with methanol, mix with the aid of ultrasound
a linear gradient programme using the conditions given for 10 minutes, centrifuge and filter the supernatant liquid.
below, Reference solution (a). A 0.04 per cent w/v solution of
flow rate. 2 ml per minute, pimozide RS in methanol.
spectrophotometer set at 280 nm,
Reference solution (b). A solution containing 0.004 per cent
injection volume. 10 Ill.
w/v of pimozide RS and 0.002 per cent w/v of mebendazole
Time Mobile phase A Mobile phase B RS in methanol.
(min.) (per cent v/v) (per cent v/v)
Use chromatographic system as described under Related
o 80 20 substances.
10 70 30
Inject reference solution (b). The test is not valid unless the
15 70 30 resolution between the peaks due to pimozide and mebendazole
16 80 20 is not less than 5.0.
30 80 20 Inject the test solution and reference solution (a).
Inject reference solution (b). The test is not valid unless the Calculate the content ofCzsHz9FzN30 in the tablets.
reSolution between the peaks obtained with mebendazole and
pimozide is not less than 5.0. Storage. Store protected from light.

Inject the test solution and reference solution (a). In the


chromatogram obtained with the test solution the area of any
secondary peak is not more than the area ofthe principal peak Pindolol
in the chromatogram obtained with reference solution (a) (0.5
per cent) and the sum of the areas of all secondary peaks is
not more than 1.5 times the area of the principal peak in the
chromatogram obtained with reference solution (a) (0.75 per
cent).
Uniformity of content (For tablets containing 10 mg or less).
Comply with the test stated under Tablets. Mol. wt. 248.3
Pindolol is (RS)-1-indol-4-yloxy-3-isopropylaminopropan-2-
Test solution. Shake one tablet with 7 ml of methanol for 30
01.
minutes, dilute with methanol to produce a solution containing
0.01 per cent w/v ofPimozide, mix with the aid ofultrasound Pindolol contains not less than 99.0 per cent and not more
for 10 minutes, centrifuge and filter the supernatant liquid. than 101.0 per cent of CIJIzoNzOz, calculated on the dried
basis.
Reference solution (a). A 0.01 per cent w/v solution of
pimozide RS in methanol. Category. Beta-adrenoceptor antagonist.

1914

www.webofpharma.com
IP 2010 PINDOLOL TABLETS

Dose. 10 to 30 mg daily. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method C (20 ppm).
Description. A white or almost white, crystalline powder..
Sulphated ash (2.3.18). Not more than 0.1 percent.
Identification
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Test A may be omitted if tests Band C are carried out. Test B on 1.0 g by drying in an oven at 105°.
and C may be omitted if test A is carried out.
Assay. Weigh accurately about 0.2 g, dissolve in 80 ml of
A. Determine by infrared absorption spectrophotometry (2.4.6). methanol. Titrate with 0.1 M hydrochloric acid, determining
Compare the spectrum with that obtained with pindolol RS. the end-point potentiometrically (2.4.25).
B. When examined in the range 230 nm to 360 nm (2.4.7), a 1 ml of 0.1 M hydrochloric acid is equivalent to 0.02483 ~ of
0.002 per cent w/v solution in a 0.085 per cent v/v solution of C1Ji20N20 2.
hydrochloric acid in methanol shows absorption maxima at
Storage. Store protected from light and moisture.
about 264 nm and at about 287 nm, and a shoulder at about
275 nm, absorbances at about 264 nm, 0.66 to 0.70 and at
about 287 nm, 0.34 to 0.38.
e. In the test for Related substances, the principal spot in the Pindolol Tablets
chromatogram obtained with test solution (b) corresponds to Pindolol Tablets contains not less than 90.0 per cent and not
that in the chromatogram obtained with reference solution (a). more than 11 0.0 per cent of the stated amount of pindolol,
C1Ji20N 20 2'
Tests
Usual strengths. 5 mg; 15 mg.
Appearance of solution. A 5.0 per cent w/v solution in dilute
acetic acid is clear (2.4.1) and not more intensely coloured Identification
than reference solution BYS4 (2.4.1).
A. Shake a quantity ofthe powdered tablets containing 50 mg
Related substances. Determine by thin-layer chromatography ofPindolol with 80 m1 of ether for 30 minutes, filter and dry the
(2.4.17), coating the plate with silica gel GF254. extract with anhydrous sodium sulphate. Filter the extract,
NOTE- Prepare the solutions immediately before use and remove the ether using a rotary evaporator and dry the residue
protected from light. over phosphoruspentoxide at 110° at a pressure not exceeding
2 kPa for 1 hour. The residue complies with the following test.
Solvent mixture. 99 volumes of methanol and 1 volume of
Determine by infrared absorption spectrophotometry (2.4.6).
anhydrous glacial acetic acid.
Compare the spectrum with that obtained with pindolol RS.
Mobile phase. A freshly prepared mixture of 50 volumes of
B. When examined in the range 230 nm to 360 nm (2.4.7), the
ethyl acetate, 50 volumes of methanol and 4 volumes ofstrong
final solution obtained in the Assay shows absorption maxima
ammonia solution.
at about 264 nm and 287 nm.
Test solution (a). Dissolve 0.2 g of the substance under
examination in 10 ml ofthe solvent mixture.
e. Shake a quantity ofthe powdered tablets containing 20 mg
of Pindolo1with 5 m1 ofa mixture of99 volumes of methanol
Test solution (b). Dissolve 0.2 g of the substance under and 1 volume ofglacial acetic acid for 45 minutes. Centrifuge
examination in 100 ml ofthe solvent mixture. and dilute 1 m1 of the supernatant liquid to 50 ml with the
acetic acid-methanol mixture. To 2 ml ofthis solution add 1 ml
Reference solution (a). A 0.2 per cent w/v solution ofpindolo I
of dimethylaminobenzaldehyde solution; a violet-blue colour
RS in the solvent mixture.
is produced.
Reference solution (b). A 0.006 per cent w/v solution of
pindolol RS in the solvent mixture. Tests
Apply to the plate 5 J!l of each solution. Allow the mobile Related substances. Determine by thin-layer chromatography
phase to rise 10 cm. Dry the plate immediately in a current of (2.4.17), coating the plate with silica gel GF254.
cold air and examine in ultraviolet at 254 nm without delay.
NOTE- Prepare the solutions immediately before use and
Any secondary spot in the chromatogram obtained with test
protectedfrom light.
solution (a) is not more intense than the spot in the
chromatogram obtained with reference solution (b) (0.3 per Solvent mixture. 99 volumes of methanol and 1 volume of
cent). anhydrous glacial acetic acid.

1915

www.webofpharma.com
PINDOLOL TABLETS IP 2010

Mobile phase. A freshly prepared mixture of 50 volumes of Pioglitazone Hydrochloride contains not less than 98.0 per
ethyl acetate, 50 volumes of methanol and 4 volumes of strong cent and not more than 102.0 per cent of C I9 Hzo N z 0 3S,HCl,
ammonia solution. calculated on the dried basis.
Test solution. Shake a quantity of the powdered tablets Category. Antidiabetic.
containing 20 mg ofPindolo1with 5 mI ofthe solvent mixture
Description. A white or almost white crystalline powder.
for 15 minutes, centrifuge and apply the supernatant liquid to
the plate as the last solution. Identification
Reference solution (a). Dilute 1 ml ofthe test solution to 10 ml A. Determine by infrared absorption spectrophotometry (2.4.6).
with the solvent mixture and further dilute 7 ml ofthe solution Compare the spectrum with that obtained with pioglitazone
to 100 ml with the solvent mixture. hydrochloride RS or with the reference spectrum of
Reference solution (b). Dilute 1 ml ofthe test solution to 10 ml pioglitazone hydrochloride.
with the solvent mixture and further dilute 3 ml ofthis solution B. Gives reaction A ofchlorides (2.3.1).
to 100 ml with the solvent mixture.
Tests
Apply to the plate 10 III of each solution. Allow the mobile
phase to rise 10 cm. Dry the plate in air, spray immediately with Appearance of solution. A 5.0 per cent w/v solution in
dimethylaminobenzaldehyde solution and warm at 50° for methanol is clear (2.4.1) and not more intensely coloured than
20 minutes. Any spot with R r value of about 0.1 in the reference solution BYS7 (2.4.1).
chromatogram obtained with the test solution is not more Related Substances. Detennine by liquid chromatography
intense than the spot in the chromatogram obtained with (2.4.14)
reference solution (a) (0.7 per cent). Any other secondary
spot in the chromatogram obtained with the test solution is Test solution. Dissolve 30 mg of the substance under
not more intense than the spot in the chromatogram obtained examination in 100.0 mI of methanol.
with reference solution (b) (0.3 per cent). Ignore any spot Reference solution (a). A 0.03 per cent w/v solution of
remaining on the line of application. pioglitazone hydrochloride RS in methanol.
Other tests. Comply with the tests stated under Tablets. Reference solution (b). Dilute 1.0 mI ofreference solution (a)
Assay. Weigh and powder 20 tablets. Weigh accurately a to 100.0 ml with methanol.
quantity of the powdered tablets containing about 90 mg of Chromatographic system
Pindolol and shake with 100.0 ml of methanol for 45 minutes. - a stainless steel column 25 cm x 4.6 mm, packed with
Centrifuge and dilute 15.0 ml of the supernatant liquid to octadecylsilane bonded to porous silica ( 5 Ilm),
100.0 mI with methanol and measure the absorbance of the - column temperature. 25°,
resulting solution at the maximum at about 264 nm (2.4.7). - mobile phase: a mixture of 50 volumes of 0.01 M
Calculate the content ofC'4HzoNzOz taking 338 as the specific potassium dihydrogen phosphate and 50 volumes of
absorbance at 264 nm. acetonitrile,
Storage. Store protected from light. - flow rate. 1 ml per minute,
- spectrophotometer set at 225 nm,
- injection volume. 20 Ill.
Inject reference solution (a). The test is not valid unless the
column efficiency is not less than 2500 theoretical plates, the
Pioglitazone Hydrochloride tailingfactQr is not more than 2.0 and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
Inject the blank, the test solution and reference solution (b).
In the chromatogram obtained with the test solution, the area
, Hel
of any secondary peak is not more than 0.5 times the area of
the peak in the chromatogram obtained with reference solution
(b) (0.5 per cent) and the sum ofthe areas ofall the secondary
peaks is not more than the area ofthe peak in the chromatogram
CI9HzoNz03S,HCl Mol.Wt. 392.9 obtained with reference solution (b) (1.0 per cent ).
Pioglitazone Hydrochloride is (RS)-5-{4-[2-(5-ethyl-2- Heavy metals (2.3.13). 1.0 g complies with the limit test for
pyridinyl)ethoxy]benzyl}thiazolidine-2,4-dione hydrocWoride. heavy metals, Method B (20 ppm ).

1916

www.webofpharma.com
IP 2010 PIOGLITAZONE TABLETS

Sulphated ash (2.3.18). Not more than 0.1 per cent. Test solution: Weigh and powder 20 tablets. Transfer a quantity
of the powder equivalent to 30 mg of pioglitazone and add
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
about 150 ml of methanol. Mix with the aid ofultrasound at a
on 1.0 g by dying in an oven at 105°.
temperature not exceeding 30° for 15 minutes and shake for 20
Assay. Determine by liquid chromatography (2.4.14) as minutes. Dilute to 250 ml with methanol and mix. Filter through
described under the test for Related substances. a membrane filter and discard the first 5 ml. Use the filtrate as
Inject the test solution and reference solution (a). the test solution.

Calculate the content ofC l9 H zo N z0 3S,HCl. Reference solution. A solution of pioglitazone hydrochloride
RS in methanol containing 0.00012 per cent w/v of
pioglitazone.
Chromatographic system
- a stainless steel column 25 cm x 4.6 rom, packed with
Pioglitazone Tablets octadecylsilane bonded to porous silica (5 I!m),
Pioglitazone Hydrochloride Tablets - mobile phase: a mixture of 50 volumes of 0.01 M
potassium dihydrogen phosphate and 50 volumes of
Pioglitazone Tablets contain not less than 90.0 per cent and
acetonitrile,
not more than 110.0 per cent of the stated amount of
- flow rate. 1 ml per minute,
pioglitazone, CI9HzoNz03 S.
- spectrophotometer set at 225 nm,
Usual strengths. 15 mg; 30 mg; 45 mg. - injection volume. 20 I!l.
Inject the test solution and the reference solution. In the
Identification
chromatogram obtained with the test solution, the area ofany
A. Shake a quantity of the powdered tablets containing 0.1 g secondary peak is not more than 0.5 times the area ofthe peak
of pioglitazone with 40 ml of methanol, filter and evaporate in the chromatogram obtained with the reference solution (0.5
the filtrate to dryness. The residue complies with the following per cent) and the sum of the areas of all the secondary peaks
test. - is not more than twice the area ofthe peak in the chromatogram
obtained with the reference solution (2.0 per cent).
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with pioglitazone Other tests. Comply with the tests stated under Tablets.
hydrochloride RS or with the reference spectrum of
Assay. Determine by liquid chromatography (2.4.14).
pioglitazone hydrochloride.
Solvent mixture. A mixture of 35 volumes of water and 65
B. In the Assay, the principal peak in the chromatogram
volumes of acetonitrile.
obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity ofthe powder containing about 30 mg ofpioglitazone,
Tests add about 20 ml of water and mix with the aid of ultrasound.
Add about 150 ml ofthe solvent mixture and mix with the aid
Dissolution (2.5.2).
ofultrasound for 15 minutes. Dilute to 250.0 ml with the solvent
Apparatus No. 1 mixture.
Medium. 900 ml of 0.1 M hydrochloric acid,
Reference solution. A 0.015 per cent w/v solution of
Speed and time. 75 rpm and 45 minutes.
pioglitazone hydrochloride RS in the solvent mixture.
Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance of the filtered solution, suitably diluted with Chromatographic system
the medium if necessary, at the maximum at about 270 nm - a stainless steel column 25 cm x 4.6 rom, packed with
(2.4.7). Calculate the content ofC19HzoNz03S in the medium octadecylsilane bonded to porous silica ( 5 I!m) ,
from the absorbance obtained from a solution of known mobile phase: a mixture of35 volumes ofa buffer solution
concentration ofpioglitazone hydrochloride RS in the same prepared by dissolving 1.36 g of potassium dihydrogen
medium. orthophosphate and 1.15 g of diammonium hydrogen
phosphate in 1000 ml of water, adding 1.0 ml of
D. Not less than 70 per cent of the stated amount of triethylamine and adjusting the pH to 5.0 with
CI9HzoNz03S, orthophosphoric acid, and 65 volumes of acetonitrile,
Related substances. Determine by liquid chromatography flow rate. 1.5 ml per minute,
(2.4.14). spectrophotometer set at 270 urn,

1917

www.webofpharma.com
PIPERACILLIN IP 2010

- injectionvolume.20.gl. Solvent mixture. 25 volumes of acetonitrile and 75 volumes


Inject the reference solution. The test is not valid unless the on .12 per cent w/v solution ofsodium dihydrogen phosphate.
tailing factor is not more than 2.0 and the relative standard Test solution (a). Dissolve 25 mg of the substance under
deviation forreplicate injections is not more than 2.0 per cent. examination in 50 m1 ofthe solvent mixture.
Inject the test solutioIl and the. reference solution. Test solution (b). Dissolve 40 mg of the substance under
,

Calculate the content ofCI9HzoNz03 S in the tablets. examination in 20 ml ofthe solvent mixture.
Labelling, The label state.s the strength in terms of the Reference solution (a). A 0.05 per cent w/v solution of
equivalent amount ofpioglitazone. piperacillin RS in the solvent mixture.
Reference solution (b). Dilute 1.0 ml ofreference solution (a)
to 25 ml with the solvent mixture.
Reference solution (c). Dissolve 10 mg of piperacillin RS
Piperacillin and 10 mg of anhydrous ampicillin RS (piperacillin impurity
A RS) in 50 ml solvent mixture.
Reference solution (d). Dilute 1.0 ml ofreference solution (a)
to 100 m1 with the solvent mixture. Dilute 1.0 ml ofthis solution
to 50 ml with the solvent mixture.
Chromatographic system
. H2 0
a stainless steel column 25 em x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 gm),
mobile phase: A. a mixture of576 volumes of water, 200
volumes of 3.12 per cent w/v solution of sodium
dihydrogen phosphate and 24 volumes of an 8.0 per
Mol.Wt. 535.6 cent w/v solution of tetrabutylammonium hydroxide,
Piperacillin is (6R)-6-[R-2-(4-ethyl-2,3-dioxopiperazine-l- adjusted to pH 5.5 with dilute phosphoric acid or dilute
carboxamido)-2-phenylacetamido]penicillanic acid sodium hydroxide solution; add 200 volumes of
monohydrate. acetonitrile,
B. a mixture 126 volumes of water, 200
Piperacillin contains not less than 96.0 per cent and not more
volumes of a 3.12 per cent w/v solution of sodium
than 102.0 per cent of CZ3Hz7Ns07S, calculated on the
dihydrogen phosphate and 24 volumes of an 8.0 per
anhydrous basis.
cent w/v solution of tetrabutylammonium hydroxide,
Category. Penicillin antibacterial. adjusted to pH 5.5 with dilute phosphoric acici or dilute
Description. A white oralmost white powder. sodium hydroxide solution; add 650 volumes of
acetonitrile, .
a linear gradient programme using the conditions given
Identification
below,
Determine by infrared absorption spectrophotometry (2.4.6). flow rate. 1 ml per minute,
Compare the spectrum with that obtained with piperacillin spectrophotometer set at 220 nm,
RS or with the reference spectrum ofpiperacillin. injection volume. 20 gl.
Time Mobile phase A Mobile phaseB
Tests (in min.) (per cent v/v) (per cent v/v)
Appearance of solution. A 10 per cent w/v solution in carbon O-tg 88 12
dioxide-free water is clear (2.4.1) and not more intensely tg -(tg +30) 88~0 12~100
coloured than the reference solution BS8 (2.4,1). (tg+30 )-( tg-45) 0~88 100~12
Specific optical rotation (2.4.22). + 165 to + 175 , determined
0 0
where, tg = retention time ofpiperacillin determined with
in a 1.0 per centw/v solution in methanol. reference solution (b)
Related substances. Determine by liquid chromatography
Inject reference solution (b), (c) and (d). The test is not valid
(2.4.14).
unless the. resolution between the peaks due to piperacillin
NOTE-Use freshly prepared solutions. impurity A and piperacillin in the chromatogram obtained with

1918

www.webofpharma.com
IP 2010 PIPERACILLIN INTRAVENOUS INFUSION

reference solution (c) is not less than 10; signal-to-noise ratio Usual strengths. 1 g per vial; 2 g per vial; 4 g per vial.
is not less than 3 for the principal peak in the chromatogram
The contents of the sealed container comply with the
obtained with reference solution (d); mass distribution ratio
requirements stated under Parental Preparations (Powder
is (2:3) for the peak due to piperacillin in the chromatogram
for Injections) and with the following requirements.
obtained with reference solution (c). The area ofany secondary
peak is not more than twice the area of the principal peak in
Identification
the chromatogram obtained with reference solution (b) (2.0
per cent). A. Dissolve a quantity ofthe powder containing about 0.25 g
ofanhydrous piperacillin in 20 ml of water, add 0.5 ml of dilute
N,N-Dimethylaniline (2.3 .21). Not more than 20 ppm.
hydrochloric acid, mix well, allow to stand until a precipitate
Heavy metals (2.3.13). 1.0 g complies with the limit test for appears and filter. Determine by infrared absorption
heavy metals, Method B (20 ppm). spectrophotometry (2.4.6). Compare the spectrum with that
Water. (2.3.43). 2.0 per cent to 4.0 per cent, determined on obtained with piperacillin RS or with the reference spectrum
0.5g. ofpiperacillin.

Assay. Determine by liquid chromatography (2.4.14), as B. In the Assay, the principal peak in the chromatogram
described in the test for Related substances. obtained with the test solution corresponds to the peak in the
chromatogram obtained with reference solution (a).
Inject reference solution (a). The test is not valid unless the
relative standard deviation for replicate injections is not more C. Gives the reaction ofsodium (2.3.1).
than 1.0 per cent.
Tests
Inject test solution (a) and reference solution (a).
pH (2.4.24).4.8 to 6.8, detennined in a 10.0 percent w/v solution.
Calculate the content of C23H27N507S,
Related substances. Determine by liquid chromatography
Storage. Store protected from light and moisture.
(2.4.14).
Test solution. Disperse a quantity ofpowder containing about
39 mg ofanhydrous piperacillin in 100 ml ofthe mobile phase.
Piperacillin Intravenous Infusion
Reference solution (a). A 0.0008 per cent w/v solution of
Piperacillin Intravenous Infusion is a sterile solution of piperacillin RS in the mobile phase.
Piperacillin Sodium in Water for Injections. It is prepared by
Reference solution (b). A 0.0004 per cent w/v solution of
dissolving Piperacillin for Intravenous Infusion in the requisite
piperacillin RS in the mobile phase.
amount of Water for Injections before use.
Chromatographic system
The intravenous infils ion complies with the requirements
a stainless steel column 25 cm x 4.6 mm, packed with
stated under Parenteral Preparations (Injections).
octadecylsilane bonded to porous silica (5 to 10 Ilm),
(Such as Beckman Ultrasphere ODS),
Piperacillin for Intravenous Infusion
mobile phase: a mixture of 0.3 volume of 0.4 M
Piperacillin for Intravenous Infusion is a sterile material tetrabutylammonium hydroxide" 10 volumes of 0.2 M
consisting of Piperacillin Sodium or Piperacillin Sodium sodium dihydrogen orthophosphate monohydrate, 44.7
prepared by the interaction of piperacillin with sodium volumes of water and 45 volumes of methanol, cool
bicarbonate, with or without excipients. It is filled in a sealed and adjust to pH 5.5 with orthophosphoric acid,
container. . flow rate. 1 ml per minute,
spectrophotometer set at 220 nm,
The constituted solution complies with the requirements for
injection volume. 10 Ill.
Clarity of solution and Particulate matter stated under
Parental Preparations. (Injections). Inject the test solution, reference solution (a) and (b). In the
chromatogram obtained with the test solution the area of the
Storage. The constituted solution should be used immediately
peak with a relative retention time of about 0.6 is not more
after preparation but, in any case, within the period
than 3.5 times the area ofthe principal peak in the chromatogram
recommended by the manufacturer.
obtained with reference solution (b) (3.5 per cent). The area of
Piperacillin for Intravenous Infusion contains not less than any other secondary peak is not more than the area of the
94.0 per cent and not more than 115.0 per cent of the stated principal peak in the chromatogram obtained with reference
amount ofpiperacillin, C23 H 27N507S. solution (a) (2 per cent).

1919

www.webofpharma.com
PIPERACILLIN INTRAVENOUS INFUSION IP 2010

Water (2.3.43). Not more than 2.0 percent, determined on 0.5 g. Piperazine Adipate contains not less than 98.0 per cent and
not more than 101.0 per cent ofC4H ION 2, C 6H IO0 4, calculated
Bacterial endotoxins (2.2.3). Not more than 2.5 Endotoxin Units
on the anhydrous basis.
per ml ofa solution prepared in the following manner. Dissolve
a quantity in water BETto obtain a solution containing 50 mg Category. Anthelmintic.
ofpiperacillin per ml. Carry out the test using Maximum Valid Description. A white, crystalline powder.
dilution ofthis solution calculated from the declared sensitivity
of the lysate used in the test. Identification
Assay. Determine by liquid chromatography (2.4.14). Test A may be omitted iftests B, C andD are carried out. Tests .
Band C may be omitted if tests A and D are carried out.
Test solution. Disperse a quantity ofpowder containing about
39 mg ofanhydrous piperacillin in 100 ml ofthe mobile phase. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with piperazine
Reference solution (a). Dissolve 40 mg ofpiperacillin RS in
adipate RS or with the reference spectrum of piperazine
methanol and dilute to 100 ml with the mobile phase.
adipate.
Reference solution (b). A solution containing 0.01 per cent
B. In the test for Related substances, examine the plate after
w/vof anhydrous ampicillin RS and 0.02 per cent w/v of
spraying with both the ninhydrin and iodine solutions. The
piperacillin RS in the mobile phase.
principal spot in the chromatogram obtained with test solution
Chromatographic system (b) corresponds to that in the chromatogram obtained with
- a stainless steel column 25 cm x 4.6 mm, packed with reference solution (a).
octadecylsilane bonded to porous silica (5 f..l.m), (Such C. Dissolve 0.1 g in 5 ml of water, add 0.5 g of sodium
as Beckman Ultrasphere ODS), bicarbonate, 0.5 ml of a freshly prepared 5.0 per cent w/v
mobile phase: a mixture of 0.3 volume of 0.4 M solution of potassium ferricyanide and 0.1 ml of mercury,
tetrabutylammonium hydroxide, 10 volumes of 0.2 M shake vigorously for 1 minute and allow to stand for
sodium dihydrogen orthophosphate monohydrate, 44.7 20 minutes; a reddish colour slowly develops.
volumes of water and 45 volumes of methanol, cool
and adjust to pH 5.5 with orthophosphoric acid, Tests
flow rate. I ml per minute,
Appearance of solution. A 5.0 per cent w/v solution is clear
spectrophotometer set at 220 nm,
(2.4.1), and not more intensely coloured than reference solutio!1
- injection volume. 10 f..l.l.
BS8 (2.4.1).
Inject reference solution (b). The resolution between the peaks pH (2.4.24). 5.0 to 6.0, determined in a 5.0 per cent w/v solution.
due to anhydrous ampicillin and piperacillin is not less than
Related substances. Determine by thin-layer chromatography
16.
(2.4.17), coating the plate with silica gel G..
Inject the test solution and the reference solution (a).
Solvent mixture. 60 volumes of strong ammonia solution and
Calculate the content ofC23H27Ns07S, 40 volumes of ethanol.
Storage. Store protected from moisture. Mobile phase. A freshly prepared mixture of 80 volumes of
acetone and 20 volumes of strong ammonia solution.
Labelling. The label ofthe sealed container states the quantity
of active ingredient contained in it in terms ofthe equivalent Test solution (a). Dissolve I g of the substance under
amount of anhydrous piperacillin. examination in 10 ml ofthe solvent mixture.
Test solution (b). Dissolve 1 g of the substance under
examination in 100 ml ofthe solvent mixture.
Reference solution (a). A I per cent w/v solution ofpiperazine
Piperazine Adipate adipate RS in the solvent mixture.
Reference solution (b). A 0.025 per cent w/v solution of
ethylenediamine in the solvent mixture.
Reference solution (c). A 0.025 per cent w/v solution of
triethylenediamine in the solvent mixture.
Reference solution (d). A solution containing 0.025 pei cent
w/v of triethylenediamine and 1 per cent w/v ofthe substance
under examination in the solvent mixture.

1920

www.webofpharma.com
IP 2010 PIPERAZINE CITRATE

Apply to the plate 5 III of each solution. After development, vigorously for 1 minute and allow to stand for 20 minutes; a
dry the plate at 105°, spray with a 0.3 per cent w/v solution of reddish colour slowly develops.
ninhydrin in a mixture on volumes of anhydrous acetic acid
B. To 10 ml of the filtrate obtained in test A add 5 ml of
and 100 volumes of 1-butanol and then with a 0.15 per cent
hydrochloric acid and extract with three quantities, each of
w/v solution of ninhydrin in ethanol and dry at 105° for 10
10 ml, of ether, evaporate the combined ether extracts to
minutes. Any secondary spot in the chromatogram obtained
dryness; the residue, after washing with a small volume of
with test solution (a) is not more intense than the spot in the
water and drying at 105°, melts at about 152° (2.4.21).
chromatogram obtained with reference solution (b). Spray the
plate with 0.05 M iodine and allow to stand for about 10 Tests
minutes. Any spot corresponding to triethylenediamine in
the chromatogram obtained with test solution (a) is not more Other tests. Comply with the tests stated under Tablets.
intense than the spot in the chromatogram obtained with Assay. Weigh and powder 20 tablets. Weigh accurately a
reference solution (c). The test is not valid unless the quantity of the powder containing about 0.2 g of Piperazine
chromatogram obtained with reference solution (d) shows two Hydrate, shake with 10 ml of water for 1 hour, filter and wash
separated spots. Ignore any spot remaining on the line of the residue with two quantities, each of 10 ml, of water. To the
application. combined extract and washings add 5 ml of 1 M sulphuric
Heavy metals (2.3.13). Dissolve l.0 g in 20 ml ofwater, 0.5 ml of acid and 50 ml of picric acid solution, bring to boil, allow to
0.1 M hydrochloric acid and add sufficient water to produce stand for 1 hour and filter through a sintered-glass crucible
25 ml. The resulting solution complies with the limit test for (porosity No.4) and wash the residue with successive
heavy metals, Method A (20 ppm). quantities, each of 10 ml, of a mixture of equal volumes of a
saturated solution ofpicric acid and water until the washings
Sulphated ash(2.3.18). Not more than 0.1 per cent.
are free from sulphate. Wash with five quantities, each of
Water (2.3.43). Not more than 0.5 per cent, determined on 10 ml, of ethanol and dry to constant weight at 105°.
l.Og. 1 g ofthe residue is equivalent to 0.3567 g ofC4HIQN2 , 6H20.
Assay. Weigh accurately about 0.2 g, dissolve in 10 ml of Storage. Store protected from moisture.
anhydrous glacial acetic acid with gentle heating and dilute
to 70 ml with the same solvent. Titrate with 0.1 Mperchloric Labelling. The label states the strength in terms of the
acid, using 0.25 ml of 1-naphtholbenzein solution as indicator equivalent amount of piperazine hydrate.
and titrating until the colour of the solution changes from
brownish yellow to green. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.01161 g of Piperazine Citrate
C4HIQN2, C6HIQ 0 4.
Storage. Store protected from moisture.
HO GOOH
, HOOG~GOOH
Piperazine Adipate Tablets 3 2
Piperazine Adipate Tablets contain not less than 92.5 per cent
Mol. Wt. 642.7 (anhydrous)
and not more than 107.5 per cent of the stated amount of
piperazine hydrate, C4HIQNz, 6H20. Piperazine Citrate is a salt of piperazine with citric acid
containing a variable amount ofwater of crystallisation.
Usual strength. The equivalent of 300 mg of piperazine
hydrate. (144 mg of Piperazine Adipate is approximately Piperazine Citrate contains not less than 98.0 per cent and not
equivalent to 120 mg ofPiperazine Hydrate). more than 101.0 per cent of(C4HIQN2)3, 2C6Hg0 7, calculated on
the anhydrous basis.
Identification Category. Anthelmintic.
A. Extract a quantity ofthe powdered tablets containing 1 g of Dose. For an adult, in the treatment ofthreadworm infestation,
Piperazine Hydrate with 20 ml of water and filter. Dilute 1mlof equivalent of 1 to 2 g of Piperazine Hydrate daily, in divided
the filtrate to 5 ml with water, add 0.5 g ofsodium bicarbonate, doses for 7 days; in the treatment of roundworm infestation,
0.5 ml of a freshly prepared 5.0 per cent w/v solution of equivalent of4.5 g ofPiperazine Hydrate as a single dose. For
potassium ferricyanide and 0.1 ml of mercury, shake a child, in the treatment of threadworm infestation, the

1921

www.webofpharma.com
PIPERAZINE CITRATE IP 2010

equivalent of 40 mg of Piperazine Hydrate per kg of body Apply to the plate 5 Jll of each solution. After development,
weight daily, in divided doses for 7 days; inthe treatment of dry the plate at 105°, spray with a 0.3 per cent w/v solution of
roundworm infestation, as a sillgle dose, the equivalent of 120 ninhydrin in a mixture on volumes of anhydrous acetic acid
mg of Piperazine Hydrate per kg of body weight, up to a and 100 volumes of 1-butanol and then with a 0.15 per cent
maximum dose of4 g. w/v solution of ninhydrin in ethanol and dry at 105° for
(150 mg ofPiperazine Citrate or 132 mg ofanhydrous piperazine 10 minutes. Any secondary spot in the chromatogram obtained
citrate is approximately equivalent to 120 mg of Piperazine with test solution (a) is not more intense than the spot in the
Hydrate). chromatogram obtained with reference solution (b). Spray the
plate with 0.05 M iodine and allow to stand for about
Description. A white, granular powder; almost odourless. 10 minutes. Any spot corresponding to triethylenediamine in
Identification the chromatogram obtained with test solution (a) is not more
intense than the spot in the chromatogram obtained with
Test A may be omitted if tests Band C are carried out. Test B reference solution (c). The test is not valid unless the
may be omitted if tests A and C are carried out. chromatogram obtained with reference solution (d) shows two
A. Determine by infrared absorption spectrophotometry (2.4.6). separated spots. Ignore any spot remaining on the line of
Compare the spectrum with that obtained with piperazine application.
citrate RS or with the reference spectrum ofpiperazine citrate. Heavy metals (2.3.13). 1.0 g complies with the limit test for
B. In the test for Related substances, examine the plate after heavy metals, Method A (20 ppm).
spraying with both the ninhydrin solutions. The principal Sulphated ash (2.3.18). Not more than 0.1 per cent.
spot in the chromatogram obtained with test solution (b)
Water (2.4.19). 10.0 to 14.0 per cent, determined on 0.3 g.
corresponds to that in the chromatogram obtained with
reference solution (a). Assay. Weigh accurately about 0.2 g, dissolve in 10 ml of
anhydrous glacial acetic acid with gentle heating and dilute
C. A 10 per cent w/v solution gives the reactions of citrates
to 70 ml with the same solvent. Titrate with 0.1 Mperchloric
(2.3.1).
acid, using 0.25 ml of 1-naphtholbenzein solution as indicator
Tests and titrating until the colour of the solution changes from
brownish yellow to green. Carry out a blank titration.
Appearance of solution. A 5.0 per cent w/v solution is clear
(2.4.1), and not more intensely coloured than reference solution 1 ml of 0.1 M perchloric acid is equivalent to 0.01071 g of
BS8 (2.4.1). (C4HIONz)3,2C6H807'
pH (2.4.24).5.0 to 6.0, determined in a 5.0 per cent w/v solution. Storage. Store protected from light and moisture.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
Solvent mixture. 60 volumes of strong ammonia solution and Piperazine Citrate Syrup
40 volumes of ethanol. Piperazine Citrate Oral Solution; Piperazine Citrate Elixir
Mobile phase. A freshly prepared mixture of 80 volumes of Piperazine Citrate Syrup is a solution ofPiperazine Citrate in a
acetone and 20 volumes of strong ammonia solution. suitable flavoured Vehicle.
Test solution (a). Dissolve 1 g of the substance under
Piperazine Citrate Syrup contains the equivalent of not less
examination in 10 ml ofthe solvent mixture.
than 92.5 per cent and not more than 107.5 per cent of the
Test solution (b). Dissolve 1 g of the substance under stated amount ofpiperazine hydrate, C4H ION z, 6H zO.
examination in 100 ml ofthe solvent mixture.
Usual strength. The equivale.nt of 750 rug of piperazine
Reference solution (a). A 1.0 per cent w/v solution of hydrate in 5 ml.
piperazine citrate RS in the solvent mixture.
(125 mg ofPiperazine Citrate or 110 mg ofanhydrous piperazine
Reference solution (b). A 0.025 per cent w/v solution of citrate is approximately equivalent to 100 mg of Piperazine
ethylenediamine in the solvent mixture. Hydrate).
Reference solution (c). A 0.025 per cent w/v solution of
triethylenediamine in the solvent mixture. Identification
Reference solution (d). A solution containing 0.025 per cent A. To 1 ml add 5 ml of2 M hydrochloric acid and, with stirring,
w/v of triethylenediamine and 1 per cent w/v ofthe substance 1 ml of a freshly prepared 50 per cent w/v solution of sodium
under examination in the solvent mixture. nitrite, cool in ice for 15 minutes, induce crystallisation, wash

1922

www.webofpharma.com
IP 2010 PIPERAZINE HYDRATE

the crystalline precipitate with water and dry at 105°. The treatment of threadworm infestation, 40 mg per kg of body
crystals melt at about 159° (2.4.21). weight daily, in divided doses for 7 days. In the treatment of
roundworm infestation, as a single dose, 120 mg per kg of
B. Warm 10 ml with activated charcoal and filter. Boil a portion
body weight, up to a maximum dose of4 g.
of the filtrate with an excess of mercuric sulphate solution,
filter, boil the filtrate and add 0.25 ml of dilute potassium Description. Colourless, glassy deliquescent crystals.
permanganate solution; the permanganate solution is
decolorised and a white precipitate is produced. Identification
C. AcidifY a portion of the filtrate obtained in test B with 1 M Test A may be omitted iftests Band C are carried out. Tests B
sulphuric acid, add 0.25 ml of dilute potassium permanganate and C may be omitted if test A is carried out.
solution, warm until the colour is discharged and add an excess A. Detennine by infrared absorption spectrophotome1:Iy (2.4.6).
of bromine water; a white precipitate is produced either Compare the spectrum with that obtained with piperazine
immediately or on cooling. hydrate RS.

Tests B. In the test for Related substances, examine the plate after
spraying with both the ninhydrin solutions. The principal spot
Other tests. Complies with the tests stated under Oral Liquids. in the chromatogram obtained with test solution (b)
corresponds to that in the chromatogram obtained with
Assay. Weigh accurately a quantity containing about 0.2 g of
reference solution (a).
Piperazine Hydrate, add 3.5 ml of 0.5 M sulphuric acid and
10 ml of water, add 100 ml ofpicric acid solution, heat on a C. Dissolve 0.2 g in 5 ml of dilute hydrochloric acid, add 0.5 g
water-bath for 15 minutes, allow to stand for I hour and filter of sodium nitrite and heat to boiling. Cool in ice for 15 minutes,
through a sintered-glass crucible (porosity No.4). Wash the scratching the walls of the container with a glass rod and
residue with successive quantities, each of I 0 ml, ofa mixture filter. The crystals, after washing with 10 ml ofice-cold water
of equal volumes of a saturated solution of picric acid and and drying at 105°, melt at about 159° (2.4.21).
water until the washings are free from sulphate. Wash the
residue with five quantities, each of 10 ml, of ethanol and dry
Tests
to constant weight at 105°. Appearance ofsolution. A 5.0 per cent w/v solution in carbon
1 g ofthe residue is equivalent to 0.3567 g ofC4HIQNz, 6H zO; dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution BS8 (2.4.1).
Determine the weight per ml ofthe syrup (2.4.29), and calculate
pH (2.4.24). 10.5 to 12.0, determined in a 5.0 per cent w/v
the content ofC4HIQNz, 6H zO, weight in volume.
solution in carbon dioxide:free water.
Storage. Store protected from light.
Related substances. Determine by thin-layer chromatography
Labelling. The label states the strength in terms of the (2.4.17), coating the plate with silica gel G.
equivalent amount of piperazine hydrate.
Solvent mixture. 60 volumes of strong ammonia solution and
40 volumes of ethanol.
Mobile phase. A freshly prepared mixture of 80 volumes of
Piperazine Hydrate acetone and 20 volumes of strong ammonia solution.
Test solution (a). Dissolve 1 g of the substance under
H examination in 10 ml ofthe solvent mixture.

eN)N
Test solution (b). Dissolve 1 g of the substance under
examination in 100 ml ofthe solvent mixture.
H Reference solution (a). A 1 per cent w/v solution ofpiperazine
hydrate RS in the solvent mixture.
C4HIQNz,6HzO Mol. Wt.194.2
Reference solution (b). A 0.025 per cent w/v solution of
Piperazine Hydrate contains not less than 98.0 per cent and ethylenediamine in the solvent mixture.
not more than 10 I. 0 per cent ofC4H IQNz, 6H zO.
Reference solution (c). A 0.025 per cent w/v solution of
Category. Anthelmintic. triethylenediamine in the solvent mixture.
Dose. For an adult, in the treatment ofthreadworm infestation, Reference solution (d). A solution containing 0.025 per cent
1 to 2 g daily, in divided doses for 7 days; in the treatment of w/v of triethylenediamine and 1 per cent w/v of the substance
roundworm infestation, 4 g as a single dose. For a child, in the under examination in the solvent mixture.

1923

www.webofpharma.com
PIPERAZINE HYDRATE IF 2010

Apply to the plate 5 III of each solution. After development, Description. A white, crystalline powder; odourless or almost
dry the plate at 105°, spray with a 0.3 per cent w/v solution of odourless.
ninhydrin in a rriixture oD volumes ofanhydrous acetic acid
and 100 volumes off-butanol and then with a 0.15 per cent Identification
w/v solution of ninhydrin in ethanol and dry at 105° for
10 minutes. Any secondary spot in the chromatogram obtained A. Dissolve 0.2 g in 5 ml of dilute hydrochloric acid, add
with test solution (a) is not more intense than the spot in the 0.5 g of sodium nitrite and heat to boiling. Cool in ice for
chromatogram obtained with reference solution (b). Spray the 15 minutes, scratching the walls ofthe container with a glass
plate with 0.05 M iodine and allow to stand for about rod and filter. The crystals, after washing with 10 ml of ice-
10 minutes. Any spot corresponding to triethylenediamine in cold water and drying at 105°, melt at about 159° (2.4.21).
the chromatogram obtained with test solution (a) is not more B. Dissolve 0.1 g in 5 ml of water, add 0.5 g of sodium
intense than the spot in the chromatogram obtained with bicarbonate, 0.5 ml of a freshly prepared 5.0 per cent w/v
reference solution (c). The test is not valid unless the solution of potassium ferricyanide and 0.1 ml of mercury,
chromatogram obtained with reference solution (d) shows two shake vigorously for 1 minute and allow to stand for
separated spots. Ignore any spot remaining on the line of 20 minutes; a reddish colour slowly develops.
application.
C. Gives the reactions of phosphates (2.3.1).
Heavy metals (2.3.13).1.0 g complies with the limit test for
heavy metals, Method A (20 ppm). Tests
Sulphated ash (2.3.18). Not more than 0.1 percent. pH (2.4.24). 6.0 to 6.5, determined in a 1.0 per cent w/v solution.
Assay. Weigh accurately about 0.2 g, dissolve in 10 ml of Heavy metals (2.3.13). Dissolve 1.0 g in 20 ml of2 M acetic
anhydrous glacial acetic acid with gentle heating and dilute acid and add sufficient water to produce 25 ml. The solution
to 70 ml with the same solvent. Titrate with 0.1 M perchloric complies with the limit test for heavy metals, Method A
acid, using 0.25 ml of 1-naphtholbenzein solution as indicator (20 ppm).
and titrating until the colour of the solution changes from
brownish yellow to green. Canoy out a blank titration. Water (2.3.43). 8.0 to 9.5 percent, determined on 0.25 g.

1 ml of 0.1 M perchloric acid is equivalent to 0.009705 g of Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
C4H ION2,6H20. 3.5 ml of0.5 M sulphuric acid and 10 ml ofwater. Add 100 ml
of picric acid solution, heat on a water-bath for 15 minutes
Storage. Store protected from light and moisture. and allow to stand for 1 hour. Filter through a sintered-glass
crucible (porosity No.4) and wash the residue with successive
quantities, each of 10 ml, of a mixture of equal volumes of a
Piperazine Phosphate saturated solution ofpicric acid and water until the washings
are free from sulphate. Wash the residue with five quantities,
C4H ION2, H3P04 , H20 Mol. Wt. 202.2 each of 10 ml, of ethanol and dry to constant weight at 105°.
Piperazine Phosphate contains not less than 98.5 per cent 1 g ofthe residue is equivalent to 0.3382 g ofC 4H ION 2 , H3P04 •
and not more than 100.5 per cent ofC 4H ION 2 , H3P0 4 ,
Storage. Store protected from moisture.
calculated on the anhydrous basis.
Category. Anthelmintic.
Dose. For an adult, in the treatment ofthreadworm infestation,
the equivalent of 1 to 2 g ofPiperazine Hydrate daily, in divided
doses for 7 days; in the treatment of roundworm infestation, Piperazine Phosphate Tablets
the equivalent of4.5 g ofPiperazine Hydrate as a single dose.
If the tablets are intended to be chewed before swallowing
For a child, in the treatment of threadworm infestation, the
they may contain suitable flavouring agents.
equivalent of 40 mg of Piperazine Hydrate per kg of body
weight daily, in divided doses for 7 days; in the treatment of Piperazine Phosphate Tablets contain not less than 92.5 per
roundworm infestation, as a single dose, the equivalent of 120 cent and not more than 107.5 per cent of the stated amount of
mg of Piperazine Hydrate per kg of body weight, up to a piperazine phosphate, C4H ION 2, H3P04, H 20.
maximum dose of4 g.
Usual strengths. The equivalent of260 mg; 520 mg Piperazine
(125 mg ofpiperazine phosphate is approximately equivalent Hydrate (125 mg of piperazine phosphate is approximately
to 120 mg ofPiperazine Hydrate). equivalent to 120 mg ofPiperazine Hydrate).

1924

www.webofpharma.com
IP 2010 PlRACETAM

Identification Piracetam contains not less than 98.0 per cent and not more
than 102.0 per cent ofC 6HIQN20 2, calculated on the dried basis.
Extract a quantity of the powdered tablets containing 1 g of
Piperazine Phosphate with 20 rnl ofwater and filter. The filtrate Description. A white or almost white powder.
complies with the following tests.
Identification
A. To 4 ml of the filtrate, add 1 ml of hydrochloric acid, 0.5 g
Determine by infrared absorption spectrophotometry (2.4.6).
ofsodium nitrite and heat to boiling. Cool in ice for 15 minutes,
Compare the spectrum with that obtained with piracetam RS
scratching the walls of the container with a glass rod and
or with the reference spectrum ofpiracetam.
filter. The crystals, after washing with 10 ml ofice-cold water
and drying at 105°, melt at about 159° (2.4.21). Tests
B. Dilute 1 ml of the filtrate to 5 ml with water, add 0.5 g of Appearance of solution. A20.0 per cent w/v solution in water
sodium bicarbonate, 0.5 ml of a freshly prepared 5.0 per cent is clear (2.4.1) and colourless (2.4.1).
w/v solution ofpotassiumferricyanide and 0.1 ml ofmercwy,
Related substances. Determine by liquid chromatography
shake vigorously for 1 minute and allow to stand for
(2.4.14).
20 minutes; a reddish colour slowly develops.
Solvent mixture. A mixture of 10 volumes of acetonitrile and
C. Gives the reactions of phosphates (2.3.1). 90 volumes ofwater.
Tests Test solution (a). Dissolve 50.0 mg of the substance under
examination in 100.0 ml ofthe solvent mixture.
Disintegration. The test does not apply to Piperazine
Phosphate Tablets intended to be chewed before swallowing. Test solution (b). Dilute 10.0 ml oftest solution (a) to 50.0 ml
with the solvent mixture.
Other tests. Comply with the tests stated under Tablets.
Reference solution (a). Dissolve 5 mg ofthe substance under
Assay. Weigh and powder 20 tablets. Weigh accurately a examination and 10 III of 2-pyrrolidone in sufficient solvent
quantity of the powder containing about 0.15 g of Piperazine mixture to produce 100.0 ml.
Phosphate, shake with 10 ml of water for 1 hour, filter and
Reference solution (b). Dilute 1.0 ml of test solution (a) to
wash the residue with two quantities, each of 10 ml, ofwater.
100.0 ml with the solvent mixture. Dilute 5.0 ml ofthis solution
To the combined extract and washings add 5 ml of 1 M
to 50.0 ml with the solvent mixture.
sulphuric acid and 50 ml ofpicric acid solution, boil, allow
the mixture to stand for several hours and filter through a Reference solution (c). Dissolve 50.0 mg ofpiracetam RS in
sintered glass crucible (porosity No.4). Wash the residue 100.0 ml ofthe solvent mixture. Dilute 10.0 ml ofthis solution
with successive quantities, each of 10 ml, ofa mixture ofequal to 50.0 ml with the solvent mixture.
volumes of a saturated solution ofpicric acid and water until Chromatographic system
the washings are free from sulphate. Wash the residue with - a stainless steel column 25 cm x 4.6 mm, packed with end
five quantities, each of 10 ml,. ofethanol (95 per cent) and dry capped octadecylsilane bonded to silica (5 Ilm),
to constant weight at 105°. - mobile phase: a mixture of 10 volumes of acetonitrile
1 g ofthe residue is equivalent to 0.3714 g ofC 4HIQN2 , H 3P0 4, and 90 volumes of 0.1 per cent w/v solution of
H20. dipotassium hydrogen phosphate, adjusted to pH 6.0
Storage. Store protected from moisture. with orthophosphoric acid,
- flow rate. 1 ml per minute,
Labelling. The label states, where applicable, that the tablets - spectrophotometer set at 205 nm,
are to be chewed before swallowing. - injection volume. 20 Ill.
Inject reference solution (a). The test is not valid unless the
resolution between the peaks due to 2-pyrrolidone and
Piracetam piracetam is not less than 3.0 and the symmetry factor is not
more than 2.0 for the peak due to piracetam.
Inject test solution (a) and reference solution (b). Run the
chromatogram for 8 times the retention time ofpiracetam. In
the chromatogram obtained with the test solution (a), the area
ofany secondary peak is not more than the area ofthe principal
C6HIQN20 2 Mol.Wt.142.2 peak in the chromatogram obtained with the reference solution
Piracetam is 2-(2-oxopyrrolidin-l-yl)acetarnide . (b) (0.1 per cent) and the sum ofareas of all the secondary

1925

www.webofpharma.com
PIROXICAM IP 2010

peaks is not more than 3 times the area ofthe principal peak in acid shows absorption maxima at about 242 urn and 334 urn
the chromatogram obtained with reference solution (b) (0.3 and a minimum at about 270 urn; absorbance at about 334 urn,
per cent). Ignore any peak with an area less than 05 times the aboutO.8T
area of the principal peak in the chromatogram obtained with
C. Determine by thin-layer chromatography (2.4.17), coating
reference solution (b) (0.05 per cent).
the plate with silica gel GF254.
Heavy Metals (2.3.13). 2.0 g complies with limit test for heavy
Mobile phase. A mixtureof·95 volumes of toluene and
metals, Method B (10 ppm).
5 volumes of acetic acid.
Sulphated ash (2.3.1 8). Not more than 0.1 per cent.
Test solution. Dissolve 0.1 g of the substance under
Loss on drying (2.4.19). Not more than 1.0 per cent, determined examination in 100 ml of a mixture of equal volumes of
on 1.0 g by drying in an oven at 105°. chloroform and methanol.
Assay. Determine by liquid chromatography (2.4.14) as Reference solution. A 0.1 per cent w/v solution of piroxicam
described in the test for Related substances. RS in a mixture ofequal volumes of chloroform and methanol.
Inject reference solution (c). The test is not valid unless the Apply to the plate 20 III of each solution. After development,
relative standard deviation for replicate injections is not more dry the plate in air and examine in ultraviolet light at 254 urn.
than 2.0 per cent. The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Inject test solution (b) and reference solution (c).
with the reference solution.
Calculate the content ofC 6H ION 20 2 •
Storage. Store protected from light and moisture. Tests
Related substances. Determine by liquid chromatography
(2.4.14).
Piroxicam Test solution. Dissolve about 75 mg of the substance under
examination in 50.0 ml of acetonitrile.
Reference solution. Dilute 1.0 ml ofthe test solution to 10.0 ml
with acetonitrile. Dilute 1.0 ml ofthis solution to 50.0 ml with
acetonitrile.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm packed with
endcapped octadecylsilane bonded to porous silica (5
Mol. Wt. 331.4 /lill),
column temperature. 40°,
Piroxicam is 4-hydroxy-2-methyl-N-(2-pyridyl)-2H-l,2-
- mobile phase: a mixture of 40 volumes of acetonitrile
benzothiazine-3-carboxamide-l, I-dioxide.
and 60 volumes of a 0.7 per cent w/v solution of
Piroxicam contains not less than 97.0 per cent and not more potassium dihydrogen phosphate, adjusted to pH 3.0
than 103.0 per cent of ClsH13N304S, calculated on the with orthophosphoric acid,
anhydrous basis. flow rate. 1 ml per minute,
Category. Analgesic; antiinflammatory; antipyretic. - spectrophotometer set at 230 nm,
injection volume. 20 Ill.
Dose. 10 to 20 mg daily.
Inject the reference solution. The test is not valid unless the
Description. An off-white to light tan or light yellow powder; symmetry factor ofthe peak due to piroxicam impurity B is not
odourless. more than 5.0. The relative retention time with reference to
piroxicam for piroxicam impurity B is about 0.85.
Identification
Inject the test solution and the reference solution. Run the
A. Detennine by infrared absorption spectrophotometry (2.4.6). chromatogram 5 times the retention time ofthe principal peale.
Compare the spectrum with that obtained with piroxicam RS In the chromatogram obtained with the test solution, the area
or with the reference spectrum ofpiroxicam. ofany secondary peak is not more than the area ofthe principal
B. When examined in the range 230 nm to 360 nm (2.4.7), a peak in the chromatogram obtained with reference solution
0.001 per cent w/v solution in 0.01 M methanolic hydrochloric (b) (0.2 per cent), the sum of all the secondary peaks is not

1926

www.webofpharma.com
IP 2010 PIROXICAM CAPSULES

more than twice the area of the principal peak in the Test solution. Dissolve a portion of the contents of the
chromatogram obtained with reference solution (b) (0.4 per capsules in sufficient of a mixture of equal volumes of
cent). Ignore any peak with an area less than 0.1 times the area chloroform and methanol to obtain a solution containing
of the principal peak in the chromatogram obtained with about 0.1 per cent w/v ofPiroxicam. Shake for 10 minutes, filter
reference solution (b) (0.02 per cent). and use the filtrate.
Heavy metals (2.3.13). 0.4 g complies with the limit test for Reference solution. A 0.1 per cent w/v solution of piroxicam
heavy metals, Method B (50 ppm). RS in a mixture ofequal volumes of chloroform and methanol.
Sulphated ash (2.3.18). Not more than 0.3 percent. Apply to the plate 20 III of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Water (2.3.43). Not more than 0.5 per cent, determined on
The principal spot in the chromatogram obtained with the test
2.0g.
solution corresponds to that in the chromatogram obtained
Assay. Determine by liquid chromatography (2.4.14). with the reference solution.
Test solution. A 0.005 per cent w/v solution of the substance Tests
under examination in 0.1 M methanolic hydrochloric acid.
Dissolution (2.5.2).
Reference solution. A 0.005 per cent w/v solution ofpiroxicam Apparatus No.2,
RS in 0.1 M methanolic hydrochloric acid.
Medium. 900 ml of 0.1 M hydrochloric acid,
Chromatographic system Speed and time. 100 rpm and 45 minutes.
- a stainless steel column 25 cm x 4.6 mm, packed with
Withdraw 10 ml of the medium and filter. Measure the
octadecylsilane bonded to porous silica (5 Ilm),
absorbance ofthe filtrate (2.4.7), suitably diluted ifnecessary,
mobile phase: a mixture of45 volumes of methanol and
at the maximum at about 242 nm. Calculate the content of
55 volumes of a buffer solution prepared by diluting a
ClsH13N304S, in the medium taking 352 as the specific
mixture of 7.72 g of anhydrous citric acid in 400 ml of
absorbance at 242 run.
water and 5.35 g ofsodiumphosphate in 100 ml of water
to 1000 ml with water; D. Not less than 75 per cent of the stated amount of
flow rate. 1.2 ml per minute, ClsH13N304S.
spectrophotometer set at 254 run, Uniformity of content. (For capsules containing 10 mg or
injection volume. 20 Ill. lessJ- Comply with the test stated under Capsules.
Inject the reference solution. The test is not valid unless the Test solution. Dissolve the contents ofa capsule in 100.0 ml of
relative standard deviation for replicate injections is not more 0.1 M methanolic hydrochloric acid and filter. Dilute further
than2.0 per cent. if necessary.
Inject alternately the test solution and the reference solution. Determine by liquid chromatography (2.4.14) using the
Calculate the content OfC1SH13N304S. chromatographic system and the reference solution described
under Assay.
Storage. Store protected from light and moisture.
Calculate the content ofCIsHI3N304S in the capsule.
Water (2.3.43). Not more than 8.0 per cent, determined on
0.25 g.
Piroxicam Capsules Other tests. Comply with the tests stated under Capsules.
Assay. Determine by liquid chromatography (2.4.14).
Piroxicam Capsules contain not less than 92.5 per cent and·
not more than 107.5 per cent ofthe stated amount ofpiroxicam, Test solution. Dissolve an accurately weighed quantity of the
ClsH13N304S. mixed contents of the capsules containing about 50 mg of
Piroxicam in 100.0 ml of 0.1 Mmethanolic hydrochloric acid.
Usual strengths. 10 mg; 20 mg.
Further dilute 1.0 ml ofthe solution to 10.0 ml with the same
solvent.
Identification
Reference solution. A 0.005 per cent w/v solution ofpiroxicam
Determine by thin-layer chromatography (2.4.17), coating the RS in 0.1 M methanolic hydrochloric acid.
plate with silica gel GF254. Chromatographic system
Mobile phase. A mixture of 95 volumes of toluene and - a stainless steel column 25 cm x 4.6 mm, packed with
5 volumes of acetic acid. octadecylsilane bonded to porous silica (5 Ilm),

1927

www.webofpharma.com
PLASTER OF PARIS IP 2010

- mobile phase: a mixture of45 volumes of methanol and retain their sharpness of outline and do not crumble under
55 volumes of a buffer solution prepared by diluting a pressure.
mixture of7.72 g of anhydrous citric acid in 400 ml of Loss on ignition (2.4.20). 4.5 to 8.0 per cent, detennined by
water and 5.35 g ofsodium phosphate in 100 ml of water
igniting to constant weight at red heat.
to 1000 ml with water,
- flow rate. 1.2 ml per minute, Storage. Store protected from moisture.
- spectrophotometer set at 254 nm,
- injection volume. 20 Ill.
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
Poloxamers
than 2.0 per cent.
Inject altemately the test solution and the reference solution.
b is atleast 15 and (CHzCHzO)a+c is varied from 20 to 90% by weight
Calculate the content ofCIsH13N304S in the capsules.
Storage. Store protected from light and moisture. Mol. Wt. ranges from 1000 to > 16000
Poloxamers are polyethyleneprQpylpropyleneglycols.
Synthetic block copolymer of ethylene oxide and propylene
Plaster of Paris oxide represented by the following general fonnula:

Dried Calcium Sulphate; Exsiccated Calcium Sulphate Poloxamer Ethylene Propylene Content of Average
Type oxide oxide oxyethylene relative
CaS04, YzHzO Mol. Wt. 145.1 units (per molecular
units
Plaster of Paris is prepared by heating powdered gypsum, (a) (b) cent) mass
CaS04,YzHzO, at about 1500 in a controlled manner such that it
is substantially converted into the hemihydrate, CaS04,YzHzO, 124 10-15 18-23 44.8-48.6 2090-2360
with minimum production ofthe anhydrous phases ofcalcium 188 75-85 25-30 79.9-83.7 7680-9510
sulphate. It may contain suitable accelerators or decelerators.
237 60-68 35-40 70.5-74.3 6840-8830
Category. Surgical aid.
338 137-146 42-47 81.4-84.9 12700-17400
Description. A white or almost white powder; odourless or
407 95-105 54.60 71.5-74.9 9840-14600
almost odourless; hygroscopic.
A suitable antioxidant may be added.
Identification
Description. Poloxamer 124 is a colourless or ahnost colourless
Gives the reactions of calcium salts and of sulphates (2.3.1). liquid and poloxamers 188,237,338,407 are a white or ahnost
white, waxy powder, microbeads or flakes.
Tests
pH (2.4.24). 6.5 to 9.0, detennined in a20.0 percentw/v slurry Identification
in water.
Test A may be omitted if tests Band C are carried out. Tests B
Acid insoluble Dlatter: Dissolve 0.5 gin 30 ml ofa mixtureof and C may be omitted if test A is carried out.
1 volume of hydrochloric acid and 2 volumes of water and
evaporate to dryness in a dish on a water-bath. Heat for A. Detennine by infrared absorption spectrophotometry (2.4.6).
2 hours at 1200 and again add 20 ml ofthe acid mixture. Wann Compare the spectrum with that obtained with poloxamer RS
for a few minutes and filter. Wash the residue with wann water or with the reference spectrum ofpoloxamer.
until free from chlorides, dry, ignite and weigh. The residue B. Average relative molecular mass.
weighs not more than 5 mg (1.0 per cent).
C. Oxypropylene: oxyethylene ratio.
Setting properties. 20 g mixed with 10 ml of wate,; at 150 to 200
in a cylindrical mould about 2.4 cm in diameter sets in not less Tests
than 4 minutes and not more than 6 minutes. The mass thus
formed, after standing for 3 hours, possesses sufficient Melting point (2.4.21). About 500 for poloxamers 188, 237, 338
hardness to resist pressure of the fingers at the edges, which and 407.

1928

www.webofpharma.com
IP 2010 POLYETHYLENE GLYCOL 1500

Appearance ofsolution. A 10.0 per cent w/v solution in carbon The ethylene oxide is not more than 0.5 times the area of the
dioxide-ji-ee water (solution A) is not more intensely coloured corresponding peak in the chromatogram obtained with the
than reference solution BYS7 (2.4.1). reference solution (1 ppm); propylene oxide is not more than
0.5 times the area of the corresponding peak in the
pH (2.4.24).5.0 to 7.5, determined in solution A.'
chromatogram obtained with the reference solution (5 ppm)
Ethylene oxide, propylene oxide and dioxan. Determine by gas and dioxan is not more than 0.5 times the area of the
chromatography (2.4.13). corresponding peak in the chromatogram obtained with the
Ethylene oxide stock solution. Disperse about 0.5 g of reference solution (10 ppm).
ethylene oxide RS with 50.0 ml of dimethyl sulphoxide. Average relative molecular mass. Weigh 15 g ofthe substance
Ethylene oxide solution. Dilute 1.0 ml of the ethylene oxide under examination into a 250-ml ground glass stoppered flask,
stock solution to 250.0 ml with dimethyl sulphoxide. add 25.0 ml of phthalic anhydride solution and a few glass
beads and swirl to dissolve. Boil gently under a reflux
Propylene oxide stock solution. Introduce about 7 ml of condenser for 60 minutes, cool and add 2 quantities, each of
dichloromethane into a volumetric flask, add 0.5 g ofpropylene 10 ml, ofpyridine, through the condenser. Add 10 ml ofwater,
oxide and dilute to 10.0 ml with dichloromethane. Dilute 0.5 mix and allow to stand for 10 minutes. Add 40.0 ml of 0.5 M
mlofthis solution to 50.0 ml with dimethyl sulphoxide. sodium hydroxide and 0.5 ml of a 1.0 per cent w/v solution of
Propylene oxide solution. Dilute 1.0 ml ofthe propylene oxide phenolphthalein in pyridine. Titrate with 0.5 M sodium
stock solution to 50.0 ml with dimethyl sulphoxide. hydrOXide to a light pink endpoint that persists for 15 second
and record the volume ofsodium hydroxide used (S). Prepare
Dioxan solution. Introduce 0.1 g of dioxan into a flask and
a blank Record the volume of sodium hydroxide used (B).
dilute to 50.0 ml with dimethyl sulphoxide. Dilute 2.5 ml ofthis
Calculate the average relative molecular mass using the
solution to 100.0 ml with dimethyl sulphoxide.
following expression:
Mixture solution. Dilute a mixture of 6.0 ml of the ethylene
Oxypropylene:oxyethylene ratio. Nuclear magnetic resonance
oxide solution, 6.0 ml ofthe propylene oxide solution and 2.5
spectrometry (2.434).
ml ofthe dioxan solution to 25.0 ml with dimethyl sulphoxide.
Use a 10.0 per cent w/v solution of the substance under
Test solution. To 1.0 g ofthe substance under examination in
examination in deuterated chloroform. Record the average
a head-space vial, add 4.0 ml ofdimethyl sulphoxide and close
area of the doublet appearing at about 1.08 ppm due to the
the vial immediately.
methyl groups ofthe oxypropylene units (A I) and the average
Reference solution. To 1.0 g ofthe substance under examination area of the composite band from 3.2 ppm to 3.8 ppm due to
in a head-space vial, add 2.0 ml of dimethyl sulphoxide and CH20 groups ofboth the oxyethylene and oxypropylene units
2.0 ml ofthe mixture solution. Close the vial immediately. and the CHO groups of the oxypropylene units (A 2) with
Chromatographic system reference to the internal standard. Calculate the percentage
- a fused silica column 50 m x 032 mm, packed with of oxyethylene, by weight, in the sample under examination
poly(dimethyl)(diphenyl)siloxane (film thiclmess 5 J.l.m), using the following expression:
- temperature: Total ash (23.19). Not more than 0.4 percent.
column 70 0 to 240°,
Water (2.3.43). Not more than 1.0 per cent, determined on
inlet port and detector at 250 0 ,
LOg.
- a flame ionisation detector,
- flow rate. 1.4 ml per minute using nitrogen as the carrier Storage. Store protected from moisture.
gas, Labelling. The label states the type ofpoloxamer.
Head-space conditions
- equilibration temperature 1100 ,
- equilibration time 30 minutes,
- transfer line temperature 140 0 , Polyethylene Glycol 1500
- pressurisation time 1 minute,
- injection time 0.05 minute. Macrogol1500

Inject 1 J.l.I of the gaseous phase of the test solution and the Polyethylene Glycol 1500 is a mixture ofthe polycondensation
reference solution. The relative retention time with reference products of ethylene oxide and water obtained under
to ethylene oxide is about 6 minutes, for propylene oxide is controlled conditions. It is represented by the formula
about 1.3, for dichloromethane is about 1.6, for dioxan is about HOCH2[CH20CH2]nCH20H, where n is between 28 and 36.
3.0, for dimethyl sulphoxide is about 3.7. Category. Pharmaceutical aid (ointment base).

1929

www.webofpharma.com
POLYETHYLENE GLYCOL 1500 IP 2010

Description. A white or creamy white, soft, wax-like solid; Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium
odour, faint and characteristic. carbonate, add 10 ml of bromine solution and mix thoroughly.
Evaporate to dryness on a water-bath, gently ignite and
Tests dissolve the cooled residue in 16 ml of brominated
Appearance of solution (2.4.1). A25.0 per centw/v solution is hydrochloric acid and 45 ml of water. Remove the excess of
not more intensely coloured than reference solution BYS6. bromine with 2 ml of stannous chloride solution AsT. The
resulting solution complies with the limit test for arsenic
pH (2.4.24). 4.0 to 7.0, determined ina5.0 percentw/v solution. (3 ppm).
Freezing point (2.4.11).42° to 46°. Heavy metals (2.3.13). Dissolve 4.0 g in 5 ml ofa 1.0 per cent
Hydroxyl value (2.3.27).70 to 86, determined on 5.0 g. w/v solution of hydrochloric acid and sufficient water to
produce 25 ml. The solution complies with the limit test for
Viscosity (2.4.28). 25 mm2s- 1 to 32 mm2s-1, determined at 100°
heavy metals, Method A (5 ppm).
by Method A using a V-tube viscometer (size D).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium
carbonate, add 10 ml of bromine solution and mix thoroughly. Storage. Store protected from moisture.
Evaporate to dryness on a water-bath, gently ignite and
dissolve the cooled residue in 16 ml of brominated
hydrochloric acid and 45 ml of water. Remove the excess of
bromine with 2 ml of stannous chloride solution AsT. The Polyethylene Glycol 6000
resulting solution complies with the limit test for arsenic
(3 ppm).
Macrogo16000

Heavy metals (2.3.13). Dissolve 4.0 g in5 ml ofa 1.0 percent Polyethylene Glycol 6000 is a mixture ofthe polycondensation
w/v solution of hydrochloric acid and sufficient water to products of ethylene oxide and water obtained under
produce 25 m!. The solution complies with the limit test for controlled conditions. It is represented by the formula
heavy metals, MethodA(5 ppm). HOCH2[CH20CH2]nCH20H, where n is between 112 and 158.

Sulphated ash (2.3.18). Not more than 0.1 percent. Category. Pharmaceutical aid (ointment base and tablet
excipient).
Storage. Store protected from moisture.
Description. A white to creamy white, wax-like solid or flakes;
odour, faint and characteristic.

Polyethylene Glycol 4000 Tests


Macrogo14000 Appearance ofsolution (2.4.1). A 15.0 per cent w/v solution is
Polyethylene Glycol 4000 is a mixture ofthe polycondensation not more intensely coloured than reference solution BYS6.
products of ethylene oxide and water obtained under pH (2.4.24). 4.5 to 7.5, determined in a5.0percentw/v solution.
controlled conditions. It is represented by the formula
Freezing point (2.4.11). 56° to 60°.
HOCH2[CH20CH2]IICH20H, where n is between 69 and 84.
Viscosity (2.4.28). 250 mm2s- 1 to 390 mm 2s- l , determined at
Category. Phannaceutical aid (ointment base).
100° by Method A using a V-tube viscometer (size F).
Description. A creamy white, hard, wax-like solid, powder or
Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydroussodium
flakes; odour, faint llndcharacteristic.
carbonate, add 10 ml of bromine solution and mix thoroughly.
Tests Evaporate to dryness on a water-bath, gently ignite and
dissolve the cooled residue in 16 ml of brominated
Appearance of solution (2.4.1). A20.0 per cent w/v solution is hydrochloric acid and 45 ml of water. Remove the excess of
not more intensely coloured than reference solution BYS6. bromine with 2 ml of stannous chloride solution AsT. The
pH (2.4.24). 4.5 to 7.5, determined in a 5.0 per cent w/v solution. resulting solution complies with the limit test for arsenic
(3 ppm).
Freezing point (2.4.11). 53° to 56°.
Heavy metals (2.3.13). Dissolve 4.0 g in 5 ml ofa 1.0 percent
Hydroxyl value (2.3.27). 30 to 36, detennined on 20.0 g. w/v solution of hydrochloric acid and sufficient water to
Viscosity (2.4.28).76 mm2s-1 to 110 mm2s- l , determined at 100° produce 25 ml. The resulting solution complies with the limit
by Method A using a V-tube viscometer (size E). test for heavy metals, Method A (5 ppm).

1930

www.webofpharma.com
IP 2010 POLYOXYL 40 HYDROGENATED CASTOR OIL

Sulphated ash (2.3.18). Not more than 0.1 per cent. filtrate and rinsing in a 50-ml Nessler cylinder, dilute with water
to 40 ml, and mix.
Storage. Store protected from moisture.
To each of the cylinder containing the standard solution and
the test solution, add 2 ml of pH 3.5 acetate buffer, then add
1.2 ml of thioacetamide- glycerine base, dilute with water to
Polyoxyl 35 Castor Oil 50 ml, mix, allow to stand for 2 minutes, and view downward
Polyoxy135 Castor Oil contains mainly the tri-ricinoleate ester over a white surface: the colour ofthe solution obtained from
ofethoxylated glycerol, with smaller amounts ofpolyethylene the test solution is not more intense than that of the solution
glycol ricinoleate and the corresponding free glycols. It results obtained from the standard solution, produced by treating 2
from the reaction of glycerol ricinoleate with about 35 moles mlof lead standard solution (l ppm) in a similar manner.
of ethylene oxide. Sulphated ash (2.3.18). Not more than 0.3 per cent.
Water (2.3.43). Not more than 3.0 per cent, determined on 1 g.
Identification
Storage. Store protected from moisture.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with polyoxyl 35
castor oil RS or with the reference spectrum of polyoxyl 35
castor oil. Polyoxyl 40 Hydrogenated Castor Oil
B. Dissolve about 0.1 g in 10 ml of ethanolic potassium Polyoxy140 Hydrogenated Castor Oil contains mainly the tri-
hydroxide solution, boil for about 3 minutes and evaporate to hydroxystearate ester of ethoxylated glycerol, with smaller
dryness. The residue dissolve with 5 ml of water, yielding a amounts of polyethylene glycol tri-hydroxystearate and of
clear solution. Add a few drops of glacial acetic acid; a white the corresponding free glycols. It results from the reaction of
precipitate is formed. glycerol tri-hydroxystearate with about 40 to 45 moles of
ethylene oxide.
C. To a 5 .0 per cent w/v solution, add bromine solution,
dropwise, the bromine is decolorized. Category. Pharmaceutical aid.
Identification
Tests
A. Dissolve 0.1 gin 1 ml of water, add 9 ml of5.0percentw/v
Weight per ml (2.4.29). 1.05 to 1.06. solution of sodium chloride and heat on a water-bath, the
Viscosity (2.4.28). 600 to 850 centipoises at 25°, a capillary solution becomes turbid at a temperature between 700 and 85°.
viscometer being used.
B. Dissolve about 0.1 g in 10 ml of ethanolic potassium
Acid value (2.3.23) Not more than 2.0.· hydroxide solution, boil for about 3 minutes, and evaporate
to dryness. Dissolve the residue with 5 ml of water, yielding a
Hydroxyl value (2.3.27). 65 to 80.
clear solution. Add a few drops of glacial acetic acid; a white
Saponification value (2.3.37).60 to 75. precipitate is fonned.
Iodine value (2.3.28).25 to 35. Tests
Heavy metals (2.3 .13). Weigh about 2 g ofthe substance under Congealing temperature (2.4.10). 16° to 26°.
examination in a crucible and add sufficient sulphuric acid to
wet the substance, carefully ignite at a low temperature until Acid value (2.3.23). Not more than 2.0.
thoroughly charred. Add to the carbonized mass 2 ml of nitric Hydroxyl value (2.3.27).60 to 80.
acid and 5 drops of sulphuric acid, and heat cautiously until
Saponification value (2.3 .37).45 to 69.
white fumes no longer are evolved. Ignite, preferably in a
muffle furnace, at 500° to 600 0 , until the carbon is completely Iodine value (2.3.28). Not more than 2. O.
burned off. Cool, add 4 ml of dilute hydrochloric acid, cover, Heavy metals (2.3.13). Weigh 2 g of the substance under
digest on a steam-bath for 15 minutes, and slowly evaporate examination in a crucible and add sufficient sulphuric acid to
on a steam-bath to dryness. Moisten the residue with 1 drop wet the substance, carefully ignite at a low temperature until
of hydrochloric acid, add 10 ml ofhot water, and digest for 2 thoroughly charred. Add to the carbonized mass 2 ml of nitric
minutes. Add dilute ammonium hydroxide dropwise until the acid and 5 drops of sulphuric acid, and heat cautiously until
solution is just allcaline to litmus paper, dilute with water to 25 white fumes no longer are evolved. Ignite, preferably in a
ml, and adjust at pH 3.0 with dilute acetic acid, filter. Rinse muffle furnace, at 5000 to 600 0 , until the carbon is completely
the crucible and the filter with 10 ml of water, combine the burned off. Cool, add 4 ml of dilute hydrochloric acid, cover,

1931

www.webofpharma.com
POLYSORBATE 20 IP 2010

digest on a stearn bath for 15 minutes, and slowly evaporate B. Dissolve 0.1 ginS mlofchlorojorm, add 0.1 gofpotassium
on a stearn bath to dryness. Moisten the residue with 1 drop thiocyanate and 0.1 g of cobalt nitrate and stir with a glass
ofhydrochloric acid, add 10 ml ofhot water, and digest for 2 rod; a blue colour is produced.
minutes. Add dilute ammonium hydroxide dropwise until the
solution is just alkaline to litmus paper, dilute with water to 25 Tests
ml, and adjust with diluteacetic acid to a pH between 3.0 and pH (2.4.24). 5.0 to 7.0, determined in a 5.0 per cent w/v solution
4.0, using short-range pH indicator paper as an external in carbon dioxide-free water.
indicator. Filter if necessary, rinse the crucible and the filter
with 10 ml ofwater, combine the filtrate and rinsing in a 50-rnl Acid value (2.3.23). Not more than 2.0, determined on 5.0 g.
Nessler cylinder, dilute with water to 40 ml, and mix. Hydroxyl value (2.3.27). 96 to 108, determined on 2.0 g.
To each of the cylinder containing the standard solution and Saponification value (2.3.37). 40 to 50, using 15 ml of0.5 M
the test solution, add 2 ml of acetate buffer pH 3.5, then add ethanolic potassium hydroxide and diluting with 50 ml of
1.2 ml of thioacetamide- glycerine base, dilute with water to water before carrying out the titration.
50 ml, mix, allow to stand for 2 minutes, and view downward
Iodine value (2.3.28). Not more than 5.0, determined by the
over a white surface: the colour ofthe solution obtained from
iodine bromide method.
the test solution is not more intense than that of the solution
obtained from the standard solution, produced by treating 2 Heavy metals (2.3.13). 2.0 g complies with the limit test for
mlof lead standard solution (l ppm) in a similar manner. heavy metals, Method B (l0 ppm).
Sulphated ash (2.3.18). Not more than 0.3 per cent. Reducing impurities. Dissolve 2.0 g in 25 ml ofhot water, add
25 ml of 1 M sulphuric acid and 0.1 ml ofjerroin solution and
Water (2.3.43). Not more than 3.0 per cent, determined on 1 g.
titrate with 0.01 M eerie ammonium sulphate shaking
Storage. Store protected from moisture. continuously until the colour changes from red to greenish
blue persists for 30 seconds. Carry out a blank titration. Not
more than 2.0 ml of 0.01 M eerie ammonium sulphate is
required.
Polysorbate 20
Sulphated ash. Not more than 0.2 per cent, determined by the
Polyoxyethylene 20 Sorbitan Monolaurate following method. Weigh accurately about 2.0 g in a silica or
platinum crucible, add 0.5 ml ofsulphuric acid and heat on a
water-bath for 2 hours. Carefully ignite at a low temperature
until thoroughly chaned. Add to the carbonised mass 2 ml of
nitric acid and 0.25 ml ofsulphuric acid, cautiously heat until
white fumes are evolved and then ignite at 6000 until the. carbon
is completely burnt off. Allow to cool, weigh and repeat the
operation for periods of 15 minutes until two successive
weighings do not differ by more than 0.5 mg.
Water (2.3.43). Not more than 3.0 per cent, determined on 1.0 g.
Polyoxyethylene 20 Sorbitan Monolaurate
Storage. Store protected from light and moisture.
Polysorbate 20 is a mixture ofpartial lauric esters of D-glucitol
and its anhydridescopolyrnerised with approximately 20 moles
ofethylene oxide for each mole of D-glucitol and its anhydrides.
The lauric acid used for the esterification may contain other Polysorbafe80
fatty acids.
Polyoxyethylene 80 Sorbitan Monooleate
Category. Pharmaceutical aid (non-ionic surfactant).
Description. A clear or slightly opalescent, oily, yellowish or
brownish yellow liquid; odour, faint and characteristic.

Identification

A. Determine by infrared absorption spectrophotometry (2.4.6).


Compare the spectrum with that obtained with polysorbate
RS or with the reference spectrum of polysorbate. w+x+y+z =20

1932

www.webofpharma.com
IP 2010 POTASSIUM CHLORIDE

Polysorbate 80 is a mixture ofpartial oleic esters of D-glucitol Potassium Chloride


and its anhydrides copolymerised with approximately 20 moles
ofethylene oxide for each mole ofD-glucitol and its anhydrides. KCl Mol. Wt. 74.6
Category. Pharmaceutical aid (non-ionic surfactant). Potassium Chloride contains not less than 99.0 per cent and
not more than 100.5 per cent of KC1, calculated on the dried
Description. Aclear or almost clear, oily, yellowish or brownish basis.
yellow liquid; odour, faint and characteristic.
Category. Electrolyte replenisher.
Identification
Dose. Prophylactic; 2 to 4 g (approximately 25 to 50 mmol)
A. Determine by infrared absorption spectrophotometry (2.4.6). daily; therapeutic, in established potassium depletion, 10 to
Compare the spectrum with that obtained with polysorbate 15 g (approximately 135 to 200 mmol) daily.
RS or with the reference spectrum of polysorbate.
Description. Colourless crystals or a white, crystalline powder;
B. Dissolve 0.1 gin 5 ml of chloroform, add 0.1 g ofpotassium odourless.
thiocyanate and 0.1 g of cobalt nitrate and stir with a glass
rod; a blue colour is produced. Identification
C. To 2 ml of a 5 per cent w/v solution add 0.5 ml of bromine Dissolve 109 in 100 ml of carbon dioxide-fi-ee water prepared
water; the bromine is decolourised. from distilled water (solution A). The solution gives the
Tests reactions of potassium salts and of chlorides (2.3.1).

pH (2.4.24).6.0 to 8.0, determined in a 5.0 per cent w/v solution Tests


in carbon dioxide-free water.
Acid value (2.3.23). Not more than 2.0, determined on 5 g. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Hydroxyl value (2.3.27). 65 to 80, determined on 2.0 g.
Acidity or alkalinity. 5.0 g dissolved in 50 rnl ofcarbon dioxide-
Saponification value (2.3.37). 45 to 55, using 15 ml of 0.5 M free water requires not more than 0.5 ml of 0.01 M sodium
ethanolic potassium hydroxide and diluting with 50 ml of hydroxide or 0.01 M hydrochloric acid for neutralisation to
water before carrying out the titration. bromothymol blue solution.
Iodine value (2.3.28). 18 to 24, determined by the iodine bromide
Arsenic (2.3.10). Dissolve 10.0 g in 50 ml of water and add
method.
10 ml of stannated hydrochloric acid AsT. The resulting
Reducing impurities. Dissolve 2.0 g in 25 ml ofhot water, add solution complies with the limit test for arsenic (1 ppm).
25 ml of 1 M sulphuric acid and 0.1 ml offerroin solution and
Barium. To 5 ml of solution A add 5 ml of water and 1 ml of
titrate with 0.01 M eerie ammonium sulphate shaking
1 M sulphuric acid; the solution, after not less than 15 minutes,
continuously until the colour changes from red to greenish
is not more opalescent than a mixture of5 ml ofsolution A and
blue persists for 30 seconds. Carry out a blank titration. Not
6 ml of water.
more than 5.0 ml of 0.01 M eerie ammonium sulphate is
required. Heavy metals (2.3.13). Dissolve 2.0 g in 10 ml of water to
Heavy metals (2.3.13). 2.0 g complies with the limit test for
which are added 2 ml of dilute acetic acid and 13 ml of water.
heavy metals, Method B (10 ppm). The solution complies with the limit test for heavy metals,
Method A (10 ppm).
Sulphated ash. Not more than 0.2 per cent, determined by the
following method. Weigh accurately about 2.0 g in a silica or Calcium and magnesium. Dissolve 1gin 20 ml of water, add
platinum crucible, add 0.5 ml of sulphuric acid and heat on a 1 ml of dilute ammonia solution and 1ml ofsodium phosphate
water-bath for 2 hours. Carefully ignite at a low temperature solution; the solution remains clear.
until thoroughly charred. Add to the carbonised mass 2 ml of Iron (2.3.14). 20 ml ofsolutionA complies with the limit test for
nitric acid and 0.25 ml of sulphuric acid, cautiously heat until iron (20 ppm).
white fumes are evolved and then ignite at 6000 until the carbon
Bromides. Dilute 1.0 ml ofsolution Ato 50.0 mlwith water. To
is completely burnt off. Allow to cool, weigh and repeat the
5.0 ml of the solution add 2.0 ml of phenol red reagent and
operation for periods of 15 minutes until two successive
. 1.0 ml of a 0.0 I per cent solution of chloramine T and mix
weighings do not differ by more than 0.5 mg.
immediately. After exactly 2 minutes, add 0.15 ml of 0.1 M
Water (2.3.43). Not more than 3.0 percent, determined on 1.0 g. sodium thiosulphate, mix and dilute to 10.0 ml with water. The
Storage. Store protected from light and moisture. absorbance ofthe solution measured at about 590 nm (2.4.7),

1933

www.webofpharma.com
POTASSIUM CHLORIDE IP 2010

using water as the blank, is not more than that of a standard Potassium Chloride and Dextrose
prepared at the same time and in the same manner, using 5.0 ml
of a 0.0003 per cent w/v solution of potassium bromide Injection
(0.1 percent). Potassium Chloride in Dextrose Injection; Potassium CWoride
Iodides. Moisten 5 g by adding dropwi~e, a solution freshly and Dextrose Intravenous Infusion; Potassium Chloride and
prepared by mixing 25 ml of iodide-free starch solution, 2 ml Glucose Intravenous Infusion
of 0.5 M sulphuric acid, 0.15 ml of sodium nitrite solution Potassium Chloride and Dextrose Injection is a sterile solution
and 25 ml of water and examine the mixture in daylight; the of Potassium Chloride and Dextrose in Water for Injections.
substance shows no blue colour after 5 minutes.
Potassium Chloride and Dextrose Injection contains not less
Sulphates (2.3.17). 0.5 g complies with the limit test for than 95.0 per cent and not more than 110.0 per cent of the
sulphates (300 ppm). stated amount of potassium chloride, KCl, and not less than
95.0 per cent and not more than 105.0 per cent of the stated
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
amount ofdextrose, C 6H 1Z0 6• It contains no antimicrobial agent.
on 1.0 g by drying in an oven at 105°.
Assay. Weigh accurately about 0.15 g, dissolve in 50 ml of Usual strengths: Injections containing the following amounts
ofpotassium chloride, KCl, and dextrose, C 6H IZ06.
water and titrate with 0.1 M silver nitrate using potassium
chromate solution as indicator. per cent w/v of per cent w/v of'
1 ml of 0.1 M silver nitrate is equivalent to 0.007455 g ofKCl. Potassium Chloride Dextrose
(KCl) (C6H 1Z0 6)
Potassium Chloride intended for use in the manufacture of
dialysis solutions complies with the following additional 0.Q75 5
requirement. 0.15 5
Aluminium. Dissolve 4.0 g in 100 ml of water and add 10 ml of 0.225 5
acetate buffer pH 6.0. Extract with successive quantities of 0.30 5
20, 20 and 10 ml of a 0.5 per cent w/v solution of
8-hydroxyquinoline in chloroform and dilute the combined Description. A clear, colourless or faintly straw-coloured
extracts to 50 ml with chloroform. Use as the standard solution solution.
a mixture of2 ml of aluminium standard solution (2 ppm AI),
10 ml of acetate bufferpH6.0 and 98 ml of water treated in the Identification
same manner and as the blank solution a mixture of 10 ml of
A. To I ml add 0.05 mlofpotassium cupri-tartrate solution;
acetate bufferpH 6.0 and 100 ml of water treated in the same
the solution remains blue and clear. Heat to boiling, a copious
manner. Measure the fluorescence of the test and standard
red precipitate is formed.
solutions (2.4.5), using an excitation wavelength of about
392 urn and emission wavelength ofabout 518 urn and setting B. Gives reaction B of potassium salts and reaction A of
the instrument to zero with the blank solution in each case. chlorides (2.3.1).
The fluorescence of the test is not greater than that of the
standard solution (1 ppm). Tests
Potassium chloride intended for use in the manufacture of pH (2.4.24).3.5 to 6.5.
Parenteral Preparations or for the preparation of
liaemlJdzalyszsslJlution complies· wliliiliejolllJwing 5-Hydroxymethylfurfural and Related substances. Dilute a
additional requirement. volume containing 1.0 g of Dextrose to 500.0 ml with water
and measure the absorbance of the resulting solution at the
Sodium. Not more than 0.1 per cent, determined by atomic maximum at about 284 urn, absorbance at about 284 urn (2.4.7);
absorption spectrophotometry (2.4.2), using a 1.0 per cent not more than 0.25.
w/v solution and measuring at 589 urn. Use sodium solution
AAS, suitably diluted with water, for the standard solutions. Heavy metals (2.3 .13). Evaporate a volume containing 4.0 g of
Dextrose to 10 ml and add 2 ml of dilute acetic acid and
Storage. Store protected from moisture. sufficient water to produce 25 ml. The solution complies with
the limit test for heavy metals, Method A (5 ppm).
Labelling. The label states whether or not the material is
suitable for use in the manufacture ofParenteral Preparations Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
or for the preparation ofhaemodialysis or dialysis solutions. perrnI.

1934

www.webofpharma.com
IP 2010 POTASSIUM CHLORIDE, SODIUM CHLORIDE AND DEXTROSE INJECTION

Other tests. Complies with the tests stated under Parenteral per cent w/v of per cent w/v of per cent w/v of
Preparations (Injections). Potassium Sodium Dextrose
Chloride Chloride
Assay. For potassium chloride - Titrate an accurately
(KCI) (NaCl) (C6H12 06J
measured volume containing 0.1 g ofPotassium Chloride with
0.1 M silver nitrate using potassium chromate solution as 0.075 0.250 5
indicator. 0.075 0.330 5
1 ml of 0.1 Msilver nitrate is equivalent to 0.007455 g ofKCl. 0.075 0.450 5
0.150 0.225 5
For dextrose - To an accurately measured volume
0.150 0.225 10
containing 2 g to 5 g of Dextrose, add 0.2 ml of 5 M ammonia
and sufficient water to produce 100.0 ml. Mix well, allow to 0.150 0.330 5
stand for 30 minutes and measure the optical rotation in a 0.150 0.450 5
2-dm tube (2.4.22). The observed rotation in degrees multiplied 0.150 0.900 5
by 0.9477 represents the weight, in g, ofdextrose, C 6H 1Z0 6, in 0.225 0.225 5
the volume taken for assay. 0.225 10
0.225
Storage. Store in single dose containers. 0.225 0.330 5
Labelling. The label states (1) the strength as the percentages 0.225 0.450 5
w/v ofPotassium Chloride and Dextrose; (2) the total osmolar 0.300 0.225 5
concentration in mOsmol per litre; (3) where the contents are 0.300 0.330 5
less than 100 ml, or where the label states that the Injection is 0.300 0.450 5
not for direct use but is to be diluted before use, the label 5
0.300 0.900
alternatively may state the total osmolar concentration in
mOsmol per ml; (4) the content ofpotassium in millirnoles; (5) Identification
that the injection containing visible particles in the solution
should not be used. A. To 1 ml add 0.05 ml ofpotassium cupri-tartrate solution;
the solution remains blue and clear. Heat to boilIng, a copious
red precipitate is formed.
B. Gives reaction A of potassium salts, reaction B of sodium
salts and reaction A of chlorides (2.3.1)
Potassium Chloride, Sodium Chloride Tests
and Dextrose Injection
pH (2.4.24).3.5 to 6.5, determined on a portion diluted with
Potassium Chloride in Dextrose and Sodium Chloride water, ifnecessary, to a concentration ofnot more than 5.0 per
Injection; Potassium Chloride, Sodium Chloride cent of dextrose.
and Dextrose Intravenous Infusion; Potassium 5-Hydroxymethylfurfural and Related substances. Dilute a
Chloride, Sodium Chloride and Glucose Intravenous volume containing 1.0 g of Dextrose to 500.0 ml with water
Infusion and measure the absorbance of the resulting solution at the
maximum at about 284 urn, absorbance at about 284 urn (2.4.7);
Potassium Chloride, Sodium Chloride and Dextrose Injection
not more than 0.25.
is a sterile solution of Potassium Chloride, Sodium Chloride
and Dextrose in Water for Injections. Heavy metals (2.3.13). Evaporate a volume containing 4.0g of
Dextrose to 10 ml and add 2 ml of dilute acetic acid and
Potassium Chloride, Sodium Chloride and Dextrose Injection sufficient water to produce 25 ml. The solution complies with
contains not less than 95.0 per cent and not more than the limit test for heavy metals, Method A (5 ppm).
110.0 per cent ofthe stated amounts ofsodium, Na, potassium,
K, and chloride, Cl, and not less than 95.0 per cent and not Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
more than 105.0 per cent of the stated amount of dextrose, perml.
C 6H 1Z 0 6 • It contains no antimicrobial agent. Other tests. Complies with the tests stated under Parenteral
Usual strengths: Injections containing the following amounts Preparations (Injections).
of potassium chloride, KCl, sodium chloride, NaCl, and Assay. For sodium - Dilute appropriately with water and
dextrose, C 6H 1Z0 6 • determine by Method A for flame 'photometry (2.4.4), or by

1935

www.webofpharma.com
POTASSIUM CHLORIDE, SODIUM CHLORIDE AND DEXTROSE INJECTION IP 2010

Method A for atomic absorption spectrophotometry (2.4.2), Category. Systemic alkaliniser.


measuring at 589 nm and using sodium solution FP or sodium
Dose. 4 to 109.
solution AAS respectively, suitably diluted with water for the
standard solutions. Description. White, granular crystals or a crystalline powder;
odourless; hygroscopic,
For potassium - Dilute appropriately with water and
determine by Method A for flame photometry (2.4.4), or by Identification
Method A for atomic absorption spectrophotometry (2.4.2),
measuring at 767 nm and using potassium solution FP or A. Dissolve 10 g in 100 ml of carbon dioxide-free water
potassium solution AAS respectively, suitably diluted with prepared from distilled water (solution A). 0.5 ml of solution
water for the standard solutions. A gives the reactions of potassium salts (2.3.1).
For total chlorides - To 20.0 ml add 30 ml of water, 50.0 ml of B. To 1 ml of solution A add 4 ml of water; the solution gives
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the the reactions of citrates (2.3.1).
precipitate with water slightly acidified with nitric acid and
titrate the excess of silver nitrate with 0.1 M ammonium
Tests
thiocyanate using ferric ammonium sulphate solution as Appearance of solution. Solution A is clear (2.4.1), and
indicator until a reddish yellow colour is produced. Carry out colourless (2.4.1).
a blank titration.
Acidity or alkalinity. To 10 ml of solution A add 0.1 mlof
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g oftotal dilute phenolphthalein solution; not more than 0.2 ml of
chloride, calculated as Cl. 0.1 M hydrochloric acid or 0.1 M sodium hydroxide is
For dextrose- To an accurately measured volume containing required to change the colour ofthe·solution.
2 g to 5 g ofDextrose, add 0.2 ml of5 M ammonia and sufficient Arsenic (2.3.10). Dissolve 5.0 gin 50 ml of water and add 15 ml
water to produce 100.0 ml. Mix well, allow to stand for of stannated hydrochloric acid. The resulting solution
30 minutes and measure the optical rotation in a 2-dm tube complies with the limit test for arsenic (2 ppm).
(2.4.22). The observed rotationin degrees multiplied by 0.9477
Heavy metals (2.3.13). 2.0 g complies with the limit test for
represents the weight, in g, ofdextrose, C6H 1Z0 6, in the volume
heavy metals, Method A (1 0 ppm).
taken for assay.
Sodium. Not more than 0.3 per cent, determined by atomic
Storage. Store in single dose containers.
absorption spectrophotometry (2.4.2), Method II, measuring
Labelling. The label states (1) the strength as the percentages at 589 nm. Prepare the test solution by addition of 1 ml of2 M
w/v ofPotassium Chloride, Sodium Chloride and Dextrose; (2) hydrochloric acid to 10.0 ml of solution A and diluting to
the total osmolar concentration in mOsmol per litre; (3) where 100.0 ml with distilled water. Use sodium solution AAS,
the contents are less than 100 ml, or where the label states that suitably diluted with distilled water, for the standard
the Injection is not for direct use but is to be diluted before solutions.
use, the label alternatively may state the total osmolar
Chlorides (2.3.12). Dilute 25.0 ml ofsolution A to 35 ml with
concentration in mOsmol per ml; (4) the content ofpotassium,
sodium and chloride in millimoles; (5) that the injection
water. The resulting solution complies with the limit test for
containing visible particles in the solution should not be used. chlorides (l 00 ppm).
Oxalate. Dissolve 0.5 gin 4 ml ofwater, add 3 ml ofhydrochloric
acid and 1 g of granulated zinc and heat on a water-bath for
1 minute. Allow to stand for 2 minutes, decant into 0.25 ml ofa
Potassium Citrate 1 per cent w/v solution of phenylhydrazine hydrochloride,
heat to boiling, cool rapidly, add an equal volume of
HO COOK
._~-- ,H 0 hydrochloric acid and 0.25 ml of potassium ferricyanide
2
KOOC-/ ~COOK solution, shake and allow to stand for 30 minutes. Any pink
colour produced is not more intense than that obtained by
C6HsK307,HzO Mol. Wt. 324.4 treating 4.0 ml of a 0.005 per cent w/v solution of oxalic acid
Potassium citrate is tripotassium 2-hydroxypropane-l ,2,3- at the same time and in the same manner (300 ppm, calculated
tricarboxylate monohydrate. as anhydrous oxalic acid).
Potassium Citrate contains notless than 99.0 per cent and not Sulphates (2.3.17). To 10.0 ml ofsolution A add 2 ml of 7 M
more than 101.0 per cent of C 6H sK30 7, calculated on the hydrochloric acid and dilute to 15 ml with distilled water; the
anhydrous basis. solution complies with the limit test forsulphates (150 ppm).

1936

www.webofpharma.com
IF 2010 POTASSIUM CLAVULANATE

Readily carbonisable substances. Heat 0.20 g, in powder, with Test solution. Dissolve 0.25 g of the substance under
10 ml of sulphuric acid (96 per cent w/w) in a water-bath at examination in mobile phase A and dilute to 25 ml with mobile
90° ± 1° for 60 minutes and cool rapidly. The solution is not phase A.
more intensely coloured than reference solution YS2 or GYS2
Reference solution (a). Dilute 1 ml of the test solution to 100
(2.4.1).
ml with mobile phaseA.
Water (2.3.43). 4.0 to 7.0 per cent, determined on 0.5 g, titration
Reference solution (b). A solution containing 0.01 per cent wi
being done after stirring for 15 minutes subsequent to addition
veach of lithium clavulanate RS and amoxycillin trihydrate
of the substance under examination.
RS in mobile phase A.
Assay. Weigh accurately about 0.2 g, dissolve in 20 ml of Chromatographic system
anhydrous glacial acetic acid, heat to about 50°, allow to
cool. Titrate with 0.1 M perchloric acid, using 0.25 ml of - a stainless steel column 10 cm x 4.6 mm, packed with
l-naphtholbenzein solution as indicator and titrating until a octadecylsilane bonded to porous silica (5 f.lm),
green colour is obtained. Carry out a blank titration. - column temperature. 40°,
- mobile phase: A. a 0.78 per cent w/v solution ofsodium
1 ml of 0.1 M perchloric acid is equivalent to 0.01021 g of dihydrogen phosphate adjusted to pH 4.0 with
C6HsK 30 7• phosphoric acid and filtered through a 0.5 f.lm filter,
Storage. Store protected from moisture. B. a mixture of equal volumes ofmobile
phase A and methanol,
a linear gradient programme using the conditions given
below,
Potassium Clavulanate - flow rate. 1 ml per minute,
- spectrophotometer set at 230 om,

o~rr -~
O~K - injection volume. 20 f.ll.
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent v/v)
Lj--o OH 0-4 100 o
H 4-15 100--750 050
CsHsKNOs Mol. Wt. 237.3 15 -18 50 50
Potassium Clavulanate is potassium (Z)-(2R,5R)-3-(2- 18-24 50--7100 50--7 0
hydroxyethylidene)-7-oxo-4-oxa-l-azabicyclo[3.2.O]heptane- 24- 39 100 o
2-carboxylate. Inject reference solution (b). The resolution between
Potassium Clavulanate contains not less than 96.5 per cent clavulanate (first peak) and amoxycillin (second peak) is not
and not more than 102.0 per cent of potassium clavulanate, less than 13.
CsHsKNOs, calculated on the anhydrous basis. Inject the test solution and reference solution (b). The area of
Category. Antibacterial. any individual impurity peak obtained is not more than the
Description. A white to off white, crystalline hygroscopic area of the principal peak in the chromatogram obtained with
powder. reference solution (a) (1.0 per cent). The sum of all impurity
peaks obtained is not more than twice the area ofthe principal
Identification peak in the chromatogram obtained with reference solution
(a) (2.0 per cent). Ignore any peaks with an area 0.05 times the
A. In the Assay, the principal peak in the chromatogram
area ofthe principal peak in the chromatogram obtained with
obtained with the test solution corresponds to the peak in the
reference solution (a) (0.05 per cent).
chromatogram obtained with reference solution (a).
Water (2.3.43). Not more than 1.5 percent,determinedonO.5 g.
B. Gives reaction A ofpotassium salts (2.3.1).
Assay. Determine by liquid chromatography (2.4.14).
Tests
NOTE -Prepare the solutions immediately before use.
pH (2.4.24).5.5 to 8.0, determined in 1.0 per cent w/v solution.
Test solution. Dissolve 50.0 mg of the substance under
Related substances. Determine by liquid chromatography examination in a 0.41 per cent w/v solution of sodium acetate
(2.4.14). previously adjusted to pH 6.0 with glacial acetic acid, and
NOTE - Prepare the solutions immediately before use. dilute to 50.0 ml with the same solution.

1937

www.webofpharma.com
POTASSIUM CLAVULANATE IP 2010

Reference solution (a). AO.1 per cent w/v solution of lithium Potassium Clavulanate Diluted contains not less than 91.2 per
clavulanate RS in a 0.41 per cent w/v solution of sodium cent and not more than 107.1 per cent ofthe stated amount of
acetate previously adjusted to pH 6.0 with glacial acetic potassium clavulanate, CsHsKNOs.
acid. Category. Antibacterial.
Reference solution (b). A solution containing 0.1 per cent wi
Description. A white or almost white powder, hygroscopic.
veach of lithium clavulanate RS and amoxycillin trihydrate
RS in a 0.41 per cent w/v solution ofsodium acetate previously
Identification
adjusted to pH 6.0 with glacial acetic acid.
Chromatographic system A. In the Assay, the principal peak in the chromatogram
a stainless steel column 30 cm x 4.6 mm, packed with obtained with the test solution corresponds to the peak in the
octadecy1silane bonded to porous silica (5 /lm), chromatogram obtained with reference solution (a).
- mobile phase: a mixture of 5 volumes of methanol and B. Gives reaction A ofpotassium salts (2.3.1).
95 volumes of a 1.5 per cent w/v solution of sodium
dihydrogen phosphate previously adjusted to pH 4.0 C. Depending on the diluent used, carry out one ofthe relevant
with dilute phosphoric acid, identification tests given below.
flow rate. 1 ml per minute, (i) On a watch-glass place a quantity of the substance under
spectrophotometer set at 230 nm, examination corresponding to 20 mg ofcellulose and disperse
injection volume. 10 Ill. in 4 ml of iodinated zinc chloride solution. The powder
Inject reference solution (b). The resolution between becomes violet-blue in colour.
clavulanate (first peak) and amoxycillin (second peak) is not (ii) It gives the reaction ofsilicates (2.3.1).
less than 3.5.
Inject alternately the test solution and reference solution (a). Tests
Calculate the content ofCsHsKNOs. pH (2.4.24) 4.8 to 8.0, determined on a quantity ofthe substance
1 mg of clavulanate (CsHgNO s) is equivalent to 1.191 mg of under examination containing 0.2 g ofpotassium clavulanate
CsHsKNOs. dissolved in 20 ml of carbon dioxide-free water.

Potassium Clavulanate intendedfor use in the manufacture Light absorption. When examined in the range 230 nm to 360
ofParenteral Preparations without a further procedure for nm (2.4.7), a 0.1 per cent w/v solution in 0.1 M phosphate
the removal of bacterial endotoxins complies with the buffer solution pH 7.0 shows an absorption maximum 0.40
folloWing additional requirement. measured immediately at 278 nm.

Bacterial endotoxins (2.2.3). Not more than 0.03 Endotoxin Related substances. Determine by liquid chromatography
Unit per mg of potassium clavulanate. (2.4.14).

Potassium Clavulanate intendedfor use in the manufacture Test solution. Dissolve a quantity of the substance under
of Parenteral Preparations without a further sterilisation examination containing 0.25 g of potassium clavulanate in 5
procedure complies with the folloWing additional ml ofmobile phase A, dilute to 25.0 ml with mobile phase A and
requirement. filter.

Sterility (2.2.11). Complies with the test for sterility. Reference solution (a). Dilute 1.0 ml of the test solution to
100.0 ml with mobile phase A.
Storage. Store protected from moisture.
Reference solution (b). Dissolve 10 mg of amoxycillin
Labelling. The label states whether it is intended for use in
trihydrate RS in 1 ml of the test solution and dilute to 100 ml
the manufacture of parenteral preparations.
with mobile phase A.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with
Potassium Clavulanate Diluted octadecylsilane bonded to porous silica (5 /lm),
- mobile phase: A. a 0.78 per cent w/v solution of sodium
Mol. Wt. 237.3 dihydrogen phosphate with the pH adjusted to 4.0 with
Potassium Clavulanate Diluted is a dry mixture of Potassium dilute phosphoric acid,
Clavulanate and Microcrystalline Cellulose, or Silica, colloidal B. a mixture of equal volumes of mobile
anhydrous or Silica, colloidal hydrated. phase A and methanol,

1938

www.webofpharma.com
IP 2010 POTASSIUM IODIDE

- flow rate. 1 ml per minute, Inject reference solution (b). The resolution between the
- a linear gradient programme using the conditions given clavulanate peak and amoxycillin peak is not less than 3.5.
below,
Inject alternately the test solution and reference solution (a)..
- spectrophotometer set at 230 nm,
- injection volume. 20 Ill. Calculate the content ofCsHsKNOs .
Time Mobile Mobile Comment 1 mg of clavulanate (CsHgNOs) is equivalent to 1.191 mg of
phase A phase B CsHsKNOs.
(in min.) (per cent vIv) (per cent v/v)
Storage. Store protected from moisture.
o 100 o Isocratic
Labelling. The label states the percentage contents of
4 100 o begin linear potassium clavulanate and the diluent used to prepare the
gradient mixture.
15 50 50
18 50 50 end chromatogram,
return to 100A
24 100 0 end equilibration, Potassium Iodide
begin'next K.I Mol. Wt. 166.0
chromatogram
Potassium Iodide contains not less than 99.0 per cent and
Inject reference solution (b). The resolution between the not more than 100.5 per cent ofKI, calculated on the dried
clavulanate (first peak) and amoxycillin (second peak) is not basis.
less than 13.
Category. Antithyroid; antifungal; expectorant.
Inject alternatively the test solution and reference solution
Dose. In preoperative treatment of thyrotoxicosis, 30 to 60
(a). Any individual impurity is not more than 1.0 per cent and
mg; as expectorant, 250 to 500 mg.
the sum of all impurities found is not more than 2.0 per cent.
Description. Colourless crystals or a white powder; odourless.
Water (2.3.43). Not more than 2.5 per cent, determined on
LOg. Identification
Assay. Determine by liquid chromatography (2.4.14). Dissolve 109 in 100 ml of carbon dioxide-ji-ee water (solution
Test solution. Dissolve a quantity of the substance under A). The solution gives the reactions of potassium salts, and
examination containing about 50.0 mg of potassium ofiodides (2.3.1).
clavulanate in a 0.4 per cent w/v solution of sodium acetate
Tests
with the pH previously adjusted to 6.0 with glacial acetic
acid, dilute to 50.0 ml with the same solution and filter. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Refei-ence solution (a). Dissolve 50.0 mg of lithium
clavulanate RS in a 0.4 per cent w/v solution ofsodium acetate Alkalinity. To IOmlofsolutionAaddO.2rnlofO.Ol Msulphuric
with the pH previously adjusted to 6.0 with glacial acetic acid; no colour is produced on addition of a drop of
acid and dilute to 50.0 ml with the same solution. phenolphthalein solution.
Reference solution (b). Dissolve 10 mg of amoxycillin Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and 12 ml of
trihydrate RS in 10 ml ofreference solution (a). stannated hydrochloric acid AsT. The resulting solution
complies with the limit test for arsenic (2 ppm).
Chromatographic system
- a stainless steel column 30 em x 4.6 mm, packed with Heavy metals (2.3.13). 2.0 g complies with the limit test for
octadecylsilane bonded to porous silica (10 jJ.m), heavy metals, Method A (1 0 ppm).
mobile phase: a mixture of 5 volumes of methanol and Iron (2.3.14). 20 rnl ofsolutionA complies with the limit test for
95 volumes of a 1.5 per cent w/v solution of sodium iron (20 ppm).
dihydrogen phosphate with the pH previously
Bar~um. Dissolve 0.5 g in 10 ml of water and add 1 mlofdilute
adjusted to 4.0 with dilute phosphoric acid,
sulphuric acid; no turbidity develops within one minute.
flow rate. 1 ml per minute,
spectrophotometer set at 230 urn, Cyanide. Warm 5 ml of Solution A, add one drop offerrous
injection volume. 20 Ill. sulphate solution and 0.5 ml of sodium hydroxide solution

1939

www.webofpharma.com
POTASSIUM IODIDE IP 2010

and acidify with hydrochloric acid; no blue colour is Tests


produced.
Appearance of solution. Dissolve 1.5 g in 50 m1 of distilled
Iodate. Dissolve 0.5 g in 10 ml of carbon dioxide-free water water, heat on a water-bath and add gradually 6 ml of ethanol
and add 0.15 ml of dilute sulphuric acid and a drop of iodide- (95 per cent), cool, dilute to 60 ml with distilled water and
free starch solution; no blue colour is produced within filter. The filtrate (solution A) is colourless (2.4.1).
2 minutes.
Chlorides (2.3.12). 40 ml ofsolution A complies with the limit
Sulphates (2.3.17). 1.0 g dissolved in 15 ml of water complies test for chlorides (250 ppm).
with the limit test for sulphates (150 ppm).
Sulphates (2.3.17). 10 ml ofsolution A complies with the limit
Thiosulphate. Dissolve 1 gin 10 ml of water, add 0.1 mlof test for sulphates (600 ppm).
starch solution and 0.1 ml of 0.005 M iodine; a blue colour is Water-insoluble matter. Dissolve 0.5 gin 50 ml of water, heat
produced. to boiling, filter through a tared, sintered-glass filter (porosity
Loss on drying (2.4.19). Not more than 1.0 per cent, determined No.4) and wash with water until the filtrate is colourless. The
on 1.0 g of the powdered substance by drying in an oven at weight of the residue, dried at 105° to constant weight, is not
105° for 3 hours. more than 5 mg (1.0 per cent).
Assay. Weigh accurately about 0.35 g, dissolve in about 10 ml Assay. Weigh accurately about 0.3 g, dissolve in sufficient
of water, add 35 ml of hydrochloric acid and 5 ml of chloroform. water to produce lOO.Omi. To 20.0 ml add 20 m1 of water, 1 gof
Titrate with 0.05 M potassium iodate until the purple colour potassium iodide and 10 ml of 2 M hydrochloric acid and
ofiodine disappears from the chloroform. Add the last portion titrate the liberated iodine with 0.1 M sodium thiosulphate
of the iodate solution dropwise and agitate vigorously and using starch solution, added towards the end of the titration,
continuously. Allow to stand for 5 minutes. If any colour as indicator.
develops in the chloroform layer continue the titration until 1 ml of 0.1 M sodium thiosulphate is equivalent to 0.003160 g
the chloroform is decolorised. ofKMn04'
1 ml of 0.05 M potassium iodate is equivalent to 0.0166 g of Storage. Store protected from moisture.
KI.
CA UTION - Great care should be taken in handling
Storage. Store protected from light and moisture. potassium permanganate as dangerous explosions are liable
to occur if it is brought into contact with organic or other
readily oxidisable substances, either in solution or in the
dry .condition.

Potassium Permanganate
KMn0 4 Mol. Wt. 158.0
Potassium Sorbate
Potassium Permanganate contains not less than 99.0 per cent
and not more than 100.5 per cent ofKMn04. CH 3 CH=CHCH=CHCOOK
Category. Antiseptic. Mol. Wt.150.2
Description. A dark purple or brownish black, granular powder Potassium Sorbate is potassium 2,4-hexadienoate.
or dark purple or almost black slender, prismatic crystals, having
a, metallic Illstre;odQ.urless. It decomposes on contact with Potassium Sorbate contains not less than 99.0 per cent and
celtain organic substances. not more than 101.0 per cent of C6H7KO z, calculated on the
dried basis.
Identification Category. Food preservativ.

A. A solution in water acidified with sulphuric acid and heated Description. A white or almost white powder or granules.
to 70° is decolorised by hydrogen peroxide solution (20 vol).
Identification
B. Heated to redness, it decrepitates, evolves oxygen and
Test A may be omitted if tests Band C are carried out. Test B
leaves a black residue which with water forms potassium
may be omitted if tests A and C are carried out.
hydroxide solution; the resulting solution when neutralised
with dilute hydrochloric acid gives the reactions ofpotassiurn A. Determine by infrared absorption spectrophotometry (2.4.6).
salts (2.3.1). Compare the spectrum with that obtained with potassium

1940

www.webofpharma.com
IP 2010 POVIDONE

sorbate RS or with the reference spectrum of potassium using 0.1 ml of crystal violet solution as indicator until the
sorbate. colour changes from violet to bluish-green. Carry out a blank
titration.
B. When examined in the range 230 nm to 350 nm (2.4.7), a 0.02
per cent w/v solution in water, dilute 1.0 ml ofthis solution to I ml of 0.1 M perchloric acid is equivalent to 0.01502 g of
100.0 ml with 0.1 M hydrochloric acid shows an absorption C6H7KO z.
maximum at about 264 nm and the specific absorbance at 264
Storage. Store protected from light.
nm is 1650 to 1900.
C. Dissolve 0.2 g in 2 ml of water and add 2 ml of dilute acetic
acid, filter. The solution gives reaction B ofpotassium (2.3.1).
Povidone
Tests
Polyvinylpyrrolidone; Polyvidone
Appearance of solution. A 5 per cent w/v solution in carbon
dioxide-free water (solution A) is clear (2.4.1) and not more
intensely coloured than reference solution YS5 (2.4.1).
Acidity or alkalinity. To 20 ml of solution A, add 0.1 ml of
phenolphthalein solution. Titrate with 0.1 M sodium
hydroxide or 0.1 M hydrochloric acid. Not more than 0.25 ml
is required to change th~ colour of the indicator.
n
Aldehydes. Not more than 0.15 per cent, determined by
dissolving 1.0 g in a mixture of 50 ml of 2-propanol and 30 ml
Mol. Wt. (111.2)11
of water, adjust to pH 4.0 with 1 M hydrochloric acid. and
dilute to 100 ml with water. To 10 ml ofthe solution add 1 ml of Povidone is poly(2-oxopyrrolidin-I-ylethylene) and consists
decolorisedfuchsin solution and allow to stand for 30 minutes. oflinear polymers of 1-vinylpyrrolidin-2-one. The different
Any colour produced is not more intense than that obtained types of Povidone are characterised by their viscosity in
in a solution prepared simultaneously by adding by 1 ml of solution, expressed as K-value, in the range 10 to 95.
decolorised filChsin solution to a mixture of 1.5 ml of Povidone with a nominal K-value of 15 or less has a K-value
acetaldehyde standard solution (l00 ppm C2H 4 0j, 4 ml of 2- ofnot less than 85.0 per cent and not more than 115.0 per cent
propanol and 4.5 ml of water. ofthe declared value. The K-value ofpovidone with a nominal
Heavy metals (2.3.13). Weigh in a silica crucible 2 g of the K-value ofmore than 15, or a nominal K-value range with an
substance under examination, mix with 0.5 g of magnesium average ofmore than 15, is not less than 90.0 percent and not
oxide. Ignite to dull redness until a homogeneous white or more than 107.0 per cent ofthe declared value or ofthe average
greyish-white mass is obtained. After 30 minutes of ignition if of the declared range. It contains not less than 11.5 per cent
mixture remains coloured, allow to cool, mix using a fine glass and not more than 12.8 per cent of nitrogen, N, calculated on
rod and repeat the ignition. If necessary repeat the operation. the anhydrous basis.
Heat at 8000 for about 1 hour. Dissolve the residue in 5 ml Category. Pharmaceutical aid (tablet binder, coating agent,
ofa mixture ofequal volumes of hydrochloric acid and water. dispersing and suspending agent).
Add 0.1 ml of phenolphthalein solution and then
concentrated ammonia until a pink colour is obtained. Cool, Description. A white or yellowish white powder or flakes;
add glacial acetic acid until the solution is decolourised and odourless or almost odourless; hygroscopic.
add 0.5 ml in excess. Filter if necessary and wash the filter.
Dilute to 20 ml with water, 12 ml of the resulting solution Identification
complies with the limit test for heavy metals, Method D (10 Test A may be omitted iftests B, C andD are carried out. Tests
ppm). Use lead standard solution (10 ppm pb), diluting 1 ml Band C may be omitted if tests A and D are carried out.
standard solution to 10 ml with a mixture ofequal volumes of
hydrochloric acid and water. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with povidone R8.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
B. Add 2.5 g in small portions to a suitable volume of carbon
on 1.0 g by drying in an oven at 105° for 3 hours.
dioxide-fi-ee water, stirring with a magnetic stirrer, and dilute
Assay. Weigh accurately about 0.12 g, dissolve in 20 ml of to 25 ml with the same solvent (solution A). To 0.4 ml ofsolution
anhydrous acetic acid. Titrate with 0.1 M perchloric acid A add 10 ml of water, 5 ml of2 M hydrochloric acid and 2 ml

1941

www.webofpharma.com
POVIDONE IP 2010

of potassium dichromate solution; an orange-yellow Reference solution (b). Dissolve 10 mg of I-vinylpyrrolidin-


precipitate is produced. 2-one and 0.5 g of vinyl acetate in 100.0 ml of methanol.
Dilute 1.0 rnI ofthis solution to 100.0 ml with the mobile phase.
C. To 1 ml of solution A add 0.2 ml of dimethylamino-
benzaldehyde reagent and 0.1 ml of sulphuric acid; a pink Chromatographic system
colour is produced. a stainless steel column 25 cm x 4.0 mm packed with
D. To 0.1 ml of solution A add 5 ml of water and 0.2 ml of octadecylsilane bonded to porous silica (5 Ilm),
0.05 M iodine; a red colour is produced. column temperature. 40°,
mobile phase: a mixture of 10 volumes of acetonitrile
Tests and 90 volumes of water,
Appearance of solution. Solution A is clear (2.4.1), and not flow rate. 1 ml per minute,
more intensely coloured than reference solution BS6 or BYS6 - spectrophotometer set at 235 nm,
(2.4.1). - injection volume. 50 Ill.

Heavy metals. Mix 2.0 g with 0.5 g of magnesium oxide in a Inject reference solution (a) and (b). The test is not valid unless
silica crucible. Ignite to dull red heat until a homogeneous the resolution between the peaks due to I-vinylpyrrolidin-2-
white or greyish white mass is produced. Heat at 800° for one and vinyl acetate is not less than 2.0 in the chromatogram
about 1 hour, dissolve the residue using two quantities, each obtained with reference solution (b) and the relative standard
of 5 ml, of 5 M hydrochloric acid, add 0.1 ml of deviation for replicate injections is not more than 2.0 in the
phenolphthalein solution and strong ammonia solution until chromatogram obtained with reference solution (a).
a pink colour is produced. Cool, add glacial acetic acid until Inject the test solution and reference solution (a). In the
the solution is decolorised and add a further 0.5 ml. Filter if chromatogramobtained with the test solution the area of the
necessary and dilute the solution to 20 ml with water. To 12 ml peak due to I-vinylpyrrolidin-2-one is not more than the area
ofthe resulting solution add 2 ml of acetate bufferpH 3.5, mix, of the principal peak in the chromatogram obtained with
add 1.2 ml of thioacetamide reagent, mix immediately and reference solution (a) (10 ppm).
allow to stand for 2 minutes. Any brown colour produced is
not more intense than that obtained by treating in the same Sulphated ash (2.3.18). Not more than 0.1 per cent.
manner a mixture of 2 ml ofthe test solution obtained above Water (2.3.43). Not more than 5.0 percent determined on 0.5 g.
and 10 ml of the 20 ml of solution obtained by repeating the
procedure using 2 ml of lead standard solution (10 ppm Pb) K-value. For Povidone with a stated K-value of 18 or less,
in place of the substance under examination, adding 0.5 g of prepare a 5.0 per cent w/v solution. For Povidone with a
magnesium oxide in a silica crucible and continuing as declared K-value of more than 18, prepare a 1.0 per cent w/v
described above beginning at the words "Ignite to dull red solution. Allow the solution to stand for 1 hour and carry out
heaL." (10 ppm). Method B for the determination ofviscosity (2.4.28), at 25° ±
0.2° using a size no. 1 viscometer with a minimum flow time of
Aldehydes. Boil 20.0 g in 180 rnI ofa 25 per cent v/v solution of
100 seconds. Calculate the K-value (1<.0) from the expression
sulphuric acid in a round-glass-stoppered flask under a reflux
condenser for 45 minutes and allow to cool. Fit a distillation
head, distil and collect 60 rnI ofthe distillate in 20 rnI ofa 7.0 per _ 1.5logz-1 ~300C logz + (c + 1.5c 10gz)2
K 0- +
cent w/v solution of hydroxylamine hydrochloride, previously 0.15+0.003c 0.15c + 0.003c 2
adjusted to pH 3.1 using 1 M sodium hydroxide, and cooled
in ice. Titrate with 0.1 M sodium hydroxide to pH 3.1. Carry
out a blank titration. Not more than 9.1 ml of 0.1 M sodium where c is the percentage concentration w/v of the substance
hydroxide is required (0.2 per cent, calculated as acetaldehyde, urider examiIlatioIl, calculated Oil the arihymous basts, aridi
CzILO). is the viscosity of the solution relative to that of water.

l-vinylpyrrolidin-2-one. Determine by liquid chromatography Nitrogen (2.3.30). Follow Method C, using 0.3 g, accurately
(2.4.14). weighed and 11 ml of nitrogen-free sulphuric acid. For
complete destruction of organic matter repeat the addition of
Test solution. Dissolve about 0.25 g of the substance under hydrogen peroxide (10 vol) (usually 3 to 6 times) until a clear,
examination in 10.0 ml ofthe mobile phase. light-green solution is obtained, then heat for a further 4 hours.
Reference solution (a). A 0.05 per cent w/v solution of 1-
Storage. Store protected from moisture.
vinylpyrrolidin-2-one in methanol. Dilute 1.0 ml of this
solution to 100.0 ml with methanol. Further dilute 5.0 ml of Labelling. The label states the viscosity in terms ofa K-value
this solution to 100.0 ml with the mobile phase. orK-range.

1942

www.webofpharma.com
IP 2010 POVIDONE-IODINE SOLUTION

Povidone-Iodine Loss on drying (2.4.19). Not more than 8.0 per cent, determined
on 0.5 g by drying in an oven at 105°.
Assay. Weigh accurately about 3 g, transfer to a beaker and
add 200 m1 of water. Cover the beaker and stir with a mechanical
CH-CH z stirrer at room temperature for not more than 1hour to dissolve
I ,x I as completely as possible. Titrate immediately thereafter with
(yo 0.1 M sodium thiosulphate using 3 mlof starch solution,
added towards the end of the titration, as indicator.
n 1 ml of 0.1 M sodium thiosulphate is equivalent to 0.01269 g
ofl.
Povidone-Iodine is a complex produced by interaction
Storage. Store protected from light.
between iodine and poly(2-oxopyrrolidin-l-ylethylene).
Povidone-Iodine contains not less than 9.0 per cent and not
more than 12.0 per cent ofavailable iodine, I, calculated on the
dried basis. Povidone-Iodine Solution
Category. Topical anti-infective; antiseptic. Povidone-Iodine Solution is a solution of Povidone-Iodine in
Purified Water. It may contain a small amount ofEthanoL
Description. A yellowish brown amorphous powder; odour,
slight and characteristic of iodine. Povidone-Iodine Solution contains not less than 85.0 per cent
and not more than 120.0 per cent of the stated amount of
Identification iodine, 1.
Usual strengths. 5 per cent w/v; 7.5 percent w/v; 10 per cent
A Add 0.05 ml ofa 10 per cent w/v solution to a mixture ofl ml
w/v.
of starch solution and 9 ml of water; a deep blue colour is
produced. Description. A deep brown liquid; odour, characteristic of
iodine.
B. Spread 1 ml of a 10 per cent w/v solution over an area of
about 20 cm x 20 cm on a glass plate and allow it to dry in air at Identification
room temperature and in an atmosphere of low humidity
overnight; a brown, dry, non-smearing film is formed which A Dilute 1 m1 to 20 rnl with water and add 1 ml ofthe resulting
dissolves readily in water. solution to a mixture of 1 ml of starch solution and 9 ml of
water; a deep blue colour is produced.
Tests B. Transfer 10 ml to a small flask and cover the mouth ofthe
Heavy metals (2.3.13). 1.0 g complies with the limit test for flask with a filter paper soaked in starch solution; no blue
heavy metals, Method B (20 ppm). colour is produced on the paper within 60 seconds.

Nitrogen (2.3.30).9.5 to 11.5 per cent, calculated on the dried C. Dilute 20 ml to 100 ml with water. To 10 ml add dropwise
basis, determined on about 0.3 g, by Method A 0.1 M sodium thiosulphate until the colour is just discharged.
To 5 ml ofthe resulting solution add 5 ml of 2 M hydrochloric
Iodide. Not more than 6.6 per cent, calculated on the dried acid and 2 ml of potassium dichromate solution; a red
basis, determined by the following method. Weigh accurately precipitate is produced.
about 0.5 g and dissolve in 100 ml of water. Add sufficient
sodium bisulphite solution until the colour of iodine is Tests
discharged. Add 25.0 ml of 0.1 M silver nitrate and 10 ml of
pH (2.4.24).3.0 to 6.5.
nitric acid and mix. Titrate with 0.1 M ammonium thiocyanate,
usingferric ammonium sulphate solution as indicator. Repeat Ethanol (ifpresent) (2.3.45). 90.0 to 11 0.0 per cent ofthe stated
the procedure without the substance under examination. The amount ofCzHsOH, determined by MethodA after decolorising
difference between the titrations represents the amount of the solution with just sufficient quantity of a 10 per cent w/v
silvernitrate required. 1 ml of 0.1 M silver nitrate is equivalent solution of sodium thiosulphate and treating with a few drops
to 0.01269 g ofl. From the percentage oftotal iodine, calculated of dilute sodium hydroxide solution.
on the dried basis, subtract the percentage of available iodine
Assay. To an accurately measured volume containing about
determined in the Assay and obtain the percentage of iodide.
50 mg of iodine add sufficient water to produce not less than
Sulphated ash (2.3.18). Not more than 0.2 per cent. 30 ml and titrate immediately with 0.02 Msodium thiosulphate,

1943

www.webofpharma.com
PRALIDOXIME CHLORIDE IP 2010

using 3 ml of starch solution, added towards the end of the Chloride content. 20.2 to 20.8 percent, calculated on the dried
titration, as indicator. Carry out a blank titration. basis, determined by the following method. Weigh accurately
about 0.3 g, dissolve in about 150 ml of water, add 20 ml of
1 ml of 0.02 M sodium thiosulphate is equivalent to 0.002538
glacial acetic acid and 10 drops of (4-tert-octylphenoxy)
gofI.
nonaethoxyethanol and titrate with 0.1 M silver nitrate,
Storage. Store protected from light. determining the end-point potentiometrically (2.4.25).
Labelling. The label states the quantities ofiodine and ethanol 1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g ofCl.
(if present) as percentages w/v.
Sulphated ash (2.3.18). Not more than 0.5 per cent.
Loss on drying (2.4.19). Not more than 2.0 per cent, determined
on 1.0 g by drying in an oven at 105° for 3 hours.
Pralidoxime Chloride
Assay. Weigh accurately about 0.5 g, dissolve in sufficient
water to produce 250.0 ml and mix. Dilute 5.0 ml ofthis solution
to 100.0 ml with water. Transfer 5.0 ml to a 50-ml volumetric
flask, dilute to about 40 ml with water, add 5.0 ml of 1 M
sodium hydroxide and dilute to volume with water. Within 10
minutes ofthe addition of the alkali, measure the absorbance
C7~CINZO Mol. Wt. 172.6 ofthe solution at the maximum at about 332 nm (2.4.7), using a
solution of5.0 ml of 1 M sodium hydroxide diluted with water
Pralidoxime Chloride is 2-hydroxyiminomethyl-l- to 50.0 ml as the blank.
methylpyridinium chloride.
Calculate the content of C 7H gC1NzO from the absorbance
Pralidoxime Chloride contains not less than 97.0 per cent and obtained by carrYing out the assay simultaneously using about
not more than 103.0 per cent ofC7H gClNzO, calculated on the 0.5 g, accurately weighed, of pralidoxime chloride RS.
dried basis.
Storage. Store protected from moisture.
Category. Antidote for cholinesterase inhibitors.
Dose. By intravenous injection, 1 to 2 g as a 5 per cent w/v
solution, administered over not less than 5 minutes period; by
intramuscular injection, 1 g initially, followed by 1 to 2 further Pralidoxime Chloride Injection
doses ifnecessary, upto a maximum of 12 g in 24 hours.
Pralidoxime Chloride Injection is a sterile material consisting
Description. A white to pale yellow, crystalline powder; ofPralidoxime Chloride with or without buffering agents and
odourless. other excipients. It is filled in a sealed container.

Identification The injection is constituted by dissolving the contents of the


sealed container in the requisite amount Of sterile Water for
A. Determine by infrared absorption spectrophotometry (2.4.6). Injections, immediately before use.
Compare the spectrum with that obtained with pralidoxime
The constituted solution complies with the requirements for
chloride RS or with the reference spectrum of pralidoxime
Clarity of solution and Particulate matter stated under
chloride.
Parenteral Preparations (Injections).
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Storage. The constituted solution should be used immediately
0.0005 per cent w/v solution in 0.1 Mhydrochloric acid shows
after preparation but, in any case, within the period
absorption maxima at about 242 nm and 292 nm and in
recommended by the manufacturer.
0.1 M sodium hydroxide, a maximum at about 332 nm.
Pralidoxime Chloride Injection contains not less than 90.0 per
C. To 0.1 ml of a 20 per cent w/v solution add 1 ml of a 0.6 per
cent and not more than 110.0 per cent ofthe stated amount of
cent w/v solution offerric chloride; an amber-brown colour
pralidoxime chloride, C7HgClNzO.
is produced.
Usual strength. 1g.
D. Gives the reactions of chlorides (2.3.1).
Description. A white to pale yellow powder.
Tests
The contents of the sealed container comply with the
Heavy metals (2.3.13). 1.0 g complies with the limit test for requirements stated under Parenteral Preparations (powders
heavy metals, Method A (20 ppm). for Injection) and with the following requirements:

1944

www.webofpharma.com
IP 2010 PRAVASTATIN SODIUM

Identification Pravastatin Sodium is sodium (3R,5R)-3,5-dihydroxy-7-


[(IS,2S,6S,8S,8aR)-6-hydroxy-2-methyl-8-[[(2S)-2-
A. Determine by infrared absorption spectrophotometry (2.4.6).
methylbutanoyl]oxy]-1 ,2,6,7,8,8a-hexahydronaphthalen-l-
Compare the spectrum with that obtained with pralidoxime
yl]heptanoate
chloride R8 or with the reference spectrum of pralidoxime
chloride. Pravastatin Sodium contains not less than 97.0 per cent and
not more than 102.0 per cent ofCz3H3sNa07, calculated on the
B. When examined in the range 230 nm to 360 nm (2.4.7), a
anhydrous and ethanol free basis.
0.0005 per cent w/v solution in 0.1 Mhydrochloric acid shows
absorption maxima at about 242 nm and 292 nm and in Category. Antihyperlipidaemic.
0.1 M sodium hydroxide, a maximum at about 332 nm. Description. A white to yellowish white powder or crystalline
C. To 0.1 ml ofa 20 per cent w/v solution add 1 ml ofa 0.6 per powder, hygroscopic.
cent w/v solution offerric chloride; an amber-brown colour
is produced. Identification
D. Gives the reactions ofchlorides (2.3.1). A. Determine by infi:ared absorption spectrophotometry (2.4.6).
Tests Compare the spectrum with that obtained with pravastatin
sodium RS or with the reference spectrum of pravastatin
pH (2.4.24).3.5 to 4.5, determined in a 5.0 per cent w/v solution. sodium.
Heavy metals (2.3.13). 1.0 g complies with the limit test for B. 1 ml of a 5 per cent w/v solution in carbon dioxide-free
heavy metals, Method A (20 ppm). water (Solution A) gives reaction A of sodium (2.3.1).
Bacterial endotoxins (2.2.3). Not more than 0.1 Endotoxin Unit
per mg ofpralidoxime chloride. Tests
Assay. Weigh accurately about 0.5 g, dissolve in sufficient Appearance of solution. Dilute 2.0 ml ofsolutionAto 10 ml
water to produce 250.0 rnl and mix. Dilute 5.0 ml ofthis solution with water. The solution is clear (2.4.1) and not more intensely
to 100.0 ml with water. Transfer 5.0 ml to a 50-ml volumetric coloured than the reference solution BYS6 (2.4.1).
flask, dilute to about 40 ml with water, add 5.0 ml of
1 M sodium hydroxide and dilute to volume with water. Within pH (2.4.24). 7.2 to 9.0, determined in solution A.
10 minutes ofthe addition ofthe alkali, measure the absorbance Specific optical rotation (2.4.22). + 153 0 to+159 0 , detelmined
ofthe solution at the maximum at about 332 nm (2.4.7), using a in a 0.5 per cent w/v solution in water.
solution of5.0 ml of 1 M sodium hydroxide diluted with water
to 50.0 ml as the blank. Related substances. Determine by liquid chromatography
(2.4.14).
Calculate the content of C 7H 9CINzO from the absorbance
obtained by carrying out the assay simultaneously using about Solvent mixture. 9 volumes of methanol and 11 volumes of
0.5 g, accurately weighed, of pI'alidoxime chloride RS. water.
Labelling. The label states the period within which the Test solution (a). Dissolve 100 mg of the substance under
constituted injection should be used. examination in 100 ml ofthe solvent mixture.
Test solution (b). Dilute 10 ml of test solution (a) to 100 ml
with the solvent mixture.
Pravastatin Sodium
Reference solution (a). Dissolve the contents of a vial of
pravastatin impurity A RS in 1 ml of test solution (b).
Reference solution (b). Dilute 2 ml oftest solution (a) to 100
ml with the solvent mixture. Dilute 1 ml ofthis solution to 10 ml
o
CYO
with the solvent mixture.
H3 Reference solution (c). Dissolve 12.4 mg of pravastatin
CH I H 1,1,3,3- tetramethylbutylamine RS in 100 ml of thesolvent
3
mixture.
Chromatographic system
a stainless steel column 15 cm x 4.6 rom, packed with
Mol. Wt. 446.5 octadecylsilane bonded to porous silica (5 J.l.m),

1945

www.webofpharma.com
PRAVASTATIN SODIUM IP 2010

- mobile phase: a mixture of 1 volume of glacial acetic Heavy metals (2.3.13). Dissolve 2.0 g in a mixture orI5 volumes
acid, 1 volume of triethylamine, 450 volumes of of water and 85 volumes of methanol and dilute to 20 ml with
methanol and 550 volumes of watel; the same solvent mixture and use 12ml of the solution. The
- flow rate. 1.3 ml per minute, resulting solution complies with the limit test for heavy metals,
- spectrophotometer set at 238 nm, Method B (20 ppm). Prepare the reference solution using lead
- injection volume. 10 )..Ll. standard solution (2 ppm Pb) obtained by diluting lead
standardsolution (l 00 ppm Pb) with a mixture of 15 volumes
Inject reference solution (a). The test is not valid unless the of water and 85 volumes of methanol. ~
resolution between the peaks due to pravastatin impurity A
and pravastatin is not less than 7.0. Ethanol (2.3.45). Not more than 3.0 per cent v/v (determined
by method I).
The relative retention times with reference to pravastatin for
(3R,5R)-3 ,5-dihydroxy-7-[(1 S,2S,6S,8S,8aR)-6-hydroxy-8-
Water (2.3.43). Not more than 4.0 per cent, determined on
[[(2S, 3R)-3 - hydroxy-2-methylbutanoy1] oxy]-2-methy1- 0.5 g.
1,2,6,7,8,8a-hexahydronaphthalen-1-y1] heptanoic acid (3"- Assay. Determine by liquid chromatography (2.4.14), as
hydroxypravastatin) (pravastatin impurity B), (3R,5R)-3,5- described in the test for Related substanceswith the following
dihydroxy-7-[(1 S,2S,6S,8S,8aR)-6-hydroxy-8-[[(2S,3S)-3- modification.
hydroxy-2-methylbutanoyl]oxy]-2-methyl-1 ,2,6,7 ,8,8a-
Inject the test solution (b) and reference solution (c).
hexahydronapthalen-1-yl] heptanoic acid (3" -(S)-
hydroxypravastatin) (pravastatin impurity E), (3R,5R)-3,5- Calculate the content ofCZ3H3sNa07 from the declared content
dihydroxy-7-[(lS,2S,6R,8S,8aR)-6-hydroxy-2-methyl-8-[[(2S)- of pravastatin in pravastatin 1,1,3,3-tetramethylbutylamine
2-methylbutanoyl]oxy]-1,2,6,7,8,8a-hexahydronaphthalen-1- RS.
yl]heptanoic acid (6'-epipravastatin) (pravastatin impurity 1 mg ofpravastatin is equivalent to 1.052 mg ofpravastatin
A), (lS,3S,7S,8S,8aR)-3-hydroxy-8-[2-[(2R,4R)-4-hydroxy-6- sodium.
oxotetrahydro-2H-pyran-2-yl]ethyl]-7-methyl-1 ,2,3,7,8,8a-
hexahydronaphthalen-l-yl (2S)-2-methylbutanoate Storage. Store protected from light and moisture.
(pravastatin lactone) (pravastatin impurity D ) and (3R,5R) -
3,5-dihydroxy-7-[(lS,2S,6S,8S,8aR)-6-hydroxy-2-methyl-8-
[[(2S)-2-methylpentanoyl]oxy]-1 ,2,6,7 ,8,8a- Pravastatin Tablets
hexahydronaphthalen-l-yl] heptanoic acid) (pravastatin
impurity C) and (3R, 5R)-7-[(1S,2S,6S,8S,8aR)-6,8-dihydroxy- Pravastatin Sodium Tablets
2-methyl-l ,2,6,7,8,8a-hexahydronaphthalen-l-yl]-3,5- Pravastatin Tablets contain not less than 92.5 per cent and
dihydroxyheptanoic acid (pravastatin impurity F) are about not more than 107.5 per cent of the stated amount of
0.2,0.3,0.6, 1.9 and 2.1 respectively. pravastatin sodium, C23H3sNa07.
Inject test solution (a) and reference solution (b). Run the Usual strengths. 10 mg; 20 mg; 40 mg.
chromatogram 2.5 times the retention time of the· principal
peak of pravastatin. In the chromatogram obtained with test Identification
solution (a), the area of peak due to pravastatin impurity A is A. Shake a quantity ofthe powdered tablets containing about
not more than 1.5 times the area ofthe principal peak obtained 10 mg ofPravastatin Sodium with 80 rn1 ofwater for 10 minutes,
with reference solution (b) (0.3 per cent), the area ofpeaks due dilute to 100 ml with water, filter. Dilute 1 ml to 10 ml with
to pravastatin impurities B, C, D and E is not more than the water. When examined in the range 220 to 350 nm (2.4.7),
area ofthe principal peak obtained with reference solution (b) exhibits a maximum only at 238 um.
(0.2 per cent), the area ofpeak due to pravastatin impurity F is
not more than 0.75 times the area ofthe principal peak obtained B. In the Assay, the principal peak in the chromatogram
with reference solution (b) (0.15 per cent) the area of other obtained with the test solution corresponds to the peak in the
secondary peak is not more than 0.5 times the area of the chromatogram obtained with the reference solution.
principal peak obtained with reference solution (b) (0.1 per Tests
cent) and sum of the areas of all the secondary peaks is not
more than 3 times the area ofthe principal peale obtained with Related substances. Determine by liquid chromatography
reference solution (b) (0.6 per cent). Ignore any peak with an (2.4.14).
area less than 0.25 times the area of the principal peak in the Solution A. Dissolve 4.1 g of anhydrous sodium acetate in
chromatogram obtained with reference solution (b) (0.05 per 400 ml of water, adjust the pH to 5.6 with glacial acetic acid
cent). and add sufficient water to produce 500 ml.

1946

www.webofpharma.com
IP 2010 PRAZIQUANTEL

Solution B. Mix 20 volumes of methanol with 80 volumes of Test solution. Add 20 ml of solution A to a quantity of the
solution A. powdered tablets containing about 10 mg of Pravastatin
Sodium, mix using a vortex mixer and then with the aid of
Test solution. Add 20 ml of solution A to a quantity of the
ultrasound for 15 minutes, centrifuge. Dilute 1 ml ofthe clear
powdered tablets containing about 10 mg of Pravastatin
supernatant liquid to 5 ml with solution B.
Sodium, mix using a vortex mixer and then with the aid of
ultrasound for 15 minutes, centrifuge. Dilute 1 ml ofthe clear Reference solution (a). Dissolve 12.4 mg of pravastatin
supernatant liquid to 5 ml with solution B. 1,1,3,3,-tetramethylbutylamine RS in 20 ml of solution A.
Dilute 1 ml ofthis solution to 5 ml with solution B.
Reference solution (a). Dilute 1 ml of the test solutionto 100
ml with solution B. Reference solution (b). Dissolve 2 mg of pravastatin impurity
A RS in the minimum volume of methanol and dilute to 20 ml
Reference solution (b). Dilute 1 ml ofreference solution (a) to
with the test solution.
5 ml with solution B.
Use the chromatographic system, solution A and B as
Reference solution (c). Dilute 1 ml ofreference solution (b) to
described under Related substances.
4 ml with solution B.
Inject reference solution (b). The test is not valid unless the
Reference solution (d). Dissolve 2 mg of (3R,5R)-3,5-
resolution between the peaks due to pravastatin impurity A
dihydroxy-7-[(lS,2S,6R,8S,8aR)-6-hydroxy-2-methyl-8-
and pravastatin is not less than 7.0.
[[(2S) -2-methyl bu tanoyIJoxyJ-1, 2,6,7,8, 8a-
hexahydronaphthalen-1-yUheptanoic acid RS (pravastatin Inject the test solution and reference solution (a).
impurity A RS) in the minimum volume of methanol and dilute
Calculate the content of C23H3SNa07 in the tablets using the
to 20 ml with the test solution.
declared conterit of pravastatin in pravastatin 1,1,3,3-
Chromatographic system tetramethylbutylamine RS.
- a stainless steel column 15 cm x 4.6 rom packed with
1 mg ofC23H3607 is equivalent to 1.052 mg ofC23H3sNa07'
octadecylsilane bonded to porous silica (5 f..lm) (such as
Ultrasphere ODS), . Storage. Store protected from light and moisture.
- mobile phase: a mixture of 1 volume of glacial acetic
acid, 1 volume of triethylamine, 450 volumes of
methanol and 550 volumes ofwater,
- flow rate. 1.3 ml per minute,
Praziquantel
- spectrophotometer set at 238 urn,
injection volume. 20 f..ll.
Inject reference solution (d). The test is not valid unless the
resolution between the peaks due to pravastatin impurity A
and pravastatin is not less than 7.0.
Inject the test solution, reference solution (a), (b) and (c). In
the chromatogram obtained with the test solution the area of
any secondary peak is not more than twice the area of the
principal peak in the chromatogram obtained with reference Mol.Wt. 312.4
solution (a) (2 per cent), the area of one such peak is not more Praziquantel is (11bRS)-2-(cydohexylcarbonyl)-l ,2,3,6,7,11 b-
than the area of the principal peak in the chromatogram hexahydro-4H-pyrazino[2, I-a ]isoquinolin-4-one.
obtained with reference solution (a) (1 per cent), not more
Praziquantel contains not less than97.5 per cent and not more
than a further one such peak has an area more than the area of
than 102.0 per cent of C19H24N202, calculated on the dried
the principal peak in the chromatogram obtained with reference
basis.
solution (b) (0.2 per cent) and the sum of the areas of all the
secondary peaks is not more than three times the area of the Category. Antihelminthic.
principal peak in the chromatogram obtained with reference Description. A white or almost white, crystalline powder.
solution (a) (3 per cent). Ignore any peak with an area less
than the area of the principal peak in the chromatogram Identification
obtained with reference solution (c) (0.05 per cent).
Determine by infrared absorption spectrophotometry (2.4.6).
Other tests. Comply with the tests stated under Tablets. Compare the spectrum with that obtained with praziquantel
Assay. Detennine by liquid chromatography (2.4.14). RS or with the reference spectrum ofpraziquantel.

1947

www.webofpharma.com
PRAZIQUANTEL IP 20ID

Tests Assay. Determine by liquid chromatography (2.4.14) as


described under Related substances.
Related substances. Determine by liquid chromatography
(2.4.14). Inject reference solution (b). The test is not valid unless the
relative standard deviation for replicate injections is not more
Test solution (a). Dissolve 40 mg of the substance under
than ·1.0 per cent.
examination in 10 ml ofthe mobile phase.
Inject the test solution (b) and reference solution (a).
Test solution (b). Dilute 1.0 ml oftest solution (a) to 20 ml with
the mobile phase. Calculate the content of CI9Hz4NzOz.
Reference solution (a). Dissolve 40 mg bfpraziquantel RS in Storage. Store protected from light.
10 ml ofthe mobile phase. Dilute 1.0 ml to 20 ml with the mobile
phase.
Reference solution (b). Dissolve 5.0 mg of (11 bRS)-2-benzoyl-
1,2,3,6,7,11b-hexahydro-4H-pyrazino[2, 1-a]isoquinolin-4- Praziquantel Tablets
one RS (praziquantel impurity A RS) in 25 ml of reference Praziquantel Tablets contain not less than 90.0 per cent and
solution (a). Dilute 2.0 ml of this solution to 20 ml with the not more than 110.0 per cent of the stated amount of
mobile phase. Praziquantel, CI9Hz4NzOz.
Reference solution (c). Dilute 1.0 ml oftest solution (a) to 20 Usual strength. 600 mg.
ml with the mobile phase. Dilute 5.0 ml ofthis solution to 50 ml
with the mobile phase. Identificaton
Chromatographic system
A. Determine by infrared absorption spectrophotometry (2.4.6).
- a stainless steel column 25 cm x 4.0 mm, packed with
Compare the spectrum with that obtained with praziquantel
octadecylsilane bonded to porous silica (5 Ilm),
RS or with the reference spectrum of praziquantel.
- mobile phase: a mixture of 45 volumes of acetonitrile
and 55 volumes of water, B. Determine by thin-layer chromatography (2.4.17), coating
- flow rate. 1 ml per minute, the plate with silica gelG.
- spectrophotometer set at 210 run, Mobile phase. Ethyl acetate.
- injection volume. 20 Ill.
Test solution. Shake a quantity of the powdered tablets
Inject reference solutions (b). The test is not valid unless the containing 30 mg of praziquantel in 5 ml of methanol for 5
resolution between the peaks corresponding to praziquantel minutes, centrifuge and use the clear supernatant liquid.
impurity A and praziquantel is not less than 3.0.
Reference solution. A 0.6 per cent w/v solution ofpraziquantel
Inject test solution (a), reference solution (b) and (c). Run the RS in methanol.
chromatogram 5 times the retention time ofthe principal peale.
In the chromatogram obtained with test solution (a), the area Apply to the plate 10 III of each solution. After development,
ofany secondary peak is not more than the area ofthe principal dry the plate in air and examine under ultraviolet light at 254
peak in the chromatogram obtained with reference solution urn. The principal spot in the chromatogram obtained with the
(c) (0.5 per cent). The area of one such peak is not more than test solution corresponds to that in the chromatogram obtained
0.4 times the area of the principal peak in the chromatogram with the reference solution.
obtained with reference solution (c) (0.2 per cent). The sum of
the areas of all other secondary peaks is not more than the
Tests
area of the principal peak in the chromatogram obtained with Dissolution (2.5.2).
reference solution (b) (0.5 per cent). Ignore any peak with an
area 0.1 times the area ofthe principal peale in the chromatogram Apparatus No.1,
obtained with reference solution (b) (0.05 per cent). Medium. 900 ml of 0.1 M hydrochloric acid containing 2.0 mg
per ml of sodium lauryl sulphate,
Heavy metals (2.3.13). 1.0 g complies with the limit test for Speed and time. 50 rpm and 60 minutes.
heavy metals, Method B (20 ppm).
Withdraw a suitable volume ofthe medium and filter. Measure
Sulphated ash (2.3.18). Not more than 0.1 per cent. the absorbance ofthe filtrate at the maximum at about 263 urn
Loss on drying (2.4.19). Not more than 0.5 per cent, determined (2.4.7). Calculate the content of CI9Hz4NzOz in the medium
on 1.0 g by drying in an oven at 50° over diphosphorus from the absorbance obtained from a solution of known
pentoxide at a pressure not exceeding 0.7 kPa for 2 hours. concentration of praziquantel RS in the same medium.

1948

www.webofpharma.com
IP 2010 PRAZOSlN HYDROCHLORIDE

D. Not less than 75 per cent of the stated amount of Description. A white or almost white powder; odourless or
C19H24N202' almost odourless.
Other tests. Comply with the tests stated under Tablets.
Identification
Assay. Determine by liquid chromatography (2.4.14).
Test A may be omitted iftests B, C andD are carried out. Tests
Test solution. Weigh and powder 20 tablets. Weigh accurately Band C may be omitted if tests A and D are carried out.
a quantity of the powder containing 150 mg ofPraziquantel,
add 70 ml ofmobile phase, sonicate for 5 minutes, dilute with A. Determine by infrared absorption spectrophotometry (2.4.6).
mobile phase to 100 ml, mix and filter. Dilute 3.0 ml of the Compare the spectrum with that obtained with prazosin
solution to 25 ml with the mobile phase and mix. hydrochloride RS.

Reference solution. A 0.018 per cent w/v solution of B. Prepare a 0.05 per cent w/v solution ofthe substance under
praziquentel RS in the mobile phase. examination in a 0.1 per cent v/v solution of hydrochloric
acid in methanol. Dilute separately 1 ml and 5 ml ofthis solution
Chromatographic System to 100 ml with the same acid solution (solutions A and B
- a stainless steel colurnn25 cm x 4.0 mID, packed with respectively). When examined in the range 220 nm to 280 nm
octadecylsilane bonded to porous silica (10 flm), (2.4.7), solution A shows an absorption maximum at about
- mobile phase: a mixture of 60 volumes of acetonitrile 247 nm; absorbance at the maximum, 0.66 to 0.70. When
and 40 volumes of water, examined in the range 280 nm to 400 nm, solution B exhibits
- flow rate. 1.5 ml per minute, two maxima at about 330 nm and 343 nm; absorbances at the
- spectrophotometer set at 210 nm, two maxima, 0.65 to 0.70 and 0.60 to 0.66 respectively.
- injection volume. 10 fll.
C. Determine by thin-layer chromatography (2.4.17), coating
Inject the reference solutIon. The test is not valid unless the the plate with silica gel GF254.
tailing factor is not more than 1.5 and the relative standard
deviation for replicate injections is not more than 1.0 per cent. Solvent mixture. A mixture of 1 volume of diethylamine, 10
volumes of methanol and 10 volumes of methylene chloride.
Inject the test solution and the reference solution.
Mobile phase. A mixture of 5 volumes of diethylamine and 95
Calculate the content OfC19H24N202. in the tablets. volumes of ethyl acetate.
Storage. Store protected from light. Test solution. Dissolve 10 mg of the substance under
examination in 10 ml ofsolvent mixture.
Reference solution. A 0.1 percent w/v solution of prazosin
hydrochloride RS in solvent mixture.
Prazosin Hydrochloride
Apply to the plate 10 fll of each solution. Allow the mobile
phase to rise 8 em. Dry the plate in warm air and examine under
ultra-violet light at 254 nm. The principal spot in the chroma
togram obtained with the test solution corresponds to that in
the chromatogram obtained with the reference solution.
D. A 0.1 per cent w/v solution gives the reactions of chlorides
(2.3.1).

Tests

Mol. Wt.4l9.9 Iron.To 1.0 g add dropwise about 1.5 ml of nitric aCid,heat
cautiously on a water-bath until fumes are no longer evolved.
Prazosin Hydrochloride is 2-[4-(2-furoyl)piperazin-l-yl]-6,7- Ignite by slowly raising the temperature from 150° to 1000°,
dimethoxyquinazolin-4-ylaminehydrochloride. maintaining the final temperature for 1 hour. Cool, dissolve
Prazosin Hydrochloride contains not less than 98.5 per cent the residue in 20 ml of 2 M hydrochloric acid, evaporate to
and not more than 101.0percentofC19H21Ns04,HCl,calculated about 5 ml, dilute to 25 ml with 2 M hydrochloric acid and
on the anhydrous basis. examine the resulting solution by atomic absorption
spectrophotometry (2.4.2), measuring at 248 nm using an iron
Category. Antihyperten,sive.
hollow-cathode light as a source ofradiation, an air-acetylene
Dose. 500 flg to 1 mg two to three times daily. flame and iron standard solution (8 ppm Fe), diluted as

1949

www.webofpharma.com
PRAZOSIN HYDROCHLORIDE IP 2010

necessary with water to prepare. the. standard solutions acid to produce 50 rril. The resulting solution complies with
(100 ppm). the limit test for heavy metals, Method B (50 ppm).
Reserve about 10 ml of the solution for the Nickel test. Sulphated ash (2.3.18). Not more than 0.2 per cent.
Nickel. Examine the final solution prepared in the test for Iron Water (2.3.43). Not more than 0.5 per cent w/w, using 2.0 g
by atorriiC absorption spectrophotometry (2.4.2), measuring dissolved in a mixture of equal volumes of dichloromethane
at 232 urn using a nickel hollow-cathode light as a source of and methanol.
radiation, an air-acetylene flame and using nickel standard
Assay. Weigh accurately about 0.35 g, dissolve in 20 ml of
solution (10 ppm Ni},diluted as necessary with water to
anhydrous formic acid and 30 ml of acetic anhydride. Titrate
prepare the standard solutions (50 ppm).
with 0.1 M perchloric acid, determining the end point
Related substances. Determine by liquid chromatography potentiometrically (2.4.25). Carry out a blank titration.
(2.4.14).
1 ml of 0.1 M perchloric acid is equivalent to 0.04199 g of
Test solution. Dissolve 50.0 mg of the substance under C19H21Ns04, HCL
examination in the mobile phase and dilute to 50 ml with the
Storage. Store proteCted from light.
mobile phase.
Reference solution (a). Dilute 1 ml of the test solution to
100 ml with the mobile phase. Further dilute 1 rril ofthe solution
to 10 ml with the mobile phase. Prazosin Tablets
Reference solution (b). Dissolve 8 mg of metoclopramide Prazosin Hydrochloride Tablets
hydrochloride RS in 1 ml of the test solution and dilute to
Prazosin Tablets contain not less than 90.0 per cent and not
25 rril with the mobile phase. Further dilute 1 ml ofthe solution
to 10 ml with the mobile phase. more than 11 0.0 per cent of the stated amount of prazosin,
C19H21Ns04.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with Usual strengths. The equivalent of500 ~g; 1 mg; 2 mg; 5 mg
octadecylsilane bonded to porous silica (5 ~m), ofprazosin.
- mobile phase: a mixture of50 volumes of methanol and
50 volumes of a solution containing 3.484 g per litre of
Identification
sodium pentanesulphonate and 3.p4 g per litre of Shake a quantity of the powdered tablets containing about
tetramethylammonium hydroxide adjusted to pH 5.0 10 mg ofprazosin with a mixture of 10 ml of dichloromethane
with glacial acetic acid, and 10 ml of 0.05 M potassium hydroxide, filter the organic
- flow rate. 1 ml per minute, layer through cotton, evaporate to dryness and drythe residue
- spectrophotometer set at 254 nm, at 60° at a pressure not exceeding 2 kPa at 60° for 2 hours.
- injection volume. 20 ~L
On the residue determine by infrared absorption
InjeCt reference solution (b). The retention times are: prazocine spectrophotometry (2.4.6). Compare the spectrum with that
about 9 minutes and metoclopramide abOut 5 minutes.The obtained with prazosin hydrochloride RS or with the reference
test is not valid unless the resolution between the peaks spectrum of prazosin.
corresponding to prazocine and metoclopramide is at least 8.
Inject the test solution and reference solution (a). Continue
Tests
the chromatography of the test solution for 6 times the Related substances. Determine by thin-layer chromatography
retention time of the principal peak. Any secondary peak (2.4.17), coating the plate with silica gel HF254.
obtained with the test solution is not more than twice the area
of the principal peak in the chromatogram obtained with Mobile phase. A mixture of 95 volumes of ethyl acetate and
reference solution (a) (0.2 per cent).The sum ofall the impurities 5 volumes of diethylamine.
is not more than five times the area ofthe principal peale in the Test solution. Shake a quantity of the powdered tablets
chromatogram obtained with reference solution (a) (0.5 per containing 5 mg ofprazosin with 2 rril ofa mixture ofl 0 volumes
cent). Ignore any peale with an area 0.5 times the area of the of dichloromethane, 10 volumes of methanol and 1 volume of
principal peak in the chromatogram obtained with reference diethylamine, centrifuge a~d pass the supernatant liquid
solution (a) (0.05 per cent). through a 0.5 ~m PTFE (Polytetrafluoroethylene) filter.
Heavy metals (2.3.13). Determine on the residue obtained in Reference solution (a). Dilute 1 volume of test solution to
the test for Sulphated ash. Dissolve in sufficient 2 M nitric 200 volumes with the same solvent mixture

1950

www.webofpharma.com
IF 2010 PREDNISOLONE

Reference solution (b). Dilute 2 volumes ofreference solution Prednisolone


(a) to 5 volumes with the same solvent mixture.

Apply to the plate 20 III of each solution. Any secondary spot


in the chromatogram obtained with the test solution is not OH
more intense than the spot in the chromatogram obtained -OH
with reference solution (a) and not more than two such spots
are more intense than the spot in the chromatogram obtained
with reference solution (b).

Uniformity of content. Comply with the tests stated under


o
Tablets.
Mol. Wt. 360.4
Determine by liquid chromatography (2.4.14). Prednisolone is 1113,17a,21-trihydroxypregna-l ,4-diene-3,20-
Test solution. Shake one tablet for 1 hour in a suitable volume dione.
of a mixture of96 volumes of methanol, 2 volumes of glacial Prednisolone contains not less than 96.0 per cent and not
acetic acid and 2 volumes of water to produce a solution more than 104.0 per cent of C Z1 HZ 80 S, calculated on the dried
containing 0.002 per cent w/v solution ofprazosin, centrifuge basis.
and use the supernatant liquid.
Category. Adrenocortical steroid.
Reference solution. A 0.0022 per cent w/v solution ofprazosin
Dose. Upto 30 mg daily, in divided doses.
hydrochloride RS in the same solvent mixture.
Description. A white or almost white, crystalline powder;
Chromatographic system hygroscopic.
a stainless steel column 20 cm x 4 rom, packed with porous
silica particles, (5 Ilm), Identification
mobile phase: 0.01 per cent w/v solution of diethylamine
Test A may be omitted if tests Band C are carried out. Test C
in a mixture of 96 volumes of methanol, 2 volumes of
may be omitted if tests A and B are carried out.
glacial acetic acid and 2 volumes of water,
flow rate. 1 ml per minute, A. Determine by infrared absorption spectrophotometry (2.4.6).
spectrophotometer set at 254 om, Compare the spectrum with that obtained with prednisolone
injection volume. 20 Ill. RS or with the reference spectrum of prednisolone.

Calculate the content OfC19HzINs04 in the tablet. B. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G.
Other tests. Comply with the tests stated under Tablets.
Solvent mixture. A mixture of 90 volumes of acetone and
Assay. Determine by liquid chromatography (2.4.14). 10 volumes offormamide.

Test solution. Weigh and powder 20 tablets. Shake a quantity Mobile phase. Chloroform.
of the powdered tablets containing about 2. mg of prazosin Test solution. Dissolve 25 mg of the substance under
with 100 ml in a mixture of96 volumes of methanol, 2 volumes examination in 10 ml ofthe solvent mixture.
of glacial acetic acid and 2 volumes of water for 30 minutes,
centrifuge and use the supernatant liquid: Reference solution (a). Dissolve 25 mg ofprednisolone RS in
10 ml ofthe solvent mixture.
Reference solution. A 0.0022 per cent w/v solution ofprazosin
Reference solution (b). Mix equal volumes ofthe test solution
hydrochloride RS in a mixture of 96 volumes of methanol,
and reference solution (a).
2 volumes of glacial acetic acid and 2 volumes of water.
Place the dry plate in a tank containing a shallow layer of the
The chromatographic conditions as described under solvent mixture, allow the solvent mixture to ascend to the
Uniformity of content may be used. top, remove the plate from the tank and allow the solvent to
Calculate the content OfC19HzINs04 in the tablets. evaporate. Use within 2 hours, with the flow of the mobile
phase in the direction in which the aforementioned treatment
Storage. Store protected from light. was done.
Labelling. The label states the strength in terms of the Apply to the plate 2 III of each solution. Allow the mobile
equivalent amount of prazosin. phase to rise 12 cm. Dry the plate in a current ofwarm air, allow

1951

www.webofpharma.com
PREDNISOLONE IP 2010

the solvent to evaporate, heat at 120° for 15 minutes and spray Inject reference solution (a). The retention times are:
the hot plate with ethanolic sulphuric acid (20 per cent v/v). prednisolone, about 14 minutes and hydrocortisone about
Heat at 120° for a further 10 minutes, allow to cool and examine 15.5 minutes. The test is not valid unless the resolution between
in daylight and in ultraviolet light at 365 nrn. The principal the peaks corresponding to prednisolone and hydrocortisone
spot in the chromatogram obtained with the test solution is at least 2.2. If necessary, adjust the concentration of
corresponds to that in the chromatogram obtained with tetrahydrofitran in the mobile phase.
reference solution (a). The principal spot in the chromatogram
Inject separately the solvent mixture ofthe test solution as a
obtained with reference solution (b) appears as a single,
blank, the test solution and reference solution (b). Continue
compact spot. A.
the chromatography of the test solution for 4.5 times the
C. Dissolve 2 mg in 2 ml of sulphuric acid by shaking and retention time of the principal peak. In the chromatogram
allow to stand for 5 minutes; an intense red colour is produced obtained with the test solution, the area of any peak other
with a reddish brown fluorescence when examined in ultraviolet than the principal peak, is not more than the area ofthe principal
light at 365 nrn. Pour the solution into 10 ml of water and mix; peak in the chromatogram obtained with reference solution
the colour fades and there is a yellow fluorescence under (b) (1.0 per cent) and not more than one such peak has an area
ultra-violet light (365 nrn). more than half the area of the principal peak in the
chromatogram obtained with reference solution (b) (0.5 per
Tests cent); the sum of the areas of all the peaks other than the
principal peak is not more than 2.0 times the area ofthe principal
Specific optical rotation (2.4.22). +96.0° to +102°, determined peak in the chromatogram obtained with reference solution
in a 1.0 per cent w/v solution in dioxan. (b) (2 per cent). Ignore any peak obtained with the blank run
Light absorption (2.4.7). Absorbance ofa 0.001 per cent w/v and any peak with an area less than 0.05 times that of the
solution in ethanol (95 per cent) at the maximum at about principal peak in the chromatogram obtained with reference
240 nrn, 0.40 to 0.43. solution (b).
Related substances. Determine by liquid chromatography Sulphated ash (2.3.18). Not more than 0.1 per cent.
(2.4.14). Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Test solution. Dissolve 25 mg of the substance under on 1.0 g by drying in an oven at 105° for 3 hours.
examination in 2 ml of tetrahydrofilran and dilute to 10m1 with
Assay. Weigh accurately about 0.1 g and dissolve in sufficient
water.
ethanol to produce 100.0 ml. Dilute 2.0 ml of this solution to
Reference solution (a). Dissolve 2 mg of prednisolone RS 100.0 ml with ethanol. Measure the absorbance ofthe resulting
and 2 mg of hydrocortisone RS in the mobile phase and dilute solution at the maximum at about 243.5 nrn. Calculate the
to 100 ml with the mobile phase. content of CZIHzsOs taking 415 as the specific absorbance at
Reference solution (b). Dilute 1 ml of the test solution to 243.5nrn.
100 ml with the mobile phase. Storage. Store protected from light and moisture.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
base deactivated end-capped octadecylsilane bonded Prednisolone Tablets
to porous silica (5 I!m),
column temperature. 45°, Prednisolone Tablets contain not less than 90.0 per cent and
- mobile phase: a mixture of 220 ml of tetrahydrofitran not more than 110.0 per cent of the stated amount of
and 700 ml of water, allowed to equilibrate, diluted to prednisolone, CZIHzsOs.
1000 ml with water and mixed again, Usual strengths. 5 mg; 10 mg; 20 mg.
- flow rate. 1 ml per minute,
- spectrophotometer set at 254 nrn, Identification
- injection volume. 20 I!l. Extract a quantity ofthe powdered tablets containing 30 mg of
Equilibrate the column with the mobile phase for about 30 Prednisolone with 10 ml of chloroform, filter and evaporate
minutes. the filtrate to dryness. The residue complies with the following
tests.
Adjust the sensitivity of the system so that the height of the
principal peak in the chromatogram obtained with reference A. Determine by infrared absorption spectrophotometry (2.4.6).
solution (b) is not less than 50 per cent of the full scale of the Compare the spectrum with that obtained with prednisolone
recorder. RS or with the reference spectrum of prednisolone.

1952

www.webofpharma.com
IP 2010 PREDNISOLONE TABLETS

B. Determine by thin-layer chromatography (2.4.17), coating mobile phase: a mixtl.J[e of 220 ml of tetrahydrofilran
the plate with silica gel G. and 700 ml of water, allowed to equilibrate, diluted to
Solvent mixture. A mixture of 90 volumes of acetone and 1000 ml with water and mixed again,
10 volumes offormamide. flow rate. 1 ml per minute,
spectrophotometer set at 254 nm,
Mobile phase. Chloroform. injection volume. 20 !-tl.
Test solution. Dissolve 25 mg of the residue in 10 ml of the Equilibrate the column with the mobile phase for about
solvent mixture. 30 minutes.
Reference solution (a). Dissolve 25 mg ofprednisolone RS in Adjust the sensitivity of the system so that the height of the
10 ml ofthe solvent mixture. principal peak in the chromatogram obtained with reference
Reference solution (b). Mix equal volumes ofthe test solution solution (b) is not less than 50 per cent of the full scale of the
and reference solution (a). recorder.
Place the dry plate in a tank containing a shallow layer of the Inject reference solution (a). The retention times are:
solvent mixture, allow the solvent mixture to ascend to the prednisolone, about 14 minutes and hydrocortisone about
top, remove the plate from the tanle and allow the solvent to 15.5 minutes. The test is not valid unless the resolution between
evaporate. Use within 2 hours, with the flow of the mobile the peaks corresponding to prednisolone and hydrocortisone
phase in the direction in which the aforementioned treatment is not less than 2.2. If necessary, adjust the concentration of
was done. tetrahydrojitran in the mobile phase.
Apply to the plate 2 !-tl of each solution. Allow the mobile Inject separately the solvent mixture of the test solution as a
phase to rise 12 cm. Dry the plate in a current ofwarm air, allow blank, the test solution and reference solution (b). Continue
the solvent to evaporate, heat at 120° for 15 minutes and spray the chromatography of the test solution for 4.5 times the
the hot plate with ethanolic sulphuric acid (20 per' cent v/v). retention time of the principal peale In the chromatogram
Heat at 120° for a further 10 minutes, allow to cool and examine obtained with the test solution, the area of any secondary
in daylight and in ultraviolet light at 365'nm. The principal peak is not more than the area of the principal peak in the
spot in the chromatogram obtained with the test solution chromatogram obtained with reference solution (b) (1.0 per
corresponds to that in the chromatogram obtained with cent) and the sum of the areas of all the peaks other than the
reference solution (a). The principal spot in the chromatogram principal peak is not greater than three times the area of the
obtained with reference solution (b) appears as a single, principal peak in the chromatogram obtained with reference
compact spot. solution (b) (3.0 per cent). Ignore any peak obtained with the
blanle run and any peak with an area less than 0.05 times that
Tests of the principal peak in the chromatogram obtained with
Related substances. Determine by liquid chromatography reference solution (b) (0.05 per cent) and any peak with a
(2.4.14). retention time of3 minutes or less.
Test solution. Shake a quantity of the powdered tablets Uniformity of content. (For tablets containing 10 mg or less)
containing 10 mg ofPrednisolone with 25 ml of methanol for - Comply with the test stated under Tablets.
10 minutes and mix with the aid of ultrasound for 2 minutes; Powder one tablet, add 50 ml of ethanol (95 per cent), shake
filter the extract (Whatman GF/F is suitable), washthe filter for 30 minutes, add sufficient ethanol (95 per cent) to produce
with two 10-ml quantities of methanol, combine the filtrate 100.0 ml. Centrifuge and pipette a suitable volume ofthe
and washings and evaporate to dryness using a rotary supernatant liquid containing 0.5 mg of Prednisolone and
evaporator and a warm water-bath, dissolve the residue in dilute to 50.0 ml with ethanol (95 per cent). Measure the
10 ml of tetrahydrofilran and dilute to 20 ml with water. absorbance ofthe resulting solution at the maximum at about
Reference solution (a). Dissolve 2 mg of prednisolone RS 240 nm (2.4.7). Calculate the content of CZIHzsOs taking
and 2 mg of hydrocortisone RS in the mobile phase and dilute 415 specific absorbance at 240 nm.
to 100 ml with the mobile phase. Dissolution (2.5.2).
Reference solution (b). Dilute 1 ml of the test solution to Apparatus. No.1,
100 ml with a 50 per cent v/v solution of tetrahydrofitran. Medium. 900 ml of water,
Chromatographic system Speed and time. 50 rpm and 30 minutes.
- a stainless steel column 25 cm x 4.6 mm, packed with Withdraw a suitable volume ofthe medium, filter and dilute a
octadecylsilane bonded to porous silica (5 !-tm), suitable volume ofthe filtrate with the same solvent. Measure
- column temperature. 45°, the absorbance ofthe filtrate at the maximum at about240 nm

1953

www.webofpharma.com
PREDNISOLONE TABLETS IP 2010

(2.4.7). Calculate thecontentof~21H2s0sin the medium from Prednisolone Acetate contains not less than 97.0 per cent and
the absorbance obtained from a solution of known not more than 103.0 per cent of C23H3006, calculated on the
concentration of prednisolone RS. dried basis.
D. Not less than 70 per cent ofthe stated amount ofC21H2s0s. Category. Antiinflammtory; immunosuppressant.
Other tests. Comply with the tests stated under Tablets. Description. A white or almost white, crystalline powder.
Assay. Determine by liquid chromatography (2.4.14).
Identification
Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing about 5 mg of Test A may be omitted iftests B, C andD are carried out. Tests
Prednisolone, add 58 ml of methanol, shake for 10 minutes B, C and D may be omitted if test A is carried out.
and add sufficient water to produce 100.0 ml. Mix well and
A. Determine by infrared absorption spectrophotometry (2.4.6).
filter.
Compare the spectrum with that obtained with prednisolone
Reference solution (a). A solution containing 0.005 per cent acetate RS or with the reference spectrum of prednisolone
w/v of prednisolone RS and 0.0075 per cent w/v of acetate.
dexamethasone (internal standard) in a mixture of58 volumes
of methanol and 42 volumes of water. B. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel F254.
Reference solution (b). Prepare in the same manner as the test
solution but adding 10 ml ofa 0.075 per cent w/v solution of Solvent muture. 10 volumes of methanol and 90 volumes of
dexamethasone in methanol and 48 ml of methanol in place of dichloromethane.
58 mlofmethanol. Mobile phase. Add a mixture of 1.2 volumes of water and 8
Chromatographic system volumes of methanol to a mixture of 15 volumes of ether and
- a stainless steel column 20 cm x 4.6 mm, packed with 77 volumes of dichloromethane.
octadecylsilyl silica gel (5 ~m), Test solution. Dissolve 10 mg of the substance under
- mobile phase: a mixture of 42 volumes of water and examination in 10.0 ml ofthe solvent mixture.
58 volumes of methanol,
- flow rate. 1 ml per minute, Reference solution (a). A 0.1 per cent w/v solution of
- spectrophotometer set at 254 nm, prednisolone· acetate RS in the solvent mixture.
- injection volume. 20 ~l. Reference solution (b). Dissolve 10 mg of prednisolone
The assay is not valid unless the resolution factor between pivalate RS in 10.0 ml ofreference solution (a).
the peaks due to prednisolone and dexamethasone is greater Apply to the plate 5 ~l of each solution. Allow the mobile
than 2.5 and the column efficiency, determined using the peak phase to rise 15 cm. Dry the plate in air and examine in ultraviolet
due to prednisolone in the chromatogram obtained with the light at 254 nm. Spray with alcoholic solution ofsulphuric
test solution is greater than 15,000 theoretical plates per metre. acid. Heat at 105° for 10 minutes or until the spots appear.
Calculate the content ofC21H2s0s in the tablets. Allow to cool and examine under ultraviolet light at 365 urn.
Storage. Store protected from light. The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
with reference solution (a). The test is not valid unless the
chromatogram obtained with reference solution (b) shows two
Prednisolone Acetate clearly separated spots.
C. Add about 2 mg ofthesubstance under examination in 2 ml
of sulphuric acid and shake to dissolve. Within 5 minutes, an'
intense red colour develops. When examined under ultraviolet
light at 365 urn, a reddish-brown fluorescence is seen. Add the
solution to 10 ml of water and mix. The colour fades and there
is greenish-yellow fluorescence in ultraviolet light at 365 urn.
D. It gives the reaction ofacetyl (2.3.1).

Tests
~3H3006 Mol.Wt. 402.5
Prednisolone Acetate is 11~, 17a-dihydroxy-3,20- Specific optical rotation (2.4.22). + 128° to + 137°, determined
dioxopregna-l ,4~dien-2l-yl acetate. in a 0.35 per cent w/v solution in methanol.

1954

www.webofpharma.com
IP 2010 PREDNISOLONE SODIUM PHOSPHATE

Related substances. Determine by liquid chromatography than 0.25 times the area of the principal peak in the
(2.4.14). chromatogram obtained with reference solution (b) (0.05 per
cent).
NOTE-Prepare the solutions immediately before use.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Solvent mixture. Equal volumes of acetonitrile and buffer
on 1.0 g by drying in an oven at 105°.
solution pH 4 prepared by mixing 1 volume of dilute
hydrochloric acid, 5 volumes of a 6.81 per cent w/v solution Assay. Dissolve 0.1 gin 100.0 ml of ethanol (95 per cent).
of sodium acetate, 15 volumes ofa 3.73 percent w/v solution Dilute 2 ml of this solution to 100 ml with the same solvent.
of potassium chloride and 79 volumes of water. Measure the absorbance at the maximum at about 243 urn
(2.4.7). Calculate the content of CZ3H3006 taking 370 as the
Test solution. Dissolve 25 mg of the substance under
specific absorbance at 243 nm.
examination in 10 ml of methanol.
Storage. Store protected from light.
Reference solution (a). Dissolve 2 mg each of prednisolone
acetate RS and hydrocortisone acetate RS (prednisolone
impurity A RS) in 100 ml ofthe solvent mixture.
Reference solution(b). Dilute 1.0 ml ofthe test solution to 100 Prednisolone Sodium Phosphate
ml with the solvent mixture. Dilute 2.0 ml ofthis solution to 10
ml with the solvent mi;l£ture. oII
Chromatographic system O-P-ONa
I
a stainless steel column 25 cm x 4.6 mm, packed with -OH ONa
endcapped octadecylsilane bonded to porous silica (5
j.ll11),
column temperature. 40°,
mobile phase: a mixture of 35 volumes of acetonitrile o
and 65 volumes of water,
flow rate. 1 ml per minute, CzIHz7Naz08P Mol. Wt. 484.4
spectrophotometer set at 254 urn,
injection volume. 20 Ill. Predisolone Sodium Phosphate is 11~,17a, 21-
trihydroxypregna-l,4-diene-3,20-dione-2l-
The relative retention time with reference to prednisolone (dihydrogenphosphate) disodium salt.
acetate for prednisolone acetate impurity B is about 0.4, for
Prednisolone Sodium Phosphate contains not less than 96.0
prednisolone acetate impurity A is about 1.1, for prednisolone
per cent and not more than 102.0 per cent of CZIHz7Naz08P,
acetate impurity C is about 2.
calculated on the dried basis.
Inject reference solution (a). The test is not valid unless the
Category. Antiinflammtory; immunosuppressant.
resolution between the peaks due to prednisolone acetate
and prednisolone acetate impurity A is not less than 2.0.
Identification
Inject the test solution and reference solution (b). Run the
chromatogram 2.5 times the retention time of the principal A. Determine by infrared absorption spectrophotometry (2.4.6).
peak. In the chromatogram obtained with the test solution the Compare the spectrum with that obtained with prednisolone
area of secondary peak corresponding to prednisolone acetate sodium phosphate RS. If the spectra obtained in the solid
impurity A and B is not more than 5 times the area of the state show differences, dissolve the substance under
principal peak in the chromatogram obtained with reference examination and the reference substance separately in the
solution (b) (1.0 per cent), the area of secondary peak minimum volume of ethanol (95 per cent) evaporate to dryness
corresponding to prednisolone acetate impurity C is not more on a water-bath and record the spectra again using the residues.
than 2.5 times the area ofthe principal peak in the chromatogram B. To about 40 mg add 2 ml of sulphuric acid and heat gently
obtained with reference solution (b) (0.5 per cent). The area of until white fumes are evolved. Add nitric acid dropwise,
any other secondary peak is not more than 0.5 times the area continue the heating until the solution is almost colourless
of the principal peak in the chromatogram obtained with and cool. Add 2 ml of water, heat until white fumes are again
reference solution (b) (0.1 per cent) and the sum of the areas evolved, cool, add 10 ml of water and neutralise to red litmus
ofall secondary peaks is not more than 10 times the area of,the paper with dilute ammonia solution. The solution complies
principal peak in the chromatogram obtained with reference with reaction A of sodium salts and reaction B ofphosphates
solution (b) (2.0 per cent). Ignore any peak with an area less (2.3.1).

1955

www.webofpharma.com
PREDNISOLONE SODillM PHOSPHATE IP 2010

Tests than half the area of the principal peak in the chromatogram
obtained with reference solution (b) (1 per cent); the sum of
pH (2.4.24). 7.5 to 10.5 determined in 1.0 per cent w/v solution. the areas of all the peaks other than the principal peak is not
Specific optical rotation (2.4.22). +95.0° to +102.0°, determined greater than 1.5 times the area of the principal peak in the
in a 1.0 per cent w/v solution in a mixture of 9 volumes of chromatogram obtained with reference solution (b) (3 per
phosphate bufferpH 7.0 and 1 volume of carbon dioxide-free cent). Ignore any peak due to the solvent and any peak with
water. an area less than 0.025 times the area of the principal peak in
the chromatogram obtained with reference solution (b).
Related substances. Determine by liquid chromatography
(2.4.14). Inorganic phosphate. Not more than 1.0 per cent ofphosphate,
P04•
Test solution. Dissolve 62.5 mg of the substance under
examination in the mobile phase and dilute to 25.0 ml with the Dissolve 50 mg in sufficient water to produce 100 ml. To 10 ml
mobile phase. ofthe resulting solution add 5 ml of molybdovanadic reagent,
mix and allow to stand for 5 minutes. Any yellow colour in the
Reference solution (a). Dissolve 25 mg of prednisolone
solution is not more intense than that produced in a standard
sodium phosphate RS and 25 mg of prednisolone RS in the
prepared at the same time in the same manner using 10 ml of
mobile phase and dilute to 25.0 ml with the same solvent.
phosphate standard solution (5 ppm PO,J.
Dilute 1.0 ml ofthe solution to 25.0 ml with the mobile phase.
Water (2.3.43). Not more than 6.5 per cent, determined on
Reference solution (b). Dilute 1.0 ml of the test solution to
1.0 g.
50.0 ml with the mobile phase.
Assay.· Weigh accurately about 0.1 g, dissolve in sufficient
Chromatographic system
water to produce 100.0 ml and mix. Dilute 5.0 ml ofthe resulting
- a stainless steel column 15 cm x 4.6 IDm, packed with
solution to 250.0 ml with water. Measure the absorbance at
octadecylsilane bonded to porous silica or ceramic
the maximum at about 247 nm (2.4.7). Calculate the content of
microparticles (5 /lm),
CZJHz7NazOsP taking 312 as the specific absorbance at 247 nm.
- mobile phase: mix 1.36 g of potassium dihydrogen
phosphate and 0.6 g of hexylamine and allow to stand Storage. Store protected from light.
for 10 minutes and dissolve in 185 ml of water, add 65 ml
of acetonitrile,
- flow rate. 1 ml per minute,
- spectrophotometer set at 254 nm,
Prednisolone Sodium Phosphate
- injection volume. 20 Ill. Injection
Inject reference solution (b). Adjust the sensitivity of the Prednisolone Sodium Phosphate Injection is a sterile solution
system so that the height of the principal peak in the of Prednisolone Sodium Phosphate in Water for Injections.
chromatogram obtained with reference solution (b) is 70 per
Prednisolone Sodium Phosphate Injection contains not less
cent to 90 per cent ofthe full scale of the recorder.
than 90.0 per cent and not more than 110.0 per cent of the
Equilibrate the column with the mobile phase for about 30 stated amount of prednisolone phosphate, CZIHz90SP.
minutes.
Usual strength. 20 mg per ml.
Inject reference solution (a). The retention times are:
Description. A clear, colourless liquid.
prednisolone sodium phosphate, about 6.5 minutes and
prednisolone, about 8.5 minutes. The test is not valid unless Identification
the resolution between the peaks due to prednisolone sodium
phosphate and prednisolone is at least 4.5; ifthis resolution is A. In the Assay, the chromatogram obtained with the test
not achieved, increase the concentration. of acetonitrile or solution corresponds to the peak due to prednisolone sodium
increase the concentration of water in the mobile phase. phosphate in the chromatogram obtained with reference
solution (a).
Inject separately the test solution and reference solution (b).
Continue the chromatography for three times the retention B. To a volume containing 0.2 mg of Prednisolone Sodium
time ofthe principal peak. III the chromatogram obtained with Phosphate slowly add 1 ml of sulphuric acid. and allow to
the test solution, the area of any peak other than that from the stand for 2 minutes. A deep red colour is produced.
principal peak is not greater than the area ofthe principal peak
in the chromatogram obtained with reference solution (b) (2 Tests
per cent) and not more than one such peak has an area greater pH (2.4.24).7.0 to 8.0.

1956

www.webofpharma.com
IP 2010 PREDNISONE

Bacterial endotoxins (2.2.3). Not more than 5.0 Endotoxin Units Category. Adrenocortical steroid.
per mg of prednisolone phosphate.
Dose. 5 to 60 mg daily, in divided doses.
Other tests. Complies with the tests stated under Parenteral
Description. A white or almost white, crystalline powder;
Preparations (Injections).
odourless.
Assay. Detennine by liquid chromatography (2.4.14).
Test solution. Dilute an accurately measured volume of the Identification
injection to obtain a solution containing 0.001 per cent w/v of Tests A and B may be omitted iftests C and D are carried out.
prednisolone phosphate. Tests C and D may be omitted if tests A and B are carried out.
Reference solution (a). Weigh accurately about 10 mg of A. Detennine by infrared absorption spectrophotometry (2.4.6).
prednisolone sodium phosphate RS, dissolve in sufficient Compare the spectrum with that obtained with prednisone RS
water to produce 100.0 ml (solution A) and dilute 10.0 ml of or with the reference spectrum of prednisone.
the solution to 100.0 ml with watel:
B. Determine by thin-layer chromatography (2.4.17), coating
Reference solution (b). Add 10 ml of a 0.01 per cent w/v the plate with silica gel G.
solution of betamethasone sodium phosphate RS in water to
10 ml of solution A and dilute to 100 ml with water. Solvent mixture. A mixture of 90 volumes of acetone and
10 volumes offormamide.
Chromatographic system
a stainless steel column 20 cm x 4.6 rom, packed with Mobile phase. Chloroform.
octadecylsilane bonded to porous silica or ceramic Test solution. Dissolve 25 mg of the substance under
microparticles (l0 /lm) (such as Spherisorb ODS 1), examination in 10 ml ofthe solvent mixture.
mobile phase: a mixture of45 volumes of methanol and
55 volumes of citro-phosphate buffer pH 5.0, Reference solution (a). Dissolve 25 mg of prednisone RS in
flow rate. 2.0 ml per minute, 10 ml ofthe solvent mixture.
spectrophotometer set at 247 om, Reference solution (b). Mix equal volumes ofthe test solution
injection volume. 10 Ill. and reference solution (a).
Inject reference solution (b). The test is not valid unless the Place the dry plate in a tame containing a shallow layer of the
resolution between the peaks due to betamethasone sodium solvent mixture, allow the solvent mixture to ascend to the
phosphate and prednisolone sodium phosphate is at least 2.5. top, remove the plate from the tame and allow the solvent to
Inject alternatively the test solution and reference solution (a). evaporate. Use within 2 hours, with the flow of the mobile
phase in the direction in which the aforementioned treatment
Calculate the content ofCzIHz90sP in the injection. was done.
Storage. Store protected from light, in a single-dose or in multi- Apply to the plate 2 III of each solution. Allow the mobile
dose containers. phase to rise 12 cm. Dry the plate in a current ofwarm air, allow
the solvent to evaporate, heat at 120° for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Prednisone Heat at 120° for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with
reference solution (a). The principal spot in the chromatogram
obtained with reference solution (b) appears as a single,
compact spot.
C. Dissolve 2 mg in 2 ml of sulphuric acid and allow to stand
o for 5 minutes; an orange colour is produced within 5 minutes,
which exhibits a blue fluorescence in ultraviolet light at 365
CZIHz60S Mol. Wt. 358.4 om. Pour the solution into 10 ml of water; the colour changes
Prednisone is 17a.,21-dihydroxypregna-I,4-diene-3,11,20- first to yellow and then fades gradually but the blue
trione. fluorescence in ultraviolet light remains.
Prednisone contains not less than 96.0 per cent and not more D. Dissolve 1 mg in 1 ml of ethanol (95 per cent), evaporate to
than 104.0 per cent ofCzlHz60s, calculated on the dried basis. dryness at a pressure not exceeding 0.7 kPa, add 5 ml of 1 M

1957

www.webofpharma.com
PREDNISONE IP 2010

sodium hydroxide and heat at 70° for 30 minutes; not more Equilibrate the column with the mobile phaseB for at least
than a slight yellow colour is produced (distinction from 30 minutes and then with mobile phase A foTS minutes: For
cortisone acetate). subsequent chromatograms, use the conditions described from
40.0 to 52.0 minutes.
Tests
Specific optical rotation (204.22). +167° to +175°, determined Adjust the sensitivity of the system so that the height of the
in a 1.0 per cent w/v solution in dioxan. principal peak in the chromatogram obtained with reference
solution (b) is not less than 50 per centofthe full scale of the
Light absorption (204.7). Absorbance ofa 0.001 per cent w/v
recorder.
solution in methanol at the maximum at about 240 om, 0040 to
0043; the ratio of the absorbance at the maximum at about Inject reference solution (a). The retention times are:
240 nm to that at about 263 om, 1.85 to 2.05. prednisone, about 19 minutes and prednisolone about
Related substances. Determine by liquid chromatography 23 minutes. The test is not valid unless the resolution between
(2.4.14). the peaks corresponding to prednisone and prednisolone is
at least 2.7. Ifnecessary, adjust the concentration ofacetonitrile
Test solution. Dissolve 25 mg of the substance under in mobile phase A.
examination in methanol and dilute to 10 ml with the same
solvent. Inject separately methanol as a blank, the test solution and
reference solution (b). In the chromatogramobtained with the
Reference solution (a). Dissolve 2.0 mg of prednisolone RS
test solution: the area of any peak other than the principal
and 2.0 mg ofprednisone RS in methanol and dilute to 100 ml
peak is not greater than 0.25 times the area of the principal
with the same solvent.
peak in the chromatogram obtained with reference solution
Reference solution (b). Dilute 1 ml of the test solution to (b) (0.25 per cent); the sum ofthe areas of all the peaks, apart
100 ml with methanol. from the principal peak, is not greater than 0.75 times the area
Chromatographic system of the principal peak in the chromatogram obtained with
- a stainless steel column 25 cm x 4.6 mm, packed with reference solution (b) (0.75 per cent). Ignore any peak due to
octadecylsilyl silica gel (5 /-lm), the blank run and any peak with an area less than 0.05 times
column temperature. 45°, the area ofthe principal peak in the chromatogram obtained
mobile phase: A. a mixture of 100 ml of acetonitrile, with reference solution (b).
200 m1 of methanol and 650 ml of water, allowed to
Sulphated ash (2.3.18). Not more than 0.1 per cent.
equilibrate, diluted to 1000 m1 with water and mixed again,
B. acetonitrile, Loss on drying (2,4.19). Not more than 1.0 percent, determined
-flow rate. 2.5 ml per minute, on 1.0 g by drying in an oven at 105° for 3 hours.
a linear gradient programme using the conditions given
Assay. Weigh accurately about 0.1 g and dissolve in sufficient
below,
spectrophotometer set at 254 om,
ethanol to produce 100.0 m1. Dilute 2.0 ml of this solution to
100.0 ml with ethanol. Measure the absorbance ofthe resulting
injection volume. 20 /-l1.
solution at the maximum at about 238 om. Calculate the content
Time Mobile Mobile Comment of C Z1 HZ60 Staking 425 as the specific absorbance at 238 om.
Phase A PhaseB
(min) (per cent v/v) (per cent v/v) Storage. Store protected from light.
o 100 o Isocratic
25 100 o begin linear
gradient
40 40 60 end chromatogram, Prednisone Tablets
change to 100B
41 o 100 being treatment Prednisone Tablets contain not less than 90.0 per cent and
withB not more than 110.0 per cent of the stated amount of
46 o 100 end treatment, prednisone, CZIHz60S'
return to 100A
Usual strengths. 5 mg; 10 mg.
47 100 o begin equilibration
with A Identification
52 100 o end equilibration,
begin next Shake a quantity ofthe powdered tablets containing 30 mg of
chromatogram Prednisone with 10 m1 of chloroform, filter and evaporate the

1958

www.webofpharma.com
IP 2010 PREDNISONE TABLETS

filtrate to dryness. The residue complies with the following Chromatographic system
tests. - a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to poroussilica(5 Ilm),
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with prednisone RS column temperature. 45°,
or with the reference spectrum of prednisone. mobile phase: A. a mixture of 100 ml of acetonitrile,
200 ml of methanol and 650 ml of water, allowed to
B. Determine by thin-layer chromatography (2.4.17), coating equilibrate, diluted to 1000 ml with water and mixed again,
the plate with silica gel G.
B. acetonitrile,
Solvent mixture. A mixture of 90 volumes of acetone and flow rate. 2.5 ml per minute,
10 volumes offormamide. a linear gradient programme using the conditions given
Mobile phase. Chloroform. below,
spectrophotometer set at 254 run,
Test solution. Dissolve 25 mg of the residue in 10 ml of the
solvent mixture. injection volume. 20 Ill.

Reference solution (a). Dissolve 25 mg of prednisone RS in Time Mobile Mobile Comment


10 ml ofthe solvent mixture. Phase A Phase B
(min) (per cent v/v) (per cent v/v)
Reference solution (b). Mix equal volumes ofthe test solution
0 100 0 Isocratic,
and reference solution (a).
25 100 0 begin linear
Place the dry plate in a tank containing a shallow layer of the gradient
solvent mixture, allow the solvent mixture to ascend to the end chromatogram,
40 40 60
top, remove the plate from the tank and allow the solvent to change to 100B
evaporate. Use within 2 hours, with the flow of the mobile
phase in the direction in which the aforementioned treatment 41 0 100 begin treatment
was done. withB

Apply to the plate 2 III of each solution. Allow the mobile 46 0 100 end treatment,
phase to rise 12 cm. Dry the plate in a current ofwarm air, allow return to 100A
the solvent to evaporate, heat at 120° for 15 minutes and spray 47 100 0 begin equilibration
the hot plate with ethanolic sulphuric acid (20 per cent v/v). with A
Heat at 120° for a further 10 minutes, allow to cool and examine
52 100 0 end equilibration,
in daylight and in ultraviolet light at 365 nm. The principal
begin next
spot in the chromatogram obtained with the test solution
chromatogram
corresponds to that in the chromatogram obtained with
reference solution (a). The principal spot in the chromatogram Equilibrate the column with the mobile phase B for at least
obtained with reference solution (b) appears as a single, 30 minutes and then with mobile phase A for 5 minutes. For
compact spot. subsequent chromatograms use the conditions described from
40.0 to 52.0 minutes.
Tests Adjust the sensitivity of the system so that the height of the
Related substances. Determine by liquid chromatography principal peak in the chromatogram obtained with reference
(2.4.14). solution (b) is not less than 50 per cent of the full scale of the
recorder.
Test solution. Shake a quantity of the powdered tablets
containing 25 mg of Prednisone with 10 ml of methanol for Inject reference solution (a). The retention times are:
10 minutes, mix with the aid of ultrasound for 2 minutes and prednisone, about 19 minutes and prednisolone about
filter the extract (Whatman GFIF is suitable). 23 minutes. The test is not valid unless the resolution between
the peaks corresponding to prednisone and prednisolone is
Reference solution (a). Dissolve 2.0 mg of prednisolone RS at least 2.7. Ifnecessary, adjust the concentration ofacetonitrile
and 2.0 mg ofprednisone RS in methanol and dilute to 100 ml in mobile phase A.
with the same solvent.
Inject separately methanol as a blank, the test solution and
Reference solution (b). Dilute 1 ml of the test solution to reference solution (b). In the chromatogram obtained with the
100 ml with methanol. test solution: the area of any peak other than the principal

1959

www.webofpharma.com
PREDNISONE TABLETS IP 2010

peak is not greater than 0.25 times the area of the principal standard solution and dilute to 50.0 ml with a 50 per cent v/v
peak in the chromatogram obtained with reference solution solutIon of methanol.
(b) (0.25 per cent); the sum ofthe areas of all the peaks, apart Chromatographic system
from the principal peak, is not greater than 0.75 times the area - a stainless steel column 25 cm x 4 mm, packed with
of the principal peak in the chromatogram obtained with octadecylsilane bonded to porous silica (3 to 10 J..Lm),
reference solution (b) (0.75 per cent). Ignore any peak due to - mobile phase: a suitable filtered mixture of688 volumes
the blank run and any peak with an area less than 0.05 times of water, 250 volumes ofperoxide-free tetrahydrofuran
the area of the principal peak in the chromatogram obtained and 62 volumes of methanol such that at a flow rate of
with reference solution (b). 1 ml per minute the retention times of prednisone and
Uniformity of content. Comply with the test stated under acetanilide are about 8 and 6 minutes respectively,
Tablets. - spectrophotometer set at 254 nm,
- injection volume. 10 J..Ll.
Powder one tablet, add 50 ml of ethanol (95 per cent), shake
for 30 minutes, add sufficient ethanol (95 per cent) to produce Inject the reference solution. Adjust the operating parameters
100.0 ml. Centrifuge and pipette a suitable volume of the such that the peak obtained is about 50 per cent of the full
supernatant liquid equivalent to 0.5 mg of Prednisone and scale. The relative standard deviation for replicate injections
dilute to 50.0 ml with ethanol (95 per cent). Measure the is not more than 2.0 per cent and the resolution between
absorbance ofthe resulting solution at the maximum at about prednisone and the internal standard is not less than 3.0.
240 nm (2.4.7). Calculate the content ofCzIHz60s taking 415 as Inject the test solution and the reference solution. Calculate
specific absorbance at 240 om. the content of CZIHz60S in the tablets.
Dissolution (2.5.2). Storage. Store protected from light.
Apparatus No.1,
Medium. 900 ml of water,
Speed and time. 50 rpm and 30 minutes.
Pregabalin
Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance ofthe filtrate at the maximum at about 240 nm NHz
CH r
(2.4.7). Calculate the content ofCzIHz60s in the medium from I3 ~
the absorbance obtained from a solution of known ~COOH
H 3C
concentration of prednisone RS.
CgH 17NOz Mol. Wt. 159.2
D. Not less than 70 per cent ofthe stated amount OfCZIHz60s.
Pregabalin is (S)-4-amino-3-(2-methylpropyl)butyric acid.
Other tests. Comply with the tests stated under Tablets.
Pregabalin contains not less than 98.0 per cent and not more
Assay. Determine by liquid chromatography (2.4.14).
than 102.0 per cent ofC gH 17NOz, calculated on the dried basis.
Internal standard solution. Dissolve an accurately weighed
Category. Anticonvulsant.
quantity of acetanilide in a 50 per cent v/v solution of
methanol to obtain a solution having a known concentration Description. A white to off-white powder.
of0.1 1 mg per ml.
Identification
Test solution. Weigh and powder 20 tablets. To an accurately
weighed quantity of the powder containing about 20 mg of Determine by infrared absorption spectrophotometry (2.4.6).
Prednisone, aM 5mlofwater, mixWltl1 the aId ofuitras()und Compare the spectrum with that obtained with pregabalinR8
for one minute, add 50 ml of methanol and mix with the aid of or with the reference spectrum of pregabalin.
ultrasound for I minute. Dilute with water to 100.0 rnl and mix.
To 5.0 m1 ofthis solution add 5.0 ml ofinternal standard solution Tests
and dilute to 50.0 ml with a 50 per cent v/v solution of methanol
and mix. Filter through a 5 J..Lm filter and discard the fIrst 20 ml Specific optical rotation (2.4.22). +10° to +12.0°, determined
ofthe filtrate. on 1.0 per cent w/v solution.
--:::--:;;----'-·--'--;-~~~;--;----'---'--__,____--'-______;___.__,..___._--'-___;--'-......,.-Enantiomeric-purity.,...Determine-by~liquidc0hr0mat0graphy--'---'-
Reference solution. Weigh accurately a suitable quantity of
prednisone RS and dissolve in a 50 per cent v/v solution of (2.4.14).
methanol to obtain a solution having a concentration of about Merfey's reagent. Dissolve about 0.1502 g ofmerfey's reagent
0.2 mg per ml. To 5.0 rnl ofthis solution add 5.0 ml ofinternal in 50 ml of acetone.

1960

www.webofpharma.com
IP 2010 PREGABALIN CAPSULES

NOTE--8tore the reagent at 2° to 8°. Loss on drying (2.4.19). Not more than 0.5 per cent determined
on 1.0 g by drying in an oven at 105° for 3 hours.
Test solution (aJ. Dissolve about 100 mg of the substance
under examination in 100.0 ml of water. Assay. Detennine by liquid chromatography (2.4.14).
Test solution (b). To 2.0 ml of test solution (a), add 0.5 ml of Test solution. Dissolve about 200 mg of the substance under
merfey's reagent and 0.5 ml of 1 M sodium bicarbonate in the examination in 100.0 ml ofthe mobile phase.
test tube. Heat at 40° for 60 minutes. Allow to equilibrate to
Reference solution. A 0.2 per cent w/v solution ofpregabalin
room temperature.
RS in the mobile phase.
Chromatographic system
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm packed with a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 Ilm) (Such
octadecylsilane bonded to porous silica (5 Ilm) (Such
as Hypersil BDS C18),
as Inertsil ODS-3),
mobile phase: a mixture of62 volumes ofa buffer solution
mobile phase: a mixture of 95 volumes of water and 5
prepared by diluting about 7.18 ml of triethylamine to
volumes of acetonitrile,
1000 ml water, adjusted to pH 3.0 with orthophosphoric
- flow rate. 1 ml per minute,
acid and 38 volumes of acetonitrile,
spectrophotometer set at 205 nm,
flow rate. 2 ml per minute,
injection volume. 20 Ill.
spectrophotometer set at 340 nm,
- injection volume. 20 Ill. Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0 and the relative standard
The retention time for S- Pregabalin is about 9 minutes and R-
deviation ofreplicate injections is not more than 2.0 per cent.
Pregabalin is about 12 minutes.
Inject the test solution and the reference solution.
Inject test solution (b). The area of peak due to R-pregabalin
is not more than 0.5 per cent the area of the S-pregabalin. Calculate the content of CgH 17N02 •

Related substances. Determine by liquid chromatography Storage. Store protected from light and moisture.
(2.4.14).
NOTE-Prepare the solutions immediately before use.
Pregabalin Capsules
Solvent mixture. 95 volumes of water and 5 volumes of
acetonitrile. Pregabalin Capsules contain not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
Test solution. Dissolve about 50 mg of the substance under
pregabalin, CgH 17N0 2•
examination in 50.0 ml ofthe solvent mixture.
Usual strengths. 25 mg; 50 mg; 75 mg; 100 mg; l50mg; 225
Reference solution. Dilute 5.0 ml ofthe test solution to 100 ml
mg;300mg.
with the solvent mixture. Further dilute 5.0 ml ofthis solution
to 50 ml with the solvent mixture.
Identification
Use chromatographic system as described under Assay.
In the Assay, the principal peak in the chromatogram obtained
Inject the reference solution. The test is not valid unless the with the test solution corresponds to the principal peak in the
relative standard deviation of replicate injections is not more chromatogram obtained with the reference solution.
than 5.0 per cent.
Tests
Inject the test solution and the reference solution. In the
chromatogram obtained with the test solution, the area of any Dissolution (2.5.2);
secondary peak is not more than the area ofprincipal peak in
Apparatus No.1,
the chromatogram obtained with reference solution (0.5 per
cent) and the sum of areas of all the secondary peaks is not Medium. 900 ml of O. 05 M hydrochloric acid,
more than twice the area of the principal peak in the Speed and time. 50 rpm and 30 minutes.
chromatogram obtained with reference solution (1.0 per cent). Withdraw a suitable volume ofthe medium and filter.
Heavy Metals (2.3.13). 1 g complies with the limit test for Determine by liquid chromatography (2.4.14).
heavy metals, Method B (20 ppm).
Test solution. Use the filtrate and dilute, if necessary, with the
Sulphated ash (2.3.18). Not more than 0.1 percent. dissolution medium.

1961

www.webofpharma.com
PREGABALIN CAPSULES IP 2010

Reference solution. A 0.0083 per cent w/v solution of Inject reference solution (a). The test is not valid unless the
pregabalin RS in the dissolution medium. theoretical plates is not less than 3000, the tailing factor is not
more than 2.0 and the relative standard deviation of replicate
Use chromatographic system as described under Assay using
injections is not more than 2.0 per cent.
injection volume 100 Ill.
Inject the reference solution. The test is not valid unless the Inject reference solution (a) and the test solution. The relative
theoretical plates of the principal peak is not less than 2000, retention time with reference to lactam peak for lactose
tailing factor is not more than 2.0 and the relative standard conjugate impurity is about 0.78. In the chromatogram
deviation ofreplicate injections is not more than 2.0 per cent. obtained with the test solution, the area of the peak due to
lactam multiplied with relative response factor, 0.07 is not more
Inject the test solution and the reference solution. than 0.5 times the area ofthe principal peak in the chromatogram
Calculate the content ofCsH 17NO z. obtained with reference solution (a) (0.5 per cent), the area of
lactose conjugate impurity peak multiplied with relative
D. Not less than 70 per cent ofthe stated amount ofCsH 17NO z. response factor, 0.05 is not more than 1.5 times the area ofthe
Related substances. Determine by liquid chromatography principal peak in the chromatogram obtained with reference
(2.4.14). solution (a) (1.5 per cent). The area of any other secondary
peak is not more than the area of the principal peak in the
Solvent mixture. Dissolve 1.2 g of potassium dihydrogen chromatogram obtained with reference solution (a)'(1.0 per
phosphate in 1000 ml of water, adjust the pH to 6.9 with dilute cent) and the sum of areas of all the secondary peaks is not
potassium hydroxide. more than 2.5 times the area of the principal peak in the
Test solution. Mix the content of 10 capsules. Weigh and chromatogram obtained with reference solution (a) (2.5 per
disperse a quantity containing about 15 mg of Pregabalin, in cent). Ignore any peak with an area less than 0.05 times ofthe
about 25 ml ofthe solvent mixture, sonicate for 30 minutes and area of principal peak in the chromatogram obtained with
dilute to 100.0 ml with the solvent mixture. reference solution (a) (0.05 per cent).
Reference solution (a). A 0.015 per cent w/v solution of Other tests. Comply with the tests stated under Capsules.
pregabalin RS in the solvent mixture.
Assay. Determine by liquid chromatography (2.4.14).
Reference solution (b). Dissolve 4.5 mg of lactam RS in 10.0
Test solution. Mix the contents of 20 capsules. Disperse a
m1 ofthe solvent mixture.
quantity of powder containing about 200 mg of Pregabalin
Reference solution (c). Dissolve 750 mg ofpregabalin RS in with 35 ml of the mobile phase, sonicate for 10 minutes and
30 m1 ofsolvent mixture. Add about 5 ml ofreference solution dilute to 50 ml with the mobile phase.
(b) and dilute to 50.0 ml with the solvent mixture.
Reference solution. A 0.4 per cent w/v solution ofpregabalin
Chromatographic system RS in the mobile phase.
a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 Ilin) (Such Chromatographic system
as Inertsil ODS-3V), a stainless steel column 25 cm x 4.6 mm, packed with
mobile phase: A. a mixture of90 volumes ofthe solvent octadecylsilane bonded to porous silica (5 Ilm) (Such
mixture and 10 volumes of acetonitrile, as Inertsil ODS-3),
B. acetonitrile, mobile phase: a mixture of 92 volumes of solution
- a linear gradient programme using the conditions given containing 2.72 g of potassium dihydrogen
below, orthophosphate in 900 ml of water, add 2 ml of
flow rate. 1 ml per minute, triethylamine and adjusted to pH 6.0 with
spectrophotometer set at 210 nm, orthophosphoric acid, 5 volumes of methanol and 3
injection volume. 20 Ill. volumes of acetonitrile,
flow rate. 1 ml per minute,
Time Mobile phase A Mobile phase B spectrophotometer set at 205 nm,
(in min.) (per cent v/v) (per cent v/v) injection volume. 20 Ill.
o 100 o Inject the reference solution. The test is not valid unless the
5 100 o theoretical plates are not less than 2000, tailing factor is not
25 35 65 more than 2.0 and the relative standard deviation of replicate
30 100 o injections is not more than 2.0 per cent.
40 100 o Inject the test solution and the reference solution.

1962

www.webofpharma.com
IP 2010 PRIMAQUINE PHOSPHATE

Calculate the content of CgH 17NO z in the capsules. Foreign matter. Examined under a microscope using a mixture
of equal volumes ofglycerol and water, not more than traces
Storage. Store protected from moisture, at a temperature not
exceeding 30°. of matter other than starch granules are present.
Sulphated ash (2.3.18). Not more than 0.6 per cent.
Labelling. The label states the strength in terms of the
equivalent amount ofPregabalin. Loss on drying (2.4.19). NOhum'ethan 15.0 percent, determined
on 1.0 g by drying in an oven at 130° for 90 minutes.
Microbial contamination (2.2.9). Total viable aerobic count is
not more than 103bacteria and 10z fungi per gram, determined
Pregelatinised Starch by plate count. It complies with the test for Escherichia coli.
Pregelatinised Starch is prepared from maize starch, potato Labelling. The label states the type of starch used as starting
starch or rice starch by mechanical processing in the presence material.
of water, with or without heat, to rupture all or part of the
starch granules and subsequent drying. It contains no added
substances but it may be modified to render it compressible
and to improve its flow characteristics. Primaquine Phosphate
Description. A white or yellowish-white powder.

Identification
A. Microscopic- In a mixture of equal volumes of glycerol
and water, it presents irregular, translucent, white or yellowish-
white flakes or pieces with an uneven surface. Under polarised
light (between crossed nicol prisms), starch granules with a
distinct black cross intersecting at the hilum may be seen.
ClsHzIN30,2H3P04 Mol. Wt. 455.3
E. Disperse 0.5 g in2 ml ofwater without heating, add 0.05 ml Primaquine Phosphate is (RS)-8-(4-amino-l-
of iodine solution; a reddish-violet to blue colour is produced. methylbutylamino)-6-methoxyquinoline diphosphate.
Tests Primaquine Phosphate contains not less than 98.5 per cent
and not more than 101.5 per cent of ClsHzIN30,2H3P04,
pH (2.4.24). 4.5 to 7.0, determined in 3.0 percent w/v solution calculated on the dried basis.
in carbon dioxide-free water.
Category. Antimalarial.
Oxidising substances. Transfer 4.0 g to a glass-stoppered,
125 ml conical flask and add 50.0 ml of a mixture of equal Dose. The equivalent of 15 mg ofprimaquine, once a day for
volumes of water and methanol. Insert the stopper and swirl 14 days.
for 5 minutes. Transfer to a glass-stoppered 50 ml centrifuge Description. An orange, crystalline powder; odourless or
tube and centrifuge. Transfer 30.0 ml ofthe clear supernatant almost odourless.
liquid to a glass-stoppered 125-ml conical flask. Add I mlof
glacial acetic acid and 0.5 g to 1.0 g of potassium iodide. Identification
Insert the stopper, swirl, and allow to stand for 30 minutes in
Test A may be omitted iftests B, C andD are carried out. Tests
the dark. Add I ml ofstarch solution and titrate with 0.002 M
Band C may be omitted if tests A and D are carried out.
sodium thiosulphate until the starch-iodine colour disappears.
Carry out a blank titration. Not more than 1.4 ml of O. 002 M A. Determine by infrared absorption spectrophotometry (2.4.6).
sodium thiosulphate is required (0.002 per cent, calculated as Compare the spectrum with that obtained with primaquine
HzOz). phosphate RS or with the reference spectrum of primaquine
phosphate.
I ml of0.002 M sodium thiosulphate is equivalent to 0.034 mg
of oxidising substances, calculated as HzO z. E. When examined in the range 300 nm to 450 nm (2.4.7), a
0.015 per cent w/v solution in 0.01 Mhydrochloricacidshows
Sulphur dioxide (2.3.40). Not more than 50 ppm.
absorption maxima at about 332 nm and 415 nm; absorbance
Iron (2.3.14). Dissolve the residue obtained in the test for at about 332 nm,aboutO.68 to 0.78 and at about 41 5 nm, 0.41 to
Sulphated ash in 20 ml ofdilute hydrochloric acid, filter. 10 ml 0.53. Dilute 5 ml of the solution to 50 ml with 0.01 M
ofthe filtrate complies with the limit test for iron (20 ppm). hydrochloric acid. When examined in the range 215 urn to

1963

www.webofpharma.com
PRIMAQUINE PHOSPHATE IP 2010

310 nm, the resulting solution shows absorption maxima at The test is not valid unless in the chromatogram obtained
about 225 urn, 265 urn and 282 urn; absorbances at the maxima with reference solution (c) there is a peak just before the
are 0.74 to 0.77, 0.50 to 0.53 and 0.50 to 0.52, respectively. principal peak with an area of about 6 per cent of that of the
C. Determine by thin-layer chromatography (2.4.17), coating principal peak and the resolution between these peaks is not
the plate with silica gel F254. less than 2.0.

Mobile phase. A mixture of 60 volumes of chloroform, Inject each solution and record the chromatograms for at least
40 volumes of methanol and 1 volume of strong ammonia twice the retention time ofprimaquine. The sum ofthe areas of
solution. any secondary peaks in the chromatogram ob~ained with the
test solution is not greater than the area of the principal peak
Test solution. DissolveO.2 g in 5 m1 of water, dilute to 10 ml
in the chromatogram obtained with reference solution (a).
with methanol and dilute 1 volume ofthe resulting solution to
Ignore any peak the area of which is less than that of the
10 volumes with methanol (50 per cent).
principal peak in the chromatogram obtained with reference
Reference solution. Dissolve 20 mg ofprimaquinephosphate solution (b).
RS in 5 ml of water, dilute to 10 ml with methanol.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Apply to the plate 5 j..Ll of each solution. After development, on 1.0 g by drying in an oven at 105°.
dry the plate in air and examine in ultraviolet light at 254 nm.
Assay. Weigh accurately about 0.2 g, dissolve in 40 ml of
The principal spot in the chromatogram obtained with the test
anhydrous glacial acetic acid with gentle heating. Titrate
solution corresponds to that in the chromatogram obtained
with 0.1 M perchloric acid, determining the end point
with the reference solution.
potentiometrically (2.4.25). Carry out a blank titration.
D. Dissolve 50 mg in 5 ml of water, add 2 ml of 2 M sodium
hydroxide and shake with two quantities, each of 5 ml, of 1 ml of 0.1 M perchloric acid is equivalent to 0.02277 g of
chloroform. The aqueous layer, acidified by the addition of ClsHzIN30,2H3P04.
nitric acid, gives reaction B of phosphates (2.3.1). Storage. Store protected from light and moisture.
Tests
pH (2.4.24). 2.5 to 3.5, determined in a 1.0 per centw/v solution.
Related substances. Determine by liquid chromatography Primaquine Tablets
(2.4.14). Primaquine Phosphate Tablets
Test solution. Add 0.2 ml ofstrong ammonia solution to 1 ml
Primaquine Tablets contain not less than 90.0 per centand not
of a 1.0 per cent w/v solution of the substance under
more than 110.0 per cent ofthe stated amount ofprimaquine,
examination, shake with 10 ml ofthe mobile phase and use the
ClsHz1N30. The tablets are coated.
clear, lower layer.
Usual strength. The equivalent of2.5 mg of primaquine (13
Reference solution (a). Dilute 3 ml of the test solution to
mg of Primaquine Phosphate is approximately equivalent to
100 ml with the mobile phase.
7.5 mg ofprimaquine).
Reference solution (b). Dilute 1 m1 ofthe test solution to 10m1
with the mobile phase and further dilute 1 ml ofthe resulting Identification
solution to 50 ml with the same solvent.
A. Dissolve a quantity of the powdered tablets containing
Reference solution (c). Add 0.2 ml ofstrong ammonia solution 60 mg ofprimaquine in a mixture of 10 m1 ofwater and 2 m1 of
to 1 m1 of a 1.0 per cent w/v solution of the primaquine 2 M sodillll1 hydroxide and extractwith two quantities, each
phosphate RS, shake with 10 ml of the mobile phase and use of 20 ml, of chloroform. Wash the chloroform extracts with
the clear, lower layer. water, dry over anhydrous sodium sulphate, evaporate to
Chromatographic system dryness and dissolve the residue in 2 ml of chloroform. The
- a stainless steel column 20 em x 4.6 mm, packed with residue complies with the following test.
porous silica particles (10 j..Lm),
Determine by infrared absorption spectrophotometry (2.4.6).
- mobile phase: a mixture of 45 volumes of chloroform,
Compare the spectrum with that obtained with primaquine
45 volumes of hexane, 10 volumes of methanol and
phosphate RS or with the reference spectrum of primaquine.
0.1 volume of strong ammonia solution,
- flow rate. 3 ml per minute, B. Extract a quantity ofthe powdered tablets containing 25 mg
- spectrophotometer set at 261 urn, of primaquine with 10 ml of water and filter. To 2 m1 of the
- injection volume. 20 j..L1. filtrate add 3 ml ofwater and 1 ml ofa 5 percentw/v solution

1964

www.webofpharma.com
IP 2010 PROBENECID

of eerie ammonium sulphate in 2 M nitric acid; a deep violet Identification


colour is produced immediately.
Test A may be omitted iftests Band C are carried out. Tests B
Tests and C may be omitted if test A is carried out.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Uniformity of content. Comply with the tests stated under
Compare the spectrum with that obtained with probenecidRS
Tablets.
or with the reference spectrum of probenecid.
Transfer one tablet into a suitable container, add 5 ml of
B. When examined in the range 210 nm to 360 nm (2.4.7), a
hydrochloric acid, disperse the tabletin about 25 g ofcrushed
0.001 per cent w/v solution in a mixture of9 volumes ofethanol
ice and add sufficient water to produce 50.0 ml. Carry out the
(95 per cent) and 1 volume of 0.1 M hydrochloric acid shows
nitrite titration (2.3 .31), using 0.01 M sodium nitrite as titrant.
absorption maxima at about 223 urn and 248 nm; absorbance
Carry out a blank titration.
at about 248 nm is about 0.33.
1 ml of 0.01 M sodium nitrite is equivalent to 0.002594 g of
C. Dissolve 0.2 g in about 0.6 ml of2 M ammonia and add 3 ml
ClsH21N30.
of 1.7 per cent w/v solution of silver nitrate; a white precipitate
Other tests. Comply with the tests stated under Tablets. is produced which is soluble in an excess of dilute ammonia
Assay. Weigh and powder 20 tablets. Weigh accurately a solution.
quantity ofthe powder containing about 0.15 g ofprimaquine,
Tests
dissolve in 20 ml of water, add 5 ml of2 M sodium hydroxide
and extract with four quantities, each of25 ml of chloroform. Appearance of solution. A 10.0 per cent w/v solution i.n 2 M
Combine the chloroform extracts and evaporate to a volume sodium hydroxide is clear (2.4.1), and not more intensely
of about 10 ml. Add 40 ml of anhydrous glacial acetic acid, coloured than reference solution YS6 (2.4.1).
Titrate with 0.1 M perchloric acid, determining the end point Acidity. Add 2.0 g to 100 ml of water, heat on a water-bath for
potentiometrically (2.4.25). Carry out a blank titration. 30 minutes, cool, filter and dilute with water to 100.0 mI. Titrate
1 ml of 0.1 M perchloric acid is equivalent to 0.01297 g of 50.0 ml of the solution with 0.1 M sodium hydroxide using
ClsH21N30. phenolphthalein solution as indicator. Not more than 0.5 ml
of 0.1 M sodium hydroxide is required to change the colour of
Storage. Store protected from moisture.
the solution.
Labelling. The label states the strength in terms of the
Related substances. Determine by thin-layer chromatography
equivalent amount ofprimaquine.
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of55 volumes of toluene, 20 volumes
of di-isopropyl ether, 15 volumes of chloroform and
Probenecid 10 volumes of glacial acetic acid.
Test solution. Dissolve 0.1 g of the substance under
examination in 10 ml of acetone.
Reference solution. A 0.005 per cent w/v solution of the
substance under examination in acetone.
Apply to the plate 20 III of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 urn.
Any secondary spot in the chromatogram obtained with the
Cl3Hl9N04S Mol. Wt. 285.4
test solution is not more intense than the spot in the
Probenecid is 4-(dipropylsulphamoyl)benzoic acid. chromatogram obtained with the reference solution.
Probenecid contains not less than 99.0 per cent and not more Heavy metals (2.3.13).1.0 g complies with the limit test for
than 101.0 per cent of C 13H I9N04S, calculated on the dried heavy metals, Method B (20 ppm).
basis.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Category. Uricosuric agent and for delaying renal excretion of
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
penicillins and cephalosporins.
on 1.0 g by drying in an oven at 105°.
Dose. 250 mg to 500 mg twice daily.
Assay. Weigh accurately about 0.5 g and dissolve in 100 ml of
Description. A white or almost white, crystalline powder. ethanol (95 per cent), shaking well and heating gently if

1965

www.webofpharma.com
PROBENECID TABLETS IP 2010

necessary. Cool and titrate with 0.1 M sodium hydroxide, Medium. 900 ml ofphosphate bufferpH 7.6,
using bromothymol blue solution as indicator. Carry out a Speed and time. 50 rpm and 30 minutes.
blank titration.
Withdraw a suitable volume of the medium and filter, Dilute
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02854 g of 4.0 ml ofthe filtrate to 100.0 ml with 0.1 M sodium hydroxide.
CI3HI9N04S. Measure the absorbance of the resulting solution at the
Storage. Store protected from moisture. maximum at about 244 nm (2.4.7). Similarly measure the
absorbance of a solution of a known concentration of
probenecid RS. Calculate the content of C 13H I90 4S.
D. Not less than 80 per cent of the stated amount of
Probenecid Tablets CI3HI9N03S,
Probenecid Tablets contain not less than 95.0 per cent and Other tests. Comply with the tests stated under Tablets.
not more than 105.0 per cent of the stated amount of
probenecid, CI3HI9N04S. Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.2 g ofProbencid,
Usual strength. 500 mg. add 200 ml of ethanol (95 per cent) and 5 ml of 1 M
hydrochloric acid, heat on a water-bath at 70 0 for 30 minutes,
Identification
shaking occasionally. Cool, add sufficient ethanol (95 per
A.Triturate a quantity of the powdered tablets containing 0.5 cent) to produce 250.0 ml and filter. To 5.0 ml ofthe filtrate add
g of Probenecid with ethanol (95 per cent), filter and 5 ml of 0.1 M hydrochloric acid, dilute to 250.0 ml with ethanol
concentrate the filtrate by evaporation on a water-bath. Cool, (95 per cent) and measure the absorbance of the resulting
filter and recrystallise the residue from ethanol (50 per cent). solution at the maximum at about 248 nm (2.4.7). Calculate the
The residue complies with the following test. content of C 13H I9N04S taking 332 as specific absorbance at
248nm.
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with probenecidRS Storage. Store protected from moisture.
or with the reference spectrum of probenecid.
B. When examined in the range 210 nm to 360 nm (2.4.7), the
final solution obtained in the Assay shows absorption maxima Procainamide Hydrochloride
at about 225 nm and 248 nm.
C. The residue obtained in test A melts at about 1990 (2.4.21). ° (CH
3

Tests ~JlN~N,--/,CH3
Related substances. Determine by thin-layer chromatography ~ H .HCI
H2N
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of55 volumes of toluene, 20 volumes C 13Hz1 N30,HCl Mol. Wt. 271.8
of di-isopropyl ether, 15 volumes of chloroform and Procainamide Hydrochloride is 4-amino-N-[2-(diethylamino)
10 volumes of glacial acetic acid. ethyl)benzamide hydrochloride.
Test solution. Extract a quantity of the powdered tablets Procainamide Hydrochloride contains not less than 98.0 per
containing 0.2 g ofProbenecid with 20 ml of acetone, filter and cent and not more than 101.0 per cent of C 13 Hz1 N 30,HC1,
use the filtrate. calculated on the dried basis.
Reference solution. Dilute 1 ml of the test solution to 200 ml Category. Antiarrhythmic.
with acetone.
Dose. Orally, 500 mg to 1.5 g daily, in divided doses; by slow
Apply to the plate 20 III of each solution. After development, intravenous injection
dry the plate in air and examine in ultraviolet light at 254 nm.
Description. A white or very slightly yellow, crystalline
Any secondary spot in the chromatogram obtained with the
powder; hygroscopic.
test solution is not more intense than the spot in the
chromatogram obtained with the reference solution. Identification
Dissolution (2.5.2). Test A may be omitted iftests B, C andD are carried out. Tests
Apparatus. No.1, B, C and D may be omitted if test A is carried out.

1966

www.webofpharma.com
IP 2010 PROCAINAMIDE INJECTION

A. Determine by infrared absorption spectrophotometry (2.4.6). Procainamide Injection


Compare the spectrum with that obtained with procainamide
hyc/rochloride RS or with the reference spectrum of Procainamide Hydrochloride Injection
procainamide hydrochloride.
Procainamide Injection is a sterile solution of Procainamide
B. When examined in the range 230 nm to 360 nm (2.4.7), a Hydrochloride in Water for Injections. It may contain Sodium
0.001 per cent w/v solution in 0.1 M sodium hydroxide shows Metabisulphite as a stabilising agent.
an absorption maximum at about 273 nm; absorbance at about Procainamide Injection contains not less than 95.0 per cent
273 nm, 0.58 to 0.61. and not more than 105.0 per cent of the stated amount of
C. Dilute I ml of a solution, prepared by dissolving 2.5 g in procainamide hydrochloride, C 13Hz1 N 30, HC1.
sufficient carbon dioxide-free water to produce 25 ml, to 2 ml Usual strength. 100 mg per m1.
with water. 1 ml ofthis solution gives the reaction ofprimary
aromatic amines (2.3.1). Identification
D. A 2 per cent w/v solution gives reaction A of chlorides A. Dilute a volume containing 0.25 g of Procainamide
(2.3.1). Hydrochloride to 25 ml with water, make alkaline with 5 M
sodium hydroxide and extract with two quantities, each of
Tests 5 ml, of chloroform. Filter the combined extracts through
anhydrous sodium sulphate, evaporate the filtrate to dryness
Appearance of solution. A 10.0 per cent w/v solution in carbon
using a rotatory evaporator and dissolve the residue in 5 ml of
dioxide-free water is clear (2.4.1), and not more intensely
chloroform.
coloured than reference solution BS6 (2.4.1).
Determine by infrared absorption spectrophotometry (2.4.6).
pH (2.4.24). 5.6to 6.3, deterrnined in a 10.0 per cent w/v solution.
Compare the spectrum with that obtained with procainamide
Related substances. Determine by thin-layer chromatography hydrochloride RS or with the reference spectrum of
(2.4.17), coating the plate with silica gel GF254. procainamide.
Mobile phase. A mixture of 60 volumes of 1-butanol, B. Dilute a suitable volume of the injection with water to
30 volumes of water and 15 volumes of glacial acetic acid. produce a solution containing 0.0005 per cent w/v of
Procainamide Hydrochloride. Absorbance of the resulting
Test solution. Dissolve 0.1 g of the substance under solution at about 280 nm, about 0.30 (2.4.7).
examination in 10 ml of ethanol (95 per cent).
C. Gives the reactions ofchlorides (2.3.1).
Reference solution. A 0.005 per cent w/v solution of the
substance under examination in ethanol (95 per cent).
Tests
Apply to the plate 5 III of each solution. After development,
pH (2.4.24). 4.0 to 5.5.
dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the Related substances. Determine by thin-layer chromatography
test solution is not more intense than the spot in the (2.4.17), coating the plate with silica gel GF254.
chromatogram obtained with the reference solution.
Mobile phase. A mixture of 60 volumes of 1-butanol,
Heavy metals (2.3.13). 1.0 g complies with the limit test for 30 volumes of water and 15 volumes of glacial acetic acid.
heavy metals, Method B (20 ppm).
Test solution. Dilute a volume ofthe injection containing 0.1 g
Sulphated ash (2.3.18). Not more than 0.1 per cent. ofProcainamide Hydrochloride to 5 ml with methanol.

Loss on drying (2.4.19). Not more than 0.5 per cent, detennined Reference solution. Dilute 1 volume of the test solution to
on 1.0 g by drying in an oven at 105°. 100 volumes with methanol.

Assay. Weigh accurately about 0.25 g, dissolve in 75 ml of Apply to the plate 5 III of each solution. After development,
water and 10 ml of hydrochloric acid and carry out the nitrite dry the plate in air and examine in ultraviolet light at 254nm.
titration (2.3.31). Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the
I ml of 0.1 M sodium nitrite is equivalent to 0.02718 g of chromatogram obtained with the reference solution.
C13Hz1N30, HCL
Other tests. Complies with the tests stated under Parenteral
Storage. Store protected from light. Preparations (Injections).

1967

www.webofpharma.com
PROCAINAMIDE TABLETS IP 2010

Assay. To a volume containing about 0.25 g of Procainamide Other tests. Comply with the tests stated under Tablets.
Hydrochloride add 45 ml of 6 M hydrochloric acid and boil Assay. Weigh and powder 20 tablets. Weigh accurately a
for 1 minute. Carry out the nitrite titration (2.3.31). quantity ofthe powder containing about 0.25 g ofProcainamide
1 ml of 0.1 M sodium nitrite is equivalent to 0.02718 g of Hydrochloride, add 100 ml of 6 M hydrochloric acid, shake
C13H21N30, HCl. for 15 minutes and boil for 1 minute. Carry out the nitrite
titration (2.3.31).
Storage. Store protected from light.
1 ml of 0.1 M sodium nitrite is equivalent to 0.02718 g of
C13H21N30, HCl.
Storage. Store protected from light and moisture.
Procainamide Tablets
Procainamide Hydrochloride Tablets
Procainamide Tablets contain not less than 95.0 per cent and Procaine Hydrochloride
not more than 105.0 per cent of the stated amount of
procainamide hydrochloride, C 13H2J N 30,HCl.
Usual strength. 250 mg.
, Hel
Identification
A. Shake a quantity ofthe powdered tablets containing 0.25 g
of Procainamide Hydrochloride with 25 ml of water, make
C13H20N202,HCl Mol. Wt. 272.8
alkaline with 5 M sodium hydroxide and extract with two
quantities, each of 5 ml, of chloroform. Filter the combined Procaine Hydrochloride is 2-(diethylamino)ethy14-
extracts through anhydrous sodium sulphate, evaporate the aminobenzoate hydrochloride.
filtrate to dryness using a rotatory evaporator and dissolve Procaine Hydrochloride contains not less than 99.0 per cent
the residue in 5 ml of chloroform. and not more than 101.0 per cent ofC13H2oN202,HCl, calculated
Determine by infrared absorption spectrophotometry (2.4.6). on the dried basis.
Compare the spectrum with that obtained with procainamide Category. Local anaesthetic.
hydrochloride RS or with the reference spectrum of
procainamide. Dose. To be adjusted according to the site of operation and
response of the patient.
B. Triturate a quantity ofthe powdered tablets containing 1 g
ofProcainamide Hydrochloride with 10 ml of water and filter. Description. Colourless crystals or a white, crystalline powder;
The filtrate gives the reactions of chlorides (2.3.1). odourless.

Identification
Tests
Test A may be omitted if tests B, C, D and E are carried out.
Related substances. Determine by thin-layer chromatography
Tests B, C and D may be omitted if tests A and E are carried
(2.4.17), coating the plate with silica gel GF254.
out.
Mobile phase. A mixture of 60 volumes of I-butanol,
A. Determine by infrared absorption spectrophotometry (2.4.6).
30 volumes of water and 15 volumes of glacial acetic acid.
Compare the spectrum with that obtained with procaine
Test solution. Shake a quantity of the powdered tablets hydrochloride RS or with the reference spectrum of procaine
containing 0.4 g ofProcainamide Hydrochloride with 20 ml of hydrochloride.
methanol (90 per cent) for 15 minutes and filter.
B. To 0.2 ml ofa 5 per cent w/v solution add 2 ml of water and
Reference solution. Dilute 1 volume of the test solution to 0.5 ml of 1 M sulphuric acid, shake and add 1 ml of a 0.1 per
100 volumes with methanol. cent w/v solution of potassium permanganate; the colour is
Apply to the plate 5 III of each solution. After development, immediately discharged.
dry the plate in air and examine in ultraviolet light at 254 nrn. C. To about5 mg add 0.5 ml ofjUming nitric acid, evaporate to
Any secondary spot in the chromatogram obtained with the dryness on a water-bath, cool, dissolve the residue in 5 ml of
test solution is not more intense than the spot in the acetone and add 1 ml of 0.1 M ethanolic potassium
chromatogram obtained with the reference solution. hydroxide; only a brownish red colour develops.

1968

www.webofpharma.com
IP 2010 PROCAINE AND ADRENALINE INJECTION

D. Dilute 1 mlofa5 percentw/v solution to 100ml with water; Procaine and Adrenaline Injection contains not less than
2 ml of the solution gives the reaction of primary aromatic 1.9 per cent and not more than 2.1 per cent w/v of procaine
arnines (2.3.1). hydrochloride, C13HzoNzOz, HCI and the equivalent ofnot less
E. Gives reaction A ofchlorides (2.3.1). than 0.00175 percent and not more than 0.00225 per cent w/v
ofadrenaline, C9H I3N0 3.
Tests Description. A clear, colourless solution.
Appearance of solution. A 5.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and colourless (2.4.1).
Identification

pH (2.4.24). 5.0to 6.5, determined in a 2.0 per cent w/v solution. A. To 5 ml add 5 ml of water and 10 ml ofpicric acidsolution,
shake gently and set aside for 1 hour; the crystalline precipitate,
Related substances. Determine by thin-layer chromatography after washing with water and drying at 100°, melts at about
(2.4.17), coating the plate with silica gel GF254. 134° (2.4.21).
Mobile phase. A mixture of 80 volumes of dibutyl ether, B. To 5 ml add 1 m1 of hydrochloric acid, cool to 0°, add 5 ml of
16 volumes of n-hexane and 4 volumes of glacial acetic acid. sodium nitrite solution and pour the mixture into 2 ml of
Test solution. A 10 per cent w/v solution of the substance 2-naphthol solution containing 1 g of sodium acetate; an
under examination in water. orange-red colour is produced.

Reference solution. A 0.005 per cent w/v solution of C. To 10 ml add 4 ml of disodium hydrogen phosphate solution
4-aminobenzoic acid in water. and sufficient 0.1 M iodine to produce a distinct brown colour.
Add 0.1 M sodium thiosulphate to remove the excess of
Apply to the plate 5 III of each solution. After development,
iodine; a pink colour is produced.
dry the plate at 105° for 10 minutes and examine in ultraviolet
light at 254 nm. Any secbndary spot in the chromatogram
obtained with the test solution is not more intense than the Tests
spot in the chromatogram obtained with the reference solution. pH (2.4.24). 3.0 to 5.5.
The principal spot remains on the line of application.
Other tests. Complies with the tests stated under Parenteral
Heavy metals (2.3.13).2.0 g complies with the limit test for Preparations (Injections).
heavy metals, Method A (10 ppm).
Assay. For procaine hydrochloride - To 10.0 ml of the
Sulphated ash (2.3.18). Not more than 0.1 per cent. injection add 0.5 g ofsodium carbonate and extract with three
Loss on drying (2.4.19). Not more than 0.5 per cent, determined quantities, each of20 ml, ofa mixture of 1 volume of2-propanol
on 1.0 g by drying in an oven at 105°. and 3 volumes of chloroform until complete extraction of
procaine is effected. Shake the combined extracts with 5 ml of
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of 2 M water, wash the water with the solvent mixture and add the
hydrochloric acid, add 3 g ofpotassium bromide, cool in ice washing to the combined extracts. Shake the combined extracts
and titrate slowly with 0.1 M sodium nitrite, stirring constantly. and washings with 10.0 ml of 0.1 M hydrochloric acid,
Determine the end point potentiometrically (2.4.25).
separate the acid layer, wash the combined extracts and
1 ml of 0.1 M sodium nitrite is equivalent to 0.02728 g of washings with 5 ml of water, add the aqueous extract to the
C13HzoNzOz, HCl. separated acid layer and titrate with 0.1 M sodium hydroxide,
using methyl red-methylene blue solution as indicator.
Storage. Store protected from light.
1 m1 of 0.1 M hydrochloric acid is equivalent to 0.02728 g of
CI3HzoNzOz,HCl.
For adrenaline To 10.0 m1 of the injection add 20 mg of
Procaine and Adrenaline Injection sodium metabisulphite, 0.1 ml of ferrous sulphate-citrate
Procaine Hydrochloride and Adrenaline Bitartrate solution, 1 ml of glycine buffer solution and mix. Allow to
stand for 10 minutes, extract with 10 ml of ether, allow to
Injection; Procaine Hydrochloride and Epinephrine
separate, reject the ether and 'measure- the absorbance of a
Bitartrate Injection
4-cm layer of the solution at about 540 nm (2.4.7). Calculate
Procaine and Adrenaline Injection is a sterile solution of the content of adrenaline, C9H 13N0 3, from a reference curve
Procaine Hydrochloride and Adrenaline Bitartrate in Water prepared by treating suitable aliquots of a solution of
for Injections. adrenaline bitartrate RS in the same manner.

1969

www.webofpharma.com
PROCAlNE PENICILLIN IP 2010

1 mg of adrenaline bitartrate is equivalent to 0.0005497 g of Water (2.3.43).2.8 to 4.2 per cent, determined on 0.5 g.
C9H I3N03.
Assay. Determine the contents of benzyl penicillin and
Storage. Store protected from light. procaine by liquid chromatography (2.4.14).
Labelling. The label states the strength as "Procaine Test solution. Weigh accurately 70 mg ofthe substance under
Hydrochloride, 2 per cent w/v; Adrenaline, 0.002 per cent examination and dissolve in 30 ml ofthe mobile phase with the
w/v". aid of ultrasound for about 2 minutes. Dilute to 50.0 ml with
the mobile phase.
Reference solution (a). Weigh accurately about 56 mg of
Procaine Penicillin benzyl penicillin potassium RS and 40 mg of procaine
hydrochloride RS and dissolve in 25 ml of the mobile phase
with the aid of ultrasound for about 2 minutes. Dilute to
50.0 ml with the mobile phase.
Reference solution (b). Mix 1 volume of a 0.24 per cent w/v
solution of phenoxymethylpenicillin potassium RS in the
mobile phase with 3 volumes of reference solution (a).
CI3H20N202,CI6HlSN204S,H20 Mol. Wt. 588.7
Chromatographic system
Procaine Penicillin is 2-diethylaminoethyI4-aminobenzoate
a stainless steel column 30 cm x 4 mm, packed with
(6R)-6-(2-phenylacetamido)penicillanate monohydrate.
octadecylsilane bonded to porous silica (l0 /lm),
Procaine Penicillin contains not less than 51 per cent and not mobile phase: a mixture of 50 volumes of a solution
more than 59.6 per cent of penicillin G, calculated as prepared by mixing 14 g of potassium dihydrogen
CI6HISN204S, and not less than 37.5 per cent and not more phosphate in 6.5 g of tetrabutylammonium hydroxide
than 43.0 per cent ofprocaine, C13H20N202, both calculated on (40 per cent) and adjusting the pH to 7.0 with 1 M
the anhydrous basis. potassium hydroxide or dilute phosphoric acid,
25 volumes of acetonitrile and 25 volumes of water,
Category. Antibacterial.
flow rate. 1 ml per minute,
Dose. By intramuscular injection, 300 to 900 mg daily (300 mg spectrophotometer set at ~35 om,
ofProcaine Penicillin is approximately equivalent to 200 mg of injection volume. 20 /ll.
benzylpenicillin).
Inject reference solution (b). The resolution between benzyl
Description. A white, crystalline powder. penicillin potassium and phenoxymethylpenicillin potassium
is not less than 2. The relative retention time ofprocaine with
Identification reference to benzyl penicillin is about 2.2.
A. Determine by infrared absorption spectrophotometry (2.4.6). Inject the test solution and reference solution (a). Record the
Compare the spectrum with that obtained with procaine
peak responses of the main peaks of benzyl penicillin and
penicillin RS or with the reference spectrum of procaine
procaine. Calculate the content of benzyl penicillin,
penicillin.
Cl6HlSN204S and the content of procaine, Cl3H20N202 using
B. In the Assay, the principal peak in the chromatogram the factor of 0.866 for converting the content of procaine
obtained with the test solution corresponds to the peak in the hydrochloride to that of procaine.
chromatogram obtained with the reference solution (a).
Procaine Penicillin intended for use in the manufacture of
C. A turbid solution of 0.1 gin 2 ml of 2 M hydrochloric acid Parenteral Preparations without afitrther procedure for the
gives the reaction ofprimary aromatic amines (2.3.1). removal ofbacterial endotoxins complies with the following
additional requirement.
Tests
Bacterial endotoxins (2.2.3). Not more than 0.1 0 Endotoxin
pH (2.4.24).5.0 to 7.5, determined in a solution prepared by
Unit per mg.
shaking 50 mg in sufficient carbon dioxide-free water to
produce 15 ml until dissolution is complete. Procaine Penicillin intended for use in the manufacture of
Specific optical rotation (2.4.22). +165° to +180°, determined Parenteral Preparations without a further sterilisation
in a solution prepared by dissolving 0.25 g in sufficient of a procedure complies with the follOWing additional
mixture of 3 volumes of acetone and 2 volumes of water to requirement.
produce 25 ml. Sterility. Complies with the test for sterility (2.2.11).

1970

www.webofpharma.com
IP 2010 FORTIFIED PROCAINE PENICILLIN INJECTION

Storage. Store protected from moisture. If the material is B. In the Assay, the principal peak in the chromatogram
intended for use in the manufaGture ofparenteral preparations, obtained with the test solution corresponds to the peak in the
the container should be sterile and sealed so as to exclude chromatogram obtained with the reference solution.
micro-organisms.
C. Give the reaction of primary aromatic amines (2.3.1),
Labelling. The label states (1) where applicable, that the producing a bright, orange-red precipitate.
contents are free from bacterial endotoxins; (2) where
applicable, that the contents are sterile. Tests
Water (2.3.43). Not more than 3.5 per cent, determined on
0.5g.
Assay. Determine by liquid chromatography (2.4.14).
Fortified Procaine Penicillin Injection Test solution. Weigh accurately a quantity of the mixed
Procaine Penicillin with Benzylpenicillin Injection contents of 10 containers equivalent to about 70 mg of
procaine penicillin and dissolve in 30 ml of the mobile phase
Fortified Procaine Penicillin Injection is a sterile mixture offive with the aid of ultrasound for about 2 minutes. Dilute to
parts of Procaine Penicillin and one part of Benzylpenicillin 50.0 ml with the mobile phase.
Potassium or Benzylpenicillin Sodium, together with suitable
dispersing and buffering agents. It is filled in a sealed container. Reference solution (a). Weigh accurately about 56 mg of
benzylpenicillin potassium RS and 40 mg of procaine
The injection is constituted by suspending the contents of hydrochloride RS and dissolve in 25 ml of the mobile phase
the sealed container in the requisite amount of sterile Water with the aid of ultrasound for about 2 minutes. Dilute to
for Injections, immediately before use. 50.0 ml with the mobile phase.
Storage. The constituted injection should be used within Reference solution (b). Mix 1 volume of a 0.24 per cent w/v
24 hours (4 days ifa buffering agent is present) ofpreparation solution of phenoxymethylpenicillin potassium RS in the
when stored at a temperature not exceeding 20° or within mobile phase with 3 volumes ofreference solution (a).
7 days (14 days if a buffering agent is present) when stored at
Chromatographic system
a temperature between 2° and 8°.
a stainless steel column 30 cm x 4 mm, packed with
Fortified Procaine Penicillin Injection contains a quantity of octadecylsilane bonded to porous silica (10 Jlm),
total penicillins calculated as CI6H17NzNa04S and equivalent mobile phase: a mixture of 50 volumes of a solution
to not less than 60.0 per cent and not more than 74.0 per cent containing 1.4 per cent w/v of potassium dihydrogen
ofthe stated amounts ofprocaine penicillin and benzylpenicillin orthophosphate and 0.65 per cent w/v tetrabutyl
potassium or benzylpenicillin sodium and a quantity of ammonium hydroxide adjusted to pH 7.0 with 1 M
procaine, CI3HzoNzOz, equivalent to not less than 36.0 per cent potassium hydroxide or orthophosphoric acid,
and not more than 44.0 per cent of the stated amount of 25 volumes of acetonitrile and 25 volumes of water,
procaine penicillin. flow rate. 1 ml per minute,
spectrophotometer set at 235 nm,
Usual strengths. Procaine Penicillin, 300 mg (300,000 Units)
injection volume. 20 Jll.
and Benzylpenicillin Potassium or Benzylpenicillin Sodium 60
mg (100,000 Units), Procaine Penicillin 3 g (3,000,000 Units) Inject reference solution (b). The resolution between
and Benzylpenicillin Potassium or Benzylpenicillin Sodium, benzylpenicillin potassium and phenoxymethylpenicillin
600 mg (1,000,000 Units). potassium is not less than 2. The relative retention time of
procaine with reference to benzylpenicillin is about 2.2.
Description. A white or almost white powder.
Inject the test solution and reference solution (a). Record the
The contents of the sealed container comply with the tests peak responses of the main peaks of benzyl penicillin and
stated under Parenteral Preparations (Powders for procaine. Calculate the total penicillin content as
Injection) and with the following requirements. benzylpenicillin, CI6HISNz04S and the content of procaine,
CI3HzoNzOz using the factor of0.866 for converting the content
Identification of procaine hydrochloride to that of procaine.
A. Dissolve 10 mg in 10 ml of water and add 0.5 ml of neutral Labelling. The label states (1) the quantities of Procaine
red solution. Add sufficient 0.01 M sodium hydroxide to give Penicillin and Benzylpenicillin Potassium or Benzylpenicillin
a permanent orange colour and then add 1 mlofpenicillinase Sodium contained in it; (2) the names ofany added dispersing
solution; a red colour is produced rapidly. and buffering agents; (3) "for intramuscular injection only".

1971

www.webofpharma.com
PROCHLORPERAZINE MALEATE IP 2010

Prochlorperazine Maleate Apply 4 f.!l of each solution.


D. Triturate 0.2 g with a mixture oB ml of water and 1 m1 of10
M sodium hydroxide and shake with three quantities, each of
5 ml, of ether. To 0.1 ml ofthe aqueous layer add a solution of
,[(COOH] 10 mg of resorcinol in 3 ml of sulphuric acid and heat on a
water-bath for 15 minutes; no colour develops. To the
COOH remainder ofthe aqueous layer add 2 ml of bromine solution,
2 heat on a water-bath for 15 minutes, then heat to boiling and
cool. To 0.1 ml of the solution add a solution of 10 mg of
resorcinol in 3 ml of sulphuric acid and heat on a water-bath
C2oH24CIN3S,2CJ!404 Mol. Wt. 606.1 for 15 minutes; a blue colour develops.
Prochlorperazine Maleate is 2-chloro-1 0-[3-(4-
methylpiperazin-I-yl)propyl]phenothiazine di(hydrogen
Tests
maleate). pH (2.4.24). 3.0 to 4.0, determined in a freshly prepared saturated
Prochlorperazine Maleate contains not less than 98.0 per cent solution.
and not more than 101.0 per cent of C2oH24CIN3S, 2C 4H40 4, Related substances. Complies with the test for Related
calculated on the dried basis. substances in Phenothiazines (2.3.5), using mobile phase A.
Category. Antipsychotic; antiemetic. NOTE- Prepare the test solution immediately before use.
Dose. As antipsychotic, 15 to 100 mg daily, in divided doses; Sulphated ash (2.3.18). Not more than 0.1 percent.
as antiemetic, 10 to 30 mg daily, in divided doses. Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Description. A white or pale yellow, crystalline powder; on 1.0 g by drying in an oven at 105°.
practically odourless. Assay. Weigh accurately about 0.2 g, in powder, dissolve in
50 ml of anhydrous glacial acetic acid, heating gently on a
Identification water-bath, allow to cool. Titrate with 0.1 M perchloric acid,
Test A may be omitted iftests B, C andD are carried out. Tests determining the end-point potentiometrically (2.4.25). Carry .
B, C and D may be omitted if test A is carried out. out a blank titration.
A.To 20 mg add 5 ml of water and I m1 of 1 M sodium hydoxide, I ml of 0.1 M perchloric acid is equivalent to 0.03031 g of
shake and extract with 10 ml of ether. Wash the ether extract ~OH24CIN3S,2CJ!404.
with 5 ml of water, dry with anhydrous sodium sulphate, Storage. Store protected from light and moisture.
evaporate to dryness and dissolve the residue in 0.2 ml of
chloroform.
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with
Prochlorperazine Tablets
prochlOlperazine maleate RS or with the reference spectrum Prochlorperazine Maleate Tablets
ofprochlorperazine.
Prochlorperazine Tablets contain not less than 92.5 per cent
B. When examined in the range 230 nm to 360 nm (2.4.7), a and not more than 107.5 per cent of the stated amount of
0.001 per cent w/v solution in ethanol (95 per cent) containing prochlorperazine maleate, C2oH24CIN3S, 2C4~04'
0.01 per cent v/vof strong ammonia solution shows an
Usual strengths. 5 mg; 25 mg.
absorption maximum at about 258 nm and a less well-defined
maximum at about 313 nm; absorbance at about 258 nm, about Identification
0.6.
A.To a quantity of the powdered tablets containing 40 mg of
C. Complies with the test for identification ofphenothiazines Prochlorperazine Maleate add 10 ml of water and 2 ml of 1 M
(2.3.3). sodium hydroxide, shake and extract with 15 ml of ether. Wash
Test solution. A solution containing 0.1 per cent w/v of the the ether with 5 ml of water, dry with anhydrous sodium
substance under examination in a mixture ofequal volumes of sulphate, evaporate to dryness and dissolve the residue in
methanol and chloroform. 0.4 ml of chloroform.
Reference solution. A 0.1 per cent w/v solution of Detelmine by infrared absorption spectrophotometry (2.4.6).
prochlorperazine maleate RS in the same solvent mixture. Compare the spectrum with that obtained with

1972

www.webofpharma.com
IP 2010 PROCHLORPERAZlNE MESYLATE

prochlorperazine maleate RS or with the reference spectrum ethanol, dilute 10.0 ml ofthis solution to 50.0 ml with ethanol
ofprochlorperazine. and measure the absorbance of the resulting solution at the
maximum at about 258 nm (2.4.7). Calculate the content of
B. To a quantity of the powdered tablets containing 5 mg of
CZOHZ4ClN3S, 2C4H40 4 taking 620 as the specific absorbance at
Prochlorperazine Maleate add 5 ml ofsulphuric acid and allow
258nm.
to stand for 5 minutes; a red colour is produced.
Storage. Store protected from light and moisture.
C. Shake a quantity of the powdered tablets containing 0.2 g
ofProchlorperazine Maleate with 2 ml of water and 1 ml of5 M
sodium hydroxide, mix and extract with three quantities, each
of 10 ml, of ether. Dry the combined extracts with anhydrous
sodium sulphate, filter, evaporate the filtrate to dryness and
Prochlorperazine Mesylate
dissolve the residue in 10 ml of methanol and add a solution
of 0.15 g ofpicric acid in 10 ml of methanol. The precipitate,
after washing with a small quantity of methanol, melts at about
255°, with decomposition (2.4.21).

Tests
Related substances. Comply with the test for Related,
substances in Phenothiazines (2.3.5), using mobile phase A.
NOTE- Prepare the following solutions freshly. Mol. Wt. 566.2
Test solution. Extract a quantity of the powdered tablets Prochlorperazine Mesylate is 2-chloro-1 0-[3-(4-
containing 0.1 g of Prochl.orperazine Maleate with 10 ml of methylpiperazin-1-yl)propyl]phenothiazine
methanol containing 0.5 per cent v/v of strong ammonia di(methanesulphonate).
solution and filter. Prochlorperazine Mesylate contains not less than 98.0 per
Reference solution. Dilute 1.0 ml of the test solution to cent and not more than 101.0 per cent ofCzoHz4ClN3S,2CH4S03,
200.0 ml with the same solvent. calculated on the dried basis.
Apply to the plate 20 JlI of each solution. Category. Antipsychotic; antiemetic.
Uniformity ofcontent. (For tablets containing 10 mg or less) Dose. As antipsychotic, by intramuscular injection, 12.5 to 25
- Comply with the test stated under Tablets. mg; twice or thrice daily; as antiemetic, by intramuscular
NOTE- Protect the solutions from light throughout the injection, 12.5 mg.
Assay. Description. A white or almost white powder; odourless.
Crush one tablet and extract with three quantities, each of
Identification
10 ml, of ethanol containing 1 per cent v/v of strong ammonia
solution. Filter the extracts and to the combined filtrates add A. Determine by infrared absorption spectrophotometry (2.4.6).
sufficient ethanol to produce 50.0 ml. Dilute 10.0 ml of this Compare the spectrum with that obtained with
solution to 100.0 ml with ethanol. Dilute further with ethanol, prochlOlperazine mesylate RS or with the reference spectrum
if necessary, to give a final solution containing 10 Jlg of ofprochlorperazine mesylate.
Prochlorperazine Maleate per ml and measure the absorbance
B. When examined in the range 230 run to 360 nm (2.4.7), a
ofthe resulting solution at the maximum at about 258 nm (2.4.7).
0.001 per cent w/v solution in ethanol containing 0.01 per
Calculate the content of CZOHZ4ClN3S, 2C4H40 4 taking 620 as
cent v/v of strong ammonia solution shows an absorption
the specific absorbance at 258 nm.
maximum at about258 nm and a less well-defined maximum at
Other tests. Comply with the tests stated under Tablets. about 313 nm; absorbance at about 258 nm, about 0.6.
Assay. Protect the solutions from light throughout the Assay. C. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand
Weigh and powder 20 tablets. Weigh accurately a quantity of for 5 minutes; a red colour is produced.
the powder containing about 25 mg of Prochlorperazine D. Mix 50 mg with 0.2 g ofpowdered sodium hydroxide, heat
Maleate and extract with three quantities, each of 10 ml, of to fusion and continue the heating for a few seconds longer.
ethanol containing 1 per cent v/v of strong ammonia solution. Cool, add 0.5 ml of water and a slight excess of 2 M
Filter the extracts and to the combined filtrates add sufficient hydrochloric acid and warm; sulphur dioxide is evolved which
ethanol to produce 100.0 ml. Dilute 10.0 ml to 50.0 ml with turns moistened starch iodate paper blue.

1973

www.webofpharma.com
PROCHLORPERAZINE MESYLATE IP 2010

Tests B. To a volume containing 5 mg ofProchlorperazine Mesylate


add carefully 2 ml of sulphuric acid and allow to stand for
pH (2.4.24). 2.0 to 3.0, determined in a 2.0 per cent w/v solution. 5 minutes; a red colour is produced.
Related substances. Complies with the test for Related
Tests
substances in Phenothiazines (2.3.5), using mobile phase A.
pH (2.4.24).5.5 to 6.5.
Test solution. Dissolve the substance under examination in
methanol containing 0.5 per cent v/v of strong ammonia Related substances. Carry out the test for Related substances
solution. in Phenothiazines (2.3.5), using mobile phase A.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Test solution. Use the injection under examination.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined Reference solution (a). Dilute 1.0 ml of the test solution to
on 1.0 g by drying in an oven at 100° at a pressure not exceeding 40 ml with methanol containing 0.5 per cent v/v of strong
0.7kPa. ammonia solution immediately before use.
Assay. Weigh accurately about 0.5 g, dissolve in 10 ml of Reference solution (b). Dilute 1.0 ml of the test solution to
water, add 5 ml of 1 M sodium hydroxide and extract with 200.0 ml with the methanol-ammonia mixture immediately
successive quantities of 50, 25, 25 and 25 ml of ether. Wash before use.
the combined ether extracts with 5 ml of water, shake the "Any secondary spot in the chromatogram obtained with the
washings with 5 ml of ether, add the ether to the combined test solution is not more intense than the spot in the
ether extracts and evaporate to dryness. Add 2 ml of ethanol, chromatogram obtained with reference solution (a) and not
evaporate to dryness. Titrate with 0.1 M perchloric acid, more than one such spot is more intense than the spot in the
using 1-naphtholbenzein solution as indicator. Carry out a chromatogram obtained with reference solution (b).
blank tih·ation.
Other tests. Complies with the tests stated under Parenteral
I ml of 0.1 M perchloric acid is equivalent to 0.02831 g of Preparations (Injections).
C2oH24ClN3S,2CH4S03.
Assay. Protect the solutions from light throughout the Assay.
Storage. Store protected from light and moisture.
To an accurately measured volume containing about 25 mg of
Prochlorperazine Mesylate add sufficient ethanol containing
0.01 per cent v/v of strong ammonia solution to produce
Prochlorperazine Injection 200.0 ml. Dilute 5.0 ml of this solution to 100.0 ml with the
ammoniacal ethanol and measure the absorbance of the
Prochlorperazine Mesylate Injection resulting solution at the maximum at about 258 nm (2.4.7).
Prochlorperazine Injection is a sterile solution of Calculate the content ofC2oH24ClN3S, 2CH4S03taking 635 as
Prochlorperazine Mesylate in Water for Injections free from the specific absorbance at 258 nm.
dissolved air. Storage. Store protected from light.
Prochlorperazine Injection contains not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
prochlorperazine mesylate, C2oH24ClN3S,2CH4S03'
Usual strength. 12.5 mg perml. Procyclidine Hydrochloride
Identification.
A. To a volume containing 0.1 g ofProchlorperazine Mesylate
add 20 ml of water and 2 ml of 10Msodium hydroxide. Shake
and extract with 25 ml of ether. Wash the ether layer with two , Hel
quantities, each of5 ml, of water, dry with anhydrous sodium
sulphate and evaporate to dryness. l)issolve the residue in
1 ml of chloroform.
o
Detelmine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with C I9H29NO,HCI Mol. wt. 323.9
prochlorperazine mesylate RS h'eated in the same manner or Procyclidine Hydrochloride is (RS)-I-cyclohexy1-1-phenyl-3-
with the reference spectrum ofprochlorperazine. pyrrolidin-1-ylpropan-1-01 hydrochloride.

1974

www.webofpharma.com
IP 2010 PROCYCLIDINE TABLETS

Procyclidine Hydrochloride contains not less than 99.0 per Test solution. Add 5 ml of 1.25 M sodium hydroxide to 20 ml
cent and not more than 101.0 per cent ofC I9 H29NO,HCI, of a 0.015 per cent w/v solution of the substance under
calculated on the dried basis. examination and mix. Extract with two quantities, each of
20 ml, of ether, add to the combined extracts 5 ml ofa 0.06 per
Category. Antiparkinsonian.
cent w/v solution of triphenylethylene (internal standard) in
Dose. Initially, 2.5 mg thrice daily, after meals increasing at ether, shake with anhydrous sodium sulphate and filter.
intervals oftwo to three days by 2.5 to 5 mg daily; maximum, Evaporate the filtrate and dissolve the residue in 1 ml of ether.
30 mg daily.
Reference solution (a). Prepare in the same manner as the test
Description. A white, crystalline powder, odourless or almost solution but using 20 ml of a 0.5 per cent w/v solution of the
odourless. substance under examination and omitting the addition ofthe
internal standard solution.
Identification
Reference solution (b). Prepare in the same manner as the test
A. Determine by infrared absorption spectrophotometry (2.4.6). solution but using 20 ml of a 0.5 per cent w/v solution of the
Compare the spectrum with that obtained with procyclidine substance under examination.
hydrochloride RS. Chromatographic system
B. Dissolve 0.25 gin 10 ml of water, make allcaline with 5 M a glass column 1.5 m x 4 mm, packed with acid-washed,
ammonia and extract with three quantities, each of 10 ml, of silanised diatomaceous support (such as HP
ether. Dry the combined ethereal extracts over anhydrous chromosorb W) coated with 10 per cent w/w ofmodified
sodium sulphate, filter, remove the ether and scratch the polyethylene glycol 20M (such as SP-1000) and 2 per
residue with a glass rod to induce solidification. The residue cent w/w of potassium hydroxide,
melts at about 85° (2.4.21). temperature:
column. 240°,
C. Gives the reactions ofchlorides (2.3.1).
inlet port and detector at 240 0 ,
Tests - flow rate. 30 ml per minute ofthe carrier gas.

pH (2.4.24). 4.5 to 6.5, determined in a 1.0 per centw/v solution. The ratio of the sum of the areas of any secondary peaks to
the area of the peak due to the internal standard in the
Related substances. A. Determine by thin-layer chromatogram obtaihed with reference solution (b) is not more
chromatography (2.4.17), coating the plate with silica gel than the ratio ofthe area ofthe principal peak to the area ofthe
GF254. internal standard peak in the chromatogram obtained with the
Mobile phase. A mixture oflOO volumes of ether and 1volume test solution.
of strong ammonia solution. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Test solution. Dissolve 0.2 g of the substance under Loss on drying (2.4.19). Not more than 0.5 per cent, determined
examination in 10 ml of chloroform. on 1.0 g by drying in an oven at 105°.
Reference solution (a). A 0.004 per cent w/v solution of Assay. Weigh accurately about 0.7 g, dissolve in 75 ml of
1-phenyl-3-pyrrolidinopropan-1-one hydrochloride RS in anhydrous glacial acetic acid, warm if necessary to effect
chloroform. solution and cool. Add 10 ml of mercuric acetate solution.
Reference solution (b). A 0.01 per cent w/v solution of the Titrate with 0.1 M perchloric acid, using crystal violet
substance under examination in chloroform. solution as indicator. Carry out a blank titration.

Apply to the plate 5 III of each solution. After development, 1 ml of 0.1 M perchloric acid is equivalent to 0.03239 g of
dry the plate at 105° for 15 minutes and examine in ultraviolet C I9H29NO,HCI.
light at 254 nm. Any spot corresponding to 1-phenyl-3- Storage. Store protected from moisture.
pyrrolidinopropan-l-hydrochloride-l-one in the chromatogram
obtained with the test solution is not more intense than the
spot in the chromatogram obtained with reference solution
(a). Spray the plate with dilute potassium iodobismuthate
Procyclidine Tablets
solution. Any secondary spot in the chromatogram obtained Procyc1idine Hydrochloride Tablets
with the test solution is not more intense than the spot in the
Procyclidine Tablets contain not less than 90.0 per cent and
chromatogram obtained with reference solution (b).
not more than 110.0 per cent of the stated amount of
B. Determine by gas chromatography (2A.13). procyclidine hydrochloride, C I9H 29NO,HCl.

1975

www.webofpharma.com
PROCYCLIDINE TABLETS IP 2010

Usual strengths. 2.5 mg; 5 mg. to settle. Transfer 5.0 ml of the supernatant solution to a
separating funnel, extract with 20.0 ml of chloroform and filter
Identification the extract, discarding the first 5 mlofthe filtrate. Measure the
A. Dissolve a quantity of the powdered tablets containing absorbance of the filtrate at the maximum at about 405 nm
about 25 mg ofProcyclidine Hydrochloride in 10 ml of water, (2.4.7), using as the blank a solution prepared by treating
shake with 20 ml of ether and discard the ether layer. Make the 0.5 ml of water and 4.5 ml ofa 0.025 per cent w/v solution of
aqueous layer alkaline with 2 M sodium hydroxide and extract bromocresol pUiple in the same manner beginning at the
with two quantities, each of20 ml, ofether. Wash the combined words "extract with 20 ml of chloroform ".
ether extracts with two quantities, each of 10 ml, of water, dry Calculate the content of C 19Hz9NO.HCI from the absorbance
by shaking with anhydrous sodium sulphate, filter and obtained by repeating the operation using procyclidine
evaporate the filtrate to dryness. If necessary, induce hydrochloride RS in place of the powdered tablets.
crystallization by scratching with a glass rod.
Storage. Store protected from moisture.
On the residue, determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with procyclidine hydrochloride RS.
B. The powdered tablets give the reactions ofchlorides (2.3.1). Progesterone Injectable Suspension
Tests Progesterone Injectable Suspension is a sterile suspension of
Progesterone in Water for Injections.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Progesterone Injectable Suspension contains not less than
93.0 per cent and not more than 107.0 per cent of the stated
Mobile phase. A mixture ofl 00 volumes of ether and 1volume
amount of progesterone, CZIH300Z'
of strong ammonia solution.
Usual Strengths. 25 mg per ml; 50 mg per ml; 100 mg per ml;
Test solution. Shake a quantity of the powdered tablets
200 mg per m!.
containing 25 mg ofProcyclidine Hydrochloride with 5 ml of
chloroform and filter. Identification
Reference solution (a). A 0.001 per cent w/v solution of
A. Filter a volume of suspension containing about 100 mg of
I-phenyl-3-pyrrolidinopropan-l-one hydrochloride RS in
progesterone, through a medium-porosity, sintered-glass
chloroform.
crucible, filter again ifthe fluid is not clear. Wash with several
Reference solution (b). Dilute 1 volume of test solution to 5.0 ml portions of water, evaporate on a steam bath, dry the
200 volumes with chloroform. residue at 105°. On the residue, determine by infrared absorption
Apply to the plate 20 /!l of each solution. After development, spectrophotometry (2.4.6). Compare the spectrum with that
dry the plate at 105° for 15 minutes and examine in ultraviolet obtained with progesterone RS or with the reference spectrum
light at 254 nm. Any spot corresponding to I-phenyl-3- of progesterone.
pyrrolidinopropan-l-one in the chromatogram obtained with B. Melting point (2.4.21). 126° to 131°.
the test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a) (0.2 per Tests
cent). Spray the plate with dilute potassium iodobismuthate
solution. Any secondary spot in the chromatogram obtained pH (2.4.24). 4.0 to 7.5.
with the test solution is not more intense than the spot in the Other tests. Complies with the tests stated under Parenteral
chromatogram obtained with reference solution (b) (0.5 per Preparations (Injections).
cent). Ignore any spot due to excipients on the line of
Assay. Detennine by liquid chromatography (2.4.14).
application.
Solvent mixture. 85 volumes of ethanol (95 per cent) and 15
Other tests. Comply with the tests stated under Tablets.
volumes of water.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Internal standard solution. A 0.66 per cent w/v solution of
quantity ofthe powder containing about 2.5 mg ofProcyclidine
methyltestosterone RS in the solvent mixture.
Hydrochloride, transfer to a 100-ml volumetric: flask, add
10.0 ml of water and mix, and dilute to volume with a 0.025 per Test solutiO/i. Shake aVolume Ofsl.lspellsioll cOlltairiingabollt
cent w/v solution of bromocresol purple in 2 per cent v/v 25 mg ofprogesterone with 16 ml ofthe solvent mixture into a
solution ofglacial acetic acid. Allow the undissolved particles polytef-lined, screw-capped, 25 ml test tube until the solution

1976

www.webofpharma.com
IP 2010 PROGUANIL TABLETS

is clear. Add 2.0 ml ofthe internal standard solution and dilute A. Detelmine by infrared absorption spectrophotometry (2.4.6).
to 25 ml with the solvent mixture. Compare the spectrum with that obtained with proguanil
Reference solution. A 0.25 per cent w/v solution of hydrochloride RS or with the reference spectrum ofproguanil
progesterone RS in the solvent mixture. To 4.0 ml of this hydrochloride.
solution, add 2.0 ml ofthe internal standard solution and dilute B. To 10 ml of a saturated solution add 0.25 ml of potassium
to 10 ml with the solvent mixture. ferrocyanide solution; a white precipitate is produced which
dissolves on addition of a few drops of dilute nitric acid.
Chromatographic system
- a stainless steel column 100 cm x 2.1 mm, packed with C. Dissolve 5 mg in 5 ml ofa wann 1.0 percentw/v solution of
octadecylsilane chemically bonded to porous silica (30- cetrimide and add 1 ml of 5 M sodium hydroxide and 1 ml of
50llm), bromine solution; a deep red colour is produced.
- mobile phase: a mixture ofn volumes of water and 28 D. Gives the reactions ofchlorides (2.3.1).
volumes of isopropyl alcohol,
- flow rate. 1 ml per minute, Tests
- spectrophotometer set at 254 nm,
Acidity or alkalinity. To 35 ml of water maintained at 60° to
- injection volume. 5 Ill.
65° add 0.2 ml of methyl red solution, neutralise with 0.01 M
Inject the reference solution. The test is not valid unless the sodium hydroxide or 0.01 M hydrochloric acid, add 0.4 g of
resolution between the peaks due to progesterone and the substance under examination and stir until dissolved. The
methyltestosterone is not less than 3.5 and the relative resulting solution is not acidic and requires for neutralisation
standard deviation for replicate injections is not more than 1.5 not more than 0.2 ml of 0.01 M hydrochloric acid.
per cent.
4-Chloroaniline. Dissolve 0.1 g in 1 ml of 2 M hydrochloric
Inject the test solution and the reference solution. acid, add sufficient water to produce 20 ml, cool to 5°, add 1
ml of 0.05 M sodium nitrite, allow to stand at 5°for 5 minutes,
Calculate the content of CZIH300z in the suspension.
add 2 ml ofa 5 per cent w/v solution of ammonium sulphamate
Storage. Store in single dose or multiple dose containers, and allow to stand for 10 minutes. Add 2 ml of a 0.1 per cent
preferably of Type 1 glass. w/v solution of N-(l-naphthyl)ethylenediamine
dihydrochloride, dilute to 50 ml with water and allow to stand
for 30 minutes. Any magenta colour produced is not more
intense than that obtained by treating in the same manner and
Proguanil Hydrochloride at the same time 20 ml of a solution containing 1.25 Ilg of
Chloroguanide Hydrochloride 4-chloroaniline.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
on 1.0 g by drying in an oven at 105°.
Assay. Weigh accurately about 0.15 g, dissolve in 25 ml of
anhydrous glacial acetic acid and 10 ml of mercuric acetate
CjlHj6CINs, HCl Mol. Wt. 290.2 solution. Titrate with 0.1 M perchloric acid, determining the
end-point potentiometrically (2.4.25). Carry out a blank titration.
Proguanil Hydrochloride is 1-(4-chlorophenyl)-5-
isopropylbiguanide hydrochloride. I ml of 0.1 M perchloric acid is equivalent to 0.01451 g of
CjlH16ClNS, HCI.
Proguanil Hydrochloride contains not less than 99.0 per cent
and not more than 101.0 per cent ofCjjH16ClNs, HCl, calculated Storage. Store protected from light and moisture.
on the dried basis.
Category. Antimalarial.
Proguanil Tablets
Dose. Suppressive, 100 to 300 mg daily.
Description. A white, crystalline powder; odourless.
Proguanil Hydrochloride Tablets; Chloroguanide
Hydrochloride Tablets
Identification
Proguanil Tablets contain not less than 95.0 per cent and not
Test A may be omitted iftests B, C andD are carried out. Tests more than 105.0 per cent of the stated amountofproguanil
Band C may be omitted if tests A and D are carried out. hydrochloride, CjjHj6ClNs, HCI.

1977

www.webofpharma.com
IP 2010
PROGUANIL TABLETS

Usual strength. 100 mg. Calculate the content ofCIIHI6ClNs,HCl from the absorbance
obtained by repeating the operation using 10 ml ofa 0.01 per
Identification cent w/v solution of proguanil hydrochloride RS beginning
A. Boil a quantity ofthe powdered tablets containing 0.5 g of at ~he words "add 70 ml ofwater,..,.".
Proguanil Hydrochloride with 5 ml of dilute hydrochloric acid, Storage. Store protected from light and moisture.
cool and filter. To the filtrate add a slight excess of sodium
hydroxide solution, extract with 30 ml of ether and evaporate
the ethereal extract; the residue, after drying at 105°, melts at
about 131° (2.4.21). Promazine Tablets
Dissolve the residue obtained in testA in the minimum quantity
Promazine Hydrochloride Tablets
of dilute hydrochloric acid; the solution diluted with water
to about 40 ml and neutralised if necessary, with cautious Promazine Tablets contain not less than 92.5 per cent and not
a4dition of dilute ammonia solution, complies with the more than 107.5 per cent of ~he stated amount of promazine
following tests. hydrochloride, CI7HzoNzS,HCl.
B. To 10 ml add 0.25 ml ofpotassiumferrocyanide solution; a Usual strengths. 25 mg; 50 mg; 100 mg.
white precipitate is produced which dissolves on addition of
a few drops of dilute nitric acid. Identification
C. To 0.5 ml add 5 ml of a warm 1.0 per cent w/v solution of A. To a quantity ofthe powdered tablets containing 40 mg of
cetrimide and add 1 ml of5 M sodium hydroxide and 1 ml of Promazine Hydrochloride, add 10 ml of water and 2 ml of1 M
bromine solution; a deep red colour is produced. sodium hydroxide, shake and extract with 15 ml of ether. Wash
the extract with 5 ml of water, dry with anhydrous sodium
Tests
sulphate, evaporate to dryness and dissolve the residue in
4-Chloroaniline. To a quantity of the powdered tablets 0.4 ml of chloroform. On the residue, determine by infrared
containing about 0.1 g ofProguanil Hydrochloride add 5 ml of absorption spectrophotometry (2.4.6). Compare the spectrum
ethanol (95 per cent) and shake for 10 minutes. Add 2.5 mlof with that obtained with promazine hydrochloride RS or with
2 M hydrochloric acid and 15 ml of water, mix and filter the reference spectrum ofpromazine hydrochloride.
through a wetted filter paper, washing the filter with 5 ml of B. To a quantity of the powdered tablets containing 5 mg of
water. Cool to 5°, add 1 ml of 0.05 M sodium nitrite, allow to Promazine Hydrochloride, add 5 ml ofsulphuric acid and allow
stand at 5° for 5 minutes, add 2 ml ofa 5 per cent w/v solution to stand for 5 minutes. An orange colour is produced.
of ammonium sulphamate and allow to stand for 10 minutes.
Add 2 ml of a 0.1 per cent w/v solution of C. Triturate a quantity of the powdered tablets containing 0.2
N-(l-naphthyl)ethylenediamine dihydrochloride, dilute to g ofPromazine Hydrochloride with 4 rnl of methanol, centrifuge,
50 ml with water and allow to stand for 30 minutes. Any filter the supernatant liquid, heat almost to boiling, immediately
magenta colour produced is not more intense than that add 2 ml ofa boiling 3.5 per cent w/v solution ofpicric acid in
obtained by treating in the same manner and at the same time methanol and boil for 2 minutes. Cool in ice, filter, wash the
20 ml of a solution containing 1.25 Ilg of 4-chloroaniline. crystals with three quantities of methanol, dissolve in 10 ml of
hot methanol and repeat the crystallisation and washing. The
Other tests. Comply with the tests stated under Tablets.
rust red crystals so obtained, after drying at 105° for 1 hour,
Assay. Weigh and powder 20 tablets. Weigh accurately a melts at about 144° (2.4.21).
quantity of the powder containing about 0.1 g of Proguanil
Hydrochloride, add 5 ml of water and warm on a water-bath Tests
with stirring until a smooth paste is obtained. Add 50 ml of
water, continue warming for 10 minutes, cool, add sufficient Related substances. Comply with the test for Related
water to produce 100.0 rnl and filter. Dilute 10.0 ml ofthe filtrate substances in Phenothiazines (2.3.5), using mobile phase (a)
to 100.0 ml with water and to 10.0 ml ofthe resulting solution and the following freshly prepared solutions.
add 70 ml of water, 5 ml of a 20 per cent w/v solution of Test solution. Extract a quantity of the powdered tablets
cetrimide and 1 ml of 2-propanol. Adjust the temperature of containing 0.1 g of Promazine Hydrochloride with 10 ml of
the solution to 20° and add 2 ml of alkaline sodium methanol and filter.
hypobromite solution and sufficient water to produce
Reference solution. Dilute 1 ml oftestsolutiont0200ml with
100.0 rnl. Allow to stand at 20° for 25 minutes and measure the
methanol.
absorbance ofthe resulting solution at the maximum at about
480 nm (2.4.7). Other tests. Comply with the tests stated under Tablets.

1978

www.webofpharma.com
IP 2010
PROMETHAZINE INJECTION

Assay. Heat the solution to boiling; it becomes orange and an orange-


NOTE-earlY out the test protectedfi-om light. red precipitate is produced.

Weigh and powder 20 tablets. Weigh accurately a quantity of D. Gives reaction B ofchlorides (2.3.1).
the powder containing about 50 mg of Promazine
Hydrochloride and triturate with 10 ml of 2 M hydrochloric
Tests
acid and add 200 ml of water. Shake for 15 minutes, add pH (2.4.24). 4.0 to 5.0, determined in a 10.0 per cent w/v solution
sufficient water to produce 500 ml and centrifuge about 50 ml prepared immediately before use. .
ofthe mixture. To 5 ml ofthe clear, supernatant liquid add 10 m1
of 0.1 M hydrochloric acid and sufficient water to produce Related substances. Carry out the test for Related substances
100 mi. Measure the absorbance (2.4.7) ofthe resulting solution in Phenothiazines (2.3.5), protected from bright light using
at the maximum at about 251 urn. mobile phase B.

Calculate the content of C 17H zoN zS,HCI taking 935 as the


Prepare the following solutions immediately before use.
specific absorbance at 251 urn. Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml ofa mixture of95 volumes of methanol
and 5 volumes of diethylamine.
Reference solution (aj. A 0.01 per cent w/v solution of the
Promethazine Hydrochloride substance under examination in the same solvent mixture.
Reference solution (bj. A 0.02 per cent w/v solution of
isopromethazine hydrochloride RS in the same solvent
mixture.
, Hel Apply to the plate 10 )11 of each solution.
Any spot corresponding to isopromethazine in the
chromatogram obtained with the test solution is not more
intense than the spot in the chromatogram obtained with
Mol. Wt. 320.9 reference solution (b) and any other secondary spot is not
more intense than the spot in the chromatogram obtained
Promethazine Hydrochloride is (RS)-dimethyl(2-
with reference solution (a).
phenothiazin-1 O-ylpropyl)amine hydrochloride.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Promethazine Hydrochloride contains not less than 98.5 per
cent and not more than 101.0 per cent of C 17H zoN zS, HCI, Loss on drying (2.4.19). Not more than 0.5 per cent, determined
calculated on the dried basis. on 1.0 g by drying in an oven at 105°.
Category. Antiemetic. Assay. Weigh accurately about 0.25 g, dissolve in a mixture of
5 ml of 0.01 M hydrochloric acid and 50 ml of ethanol (95 per
Dose. 20 to 50 mg daily, in single or divided doses.
cent) and titrate with 0.1 M sodium hydroxide, determining
Description. A white or faintly yellowish, crystalline powder. the end-point potentiometrically (2.4.25). Record the volume
added between the two inflections.
Identification
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03209 g of
Test A may be omitted iftests B, C andD are carried out. Tests C 17HzoNzS, HCI.
B, C and D may be omitted if test A is carried out. Storage. Store protected from light and moisture.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with promethazine
hydrochloride RS or with the reference spectrum of
promethazine hydrochloride.
Promethazine Injection
B. Complies with the test for Identification ofPhenothiazines
(2.3.3). Promethazine Hydrochloride Injection
C. Dissolve 0.1 gin 3 ml of water and add 1 ml of nitric acid Promethazine Injection is a sterile solution of Promethazine
dropwise; a precipitate is produced which dissolves rapidly Hydrochloride in Water for Injections free from dissolved air.
to give a red solution which becomes orange and then yellow. It may contain suitable stabilising agents.

1979

www.webofpharma.com
PROMETHAZINE INJECTION IP 2010

Promethazine Injection contains not less than 95.0 per cent chromatogram obtained with the test solution is not more
and not more than 105.0 per cent of the stated amount of intense than the spot in the chromatogramI obtained with
promethazine hydrochloride, C 17H2oN2S, HCl. reference solution (a) and not more than one such spot is
more intense than the spot in the chromatogram obtained
Usual strengths. 25 mg in 1 ml; 50 mg in 2 ml.
with reference solution (b).
Identification Other tests. Complies with the tests stated under Parenteral
A. To a volume containing 0.1 g ofPromethazine Hydrochloride Preparations (Injections).
add 20 ml of water and 2 ml of 10Msodium hydroxide. Shake Assay. Carry out the following procedure protected from
and extract the mixture with 25 ml of ether. Wash the ether light.
layer with two quantities, each of 5 ml, of water, dry with
anhydrous sodium sulphate and evaporate to dryness. To an accurately measured volume containing about 25 mg of
Dissolve the residue in 1 ml of chloroform. Promethazine Hydrochloride add sufficient 0.01 M
hydrochloric acid to produce 100.0 ml. Dilute 10.0 ml to
Determine by infrared absorption spectrophotometry (2.4.6). 100.0 ml with 0.01 Mhydrochloric acid, dilute 1O.0ml ofthis
Compare the spectrum with that obtained with promethazine solution to 50.0 ml with 0.01 M hydrochloric acid and measure
hydrochloride RS treated in the same manner or with the the absorbance of the resulting solution at the maximum at
reference spectrum ofpromethazine. about 249 nm (2.4.7). Calculate the content ofC 17H 2oN 2S, HCl
B. To a volume containing 0.2 g ofPromethazine Hydrochloride taking 910 as the specific absorbance at 249 om.
add sufficient potassium carbonate to saturate the solution, Storage. Store protected from light.
extract with two quantities, each of 10 ml, of ether and
evaporate the combined extracts to dryness. Dissolve the
residue in 2 ml of methanol and pour into a solution of0.4 g of
picric acid in 10 ml of methanol, previously warmed to about Promethazine Syrup
50°. Cool, scratch the sides ofthe tube to induce crystallisation,
Promethazine Hydrochloride Syrup; Promethazine
allow to stand for 3 to 4 hours and filter. The residue, after
washing with methanol and drying, melts at about 160° (2.4.21). Hydrochloride Oral Solution; Promethazine Oral Solution

C. To a volume containing 5 mg ofPromethazine HydrocWoride Promethazine Syrup is a solution containing Promethazine


add carefully 2 ml of sulphuric acid and allow to stand for Hydrochloride in a suitable flavoured vehicle.
5 minutes; a red colour is produced. Promethazine Syrup contains not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
Tests promethazine hydrochloride, C 17H 2oN 2S, HCl.
pH (2.4.24).5.0 to 6.0. Usual strength. 5 mg per 5 ml.
Related substances. Carry out the test for Related substances
in Phenothiazines (2.3.5), using mobile phase B. Identification
Test solution. Dilute a volume ofthe injection with a mixture of Carry out the method for identification of phenothiazines
95 volumes of methanol and 5 volumes of diethylamine to (2.3.3).
contain 1 per cent w/v of Promethazine Hydrochloride. Test solution. A solution prepared by diluting a suitable volume
Reference solution (a). Dilute 1 volume of the test solution to of the syrup with water to contain 0.08 per cent w/v of
40 volumes with the same solvent mixture. Promethazine Hydrochloride. Apply 5 f.11 to the plate.

Reference solution (b). Dilute 1 volume of the test solution to Tests


200 volumes with the same solvent mixture.
Other tests. Complies with the tests stated under Oral Liquids.
Reference solution (c). A 0.01 per cent w/v solution of
Assay. Carry out the following procedure protected from
isopromethazine hydrochloride RS in the same solvent
light.
mixture.
Weigh accurately a quantity containing about 10 mg of
Apply separately to the plate 10 f.1l of each solution.
Promethazine Hydrochloride, add 25 ml of water and 5 ml ofa
Any spot corresponding to isopromethazine in the 5 per cent w/v solution of sodium hydroxide. Extract the
chromatogram obtained with the test solution is not more mixture with two quantities, each of 50 ml, of chloroform,
intense than the spot in the chromatogram obtained with shaking vigorously for 1 minute each time, evaporate the
reference solution (c). Any other secondary spot in the combined extracts to dryness at .about 30° at a pressure of

1980

www.webofpharma.com
IP 2010 PROMETHAZINE TABLETS

2 kPa and dissolve the residue in sufficient 0.1 M hydrochloric C. To a quantity of the powdered tablets containing 5 mg of
acidto produce 50.0 ml (solution A). Dilute 10 ml ofsolution A Promethazine Hydrochloride add 5 ml of sulphuric acid and
to 50.0 ml with water (solution B). To a further 10 ml ofsolution allow to stand for 5 minutes; a red colour is produced.
A add 5 ml ofperoxyacetic acid solution, allow to stand for
10 minutes and add sufficient water to produce 50.0 m1 (solution Tests
C). Measure the absorbance of solution C at the maximum at Related substances. Carry out the test for Related substances
about 336 run (2.4.7), using solution B as the blank and measure in Phenothiazines (2.3.5), using mobile phase B.
the absorbance of solution B at the same wavelength using
Test solution. A solution freshly prepared by extracting a
water as the blank. Repeat the procedure using a 0.02 per cent
quantity of the powdered tablets containing 0.1 g of
w/v solution of promethazine hydrochloride RS in 0.1 M
Promethazine Hydrochloride with 10 ml of a mixture of
hydrochloric acid in place of solution A and beginning at the
95 volumes of methanol and 5 volumes of diethylamine and
words "Dilute 10 ml of......".
filtering.
Determine the weight per ml ofthe symp (2.4.29), and calculate
Reference solution (a). Dilute 1.0 ml of the test solution to
the content ofC 17HzoN zS, HCI, weight in volume. The result is
40 ml with the same solvent.
not valid unless the absorbance of solution B is less than
0.10. Reference solution (b). Dilute 1.0 ml of the test solution to
200.0 mlwith the same solvent.
Storage. Store protected from light and moisture.
Reference solution (c). A 0.01 per cent w/v solution of
isopromethazine hydrochloride RS in the same solvent.
Apply to the plate 10 III of each solution. Any spot
Promethazine Tablets corresponding to isopromethazine in the chromatogram
obtained with the test solution is not more intense than the
Promethazine Hydrochloride Tablets spot in the chromatogram obtained with reference solution
Promethazine Tablets contain not less than 92.5 per cent and (c). Any other secondary spot in the chromatogram obtained
not more than 107.5 per cent of the stated amount of with the test solution is not more intense than the spot in the
promethazine hydrochloride, C 17HzoN zS, HCl. The tablets are chromatogram obtained with reference solution (a) and not
coated. more than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b).
Usual strengths. 10 mg; 25 mg; 50 mg.
Uniformity of content. (For tablets containing 10 mg or less)
Identification - Comply with the test stated under Tablets.
Crush one tablet; add 1 ml of dilute hydrochloric acid and 30
A. To a quantity of the powdered tablets containing 40 mg of ml ofwater and shake for 15 minutes. Add sufficient water to
Promethazine Hydrochloride add 10 ml of water and 2 ml of produce 50.0 ml and centrifuge. Complete the procedure
1 M sodium hydroxide, shake and extract with 15 ml of ether. described in the Assay beginning at the words "To 5.0 ml of
Wash the ether extract with 5 ml ofwater, dry with anhydrous . the clear supernatant liquid....".
sodium sulphate, evaporate to dryness and dissolve the
residue in 0.4 ml ofchloroform. Other tests. Comply with the tests stated under Tablets.

Determine by infrared absorption spectrophotometry (2.4.6). Assay. earlY out the following procedure protected froin
Compare the spectrum with that obtained with promethazine light.
hydrochloride RS treated in. the same manner or with the Weigh and powder 20 tablets. Weigh accurately a quantity of
reference spectrum ofpromethazine. the powder containing about 50 mg of Promethazine
B. Dissolve a quantity of the powdered tablets containing Hydrochloride with 10 ml of 2 M hydrochloric acid and add
0.2 g of Promethazine Hydrochloride in 2 ml of water, filter, 200 ml ofwater. Shake for 15 minutes, add sufficient water to
add sufficient potassium carbonate to saturate the solution, produce 500.0 ml and centrifuge about 50 ml ofthe mixture. To
extract with two quantities, each of 10 ml, of ether and 5.0 ml of the clear supernatant liquid add 10 ml of 0.1 M
evaporate the combined extracts to dryness. Dissolve the hydrochloric acid and sufficient water to produce 100.0 ml.
residue in 2 ml ofmethanol and pour into a solution of0.4 g of Measure the absorbance of the resulting solution at the
picric acid in 10 ml of methanol, previously warmed to about maximum at about 249 nm (2.4.7). Calculate the content of
50°. Cool, scratch the sides ofthe tube to induce crystallisation, C 17HzoN zS, HCI taking 910 as the specific absorbance at
allow to stand for 3 to 4 hours and filter. The residue, after 249 run.
washing with methanol and drying, melts at about 160° (2.4.21). Storage. Store protected from light and moisture.

1981

www.webofpharma.com
PROMETHAZINE THEOCLATE IP 2010

Promethazine Theoclate E. Fuse 50 mg ofthe residue obtained in test D with 0.5 g of


anhydrous sodium carbonate, boil the residue with 5 ml of
water, acidify to litmus paper with nitric acid and filter. The
filtrate gives reaction A ofchlorides (2.3.1).

Tests
CWorides (2.3.12). Shake 1.5 g with 50 ml ofwater for 2 minutes
and filter. 25 ml of the filtrate complies with the limit test for
chlorides (330 ppm).
Mol. Wt. 499.0 Related substances. Carry out the test for Related substances
Promethazine Theoclate is the (RS)-dimethyl(2- in Phenothiazines (2.3.5), protected from bright light, using
phenothiazin-10-ylpropyl)amine salt of8- mobile phase B.
chlorotheophylline. NOTE-Prepare the following solutions immediately before
Promethazine Theoclate contains not less than 98.0 per cent use.
and not more than 101.0 per cent of C 17H zoN zS, C7H 7C1N40 z, Test solution. Dissolve 0.2 g of the substance under
calculated on the dried basis. examination in 10 ml of a mixture of95 volumes of methanol
Category. Antiemetic. and 5 volumes of diethylamine.
Dose. 25 to 50 mg daily, in single or divided doses. Reference solution (a). A 0.01 per cent w/v solution of the
substance under examination in the same solvent mixture.
Description. A white or almost white powder; odourless or
almost odourless. Reference solution (b). A 0.02 per cent w/v solution of
isopromethazine hydrochloride RS in the same solvent
Identification mixture.
Test A may be omitted if tests B, C, D and E are carried out. Apply to the plate 10 III of each solution. Any spot
Tests B, C and D may be omitted if tests A and E are carried corresponding to isopromethazine in the chromatogram
out. obtained with the test solution is not more intense than the
spot in the chromatogram obtained with reference solution
A. Shake 0.15 g with 2.5 ml of water, add 1 ml of5 M ammonia
(b) and any other secondary spot is not more intense than the
and extract with 30 ml of ether. Wash the ether extract with
spot in the chromatogram obtained with reference solution (a).
10 ml of water, dry with anhydrous sodium sulphate and
evaporate to dryness. Dissolve the residue in 1 ml of Sulphated ash (2.3.18). Not more than 0.1 percent.
chloroform.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Determine by infrared absorption spectrophotometry (2.4.6). on 1.0 g by drying in an oven at 105°.
Compare the spectrum with that obtained with promethazine
Assay. Weigh accurately about 1.0 g and dissolve in 200 ml of
RS or with the reference spectrum ofpromethazine.
acetone. Titrate with 0.1 M perchloric acid, using 3 ml of a
B. When examined in the range 230 nrn to 360 nrn (2.4.7), a saturated solution of methyl orange in acetone as indicator.
0.0007 per cent w/v solution in ethanol containing 0.01 per Carry out a blank titration.
cent v/v of strong ammonia solution shows an absorption
maximum at about 255 nm; absorbance is a.bout 0.53. I ml of 0.1 M perchloric acid is equivalent to 0.04990 g of
C17HzoNzS,C7H7ClN40Z.
C. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand
for 5 minutes; a red colour is produced. Storage. Store protected from light and moisture.

D. Shake 0.4 g with 10 ml of water, add 4 rlil of5 M ammonia,


shake with two quantities, each oBO ml, of ether and add 4 ml
of hydrochloric acid to the aqueous solution; a white Promethazine Theoclate Tablets
precipitate is produced. Filter, wash with water and dry at
Promethazine Theoclate Tablets contain not less than 92.5 per
105°. Dissolve 10 mg of the residue in 1 ml of hydrochloric
cent and not more than 107.5 per cent of the stated amount of
acid, add 0.1 g of potassium chlorate and evaporate to
promethazine theoclate, C 17H zoN zS, C7H7ClN40 Z•
dryness; a reddish residue remains which becomes purple on
exposure to the vapour of ammonia. Usual strengths. 25 mg; 50 mg.

1982

www.webofpharma.com
IP 2010 PROPANE

Identification Reference solution (c). A 0.01 per cent w/v solution of


isopromethazine hydrochloride RS in the same solvent
A. To a quantity of the powdered tablets containing 40 mg of mixture.
Promethazine Theoclate add 10 ml of water and 2 ml of 1 M
sodium hydroxide, shake and extract with 15 ml of ether. Wash Apply to the plate 10 /ll of each solution. Any spot
the ether extract with 5 ml of water, dry with anhydrous sodium corresponding to isopromethazine in the chromatogram
sulphate, evaporate to dryness and dissolve the residue in obtained with the test solution is not more intense than the
0.4 ml of chloroform. spot in the chromatogram obtained with reference solution
(c). Any other secondary spot in the chromatogram obtained
Determine by infrared absorption spectrophotometry (2.4.6). with the test solution is not more intense than the spot in the
Compare the spectrum with that obtained with promethazine chromatogram obtained with reference solution (a) and not
theoclate RS treated in the same manner or with the reference more than one such spot is more intense than the spot in the
spectrum ofpromethazine. chromatogram obtained with reference solution (b).
B. Dissolve a quantity of the powdered tablets containing Other tests. Comply with the tests stated under Tablets.
0.2 g of Promethazine Theoclate in 2 ml of water, filter, add
Assay. CarlY out the following procedure protected from
sufficient potassium carbonate to saturate the solution, extract
light.
with two quantities, each of 10 ml, of ether and evaporate the
combined extracts to dryness. Dissolve the residue in 2 ml of Weigh and powder 20 tablets. Weigh accurately a quantity of
methanol and pour into a solution of 0.4 g of picric acid in the powder containing about 50 mg ofPromethazine Theoclate,
10 ml of methanol, previously warmed to about 50°. Cool, add 5 ml of water and 1 m1 of strong ammonia solution and
scratch the sides ofthe tube to induce crystallisation, allow to allow to stand for 5 minutes. Add 50 ml of ethanol, shake for
stand for 3 to 4 hours and filter. The residue, after washing 5 minutes and filter, washing the residue with five quantities,
with methanol and drying, melts at about 160° (2.4.21). each of 5 ml, of ethanol. Add sufficient ethanol to the filtrate
to produce 100.0 ml. Dilute 10.0 ml to 100.0 ml with ethanol
C. To a quantity of the powdered tablets containing 5 mg of
and dilute 10.0 ml of this solution to 100.0 ml with ethanol.
Promethazine Theoclate add 5 m1 of sulphuric acid and allow
Measure the absorbance of the resulting solution at the
to stand for 5 minutes; a red colour is produced.
maximum at about 255 om (2.4.7). Calculate the content of
D. Extract a quantity ofthe powdered tablets containing 0.2 g C 17H zoN zS, C7H 7ClN40 z taking 755 as the specific absorbance
of Promethazine Theoclate with chloroform, filter and at255 om.
evaporate the filtrate to dryness. Shake the residue with 10 ml
Storage. Store protected from light and moisture.
of water, add 4 ml of 5 M ammonia and extract with two
quantities, each of30 ml, of ether. Wash the combined extracts
with 10 ml of water. Combine the aqueous layer and washings,
add 4 ml of hydrochloric acid; a white precipitate is produced. Propane
Filter, wash the residue with water and dry at 105°. Dissolve
10 mg ofthe residue in 1 ml of hydrochloric acid, add 0.1 g of /"'--..
potassium chlorate and evaporate to dryness; a reddish H3C CH3
residue remains which becomes purple on exposure to the C3Hg Mol. Wt. 44.1
vapour ofammonia.
Propane is dimethylmethane.
Tests Propane contains not less than 98.0 per cent of C3H g•

Related substances. Carry out the test for related substances CAUTION-Propane is highly flammable and explosive.
in phenothiazines (2.3.5), using mobilephase B.
Identification
Test solution. A solution freshly prepared by extracting a
Determine by infrared absorption spectrophotometry (2.4.6),
quantity of the powdered tablets containing 0.1 g of
the solution give maxima at the wavelength of3.4 /lm, 6.8 /lm
Promethazine Theoclate with 10 ml ofa mixture of95 volumes
and7.2/lm.
of methanol and 5 volumes of diethylamine and filtering.
Reference solution (a). Dilute 1 volume ofthe test solution to Tests
40 volumes with the same solvent mixture.
Water(2.3.43). Not more than 0.001 per cent with the following
Reference solution (b). Dilute 1 volume ofthe test solution to modifications (a) provide the closed-system titrating vessel
200 volumes with the same solvent mixture. with an opening through which passes a coarse-porosity gas

1983

www.webofpharma.com
PROPANE IP 2010

dispersion tube connected to a sample cylinder; (b) dilute the prepared from crushed firebrick and calcined or burned
reagent with anhydrous methanol to give a water equivalence with a clay binder ab~ve 900 0 with subsequent acid-
factor ofbetween 0.2 and 1.0 mg per ml, age this diluted solution wash. It may be silanized),
for not less than 16 hours before sanitation; (c) obtain a 100 g a thermal-conductivity detector,
sample as directed under inhalation preparation, and introduce - flow rate. 50 ml per minute using nitrogen as carrier gas.
the sample into the titration vessel through the gas dispersion
Inject the test solution. The test is not valid unless"the relative
tube at a rate of about 100 ml of gas per minute.
standard deviation for replicate injections is not more than
High-boiling residues. Not more than 5 flg per ml, determined 1.0.
by the following method. Prepare a cooling coil from copper
Inject 2 fll of test solution.
tubing (about 6 mm outside diameter x about 6.1 m long) to fit
into a vacuum-jacketed flask. Immerse the cooling coil in a Calculate the· percentage purity by dividing 100 times the
mixture ofdry ice and acetone in a vacuum-jacketed flask, and propane response by the sum of all of the responses in the
connect one end of the tubing to the propellant sample chromatogram.
cylinder. Carefully open the sample cylinder valve, flush the Storage. Store protected from moisture and prevent exposure
cooling coil with about 50 ml of the propellant, and discard to excessive heat.
this portion of liquefied propellant. Continue delivering
liquefied propellant from the cooling coil, and collect it in a
previously chilled 1000-ml sedimentation cone until the cone
is filled to the 1OOO-mi mark. Allow the propellant to evaporate,
Propionic Acid
using a warm water bath maintained at about 400 to reduce
evaporating time. When all ofthe liquid has evaporated, rinse H3C~OH
the sedimentation cone with two 50-ml portions of pentane, o
and combine the rinsings in a tared 150-ml evaporating dish.
Transfer 100 ml ofthe pentane solvent to a second tared 150- C3H60 z Mol. Wt. 74.1
ml evaporating dish, place both evaporating dishes on a water Propionic Acid contains not less than 99.5 per cent and not
bath, evaporate to dryness, and heat the dishes in an oven at more than I00.5 per cent ofC3H60 z.
1000 for 60 minutes. Cool the dishes in a desiccator, and weigh.
Repeat the heating for IS-minute periods until successive Tests
weighings are within 0.1 mg, and calculate the weight of the Weight per ml (2.4.29). 0.988 to 0.993.
residue obtained from the propellant as the difference between
the weights of the residues in the two evaporating dishes. Distilling range (2.4.8). 138,SO to 142'so.

Acidity of residue. Add 10 ml of water to the residue obtained Heavy metals (2.3 .13). To the residue obtained in the test for
in the test for High boiling residues, mix by swirling for about Nonvolatile residue, .add 8 ml of 0.1 M hydrochloric acid,
30 seconds, add 2 drops of methyl orange solution, insert the warm gently until solution is complete, dilute with water to
stopper in the tube, and shake vigorously; no pink or red 100 ml. 10 ml ofthe solution complies with the limit test for
color appears in the aqueous layer. heavy metals, Method D (10 ppm).

Sulphur. Carefully open the container valve to produce a Limit of nonvolatile residue. Evaporate 20 g in a tared dish,
moderate flow of gas. Do not direct the gas stream toward the and dry at 105 0 for 1 hour, the weight ofthe residue does not
face, but deflect a portion of the stream toward the nose, the exceed 2 mg.
odour free from the characteristic odour of sulphur Readily oxidizable substances. Dissolve 15 g of sodium
compounds. hydroxide in 50 ml of water, cool, add 6 ml of bromine, stirring
Assay. Detennine by Gas chromatography (2.4.13). to effect complete solution, and dilute with water to 2000 ml.
Transfer 25.0 ml ofthis solution to a glass-stoppered, 250-ml
Test solution. Connect 1 Propane cylinder to the chromatograph conical flask containing 100 ml of water, and add 10 ml of20.0
through a suitable sampling valve and a flow control valve percentw/vsolution sodium acetate and 10.0mlofPropionic
downstream from the sampling valve. Flush the liquid sample Acid. Allow to stand for 15 minutes, add 5 ml of25.0 per cent
through the sampling valve, taking care to avoid entrapment w/v solution of potassium iodide and 10 ml of hydrochloric
of gas or air in the sampling valve. acid, and titrate with 0.1M sodium thiosulphate just to the
Chromatographic system disappearance ofthe brown color. Carry out a blank titration.
an aluminum column 6 m x 3 mm, packed with 10 per cent The difference between the volume of 0.1 M sodium
of liquid phase G30 ( Tetraethylene glycol dimethyle thiosulphate required for the blank and that required for the
ether) on non-acid-washed support SIC ( A support test solution is not more than 2.2 ml.

1984

www.webofpharma.com
IP 2010 PROPOFOL

Aldehydes. Transfer 10.0 ml to a glass-stoppered, 250-ml 2, 6-bis(l-methylethyl) benzene-1, 4-dione RS (propofol


conical flask containing 50 ml of water and 10.0 ml of 1.25 per impurity J RS) in 50 ml of hexane.
cent w/v solution of sodium bisulphite, insert the stopper,
Reference solution (b). Dilute 0.1 ml of propofol for peak
and shake vigorously. Allow the mixture to stand for 30
identification RS (containing 3,3' ,5,5' -tetrakis(1-
minutes, then titrate with 0.1 M iodine to the same brownish
methylethyl)biphenyl-4,4'-diol (propofol impurities E) and 2-
yellow endpoint obtained with a blank treated with the same
(1-methylethoxy)-1,3-bis(1-methylethyl)benzene (propofol
quantities of the same reagents. The difference between the
impurities G) to 1.0 ml with hexane.
volume of 0.1 M iodine required for the blank and that required
for the test solution is not more than 1.75 ml. Reference solution (c). Dilute 1.0 ml oftest solution (a) to 100
ml with hexane. Dilute 1.0 ml of this solution to 10 ml with
Assay. Weigh accurately about 1.5 g and dissolve with 100 ml
hexane.
ofrecently boiled and cooled water in a 250-ml conical flask,
and titrate with 0.5 M sodium hydroxide using dilute Reference solution (d). A 0.24 per cent w/v solution of
phenolphthalein solution as indicator , propofol RS in hexane.
1 ml of 0.5 M sodium hydroxide is equivalent to 0.03704 g of Chromatographic system
C3H60 Z• - a stainless steel column 20 cm x 4.6 rom, packed with
Storage. Store protected from moisture. silica gel (5Ilm),
- mobile phase: a mixture of 1 volume of anhydrous
ethanol, 7.5 volumes of acetonitrile and 990 volwnes
of hexane,
Propofol - flow rate. 2 ml per minute,
- spectrophotometer set at 275 nm,
- injection volume. 10 Ill.
Inject reference solution (a). The test is not valid unless the
resolution between the peaks due to propofol impurity J and
propofol is not less than 4.0. The relative retention times with
reference to propofol for propofol impurity G is about 0.5, for
Mol. Wt. 178.3
oxydibenzene (propofol impurity G) is about 0.6, for 2-(1-
Propofol is 2,6-diisopropylphenol. methylethenyl)-6-(1-methylethyl)phenol (propofol impurity B)
Propofol contains not less than 98.0 per cent and not more is about 0.7, for 4-hydroxy-3,5-bis(1-methylethyl)benzoic acid
than 102.0 per cent ofC1zHlsO. (propofol impurity F) is about 2.3, for 2,5-bis(1-
methylethyl)phenol (propofol impurity D) is about 2.5 , for 1-
Category. General anaesthetic. methylethy14-hydroxy-3,5-bis(1-methylethyl)benzoate
Description. A colourless or very light yellow, clear liquid. (propofol impurity P) is about 2.9, for 2,4-bis(1-
methylethyl)phenol (propofol impurity A) is about 3.0, for 2-
Identification (l-methylethyl)phenol (propofol impurity C) is about 3.4, for
propofol impurity E is about 4.0, for 3-(1-methylethyl)phenol
Determine by infrared absorption spectrophotometry (2.4.6). (propofol impurity F) is about 5.8 and for 4-(1-
Compare the spectrum with that obtained with propofol RS or methylethyl)phenol (propofol impurity H) is about 6.4. I

with the reference spectrum ofpropofol.


Inject test solution (a), reference solution (a), (b) and (c). Run
Tests the chromatogram 7 times the retention time of the principal
Refractive index (2.4.27). 1.5125 to 1.5145. peak. In the chromatogram obtained with test solution (a), the
area ofpeak due to propofol impurity G is not more than twice
Related substances. Determine by liquid chromatography the area of the principal peak in the chromatogram obtained
(2.4.14). with reference solution (c) (0.2 per cent); the area ofthe peak
Test solution (a). Dissolve 1.0 g of the substance under due to propofol impurity E is not more than 0.1 times the area
examination in 10 ml of hexane. of the principal peak in the chromatogram obtained with
reference solution (c) (0.01 per cent). The area of any
Test solution (b). Dissolve 0.24 g of the substance under
secondary peak is not more than 0.5 times the area of the
examination in 100 ml of hexane.
principal peak in the chromatogram obtained with reference
Reference solution (a). A solution containing 5 mg of the solution (c) (0.05 per cent). The sum of the areas of all the
substance under examination and 15 III containing 15 mg of secondary peaks is not more than 3 times the area of the

1985

www.webofpharma.com
PROPOFOL IP 2010

principal peak in the chromatogram obtained with reference Propofol Injection


solution (c) (0.3 per cent). Ignore any peak other than propofol
impurity E with an area less than 0.3 times the area of the Propofol Injection is a sterile emulsion ofPropofol in a suitable
principal peak in the chromatogram obtained with reference base.
solution (c) (0.03 per cent)~ Propofol Injection contains not less than 95.0 per cent and
Impurities J, K, Land O. Determine by gas chromatography not more than 105.0 per cent ofthe stated amount ofpropofol,
(2.4.13). C1zH1sO.
Test solution. Dissolve 40 mg of the substance under UsmH strength. 10 mg per ml.
examination in 10 ml of dichloromethane.
Identification·
Reference solution (a). Dilute 1.0 ml ofthe test solution to 100
ml with dichloromethane. Further dilute 1.0 ml ofthis solution A. Extract a volume ofthe injection containjng 0.1 g ofPropofol
to 10 ml with dichloromethane. with three 25 ml quantities of hexane and filter the combined
extracts using a glass fibre filter (such as Whatrnan GF/C).
Reference solution (b). Dilute 5 III containing 5 mg ofpropofol Extract the filtrate with two 20 ml quantities of methanol (90
impurity J RS to 100 ml with dichloromethane. Dilute 1.0 ml of per cent) and evaporate the combined extracts under reduced
this solution to 25 ml with dichloromethane. pressure. Dissolve the residue obtained in 2 ml of ethanol and
Reference solution (c). Dissolve 4 mg ofpropofol RS in 1.0 ml evaporate to dryness under reduced pressure. Dry the residue
of reference solution (b). at 50° over phosphorus pentoxide for 1 hour. On the residue,
determine by infrared absorption spectrophotometry (2.4.6).
Chromatographic system
Compare the spectrum with that obtained with propofol RS or
a fused-silica capillary column 30 m x 0.32 mm, coated
with the reference spectrum of propofol.
with polymethylphenylsiloxane (0.5 Ilm),
- temperature: B. In the Assay, the principal peak in the chromatogram
Column 80° from 0-3 minutes, 80°-120° from 3-25 minutes obtained with the test solution corresponds to the peak in the
and 210° from 25-40 minutes, chromatogram obtained with the reference solution.
Injection port at 100° and detector at 270°,
- a flame ionisation detector, Tests
- flow rate. 1.7 ml per minute using nitrogen as carrier gas. pH (2.4.24).6.0 to 8.5.
Inject 1 III ofreference solution (c). The test is not valid unless Propofol quinone and propofol dimer. Determine by liquid
the peak- to- valley ratio isnot lessthan3.0 where Hp is the chromatography (2.4.14).
height above the baseline ofthe peak due to propofol impurity
J and HI' is the height above the baseline ofthe lowest point of Test solution. Dilute a volume ofinjection containing 80 mg of
the curve separating this peak from the peak due to propofol. Propofol in 100 ml ofpropan-2-ol.
The relative retention time with reference to propofol for 1-(1- Reference solution. A solution containing 0.0008 per cent w/v
methylethoxy)-2-( l-methylethyl)benzene (propofol impurity K) ofpropofol RS, 0.00008 per cent w/v of2, 6-bis(l-methylethyl)-
is about 0.76, for 2,2-dimethyl-4-(l-methylethyl)-1,3- 1,4-benzoquinone RS (propofol impurity J RS) and 0.0002
benzodioxole (propofol impurity L) is about 0.81 ,forpropofol per cent w/v of 3,3',5,5'-tetrakis (l-methylethyl)biphenyl-
impurity J is about 1.01 and for 2-(1-methylethyl)-6- 4,4'-diol RS (propofol dimer RS) in propan-2-o1 containing
propylphenol (propofol impurity 0) is about 1.03. 6.8 per cent v/v of water.
Inject 1 III ofthe test solution, reference solution (a) and (c). In Chromatographic system
the chromatogram obtained with the test solutioIl the area of a stainless steel column 10 cm x 4.6 mrn, packed with
the peak due to propofol impurities J, K, Land 0, for each octadecylsilane bonded to porous silica (5 Ilm) (such as
impurity, is not more than 0.5 times the area of the principal HypersilODS),
peak in the chromatogram obtained with reference solution mobile phase: a mixture of 40 volumes of
(a) (0.05 percent). tetrahydrojUran and 60 volumes of water,
Assay. Determine by liquid chromatography (2.4.14) as flow rate. 2 ml per minute,
described under Related substances. - spectrophotometer set at 254 nm,
- injection volume. 20 Ill.
Inject test solution (b) and reference solution (d).
Inject the reference solution. The relative retention time with
Calculate the content ofC 12H 1sO. reference to propofol for propofol impurity J is about 0.8 and
Storage. Store protected from light under an inert gas. for propofol dimer is about 2.5.

1986

www.webofpharma.com
IP 2010 PROPRANOLOL HYDROCHLORIDE

Inject the test solution and the reference solution. Run the - spectrophotometer set at 210 nm,
chromatogram three times the retention time of the principal - injection volume. 250 Jll.
peale. In the chromatogram obtained with the test solution the
Inject the test solution and the reference solution.
area ofthe peak due to propofol quinone (propofol impurity 1)
is not more than the areaof'the corresponding peak in the Calculate the content of lysolecithin.
chromatogram obtained with the reference solution (0.1 per Bacterial endotoxins (2.2.3). Not more than 1.65 Endotoxin
cent) and the area of the peak due to propofol dimer is not Units per ml.
more than the area of the corresponding peak in the
Other tests. Complies. with the tests stated under Parenteral
chromatogram obtained with the reference solution (0.25 per
Preparations (Injections).
cent).
Assay. Determine by liquid chromatography (2.4.14).
Globule size. Not more than 5000 globules more than or equal
to 2 Jlm per ml ofa 0.01 per cent vIv dilution in a filtered 0.9 per Test solution. Dilute a volume of injection containing 80 mg of
cent w/v solution of sodium chloride. By means of suitable Propofol to 100 ml with the propan-2-ol.
equipment, pass the solution through a 50 Jlm aperture and Reference solution (a). A 0.08 per cent wIv solution ofpropofol
count the number of globules by monitoring the change in RS in in propan-2-o1 containing 6.8 per cent v/v of water.
electrical current between two electrodes immersed on either
side of the aperture (such as Coulter Counter). Repeat the Reference solution (b). A solution containing 0.0008 per cent
procedure using the 0.9 per cent w/v solution of sodium w/v of propofol RS and 0.00008 per cent w/v of propofol
chloride only to determine the background count. impurity J RS in propan-2-o1 containing 6.8 per cent v/v of
water.
Free fatty acid. Not more than 7 millimoles per litre, when
determined by the following method. The chromatographic system described under Propofol
quinone and propofol dimer may be used but using a detection
To 5 ml ofa well shaken injection add 25 ml ofa mixture of40 wavelength of275 run and injecting 20 Jll of each solution.
volumes ofpropan-2-ol, 10 volumes ofn-heptane and 1 volume
of 0.5 sulphuric acid and shake for 1 minute. Allow to stand Inject reference solution (b). The test is not valid unless the
for 10 minutes, add 15 ml ofn-heptane and 15 ml ofwater. Mix resolution between the peaks due to propofol and propofol
by inverting the container 10 times and allow to stand for 15 impurity J is not less than 2.5.
minutes. To 15 ml of the upper phase add 5 ml of nile blue A Inject the test solution and reference solution (b).
solution and pass a stream of nitrogen, previously passed
Calculate the content ofC1zHIsO in the injection.
through 0.1 M sodium hydroxide, through the solution. Titrate
with 0.02 M sodium hydroxide, using a microburette. Calculate Storage. Store at a temperature not exceeding 30°. It should
the content offree fatty acid from a calibration curve prepared not be allowed to freeze.
from quantities of 1, 2, 4, 6 and 8 ml of a 1.282 per cent w/v
solution ofpalmitic acid in n-heptane, each diluted to 50 ml
with n- heptane. Carry out the method described above, using
5 ml ofeach solution and beginning at the words 'add 25 mlof
Propranolol Hydrochloride
a mixture'.
Lysolecithin. Not more than 0.2 per cent w/v.
Determine by liquid chromatography (2.4.14).
Test solution. Dilute a volume of injection containing 0.1 g of
Propofol in 100 ml ofthe mobile phase.
Reference solution. A 0.01 per cent w/v solution oflysolecithin CI6HzINOz,HCl Mol. Wt. 295.8
in the mobile phase.
Propranolol Hydrochloride is (RS)-I-[( I-methylethyl)amino]-
Chromatographic system 3-(naphthalen-l-yloxy)propan-2-01 hydrochloride.
- a stainless steelcolurnn 15 cm X 4.6 mm, packed with
Propranolol Hydrochloride contains not less than 99.0 per
octadecylsilane bonded to porous silica (5 Jlm) (Such
cent and not more than 101.0 per cent of CI6HzINOz, HCl,
as Nucleosil C 18),
calculated on the dried basis.
- mobile phase: a mixture of 1.4 volumes of
orthophosphoric acid, 40 volumes of water and 60 Category. Antihypertensive, antianginal and antiarrhythmic
volumes of acetonitrile, agent.
- flow rate. 1.2 ml per minute,

1987

www.webofpharma.com
PROPRANOLOL HYDROCHLORIDE IP 2010

Dose. Orally, 20 mg to 2 g daily, in divided doses; the initial is not more than twice the area of the principal peak in the
dose should not exceed 40 mg; by slow intravenous injection, chromatogram obtained with reference solution (0.4 per cent).
3 to 10mg.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Description. A white or almost white powder.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105°.
Identification
Assay. Weigh accurately about 0.25 g, dissolve in 25 mlof
A. Determine by infrared absorption spectrophotometry (2.4.6). ethanol (95 percent) and titrate with 0.1 Msodium hydroxide,
Compare the spectrum with that obtained with propranolol determining the end-point potentiometrically (2.4.25).
hydrochloride RS or with the reference spectrum of
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02958 g of
propranolol hydrochloride.
CI6HzINOz, HCl.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Storage. Store protected from moisture.
0.002 per cent w/v solution in methanol shows absorption
maxima at about 290 nm, 306 nm and 319 nm.
C. Gives reaction A ofchlorides (2.3.1).
Propranolol Injection
Tests Propranolol Hydrochloride Injection
Appearance of solution. A 10.0 per cent w/v solution in Propranolol Injection is a sterile solution of Propranolol
methanol is clear (2.4.1), and not more intensely coloured Hydrochloride in Water for Injections containing Citric Acid.
than degree 60fthe appropriate range of reference solutions Propranolol Injection contains not less than 90.0 per cent and
(2.4.1). not more than 110.0 per cent of the stated amount of
pH (2.4.24).5.0 to 6.0, determined in a 1.0 per centw/v solution. propranolol hydrochloride, CJ6HzJNOz, HCI.

Specific optical rotation (2.4.22). -1.0° to +1.0°, determined in Usual strength. 1 mgpermI.
a 4.0 per cent w/v solution. .
Identification
Related substances. Determine by liquid chromatography
A. Make alkaline with 1 M sodium hydroxide a volume
(2.4.14).
containing 10 mg of Propranolol Hydrochloride and extract
Test solution. Dissolve 20 mg of the substance under with three quantities, each of5 ml, of ether. Wash the combined
examination in 10.0 ml ofthe mobile phase. extracts with water until the washings are freefrorrialkali, dry
with anhydrous sodium sulphate, filter, evaporate the filtrate
Reference solution. Dilute 2.0 ml of the test solution to 100.0
ml with the mobile phase. Dilute 1.0 ml ofthis solution to 10.0 to dryness and dry the residue at 50° at a pressure of2 kPa for
ml with the mobile phase. 1 hour.
On the residue determine by infrared absorption
Chromatographic system
spectrophotometry (2.4.6). Compare the spectrum with that
- a stainless steel column 25 cm x 4.6 mm, packed with
obtained with propranolol hydrochloride RS treated in the
octadecylsilane bonded to porous silica (5 ~m),
same manner or with the reference spectrum of propranolol.
- mobile phase: dissolve 1.6 g of sodium laurylsulphate
and 0.31 g of tetrabutylammonium dihydrogen B. When examined in the range 230 nm to 360 nm (2.4.7),the
phosphate in a mixture of 1 ml of sulphuric acid, 450 ml solution obtained in the Assay shows absorption maxima at
of water and 550 ml of acetonitrile, adjust to pH 3.3 about 290 nm, 306 nm and 319 nm.
using dilute sodium hydroxide solution,
- flow rate. 1.8 ml per minute, Tests
- spectrophotometer set at292 nm, pH (2.4.24).3.0 to 3.5.
injection volume. 20 ~I.
Related substances. Determine by liquid chromatography
Inject the test solution and the reference solution. Run the (2.4.14).
chromatogram 7 times the retention time ofthe principal peak.
Test solution. Dilute a volume of injection containing about
In the chromatogram obtained with the test solution the area
100 mg ofPropranolol HydrocWoride in 100 ml of acetonitrile.
of any secondary peak is not more than 0.5 times the area of
the principal peak in the chromatogram obtained with reference Reference solution. Dilute 1 ml of the test solution to 500 ml
solution (0.1 per cent) and the sum ofall the secondary peaks with the mobile phase.

1988

www.webofpharma.com
IP 2010 PROPRANOLOL TABLETS

Chromatographic system to dryness and dry the residue at 500 at a pressure of2 kPa for
- a stainless steel column 20 cm x 5 mm, packed with 1 hour.
endcapped octadecylsilane bonded to porous silica
On the residue, determine by infrared absorption
(5 /lm) (Such as Hypersil ODS),
spectrophotometry (2.4.6). Compare the spectrum with that
- mobile phase: a mixture of 1.15 g of sodium dodecyl
obtained with propranolol hydrochloride RS treated in t he
sulphate, 10 ml of a mixture of 1 volume of sulphuric same manner or with the reference spectrum of propranolol.
acid and 9 volumes of water, 20 ml of a 1.7 percent
w/v solution of tetrabutylammonium dihydrogen B. When examined in the range 230 nm to 360 nm (2.4.7), the
orthophosphate, 370 ml of water and 600 ml of final solution obtained in the Assay shows absorption maxima
acetonitrile, adjusted to pH 3.3 with 2 M sodium at about 290 nm, 306 nm and 319 nm.
hydroxide,
flow rate. 1.8 ml per minute, Tests
- spectrophotometer set at 292 nm, Uniformity of content. (For tablets containing 10 mg or less)
- injection volume. 10 ~Ll. - Comply with the test stated under Tablets.
Inject the test solution and the reference solution. Run the Transfer one tablet to a 100-ml volumetric flask, add 5 ml of
chromatogram 5 times the retention time ofthe principal peak. dilute hydrochloric acid and allow to stand, swirling
In the chromatogram obtained with the test solution, the area occasionally, until it is disintegrated. Add about 70 ml of
ofany secondary peak is not more than the area ofthe principal methanol and shake well for about 1 minute. Dilute to volume
peak in the chromatogram obtained with the reference solution with methanol, mix and centrifuge an aliquot of the solution.
(0.2 per cent) and the sum of the areas of all the secondary Dilute a suitable volume of the clear solution with methanol
peaks is not more than four times the area ofthe principal peak to produce a solution containing 20 /lg of Propranolol
in the chromatogram obtained with the reference solution (0.8 Hydrochloride per ml. Measure the absorbance ofthe resulting
per cent). solution at the maximum at about 290 nm (2.4.7), using
Other tests. Complies with the tests stated under Parenteral methanol as the blank. Calculate the content of CI6HzINOz,
Preparations (Injections). HCI taking 206 as the specific absorbance at 290 nm.
Assay. To an accurately measured volume containing about Dissolution (2.5.2).
2 mg ofPropranolol Hydrochloride add sufficient methanol to Apparatus No.1,
produce 100.0 ml and measure the absorbance ofthe resulting Medium. 900 mltf 0.1 M hydrochloric acid,
solution at the maximum at about 290 urn (2.4.7). Calculate the Speed and time. 100 rpm and 45 minutes.
content of CI6HzINOz, HCI taking 206 as the specific
absorbance at 290 nm. Withdraw a suitable volume ofthe medium and filter. Dilute a
suitable volume ofthe filtrate with the same solvent. Measure
Storage. Store protected from light, in a single dose containers. the absorbance of the resulting solution at the maximum at
about 290 nm (2.4.7). Calculate the content ofC,6Hz,NOz,HCI
in the medium taking 206 as the specific absorbance at 290 urn.
D. Not less than 75 per cent ofthe stated amount ofC,6HzINOz,
Propranolol Tablets HCI.
Propranolol Hydrochloride Tablets Related substances. Determine by liquid chromatography
Propranolol Tablets contain not less than 92.5 per cent and (2.4.14).
not more than 107.5 per cent of the stated amount of Test solution. Disperse a quantity of powdered tablets
propranolol hydrochloride, CI6HzlNOz, HCI. containing about 100 mg ofPropranolol HydrochlOlide in 100.0
Usual strengths. 10 mg; 40 mg. ml of methanol and filter.
Reference solution. Dilute 1 ml of the test solution to 500 ml
Identification with the mobile phase.
A.Suspend a quantity of the powdered tablets containing Chromatographic system
0.1 g of Propranolol Hydrochloride in 20 ml of water, filter, a stainless steel column 20 cm x 5 mm, packed with
make the filtrate alkaline with 1 M sodium hydroxide and extract endcapped octadecylsilane bonded to porous silica
with three quantities, each of 10 ml, ofether. Wash the combined (5 /lm) (Such as Hypersil ODS),
extracts with water until the washings are free from alkali, dry mobile phase: a mixture of 1.15 g of sodium dodecyl
with anhydrous sodium sulphate, filter, evaporate the filtrate sulphate, 10 ml of a mixture of 1 volume of sulphuric

1989

www.webofpharma.com
PROPYL GALLATE IP 2010

acid and 9 volumes of water, 20 ml of a 1.7 per cent B. Dissolve 10 mg in 50 ml of water, cool and add 5 ml of dilute
w/v solution of tetrabutylammonium dihydrogen ammonia solution; a red colour is produced which becomes
orthophosphate, 370 ml of water and 600 ml of brown on standing and is restored on shaking.
acetonitrile, adjusted to pH 3.3 with 2 M sodium C. Dissolve 5 mg in 50 ml of water and add 0.05 mlofjerric
hydroxide, chloride test solution; a bluish black colour is produced.
- flow rate. 1.8 ml per minute,
- spectrophotometer set at 292 nm, Tests
- injection volume. 10 Ill.
Chlorides (2.3.12). Shake 1.0 gwith 50 ml ofwater for 5 minutes
Inject the test solution and the reference solution. Run the
and filter. 25 ml of the filtrate complies with the limit test for
chromatogram 5 times the retention time ofthe principal peak.
chlorides (500 ppm).
In the chromatogram obtained with the test solution, the area
ofany secondary peak is not more than the area ofthe principal Sulphates (2.3.17). Shake 0.8 g with 100 ml of water for
peak in the chromatogram obtained with the reference solution 5 minutes and filter. 15 ml ofthe filtrate complies with the limit
(0.2 per cent) and the sum of the areas of all the secondary test for sulphates (0.12 per cent).
peaks is not more than four times the area ofthe principal peak Sulphated ash (2.3 .18). Not more than 0.1 per cent.
in the chromatogram obtained with the reference solution (0.8
per cent). Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105°.
Other tests. Comply with the tests stated under Tablets.
Storage. Store protected from light and moisture, in non-
Assay. Weigh and powder 20 tablets. Weigh accurately a metallic containers.
quantity ofthe powder containing about 20 mg ofPropranolol
Hydrochloride and shake with 20 ml of water for 10 minutes.
Add 50 ml of methanol, shake for a further 10 minutes, add
sufficient methanol to produce 100.0 ml and filter. Dilute
10.0 ml ofthe filtrate to 50.0 ml with methanol and measure the Propylene Glycol
absorbance of the resulting solution at the maximum at about
1,2-Propanediol
290 nm (2.4.7). Calculate the content ofC16HzlNOz, HCI taking
206 as the specific absorbance at 290 nm.
'ii',
Storage. Store protected from light and mOIsture.

Propyl Gallate C3H sOz Mol. Wt. 76.1


Propylene Glycol is (RS)-propane-l ,2-diol.
o
HO~O~CH3 Category. Pharmaceutical aid (humectant; solvent).
Description. A clear, colourless, viscous liquid; practically
HO~ odourless; hygroscopic.
OH
Identification
Mol. Wt. 212.2
A. To 0.5 ml ofa 0.01 percentw/v solution, cooled in ice, add
Propyl Qallate is propy13,4,5-trihydroxybenzoate. 5 ml ofa cooled mixture of 10 ml ofwater and 90 ml ofsulphuric
Category. Pharmaceutical aid (antioxidant). acid. Heat for 10 minutes on a water-bath at 70°, cool and add
0.2 ml ofa 3 per cent w/v solution of ninhydrin in a 2.5 per cent
Description. A white or creamy white, crystalline powder;
w/v solution of sodium metabisulphite; a violet colour slowly
odourless or almost odourless.
appears.
Identification B. Heat 0.15 ml with 0.1 g of boric acid; a pleasant odour
develops.
A. When examined in the range 230 nm to 360 nm (2.4.7), a
0.001 per cent w/v solution in methanol shows an absorption C. Add 1 ml to 0.5 g ofpotassiumbisulphate and heat gently;
maximum only at about 275 nm; absorbance at about 275 nm, a fruity odour develops and when the solution is heated to
about 0.49. dryness, no sharp, acrid smell of acrolein is perceptible.

1990

www.webofpharma.com
IP 2010 PROPYLPARABEN

Tests diethylene glycol. The test is not valid unless in the


chromatogram obtained with the reference solution, the
Appearance of solution. The substance under examination is resolution between the peaks due to propylene glycol (first
clear (2.4.1), and colourless (2.4.1). peak) and ethylene glycol (second peak) is not less than 1.0.
Acidity. Mix 10 ml with 40 m1 of water and add 0.1 mlof No peaks corresponding to ethylene glycol and diethylene
bromothymol blue solution. The solution is greenish yellow glycol are obtained in the chromatogram obtained with the
and not more than 0.05 ml of 0.1 M sodium hydroxide is test solution.
required to change the colour to blue.
Sulphated ash (2.3.18). Not more than 0.01 per cent w/w,
Boiling range (2.4.8). 184° to 189°. detennined by the following method. Heat 50 g until it burns,
Relative density (2.4.29). 1.035 to 1.040. and ignite. Allow to cool, moisten the residue with sulphuric
acid and ignite; repeat the operations.
Refractive index (2.4.27). 1.431 to 1.433.
Water (2.3.43). Not more than 0.2 per cent, determined on
Heavy metals (2.3.13). Dilute 3 ml to 12 ml with water. The
5.0g.
resulting solution complies with the limit test for heavy metals,
Method D (5 ppm). Use lead standard solution (l ppm Pb) to Storage. Store protected from moisture.
prepare the standard.
Oxidising substances. To 10 ml add 5 ml of water, 2 m1 of
potassium iodide solution and 2 m1 of 1 M sulphuric acid and Propylparaben
allow to stand in a ground-glass-stoppered flask protected
from light for 15 minutes. Titrate the liberated iodine with Propyl Hydroxybenzoate
0.05 M sodium thiosulphate using 1 ml of starch solution,
added towards the end of the titration, as indicator. Not more
than 0.2 ml of 0.05 M sodium thiosulphate is required.
Reducing substances. Mix 1 rnl with 1ml of6 M ammonia and
heat on a water-bath at 60° for 5 minutes; the solution is not
yellow. Immediately add 0.15 ml of 0.1 M silver nitrate; the
solution does not change its appearance within 5 minutes. Mol. Wt. 180.2

Ethylene glycol and diethylene glycol. Determine by gas Propylparaben is propyI4-hydroxybenzoate.


chromatography (2.4.13). Propylparaben contains not less than 99.0 per cent and not
Test solution. Dissolve 2 g ofthe substance under examination more than 101.0 per cent ofC IOH I20 3, calculated on the dried
in sufficient ethanol (95 per cent) to produce 100 ml. basis.

Reference solution. Dissolve 2 g of the substance under Category. Pharmaceutical aid (antimicrobial preservative).
examination, 0.02 g ofethylene glycol and 0.02 g ofdiethylene Description. A white, crystalline powder; odourless.
glycol in ethanol (95 per cent) and dilute to 100 ml with the
same solvent. Identification
Chromatographic system A. When examined in the range 230 nm to 360 urn (2.4.7), a
- a glass column 1.5 m x 3 mm, packed with 12 per cent 0.0005 per cent w/v solution in ethanol (95 per cent) shows
Sorbitol ~)ll untreated siliceous earth (Such as an absorbance maximum at about 258 nm; absorption at
Chromosorb W-NAW (SINS), 258 urn, 0.44 to 0.47.
temperature:
B. To about 10 mg in a test-tube add 1 ml ofsodium carbonate
column: 165°,
solution, heat to boiling for 30 seconds and cool (solution A).
inlet port and detector at 260°,
To a further 10 mg in a test-tube add 1 rnl ofsodium carbonate
- flow rate. 30 ml per minute ofthe carrier gas.
solution; the substance partly dissolves (solution B). Add at
Inject 3 ).11 or other suitable volume ofthe test solution. Record the same time to each of the solutions A and B 5 m1 of
the chromatograms adjusting the sensitivity so that the height aminophenazone solution and 1 ml ofpotassiumferricyanide
ofthe peak due to propylene glycol is more than 50 per cent of solution and mix. Solution B is yellow to orange-brown.
the full-scale deflection in the chromatograms. Inject the same Solution A is orange to red and the colour is clearly more
volume of reference solution and record the chromatograms. intense than any similar colour that may be obtained with
The order of elution is propylene glycol, ethylene glycol and solution B.

1991

www.webofpharma.com
PROPYLPARABEN IP 2010

Tests Storage. Store protected from moisture.

Appearance ofsolupon. Al 0.0 per cent w/v solution in ethanol


(95 Pier cent) is clear (2.4.1), and not more intensely coloured
than reference solution BYS6 (2.4.1). Propylthiouracil
Acidity. Dissolve 1.0 g in sufficient ethanol (95 per cent) to
produce 10 ml. To 2 ml of the solution add 3 ml of ethanol H
H3C~NyS
. . lrr
(95 per cent), 5 ml of carbon dioxide-ji-ee water and 0.1 mlof
bromocresol green solution. Not more than 0.1 ml of Od M
sodium hydroxide is required to change the colour of the NH
. solution.
o
Related substances. Detelmine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. C7H ION 20S Mol. Wt. 170.2

Mobile phase. A mixture of 1 volume ofglacial acetic acid, 30 Propylthiouracil is 2,3-dibydro-6-propyl-2-thioxopyrimidin-


volumes of water and 70 volumes of methanol. 4(1H)-one.

Test solution. Dissolve 0.1 g of the substance under Propylthiouracil contains not less than 98.0 per cent and not
examination in 10.0 ml of acetone. more than 100.5 per cent ofC 7H ION 20S, calculated on the dried
basis.
Reference solution (a). Dilute 0.5 ml of the test solution to
100.Oml with acetone. Category. Antithyroid.

Reference solution (b). Dissolve 10 mg of ethyl Dose. 300 to 450 mg daily, in divided doses.
parahydroxybenzoate RS in 1.0 ml of the test solution and Description. A white or pale cream-coloured crystals or
dilute to 10.0 ml with acetone. crystalline powder; odourless.
Apply 2 !!l of each solution. Allow the mobile phase to rise 15
cm. Dry the plate in air and examine in ultraviolet light at 254 Identification
nm. In the chromatogram obtained with the test solution, any Test A may be omitted if tests Band C are carried out. Tests B
secondary spot is not more intense than the cOlTesponding and C may be omitted if test A is carried out.
spot in the chromatogram obtained with reference solution (a)
(0.5 per cent) The test is not valid unless the chromatogram A. Detenmne by infrared absorption spectrophotometry (2.4.6).
obtained with reference solution (b) shows two Clearly Compare the spectrum with that obtained with
separated principal spots. propylthiouracil RS or with the reference spectrum of
propylthiouracil.
Chlorides (2.3.12). Heat 2.0 gwith 100 ml of water, cool, add
sufficient water to restore the original volume, and filter. 25 ml B. Examine the chromatograms obtained in the test for Related
of the filtrate complies with the limit test for chlorides substances in ultraviolet light at 254 nm before exposure of
(500 ppm). the plate to iodine vapour. The principal spot in the
chromatogram obtained with test solution (b) cOlTesponds to
Sulphates.To 10 ml of the filtrate obtained in the test for that in the chromatogram obtained with reference solution
Chlorides add 0.15 ml of dilute hydrochloric acid and 0.1 m1 (b).
of barium chloride solution; no turbidity is produced within
10 minutes. C. To about 20 mg add 8 ml of bromine water and shake for a
few minutes. Boil until the mixture is decolorised, allow to cool
Sulphated ash (2.3.18). Not more than 0.1 per cent.
and filter. Add 2 ml of barium chloride solution; a white
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined precipitate is produced. Add 5 ml of 2 M sodium hydroxide;
on 1.0 g by drying over silica gel for 5 hours. the precipitate does not become violet.
Assay. To 1.0 g add 20.0 ml of 1 M sodium hydroxide. Heat at
Tests
about 70° for 1 hour. Cool rapidly in an ice bath. Titrate the
excess sodium hydroxide with 0.5 M sulphuric acid, Related substances. Detennine by thin-layer chromatography
continuing the titration until the second point ofinflexion and (2.4.17), coating the plate with silica gel GF254.
detelmining the end-point potentiometrically (2.4.25).
Mobile phase. A mixture of 100 volumes of chloroform,
1 ml of 1 M sodium hydroxide is equivalent to 0.1802 g of 12 volumes of 2-propanol and 0.2 volume of glacial acetic
CIOH I20 3• acid.

1992

www.webofpharma.com
IP 2010 PROPYLTHIOURACIL TABLETS

Test solution (a). Dissolve 0.1 g of the substance under Identification


examination in 10 ml ofmethanol.
A. Shake a quantity ofthe powdered tablets containing 50 mg
Test solution (b). Dissolve 0.1 g of the substance under of Propylthiouracil with 20 ml of methanol for 10 minutes,
examination in 100 ml ofmethanol. filter and evaporate to dryness.
Reference solution (a). A 0.01 per cent w/v,solution of the On the residue, determine by infrared absorption
substance under examination in methanol. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with propylthiouracil RS or with the reference
Reference solution (b). A 0.1 per cent w/v solution of
propylthiouracil RS in methanol. spectrum ofpropylthiouracil.
B. Shake a quantity ofthe powdered tablets containing 50 mg
Reference solution (c). A 0.0005 per cent w/v solution of
of Propylthiouracil with 60 ml of methanol for 20 minutes,
thiourea in methanol.
dilute to 100 ml with methanol and filter. Dilute 5 ml of the
Apply to the plate 10 III of each solution. After development, filtrate to 250 ml with methanol. When examined in the range
dry the plate in air and examine in ultraviolet light at 254 nm. 230 nm to 360 nm (2.4.7), the resulting solution shows an
Expose the plate to iodine vapour for 10 minutes. By both absorption maximum only at about 274 nm.
methods of visualisation, any spot corresponding to thiourea
C. Extract a quantity of the powdered tablets in a continuous
in the chromatogram obtained with test solution (a) is not
extraction apparatus (2.1.8) with ether and evaporate the
more intense than the spot in the chromatogram obtained
solution to dryness. The residue, after drying at 105°, melts at
with reference solution (c) and any other secondary spot is
about 219° (2.4.21).
not more intense than the spot in the. chromatogram obtained
with reference solution (a). Tests
Arsenic (2.3.1 0). Dissolve 2.0 g in 50 ml ofwater and add 10 rnl Related substances. Detennine by thin-layer chromatography
of stannated hydrochloric acid AsT. The resulting solution (2.4.17), coating the plate with silica gel GF254.
complies with the limit test for arsenic (5 ppm).
Mobile phase. A mixture of 100 volumes of chloroform,
Heavy metals (2.3.13). 1.0 g complies with the limit test for 12 volumes of 2-propanol and 0.2 volume of glacial acetic
heavy metals, Method C (20 ppm). acid.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Test solution. Shake a quantity of the powdered tablets
containing 50 mg ofPropylthiouracil with 5 ml ofmethanol for
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
15 minutes, filter and use the filtrate.
on 1.0 g by drying in oven at 105°.
Reference solution (a). Dilute 1 volume ofthe test solution to
Assay. Weigh accurately about 0.3 g, add 30 ml of water and 100 volumes with methanol.
30.0 ml (n] ml) of0.1 M sodium hydroxide, boil and shake until
solution is complete. Add 50 ml of 0.1 M silver nitrate with Reference solution (b). A 0.001 per cent w/v solution of
stirring, boil gently for 5 minutes, cool and titrate with 0.1 M thiourea in methanol.
sodium hydroxide, determining the end-point Apply to the plate 10 III of each solution. After development,
potentiometrically (2.4.25) (n2 ml). Record the total volume dry the plate in air and examine in ultraviolet light at 254 mTI.
(n] + n2 ml) of 0.1 M sodium hydroxide added. Expose the plate to iodine vapour for 10 minutes. By both
1 ml of0.1 M sodium hydroxide is equivalent to 0.008511 g of methods of visualisation, any spot corresponding to thiourea
C7H ION20S. in the chromatogram obtained with the test solution is not
more intense than the spot in the chromatogram obtained
Storage. Store protected from light and moisture. with reference solution (b) and any other secondary spot is
not more intense than the spot in the chromatogram obtained
with reference solution (a).
Other tests. Comply with the tests stated under Tablets
Propylthiouracil Tablets Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.15 g of
Propylthiouracil Tablets contain not less than 92.5 per cent
Propylthiouracil, dissolve in a mixture of20 ml of0.1 M sodium
and not more than 107.5 per cent of the stated amount of
hydroxide and 75 ml ofwater with the aid ofgentle heat. Cool,
propylthiouracil, C7H ION 20S.
add 4 g of sodium acetate, just acidity the solution to litmus
Usual strength. 50 mg. paper with 6 M acetic acid, add 0.5 ml of a freshly prepared

1993

www.webofpharma.com
PROPYPHENAZONE IP 2010

0.5 per cent w/v solution of 1,5-diphenylcarbazone in ethanol Tests


(95 per cent) and titrate with 0.02 M mercuric nitrate until a
pinkish violet colour persists for 2 to 3 minutes. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
I ml of 0.02 M mercuric nitrate is equivalent to 0.006808 g of
Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of
C7H ION zOS.
phenolphthalein solution; the solution is colourless. Add
Storage. Store protected from light and moisture. 0.2 ml of 0.01 M sodium hydroxide; the solution is pink. Add
0.4 ml of 0.01 M hydrochloric acid; the solution becomes
colourless. Add 0.2 ml of methyl red solution; the solution is
orange or red.
Propyphenazone
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel HF254.
Mobile phase. A mixture of 45 volumes of cyclohexane,
45 volumes of ethyl acetate and 10 volumes of I-butanol.
Test solution (a). An 8 per cent w/v solution ofthe substance
under examination in methanol.
Test solution (b). A 1.6 per cent w/v solution of the substance
under examination in methanol.
Reference solution (a). A 0.016 per cent w/v solution of the
Mol. Wt. 230.3 substance under examination in methanol.

Propyphenazone is 4-isopropyl-2,3-dimethyl-l-phenyl-3- Reference solution (b). A 1.6 per cent w/v solution of
pyrazolin-5-one. propyphenazone RS in methanol.

Propyphenazone contains not less than 99.0 per cent and not Apply to the plate 5 III of each solution. After development,
more than 101.0 per cent ofC 14H IsNzO, calculated on the dried dry the plate in a current ofhot air for 15 minutes and examine
basis. in ultraviolet light at 254 nm. Spray the plate with a mixture of
equal volumes of potassium ferricyanide solution and ferric
Category. Analgesic; antipyretic. chloride solution. By both methods of visualisation, any
Dose. 1.5 to 3 g daily, in divided doses. secondary spot in the chromatogram obtained with test
solution (a) is not more intense than the spot in the
Description. A white or slightly yellowish, crystalline powder;
chromatogram obtained with reference solution (a).
odourless.
Arsenic (2.3.10). Mix 1.0 g with 10 ml ofa 2 per cent w/v
Identification solution of magnesium nitrate in ethanol in a silica or platinum
dish, evaporate on a water-bath and heat gradually in order to
Test A may be omitted iftests Band C are carried out. Tests B
incinerate. If the material remains incompletely carbonised,
and C may be omitted if test A is carried out.
moisten with a small quantity of nitric acid and ignite again.
A. Determine by infrared absorption spectrophotometry (2.4.6). Cool, add 3 ml of hydrochloric acid and heat on a water-bath
Compare the spectrum with that obtained with to dissolve the residue. The resulting solution complies with
propyphenazone RS or with the reference spectrum of the limit test for arsenic (10 ppm).
propyphenazone.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
B. In the test for Related substances, the principal spot in the heavy metals, Method B (l0 ppm).
chromatogram obtained with test solution (b) corresponds to
Sulphated ash (2.3.18). Not more than 0.1 per cent.
that in the chromatogram obtained with reference solution
(b). Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 60° over phosphoruspentoxide
C. Dissolve 2 g in sufficient of a mixture of equal volumes of
ethanol (95 per cent) and carbon dioxide-free water to at a pressure of 1.5 to 2.5 kPa for 4 hours.
produce 50 ml (solutionA). To 1 ml ofsolutionA add O.lml of Assay. Weigh accurately about 0.2 g of the dried material,
ferric chloride solution; a brownish red colour is produced dlssoive· in· 3()11110f anhjid,;ous glacial acetic acid.· titraTe
which becomes yellow on addition ofl ml of2 M hydrochloric with 0.1 M perchloric acid, determining the end-point
acid. potentiometrically (2.4.25). Carry out a blank titration.

1994

www.webofpharma.com
IP 2010 PROTAMINE SULPHATE

1 m1 of 0.1 M perchloric acid is equivalent to 0.02303 g of Iron (2.3.14). Dissolve 2.0 g in water with the aid ofheat and
C1JflSN20. dilute to 20 ml with water. The resulting solution complies
with the limit test for iron (20 ppm).
Storage. Store protected from moisture.
Mercury. Add 20 ml of a mixture of equal volumes of nitric
acid and sulphuric acid to 2.0 g in a 250-ml flask fitted with a
ground-glass stopper, boil under a reflux condenser for 1 hour,
Protamine Sulphate cool and carefully dilute with water. Boil until nitrous fumes
are no longer evolved. Cool, carefully dilute the solution to
Protamine Sulphate is a purified mixture of the su1phates of 200 ml with water, mix and filter. Transfer 50 ml ofthe filtrate to
basic peptides prepared from the sperm or mature testes of a separating funnel. Shake with successive small quantities of
suitable species of fish. It binds with heparin in solution, chloroform until the chloroform layer remains colourless. To
inhibiting its anticoagulant activity. It is prepared in conditions the aqueous layer add 25 ml of 1 M sulphuric acid, 115 ml of
designed to minimise the degree of Microbial contamination. water and 10 ml ofa 20 per cent w/v solution of hydroxylamine
Each mg of Protamine Sulphate precipitates not less than hydrochloride. Titrate with dithizone solution; after each
100 Units of heparin sodium RS, calculated on the dried basis. addition, shake the mixture 20 times and towards the end-
point of the titration allow to separate and discard the
Category. Heparin antidote. chloroform layer. Titrate until a greenish blue colour is
Dose. By slow intravenous injectiCln over a 1O-minute period, produced. Calculate the content of mercury using the
1 mg for every 100 Units ofheparin remaining in the patient if equivalent ofmercury in Ilg per ml oftitrant determined in the
given within 15 minutes; maximum 50 mg. standardisation ofthe dithizone solution (10 ppm).
Description. A white or almost white powder; hygroscopic. Nitrogen (2.3.30). 21.0 to 26.0 per cent, calculated on the dried
basis, determined by Method C.
Identification Sulphates. 16 to 24 per cent, determined by the following
A. Produces a precipitate under the conditions of the Assay. method. Dissolve 0.15 gin 15 ml of water in a beaker, add 5 ml
of 2 M hydrochloric acid and heat to boiling. Slowly addto
B. Dissolve 0.2 g in 5 ml of water and dilute to 10 ml with the the boiling solution 10 ml of barium chloride solution. Cover
same solvent (solution A). To 0.5 ml ofsolution A add 4.5 ml of and heat on a water-bath for 1 hour. Filter and wash the
water, 1 ml of a 10 per cent w/v solution of sodium hydroxide precipitate several times with small quantities of hot water.
and 1 m1 ofa 0.02 per cent w/v solution of I-naphthol and mix. Dry and ignite the residue to constant weight at 600°.
. Cool to 5° and add 0.5ml of alkaline sodium hypobromite
1 g ofthe residue is equivalent to 0.4117 g ofS04 •
solution; an intense red colour is produced.
Abnormal toxicity (2.2.1). Complies with the test for abnormal
C. Heat 2 ml ofso1utionAon a water-bath at 60°, add 0.1 ml of
toxicity, using 0.5 mg dissolved in 0.5 ml ofwater for injections.
mercuric sulphate solution and mix; no precipitate is produced.
Cool the mixture in ice; a white precipitate is produced. Loss on drying (2.4.19). Not more than 5.0 per cent, determined
on 1.0 g by drying in an oven at 105° for 3 hours.
D. Gives reactionAofsulphates (2.3.1).
Assay. Prepare solutions of the substance under examination
Tests in water containing (1) 0.015 per cent w/v (2) 0.01 per cent
w/v and (3) 0.005 per cent w/v. Titrate each ofthe solutions in
Appearance ofsolution. To 2.5 ml ofsolution A add 7.5 mlof duplicate with a 174 ill per ml or a suitable dilution of heparin
water. The resulting solution is not more opalescent than sodium RS using the following procedure. Introduce an
opalescence standard OS2 (2.4.1), and not more intensely accurately measured volume ofthe solution to be titrated, for
.coloured than reference solution BYS6 (2.4.1). example 1.5 m1, into the cell of a suitable spectrophotometer,
Specific optical rotation (2.4.22). -65.0° to -85.0°, determined set the instrument at a suitable wavelength (none is critical) in
at 20° in a 1.0 per cent w/v solution in 0.1 M hydrochloric the visible range and add the titrant in small volumes until
acid. there is a sharp increase in the absorbance. Note the volume
of titrant added.
Light absorption (2.4.7). Dilute 2.5 ml ofsolution A to 5.0 ml
Carry out three independent assays. For each individual
with water. Absorbance of the resulting solution at 260 to
titration, calculate the number ofUnits of heparin titrated per
280 nm is not more than 0.1.
mg of the substance under examination. Calculate the result
Heavy metals (2.3.13). 1.0 g complies with the limit test for ofthe assay as the average ofthe 18 values. Test the linearity
heavy metals, Method B (20 ppm). of the response by standard statistical methods. The assay is

1995

www.webofpharma.com
PROTAMINE SULPHATE INJECTION IP 2010

not valid unless the standard deviations calculated for the Light absorption. Dilute the injection, ifnecessary, with water
results obtained with each test solution are less than 5 per to produce a solution containing 1 per cent w/v solution of
cent of the average result. Protamine Sulphate. Absorbance of the resulting solution at
Protamine Sulphate intended for use in the manufacture of 260 to 280 urn, not more than 0.1 (2.4.7).
Parenteral Preparations without a further appropriate Bacterial endotoxins (2.2.3). Not more than 7.0 Endotoxin Units
procedure for the removal of bacterial endotoxins· complies per mg of protamine sulphate.
with the following additional requirement.
Abnormal toxicity. Complies with the test for abnormal toxicity
Bacterial endotoxins (2.2.3). Not more than 7.0 Endotoxin Units (2.2.1), using a volume containing 0.5 mg of Protamine
per mg ofprotamine sulphate~ Sulphate~
Protamine Sulphate intended for use in the mr;lnufacture of Other tests. Complies with the tests stated under Parenteral
Parenteral Preparations without a fitrther appropriate Preparations (Injections).
sterilisation procedure complies with the following
additional requirement. Assay. Prepare solutionsofthe substance under examination
in water containing (1) 0.015 per cent w/v (2) 0.01 per cent
Sterility. Complies with the test for sterility (2.2.11). w/v and (3) 0.005 per cent w/v. Titrate each ofthe solutions in
Storage. Store protected from moisture. If it is intended for duplicate with a 174 IV per ml or a suitable dilution of heparin
use in the manufactUre ofParenteral Preparations, the container sodium RS using the following procedure. Introduce an
should be sterile and sealed so as to exclude micro-organisms. accurately measured volume of the solution to be titrated, for
Labelling. The label states whether or not the contents are example 1.5 ml, into the cell of a suitable spectrophotometer,
intended for use in the manufacture ofParenteral Preparations. set the instrument at a suitable wavelength (none is critical) in
the visible range and add the titrant in small volumes until
there is a sharp increase in the absorbance. Note the volume
of titrant added.
Protamine Sulphate Injection
Carry out three independent assays. For each individual
Protamine Sulphate Injection is a sterile solution ofProtamine titration, calculate the number ofUnits of heparin titrated per
Sulphate in Water for Injections. mg of the substance under examination. Calculate the result
Protamine Sulphate Injection contains not less than 80.0 per ofthe assay as the average ofthe 18 values. Test the linearity
cent of the stated amount of protamine sulphate. of the response by standard statistical methods. The assay is
not valid unless the standard deviations calculated for the
Usual strengths. 50 mg in 5 ml; 100 mg in 10 ml.
results obtained with each test solution are less than 5 per
Identification cent of the average result.
A. Produces a precipitate under the conditions of the Assay. Storage. Store protected from light, in single dose containers.
B. Dilute a suitable volume with water to give a solution Labelling. The label states (1) that the dose is calculated from
containing 0.2 per cent w/v solution of Protamine Sulphate. the results of determinations of the amount required to
To 5 ml of this solution add 1 ml of a 10 per cent w/v solution produce an acceptable blood-clotting time in the patient; (2)
of sodium hydroxide and 1 ml of a 0.02 per cent w/v solution the approximate number of Units of heparin activity 1 ml is
of I-naphthol and mix. Cool to 5° and add 0.5 ml of alkaline capable of neutralising.
sodium hypobromite solution; an intense red colour is
produced.
C. Heat 2 ml on a water-bath at 60°, add 0.1 ml of mercuric Prothionamide
sulphate solution and mix; no precipitate is produced. Cool
the mixture in ice; a white precipitate is produced.
D. Gives reaction A ofsulphates (2.3.1).
Tests
pH (2.4.24).2.5 to 3.5.
Optical rotation (2.4.22). -0.52°to -0.68°, determined in a
solution prepared by diluting the injection with 0.5 M
C9H 1zNzS Mol. wt.180.3
hydrochloric acid so as to contain 0.8 per cent w/v ofProtamine
Sulphate. Prothionamide is 2-propyl-4-pyridinecarbothioamide.

1996

www.webofpharma.com
IP 2010 PROTHIONAMIDE TABLETS

Prothionamide contains not less than 99.0 per cent and not reference solution (0.5 per cent) and the sum ofthe areas ofall
more than 101.0 per cent ofCsHIQN2 S ,calculated on the dried such peaks is not more than twice the area of the principal
basis. peak in the chromatogram obtained with the reference solution
(1.0 percent).
Category. Antitubercular.
Heavy Metals (2.3.13). 2.0 g complies with the limit test for
Description. A yellow, crystalline powder.
heavy metals, Method B (10 ppm).
Identification Sulphated ash (2.3.18). Not more than 0.1 per cent.
A. Detennine by infrared absorption spectrophotometry (2.4.6). Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
Compare the spectrum with that obtained with prothionamide on 1.0 g by drying in an oven at 105° for 3 hours.
RS. Assay. Determine by liquid chromatography (2.4.14) as
B. When examined in the range 230 nm to 350 nm (2.4.7), a described under Related substances using the following
0.002 per cent w/v solution in ethanol (95 per cent) shows an solutions.
absorption maximum only at about 291 nm. The absorbance at Test solution. Dissolve 50.0 mg of the substance under
291 nm is about 0.78. examination in the mobile phase and dilute to 100.0 m1 with the
mobile phase. Dilute 5.0 ml ofthis solution to a 50.0 ml with the
Tests mobile phase.
Acidity. Dissolve 2.0 g in 20 ml of methanol, heating to about Reference solution. A 0.05 per cent w/v solution of
50°, and add 20 ml of water. Cool slightly, shake until prothionamide RS in the mobile phase. Dilute 5.0 ml of the
crystallisation occurs, if any and allow to cool to room solution to 50.0 ml with the mobile phase.
temperature. Add 60 ml of water and titrate with 0.1 M sodium
. Inject the reference solution. The test is not valid unless the
hydroxide using 0.2 ml of cresol redsolution as indicator. Not
tailing factor is not more than 2.0, the column efficiency is not
more than 0.2 ml of 0.1 M sodium hydroxide is required to
less than 5000 theoretical plates and the relative standard
change the colour of the indicator to red.
deviation for replicate injections is not more than 2.0 per cent.
Related substances. Detennine by liquid chromatography
Inject the test solution and the reference solution.
(2.4.14).
Calculate the content ofC 9H I2N 2S.
Test solution. Dissolve 50 mg of the substance under
examination in the mobile phase and dilute to 100 ml with the Storage. Store protected from light and moisture.
mobile phase.
Reference solution. A 0.025 per cent w/v solution of
prothionamide RS in the mobile phase. Dilute 1 ml of the
solution to 100 ml with the mobile phase. Prothionamide Tablets
Chromatographic system Prothionamide Tablets contain not less than 90.0 per cent not
a stainless steel column 25 cm x 4.6 mm, packed with more than 110.0 per cent ofthe stated amount ofprothionamide,
octadecylsilane bonded to porous silica (5 !-tm), C9H I2N2S.
mobile phase: 60 volumes of a buffer solution prepared
Usual strength. 250 mg.
by mixing 2.0 ml of triethylamine with 1000 ml of water,
adjusting the pH to 6.0 with dilute orthophosphoric
Identification
acid and 40 volumes of acetonitrile,
flow rate. I ml per minute, A. Extract a quantity of the powdered tablets containing 25
spectrophotometer set at 290 nm, mg ofProthionamide with 5 rnl of methanol, filter and evaporate
- injection volume. 20 !-tl. the filtrate to dryness. The residue complies with the following
Inject the reference solution.The test is not valid unless the test.
relative standard deviation for replicate injections is not more Detennine by infrared absorption spectrophotometry (2.4.6).
than 2.0 per cent. Compare the spectrum with that obtained with prothionamide
Inject alternatively the test solution and the reference solution. RS.
In the chromatogram obtained with the test solution the area B. In the Assay, the principal peak in the chromatogram
of any individual impurity peak is not more than the area of obtained with the test solution corresponds to the peak in the
the principal peak in the chromatogram obtained with the chromatogram obtained with the reference solution.

1997

www.webofpharma.com
PROTHIONAMIDE TABLETS IP 2010

Tests of water and adjusting the pH to 6.0 with dilute


orthophosphoric acid, and 40 volumes of acetonitrile,
Dissolution (2.5.2).
- flow rate. 1 ml perminute,
Apparatus No.2, - spectrophotometer set at 290 nm,
Medium. 900 ml 0.1 M hydrochloric acid, - injection volume. 20 jll.
Speed and time. 100 rpm and 30 minutes.
Inject the reference solution. The test is not valid unless the
Withdraw a suitable volume ofthe medium and filter. Measure tailing factor is not more than 2.0 the column efficiency is not
the absorbance of the filtered solution, suitably diluted with less than 5000 theoretical plates and the relative standard
the dissolution medium ifnecessary, at the maximum at about deviation for replicate injections is not more than 2.0 per cent.
290 nm (2.4.7). Calculate the content ofC9H 1zNzS in the medium
Inject the test solution and the reference solution.
from the absorbance obtained from a solution of known
concentration of prothionamide RS in the same medium. Calculate the content ofC9H 1zN zS in the tablets.
D. Not less than 75 per cent ofthe stated amount ofC 9H 12N zS. Storage. Store protected from light and moisture.
Related substances. Determine by liquid chromatography
(2.4.14) as described under Assay using the following
solutions. Protriptyline Hydrochloride
Test solution. Weigh accurately a quantity of the powdered
tablets containing 50 mg of Prothionamide disperse in the
mobile phase, shake, dilute to 100 ml with the mobile phase
and filter. , Hel
Reference solution. A solution containing 0.025 per cent w/v
ofprothionamide RS in the mobile phase. Dilute 1.0 ml ofthe
solution to 100.0 ml with the mobile phase.
Inject the reference solution. The test is not valid unless the Mol. Wt. 299.8
relative standard deviation for replicate injections is not more
Protriptyline Hydrochloride is 3-(5H-dibenzo[a,d]cyc1ohept-
than 2.0 per cent.
5-yl)propyl(methyl)amine hydrochloride.
Inject alternatively the test solution and the reference solution.
Protriptyline Hydrochloride contains not less than 99.0 per
In the chromatogram obtained with the test solution the area
cent and not more than 101.0 per cent ofC I9Hz1 N,HCl, calculated
of any individual impurity peak is not more than the area of
on the dried basis.
the peak in the chromatogram obtainxd with the reference
solution (0.5 per cent) and the sum of the areas of all such Category. Tricyclic antidepressant.
impurities is not more than twice the area of the peak in the Description. A white to yellowish white powder.
chromatogram obtained with the reference solution (1.0 per
cent). Identification
Other tests. Comply with the tests stated under Tablets. A. Dissolve 0.1 gin 10 ml of water, make alkaline with 1M
Assay. Determine by liquid chromatography (2.4.14). sodium hydroxide, extract with 5 ml of chloroform, dry with
anhydrous sodium sulphate and evaporate the solvent using
Test solution. Weigh and powder 20 tablets. Weigh accurately
a current of nitrogen. On the oily residue, determine by the
a quantity of the powder containing about 50 mg of
infrared absorption spectrophotometry (2.4:6). Compare the
Prothionamide, disperse in the mobile phase, shake and dilute
spectrum with that obtained with protriptyline hydrochloride
to 100.0 ml with the mobile phase. Dilute 5.0 ml ofthe resulting
RS or with the reference spectrum of protriptyline
solution to 50.0 ml with the mobile phase.
hydrochloride.
Reference solution. A 0.05 per cent w/v solution of
B. Gives reaction A ofchlorides (2.3.1).
prothionamide RS in the mobile phase. Dilute 5.0 ml of the
solution to 50.0 ml with the mobile phase. Tests
Chromatographic system
pH (2.4.24). 5.0to 6.5,determinedina 1.0percentw/v solution.
- a stainless steel Golumn 25 cm x 4.6 mID, packed with
octadecylsilane bonded to porous silica (5 jlm), Loss on drying (2.4.19). Not more than 0.5 per cent, determined
- mobile phase: a mixture of60 volumes ofa buffer solution on 1.0 g by drying in an oven at 60° at a pressure not exceeding
prepared by mixing 2.0 ml of triethylamine with 1000 ml 0.7kPa.

1998

www.webofpharma.com
IP 2010 PSEUDOEPHEDRINE HYDROCHLORIDE

Sulphated ash (2.3.18). Not more than 0.1 per cent. Pseudoephedrine Hydrochloride
Assay. Weigh accurately about 0.7 g, dissolve in 0.5 ml of
acetic anhydride and 20 ml of anhydrous glacial acetic acid, H pH

~
add 10 m1 of mercuric acetate solution. Titrate with 0.1 M CH3
perchloric acid, using clystal violet solution as indicator. I .0 H
' NHCH I HCI
3
Carry out a blank titration.
1 ml of O.lM perchloric acid is equivalent to 0.02998 g of CIOH1sNO,HCl Mol. Wt. 201.7
C I9H21 N,HCl.
Pseudoephedrine Hydrochloride is (lS,2S)-2-methylamino-
I-phenylpropan-1-01 hydrochloride.

Protriptyline Tablets Pseudoephedrine Hydrochloride contains not less than


99.0 per cent and not more than 101.0 per cent ofCIOHlsNO,
Protriptyline Hydrochloride Tablets HCl, calculated on the dried basis.
Protriptyline Tablets contain not less than 90.0 per cent and Category. Sympathomimetic.
not more than 110.0 per cent of the stated amount of
Dose. 60 mg three to four times daily.
protriptyline hydrochloride, C 19H 21 N, HCl.
Description. A white, crystalline powder; odourless or almost
Usual strength. 5 mg.
odourless.
Identification
Identification
A. When examined in the range 230 to 350 nm (2.4.7), a solution
obtained in the Assay exhibits a maximum only at 292 nm. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that· obtained with
B. Shake a quantity of the powdered tablets containing about pseudoephedrine hydrochloride RS or with the reference
5 mg ofProtriptyline Hydrochloride with 20 m1 of methanol spectrum of pseudoephedrine hydrochloride.
and filter. To 1 ml ofthe filtrate add 1 ml of a 2.5 per cent w/v
solution of sodium hydrogen carbonate, 1 ml of a 2 per cent B. When examined in the range 230 nm to 360 nm (2.4.7), a
w/v solution of sodiumperiodate and 1 ml ofa 0.3 per cent wi 0.1 per cent w/v solution shows absorption maxima at about
v solution ofpotassium permanganate. Allow to stand for 15 251 nm, 257 nm and 263 nm; absorbances at the maxima, about
minutes, acidify with 1 M sulphuric acid and extract with 10 0.75, about 0.98 and about 0.78, respectively.
ml of 2,2,4-trimethylpentane, washing the extract with 10 ml C. A 5 per cent w/v solution gives the reactions of chlorides
of 0.25 M sulphuric acid. The absorbance oftrimethyIpentane (2.3.1).
.solution does not exhibit a maxima at 265 nm (2.4.7) (distinction
from amitriptyline and nortriptyline). Tests
C. Triturate a quantity of the powdered tablets containing Appearance of solution. A 5.0 per cent w/v solution is not
about 0.1 g of Protriptyline Hydrochloride with 10 ml of more than very faintly opalescent and colourless (2.4.1).
chloroform, filter and evaporate the filtrate to dryness.
pH (2.4.24). 4.6 to 6.0, determined in a 5.0 per cent w/v solution.
Dissolve part ofthe residue in 3 ml of water and add 0.05 ml of
a 2.5 per cent w/v solution of quinhydrone in methanol. A red Specific optical rotation (2.4.22). +61.0° to +62.so, determined
colour develops. in a 5.0 per cent w/v solution using a 2-dm tube.

Tests Related substances. Determine by liquid chromatography


(2.4.14).
Other tests. Comply with the tests stated under Tablets.
Test solution. Dissolve about 50 mg of the substance under
Assay. Triturate 10 tablets for 15 minutes with 100 ml of a examination in the mobile phase and dilute to 25.0 ml with the
solution prepared by mixing 1 volume of 1 M hydrochloric mobile phase.
acid and 9 volumes of methanol , transfer to a graduated
flask using sufficient ofthe solvent mixture to produce 250 ml, Reference solution (a). Dissolve 20 mg of ephedrine
mix and filter. Dilute a volume ofthe filtrate containing about 1 hydrochloride RS (pseudoephedrine impurity A RS) in 20.0
mg ofProtriptyline Hydrochloride to 100 ml with the solvent ml ofthe mobile phase. Dilute 1.0 ml ofthis solution to 50.0 ml
mixture and measure the absorbance at the maximmn at 292 nm with the mobile phase.
(2.4.7). Calculate the content ofC I 9H2 IN, HCl taking 465 as the Reference solution (b). Dilute 1.0 ml of the test solution to
absorbance. 200.0 ml with the mobile phase.

1999

www.webofpharma.com
PSEUDOEPHEDRINE SYRUP IP 2010

Reference solulion (c). Dissolve 10. mg of ephedrine Pseudoephedrine Syrup contains not less than 90.0 percent
hydrochloride RS (pseudoephedrine impurity A RS) in 5 ml and not more than 110.0 per cent of the. stated amount of
ofthe test solution and dilute to 100 ml with the mobile phase. pseudoephedrine hydrochloride, CIOH1SNO, HCI.
Chromatographic system Usual strength. 30 mg per 5 ml.
- a stainless steel column 25 cm x 4.6 mm, packed with
phenylsilane bonded to porous silica (5 /lm), Identification
mobile phase: a mixture of 6 volumes of methanol and
94 volumes of 1.2 per cent w/v solution of ammonium A. Shake a quantity of the syrup containing 120 mg of
acetate, adjusted to pH 4.0 with glacial acetic acid, Pseudoephedrine Hydrochloride with two quantifies, each of
flow rate. 1 ml per minute, 30 ml, of ether, and discard the ether layer. Add 4 ml of
- spectrophotometer set at 257 nm, 1 M sodium hydroxide to the aqueous layer and extract with
- injection volume. 20 /ll. two quantities, each of 10 ml, of ether. Dry the combined ether
extracts with anhydrous sodium sulphate, filter and evaporate
Inject the test solution, reference solution (a), (b) and (c). Run the filtrate to dryness.
the chromatogram 1.5 times the retention time of
pseudoephedrine. The relative retention time with reference On the residue, determine by infrared absorption
to pseudoephedrine for pseudoephedrine impurity A is about spectrophotometry (2.4.6). Compare the spectrum with that
0.89. The test is not validunless the resolution between the obtained with pseudoephedrine hydrochloride RS treated in
peaks due to pseudoephedrine impurity A and the saame manner or with the reference spectrum of
pseudoephedrine is not less than 2.0. In the chromatogram pseudoephedrine.
obtained with the test solution the area of the peak due to B. The residue obtained in testA melts at about U8° (2.4.21).
pseudoephedrine impurity A is not more than the area of the
principal peak in the chromatogram obtained with reference C. Dissolve 50 mg ofthe residue obtained in testA in 10 ml of
solution (a) (1.0 per cent), the area of any other secondary 0.1 M hydrochloric acid; it is dextro-rotatory.
peak is not more than the area of the principal peak in the
chromatogram obtained with reference solution (b) (0.5 per Tests
cent). The sum of all the secondary peaks other than
Other tests. Complies with the tests stated under Oral Liquids.
pseudoephedrine impurity A is not more than twice the area of
the principal peale in the chromatogram obtained with reference Assay. Determine by liquid chromatography (2.4.14).
solution (b) (1.0 per cent). Ignore any peak with an area less Test solution. To an accurately measured volume of the syrup
than 0.1 times the area oftheprincipal peale in the chromatogram containing about 0.12 g of Pseudoephedrine Hydrochloride,
obtained with reference solution (b) (0.05 per cent). add 50 ml of 0.1 M hydrochloric acid mix, add sufficient
Sulphated ash (2.3 .18). Not more than 0.1 per cent. 0.1 M hydrochloric acid to produce 100.0 ml, filter and use
the filtrate.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105° for 3 hours. Reference solution. A 0.12 per cent w/v solution of
pseudoephedrine hydrochloride RS in 0.1 M hydrochloric
Assay. Weigh accurately about 0.5 g, dissolve in a mixture of acid.
50 ml of anhydrous glacial acetic acid and 10 ml of mercuric
acetate solution. Titrate with 0.1 M perchloric acid, using Chromatographic system
clystal violet solution as indicator. Carry out a blank titration. a stainless steel column 25 cm x 4.2 mm, packed with
octadecylsilane bonded to porous silica (5 /lm),
1 ml of 0.1 M perchloric acid is equivalenttoO.020l7g of mobile phase: a mixture of 85 volumes of ethanoland
CIOH1SNO, HCI. 15 volumes ofa 0.4 per cent w/v solution of ammonium
Storage. Store protected from light and moisture. acetate,
flow rate. 1.5 ml per minute,
spectrophotometer set at 254 nm,
injection volume. 20 /li.
Inject the reference solution and record the chromatogram.
Pseudoephedrine Syrup The test is not valid unless the relative standard deviation is
Pseudoephedrine Hydrochloride Syrup not more than 2.0 per cent and the tailing factor is not more
than 1.5.
Pseudoephedrine Syrup is a solution of Pseudoephedrine
Hydrochloride in a suitable flavoured vehicle. Inject altemately the test solution and the reference solution..

2000

www.webofpharma.com
IP 2010 PSORALEN

Determine the weight per ml ofthe syrup (2.4.29), and calculate Apply to the plate 10 /-Ll of each solution. After development,
the content ofCIOHlsNO, HCl weight in volume. dry the plate in a current of warm air, spray with a solution
containing 0.3 g of ninhydrin in a mixture ofl 00 mIl-butanol
Storage. Store protected from light and moisture.
and 3 ml ofglacial acetic acid and heat at 1200 for 20 minutes.
Any secondary spot in the chromatogram obtained with test
solution (a) is not more intense than the spot in the
Pseudoephedrine Tablets chromatogram obtained with reference solution (a). Ignore
any yellow spot near the line of application.
Pseudoephedrine Hydrochloride Tablets
Other tests. Comply with the tests stated under Tablets.
Pseudoephedrine Tablets contain not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of Assay. Determine by liquid chromatography (2.4.14).
pseudoephedrine hydrochloride, CIOH1SNO, HCI. Test solution. Weigh and powder 20 tablets. To a quantity of
Usual strength. 60 mg. the powdered tablets containing about 0.12 g of
Pseudoephedrine Hydrochloride, add 50 ml of 0.1 M
Identification hydrochloric acid mix with the aid ofultrasound for 15 minutes,
add sufficient methanol to produce 100.0 ml, filter and use the
A. Shake a quantity ofthe powdered tablets containing 60 mg
filtrate.
of Pseudoephedrine Hydrochloride with 10 ml of water and
filter. Shake the filtrate with 10 ml of ether, discard the ether Reference solution. A 0.12 per cent w/v solution of
layer. Add 1 ml of 1 M sodium hydroxide to the aqueous layer pseudoephedrine hydrochloride RS in methanol (50 per
and extract with two quantities, each of 10 ml, of ether. Dry the cent).
combined ether extracts with anhydrous sodium sulphate, Chromatographic system
filter and evaporate the filtrate to dryness. - a stainless steel column 20 cm x 4.6 rom, packed with
On the residue, determine by infrared absorption octadecylsilane bonded to porous silica (10 /-Lm),
spectrophotometry (2.4.6). Compare the spectrum with that - mobile phase: 0.005 M dioctyl sodium sulphosuccinate
obtained with pseudoephedrine hydrochloride RS treated in in a mixture of 65 volumes of methanol, 35 volumes of
the same manner or with the reference spectrum of water and 1 volume of glacial acetic acid,
pseudoephedrine. - flow rate; 1.5 ml per minute,
- spectrophotometer set at 258 om,
B. In the test for Related substances, the principal spot in the
- injection volume. 20 /-Ll.
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution (b). Inject alternately the test solution and the reference solution.

Tests Calculate the content ofC IOH 1sNO,HCI in the tablets.


Storage. Store protected from light and moisture.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
Mobile phase. A mixture of 40 volumes of butyl acetate,
20 volumes of acetone, 20 volumes of I-butanol, 10 volumes Psoralen
of 5 M ammonia and 10 volumes of methanol.
Test solution (a). Add 25 ml of methanol to a quantity ofthe
powdered tablets containing 0.5 g of Pseudoephedrine
Hydrochloride, shake for 5 minutes, filter, wash the filter with
methanol and evaporate the combined filtrate and washings Mol. Wt. 186.2
to dryness. Dissolve the residue as completely as possible in Psoralen is 7H- furo[3 ,2-g][ 1]benzopyran-7-one, obtained
5 ml of methanol, centrifuge and use the supernatant liquid. from the fruits of Psoralea carylifolia Linn. (Fam. Legumi-
Test solution (b). Dilute 1 volume of the test solution to nosae) and from the leaves of Ficus carica (Fam.
10 volumes with methanol. Urticaceae) or prepared by synthesis.

Reference solution (a). Dilute 1 volume ofthe test solution to Psoralen contains not less than 95.0 per cent and not more
100 volumes with methanol. than 101.0 per cent ofC Il H 60 3 , calculated on the dried basis.

Reference solution (b). A 1.0 per cent w/v solution of Category. Topical pigmenting agent.
pseudoephedrine hydrochloride RS in methanol. Description. Colourless needles; odourless.

2001

www.webofpharma.com
PSORALEN IP 2010

Identification Pyrantel Pamoate


A. Dissolve 1 mg in 5 ml of ethanol (95 per cent) and add Pyrantel Embonate
15 ml ofamixture containing 43 volumes of water, 5 volumes
of acetic acid and 3 volumes of propylene glycol; a blue COOH
fluorescence is visible in ultraviolet light at 365 nm.
B. Dissolve 1 mg in 2 ml of ethanol (95 per cent) and add OH
0.1 ml of 0.1 M sodium hydroxide; a yellow fluorescence is
visible in ultraviolet light at 365 nm. OH
Tests
COOH
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G CIIHI4NzS, CZ3HI606 Mol. Wt. 594.7

Mobile phase. A mixture of 90 volumes of benzene and 10 Pyrantel Pamoate is 1,4,5,6-tetrahydro-1-methyl-2-[(E)-2-(2-


volumes of ethyl acetate. thienyl)vinyl]pyrimidine hydrogen; 4,4'-methylene bis(3-
hydroxynaphthalene-2-carboxylate).
Test solution. Dissolve 0.2 g of the substance under
Pyrantel Pamoate contains not less than 98.0 per cent and not
examination in 10 ml of chloroform.
more than 102.0 per cent of C34H30Nz06S, calculated on the
Reference solution. Dilute 1 volume ofthe test solution to 100 dried basis.
volumes with chloroform. Category. Anthelmintic.
Apply to the plate 5 /-ll of each solution. After development, Dose. 10 mg per kg
dry the plate in air and examine in ultraviolet light at 365 nm. Description. A pale yellow or yellow powder.
Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the Identification
chromatogram obtained with the reference solution.
Determine by infrared absorption spectrophotometry (2.4.6).
Sulphated ash (2.3.18). Not more than 0.1 percent. Compare the spectrum with that obtained with pyrantel
Loss on drying (2.4.19). Not more than 1.0 per cent, determined pamoate RS or with the reference spectrum of pyrantel
on 1.0 g by drying in an oven at 105°. pamoate.

Assay. Weigh accurately about 0.1 g and dissolve in sufficient Tests


methanol to produce 100.0 ml. Dilute 2.0 ml ofthis solution to
Related substances. Determine by liquid chromatography
100.0 ml with methanol and measure the absorbance of the
(2.4.14).
resulting solution at the maximum at about 247 nm(2.4.7).
NOTE- Prepare the solutions immediately before use and
Calculate the content ofC 11 H60 3from the absorbance obtained protectedii'om light.
by repeating the operation using a final solution of 20 /-lg per
Solvent mixture. Mix 25 volumes of glacial acetic acid, 25
ml ofpsoralen RS in methanol in place ofthe substance under
volumes of water and 10 volumes of diethylamine with cooling.
examination.
Test solution. Dissolve 80 mg of the substance under
Storage. Store protected from light and moisture. examination in 7 ml ofthe solvent mixture and dilute to 100.0
ml with mobile phase. .
Reference solution (a). Dissolve 10.0 mg of 1-methyl-2-[(Z)-
2-(thiophen-2-yl)-1,4,5,6-tetrahydropyrimidine RS (pyrantel
impurity A RS) in the solvent mixture, add 2.5 ml of the test
solution and dilute to 50.0 ml with the mobile phase. Dilute 2.0
ml ofthis solution to 100.0 ml with the mobile phase.
Reference solution (b). Dilute 1.0 ml of the test solution to
200.0 ml with the mobile phase.
Chromatographic system
a stainless steel column 25 cm x 4.6 rom, packed with
silica gel (5 /-lm),

2002

www.webofpharma.com
IP 2010 PYRANTEL PAMOATE ORAL SUSPENSION

- mobile phase: 7.2 volumes of the solvent mixture and for 10 minutes. Allow to cool (a clear solution is not obtained).
92.8 volumes of acetonitrile, Titrate with 0.1 M perchloric acid, determining the end-point
- flow rate. I ml per minute, potentiometrically (2.4.25). Carry out a blank titration. .
- spectrophotometer set at 288 mn,
1 ml of 0.1 M perchloric acid is equivalent to 0.05947 g of
- injection volume. 20 Ill.
C34H30Nz06S,
Inject reference solution (a). Run the chromatogram for 4 times
Storage. Store protected from light.
the retention time of pyrantel. The test is not valid unless the
resolution between the peaks due to pyrantel and pyrantel
impurity A is at least 4.0. The relative retention times with
reference to pyrantel pamoate are about 0.5 for pamoic acid,
Pyrantel Pamoate Oral Suspension
about 1.3 for pyrantel impurity A and about 1.8 for ((E)-N- Pyrantel Pamoate Oral Suspension is a suspension ofPyrantel
{methylamino)propyl]-3-(thiophen-2-yl)prop-2-enamide) Pamoate in a suitable flavoured aqueous vehiCle.
pyrantel impurity B.
Pyrantel Pamoate Oral Suspension contains not less than 90.0
Inject the test solution, reference solutions (a) and (b). For the per cent and not more than 110.0 per cent ofthe stated amount
calculation of content, multiply the peak area of pyrantel ofpyrantel, CIIHI4NzS.
impurity B by 0.4. In the chromatogram obtained with the test
solution, the area ofthe peak due to pyrantel impurity A is not Usual strength. 50 mg per ml
more than the corresponding peak in the chromatogram Identification
obtained with reference solution (a) (0.5 per cent); the area of
the peak due to pyrantel impurity B is not more than the area A. Determine by thin-layer chromatography (2.4.17), coating
of the principal peak in the chromatogram obtained with the plate with silica gel H.
reference solution (b) (0.2 per cent); the area any other Mobile phase. The upper layer ofa mixture obtained by shaking
secondary peak is not more than 0.2 times the area of the 20 volumes of methyl isobutyl ketone, 10 volumesofJormic
principal peak in the chromatogram obtained with the reference acid and 10 volumes of water.
solution (b) (0.1 per cent) and the sum of the areas of all the
secondary peaks other than those of pyrantel impurities A Test solution. Dilute a known volume of the oral suspension
and B is not more than 0.6 times the area ofthe principal peak with sufficient 0.05 M methanolic ammonia to produce a
in the chromatogram obtained with the reference solution (b) solution containing about 8 mg ofpyrantel per ml. Shake the
(0.3 per cent). Ignore any peak with an area less than 0.1 times mixture by mechanical means and centrifuge. Use the clear
the area of the principal peak in the chromatogram obtained supernatant liquid.
with reference solution (b) (0.05 per cent). ReJerencesolution. A solution of pyrantel pamoate RS in
Chlorides (2.3.12). To 0.7 g, add 10 ml of dilute nitric acid 0.05 M methanolic ammonia containing 0.008 per cent w/v of
and 30 ml of water. Heat on water-bath for 5 minutes and cool. pyrantel.
The resulting solution complies with the limit test for chlorides Apply to the plate 100 III of each solution. Allow the mobile
(360 ppm). phase to rise to about 17 cm. Dry the plate in air and examine
Sulphates (2.3.17). To 0.5g, add 2.5 ml of dilute nitric acid in ultraviolet light at 365 nm. The principal spot in the
and dilute to 50 ml with water. Heat on water bath for 5 minutes. chromatogram obtained with the test solution corresponds to
15 ml complies with the limit test for sulphates (0.1 per cent). that in the chromatogram obtained with the reference solution.
B. In the Assay, the major peaks in the chromatogram obtained
Iron (2.3.14). Ignite 0.54 g at 8000 for 2 hours. Dissolve the
with the test solution correspond to the peaks in the
residue in 2.5 ml of dilute hydrochloric acid with gentle heating
chromatogram obtained with the reference solution.
for 10 minutes and cool. The resulting solution complies with
the limittest for iron (75 ppm).
Tests
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method B (l0 ppm). pH (2.4.24). 4.5 to 6.0
Other tests. Complies with the tests stated under Oral Liquids.
Sulphated ash (2.3.1 8). Not more than 0.1 per cent.
Assay. Determine by liquid chromatography (2.4.14).
Loss on drying (2.4.1 9). Not more than 1.0 per cent, determined
on 1.0 g by drying in vacuum at 60 0 for 3 hours. NOTE- Carry out the test protectedfi-om light.
Assay. Weigh accurately about 0.5 g, add 10 ml of acetic Test solution. Weigh accurately a quantity of the oral
anhydride and 50 ml glacial acetic acid. Heat at 500 and stir suspension containing about 60 mg of pyrantel, disperse in

2003

www.webofpharma.com
PYRANTELPAMOATE ORAL SUSPENSION IP 2010

60 ml of water and add sufficient water to produce 100.0 m!. Dose. Up to 35 mg per kg of body weight daily, in divided
Dilute 1.0 ml ofthe well-stirred susptmsion to 25.0 ml with the doses.
mobile phase to obtain a clear solution. Mix and filter.
Description. A white or almost white, crystalline powder;
Reference solution. Weigh accurately about 20 mg ofpyrantel odourless or almost odourless.
pamoate RS and dissolve in sufficient mobile phase to produce
25.0 ml. Dilute 10.0 ml ofthe resulting solution to 100.0 ml with Identification
the mobile phase and mix.
Test A may be omitted iftests Band C are carried out. Tests B
Chromatographic system and C may be omitted if test A is carried out.
- a stainless stefllcolumn 25 cm x4.6 mill, packed with
A. Determine by infrared absorption spectrophotometry (2.4.6).
porous siiica particles (5 to 10 Ilm),
Compare the spectrum with that obtained with pyrazinamide
mobile phase: a mixture of 92.8 volumes of acetonitrile,
RS or with the reference spectrum ofpyrazinamide.
3 volumes of acetic acid, 3 volumes of water and 1.2
volumes of diethylamine, B. Dissolve 50 mg in water and dilute to 100 ml with the same
- flow rate. 1 mlper minute, solvent (solution A). Dilute 1 ml ofsolution A to 10 m!. When
- spectrophotometer set at 288 nm, examined in the range 290 om to 360 om (2.4.7), the resulting
- injection volume. 20 Ill. solution shows an absorption maximum at about 310 om. Dilute
Inject the reference solution. The resolution between pyrantel 2 ml ofsolution A to 100 ml with water. When examined in the
and pamoic acid is not less than 10.0, the column efficiency range 230 om to 290 om, the solution shows an absorption
for the pyrantel peak is not less than 8000 theoretical plates, maximum at about 268 om; absorbance at about 268 om,
the tailing factor for the pyrantel peak is not greater than 1.3 between 0.64 and 0.68.
and the relative standard deviation for replicate injections is C. Boil 20 mg with 5 ml ofsodium hydroxide solution; ammonia,
not more than 2.0 per cent. recognisable by its odour, is evolved.
Inject the test solution and t4e reference solution. Continue
Tests
the chromatography for a period not less than 2.5 times the
retention times of pyrantel base. The relative retention times Appearance ofsolution. A 1.0 per cent w/v solution in carbon
for pamoic acid and pyrantel base are about 0.6 and 1.0 dioxide-free water (solution B) is clear (2.4.1), and colourless
respectively. (2.4.1).
Determine the weight per ml (2.4.29) of the suspension and Acidity or alkalinity. To 25 ml of solution B add 0.05 mlof
calculate the content OfCIIHI4NzS, weight in volume. phenolphthalein solution and 0.2 ml of 0.01 M sodium
Storage. Store protected from light at a temperature not hydroxide; the solution is red. Add 1 ml of 0.01 Mhydrochloric
exceeding 30°. acid; the solution is colourless. Add 0.15 ml of methyl red
solution; the solution is red.
Labelling. The label states the strength in terms of the
Related substances. Determine by thin-layer chromatography
equivalent amount of pyrantel.
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 60 volumes of 1-butanol,
20 volumes of glacial acetic acid and 20 volumes of water.
Pyrazinamide Test solution. Dissolve 0.1 g of the substance under
examination in 10 ml ofa mixture of90 volumes of chloroform
and 10 volumes of methanol.
Reference solution. A 0.002 per cent w/v solution of the
substance under examination in a mixture of 90 volumes of
chloroform and 10 volumes of methanol.
Mol. Wt. 123.1 Apply to the plate 20 III of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 om.
Pyrazinamide is pyrazine-2-carboxamide. Any secondary spot in the chromatogram obtained with the
Pyrazinamide contains not less than 99.0 per cent and not test solution is not more intense than the spot in the
more than 100.5 per cent of CsHsN'lO; calculated on the chromatogram obtained with the reference solution.
anhydrous basis. Heavy metals (2.3.13). 2.0 g complies with the limit test for
Category. Antitubercular. heavy metals, Method B (10 ppm).

2004

www.webofpharma.com
IP 2010 PYRIDOXINE HYDROCHLORIDE

Sulphated ash (2.3.18). Not more than 0.1 per cent. Mobile phase. A mixture of 60 volumes of l-butanol,
20 volumes of glacial acetic acid and 20 volumes of water.
Water (2.4.19). Not more than 0.5 per cent, determined on
5.0g. Test solution. Shake a quantity of the powdered tablets
Assay. Weigh accurately about 0.3 g and transfer to the flask containing 0.1 g ofPyrazinamide with 50 mr ofa mixture of90
ofan ammonia distillation apparatus. Add 200 ml of water and volumes of chloroform and 10 volumes of methanol, filter,
75 ml ofsodium hydroxide solution. Boil gently for 20 minutes, evaporate to dryness and dissolve the residue in sufficient of
collecting the distillate in 50.0 ml of 0.05 M sulphuric acid. the same solvent mixture to produce 10 ml.
Boil vigorously to complete the distillation of the ammonia Reference solution. Dilute I volume of the test solution to
and titrate the excess of acid with 0.1 M sodium hydroxide, 500 volumes with a mixture of90 volumes of chloroform and
using methyl red solution as indicator. Repeat the operation 10 volumes of methanol.
without the substance under examination. The difference Apply to the plate 20 III of each solution. After development,
between the titrations represents the amount of acid required dry the plate in air and examine in ultraviolet light at 254 nm.
to neutralise the ammonia formed. Any secondary spot in the chromatogram obtained with the
I ml of 0.05 M sulphuric acid is equivalent to 0.01231 g of test solution is not more intense than the spot in the
CsHsN30. chromatogram obtained with the reference solution.
Storage. Store protected from moisture. Other tests. Comply with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity ofthe powder containing about 0.1 g ofPyrazinamide,
Pyrazinamide Tablets add 200 ml of water, allow to stand for 10 minutes, swirling
occasionally, mix with the aid ofultrasound for 10 minutes and
Pyrazinamide Tablets contain not less than 95.0 per cent and dilute to 500.0 ml with water. Filter and discard the first 20 ml
not more than 105.0 per cent of the stated amount of ofthe filtrate. Dilute 5.0 ml ofthe filtrate to 100.0 ml with water
pyrazinamide, CsH sN 30. and measure the absorbance of the resulting solution at the
Usual strengths. 250 mg; 500 mg; 750 mg; 1000 mg. maximum at about 268 nm (2.4.7). Calculate the content of
CsH sN 30 taking 650 as the specific absorbance at 268 nm.
Identification
Storage. Store protected from moisture.
A. Shake a quantity ofthe powdered tablets containing 0.25 g
of Pyrazinarriide with 20 ml of ethanol, filter, evaporate the
filtrate to dryness and my the residue at 105° for 30 minutes.
On the residue, determine by infrared absorption Pyridoxine Hydrochloride
spectrophotometry (2.4.6). Compare the spectrum with that VitaminB 6
obtained with pyrazinamide RS or with the reference spectrum
ofpyrazinamide. .
B. Shake a quantity ofthe powdered tablets containing 50 mg
, Hel
of Pyrazinamide with 50 ml of watel; dilute to 100 ml with
water and filter (solution A). Dilute I ml ofsolution A to 10 ml
with water. When examined in the range 290 nm to 360 nm
(2.4.7), the resulting solution shows an absorption maximum
at about 310 nm. Dilute 2 rnIofsolutionAto 100mi with water. CgH lI N03,HCI Mol. Wt. 205.6
When examined in the range 230 nm to 290 nm, the resulting
Pyridoxine Hydrochloride is 5-hydroxy-6-methylpyridine-
solution shows an absorption maximum at about 268 nm;
3,4-dimethanol hydrochloride.
absorbance at 268 nm, between 0.64 and 0.68.
Pyridoxine Hydrochloride contains not less than 99.0 per cent
C. Boil a quantity ofthe powdered tablets containing 20 mg of
and not more than 101.0 per cent ofCgH II N0 3,HCI, calculated
Pyrazinamide with 5 ml of sodium hydroxide solution;
on the dried basis.
ammonia, recognisable by its odour, is evolved.
Category. B group vitamin; sideroblastic anaemia therapy;
Tests anti-isoniazid neuropathy.
Related substances. Determine by thin-layer chromatography Dose. In deficiency states, prophylactic, upto 2 mg daily;
(2.4.17), coating the plate with silica gel GF254. therapeutic, 20 to 50 mg upto three times daily. In multi- vitamin

2005

www.webofpharma.com
PYRIDOXINE HYDROCHLORIDE IF 2010

preparations for oral use, prophylactic, 500 J.tg to 1.5 mg daily; Apply to the plate 2 J.tl of each solution. After development,
therapeutic, 1.5 to 3 mg daily. In sideroblastic anaemia, 100 to dry the plate in air and spray with a 5 per cent w/v solution of
400 mg daily, in divided doses. In isoniazid neuropathy, sodium carbonate in a mixture of 70 volumes of water and
prophylactic, 10 mg daily; therapeutic, 50 mg three times daily. 30 volumes of ethanol (95 per cent). Dry in a current of air,
spray with a 0.1 per cent w/v solution of 2,6-dichloroquinone-
Description. A white or almost white, crystalline powder;
4-chloroimide in ethanol (95 per cent) and examine
odourless.
immediately. Any secondary spot in the chromatogram
Identification obtained with test solution (a) is not more intense than the
spot in the chromatogram obtained with reference solution
Test A may be omitted iftests B, C andD are carried out. Tests (a); Ignore any spots remaining on the line of application.
B may be omitted if tests A, C and D are carried out.
Heavy metals (2.3.13). 12 ml of a 5.0 per cent w/v solution
A. Determine by infrared absorption spectrophotometry (2.4.6).
complies with the limit test for heavy metals, Method D
Compare the spectrum with that obtained with pyridoxine
(20 ppm). Use lead standard solution (1 ppm Pb) to prepare
hydrochloride RS or with the reference spectrum ofpyridoxine
the standard.
hydrochloride.
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
B. When examined in the range 250 nm to 360 nm (2.4.7), a
0.001 per cent w/v solution in 0.1 Mhydrochloric acid shows Loss on drying (2.4.19). Not more than 0.5 per cent, determined
an absorption maximum at 288 nm to 296 nm; absorbance at on 1.0 g by drying in an oven at 105°.
the maximum, 0.420 to 0.445. A solution prepared by diluting Assay. Weigh accurately about 0.15 g, dissolve in a mixture of
1 ml of a 0.1 per cent w/v solution in 0.1 M hydrochloric acid 5 ml of anhydrous glacial acetic acid and 6 ml of mercuric
to 100 ml with 0.025 M standard phosphate buffer shows acetate solution. Titrate with 0.1 M perchloric acid, using
absorption maxima at 248 nm to 256 nm and at 320 nm to crystal violet solution as indicator, until a green colour is
327 nm; absorbances at the maxima, 0.175 to 0.195 and 0.345 to produced. Carry out a blank titration.
0.365, respectively.
1 ml of 0.1 M perchloric acid is equivalent to 0.02056 g of
C. In the test for Related substances, the principal spot in the CsH II N03,HCI.
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution Storage. Store protected from moisture.
(b).
D. A 5 per cent w/v solution gives reaction A of chlorides
(2.3.1). Pyridoxine Tablets
Tests Pyridoxine Hydrochloride Tablets
Appearance of solution. A 5.0 per cent w/v solution is clear Pyridoxine Tablets contain not less than 90.0 per cent and not
(2.4.1), and not more intensely coloured than reference solution more than 110.0 per cent ofthe stated amount of pyridoxine
YS7 (2.4.1). hydrochloride, C SH II N03, HCI.

pH (2.4.24).2.4 to 3.0, determined in a 5.0 per cent w/v solution. Usual strength. 5 mg.

Related substances. Determine by thin-layer chromatography Identification


(2.4.17), coating the plate with silica gel G
A. Shake a quantity ofthe powdered tablets containing 20 mg
Mobile phase. A mixture of65 volumes of acetone, 13 volumes ofPyridoxine Hydrochloride with 50 ml of 0.025 M standard
ofdichloroinethane, 13 volumes of tetrahydrofuran and jJhosjJhcie lJUfferfor i 5 m.inutes and dllute to 16b m.l with the
9 volumes of strong ammonia solution. same solvent. Mix, filter and dilute 5 ml ofthe filtrate to 100 ml
Test solution (a). A 10 per cent w/v solution of the substance with the same solvent. When examined in the range 230 nm to
under examination in water. 360 nm (2.4.7), the resulting solution exhibits two maxima, at
about 254 nm and 324 nm.
Test solution (b). A 1 per cent w/v solution of the substance
under examination water. B. Triturate a quantity of the powdered tablets containing
20 mg of Pyridoxine Hydrochloride with 50 ml of water and
Reference solution (a). A 0.025 per cent w/v solution of the allow to stand for 20 minutes. To 1 ml ofthe supernatant liquid
substance under examination in water. add 10 ml ofa 5 per cent w/v solution of sodium acetate,lilll
Reference solution (b). A 1 per cent w/v solution ofpyridoxine of water and 1 ml of a 0.5 per cent w/v solution of 2,6-di-
hydrochloride RS in water. chloroquinone-4-chloroimide in ethanol (95 per cent) and

2006

www.webofpharma.com
IP 2010 PYRIMETHAMINE

shake; a blue colour is produced which fades rapidly and Storage. Store protected from light and moisture.
becomes brown. Repeat the operation but adding 1 ml of a
0.3 per cent w/v solution of boric acid in place of 1 ml of
water; no blue colour is produced.
Pyrimethamine
Tests
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G
Mobile phase. A mixture of65 volumes of acetone, 13 volumes
NH 2
of dichloromethane, 13 volumes of tetrahydrofuran and CI
9 volumes of strong ammonia solution.
Mol. Wt. 248.7
Test solution. Shake a quantity of the powdered tablets
Pyrimethamine is 5-(4-cWorophenyl)-6-ethylpyrimidine-2,4-
containing 40 mg of Pyridoxine Hydrochloride with 10 ml of
diamine.
water for 15 minutes, filter and use the filtrate.
Pyrimethamine contains not less than 99.0 per cent and not
Reference solution. Dilute 1 ml oftest solution to 200 ml with
more than 101.0 per cent OfCI2HI3CIN4, calculated onthe dried
water.
basis.
Apply to the plate 10 J.11 of each solution. After development,
Category. Antimalarial.
dry the plate in air and spray with a 5 per cent w/v solution of
sodium carbonate in a mixture of 70 volumes of water and Dose. Used only in combination with sulphadoxine (25 mg
30 volumes of ethanol (95 per cent). Dry it in a current of air, pyrimethamine and 500 mg sulphadoxine) or dapsone (12.5
spray with a 0.1 per cent w/v solution of 2, 6-dichloroquinone- mg pyrimethamine and 100 mg dapsone).
4-chloroimide in ethanol (95 per cent) and examine
Description. Colourless crystals or an almost white, crystalline
immediately. Any secondary spot in the chromatogram
powder; odourless.
obtained with the test solution is not more intense than the
spot in the chromatogram obtained with the reference solution. Identification
Uniformity of content. Comply with the test stated under Test A may be omitted if tests Band C are carried out. Tests B
Tablets. and C may be omitted if test A is carried out.
Powder one tablet, add 50 ml of 0.1 M hydrochloric acid and A. Determine by infrared absorption spectrophotometry (2.4.6).
heat on a water-bath for 15 minutes, swirling occasionally. Compare the spectrum with that obtained with pyrimethamine
Cool, dilute to 100.0 ml with 0.1 M hydrochloric acid, filter, RS or with the reference spectrum ofpyrimethamine.
discarding the first 20 ml of the filtrate. If necessary, dilute
quantitatively and stepwise with 0.1 M hydrochloric acid to B. In the test for Related substances, the principal spot in the
produce a solution containing 10 J.1g of the pyridoxine chromatogram obtained with test solution (b) corresponds to
hydrochloride per ml and measure the absorbance of the that in the chromatogram obtained with reference solution (b).
resulting solution at the maximum at about 290 nm (2.4.7). C. Dissolve 0.14 g in sufficient ethanol to produce 100 ml,
Calculate the content of C SH 11 N03, HCl taking 430 as the dilute 10 ml ofthis solution to 100 ml with 0.1 Mhydrochloric
specific absorbance at 290 nm. acid and dilute 10 ml ofthe solution to 100 ml with the same
Other tests. Comply with the tests stated under Tablets. solvent. When examined in the range 230 nm to 360 nm (2.4.7),
the resulting solution shows an absorption maximum at about
Assay. Weigh and powder 20 tablets. Weigh accurately a 272 nm; absorbance at about 272 nm, 0.43 to 0.46.
quantity ofthe powder containing about 20 mg ofPyridoxine
Hydrochloride, add 50 ml of 0.1 M hydrochloric ac,id and Tests
heat on a water-bath for 15 minutes, swirling occasionally.
Cool, dilute to 100.0 ml with 0.1 M hydrochloric acid and Appearance ofsolution. Dissolve 0.25 g in sufficient ofa mixture
filter, discarding the first 20 ml ofthe filtrate. Dilute 5.0 ml of of 3 volumes of dichloromethane and 1 volume of methanol
the filtrate to 100.0 ml with 0.1 M hydrochloric acid and to produce 10 ml. The resulting solution is clear (2.4.1), and
measure the absorbance of the resulting solution at the not more intensely coloured than reference solution BYS6
maximum at about 290 ~m (2.4.7). Calculate the content of (2.4.1).
C SH 11 N0 3, HCl taking 430 as the specific absorbance at Acidity or alkalinity. Shake 1.0 g with 50 ml of water for
290nm. 2 minutes and filter (solution A). To 10 ml of solution A add

2007

www.webofpharma.com
PYRIMETHAMINE IP 2010

0.05 ml ofphenolphthalein solution; the solution is colourless Identification


and not more than 0.2 ml of 0.01 M sodium hydroxide is
required to change the colour to pink. Add 0.4 ml of 0.01 M A. Shake a quantity ofthe powdered tablets containing 50 mg
hydrochloric acid and 0.05 ml of methyl red solution; the of Pyrimethamine with 50 ml of ethanol (95 per cent) for 20
solution is red or orange. . minutes, filter and evaporate the filtrate to dryness. On the
residue, determine by infrared absorption spectrophotometry
Related substances. Determine by thin-layer chromatography (2.4.6). Compare the spectrum with that obtained with
(2.4.17), coating the plate with silica gel GF254. pyrimethamine RS or with the reference spectrum of
Mobile phase. A mixture of76 volumes of toluene, 12 volumes pyrimethamine.
of glacial acetic acid, 8 volumes of I-propanol and 4 volumes
B. Extract the powdered tablets with 1 M sulphuric acid; filter
of chloroform.
and add potassium tetraiodomercurate solution to the filtrate.
Prepare the following solutions immediately before use. A creamy white precipitate is produced.
Test solution (a). Dissolve 0.1 g of the substance under C. Extract a quantity ofthe powdered tablets containing 50 mg
examination in 10 ml ofa mixture of90 volumes of chloroform ofPyrimethamine with two 10 ml quantities of chloroform and
and 10 volumes of methanol. evaporate the combined extracts to dryness. The residue melts
Test solution (b). Dissolve 0.1 g of the substance under at about 240 0 (2.4.21).
examination in 100 ml ofthe same solvent mixture.
Tests
Reference solution (a). A 0.0025 per cent w/v solution of the
substance under examination in the same solvent mixture. Related substances. Determine by thin-layer chromatography
Reference solution (b). A 0.1 per cent w/v solution of (2.4.17), coating the plate with silica gel GF254.
pyrimethamine RS in the same solvent mixture. Solvent mixture. 90 volumes of chloroform and 10 volumes of
Apply to the plate 20 !J.l of each solution. After development, methanol.
dry the plate in air and examine in ultraviolet light at 254 urn.
Mobilephase. A mixture of4 volumes of chloroform, 8 volumes
Any secondary spot in the chromatogram o15tained with test
of I-propanol, 12 volumes of glacial acetic acid and 76
solution (a) is not more intense than the spot in the
volumes of toluene.
chromatogram obtained with reference solution (a).
Test solution. Shake a quantity of the powdered tablets
Sulphates (2.3.17). 25 ml ofSolution A complies with the limit
containing 50 mg Pyrimethamine with 5 ml ofsolvent mixture
test for sulphates. Use a mixture of5.0 ml ofsulphate standard
and filter.
solution (10 ppm SO4) and 10 ml of distilled water to prepare
the standard (100 ppm). Reference solution. Dilute 1 ml ofthe test solution to 400 ml
Sulphated ash (2.3.18). Not more than 0.1 per cent. with the solvent mixture.

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Apply to the plate 20 !J.l of each solution. Allow the mobile
on 1.0 g by drying in an oven at 105 0 for 4 hours. phase to rise 10 cm. After development, dry the plate in air and
examine in ultraviolet light at 254 urn. Any secondary spot in
Assay. Weigh accurately about 0.2 g, dissolve in 25 ml of
the chromatogram obtained with the test solution is not more
anhydrous glacial acetic acid, heating gently, cool. Titrate
intense than the spot in the chromatogram obtained with the
with 0.1 M perchloric acid, determining the end-point
reference solution (0.25 per cent).
potentiometrically (2.4.25). Carry out a blank titration.
Other tests. Comply with the tests stated under Tablets.
LlTIlofO.lA1perchloric a9id is equivalent to 0.02487g of
C I2H 13ClN4. Assay. Weigh and powder 20 tablets. Weigh accurately a
Storage. Store protected from light and moisture. quantity of the powder containing about 0.025g of
Pyrimethamine, add 50 ml ofhot 0.1 M hydrochloric acid and
heat on a water-bath for 10 minutes, swirling occasionally.
Mix with the·aid of ultrasound for 30 minutes, remove and
Pyrimethamine Tablets cool to room temperature. Add sufficient 0.1 M hydrochloric
acid to produce 100 ml, filter and dilute 5 ml of the filtrate to
Pyrimethamine Tablets contain not less than 92.5 per cent and
100 ml with 0.1 M hydrochloric acid and measure the
not more than 107.5 per cent of the stated amount of
absorbance 6ftheresulting solution at the maxiIllUlTI at about
pyrimethamine, C I2H 13 ClN4.
272 urn (2.4.7). Calculate the content OfC12HI3ClN4 taking 316
Usual strength. 25 mg. as the specific absorbance at 272 urn.

2008

www.webofpharma.com
IP 2010 PYRIMETHAMINE AND SULPHADOXINE TABLETS

Pyrimethamine and Sulphadoxine Assay. Determine by liquid chromatography (2.4.14).

Tablets Test solution. Weigh accurately a quantity of the powdered


tablets containing about 25 mg ofPyrimethamine and 500 mg
Pyrimethamine and Sulphadoxine Tablets contain not less than of Sulphadoxine and shake with 35 ml of acetonitrile for
90.0 per cent and not more than 110.0 per cent of the stated 30 minutes in a 1OO-ml volumetric flask. Dilute to volume with
amounts ofpyrimethamine, C I2H 13 CIN4, and ofsulphadoxine, the mobile phase, mix and filter. To 25.0 ml ofthe filtrate add
CI2H14N404S. 2.0 ml ofsolution A prepared by dissolving 0.1 g ofphenacetin
Usual strength. Pyrimethamine 25 mg and sulphadoxine (internal standard) in 100 ml of acetonitrile and sufficient of
500 mg. the mobile phase to produce 50.0 ml.

Identification Reference solution. Weigh accurately about 25 mg of


pyrimethamine RS and 500 mg of sulphadoxine RS, add 35 ml
Determine by thin-layer chromatography (2.4.17), coating the of acetonitrile and sufficient of the mobile phase to produce
plate with silica gel G 100.0 ml and mix. To 25.0 ml add 2.0 ml ofsolution Aand add
Mobile phase. A mixture of 4 volumes of chloroform, sufficient ofthe mobile phase to produce 50.0 ml and mix well.
4 volumes of n-heptane, 1 volume of glacial acetic acid and
4 volumes ofa mixture of! volume of methanol and 19 volumes Chromatographic system
of ethanol. - a stainless steel column 30 cm x 4 mm, packed with
Test solution. Shake a quantity of the powdered tablets octadecylsilane bonded to porous silica (5 Ilm),
containing 25 mg ofPyrimethamine with 50 ml ofa 2 per cent mobile phase: a mixture of 4 volumes ofa 1 per cent v/v
w/v solution of strong ammonia solution in methanol. solution of glacial acetic acid and 1 volume of
acetonitrile,
Reference solution (a). A 0.05 per cent w/v solution of flow rate. 2 ml per minute,
pyrimethamine RS in methanol.
spectrophotometer set at 254 nm,
Reference solution (b). A 1.0 per cent w/v solution of injection volume. 20 Ill.
sulphadoxine RS in methanol.
Inject the reference solution. The test is not valid unless the
.Apply to the plate 10 III of each solution. After development, relative standard deviation for replicate injections is not more
dry the plate in air and examine in ultraviolet light at 254 llill. than 2.0 per cent. The relative retention times with reference
One ofthe principal spots in the chromatogram obtained with
to phenacetin for sulphadoxine is about 0.7 and for
the test solution corresponds to the spot in the chromatogram
pyrimethamine is about 1.3.
obtained with reference solution (a) and the other corresponds
to that in the chromatogram obtained with reference Injectthe test solution and the reference solution.
solution (b).
Calculate the content of CI2HI3ClN4 and the content of
Tests C12HI4N404S in the tablets.

Other tests. Complies with the tests stated under Tablets. Storage. Store protected from light and moisture.

2009

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Quetiapine Fumarate 2013


Quetiapine Tablets 2014
Quinalbarbitone Sodium 2014
Quinalbarbitone Tablets 2015
Quinidine Sulphate 2016
Quinidine Tablets 2017
Quinine Bisulphate 2019
Quinine Bisulphate Tablets 2020
QuinineDihydrochloride 2021
Quinine Dihydrochloride Injection 2023
Quinine Sulphate 2025
Quinine Tablets 2026
Quiniodochlor 2027
Quiniodochlor Cream 2028
Quiniodochlor Ointment 2029
QuiniodochlorTablets 2030
Quiniodochlor and Hydrocortisone Cream 2031
Quiniodochlor and Hydrocortisone Ointment 2032

2011

www.webofpharma.com
www.webofpharma.com
IP 2010 QUETIAPINE FUMARATE

Quetiapine Fumarate Reference solution (b). A 0.05 per cent w Iv solution offumariC
acid in the mobile phase.
Reference solution (c). Dissolve 1 mg of quetiapinefumarate
RS and 1.5 mg of 2-(4-Dibenzo[b,f}[1,4] thiazepine-ll-yl-
Piperazin-1-yl)-ethanol RS (quetiapine impurity A RS) in
100 ml ofthe mobile phase. Dilute 5 m1 ofthe solution to 50 ml
HXCOOH with the mobile phase.
, I
HOOC H Chromatographic system
- a stainless steel column 25 cm x 4.6 rom, packed with
octylsilane bonded to porous silica (5Ilm) (Such as C8-
2 Novapak),
- mobile phase: a mixture of 500 volumes of methanol,
400 volumes of water, 100 volumes of acetonitrile and
(C2IH2SN302S)2,C4~04 Mol. Wt. 883.1
0.4 volume of triethylamine, adjusted to pH 7.0 with
Quetiapirte Fumarate is 2-[2-(4-dibenzo[bj] [1,4]thiazepin-ll- orthophosphoric acid,
yl-l-piperazinyl)ethoxy]ethanol fumarate - flow rate. 1.5 ml per minute,
Quetiapine Fumarate contains not less than 98.0 per cent and spectrophotometer set at 254 urn,
injection volume. 20 Ill.
not more than 102.0 per cent of C42HsoN604S2,C4H404
calculated on the dried basis. Inject reference solution (c). The test is not valid unless the
Dose. 300 to 450 mg per day. resolution between the peaks corresponding to quetiapine
impurity A and quetiapine in the chromatogram obtained with
Category. Antipsychotic.
the reference solution (c) is not less than 1.5.
Description. A white to off-white powder.
Inject the test solution, reference solution (a) and (b). Run the
Identification chromatograms for three times the retention time of the
principal peak. In the chromatogram obtained with the test
A. Determine by infrared absorption spectrophotometry (2.4.6).
solution, the area ofany secondary peak is not more than 0.5
Compare the spectrum with that obtained with quetiapine
times the area ofthe peak in the chromatogram obtained with
ji/marate RS. Ifthe spectra obtained show differences, dissolve
the reference solution (a) (0.5 per cent) and the sum of the
the substance under examination and the reference substance
areas of all the secondary peaks is not more than the area of
in methanol with the aid ofultrasound. ifnecessary. Evaporate
the peak in the chromatogram obtained with the reference
the solvent under nitrogen at 50° for 2 hours and determine on
solution (a) (1.0 per cent). Ignore the peak due to fumaric acid.
the residues.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
B. In the Assay, the principal peak in the chromatogram
heavy metals, Method B (20 ppm).
obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. Sulphated ash (2.3.18). Not more than 0.1 percent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Tests
on 1.0 g by drying in an oven at 105° for 1 hour.
Fumaricacid.12.6to 13.6 percent. Assay. Determine by liquid chromatography (2.4.14).
Weigh accurately about 0.35 g, dissolve in 80 ml of
Test solution. Dissolve 50 mg of the substance under
dimethylformamide. Titrate with 0.1 M tetra butyl ammonium
examination in 50.0 ml ofthe mobile phase.
hydroxide, determining the end-point potentiometrically
(2.4.25). Carry out a blank titration. Reference solution. A 0.1 per cent wlv solution of quetiapine
ji/marate RS in the mobile phase.
1 ml of 0.1 M tetra butyl ammonium hydroxide is equivalent
to 0.0058 g offumaric acid. Use the chromatographic system described in the test for
Related substances. Determine by liquid chromatography Related substances.
(2.4.14). Inject the reference solution. The test is not valid unless the
Test solution. Dissolve 50 mg of the substance under tailing factor is not more than 2.0, the column efficiency in not
examination in 50.0 ml ofthe mobile phase. less than 2000 theoretical plates and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
Reference solution (a). A 0.001 per cent wlv solution of
quetiapine jillnarate RS in the mobile phase. Inject the test solution and the reference solution.

2013

www.webofpharma.com
QUETIAPINE TABLETS IP 2010

Calculate the content of(CzlHzsN30zS)z,C4Ht04' the mobile phase. Dilute 5.0 ml ofthis solution to 50.0 ml with
the mobile phase.
Storage. Store protected from moisture, at a temperature not
exceeding 30°. Chromatographic system
- a stainless steel column 25 cm x 4.6 rom, packed with
octadecylsilane bonded to porous silica (5 11m),
- mobile phase: a mixture of40 volumes of water and 60
Quetiapine Tablets
volumes of methanol. Add 0.4 ml of triethylamine and
Quetiapine Fumarate Tablets adjust pH to 6.8 with orthophosphoric acid,
- flow rate. 1 ml per minute,
Quetiapine Tablets contain Quetiapine Fumarate equivalent
- spectrophotometer set at 289 nm,
to not less than 90.0 per cent and not more than 110.0 per cent
injection volume. 20 Ill.
of the stated amount of quetiapine, CZIHzsN30zS.
Inject the reference solution. The test is not valid unless the
Dose. 25 mg; 50 mg; 100 mg.
relative standard deviation for replicate injections is not more
Identification than 2.0 per cent.
Inject the test solution and the reference solution.
In the Assay, the principal peak in the chromatogram of the
test solution corresponds to the peak in the chromatogram Calculate the content ofCzIHzsN30zS in the tablets.
obtained with the reference solution. Storage. Store protected from moisture.
Tests Labelling. The label states the strength in terms of the
equivalent amount of quetiapine.
Dissolution (2.5.2).
Apparatus No.1,
Medium. 900 ml of 0.1 M hydrochloric acid,
Speed and time. 50 rpm and 45 minutes.
Quinalbarbitone Sodium
Test solution. Withdraw about 10 ml of the medium and Quinalbarbital Sodium; Secobarbital Sodium;
centrifuge. Dilute the supernatant liquid suitably with 0.1 M Secobarbitone Sodium; Soluble Quinalbarbitone
hydrochloric acid to obtain a solution containing about
10 Ilg per ml, ofquetiapine.
Reference solution. Dissolve about 58 mg of quetiapine
fUmarate RS in 15 ml ofthe dissolution medium and dilute to
100.0 ml with the medium. Dilute 2.0 ml of the solution to
100.0 rnl with the same medium.
Measure the absorbance of the reference solution and test
solution at 248 nm (2.4.7) against 0.1 M hydrochloric acid as
the blank. Calculate the content of quetiapine in the medium Mol.Wt.260.3
from the absorbance obtained from a solution of known Quinalbarbitone Sodium is sodium (RS)-5-allyl-5-(I-
concentration of quetiapinefUmarate RS in the same medium. methylbutyl)barbiturate.
D. Not less than 70 per cent ofthe stated amount ofquetiapine. Quinalbarbitone Sodium contains not less than 98.5 per cent
Other tests. Comply with the tests stated under Tablets. a.nd n6t1liore than 102.0 per cent ofClzHI7NzNa03, ca.lclllated
on the dried basis.
Assay. Determine by liquid chromatography (2.4.14)
Category. Hypnotic.
Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing 250 mg of quetiapine, Dose. 100 to 200 mg.
disperse in 100.0 ml of the mobile phase, with the aid of Description. A white powder; odourless; hygroscopic.
ultrasound and dilute to 250.0 ml with the mobile phase. Dilute
5 rnl ofthe solution to 50 ml with the mobile phase. Identification
iiejerencesolution. Weigh accura.tely about 58 mg of Test A may be omitted if tests B, C, D and E are carried out.
quetiapine fumarate RS, add 30 ml of the mobile phase and Tests B, C and D may be omitted if tests A and E are carried
dissolve with aid ofultrasound. Cool and dilute to 50.0 ml with out.

2014

www.webofpharma.com
IP 2010 QUINALBARBITONE TABLETS

A. To 10ml ofa 10.0 per cent w/v solution in ethanol (95 per Quinalbarbitone Tablets
cent) add 120 ml of water and 5 ml of 2 M acetic acid, stir
vigorously, add 200 ml of water and boil until the precipitate Quinalbarbital Sodium Tablets; Quinalbarbitone
dissolves and no oily particles remain on the surface of the Sodium Tablets; Secobarbital Sodium Tablets;
liquid. Allow to cool until a haziness begins to appear in the Secobarbitone Sodium Tablets
solution, induce crystallisation, if necessary and allow the
solution to stand for 12 hours. Wash the crystals with three Quinalbarbitone Tablets contain not less than 92.5 per cent
quantities, each of 10 ml, of water and dry the residue at 80°. and not more than 107.5 per cent of the stated amount of
quinalbarbitone sodium, C12H17N2Na03. The tablets are coated.
On the residue, determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that Usual strength. 100 mg.
obtained with quinalbarbitone RStreated in the same manner.
Identification
B. To 0.5 g, add 5 ml of sodium carbonate solution and 10 ml
of nitrobenzyl chloride solution. Heat for 30 minutes on a A.Shake a quantity of the powdered tablets containing about
water-bath under reflux and allow to stand for I hour. Filter, 0.5 g ofQuinalbarbitone Sodium with 10 ml of water and filter.
wash the precipitate successively with 10 ml of dilute sodium To 10 ml ofthe filtrate add 5 ml of sodium carbonate solution
hydroxide solution and 50 ml of water; recrystallise from a and 10 ml of nitrobenzyl chloride solution. Heat for 30 minutes
mixture of equal volumes of chloroform and ethanol (95 per on a water-bath under reflux and allow to stand for 1 hour.
cent) and dry at 105°. The crystals melt atabout 156° (2.4.21) Filter, wash the precipitate successively with 10 ml of dilute
C. Complies with the test for identification of barbiturates sodium hydroxide solution and 50 ml of water; recrystallise
(2.3.2). from a mixture of equal volumes of chloroform and ethanol
(95 per cent) and dry at 105°. The crystals melt at about
D. Gives the reaction ofnon-nitrogen substituted barbiturates 156° (2.4.21) ,
(2.3.1).
B. Triturate a quantity of the powdered tablets containing
E. Ignite 1 g; the residue gives the reactions of sodium salts 0.5 g of Quinalbarbitone Sodium with 10 ml of water, filter,
(2.3.1). acidify the filtrate with acetic acid; oily drops are fonned
which may eventually clystallise. .
Tests
C. The powdered tablets give the reactions of sodium salts
Appearance of solution. A freshly prepared 10.0 per cent w/v (2.3.1).
solution in ethanol (95 per cent) is clear (2.4.1), and not more
intensely coloured than reference solution YS7 (2.4.1). Tests
pH (2.4.24). Not more than 11.0, determined on a 10.0 per cent
Other tests. Comply with the tests stated under Tablets.
w/v solution.
Assay. Weigh and digest 20 tablets with 50 ml of water until
Heavy metals (2.3.13). 0.67 g dissolved in a mixture of5 ml of
completely disintegrated and not more than a small residue
1 M sodium hydroxide and 20 ml of water complies with the
remains. Add 5 ml of 1 M sodium hydroxide, filter and wash
limit test for heavy metals, Method C (30 ppm).
the residue with sufficient water to produce 100.0 ml. Extract a
Related substances. Complies with the test for related volume of the solution containing 0.5 g of Quinalbarbitone
substances in barbiturates (2.3.4). Sodium with two quantities, each of 10 ml, of ether, washing
each ether extract with the same 3 ml of water. Add the water
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
to the aqueous liquid, acidify witn hydrochloric acid and
on 0.5 g by drying in an oven at 105°. .
extract with successive quantities, each of 15 ml, of ether until
Assay. Weigh accurately about 0.5 g, dissolve in 10 ml of complete extraction is effected. Wash the combined extracts
ethanol, add 10 ml ofsilver nitrate-pyridine reagent and titrate with two quantities, each of 2 ml, of water and extract
with 0.1 M ethanolic sodium hydroxide using 0.5 mlof the combined washings with 10 ml of ether. Add the ether to
thymolphthalein solution as indicator, until a pure blue colour the main ether layer, filter and wash the filter with ether.
is obtained. Carry out a blank titration. Evaporate the solvent and dry the residue to constant weight
at 50°.
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
0.02603 g ofCI2H17N2Na03' 1 g ofresidue is equivalent to 1.092 g ofC12H17N2Na03.
Storage. Store protected from moisture. Storage. Store protected from moisture.

2015

www.webofpharma.com
QUINIDINE SULPHATE IP 2010

Quinidine Sulphate B. To 5 ml ofa 0.1 percentw/v solution add 0.2 ml of bromine


solution and 1 ml of dilute ammonia solution; an emerald-
Quinidine Bisulphate green colour is produced.
C. To a 0.5 per cent w/v solution add an equal volume of dilute
sulphuric acid; a strong blue fluorescence is produced.
D. To 5 ml ofa 1percentw/v solution add 1 mlofsilvernitrate
solution and stir with a glass rod; after a short interval, a
white precipitate soluble in nitric acid is produced (distinction
from many other allmloids).
E. A 1 per cent w/v solution gives the reactions of sulphates
(2.3.1).
2
Tests
(CzoHz~zOz)z,HzS04,2HzO Mol. Wt. 783.0
Appearance of solution. A2.0 per cent w/v solution in 0.1 M
Quinidine Sulphate is (8R,9S)-6' -methoxycinchonan-9-01
hydrochloric acid is clear (2..4.1), and not more intensely
sulphate dihydrate. The alkaloid is obtained from the bark of
coloured than reference solution GYS4 (2.4.1).
various species of Cinchona and from Remijia pedunculata
Fluckiger (Fam. Rubiaceae) or prepared from quinine. pH (2.4.24). 6.0 to 6.8, determined in a 1.0 per cent w/v solution.
Quinidine Sulphate contains not less than 99.0 per cent and Specific optical rotation (2.4.22). +275° to +290°, determined
not more than 101.5 per cent of alkaloid monosulphates, in a 2.0 per cent w/v solution in 0.1 M hydrochloric acid.
calculated as (CzoHz4NzOz)2,H2S04 on the dried basis. Dihydroquinidine sulphate. Not more than 15.0 per cent,
Category. Antiarrhythmic. calculated on the dried basis and determined by the following
method. Dissolve 0.2 gin 20 ml of water, add 0.5 g ofpotassium
Dose. In the treatment ofcardiac arrhythmias, 200 mg daily in bromide and 15 ml of 2 M hydrochloric acid. Titrate slowly
three or four divided doses. In the treatment ofatrial fibrillation, with 0.0167 M potassium bromate using methyl red solution
200 to 400 mg every 2 to 4 hours, upto 3 g daily. as indicator until a yellow colour is obtained. Add a solution
Description. A white or almost white, crystalline powder or of0.5 g ofpotassium iodide in 200 ml of water and stopper the
needle-like crystals; odourless. flask immediately. Allow to stand in the dark for 5 minutes and
titrate with 0.1 M sodium thiosulphate using starch solution,
Identification added towards the end of the titration, as indicator. Repeat
A. Determine by thin-layer chromatography (2.4.17), coating the operation without the substance under examination.
the plate with silica gel G. I ml of 0.0167 M potassium bromate is equivalent to
0.01867 gof(C2oHz4N20z)z, H2S04.
Mobile phase. A mixture of60 volumes of toluene, 36 volumes
of ether and 15 volumes of diethylamine. Calculate the content of dihydroquinidine sulphate by
subtracting the result from the assay result.
Test solution. Dissolve 1 g ofthe substance under examination
in 100 ml of methanol. Other cinchona alkaloids. Determine by liquid
chromatography (2.4.14).
Reference solution (a). A 1 per cent w/v solution of quinidine
sulphate RS in methanol. Test solution. Dissolve 20 mg of the substance under
examination in 5 mlof the mobile phase. Heat gently, if
Reference solution (b). A 1 per cent w/v solution of each of necessary to dissolve the powder as completely as possible,
quinidine sulphate RS and quinine sulphate RS in methanol. cool, dilute to 10 ml with the mobile phase and mix.
Apply to the plate 4 III of each solution. After development, Reference solution (a). Dissolve 20 mg of quinine sulphate
dry the plate in air for 15 minutes and repeat the development. RS, with gentle heating if necessary, in 5 ml of the mobile
Dry the plate at 105° for 30 minutes, allow to cool and spray phase and dilute to 10 ml with the mobile phase.
with potassium iodoplatinate solution. The principal spot in
the chromatogram obtained with the test solution corresponds Reference solution (b). Prepare in the same manner as
to the spot in the chromatogram obtained with reference reference solution (a) but using quinidine sulphate RS in
solution (a). The test is not valid unless the chromatogram place of quinine sulphate RS.
. obtained with reference solution (b) shows two clearly Reference solution (c). Mix equal volumes of reference
separated spots. solutions (a) and (b).

2016

www.webofpharma.com
IP 2010 QUININE TABLETS

Reference solution (d). Dilute 1 volume of reference solution normalisation, ignoring any peaks the areas ofwhich are less
(a) to 10 volumes with the mobile phase and dilute 1 volume of than that of the peak in the chromatogram obtained with
the resulting solution to 50 volumes with the mobile phase. reference solution (d) (0.2 per cent). The content of
dihydroquinidine is not greater than 15 per cent, the content
Reference solution (e). A solution containing 0.1 per cent
ofany related substance eluting before quinidine is not greater
w/v of thiourea in the mobile phase.
than 5 per cent and the content of any other related substance
Chromatographic system is not greater than 2.5 per cent.
- a stainless steel column 25 cm x 4.6 mm packed with
Sulphated ash (2.3.18). Not more than 0.1 per cent.
octadecylsilane bonded to porous silica (5 11m) (Such
as Hypersil ODS 5 11m), Loss on drying (2.4.19). 3.0 per centto 5.0 per cent, determined
- mobile phase: a solution prepared by dissolving 6.8 g of on 1.0 g by drying in an oven at 130°.
potassium dihydrogen orthophosphate and 3.0 g of Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
hexylamine in 700 ml of water, adjusting the pH to 2.8 10 ml of chloroform and 20 ml of acetic anhydride. Titrate
with 1 M orthophosphoric acid, adding 60 ml of with 0.1 M perchloric acid, determining the end-point
acetonitrile and diluting to 1000 ml with water, potentiometrically (2.4.25). Carry out a blank titration.
- flow rate. 1.5 ml per minute,
spectrophotometer set at 250 urn for reference solution 1 ml of 0.1 M perchloric acid is equivalent to 0.02490 g of
(e) and 316 nm for the other solutions, (C2oH24N202)2,H2S04'
injection volume. 10 Ill. Storage. Store protected from light.
Inject separately reference solutions (b) and (e). Ifnecessary,
adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b) Quin.idine Tablets
the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Va
(the distance along the baseline between the point ofinjection . Quinidine Sulphate Tablets
and the perpendicular dropped from the maximum ofthe peak Quinidine Tablets contain not less than 95.0 per cent and not
of an unretained component) being calculated from the peak more than 105.0 per cent of the stated amount of quinidine
due to thiourea in the chromatogram obtained with reference sulphate, (C2oH24N202)2,H2S04,2H20;
solution (e).
Usual strength. 200 mg.
Inject reference solutions (a), (b), (c) and (d). The
chromatogram obtained with reference solution (a) shows a Identification
principal peak due to quinine and a peak due to dihydroquinine
A. Determine by thin-layer chromatography (2.4.17), coating
with a retention time relative to quinine of about 1.4. The
the plate'with silica gel G.
chromatogram obtained with reference solution (b) shows a
principal peak due to quinidine and a peak due to Mobile phase. A mixture of80 volumes of toluene, 20 volumes
dihydroquinidine, with a retention time relative to quinidine of acetone and 10 volumes of diethylamine.
of about 1.2. The chromatogram obtained with reference Test solution. Extract a quantity of the powdered tablets
solution (c) shows four peaks due to quinine, dihydroquinine, containing 0.1 g ofQuinidine Sulphate with 10 ml ofa mixture
quinidine and dihydroquinidine which are identified by of2 volumes of chloroform and 1 volume of ethanol (95 per
comparison of their retention times with those of the cent) and filter.
corresponding peaks in the chromatograms obtained with
Reference solution. 1.0 per cent w/v solution of quinidine
reference solutions (a) and (b).
sulphate RS in a mixture of 2 volumes of chloroform and
The test is not valid unless (a) in the chromatogram obtained 1 volume of ethanol (95 per cent).
with reference solution (c) the resolution between the peaks Apply to the plate 2 III of each solution. After development,
due to quinine and quinidine is at least 1.5 and the resolution dry the plate in air and spray with 0.05 M ethanolic sulphuric
between the peaks due to dihydroquinidine and quinine is at acid and then with dilute potassium iodobismuthate solution.
least 1.0 and (b) the signal-to-noise ratio ofthe principal peak The principal spot in the chromatogram obtained with the test
in the chromatogram obtained with reference solution (d) is at solution corresponds to that in the chromatogram obtained
least 5. with the reference solution.
Inject the test solution and allow the chromatography to B. Extract a quantity ofthe powdered tablets containing 0.1 g
proceed for 2.5 times the retention time ofthe principal peale ofQuinidine Sulphate with 20 ml of water and filter. The filtrate
Calculate the percentage content of related substances by (solution A) is dextro-rotatory.

2017

www.webofpharma.com
QUINIDINE TABLETS IP 2010

C. To 1 ml of solution A add 4 ml of water, 2 or 3 drops of flow rate. 1.5 rnl per minute,
bromine solution and 1 ml of dilute ammonia solution; an spectrophotometer set at 250 nm for reference solution
emerald green colour is produced. (e) and 316 nm for the other solutions,
D. Solution A gives the reactions ofsulphates (2.3.1). injection volume. 10 fll.
Inject separately reference solutions (b) and (e). Ifnecessary,
Tests adjust the concentration of acetonitrile in the mobile phase so
Dissolution (2.5.2). that in the chromatogram obtained with reference solution (b)
the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Vo
Apparatus No.2,
(the distance along the baseline between the point of injection
Medium. 900 ml of 0.1 M hydrochloric acid, and the perpendicular dropped from the maximum ofthe peak
Speed and time. 100 rpm and 30 minutes. of an unretained component) being calculated from the peak
Withdraw a suitable volume ofthe medium and filter. Measure due to thiourea in the chromatogram obtained with reference
the absorbance of the filtrate, suitably diluted if necessary, at solution (e).
the maximum at about 248 nm (2.4.7). Calculate the content of Inject reference solutions (a), (b), (c) and (d). The
(CzoHz4NzOz)z,HzS04,2HzO in the medium from the absorbance chromatogram obtained with reference solution (a) shows a
obtained from a solution ofknown concentration of quinidine principal peak due to quinine and a peak due to dihydroquinine
sulphate RS. with a retention time relative to quinine of about 1.4. The
D. Not less than 70 per cent of the stated amount of chromatogram obtained with reference solution (b) shows a
(CzoHz4NzOz)z,HzS04,2HzO. principal peak due to quinidine and a peak due to
dihydroquinidine, with a retention time relative to quinidine
Other cinchona alkaloids. Determine by liquid of about 1.2. The chromatogram obtained with reference
chromatography (2.4.14). solution (c) shows four peaks due to quinine, dihydroquinine,
Test solution. Mix a quantity of the powdered tablets quinidine and dihydroquinidine which are identified by
containing 50 mg of Quinidine Sulphate with 20 ml of the comparison of their retention times with those of the
mobile phase. Heat gently to dissolve the powder as completely corresponding peaks in the chromatograms obtained with
reference solutions (a) and (b).
as possible, cool, dilute to 25 ml with the mobile phase and
filter, discarding the first few ml offiltrate. The test is not valid unless (a) in the chromatogram obtained
with reference solution (c) the resolution between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate
due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile
between the peaks due to dihydroquinidine and quinine is at
phase and dilute to 10 ml with the mobile phase.
least 1.0 and (b) the signal-to-noise ratio ofthe principal peak
Reference solution (b). Prepare in the same manner as in the chromatogram obtained with reference solution (d) is at
reference solution (a) but using quinidine sulphate RS in least 5.
place of quinine sulphate RS.
Inject the test solution and allow the chromatography to
Reference solution (c). Mix equal volumes of reference proceed for 2.5 times the retention time ofthe principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
Reference solution (d). Dilute 1 volume of reference solution normalisation, ignoring any peaks the areas of which are less
(a) to 10 volumes with the mobile phase and dilute 1 volume of than that of the peak in the chromatogram obtained with
theresulting solution to 50 volumes with the mobile phase. reference solution (d) (0.2 per cent). The content of
dihydroquinidine is not greater than 15 per cent, the content
Reference solution (e). A solution containing 0.1 per cent ofany related substance eluting before quinidine,is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance
Chromatographic system is not greater than 2.5 per cent.
- a stainless steel column 25 cm x 4.6 rom packed with
Other tests. Comply with the tests stated under Tablets.
octadecylsilane bonded to porous silica (5 flm) (Such
as Hypersil ODS 5 flm), Assay. Weigh and powder 20 tablets. Weigh accurately a
mobile phase: a solution prepared by dissolving 6.8 g of quantity of the powder containing about 0.4 g of Quinidine
potassium dihydrogen orthophosphate and 3.0 g of Sulphate, dissolve as completely as possible in 40 ml of acetic
hexylamine in 700 ml of water, adjusting the pH to 2.8 anhydride with the aid of heat and cool. Titrate with 0.1 M
with 1 M orthophosphoric acid, adding 60 ml of perchloric acid, using clystal violet solution as indicator.
acetonitrile and diluting to 1000 ml with water, Carry out a blank titration.

2018

www.webofpharma.com
IP 2010 QUININE BISULPHATE

1 ml of 0.1 M perchloric acid is equivalent to 0.02610 g of B. A 5 per cent w/v solution has a blue fluorescence.
(C2oH24N202)2,H2S04,2H20. C. A 5 per cent w/v solution gives the reactions of sulphates
Storage. Store protected from light. (2.3.1).

Tests

Quinine Bisulphate pH (2.4.24).2.8 to 3.4, determined in a 1.0 per cent w/v solution.
Specific optical rotation (2.4.22). -208° to -216°, determined
Quinine Acid Sulphate
at 20° in a 3.0 per cent w/v solution in 0.1 M hydrochloric
OCH 3 acid.

~CH2
Other cinchona alkaloids. Determine by liquid
chromatography (2.4.14).
Test solution. Dissolve 20 mg of the substance under
H examination in 5 ml of the mobile phase. Heat gently, if
necessary to dissolve the powder as completely as possible,
C2oH24N202,H2S04,7H20 Mol. Wt. 548.6 cool, dilute to 10 ml with the mobile phase and mix.
Quinine Bisulphate is (8S,9R)-6' -methoxycinchonan-9-01 Reference solution (a). Dissolve 20 mg of quinine sulphate
hydrogen sulphate heptahydrate. The alkaloid is obtained RS, with gentle heating if necessary, in 5 ml of the mobile
from the bark of various species of Cinchona. phase and dilute to 10 ml with the mobile phase.
Quinine Bisulphate contains not less than 98.5 per cent and Reference solution (b). Prepare in the same manner as
not more than 101.5 per cent of alkaloid hydrogen sulphates, reference solution (a) but using quinidine sulphate RS in
calculated as C2oH24N202,H2S04 on the anhydrous basis. place of quinine sulphate RS.
Category. Antimalarial. Reference solution (c). Mix equal volumes of reference
Dose. Suppressive, 300 to 600 mg daily; therapeutic, 1.2 to 2 g solutions (a) and (b).
daily, in divided doses. Reference solution (d). Dilute 1 volume ofreference solution
Description. Colourless or faintly yellow crystals or a white (a) to 10 volumes with the mobile phase and dilute 1 volume of
or faintly yellow, crystalline powder; efflorescent in dry air. the resulting solution to 50 volumes with the mobile phase.
Reference solution (e). A solution containing 0.1 per cent
Identification
w/v of thiourea in the mobile phase.
A. Determine by thin-layer chromatography (2.4.17), coating Chromatographic system
the plate with silica gel G. - a stainless steel column 25 cm x 4.6 mm packed with
Mobile phase. A mixture of60 volumes of toluene, 36 volumes octadecylsilane bonded to porous silica (5 /lm) (Such
of ether and 15 volumes of diethylamine. as Hypersil ODS 5 /lm),
- mobile phase: a solution prepared by dissolving 6.8 g of
Test solution. Dissolve 1 g ofthe substance under examination
in 100 ml methanol. potassium dihydrogen orthophosphate and 3.0 g of
hexylamine in 700 ml ofwatel; adjusting the pH to 2.8
Reference solution (a). A 1 per cent w/v solution of quinine with 1 M orthophosphoric acid, adding 60 ml of
sulphate RS in methanol. acetonitrile and diluting to 1000 ml with water,
Reference solution (b). A 1 per cent w/v solution of each of - flow rate. 1.5 ml per minute,
qUinidine sulphate RS and quinine sulphate RS in methanol. - spectrophotometer set at 250 tim for reference solution
(e) and 316 nm for the other solutions, .
Apply to the plate 4 /ll of each solution. After development,
- injection volume. 10 Ill.
dry the plate in air for 15 minutes and repeat the development.
Dry the plate at 105° for 30 minutes, allow to cool and spray Inject separately reference solutions (b) and (e). If necessary,
with potassium iodoplatinate solution. The principal spot in adjust the concentration of acetonitrile in the mobile phase so
the chromatogram obtained with the test solution corresponds that in the chromatogram obtained with reference solution (b)
to the spot in the chromatogram obtained with reference the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Va
solution (a). The test is not valid unless the chromatogram (the distance along the baseline between the point of injection
obtained with reference solution (b) shows two clearly and the perpendicular dropped from the maximum ofthe peak
separated spots. of an unretained component) being calculated from the peak

2019

www.webofpharma.com
QUININE BISULPHATE IP 2010

due to thiourea in the chromatogram obtained with reference 1 ml of 0.1 M perchloric acid is equivalent to 0.02113 g of
solution (e). CzoHz~zOz,HzS04'
Inject reference solutions (a), (b), (c) and (d). The Storage. Store protected from light.
chromatogram obtained with reference solution (a) shows a
principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 1.4. The
chromatogram obtained with reference solution (b) shows a Quinine Bisulphate Tablets
principal peak due to quinidine and a peak due to
dihydroquinidine, with a retention time relative to quinidine Quinine Acid Sulphate Tablets
of about 1.2. The chromatogram obtained with reference Quinine Bisulphate Tablets contain not less than 95.0 per cent
solution (c) shows four peaks due to quinine, dihydroquinine, and not more than 105.0 per cent of the stated amount of
quinidine and dihydroquinidine which are identified by quinine bisulphate, CzoHz4NzOz,HzS04,7HzO. The tablets are
comparison of their retention times with those of the coated.
corresponding peaks in the chromatograms obtained with
reference solutions (a) and (b). Usual strength. 300 mg.

The test is not valid unless (a) in the chromatogram obtained Identification
with reference solution (c) the resolution between the peaks
A. Determine by thin-layer chromatography (2.4.17), coating
due to quinine and quinidine is at least 1.5 and the resolution
the plate with silica gel G.
between the peaks due to dihydroquinidine and quinine is at
least 1.0 and (b) the signal-to-noise ratio ofthe principal peak Mobile phase. A mixture of80 volumes oftoluene, 20 volumes
in the chromatogram obtained with reference solution (d) is at of acetone and 10 volumes of diethylamine.
least 5.
Test solution. Extract a quantity of the powdered tablets
Inject the test solution and allow the chromatography to containing 0.1 g ofQuinine Bisulphate with 10 m1 ofa mixture
proceed for 2.5 times the retention time ofthe principal peak. of2 volumes of chloroform and 1 volume of ethanol (95 per
Calculate the percentage content of related substances by cent) and filter.
normalisation, ignoring any peaks the areas of which are less
Reference solution. A 1.0 per cent wlv solution of quinine
than that of the peak in the chromatogram obtained with
sulphate in the same solvent mixture.
reference solution (d) (0.2 per cent). The content of
dihydroquinine is not greater than 10 per cent, the content of Apply to the plate 2 111 of each solution. After development,
any related substance eluting before quinine is not greater dry the plate in air and spray with 0.05 M ethanolic sulphuric
than 5 per cent and the content of any other related substance acid and then with dilute potassium iodobismuthate solution.
is not greater than 2.5 per cent. The principal spot in the chromatogram obtained with the test
Titratable cation. 75.3 to 79.6 per cent, calculated on the solution corresponds to that in the chromatogram obtained
anhydrous basis, determined by the following method. Titrate with the reference solution.
the combined aqueous solutions reserved in the Assay with B. Extract a quantity ofthe powdered tablets containing 0.1 g
0.1 M hydrochloric acid using 0.1 ml of phenolphthalein ofQuinine Bisulphate with 20 ml ofwater and filter (solution
solution as indicator. A). To 5 ml of solution A add 0.2 ml of bromine solution and
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01632 g of 1 ml of dilute ammonia solution; an emerald-green colour is
[CzoHz6NzOzJ2+. produced.
Sulphated ash (2.3.18). Not more than 0.1 per cent. C. Solution A is leva-rotatory.
Water (2.3.43). 19.0 to 25.0 per cent, determined on 0.2 g. D. Solution A gives the reactions of sulphates (2.3.1).
Assay. Weigh accurately about 0.45 g, dissolve in 15 mlof
water, add 25 ml of 0.1 M sodium hydroxide and extract with Tests
three quantities, each of 25 ml, of chloroform. Wash each Dissolution (2.5.2).
chloroform extract with the same 20 ml ofwater. Combine the
Apparatus No.2,
aqueous solutions and reserve for the test for Titratable cation.
Medium. 900 ml of 0.1 M hydrochloric acid,
Dry the chloroform extracts with anhydrous sodium sulphate,
Speed and time. 100 rpm and 45 minutes.
evaporate to dryness at a pressure of 2 kPa and dissolve the
residue in 50 ml ofanhydrous glacial acetic acid. Titrate with Withdraw a suitable volume ofthe medium and filter. Measure
0.1 M perchloric acid, using crystal violet solution as the absorbance of the filtrate, suitably diluted if necessary, at
indicator. Carry out a blank titration. the maximum at about 348 nm (2.4.7). Calculate the content of

2020

www.webofpharma.com
IP 2010 QUININE DIHYDROCHLORIDE

C2oH24N202,H2S04,7H20 in the medium taking 99 as the specific principal peak due to quinine and a peale due to dihydroquinine
absorbance at 348 nm. with a retention time relative to quinine of about 1.4. The
D. Not less than 70 per cent of the stated amount of chromatogram obtained with reference solution (b) shows a
CzOH24N202,H2S04,7H20. principal peak due to quinidine and a peak due to
dihydroquinidine, with a retention time relative to quinidine
Other cinchona alkaloids. Determine by liquid of about 1.2. The chromatogram obtained with reference
chromatography (2.4.14). solution (c) shows four peaks due to quinine, dihydroquinine,
Test solution. Remove any coating from the tablets and mix a quinidine and dihydroquinidine which are identified by
quantity of the powdered tablet cores containing 50 'mg of comparison of their retention times with those of the
Quinine Bisulphate with 20 ml ofthe mobile phase. Heat gently corresponding peaks in the chromatograms obtained with
to dissolve the powder as completely as possible, cool, dilute reference solutions (a) and (b).
to 25 ml with the mobile phase and filter, discarding the first The testis not valid unless (a) in the chromatogram obtained
few ml ofthe filtrate. with reference solution (c) the resolution between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile between the peaks due to dihydroquinidine and quinine is at
phase and dilute to 10 ml with the mobile phase. least 1.0 and (b) the signal-to-noise ratio ofthe principal peak
Reference solution (b). Prepare in the same manner as in the chromatogram obtained with reference solution (d) is at
reference solution (a) but using quinidine sulphate RS in least 5.
place of quinine sulphate RS. Inject the test solution and allow the chromatography to
Refel'ence solution (c). Mix equal volumes of reference proceed for 2.5 times the retention time ofthe principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
normalisation, ignoring any peaks the areas of which are less
Reference solution (d). Dilute 1 volume of reference solution than that of the peak in the chromatogram obtained with
(a) to 10 volumes with the mobile phase and dilute 1 volume of reference solution (d) (0.2 per cent). The content of
the resulting solution to 50 volumes with the mobile phase. dihydroquinine is not greater than 10 per cent, the content of
Reference solution (e). A solution containing 0.1 per cent any related substance eluting before quinine is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance .
Chromatographic system is not greater than 2.5 per cent.
a stainless steel column 25 cm x 4.6 rom packed with Other tests. Comply with the tests stated under Tablets.
octadecylsilane bonded to porous silica (5 /lm) (Such Assay. Weigh and powder 20 tablets. Weigh accurately a
as Hypersil ODS 5 /lm), quantity of the powder containing about 0.6 g of Quinine
mobile phase: a solution prepared by dissolving 6.8 g of Bisulphate, dissolve as completely as possible in 40 ml of
potassium dihydrogen orthophosphate and 3.0 g of acetic anhydride with the aid of heat and cool. Titrate with
hexylamine in 700 ml ofwatel; adjusting the pH to 2.8 0.1 M perchloric acid, using crystal violet solution as
with 1 M orthophosphoric acid, add.ing 60 ml of indicator. Carry out a blank titration.
acetonitrile and diluting to 1000 ml with water,
flow rate. 1.5 ml per minute, 1 ml of 0.1 M perchloric acid is equivalent to 0.05486 g of
spectrophotometer set at 250 nm for reference solution C2oH24N202,H2S04,7HzO.
(e) and 316 nm for the other solutions, Storage. Store protected from light.
injection volume. 10 Ill.
Inject separately reference solutions (b) and (e). Ifnecessary,
adjust the concentration of acetonitrile in the mobile phase so Quinine Dihydrochloride
that in the chromatogram obtained with reference solution (b) Quinine Acid Hydrochloride
the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Vo
(the distance along the baseline between the point ofinjection OCH 3

~CH2
and the perpendicular dropped from the maximum ofthe peak
of an unretained component) being calculated from the peak
due to thiourea in the chromatogram obtained with reference ,2HCI
solution (e).
H
Inject reference solutions (a), (b), (c) and (d). The
chromatogram obtained with reference solution (a) shows a Mol. Wt. 397.3

2021

www.webofpharma.com
QUININE DIHYDROCHLORIDE IP 2010

Quinine Dihydrochloride is the (8S,9R)-6'- method. Dissolve 0.2 g in 20 rnl of water, add 0.5 g ofpotassium
methoxycinchonan-9-01 dihydrochloride. The alkaloid is bromide and 15 ml of 2 M hydrochloric acid. Titrate slowly
obtained from the bark of various species of Cinchona. with 0.0167 M potassium bromate using methyl red solution
as indicator until a yellow colour is obtained. Add a solution
Quinine Dihydrochloride contains not less than 99.0 per cent
of0.5 g ofpotassium iodide in 200 ml of water and stopper the
and not more than 101.5 per centofalkaloid dihydrochlorides,
flask irnmediat~ly. Allow to stand in the dark for 5 minutes and
calculated as CzoHz4NzOz,2HCl on the dried basis.
titrate with 0.1 M sodium thiosulphate using starch solution,
Category. Antimalarial. added towards the end of the titration, as indicator. Repeat
Dose. Suppressive, 300 to 600 mg daily; therapeutic, 1.2 to 2 g the operation without the substance under examination.
daily, in divided doses. By slow intravenous injection, 300 to 1 m1 of 0.0167 M potassium bromate is equivalent to
600 mg. 0.01987 g of CzoHz4NzOz,2HCl. Calculate the content of
Description. A white or almost white powder; odourless. dihydroquinine dihydroch10ride by subtracting the result from
the assay result.
Identification Other cinchona alkaloids. Determine by liquid
chromatography (2.4.14).
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G. Test solution. Dissolve 20 mg of the substance under
examination in 5 ml of the mobile phase. Heat gently, if
Mobile phase. A mixture of60 volumes of toluene, 36 volumes
necessary to dissolve the powder as completely as possible,
of ether and 15 volumes of diethylamine.
cool, dilute to 10 m1 with the mobile phase and mix.
Test solution. Dissolve 1 g ofthe substance under examination
Reference solution (a). Dissolve 20 mg of quinine sulphate
in 100 ml of methanol.
RS, with gentle heating if necessary, in 5 ml of the mobile
Reference solution (a). A 1 per cent w/v solution of quinine phase and dilute to 10 m1 with the mobile phase.
sulphate RS in methanol.
Reference solution (b). Prepare in the same manner as
Reference solution (b). A 1 per cent w/v solution of each of reference solution (a) but using quinidine sulphate RS in
quinidine sulphate RS and quinine sulphate RS in methanol. place of quinine sulphate RS.
Apply to the plate 4 !J.l of each solution. After development, Reference solution (c). Mix equal volumes of reference
dry the plate in air for 15 minutes and repeat the development. solutions (a) and (b).
Dry the plate at 105° for 30 minutes, allow to cool and spray
Reference solution (d). Dilute 1 volume ofreference solution
with potassium iodopldtinate solution. The principal spot in
(a) to 10 volumes with the mobile phase and dilute 1 volume of
the chromatogram obtained with the test solution corresponds
the resulting solution to 50 volumes with the mobile phase.
to the spot in the chromatogram obtained with reference
solution (a). The test is not valid unless the chromatogram Reference solution (e). A solution containing 0.1 per cent
obtained with reference solution (b) shows two clearly w/v of thiourea in the mobile phase.
separated spots. Chromatographic system
B. To a 0.5 per cent w/v solution add an equal volume of dilute - a stainless steel column 25 cm x 4.6 rom packed with
sulphuric acid; a strong blue fluorescence is produced. octadecylsilane bonded to porous silica (5 !J.m) (Such
as Hypersil ODS 5 !J.m),
C. A 1 per cent w/v solution gives the reactions of chlorides
- mobile phase: a solution prepared by dissolving 6.8 g of
(2.3.1 ).
potassium dihydrogen orthophosphate and 3.0 g of
hexylamine in 700 ml of water; adjusting the pH to 2.8
Tests
with 1 M orthophosphoric acid, adding 60 ml of
pH (2.4.24).2.0 to 3.0, determined in a 3.0percentw/v solution. acetonitrile and diluting to 1000 ml with water
flow rate. 1.5 rnl per minute,
Specific optical rotation (2.4.22). -223° to -229°, determined
- spectrophotometer set at 250 nm for reference solution
in a 3.0 per cent w/v solution in 0.1 M hydrochloric acid.
(e) and 316 nm for the other solutions,
Barium. To 15 ml of a 2 per cent w/v solution add 1 mlof injection volume. 10 !J.l.
dilute sulphuric acid; the solution remains clear for at least Inject separately reference solutions (b) and (e). If necessary,
15 minutes. adjust the concentration of acetonitrile in the mobile phase so
Dihydroquinine dihydrochloride. Not more than 10.0 per cent, that in the chromatogram obtained with reference solution (b)
calculated on the dried basis and determined by the following the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Va

2022

www.webofpharma.com
IP 2010 QUININE DIHYDROCHLORIDE INJECTION

(the distance along the baseline between the point ofinjection anhydride and 10 ml of mercuric acetate solution. Titrate
and the perpendicular dropped from the maximum ofthe peak with 0.1 M perchloric acid, using clystal violet solution as
of an unretained component) being calculated from the peak indicator. Cany out a blank titration.
due to thiourea in the chromatogram obtained with reference
1 ml of 0.1 M perchloric acid is equivalent of 0.01987 g of
solution (e).
C2oH24N202,2HCl.
Inject reference solutions (a), (b), (c) and (d). The
Storage. Store protected from light.
chromatogram obtained with reference solution (a) shows a
.principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 1.4. The
chromatogram obtained with reference solution (b) shows a
principal peak due to quinidine and a peak due to
dihydroquinidine, with a retention time relative to quinidine Qu.inine Dihydrochloride Injection
of about 1.2. The chromatogram obtained with reference Quinine Acid Hydrochloride Injection
solution (c) shows four peaks due to quinine, dihydroquinine,
quinidine and dihydroquinidine which are identified by Quinine Dihydrochloride Injection is a sterile solution of
comparison of their retention times with those of the Quinine Dihydrochloride in Water for Injections.
corresponding peaks in the chromatograms obtained with Quinine Dihydrochloride Injection contains not less than
reference solutions (a) and (b). 95.0 per cent and not more than 105.0 per cent of the stated
The test is not valid unless (a) in the chromatogram obtained amount ofquinine dihydrochloride, C2oH24N202,2HCl.
with reference solution (c) the resolution between the peaks Usual strength. 300 mg per ml.
due to quinine and quinidine is at least 1S and the resolution
between the peaks due to dihydroquinidine and quinine is at Description. A clear, almost colourless to light yellow solution.
least 1.0 and (b) the signal-to-noise ratio ofthe principal peak
in the chromatogram obtained with reference solution (d) is at Identification
least 5.
A. Determine by thin-layer chromatography (2.4.17), coating
Inject the test solution and allow the chromatography to the plate with silica gel G.
proceed for 2.5 times the retention time ofthe principal peak.
Calculate the percentage content of related substances by Mobile phase. A mixture of80 volumes of toluene, 20 volumes
normalisation, ignoring any peaks the areas of which are less of acetone and 10 volumes of diethylamine.
than that of the peak in the chromatogram obtained with Solvent mixture. 2 volumes of chloroform and 1 volume of
reference solution (d) (0.2 per cent). The content of ethanol (95 per cent).
dihydroquinine is not greater than 10 per cent, the content of
any related substance eluting before quinine is not greater Test solution. Extract a volume of the injection containing
than 5 per cent and the content of any other related substance 0.1 g ofQuinine Dihydrochloride Bisulphate with 10 ml ofthe
is not greater than 2.5 per cent. solvent mixture and filter.

Titratable cation. 79.7 to 84.2 per cent, calculated on the dried Reference solution (a). A 1 per cent w/v solution of quinine
basis, determined by the following method. Weigh accurately sulphate RS in the solvent mixture.
about 0.4 g, dissolve in 10 ml of water, add 40 ml of methanol Reference solution (b). A solution of 1 per centw/v of each of
and titrate with 0.1 M sodium hydroxide using quinidine sulphate RS and quinine sulphate RS in the solvent
phenolphthalein solution as indicator. mixture.
1 ml of 0.1 Msodium hydroxide is equivalentto 0.01632 g of Apply to the plate 2 III of each solution. After development,
[C2oH26N202F+. dry the plate in air and spray with 0.05 M ethanolic sulphuric
Sulphates (2.3.17). 0.125 g complies with the limit test for acid and then with dilute potassium iodobismuthate solution.
sulphates ( 0.12 per cent). The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
with the reference solution (a). The test is not valid unless the
Loss on drying (2.4.19). Not more than 3.0 per cent, determined chromatogram obtained with reference solution (b) shows two
on 1.0 g by drying in an oven at 105°. clearly separated spots.
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of B. Dilute 1 ml to 20 ml with water, add 0.1 M sodium hydroxide
anhydrous glacial acetic acid and add 20 ml of acetic dropwise until the solution becomed turbid. Add 1 or 2 drops

2023

www.webofpharma.com
QUININE DffiYDROCHLORIDE INJECTION IP 2010

of 0.05 M sulphuric acid to obtain a clear solution. To this ' Inject reference solutions (a), (b), (c) and (d). The chromatogram
solution add an equal volume of dilute sulphuric acid; a obtained with reference solution (a) shows a principal peak
strong blue fluorescence is produced. due to quinine and a peak due to dihydroquinine with a
retention time relative to quinine of about 104. The
C. It gives the reactions of chlorides (2.3.1).
chromatogram obtained with reference solution (b) shows a
principal peak due to quinidine and a peak due to
Tests dihydroquinidine, with a retention time relative to quinidine
of about 1.2. The chromatogram obtained with reference
pH (2.4.24).1.5 to 3.0.
solution (c) shows four peaks due to quinine, dihydroquinine,
Other cinchona alkaloids. Determine by liquid quinidine and dihydroquinidine which are identified by
chromatography (2.4.14). comparison of their retention times with those of the
corresponding peaks in the chromatograms obtained with
Test solution. Dilute, if necessary a suitable volume of the
reference solutions (a) and (b).
injection with the mobile phase to produce a solution
containing 0.2 per cent w/v of Quinine Dihydrochloride. The test is not valid unless (a) in the chromatogram obtained
Reference solution (a). Dissolve 20 mg of quinine sulphate with reference solution (c) the resolution between the peaks
RS, with gentle heating if necessary, in 5 ml of the mobile due to quinine and quinidine is at least 1.5 and the resolution
phase and dilute to 10 ml with the mobile phase. factor between the peaks due to dihydroquinidine and quinine
is at least 1.0 and (b) the signal-to-noise ratio of the principal
Reference solution (b). Prepare in the same manner as peak in the chromatogram obtained with reference solution
reference solution (a) but using quinidine sulphate RS in (d) is at least 5.
place of quinine sulphate RS.
Inject the test solution and allow the chromatography to
Reference solution (c). Mix equal volumes of reference proceed for2.5 times theretention time of the principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
Reference solution (d). Dilute 1 volume of reference solution normalisation, ignoring any peaks the areas of which are less
(a) to 10 volumes with the mobile phase and dilute 1 volume of than that of the peak in the chromatogram obtained with
the resulting solution to 50 volumes with the mobile phase. reference solution (d) (0.2' per cent). The content of
dihydroquinine is not greater than 10 per cent, the content of
Reference solution (e). A solution containing 0.1 per cent any related substance eluting before quinine is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance
Chromatographic system is not greater than 2.5 per cent.
- a stainless steel column 25 cm x 4.6 mm packed with Other tests. Complies with the tests stated under Parenteral
octadecylsilane bonded to porous silica (5 Ilm) (Such Preparations (Injections).
as Hypersil ODS 5 Ilm),
mobile phase: a solution prepared by dissolving 6.8 g of Assay. To an accurately measured volume containing about
potassium dihydrogen orthophosphate and 3.0 g of 0.5 g ofQuinine Dihydrochloride add 20 ml of water and 5 ml
hexylamine in 700 ml ofwatel; adjusting the pH to 2.8 of sodium hydroxide solution. Extract with successive
with 1 M orthophosphoric acid, adding 60 ml of quantities, each of 10 ml, of chloroform until complete extraction
acetonitrile and diluting to 1000 ml with watel; of the alkaloid is effected, washing each extract with the same
- flow rate. 1.5 ml per minute, two quantities, each of5 ml, of water. Remove the chloroform
spectrophotometer set at 250 nm for reference solution from the combined extracts, dissolve the residue in 50 ml of
(e) and 316 nm for the other solutions, anhydrous glacial acetic acid and add 20 ml of acetic
injection volume. 10 Ill. anhydride. Titrate with 0.1 M perchloric acid, using crystal
violet solution as indicator. Carry out a blank titration.
Inject separately reference solutions (b) and (e). If necessary,
adjust the concentration of acetonitrile in the mobile phase so 1 ml of 0.1 M perchloric acid is equivalent to 0.01987 g of
that in the chromatogram obtained with reference solution (b) C2oH24N202,2HCl.
the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Vo Storage. Store protected from light.
(the distance along the baseline betWeen the point ofinjection
and the perpendicular dropped from the maximum ofthe peak Labelling. The label states that the solution mustbe diluted
of an unretained component) being cakrihitedfrom the peak to a strength not exceeding 30 mgperml before adhiinistration
due to thiourea in the chromatogram obtained with reference and that care should be taken to ensure slow intravenous
solution (e). injection.

2024

www.webofpharma.com
IP 2010 QUININE SULPHATE

Quinine Sulphate D. To 5 ml ofa 1 per centw/v solution add 1 ml of silver nitrate


solution and stir with a glass rod; after a short interval, a
white precipitate soluble in nitric acid is produced (distinction
from many other alkaloids).
OCH s E. A 1 per cent w/v solution gives the reactions of sulphates
(2.3.1). .

Dy H
CH 2 Tests
Appearance of solution. A 2.0 per cent w/v solution in 0.1 M
hydrochloric acid is clear (2.4.1), and not more intensely
2 coloured than reference solution GYS4 (2.4.1).
pH (2.4.24). 5.7 to 6.6, determined in a 1.0 per cent w/v
(C2oH24Nz02hH2S04,2H20 Mol. Wt.783.0 suspension in water.

Quinine Sulphate is (8S,9R)-6' -methoxycinchonan-9-ol Specific optical rotation (2.4.22). -237° to -245°, determined
sulphate dihydrate. The alkaloid is obtained from the bark of in a 2.0 per cent w/v solution in 0.1 M hydrochloric acid.
various species of Cinchona. Other cinchona alkaloids. Determine by liquid
Quinine Sulphate contains not less than 99.0 per cent and not chromatography (2.4.14).
more than 101.0 per cent ofalkaloid monosulphates, calculated
Test solution. Dissolve 20 mg of the substance under
as (C2oHz4N202)Z,H2S04 on the dried basis.
examination in 5 ml of the mobile phase. Heat gently, if
Category. Antimalarial. necessary to dissolve the powder as completely as possible,
Dose. Suppressive, 300 to 600 mg daily. therapeutic, 1.2 to 2 g cool, dilute to 10 ml with the mobile phase and mix.
daily, in divided doses. Reference solution (a). Dissolve 20 mg of quinine sulphate
Description. White or almost white, needle-like crystals or a RS, with gentle heating if necessary, in 5 ml of the mobile
crystalline powder. phase and dilute to 10 ml with the mobile phase.
Reference solution (b). Prepare in the same manner as
Identification reference solution (a) but using quinidine sulphate RS in
place of quinine sulphate RS.
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G. Reference solution (c). Mix equal volumes of reference
solutions (a) and (b).
Mobile phase. A mixture of40 volumes of toluene, 24 volumes
of ether and 10 volumes of diethylamine. Reference solution (d). Dilute 1 volume ofreference solution
(a) to 10 volumes with the mobile phase and dilute 1 volume of
Test solution. Dissolve 1 g ofthe substance under examination
the resulting solution to 50 volumes with the mobile phase.
in 100 ml of methanol.
Reference solution. A 1 per cent w/v solution of quinine Reference solution (e). A solution containing 0.1 per cent
sulphate RS in methanol. w/v of thiourea in the mobile phase.

Apply to the plate 5 III of each solution. After development, Chromatographic system
dry the plate in air for 15 minutes and repeat the development. a stainless steel column 25 cm x 4.6 mm packed with
Dry the plate at 105° for 30 minutes, allow to cool and spray octadecylsilane bonded to porous silica (5 Ilm) (Such
with potassium iodoplatinate solution. The principal spot in as Hypersil ODS 5 Ilm),
the chromatogram obtained with the test solution corresponds - mobile phase: a solution prepared by dissolving 6.8 g of
to the spot in the chromatogram obtained with the reference potassium dihydrogen orthophosphate and 3.0 g of
solution. hexylamine in 700 ml of water, adjusting the pH to 2.8
with 1 M orthophosphoric acid, adding 60 ml of
B. To 5 ml ofa O.1percentw/v solution add 0.2 ml of bromine acetonitrile and diluting to 1000 ml with water,
solution and 1 ml of dilute ammonia solution; an emerald- - flow rate. 1.5 ml per minute,
green colour is produced. - spectrophotometer set at 250 urn for reference solution
C. To a 0.5 per cent w/v solution add an equal volume of dilute (e) and 316 urn for the other solutions,
sulphuric acid; a strong blue fluorescence is produced. - injection volume. 10 Ill.

2025

www.webofpharma.com
QUININE SULPHATE IP 2010

Inject separately reference solutions (b) and (e). Ifnecessary, Quinine Tablets
adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b) Quinine Sulphate Tablets
the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Vo Quinine Sulphate Tablets contain not less than 95.0 per cent
(the distance along the baseline between the point of injection and not more than 105.0 per cent of the stated amount of
and the perpendicular dropped from the maximum ofthe peak quinine sulphate, (C2oH24N202)2,H2S04,2Hp. The tablets are
of an unretained component) being calculated from the peak coated.
due to thiourea in the chromatogram obtained with reference
solution (e). Usual strengths. 100 mg; 300 mg.

Inject reference solutions (a), (b), (c) and (d). The Identification
chromatogram obtained with reference solution (a) shows a
A. Determine by thin-layer chromatography (2.4.17), coating
principal peak due to quinine and a peak due to rlihydroquinine
the plate with silica gel G.
with a retention time relative to quinine of about 1.4. The
chromatogram obtained .with reference solution (b) shows a Mobile phase. A mixture of80 volumes oftoluene, 20 volumes
principal peak due to quinidine and a peak due to of acetone and 10 volumes of diethylamine.
dihydroquinidine, with a retention time relative to quinidine Test solution. Extract a quantity of the powdered tablets
of about 1.2. The chromatogram obtained with reference containing 0.1 g ofQuinine Sulphate with 10 ml ofa mixture of
solution (c) shows four peaks due to quinine, dihydroquinine, 2 volumes of chloroform and 1 volume of ethanol (95 per
quinidine and dihydroquinidine which are identified by cent) and filter.
comparison of their retention times with those of the
Reference solution. A 1 per cent w/v solution of quinine
corresponding peaks in the chromatograms obtained with
sillphate in the same solvent mixture.
solutions (a) and (b).
Apply to the plate 2 f.!l of each solution. After development,
The test is not valid unless (a) in the chromatogram obtained dry the plate in air and spray with 0.05 M ethanolic sulphuric
with reference solution (c) the resolution between the peaks acid and then with dilute potassium iodobismuthate solution.
due to quinine and quinidine is at least 1.5 and the resolution The principal spot in the chromatogram obtained with the test
between the peaks due to dihydroquinidine and quinine is at solution corresponds to that in the chromatogram obtained
least 1.0 and (b) the signal-to-noise ratio ofthe principal peak with the reference solution.
in the chromatogram obtained with reference solution (d) is at
B'. Extract a quantity ofthe powdered tablets containing 0.1 g
least 5.
ofQuinine Sulphate with 20 ml of water and filter (solution A).
Inject the test solution and allow the chromatography to To 5 ml of solution A add 0.2 ml of bromine solution and 1 ml
proceed for 2.5 times the retention time ofthe principal peak. of dilute ammonia solution; an emerald-green colour is
Calculate the percentage content of related substances by produced.
normalisation, ignoring any peaks the areas of which are less
C. Solution A is leva-rotatory.
than that of the peak in the chromatogram obtained with
reference solution (d) (0.2 per cent). The content of D. Solution A gives the reactions ofsulphates (2.3.1).
dihydroquinine is not greater than 10 per cent, the content of Tests
any related substance eluting before quinine is not greater
than 5 per cent and the content of any other related substance Dissolution (2.5.2).
is not greater than 2.5 per cent. Apparatus No.2,
Sulphated ash (2.3.18). Not more than 0.1 per cent. Medium. 900 ml of 0.1 M hydrochloric acid,
Speed and time. 100 rpm and 30 minutes.
Loss on drying (2.4.19).3.0 to 5.0 per cent, determined on
1.0 g by drying in an oven at 105°. Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance of the filtrate, suitably diluted if necessary, at
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of the maximum at about 348 urn (2.4.7). Calculate the content of
10 ml bf chloroform and add 20 ml of acetic anhydride. Titrate C2oH24N202, H 2S04, 2H20 in the medium from a solution of
with 0.1 M perchloric acid, determining the end-point known concentration of quinine sulphate RS.
potentiometrically (2.4.25). Carry out a blank titration.
D. Not less than 70 per cent of the stated amount of
1 ml of 0.1 M perchloric acid is equivalent to 0.02490 g of C2oH24Nz02, H2S04, 2H20.
(C2oH24N202)2,H2S04' Other cinchona alkaloids. Determine by liquid
Storage. Store protected from light. chromatography (2.4.14).

2026

www.webofpharma.com
IP 2010 QUINIODOCHLOR

Test solution. Remove any coating from the tablets and mix a comparison of their retention times with those of the
quantity of the powdered tablet cores containing 50 mg of corresponding peaks in the chromatograms obtained with
Quinine Sulphate with 20 ml ofthe mobile phase. Heat gently reference solutions (a) and (b).
to dissolve the powder as completely as possible, cool, dilute The test is not valid unless (a) in the chromatogram obtained
to 25 ml with the mobile phase and filter, discarding the first with reference solution (c) the resolution between the peaks
few ml ofthe filtrate. due to quinine and quinidine is at least 1.5 and the resolution
Reference solution (a). Dissolve 20 mg of quinine sulphate between the peaks due to dihydroquinidine and quinine is at
RS, with gentle heating if necessary, in 5 ml of the mobile least 1.0 and (b) the signal-to-noise ratio ofthe principal peak
phase and dilute to 10 ml with the mobile phase. in the chromatogram obtained with reference solution (d) is at
Reference solution (b). Prepare in the same manner as least 5.
reference solution (a) but using quinidine sulphate RS in Inject the test solution and allow the chromatography to
place of quinine sulphate RS. proceed for 2.5 times the retention time of the principal peak.
Reference solution (c). Mix equal volumes of reference Calculate the percentage content of related substances by
solutions (a) and (b). normalisation, ignoring any peaks the areas of which are less
Reference solution (d). Dilute 1 volume of reference solution than that of the peak in the chromatogram obtained with
(a) to 10 volumes with the mobile phase and dilute 1 volume of reference solution (d) (0.2 per cent). The content of
the resulting solution to 50 volumes with the mobile phase. dihydroquinine is not greater than 10 per cent, the content of
any related. substance eluting before quinine is not greater
Reference solution (e). A solution containing 0.1 per cent than 5 per cent and the content of any other related substance
w/v of thiourea in the mobile phase.
is not greater than 2.5 per cent.
Chromatographic system
Other tests. Comply with the tests stated under Tablets.
- a stainless steel column 25 cm x 4.6 rom packed with
octadecylsilane bonded to porous silica (5 /lm) (Such Assay. Weigh and powder 20 tablets. Weigh accurately a
as Hypersil ODS 5 /lm), . quantity of the powder containing about 004 g of Quinine
mobile phase: a solution prepared by dissolving 6.8 g of Sulphate, dissolve as completely as possible in 40 ml of acetic
potassium dihydrogen orthophosphate and 3.0 g of anhydride with the aid of heat and cool. Filter, if necessary
hexylamine in 700 ml ofwatel; adjusting the pH to 2.8 through Whatrnan No.1 filter paper and rinse with an additional
with 1 M orthophosphoric acid, adding 60 ml of 40 ml of acetic anhydride in small volumes. Titrate with 0.1 M
acetonitrile and diluting to 1000 ml with water, perchloric acid, using crystal violet solution as indicator.
flow rate. 1.5 ml per minute, Carry out a blank titration.
spectrophotometer set at 250 urn for reference solution 1 ml of 0.1 M perchloric acid is equivalent to 0.02610 g of
(e) and 316 urn for the other solutions, (C2oH2~202)2,H2S04,2H20.
injection volume. 10 /ll.
Storage. Store protected from light.
Injectseparately reference solutions (b) and (e). Ifnecessary,
adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b)
the capacity factor ofthe peak due to quinidine is 3.5 to 4.5, Va Quiniodochlor
(the distance along the baseline between the point ofinjection
and the perpendicular dropped from the maximum ofthe peak
Clioquinol; Iodochlorhydroxyquinoline;
of an unretained component) being calculated from the peak Iodochlorhydroxyquin
due to thiourea in the chromatogram obtained with reference
OH
solution (e).
Inject reference solutions (a), (b), (c) and (d). The 'hN~
~
chromatogram obtained with reference solution (a) shows a
principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 104. The CI
chromatogram obtained with reference solution (b) shows a
~HsCI1NO Mol. Wt.305.5
principal peak due to quinidine and a peak due to
dihydroquinidine, with a retention time relative to quinidine Quiniodochlor is 5-chloro-7-iodoquinolin-8-ol.
of about 1.2. The chromatogram obtained with reference Quiniodochlor contains not less than 97.0 per cent and not
solution (c) shows four peaks due to quinine, dihydroquinine, more than 103.0 per cent ofCgHsClINO, calculated on the dried
quinidine and dihydroquinidine which are identified by basis.

2027

www.webofpharma.com
QUINIODOCHLOR IP 2010

Category. Antiamoebic; topical and intestinal antiseptic. under examination and 0.5 ml ofpyridine, mix, allow to stand
Dose. 750 mg to 1.5 g daily, in divided doses. for 15 minutes and add 5 ml of hexane.
Reference solution (b). Treat a mixture of0.1 g ofthe substance
Description. A yellowish white to brownish yellow powder;
under examination and 0.5 ml ofpyridine as described for the
odoUr, faint and characteristic.
test solution.
Identification Chromatographic system
- a glass column 1.5 m x 4 mm, packed with silanised
A. Determine by infrared absorption spectrophotometry (2.4.6). diatomaceous support (100 to 120 mesh) coated with 3
Compare the spectrum with that obtained with quiniodochlor
per cent w/w ofmethyl silicone gum,
RS or with the reference spectrum of quiniodochlor.
- temperature:
B. When examined in the range 230 Dill to 360 Dill (2.4.7), a column.190°,
0.0005 per cent w/v solution in 3 M hydrochloric acid shows inlet port and detector. 240°,
an absorption maximum at about 267 Dill. - flame ionisation detector,
- nitrogen as carrier gas.
C. Bum 20 mg by the oxygen-flask method (2.3.34), using 5 ml
of 2 M sodium hydroxide as the absorbing liquid and dilute to In the chromatogram obtained with the test solution the peaks
25 ml with water. To 5 ml add 1 ml of silver nitrate solution; a following the solvent peak, in order of emergence, are due to
yellow precipitate is produced. Add 5 ml of 5 M ammonia, (a) 5-chloro-8-hydroxyquinoline, (b) 5,7-dichloro-8-hydroxy-
shake, filter and acidifY the filtrate with nitric acid; a white quinoline, (c) the internal standard, (d) quiniodochlor and (e)
precipitate is produced. 5,7-diiodo-8-hydroxyquino1ine.
In the chromatogram obtained with reference solution (b)
Tests
calculate the content of 5-chloro-8-hydroxy-quinoline, 5,7~
Acidity or alkalinity. Shake 0.5 g with 10 ml ofwater previously dichloro-8-hydroxyquinoline and 5,7-diiodo-8-hydroxy-
neutralised to phenolphthalein solution. The solution is quinoline by reference to the corresponding peaks in the
colourless and not more than 0.05 ml of 0.1 M sodium hydroxide chromatogram obtained with the test solution.
is required to change the colour of the solution to pink. The total content of the named impurities does not exceed
Free iodine. Shake 1.0 g with a solution of 1 g of potassium 3.0 per cent w/w, the content of any other impurity does not
iodide in 20 ml of water for 30 seconds, allow to stand for exceed 0.2 per cent w/w and the sum of the contents is not
5 minutes and filter. To 10 ml of the filtrate add 1 ml of 1 M more than 4.0 per cent w/w.
sulphuric acid and 2 ml of chloroform and shake. Any colour Sulphated ash (2.3.18). Not more than 0.2 per cent.
in the chloroform layer is discharged on the addition of0.1 ml
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
of 0.005 M sodium thiosulphate.
on 1.0 g by drying over phosphorus pentoxide at a pressure
Halide ions. Shake 0.5 g with 25 ml of water for 1 minute and not exceeding 0.7 kPa for 24 hours.
filter. To the filtrate add 0.5 ml of2 M nitric acid and 0.5 ml of
Assay. Weigh accurately about 0.3 g and dissolve in 25 ml of
0.1 M silver nitrate and allow to stand for 5 minutes. Any
anhydrous pyridine. Titrate with 0.1 M tetrabutylammonium
opalescence produced is not more intense than that obtained
hydroxide, determining the end-point potentiometrically
by adding 0.5 ml of 0.1 M silver nitrate to 25 ml of water
(2.4.25). Carry out a blank titration.
containing 0.5 ml of 2 M nitric acid and 0.2 ml of 0.01 M
hydrochloric acid and allowing to stand for 5 minutes. 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
0.03055 gofCgHsClINO.
Related substances. Determine by gas chromatography
(2.4.13). Storage. Store protected from light.

Test solution. Add 0.5 ml ofN,O-bis (trimethylsilyl)acetamide


to 0.5 ml ofa solution in pyridine containing 0.4 per cent w/v Quiniodochlor Cream
of each of 5-chloro-8- hydroxyquinoline, 5,7-dichloro-8-
hydroxy- quinoline and 5-chloro-7-iodo-8-hydroxyquinoline Clioquinol Cream; Iodochlorhydroxyquinoline Cream;
and 0.04 per cent w/v of the substance under examination, Iodochlorhydroxyquin Cream
mix, allow to stand for 15 minutes and add 5 ml ofa 0.05 per Quiniodochlor Cream contains Quiniodochlor in a suitable
cent w/v solution of dibutylphthalate (internal standard) in base.
hexane.
Quiniodochlor Cream contains not less than 90.0 per cent and
Reference solution (a). Add 0.5 m1 of N,O-bis not more than 110.0 per cent of the stated amount of
(trimethylsilyl)acetamide to a mixture of 0.1 g ofthe substance quiniodochlor, CgHsClINO.

2028

www.webofpharma.com
IP 2010 QUINIODOCHLOR OINTMENT

Usual strength. 4 per cent w/v. temperature:


column. 165°,
Identification inlet port 170° and detector 250°,
- flame ionization detector,
A. In the Assay, the principal peak in the chromatogram flow rate. 30 inl per minute ofhelium as the carrier gas.
obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. The test Inject the reference solution. The test is not valid unless the
solution and the reference solution prepared by using 1.0 ml resolution between the quiniodochlor and the intemal standard
of pyridine instead of the internal standard solution. peaks is not less than 3.0. The relative retention time with
reference to pyrene for quiniodochlor is about 0.6.
B. Weigh accurately a quantity containing about 25 mg of
quiniodochlor, in a 100 ml volumetric flask, add 75 ml of 25 per Inject the test solution and the reference solution.
cent v/v solution of hydrochloric acid, heat on a steam bath Calculate the content ofCgHsClINO in the Cream.
to melt the cream, shake vigorously to extract the quiniodochlor.
Storage. Store protected from light and moisture.
Cool under running water, and dilute to the volume with the
same solvent. Dilute 3.0 ml ofthe filtrate to 100.0 ml with the
same solvent.
When examined in the range 200 nm to 400 run (2.4.7), the Quiniodochlor Ointment
solution shows an absorption maximum at about 267 run.
Clioquinol Ointment; Iodochlorhydroxyquinoline
Tests Ointment; Iodochlorhydroxyquin Ointment

Other tests. Complies with the tests stated under Creams. Quiniodochlor Ointment contains Quiniodochlor in a suitable
base.
Assay. Determine by gas chromatography (2.4.13).
Quiniodochlor Ointment contains not less than 90.0 per cent
Internal standard solution. A 0.2 per cent w/v solution of and not more than 110.0 per cent of the stated amount of
pyrene in pyridine. quiniodochlor, CgHsCIINO.
Solvent mixture. 80 volumes ofpyridine and 20 volumes of n- Usual strength. 4 per cent w/v.
hexane.
Test solution. Weigh accurately a quantity containing about Identification
150 mg of quiniodochlor, in a 60-ml separator. Place the A. In the Assay, the principal peak in the chromatogram
separator on its side in a vacuum oven at a pressure of about obtained with the test solution corresponds to the peak in the
10 mm of mercury at 45° for 4 hours. Remove the separator chromatogram obtained with the reference solution. The test
from the oven, allow to cool, add 15 ml ofthe solvent mixture, solution and the reference solution prepared by using 1.0 ml
insert a polytefstopper, and mix. Transfer the mixture to a 50- of pyridine instead of the internal standard solution.
ml volumetric flask, and rinse the separator with two 15 ml
portions of the solvent mixture, shaking each time for 30 B. Weigh accurately a quantity containing about 25 mg of
seconds. Transfer both rinsings to the volumetric flask, dilute quiniodochlorin a 100-ml volumetric flask, add 75 ml of 25 per
with the solvent mixture to volume, andmix. Transfer 1.0 ml to cent v/v solution of hydrochloric acid, heat on steam bath to
a screw-capped glass vial fitted with a septum, add 1.0 ml Qf melt the ointment, shake vigorously to extract the
bis(trimethylsilyl)acetamide and 1.0 ml of internal standard quiniodochlor. Cool under running water, dilute to volume
solution, attach the cap, and mix. Heat in a water-bath at 50° with the same solvent. Filter and dilute 3.0 ml ofthis solution
for 15 minutes, and coo 1. to 100.0 ml with the same solvent.
When examined in the range 200 run to 400 nm (2.4.7), the
Reference solution. A 0.3 per cent w/v solution of
solution shows an absorption maximum at about 267 nm.
quiniodochlor RS in the solvent mixture. Transfer 1.0 ml to a
screw-capped glass vial fitted with a septum, add 1.0 ml of
bis(trimethylsilyl)acetamide and 1.0 ml of internal standard Tests
solution, attach the cap, and mix. Heat in a water-bath at 50° Other tests. Complies with the tests stated under Ointments.
for 15 minutes, and cool.
Assay. Determine by gas chromatography (2.4.13).
Chromatographic system
a glass column 1.83 m x 2 mill, packed with 3 per cent Internal standard solution. A 0.2 per cent w/v solution of
liquid phase G3 on 80 to 100-mesh support SlAB, pyrene in pyridine.

2029

www.webofpharma.com
QUINIODOCHLOR OINTMENT IP 2010

Solvent mixture. 80 volumes ofpyridine and 20 volumes of n- Usual strength. 250 mg.
hexane.
Test solution. Transfer an accurately weighed quantity of the
Identification
ointment, containing about 150 mg of Quiniodochlor, to a Triturate a quantity of the powdered tablets containing about
suitable conical flask, add 75 ml of n-hexane, and mix. Add 15 250 mg ofQuiniodochlor with 20 ml of acetone, filter and add
ml of dimethylformamide, and mix for 1 minute. Allow the 20 ml of water to the filtrate. Collect the precipitate formed on
layers to separate, and transfer the lower layer to a 50-ml a filter and dry at 105°. The residue complies with the following
volumetric flask, repeat the extraction with separate 15 ml and tests.
10 ml portions of dimethylformamide, and transfer the lower
layers to the 50-ml volumetric flask. Dilute with A. Determine by infrared absorption spectrophotometry (2.4.6).
dimethylformamide to volume, and mix. Transfer 1.0 ml ofthis Compare the spectrum with that obtained with quiniodochlor
solution to a screw-capped glass vial fitted with a septum, RS or with the reference spectrum of quiniodochlor.
and evaporate at about 60° under a stream of nitrogen to B. When examined in the range 230 nm to 360 nm (2.4.7), a
dryness. Add 1.0 ml of a mixture of internal standard solution 0.0005 per cent w/v solution in 3 M hydrochloric acid shows
to the residue, add 1.0 ml of bis(trimethylsilyl)acetamide and an absorption maximum at about 267 nm.
1.0 ml of internal standard solution, attach the cap, and mix.
Heat in a water-bath at 50° for 15 minutes and cool.
Tests
Reference solution. A 0.3 per cent w/v solution of
qUiniodochlor RS in the solvent mixture. Transfer 1.0 ml to a Disintegration (2.5.1). Maximum time, 30 minutes.
screw-capped glass vial fitted with a septum, add 1.0 ml of
Other tests. Comply with the tests stated under Tablets.
bis(trimethylsilyl)acetamide and 1.0 ml of internal standard
solution, attach the cap, and mix. Heat in a water-bath at 50° Assay. Weigh and finely powder 20 tablets. Weigh accurately
for 15 minutes, and cool. a quantity ofthe powder containing 0.125 g ofQuiniodochlor,
Chromatographic system shake with 20 ml of hot 2-methoxyethanol, decant the hot
- a glass column 1.83 m x 2 mm, packed with 3 per cent supernatant liquid through a fine filter. Repeat the extraction
liquid phase G3 on 80 to 1OO-mesh support SlAB, with two further quantities, of 20 ml and 10 ml, of
- temperature: 2-methoxyethanol, combine the filtered extracts and dilute to
column. 165°, 50.0 ml with 2-methoxyethanol. To 5.0 ml ofthis solution add
1 ml of water and sufficient of a mixture of 24 volumes of
inlet port 170° and detector 250°,
2-methoxyethanol and 6 volumes of water to produce 50.0 ml.
- flame ionization detector,
To 10.0 ml ofthe solution add 10 ml of 2-methoxyethanol and
- flow rate.· 30 ml per minute ofhelium as the carrier gas.
2 ml ofa solution prepared by dissolving 0.5 g offerric chloride
Inject the reference solution. The test is not valid unless the hexahydrate in 80 ml of 2-methoxyethanol and adding 0.1 ml
resolution between the quiniodochlor and the internal standard of hydrochloric acid and sufficient 2-methoxyethanol to
peaks is not less than 3. The relative retention time with produce 100 ml. Dilute the solution to 25.0 ml with
reference to pyrene for quiniodochlor is about 0.6. 2-methoxyethanol and measure the absorbance of the
resulting solution at the maximum at about 650 nm (2.4:7),
Inject the test solution and the reference solution.
using as blank a solution prepared by treating 10 ml of the
Calculate the content ofC 9H sCIINO in the Ointment. aqueous 2-methoxyethanol in the same manner beginning at
the words "add 10 ml of 2-methoxyethanol... ..".
Storage. Store protected from light and moisture.
Calculate the content of C 9H sCIINO from the absorbance
obtained using 10.0 ml ofa solution prepared in the following
manner. Dissolve 0.125 g of quiniodochlor RS in sufficient 2-
methoxyethanol to produce 50.0 ml, warming to effect solution;
Quiniodochlor Tablets
add 1 ml of water to 5.0 ml ofthe solution and add sufficient of
Clioquinol Tablets; Iodochlorhydroxyquinoline Tablets; the mixture of24 volumes of 2-methoxyethanol and 6 volumes
Iodochlorhydroxyquin Tablets of water to produce 50.0 ml. Using 10.0 ml of this solution
repeat the operation beginning at the words "add 10 ml of
90.0per
Quiniodochlorta1:>lets contaiiiii6t less thaii cent and 2-methoxyethanol.. .."
not more than 110.0 per cent of the stated amount of
quiniodochlor, C9HsClINO. Storage. Store protected from light.

2030

www.webofpharma.com
IP 2010 QUINI0DOCHLOR AND HYDROCORTISONE CREAM

Quiniodocblor and Hydrocortisone the filtrate with 2 M nitric acid; a white precipitate is produced
which darkens on exposure to light.
Cream
Clioquinol and Hydrocortisone Cream; Tests
Iodochlorhydroxyquinoline and Hydrocortisone Other tests. Complies with the tests stated under Creams.
Cream; Iodochlorhydroxyquin and Hydrocortisone
Cream Assay. For hydrocortisone - Determine by liquid
chromatography (2.4.14).
Quiniodochlor and Hydrocortisone Cream contains
Hydrocortisone and Quiniodochlor in a suitable base. Test solution (a). Add 30 ml of 2,2,4-trimethylpentane to a
quantity of the cream containing 10 mg of Hydrocortisone
Quiniodochlor and Hydrocortisone Cream contains not less and warm on a water-bath until the preparation has melted.
than 90.0 per cent and not more than 110.0 per cent of the Extract the warm mixture with successive quantities ono, 20
stated amount of quiniodochlor, CgHsCIINO and not less than and 20 ml of methanol (80 per cent), combine the aqueous
92.5 per cent and not more than 107.5 per cent of the stated methanolic layers, cool to about 20° and dilute to 100 ml with
amount of hydrocortisone, CZIH300s. the same solvent.
Usual Strength. Quiniodochlor 4 per cent w/w and Test solution (b). Prepare in the same manner as test solution
Hydrocortisone 1 per cent w/w. (a) but adding)O ml of a 4 per cent v/v solution of
bromobenzene in methanol to the cooled methanolic extract
Identification
and dilute to 100.0 ml with methanol (80 per cent).
A. Determine by thin-layer chromatography (2.4.17), coating
Reference solution. Dissolve 5 mg of hydrocortisone RS in 5
the plate with silanised silica gel G.
ml of a 4 per cent v/v solution of bromobenzene (internal
Mobile phase. A mixture of77 volumes of dichloromethane, standard) in methanol and dilute to 50 ml with methanol (80
15 volumes of ether, 8 volumes of methanol and 1.2 volumes per cent).
of water.
Chromatographic system .
Test solution. Disperse by warming and shaking, a quantity of a stainless steel column 25 em x 5 mm, packed with
the cream containing 2.5 g of Hydrocortisone in 10 ml of octadecylsilane bonded to porous silica (5 11m) (Such
ethanol (95 percent), cool, allow to stand at 0° for 30 minutes, as Spherisorb ODS 1),
filter and use the filtrate. - mobile phase: methanol (65 per cent),
Reference solution (a). A 0.25 per cent w/v solution of - flow rate. 1 ml per minute,
hydrocortisone RS in ethanol (95 per cent). - spectrophotometer set at 242 nm,
- injection volume. 10 Ill.
Reference solution (b). Dissolve 12.5 mg of hydrocortisone
RS in 5 ml of test solution. Calculate the content ofCzIH300s in the cream.

Apply to the plate 5 III of each solution. After development, For quiniodochlor - Weigh accurately a quantity of the
dry the plate in air and spray with alkaline tefrazoliurn blue cream containing about 25 mg ofquiniodochlor, add 80 ml of
solution. The principal spot in the chromatogram obtained a hot mixture of 6 volumes of water and 24 volumes of
with test solution corresponds to that in the ~hromatogram 2-methoxyethanol and heat on a water-bath for 5 minutes.
obtained with reference solution (a). The principal spot in the Cool in ice for 10 minutes, allow to warm to room temperature,
chromatogram obtained with reference solution (b) appears dilute to 100 ml with a mixture of 6 volumes of water and
as a single, compact spot. 24 volumes of 2-methoxyethanol, mix and filter. To 10 ml ofthe
filtrate, add 10 ml of 2-methoxyethanol and 2 ml ofa solution
B. In the Assay, the principal peak in the chromatogram due to
prepared by dissolving 0.5 g of iron(III) chloride hexahydrate
hydrocortisone obtained with test solution (b) corresponds
in 80 ml of 2-methoxyeth~nol and adding 0.1 ml of
to the peak due to hydrocortisone in the chromatogram
hydrochloric acid and sufficient 2-methoxyethanol to
obtained with reference solution.
produce 100 m!. Dilute the solution to 25 ml with
C. Fuse a quantity of the cream containing 0.1 g of 2-methoxyethanol and measure the absorbance of the
quiniodochlor with anhydrous sodium carbonate, dissolve resulting solution at the maximum at about 650 nm (2.4.7),
the fused mass in water and acidify with 2 M nitric acid. Add using in the reference cell a solution prepared by treating
silver nitrate solution; a pale yellow precipitate is produced 10 ml ofthe mixture of6 volumes of water and 24 volumes of
which is insoluble in 5 M ammonia. Add 5 M ammonia until 2-methoxyethanol in the same manner beginning at the words
the solution becomes alkaline, boil gently, filter and acidify "add 10 ml of 2-methoxyethanol...... ".

2031

www.webofpharma.com
QUINIODOCHLOR AND HYDROCORTISONE OINTMENT IP 2010

Repeat the operation beginning at the words 'add 10 ml of B. In the Assay, the principal peak in the chromatogram due to
2-methoxyethanol using 10 ml ofa solution prepared in the hydrocortisone obtained with test solution (b) corresponds
following manner. Dissolve 0.125 g of clioquinol RS in to the peak due to hydrocortisone in the chromatogram
sufficient 2-methoxyethanol to produce 50 ml; warming to obtained with reference solution.
effect solution; add 1 ml of water to 5 ml of the solution and
C. Fuse a quantity of the ointment containing 0.1 g of
add sufficient of a mixture of 6 volumes of water and
Quiniodochlor with anhydrous sodium carbonate, dissolve
24 volumes of 2-methoxyethanol to produce 50 ml.
the fused mass in water and acidify with 2 M nitric acid. Add
Calculate the content ofC 9HsCIINO in the cream. silver nitrate solution; a pale yellow precipitate is produced
Storage. Store protected from light. which is insoluble in 5 M ammonia. Add 5 M ammonia until
the solution becomes alkaline, boil gently, filter and acidify
the filtrate with 2 M nitric acid; a white precipitate is produced
which darkens on exposure to light.

Quiniodochlor and Hydrocortisone Tests


Ointment
Other tests. Complies with the tests stated under Ointments.
Clioquinol and Hydrocortisone Ointment;
Assay. For hydrocortisone Determine by liquid
Iodochlorhydroxyquinoline and Hydrocortisone
chromatography (2A.14).
Ointment; Iodochlorhydroxyquin and Hydrocortisone
Ointment Test solution (a). Add 30 ml of 2,2,4-trimethylpentane to a
quantity of the ointment containing 10 mg ofHydrocortisone
Quiniodochlor and Hydrocortisone Ointment contains
and warm on a water-bath until the preparation has 'melted.
Quiniodochlor and Hydrocortisone in a suitable base.
Extract the warm mixture with successive quantities ono, 20
Quiniodochlor and Hydrocortisone Ointment contains not less and 20 ml of methanol (80 per cent), combine the aqueous
than 90.0 per cent and not more than 110.0 per cent of the methanolic layers, cool to about 20 0 and dilute to 100 ml with
stated amount of quiniodochlor, C9HsClINO and not less than the same solvent.
92.5 per cent and not more than 107.5 per cent of the stated
Test solution (b). Prepare at the same manner as test solution
amount of hydrocortisone, CZIH300S'
(a) but adding 10 ml of a 4 per cent v/v solution of
Identification bromobenzene in methanol to the cooled methanolic extract
and dilute to 100.0 ml with methanol (80 per cent).
A. Determine by thin-layer chromatography (2.4.17), coating
Reference solution. Dissolve 5 mg of hydrocortisone RS in 5
the plate with silanised silica gel G.
ml of a 4 per cent v/v solution of bromobenzene (internal
Mobile phase. A mixture of 77 volumes of dichloromethane, standard) in methanol and dilute to 50 ml with methanol (80
15 volumes of ether, 8 volumes of methanol and 1.2 volumes per cent).
of water.
Chromatographic system
Test solution. Disperse by warming and shaking, a quantity of - a stainless steel column 25 cm x 5 mm, packed with
the ointment containing 2.5 g of Hydrocortisone in 10 ml of octadecylsilane bonded to porous silica (5 Ilm) (Such
ethanol (95 per cent), cool, and allow to stand at 0 0 for 30 as Spherisorb ODS 1),
minutes, filter and use the filtrate. mobile phase: methanol (65 per cent),
Reference solution (a). A 0.25 per cent w/v solution of flow rate. 1 ml per minute,
hydrocortisone RS in ethanol (95 per cent). spectrophotometer set at 242 run,
iI1jection volume. 10 Ill.
Referencesolution (b). Dissolve 12.5 mg of hydrocortisone
RS in 5 ml of test solution. Calculate the content ofCzIH300s in the ointment.
Apply to the plate 5 III of each solution. After development, For Quiniodochlor- Weigh accurately a quantity of the
dry the plate in air and spray with alkaline tetrazolium blue ointment containing 25 mg of Clioquinol, add 80 mlof a hot
solution. The principal spot in the chromatogram obtained mixture of24 volumes of 2-methoxy-ethanol and 6 volumes
with test solution corresponds to that in the chromatogram of water and heat on a water-bath for 5 minutes. Cool in ice for
obtained with reference solution (a). The principal spot in the 10 minutes, allow to warm to room temperature, dilute to 100
chromatogram obtained with reference solution (b) appears ml with a mixture of24 volumes of 2-methoxy-ethanol and 6
as a single, compact spot. volumes of water, mix and filter. To 10 rn1 ofthe filtrate, add 10

2032

www.webofpharma.com
IP 2010 QUINIODOCHLOR AND HYDROCORTISONE OINTMENT

ml of 2-methoxyethanol and 2 ml of a solution prepared by Repeat the operation beginning at the words 'add 10 ml of 2-
dissolving 0.5 g of iron(III) chloride hexahydrate in 80 ml of methoxyethanol using 10 ml of a solution prepared in the
2-methoxyethanol and adding 0.1 ml of hydrochloric acid following manner. Dissolve 0.125 g of clioquinol RS in
and sufficient 2-methoxyethanol to produce 100 m!. Dilute sufficient 2-methoxyethanol to produce 50 ml, warming to
the solution to 25 ml with 2-methoxyethanol and measure the effect solution; add 1 ml of water to 5 ml of the solution and
absorbance of the resulting solution at the maximum at 650 add sufficient of a mixture of 6 volumes of water and 24
urn (2.4.7), using in the reference cell a solution prepared by volumes of 2-methoxyethanol to produce 50 m!.
treating 10 m1 ofa mixture of24 volumes of 2~methoxy-ethanol
Calculate the content ofC 9HsCllNO in the ointment.
and 6 volumes of water in the same manner beginning at the
words "add 10 ml of 2-methoxyethanol..... n. Storage. Store protected from light.

2033

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Rabeprazole Sodium 2037


Rabeprazole Tablets 2037
Ramipril 2038
Ramipril Capsules 2039
Ramipril Tablets 2040
Ramipril and Hydrochlorothiazide Tablets 2041
Ranitidine Hydrochloride 2043
Ranitidine Injection 2044
Ranitidine Tablets 2045
Purified Rayon 2046
Reserpine 2047
Reserpine Injection 2048
Reserpine Tablets 2049
Ribavirin 2050
Ribavirin Inhalation Solution 2051
Riboflavin 2052
Riboflavin SodiumPhosphate 2053
Riboflavin Tablets 2054
Rifumpicin 2054
Rifampicin Capsules 2055
Rifampicin Oral Suspension 2056
Rifampicin Tablets 2058
Rifampicin and Isoniazid Tablets 2059
Rifampicin, Isoniazid and Ethambutol Tablets 2061
Rifampicin, Isoniazid and Pyrazinamide Tablets 2063
Rifampicin, Isoniazid, Pyrazinamide and Ethambutol Tablets 2065
Ritonavir 2066
Ritonavir Capsules 2067
Ritonavir Tablets 2068
Rosiglitazone Maleate 2070

2035

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Rosiglitazone Tablets 2070


Rosuvastatin Calcium 2071
Rosuvastatin Tablets 2072
Roxithromycin 2073
Roxithromycin Tablets 2075

2036

www.webofpharma.com
IP 2010 RABEPRAZOLE TABLETS

Rabeprazole Sodium Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Water (2.3.43). Not more than 7.0 per cent, determined on 0.3 g,
Assay. Determine by liquid chromatography (2.4.14).
Solvent mixture. 80 volumes of methanol, 20 volumes of water
and 0.1 volume of diethylamine.
Mol. Wt. 381.4 Test solution. Dissolve 0.1 g of the substance under
Rabeprazole sodium is 2-( {[4-(3-methoxypropoxy)-3-methyl- examination in 100.0 ml ofsolvent mixture. Dilute 5.0 ml ofthe
2-pyridinyl]methyl} sulphinyl)-IH-benzimidazole sodium. solution to 100.0 ml with the same solvent.

Rabeprazole sodium contains not less than 98.0 per cent and Reference solution. A 0.005 per cent w/v solution of
not more than 102.0 per cent ofClsHzoN303S,Na, calculated on
rabeprazole sodium RS in the solvent mixture.
the anhydrous basis. Chromatographic system
Category. Antiulcer. - a stainless steel column 25 cm x 4.6 mm packed with
octylsilane bonded to porous silica (5 !!m) (Such as
Description. A white to light yellow, crystalline powder, Hypersil keystone betabasic C s),
hygroscopic. - column temperature 40°,
mobile phase: a mixture of 72 volumes of 0.1 M phos-
Identification phate buffer pH 7.0 and 28 volumes of acetonitrile,
A. Determine by infrared absorption spectrophotometry (2.4.6). - flow rate. 1.4 ml per minute,
Compare the spectrum with that obtained with rabeprazole - spectrophotometer set at 282 nm,
sodium RS. - injection volume. 10 !!l.

B. A 10 per cent w/v solution in carbon dioxide-free water Inject the reference solution. The test is not valid unless the
gives reaction ofsodium (2.3.1). tailing factor is not more than 2.0 and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
Tests
Inject the test solution and the reference solution.
Related substances. Determine by liquid chromatography Calculate the percentage content ofClsHzoN303S,Na.
(2.4.14).
Storage. Store protected from light and moisture.
Solvent mixture. 80 volumes of methanol, 20 volumes of water
and 0.1 volume of diethylamine.
Test solution. Dissolve 50 mg of the substance under
examination in 100 ml with the solvent mixture. Rabeprazole Tablets
Reference solution (a). A 0.05 per cent w/v solution of Rabeprazole Sodium Tablets
rabeprazole sodium RS in the solvent mixture.
Rabeprazole Tablets contain not less than 90.0 per cent and
Reference solution (b). Dilute 1 ml ofreference solution (a) to not more than 110.0 per cent of the stated amount of
100 ml with solvent mixture. rabeprazole sodium, ClsHzoN303SNa. The tablets are enteric
Chromatographic system as described under Assay. coated.

Inject reference solution (b). The test is not valid unless the Usual strength. 20 mg.
tailing factor is not more than 2.0 and the column efficiency in
Identification
not less than 2000 theoretical plates.
Inject the test solution and reference solution (b). Run the In the Assay, the principal peak in the chromatogram obtained
chromatogram three times of the principal peale In the with the test solution corresponds to the peak in the
chromatogram obtained with the test solution, the area ofany chromatogram obtained with the reference solution.
secondary peak is not more than 0.5 times the area ofthe peak Tests
in the chromatogram obtained with reference solution (b)
(0.5 per cent) and the sum of areas of all the secondary peaks Dissolution (2.5.2).
is not more than 1.5 times the area ofthe peak in the chroma- Apparatus No.1,
togram obtained with the reference solution (b) (1.5 per cent). Medium. 900 ml of 0.1 M hydrochloric acid,

2037

www.webofpharma.com
RABEPRAZOLE TABLETS IP2010

Speed and time. 50 rpm and 120 minutes. Solvent mixture. 80 volumes of methanol, 20 volumes of water
and 0.1 volume of diethylamine.
Replace the 0.1 M hydrochloric acid with phosphate buffer
pH 7.4. Run the apparatus at 75 ipm for 45 minutes. Withdraw Test solution. Weigh and powder 20 tablets. Weigh accurately
a suitable volume of the medium and filter. Measure the a quantity of the powdered tablet containing 50 mg of
absorbance ofthe filtered solution immediately, suitably diluted Rabeprazole Sodium, disperse in 20 ml of 0.1 M sodium
with the dissolution medium, ifnecessary, at the maximum at hydroxide and dilute to 100.0 ml with solvent mixture, filter.
about 291 nm (2.4.7). Calculate the content ofCIsHzoN303SNa
Reference solution. Weigh accurately about 25 mg of
in the medium from the absorbance obtained from a solution
rabeprazole sodium RS, dissolve in 10 ml of 0.1 M sodium
of known concentration of rabeprazole sodium RS, prepared
hydroxide and dilute to 50.0 ml with solvent mixture.
by dissolving in minimum quantity ofa mixture of?5 volumes
of acetonitrile and 25 volumes of methanol and suitably Chromatographic system
diluted with the dissolution medium. a stainless steel column 25 cm ' 4.6 mm packed with
octadecylsilane bonded to porous silica (5 /lm),
D. Not less than 70 per cent of the stated amount of mobile phase: a mixture of65 volumes of 0.15 per cent
ClsHzoN303SNa. w/v solution of potassium dihydrogen phosphate
Related substances. Determine by liquid chromatography previously adjusted pH to 6.0 with orthophosphoric
(2.4.14). acid or sodium hydroxide solution and 35 volumes of
acetonitrile,
Solvent mixture. 80 volumes of methanol, 20 volumes of water
flow rate. 1 ml per minute,
and 0.1 volume of diethylamine.
spectrophotometer set at 280 nm,
Test solution. Weigh accurately a quantity of the powdered injection volume. 10 /ll.
tablet containing 50 mg of Rabeprazole Sodium, disperse in
Inject the reference solution. The test is not valid unless the
100 ml ofsolvent mixture and filter. tailing factor is not more than 2.0 and the relative standard
Reference solution (a). A 0.05 per cent w/v solution of deviation for replicate injections is not more than 2.0 per cent.
rabeprazole sodium RS in the solvent mixture. Inject the test solution and the reference solution.
Reference solution (b). Dilute 1 ml ofreference solution (a) to
Calculate the content ofClsHzoN303SNa.
100 ml with solvent mixture.
Storage. Store protected from light and moisture.
Chromatographic system as described under Assay.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates. Ramipril
Inject the test solution and reference solution (b). Run the
chromatogram three times of the principal peak. In the
chromatogram obtained with the test solution, the area of any
secondary peak is not more than twice the area of the peak in
the chromatogram obtained with reference solution (b)
(2.0 per cent) and the sum of areas of all the secondary peaks
is not more than 6 times the area ofthe peak in the chromato- CZ3H3ZNzOs Mol. Wt. 416.5
gram obtained with the reference solution (b) (6.0 per cent).
Ramipril is (2S,3aS,6aS)-1-[(S)-2-[[(S)-I-(ethoxycarbonyl)-
Uniformity of content (For tablets containing 10 mg or less). 3-phenylpropyl]amino]propanoyl]
Comply with the tests stated under Tablets. octahydrocyclopenta[b]pyrrole-2-carboxylic acid.
Disperse 1 tablet in sufficient 0.1 M sodium hydroxide to Ramipril contains not less than 98.0 per cent and not more
produce 0.0015 per cent w/v solution. Measure the absorbance than 101.0 per cent ofCz3H32NzOs. calculated on the dried basis.
ofthe resulting solution at the maximum at about 292 nm (2.4.7). Category. Antihypertensive.
Calculate the content ofCIsHzoN303SNafrom the absorbance
obtained from same concentration of rabeprazole sodium RS Description. A white or almost white, crystalline powder.
in the same medium. Identification
Other tests. Comply with the tests stated under Tablets. Determine by infrared absorption spectrophotometry (2.4.6).
Assay. Determine by liquid chromatography (2.4.14). Compare the spectrum with that obtained with ramipril RS.

2038

www.webofpharma.com
IP 2010 RAMIPRIL CAPSULES

Tests (0.5 per cent) and the sum of areas of all the secondary peaks
is not more than the area of the peak in the chromatogram
Appearance ofsolution. A 1.0 per cent w/v solution in methanol obtained with the reference solution (b) (1.0 per cent).
is clear (2.4.1) and colourless (2.4.1).
Sulphated ash (2.3.18). Not more than O.1percent.
Specific optical rotation (2.4.22). + 32.0° to + 38.0°, detennined
in 1.0 per cent w/v solution in 0.1 M methanolic hydrochloric Loss on drying (2.4.19). Not more than 0.2 per cent, determined
acid. on 1.0 g by drying in an oven at 60°, under vacuum, for
4 hours.
Related substances. Detennine by liquid chromatography
(2.4.14). Assay. Weigh accurately about 0.3 gm, dissolve in 25 ml of
methanol and add 25 ml of water. Titrate with 0.1 M sodium
Test solution. Dissolve 25 mg of the substance under hydroxide. Detennine the end-point potentiometrically (2.4.25).
examination in 25 ml ofmobile phase B. Carry out a blank titration.
Reference solution (a). A 0.1 per cent w/v solution of ramipril 1 ml of 0.1 M sodium hydroxide is equivalent to 0.04165 gm of
RS in the mobile phase B.
CZ3H32NzOs.
Reference solution (b). Dilute 1 ml ofreference solution (a) to
Storage. Store protected from light.
100 ml with mobile phase B.
Chromatographic system
- a stainless steel column 25 cm x 4.0 mm packed with
octadecylsilane bonded to porous silica (5 /lm),
Ramipril Capsules
- column temperature. 65°, Ramipril Capsules contain not less than 90.0 per cent and not
- mobile phase: A. dissolve 2.0 g of sodium perchlorate more than 110.0 per cent of the stated amount of ramipnl,
in a mixture of 0.5 ml of triethylamine and 800 ml of CZ3H32NzOs·
water; adjust pH to 3.6 with orthophosphoric acid and
Usual strengths. 1.25 mg; 2.5 mg; 5 mg; 10 mg.
add 200 ml of acetonitrile,
B. dissolve 2.0 g of sodium perchlorate Identification
in a mixture of 0.5 ml of triethylamine and 300 ml of
water; adjust pH to 2.6 with orthophosphoric acid and Shake a quantity of the content of the capsules containing
add 700 ml of acetonitrile, 25 mg of Ramipril with 50 ml of acetone, centrifuge for
- a linear gradient programme using the conditions given 10 minutes, filter. Evaporate the filtrate to dryness at 60° for
below, 3 hours. The residue complies with the following test.
flow rate. 1 ml per minute, Detennine by infrared absorption spectrophotometry (2.4.6).
spectrophotometer set at 210 nm, Compare the spectrum with that obtained with ramipril RS.
- injection volume. 10 /ll.
Tests
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent v/v) Dissolution (2.5.2).
o 90 ,10 Apparatus No.1,
6 90 10 Medium. 500 ml of 0.1 M hydrochloric acid,
7 75 25 Speed and time. 75 rpm and 45 minutes.
20 65 35 Withdraw a suitable volume ofthe medium and filter.
30 25 75 Detennine by liquid chromatography (2.4.14).
40 25 75 Test solution. Dilute the filtrate to get 0.00025 per cent w/v
45 90 10 solution of Ramipril with 0.1 M hydrochloric acid.
55 90 10 Reference solution. A 0.00025 per cent w/v solution oframipril
Inject reference solution (a). The test is not valid unless the RS in 0.1 M hydrochloric acid.
tailing factor is not more than 2.0 and the column efficiency in Chromatographic system as described under Assay.
not less than 2000 theoretical plates.
Inject the test solution and the reference solution.
Inject the test solution and reference solution (b). In the
chromatogram obtained with the te,st solution, the area of any Calculate'the content of CZ3H32NzOs.
secondary peak is not more than 0.5 times the area ofthe peak D. Not less than 70 per cent of the stated amount of
in the chromatogram obtained with reference solution (b) Cz3H32NZOS' '

2039

www.webofpharma.com
RAMIPRlL CAPSULES IP 2010

Uniformity of content (For capsules containing 10 mg or Evaporate the filtrate to dryness at 60° for 3 hours. The residue
less). Comply with the test stated under Capsules. complies with the following test.
Determine by liquid chromatography (2.4.14). Determine by infrared absorption spectrophotometry (2.4.6).
Test solution. Disperse one capsule in 100 ml of 0.1 M hydro- Compare the spectrum with that obtained with ramipril RS.
chloric acid, sonicate for 15 minutes. Dilute if necessary, to
Tests
produce 0.00025 per cent w/v solution of Ramipril in 0.1 M
hydrochloric acid. Dissolution (2.5.2).
Reference solution. A 0.00025 per cent w/v solution of Apparatus No.1,
ramipril RS in 0.1 M hydrochloric acid. Medium. 500 ml of 0.1 M hydrochloric acid,
Chromatographic system as described under Assay. Speed and time. 75 rpm and 45 minutes.
Inject the test solution and the reference solution. Withdraw a suitable volume ofthe medium and filter.
Calculate the content ofCz3H3zNzOs. Determine by liquid chromatography (2.4.14).
Assay. Determine by liquid chromatography (2.4.14). Test solution. Dilute the filtrate to get 0.00025 per cent w/v
Test solution. Weigh a quantity of the content of capsules solution of Ramipril with 0.1 M hydrochloric acid.
containing 25 mg of Ramipril, disperse in 100.0 ml of Reference solution. A 0.00025 per cent wIv solution of ramipril
0.1 M hydrochloric acid, mix and centrifuge. Dilute 1.0 ml of RS in 0.1 M hydrochloric acid.
the resulting solution to 100.0 ml with 0.1 M hydrochloric
Chromatographic system as described under Assay.
acid.
Inject the test solution and the reference solution.
Reference solution. A 0.00025 per cent w/v solution of ramipril
RS in 0.1 M hydrochloric acid. Calculate the content ofCz3H32NzOs.
Chromatographic system D. Not less than 70 per cent of the stated amount of
- a stainless steel column 12.5 cm X 4.6 mm packed with CZ3H3ZNzOs.
octadecylsilane bonded to porous silica (5 /lm), Uniformity of content (For tablets containing 10 mg or less).
- mobile phase: a mixture of 42 volumes of acetonitrile Comply with the test stated under Tablets.
and 58 volumes of a solution containing 1.4 per cent
Determine by liquid chromatography (2.4.14), as described
w/v solution of sodium perchlorate and 0.58 per cent
under Assay.
w/v solution of orthophosphoric acid adjusted to
pH 2.5 with triethylamine, adjust the pH ofthe mixture Test solution. Take one tablet, add 5 ml of 0.1 M hydrochloric
to 2.1 with orthophosphoric acid, acid, sonicate for 10 minutes, dilute, if necessary, with
- flow rate. 1 ml per minute, sufficient 0.1 M hydrochloric acid to produce a solution
- spectrophotometer set at 210 nm, containing 0.00025 per cent w/v of Ramipril, centrifuge and
- injection volume. 50 Ill. use the supernatant liquid.
Inject the reference solution. The test is not valid unless the Calculate the content ofCz3H32NzOs.
relative standard deviation for replicate injections is not more Assay. Determine by liquid chromatography (2.4.14).
than 2.0 per cent.
Test solution. Weigh and powder 20 tablets. Weigh a quantity
Inject the test solution and the reference solution. ofpowdered tablets containing 25 mg ofRamipril, disperse in
Calculate the content of CZ3H32NzOs. 100.0 ml of 0.1 M hydrochloric acid and centrifuge. Dilute 1.0
ml ofthe resulting solution to 100.0 ml with 0.1 Mhydrochloric
acid.
.Ramipril Tablets Reference solution. A 0.00025 per cent wIv solution of ramipril
RS in 0.1 M hydrochloric acid.
Ramipril Tablets contain not less than 90.0 per cent and not
Chromatographic system
more than 110.0 per cent of the stated amount of ramipril,
- a stainless steel column 12.5 cm X 4.6 mm packed with
Cz3H32NZOS'
octadecylsilane bonded to porous silica (5/lm),
Usual strengths. 1.25 mg; 2.5 mg; 5 lUg; 10 mg. mobile phase: a mixture of 42 volumys of acetonitrile
and 58 volumes of a solution containing 1.4 per cent
Identification
w/v solution of sodium perchlorate and 0.58 per cent
Shake a quantity ofthe powdered tablets containing 25 mg of w/v solution of orthophosphoric acid adjusted to pH
. Ramipril with 50 ml of acetone, centrifuge for 10 minutes, filter. 2.5 with triethylamine, adjust the pH of the mixture to

2040

www.webofpharma.com
IP 2010 RAMIPRIL AND HYDROCHLOROTHIAZIDE TABLETS

2.1 with orthophosphoric acid, - mobile phase: a mixture of 55 volumes of water, 45


- flow rate. 1 ml per minute, volumes of acetonitrile, and 0.1 volume of
- spectrophotometer set at 210 nm, triethylamine, adjusted to pH 3.0 with orthophoshoric
- injection volume. 50 Ill. acid,
- flow rate. 0.7 ml per minute,
Inject the reference solution. The test is not valid unless the
- spectrophotometer set at 210 nm,
relative standard deviation for replicate injections is not more
- injection volume. 20 Ill.
than 2.0 per cent.
Inject reference solution (a) and (b). The relative standard
Inject the test solution and the reference solution.
deviation for replicate injections for each peak is not more
Calculate the content of CZ3H32NzOs. than 2.0 per cent.
Storage. Store protected from moisture. Inject the test solution, reference solution (a) and (b).
Calculate the content ofCz3H32NzOs and C7HgClN304Sz.
D. Not less than 75 per cent ofthe stated amount ofCz3H32NzOs
Ramipril and Hydrochlorothiazide and C7HgClN304SZ.
Tablets Related substances. Determine by liquid chromatography
Ramipril and Hydrochlorothiazide Tablets contain not less (2.4.14).
than 90.0 per cent and not more than 110.0 per cent of the Test solution. Disperse a quantity of powdered tablets
stated amounts of ramipril, CZ3H32NzOs and containing about 10 mg ofRamipril in mobile phase A, sonicate
hydrochlorothiazide, C7HgClN304SZ. for 15 minutes and dilute to 10.0 ml with the same solvent.
Usual strength. Ramipril 2.5 mg and Hydrochlorothiazide Reference solution (a). A solution containing 0.2 per cent
12.5 mg. w/v of ramipril RS and 1.0 per cent w/v of hydrochlorothiazide
RS in mobile phase A.
Identification
Reference solution (b). Dilute 5.0 ml ofreference solution (a)
In the Assay, the principal peaks in the chromatogram obtained to 50.0 rn1 with mobile phase A. Dilute 5.0 rol ofthis solution to
with the test solution corresponds to the principal peaks in 100.0 ml with the same solvent.
the chromatogram obtained with reference solution (b).
Chromatographic system
Tests - a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm),
Dissolution (2.5.2). column temperature. 35°,
Apparatus No.1, mobile phase: A. a mixture of 60 volumes of buffer
Medium.750 ml of 0.1 M hydrochloric acid, solution prepared by ·dissolving 4 g of sodium
Speed and time. 100 rpm and 45 minutes. perchlorate in 600 ml of water, add 1.0 rol of
Withdraw a suitable volume ofthe medium and filter. triethylamine, adjusted to pH 2.6 with orthophosphoric
acid and 40 volumes of acetonitrile,
Determine by liquid chromatography (2.4.14). B. a mixture of 85 volumes of buffer
Test solution. Dilute the filtrate, if necessary, with the solution and 15 volumes of acetonitrile,
dissolution medium. a linear gradient programme using the conditions given
Reference solution (a). Dissolve a quantity of ramipril RS in below,
the mobile phase and dilute with dissolution medium to obtain flow rate. 1 ml per minute,
a solution having a known concentration similar to the test spectrophotometer set at 210 nm,
solution. injection volume. 20 Ill.
Time Mobile phase A Mobile phase B
Reference solution (b). Dissolve a quantity of
(in min) (per cent v/v) (per cent v/v)
hydrochlorothiazide RS in mobile phase and dilute with
dissolution medium to obtain a solution having a known 0-13 0 100
concentration similar to the test solution. 13 - 17 0 ~50 100 ~50
17-20 50~100 50 ~O
Chromatographic. system
- a stainless steel column 25 cm x 4.6 mm, packed with 20 - 80 100 o
octadecylsilane bonded to porous silica (5 Ilm) (Such 80 - 82 100 ~ o ~100
as Thermo quest Hypersil), 82 -87 0 100

2041

www.webofpharma.com
RAMIPRIL AND HYDROCHLOROTHIAZIDE TABLETS IP 2010

Inject reference solution (b). The test is not valid unless the disperse in water and dilute to 250 ml with the mobile phase,
relative standard deviation for replicate injections for each filter.
peak is not more than 5.0 per cent. The relative retention time
Reference solution. A 0.01 per cent w/v solution of ramipril
with reference to ramipril forramipril impurity A is about 0.9,
RS in the mobile phase.
for ramipril impurity B is about 1.2, for ramipril impurity C is
about 1.5 and for ramipril impurity D is about 1.7. ChromatographiC system
a stainless steel column 25 cm x 4.6 mm, packed with
Inject the test solution and reference solution (b). The retention
octadecylsilane bonded to porous silica (5 j.tm), (Such
time oframipril is about 30 minutes and ofhydrochlorothiazide
as Thermo quest Hypersil),
is about 10 minutes. In the chromatogram obtained with the
mobile phase: a mixture of 55 volumes of water, 45
test solution the area of each peak due to ramipril impurity A,
volumes of acetonitrile and 0.1 volume of triethylamine,
Band C is not be more than the area of the principal peak in
flow rate. 1 ml per minute,
the chromatogram obtained with reference solution (b) (1.0
spectrophotometer set at 210 nm,
per cent); the area of peak due to ramipril impurity D is not
injection volume. 10 j.tl.
more than 7 times the area· of the principal peak in the
chromatogram obtained with reference solution (b) (7.0 per Inject the reference solution. The test is not valid unless the
cent); the area of any other secondary peak is not more than theoretical plates of the principal peak is not less than 2000
the area of the principal peak in the chromatogram obtained theoretical plates and the tailing factor is not more than 2.0.
with reference solution (b) (1.0 per cent). The relative standard deviation for replicate injections is not
more than 2.0 per cent.
Uniformity of content. Comply with the test stated under
Tablets. Inject the test solution and the reference solution.
For ramipril- Determine by liquid chromatography (2.4.14). Calculate the content of C23H32N20S in the tablet.
Test solution. Disperse 1 intact tablet in water and dilute to 50 For hydrochlorothiazide-
ml with the mobile phase. Test solution. Disperse a quantity of the powdered tablets
Reference solution. Dissolve a quantity of ramipril RS in the containing about 125 mg ofHydrochlorothiazide with 50 ml of
mobile phase and dilute with the mobile phase to obtain a acetonitrile, sonicate for 15 minutes and dilute to 250 ml with
solution having a known concentration similar to the test the mobile phase. Dilute 5.0 ml ofthis solution to 20 ml with
solution. the mobile phase.
Use chromatographic system as described under Assay. Reference solution. Dissolve 12.5 mg of hydrochlorothiazide
Inject the test solution and the reference solution. RS with 10 ml of acetonitrile, sonicate and dilute to 100 ml
with the mobile phase.
Calculate the content of C23H32N20S in the tablet.
Chromatographic system
For hydrochlorothiazide-Determine by liquid a stainless steel column 10 cm x 4.6 mm, packed with
chromatography (2.4.14). octadecylsilane bonded to porous silica (5 j.tm),
Test solution. Disperse 1 tablet with sufficient amount ofthe mobile phase: a mixture of 9 volumes of 0.1 M sodium
mobile phase, sonicate for 15 minutes and dilute to 100 ml with dihydrogen orthophosphate and 1 volume of
the mobile phase. acetonitrile, adjusted to pH 3.0 with orthophosphoric
Reference solution. Dissolve a quantity of acid,
hydrochlorothiazide RS in the mobile phase and dilute with flow rate. I ml per minute,
the mobile phase to obtain a solution having a known spectrophotometer set at 254 nm,
concentration similar to the test solution. injection volume. 10 j.tl.

Use chromatographic system as described under Assay. Inject the reference solution. The test is not valid unless the
theoretical plates ofhydrochlorothiazide peak is not less than
Inject the test solution and the reference solution. 2000 and the tailing factor is not more than 2.0. The relative
Calculate the content of C7HsClN304S2 in the tablet. standard deviation for replicate injections is not more than 2.0
Other tests. Comply with the tests stated under Tablets. per cent.

Assay. For ramipril-Determine by liquid chromatography Inject the test solution and the reference solution.
(2.4.14). Calculate the content ofC7HsCIN304S2 in thetilblet.
Test solution. Weigh and powder 20 tablets: Weigh accurately Storage. Store protected from moisture, at. a temperature not
a quantity of powder containing about 25 mg of Ramipril, exceeding 30°.

2042

www.webofpharma.com
IP 2010 RANITIDINE HYDROCHLORIDE

Labelling. The label states the strength in terms of the Test solution (a). Dissolve 0.22 g of the substance under
equivalent amount oframipril and hydrochlorothiazide. examination in 10 ml of methanol.
Test solution (b). Dilute 1 ml oftest solution (a) to 100 ml with
methanol.
Ranitidine Hydrochloride Reference solution (a). Weigh accurately a quantity of
ranitidine hydrochloride RS in methanol, and dilute with
H methanol to obtain a solution containing a known
~°V"S~N~CHN02 concentration of about 0.022 per cent w/v.
H3C--N U· I , HCI
Reference solution (b). Dilute 10.0 ml ofreference solution (a)
CH 3
NHCH 3
to 20 ml with methanol.
C13H22N403S,HCl Mol. Wt. 350.9 Reference solution (c). Dilute 30.0 ml ofreference solution (a)
Ranitidine Hydrochloride is N-[2-[[[ 5-[(dimethylamino) to 100 ml with methanol.
methyl]furan-2-yl]methyl]thio]ethyl]-N-methyl-2- Reference solution (d). Dilute 5.0 ml of reference solution (a)
nitroethene-l, 1-diamine hydrochloride. to 100 ml with methanol.
Ranitidine Hydrochloride contains not less than 97.5 per cent Reference solution (e). Weigh accurately a quantity of (N,N'-
and not more than 102.0 per cent of C13H22N403S, HCl, bis[2-[[[5-(dimethylamino) methyljfilran-2-yl]methylj
calculated on the dried basis. sulphanyl]ethyl]-2-nitroethene-l, l-diamine RS (ranitidine
impurity A RS) in methanol, and dilute with methanol to obtain
Category. Antiulcer (Histamine H2-receptor antagonist).
a solution containing a known concentration of about 0.127
Dose. Orally, the equivalent of 300 to 600 mg of ranitidine per cent w/v.
daily, in divided doses; by intramuscular or slow intravenous
Reference solution (f). Weigh accurately a quantity of 2-[[[5-
injection, the equivalent of 50 mg of ranitidine every 6 to 8
[(dimethyl am ino) me thyl]furan-2-yl]methyl]
hours. (1.12 g of ranitidine hydrochloride is approximately
sulphanyljethanamine RS (ranitidine impurity B RS) in
equivalent to 1 g ofranitidine).
methanol, and dilute with methanol to obtain a solution
Description. A white to pale yellow, crystalline powder. containing a known concentration of about 0.1 per cent w/v.

Identification Apply to the plate 10 III of each solution except reference


solution (e). Apply separately an additional 10 III of the test
A. Determine by infrared absorption spectrophotometry (2.4.6). solution and on top'ofthis application, apply 10 III ofreference
Compare the spectrum with that obtained with ranitidine solution (e). After development, dry the plate in air and expose
hydrochloride RS or with the reference spectrum ofranitidine it to iodine vapours in a closed chamber until the spots are
hydrochloride. revealed. Any spot in the chromatogram obtained with the
.B. In the test for Related substances, the principal spot in the test solution (a) corresponding to the principal spot in the
chromatogram obtained with test solution (b) corresponds to chromatogram obtained with reference solution (f) is not more
that in the chromatogram obtained with reference solution (a). intense than that of the principal spot in the chromatogram
obtained with reference solution (b) (0.5 per cent) and no
C. A 5 per cent w/v solution gives the reactions of chlorides other spot in the chromatogram obtained with the test solution
(2.3.1). is more intense than the principal spot in the chromatogram
obtained with reference solution (c) (0.3 per cent). The sum of
Tests
the intensities of all the secondary spots in the chromatogram
Appearance of solution. A 1.0 per cent w/v solution is clear obtained with the test solution does not exceed 1.0 per cent.
(2.4.1), and not more intensely coloured than reference solution The test is not valid unless the chromatogram obtained with
BYS5 (2.4.1). the combined test solution and reference solution (e) shows
pH (2.4.24). 4.5 to 6.0, determined in a 1.0 per cent w/v solution two clearly separated principal spots and the chromatogram
in carbon dioxide-free water. . obtained with reference solution (d) shows a clearly visible
Related substances. Determine by thin-layer chromatography spot.
(2.4.17), coating the plate with silica gel GF254. Sulphated asb(2.3 .18). Not more than 0.1 per cent.
Mobile phase. A mixture of 25 volumes of ethyl acetate, Loss on drying (2.4.19). Not more than 0.75 percent,
15 volumes of 2-propanol, 4 volumes of strong ammonia determined on 1.0 g by drying in an oven at 60° at a pressure
solution and 2 volumes of water. not exceeding 2.75 kPa for 3 hours.

2043

www.webofpharma.com
RANITIDINE INJECTION IP 2010

Assay. Determine by liquid chromatography (2.4.14) B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the
Test solution. A 0.01l2 per cent w/v of the substance under
chromatogram obtained with the reference solution.
examination in the mobile phase.
Reference solution. 0.0112 per cent w/v of ranitidine Tests
hydrochloride RS in the mobile phase.
pH (2.4.24).6.7 to 7.3, ifthe preparation is buffered; 4.5 to 7.0,
Chromatographic system
ifthe preparation is unbuffered.
- a stainless steel column 25 cm x 4.0 rom, packed with
octadecylsilane bonded to porous silica (5 )lm), Related substances. Determine by thin-layer chromatography
- mobile phase: a mixture of85 volumes of methanol and (2.4.17), coating the plate with silica gel GF254.
15 volumes of 0.1 M ammonium acetate,
Mobile phase. A mixture of 25 volumes of ethyl acetate,
- flow rate. 2 ml per minute,
15 volumes 'of 2-propanol, 4 volumes of strong ammonia
-spectrophotometer set at 322 nm,
solution and 2 volumes of water.
- injection volume. 20 )ll.
Test solution. Dilute suitably a volume of the injection with
Inject the reference solution. The test is not valid unless the
water to produce a solution containing the equivalent of
relative standard deviation for replicate injections is not more
2.5 per cent w/v ofranitidine in water.
than 2.0 per cent.
Reference solution (a). Weigh accurately a quantity of
Inject the test solution and the reference solution.
ranitidine hydrochloride RS in water, and dilute with water
Calculate the content ofCI3H22N403S, HCI. to obtain a solution containing a known concentration of
about 0.056 per cent w/v.
Storage. Store protected from light and moisture.
Reference solution (b). Dilute 10.0 ml ofreference solution (a)
to 20 ml with water.
Reference solution (c). Dilute 5.0 ml ofreference solution (a)
Ranitidine Injection to 20 ml with water.

Ranitidine Hydrochloride Injection Reference solution (d). Dilute 6.0 ml ofreference solution (c)
to 10 ml with water.
Ranitidine Injection is a sterile solution of Ranitidine
Hydrochloride in Water for Injections and may be suitably Reference solution (e). Dilute 5.0 ml ofreference solution (b)
buffered. to 50 ml with water.

Ranitidine Hydrochloride Injection contains not less than Reference solution (f). Dilute 5.0 ml of reference solution (e)
90.0 per cent and not more than 110.0 per cent of the stated to 10 ml with water.
amount ofranitidine, C13H22N403S. Reference solution (g). A 0.127 per cent w/v of solution of
Usual strength. The equivalent of50 mg ofranitidine in 2 ml (N,N'-bis[2-[[[5-[(dimethylamino)methyl}furan-2-
(1.12 g ofranitidine hydrochloride is approximately equivalent yl]methyl]sulphanyl] ethyl]-2-nitroethene-1, 1-diamine RS
to 1 g ofranitidine). (ranitidine impurity A RS) in methanol.
Apply to the plate 10 )ll of each solution. Apply separately an
Identification additional 10 )ll of the test solution and on top of this
application, apply 10 )ll of reference solution (g). After
A. To a volume ofthe injection containing 25 mg ofranitidine
development, dry the plate in air and expose it to iodine vapours
add 20 ml of methanol, mix and evaporate to dryness. Add
in a closed chamber until the spots are revealed. The major
1 ml of light petroleum (6(J' to 80°) to the resulting residue,
secondary spot in the chromatogram obtained with the test
scratch the side of the vessel with a glass rod to induce
solution is not more intense than that of the principal spot in
crystallisation, evaporate to dryness and dry the residue at
the chromatogram obtained with reference solution (a)
60° for 10 minutes.The residue complies with the following
(2.0 per cent) and no other secondary spot in the chromatogram
test.
obtained with the test solution is more intense than the
Determine by infrared absorption spectrophotometry (2.4.6). principal spot in the chromatogram obtained with reference
Compare the spectrum with that obtained with ranitidine solution (b) (1.0 per cent). The sum ofthe intensities of all the
hydrochloride RS or with the reference spectrum ofranitidine . secondary spots in the chromatogram obtained with the test
hydrochloride. solution does not exceed 5.0 per cent.

2044

www.webofpharma.com
IP 2010 RANlTIDINE TABLETS

The test is not valid unless the chromatogram obtained with dryness and dry the residue at 60° for 10 minutes. The residue
the combined test solution and reference solution (g) shows complies with the following test.
two clearly separated principal spots and the chromatogram
Determine by infrared absorption spectrophotometry (2.4.6).
obtained with reference solution (f) shows a clearly visible
Compare the spectrum with that obtained with ranitidine
spot.
hydrochloride RS or with the reference spectrum ofranitidine
Other tests. Comply with the tests stated under Parenteral hydrochloride.
Preparations (Injections).
B. In the Assay, the principal peak in the chromatogram
Assay. Determine by liquid chromatography (2.4.14) obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution.
Test solution. Dilute a volume of the injection containing
10.0 mg ofranitidine to 100.0 ml with the mobile phase.
Tests
Reference solution. A 0.0112 per cent w/v solution of
ranitidine hydrochloride RS in the mobile phase. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Chromatographic system
- a stainless steel column 25 cm x 4.0 rom packed with Mobile phase. A mixture of 25 volumes of ethyl acetate,
15 volumes of 2-propanol, 4 volumes of strong ammonia
octadecylsilane bonded to porous silica (5 fJ.m),
- mobile phase: a mixture of85 volumes of methanol and solution and 2 volumes of water.
15 volumes of 0.1 M ammonium acetate, Test solution. Shake a quantity of the powdered tablets
- flow rate. 2 ml per minute, containing 0.45 g of ranitidine with 20 ml of methanol and
- spectrophotometer set at 322 nm, filter.
- injection volume. 20 fJ.1.
Reference solution (a). Weigh accurately a quantity of
Inject the reference solution. The test is not valid unless the ranitidine hydrochloride RS in methanol, and dilute with
relative standard deviation for replicate injections is not more methanol to obtain a solution containing a known
than 2.0 per cent. concentration of about 0.022 per cent w/v.
Inject the test solution and the reference solution. Reference solution (b). Dilute 10 ml of reference solution (a)
to 20 ml with methanol.
Calculate the content ofCI3H22N403S in the injection.
Storage. Store protected from light. Reference solution (c). Dilute 30 ml of reference solution (a)
to 100 ml with methanol.
Labelling. The label states (1) the strength in terms of the
equivalent amount of ranitidine; (2) where appropriate, that Reference solution (d). Dilute 5 ml ofreference solution (a) to
the injection is buffered. 50 ml with methanol.
Reference solution (e). Dilute 5 ml ofreference solution (a) to
100 ml with methanol.

Ranitidine Tablets Reference solution (f). Weigh accurately a quantity of (N,N'-


bis[2-[[[5-[(dimethylamino)methyl]furan-2-yl]methyl]
Ranitidine Hydrochloride Tablets sulphanyl]ethyl]-2-nitroethene-1,1-diamine RS (ranitidine
Ranitidine Tablets contain not less than 90.0 per cent and not impurity A RS) in methanol, and dilute with methanol to obtain
more than 110.0 per cent ofthe stated amount ofthe ranitidine, a solution containing a lmown concentration of about 0.127
CI3H22N403S, The tablets are coated. per cent w/v.

Usual strengths. The equivalent of 150 mg, 300 mg ofranitidine Apply to the plate 10 fJ.l of each solution. Apply separately an
(1.12 g ofranitidine hydrochloride is approximately equivalent additional 10 fJ.l of the test solution and on top of this
to 1 g ofranitidine). application, apply 10 fJ.l of reference solution (f). After
development, dry the plate in air and expose it to iodine vapours
Identification in a closed chamber until the spots are revealed. Any spot in
the chromatogram obtained with the test solution
A. Shake a quantity ofthe powdered tablets containing 25 mg corresponding to the principal spot in the chromatogram
of ranitidine with 5 ml of methanol for 5 minutes, filter and obtained with reference solution (f) is not more intense than
evaporate the filtrate to dryness. Add 1 ml of light petroleum that of the principal spot in the chromatogram obtained with
(60° to 80°) to the resulting residue, scratch the side of the reference solution (b) (0.5 per cent) and no other spot in the
vessel with a glass rod to induce crystallisation, evaporate to chromatogram obtained with the test solution is more intense

2045

www.webofpharma.com
RANITIDINE TABLETS IP 2010

than the principal spot in the chromatogram obtained with Identification


reference solution (c) (0.3 per cent). The sum bfthe intensities
of all the secondary spots in the chromatogram obtained with A. When examined under a microscope in the dry state, or
the test solution does not exceed 2.0 per cent. when mounted in ethanol (95 per cent) and water, the
following characteristics are observed. They are usually of
The test is not valid unless the chromatogram obtained with more or less uniform width. Many longitudinal parallel lines
the combined test solution and reference solution (f) shows are distributed unequally over the width in the case ofstandard
two clearly separated principal spots and the chromatogram viscose fibres, but such lines are absent or very few in fibres
obtained with reference solution (e) shows a clearly visible produced through the zinc-free process. The ends are cut
spot. more or less straight. Matt fibres contain numerous granular
Other tests. Comply with the tests stated under Tablets. particles ofapproximately 1Jl average diameter.

Assay. Determine by liquid chromatography (2.4.14) B. Treat with iodinated zinc chloride solution; the fibres
become violet.
Test solution. Weigh and powder 20 tablets. Shake 1.5 g bfthe
powder with 400 ml ofthe mobile phase, dilute to 500.0 ml with C. To 0.1 g add 10 ml ofzinc chloride-jormic acid solution,
the mobile phase, filter and dilute the filtrate with the mobile heat to 40° and allow to stand for 2 hours 30 minutes, shaking
phase to obtain a solution containing the equivalent of occasionally. The .fibres dissolve completely except for the
0.01 per cent w/v ofranitidine. matt variety where titanium dioxide particles remain.
Reference solution. A 0.0112 per cent w/v solution of D. Dissolve the residue obtained in the test for Sulphated ash
ranitidine hydrochloride RS in the mobile phase. in 5 ml of sulphuric acid with slight warming, allow to cool,
and carefully add 0.2 ml of hydrogen peroxide solution
Chromatographic system
(10 volumes). The solution does not undergo colour change
- a stainless steel column 25 cm x 4.0 mm, packed with
in case of lustrous variety of fibre, but for matt variety an
octadecylsilane bonded to porous silica (5 Jlm),
orange-yellow colour is obtained, the intensity of which
mobile phase: a mixture of85 volumes of methanol and
depends on the quantity of titanium dioxide present.
15 volumes of 0.1 M ammonium acetate,
flow rate. 2 ml per minute,
- spectrophotometer set at 322 nm,
Tests
- injection volume. 20 Jll. Colour ofextract. Take 15 g ofmaterial under examination in a
Inject the reference solution. The test is not valid unless the suitable vessel, add 150 ml of water, close the vessel and
relative standard deviation for replicate injections is not more allow to macerate for 2 hours. Decant the solution, squeeze
than 2.0 per cent. the residual liquid carefully from the sample with a glass rod,
mix and filter. The filtered extract is colourless. Compare the
Inject alternately the test solution and the reference solution. colour of the extract with water using identical tubes of
Calculate the content ofC13H22N403S in the tablets. colourless, transparent, neutral glass 12 mm in diameter
measuring 2 ml. Compare the colours in diffused daylight,
Storage. Store protected from light and moisture.
viewing horizontally against a white background.
Labelling. The label states the strength in terms of the
Acidity or alkalinity. To 25 ml offiltered extract obtained, add
equivalent amount ofranitidine.
0.1 ml of dilute phenolphthalein solution; to another 25 ml
add 0.05 ml of methyl orange solution. Neither solution shows
a pink colour.
Foreign fibres. When examined under a microscope, it is seen
Purified Rayon to consist exclusively of viscose fibres, except that
Viscose Fibre; absorbent viscose occasionally a few isolated foreign fibres may be present.
Fluorescence. Examine a layer about 5 mm in thickness under
Category. Regenerated cellulose used in surgical dressings.
ultraviolet light at 365 nm. It displays only a slight, brownish-
Description. White or very slightly yellow, purified rayon is a violet fluorescence and a few yellow particles. Not more than
fibrous form ofbleached regenerated cellulose, which can be a few isolated fibres show an intense blue fluorescence.
produced with a lustrous or matt appearance, and is soft to
Absorbancy
the touch. The fibres can be produced with average
staple length between 32 mm to 80 mm, and are practically A. Sinking time. Not more than 10 seconds, determined by
odourless. the following method.

2046

www.webofpharma.com
IP20lO RESERPINE

Apparatus solution prepared at the same time using 0.15 ml of dilute


acetic acid, 1.2 m1 of thioacetamide reagent, 1.7 ml of lead
A dry, cylindrical copper wire basket, 80 mrn high and 50 mrn in
standard solution (10 ppm Pb) and 10 ml of filtered extract.
diameter, fabricated from wire ofdiameter 004 mrn and having a
mesh aperture of 15 to 20 mm; the basketweighs 204 to 3.0 g. Sulphated ash (2.3.18). Not more than 1.5 per cent.

Method Loss on drying (204.19). Not more than 13.0 per cent, determined
on 5 g by drying in an oven at 105°.
Weigh the basket to the nearest 10 mg. Take five samples,
each ofapproximately 1g, from different places in the material
under examination, place loosely in the basket and weigh the
packed basket to the nearest 10 mg. Hold the basket with its
long axis in the horizontal position and drop it from a height of Reserpine
about 10 mm into water at 25° contained in a beaker at least
12 cm in diameter and filled to a depth of 10 cm. Measure with
a stopwatch the time taken by the basket to sink below the
surface of the water. Repeat the procedure on two further
samples and calculate the average value.
B. Water-holding capacity. Not less than 18.0 g per g,
determined by the following method.
After the sinking time has been recorded in test A, remove the
basket from the water, allow it to drain for 30 seconds with its
long axis in the horizontal position over the beaker, transfer it
to a tared beaker and weigh to the nearest 10 mg. Calculate the
weight of water retained bythe sample. Repeat the procedure C33H4oN209 Mol. Wt. 608.7
on two further samples and calculate the average value.
Reserpine is methyl11,17a.-dimethoxy-18~-[(3,4,5-tri­
Colouring matter. Slowly extract 109 in a narrow percolator methoxybenzoyl)oxy]-3~,20a.-yohimbane-16~-carboxy1ate.
with ethanol (95 per cent) until 50 ml of extract is obtained.
Reserpine contains not less than 99.0 per cent and not more
The extract is not more intensely coloured than reference
than 101.0 per cent oftotal alkaloids and not less than 98.0 per
solution YS5 or GYS6, (204.1) or a solution prepared in the
cent and not more than 102.0 per cent ofreserpine, C33H4oN209'
following manner. To 3.0 ml ofCSS add 7.0 ml ofa solution of
both calculated on the dried basis.
hydrochloric acid containing 1 per cent w/v of hydrochloric
acid and dilute 0.5 ml of the resulting solution to 10 ml with Category. Antihypertensive.
the same solution of hydrochloric acid. Dose. 500 flg daily.
Ether-soluble substances. Not more than 0.5 per cent, Description. White to slightly yellow small crystals or a
determined by the following method. Extract 5 g with ether in crystalline powder which darkens slowly on exposure to light.
a continuous extraction apparatus such as a Soxhlet apparatus,
for 4 hours in such a way that the rate is at least four extractions Identification
per hour. Evaporate the ether and dry the residue to constant
weight at 105°. Test A may be omitted if tests B, C, D and E are carried out.
TestsB, C, D and E may be omitted if test A is carried out.
Water-soluble substances. Not more than 0.7 per cent,
determined by the following method. BoilS g with 500 ml of A. Determine by infrared absorption spectrophotometry (204.6).
water for 30 minutes, stirring frequently and replacing the Compare the spectrum with that obtained with reserpine RS
water lost by evaporation. Decant the liquid into a beaker, or with the reference spectrum of reserpine.
squeeze the residual liquid from the material carefully with a
B. Dilute 1 ml of a 0.2 per cent w/v solution in chloroform to
glass rod, mix the liquids and filter the extract whilst hot. 100.0 m1 with ethanol (95 per cent). When examined
Evaporate 400 ml ofthe filtrate and dry the residue to constant immediately after preparation, in the range 230 nm to 360 nm
weight at 105°.
(204.7), the resulting solution shows an absorption maximum
Hydrogen sulphide. To 10 ml of the filtered extract obtained at about 268 nm; absorbance at about 268 nm, about 0.55.
in the Colour ofextract, add 1.9 ml ofwater,0.15 ml of dilute Over the range 288 nm to 295 nm, the spectrum exhibits a
acetic acid and 1 ml of lead acetate solution. After 2 minutes, slight minimum and then a shoulder or a slight maximum;
the solution is not more intensely coloured than a reference absorbance over this range, about 0.34.

2047

www.webofpharma.com
RESERPINE IP 2010

C. To about 1 mg add 0.1 ml of a 0.1 per cent w/v solution of Reserpine Injection
sodium molybdate in sulphuric acid; a yellow colour is
produced which changes to blue within 2 minutes. Reserpine Injection is a sterile solution of Reserpine in Water
for Injections prepared with the aid of a suitable acid. It may
D. To 1 mg add 0.2 ml of a freshly prepared 1 per cent w/v
contain suitable antioxidants.
solution of vanillin in hydrochloric acid; a pink colour
develops within 2 minutes. Reserpine Injection contains not less than 90.0 per cent and
not more than 110.0 per cent ofthe stated amount ofreserpine,
E. Mix about 0.5 mg with 5 mg of 4-dimethylaminobenzal-
C33~oNz09'
dehyde and 0.2 ml of glacial acetic acid and add 0.2 ml of
sulphuric acid; a green colour is produced. Add 1 ml of Usual strengths. 1 mg per ml; 2.5 mg per ml.
glacial acetic acid; the colour changes to red.
Identification
Tests
Extract a suitable volume ofthe injection containing 10 mg of
Specific optical rotation (2.4.22). -116° to -128°, determined Reserpine with 10 ml of chloroform and evaporate the
in a solution prepared immediately before use by dissolving chloroform layer to dryness. The residue complies with the
0.25 g in sufficient chloroform to produce 25 ml. following tests.
Oxidation products. Absorbance ofa 0.02 per cent wIv solution
Test A may be omitted if tests B, C, D and E are carried out.
in glacial acetic acid at about 388 nm, measured immediately
Tests B, C, D and E may be omitted if test A is carried out.
after preparation, not more than 0.10 (2.4.7).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Compare the spectrum with that obtained with reserpine RS
Loss on drying (2.4.19). Not more than 0.5 per cent, determined or with the reference spectrum of reserpine.
on 0.5 g by drying in an oven over phosphorus pentoxide at
60° at a pressure not exceeding 0.7 kPa for 3 hours. B. Dilute 1 ml of a 0.2 per cent w/v solution in chloroform to
100 ml with ethanol (95 per cent). When examined immediately
Assay. For total alkaloids - Weigh accurately about 0.5 g after preparation, in the range 230 nm to 360 nm (2.4.7), the
and dissolve in a mixture of40 ml of anhydrous glacial acetic resulting solution shows an absorption maximum at about
acid and 6 ml of acetic anhydride. Titrate with 0.1 Mperchloric 268 urn; absorbance at about 268 nin, about 0.55. Over the
acid, determining the end-point potentiometrically (2.4.25). range 288 urn to 295 urn, the spectrum exhibits a slight minimum
Carry out a blank titration. and then a shoulder or a slight maximum; absorbance over
1 ml of 0.1 M perchloric acid is equivalent to 0.06087 g of this range, about 0.34.
total alkaloids. C. To about 1 mg add 0.1 ml of a 0.1 per cent w/v solution of
For reserpine, C33H40N209 - Carry out the following sodium molybdate in sulphuric acid; a yellow colour is
procedure protected from light. Weigh accurately about produced which changes to blue within 2 minutes.
25.0 mg, moisten with 2 ml of ethanol (95 per cent), add 2 ml D. To 1 mg add 0.2 ml of a freshly prepared 1 per cent w/v
of 0.25 M sulphuric acid and 10 ml of ethanol (95 per cent) solution of vanillin in hydrochloric acid; a pink colour
and warm gently to dissolve. Cool, dilute to 100.0 ml with develops within 2 minutes.
ethanol (95 per cent) and dilute 5.0 ml to 50.0 ml with the
same solvent (solution A). Transfer 10.0 ml to a boiling tube, E. Mix about 0.5 mg with 5 mg of 4-dimethylaminobenzal-
add 2 ml of 0.25 M sulphuric acid and 2 ml ofa freshly prepared dehyde and 0.2 ml of glacial acetic acid and add 0.2 ml of
0.3 per cent w/v solution of sodium nitrite, mix and heat in a sulphuric acid; a green colour is produced. Add 1 ml of
water-bath at 55° for 35 minutes. Cool, add 1 ml of a freshly glacial acetic acid; the colour changes to red.
prepared 5 per cent w/v solution of sulphamic acid and dilute
to 25.0 ml with ethanol (95 per cent). Measure the absorbance Tests
ofthe resulting solution atthe maximum at about 388 urn (2.4.7),
pH (2.4.24). 3.0t04.0.
using as the blank a solution prepared by treating a further
10.0 ml of solution A in the same manner and at the same time Other tests. Comply with the tests stated under Parenteral
but omitting the sodium nitrite solution. Preparations (Injections).
Calculate the content of C33H4oNz09 from the absorbance Assay. Protect the solutions from light throughout the assay.
obtained by repeating the operation using reserpine RS in
Measure accurately a volume ofthe injection containing about
place of the substance under examination.
10 mg ofReserpine and dilute with a 2 per cent w/v solution of
Storage. Store protected from light. citric acid to 100.0 ml. Extract 10.0 ml of this solution for

2048

www.webofpharma.com
IP 2010 RESERPINE TABLETS

2 minutes with three quantities, each of 15 ml, of chloroform. each of 5 ml, of chloroform, filter the extracts through a plug
Wash the combined extracts with 10 ml of a 1 per cent w/v of cotton moistened with chloroform. Wash the chloroform
solution of sodium bicarbonate, add sufficient chloroform to extracts with to ml of a 1 per cent w/v solution of sodium
produce 50.0 ml, mix and evaporate to.O ml to dryness on a bicarbonate and evaporate the chloroform extracts to dryness
water-bath. Dissolve the residue in 10 ml of ethanol (95 per on a water-bath. For tablets containing upto 250 flg of
cent), add 2 ml of 0.25 M sulphuric acid and 10 ml of ethanol Reserpine per tablet, dissolve the residue in 10.0 ml of ethanol
(95 per cent) and warm gently to dissolve. Cool, dilute to (95 per cent). For tablets containing more than 250 flg of
100.0 ml with ethanol (95 per cent) and dilute 5.0 ml to 50.0 ml Reserpine per tablet, dissolve the residue in a suitable volume
with the same solvent (solution A). Transfer to.O ml to a boiling of ethanol (95 per cent) to give a concentration of250 flg of
tube, add 2 ml of 0.25 M sulphuric acid and 2 ml of a freshly Reserpine per 10.0 ml; add 2 ml of 0.25 M sulphuric acid and
prepared 0.3 per cent w/v solution of sodium nitrite, mix and 2 ml of a freshly prepared 0.3 per cent w/v solution of sodium
heat in a water-bath at 55° for 35 minutes. Cool, add 1 mlofa nitrite, mix and heat in a water-bath at 55° for 35 minutes. Cool,
freshly prepared 5 per cent w/v solution of sulphamic acid add 1 ml of a freshly prepared 5 per cent w/v solution of
and dilute to 25.0 ml with ethanol (95 per cent). Measure the sulphamic acid and dilute to 25.0 ml with ethanol (95 per
absorbance ofthe resulting solution at the maximum at about cent). Measure the absorbance of the resulting solution at
388 nm (2.4.7), using as the blank a solution prepared by the maximum at about 388 nm (2.4.7), using as the blank a
treating a further to. 0 ml ofsolution A in the same manner and solution prepared by treating a further 10.0 ml ofsolution A in
at the same time but omitting the sodium nitrite solution. the same manner and at the same time but omitting the sodium
Calculate the content of C33H40Nz09 from the absorbance nitrite solution.
obtained by repeating the operation using reserpine RS in Calculate the content of C33H40Nz09 in the tablet from the
place of the substance under examination. absorbance obtained by repeating the operation using
Storage. Store protected from light in single dose (or if reserpine RS in place of the substance under examination.
stabilising agents are present, in multiple dose) containers.
Other tests. Comply with the tests stated under Tablets.

Assay. Protect the solutions from light throughout the assay.

Weigh accurately a. quantity of the powdered tablets


Reserpine Tablets containing about 1 mg ofReserpine, add 10 ml ofa 2 per cent
Reserpine Tablets contain not less than 90.0 per cent and not w/v solution of citric acid and extract for 2 minutes with three
more than 110.0 per cent of the stated amount of reserpine, quantities, each of 15 ml, of chloroform. Wash the combined
extracts with to m1 of a 1 per cent w/v solution of sodium
C33~oNz09'
bicarbonate, add sufficient chloroform to produce 50.0 ml
Usual strengths. 100 flg; 250 flg; 500 flg; 1 mg. and evaporate to.O m1 to dryness on a water-bath. Dissolve
the residue in 10 m1 of ethanol (95 per cent) and add 2 m1 of
Identification 0.25 M sulphuric acid and 10 m1 of ethanol (9Jper cent) and
warm gently to dissolve. Cool, dilute to 100.0 m1 with ethanol
A. Powder a few tablets and extract with chloroform. Evaporate
(95 percent) and dilute 5.0 ml to 50.0 ml with the same solvent
the extract to dryness, add 0.1 ml ofa 0.1 per cent w/v solution
(solution A). Transfer 10.0 m1 to a boiling tube, add 2 ml of
of sodium molybdate in sulphuric acid; a yellow colour is
0.25 M sulphuric acid and 2 ml of a freshly prepared 0.3 per
produced which changes to blue within 2 minutes.
cent w/v solution of sodium nitrite, mix and heat in a water-
B. Powder a few tablets and extract with chloroform. Evaporate bath at 55° for 35 minutes. Cool, add 1 m1 ofa freshly prepared
the extract to dryness, add 0.2 ml of a freshly prepared I per 5 per cent w/v solution of sulphamic acid and dilute to 25.0 ml
cent w/v solution of vanillin in hydrochloric acid; a pink with ethanol (95 per cent). Measure the absorbance of the
colour develops within 2 minutes. resulting solution at the maximum at about 388 nm (2.4.7),
using as the blank a solution prepared by treating a further
Tests to.O m1 ofsolution A in the same manner and at the same time
but omitting the sodium nitrite solution.
Uniformity of content. Protect the solutions from light
throughout the test. Calculate the content of C33H40Nz09 from the absorbance
Comply with the test stated under Tablets. obtained by repeating the operation using reserpine RS in
place of the substance under examination.
Powder one tablet, disperse in 10 ml ofa 2 per cent w/v solution
of citric acid and extract for 2 minutes with three quantities, Storage. Store protected from light.

2049

www.webofpharma.com
RlBAVIRlN IP 2010

Ribavirin Chromatographic system


a stainless steel column 10 cm x 7.8 mm, packed with
strong cation-exchange resin (9 Ilm),
- column temperature. 40°,
mobile phase. water, adjusted to pH 2.5 with sulphuric
acid,
- flow rate. ImlpermiIiute,
- spectrophotometer set &t 207 nm,
- injection volume. 10 Ill.
Inject reference solution (b). The test is not valid unless the
peak-to-valley ratio is not less than 1.2, where H p is the height
above the baseline ofthe peak due to ribavirin impurity F and
H v is the height above the baseline of the lowest point of the
Mol.Wt. 244.2 curve separating this peak from the peak due to ribavirin.

Ribavirin is 1-~-D-ribofuranosyl-lH-l ,2,4-triazole-3- Inject test solution (a), reference solution (a) and (b). Record
carboxamide. the chromatogram eleven times the retention time of the
principal peak. The relative retention time with reference to
Ribavirin contains not less than 98.0 per cent and not more ribavirin for 1-(5-0-acetyl-~-D-ribofuranosyl)-IH-l,2,4-
than 102.0 per cent ofCsHJ2N40s, calculated on the dried basis. triazole-3-carboxarnide (5'-O-acetylribavirin) (ribavirin impurity
Category. Antiviral. F ) is about 1.2. The peak area corresponding to ribavirin
impurity F is not more than the area ofthe principal peak in the
Description. A white or almost white crystalline powder.
chromatogram obtained with reference solution (a) (0.1 per
cent). The area of each secondary peak corresponding to 1-~­
Identification
D-ribofuranosyl-lH-l ,2,4-triazole-3-carboxylic acid (ribavirin
Determine by infrared absorption spectrophotometry (2.4.6). impurity A), l-a-D-ribofuranosyl-lH-l,2,4-triazole-3-
Compare the spectrum with that obtained with ribavirin RS or carboxamide (ribavirin impurity B), IH-l,2,4-triazole-3-
with the reference spectrum ofribavirin. carboxylic acid (ribavirin impurity C), H-l,2,4-triazole-3-
carboxamide (ribavirin impurity D), 1-(5-0-benZoyl~a -D-
Tes'ts ribofuranosyl)-IH-l,2,4-triazole-3-carboxamide (5' -0-
benzoylribavirin) (ribavirin impurity E), 1- ~ -D-ribofuranosyl-
pH (2.4.24). 4.0 to 6.5, determined in a 2.0 per cent w/v solution IH-l ,2,4-triazole-5-carboxamide (N-isomer) (ribavirin impurity
in carbon dioxide-free water. G) is not more than the area of the principal peak in the
Specific optical rotation (2.4.22). - 33° to - 37°, determined in chromatogram obtained with reference solution (a) (0.1 per
a 1.0 per cent w/v solution, use freshly prepared solution. cent). The sum of areas ofall the secondary peaks is not more
than twice the area ofthe principal peak in the chromatogram
Related substances. Determine by liquid chromatography obtained with reference solution (a) (0.2 per cent). Ignore any
(2.4.14). peak with an area less than 0.5 times the area ofthe principal
Test solution (a). Dissolve 50 mg of the substance under peak in the chromatogram obtained with reference solution
examination in 100 ml ofthe mobile phase. (a) (0.05 per cent).

Test solution (b). Dissolve 25 mg of the substance under Heavy metals (2.3.13). Dissolve 4.0 g in 20 ml of water with
examination in100 ml ofthe mobile phase. Dilute 10 ml ofthe heating ifnecessary and use 12 ml ofthe solution. The resulting
solution to 100 ml with the mobile phase. solution complies with the limit test for heavy metals, Method
B(10ppm).
Reference solution (a). Dilute 1 ml of test solution (a) to 100
ml with the mobile phase. Dilute 1 ml ofthis solution to 10 ml Sulphated ash (2.3.18). Not more than 0.1 per cent.
with the mobile phase. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105° for 5 hour.
Reference solution (b). Dissolve the contents of a vial of
ribavirinfor system suitability RS in 2 ml Of the mobile phase. Assay. Determine by liquid chromatography (2.4.14) same as
described under Related substances with the following
Reference solution (c). Dissolve 25 mg of ribavirin RS in 100
modification.
ml pfthe mobile phase. Dilute 10 ml ofthe solution to 100 ml
with the mobile phase. Inject test solution (b) and reference solution (c).

2050

www.webofpharma.com
IP 2010 RlBAVIRlN INHALATION SOLUTION

Calculate the content ofCsHl2N40s. spot in the chromatogram obtained with the test solution is
similar in position, colour and size to that in the chromatogram
Storage. Store protected from light.
obtained with the reference solution.
B. In the Assay, the retention time ofthe principal peak in the
chromatogram obtained with the test solution corresponds to
Ribavirin Inhalation Solution that of the principal peak in the chromatogram obtained with
the reference solution.
Ribavirin Solution for Inhalation
Catogery. Antiviral. Tests
Ribavirin Inhalation Solution is a sterile solution ofRibavirin Related substances. Determine by liquid chromatography
in Water for Injections. It is prepared by dissolving Ribavirin (2.4.14).
for Inhalation in the requisite amount ofWater for Injections.
Test solution. A 0.05 per cent w/v solution of ribavirin in the
mobile phase.
Ribavirin for Inhalation
Reference solution (a). A 0.000125 per cent w/v solution of
Ribavirin for Inhalation is a sterile ribavirin consisting of ribavirin in the mobile phase.
Ribavirin with or without excipients. It is filled in a sealed
container.
Reference solution (b). A 0.05 per cent w/v solution of
ribavirin RS in the mobile phase.
The inhalation solution is constituted by dissolving the
contents of the sealed container in the requisite amount of Chromatographic system
sterile water for injection, immediately before use. a stainless steel column 10 cm x 7.8 mm, packed with
strong cation-exchange resin of sulphonated, cross-
The constituted solution complies with the requirements for linked styrene-divinylbenzene co-polymer in the
Clarity of solution and particulate matter stated under hydrogen form (7 to 11 Ilm)(Such asArninexHPAH),
Inhalation Preparations. column temperature. 40°,
Storage. The constituted solution should be used immediately mobile phase: water, adjusted to pH 2.5 with sulphuric
after preparation but, in any case, within the period acid,
recommended by the manufacturer. flow rate. 1 ml per minute,
- spectrophotometer set at 207 urn,
Ribavirin Inhalation Solution contains not less than 95.0 per
- injection volume. 10 Ill.
cent and not more than 105.0 per cent ofthe stated amount of
CSH 12N40 s. Inject reference solution (b). The test is not valid unless the
tailing factor for the principal peak is not less than 0.7 and not
The content of the sealed container complies with the
more than 1.5.
requirements stated under Inhalation Preparations and with
the following requirements. Inject the test solution and reference solution (a). In the
chromatogram obtained with the test solution, the area ofany
Identification secondary peak is not more than the area of principal peak in
the chromatogram obtained with reference solution (a) (0.25
A. Determine by thin-layer chromatography (2.4.17), coating per cent) and the sum of the areas of all the secondary peaks
the plate with silica gel G. is not more than 4 times the area of the principal peak in the
Mobile phase. A mixture of 20 volumes of 0.1 M ammonium chromatogram obtained with reference solution (a) (1.0 per
chloride and 90 volumes of acetonitrile. cent).
Test solution. Dissolve ribavirin inhalation solution containing Sterility (2.2.11). Complies with the test for sterility.
about 0.1 g ofRibavirin in 10 ml of water.
Assay. Determine by liquid chromatography (2.4.14).
Reference solution. A 1.0 per cent w/v solution of ribavirin
Test solution. Reconstitute the contents of one container as
RS in water.
instructed on the label using an accurately measured volume
Apply to the plate 10 III of each solution. After development, of diluent, dilute a suitable volume of the solution to 200
dry the plate in air for 15 minutes, and spray the plate with the volumes with the mobile phase to produce a solution
mixture of0.5 ml of anisaldehyde, 0.5 ml ofsulphuric acid, 0.1 containing 0.1 per cent w/v of Ribavirin, mix and dilute 5
ml of glacial acetic acid and 9 ml of ethanol (95 per cent) and volumes of the resulting solution to 20 volumes with the
heat at 105° for 40 minutes and allow to cool.The principal mobile phase.

2051

www.webofpharma.com
RlBOFLAVINE IP 2010

Reference solution. A 0.025 per cent w/v solution of ribavirin Dose. Prophylactic, 1 to 4 mg daily; therapeutic, 5 to 10 mg
RS in the mobile phase. daily.
Chromatographic system Description. A yellow to orange-yellow, crystalline powder;
- a stainless steel column 10 cm x 7.8 mm, packed with odour, slight.
strong cation-exchange resin of sulphonated, cross-
linked styrene-divinylbenzene co-polymer in the Identification
hydrogen form (7 to 11 J.Lm) (Such as Aminex HPAH),
A. Determine by infrared absorption spectrophotometry (2.4.6).
- column temperature. 40°,
Compare the spectrum with that obtained with riboflavine RS
- mobile phase: water, adjusted to pH 2.5 with sulphuric
or with the reference spectrum ofriboflavine.
acid,
- flow rate. 1 ml per minute, B. Dissolve about 1 mg in 100 ml of water. The solution has a
spectrophotometer set at 207 urn, pale greenish yellow colour by transmitted light and an intense
- injection volume. 10 J.Ll. yellowish green fluorescence by reflected light, which
disappears on addition of mineral acids or allcalis.
Inject the reference solution. The test is not valid unless the
tailing factor is not less than 0.7 and not more than 1.5. Tests
Inject the test solution and the reference solution.
pH (2.4.24).5.5 to 7.2, determined in a saturated solution.
Calculate the content of CgHI2N40s in inhalation solution. Specific optical rotation (2.4.22). -115° to -135°, determined
Labelling. The label of the sealed container states (1) the in a 0.5 percentw/v solution in carbonate-free 0.05 Msodium
procedure for preparing the solution, (2) the delivery system hydroxide. Measure the angle ofrotation within 30 minutes of
to be used for the constituted solution (3) the conditions preparing the solution.
under which the constituted solution should be stored. Light absorption (2.4.7). Dilute a suitable volume ofthe final
solution obtained in the Assay with an equal volume of water.
When examined in the range 210 urn to 460 urn, the resulting
solution exhibits maxima at about 223 urn, 267 urn, 373 nm and
Riboflavin 444 nm; the ratio of the absorbance at the maximum at about
373 urn to that at about 267 urn, 0.31 to 0.33 and the ratio ofthe
Lactoflavin; Vitamin B 2
absorbance at the maximum at about 444 nm to that at about
267 urn, 0.36 to 0.39.
Lumiflavine. Shake 25 mg with 10 ml ofchlorofOrm for 5 minutes
and filter. The filtrate is not more intensely coloured than
reference solution BYS6 (2.4.1).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 1.5 per cent, determined
on 1.0 g by drying in an oven at 105°.
Assay. Carry out the procedure in subdued light.
Weigh accurately about 65 mg and transfer to an amber-glass
500-ml volumetric flask, suspend in 5 ml of water, ensuring
that it is completely wetted. Dissolve in 5 ml of 2 M sodium
hydroxide. As soon as dissolution is complete add 100 ml of
Mol. Wt. 376.4
water and 2.5 ml of glacial acetic acid and dilute to 500.0 ml
with water. To 20.0 ml of this solution add 3.5 ml ofa 1.4 per
Riboflavin is 3,1O-dihydro-7,8-dimethyl-l0-[(2S,3S,4R)- cent w/v solution of sodium acetate and dilute to 200.0 ml
2,3,4,5-tetrahydroxypentyl]benzo[g]pteridine-2,4-dione. with water. Measure the absorbance ofthe resulting solution
Riboflavin contains not less than98.0 per cellt and not more atthe maximum at about 444 nm (2.4.7).
thali 101.0 per celit of CI7H2oN406, calCulated on the dried Calculate thec()ntetlt ()fC17H2oN406 taking 328 as the specific
basis. absorbance at 444 nm.
Category. B-group vitamin. Storage. Store protected from light.

2052

www.webofpharma.com
IP 2010 RIBOFLAVINE SODIUM PHOSPHATE

Riboflavin Sodium Phosphate Tests


Riboflavine-5-phosphate (Sodium Salt); Vitamin B2 pH (2.4.24). 4.0 to 6.3, determined in a 2.0 per cent w/v
Sodium Phosphate solution.
Specific optical rotation (2.4.22). +38.0° to +42.0°, determined
in a 1.5 per cent w/v solution in 5 M hydrochloric acid.
Heavy metals (2.3.13). To 2.0 g in a silica crucible add 2 ml of
nitric acid dropwise followed by 0.25 ml of sulphuric acid.
Heat cautiously until white fumes are evolved and ignite. Extract
the cooled residue with two quantities, each of 2 ml, of
hydrochloric acid and evaporate the extracts to dryness.
Dissolve the residue in 2 ml of 2 M acetic acid and dilute to
,2H zO 20 ml with water. 12 ml of the solution complies with the
limit test for heavy metals, Method D (10 ppm). Use 1.0 ml
of lead standard solution (10 ppm Pb) to prepare the
standard.
Lumiflavine. Shake 35 mg with 10 ml of dichloromethane for
Mol. Wt. 514.4 5 minutes and filter. The filtrate is not more intensely coloured
than reference solution BYS6 (2.4.1).
Riboflavin Sodium Phosphate is monosodium 3, lO-dihydro-
7,8-dimethyl-1 0-[(2S,3S,4R)-2,3,4-trihydroxypentyl] Inorganic phosphate. Not more than 1.5 per cent, determined
benzo[g]pteridine-2,4-dione 5-phosphate dihydrate. by the following method. Dissolve 0.1 g in sufficient water to
Riboflavin Sodium Phosphate contains the equivalent of not produce 100 mI. Dilute 5 rnl with 10 rnl ofwater and add 5 mI of
less than 73.0 per cent and not more than 79.0 per cent of buffered cupric sulphate solution pH 4.0,2 ml of a 3 per cent
CI7H2oN406, calculated on the dried basis. w/v solution of ammonium molybdate, 1 ml of a freshly
prepared solution containing 2 per cent w/v of 4-methyl-
Category. B-group vitamin. aminophenol sulphate and 5 per cent w/v of sodium meta-
Dose. Prophylactic, the equivalent of 1 to 4 mg ofriboflavine bisulphite and 1 ml of a 3 per cent v/v solution ofperchloric
daily; therapeutic, the equivalent of 5 to 10 mg ofriboflavine acid. Add sufficient water to produce 25 ml, mix and measure
daily. (1.37 g ofriboflavine sodium phosphate is approximately the absorbance of the resulting solution at the maximum at
equivalent to·1 g ofriboflavine). about 800 nm (2.4.7), within 15 minutes of its preparation,
using as the blank a solution prepared in the same manner but
Description. A yellow to orange-yellow, crystalline powder;
omitting the substance under examination. The absorbance is
hygroscopic.
not greater than that produced by repeating the operation
using a solution prepared in the same manner using 15 ml of
Identification phosphate standard solution (5 ppm PO,J and beginning at
A. When examined in the range 230 nm to 360 nm (2.4.7), a the words "add 5 ml of buffered cupric sulphate solution
0.001 per cent w/v solution in phosphate bufferpH 7.0 shows pH 4.0,"
an absorption maximum at about 267 nm; absorbance at about Loss on drying (2.4.19). Not more than 8.0 per cent, determined
267 nm, 0.58 to 0.64. on 0.5 g by drying in an oven at 100° at a pressure not exceeding
B. Dissolve about 10 mg in sufficient 2 M sodium hydroxide 0.7 kPa for 5 hours.
to produce 100 rnl, expose 1 ml to ultraviolet light at 254 nm for Assay. Carry out the procedure protectedfrom light.
5 minutes, add sufficient 5 M acetic acid to make the solution
acidic to litmus paper and shake the mixture with 2 ml of Weigh accurately about 0.1 g, dissolve in 150 ml of water, add
dichloromethane; the lower layer exhibits a yellow 2 ml of glacial acetic acid and dilute to 1000.0 ml with water.
fluorescence. To 10.0 m1 add 3.5 ml ofa 1.4 per cent w/v solution ofsodium
acetate, dilute to 50.0 ml with water and measure the
C. To 0.5 g add 10 ml of nitric acid, evaporate the mixture to
absorbance ofthe resulting solution at the maximum at about
dryness on a water-bath, ignite the residue until the carbon is
444 nm (2.4.7). Calculate the percentage content ofC17H2oN406
removed, dissolve the final residue in 5 ml of water and filter.
taking 328 as the specific absorbance at 444 nm.
The filtrate gives the reactions of sodium salts and reaction B

of phosphates (2.4.1). Storage. Store protected from light.

2053

www.webofpharma.com
RIBOFLAVINE TABLETS IP 2010

Riboflavin Tablets Rifampicin


Lactoflavin Tablets; Vitamin B z Tablets Rifampin
Riboflavin Tablets contain not less than 90.0 per cent and not
more than 115.0 per cent ofthe stated amount of riboflavine, CH
I 3 CH 3
C17HzoN406. HO,
~
I
Usual strengths. 2 mg; 5 mg. H3C O OH
y CH 3
0
CH 3
Identification 0 NH
H3CO- 'CH 3 OH OH
Shake a quantity of the powdered tablets containing 1 mg
~ H3C
of Riboflavine with 100 ml of water and filter; the filtrate ~
has a pale greenish yellow colour by transmitted light 0 //
and an intense yellowish green fluorescence by reflected ,;;N'Nl
light, which disappears on addition of mineral acids or
alkalis.
OH
~N'CH3
Tests
C43HssN40JZ Mol. Wt. 823.0
Uniformity of content. Comply with the test stated under
Rifampicin is (12Z, 14E,24E)-(28,168,178,18R,19R,20R,
Tablets.
218,22R,23S)-1,2-dihydro-5,6,9,17,19-pentahydroxy-
Powder one tablet, add a mixture of 2.5 ml of glacial acetic 23-methoxy-2,4,12,16,18,20,22-heptamethyl-8-(4-methyl-
acid and 50 ml of water and heat on a water-bath for piperazin-l-yliminomethyl)-l, ll-dioxo-2,7-(epoxypentadeca-
1 hour with occasional stirring. Dilute with 50 ml of water, 1,11, l3-trienimino)naphtho[2, I-b ]furan-21-yl acetate.
add 15 ml of 1 M sodium hydroxide with continuous Rifampicin contains not less than 97.0 per cent and not more
stirring and then sufficient water to produce a solution than 102.0 per cent of C43HssN40JZ, calculated on the dried
containing 10 llg of Riboflavine per ml. Filter and discard basis.
the first few ml of the filtrate. On the clear filtrate carry out
the Assay beginning at the words "Measure the Category. Antitubercular.
absorbance....". Dose. For an adult, 450 to 600 mg (about 10 mg per kg) daily
preferably before breakfast. For a child, upto 20 mg per kg
Other tests. Compiy with the tests stated under Tablets. daily to a maximum of600 mg.
Assay. earlY out the procedure in subdued light. Description. A brick-red to reddish brown, crystalline powder;
practically odourless.
Weigh and powder 20 tablets. Weigh accurately a quantity of
the powder containing about 10 mg of Riboflavine, add a Identification
mixture of5 ml ofglacial acetic acid and 100 ml ofwater and
heat on a water-bath for 1 hour with occasional shaking. Dilute A. Determine by infrared absorption spectrophotometry (2.4.6).
with 50 ml ofwater, cool, add 30 ml of 1 M sodium hydroxide Compare the spectrum with that obtained with rifampicin R8
with continuous stirring. Add sufficient water to produce or with the reference spectrum ofrifampicin.
1000.0 mI, mix and filter, discarding the first few mI ofthe filtrate. B. In the Assay, the principal peak in the chromatogram
Measure the absorbance ofthe filtrate at the maximum at about obtained with the test solution corresponds to the peak in the
444 nm (2.4.7). chromatogram obtained with the reference solution.

Calculate the content ofC17HzoN406 taking 328 as the specific Tests


absorbance at 444 nm.
pH (2.4.24). 4.5 to 6.5, determined in a 1.0 per cent w/v
Storage. Store protected from light. suspension.
Related substances. Determine by liquid chromatography
(2.4.14).
NOTE - Prepare the solutions immediately before use.

2054

www.webofpharma.com
IP 2010 RIFAMPICIN CAPSULES

Solvent mixture. A mixture of 10 volumes of a 21.01 per cent of a 17.42 per cent w/v solution of dipotassium hydrogen
w/v solution of citric acid, 23 volumes of a 13.61 per cent phosphate, 640 volumes of water and 250 volumes of
w/v solution ofpotassium dihydrogen phosphate, 77 volumes acetonitrile.
of a 17.42 per cent w/v solution of dipotassium hydrogen
Test solution. Dissolve 20.0 mg of the substance under
phosphate, 640 volumes of water and 250 volumes of
examination in 10.0 ml of acetonitrile and filter. Dilute 5.0 ml of
acetonitrile.
the filtrate to 100.0 ml with the solvent mixture.
Test solution. Weigh accurately a quantity of powder
Reference solution. A solution containing 0.2 per cent w/v of
containing 20 mg of Rifampicin, add 10 ml of acetonitrile,
rifampicin RS in acetonitrile. Dilute 5.0 ml of this solution to
shake and filter. Dilute 5 ml of the filtrate to 50 ml with the
100.0 ml with the solvent mixture.
solvent mixture.
Reference solution. A solution containing 0.02 per cent w/v of Use the chromatographic system and the system suitability
rifampicin quinone RS in acetonitrile. To 1 ml ofthe solution, parameters described under the test for Related substances.
add 1 ml of the test solution and dilute to 100 ml with the Inject the reference solution. The test is not valid unless the
solvent mixture. relative standard deviation for replicate injections is not more
Chromatographic system than 2.0 per cent.
a stainless steel column 10 cm x 4.6 mm, packed with Inject the test solution and the reference solution.
octylsilane bonded to porous silica (5 /lm),
- mobile phase: a mixture of 65 volumes of a solution Calculate the content ofC43H58N4012.
containing 0.1 per cent v/v of orthophosphoric acid, Storage. Store protected from light, in an atmosphere of
0.19 per cent w/v of sodium perchlorate, 0.59 per cent nitrogen.
w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate and 35 volumes of acetonitrile,
flow rate. 1.5 ml per minute,
spectrophotometer set at 254 mn, Rifampicin Capsules
injection volume. 20 Ill.
Rifampin Capsules
Inject the reference solution. The test is not valid unless the
resolution between rifampicin and rifampicin quinone is not Rifampicin Capsules contain not less than 92.5 per cent and
less than 4. not more than 107.5 per cent ofthe stated amount ofrifampicin,
C43H58N4012.
In the chromatogram obtained with the test solution the area
of any peak due to rifampicin quinone is not more than Usual strengths. 150 mg; 300 mg; 450 mg; 600 mg.
1.5 times the area ofthe peak due to rifampicin quinone in the
chromatogram obtained with the reference solution (1.5 per Identification
cent); the area ofany peak, other than the peak due to rifampicin A. Shake a quantity ofthe contents ofthe capsules containing
and rifampicin quinone, is not more than the area ofthe peak 0.15 g ofRifampicin with 5 ml of chloroform, filter and evaporate
due to rifampicin in the chromatogram obtained with the the filtrate to dryness. The residue complies with the following
reference solution (1.0 per cent) and the sum of the areas of test.
any such peaks is not more than 3.5 times the area ofthe peak
due to rifampicin in the chromatogram obtained with the Determine by infrared absorption spectrophotometry (2.4.6)..
reference solution (3.5 per cent). Compare the spectrum with that obtained with rifampicin RS
or with the reference spectrum ofrifampicin.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm). B. In the Assay, the principal peak in the chromatogram
Sulphated ash (2.3.18). Not more than 0.1 per cent. obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 80 0 at a pressure not exceeding Tests
0.7 kPa for 4 hours.
Assay. Determine by liquid chromatography (2.4.14). Related substances. Determine by liquid chromatography
(2.4.14).
NOTE - Prepare the solutions immediately before use.
NOTE - Prepare the solutions immediately before use.
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent
w/v solution of citric acid, 23 volumes of a 13.61 per cent Solvent mixture. A mixture of 10 volumes ofa 21.01 per cent
w/v solution ofpotassium dihydrogen phosphate, 77 volumes w/v solution of citric acid, 23 volumes of a 13.61 per cent

2055

www.webofpharma.com
RIFAMPICIN CAPSULES IP 2010

w/v solution ofpotassium dihydrogen phosphate, 77 volumes Dissolution (2.5.2).


of a 17.42 per cent w/v solution of dipotassium hydrogen Apparatus No.2,
phosphate, 640 volumes of water and 250 volumes of
Medium. 900 ml of 0.1 M hydrochloric acid,
acetonitrile.
Speed and time. 100 rpm and 45 minutes.
Test solution. Shake a quantity ofthe contents ofthe capsules
Withdraw a suitable volume ofthe medium and filter promptly
containing 200 mg of Rifampicin, with 100 ml of acetonitrile
through a membrane filter disc having an average pore diameter
and filter. Dilute 5 ml ofthe filtrate to 50 ml with the solvent
not greater than 1.0 f.Lm, rejecting the first 1 ml ofthe filtrate.
mixture.
Dilute the filtrate, ifnecessary, with the same solvent. Measure
Reference solution (a). A solution containing 0.02 per cent the absorbance (2.4.7) ofthe resulting solution at the maximum
w/v of rifampicin RS in acetonitrile. Dilute 1 ml ofthis solution at about 475 nm. Calculate the content ofC43HssN4012 in the
to 100 ml with the solvent mixture. medium from the absorbance obtained from a solution of
Reference solution (b). A solution containing 0.01 per cent Imown concentration of rifampicin RS.
w/v each of rifampicin RS and rifampicin quinone RS in D. Not less than 75 per cent of the stated amount of
acetonitrile. Dilute 5 ml of this solution to 50 ml with the C43HssN4012'
solvent mixture.
Other tests. Comply with the tests stated under Capsules.
Chromatographic system
- a stainless steel column 10 cm x 4.6 mm, packed with Assay. Determine by liquid chromatography (2.4.14).
octylsilane bonded to porous silica (5 f.Lm), Test solution. Shake a quantity ofthe contents of the capsules
- column temperature. 30°, containing about 300.0 mg of Rifampicin, with 200.0 ml of
- mobile phase: a mixture of 65 volumes of a solution acetonitrile, filter. Dilute 10.0 ml ofthe filtrate to 50.0 ml with
containing 0.1 per cent v/v of orthophosphoric acid, acetonitrile. Dilute 5.0 ml of this solution to 50.0 ml with the
0.19 per cent w/v of sodium perchlorate, 0.59 per cent solvent mixture.
w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate and 35 volumes of acetonitrile, Reference solution (a). A 0.15 per cent w/v solution of
- flow rate. 1.5 ml per minute, rifampicin RS in acetonitrile. Dilute 10.0 ml ofthis solution to
- spectrophotometer set at 254 urn, 50.0 ml with acetonitrile. Dilute 5.0 ml ofthe resulting solution
injection volume. 20 f.Ll. to 50.0 ml with the solvent mixture.

Inject reference solution (b). The test is not valid unless the Reference solution (b). A solution containing 0.01 per cent
resolution between rifampicin and rifampicin quinone is not w/v each of rifampicin RS and rifampicin quinone RS in
less than 4, the tailing factor is not more than 2.0 and the acetonitrile. Dilute 5ml of this solution to 50 ml with the
column efficiency is not less than 2000 theoretical plates. solvent mixture.

Inject the test solution and reference solution (a). The relative Use the chromatographic system and system suitability
retention times are 1.0, 0.55, 1.25 and 2.61 for rifampicin, parameters, as described under the test for Related substances.
rifampicin quinone, rifampicin N-oxide and 3-formylrifamycin Inject reference solution (a). The test is not valid unless the
SV respectively. The response factors are 1.00, 1.19, 1.03 and relative standard deviation for replicate injections is not more
1.25 for rifampicin, rifampicin quinone, rifampicin N-oxide and than 2.0 per cent.
3-formylrifamycin SV respectively.
Inject the test solution and reference solution (a).
In the chromatogram obtained with the test solution the area
of any peak due to rifampicin quinone should not be more Calculate the content ofC43HssN4012 in the capsules.
than 4 times the area ofthe principal peak in the chromatogram
Storage. Store protected from light and moisture.
obtained with the reference solution, the area of any peak due
to rifampicin N-oxide should not be more than 1.5 times the
. area ofthe principal peak in the chromatogram obtained with
the reference solution and the area of any peak due to
3-formylrifamycin SV should not be more than the area ofthe Rifampicin Oral Suspension
principal peak in the chromatogram obtained with the reference
Rifampicin oral suspension contains not less than 90.0 per
solution. In the chromatogram obtained with the test solution
cent and not more than 110.0 per cent ofthe stated amount of
the area of allY unknowllpealc shoulclllot more than the
rifampicin, C43HssN4012.
area ofthe principal peak in the chromatogram obtained with
the reference solution. Usual strengths. 10 mg per ml; 20 mg per ml.

2056

www.webofpharma.com
IP 2010 RIFAMPICIN ORAL SUSPENSION

Identification - flow rate. 1.5 ml per minute,


- spectrophotometer set at 254 nm,
A. To a quantity containing 0.1 g of Rifampicin add 30 m1 of
injection volume. 20 Ill.
water and shake with two quantities, each of 50 ml, of
chloroform. Dry the combined extracts with anhydrous sodium Inject reference solution (b). The test is not valid unless the
sulphate, filter and evaporate the filtrate to dryness at a resolution between rifampicin and rifampicin quinone is not
temperature not exceeding 70°. Wash the residue with 1 ml of less than 4; the tailing factor is not more than 2.0 and the
ether and illy at 70°. The residue complies with the following column efficiency is not less than 2000 theoretical plates.
test. Inject the test solution and reference solution (a). The relative
Determine by infrared absorption spectrophotometry (2.4.6). retention times are 1.0, 0.55, 1.25 and 2.61 for rifampicin,
Compare the spectrum with that obtained with rifampicin RS rifampicin quinone, rifampicin N-oxide and 3-formylrifamycin
or with the reference spectrum ofrifampicin. SV respectively. Multiply the areas of each Imown impurity
by their response factor. The response factors are 1.00, 1.19,
B. In the Assay, the principal peak in the chromatogram
1.03, 1.22 and 1.25 for lifampicin, rifampicin quinone, rifampicin
obtained with the test solution con'esponds to the peak in the
N-oxide, Isonicotinyl hydrazone and 3-formylrifamycin SV
chromatogram obtained with the reference solution.
respectively.
Tests In the clu'omatogram obtained with the test solution the area
pH (2.4.24). 4.2 to 4.8. of any peak due to rifampicin quinone should not be more
than 1.5 times the area ofthe principal peak in the chromatogram
Related substances. Determine by liquid chromatography obtained with the reference solution, the area of any peak due
(2.4.14). to rifampicin N-oxide should not be more than the area ofthe
NOTE - Prepare the solutions immediately before use. principal peak in the chromatogram obtained with the reference
solution and the area ofany peak due to 3-fonnylrifamycin SV
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent
should not be more than 5.0 times the area of the principal
w/v solution of citric acid, 23 volumes of a 13.61 per cent
peak in the chromatogram obtained with the reference solution.
w/v solution ofpotassium dihydrogen phosphate, 77 volumes
In the chromatogram obtained with the test solution the area
of a 17.42 per cent w/v solution of dipotassium hydrogen
of any unlmown peak should not be more than the area ofthe
phosphate, 640 volumes of water and 250 volumes of
principal peak in the chromatogram obtained with the reference
acetonitrile.
solution.
Test solution. Add 5 m1 of water to a quantity of the oral
Other tests. Comply with the tests stated under Oral
suspension containing 20 mg of Rifampicin and extract with
Suspensions.
four quantities, each of 10 ml, of dichloromethane, filter the
combined extracts and evaporate to dryness at a temperature Assay. Determine by liquid chromatography (2.4.14).
not exceeding 40°. Dissolve the residue in 10 ml of acetonitrile. Test solution. Weigh accurately a quantity of the oral
Dilute 5 ml of the resulting solution to 50 ml with the solvent suspension containing about 150 mg of rifampicin and dilute
mixture. to 100.0 ml with acetonitrile. Dilute 10.0 ml ofthis solution to
Reference solution (a). A solution containing 0.02 per cent 50.0 ml with acetonitrile. Dilute 10.0 ml ofthe resulting solution
w/v of rifampicin RS in acetonitrile. Dilute 1 ml ofthis solution to 50.0 ml with the solvent mixture.
to a 100 ml with the solvent mixture.
Reference solution (a). A 0.15 per cent w/v solution of
Reference solution (b). A solution containing 0.01 per cent rifampicin RS in acetonitrile. Dilute 10.0 ml ofthe solution to
w/v each of rifampicin RS and rifampicin quinone RS in 50.0 m1 with acetonitrile. Dilute 10.0 ml ofthe resulting solution
acetonitrile. Dilute 5 ml of this solution to 50 ml with the to 50.0 ml with the solvent mixture.
solvent mixture.
Reference solution (b). A solution containing 0.01 per cent
Chromatographic system w/v of each of rifampicin RS and rifampicin quinone RS in
- a stainless steel column 10 cm x 4.6 mm, packed with acetonitrile. Dilute 5 ml of this solution to 50 ml with the
octylsilane bonded to porous silica (5 Ilm), solvent mixture.
- column temperature. 30°,
mobile phase: a mixture of 65 volumes of a solution Use the chromatographic system and the system suitability
containing 0.1 per cent v/v of orthophosphoric acid, parameters as described under the test for Related substances.
0.19 per cent w/v of sodium perchlorate, 0.59 per cent Inject reference solution (a). The test is not valid unless the
w/v of citric acid and 2.09 per cent w/v of potassium relative standard deviation for replicate injections is not more
dihydrogen phosphate and 35 volumes of acetonitrile, than 2.0 per cent.

2057

www.webofpharma.com
RIFAMPICIN TABLETS IP 2010

Inject the test solution and reference solution (a). Chromatographic system
a stainless steel column 10 cm x 4.6 mm, packed with
Determine the weight per ml of the suspension (2.4.29) and
octylsilane bonded to porous silica (5 Ilm),
calculate the content ofC43HssN4012 weight in volume.
column. temperature. 30°,
Storage. Store protected from light and moisture. mobile phase: a mixture of 65 volumes of a solution
containing 0.1 per cent v/v of orthophosphoric acid,
0.19 per cent w/v of sodium perchlorate, 0.59 per cent
w/v of citric acid and 2.09 per cent w/v of potassium
Rifampicin Tablets dihydrogen phosphate and 35 volumes of acetonitrile,
Rifampin Tablets flow rate. 1.5 ml per minute,
spectrophotometer set at 254 nm,
Rifampicin Tablets contain not less than 92.5 per cent and not
injection volume. 20 Ill.
more than 107.5 per cent of the stated amount of rifampicin,
C43HssN4012. Inject reference solution (b). The test is not valid unless the
resolution between rifampicin and rifampicin quinone is not
Usualstrength.150mg.
less than 4, the tailing factor is not more than 2.0 and the
Identification column efficiency is not less than 2000 theoretical plates.
Inject the test solution and reference solution (a). The relative
A. Shake a quantity ofthe powdered tablets containing 0.15 g
retention times are 1.0, 0.55, 1.25 and 2.61 for rifampicin,
ofRifampicin with 5 ml of chloroform, filter and evaporate the
rifampicin quinone, rifampicin N-oxide, and 3-formylrifamycin
filtrate to dryness. The residue complies with the following
SV respectively. The response factors are 1.00, 1.19, 1.03 and
test.
1.25 for rifampicin, rifampicin quinone, rifampicin N-oxide and
Determine by infrared absorption spectrophotometry (2.4.6). 3-formylrifamycin SV respectively.
Compare the spectrum with that obtained with rifampicin RS
In the chromatogram obtained with the test solution the area
or with the reference spectrum ofrifampicin.
of any peak due to rifampicin quinone should not be more
B. In the Assay, the principal peak in the chromatogram than 4 times the area ofthe principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the obtained with the reference solution (4 per cent), the area of
chromatogram obtained with the reference solution. any peak due to rifampicin N-oxide should not be more than
1.5 times the area of the principal peak in the chromatogram
Tests obtained with the reference solution (l.5 per cent) and the
area of any peak due to 3-formyl rifamycin SV should not be
Related substances. Determine by liquid chromatography
more than the area of the principal peak in the chromatogram
(2.4.14).
obtained with the reference solution (l per cent). In the
NOTE Prepare the solutions immediately before use. chromatogram obtained with the test solution the area of any
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent unknown peak should not be more than the area of the
w/v solution of citric acid, 23 volumes of a 13.61 per cent principal peak in the chromatogram obtained with the reference
solution w/v ofpotassiurn dihydrogen phosphate, 77 volumes solution.
of a 17.42 per cent w/v solution of dipotassium hydrogen Dissolution (2.5.2).
phosphate, 640 volumes of water and 250 volumes of Apparatus No.2,
acetonitrile. Medium. 900 ml of 0.1 M hydrochloric acid,
Test solution. Weigh accurately a quantity of the powdered Speed and time. 100 rpm and 45 minutes.
tablets containing 200 mg ofRifampicin, dissolve in 100 ml of
Withdraw a suitable volume ofthe medium and filter promptly
acetonitrile, filter. Dilute 5 ml ofthe filtrate to 50 ml with the
through a membrane filter disc having an average pore diameter
solvent mixture.
not greater than 1.0 j.lm, rejecting the first Iml afthe filtrate.
Reference solution (a). A solution containing 0.02 per cent Dilute the filtrate, ifnecessary, with the same solvent. Measure
w/v of rifampicin RS in acetonitrile. Dilute 1 ml ofthis solution the absorbance (2.4.7) ofthe resulting solution at the maximum
to a 100 ml with the solvent mixture. at about 475 nm. Calculate the content ofC43HssN4012 in the
medium from the absorbance obtained from a solution of
Reference solution (b). A solution containing 0.01 per cent
known concentration of rifampicin RS.
w/v of each of rifampicin RS and rifampicin quinone RS in
acetonitrile. Dilute 5 ml of this solution to 50 ml with the D. Not less than 75 per cent of the stated amount of
solvent mixture. C43HssN4012.

2058

www.webofpharma.com
IP 2010 RIFAMPICIN AND ISONIAZID TABLETS

Other tests. Comply with the tests stated under Tablets. phosphate, 640 volumes of water and 250 volumes of
Assay. Determine by liquid chromatography (2.4.14).
acetonitrile.
Test solution. Weigh accurately a quantity of the powdered
Test solution. Weigh and powder 20 tablets. Weigh accurately
tablets containing 200 mg ofRifampicin in 100 ml of acetonitrile
a quantity of the powder containing about 300.0 mg of
and filter. Dilute 5 ml of the filtrate to 50 ml with the solvent
Rifa~picin, dissolve in 200.0 ml of acetonitrile,filter. Dilute
mixture.
10.0 ml ofthe filtrate to 50.0 ml with acetonitrile. Dilute 5.0 ml
ofthis solution to 50.0 ml with the solvent mixture. Reference solution (a). Dissolve rifampicin RS in acetonitrile
to obtain a solution containing 0.2 mg per ml. Dilute 1 mlof
Reference solution (a). A 0.15 per cent w/v solution of
this solution to 100 ml with the solvent mixture.
rifampicin RS in acetonitrile. Dilute 10.0 ml ofthis solution to
50.0 ml with acetonitrile. Dilute 5.0 ml ofthe resulting solution Reference solution (b). A solution containing 0.01 per cent
to 50.0 ml with the solvent mixture. w/v each of rifampicin RS and rifampicin quinone RS in
R~rerence solution (b). A solution containing 0.01 per cent acetonitrile. Dilute 5 ml of this solution to 50 ml with the
solvent mixture.
w/v each of rifampicin RS and rifampicin quinone RS in
acetonitrile. Dilute 5.0 ml of this solution to 50.0 ml with the Chromatographic system
solvent mixture. - a stainless steel column 10 cm x 4.6 rom, packed with
octylsilane bonded to porous silica (5 /lm),
Use the chromatographic system and system suitability
- column temperature 30°,
parameters described under the test for Related substances.
- mobile phase: a mixture of 65 volumes of a solution
Inject reference solution (a). The test is not valid unless the containing 0.1 per cent v/v of orthophosphoric acid,
relative standard deviation for replicate injections is not more 0.19 per cent w/v of sodium perchlorate, 0.59 per cent
than 2.0 per cent. w/v of citric acid and 2.09 per cent w/v of potassium
Inject the test solution and reference solution (a). dihydrogen phosphate and 35 volumes of acetonitrile,
- flowrate. 1.5 ml per minute,
Calculate the content of C43HssN4012 in the tablets. - spectrophotometer set at 254 urn,
Storage. Store protected from light and moisture. injection volume. 20 Ill.
Inject reference solution (b). The test is not valid unless the
resolution between rifampicin and rifampicin quinone is not
Rifampicin and Isoniazid Tablets less than 4, the tailing factor is not more than 2.0 and the
column efficiency is not less than 2000 theoretical plates.
Rifampin and Isonicotinylhydrazid Tablets Inject the test solution and reference solution (a). The relative
Rifampicin and Isoniazid Tablets contain not less than retention times are 1.0,0.55, 1.25, 1.51 and 2.61 for rifampicin,
90.0 per cent and not more than 110.0 per cent of the stated rifampicin quinone, rifampicin N-oxide,
amounts ofrifampicin, C43HssN4012 and isoniazid, C6H 7N 30. 3-formylrifamycin SV isonicotinyl hydrazone and
3-formylrifamycin SV respectively. Multiply the area ofeach
Usual strength. Rifampicin 50 mg and Isoniazid 150 mg.
known impurity by its response factor. The response factors
Identification are 1.00, 1.19, 1.03, 1.22 and 1.25 for rifampicin, rifampicin
quinone, rifampicin N-oxide, isonicotinyl hydrazone and
A. In the Assay, the chromatogram obtained with the test
3-formylrifamycin SV respectively.
solution shows peaks that correspond to the peaks due to
rifampicin RS and isoniazidRS in the chromatogram obtained In the chromatogram obtained with the test solution the area
with the reference solution. of any peak due to rifampicin quinone should not be more
than 4 times the area ofthe principal peak in the chromatogram
Tests obtained with the reference solution (4 per cent), the area of
Related substances. Determine by liquid chromatography any peak due to rifampicin N-oxide should not be more than
(2.4.14). 1.5 times the area of the principal peak in the chromatogram
obtained with the reference solution (1.5 per cent), the area of
NOTE - Prepare the solutions immediately before use.
any peaks due to 3-formylrifamycin SV and isonicotinyl
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent hydrazone should not be more than 5 times the area of the
w/v solution of citric acid, 23 volumes of a 13.61 per cent principal peak in the chromatogram obtained with the reference
w/v solution ofpotassium dihydrogen phosphate, 77 volumes solution (5 per cent) and the area of any peak due to
of a 17.42 per cent w/v solution of dipotassium hydrogen 3-formylrifamycin SV should not be more than the area ofthe

2059

www.webofpharma.com
RIFAMPICIN AND ISONIAZID TABLETS IP 2010

principal peak in the chromatogram obtained with the reference Assay. Determine by liquid chromatography (2.4.14).
solution (1.0 per cent). In the chromatogram obtained with the Solvent mixture. Dissolve 1.4 g of disodium hydrogen
test solution the area of any unknown peak should not be orthophosphate anhydrous in 1000 ml of water and adjust
more than 1.5 times the area of the principal peak in the the pH to 6.8 with dilute phosphoric acid.
chromatogram obtained with the reference solution (1.5 per
cent). Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity ofthe powder containing about 40 mg ofIsoniazid,
Dissolution (2.5.2). dissolve in 100.0 ml of methanol and dilute to 500.0 m1 with the
Apparatus No.2, solvent mixture.
Medium. 900 ml of 0.1 M hydrochloric acid, Reference solution. A solution containing 0.08 per cent w/v of
Speed and time. 100 rpm and 45 minutes. rifampicin RS and 0.04 per cent w/v of isoniazid RS in
Withdraw a suitable volume ofthe mediumand filter promptly methanol. Dilute 10.0 ml of this solution to 50.0 ml with the
through a membrane filter discwith an average pore diameter solvent mixture.
not greater than 0.8 Ilm. Reject the first few ml of the filtrate Chromatographic system
and dilute a suitable volume of the filtrate with the medium. a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm),
Reference stock solution. A solution containing 0.0165 per
column. temperature 30°,
cent of rifampicin RS and 0.00825 per cent of isoniazid RS in
mobile phase: A. a mixture of 96 volumes of buffer
the dissolution medium.
solution pH 6.8 prepared by dissolving 1.4 g of disodium
For rifampicin Measure the absorbance of the sample hydrogen orthophosphate anhydrous in 1000 ml of
solution and the reference stock solution suitably diluted with water and adjusting the pH to 6.8 ± 0.05 with dilute
the dissolution medium at 475 nm (2.4.7). Calculate the content phosphoric acid, and 4 volumes of acetronitrile.
ofC43HssN40J2 in the medium from the absorbance obtained B. a mixture of 45 volumes ofthe buffer
from the reference stock solution. solution and 55 volumes of acetonitrile,
For isoniazid - Determine by liquid chromatography flow rate. 1.5 m1 per minute,
(2.4.14). a linear gradient programme using the conditions given
. below,
Use the reference stock solution and sample solution suitably spectrophotometer set at 238 nm,
diluted with a solution of 0.05 .M potassium dihydrogen injection volume. 20 Ill.
orthophosphate, adjust the pH to 6.2 with 0;1 M sodium
Time Mobile phase A Mobile phase B
hydroxide to obtain the reference solution and the test
(in min.) (per cent v/v) (per cent v/v)
solution, respectively.
Chromatographic system
o 100 o
a stainless steel column 30 cm x 3.9 mm, packed with
5 100 o
octadecylsilane bonded to porous silica (5 Ilm), 6 0 100
mobile phase: a mixture of99 volumes of 0.05 Mpotassium 15 0 100
dihydrogen phosphate and 1 volume of acetonitrile 16 100 o
with the pH adjusted to 4.0 ± 0.05 with 2 per cent w/v 20 100 o
solution of phosphoric acid,
NOTE - Saturate the column with mobile phase B for about
flow rate. 1 ml per minute,
1 hour before injection.
spectrophotometer set at 254 nm,
injection volume. 20 Ill. Inject the reference solution. The tailing factor is not more
than 2.0 for rifampicin and isoniazid; the column efficiency for
Inject the reference solution. The test is not valid unless the
the isoniazid peak is not less than 3000 and for rifampicin not
tailing factor is not more than 2.0, the column efficiency innot
less than 25000 theoretical plates and the relative standard
less than ·1500 theoretical plates and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
deviation for replicate injections is not more than 2.0 per cent.
Inject alternately the test solution and the reference solution.
Inject the test solution and the reference solution.
The retention times are about 1.0 for rifampicin and about 0.3
Calculate the content ofC6H1N30 in the dissolution medium; for isoniazid.
D. Not less than 75 per cent of the stated amounts of Ca.lculaie the contents of C43HssN4012 and C 6H 7N 30 in the
C43HssN40J2 and C6H7N 30. tablets.
Other tests. Comply with the tests stated under Tablets. Storage. Store protected from moisture.

2060

www.webofpharma.com
IP 2010 RIFAMPICIN, ISONIAZIDN AND ETHAMBUTOL TABLETS

Rifampicin, Isoniazid and Ethambutol 0.19 per cent w/v of sodium perchlorate, 0.59 per cent
w/v of citric acid and 2.09 per cent w/v of potassium
Tablets dihydrogen phosphate and 35 volumes of acetonitrile,
Rifampin, Isonicotinylhydrazid and Ethambutol - flow rate. 1.5 ml per minute,
spectrophotometer set at 254 nm,
Hydrochloride Tablets
injection volume. 20 fll.
Rifampicin, Isoniazid and Ethambutol Tablets contain not less
Inject reference solution (b). The test is not valid unless the
than 90.0 per cent and not more than 110.0 per cent of the
resolution between rifampicin and rifampicin quinone is not
stated amounts ofrifampicin, C43HssN4012' isoniazid, C6H7N30
less than 4, the tailing factor is not more than 2.0 and the
and ethambutol hydrochloride CIQH24N 20 2.2HCl.
column efficiency is not less than 2000 theoretical plates.
Usual strength. Rifampicin 300 mg, Isoniazid 200 mg and
Inject the test solution and reference solution (a). The relative
Ethambutol 200 mg.
retention times are 1.0,0.55, 1.25, 1.51 and 2.61 forrifampicin,
Identification rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
Isonicotinyl hydrazone and 3-formyhifamycin SV respectively.
A. In the Assay for rifampicin and isoniazid, the principal Multiply the areas of each known impurity by its response
peaks in the chromatogram obtained with the test solution factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
correspond to the peaks in the chromatogram obtained with for rifampicin, rifampicin quinone, rifampicin N-oxide,
the reference solution. Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
B. In the Assay for ethambutol hydrochloride the principal In the chromatogram obtained with the test solution the area
peak in the chromatogram obtained with the test solution of any peak due to rifampicin quinone should not be more
corresponds to the peak in the chromatogram obtained with than 4 times the area ofthe principal peak in the chromatogram
the reference solution. obtained with the reference solution (4 per cent), the area of
Tests any peak due to rifampicin N-oxide should not be more than
1.5 times the area of the principal peak in the chromatogram
Related substances. Determine by liquid chromatography obtained with the reference solution (1.5 per cent), the area of
(2.4.14). any peak due to 3-formylrifamycin SV Isonicotinyl hydrazone
NOTE - Prepare the solutions immediately before use. should not be more than 5 times the area ofthe principal peak
in the chromatogram obtained with the reference solution
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent
( 5 per cent) and the area ofany peak due to 3-formylrifamycin
w/v solution of citric acid, 23 volumes of a 13.61 per cent
SV should not be more than the area ofthe principal peak in
w/v solution ofpotassium dihydrogen phosphate, 77 volumes
the chromatogram obtained with the reference solution (I per
of a 17.42 per cent w/v solution of dipotassium hydrogen
cent ). In the chromatogram obtained with the test solution
phosphate, 640 volumes of water and 250 volumes of
the area of any unknown peak should not be more than
acetonitrile.
1.5 times the area ofthe principal peak in the chromatogram
Test solution. Weigh accurately a quantity of the powdered obtained with the reference solution (1.5 per cent).
tablets containing 200 mg ofRifampicin, dissolve in 100 ml of
Dissolution (2.5.2).
acetonitrile and filter. Dilute 5 ml ofthis solution to 50 ml with
the solvent mixture. Apparatus No.2,
Medium. 900 ml of 0.1 M hydrochloric acid,
Reference solution (a). A solution containing 0.02 per cent
Speed and time. 100 rpm and 45 minutes.
w/v of rifampicin RS in acetonitrile. Dilute 1 ml ofthis solution
to 100 ml with the solvent mixture. Withdraw a suitable volume ofthe medium and filter promptly
through a membrane filter disc with an average pore diameter
Reference solution (b). A solution containing 0.01 per cent
not greater than 0.8 flm. Reject the first few ml ofthe filtrate.
w/v each of rifampicin RS and rifampicin quinone RS in
acetonitrile. Dilute 5 ml of this solution to' 50 ml with the Reference stock solution. A solution containing 0.0165 per
solvent mixture. cent w/v of rifampicin RS, 0.00825 per cent w/v of isoniazid
Chromatographic system RS and 0.031 per cent w/v of ethambutol hydrochloride RS in
a stainless steel column 10 cm x 4.6 mm, packed with the dissolution medium. Keep this reference stock solution in
octylsilane bonded to porous silica (5 flm), the dissolution bath during the test run.
column temperature. 30°, For rifampicin - Dilute the filtrate, if necessary, with the
mobile phase: a mixture of 65 volumes of a solution same solvent. Measure the absorbance (2.4.7) ofthe resulting
containing 0.1 per cent v/v of orthophosphoric acid, solution at the maximum at about 475 nm. Calculate the content

2061

www.webofpharma.com
RIFAMPICIN, ISONIAZID AND ETHAMBUTOL TABLETS IP 2010

ofC43H58N40lz in the medium from the absorbance obtained Reference solution. A solution containing 0.08 per cent w/v of
from suitably diluted reference stock solution. rifampicin RS and 0.04 per cent w/v of isoniazid RS in
For isoniazid- Determine by liquid chromatography (2.4.14). methanol. Dilute 10.0 ml of this solution to 50.0 ml with the
diluent.
Test solution. Suitably dilute the filtered medium with 0.05 M
Chromatographic system
potassium dihydrogen orthophosphate with the pH adjusted
to 6.2 with 0.1 M sodium hydroxide. - a stainless steel column 25 cm x 4.6 mm, packed with
octadecy1si1ane bonded to porous silica (5 11m),
Reference solution. Suitably dilute the reference stock solution -column temperature. 30°,
with 0.05 M potassium dihydrogen orthophosphate with the mobile phase: A. a mixture of 96 volumes of buffer
pH adjusted to 6.2 with 0.1 M sodium hydroxide. solution pH 6.8 (diluent) prepared by dissolving 1.4 g of
Chromatographic system disodium hydrogen orthophosphate anhydrous in
a stainless steel column 30 cm x 4.6 rom, packed with 1000 ml of water, and adjusting the pH to 6.8 ± 0.05 with
octadecylsilane bonded to porous silica (5 11m), dilute phosphoroic acid, and 4 volumes of acetonitrile,
- mobile phase: a mixture of99 volumes of0.05 M potassium B: a mixture of 45 volumes of the buffer
dihydrogen phosphate and 1 volume of acetonitrile solution and 55 volumes of acetonitrile,
previously adjusted to pH 4.0 with a 2 per cent w/v flow rate. 1.5 ml per minute,
solution of phosphoric acid, a linear gradient programme using the conditions given
flow rate. 1 ml per minute, below,
spectrophotometer set at 254 nm, spectrophotometer set at 238 nm,
injection volume. 20 Ill. injection volume. 20 Ill.
Inject the reference solution. The test is not valid unless the Time Mobile phase A Mobile phase B
tailing factor is not more than 2.0, the column efficiency in not (in min.) (per cent v/v) (per cent v/v)
less than 1500 theoretical plates and the relative standard o 100 o
deviation for replicate injections is not more than 2.0 per cent.
5 100 o
Inject the test solution and the reference solution.
6 0 100
Calculate the content ofC 6H 7N 30 in the dissolution medium. 15 0 100
For ethambutol hydrochloride - Determine by liquid 16 100 o
chromatography (2.4.14).
22 100 o
Test solution. Suitably dilute the filtered medium with 0.05 M
NOTE - Saturate the column with mobile phase B for about
potassium dihydrogen orthophosphate adjusted to pH 6.2
1 hour.
with 0.1 M sodium hydroxide.
Inject the reference solution. The test is not valid unless the
Reference solution. Suitably dilute the reference stock solution
tailing factor is not less than 2.0 for rifampicin and isoniazid,
with 0.05 M potassium dihydrogen orthophosphate with the
the column efficiency determined from isoniazid peak is not
pH adjusted to 6.2 with 0.1 M sodium hydroxide.
less than 3000 and that from rifampicin is not less than 25000
Use the chromatographic system described under the Assay theoretical plates respectively, and the relative standard
of ethambutol hydrochloride. deviation for replicate injections is not more than 2.0 per cent.
Calculate the content of CIOHz4NzOz.2HCl in the dissolution The relative retention times are about 1.0 for rifampicin and
medium. about 0.3 for isoniazid.

D. Not less than 75 per cent of the stated amounts of Calculate the contents of C43H58N40lZ and C 6H 7N30 in the
C43H58N401Z, C6H 7N 30 and CIOHz4NzOz.2HCl. tablets.
Other tests. Comply with the tests stated under Tablets. For ethambutol hydrpchloride Determine by liquid
chromatography (2.4.14).
Assay. For rifampicin and isoniazid - Determine by liquid
chromatography (2.4.14). Test solution. Weigh accurately a quantity of the powdered
tablets containing about 60mg of Ethambutol Hydrochloride
Test solution. Weigh and powder 20 tablets. Weigh accurately
and dissolve in 100.0 ml ofthe diluent.
a quantity ofthe powdered tablets containing about 40 mg of
Isoniazid dissolve in 100.0 ml of methanol, dilute to 500.0 ml Reference solution. A 0.06 per cent w/v solution of ethambutol
with the diluent and mix. hydrochloride RS in the diluent.

2062

www.webofpharma.com
IP 2010 RIFAMPICIN, ISONIAZID AND PYRAZINAMIDE TABLETS

Chromatographic system Test solution. Weigh accurately a quantity of the powdered


- a stainless steel column 15 cm x 4.6 mm, packed with tablets containing about 200 mg of Rifampicin, dissolve in
nitrile groups chemically bonded to porous silica 100 ml of acetonitrile and filter. Dilute 5 ml ofthis solution to
particles 5 (/-lm) (such as Zorbax SB CN), 50 ml with the solvent mixture.
mobile phase: a mixture of 50 volumes of acetonitrile,
Reference solution (a). A solution containing 0.02 per cent
and 50 volumes ofa buffer solution pH 7.0 prepared by
w/v of rifampicin RS in acetonitrile. Dilute Iml ofthis solution
dissolving 1 ml of triethylamine in 1000 ml of water and
to a 100 ml with the solvent mixture.
adjusting the pH to 7.0 with dilute phosphoric acid,
flow rate. 1 ml per minute, , Reference solution (b). A solution containing 0.01 per cent
spectrophotometer set at 200 urn, w/v each of rifampicin RS and rifampicin qUinone RS in
injection volume. 20 /-ll. acetonitrile. Dilute 5 ml of this solution to 50 ml with the
solvent mixture.
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more Chromatographic system
than 2.0 per cent, the tailing factor is not more than 3.0 and the - a stainless steel column 10 cm x 4.6 mm, packed with
column efficiency determined from Ethambutol hydrochloride octylsilane bonded to porous silica (5 /-lm),
peak is not less than 1500 theoretical plates. column temperature. 30°,
Inject the test solution and the reference solution. mobile phase: a mixture of 65 volumes of a solution
containing 0.1 per cent v/v of orthophosphoric acid,
Calculate the content ofCIOH24N202.2HCl in the tablets. 0.19 per cent w/v of sodium perchlorate, 0.59 per cent
Storage. Store protected from moisture. w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate, 35 volumes of acetonitrile,
flow rate. 1.5 ml per minute,
spectrophotometer set at 254 nm,
Rifampicin, Isoniazid and - injection volume. 20 /-ll.
Pyrazinamide Tablets Inject reference solution (b). The test is not valid unless the
resolution between rifampicin and rifampicin quinone is not
Rifampin, Isonicotinylhydrazid and Pyrazinamide Tablets
less than 4, the tailing factor is not more than 2.0 and the
Rifampicin, .Isoniazid and Pyrazinamide Tablets contain not column efficiency is not less than 2000 theoretical plates;
less than 90.0 per cent and not more than 110.0 per cent ofthe
Inject the test solution and reference solution (a). The relative
stated amounts ofrifampicin, C43HssN4012' isoniazid, C6 H7N30
and pyrazinamide CsHsN30. retention times are 1.0,0.55, 1.25, 1.51 and 2.61 for rifampicin,
rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
Usual strength. Rifampicin 120 mg, Isoniazid 50 mg and Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
Pyrazinamide 300 mg. Multiply the area of each known impurity by its response
factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
Identification for rifampicin, rifampicin quinone, rifampicin N-oxide,
Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
In the Assay of rifampicin, isoniazid and pyrazinamide, the
principal peaks in the chromatogram obtained with the test In the chromatogram obtained with the test solution the area
solution correspond to the peaks in the chromatogram of any peak due to rifampicin quinone should not be more
obtained with the reference solution. than 4 times the area ofthe principal peak in the chromatogram
obtained with the reference solution (4 per cent), the area of
Tests any peak due to rifampicin N-oxide should not be more than
Related substances. Determine by liquid chromatography 1.5 times the area of the principal peak in the chromatogram
(2.4.14). obtained with the reference solution (1.5 per cent), the area of
any peak due to 3-formylrifamycin SV Isonicotinyl hydrazone
NOTE - Prepare the solutions immediately before use.
should not be more than 5 times the area ofthe principal peak
Solvent mixture. A mixture of 10 volumes ofa 21.01 per cent in the chromatogram obtained with the reference solution
w/v solution of citric acid, 23 volumes of a 13.61 per cent (5 per cent) and the area ofany peak due to 3-formylrifamycin
w/v solution ofpotassium dihydrogen phosphate, 77 volumes SV should not be more than the area of the principal peak in
of 17.42 per cent w/v solution of dipotassium hydrogen the chromatogram obtained with the reference solution ( 1 per
phosphate, 640 volumes of water and 250 volume,S of cent ). In the chromatogram obtained with the test solution
acetonitrile. the area of any unknown peak should not be more than

2063

www.webofpharma.com
RIFAMPICIN, ISONIAZID AND PYRAZINAMIDE TABLETS IP 2010

1.5 times the area of the principal peak in the chromatogram The test is not valid unless in the chromatogram obtained
obtained with the reference solution (1.5 per cent). with reference solution (b) the resolution between isonicotinic
acid and pyrazinamide is not more than 2.5 and between
Dissolution (2.5.2).
pyrazinamide and isoniazid is not more than 4.0.
Apparatus No.2, Calculate the contents ofC6H7N30 and CsHsN 30 in the medium.
Mediuni. 900 nil ofa solution containing 2 g ofsodium chloride
and7.0 ml of hydf"Ochloric acid in 1000 illl of water, with a pH D. Not less than 75 per cent of the stated amounts of
of about 1.2, C43HssN4012, C6H7N30 and CsHsN30.
Speed and time. 100 rpm and 45 minutes. Other tests. Comply with the tests stated under Tablets.

Withdraw a suitable volume ofthe medium and filter through Assay. For rifampicin, isoniazid and pyrazinamide -
a membrane filter disc with an average pore diameter not greater Detennine by liquid chromatography (2.4.14).
than 0.8 flm. Reject the first few ml ofthe filtrate. Test solution. Weigh and powder20 tablets. Weigh accurately
a quantity of the powdered tablets containing about 40 mg of
Reference stock solution. A solution containing 0.0165 per
Isoniazid, dissolve in 100.0 ml of methanol, dilute to 500 ml
cent w/v of rifampicin RS, 0.00825 per cent w/v of isoniazid
with the buffer solution pH 6.8 and mix.
RS and 0.04375 per cent w/v of pyrazinamide RS in the
dissolution medium and filtered. Keep this reference stock Reference solution. A solution containing 0.08 per cent w/v of
solution in the dissolution bath during the test run. rifampicin RS, 0.04 per cent w/v of isoniazid RS and 0.2 per
cent w/v of pyrazinamide RS in methanol. Dilute 10.0 ml of
For rifampicin ~ Dilute the filtrate, if necessary, with the .this solution to 50.0 ml with the buffer solution pH 6.8.
same solvent. Measure the absorbance (2.4.7) ofthe resulting
Chromatographic system
solution atthe maximum at about 475 urn. Calculate the content
a stainless steel column 25 cm x 4.6 mm, packed with
ofC43HssN40,2 in the medium from the absorbance obtained
octadecylsilane bonded to porous silica (5 flm),
from suitably diluted reference stock solution.
column. temperature 30°,
For isoniazid and pyrazinamide Determine by liquid mobile phase: A. a mixture of 96 volumes of buffer
chromatography (2.4.14). solution pH 6.8 (diluent) prepared by dissolving 1.4 g of
dis odium hydrogen orthophosphate anhydrous in
NOTE - Use this solution within one hourfrom preparation.
1000 ml of water, adjusted to pH 6.8 ± 0.05 with dilute
Testsolution. To 15.0 ml ofthe filtered medium, add 15 ml of phosphoroic acid and 4 volumes of acetonitrile,
1 M dibasic potassium phosphate, dilute to 100.0 ml with the B. a mixture of 45 volumes ofthe buffer
mobile phase and mix. solution and 55 volumes of acetonitrile,
flow rate. 1.5 ml per minute,
Reference solution (a). To 15.0 ml of the reference stock
a linear gradient programme using the conditions given
solution, add 15 ml of 1 M dibasic potassium phosphate,
below,
dilute to 100.0 ml with the mobile phase and mix.
spectrophotometer set at 238 nm,
Reference solution (b). To 10.0 ml of a 0.0125 per cent w/v injection volume. 20 fll.
solution of isonicotinic acid in the dissolution medium, add Time Mobile phase A Mobile phase B
4.0 ml ofthe reference stock solution and 15 ml of 1 M dibasic (in min.) (per cent v/v) (per cent v/v)
potassium phosphate, dilute to 100.0 ml with the mobile phase o 100 o
and mix. 5 100 o
Chromatographic system 6 0 100
a stainless steel column 30 cm x 4.6 mm, packed with 15 0 100
octylsilane bonded to porous silica (5 flm), 16 100 o
mobile phase: a mixture of 860 volumes of water, 22 100 o
100 volumes of 1 M monobasic potassium phosphate NOTE - Saturate the column with mobile phase (B) for
and 40 volumes of acetonitrile, about 1 hour.
flow rate. 1ml per minute,
Inject the test solution and the reference solution. The test is
- spectrophotometer set at 254 nm,
not valid unless the tailing factor is not less than 2.0 for
injection volume. 50 fll.
rifampicin, isoniazid and pyrazinamide, the column efficiencies
Inject the test solution and reference solutions (a) and (b). determined from Isoniazid, pyrazinamide and that from
The relative retention times are about 0.7 for isonicotinic acid, rifampicin are not less than 3000, 5000 and 25000 theoretical
1.0 forpyrazinainide and 1.8 for iSoniazid. plates respectively, and the relative standard deviation for

2064

www.webofpharma.com
IP 2010 RIFAMPICIN, ISONIAZID, PYRAZINAMIDE AND ETHAMBUTOL TABLETS

replicate injections is not more than 2.0 per cent. The relative Reference solution (b). A solution containing 0.01 per cent
retention times are about 1.8 for rifampicin, about 0.7 for w/v each of rifampicin RS and rifampicin quinone RS in
isoniazid and about 1.0 for pyrazinamide. acetonitrile. Dilute 5 ml of this solution to 50 ml with the
Inject the test solution and the reference solution. solvent mixture.

Calculate the contents ofC43HssN4012, C6H7N30 and CsH sN 30 Chromatographic system


in the tablets. a stainless steel column 10 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 llm),
Storage. Store protected from moisture. column temperature. 30°,
- mobile phase: a mixture of 65 volumes of a solution
containing 0.1 per cent v/v of orthophosphoric acid,
Rifampicin, Isoniazid, Pyrazinamide 0.19 per cent w/v of sodium perchlorate, 0.59 per cent
and Ethambutol Tablets w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate, 35 volumes of acetonitrile,
Rifampin, Isonicotinylhydrazid, Pyrazinamide and - flow rate. 1.5 ml per minute,
Ethambutol Hydrochloride Tablets spectrophotometer set at 254 urn,
injection volume. 20 lll.
Rifampicin, Isoniazid, Pyrazinamide and Ethambutol Tablets
contain not less than 90.0 per cent and not more than 110.0 per Inject reference solution (b). The test is not valid unless the
cent ofthe stated amounts ofrifampicin, C43HssN4012' isoniazid, resolution between rifampicin and rifampicin quinone is not
C 6H 7N 30, pyrazinamide, C sH sN 30 and ethambutol less than 4, the tailing factor is not more than 2.0 and the
hydrochloride, CIOH24N202.2HCl. column efficiency is not less than.2000 theoretical plates.
Usual strength. Rifampicin 150 mg, Isoniazid 75 mg, Inject the test solution and reference solution (a). The relative
Pyrazinamide 400 mg and Ethambutol 275 mg. retention times are 1.0,0.55, 1.25, 1.51 and 2.61 for rifampicin,
rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
Identification Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
A. In the Assay for rifampicin, isoniazid and pyrazinamide, the Multiply the area of each known impurity by its response
principal peaks in the chromatogram obtained with the test factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
solution correspond to the peaks in the chromatogram for rifampicin, rifampicin quinone, rifampicin N-·oxide,
obtained with the reference solution. Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.

B. In the Assay for ethambutol hydrochloride, the principal In the chromatogram obtained with the test solution the area
peak in the chromatogram obtained with the test solution of any peak due to rifampicin quinone should not be more
corresponds to. the peak in the chromatogram obtained with than 4 times the area ofthe principal peak in the chromatogram
the reference solution. obtained with the reference solution (4 per cent), the area of
any peak due to rifampicin N-oxide should not be more than
Tests 1.5 times the area of the principal peak in the chromatogram
obtained with the reference solution (1.5 per cent), the area of
. Related substances. Determine by liquid chromatography
any peak due to 3-fonnylrifamycin SV Isonicotinyl hydrazone
(2.4.14).
should not be more than 5 times the area ofthe principal peak
NOTE - Prepare the solutions immediately before use. in the chromatogram obtained with the reference solution
Solvent mixture. A mixture of 10 volumes ofa 21.01 per cent ( 5 per cent) and the area ofany peak due to 3-formylrifamycin
w/v solution of citric acid, 23 volumes of a 13.61 per cent SV should not be more than the area ofthe principal peak in
w/v solution ofpotassium dihydrogen phosphate, 77 volumes the chromatogram obtained with the reference solution (l per
of a 17.42 per cent w/v solution of dipotassium hydrogen cent ). In the chromatogram obtained with the test solution
phosphate, 640 volumes of water and 250 volumes of the area of any unknown peak should not be more than
acetonitrile. 1.5 times the area of the principal peak in the chromatogram
Test solution. Weigh accurately a quantity ofpowdered tablets obtained with the reference solution (1.5 per cent).
containing 200 mg of Rifampicin, dissolve in 100 ml of Dissolution (2.5.2).
acetonitrile and filter. Dilute 5 ml ofthis solution to 50 ml with Apparatus No.1,
the solvent mixture. Medium. 900 ml of sodium phosphate bufferpH 6.8 prepared
Reference solution (a). Dissolve 20 mg of rifampicin RS in by dissolving 7 g of dibasic sodium phosphate anhydrous in
100 ml of acetonitrile. Dilute 1 ml ofthis solution to a 100 ml 5000 ml of water and adjusting to pH 6.8 with phosphoric
with the solvent mixture. acid,

2065

www.webofpharma.com
RIFAMPICIN, ISONIAZID, PYRAZINAMIDE AND ETHAMBUTOL TABLETS IP 2010

Speed and time. 100 rpm and 45 minutes. efficiencies for isoniazid, pyrazinamide and rifampicin are not
Withdraw a suitable volume ofthe medium and filter through less than 3000, 5000 and 25000 theoretical plates respectively.
a membrane filter disc with an average pore diameter not greater Inject the test solution and the reference solution. The relative
than 0.8Ilm. Reject the first few ml ofthe filtrate. Determine the retention time for rifampicin is 1.8, for isoniazid, 0.7 and for
amounts of rifampicin, C43Hs8N401Z, isoniazid, C 6H 7N 30, pyrazinamide, 1.0.
pyrazinamide, CsHsNP and ethambutol hydrochloride, Calculate the contents OfC43Hs8N401Z, C6H 7N 30 and CsH sN 30
CIQHz4NzOz.2HCl by using the methods described in the Assay in the tablets.
for rifampicin, isoniazid, pyrazinamide and ethambutol
hydrochloride. For ethambutol hydrochloride - Determine by liquid
chromatography (2.4.14).
D. Not less than 75 per cent of the stated amounts of
C43Hs8N401Z, C6H7N 30, CsH sN 30 and CIQHZ4NzOz.2HCI. Test solution. Weigh accurately a quantity of the powdered
tablets containing about 60 mg of Ethambutol Hydrochloride
Other tests. Comply with the tests stated under Tablets. and dissolve in 100 ml ofthe solvent mixture.
Assay. For rifampicin, isoniazid and pyrazinamide - Reference solution. A 0.06 per cent w/v solution of ethambutol
Determine by liquid chromatography (2.4.14).
hydrochloride RS in the solvent mixture.
Test solution. Weigh and powder 20 tablets. Weigh accurately
Chromatographic system
a quantity ofthe powdered tablets containing about 40 mg of
- a stainless steel column 15 cm x 4.6 mm, packed with
Isoniazid, dissolve in 100 ml of methanol, dilute to 500 ml with
nitrile groups chemically bonded to porous silica
the buffer solution pH 6.8, mix and filter.
particles (5Ilm),
Reference solution. A solution containing 0.08 per cent w/v of - mobile phase: a mixture of 50 volumes of acetonitrile,
rifampicin RS, 0.04 per cent w/v of isoniazid RS and 0.2 per and 50 volumes of buffer solution pH 7.0 (diluent)
cent w/v of pyrazinamide RS in methanol. Dilute 10.0 ml of prepared by dissolving 1 ml of triethylamine in 1000 ml
this solution to 50.0 ml with the buffer solution pH 6.8. of water and adjusting the pH to 7.0 with dilute
Chromatographic system phosphoric acid,
a stainless steel column 25 cm X 4.6 mm, packed with - flow rate. I ml per minute,
octadecylsilane bonded to porous silica (5 Ilm), - spectrophotometer set at 200 nm,
- column. temperature 30°, - injection volume. 50 Ill..
- mobile phase: A. a mixture of 96 volumes of buffer Inject the reference solution. The test is not valid unless the
solution pH 6.8 prepared by dissolving 1.4 g of disodium relative standard deviation for replicate injections is not more
hydrogen orthophosphate anhydrous in 1000 ml of than 2.0 per cent, the tailing fac:tor is not more than 3.0 and the
water, adjusting to pH 6.8 ± 0.05 with dilute phosphoric column efficiencydeterminedfrom ethambutol hydrochloride
acid and 4 volumes of acetonitrile, peak is not less than 1500 theoretical plates.
B. a mixture of 45 volumes ofthe buffer
solution and 55 volumes of acetonitrile, Calculate the content ofCIQHz4NzOz.2HCl in the tablets.
- flow rate. 1.5 ml per minute, Storage. Store protected from moisture.
- a linear gradient programme using the conditions given
below,
- spectrophotometer set at 238 nm,
- injection volume. 20 Ill.
Ritonavir
Time Solution A SolutionB
(in min.) (per cent v/v) (per cent v/v)
o 100 o
5 100 o
6 0 100
15 0 100
16 100 o
22 100 o
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
than 2.0 per cent, the tailing factor is not more than 2.0 for
rifampicin, isoniazid and pyrazinamide and the column Mol. Wt. 721.0

2066

www.webofpharma.com
IP 2010 RlTONAVIR CAPSULES

Ritonavir is (5S,8S,lOS,IIS)-1 0-hydroxy-2-methyl- dissolving 3.4 g ofsodium acetate and 0.94 g ofsodium
5-(1-methylethyl)-I-[2-(1-methylethyl)-4-thiazolyl]-3,6-dioxo- hexanesulphonate in 1000 ml of water and adjusting
8,11-bis(phenylmethyl)-2,4,7, 12-tetraazatridecan-13-oic acid the pH to 4.0 with hydrochloric acid,
5-thiazolylmethyl ester. - flow rate. I ml per minute,
- spectrophotometer set at 239 urn,
Ritonavir contains not less than 98.0 per cent and not more
- injection volume. 20 Ill.
than 102.0 per cent of C37H48N60SS2, calculated on the
anhydrous basis. Inject the reference solution. The test is not valid unless the
column efficiency determined from the ritonavir peak is not
Category. Antiretroviral.
less than 1000 theoretical plates, the tailing factor is not more
Dose. 100 mg twice daily. than 2.0, and the relative standard deviation for replicate
Description. A white to off-white powder. injections is not more than 2.0 per cent.
Inject the test solution and the reference solution.
Identification
Calculate the content ofC37H48N60sS2.
A. Determine by infrared absorption spectrophotometrY (2.4.6).
Storage. Store protected from light.
Compare the spectrum with that obtained with ritonavir RS or
with the reference spectrum of ritonavir.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak due Ritonavir Capsules
to ritonavir in the chromatogram obtained with the reference Ritonavir Capsules contain not less than 90.0 per cent and not
solution. more than 110.0 per cent of the stated amount of ritonavir,
C.Meltingpoint.119°to 123°(2.4.21). C37~8N60SS2'
Usual strength.· 100 mg.
Tests
Specific optical rotation (2.4.22). +7.0° to + 10.5°, determined
Identification
in a 0.2 per cent w/v solution in methanol. A. In the test for Assay, the principal spot in the chromatogram
Related substances. Determine by liquid chromatography obtained with the test solution corresponds to that in the
(2.4.14), as described in the Assay and calculate the percentage chromatogram obtained with the reference solution.
ofeach impurity peak in the chromatogram obtained with the B. When examined in the range 200 urn to 350 urn (2.4.7), a
test solution. Not more than 0.5 per cent of any individual 0.001 per cent w/v solution in methanol shows absorption
impurity and not more than 1.0 per cent oftotal impurities is maxima at the same wavelengths as the reference solution.
found.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Tests
heavy metals, Method B (20 ppm). Dissolution (2.5.2).
Sulphated ash (2.3.18). Not more than 0.1 per cent Apparatus No.1,
Water (2.3.43). Not more than 0.5 per cent, determined on 2.0 g. Medium. 900 ml of0.1 M hydrochloric acidwith 25 roM poly-
oxyethylene 10 lauryl ether prepared by dissolving
Assay. Determine by liquid chromatography (2.4.14). 15.65 g ofpolyoxyethylene 10 lauryl ether in 950 ml ofwater.
Test solution. Dissolve 25.0 mg of the substance under Add 8.5 ml of hydrochloric acid and dilute to 1000 ml with
examination in sufficient of a mixture of 45 volumes of water,
acetonitrile and 55 volumes of water to produce 100.0 ml. Speed and time. 50 rpm and 90 minutes.
Reference solution. A 0.025 per cent w/v solution ofritonavir Withdraw a suitable volume ofthe medium and filter.
RS in a mixture of45 volumes ofacetonitrile and 55 volumes
Deterrriine by liquid chromatography (2.4.14).
ofwater.
Test solution. Use the filtrate and if necessary, dilute with the
Chromatographic system
dissolution medium.
- a stainless steel column 15 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 /lm), Reference solution. A 0.1 per cent w/v solution of ritonavir
- mobile phase: a mixture of 45 volumes of acetonitrile RS in methanol. Dilute 5 ml of the solution to 50 ml with the
and 55 volumes of a buffer solution prepared by dissolution medium.

2067

www.webofpharma.com
RITONAVIR CAPSULES IP 2010

Use the chromatographic system described under Assay. chromatogram obtained with the reference solution (b)
Inject the test solution and the reference solution. (5.0 per cent).

D. Not less than 70 per cent of the stated amount of Other tests. Comply with the tests stated under Capsules.
C37HtsN60sS2. Assay. Determine by liquid chromatography (2.4.14).
Related substances. Determine by liquid chromatography Test solution. Weigh accurately a quantity of the mixed
(2.4.14). contents of20 capsules containing about 25 mg ofRitonavir,
Solvent mixture. 40 volumes of 0.03M monobasic potassium disperse in 100.0 ml of methanol and filter.
phosphate buffer and 60 volumes of acetonitrile. Reference solution. A 0.025 per cent w/v solution ofritonavir
Test solution. Weigh accurately a quantity of the mixed RS in methanol.
contents of20 capsules containing about 25 mg of Ritonavir, Chromatographic system
dissolve in 50 ml ofthe solvent mixture and filter. a stainless steel column 5 cm x 4.6 rom, packed with
Reference solution (a). A 0.05 per cent w/v solution of octadecylsilane bonded to porous silica (3.5 /lm),
ritonavir RS in the solvent mixture. column temperature 45°,
mobile phase: a mixture of 55 volumes ofa buffer solution
Reference solution (b). Dilute 1 ml of the solution to 100 ml prepared by dissolving 3.4 g of sodium acetate
with the solvent mixture.
trihydrate and 0.94 g of sodium l-hexanesulphonate in
Chromatographic system 1000 ml of water and adjusting the pH to 4.0 with
a stainless steel column 15 cm x 4.6 rom, packed with hydrochloric acid, and 45 volumes of acetonitrile,
butyl silane chemically bonded to porous silica (3 /lm) - flow rate. 2.5 ml per minute,
(Such as ACE C4), - spectrophotometer set at 240 urn,
- mobile phase: A. a mixture of69 volumes of0.03 M mono- - injection volume. 10 Ill.
basic potassium phosphate buffer prepared by
Inject the reference solution. Run the chromatogram 1.5 times
dissolving 4.1 g of monobasic potassium phosphate
the retention time (about 3 minutes)ofthe principal peak The
in 1000 ml of water, 18 volumes ofacetonitrile, 8 volumes
test is not valid unless the tailing factor is not more than 2.0,
of tetrahydrofuranand 5 volumes of n-butanol,
the column efficiency in not less than 2000 theoretical plates
B. a mixture of40 volumes of0.03 M mono-
and the relative standard deviation for replicate injections is
basic potassium phosphate buffer, 47 volumes of
not more than 2.0 per cent.
acetonitrile, 8 volumes of tetrahydrojitran and 5 volumes
of n-butanol, Inject the test solution and the reference solution.
flow rate. 1 ml per minute, Calculate the content of C37H4sN60sS2 in the capsules.
a linear gradient programme using the conditions given
below, Storage. Store protected from light in a refrigerator (2° to 8°).
spectrophotometer set at 240 nm,
injection volume. 50 Ill.
Time mobile phase A mobile phase B
(in min.) (per cent v/v) (per cent v/v) Ritonavir Tablets
o 100 o Ritonavir Tablets contain not less than 90.0 per cent and not
60 100 o more than 110.0 per cent of the stated amount of ritonavir,
C37HtsN60sS2.
120 0 100
155 100 o Usual strength. 100 mg.

Inject the reference solution (a). The test is not valid unless Identification
the tailing factor is not more than 2.0 arid the column efficiency
in not less than 5000 theoretical plates. In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the principal peak in the
. Inject the test solution and reference solution (b). In the
chromatogram obtained with the reference solution.
chromatogram obtained with the test solutiQn, the area ofany
secondary peak is not more than 2.5 times the area ofthe peak Tests
in the chromatogram obtained with the reference solution (b)
(2.5 per cent) and the sum of areas of all the secondary peaks Dissolution (2.5.2)
is not more than 5 times the area of the peak in the Apparatus No.1,

2068

www.webofpharma.com
IP 2010 RITONAVIR TABLETS

Medium. 900 ml ofa solution prepared by dissolving 15.7 g of - injection volume. 100 f..11.
polyoxyethylene 1O-lauryl ether in 1000 ml of a 0.85 per cent Time Mobile phase A Mobile phase B
v/v solution of hydrochloric acid, (in min.) (per cent v/v) (per cent v/v)
Speed and time. 100 rpm and 60 minutes.
o 100 o
Withdraw a suitable volume ofthe medium and filter.
60 100 o
Determine by liquid chromatography (2.4.14) 120 0 100
Test solution. The filtrate obtained as given.above. Dilute the 121 100 o
filtrate if necessary, with the dissolution medium.
155 100 o
Reference solution. A 0.1 per cent w/v solution of ritonavir
Inject reference solution (a). The test is not valid unless the
RS in methanol. Dilute 5 ml ofthe solution to 50 ml with the
column efficiency is not less than 2000 theoretical plates and
dissolution medium.
the tailing factor is not more than 2.0.
Use the chromatographic system described under Assay.
Inject the test solution and reference solution (b). In the
Inject the reference solution. The test is not valid unless the chromatogram obtained with the test solution, the area ofany
tailing factor is not more than 2.0 and the relative standard secondary peak is not more than 2.5 times the area ofthe peak
deviation ofreplicate injections is not more than 2.0 per cent. in the chromatogram obtained with the reference solution (b)
Inject the test solution and the reference solution. (2.5 per cent) and the sum of areas of all the secondary peaks
is not more than 5 times the area of the peak in the
Calculate the content of C37H48N60sSz. chromatogram obtained with the reference solution (b)
D. Not less than 75 per cent of the stated amount of (5.0 percent).
C37~8N60SSZ. Other tests. Comply with the tests stated under Tablets.
Related substances. Determine by liquid chromatography Assay. Determine by liquid chromatography(2.4.14)
(2.4.14).
Test solution. Weigh accurately a quantity of the powdered
Solvent mixture. A mixture of40 volumes of 0.03 Mmonobasic tablets containing 25 mg ofRitonavir and disperse in 100.0 ml
potassium phosphate and 60 volumes of acetonitrile. of methanol.
Test solution: Shake a quantity of the powdered tablets Reference solution. A 0.025 per cent w/v solution of ritonavir
containing 25 mg ofRitonavir with 50 m1 ofthe solvent mixture. RS in methanol.
RefereYice solution (a). A 0.05 per cent w/v solution of
Chromatographic system
ritonavir RS in the solvent mixture. a stainless steel column 5 cm x 4.6 mm, packed with
Reference solution (b). Dilute 1 ml ofreference solution (a) to phenyl stationary phase bonded to porous silica (3 f..1m),
100 ml with the solvent mixture. - column temperature 45°,
mobile phase: a mixture of55 volumes ofa buffer solution
Chromatographic system
prepared by dissolving 3.4 g of sodium acetate
a stainless steel column 15 cm x 4.6 rom, packed with
trihydrate and 0.94 g of sodium l-hexanesulphonate
silica gel consisting of porous spherical particles with
to 1000 ml with water and adjusting the pH to 4.0 with
chemically bonded with nitrile group (3f..1m) (such as
dilute hydrochloric acid, and 45 volumes of
YMCC4),
acetonitrile.
mobile phase: A. a mixture of69 volumes of 0.03 M mono-
flow rate. 2.5 ml per minute,
basic potassium phosphate solution, 18 volumes of
spectrophotometerset at 239 nm,
acetonitrile, 8 volumes of tetrahydrofuran and
injection volume. 10 f..11.
5 volumes of n- butanol,
B. a mixture of40 volumes of 0.03 M mono- Inject the reference solution. The test is not valid unless the
basic potassium phosphate solution, 47 volumes of relative standard deviation for replicate injections is not more
acetonitrile, 8 volumes of tetrahydrofuran and than 2.0 per cent.
5 volumes of n-butanol, Inject the test solution and the reference solution.
flow rate. 1 ml per minute,
a linear gradient programme using the conditions given Calculate the content of C37H48N60sSz in the tablets.
below, Storage. Store protected from moisture, at a temperature not
spectrophotometer set at 240 nm, exceeding 30°.

2069

www.webofpharma.com
ROSIGLITAZONE MALEATE IP 2010

Rosiglitazone Maleate Assay. Determine by liquid chromatography (2.4.14).


Test solution. Dissolve 20 mg of the substance under
(COOH examination in 100.0 ml ofmobile phase.
Reference solution. A 0.020 per cent w/v solution of
COOH rosiglitazone maleate RS in mobile phase.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm packed with
ClsHI9N303S,CJ1404 Mol. Wt. 473.5 octadecylsilane bonded to porous silica (5 J.!m),
Rosiglitazone Maleate is (RS)-5- {p-[2-(methyl-2-pyridylamino) - mobile phase: a mixture of65 volumes of buffer solution
ethoxy]benzyl}-2,4-thiazolidinedione maleate (1: 1). prepared by dissolving 1.36 g ofpotassium dihydrogen
Rosiglitazone Maleate contains not less than 98.0 per cent orthophosphate in 1000 ml of water and adjust the pH
and not more than 102.0 per cent of ClsHI9N303S,C4H404, to 3.0 with dilute phosphoric acid, 25 volumes of
calculated on the anhydrous basis. acetonitrile and 10 volumes of methanol,
flow rate. 1 ml per minute,
Category. Antidibatic spectrophotometer set at 235 nm,
Description. A white to off-white crystalline powder. - injection volume. 10 J.!l.
Inject the reference solution. The test is not valid unless the
Identification
relative standard deviation for replicate injections is not more
Determine by infrared absorption spectrophotometry (2.4.6). than 2.0 per cent.
Compare the spectrum with that obtained with rosiglitazone
Inject the test solution and the reference solution.
maleate RS.
Calculate the content ofClsHI9N303S,C4H404.
Tests
Storage. Store protected from light.
Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Dissolve 25 mg of the substance under
examination in 50 ml ofmobile phase.
Rosiglitazone Tablets
Reference solution (a). A 0.05 per cent w/v solution of
rosiglitazone maleate RS in the mobile phase. Rosiglitazone Maleate Tablets
Reference solution (b). Dilute 1 ml ofreference solution (a) to Rosiglitazone Tablets contains not less than 90.0 per cent and
100 ml with mobile phase. not more than 110.0 per cent of the stated amount of
rosiglitazone, ClsH19N303S,
Use the chromatographic system described under Assay.
Usual strengths. 1 mg; 2 mg; 4 mg.
Inject reference solution (b). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
Identification
not less than 2000 theoretical plates.
Inject the test solution and reference solution (b). In the In the Assay, the principal peak in the chromatogram obtained
chromatogram obtained with the test solution, the area of any with the test solution corresponds to the peak in the
secondary peak is not more than 0.5 times the area ofthe peak chromatogram obtained with the reference solution.
in the chromatogram obtained with reference solution (b)
(0.5 per cent) and the sum of areas of all the secondary peaks
Tests
is not more than the area of the peak in the chromatogram Dissolution (2.5.2).
obtained with the reference solution (b) (1.0 per cent). Ignore
Apparatus No.1,
any peak (due to maleic acid) with a relative retention time of
Medium. 900 ml of 0.1 M hydrochloric acid,
about 0.4.
Speed and time. 75 rpm for 30 minutes.
Heavy metals (2.3.13). Lgcomplies with the limit test for heavy
metals, Metboc1BJ20ppm). Withdraw a suitable volume ofthe medium and filter promptly
through a membrane filter disc having an average pore diameter
Sulphated ash (2.3.18). Not more than 0.1 per cent. not greater than 1.0 J.!m, rejecting the first 1 ml ofthe filtrate.
Water (2.3.43). Not more than 0.5 per cent, determined on 1.0 g. Dilute the filtrate, ifnecessary, with the same solvent. Measure

2070

www.webofpharma.com
IP 2010 ROSUVASTATIN CALCIUM

the absorbance of the resulting solution at the maximum at - mobile phase: a mixture of 65 volumes of 0.01 M
about 318 urn (2.4.7). Similarly measure the absorbance of a potassium hydrogen phosphate adjusted to pH 3.0 with
standard solution of known concentration of rosiglitazone orthophosphoric acid, 25 volumes of acetonitrile and
maleate RS and calculate the content ofClsH,9N303S. 10 volumes of methanol,
D. Not less than 70 per cent of the stated amount of - flow rate. 1 ml per minute,
ClsH19N303S. - spectrophotometer set at 235 nm,
- injection volume. 10 Ill.
Related substances. Determine by liquid chromatography
(2.4.14). Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0, the column efficiency in not
Test solution. Weigh accurately a quantity ofpowdered tablets less than 4000 theoretical plates and the relative standard
containing 200 mg of Rosiglitazone, disperse in 100.0 ml of deviation for replicate injections is not more than 2.0 per cent.
mobile phase. Centrifuge and use clear supernatant liquid.
Inject the test solution and the reference solution.
Reference solution (a). A 0.2 per cent w/v solution of
rosiglitazone maleate RS in the mobile phase. Calculate the content ofClsH19N303S.

Reference solution (b). Dilute 1 ml ofreference solution (a) to Storage. Store protected from light and moisture.
100 ml with mobile phase.
Use the chromatographic system described under Assay.
Inject reference solution (b). The test is not valid unless the
Rosuvastatin Calcium
tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates.
F
Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any
secondary peak is not more than the area of the peak in the
chromatogram obtained with reference solution (b) (1.0 per
cent) and the sum of areas of all the secondary peaks is not Ca++ N/' ~
,
OH OH 0
0-
more than twice the area of the peak in the chromatogram
obtained with the reference solution (b) (2.0 per cent). Ignore H3C'N ANI CH 3
any peak (due to maleic acid) with a relative retention time of I
about 0.4. S02CH 3 CH 3
2
Uniformity of content. Comply with the tests stated under
Tablets. (C22H27FN306S)2.Ca Mol. Wt.l00l.l
Disperse 1 tablet in 0.1 M hydrochloric acid to produce Rosuvastatin Calcium is (E)-(3R,5S)-7- {4-(4-fluorophenyl)-
0.004 per cent w/v solution. Measure the absorbance of the 6-isopropyl-2-{methyl(methylsulphonylamino)]pyrimidin-
resulting solution at the maximum at about 318 nm (2.4.7). 5-yl} -3,5-dihydroxyhepten-6-oic acid calcium.
Calculate the content of CIsHI9N303S from the absorbance
Rosuvastatin Calcium contains not less than 98.0 per cent
obtained from same concentration of rosiglitazone maleate
and not more than 102.0 per cent of (C22H27FN306S)2.Ca.
RS in the same medium.
calculated on the anhydrous basis.
Other tests. Comply with the tests stated under Tablets.
Category. Antihyperlipidaemic.
Assay. Determine by liquid chromatography (2.4.14).
Description. An off- white to creamish white crystalline
Test solution. Weigh and powder 20 tablets. Weigh accurately powder.
a quantity of powdered tablets containing 20 mg of
RosiglitaZone, disperse in 100.0 ml ofmobile phase. Centrifuge Identification
and use clear supernatant liquid. A. Determine by iDfrared absorption spectrophotometry(2.4.6).
Reference solution. A 0.020 per cent w/v solution of Compare the spectrum with that obtained with fosuvastatin
rosiglitaione maleate RS in mobile phase. calcium RS. ..
Chromatographic system B. Dissolve 20 mg in 25 ml of methanol, add 2 drops of methyl
- a stainless steel column 25 cm x 4.6 mm packed with red indicator neutralise with 6 M ammonium hydroxide. Add
octadecysilane bonded to porous silica (5 11m), 3 M hydrochloric acid until the solution is acidic to the

2071

www.webofpharma.com
ROSUVASTATIN CALCIUM IF 2010

indicator. Add ammonium oxalate solution, a white precipitate Inject the reference solution. The test is not valid unless the
is obtained. relative standard deviation for replicate injections is not more
than 2.0 per cent.
Tests
Inject the test solution and the reference solution.
Related substances. Determine by liquid chromatography Calculate the content of(C22H27FN)06S)2.Ca.
(2.4.14).
Storage. Store protected from light and moisture.
Test solution. Dissolve 50 mg of the substance under
examination in 100 ml ofthe mobile phase.
Reference solution (a). A 0.05 per cent w/v solution of
rosuvastatin calcium RS in the mobile phase. Rosuvastatin Tablets
Reference solution (b). Dilute 1 ml ofreference solution (a) to Rosuvastatin Calcium Tablets
100 ml with mobile phase. Rosuvastatin Tablets contain not less than 90.0 per cent and
Chromatographic system not more than 110.0 per cent of the stated amount of
- a stainless steel column 25 cm x 4.6 mrn packed with rosuvastatin, C44Hs4F2N6012S2.
octadecylsilane bonded to porous silica (5 ~m), Usual strengths. 5 mg; 10 mg; 20 mg.
- mobile phase: a mixture of 50 volumes of 0.2 per cent
w/v acetic acid in water, 25 volumes of acetonitrile Identification
and 25 volumes of methanol,
In the Assay, the principal peak in the chromatogram obtained
flow rate. 1 ml per minute,
with the test solution corresponds to the peak in the
spectrophotometer set at 248 nm,
chromatogram obtained with the reference solution.
injection volume. 20 ~l.
Inject reference solution (a). The test is not valid unless the Tests
tailing factor is not more than 2.0 and the column efficiency in Dissolution (2.5.2).
not less than 2000 theoretical plates.
Apparatus No.1,
Inject the test solution and reference solution (b). In the Medium. 900 ml ofphosphate buffer pH 6.8,
chromatogram obtained with the test solution, the area of any Speed and time. 50 rpm and 30 minutes.
secondary peak is not more than 0.5 times the area ofthe peak
in the chromatogram obtained with reference solution (b) Withdraw a suitable volume ofthe medium and filter.
(0.5 per cent) and the sum of areas of all the secondary peaks Determine by liquid chromatography (2.4.14).
is not more than twice the area ofthe peak in the chromatogram
Test solution. The filtrate obtained as given above.
obtained with the reference solution (b) (2.0 per cent).
Reference solution. A 0.05 per cent w/v solution of
Water (2.3.43). Not more than 8.0 per cent, determined on 0.3 g.
rosuvastatin calcium RS in the mobile phase. Dilute 2 ml of
Assay. Determine by liquid chromatography (2.4.14). the solution to 100 ml with Dissolution medium.
Test solution. Dissolve 50 mg of the substance under Chromatographic system as described under Assay.
examination in 100.0 ml ofmobile phase. Dilute 5.0 ml ofthe
solution to 25.0 ml with mobile phase, mix and filter. Calculate the content ofC44Hs4F2N6012S2.
D. Not less than 70 per cent of the stated amount of
Reference solution. A 0.05 per cent w/v of rosuvastatin
calcium RS in mobile phase. Dilute 5.0 ml of the solution to C44Hs4F2N6012S2.
25.0 ml with mobile phase. Uniformity of content. Comply with the tests stated under
Chromatographic system Tablets.
- a stainless steel column 25 cm x 4.6 mm packed with Determine by liquid chromatography (2.4.14), as described
octadecylsilane bonded to porous silica (5 ~m), under Assay.
- mobile phase: a mixture of 50 volumes of 0.2 per cent
Test solution. Disperse one tablet in 100 ml ofthe mobile phase.
acetic acid in water, 25 volumes of acetQn.itrile and
Dilute 5 ml of the solution to 10 mlwith mobile phase and
25 volumes of methanol, filter and degas.
filter.
flow rate. 1 ml per minute,
spectrophotometer set at 248 nm, Related substances. Determine by liquid chromatography
injection volume. 20 ~l. (2.4.14).

2072

www.webofpharma.com
IP 2010 ROXITHROMYCIN

Test solution. Weigh and powder 20 tablets. Weigh accurately Roxithromycin


a quantity of powdered tablet containing 25 mg of
Rosuvastatin, disperse in 50 ml of mobile phase and filter.
Reference solution (a). A 0.05 per cent w/v solution of
rosuvastatin calcium RS in the mobile phase.
Reference solution (b). Dilute 1 ml ofreference solution (a) to
100 ml with mobile phase.
Chromatographic system as described under Assay.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates.
Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any
secondary peak is not more than 1.5 times the area ofthe peak
Mol. Wt. 837.0
in the chromatogram obtained with reference solution (b)
(1.5 per cent) and the sum of areas of all the secondary peaks Roxithromycin is (3R,4S,5S,6R,7R,9R,IIS,12R,13S,14R)-
is not more than 3.0 times the area ofthe peak in the chromato- 4-[(2,6-dideoxy-3-C-methyl-3-0-methyl-a-L-ribo-hexo-
gram obtained with the reference solution (b) (3.0 per cent). pyranosyl)oxy]-14-ethyl-7, 12,13-trihydroxy-
10-[(E)-[(2-methoxyethoxy)methoxy]imino]-3,5,7,9,11,
Other tests. Comply with the tests stated under Tablets.
13-hexamethyl-6-[[3,4,6-trideoxy-3-(dimethylamino)-
Assay. Determine by liquid chromatography (2.4.14). f3-D-xylo-hexopyranosyl]oxy]oxacyclotetradecan-2-one;
Test solution. Weigh and powder 20 tablets. Weigh accurately erythromycin 9-(E)-[0-[(2-methoxyethoxy)methyl]oxime].
a quantity of powdered tablet containing 25 mg of Rosu- Roxithromycin contains not less than 96.0 per cent and not
vastatin, disperse in 100.0 ml ofmobile phase. Dilute 5.0 ml of more than 102.0 per cent of C41H76N201s, calculated on the
the solution to 25.0 ml with mobile phase and filter. anhydrous basis.
Reference solution. A 0.025 per cent w/v solution of
Category. Antibacterial.
rosuvastatin calcium RS in mobile phase. Dilute 5.0 ml ofthe
solution to 25.0 ml with mobile phase. Dose. 150 mg twice a day.
Chromatographic system Description. A white, crystalline powder.
- a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 Ilm), Identification
- column temperature 30°,
A. Determine by infrared absorption spectrophotometry (2.4.6).
- mobile phase: a mixture of 585 volumes of a buffer Compare the spectrum with that obtained with roxithromycin
solution prepared by dissolving 1.54 g of ammonium
RS.
acetate in 900 ml water, adjust pH to 4.0 with glacial
acetic acid and dilute to 1000 ml with water, 360 volumes B. In the Assay, the principal peak in the chromatogram
of acetonitrile and 50 volumes of tetrahydrojitrane, obtained with the test solution corresponds to the peak in the
- flow rate. 1.5 ml per minute, chromatogram obtained with reference solution (a).
- spectrophotometer set at 248 nm, Tests
- injection volume. 20 Ill.
Appearance ofsolution. A 1.0 per cent w/v solution in methanol
Inject the reference solution. The test is not valid unless the
is clear (2.4.1) and colourless (2.4.1).
column efficiency is not less than 2000 theoretical plates, the
tailing factor is not more than 2.0 and the relative standard Specific optical rotation (2.4.22). - 93.0° to - 96.0°, determined
deviation for replicate injections is not more than 2.0 per cent. in a 1.0 per cent w/v solution in acetone.
Inject the test solution and the reference solution. Related substances. Determine by liquid chromatography
(2.4.14).
Calculate the content ofC44Hs4F2N6012S2.
Solvent mixture. Mix 30 volumes of acetonitrile and
Storage. Store protected from light and moisture.
70 volumes of a 4.8 per cent w/v solution of ammonium
Labelling. The label states the strength in terms of the dihydrogen phosphate and adjust the pH to 5.3 with dilute
equivalent amount of Rosuvastatin. sodium hydroxide solution.

2073

www.webofpharma.com
ROXITHROMYCIN IP 2010

Test solution. Dissolve 50.0 mg of the substance under solution (b) (1.0 per cent). The area of any other individual
examination in 25.0 ml ofthe solvent mixture. impurity peak is not more than 0.5 times the area ofthe principal
peak in the chromatogram obtained with reference solution
Reference solution (a). A 0.2 per cent w/v solution of
(b) (0.5 per cent). The sum ofthe areas of all the peaks is not
roxithromycin RS in the solvent mixture.
more than 3 times the area of the principal peak in the
Reference solution (b). Dilute 1 ml ofreference solution (a) to chromatogram obtained with reference solution (b}(3.0 per
100 ml with the solvent mixture. cent). Ignore any peak with an area 0.05 times the area ofthe
Reference solution (c). Dissolve 2 mg of erythromycin 9-(E)- principal peak in the chromatogram obtained with reference
[0-[[(2-methoxyethoxy)methoxy]methyl]oxime] (impurity solution (b) (0.05 per cent). Ignore any peak due to toluene
A) in 10 ml ofreference solution (a). Further dilute 1 mlofthis identified using reference solution (d).
solution to 10 ml with reference solution (a). Heavy metals (2.3.13). Dissolve 2.0 g in a mixture of15 volumes
Reference solution (d). Dilute 1.0 ml of toluene to 100 ml with of water and 85 volumes of acetone and dilute to 20 ml with
acetonitrile. Dilute 0.2 ml of this solution to 200 ml with the the same solvent mixture. 12 ml ofthis solution complies with
solvent mixture. the limit test for heavy metals, MethodA(10ppm). Use 1 mlof
lead standard solution (l0 ppm lead) prepared by diluting
Chromatographic system
lead standard solution (100 ppm lead) with a mixture of
- a stainless steel column 15 cm x 4.6 mm, packed with
15 volumes of water and 85 voiumes of acetone.
spherical end-capped octadecylsilane bonded to
porous silica (5 /lm), with a 10 nm pore size and a carbon Sulphated ash (2.3.18). Not more than 0.1 percent.
loading of about 19 per cent,
Water (2.3.43). Not more than 3.0 per cent, determined on
- column temperature. 15°,
0.2g.
- mobile phase: A. a mixture of26 volumes of acetonitrile
and 74 volumes of a 5.97 per cent w/v solution of Assay. Determine by liquid chromatography (2.4.14).
ammonium dihydrogen phosphate, adjusted to pH 4.3
Solvent mixture. Mix 30 volumes of acetonitrile and
with dilute sodium hydroxide solution,
70 volumes of a 4.8 per cent w/v solution of ammonium
B. a mixture ono volumes of water and dihydrogen phosphate and adjust the pH to 5.3 with dilute
70 volumes of acetonitrile,
sodium hydroxide solution.
- flow rate. 1. 1 ml per minute,
- a linear gradient programme using the conditions given Test solution. Dissolve 50.0 mg of the substance under
below, examination in 25.0 ml ofthe solvent mixture.
- spectrophotometer set at 205 nm, Reference solution (a). A 0.2 per cent w/v solution of
- injection volume. 20 Ill. roxitliromycin RS in the solvent mixture.
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent vIv) Reference solution (b). Dissolve 2 mg of erythromycin
9-(E)-[0-[[(2-methoxyethoxy)methoxy]methyl]oxime]
o 100 o (impurity A) in 10.0 ml ofreference solution (a). Further dilute
50 100 o 1.0 ml ofthis solution to 1O~O ml with reference solution (a).
80 90 10
Chromatographic system
100 100 o a stainless steel column 15 cm x 4.6 mm, packed with
Inject reference solution (c). Relative retention time between spherical end-capped octadecylsilane bonded to
roxithromycin and erythromycln9-(E)-[O-[[(2-methoxyethoxy) porous silica (5 /lm), with a 10 nm pore size and a carbon
methoxy]methyl] oxime] (impurity A) is about 1.15. The test is loading of about 19 per cent,
not valid unless the peak-to-valley ratio Hpl HI' is not more - column temperature 15°,
than 2.0, where H p = height above the baseline ofthe peak due - mobile phase: mix 307 volumes of acetonitrile and 693
to impurity A and HI' = height above the baseline ofthe lowest volumes of a 4.91 per cent w/v solution of ammonium
point of the curve separating this peak from the peak due to dihydrogen phosphate adjusted to pH 5.3 with dilute
roxithromycin. . sodium lJydroxide solution,
flow rate.l.5 ml per minute,
Inject the test solution and reference solutions (b) and (d). In
- spectrophotometer set at 205 rirri,
the chromatogralll obtained with the test solution the area of
- injection volume. 20 iiI.
the impurity A peak obtained immediately after the peak
obtained with roxithromycin is not more than the area of the Inject reference solution (b). The test is not valid unless the
principal peak in the chromatogram obtained with reference peak-to-valley ratio Hpl HI' is not less than 1.5.

2074

www.webofpharma.com
IP20l0 ROXITHROMYCIN TABLETS

Inject the test solution and reference solution (a) Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing about 0.2 g of
Calculate the content ofC14Hz6Nz015'
Roxithromycin, add 80 ml ofthe solvent mixture and mix. Dilute
Storage. Store protected from light and moisture. to 100.0 ml with the solvent mixtUre and filter.
Reference solution (a). A 0.2 per cent w/v solution of
roxithromycin RS in the solvent mixture.
Roxithromycin Tablets Reference solution (b). Dissolve 2 mg of elythromycin 9-(£)-
Roxithromycin Tablets contain not less than 90.0 per cent and [0-[[(2-methoxyethoxy)methoxy]methyl]oxime] (impurity
not more than 110.0 per cent of the stated amount of A) in 10 ml ofreference solution (a). Further dilute 1 ml ofthis
roxithromycin, C 4 IH76N zO I 5. solution to 10 ml with reference solution (a).

Usual strengths. ISO mg; 300 mg. Chromatographic system


- a stainless steel column 15 em x 4.6 mm, packed with
Identification spherical end-capped octadecylsilane bonded to
porous silica (5 /lm), with a 10 nm pore size and a carbon
In the Assay, the principal peak in the chromatogram obtained
loading of about 19 per cent,
with the test solution corresponds to the peak in the
- column temperature. 15°,
chromatogram obtained with the reference solution.
- mobile phase: mix 307 volumes of acetonitrile and
Tests 693 volumes of a 4.91 per cent w/v solution of
ammonium dihydrogen phosphate adjusted to pH 5.3
Dissolution (2.5.2). with dilute sodium hydroxide solution,
Apprartus No.1, - flow rate. 1.5 ml per minute,
Medium. 900 ml ofphosphate bujferpH 6.0, - spectrophotometer set at 205 urn,
Speed and time. 50 rpm and 45 minutes. - injection volume. 20 /ll.
Withdraw a suitable volume ofthe medium and filter. Inject reference solution (b). The relative retention time
between roxithromycin and erythromycin-(E)-[O-[[(2-
Determine by liquid chromatography (2.4.14).
methoxyethoxy)methoxy]methyl]oxime] (impurity A) is about
Test solution. The filtrate obtained as given above. 1.15. The test is not valid unless the peak-to-valley ratio
Reference solution. A 0.0166 per cent w/v solution of Hpl ~, is not more than 1.5, where Hp = height above the
roxithromycin RS in the dissolution medium. baseline of the peak due to impurity A and ~, = height above
the baseline of the lowest point of the curve separating this
Chromatographic system as described under Assay. peak from the peak due to roxithromycin.
Inject the test solution and the reference solution.
Inject reference solution (a). The test is not valid unless the
Calculate the content OfC41H76NzOl5 in the medium. relative standard deviation for replicate injections is not more
D. Not less than 70 per cent of the stated amount of than 2.0 per cent.
C41H76Nz015. Inject the test solution and reference solution (a).
Other tests. Comply with the tests stated under Tablets.
Calculate the content of C41H76NzOl5 in the tablets.
Assay. Determine by liquid chromatography (2.4.14).
Storage. Store protected from light and moisture.
Solvent mixture. Mix 30 volumes of acetonitrile and
70 volumes of a 4.8 per cent w/v solution of ammonium Labelling. If the tablets are dispersible, the label states that
dihydrogen phosphate and adjust the pH to 5.3 with dilute the tablets should be dispersed in water immediately before
sodium hydroxide solution. use.

2075

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

s
Saccharin 2081
Saccharin Sodium 2082
Salbutamol 2083
Salbutamol Inhalation 2084
Salbutamol Sulphate 2085
SalbutamolInjection 2087
Salbutamol Syrup 2088
Salbutamol Tablets 2088
SalicylicAcid 2089
Salmeterol Xinafoate 2090
Salmetrol and Fluticasone Propionate Inhalation 2091
Salmetrol and Fluticasone Propionate Powder for Inhalation 2091
Saquinavir 2092
SaquinavirMesylate 2093
SaquinavirMesylate Tablets 2094
Secnidazole 2095
Secnidazole Tablets 2096
Serratiopeptidase 2097
Serratiopeptidase Tablets 2098
ColloidalSiliconDioxide 2099
Sildenafil Citrate 2100
Sildenafil Tablets 2101
SilverNitrate 2102
Simvastatin 2103
SimvastatinTablets 2104
SodiumAcetate 2105
SodiumAlginate 2106
SodiumAminosalicylate 2107
SodiumAminosalicylate Tablets 2108
SodiumAscorbate 2109
Sodium Benzoate 2110

2077

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Sodium Bicarbonate 2111


Sodium Bicarbonate Injection 2111
Sodium Carbonate 2112
Sodium CWoride 2112
Sodium Chloride and Dextrose Injection 2113
Sodium Chloride and Fructose Injection 2114
Compound Sodium Chloride and Dextrose Injection 2115
Sodium CWoride Hypertonic Injection 2116
Sodium CWoride Injection 2116
Compound Sodium Chloride Injection 2117
Compound Sodium Chloride Solution 2117
Sodium CWoride Irrigation Solution 2118
Sodium Citrate 2118
Sodium Diatrizoate 2119
Sodium Diatrizoate Injection 2120
Sodium Dihydrogen Phosphate Dihydrate 2121
Sodium Fluoride 2122
Sodium Formaldehyde Sulphoxylate 2122
Sodium Fusidate 2123
Sodium Hydroxide 2124
Sodium Lactate Injection 2125
Compound Sodium Lactate and Dextrose Injection 2125
HalfStrength Compound Sodium Lactate and Dextrose Injection 2127
Modified Compound Sodium Lactate and Dextrose Injection 2128
Compound Sodium Lactate Injection 2129
Compound Sodium Lactate Solution for Irrigation 2130
SodiumLauryl Sulphate 2131
Sodium Metabisulphite 2132
SodiumMethylparaben 2132
Sodium Phosphate 2133
SodiumPropylparaben 2134
Sodium Salicylate 2134
Sodium Starch Glycollate 2137

2078

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Sodium Stibogluconate 2136


Sodium Stibogluconate Injection 2137
Sodium Thiosulphate 2138
Sodium Thiosulphate Injection 2138
Sodium Valproate 2139
Sodium Valproate Oral Solution 2140
Sodium Valproate Tablets 2140
SorbicAcid 2141
Sorbitan Oleate 2142
Sorbitol 2143
.Sorbitol Solution (70 Per cent) (Crystallising) 2144
Sorbitol Solution (70 Per cent) (Non-Crystallising) 2144
Spironolactone 2147
Spironolactone Tablets 2148
Stavudine 2149
Stavudine Capsules 2151
Stavudine Oral Solution 2152
Stavudine and Lamivudine Tablets 2153
Stearic Acid 2154
StearylAlcohol 2155
Stilboestrol 2156
Stilboestrol Tablets 2156
Streptokinase 2157
Streptokinase Injection 2159
Streptomycin Sulphate 2161
Streptomycin Injection 2162
Streptomycin Tablets 2163
Succinylcholine Chloride 2164
Succinylcholine Injection 2165
Sucralose 2165
Sucrose 2166
Sulphacetamide Sodium 2167
Sulphacetamide Eye Drops 2168

2079

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Sulphadiazine 2169
Sulphadiazine Tablets 2170
Sulphadoxine 2170
Sulphamethizole 2171
Sulphamethoxazole 2172
Sumatriptan 2173
Sumatriptan Injection 2174

2080

www.webofpharma.com
IP 2010 SACCHARIN

Saccharin acid and dilute to 50 ml with water. Shake the solution with 4
quantities, each of 50 ml, of dichloromethane. Combine the
lower layers, dry over anhydrous sodium sulphate and filter.
o

~
S~O Wah the filter and the sodium sulphate with 10 ml of
-:?' I \NH dichloromethane. Combine the solution and the washings
~ and evaporate almost to dryness in a water-bath at a
temperature not exceeding 40°. With a small quantity of
o dichloromethane transfer quantitatively the residue into a
suitable 10-ml tube, evaporate to dryness in a current of
C7HsN03S Mol. Wt. 183.2
nitrogen and dissolve the residue in 1.0 ml of the internal
Saccharin is 1,2-benzisothiazol-3(2H)-one 1, I-dioxide. standard solution.
Saccharin contains not less than 98.0 per cent and not more Blank solution. Evaporate 200 ml of dichloromethane to
than 101.0 per cent ofC 7HsN03S, calculated on the dried basis. dryness in a water-bath at a temperature not exceeding 40°.
Category. Pharmaceutical aid (sweetening agent). Dissolve the residue in I ml of dichloromethane.
Reference solution. Dissolve 20 mg of o-toluenesulphonamide
Description. White crystals or a white, crystalline powder;
and 20 mg of p-toluene sulphonamide in dichloromethane
odourless or with a faint, aromatic odour.
and dilute to 100 ml with the same solvent. Dilute 5 mlofthe
Identification solution to 50 ml with dichloromethane. Evaporate 5 mlofthe
final solution to dryness in a current of nitrogen. Dissolve the
Test A may be omitted iftests B, C andD are carried out. Tests residue in 1 ml of the internal standard solution.
B, C and D may be omitted if test A is carried out.
Chromatographic system
A. Determine by infrared absorption spectrophotometry (2.4.6). a fused silica column 10 m x 0.53 rom packed with
Compare the spectrum with that obtained with saccharin RS. polymethylphenylsiloxane (film thickness 2 /lm),
- temperature:
B. Mix 20 mg with 20 mg of resorcinol, add 05 ml ofsulphuric
acid and heat over a small flame until- a dark green colour is column.! 80°,
produced; allow to cool and add 10 ml of water and an excess inlet port and detector. 250°,
of 2 M sodium hydroxide; a fluorescent green liquid is flame ionization detector,
produced. flow rate. 10 ml per minute ofnitrogen (carrier gas),
split ratio. 1:2.
C. Dissolve 0.1 g in 5 ml of a 10 per cent w/v solution of
sodium hydroxide, evaporate to dryness and gently fuse the Inject 1 /ll of each solution. The order of elution is
residue over a small flame until ammonia is no longer evolved. o-toluenesulphonamide, p-toluenesulphonamide, caffeine.
Allow to cool, dissolve in 20 ml of water, make the solution The test is not valid unless the resolution between the peaks
just acidic to litmus paper, filter and add 0.05 ml of ferric due to o-toluenesulphonamide and p-toluenesulphonamide
chloride solution; a violet colour is produced. in the chromatogram obtained with the reference solution is
D. A saturated solution is acidic to litmus paper. not less than 1.5 and the chromatogram obtained with the
blank solution does not show any peak with the same retention
Tests times as the internal standard, o-toluenesulphonamide and
p-toluenesulphonamide.
Appearance ofsolution. Dissolve 5.0 g in 20 ml ofa 20 per cent
w/v solution of sodium acetate and dilute to 25 ml with the In the chromatogram obtained with the test solution the ratio
same solution; the solution is clear (2.4.1), and colourless of the area of the peak due to o-toluenesulphonamide to that
(2.4.1). of the internal standard is not greater than the corresponding
ratio in the chromatogram obtained with the rference solution
Related substances. Determine by gas chromatography (lOppm).
(2.4.13).
In the chromatogram obtained with the test solution the ratio
Internal standard solution. Dissolve 25 mg of caffeine in of the area of the peak due to p-toluenesulphonamide to that
dichloromethane and dilute to 100 ml with the same solvent. of the internal standard is not greater than the corresponding
Test solution. Suspend 10.0 g ofthe substance under examina- ratio in the chromatogram obtained with the reference solution
tion in 20 ml of water and dissolve using about 5 ml of strong (10 ppm).
sodium hydroxide solution. Ifnecessary, adjust the pH of the Arsenic (2.3.10). Mix 5.0 g with 3.0 g of anhydrous sodium
solution to 7.8 with 1 M sodium hydroxide or 1 M hydrochloric carbonate, add 10 ml of bromine solution and mix thoroughly.

2081

www.webofpharma.com
SACCHARIN SODIUM IP 2010

Evaporate to dryness on a water-bath, gently ignite,and add Identification


to the cooled residue a mixture of 16 ml of brominated
hydrochloric acid AsT and 5 ml of bromine solution. Add Test A may be omitted if tests B, C, D and E are carried out.
40 ml of water and boil gently, adding sufficient bromine Tests B, C and D may be omitted if tests A and E are carried
solution from time to time to maintain a slight excess. Filter out.
and remove the excess ofbromine with a sufficient quantity of A. Detennine by infrared absorption spectrophotomeuy (2.4.6).
stannous chloride solution AsT. The resulting solution Compare the spectrum with that obtained with saccharin
complies with the limit test for arsenic (2 ppm). sodium RS.
Heavy metals (2.3.13). 1.2 g complies with the limit test for B. Dissolve 0.1 g in 5 ml of a 10 per cent w/v solution of
heavy metals, Method D (20 ppm). Use leadstandardsolution sodium hydroxide, evaporate to dryness and gently fuse the
(2 ppm Pb). residue over a small flame until ammonia is no longer evolved.
Readily carbonisable substances. Dissolve 0.2 g in 5 ml of Allow to cool, dissolve in 20 ml of water, make the solution
sulphuric acid (96 per cent w/w) and maintain at about 50° just acidic to litmus paper, filter and add 0.05 ml of ferric
for 10 minutes. The solution is not more intensely coloured chloride solution; a violet colour is produced.
than reference solution BYS6 (2.4.1). C. Mix 20 mg with 20 mg of resorcinol, add 0.5 mlofsulphuric
Sulphated ash (2.3.18). Not more than 0.1 per cent. acid and heat over a small flame until a dark green colour is
produced; allow to cool and add 10 ml of water and an excess
Loss on drying (2.4.19). Not more than LOper cent, detennined of 2 M sodium hydroxide; a fluorescent green liquid is
on 1.0 g by drying in an oven at 105° for 4 hours. produced.
Assay. Weigh accurately about 0.5 g, dissolve in 75 ml ofhot D. To 5 mlofa 10 per centw/v solution add 3 mlof2 Mhydro-
water, cool quickly and titrate with 0.1 M sodium hydroxide chloric acid; a white precipitate is produced. The precipitate,
using phenolphthalein solution as indicator. after washing with water and drying at 105° melts at 226° to
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01832 g of 230° (2.4.21).
C7HsN03S. E. 0.5 ml of a 10 per cent w/v solution gives reaction B of
Storage. Store protected from moisture. sodium salts (2.3.1).

Tests
Appearance of solution. Solution A is clear (2.4.1), and
Saccharin Sodium colourless (2.4.1).
Acidity or alkalinity. Dissolve 5.0 g in sufficient carbon
Soluble Saccharin dioxide-free water to produce 50 ml (solution A). To 10 ml of
solution A add 5 ml of 0.005 M sulphuric acid, heat to boiling,
cool and add 0.1 ml of phenolphthalein solution. Not less
o than 4.5 ml and not more than 5.5 ml of 0.01 M sodium

~
s-:-o
-;:/' , hydroxide is required tochange the colour of the solution to
~ I N-Na pink.

o Related substances. Detennine by gas chromatography


(2.4.13).
Internal standard solution. Dissolve 25 mg of caffeine in
C7~a03S Mol. Wt. 205.2 dichloromethane and dilute to 100 ml with the same solvent.
Saccharin Sodium is the sodium salt of 1,2-benzisothiazol- Testsolution. Suspend 10.Og of the substancel ul1<ier
3(2H)-3-one 1, I-dioxide. examination in 50 ml of water. If necessary, adjust the pH of
Saccharin Sodium contains not less than 99.0 per cent and not the solution to 7.8 with 1 M sodium hydroxide or 1 M hydro-
more than 101.0 per cent ofC7H4NNa03S, calculated on the chloric acid and dilute to 50 m1 with water. Shake the solution
anhydrous basis. with 4 quantities, each of50 ml, of dichloromethane. Combine
the lower layers, dry over anhydrous sodium sulphate and
Category. Phannaceutical aid (sweetening agent). filter. Wah the filter and the sodium sulphate with 10 ml of
Description. A white, crystalline powder or colourless crystals; dichloromethane. Combine the solution and the washings
efflorescent in dry air. and evaporate almost to dryness in a water-bath at a

2082

www.webofpharma.com
IP 2010 SALBUTAMOL

temperature not exceeding 40°. With a small quantity of and'remove the excess ofbromine with a sufficient quantity of
dichloromethane transfer quantitatively the residue into a stannous chloride solution AsT. The resulting solution
suitable 10-ml tube, evaporate to dryness in a current of complies with the limit test for arsenic (2 ppm).
nitrogen and dissolve the residue in 1,0 ml of the internal
Readily carbonisable substances. Dissolve 0.2 g in 5 ml of
standard solution.
sulphuric acid (96 per cent w/w) and maintain at about 50°
Blank solution. Evaporate 200 ml of dichloromethane to for 10 minutes. The' solution is not more intensely coloured
dryness in a water-bath at a temperature not exceeding 40°. than reference solution BS6 (2.4.1).
Dissolve the residue in 1 ml of dichloromethane.
Heavy metals (2.3.13). 12 ml ofsolution A complies with the
Reference solution. Dissolve 20 mg of o-toluenesulphonamide limit test for heavy metals, Method D (20 ppm). Use lead
and 20 mg of p-toluene sulphonamide in dichloromethane standard solution (2 ppm Pb).
and dilute to 100 ml with the same solvent. Dilute 5 ml of this
Water (2.3.43). Not more than 15.0 per cent, determined on
solution to 50 ml with dichloromethane. Evaporate 5 mlofthe
0.2g.
final solution to dryness in a current of nitrogen. Dissolve the
residue in 1 ml of the internal standard solution. Assay. Weigh accurately about 0.15 g, dissolve in 50 ml of
anhydrous glacial acetic acid, with slight heating ifnecessary.
Chromatographic system Titrate with 0.1 M perchloric acid, determining the end-point
a fused silica column 10m x 0.53 mm packed with potentiometrically (2.4.25). Carry out a blank titration.
polymethylphenylsiloxane (film thickness 2 !!m),
temperature: 1 ml of 0.1 M perchloric acid is equivalent to 0.02052 g of
column.180°, C7H;NNa03S,
inlet port and detector. 250 0 Storage. Store protected from moisture.
flame ionization detector,
flow rate. 10 ml per minute ofnitrogen (carrier gas),
split ratio. 1:2.
Inject 1 !!1 of each solution. The order of elution is Salbutamol
o-toluenesulphonamide, p-toluenesulphonamide, caffeine.
Albuterol
The test is not valid unless the resolution between the peaks
due to .o-toluenesulphonamide and p-toluenesulphonamide OH
in the chromatogram obtained with the reference solution is
not less than 1.5 and the chromatogram obtained with the
blank solution does not show any peak with the same retention
times as the internal standard, o-toluenesulphonamide and HO
p-toluenesulphonamide.
OH
In the chromatogram obtained with the test solution the ratio
of the area of the peak due to o-toluenesulphonamide to that Mol. Wt. 239.3
of the internal standard is not greater than the corresponding
Salbutamol is (RS)-I-( 4-hydroxy-3-hydroxymethylphenyl)-
ratio in the chromatogram obtained with the reference solution
2-(tert-butylamino)ethanol.
(10 ppm).
Salbutamol contains not less than 98.0 per cent and not more
In the chromatogram obtained with the test solution the ratio than 101.0 per cent ofC 13Hz1 N0 3, calculated on the dried basis.
of the area of the peak due to p-t6luenesulphonamide to that
of the internal standard is not greater than the corresponding Category. Beta-adrenoceptor agonist.
ratio in the chromatogram obtained with the rference solution Dose. Orally, 6 to 16 mg daily, in divided doses; by inhalation,
. (10 ppm). for chronic bronchial asthma or as a prophylactic, 2 inhalations
Arsenic (2.3.10). Mix 5.0 g with 3 g of anhydrous sodium of 100 !!g, 3 or 4 times daily; for the relief of acute
carbonate, add 10 ml of bromine solution and mix thoroughly. bronchospasm, 1 or 2 inhalations of 100 !!g as a single dose
Evaporate to dryness on a water-bath, gently ignite and add when required, up to a maximumof8 inhalations in 24 hours;
to the cooled residue a mixture of 16 ml of brominated by subcutaneous or intramuscular injection, 500 !!g, repeated
hydrochloric acid AsT and 5 ml of bromine solution. Add every 4 hours, if necessary; by slow intravenous injection,
40 ml of water and boil gently, adding sufficient bromine 250 !!g, repeated if necessary.
solution from time to time to maintain a slight excess. Filter Description. A white or almost white, crystalline powder.

2083

www.webofpharma.com
SALBUTAMOL IF 2010

Identification Inject the test solution and the reference solution. Run the
chromatogram 25 times the retention time ofthe principal peak.
Test A may be omitted iftests B, C andD are carried out. Tests In the chromatogram obtained with the test solution the area
B, C and D may be omitted if test A is carried out. ofany secondary peale is not more than the area ofthe principal
A. Detennine by infrared absorption spectrophotometry (2.4.6), peak in the chromatogram obtained with the reference solution
Compare the spectrum with that obtained with salbutamol RS (0.3 per cent) and the sum of areas of all the secondary peaks
or with the reference spectrum of salbutamol. is not more than 3.3 times the area ofthe principal peak in the
chromatogram obtained with the reference solution (1.0 per
B. When examined in the range 230 nm to 360 run (2.4.7), a
cent). Ignore any peak with an area less than 0.05 per cent the
0.008 per cent w/v solution in 0.1 Mhydrochloric acid shows
area ofthe principal peak in the chromatogram obtained with
an absorption maximum only at about 276 nm; absorbance at
the reference solution.
about 276 run, about 0.53 to 0.60.
C. In the test for Related substances, the principal spot in the Boron. To 50 mg add 5 ml ofa solution containing 1.3 per cent
chromatogram obtained with reference solution (a) w/v of anhydrous sodium carbonate and 1.7 per cent w/v of
corresponds to that in the chromatogram obtained with potassium carbonate, evaporate to dryness on a water-bath
reference solution (b). and dry at 120°. Ignite the residue rapidly until the organic
matter has been destroyed, allow to cool, add 0.5 ml of water
D. Dissolve 10 mg in 50 ml of a 2 per cent w/v solution of and 3.0 ml ofa freshly prepared 0.125 per cent w/v solution of
borax, add 1ml ofa 3 per cent w/v solution of4-aminophenazone, curcumin in glacial acetic acid. Evaporate to dryness and
10 ml of a 2 per cent w/v solution of potassium ferricyanide allow to cool. Add 3 ml ofa mixture prepared by adding 5 ml of
and 10 ml of chloroform, shake and allow to separate; an sulphuric acid, slowly and with stirring, to 5 ml of glacial
orange-red colour is produced in the chlorofonn layer. acetic acid. Mix and allow to stand for 30 minutes. Add
Tests sufficient ethanol (95 per cent) to produce 100.0 ml, filter and
measure the absorbance ofthe filtrate at 555 run (2.4.7). Prepare
Appearance ofsolution. A2.0 per cent w/v solution in methanol a reference solution in the following manner. Dissolve 0.572 g
is clear (2.4.1), and not more intensely coloured than reference of boric acid in 1000.0 ml of water. Dilute 1.0 ml to 100.0 ml
solution BYS5 (2.4.1). with water. To 2.5 ml of this solution add 5 ml of a solution
Related substances. Detennine by liquid chromatography containing 1.3 per cent w/v of anhydrous sodium carbonate
(2.4.14). and 1.7 per cent w/v of potassium carbonate and treat this
mixture in the same manner as described above beginning at
Test solution. Dissolve 0.1 g of the substance under
the words "Evaporate to dryness; ..". The absorbance of the
examination in 50.0 ml ofthe mobile phase.
solution prepared from the substance under examination is
Reference solution. A solution containing 0.02 per cent w/v not more than that ofthe reference solution (50 ppm).
each of salbutamol RS and (lRS)-2-[(l,1-
Sulphated ash (2.3.18). Not more than 0.1 percent.
dimethylethyl)amino]-1-(4- hydroxyphenyl)ethanol RS
(salbutamol impurity A RS) in the mobile phase. Dilute 2.0 ml Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
ofthis solution to 100.0 ml with the mobile phase. on 1.0 g by drying in an oven at 105°.
Chromatographic system Assay. Weigh accurately about 0.2 g, dissolve in 30 ml of
- a stainless steel column 15 cm x 3.9 mrn, packed with anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
endcapped octylsilane bonded to porous silica (5 /lm), acid, detennining the end-point potentiometrically (2.4.25).
mobile phase: a mixture of 22 volumes of acetonitrile Carry out a blank titration.
and 78 volumes of a solution containing 0.29 per cent 1 ml of 0.1 M perchloric acid is equivalent to 0.02393 g of
w/v of sodium heptanesulphonate and 0.25 per cent
CI3HzIN03.
w/v of potassium dihydrogen phosphate, adjusted to
pH 3.7 with orthophosphoric acid, Storage. Store protected from light.
- flow rate; 1 ml per minute,
spectrophotometer set at 220 run,
injection volume. 20 Ill. Salbutamol Inhalation
Inject the ref~rence solution. The test is not valid llnless the
Salbutamol Inhalation Aerosol; Albuterol Inhalation
r~s()lllti<:>.n _~e_t\\lee!1the peaks_~ue!()~ll('U()_s~llJllt~IIloLan~
salbutamol impurity A is not less than 3.0. The relative retention
Aerosol
time with reference to salbutamo1 for salbutamol impurity A is . Salbutamol Inhalation is a suspension ofmicrofine Salbutamol
about 1.3. or Salbutamol Sulphate in a suitable liquid in a suitable

2084

www.webofpharma.com
IP 2010 SALBUTAMOL SULPHATE

pressurised container. It may contain suitable pharmaceutical· Assay. Carry out the test for Content of active ingredient
aids such as surfactants, stabilising agents etc. delivered per actuation stated under Inhalation Preparations
(Pressurised metered-dose Preparations).
Salbutamol Inhalation delivers not less than 80.0 per cent and
not more than 120.0 per cent of the sfated amount of Use 30 ml of ethanol for preparations containing salbutamol
salbutamol, C 13H21 N0 3, per inhalation, by actuation ofthe valve. and 30 ml ofa mixture ofequal volumes of ethanol and water
for preparations containing salbutamol sulphate and dilute
Usual strength. 100 Jlg in each metered-dose.
the final solution and washings to 200.0 ml with ethanol. Dilute
a suitable volume of this solution with ethanol to produce a
Identification solution containing 10 Jlgofsalbutamolperml. To 20ml ofthe
A. Discharge the container a sufficient number oftimes into a solution in a separating funnel add 180 ml of water and in the
mortar to obtain about 2 mg of salbutamol, grind the residue following order, 4 ml of N,N-dimethyl-4-phenylenediamine
thoroughly with 0.1 g of potassium bromide, add a further sulphate solution and 4 ml of a freshly prepared 8 per cent
0.2 g ofpotassium bromide and mix thoroughly. w/v solution of potassium ferricyanide. Mix, allow to stand
for 15 minutes in subdued light and extract with two quantities,
On the resultant dispersion determine by infrared absorption each of 10 ml, of chloroform. Filter the extracts through a plug
spectrophotometry (2.4.6). Compare the spectrum with that of cotton wool, dilute to 25 ml with chloroform and measure
obtained with salbutamol RS or with the reference spectrum the absorbance of the resulting solution at 605 nm (2.4.7).
of salbutamol. Calculate the content of C 13H 21 N0 3 in the solution from the
B. In the test for Related substances, the principal spot in the absorbance obtained by repeating the operation using a
chromatogram obtained with reference solution (a) suitable quantity ofa 0.001 per cent w/v solution ofsalbutamol
corresponds to that in the chromatogram obtained with RS in ethanol.
reference solution (b). Calculate the amount OfCl3H21N03 delivered per actuation of
the valve.
Tests Determine the content of active ingredient a second and third
Related substances. Determine by thin-layer chromatography time by repeating the procedure on the middle ten and on the
(2.4.17), coating the plate with silica gel G last ten successive combined actuations of the valve. For
each of the three determinations the average content of
Mobile phase. A mixture of 50 volumes of ethyl acetate, Cl3H21N03 delivered per actuation of the valve meets the
30 volumes of 2-propanol, 16 volumes of water and 4 volumes requirements.
of strong ammonia solution.
Storage. Store protected from light and moisture at a
Test solution. Discharge the inhaler a sufficient number of temperature not exceeding 30°.
times into a small, dry beaker to obtain 10 mg of Salbutamol
and dissolve the residue in 0.5 ml of methanol. Labelling. The label states whether the preparation contains
Salbutamol or Salbutamol Sulphate.
Reference solution (aJ. Dilute 1.0 volume of test solution (a)
When the active ingredient is Salbutamol Sulphate, the
to 200 volumes with methanol.
quantity is stated in terms of the equivalent amount of
Reference solution (bJ. A 0.010 per cent w/v solution of salbutamol.
salbutamol RS in methanol.
Apply to the plate 5 Jll of each solution. After development,
dry the plate in air until the odour of the solvent is no longer
detectable, place it for a few minutes in an atmosphere saturated Salbutamol Sulphate
with diethylamine and spray with diazotised sulphanilic acid Albuterol Sulphate
solution. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the (CI3H21N03)2,H2S04 Mol. Wt. 576.7
chromatogram obtained with reference solution (a). Ignore Salbutamol Sulphate is (RS)-1-(4-hydroxy-3-hydroxymethyl-
any spot with an R r value higher than 0.85. phenyl)-2-(tert-butylamino)ethanol sulphate.
Other tests. Complies with the tests stated under Inhalation Salbutamol Sulphate contains not less than 98.0 per cent and
Preparations (Pressurised metered-dose Preparations). not more than 101.0 per cent of(C 13H 21 N03)2, H 2S04, calculated
on the dried basis.
Follow the procedure described under Assay wherever the
amount of active substance is to be determined in any test. Category. Beta-adrenoceptor agonist.

2085

www.webofpharma.com
SALBUTAMOLSULPHATE IP 2010

Dose. Orally, the equivalent of6 to 16 mg ofsalbutamol daily, (salbutamol impurity A RS) in the mobile phase. Dilute 2.0 ml
in divided doses; by slow intravenous injection, the equivalent of this solution to 100.0 ml with the mobile phase.
of 250 Ilg of salbutamol or by intravenous infusion, the Chromatographic system
equivalent of 3 to 20 Ilg of salbutamol per minute. (1 mg of a stainless steel column 15 cm x 3.9 mm, packed with
Salbutamol Sulphate is approximately equivalent to 830 Ilg of endcapped octylsilanebonded to porous silica (5 Ilm),
salbutamol). mobile phase: a mixture of 22 volumes of acetonitrile
Description. A white or almost white, crystalline powder. and 78 volumes of a solution containing 0.29 per cent
w/v of sodium heptanesulphonate and 0.25 per cent
Identification w/v of potassium dihydrogen phosphate, adjusted to
pH 3.7 with orthophosphoric acid,
Test A may be omitted if tests B, C, D andE are carried out.
- flow rate. 1 ml per minute,
Tests Band C may be omitted if tests A, D and E are carried
- spectrophotometer set at 220 run,
out.
injection volume. 20 Ill.
A. Detennine by infrared absorption spectrophotometry (2.4.6),
Inject the reference solution. The test is not valid unless the
Compare the spectrum with that obtained with salbutamol
resolution between the peaks due to due to salbutamol and
sulphate RS or with the reference spectrum of salbutamol
salbutamol impurity Ais not less than 3.0. The relative retention
sulphate.
time with reference to salbutamol forsalbutamol impurity A is
B. When examined in the range 230 Jim to 360 nm (2.4.7), a about 1.3.
0.008 per cent w/v solution in 0.1 M hydrochloric acid shows
Inject the test solution and the reference. solution. Run the
an absorption maximum at about 276 run; absorbance at about
chromatogram 25 times the retention time ofthe principal peak.
276 urn, 0.44 to 0.51.
In the chromatogram obtained with the test solution the area
C. In the test for Related substances, the principal spot in the ofany secondary peak is not more than the area ofthe principal
chromatogram obtained with reference solution (a) peak in the chromatogram obtained with the reference solution
corresponds to that in the chromatogram obtained with (0.3 per cent) and the sum of areas of all the secondary peaks
reference solution (b). is not more than 3.3 times the area ofthe principal peak in the
D. Dissolve 10 mg in 50 ml of a 2 per cent w/v solution of chromatogram obtained with the reference solution (1.0 per
borax, add 1 ml of a 3 per cent w/v solution of 4-amino- cent). Ignore any peak with an area less than 0.05 per cent the
phenazone, lO ml of a 2 per cent w/v solution of potassium area of the principal peak in the chromatogram obtained with
ferricyanide and 10 ml of chloroform, shake and allow to the reference solution.
separate; an orange-red colour is produced in the chloroform Boron. To 50 mg add 5 m1 ofa solution containing 1.3 per cent
layer. w/v of anhydrous sodium carbonate and 1.7 per cent w/v of
E. Gives reaction A ofsulphates (2.3.1). potassium carbonate, evaporate to dryness on a water-bath
and dry at 120°. Ignite the residue rapidly until the organic
Tests matter has been destroyed, allow to cool, add 0.5 mlofwater
and 3.0 ml ofa freshly prepared 0.125 percentw/v solution of
Appearance of solution. A 1.0 per cent w/v solution in carbon
curcumin in glacial acetic acid. Evaporate to dryness and
dioxide-free water is clear (2.4.1), and not more intensely allow to cool. Add 3 m1 ofa mixture prepared by adding 5m1 of
coloured than reference solution BYS6 (2.4.1).
sulphuric acid, slowly and with stirring, to 5 ml of glacial
Acidity or alkalinity. To 10 ml ofa 1.0 per cent w/v solution in acetic acid. Mix and allow to stand for 30 minutes. Add
carbon dioxide-free water add 0.15 ml of methyl red solution sufficient ethanol (95 per cent) to produce 100.0 ml, filter and
and 0.2 ml of 0.01 M sodium hydroxide. The solution is yellow measure the absorbance ofthe filtrate at 555 run (2.4.7). Prepare
and not more than 0.4 ml of 0.01 M hydrochloric acid is a reference solution in the following manner. Dissolve 0.572 g
required to change the colour of the solution to red. of boric acid in 1000.0 ml of water. Dilute 1.0 ml to 100.0 ml
with water. To 2.5 ml of this solution add 5 m1 of a solution
Related substances. Determine by liquid chromatography
containing 1.3 per cent w/v of anhydrous sodium carbonate
(2.4.14).
and 1.7 per cent w/vofpotassium carbonate and treat this
Test solution. Dissolve 0.1 g of the substance under mixture in the same manner as described above beginning at
examination in 50.0 ml ofthe mobile phase.. the words" Evaponite to dryness::...".The absorbance onhe
R~ler~nce solution. Asolution contalnin.g()~02perceniw!v solution prepared from the substance under examination is
each of salbutamol RS and (lRS)-2-[(l,I- not more than that of the reference solution (50 ppm).
dimethylethyl)amino}-1-(4- hydroxyphenyl)ethanol RS Sulphated ash (2.3.18). Not more than O.lper cent.

2086

www.webofpharma.com
IP 2010 SALBUTAMOL INJECTION

Loss on drying (2.4.19). Not more than 0.5 per cent, determined detectable, place for a few minutes in an atmosphere saturated
on 1.0 g by drying in an oven at 105°. with diethylamine and spray with diazotised nitroaniline
Assay. Weigh accurately about 0.4 g, dissolve in 5 ml of
solution. The principal spot in the chromatogram obtained
with the test solution corresponds to that in the chromatogram
anhydrousformic acid, add 35 ml of anhydrous glacial acetic
obtained with the reference solution.
acid. Titrate with 0.1 M perchloric acid, determining the end-
point potentiometrically (2.4.25). Carry out a blank titration. C. Dilute a volume containing 0.5 mg of salbutamol to 50 ml
with water, add 1 ml of dilute ammonia solution, 1 ml ofa 3 per
1 ml of 0.1 M perchloric acid is equivalent to 0.05767 g of
cent w/v solution of 4-aminophenazone, 10 ml of a 2 per cent
(CI3H2IN03)2, H2S04 •
w/v solution of potassium ferricyanide and 10 ml of
Storage. Store protected from light. chloroform. Shake and allow to separate; an orange-red colour
is produced in the chloroform layer.
D. A volume containing 1 mg ofsalbutamol gives the reactions
Salbutamol Injection ofsulphates (2.3.1).
Albuterol Sulphate Injection; Salbutamol Sulphate Tests
Injection
pH (2.4.24).3.4 to 5.0.
Salbutamol Injection is a sterile solution of Salbutamol
Sulphate in Water for Injections containing suitable stabilising Related substances. Determine by liquid chromatography
agents. (2.4.14).

Salbutamol Injection contains not less than 90.0 per cent and Test solution. Dilute a volume of injection containing about 5
not more than 110.0 per cent of the stated amount of mg ofSalbutamol in 100.0 ml ofthe mobile phase.
salbutamol, C 13 H2I N0 3. Reference solution (a). Dilute 1.0 ml of the test solution to
Usual strengths. The equivalent of250 Ilg of salbutamol per 100.0 ml with the mobile phase.
rnl; the equivalent of500 Ilg ofsalbutamol pennI; the equivalent Reference solution (b). A solution containing 0.0004 per cent
of5 mg ofsalbutamol in 5 ml (for intravenous infusion). (l mg w/v of (lRS)-2-[(l,1-dimethylethyl)amino]-1-(4-
ofSalbutamol Sulphate is approximately equivalent to 830 Ilg hydroxyphenyl)ethanol RS (salbutamol impurity A RS) and
ofsalbutamol). 0.0005 per cent w/v of salbutamol sulphate RSin the mobile
phase.
Identification
Chromatographic system
A. Dilute a volume with sufficient 0.1 M hydrochloric acid to a stainless steel column 15 cm x 3.9 mm, packed with
produce a solution containing 0.008 per cent w/v ofsalbutamol. endcapped octylsilane bonded to porous silica (5 Ilm)
When examined in the range 230nm to 360 nm (2.4.7), the (such as Waters Symmetry C8),
resulting solution shows an absorption maximum only at about mobile phase: a mixture of 22 volumes of acetonitrile
276nm. and 78 volumes of a solution containing 0.29 per cent
B. Determine by thin-layer chromatography (2.4.17), coating w/v of sodium heptanesulphonate and 0.25 per cent
the plate with silica gel G w/v of potassium dihydrogen phosphate, adjusted to
pH 3.7 with orthophosphoric acid,
Mobile phase. A mixture of 50 volumes of ethyl acetate,
flow rate. 1 ml per minute,
30 volumes of 2-propanol, 16 volumes of water and 4 volumes
spectrophotometer set at 220 nm,
of strong ammonia solution.
injection volume. 20 Ill.
Test solution. Evaporatea suitable volume ofthe injection to
Inject reference solution (b). The test is not valid unless the
dryness using a rotary evaporator, wash the residue with four
resolution between the two principal peaks is not less than
quantities, each of5 ml, of ethanol, filter, evaporate the filtrate
3.0.
to. dryness and dissolve the residue in sufficient water to
produce a solution containing the equivalent of 0.1 per cent Inject the test solution and reference solution (a). Run the
w/v of salbutamol. chromatogram 25 times the retention time ofthe principal peak.
In the chromatogram obtained with the test solution the area
Reference solution. A 0.12 per cent w/v solution of salbutamol of any secondary peak is not more than 0.5 times the area of
sulphate RS in water..
the principal peale in the chromatogram obtained with reference
Apply to the plate 2 III of each solution. After development, solution (a) (0.5 per cent) and the sum of all the secondary
dry the plate in air until the odour of solvent is no longer peaks is not more than twice the area of the principal peak in

2087

www.webofpharma.com
SALBUTAMOL SYRUP IP 2010

the chromatogram obtained with reference solution (a) (2.0 Assay. To an accurately measured volume containing about
per cent). 4 mg of salbutamol add 25 ml of 0.05 M sulphuric acid and
extract with two quantities, each of50 ml, of ether. Collect the
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections). aqueous layers into a 250-ml volumetric flask and combine the
ether extracts. Wash the combined ether extracts with 50 ml of
Assay. Dilute a volume, accurately measured, containing about water and add the aqueous layer to the solution in the 250 ml
0.15 mg ofsalbutamol with sufficient water to produce 80 ml, volumetric flask. Discard the ether extracts and dilute the
add 4 ml of a 5 per cent w/v solution ofsodium bicarbonate, aqueous solution with sufficient water to produce 250.0 ml.
4 ml of N,N-dimethyl-4-phenylenedial~inesulphate solution To 10.0 ml of this solution add sufficient water to produce
and 4 ml of a freshly prepared 8 per cent w/v solution of 80 ml and add 4 ml of a 5 per cent w/v solution of sodium
potassiumferricyanide. Mix, allow to stand for 15 minutes, bicarbonate, 4 ml of N,N-dimethyl-4-phenylenediamine
protected from light. Extract with two quantities, each of 10 ml, sulphate solution and 4 ml of a freshly prepared 8 per cent
of chloroform. Filter the extracts through a plug of cotton w/v solution of potassium ferricyanide. Mix, allow to stand
wool and dilute to 25.0 ml with chloroform. Measure the for 15 minutes, protected from light. Extract with two quantities,
absorbance of the resulting solution at 605 run (2.4.7). each of 10 ml, of chloroform. Filter the extracts through a plug
Calculate the content of C 13 H Z1 N03 from the absorbance ofcotton wool and dilute to 25.0 ml with chloroform. Measure
obtained by repeating the operation using 10.0 ml of a the absorbance ofthe resulting solution at 605 run (2.4.7).
0.0018 per cent w/v solution of salbutamol sulphate. Calculate the content of C 13 H Z1 N0 3 from the absorbance
Storage. Store protected from light, in single dose containers obtained by repeating the operation using 10.0 ml of a
in which the air has been displaced by nitrogen or other 0.0018 per cent w/v solution of salbutamol sulphate.
suitable inert gas. Storage. Store protected from light.
Labelling. The label states the strength in terms of the Labelling. The label states the strength in terms of the
equivalent amount of salbutamol in a suitable dose-volume. equivalent amount of salbutamol in a suitable dose-volume.

Salbutamol Syrup
Salbutamol Tablets
Albuterol Sulphate Syrup; Salbutamol Sulphate Syrup
Albuterol Sulphate Tablets; Salbutamol Sulphate Tablets
Salbutamol Syrup contains Salbutamol Sulphate equivalent
to not less than 90.0 per cent and not more than 110.0 per cent Salbutamol Tablets contain not less than 90.0 per cent and not
of the stated amount of salbutamol, C 13HZ1 N03 • more than 110.0 per cent ofthe stated amount of salbutamol,
C13Hz IN03 •
Usual strength. The equivalent of2 mg ofsalbutamol in 5 ml.
(1 mg of Salbutamol Sulphate is approximately equivalent to Usual strengths. The equivalent of2 mg; 4 mg ofsalbutamol.
830 f.!g ofsalbutamol). (1 mg of Salbutamol Sulphate is approximately equivalent to
830 f.!g ofsalbutamol).
Identification
Identification
A. To 5 ml add 50 ml ofa 2 per cent w/v solution of borax, 1 ml
of a 3 per cent w/v solution of 4-aminophenazone, 10 ml of a A. Carry out the method described under Related substances
2 per cent w/v solution ofpotassiumferricyanide and 10 ml of applying separately to the plate 2 f.!l of each ofthe following
chloroform. Shake and allow to separate; an orange-red colour solutions. For the test solution shake a quantity of the
is produced in the chloroform layer. powdered tablets containing 10 mg of salbutamol with 10 ml
of methanol (80 per cent) and filter. The reference solution
B. To 5 ml add sufficient 1 M sodium hydroxide to make the
containS 0.12 per cent 'ii/v ofSalbutamol sulphate RS The
solution alkaline, add 1 ml of alkaline borate buffer pH 9.2
principal spot in the chromatogram obtained with the test
and 1 ml ofa 0.04 per cent w/v solution of2, 6-dichloroquinone
solution corresponds to that in the chromatogram obtained
chlorimide in ethanol (95 per cent); a blue colour develops.
with the reference solution.
Tests B. Shake a quantity ofthe powdered tablets containing 8 mg

pH (2.4.24).3.4 to 4.5.
ofsalbutalIlol witll50 1Il1 ofa 2per cent vv!v
solution oflJortix,
add 1 m1 of a 3 per cent w/v solution of 4-aminophenazone,
Other tests. Complies with the tests stated under Oral Liquids. 10 ml of a 2 per cent w/v solution of potassium ferricyanide

2088

www.webofpharma.com
IP 2010 SALICYLIC ACID

and 10 ml of chloroform. Shake and allow to separate; an flow rate. 2 ml per minute,
orange-red colour is produced in the chloroform layer. spectrophotometer set at 276 nm,
injection volume. 20 /-Ll.
C. Shake a quantity of the powdered tablets containing 4 mg
of salbutamol with 10 ml of water and filter; the filtrate gives The test is not valid unless resolution between two principal
the reactions of sulphates (2.3.1). peaks in the chromatogram obtained with reference solution
(b) is at least 1.5.
Tests Calculate the content ofC,3H2IN03 in the tablet.
Related substances. Determine by thin-layer chromatography Other tests. Comply with the tests stated under Tablets.
(2.4.17), coating the plate with silica gel G
Assay. Detelmine by liquid chromatography (2.4.14).
Mobile phase. A mixture of 50 volumes of ethyl acetate,
30 volumes of 2-propanol, 16 volumes of water and 4 volumes
Test solution. Shake 10 tablets or a sufficient number oftablets
containing 5.0 mg ofsalbutamol with about 60 ml of water for
of strong ammonia solution.
1 hour, add sufficient water to produce 250.0 ml, mix and
Test solution. Shake a quantity of the powdered tablets centrifuge 10 ml ofthe mixture and use the supernatant liquid.
containing 10 mg of salbutamol with 1 ml of water for
15 minutes, centrifuge and use the supernatant liquid. Reference solution (a). A 0.0024 per cent wlv of salbutamol
sulphate RS in water..
Reference solution. A 0.006 per cent wlv solution of
salbutamol sulphate RS in water. Reference solution (b). A solution containing 0.0024 per cent
wlv of 2-tert-butylamino-I-(4-hydroxy-3-methylphenyl)
Apply to the plate 20 /-Ll of each solution. After development, ethanol sulphate RS and 0.0024 per cent wlv of salbutamol
dry the plate in air until the odour of the solvent is no longer sulphate RS in methanol (10 per cent).
detectable, place it for a few minutes in an atmosphere saturated
with diethylamine and spray with diazotised sulphanilic acid Follow the chromatographic procedure described under
solution. Any secondary spot in the chromatogram obtained Uniformity ofcontent. The test is not valid unless the resolution
with the test solution is not more intense than the spot in the between the two principal peaks in the chromatogram obtained
chromatogram obtained with the reference solution. Ignore with reference solution (b) is at least 1.5.
any pink spot near the line of application. Calculate the content ofC 13 H 2I N0 3in the tablets.
Uniformity of content. Comply with the test stated under Storage. Store protected from light.
Tablets.
Labelling. The label states the strength in terms of the
Determine by liquid chromatography (2.4.14). equivalent amount of salbutamol.
Test solution. Add 50 ml of water to one tablet, shake for
1 hour, add sufficient water to produce 100.0 ml, mix and
centrifuge. Dilute further with water, ifnecessary, to produce
a solution containing 0.002 per cent wlv of salbutamol. Salicylic Acid
Reference solution (a). A 0.0024 per cent. wlv of salbutamol
sulphate RS in water. COOH
Reference solution (b). A 0.048 per cent wlv of 2-tert- ~OH
butylamino-l-(4-hydroxy-3-methylphenyl) ethanol sulphate
RS and 0.048 per cent wlv of salbutamol sulphate RS in
methanol (10 per cent).
V
C7H60 3 Mol. Wt. 138.1
Chromatographic system
Salicylic Acid is 2-hydroxybenzoic acid.
a stainless steel column 20 cm x 5 mm, packed with
spherical particles ofsilica, 5 /-Lm in diameter, the surface Salicylic Acid contains not less than 99.0 per cent and not
of which has been modified with chemically-bonded more than 100.5 per cent of C7H60 3, calculated on the dried
nitrile groups (such as Spherisorb CN), basis.
mobile phase: a mixture of 65 volumes of water,
Category. Keratolytic.
30 volumes of 0.05 M ammonium acetate and 5 volumes
of 2-propanol, the pH of the mixture being adjusted to Description. White or colourless, acicular crystals or a white,
4.5 with glacial acetic acid, crystalline powder.

2089

www.webofpharma.com
SALICYLIC ACID IP 2010

Identification Salmeterol Xinafoate


Test A may be omitted if tests Band C are carried out. Test B OH
may be omitted if tests A and C are carried out. OH H ~ ~COOH
HO~N~o~
A. Determine by infrared absorption spectrophotometry (2.4.6). HO)lJ .
VV
Compare the spectmm with that obtained with salicylic acid
RS or with the reference spectIUm of salicylic acid. C2sH37N04, C II H s0 3 Mol. Wt. 603.7
B. Dissolve about 30 mg in 5 ml of 0.05 M sodium hydroxide, Salmeterol Xinafoate is (RS)-4-hydroxy-d-[[[6-(4-phenyl-
neutralise if necessary and dilute to 20 ml with water. 1 ml of butoxy)hexyl]amino]methyl]-1 ,3-benzenedimethanol
the solution gives reaction A of salicylates (2.3.1). I-hydroxy-2-naphthoate.
C.Meltingpoint.158°to 161°(2.4.21). Salmeterol Xinafoate contains not less than 97.0 per cent and
not more than 102.0 per cent ofC36H4sN07, calculated on the
Tests anhydrous basis.
Appearance of solution, Dissolve 1.0 gin 10 ml of ethanol Category. Bronchodilator.
(95 per cent). The resulting solution is clear, and colourless
Description. A white to off-white powder.
(2.4.1).
Heavy metals (2.3.13). Dissolve 2.0 g in 15 ml of ethanol Identification
(95 per cent) and add 5 ml of water. 12 ml of the resulting Determine by infrared absorption spectrophotometry (2.4.6).
solution complies with the limit test for heavy metals, Method Compare the spectIUm with that obtained with salmeterol
D (20 ppm). Use lead standard solution (100 ppm Ph) diluted xinafoate RS or with the reference spectIUm of salmeterol
with a mixture of 3 volumes of ethanol (95 per cent) and xinafoate.
1 volume of water to contain 2 Ilg ofPb per ml to prepare the
standard. Tests
Iron (2.3.14). Boil 12.0 g with 14 rnl of dilute ammonia solution Related substances. Determine by liquid chromatography
and 35 ml of water. Cool and adjust the pH 5.0 to 6.0 by the (2.4.14).
dropwise addition of dilute ammonia solution or dilute
Test solution. Dissolve 50 mg of the substance under
sulphuric acid and dilute to 50 ml with water, if necessary.
examination in 50 ml ofthe mobile phase.
Any pink colour produced is not more intense than that
obtained by boiling 2.0 g with 1 ml of iron standard solution Reference solution. A 0.002 per cent w/v solution ofsalmeterol
(20 ppm Fe), 2 ml of dilute ammonia solution and 45 ml of xinafoate RS in the mobile phase.
water, adjusting the pH 5.0 to 6.0 and diluting to 50 ml with Chromatographic system
water (2 ppm). . - fl stainless steel column 25 cm x 4.6 mm, packed with
Chlorides (2.3.12). Dissolve 5.0 g in 50 ml ofboiling distilled octadecylsilane bonded to porous silica (5 Ilm),
water, cool and filter (solution A). 20 ml ofsolution A complies - mobile phase: a mixture of60 volumes ofa buffer solution
with the limit test for chlorides (125 ppm). prepared by dissolving 1.36 g ofpotassium dihydrogen
phosphate in 1000 ml of water and adjusting the pH to
Sulphates (2.3.17). 7.5 ml ofsolution A complies with the limit
7.0 with triethylamine and 40 volumes of acetonitrile,
test for sulphates (200 ppm).
- flow rate. 1 ml per minute,
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined - spectrophotometer set at 220 TIm,
on 2.0 g. - injectionvolume. 10 Ill.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Inject the reference solution. The test is not valid unless the
on 1.0 g by drying over phosphorus pentoxide in a desiccator. tailing factor is not more than 2.0 and the column efficiency in
not less than 4500 theoretical plates.
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of
ethanol (95 per cent), add 20 ml of water and titrate with Inject the test solution. Any individual impurity is not more
0.1 M sodium hydroxide, using phenol red solution as than 0.5 per cent and the sum of all the impurities found is not
indicator, until a reddish violet colour is obtained. more than 1.0 per cent.
1 ml of 0.1 M sodium hydroxide is equivalenttoO:01381 gaf Heavy metals (2.3.13). 1 g complies with limit test for heavy
C7H 60 3 • metals, Method B (20 ppm).

Storage. Store protected from light. Sulphated ash (2.3.18). Not more than 0.1 per cent.

2090

www.webofpharma.com
IP 2010 SALMETEROL AND FLUTICASONE PROPIONATE POWDER FOR INHALATION

Water (2.3.43). Not more than 0.5 per cent, determined on Reference solution (b). A solution containing 0.5 mg of
0.5g. fluticasone propionate per ml prepared by dissolving 10 mg of
Assay. Weigh accurately about 0.2 g and dissolve in 50 ml of
jluticasone propionate RS in 10 ml acetonitrile and adding
sufficient ofthe mobile phase to produce 20 ml.
glacial acetic acid. Titrate with 0.1 M perchloric acid,
determining the end-point potentiometrically (2.4.25). Cany Reference solution (c). Dilute suitable volumes of reference
out a blank titration. solution (a) and reference solution (b) with the mobile phase
to obtain a solution containing 5 Ilg Salmeterol and 50 Ilg
1 m1 of 0.1 M perc/doric acid is equivalent to 0.06038 g of
Fluticasone propionate per ml or quantities as per the label
C36ILsN07 •
claim.
Storage. Store protected from light.
Chromatographic system
a stainless steel column 15 cm x 3.9 mm, packed with
octylsilyl silica gel (5 Ilm),
Salmeterol and Fluticasone Propionate column temperature 40°,
- mobile phase: a mixture of45 volumes ofa buffer solution
Inhalation prepared by dissolving 1.15 g ammonium dihydrogen
Salmeterol and F1uticasone Propionate Inhalation is a orthophosphate to 1000 ml of water and adjusting the
suspension ofmicrofine Salmeterol Xinafoate and Fluticasone pH to 3.5 with orthophosphoric acid, 25 volumes of
Propionate in a suitable liquid filled in a suitable pressurised acetonitrile and 30 volumes of methanol,
container. It may contain suitable pharmaceutical aids such as flow rate. 2 ml per minute,
surfactants, stabilizing agents. - spectrophotometer set at 220 nm,
- inject 200 Ill.
Salmeterol and Fluticasone Propionate Inhalation delivers not
less than 80.0 per cent and not more than 120.0 per cent ofthe Inject reference solution (c). The test is not valid unless the
stated amounts of salmeterol, C 2s H 37N0 4 and fluticasone column efficiency for salmeterol and fluticasone propionate
propionate, C2sH31F30SS, per inhalation by actuation of the peak is not less than 1000 and 2500 theoretical plates
valve. respectively and the tailing factor is not more than 2.0 for each
peak and the relative standard deviation for replicate injections
Identification for each component is not more than 2.0 per cent.

In the Assay, the principal peaks in the chromatogram obtained Inject the test solution and reference solution (c).
with test solution correspond to the peaks in the Calculate the contents of C2s H37N0 4 and C2sH31F30SS in the
chromatogram obtained with reference solution (c). solution and the contents of C2sH37N04 and C2sH31FjOsS
delivered per actuation of the valve.
Tests
Determine the contents of the active ingredients a second
Other tests. Comply with the tests stated under Inhalation and third time by repeating the procedure on the middle ten
Preparations (Pressurised Metered-dose Preparations). and on the last ten successive combined actuations of the
valve. For each of the three determinations the average
Follow the procedure described under Assay with suitable
contents ofC 2s H37N04and C2sH31F30SS delivered per actuation
dilution of the reference solution wherever the amount of
of the valve meet the requirements.
active substance is to be determined in any test.
Storage. Store protected from moisture at a temperature not
Assay. CaD')' out the test for Content of active ingredient
exceeding 30°.
delivered per actuation stated under Inhalation Preparations
(Pressurised Metered-dose Preparations). Labelling. The label states the amounts of active ingredients
delivered per inhalation.
Determine by liquid chromatography (2.4.14).
Test solution. Prepare using the mobile phase as described
under the test for Content of active ingredient delivered per Salmeterol and Fluticasone Propionate
actuation stated under Inhalation Preparations (Pressurised
Metered-dose Preparations). Powder for Inhalation
Reference solution (a). A solution containing 0.5mg of Salmeterol and Fluticasone Propionate Powder for Inhalation
salmeterol per ml prepared by dissolving 10 mg of salmeterol consists of Fluticasone Propionate and Salmeterol Xinafoate
xinafoate RS in 10 ml acetonitrile and adding sufficient ofthe in microfine powder either alone oradmixed with Lactose in a
mobile phase to produce 20 ml. pre-metered unit for use in a suitable powder inhaler.

2091

www.webofpharma.com
SALMETEROL AND FLUTICASONE PROPIONATE POWDER FOR INHALATION IP 2010

Salmeterol and Fluticasone Propionate Powder for Inhalation Inject reference solution(c). The test is not valid unless the
contains not less than 90.0 per cent and not more than column efficiency determined from the salmeterol and
125.0 per cent ofthe stated amounts ofsalmeterol C2sH37N04 fluticasone propionate peak is not less than 1000 and 2500
and fluticasone propionate, C2sH31F30SS per unit dose. theoretical plates respectively, the tailing factor for each of
salmeterol and fluticasone propionate peaks is not more than
2.0 and the relative standard deviation for replicate injections
Identification
is not more than 2.0 per cent.
In the Assay, the principal peaks in the chromatogram obtained Inject the test solution and reference solution (c).
with the test solution correspond to the peaks in the
chromatogram obtained with reference solution (c). Calculate the contents ofC2sH37N04 and C 2s H 31 F 30SS per unit.
Storage. Store protected from moisture, at temperature not
Tests exceeding 30°.
Labelling. The label states the quantities ofactive ingredients
Other tests. Complies with the tests stated under the Inhalation
per pre-metered unit.
Preparations (Powders for Inhalation).

Follow the procedure described under Assay with suitable


dilution of the reference solution wherever the amount of
active substance is to be determined in any test.
Saquinavir
Assay. Determine by liquid chromatography (2.4.14).

Test solution. Dissolve a quantity of the mixed contents of


20 capsules in sufficient ofthe mobile phase to get a solution
containing 5 Ilg per ml ofSalmeterol.

Reference solution (a). A solution containing 0.5 mg of


salmeterol per ml prepared by dissolving 10 mg of salmeterol
xinafoate RS in 10 ml acetonitrile and adding sufficient ofthe
mobile phase to produce 20 ml.

Reference solution (b). A solution containing 0.5 mg of


fluticasone propionate per ml prepared by dissolving 10 mg Mol. Wt. 670.8
ofjluticasone propionate RS in 10 ml acetonitrile and adding
Saquinavir is (S)-N-[( fJS)-a-{(IR)-2-[(3S,4aS,8aS)-
sufficient ofthe mobile phase to produce 20 ml.
3-(tert-butylcarbamoyl)octahydro-2(1H)-isoquinolyl]-
Reference solution (c). Dilute suitable volumes of reference 1-hydroxyethyl}phenethyl]-2-quinaldamidosuccinamide.
solution (a) and reference solution (b) with the mobile phase Saquinavir contains not less than 98.5 per cent and not more
to obtain a solution containing 5 Ilg Salmeterol and 50 Ilg than 101.0 per cent ofC38HsoN60s, calculated on the anhydrous
Fluticasone propionate per ml or quantities as per the label basis.
claim..
Category. Antiretroviral.
Chromatographic system
Dose. 1 g twice daily.
- a stainless steel column 15 cm x 3.9 mm, packed with.
octylsilyl silica gel (5 Ilm), Description. A white crystalline powder.
column temperature 40°,
Identification
mobile phase: a mixture of45 volumes ofa buffer solution
prepared by dissolving 1.15 g ammonium dihydrogen A. Determine by infrared absorption spectrophotometry (2.4.6),
orthophosphate to 1000 ml of water and adjusting the Compare the spectrum with that obtained with saquinavir RS
pH to 3.5 with orthophosphoric acid, 25 volumes of or with the reference spectrum of saquinavir.
acetonitrile and 30 volumes of methanol, B. In the Assay, the principal peak in the chromatogram
flow rate. 2 ml per minute, obtained with the test solution corresponds to the peak due
spectrophotometer set at 220 nm, to saquinavir in the chromatogram obtained with reference
injection volume. 200 Ill. solution (a).

2092

www.webofpharma.com
IP 2010 SAQUINAVIR MESYLATE

C. When examined in the range 220 nm to 350 nm (2.4.7), a Inject reference solution (b). The relative retention times are
0.002 per cent w/v solution in methanol shows absorption about 0.89 for saquinavir-related compound A ami about 1.0
maxima at about 239 nm and 290 nm. for saquinavir. The test is not valid unless the resolution
between the peaks due to saquinavir-related compound A
Tests and saquinavir is not less than 1.5, the column efficiency
determined from the saquinavir peak is not less than 500
Specific optical rotation (2.4.22). -50.00 to-55.0°, determined theoretical plates and the relative standard deviation for
in a 0.5 per cent w/v solution in methanol. replicate injections is not more than 2.0 per cent.
Related substances. Determine by liquid chromatography Separately inject the test solution and reference solution (a).
(2.4.14), as described in the Assay using the test solution and Record the chromatograms up to three times the retention
reference solution (c). time of the principal peak and measure the responses for the
Inject reference solution (c). Calculate the amount of related principal peak.
substance by area normalisation method. In the chromatogram Calculate the content of C3sHsoN60s.
obtained with the test solution, the area of any peak other
Storage. Store protected from light.
than the principal peak is not greater than the area of the
principal peak obtained with reference solution (c) (0.1 per
cent) and the sum of the areas of all such peaks is not greater
than 5 times the area ofthe principal peak in the chromatogram
obtained with reference solution (c) (0.5 per cent). Saquinavir Mesylate
Heavy metals (2.3.13). 2.0 g complies with the limit tests for
heavy metals, Method B (10 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Water (2.3.43). Not more than 2.0 per cent, determined on
0.5g.
Assay. Detennine by liquid chromatography (2.4.14).
Test solution. Dissolve 25.0 mg of the substance under
examination in 100.0 ml ofthe mobile phase. C3sHsoN60s,C~03S Mol. Wt. 767.0
Reference solution (a). A 0.025 per cent w/v solution of Saquinavir mesylate is (S)-N-[( as)-Cf..-{ (IR)-2-[(3S,4aS,8aS)-
saquinavir RS in the mobile phase. 3-(tert-butylcarbamoyl)octahydro-2(1H)-isoquinolyl]-
I-hydroxyethyl}phenethyl]-2-quinaldamidosuccinamide
Reference solution (b). Dissolve suitable quantities of
methanesulphonate.
saquinavir-related compound A RS and saquinavir RS in the
mobile phase to obtain a solution containing 2 /lg per ml of Saquinavir Mesylate contains not less than 98.5 per cent and
saquinavir-related compound A and 0.25 mg per ml of not more than 101.0 per cent ofC3sHsoN60s, CH40 3S, calculated
saquinavir. on the anhydrous basis.
Reference solution (c). A 0.000025 per cent w/v solution of Category. Antiretroviral.
saquinavir RS in the mobile phase. Dose. 1 g twice daily.
Chromatographic system Description. A white or almost white powder.
a stainless steel column 25 cm x 4.6 mrn, packed with
octadecylsilane bonded to porous silica (5 /lm), Identification
mobile phase: a mixture· of 20 volumes of methanol,
50 volumes of acetonitrile and 30 volumes of a buffer A. Determine by infrared absorption spectrophotometry (2.4.6),
prepared by dissolving 4 g of sodium dihydrogen Compare the spectrum with that obtained with saquinavir
phosphate in 1000.0 ml of water to which I ml of mesylate RS or with the reference spectrum of saquinavir
diethylamine and 1 g of sodium octane sulphonate has mesylate.
been added, B. In the Assay, the principal peak in the chromatogram
flow rate. I ml per minute, obtained with the test solution corresponds to the peak due
spectrophotometer set at 210 urn, to saquinavir mesylate in the chromatogram obtained with
injection volume. 20 Ill. reference solution (a).

2093

www.webofpharma.com
SAQUINAVIR MESYLATE IP 2010

Tests triethylamine with water to make 1000 ml and adjusting


the pH of the solution to 25 with phosphoric acid,
Specific optical rotation (2.4.22). -66.8° to-69.6°, determined flow rate. 1 ml per minute,
in a 0.5 per cent w/v solution in methanol at 436 run. spectrophotometer set at 210 nm,
Related substances. Determine by liquid chromatography injection volume. 20 ).Ll.
(2.4.14), as described in the Assay using the test solution and
Inject reference solution (b). The relative retention times are
reference solution (c).
about 0.89 for saquinavir-related compound A and about 1.0
Inject reference solution (c). Calculate the amount of related for saquinavir mesylate. The test is not valid unless the
substances by area normalisation method. In the resolution between the peaks due to saquinavir related
chromatogram obtained with the test solution, the area of any compound A and saquinavir mesylate is not less than 1.5, the
peak other than the principal peak is not greater than the area column efficiency determined from the saquinavir mesylate
of the principal peak in the' chromatogram obtained with peak is not less than 500 theoretical plates and the relative
reference solution (c) (0.1 per cent) and the sum ofthe areas of standard deviation for replicate injections is not more than
all such peaks is not greater than 5 times the area of the 2.0 per cent.
principal peak in the chromatogram obtained with reference
Separately inject the test solution and reference solution (a).
solution (c) (0.5 per cent).
Record the chromatograms upto three times the retention time
Heavy metals (2.3.13). 1.0 g complies with the limit test for of the principal peak and measure the responses for the
heavy metals, Method B (20 ppm). principal peak.
Methanesulphonic acid. 11.9 to 13.1 per cent w/w, calculated Calculate the content ofC3sHsoN60s, CH40 3S.
on the anhydrous basis, determined by the following method.
Storage. Store protected from light.
Weigh accurately about 0.1 g of the substance under
examination, dissolve in 50 ml of methanol. Titrate with
0.1 M sodium hydroxide, determining the end-point potentio-
metrically (2.4.25). Carry out a blank titration.
Saquinavir Mesylate Tablets
1 ml of 0.1 M sodium hydroxide is equivalent to 0.00961 g of
CH3S03H. Saquinavir Mesylate Tablets contain not less than 90.0 per
cent and not more than 110.0 per cent ofthe stated amount of
Sulphated ash (2.3.18). Not more than 0.1 per cent.
saquinavir C3sHsoN60s.
Water (2.3.43). Not more than 1.0percent,determinedon 0.5 g.
NOTE -Pelform the tests and assay using low-actinic
Assay. Determine by liquid chromatography (2.4.14). glassware.
Test solution. Dissolve 25.0 mg of the substance under Usual strength. 200 mg.
examination in 100.0 ml ofthe mobile phase.
Reference solution (a). A 0.025 per cent w/v solution of Identification
saquinavirmesylate RS in the mobile phase. A. In the Assay, the principal spot in the chromatogram
Reference solution (b). Dissolve suitable quantities of obtained with the test solution corresponds -to that in the
saquinavir-related compound A RS and saquinavir mesylate chromatogram obtained with the reference solution.
RS in the mobile phase to obtain a solution containing 2 ).Lg B. When examined in the range 200 nm to 400 run (2.4.7),1 ml
per ml ofsaquinavir- related compound A and 0.25 mg per ml of a 0.1 per cent w/v solution in methanoi diluted to 100 ml
of saquinavir mesylate. with citrate buffer pH 3.0 (see under Dissolution), shows
Reference solution (c). A 0.000025 per cent w/v solution of absorption maxima at the same wavelengths as shown by the
saquinavir mesylate RS in the mobile phase. reference solution.
NOTE - Store the btif.fer solution protectedfrom light. Make
Tests
adjustments ifnecessaly for system suitability.
Chromatographic system Dissolution (2.5.2).
- a stainless steel column 25 cm x 4.6 mm, packed with Apparatus. No 1
octadecylsi1ane bonded to porous silica (5 ).Lm), Medium. 900 ml of citrate bufferpH3.0 prepared by dissolving
mobile phase: a mix.ture of 25 volumes of 5.82 mg of anhydrous dibasic sodium phosphate and 16.7 mg
tetrahydrofuran, 5 volumes of acetonitrile and of citiric acid monohydrate in 1000 ml of water and adjusting
17 volumes of a buffer prepared by mixing 10. ml of the pH to 3.0 with orthophosphoric acid.

2094

www.webofpharma.com
IP 2010 SECNIDAZOLE

Speed and time. 50 rpm and 45 minutes. Calculate the content of C3sHsoN60S in the tablet.
Withdraw a suitable volume ofthe medium and filter promptly. Other tests. Comply with the tests stated under Tablets.
Dilute the filtrate, ifnecessary, with the medium. Measure the
Water (2.3.43). Not more than 6.0 per cent, determined on
absorbance (2.4.7) ofthe resulting solution at the maximum at
0.5 g.
about 240 nm. Calculate the content of C38HsoN60S in the
medium from the absorbance obtained from a solution of Assay. Determine by liquid chromatography (2.4.14).
lmown concentration of saquinavir mesylate RS. Test solution. Weigh and powder 20 tablets. Weigh accurately
D. Not less than 75 per cent of the stated amount of a quantity of the powder containing 100 mg of saquinavir,
C3sHsoN60S. dissolve in 100.0 ml ofmobile phase and filter. Dilute 5.0ml of
the filtrate to 20.0 ml with the mobile phase.
Related substances. Determine by liquid chromatography
(2.4.14). Reference solution. A 0.1 per cent w/v solution of saquinavir
mesylate RS in the mobile phase. Dilute 5.0 ml ofthe solution
Test solution. Weigh and powder 20 tablets Weigh accurately
to 20.0 ml with the mobile phase.
a quantity of the powder containing 100 mg of saquinavir in
100 ml ofthe mobile phase and filter. Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
Reference solution (a). A 0.1 per cent w/v solution of
octadecylsilane bonded to porous silica (5 flm),
saquinavir mesylate RS in the mobile phase.
- mobile phase: a mixture of 14 volumes of a solution
Reference solution (b). Dilute 1 ml ofreference solution (a) to prepared by diluting 10.0 ml of triethylamine to 1000 ml
100 ml with the mobile phase. with water, adjusting the pH to 2.5 with orthophosphoric
Chromatographic system
acid and filtering, 5 volumes of tetrahydrofitran and
1 volume of acetonitrile.
a stainless steel column 25 cm x 4.6 mm, packed with
- flow rate. 1.5 ml per minute,
octadecylsilane bonded to porous silica (5 flm),
- spectrophotometer set at 210 nm,
- mobile phase: a mixture of 14 volumes of triethylamine
- injection volume. 20 fll.
phosphate solution prepared by diluting 10 ml of
triethylamine to 1000 ml with water and adjusting the Inject the reference solution. The test is not valid unless the
pH to 2.5 with orthophosphoric acid, 5 volumes of tailing factor is not more than 2.0, the column efficiency in not
tetrahydrofuran and 1 volume of acetonitrile, less than 2000 theoretical plates and the relative standard
- flow rate. 1 ml per minute, deviation for replicate injections is not more than 2.0 per cent.
- spectrophotometer set at 210 nm,
Inject the test solution and the reference solution.
- injection volume: 20 ~Ll.
Calculate the content of C3sHsoN60S in the tablets.
Inject reference solution (a). The test is not valid unless the
column efficiency is not less than 3000 theoretical plates and Storage. Store protected from moisture.
the tailing factor is not more than 2.0. Labelling. The label states the strength in terms of the
Inject the test solqtion and reference solution (b). Run the equivalent amount of saquinavir.
chromatogram for 5 times the retention time (about 12 minutes)
of the principal peale. In the chromatogram obtained with the
test solution, the area of any secondary peak is not more than
0.2 times the area of the peak in the chromatogram obtained Secnidazole
with the reference solution (b) (0.2 per cent) and the sum of
areas of all the secondary peaks is not more than the area of
the peak in the chromatogram obtained with the reference
solution (b) (1.0 per cent).
Uniformity of content. Comply with the test stated under
Tablets.
Determine by liquid chromatography (2.4.14), as described
under Assay.
Mol. Wt. 185.2
Test solution. Disperse one tablet in 500 ml ofthe mobile phase
and filter. Dilute 5 ml of the filtrate to 20 ml with the mobile Secnidazole is (RS)-I-(2-methyl-5-nitroimidazol-l-yl)propan-
. phase. 2-01.

2095

www.webofpharma.com
SECNIDAZOLE IP 2010

Secnidazole contains not less than 98.0 per cent and not more is not more than 5 times the area of the peak in the
than 101.0 per cent of C7HIIN303, calculated on the anhydrous chromatogram obtained with the reference solution (a)
basis. (0.5 per cent). Disregard any peak which is 0.1 times the area
Category. Antiamoebic. of the principal peak in the chromatogram obtained with
reference solution (a) (0.01 per cent).
Dose. Adults - 2g; Children - 30 mg per kg as single dose.
Heavy metals (2.3.13).1.0 g complies with the limit test for
Description. A white to yellowish white, crystalline powder. heavy metals, Method A (20 ppm).

Identification Sulphated ash (2.3.18). Not more than 0.1 per cent.
Water (2.3.43). 4.0 to 5.0 percent, determined on 0.4 g.
A. When examined in the range 230 nm to 360 nm (2.4.7), a
0.001 per centw/v solution in 0.1 Mhydrochloric acid shows Assay. Determine by liquid chromatography (2.4.14).
an absorption maximum at about 277 nm, 0.325 to 0.355.
Test solution. Dissolve 25 mg of the substance under
B. Heat about 10 mg in a water-bath with 10 mg ofzincpowder, examination in 25.0 ml ofthe mobile phase. Dilute 5.0 ml ofthe
1 ml of water and 0.25 ml of 2 M hydrochloric acid for solution to 100.0 ml with the same solvent.
5 minutes and cool. The solution gives the reaction ofprimary
aromatic amines (2.3.1).
Reference solution. A 0.005 per cent w/v solution of
secnidazole RS in mobile phase.
Tests Chromatographic system
Appearance of solution. A 5 per cent w/v solution in a stainless steel column 25 cm x 4.6 mm packed with
1 M hydrochloric acid is not more opalescent than octadecylsilane bonded to porous silica (5 Ilm),
opalescence standard OS2 (2.4.1) and not more intensely mobile phase: a mixture of35 volumes of methanol and
coloured than reference solution GYS4 (2.4.1). 65 volumes of 0.14 per cent w/v solution of potassium
dihydrogen phosphate,
Related substances. Determine by liquid chromatography
flow rate. 1 ml per minute,
(2.4.14).
spectrophotometer set at 318 nm,
Test solution. Dissolve 50 mg of the substance under - injection volume. 20 Ill.
examination in 100.0 ml ofthe mobile phase.
Inject the reference solution. The test is not valid unless the
Reference solution (a). Dilute 1.0 ml of the test solution to tailing factor is not more than 2.0 and the relative standard
100.0 ml with mobile phase. Dilute 1.0 ml of the solution to deviation for replicate injections is not more than 2.0 per cent.
10.0 ml with the same solvent.
Inject the test solution and the reference solution.
Reference solution (b). A solution containing 0.005 per cent
w/v each of 2-methyl-5-nitroimidazole RS and secnidazole Calculate the content of C7HIIN303.
RS in the mobile phase. Dilute 1.0 ml of the solution to Storage. Store protected from light and moisture.
100.0 ml with the same solvent.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 Ilm), Secnidazole Tablets
- mobile phase: a mixture of35 volumes of methanol and Secnidazole Tablets contain not less than 95.0 per cent and
65 volumes of 0.14 per cent w/v solution of potassium not more than 110.0 per cent of the stated amount of
dihydrogen phosphate, secnidazole, C7HIIN303'
- flow rate. 1 ml per minute,
- spectrophotometer set at 318 nm, Usual strengths. 1 g; 2 g.
injection volume. 10 Ill.
Identification
Inject reference solution (b). Tlie test is not valid unless the
resolution between 2-methyl-4-nitroimidazole and secnidazole In the Assay, the principal peak in the chromatogram obtained
is not less than 1.5. with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution.
Inject the test solution and reference solution (a). In the
chmmaj:ogram obtained with the test solution, the area ofany Tests
secondary peak is not more than 2 times the area ofthe peak in
the chromatogram obtained with reference solution (a) Dissolution (2.5.2).
(0.2 per cent) and the sum of areas of all the secondary peaks Apparatus. No.1,

2096

www.webofpharma.com
IP 2010 SERRATIOPEPTIDASE

Medium. 900 ml of 0.1 M hydrochloric acid. spectrophotometer set at 228 nm,


Speed and time. 100 rpm and 30 minutes. injection volume. 20 Ill.
Withdraw a suitable volume ofthe medium and filter promptly Inject the reference solution. The test is not valid unless the
through a membrane filter disc having an average pore diameter column efficiency is not less than 2000 theoretical plates, the
not greater than 1.0 Ilm, rejecting the first I ml of the filtrate. tailing factor is not more than 2.0 and the relative standard
Dilute the filtrate, ifnecessary, with the same solvent. Measure deviation of replicate injections is not more than 2.0 per cent.
the absorbance of the resulting solution at the maximum at Inject the test solution and the reference solution.
about 277 urn (2.4.7). Calculate the content ofC7HIIN303 in the
medium from the absorbance obtained from a solution of Calculate the content ofC7HIIN303.
known concentration of secnidazole RS in the same medium. Storage. Store protected from light and moisture.
D. Not less than 80 per cent ofthe stated amount ofC7HIIN303' Labelling. The label states the strength of secnidazole.
Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Weigh accurately a quantity ofpowdered tablets Serratiopeptidase
containing 50 mg ofSecnidazole, disperse in 100 ml ofmobile Serratiopeptidase is a proteolytic enzyme for oral use which is
phase and filter. produced by bacteria of genus Serratia.
Reference solution (a). A 0.05 per cent w/v solution of Serratiopeptidase contains not less than 2000 units and not
secnidazole RS in the mobile phase. more than 2600 units of serratiopeptidase activity per mg.
Reference solution (b). Dilute 1 ml ofreference solution (a) to Category. Prolytic enzyme.
100 ml with mobile phase.
Description. A grayish white to pale brown pow4er with
Chromatographic system as described under Assay.
characteristic odour.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in Identification
not less than 2000 theoretical plates.
Dissolve 0.4 g of serratiopeptidase in 100 ml of acetic acid
Inject the test solution and reference solution (b). In the sodium acetate buffer solution pH5.0, transfer exactly 1 mlof
chromatogram obtained with the test solution, the area of any this solution into 3 test tubes, refer them as A, Band C. Add 1
secondary peak is not more than the area of the peak in the ml of water in tube A. In tube Band C add 1 ml of 0.04 ml of
chromatogram obtained with reference solution (b) (1.0 per disodium edetate solution. Mix gently and allow them to stand
cent) and the sum of areas of all the secondary peaks is not at 4±10, for 1 hour. Add 2 ml of water in tube A and C. Add 2 ml
more than 2.0 times the area ofthe peak in the chromatogram of 0.04 M zinc chloride solution in tube B. Mix gently and
obtained with the reference solution (b) (2.0 per cent). allow them to stand at4±1 0, for 1 hour. Pipette 1 ml ofA and B
Water (2.3.43). Not more than 6.5 per cent, determined on 0.5 g. and make up the volume to 200 ml with borate bufferpH9.0.
Pipette 1 ml ofC and make up the volume to 50 ml with borate
Assay. Determine by liquid chromatography (2.4.14). bufferpH 9. O. Proceed with these solutions as sample solutions
Test solution. Weigh and powder 20 tablets. Weigh accurately as directed in the assay. The activities of solution A and Bare
a quantity ofpowdered tablet containing 50 mg ofSecnidazole, almost same and the activity ofC is not more than 5 per cent of
disperse in 100.0 ml ofmobile phase and filter.. Dilute 5.0 ml of that of activity of A.
the filtrate to 50.0 ml with mobile phase.
Tests
Reference solution. A 0.005 per cent w/v solution of
secnidazole RS in mobile phase. Arsenic (2.3.10). Dissolve 0.4 g in 50 ml of water, and add 10
Chromatographic system ml of stannated hydrochloric acid. The resulting solution
a stainless steel column 25 cm x 4.6 mm packed with complies with the limit test for arsenic (5 ppm).
octadecylsilane chemically bonded to porous silica Heavy metals (2.3.13). 1.0 g complies with the limit test for
(5Ilm), heavy metals, MethodB (50 ppm).
mobile phase: a mixture of 85 volumes of
Sulphated Ash (2.3 .18). Not more than 1.5 per cent.
0.01 M potassium dihydrogen orthophosphate and
15 volumes of acetonitrile, Loss on drying (2.4.19). Not more than 7.0 per cent, determined
flow rate. 1 ml per minute, on 1 g by drying in an Qven at 105° for 4 hours.

2097

www.webofpharma.com
SERRATIOPEPTIDASE IP 2010

Microbial contamination(2.2.9). Total viable aerobic count, minutes and filter. Take 2 ml ofthe filtrate, add 50 ml of0.6 per
not more than 100 micro-organisms perml,determined by plate cent w/v sodium carbonate solution. Add 1 ml of diluted
count. 1 ml is free from Escherichia coli. folin s reagent mix and allow stand at 37° for 30 minutes.
Measure the absorbance at 660 mn (2.4.7).
Assay. Unit defmition: one serratiopeptidase unit is defined
as the amount of enzyme required to liberate 1 I!m of free Calculate the serratiopeptidase IV/mg by using concentration
tyrosine per minute under the specified assay conditions. of tyrosine from graph considering the reaction time and
sodium borate hydrochloric acid buffer pH 9. O. Dissolve 19 dilution.
gsodium borate in 900.ml of water. Adjust the pH 9.0 with 1 Storage. Store protected from light and moisture, at temperature
M hydrochloric acid and dilute with water to 1000 ml. not exceeding 30°.
Substrate solution. Dissolve 1.2 g dried casein in 100 ml of
sodium borate hydrochloric acid buffer pH 9.0. Keep it on
boiling water-bath for 1-2 minutes to get clear solution. Cool Serratiopeptidase Tablets
and filter through cotton and dilute with sodium borate
hydrochloric acid buffer pH 9. a to 200 ml. Serratiopeptidase Tablets contain not less than 90.0 per cent
of the stated amount of Serratiopeptidase.
Protein precipitating solution. To 18 g of trichloroacetic acid,
add 30 g of sodium acetate and 20 ml of glacial acetic acid, Usual strengths. 5 mg; 10 mg.
and dilute with water to 100 ml.
Tests
Dilute folin s reagent. 1 ml of folin s reagent, add 2 ml of
water. Other tests. Comply with the tests stated under Tablets.

Reference tyrosine solution. Dissolve 10 mg of tyrosine in 1 Assay. The tyrosine units are dissolved in sodium carbonate
ml of 1 M hydrochloric ac:id and dilute to 100 ml with sodium solution, which is alkaline in nature. The Folin s ciocalteau
borate hydrochloric acid buffer pH 9.0. reagent helps in colour development where the tyrosine units
bind to the copper molecule in the reagent and causes the
Reference tyrosine curve. To 1, 2, 3, 4 and 5 ml of reference reduction of phosphomolybdate which is present in the
tyrosine solution, add 5, 4, 3, 2 and 1 ml of sodiul1~ borate reagent. There is formation of tyrosine-copper molybdate
hydrochloric acid buffer pH 9.0 respectively. Add 5 ml of coloured complex. The intensity of colour depends upon the
protein precipitating reagent in each tube. To 2 ml of these tyrosine units present which is read at 660 nm.
solutions, add 5 ml of sodium carbonate solution. Add 1 ml of
dilutedfolins reagent mix and allow to stand at 37° for 30 Disodium tetraborate buffer. Dissolve 19.0 gm of disodium
minutes, measure the absorbance at 660 nm (2.4.7). Plot a graph tetraborate in 900 ml of water, adjusted to pH 9.0 with 1 M
of I!m of tyrosine per system against the absorbance. hydrochloric acid and dilute to 1000 ml with water.

Stock test solution. Weigh about 0.1 g ofthe substance under Casein hammerstein substrate. Dissolve 1.2 gm of Casein
examination, dissolve in 100 ml ofsodium borate hydrochloric Hammerstein to 100 ml ofthe sodium tetraborate buffer. Allow
acid bufferpH 9. a(solution A). Mix and keep it for 5 minutes. the casein to dissolve and form homogeneous solution. Boil
Take 1 ml ofsolution A and dilute to 200 ml with sodium borate the solution in boiling water-bath for 2 minutes. After removing
hydrochloric acid buffer pH 9.0 (solution B). from boiling water-bath, cool the casein substrate immediately
in ice cold water. Filter this solution through cotton plug to
Test solution. Pipet 1.0 ml of stock test solution, allow to get clear solution and dilute to 200 ml with water.
stand in a water-bath at 37° for 5 minutes. Add 5 mlofsubstrate
solution shake immediately and allow to stand in water-bath
NOTE-Always add casein to the buffer while stirring. Do
at 37° for 20 minutes. Add 5 ml of protein precipitating not add buffer to casein as this will cause lumps and not go
solution. Mix and allow to stand in water-bath at 37° for 30 into solution completely and giving incorrect values.
minutes and filter. Take 2 ml ofthe filtrate, add 5 ml of 0.6 per Trichloroacetic acid reagent. Dissolve 18 gm of
cent w/v sodium carbonate solution. Add 1 ml of diluted trichloroacetic Acid, 30 gm of anhydrous sodium acetate
folin s reagent mix and allow to stand at 37° for 30 minutes. and 20 ml of glacial acetic acid in 1000 ml of water.
Measure the absorbance at 660 nm (2.4.7). NOTE-TCA should be in the crystal form and not in liquid
Blank solution. Pipet 1.0 ml of stock test solution, allow to form as this will effect the quantity weighed and thereby
stand in a 'Yater~bathat3rfor5 ll1inutes, add5 1l1l ofpr:ot~il1 interfere with the complete precipitation of casein. Use
precipitating solution, shake immediately and allow to stand Sodium acetate anhydrous because if, sodium acetate
in water-bath at 37° for 20 minutes. Add 5 ml of substrate trihydrate should be used, the trihydrate forms gives lower
solution. Mix and allow to stand in water-bath at 37° for 30 results.

2098

www.webofpharma.com
IP 2010 COLLOIDAL SILICON DIOXIDE

Diluted Folin 's Ciocalteau Reagent. Dilute 1 ml of Fo1in's


Ciocalteau reagent with 2 ml of water to get 3 ml ofthe reagent. Serratiopeptidase Units/Tablets = AI - A 2 x 176 x x1-
NOTE-Folin's Ciocalteau reagent should be ofthe protein A 3 -A 4 20
estimation grade and not the indicator grade. Folin 's reagent 100 25 100 .
- - - - x - x - x Avg.wt.mgm
. should be diluted to just before addition to the tubes since it SpI.Wt.(g) 5 2
is sensitive to light. Do not prepare this solution at the
% of LA = Serratiopeptidase Units/Tablet x 100
beginning ofthe assay.
L.A.
Tyrosine reference solution. Dissolve 160 mg of tyrosine in
100 ml of 0.2 M hydrochloric acid. Where, 176 Conversion Co-efficient of Tyrosine to
Dilute 1 ml of this solution to 100 ml with 0.2 Mhydrochloric Serratiopeptidase
acid. Use 2 ml ofthis solution for colour development. 20 Reaction time [minutes]
AI Absorbance of test solution
Trisodium phosphate buffer pH 6.8. Weigh 19 g of trisodium
phosphate and 6.4 ml of hydrochloric acid in 1000 ml of water, A2 Absorbance of blank solution
adjusted to pH 6.8. A3 Absorbance of tyrosine reference solution.
A4 Absorbance ofO.2M hydrochloric acid
Test solution. Weigh and powder 20 tablets. Disperse a quantity
L.A. = 20000 Units/tablet.
ofpowder containing about 120 mg ofSerratiopeptidase with
100 ml of trisodium phosphate btifferpH 6.8. Stir on magnetic Storage. Store protected from moisture, at a temperature not
stirrer for 1 hour. Further dilute 5.0 ml to 25 ml with 5.0percent exceeding 25°.
ammonium sulphate solution, stir for 5 minutes and filter. Labeling. The label states the strength in terms of the
Dilute 2 ml of the filtrate to 100 ml with sodium tetraborate equivalent fo Enzyme activity units of Serratiopeptidase.
btiffer. 1 ml of resulting solution is used for arialysis. Set the
water bath to 37°.
For each sample, keep four test tubes. Mark two test tubes as
TEST and two as BLANK. Add 5 ml ofCasein Su,bstrate to the
Colloidal Silicon Dioxide
tubes marked as TEST and 5 ml. ofTCA to the test tube marked ColloidalAnhydrous Silica
as BLANK. Prewarm these tubes for five minutes along with
Si02 . Mol. Wt. 60.1
the tubes containing the sample dilutions. Add 1 ml ofenzyme
solution to all these tubes and vertex the tubes for exactly 10 Colloidal Silicon Dioxide is a submicroscopic finned silica
seconds. Keep the test tubes in the water-bath at 37° for 20 prepared by the vapour-phase hydrolysis of a silicon
minutes for the reaction. After exactly 20 minutes, add 5 ml of compound.
TCA to tubes marked as TEST and 5 ml ofCasein substrate to Colloidal Silicon Dioxide contains not less than 99.0 per cent
the tube marked as BLANK. Vertex all the tubes exactly for 30 and not more than 100.5 per cent of Si02 , calculated on the
seconds. Keep the tubes in the water-bath at 37° for 30 minutes ignite basis.
for precipitation, filter.
Category. Pharmaceutical aid (tablet excipient).
NOTE- For tablets, the settlement may take a longer time
than 30 minutes. So filter the solution after the settlement is Description. A light, fine, white, amorphous powder. It has a
clearly seen (Vertex in between for better precipitation). particle size of about 15 nm.
Mark tubes as TEST, BLANK, TYROSINE and TYROSINE Identification
BLANK.
About 20 mg gives the reaction of silicates (2.3.1).
Add the following respectively;
TEST - 2 m1 ofthe TEST filtrate. Tests
BLANK- 2 ml ofBLANK filtrate. pH (2.4.24).3.5 to 5.5, determined in a suspension of 1.0 g in
30 ml of carbon dioxide-free water.
TYROSINE - 2 ml ofTYROSINE standard solution.
TYROSINE BLANK- 0.2 M hydrochloric acid. Arsenic (2.3.10). To 2.5 g contained in a round-bottomed flask
add 50 ml of 3 M hydrochloric acid and heat under a reflux
Add 5 ml of 6 per cent sodium carbonate solution to all tubes. condenser for 30 minutes. Cool, filter with the aid ofsuction
Add 1 ml of diluted Folin's reagent to all the tubes. Keep the and transfer the filtrate to a 1OO-ml volumetric flask. Wash the
tubes at 37° for 30 minutes for colour development. Measure filter with several portions of hot water and add the washings
the absorbance at about 660 nm (2.4.7). to the volumetric flask. Cool, dilute to volume with water and

2099

www.webofpharma.com
COLLOIDAL SILICON DIOXIDE IF 2010

mix. To 50.0 ml ofthe solution add 3 ml of hydrochloric acid; Sildenafil Citrate is 5-[2-ethoxy-5-(4-
the resulting solution complies with the limit test for arsenic methylpiperazinylsulphonyl)phenyl]-I-methyl-3-n-propyl-
(8 ppm). 1,6-dihydro-7H-pyrazolo[4,3-dJpyrirnidin-7-one citrate.
Heavy metals (2.3.13). Suspend 2.5 g in sufficient water to Sildenafil Citrate contains not less than 98.0 per cent and not
produce a semi-fluid slurry and dry at 140°. When the dried more than 102.0 per cent ofCzsH3sN601lS, calculated on the
substance is white, break up the mass using a glass rod, add dried basis.
25 ml of 1 M hydrochloric acid, boil gently for 5 minutes, Category. Used in erectyle dysfunction.
stirring frequently with the glass rod, centrifuge for 20 minutes
Description. A white to offwhite powder.
and filter the supernatant liquid through a membrane filter. To
the residue in the centrifuge tube add 3 ml of 2 M hydrochloric Dose. 50 mg per day.
acid and 9 ml of water, boil, centrifuge for 20 minutes and filter Identification
the supernatant liquid through the same membrane filter. Wash
the residue with small quantities of watel; combine the filtrates A. Determine by infrared absorption spectrophotometry (2.4.6).
and washings and dilute to 50.0 ml with water. To 20.0 ml of Compare the spectrum that obtained with sildenafil citrate
the solution add 50 mg of L-ascorbic acid and 1 ml of strong RS or with the reference spectrum of sildenafil citrate.
ammonia solution, neutralise with 2 M ammonia and dilute to B. Complies with the test for Citrate (2.3.1).
25 ml with water. 12 ml ofthe solution complies with the limit
test for heavy metals, Method D (25 ppm). Use lead standard Tests
solution (1 ppm Pb) to prepare the standard.
Citric acid. 28.0 per cent to 34.0 per cent.
Chlorides (2.3.12). To 1.0 g add a mixture of20 ml of 2 Mnitric
Accurately weigh about 0.2 g of the substance under
acid and 30 ml of water, heat on a water-bath for 15 minutes,
examination and transfer into a 250-ml conical flask, add 50 ml
shaking frequently, dilute to 50 ml with water if necessary,
of methanol and sonicate to dissolve, then add 50 ml of water.
filter and cool. The filtrate complies with the limit test for
Titrate with 0.1 M sodium hydroxide solution using
chlorides (250 ppm).
phenolphthalein indicator until the colour changes from
Loss on ignition (2.4.20). Not more than 5.0 per cent, colourless to pink.
determined on 0.2 g by igniting at 900° in a platinum crucible
1 ml of 0.1 M sodium hydroxide is equivalent to 0.006403 g of
for 2 hours.
C6Hs0 7•
Assay. To the residue obtained in the test for Loss on ignition Related substances. Determine by liquid chromatography
add 0.2 ml of sulphuric acid and sufficient ethanol (95 per (2.4.14).
cent) to moisten the residue completely, add 6 ml of
NOTE~Use freshly prepared solution.
hydrofluoriC acid and evaporate to dryness on a hot plate at
95° to 105°, avoiding loss from sputtering. Wash the sides of Solvent mixture. 30 volumes of water and 70 volumes of
the dish with 6 ml of hydrofluoric acid, evaporate to dryness acetonitrile.
in a well-ventilated hood, ignite at 1000°, allow to cool in a Test solution. Dissolve 50 mg of the substance under
desiccator and weigh. The difference between the weight of examination in 100.0 ml ofthe solvent mixture.
the final residue and that ofthe residue obtained in the test for Reference solution (a). A 0.05 per cent w/v solution of
Loss on ignition represents the amount of SiOz in the amount sildenafil citrate RS in the solvent mixture.
of the substance taken for the test for Loss on ignition.
Reference solution (b). Dilute 10.0 ml ofreference solution (a)
Storage. Store protected from light. to 100.0 ml with the solvent mixture. Further dilute 3.0 ml of
this solution to 100.0 ml with the solvent mixture.
Chromatographic system
Sildenafil Citrate a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 /-lm) (such as
o Hypersil ODS C18),
(COOH - mobile phase: A. dissolve 3.85 g of ammonium acetate
• HO-C-COOH
in 1000 ml of water, adjusted to pH 7.5 with ammonia
solution,
t cooH B. acetonitrile,
- a linear gradient programme using the conditions given
below,
Mol. Wt. 666.7 flow rate. 1 ml per minute,

2100

www.webofpharma.com
IP 2010 SILDENAFIL TABLETS

- spectrophotometer set at 240 nm, mobile phase: a mixture of50 volumes ofa buffer solution
injection volume. 20 /ll. prepared by dissolving 3.85 g of ammonium acetate in
Time Mobile phase A Mobile phase B 1000 ml of water, adjusted to pH 7.5 with ammonia
(in min.) (per cen v/v) (per cent v/v) solution and 50 volumes of acetonitrile,
- flow rate. 1 ml per minute,
o 65 35
spectrophotometer set at 240 nm,
15 65 35
- injection volume. 20 /ll.
30 20 80
Inject the reference solution. The test is not valid unless the
40 20 80
theoretical plates is not less than 2000, the tailing factor is not
45 65 35 more than 2.0 and the relative standard deviation for replicate
50 65 35 injections is not more than 2.0 per cent.
Inject reference solution (b). The test is not valid unless the Inject the test solution and the reference solution.
relative standard deviation for replicate injections is not more
Calculate the content ofC2sH3sN6011S.
than 5.0 per cent. The relative retention time with reference to
sildenafil for N-oxide is about 0.29 and for chlorosulphonyl is Storage. Store protected from light and moisture.
about 1.6.
Inject the test solution and reference solution (b). In the
chromatogram obtained with test solution the area ofthe peak SildenafIl Tablets
due to N-oxide impurity is not more than 1.67 times the area of
the principal peak in the chromatogram obtained with reference Sildenafil Citrate Tablets
solution (b) (0.5 per cent), the area of peak due to Sildenafil Tablets contain not less than 90.0 per cent and not
chlorosulphonyl is not more than 0.67 times the area of the more than 110.0 per cent of the stated amount of sildenafil
principal peak in the chromatogram obtained with reference citrate, C2sH3SN6011 S.
solution (b) (0.2 per cent). The area of any other secondary
peak is not more than the area of the principal peak in the Usual Strengths. 25 mg; 50 mg; 100 mg.
chromatogram obtained with reference solution (b) (0.3 per Identification
cent). The sum of areas of all the secondary peaks is not more
than 3.3 times the area ofthe principal peale in the chromatogram In the Assay, the principal peak in the chromatogram obtained
obtained with reference solution (b) (1.0 per cent). Ignore any with the test solution corresponds to the peak in the
peak corresponding to citric acid. chromatogram obtained with the reference solution.
Heavy Metals (2.3.13). 1.0 g complies with the limit test for Tests
heavy metals, Method B (20 ppm).
Dissolution (2.5.2).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Apparatus No.1,
Loss on drying (2.4.19). Not more than 1.0 per cent, determined Medium. 900 ml of O. 01 M Hydrochloric acid,
on 1.0 g by drying in oven at 105° for 3 hours. Speed and time. 100 rpm and 30 minutes.
Assay. Determine by liquid chromatography (2.4.14). Withdraw a suitable volume ofthe medium and filter. Measure
NOTE-Use freshly prepared solution. the absorbance of the filtered solution, suitably diluted with
the medium if necessary, at the maximum at about 294 nm
Solvent mixture. 30 volumes of water and 70 volumes of
(2.4.7). Calculate the content of C2sH3SN60 IIS in the medium
acetonitrile.
from the absorbance obtained from a solution of known
Test solution. Dissolve 50 mg of the substance under concentration of sildenafil citrate RS in the same medium.
examination in 50.0 ml ofthe solvent mixture. Dilute 5.0 ml of
D. Not less than 70 per cent of the stated amount of
this solution to 50.0 ml with the solvent mixture.
C2sH3SN6011 S.
Reference solution. A 0.1 per cent w/v solution of sildenafil
citrate RS in the solvent mixture. Dilute 5.0 ml ofthis solution Related substances. Determine by liquid chromatography
to 50.0 ml with the solvent mixture. (2.4.14)

Chromatographic system
Solvent mixture. 50 volumes ofmobile phase A and 50 volumes
- a stainless steel column 25 cm x 4.6 mm, packed with of methanol.
octadecylsilane bonded to porous silica (5 /lm) (such as Test solution. Weigh and powder 20 tablets. Disperse a quantity
Hypersil ODS CI8), ofpowder containing about 50 mg of sildenafil with 35 ml of

2101

www.webofpharma.com
SILDENAFIL TABLETS IP 2010

the solvent mixture, sonicate for 20 minutes and dilute to 50 ml Test solution.N'Ieigh and powder 20 tablets. Disperse a quantity
with the solvent mixture, filter. ofpowder containing about 50 mg of sildenafil with 35 mlof
the solvent mixture, sonicate for 20 minutes and dilute to 50.0
Reference solution (a). A 0.028 per cent w/v solution of
ml with the solvent mixture, filter.
sildenafil citrate RS in the solvent mixture.
Reference solution. A 0.14 per cent w/v solution of sildenajil
Reference solution (b). Dilute 5.0 ml ofreference solution (a)
citrate RS in the solvent mixture.
to 100 ml with the solvent mixture.
Chromatographic system
Chromatographic system
a stainless steel column 25 cm x 4.6 rom, packed with
a stainless steel column 25 cm x 4.6 rom, packed with
silica (5 Ilm) (Such as Zorbax Eclipse XDB C18),
octadecylsilane chemically bonded to porous silica (5
- mobile phase: a mixture of 30 volumes of solution
Ilm) (Such as Zorbax Eclipse XDB C 18),
containing 3.85 g of ammonium acetate and 1.0 ml of
- column temperature. 40°,
triethylamine in 1000 ml of water, adjusted to pH 5.5
mobile phase: A. a solution containing 3.85 g of
with acetic acid and 70 volumes of methanol,
ammonium acetate and 1.0 ml of triethylamine in 1000
flow rate. 1 ml per minute,
of water, adjusted to pH 5.5 with acetic acid,
spectrophotometer set at 294 nm,
B. methanol,
injection volume. 20 Ill.
a linear gradiept programme using the conditions given
below, Inject the reference solution. The test is not valid unless the
flow rate. 1 ml per minute, theoretical plates is not less than 2000, the tailing factor is not
spectrophotometer set at 294 nm, more than 2.0 and the relative standard deviation for replicate
injection volume. 10 Ill. injections is not more than 5.0 per cent.
Time Mobile Phase A Mobile Phase B Inject the reference solution the test solution.
(in min.) (per cent lv/v) (per cent lv/v)
Calculate the content of C2sH3sN60 I IS in the tablet.
o 47 53
Storage. Store protected from moisture.
20 47 53
Labelling. The label states the strength in terms of the
35 10 90 equivalent amount of sildenafil.
45 . 10 90
50 47 53
ill 47 53
Silver Nitrate
Inject reference solution (b). The test is not valid unless the
relative standard deviation for replicate injections is not more Mol. Wt. 169.9
than 5.0 per cent. Silver Nitrate contains not less than 99.0 per cent and not
Inject reference solution (b) and the test solution. In the . more than 100.5 per cent ofAgN03.
chromatogram obtained with the test solution, the area of any Category. Local anti-infective.
secondary peak is not more than 0.8 times the area of the
principal peak in the chromatogram obtained with reference Description. Colourless crystals or a white, crystalline powder.
solution (b) (0.8 per cent), the area of peak cOlTesponding to
Identification
N-oxide impurity at relative retention time 0.54 is not more
than the area of the principal peak in the chromatogram A. Gives the reactions of silver salts (2.3.1).
obtained with reference solution (b) (1.0 per cent) and the sum
B. 10 mg gives reaction A ofnitrates (2.3.1).
of areas of all the secondary peaks is not more than 2.0 times
the area of the peak in the chromatogram obtained with
Tests
reference solution (b) (2.0 per cent). Ignore the peak due to
citric acid at retention time about 2.4 minutes. Appearance of solution. A4.0 per cent w/v solution (solution
A) is clear (2.4.1), and colourless (2.4.1).
Other tests. Comply with the tests stated under Tablets.
Acidity or alkalinity. To 2 ml of solution A add 0.1 mlof
Assay. Detennine by liquid chromatography (2.4.14).
bromocresol green solution; the solution is blue. To 2 ml of
Solvent mixture. 50 volumes ofmobile phase and 50 volumes this solution add 0.1 ml ofphenol red solution; the solution is
of methanol. yellow.

2102

www.webofpharma.com
IP 2010 SIMVASTATIN

Aluminium, bismuth, copper and lead. Dissolve 1.0 g in a Specific optical rotation (2.4.22). + 285 0 to + 3000 , determined
mixture of4 ml of strong ammonia solution and 6 ml of water; in a 0.5 per cent w/v solution in acetonitrile.
the resulting solution is clear (2.4.1), and colourless (2.4.1).
Related substances. Determine by liquid chromatography
Foreign salts. Not more than 0.3 per cent, determined by the (2.4.14), as described under the Assay.
following method. To 30 ml of solution A add 7.5 ml of
Inject test solution (a). Run the chromatogram 5 times the
2 M hydrochloric acid, shake vigorously, heat for 5 minutes
retention time ofthe principal peak. The relative retention time
on a water-bath, filter and evaporate 20 ml of the filtrate to
with reference to simvastatin for (3R,5R)-7-[(lS,2S,6R,8S,8aR)-
dryness on a water-bath. Dry the residue at 105° and weigh.
8-[(2,2-dimethylbutanoyl)oxy]-2,6-dimethyl-l ,2,6,7,8,8a-
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of hexahydronaphthalen-l-y1]-3 ,5-dihydroxyheptanoic acid
water, add 2 ml of2 M nitric acid and 2 ml ofjerric ammonium (hydroxy acid) (simvastatin impurity A) is about 0.45, for
sulphate solution and titrate with 0.1 M ammonium lovastatin (simvastatin impurity E) and epilovastatin
thiocyanate until a reddish yellow colour is produced. (simvastatin impurity F) is about 0.6, for (lS,7S,8S,8aR)-8-[2-
1 mlof 0.1 M ammonium thiocyanate is equivalent to [(2R,4R)-4-hydroxy-6-oxotetrahydro-2H-pyran-2-yl]ethyl]-7-
0.01699 gofAgN03. methyl-3-methylene-l ,2,3,"7,8,8a-hexahydronaphthalen-l-yl
2,2-dimethylbutanoate) (simvastatin impurity G) is about 0,8,
Storage. Store protected from light and moisture, in non- for (lS,3R, 7 S,8S,8aR)-8-[2-[(2R,4R)-4-(acetyloxy)-6-
metallic containers. oxotetrahydro-2H-pyran-2-yl]ethyl]-3,7-dimethyl-l ,2,3,7,8,8a-
hexahydronaphthalen-l-yl 2,2-dimethylbutanoate (acetate
Simvastatin ester) (simvastatin impurity B) is about 2.38, for
(lS,3R,7S,8S,8aR)-3,7-dimethyl-8-[2-[(2R)-6-oxo-3,6-dihydro-
2H-pyran-2-yl]ethyl]-1 ,2,3,7,8,8a-hexahydronaphthalen-l-yl
2,2-dimethylbutanoate (anhydrosimvastatin) (simvastatin
impurity C) is about 2.42 and for (2R,4R)-2-[[(lS,2S,6R,8S,8aR)-
8-[(2,2-dimethylbutanoyl)oxy]-2,6-dimethyl-l,2, 6,7,8,8a-
hexahydronaphthalen-l-yl]ethyl]-6-oxotetrahydro-2H-pyran-
4 - y 1(3 R, 5 R) -7 - [( 1 S, 2 S, 6 R, 8 S, 8 aR) - 8 - [( 2,2-
dimethylbutanoyl)oxy]-2,6-dimethyl-l ,2,6,7 ,8,8a-
hexahydronaphthalen-l-yl]-3,5-dihydroxyheptanoate (dimer)
(simvastatin impurity D) is about 3.8.
Inject test solution (a) and reference solution (b). In the
~SH380S Mol.Wt.4l8.6 chromatogram obtained with test solution (a) the area of the
Simvastatin is (lS,3R, 7S,8S,8aR)-8-[2-[(2R,4R)-4-hydroxy-6- .secondary peaks corresponding to lovastatin and
oxotetrahydro-2H-pyran-2-yl]ethyl]-3,7-dimethyl- epilovastatin is not more than twice the area of the principal
1,2,3,7,8,8a-hexahydronapthalen-l-y12,2-dimethylbutyrate. peak in the chromatogram obtained with reference solution
(b) (LOper cent), the area of any secondary peak other than
Simvastatin contains not less than 97.0 per cent and not more
lovastatin and epilovastatin is not more than 0.8 times the
than 102.0 per cent ofC2sH380S, calculated on the dried basis.
area of the principal peak in the chromatogram obtained with
A suitable antioxidant may be added.
reference solution (b) (0.4 per cent). The sum ofthe areas ofall
Category. Antihyperlipidaemic. the secondary peaks other than lovastatin and epilovastatin
Dose. 5 mg to 10 mg per day. is not more than twice the area of the principal peak in the
Description. A white or almost white, crystalline powder. chromatogram obtained with reference solution (b) (1.0 per
cent). Ignore any peak with an area less than 0.1 times the area
Identification of the principal peak in the chromatogram obtained with
reference solution (b) (0.05 per cent).
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with simvastatin RS Heavy metals (2.3.13).). 1.0 g complies with the limittest for
or with the reference spectrum of simvastatin. heavy metals, Method B (20 ppm).

Tests Sulphated ash (2.3 .18). Not more than 0.1 per cent.

Appearance of solution. A 1.0 per cent w/v solution in Loss on drying (2.4.19). Not more than 0.5 per cent, determined
methanol is clear (2.4.1) and not more intensely coloured than on 1.0 g by drying in an oven at 60 0 for 3 hours under high
reference solution BYS6 (2.4.1). vacuum.

2103

www.webofpharma.com
SIMVASTATIN IP 2010

Assay. Determine by liquid chromatography (2.4.14). Simvastatin Tablets


NOTE-Prepare the solutions immediately before use. Simvastatin Tablets contain not less than 90.0 per cent and
Solvent mixture. 40 volumes ofa 0.14 per centw/v solution of not more than 110.0 per cent of the stated amount of
potassium dihydrogen phosphate, adjusted to pH 4.0 with simvastatin, C2sH3s0s.
orthophosphoric acid and 60 volumes of acetonitrile. Usual strengths. 10 mg; 20 mg; 40 mg.
Test solution (a). Dissolve 75 mg of the substance under
e~amination in 50.0 ml ofthe solvent mixture.
Identification
Test solution (b). Dissolve 40 mg of the substance under Shake a quantity ofthe powdered tablets containing about 50
examination in 50.0 inl ofthe solvent mixture. mg of Simvastatin with 20 ml of dichloromethane, filter and
evaporate the filtrate to dryness using a rotary evaporator
Reference solution (a). Dissolve 1 mg each of simvastatin RS and a water-bath at 40°. On the residue, determine by infrared
and lovastatin RS in 50.0 ml ofthe solvent mixture. absorption spectrophotometry (2.4.6). Compare the spectrum
Reference solution (b). Dilute 0.5 ml of test solution (a) to with that obtained with simvastatin RS or with the reference
100.0 ml with the solvent mixture. spectrum of simvastatin.

Reference solution (c). Dissolve 40 mg of simvastatin RS in Tests


50.0 m1 ofthe solvent mixture.
Dissolution (2.5.2).
Chromatographic system
- a stainless steel column 3.3 cm x 4.6 mm, packed with Apparatus No.1,
end-capped octadecylsilane bonded to porous silica (3 Medium. 900 ml of 0.01 M sodium dihydrogen orthophosphate
J.UTI), containing 0.5 per cent w/v of sodium dodecyl sulphate,
mobile phase: A. a mixture 50 volumes of acetonitrile adjusted to pH 7.0 with 1 M sodium hydroxide,
and 50 volumes of a 0.1 per cent v/v solution of Speed and time. 50 rpm and 30 minutes.
orthophosphoric acid, Withdraw a suitable volume ofthe medium and filter through
B. 0.1 per cent v/v solution of a membrane filter and transfer 10 ml of the filtrate into a
orthophosphoric acid in acetonitrile, centrifuge tube containing 0.1 g of pre-washed manganese
a linear gradient programme using the conditions given (IV) oxide. Shake the tube for 30 minutes, or until the
below, manganese (IV) oxide is completely dispersed, centrifuge and
flow rate. 3.0 m1 per minute, measure the absorbance (2.4.7) ofthe clear supernatant liquid,
spectrophotometer set at 238 mn, suitably diluted if necessary with dissolution medium at the
injection volume. 5 J..tl. maximum at about 257 mn and the minimum at about 257 mn.
Time Mobile phase A Mobile phase B Calculate the content ofsimvastatin, C2sH3s0s.in the medium
(min) (per cent v/v) (per cent v/v) from the absorbance obtained from a solution of known
concentration of simvastatin RS, using the differences in
0-4.5 100 o absorbance at 247 nm and at 257 mn.
4.5-4.6 100 --795 o --75 D. Not less than 70 per cent ofthe stated amount ofC2sH3s0s.
4.6 - 8.0 95 --725 5 --775
Related substances. Determine by liquid chromatography
8.0-11.5 25 75 (2.4.14).
11.5 -11.6 25 --7100 75 --70 NOTE-Prepare the solutions immediately before use.
11.6-13 100 o Buffer solution. A 0.51 per cent w/v solution of sodium
Inject reference solution (a). The test is not valid unless dihydrogen orthophosphate, adjusted to pH 4.5 with 25 per
resolution between the peaks due to lovastatin and cent v/v solution of orthophosphoric acid.
epilovastatin and simvastatin is not less than 5.0. SolutionA. A mixture ofl volume of0.3 per cent v/v solution
Inject test solution (b) and reference solution (c). of glacial acetic acid, adjusted to pH 4.0 with 1 M sodium
hydroxide and 4 volumes of acetonitrile.
Calculate the content of simvastatin.
Test solution. Shake a quantity Of the powdered tablets
Storage. Store protected from light and moisture, under containing about 25 mg ofSimvastatin with 20 ml ofsolution
nitrogen. A for 15 minutes, dilute to 100 m1 and filter.

2104

www.webofpharma.com
IP 2010 SODIUM ACETATE

Reference solution (a). Dilute 1 ml of the test solution to 500 SolutionA. Add 3 ml ofglacial acetic acid to 900 ml of water;
ml with solution A. adjust the pH to 4.0 with 1 M sodium hydroxide and add
Reference solution (b). A solution of 0.002 per cent w/v each sufficient water to produce 1000 ml. Mix 1 volume of this
of simvastatin RS and lovastatin RS in solution A. solution with 4 volumes of acetonitrile.
Chromatographic system Test solution. Disperse a quantity ofwhole tablets containing
- a stainless steel column 10 cm x 4.6 mm packed with about 0.16 g ofSimvastatin in water and mix with the aid elf
octadecylsilane bonded to porous silica (3 11m) (such as ultrasound and shaking. Add sufficient of solution A to
HypersilODS), produce about 75 ml, mix with the aid ofultrasound and shaking
- mobile phase: A. a mixture of40 volumes of acetonitrile for 15 minutes, dilute to 100 ml with solution A and centrifuge.
and 60 volumes of buffer solution, Dilute 1 ml ofthe clear supernatant solution with sufficient of
B. a mixture of 20 volumes of buffer solution A to produce a solution containing 0.01 per cent
solution and 80 volumes of acetonitrile, w/v ofSimvastatin.
- a linear gradient programme using the conditions given Reference solution. A 0.01 per cent w/v solution ofsimvastatin
below, RS in solution A.
flow rate. 1.5 ml per minute,
spectrophotometer set at 238 nm, Chromatographic system
injection volume. 20 Ill. a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 11m) (such as
Time Mobile phase A Mobile phase B
HypersilODS),
(in min.) (per cent v/v) (per cent v/v)
column temperature. 45°,
0-5 85 15 mobile phase: a mixture of7 volumes ofa buffer solution
5 -30 85 -70 15 -7100 prepared by dissolving 5.1 g of sodium dihydrogen
30-45 0 100 orthophosphate in 900 ml of water, adjusted to pH 4.5
45~46 0 -785 100 -715 with orthophosphoric acid or 1 M sodium hydroxide
46-50 85 15 and add sufficient water to produce 1000 ml and 13
volumes of acetonitrile,
Inject reference solution (b). The test is not valid unless the
flow rate. 1.5 mlperminute,
resolution between the peaks due to simvastatin and lovastatin
spectrophotometer set at 238 urn,
is not less than 3.0. The relative retention time with reference
injection volume. 20 Ill.
to simvastatin for simvastatin hydroxy acid (simvastatin
impurity A) is about 0.52, for lovastatin (simvastatin impurity . Inject the reference solution. The test.is not valid unless the
E) is about 0.71, for epilovastatin (simvastatin impurity F) is tailing factor ofthe principal peak is not more than 2.0.
about 0.74, for anhydrosimvastatin (simvastatin impurity C) is Inject the test solution and the reference solution.
about 1.74 and for simvastatin dimer (simvastatin impurity D)
is about 2.42. Calculate the content of CZSH380S in the tablets.

Inject the test solution and reference solution (a). In the


chromatogram obtained with the test solution the area ofpeaks
corresponding to lovastatin and epilovastatin is not more than Sodium Acetate
five times the area of the principal peak in the chromatogram
obtained with reference solution (a) (1.0 per cerit), the area of CHaCOONa,3H20
any secondary peak other than lovastatin and epilovastatin is Mol. Wt. 136.1
not more than twice the area of the principal peak in the
Sodium Acetate contains not less than 99.0 per cent and not
chromatogram obtained with reference solution (a) (0.4 per
more than 101.0 percent ofC zH 3NaOz, calculated on the
cent). The sum of the areas of all the secondary peaks other
dried basis.
than lovastatin and epilovastatin is not more than five times
the area of the principal peak in the chromatogram obtained Category. Pharmaceuticalaid (for peritoneal dialysis fluids).
with reference solution (a) (1.0 per cent). Ignore any peak with Description. Colourless crystals or a white, crystalline powder.
an area less than 0.25 times ofthe area ofthe principal peak in
the chromatogram obtained with reference solution (a) (0.05 . Identification
per cent).
Dissolve 10.0 g in sufficient carbon dioxide-free water to
Other tests. Comply with the tests stated under Tablets. produce 100.0 ml (solution A). 1 ml ofsolution A gives reaction
Assay. Determine by liquid chromatography (2.4.14). B of acetates and reaction A of sodium salts (2.3.1).

2105

www.webofpharma.com
SODIUM ACETATE IP 2010

Tests aluminium standard solution (2 ppm Al), 10 ml of acetate


buffer pH 6.0 and 98 ml of water treated in the same manner
Appearance of solution. Solution A is clear (2.4.1), and and as the blank solution a mixture of 10 ml of acetate buffer
colourless (2.4.1). pH6.0 and 100 ml of water treated in the same manner. Measure
pH (2.4.24).7.5 to 9.0, determined in a 5.0per cent w/v solution. the fluorescence (2.4.5) of the test solution and the standard
solution, using an excitation wavelength of about 392 nm and
Arsenic (2.3.1 0). Dissolve 5.0 g in 50ml of water and add 15 ml
emission wavelength of about 518 nm, and setting the
of stannated hydrochloric acid AsT. The resulting solution
instrument to zero with the blank solution in each case. The
complies with the limit test for arsenic (2 ppm).
fluorescence ofthe test solution is not greater than that ofthe
Calcium and magnesium. Not more than 50 ppm, calculated standard solution (0.2 ppm).
as Ca, determined by the following method. Mix 200 ml of
Storage. Store protected from light.
water with 10mlofammonia bufferpH10.0,0.1 gofmordant
black 11 mixture and 2 ml of 0.05 M zinc chloride. Add Labelling. The label states whether or not the material is
dropwise 0.02 M disodium edetate until the colour changes intended for use in the manufacture of dialysis solutions.
from violet to blue. To this solution add 109 ofthe substance
under examination, shake to dissolve and titrate with
0.02 M disodium edetate until the blue colour is restored. Not
more than 0.65 ml of 0.02 M disodium edetate is required. Sodium Alginate
Heavy metals (2.3.13).12 ml of solution A complies with the
limit test for heavy metals, Method D (10 ppm). Sodium Polymannuronate
Iron (2.3.14). 20 ml ofsolution A complies with the limittestfor Sodium Alginate consists mainly ofthe sodium salt of alginic
iron (20 ppm). acid which is a mixture of polyuronic acids [(C6H s0 6)n]
composed of residues ofD-mannuronic acid and L-guluronic
Chlorides (2.3.12). 10 ml ofsolutionA complies with the limit
acids and is obtained mainly from algae belonging to the order
test for chlorides (250 ppm).
Phaeophyceae.
Sulphates (2.3.17). 15 ml ofsolution A complies with the limit
Category. Pharmaceutical aid (viscosity-increasing agent).
test for sulphates (225 ppm).
Description. A cream-coloured to pale yellowish brown
Reducing substances. Dissolve 1.0 g in 100 ml ofboiling water,
powder; almost odourless.
add 5 ml of1 M sulphuric acid and 0.5 ml of 0.002 Mpotassium
permanganate, mix and boil gently for 5 minutes; the pink Identification
colour is not completely discharged.
A. Dissolve 0.2 gwith shaking in 20ml of water and to 5 ml of
Loss on drying (2.4.19).39.0 to 40.5 percent, determined on
the resulting solution add 1 ml of calcium chloride solution;
0.2 g by drying in an oven at 130°. Place the substance under
a voluminous gelatinous precipitate is produced.
examination in the oven while the oven is still cold.
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of B. To 10 ml of the solution obtained in test A add 1 ml of
anhydrous glacial acetic acid, add 5 ml of acetic anhydride, 1 M sulphuric acid; a gelatinous mass is produced.
mix and allow to stand for 30 minutes. Titrate with C. To 5 mg add 5 ml of water, 1 ml ofa freshly prepared 1 per
0.1 M perchloric acid, using 0.3 ml of 1-naphtholbenzein cent w/v solution of naphthalene-1,3-diol in ethanol (95 per
solution as indicator, until a green colour is produced. Carry cent) and 5 ml of hydrochloric acid. Boil for 3 minutes, cool,
out a blank titration. add 5 ml of water and shake with 15 ml of di-isopropyl ether.
1 ml of 0.1 M perchloric acid is equivalent to 0.00820 g of The upper layer exhibits a deeper bluish red colour than the
CzH3Na02. upper layer obtained by repeating the procedure without the
substance under examination.
Sodium Acetate intended for use in the preparation ofdialysis
solutions complies with the following additional requirement D. The residue obtained in the test for Sulphated ash gives
reaction A ofsodium salts (2.3.1).
Aluminium. Dissolve 20 g in 100 ml of water and adjust to
pH 6.0 by the addition of about 10 ml of lA{hydrqchloric Tests
acid. Extract with successive quantities of20, 20 and 10 ml of
a 0.5 per cent w/v solution of 8-hydroxyquinoline in Heavy metais (2.3.13). 0.5 g complies with the limiftestfor
chloroform and dilute the combined extracts to 50 ml with heavy metals, MethodB (40 ppm), using nitric acid instead
chloroform. Use as the standard solution a mixture of0.4 ml of of sulphuric acid for wetting the sample.

2106

www.webofpharma.com
IP 2010 SODIUM AMINOSALICYLATE

Chloride. Not more than 1.0 per cent, determined by the Tests
following method. Shake 2.5 g with 50 ml of2 M nitric acid for
1 hour, dilute to 100 ml with 2M nitric acid and filter. To 50 ml of pH (2.4.24).6.5 to 8.5, determined in a2.0 percentw/v solution.
the filtrate add 10.0 ml of 0.1 M silver nitrate and 5 ml of 3-aminophenol. Determine by liquid chromatography (2.4.14).
toluene. Titrate with 0.1 M ammonium thiocyanate using 2 ml
offerric ammonium sulphate solution as indicator and shaking Test solution. Dissolve 50.0 mg of the substance under
vigorously towards the end-point. examination in 100 ml ofthe mobile phase.

1 ml of 0.1 M silver nitrate is equivalent to 0.00355 g ofCI. Reference solution (a). Dissolve 25.0 mg of 3-aminophenol
RSin 100.0 ml ofthe mobile phase (solution A). Dilute5.0mlof
Microbial contamination (2.2.9). Not more than 103 micro-
the solution to 100.0 ml with the mobile phase. Dilute 5.0 ml of
organisms per gram. It complies with the test for Escherichia
the resulting solution to 50.0 ml with the mobile phase.
coli and Salmonella.
Reference solution (b). Dissolve 25.0 mg of sodium
Sulphated ash (2.3.18). 30.0 to 36.0 per cent, determined on
aminosalicylate RS in 100.0 ml of the mobile phase. Dilute
0.1 g and calculated on the dried basis.
5.0 ml ofthis solution and 5 ml of solution A to 100.0 ml with
Loss on drying (2.4.19). Not more than 15.0 per cent, the mobile phase.
determined on 0.2 g by drying in an oven at 105° for 4 hours.
Chromatographic system
Storage. Store protected from light. - a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous base deactivated
silica (5 11m) (such as Hypersil BDS),
mobile phase: dissolve 6.0 g of disodium hydrogen
Sodium Aminosalicylate orthophosphate and 6.6 g of sodium dihydrogen
orthophosphate dihydrate in 1600 ml with water, add
Sodium PAS 19 ml of tetra n-butyl ammonium hydrOXide (20 per
cent solution) and make the volume to 1700 ml with
COON a water and add 300 ml of methanol,

~OH
flow rate. 1.5 ml per minute,

Y ,2H zO
spectrophotometer set at 280 urn,
injection volume. 20 Ill.
Inject reference solution (a). The relative standard deviation
NH z
for replicate injections is not more than 5.0 per cent
Inject reference solution (b). The test is not valid unless the
Mol. Wt. 211.2
resolution between m-aminophenol and sodium
Sodium Aminosalicylate is sodium 4-amino-2-hydroxy- aminosalicylate is at least 5.0.
benzoate dihydrate.
Inject the test solution and reference solution (a). In the
Sodium Aminosalicylate contains not less than 99.0 per cent chromatogram obtained with the test solution, the area of the
and not more than 101.0 per cent ofC7H6NNa03' calculated on peak corresponding to 3-aminophenol is not more than the
the anhydrous basis. area ofthe corresponding peak in the chromatogram obtained
Category. Antitubercular. with reference solution (a) (0.25 per cent).

Dose. 10 to 15 g daily, in divided doses. Hydrogen Sulphide and Sulphur dioxide. Dissolve 0.5 gin
5 ml of 1 M sodium hydroxide, add 6 ml of 3 M hydrochloric
Description. A white to cream coloured crystalline powder.
acid and stir vigorously. No odour of hydrogen sulphide or
sulpher dioxide is perceptible, and not more than a faint odour
Identification
of Amyl Alcohol is perceptible. A piece of moistened lead
A. Determine by infrared absorption spectrophotometry (2.4.6). acetate paper held over the mixture does not become
Compare the spectrum with that obtained with sodium discoloured.
aminosalicylate RS or with the reference spectrum ofsodium Arsenic (2.3.1 0). Mix 5.0 g with 10 ml of bromine solution and
aminosalicylate.
evaporate to dryness on a water-bath, i~ite gently, dissolve
B. A 5 per cent w/v solution complies with the tests for sodium the cooled residue, ignoring any carbon, in 50 ml of water and
salts (2.3.1). 14 ml of brominated hydrochloric acid AsT and remove the

2107

www.webofpharma.com
SODIUM AMINOSALICYLATE IP 2010

excess bromine with a few drops of stannous chloride solution Sodium Aminosalicylate Tablets
AsT. The resulting solution complies with the limit test for
arsenic (2 ppm). Sodium PAS Tablets

Heavy metals (2.3.13). 0.66 g complies with the limit test for Sodium aminosalicylate tablet contains not less than 95.0 per
heavy metals, Method B (30 ppm). cent not more than 105.0 per centofC7H6NNa03.2HzO..

Chlorides (2.3.12). Dissolve 1.0 g in 10 rn1 ofwater, add 3 rn1 of Usual strengths. 500 mg; 1 g.
acetic acid and filter, dilute the filtrate to 50 ml with water.
Identification
10 ml ofthe solution complies with the limit test for chlorides
(0.25 per cent). Digest a quantity of powdered tablets containing about 3.0 g
Sulphates (2.3.17).0.1 g complies with the limit test for ofsodium aminosalicylate, with 40 ml of water, and filter. Add
sulphates (0.15 per cent). to the filtrate 15 ml of 1M acetic acid, and allow it to stand
until precipitation has occurred. Collect the precipitate on a
Loss on drying (2.4.19). 16.0 to 17.5 per cent, determined on filter, wash well with water and dry at 105°C for 30 min. The
0.5 g by drying in an oven at 105°. residue complies with the following tests.
Assay. Determine by liquid chromatography (2.4.14). A. Place about 1 g of the residue in a small, round-bottom
flask, and add 10 ml of acetic anhydride. Heat the flask on a
Test solution. Dissolve 50.0 mg of the substance under
steam bath for 30 minutes, and add 40ml of water, mix, filter,
examination in 100.0 ml ofthe mobile phase. Dilute 5.0 ml of
cool, and allow to stand until diacetyl derivative crystallizes,
this solution to 100.0 ml with the mobile phase.
wash well with water, and dry at 105° for 1 hour. The diacetyl
Reference solution (a). Dissolve 50.0 mg of sodium derivative so obtained melts at 191° to 197°.
aminosalicylate RS in 100.0 ml of the mobile phase (solution
B. Shake 0.1 g ofthe residue with 10 ml ofwatel; and filter. To
A). Dilute 5.0 ml ofthis solution to 100.0 ml with the mobile
5 ml of the filtrate add 1 drop offerric chloride solution. A
phase.
violet colour is produced.
Reference solution (b). Dissolve 25.0 mg of 3-aminophenol
C. In the Assay, the principal peak in the chromatogram
RS in 100.0 ml ofthe mobile phase. Dilute 5.0 rn1 ofthis solution
obtained with the test solution corresponds to the peak in the
and 5 ml ofsolution A to 100.0 ml with the mobile phase.
chromatogram obtained with the reference solution.
Chromatographic system
- a stainless steel column 25 cm X 4.6 rom, packed with Tests
octadecylsilane bonded to porous silica (5 ).tm), 3-aminophenol. Determine by liquid chromatography (2.4.14).
- mobile phase: dissolve 6.0 g of disodium hydrogen
orthophosphate and 6.6 g of sodium dihydrogen Test solution. Weigh and powder 20 tablets. Weigh a quantity
orthophosphate dihydrate in 1600 ml with water, add of the powder containing about 50 mg of Sodium
19 ml of tetra n-butyl ammonium hydroxide (20 per Aminosalicylate and dissolve in 100 ml ofthe mobile phase.
cent solution) and make the volume to 1700 ml with Reference solution (a). A 0.024 per cent w/v solution of
water and add 300 ml of methanol, m-aminophenol RS in the mobile phase. Dilute 5 ml to 100 ml
- flow rate. 1.5 ml per minute, with the mobile phase. Dilute 5 ml ofthe resulting solution to
- spectrophotometer set at 254 nm, 50 ml with the mobile phase.
- injectionvolume. 20 ).tl.
Reference solution (b). A 0.024 per cent w/v solution of
Inject reference solution (a). The tailing factor is not more sodium aminosalicylate RS in the mobile phase.
than 2.0. The column efficiency in not less than 3000 theoretical
plates. The relative standard deviation for replicate injections Reference solution (c). Mix 5 ml each ofreference solution (a)
is not more than 5.0 per cent. and reference solution (b) and dilute to 100 ml with the mobile
phase.
Inject reference solution (b). The test is not valid unless the
Chromatographic system
resolution between 3-aminophenol and sodium
- a stainless steel column 25 cm x 4.6 mm, packed with
aminosalicylate is at least 5.0.
octadecylsilane bonged tQ PQrous .pase .de().ctivated
Inject alternately the test solution and the reference solution. silica (5 ).tm) (such as HypersilBDS),
- mobile phase: dissolve 6.0 g of disodium hydrogen
Calculate the content of C7H6NNa03'
orthophosphate and 6.6 g of sodium dihydrogen
Storage. Store protected from light and moisture. orthophosphate dihydrate in 1600 ml with water, add

2108

www.webofpharma.com
IP 2010 SODIUM ASCORBATE

19 ml of tetra n-butyl ammonium hydroxide (20 per Assay. Determine by liquid chromatography (2.4.14).
cent aqueous solution) and make the volume to 1700 ml Test solution. Weigh and powder 20 tablets. Weigh accurately
with water, add 300 ml of methanol, mix well and degas,
a quantity of the powder containing about 0.5 g of sodium
flow rate. 1.5 ml per minute,
aminosalicylate, add sufficient mobile phase to produce
spectrophotometer set at 280 nm,
100.0 ml and filter. Dilute 5.0 ml ofthis solution to 50.0 ml with
injection volume. 20 Ill.
the mobile phase.
Inject reference solution (a). The relative standard deviation
Reference solution. A 0.05 per cent w/v solution of sodium
for replicate injections is not more than 5.0 per cent.
aminosalicylate RS in the mobile phase.
Inject reference solution (c). The resolution between
Chromatographic system
3-aminophenol and sodium aminosalicylate is not less
a stainless steel column 25 cm x 4.6 mm, packed with
than 5.0.
octadecylsilanebonded to porous silica (5 Ilm),
Inject the test solution and reference solution (a). In the mobile phase: dissolve 6.0 g of disodium hydrogen
chromatogram obtained.with the test solution, the area of the orthophosphate and 6.6 g of sodium dihydrogen
peak corresponding to 3-aminophenol is not more than the orthophosphate dihydrate in 1600 ml with water, add
area ofthe peak in the chromatogram obtained with reference 19 ml of tetra n~butyl ammonium hydroxide (20 per
solution (a)( 1.0 per cent): cent aqueous solution) and make the volume to 1700 ml
with water, add 300 ml of methanol, mix well and degas,
Dissolution (2.5.2). flow rate. 1.5 ml per minute,
Apparatus. No.2, spectrophotometer set at 254 nm,
Medium. 900 m1 water, injection volume. 20 Ill.
Speed and time. 100 rpm for 45 minutes. Inject the reference solution. The tailing factor is not more
Withdraw a suitable volume ofthe medium and filter. than 2.0 and the relative standard deviation for replicate
injections is not more than 2.0 per cent
Determine by liquid chromatography (2.4.14).
Inject alternately the test solution and the reference solution.
Test solution. The filtrate diluted with the mobile phase to
produce a 0.055 per cent w/v solution. Calculate the content OfC7H6NNa03. 2H20 in the tablets.

Reference solution. A 0.055 per cent w/v solution of sodium Storage. Store protected from moisture.
aminosalicylate RS in the mobile phase.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with Sodium Ascorbate
octadecylsilane bonded to porous silica (5 Ilm),
mobile phase: dissolve 6.0 g of disodium hydrogen
CHzOH
orthophosphate and 6.6 g of sodium dihydrogen I

orthophosphate dihydrate in 1600 ml with water, add


19 ml of tetra n-butyl ammonium hydroxide (20 per
cent aqueous solution) and make the volume to 1700 ml
with water, add 300 ml of methanol, mix well and degas, Nad
RO
HCOH

OH
flow rate. 1.5 ml per minute,
spectrophotometer set at 254 nm, Mol. Wt. 198.1
injection volume. 20 Ill.
Sodium Ascorbate is L-ascorbic acid monosodium salt.
Inject the reference solution. The tailing factor is not more
than 2.0 and the relative standard deviation for replicate Sodium Ascorbate contains not less than 99.0 per cent and
injections is not more than 2.0 per cent not more than 101.0 per cent ofC6H7Na06, calculated on the
dried basis.
Inject alternatively the test solution and the reference solution.
Category. Vitamin C.
Calculate the content ofC7H6NNa03.2H20 in the medium.
Dose. The equivalent of up to 1 g of ascorbic acid daily.
D. Not less than 75 per cent of the stated amount of
Description. White or faintly yellow crystals or a crystalline
C7H6NNa03.2H20.
powder; odourless or almost odourless. It darkens gradually
Other tests. Comply with the tests stated under Tablets. on exposure to light.

2109

www.webofpharma.com
SODIUM ASCORBATE IP 2010

Identification Category. Phannaceutical aid(preservative).

Test A may be omitted iftests B, C and D are carried out. Test Description. A white, crystalline or granular powder or flakes;
Band C may be omitted if tests A and D are carried out. odourless or with a faint odour; hygroscopic.
A. Detennine by infrared absorption spectrophotometry (2.4.6),
Compare the spectrum with that obtained with sodium
Identification
ascorbate RS. A. To a 10 per cent w/v solution add ferric chloride test
B. T04mlofa2percentw/vsolutionadd 1 mlofO.1 Mhydro- solution; a buff coloured precipitate is fonned. Add dilute
chloric acid, add a few ml of 2,6-dichlorophenol-indophenol hydrochloric acid; a white crystalline precipitate is produced.
solution; the solution is decolorised. B. Gives the reactions of sodium salts and reactions Band C
C. To 4mlofa2percentw/v solution add 1 mlofO.1 Mhydro- ofbenzoates (2.3.1).
chloric acid, add 1 drop of a freshly prepared 5 per cent w/v
solution of sodium nitroprusside and 2 ml of dilute sodium Tests
hydroxide solution. Add 0.6 ml of hydrochloric acid dropwise
and stir; the yellow colour turns blue. Appearance ofsolution. A 10.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely
D. A 2 per cent w/v solution gives the reactions of sodium coloured than reference solution YS6 (2.4.1).
salts (2.3.1).
Acidity or alkalinity. To 20 ml ofa 5.0 per cent w/v solution in
Tests carbon dioxide-free water add 0.2 ml of phenolphthalein
Appearance of solution. A5.0 per cent w/v solution in carbon solution. Not more than 0.2 ml of 0.1 M hydrochloric acid or
dioxide-free water is clear (2.4.1) and not more intensely 0.2 ml of 0.1 M sodium hydroxide is required to change the
coloured than reference solution BYS7 (2.4.1). colour of the solution.
pH (2.4.24). 7.0 to 8.0, detennined in a 10.0 per centw/v solution. ~rsenic (2.3.10). Mix 5.0 gwith 10ml of bromine solution and
Specific optical rotation (2.4.22). +103° to +108°, detennined evaporate to dryness on a water-bath. Ignite gently, dissolve
in a 10.0 per cent w/v solution. the cooled residue, ignoring any carbon, in 50 ml of water and
14 ml of brominated hydrochloric acid AsT, and remove the
Heavy metals (2.3.13). 1.0 g complies with the limit test for
excess of bromine with 2 ml of stannous Chloride solution
heavy metals, Method B (20 ppm).
AsT. The resulting solution complies with the limit test for
Loss on drying (2.4.19). Not more than 0.25 per cent, arsenic (2 ppm).
detennined on 1.0 g by drying over phosphorus pentoxide at
a pressure not exceeding 0.7 kPa for 24 hours. Heavy metals (2.3.13). Dissolve 2.0 g in 45 rnl of water and 5 ml
of hydrochloric acid. 25 ml of the solution complies with the
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
limit test for heavy metals, Method B (20 ppm).
100 ml of carbon dioxide-free water and 25 rnl of1 M sulphuric
acid. Titrate immediately with 0.05 M iodine, using 1 ml of Chlorinated compounds. Dissolve 0.33 g in 5 ml of
starch solution as indicator, until a persistent violet-blue 0.5 M sodium carbonate, evaporate to dryness and heat the
colour is obtained. residue until completely charred, keeping the temperature
1 ml of 0.05 M iodine is equivalentto 0.009905 g ofC6H7Na06. below 400°. Extract the residue with a mixture of 10 ml of water
and 12 ml of dilute nitric acid and filter; the filtrate complies
Storage. Store protected from light.
with the limit test for chlorides (2.3.12).
Loss on drying (2.4.19). Not more than 2.0 per cent, detennined
Sodium Benzoate on 1.0 g by drying in an oven at 105°.

COONa Assay. Weigh accurately about 0.25 g, dissolve in 20 ml of


anhydrous glacial acetic acid, wanning to 50° if necessary,

C7HsNaOz
6 Mol. Wt. 144.1
cool. Titrate with 0.1 M perchloric acid, using 0.05 ml of
1-naphtholbenzein solution as indicator. Carry out a blank
titration.
1 ml·ofO.1 M perchloric acid is equivalent to 0.01441-g of
Sodium Benzoate contains not less than 99.0 per cent and not C7HsNaOz.
more than 100.5 per cent ofC7HsNaOz, calculated on the dried
basis. Storage. Store protected from light.

2110

www.webofpharma.com
IP 2010 SODIUM BICARBONATE INJECTION

Sodium Bicarbonate neutralise with hydrochloric acid and dilute to 15 ml with


distilled water. The resulting solution complies with the limit
Sodium Hydrogen Carbonate test for sulphates (150 ppm).
NaHC0 3 Mol. Wt. 84.0 Assay. Weigh accurately about 1.5 g, dissolve in 50 ml of
carbon dioxide-free water and titrate with 1 M hydrochloric
Sodium Bicarbonate contains not less than 99.0 per cent and acid using 0.2 ml of methyl orange solution as indicator.
not more than 101.0 per cent ofNaHC03 •
1 ml of 1 M hydrochloric acid is equivalent to 0.08401 g of
Category. Electrolyte replenisher; systemic alkaliser. NaHC03 •
Dose. 1 to 4 g. Storage. Store protected from moisture.
Description. A white, crystalline powder or small, opaque,
monoclinic crystals. It gradually forms sodium carbonate on
heating in the dry state or in solution. Sodium Bicarbonate Injection
Identification Sodium Bicarbonate Intravenous Infusion
A. To 5 ml of a 5.0 per cent w/v solution in carbon dioxide- Sodium Bicarbonate Injection is a sterile solution of Sodium
free water (solution A) add 0.1 m1 ofphenolphthalein solution; Bicarbonate in Water for Injections.
a pale pink colour is produced. On heating, a gas is evolved
Sodium Bicarbonate Injection contains not less than 94.0 per
and the solution becomes red.
cent and not more than 106.0 per cent ofthe stated amount of
B. Gives reaction A ofbicarbonates (2.3.1). sodium bicarbonate, NaHC0 3 •
C. Solution A gives the reactions of sodium salts (2.3.1). Category. Electrolyte replenisher; systemic allcaliser.
Usualstrengths. 1.4,4.2,5.0,7.5 and 8.4 percentw/v.
Tests
Description. A clear, colourless solution.
Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1). Identification
Arsenic (2.3.10). Dissolve 5.0 gin 50 rnl ofwater, add 15 ml of A. The residue on evaporation, when moistened with
brominated hydrochloric acid AsT and remove the excess of hydrochloric acid and introduced on a platinum wire into a
bromine with a few drops ofstannous chloride solution AsT. flame, imparts a yellow colourto the flame.
The resulting solution complies with the limit test for arsenic
B. Gives reaction A of sodium salts and the reactions of
(2 ppm).
bicarbonates (2.3.1).
Calcium. A2.0 per centw/v solution, when boiled for 5 minutes,
is clear. Tests
Heavy metals (2.3.13). Mix 4.0 g wi1h 5 rnl ofwater and 18 rnl of pH (2.4.24). 7.0 to 8.5.
dilute hydrochloric acid, heat to boiling and maintain that Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
temperature for 1 minute. Add 0.05 ml of phenolphthalein perrnl.
solution and sufficient 5 M ammonia dropwise to give the
Other tests. Complies with the tests stated under Parenteral
solution a faint pink colour, cool and dilute to 25 ml with water.
Preparations (Injections).
The solution complies with the limit test for heavy metals,
MethodA(5 ppm). Assay. Titrate a volume containing 1.5 g ofSodium Bicarbonate
with 1 M hydrochloric acid using methyl orange solution as
Iron (2.3.14). Dissolve 2.0 g in 20 ml of water and 4 ml of indicator.
hydrochloric acid and dilute to 40 ml with water; the resulting
solution complies with the limit test for iron (20 ppm). 1 ml of 1 M hydrochloric acid is equivalent to 0.08401 g of
NaHC03 •
Carbonate. pH ofsolution A, when freshly prepared, not more
Storage. Store in single dose containers.
than 8.6.
Labelling. The label states (1) the strength as the percentage
Chlorides (2.3.12).1.25 g dissolved in 15 ml ofwater and 2 ml
w/v of Sodium Bicarbonate; (2) the approximate
of nitric acid complies with the limit test for chlorides
concentrations, in millimoles per litre, ofthe sodium ions and
(200 ppm).
the bicarbonate ions; (3) that an injection containing visible
Sulphates (2.3.17). Suspend 1.0 g in 10 ml ofdistilled water, particles in the solution should not be used.

2111

www.webofpharma.com
SODIUM CARBONATE IP 2010

Sodium Carbonate Assay. Weigh accurately about l.0 g,dissolve in 25 ml of


water and titrate with 1 M hydrochloric acid using methyl
Na2C03 Mol. Wt. 106.0 (anhydrous) orange solution as indicator.
Na2C03,H20 Mol.Wt.124.0 (monohydrate) 1 ml of 1 M hydrochloric acid is equivalent to 0.05299 g of
Sodium Carbonate is anhydrous or contains one molecule Na2C03'
ofwater of hydration. Storage. Store in tightly-closed, non-metallic containers.
Sodium Carbonate contains not less than 99.5 per cent and Labelling. The label states whether the material is anhydrous
not more than 100.5 per cent of Na2C03, calculated on the or monohydrate.
dried basis.
Category. Pharmaceutical aid (alkalising agent)
Description. Anhydrous - A white or almost white, slightly Sodium Chloride
granular powder, hygroscopic.
NaCI Mol. Wi. 58.4
Monohydrate - A white, crystalline powder or colourless
crystals. Sodium Chloride contains not less than 99.0 per cent and not
more than 100.5 per cent ofNaCl, calculated on the dried basis.
Identification
Category. Pharmaceutical aid (tonicity agent); fluid and
A. Dissolve 1 g in water and dilute to 10 ml with water; the electrolyte replenisher.
solution is strongly alkaline. Description. White or colourless crystals or a white crystalline
B. The solution prepared for test A gives reaction A of powder.
carbonates and reactions of sodium salts (2.3.1)
Identification
Tests
A. Gives the reactions of chlorides (2.3.1).
Appearance ofsolution. Dissolve 2.0 g in 10 ml of water. The
resulting solution is clear and not more intensely coloured B. A 20 per cent w/v solution in carbon dioxide-free water
thanreference solution YS6, (2.4.1). prepared from distilled water (solution A) gives the reactions
ofsodium salts (2.3.1).
Alkali hydroxides and bicarbonates. Dissolve 0.4 g in 20 ml of
water, add 20 ml of barium chloride solution and filter. To Tests
10 ml of the filtrate add 0.1 ml of phenolphthalein solution.
The solution does not become red. Heat the remainder ofthe Appearance of solution. Solution A is clear (2.4.1), and
filtrate to boiling for 2 minutes. The solution remains clear. colourless (2.4.1).
Heavy metals (2.3.13). Dissolve 2.0 g in portions in a mixture of Acidity or alkalinity. To 20 ml of solution A add 0.1 ml of
5 ml ofhydrochloric acid and 25 ml ofwater. Heat the solution bromothymol blue solution; not more than 0.5 ml of
to boiling and cool. Add dilute sodium hydroxide solution 0.01 M hydrochloric acid or of 0.01 M sodium hydroxide is
until the solution is neutral. Dilute to 50 ml with water (solution required to change the colour of the solution.
A). 12 ml ofthe resulting solution complies with the limit test Arsenic (2.3.10). Dissolve 10.0 gin 50 ml of water and 12 mlof
for heavy metals, Method A (50 ppm). Use lead standard stannated hydrochloric acid AsT. The resulting solution
solution (2 ppm Pb) for the standard. complies with the limit test for arsenic (1 ppm).
Iron (2.3 .14). Dilute 5 ml ofsolution A to 10 ml with water. The
Barium. Dissolve 2 g in 10 ml of water, and add 2 ml of dilute
solution complies with the limit test for iron (50 ppm).
sulphuric acid; no turbidity is produced within 2 hours.
Chlorides (2.3.12). Dissolve 0.4 g in water, add 4 ml ofdilute
Bromide. To 0.5 ml ofsolution A add 4.0 ml of water, 2.0 ml of
nitric acidand dilute to 15 ml with water. The solution complies
phenol red reagent and l.Oml of 0.01 per cent w/v solution of
with the limit test fOfchlorides (125 ppm).
chloramine Tand mix immediately. After exactly 2 minutes,
Sulphates (2.3.17). 15 ml ofsolution A complies with the limit add 0.15 ml of 0.1 M sodium thiosulphate, mix and dilute to
test for sulphates, (250 ppm). 10.0 ml with water. The absorbance ofthe solution measured
Loss on drying (2.4.19). Not more thanl.O per cent (for a:t!l1J()ut~9QI!Ill C2,4})'lIsiilg ~ateras theblailk, is notIllore
anhydrous form) or between 12.0 per cent and 15.0 per cent than that of the standard solution prepared at the same time
(for monohydrate form), determined on 2.0 g by drying in an and in the same manner, using 5.0ml ofa 0.0003 percentw/v
oven at 300°. solution of potassium bromide (100 ppm).

2112

www.webofpharma.com
IP 2010 SODIUM CHLORIDE AND DEXTROSE INJECTION

Calcium and magnesium. Not more than 50 ppm, calculated the combined extracts to 50 ml with chloroform. Use as the
as Ca, determined by the following method. Dissolve 20.0 gin blank solution a mixture of 10 ml of acetate bufferpH 6.0 and
200 ml of water, and add 0.1 ml of hydrochloric acid, 5 ml of 100 ml of water treated in the same manner and as the standard
strong ammonia-ammonium chloride solution, 5 drops of solution a mixture of 2 ml of aluminium standard solution
eriochrome black T solution and titrate with 0.005 M disodium (2 ppm AI), 10 ml of acetate bufferpH 6.0 and 90 ml of water
edetate to a blue end-point. treated in the same manner. Measure the fluorescence of the
test solution and of the standard solution (2.4.5), using an
1 ml of 0.005 M disodium edetate is equivalent to 0.0002004 g
excitation wavelength of392 nm and a secondary filter with a
ofCa.
transmission band centered at 518 nm, or a monochromator
Ferrocyanide. Dissolve 2.0 g in 6 ml ofwater and add 0.5 ml of set to transmit at this wavelength, and setting the instrument
a mixture of5 ml ofa 1per cent wIv solution ofjerric ammonium to zero with the blank solution in each case. The fluorescence
sulphate in a 0.25 per cent w/v solution of sulphuric acid, and of the test solution is not greater than that of the standard
95 ml ofa 1 per cent w/v solution offerrous sulphate; no blue solution.
colour is produced within 10 minutes.
Storage. Store protected from light.
Heavy metals (2.3.13). 4.0 g in 2 ml of dilute acetic acid and
Labelling. The label states whether or not the material is
sufficient water to produce 25 ml. The solution complies with
suitable for use in the manufacture of parenteral preparations
the limit test for heavy metals, Method A (5 ppm).
or for the preparation of dialysis solutions.
Iodide. Moisten 5 g by adding dropwise, a solution freshly
prepared by mixing 25 ml of iodide-free starch solution 2 ml of
0.5 M sulphuric acid, 0.15 ml of sodium nitrite solution and
25 ml of water and examine the mixture in daylight; the
substance shows no blue colour after 5 minutes.
Sodium Chloride and Uextrose
Iron (2.3.14). 2.0 g dissolved in 20 ml of water complies with
Injection
the limit test for iron (20 ppm). Sodium Chloride and Dextrose Intravenous Infusion;
Sulphates (2.3.17). 2.5 ml of solution A diluted to 15 ml with Sodium Chloride and Glucose Injection; Sodium Chloride
water complies with the limit test for sulphates (300 ppm). and Glucose Intravenous Infusion
Loss on drying (2.4.19). Not more than 1.0 per cent, determined Sodium Chloride and Dextrose Injectionis a sterile solution of
on 1.0 g by drying in an oven at 105° for 3 hours. Sodium Chloride and pextrose in Water for Injections.
Assay. Weigh accurately about 0.1 g and dissolve in 50 ml of Sodium Chloride and Dextrose Injection contains not less than
water in a glass-stoppered flask. Add 50.0 ml of 0.1 M silver 95.0 per cent and not more than 105.0 per cent of the stated
nitrate, 5 ml of 2 M nitric acid and 2 ml of dibutyl phthalate, amounts ofsodium chloride, NaCl, and dextrose, C6H 120 6 •
shake well and titrate with 0.1 M ammonium thiocyanate using
Usual strengths. Injections containing the following amounts
2 ml offerric ammonium sulphate solution as indicator, until
ofSodium Chloride, NaCI and Dextrose, C6H 1Z0 6•
the colour becomes reddish yellow.
1 ml of 0.1 M silver nitrate is equivalentto 0.005844 gofNaCl. Per centage Percentage Percentage Percentage
w/vof w/vof w/vof w/vof
Sodium Chloride intended for use in the manufacture of
sodium dextrose sodium dextrose
parenteral preparations or in the manufacture of dialysis
chloride (C6H 1Z0 6) chloride (C6H 1Z0 6)
solutions complies with the following additional requirement.
(NaCl) (NaCl)
Potassium. Not more than 0.1 per cent, determined by flame
0.11 5
photometry (2.4.4), using a 1.0 per cent w/v solution and
measuring at 768 nm. Use suitable dilutions in water of 0.18 5 0.45 5
potassium solution FP for the standard solution. 0.2 5 0.45 10
Sodium Chloride intended for use in the preparation ofdialysis 0.225 5 0.9 2.5
solutions complies with the fpllowing additionai requirement. 0.3 5 0.9 5
Aluminium. Not more than 0.2 ppm, determined by the 0.33 5 0.9 10
following method. Dissolve 20 g in 100 ml of water and add 0.45 2.5 0.9 25
10 ml of acetate buffer pH 6.0. Extract the resulting solution
with successive quantities of20, 20 and 10 ml ofa 0.5 per cent Description. A clear, colourless or faintly straw-coloured
w/v solution of 8-hydroxyquinoline in chloroform and dilute solution.

2113

www.webofpharma.com
SODIUM CHLORIDE AND DEXTROSE INJECTION IP 2010

Identification Sodium Chloride and Fructose Injection contains not less than
95.0 per cent and not more than 105.0 per cent of the stated
A. To 1 ml add 0.05 mlofpotassium cupri-tartrate solution; amounts of sodium chloride, NaCI, and fructose, C6H 1Z0 6• It
the solution remains blue and clear. Heat to boiling, a copious
contains .no antimicrobial agent.
red precipitate is formed.
Usual strengths. Injections containing the following amounts
B. Gives reaction B ofsodium salts and reaction A ofchlorides
ofSodium Chloride, NaCl, and Fructose, C6H 1z06•
(2.3.1).
Per cent Per cent Per cent Per cent
Tests w/v of w/vof w/v of w/vof
pH (2.4.24).3.5 to 6.5. Sodium Fructose Sodium Fructose
Chloride Chloride
5-Hydroxymethylfurfural and related substances. Dilute a (NaCl) (C6H 1Z0 6) (NaCI)
volume containing 1.0 g ofdextrose, C6H 1Z0 6, to 500 ml with
water and measure the absorbance of the resulting solution 0.11 5 0.45 2.5
at the maximum at about 284 nm (2.4.7); absorbance at about 0.18 5 0.45 10
284 nm, not more than 0.25. 0.225 5 0.9 2.5
Bacterial endotoxins (2.2.3). Not more than 10 Endotoxin Units 0.33 5 0.9 5
per g of dextrose. 0.45 5 0.9 10
Other tests. Complies with the tests stated under Parenteral
Description. A clear, colourless or faintly straw-coloured
Preparations (Intravenous Infusions).
solution.
Assay. For sodium chloride- Titrate an accurately measured
volume containing 0.1 g ofSodium Chloride with 0.1 M silver Identification
nitrate using potassium chromate solution as indicator.
A. To 1 ml add 0.05 ml ofpotassium cupri-tartrate solution;
I ml of0.1 M silver nitrate is equivalentto 0.005844 g ofNaCl. the solution remains blue and clear. Heat to boiling, a copious
For dextrose- To an accurately measured volume containing red precipitate is formed.
2 to 5 g of anhydrous dextrose, C6H 1Z 0 6, add 0.2 ml of B. The solution prepared in the Assay is laevo-rotatory.
5 M ammonia and sufficient water to produce 100.0 ml. Mix
well, allow to stand for 30 minutes and measure the optical C. Gives reaction B ofsodium salts and reaction A ofchlorides
rotation in a 2-dm tube (2.4.22). The observed rotation in (2.3.1).
degrees multiplied by 0.9477 represents the weight, in g, of
dextrose, C6H J2 0 6, in the volume ofthe injection taken for assay. Tests
Storage. Store in single dose containers. On keeping, small pH (2.4.24).3.0 to 6.0.
solid particles may separate from a glass container. 5-Hydroxymethylfurfural and related substances. Dilute a
Labelling. The label states (I) the strength as the percentages volume containing 1.0 g of Fructose to 500.0 ml with water
w/v of Sodium Chloride and Dextrose; (2) that a solution and measure the absorbance of the resulting solution at the
containing visible particles must not be used. maximum at about 284 nm (2.4.7); absorbance at about 284 nm,
When the preparation is intended for intravenous infusion, not more than 0.50.
the label states the approximate concentrations, in millimoles Heavy metals (2.3.13). Evaporate a volume containing 4.0 g of
per litre, of the sodium and chloride ions and the number of Fructose to 10 ml and add 2 ml of dilute acetic acid and
grams per litre ofdextrose, C 6H 1Z0 6 • sufficient water to produce 25 ml. The solution complies with
the limit test for heavy metals, Method A (5 ppm).
Other tests. Complies with the tests stated under Parenteral
Sodium Chloride and Fructose Preparations (Injections).
Injection Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
perrol.· ..
Sodium Chloride and Fructose Intravenous Infusion;
Sodium Chloride and Fructose Infusion; Fructose and Assay. For sodium chloride "- Titrate an accurately measured
Sodium CWoride Injection. volume containing about 0.1 g ofSodium Chloride with 0.1 M
silver nitrate using potassium chromate solution as indicator.
Sodium Chloride and Fructose Injection is a sterile solution of
Sodium Chloride and Fructose in Water for Injections. 1 ml of0.1 M silver nitrate is equivalentto 0.005844 g ofNaCl.

2114

www.webofpharma.com
IP 2010 COMPOUND SODIUM CHLORIDE AND DEXTROSE INJECTION

Forfructose - To an accurately measured volume containing C. After evaporation to one half of its original volume, the
about 5.0 g of Fructose, add 0.2 ml of 6 M ammonia and solution gives reaction A ofpotassium salts and reaction B of
sufficient water to produce 100.0 ml. Mix well, allow to stand calcium salts (2.3.1).
for 30 minutes and measure the optical rotation in a 2-dm tube
(2.4.22). The observed rotation in degrees multiplied by 0.5427 Tests
represents the weight, in g, offructose, C6H 120 6, in the volume
pH (2.4.24). 3.5 to 6.5.
taken for assay.
5-Hydroxymethylfurfural and related substances. Dilute a
Storage. Store in· single dose containers. On keeping, small volume containing 1.0 g of Dextrose to 500.0 ml with water
solid particles may separate from glass containers. and measure the absorbance of the resulting solution at the
Labelling. The label states (1) the strength as the percentages maximum at about 284 nm; absorbance at about 284 nm, not
w/v ofSodium Chloride and Fructose; (2) when the preparation more than 0.25 (2.4.7).
is intended for intravenous infusion, the approximate Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
concentrations, in millimoles per litre, of the sodium and perml.
chloride ions and the number ofgrams per litre offructose; (3)
that a solution containing visible particles should not be used. Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Assay. For sodium chloride - Dilute suitably with water and
determine by Method A for flame photometry (2.4.4), or by
Method A for atomic absorption spectrophotometry (2.4.2),
Compound Sodium Chloride and measuring at 589 nm and using sodium solution FP or sodium
Dextrose Injection solution AAS respectively, suitably diluted with water for the
standard solutions.
Compound Sodium Chloride and Dextrose Intravenous
1 g ofNa is equivalent to 2.542 g ofNaCl.
Infusion .
For potassium chloride - Dilute suitably with water and
Compound Sodium Chloride and Dextrose Injection is a sterile determine by Method A for flame photometry (2.4.4), or by
solution containing 0.86 per cent w/v of Sodium Chloride, Method A for atomic absorption spectrophotometry (2.4.2),
0.03 per cent w/v ofPotassium Chloride, 0.033 per cent w/v of measuring at 767 nm and using potassium solution FP or
Calcium Chloride and 5. per cent w/v ofDextrose in Water for potassium solution AAS respectively, suitably diluted with
Injections. water for the standard solutions.
Compound Sodium Chloride and Dextrose Injection contains I g ofK is equivalent to 1.907 g ofKCl.
not less than 0.82 per cent and not more than 0.90 per cent
w/v of sodium chloride, NaCI, not less than 0.0285 per cent For calcium chloride - To 50.0 ml add 5.0 ml of
and not more than 0.0315 percent w/v ofpotassium chloride, 0.01 M magnesium sulphate and 5 ml of ammonia buffer
KCI, not less than 0.030 per cent and not more than 0.036 per pH 10.9 and titrate with 0.01 M disodium edetate using
cent w/v of calcium chloride, CaCh,2H 20, and not less than eriochrome black T mixture as indicator. From the volume of
0.523 per cent and not more than 0.580per cent w/v ofchloride, 0.01 M disodium edetate required subtract the volume of
CI, and not less than 4.75 per cent and not more than 5.25 per 0.01 M magnesium sulphate added.
cent w/v of dextrose, C6H I2 0 6• It contains no antimicrobial 1 ml ofthe remainder of O. 01 M disodium edetate is equivalent
agent. to 0.00147 gofCaCh,2H20.
Category. Fluid and electrolyte replenisher. For total chloride- To 20.0 ml add 30 ml of water, 50.0 ml of
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the
Description. A clear, colourless or faintly straw-coloured
precipitate with water slightly acidified with nitric acid and
solution.
titrate the excess of silver nitrate with 0.1 M ammonium
thiocyanate using ferric ammonium sulphate solution as
Identification indicator until a reddish yellow colour is produced. Carry out
A. To 1 ml add 0.05 ml ofpotassium cupri-tartrate solution; a blank titration.
the solution remains blue and clear. Heat to boiling, a copious 1 ml of 0.1 M silver nitrate is equivalentto 0.003545 goftotal
red precipitate is formed. Chloride, calculated as Cl.
B. Gives reaction B ofsodium salts and reaction A ofchlorides For dextrose - To an accurately measured volume containing
(2.3.1). 2 g to 5 g ofDextrose, add 0.2 ml of5 M ammonia and sufficient

2115

www.webofpharma.com
SODIUM CHLORIDE HYPERTONIC INJECTION IP 2010

water to produce 100.0 ml.Mix well, allow to stand for sulphate solution as indicator, until the colour becomes reddish
30 minutes and measure the optical rotation in a 2-dm tube yellow.
(2.4.22). The observed rotation in degrees multiplied by 0.9477 1ml of 0.1 M silver nitrate is equivalentto 0.005844 g ofNaCl.
represents the weight, in g, ofdextrose, C6H 1Z0 6, in the volume
taken for assay. Storage. Store in single dose containers of glass or plastic.
On keeping, small solid particles may separate from a glass
Storage. Store in single dose containers of glass or plastic at container.
a temperature not exceeding 30°. On keeping, small solid
particles may separate from the solution i~ glass containers. Labelling. The label states{l) the strength as the percentage
w/v ofSodiumChloride; (2) that a solution containing visible
Labelling. The label states (1) the.strength as the percentages solid particles must not be used.
w/v ofSodium Chloride, Potassium Chloride, Calcium Chloride
and Dextrose; (2) that the injection contains, in millimoles per When the preparation is .intended for intravenous. infusion,
litre, the following approximate amounts ofthe ions. sodium, the label states that the injection contains approximately
147.5, potassium, 4, calcium, 4.5, and Chloride, 156; (3)the 270 millimoles each ofsodium and chloride ions per litre.
total osmolar concentration in mOsmol per litre; (4) that the
injection should not be used if it contains visible partiCles.
Sodium Chloride Injection
Sodium Chloride Intravenous Infusion
Sodium Chloride Hypertonic Injection Sodium Chloride Injection is a sterile 0.9 per cent w/v solution
of Sodium Chloride in Water for Injections. It contains no
Hypertonic Saline antimicrobial agent.
Sodium Chloride Hypertonic Injection is a sterile 1.6 per cent Sodium Chloride Injection contains not less than 0.85 per cent
, w/v solution of Sodium Chloride in Water for Injections. It w/v and not more than 0.95 per cent w/v of sodium Chloride,
contains no antimicrobial agent. NaCl.
Sodium Chloride Hypertonic Injection contains not less than Description. A clear, colourless solution.
1.52 per cent w/v and not more than 1.68 per cent w/v of
sodium chloride, NaCI. Identification
Description. A clear, colourless solution. Gives the reactions ofsodium salts and reaction A ofchlorides
(2.3.1).
Identification
Tests
Gives the reactions ofsodium salts and reaction A ofchlorides
(2.3.1). pH (2.4.24).4.5 to 7.0.

Tests Heavy metals (2.3.13). Evaporate 67 mI to about 20 mI, add 2 mI


of dilute acetic acid and dilute to 25 ml with water. The solution
pH (2.4.24). 5.0to 7.5. complies with the limit test for heavy metals, Method A
(0.3 ppm).
Heavy metals (2.3.13). Evaporate 40 ml to about 20 mI, add 2 ml
of dilute acetic acid and dilute to 25 ml with water. The solution Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
complies with the limit test for heavy metals, Method A perml.
(0.5 ppm).
Other tests. Complies with the tests stated under Parenteral
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit Preparations (Intravenous Infusions).
perml.
Assay. To an accurately measured volume containing about
Other tests. Complies with the tests stated under Parenteral 0.16 g ofSodium Chloride in a glass-stoppered flask add 50 ml
Preparations (Intravenous Infusions). of water and 50.0 ml of 0.1 M silver nitrate, 5 ml of2 M nitric
acid and 2 ml of dibutylphthalate. Shake well and titrate with
Assay. To an accurately measured volume containing about
O;lMammonium thiocyanate using2 ml ofjerric ammonium
6.1615 ofSodium Chloride ill a glass-stoppered flask add 50 mI
sulphate solution as indicator, until the colour becomes reddish
6fwater arid 50.0 rril ofO.l MsilvernTtrate,Srrilof 2.MtiiiiTc
yellow.
acid and 2 ml of dibutylphthalate. Shake well and titrate with
0.1 M ammonium thiocyanate using 2 ml ofjerric ammonium 1ml of 0.1 M silver nitrate is equivalentto 0.005844 g ofNaCI.

2116

www.webofpharma.com
IP 2010 COMPOUND SODIUM CHLORIDE SOLUTION

Storage. Store in single dose containers of glass or plastic. 1 g ofNa is equivalent to 2.54 g ofNaCl.
On keeping, small solid particles may separate from a glass
For potassium Chloride - Dilute appropriately with water
container.
and determine by Method A for flame photometry (2.4.4),
Labelling. The label states (1) the strength as the percentage measuring at 767 nm or by Method A for atomic absorption
w/v of Sodium Chloride; (2) that a solution containing visible spectrophotometry (2.4.2), using potassium solution Fp,
solid particles must not be used. suitably diluted with water for the standard solutions.
When the preparation is intended for intravenous infusion, 1 g ofK is equivalent to 1.007 g ofKCl.·
the label states that the injection contains approximately
For Calcim Chloride - To 50.0 mladd 5.0 ml of 0.01 M
150 millimoles each ofsodium and chloride ions per litre.
magnesium sulphate and 5 ml of ammonia bufferpH 10.9 and
titrate with O.OlM disodium edetate using mordant black II
mixture as indicator. From the volume of 0.01 M disodium
Compound Sodium Chloride Injection edetate required subtract the volume of O.OlM magnesium
sulphate added.
Ringer's Injection
1 ml ofthe remainder of 0.01 M disodium edetate is equivalent
Compound Sodium Chloride Injection isa sterile solution to 0.00147 g ofCaClz, 2H20.
containing 0.86 per cent w/v ofSodium Chloride, 0.03 per cent
Storage. Store in single dose containers of glass or plastic.
w/v ofPotassium Chloride and 0.033 per cent w/v ofCalcium
On keeping, small solid particles may separate from a glass
Chloride in Water for Injections. It contains no antimicrobial
agent. container.
Labelling. The label states (1) the strength as the percentages
Compound Sodium Chloride Injection contains not less than
w/v of Sodium Chloride, Potassium Chloride and Calcium
0.82 per cent wIV and not more than 0.90 per cent w/v of
Chloride; (2) that a solution containing visible solid particles
sodium chloride, NaCl, not less than 0.0285 percent w/v and
must not be used.
not more than 0.0315 per cent w/v ofpotassium chloride, KCl,
and not less than 0.030 per cent w/v and not more than When the preparation is intended for intravenous infusion,
0.036 per centw/v ofcalcium chloride, CaClz, 2H20. the label states that the Injection contains in millimolesper
litre, the following approximate amounts ofions; sodium, 147.5;
Description. A clear, coloUrless solution.
potassium, 4; calcium, 2.25 and chloride, 156.
Identification
Gives reaction B of sodium salts and reaction A of chlorides
(2.3.1). When concentrated to one halfof its original volume,
it gives reaction A ofpotassium salts and reaction B ofcalcium
Compound Sodium Chloride Solution
salts (2.3.1). Ringer's Solution
Tests Compound Sodium Chloride Solution is a solution containing
0.86 per cent w/v of Sodium Chloride, 0.03 per cent w/v of
pH (2.4.24). 5.0to 7.5. Potassium Chloride and 0.033 per cent w/v ofCalcium Chloride
Heavy metals (2.3 .13). Evaporate 67 ml to about 20 ml, add 2 ml in Purified Water. The solution may be clarified by filtration.
of dilute acetic acid and dilute to 25 ml with water. The solution Compound Sodium Chloride Solution contains not less than
complies with the limit test for heavy metals, Method A 0.82 per cent w/v and not more than 0.90 per cent of sodium
(O.3'ppm). chloride, NaC!, not less than 0.025 per cent and not more than
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin 0.035 per cent w/v of potassium chloride, KCl, and not less
Unitperml. than 0.030 per cent and not more than 0.036 per cent wlv of
calcium chloride, CaC12 , 2H20.
Other tests. Complies with the tests stated under Parenteral
Preparations (Intravenous Infusions). Category. Irrigation solution (forextemal use).

Assay. For sodium chloride -Dilute appropriately with water Description. A clear, colourless solution.
and determine by Method A for flame photometry (2.4.4),
Identification
measuring at 589 urn or by Method A for atomic absorption
spectrophotometry (2.4.2), using sodium solution FP, suitably Gives reaction B of sodium salts and reaction A of chlorides
diluted with water for the standard solutions. (2.3.1). When concentrated to one half of its original volume,

2117

www.webofpharma.com
COMPOUND SODIUM CHLORIDE SOLUTION IP 2010

it gives reaction A ofpotassium salts and reaction B ofcalcium Identification


salts (2.3.1).
Gives the reactions ofsodium salts and reaction A ofchlorides
Tests (2.3.1).

pH (2.4.24). 5.0to 7.5. Tests


Heavy metals (2.3.13). Evaporate 67 ml to about 20 rnI, add 2 rnl
pH (2.4.24). 4.5 to 7.0.
ofdilute acetic acid and dilute to 25 ml with water. The solution
complies with the limit test for heavy metals, Method A Heavy metals (2.3.13). Evaporate 67 ml to about20 ml, add 2 rnl
(0.3 ppm). ofdilute acetic acidand dilute to 25 rnl with water. The solution
complies with limit test for heavy metals, MethodA( 0.3 ppm).
Assay. For sodium chloride - Dilute appropriately with water
and determine by Method A for flame photometry (2.4.4), Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
measuring at 589 nm or by Method A for atomic absorption . perml.
spectrophotometry (2.4.2), using sodium solution Fp, suitably Other tests. Complies with the tests stated under Parenteral
diluted with water for the standard solutions. Preparations (Injections).
1 g ofNa is equivalent to 2.54 g ofNaCI. Assay. Titrate an accurately measured volume containing
For potassium chloride - Dilute appropriately with water about 0.135 g of Sodium Chloride with 0.1 M silver nitrate
and determine by Method A for flame photometry (2.4.4), using potassium chromate solution as indicator.
measuring at 767 nm or by Method A for atomic absorption 1 ml of0.1 M silver nitrate is equivalentto 0.005844 g ofNaCI.
spectrophotometry (2.4.2), using potassium solution Fp,
suitably diluted with water for the standard solutions. Storage. Store in single dose containers. The container may
be designed to empty rapidly and may contain a volume of
1 g ofK is equivalent to 1.007 g ofKCl. more than 1 litre.
For calcium Chloride - To 50.0 ml add 5.0 ml of 0.01 M Labelling. The label states (1) the strength as the percentage
magnesium sulphate and 5 ml ofammonia bufferpH 10.9 and w/v of Sodium Chloride; (2) that the solution should not be
titrate with 0.01 M disodium edetate using mordant black II used if it contains visible particles; (3) 'For Irrigation only'
mixture as indicator. From the volume of 0.01 M disodium and 'Not for Injection'; (4) that once the container is opened,
edetate required subtract the volume of O.OIM magnesium the unused portion should be discarded.
sulphate added.
1 ml ofthe remainder of O. 01 M disodium edetate is equivalent
to 0.00147 g ofCaClz, 2HzO.
Sterility (2.2.11). Complies with the test for sterility. Sodium Citrate
Storage. Store protected from light. Trisodium Citrate
Labelling. The label states the strength as the percentages
w/v of Sodium Chloride, Potassium Chloride and Calcium
Chloride. If the contents ofthe container are sterile, the label HJC-0
GOONa ,
2H 20
states (1) Sterile Compound Sodium Chloride Solution; (2) NaOOG GOONa .
that the solution is not intended for injection.
C6HsNa307,2HzO Mol. Wt. 294.1
Sodium Citrate is trisodium 2-hydroxypropane-
Sodium Chloride Irrigation Solution 1,2,3-tricarboxylate dihydrate.
Sodium Citrate contains not less than 99.0 per cent and not
Sodium Chloride Irrigation Solution is a sterile solution
more than 101.0 per cent of C6HsNa307, calculated on the
containing 0.9 per cent w/v of Sodium Chloride in Water for
anhydrous basis.
Injections.
Category. Systemic alkalinising agent.
Sodium Chloride Irrigation Solution contains not less than
0.85 per cent w/v and not more than 0.95 per cent w/v of Dose. 1 to 10 g.
sodium Chloride, NaCl. It contains no antimicrobial agent. Description. White, granular crystals or a White, crystalline
Description. A clear, colourless solution. powder; odourless; slightly deliquescent in moist air.

2118

www.webofpharma.com
IP 2010 SODIUM DIATRIZOATE

Identification Assay. Weigh accurately about 0.15 g and dissolve in 20 ml of


anhydrous glacial acetic acid, warming ~o about 50°. Allow
A. 10.0 per cent w/v solution in carbon dioxide-free water to cool. Titrate with 0.1 M perchloric acid, using 0.25 ml of
(solution A) gives the reactions of sodium salts and reaction l-naphtholbenzein solution as indicator. Carry out a blank
A ofcitrates (2.3.1). titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.008602 g of
Tests
C6HsNa307.
Appearance of solution. Solution A is clear (2.4.1), and Storage. Store protected from light.
colourless (2.4.1).
Acidity or alkalinity. Titrate 20 ml of solution A with
0.05 M sulphuric acid or 0.1 M sodium hydroxide using
Sodium Diatrizoate
thymol blue solution as indicator; not more than 0.5 ml of
0.05 M sulphuric acid or 0.1 M sodium hydroxide is required. Diatrizoate Sodium
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 15 ml
of stannated hydrochloric acid AsT. The resulting solution
complies with the limit test for arsenic (2 ppm).
Heavy metals (2.3.13). Dissolve 2.0 g in lOrnlofwater, 5 mlof
dilute hydrochloric acid and sufficient water to produce
25 ml. The solution complies with the limit test for heavy metals,
Method A ( 10 ppm).
CIIHsI3N2Na04 Mol. Wt. 635.9
Chlorides (2.3.12). 25 ml ofsolution A complies with the limit
test for chlorides (100 ppm). Sodium Diatrizoate is sodium 3,5-diacetamido-2,4,6-triiodo-
benzoate.
Oxalate. Dissolve 0.5 g in 4 rnl ofwater, add 3 rnl ofhydrochloric
acid and 1 g of zinc, in granules, and heat on a water-bath for Sodium Diatrizoate contains not less than 98.0 per cent and
1 minute. Allow to stand for 2 minutes, decant the liquid into a not more than 101.0 per cent ofC II H sI3N 2N aO4, calculated on
test-tube containing 0.25 ml of a 1 per cent w/v solution of the anhydrous basis. . .
phenylhydrazine hydrochloride and heat to boiling. Cool Category. Urographic radio-opaque substance.
rapidly, transfer to a graduated cylinder a,nd add an equal
Dose. To be decided by the physician in accordance with the
volume of hydrochloric acid and 0.25 ml of potassium
needs of the patient.
ferricyanide solution. Shake and allow to stand for 30 minutes.
Any pink colour produced is not more intense than that Description. A white powder; odourless or almost odourless.
obtained by treating at the same time and in the same manner
4 ml of a 0.005 per cent w/v solution of oxalic acid (300 ppm, Identification
calculated as anhydrous oxalic acid). Tests A andD may be omitted iftests B, C, E andFare carried
Sulphates (2.3.17). To 10 ml of solution A add 2 ml of out. Tests B, C and F may be omitted if tests A, D and E are
7 M hydrochloric acid and dilute to 15 ml with distilled water; carried out.
the resulting solution complies with the limit test for sulphates A. Determine by infrared absorption spectrophotometry (2.4.6).
(150 ppm). Compare the spectrum with that obtained with sodium
Tartrates. To a solution of 1 g in 2 ml of water in a test-tube, diatrizoate RS or with the reference spectrum of sodium
add 1 ml of a 10 per cent w/v solution of potassium acetate diatrizoate.
and 1 ml of 6 M acetic acid. Scratch the walls ofthe test-tube B. Determine by thin-layer chromatography (2.4.17), coating
with a glass rod; no crystalline precipitate is formed. the plate with silica gel GF254.
Readily carbonisable substances. Heat 0.2 g, in powder, with Mobile phase. A mixture of 20 volumes of chloroform,
10 ml of sulphuric acid (96 per cent w/w) in a water-bath at 10 volumes of methanol and 2 volumes of strong ammonia
90° ± 1° for J hour and cool rapidly. The solution is not more solution.
intensely coloured than reference solution YS2 or GYS2 (2.4.1).
Test solution. Dissolve 0.1 g of the substance under
Water (2.3.43). 11.0 to 13.0 per cent, determined on 0.3 g and examination in 100 ml of0.08 per cent w/v solution ofsodium
after stirring for 15 minutes before titrating. hydroxide in methanol.

2119

www.webofpharma.com
SODIUM DIATRIZOATE IP20IO

Reference solution. A 0.1 per cent w/v of sodium diatrizoate manner using 2.0 ml of a 0.025 per cent w/v solution of
RS in 0.08 per, cent w/v solution of sodium hydroxide in potassium iodide and 22 ml of water (220 ppm).
methanol~
Heavy metals. Dissolve 1.0 g in 20.0 ml of water and 5 ml of
Apply to the plate 10 'Ill of each solution. After development, 1 M sodium hydroxide, transfer the solution to a 50-ml Nessler
dry the plate in air and examine in ultraviolet light at 254 nm. cylinder, dilute with water to 40 ml and mix. Add 10 ml of
The principal spot in the chromatogram obtained with the test sodium sulphide solution, shake and allow to stand for
solution corresponds to that in the chromatogram obtained 5 minutes (20 ppm), the colour of the solution when viewed
with the reference solution. downward over a white surface is not,more intense than that
produced by treating 2.0 ml of lead standard solution
C. Heat 0.5 g in a crucible; violet vapours ofiodine are evolved.
(10 ppm Ph) in the same manner in place of the substance
D. To 20 mg add 5 ml of1 M sodium hydroxide and boil gently under examination..
under a reflux condenser for 10 minutes. Cool, add 5 ml of
Water (2.3.43). 4.0 to 7.0 percent,determined on 0.4 g.
2 M hydrochloric acid and cool in ice for 5 minutes. Add 4 ml
of a 1 per cent w/v solution of sodium nitrite, cool in ice for Assay. Weigh accurately about 0.4 g in a glass-stoppered
5 minutes, add 0.3 g of sulphamic acid, shake gently until conical flask, add 12 ml of 5 M sodium hydroxide and 1 g of
effervescence ceases and add 2 ml ofa 0.4 per cent w/v solution zinc powder and boil under a reflux condenser for 30 minutes.
of N-(l-naphthyl) ethylenediamine dihydrochloride; an Cool, rinse the condenser with 30 ml of water, filter through
orange-red colour is produced. cotton and wash the flask and filter with two quantities, each
of20 ml, of water. To the combined filtrate and washings add
E. Heat 0.5 g with 1 ml of sulphuric acid on a water-bath until 80 ml of hydrochloric acid, cool and titrate with
a pale violet solution is produced, add 2 ml of ethanol (95 per 0.05 Mpotassium iodate until the dark brown colour becomes
cent) and heat again; odour of ethyl acetate is produced. pale brown. Add 5 ml of chloroform and continue the titration,
F. Gives the reactions of sodium salts (2.3.1). shaking well after each addition, until the chloroform becomes
colourless.
Tests 1 ml of 0.05 Mpotassium iodate is equivalent to 0.02120 g of
pH (2.4.24).7.5 to 9.5, determined in a 50 per centw/v solution. CIIHsI3N2Na04.
Storage. Store protected from light.
Free amine. Place 1.0 g in a 50-ml glass-stoppered volumetric
flask, add 5 ml of water, 10 ml of 0.1 M sodium hydroxide and
25 ml of dimethyl sulphoxide. stopper the flask, mix the
contents gently and cool in ice, protected from light. After Sodium Diatrizoate Injection
5 minutes, slowly add 2 ml of hydrochloric acid, mix and allow
Diatrizoate Sodium Injection
to stand for 5 minutes. Add 2 ml ofa 2 per cent w/v solution of
sodium nitrite, mix and allow to stand for 5 minutes. Add 1 ml Sodium Diatrizoate Injection is a sterile solution of Sodium
ofan 8 per cent w/v solution of sulphamic acid, mix and allow Diatrizoate in Water for Injections. It may contain small
to stand for 5 minutes. Add 2 ml of a 0.1 per cent w/v solution amounts ofsuitable buffers, stabilisers and antimicrobial agents
of N-(l-naphthyl)ethylenediamine dihydrochloride in a but the preparation intended for intravenous administration
70 per cent w/v solution of 1,2-propanediol and mix. Rem~ve contains no antimicrobial preservative.
the flask from the ice and allow to stand in water at 25° ± 2° for Sodium Diatrizoate Injection contains not less than 95.0 per
10 minutes, with occasional shaking. Add sufficient dimethyl cent and not more than 105.0 per cent of the stated amount of
sulphoxide to produce 50 ml and mix. Within 5 minutes, measure sodium diatrizoate, CIIHsI3N2Na04'
the absorbance of the resulting solution at the maximum at
about 470 nm (2.4.7), using as the blank a solution prepared Usual strengths. 25 per cent w/v; 40 per cent w/v.
by treating 5 ml of water in the same manner; absorbance, not Identification
more than 0.40, calculated on the anhydrous basis.
Evaporate a volume ofthe injection containing 1 g ofSodium
Free iodine and iodide. Dissolve 2.0 g in 24 ml of water taken Diatrizoate to dryness. The residue complies with following
in a 50-ml glass-stoppered centrifuge tube. Add 5 ml of toluene tests.
and 5 ml of 1 M sulphuric acid, shake well and centrifuge; no
red colour appears in the toluene layer. To the mixture add 1 ml A; Heat 0.5 g ofresidue in a crucible; violet vapours ofiodine
of a. 2 per cent w/v solution of sodium nitrite, shake· arid are evolved.
centrifuge; any red colour in the toluene layer is not more B. To 20 mg ofthe residue add 5 ml of 1 M sodium hydroxide
intense than that produced in a solution prepared in the same and boil gently under a reflux condenser for 10 minutes. Cool,

2120

www.webofpharma.com
IP 2010 SODIUM DIHYDROGEN PHOSPHATE DIHYDRATE

add 5 ml of 2 M hydrochloric acid and cod I in ice for 5 minutes. Cool, rinse the condenser with 30 ml of water, filter through
Add 4 ml ofa I per cent w/v solution of sodium nitrite, cool in cotton and wash the flask and filter with two quantities, each
ice for 5 minutes, add 0.3 g of sulphamic acid, shake gently of20 ml, of water. To the combined filtrate and washingsadd
until effervescence ceases and add 2 ml of a 0.4 per cent w/v 80 ml of hydrochloric acid, cool and titrate with
solution of N-(l-naphthyl) ethylenediamine dihydrochloride; 0.05 M potassium iodate until the dark brown colour becomes
an orange-red colour is produced. pale brown. Add 5 ml of chloroform and continue the titration,
C. Heat 0.5 g ofresidue with I ml of sulphuric acid on a water- shaking well after each addition, until the chlorofonn becomes
bath until a pale violet solution is produced, add 2 ml of ethanol colourless.
(95 per cent) and heat again; odour of ethyl acetate is 1 ml of 0.05 Mpotassium iodate is equivalent to 0.02120 g of
produced. CllHgI3N2Na04.
Storage. Store protected from light.
Tests
Labelling. The label states (1) the concentration ofthe active
pH (2.4.24). 6.6 to 7.6. ingredient; (2) whether the contents are intended for
Free amine. To a volume containing 1.0 g of Sodium Diatrizoate intravenous injection and, if so, that the unused portion
in a 50-ml glass-stoppered volumetric flask, add 5 ml of water, remaining in the container after use must be discarded.
10 ml of 0.1 M sodium hydroxide and 25 ml of dimethyl
sulphoxide. Stopper the flask, mix the contents gently and
cool in ice, protected from light. After 5 minutes, slowly add
2 ml of hydrochloric acid, mix and allow to stand for 5 minutes.
Sodium Dihydrogen Phosphate
Add 2 ml of a 2 per cent w/v solution of sodium nitrite, mix Dihydrate
and allow to stand for 5 minutes. Add 1 ml of an 8 per cent
Sodium Acid Phosphate
w/v solution of sulphamic acid,mix and· allow to stand
for 5 minutes. Add 2 ml of a 0.1 per cent w/v solution of NaH2P04,2H20 Mol. Wt. 156.0
N-(l-naphthyl)ethylenediamine dihydrochloride in a 70 per Sodium Dihydrogen Phosphate Dihydrate contains not less
cent w/v solution of 1,2-propanediol and mix. Remove the than 98.0 per cent and not more than 100.5 per cent of
flask from the ice and allow to stand in water at 25° ± 2° for NaH2P0 4, calculated on the dried basis.
10 minutes, with occasional shaking. Add sufficient dimethyl
sulphoxide to produce 50 ml and mix. Within 5 minutes, measure Category. Urinary acidifier.
the absorbance of the resulting solution at the maximum at Dose. 2 to 4 g.
about 470 nm (2.4.7), using as the blank a solution prepared
Description. Colourless crystals or a white powder; odourless.
by treating 5 ml of water in the same manner; absorbance, not
more than 0.30, calculated on the anhydrous basis. Identification
Free iodine and iodide. To a volume containing 2.0 g ofSodium A. Dissolve 10.0 g in sufficient carbon dioxide-free water to
Diatrizoate in 24 ml of water taken in a 50-ml glass~stoppered produce 100 ml (solution A). Solution A is faintly acid.
centrifuge tube add 5 ml of toluene and 5 ml of 1 M sulphuric
acid, shake well and centrifuge; no red colour appears in the B. Solution A neutralised with a 10 per cent w/v solution of
toluene layer. To the mixture add I ml of a 2 per cent w/v potassium hydroxide gives reaction A of sodium salts (2.3.1).
solution of sodium nitrite, shake and centrifuge; any red colour C. Solution A gives the reactions of phosphates (2.3.1).
in the toluene layer is not more iritense than that produced in
a solution prepared in the same manner using 2.0 ml of a Tests
0.025 per cent w/v solution ofpotas~ium iodide and 22 ml of Appearance of solution. Solution A is clear (2.4.1), and
water (220 ppm). colourless (2.4.1).
Pyrogens (2.2.8). Complies with test for pyrogens, using per pH (2.4.24). 4.2 to 4.5, determined in a mixture of5 ml ofsolution
kg ofthe rabbit's weight a volume containing 2.5 g ofSodium A and 5 ml of carbon dioxide-free water.
Diatrizoate.
Arsenic (2.3.10). Dissolve 0.5 gin 50 ml of water and add 10 ml
Other tests. Complies with the tests stated under Parenteral of stannated hydrochloric acid AsT. The resulting solution
Preparations (Injections). complies with the limit test for arsenic (2 ppm).
Assay. Weigh accurately about 0.5 g in a glass-stoppered Heavy metals (2.3.13).12 ml of solution A complies with the
conical flask, add 12 ml of 5 Msodium hydroxide and 1 gof limit test for heavy metals, Method D (10 ppm). Use lead
zinc powder and boil under a reflux condenser for 30 minutes. standard solution (l ppm Pb) to prepare the standard.

2121

www.webofpharma.com
SODIUM FLUORIDE IP 2010

Iron (2.3.14). 20 ml ofsolution A complies with the limit test for Acidity or alkalinity. Dissolve 2.5 g of potassium nitrate in
iron (20 ppm). 40 ml of solution A, dilute to 50 ml with carbon dioxide-free
water, cool to 0° and add 0.2 ml of dilute phenolphthalein
Chlorides (2.3.12).10 ml of solution A diluted to 20 ml with
solution. If the solution is colourless, not more than 1.0 ml of
water complies with the limit test for chlorides (250 ppm)
0.1 M sodiul/1 hydroxide is required to produce a red colour
Sulphates (2.3.17). To 5 ml of solution A add 0.5 ml of that persists for not less than 15 seconds. If the solution is
hydrochloric acid and dilute to 15 ml with distilled water; the red, not more than 0.25 ml of 0.1 M hydrochloric acid is
solution complies with the limit test for sulphates (300 ppm). required to change the colour of the solution. Reserve the
Reducing substances. To 5 ml of solution A add 0.25 ml of neutralised solution for the test for Fluorosilicate.
0.02 M potassium permanganate and 5 ml of 1 M sulphuric Chlorides (2.3.12). 40 ml ofsolution A complies with the limit
acid and heat in a water-bath for 5 minutes; the pink colour is test for chlorides (250 ppm).
not completely discharged.
Fluorosilicate. Heat to boiling the solution reserved in the
Disodium phosphate. Dilute 10 ml ofsolution A to 50 ml with test for Acidity or alkalinity and titrate while hot with
water and titrate with 0.05 M sulphuric acid using 0.1 M sodium hydroxide until a red colour is produced. Not
bromocresol green solution as indicator; not more than 1 ml more than 1.5 ml of 0.1 M sodium hydroxide is required.
of 0.05 M sulphuric acid is required.
Sulphates (2.3.17). Dissolve 0.25 gin 10 ml of a saturated
Loss on drying (2.4.19). 21.5 to 24.0 per cent, determined on solution of boric acid in distilled water and add 5 ml of
0.25 g by drying in an oven at 130°. distilled water and 0.6 ml of 7 M hydrochloric acid. The
Assay. Weigh accurately about 2.5 g, dissolve in 40 ml of solution complies with the limit test for sulphates (200 ppm).
water and titrate with carbonate-free 1 M sodium hydroxide, Prepare the standard by mixing together 0.6 ml of 7 M hydro-
determining the end-point potentiometrically (2.4.25). chloric acid, 5 ml of sulphate standard solution
(10 ppm SO4) and 10 ml of a saturated solution of boric acid
1 ml of 1 M sodium hydroxide is equivalent to 0.120 g of in distilled water.
NaHZP04•
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Storage. Store protected from moisture. on 1.0 g by drying in an oven at 130° for 3 hours.
Assay. Weigh accurately about 80 mg, add a mixture of5 ml of
acetic anhydride and 20 ml of anhydrous glacial acetic acid
Sodium Fluoride and heat to dissolve. Cool, add 20 ml of dioxan. Titrate with
0.1 M perchloric acid, using crystal violet solution as
NaF Mol. Wt. 42.0
indicator, until a green colour is produced. Carry out a blank
Sodium Fluoride contains not less than 98.5 per cent and not titration.
more than 100.5 per cent ofNaF, calculated on the dried basis.
1 ml of 0.1 M perchloric acid is equivalent to 0.004199 g of
Category. Preventive for dental caries. NaF.
Description. A white powder or colourless crystals. Storage. Store protected from moisture.

Identification
A. Dissolve 2.5 g in sufficient carbon dioxide-ji-ee water
without heating to produce 100 ml (solution A). To 2 ml of
Sodium Formaldehyde Sulphoxylate
solution A add 0.5 ml of calcium chloride solution; a gelatinous
white precipitate is produced which dissolves on adding 5 ml
of ferric chloride solution.
°II
N~ -O/S~OH ,2H 2 0

B. Add about 4 mg to a mixture of 0.1 ml of alizarin red S


solution and 0.1 ml of zirconyl nitrate solution and mix; the
colour changes to yellow. . Mol Wt. 154.1

C. Gives reaction A ofsodium salts (2.3.1). Sodium Formaldehyde Sulphoxylate is monosodium


hydroxymethane sulphinate dihydrate. It may contain a
Tests suitable stabilising agent such as sodium carbonate.
Appearance of solution. Solution A is clear (2.4.1), and Sodium Formaldehyde Sulphoxylate contains an amount of
colourless (2.4.1). CH3Na03S equivalent to not less than 45.0 per cent and

2122

www.webofpharma.com
IP 2010 SODIUM FUSIDATE

not more than 55.0 per cent of S02, calculated on the dried Loss on drying (2.4.19). Not more than 27.0 per cent,
basis. determined on 0.5 g by drying in an oven at 105° for 3 hours.
Category. Pharmaceutical aid (antioxidant). Assay. Weigh accurately about 1.0 g, dissolve in 25 ml of
water, add sufficient water to produce 50.0 ml and mix. To 4.0
Description. White crystals or hard white masses; odour,
ml of this solution add 100 ml of water and titrate with 0.1 M
characteristic and garlic-like.
iodine using 3 ml of starch solution, added towards the end
of the titration, as indicator.
Identification
1 ml of 0.1 M iodine is equivalent to 0.001602 g ofS0 2.
A. Dissolve about 4 g in 10 ml ofwater in a test-tube and add
Storage. Store protected from light and moisture.
1 ml of ammoniacal silver nitrate solution; metallic silver is
produced either as a finely divided grey precipitate or as a
bright metallic mirror on the inner surface of the tube.
B. Add about 50 mg to a solution of40 mg ofsalicylic acid in Sodium Fusidate
5 ml of sulphuric acid and warm very gently; a permanent
deep red colour develops.

COONa
Tests

Appearance ofsolution. A 5.0 per cent w/v solution in carbon


dioxide-free water is clear (2.4.1), and colourless (2.4.1).
Alkalinity. Dissolve 1.0 g in 50 ml ofwater and add 0.15 mlof
dilute phenolphthalein solution; not more than 3.5 ml of
0.05 M sulphuric acid is required to change the colour ofthe
solution.
pH (2.4.24). 9.5 to 10.5, determined in a2.0 per centw/v solution Mol. Wt. 538.7
in carbon dioxide-free water. Sodium Fusidate is sodium(17Z)-16~-acetoxy-3a,11a-di­
Iron (2.3.14). Ignite 1.0 g, initially at a low temperature until hydroxyfusida-17(20),24-diene-2l-oate, produced by the
thoroughly charred and finally at about 600°, preferably in a growth of certain strains of Fusidium coccineum or by any
muffle furnace, until all the carbon has been burnt off. Cool, other means.
dissolve the residue in 2 ml ofhydrochloric acid and dilute to Sodium Fusidate contains not less than 97.5 per cent and not
50 ml with water. Add about 50 mg ofammonium persulphate more than 101.0 per cent of C31 H 47Na06, calculated on the
and 5 ml ofammonium thiocyanate solution, mix and transfer anhydrous basis.
to a Nessler cylinder. The red colour of the solution is not Category. Antibacterial.
more intense than that of 1.0 ml of iron standard solution
(10 ppm) treated in the same manner. Dose. 1.5 g daily, in divided doses.

Sulphides. Dissolve 6 g in 14 ml of water in a test-tube and Description. A white or almost white, crystalline powder;
wet a strip of lead acetate paper in the clear solution; no slightly hygroscopic.
discolouration is evident within 5 minutes. Identification
Sodium sulphite. Not more than 5.0 per cent, calculated as A.Dissolve 0.1 gin 5 ml ofwater, add 5 ml ofchloroform and
Na2S03, determined by the following method. Transfer 4.0 ml
0.1 ml ofa 10 per cent w/w solution ofphosphoric acid, shake
of the solution obtained in the Assay to a flask, add 2 ml of
vigorously for 1 minute, allow to separate and filter the lower
formaldehyde solution and titrate with 0.1 M iodine that is
layer through absorbent cotton covered with anhydrous
used for the Assay, adding starch solution towards the end
sodium sulphate. Repeat the extraction with two quantities,
of the titration as indicator.
each of 5 ml, of chloroform, evaporate the combined extracts
Calculate the percentage of Na2S03 from the expression at a pressure of 2 kPa, dry the residue over phosphorus
78.775(V2- vyw, pentoxide at a pressure not exceeding 0.7 kPa for 2 hours and
dissolve in 1 ml of chloroform IR.
where VI and V 2are the volumes, in mI, of0.1 M iodine consumed
in this test and in the Assay respectively and W is the weight, On the residue determine by infrared absorption
in g, of the substance under examination taken for the Assay. spectrophotometry (2.4.6). Compare the spectrum with that

2123

www.webofpharma.com
SODIUM FUSIDATE IP 2010

obtained withfitsidic acid RS or with the reference spectrum chromatogram obtained with reference solution (b) (0.02 per
offusidic acid. cent).
B. In the test for Related substances, the principal spot in the Water (2.3.43). Not more than 2.0 per cent, determined on
chromatogram obtained with the test solution (b) corresponds 0.5g.
to that in the chromatogram obtained with reference solution
Assay. Weigh accurately about 0.2 g and dissolve in a mixture
(a).
of 15 ml of water and 20 ml of ethanol (95 per cent). Titrate
C. Ignite 1 g. The residue gives the reactions of sodium salts with 0.1 M hydrochloric acid to pH 4.1, stirring continuously,
(2.3.1). determining the end-point potentiometrically (2.4.25).
1 ml of 0.1 M hydrochloric acid is equivalent to 0.05387 g of
Tests C31H47Na06.
Appearance ofsolution. A 15.0 per cent w/v solution in carbon Storage. Store protected from light and moisture.
dioxide-free water is not more intensely coloured than
reference solution BS6 (2.4.1).
pH (2.4.24). 7.5 to 9.0, determined in a 1.25 per cent w/v solution. Sodium Hydroxide
Specific optical rotation (2.4.22). +5.0° to +8.0°, determined at Caustic Soda
20° by dissolving 1.5 g in 25 ml of water, adding 0.1 ml of
5 M ammonia and diluting to 50 ml with water. NaOH Mol. Wt. 40.0

Related substances. Determine by liquid chromatography Sodium Hydroxide contains not less than 97.0 per cent and
(2.4.14). not more than 100.5 per cent oftotal alkali, calculated as NaOH.

Test solution. Dissolve 50 mg of the substance under Category. Pharmaceutical aid (allcalising agent).
examination in 10.0 ml ofthe mobile phase. Description. White, crystalline masses supplied as sticks,
Reference solution (a). Dissolve 5 mg of 3-ketofilsidic acid pellets or slabs; deliquescent. Readily absorbs carbon dioxide.
RS in 5 ml ofthe mobile phase. To 1.0 ml of this solution add CAUTION - Great care should be exercised in handling
0.2 ml ofthe test solution and dilute to 20.0 ml with the mobile Sodium Hydroxide as it rapidly destroys tissues.
phase.
Identification
Reference solution (b). Dilute 20 III of the test solution to
100.0 ml with the mobile phase. A. Carefully dissolve 5.0 g in 12 ml of distilled water, add
17 ml of 7 M hydrochloric acid, adjust the pH to 7.0 with
Chromatographic system 1 M hydrochloric acid and add sufficient distilled water to
- a stainless steel column 12.5 cm x 5 mm, packed with produce 50 ml (solution A). 2 ml ofsolution A gives reaction A
octadecylsilane bonded to porous silica (5 11m), of sodium salts (2.3.1).
- mobile phase: a mixture of 10 volumes of methanol, 20
B. pH ofa 0.01 percentw/v solution, not less than 11.0 (2.4.24).
volumes of 1.0 per cent w/v solution of orthophosphoric
acid, 20 volumes of water and 50 volumes of Tests
acetonitrile,
- flow rate. 2 ml per minute, Appearance of solution. A 10.0 per cent w/v solution is clear
- spectrophotometer set at 235 nm, (2.4.1), and colourless (2.4.1).
- injection volume. 20 Ill. Arsenic (2.3.10). Dissolve 2.5 gin 50 ml of water, add 16ml of
Inject reference solution (a). The test is not valid unless the brominated hydrochloric acid AsT, and remove the excess of
resolution between the peaks due to 3-ketofusidic acid and bromine with a few drops of stannous chloride solution AsT.
sodium fusidate is not less than 2.5. The resulting solution complies with the limit test for arsenic
(4 ppm).
Inject the test solution, reference solution (a) and (b). The
signal-to-noise ratio of the principal peak is not less than 3. Heavy metals (2.3.13). Dissolve 1.0 ginS ml of water and 10 ml
Run the chromat()gram 3.5 times the retention time ofprincipal of 3 M hydrochloric acid, heat to boiling, cool and dilute to
peak. The sum of all the secondary peaks is not more than 4 25 ml with water. The solution complies with the lirriit test f6r
times the area of the prinCIpal peak in the c1ITolTlatogtam , heavy metals, Method A ( 20 ppm).
obtained with reference solution (a) (4.0per cent). Ignore any Iron (2.3.14). 20 ml ofsolution A complies with the limittest for
peak with an area less than the area ofthe principal peak in the iron (20 ppm).

2124

www.webofpharma.com
IP 2010 COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION

Carbonates. Not more than 2.0 per cent, calculated as Na2C03, C. Carry out reaction C of calcium salts (2.3.1); no white
determined in the Assay. precipitate is produced.
Chlorides (2.3.12). Dissolve 2.0 g in 5 ml of water, acidify with
Tests
about 8 ml of nitric acid and dilute to 20 ml with water. The
solution, without the addition of dilute nitric acid, complies pH (2.4.24).5.0 to 7.0.
with the limit test for chlorides (125 ppm).
Bacterial endotoxins (2.2.3). Not more than 2.0 Endotoxin Units
Sulphates (2.3.17). Dissolve 2.0 gin 6 ml of distilled water, per millimole.
adjust the pH to 7 with hydrochloric acid and dilute to 20 ml
Other tests. Complies with the tests stated under Parenteral
with distilled water. The resulting solution complies with the
Preparations (Intravenous Infusions).
limit test for sulphates (75 ppm).
Assay. Measure accurately 10 ml, evaporate to dryness in a
Potassium. Acidify 2.5 ml of solution A with acetic acid and
platinum dish and ignite very gently until completely
add 0.15 ml of sodium cobaltinitrite solution; no precipitate
carbonised. Boil the residue with 25.0 ml of 0.05 M sulphuric
is formed.
acid, filter and wash thoroughly with hot water. Titrate the
Assay. Weigh accurately about 2.0 g, dissolve in about 80 ml .excess of acid in the combined filtrate and washings with
of carbon dioxide-free water, add '0.3 ml of phenolphthalein 0.1 M sodium hydroxide using methyl orange solution as
solution and titrate with 1 M hydrochloric acid. Add 0.3 ml indicator.
of methyl orange solution and continue the titration with
1 ml of 0.05 M sulphuric acid is equivalent to 0.Q1l21 g of .
1 M hydrochloric acid.
C3HsNa03.
1 ml of 1 M hydrochloric acid used in the second part of the
Storage. Store in single dose containers of glass or plastic.
titration is equivalent to 0.1060 g ofNa2C03'
On keeping, small solid particles may separate from the solution
1 ml of 1 M hydrochloric acid used in the combined titrations in glass containers.
is equivalent to 0.0400 g oftotal alkali, calculated as NaOH.
Labelling. The label states (1) that the Injection is one-sixth
. Storage. Store protected from moisture, in non-metallic molar and contains, in one litre, approximately 167 millimoles
containers. each of sodium ions and of bicarbonate ions (as lactate); (2)
that the injection should not be used if the solution contains
visible solid particles.
Sodium Lactate Injection
Sodium Lactate Intravenous Infusion
Sodium Lactate Injection is a sterile solution containing Compound Sodium Lactate and
1.85 per cent w/v ofsodium lactate in Water for Injections. It is Dextrose Injection
prepared from Lactic Acid with the aid ofSodium Hydroxide
and sufficient Dilute Hydrochloric Acid to adjust the pH of Compound Sodium Lactate with Dextrose Intravenous
the solution. Infusion; Ringer-Lactate Solution with Dextrose for
Sodium Lactate Injection contains not less than 1.75 per cent Injection; Hartmann's Solution with Dextrose for
and not more than 1.95 per cent w/v of sodium lactate, Injection.
C3HsNa03. Compound Sodium Lactate and Dextrose Injection is a sterile
Category. Fluid and electrolyte replenisher. solution containing 0.24 per cent v/v ofLactic Acid (equivalent
to 0.32 per cent wIv ofsodium lactate) with 0.6 per cent w/v of
Dose. By intravenous infusion at the rate of 5 ml or less per
Sodium Chloride, 0.04 per cent w/v of Potassium Chloride,
minute.
0.027 per cent w/v of Calcium Chloride and 5 per cent w/v of
Description. A clear, colourless solution. Dextrose in Water for Injections.
Identification Compound Sodium Lactate and Dextrose Injection contains
not less than 0.27 per cent and not more than 0.32 per cent
A. When warmed with potassium permanganate, gives w/v of sodium, Na, not less than 0.019 per cent and not more
acetaldehyde, recognisable by its odour. than 0.022 percentw/v ofpotassium, K, not less than 0.37 per
B. The residue on evaporation, when moistened with cent and not more than 0.42 per cent w/v oftotal chloride, Cl,
hydrochloric acid and introduced on a platinum wire into a not less than 0.025 per cent and not more than 0.029 per cent
flame, imparts a yellow colour to the flame. w/v of calcium chloride, CaCh,2H 20, and not less than

2125

www.webofpharma.com
COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION IPlDID

0.23 per cent and not more than 0.28 per cent w/v oflactate, solution AAS respectively, suitably diluted with water for the
calculated as C3H60 3 , and not less than 4.50 per cent and not standard solutions.
more than 5.25 per cent w/v of dextrose, C6H 120 6 • It contains
For total chlorides- To 20.0 ml add 30 ml of water, 50.0 ml of
no antimicrobial agent.
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the
Category. Fluid and electrolyte replenisher. precipitate with water slightly acidified with nitric acid and
titrate the excess of silver nitrate with 0.1 M ammonium
Description. A clear, colourless or faintly straw-coloured
solution.
thiocyanate using ferric ammonium sulphate solution as
indicator until a reddish yellow colour is produced. Carry out
a blank titration.
Identification
I ml of 0.1 M silver nitrate is equivalent to 0.003545 g oftotal
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution; chloride, calculated as CL
the solution remains blue and clear. Heat to boiling, a copious
red precipitate is formed. For calcium chloride - To 50.0 ml add 5.0 ml of
0.01 M magnesium sulphate and 5 ml of ammonia buffer
B. When warmed with potassium permanganate gives pH 10.9 and titrate with 0.01 M disodium edetate using
acetaldehyde, recognisable by its odour. eriochrome black T mixture as indicator. From the volume of
C. The residue on evaporation, when moistened with 0.01 M disodium edetate required subtract the volume of
hydrochloric acid and introduced on a platinum wire into a 0.01 M magnesium sulphate added.
flame imparts a yellow colour to the flame. When viewed 1 ml ofthe remainder of 0.01 M disodium edetate is equivalent
through a suitable blue glass, the flame is tinged reddish to 0.00147 gofCaClz,2H20.
purple.
For lactate - Determine by liquid chromatography (2.4.14).
D. Gives reaction C ofcalcium saltsand reaction A ofchlorides
Test solution. The preparation under examination.
(2.3.1).
Reference solution. A 0.28 per cent w/v solution of lithium
Tests lactate RS in the mobile phase.
Chromatographic system
pH (2.4.24). 4.0 to 6.5.
a stainless steel column 20 cm x 4.6 mm, packed with
5-Hydroxymethylfurfural and Related substances. Dilute a octadecylsilane bonded to porous silica (5 /lm),
volume containing 1.0 g of Dextrose to 500.0 ml with water mobile phase: a mixture of 90 volumes of water and
and measure the absorbance of the resulting solution at the 10 volumes of a 2 per cent v/v solution of ocfylamine in
maximum at about 284 nm; absorbance at about 284 nm, not acetonitrile, the pH of which is adjusted to 7.0 with a
morethan0.25 (2.4.7). 10 per cent v/v solution of phosphoric acid,
Heavy metals (2.3.13). Evaporate a volume containing 4 g of flow rate. 2 ml per minute,
dextrose to 10 ml and add 2 ml of dilute acetic acid and spectrophotometer set at 210 nm,
sufficient water to produce 25 mL The solution complies with injection volume. 10 Ill.
the limit test for heavy metals, Method A ( 5 ppm). Inject separately the test solution and the reference solution
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit and measure the responses for the major peak.
permL Calculate the content of C3H60 3, in the injection.
Other tests. Complies with the tests stated under Parenteral For dextrose- To an accurately measured volume containing
Prepar~tions (Injections). 2 g to 5 g ofDextrose, add 0.2 ml of5 M ammonia and sufficient
Assay. For sodium - Dilute suitably with water and determine water to produce 100.0 ml. Mix well, allow to stand for
by Method A for flame photometry (2.4.4), or by Method A for 30 minutes and measure the optical rotation in a 2-dm tube
atomic absorption spectrophotometry (2.4.2), measuring at (2.4.22). The observed rotation in degrees multiplied by
589 nm and using sodium solution FP or sodium solution 0.9477 represents the weight, in g, ofdextrose, C6H 120 6, in the
AAS respectively, suitably diluted with water for the standard volume taken for assay.
solutions. Storage. Store in single dose containers of glass or plastic at
For potassium - Dilute suitably with water and determine a temperature not exceeding 30°. On keeping, small particles
by Method A for flame photometry (2.4.4), or by Method A for may separate from the solution in glass containers.
atomic absorption spectrophotometry (2.4.2), measuring at Labelling. The label states (1) that the injection contain's, in
767 nm and using potassium solution FP or potassium millimoles per litre, the following approximate amounts ofthe

2126

www.webofpharma.com
IP 2010 HALF STRENGTH COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION

ions. sodium, 131, potassium, 5, calcium, 2; bicarbonate 5-Hydroxymethylfurfural and Related substances. Dilute a
(as lactate), 29 and Chloride, 111; (2) the total osmolar volume containing 1.0 g of Dextrose to 500.0 ml with water
concentration in mOsmol per litre; (3) that the injection should and measure the absorbance of the resulting solution at the
not be used if it contains visible particles. maximum at about 284 nm; absorbance at about 284 nm, not
morethan0.25 (2.4.7).
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
Half Strength Compound Sodium perml.
Lactate and Dextrose Injection Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Half Strength Compound Sodium Lactate with Dextrose
Intravenous Infusion; Half Strength Ringer-Lactate Assay. For sodium - Dilute suitably with water and detennine
Solution with Dextrose Injection by Method A for flame photometry (2.4.4), or by Method A for
atomic absorption spectrophotometry (2.4.2), measuring at
Half Strength Compound Sodium Lactate and Dextrose 589 nm and using sodium solution FP or sodium solution
Injection is a sterile solution containing 0.12 per cent v/v of AAS respectively, suitably diluted with water for the standard
Lactic Acid (equivalent to 0.16 per cent wIv ofsodium lactate) solutions.
with 0.3 per cent w/v ofSodium Chloride, 0.02 per cent w/v of
Potassium Chloride, 0.0135 per cent w/v ofCalcium Chloride For potassium - Dilute suitably with water and detennine
and 5 per cent w/v of Dextrose in Water for Injections. by Method A for flame photometry (2.4.4), or by Method A for
atomic absorption spectrophotometry (2.4.2), measuring at
Compound Sodium Lactate and Dextrose Injection contains
767 nm and using potassium solution FP or potassium
not less than 0.135 per cent and not more than 0.16 per cent
solution AAS respectively, suitably diluted with water for the
w/v ofsodium, Na, not less than 0.0095 per cent and not more
standard· solutions.
than 0.011 per cent w/v of potassium, K, not less than
0.185 per cent and not more than 0.210 per cent w/v of total For total chlorides - To 20.0 ml add 30 ml of water, 50.0 ml of
chloride, Cl, not less than 0.0125 per cent and not more than 0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the
0.0145 per cent w/v ofcalcium chloride, CaClz,2HzO, and not precipitate with water slightly acidified with nitric acid and
less than 0.115 per cent and not more than 0.140 per cent wIv titrate the excess of silver nitrate with 0.1 M ammonium
oflactate, calculated as C3H 60 3, and not less than 4.5 per cent thiocyanate using ferric ammonium sulphate solution as
and not more than 5.25 per cent w/v of dextrose, C6H 1Z0 6• It indicator until a reddish yellow colour is produced. Carry out
contains no antimicrobial agent. a blank titration.
Category. Fluid and electrolyte replenisher. 1 ml of 0.1 Msilvernitrate is equivalent to 0.003545 g oftotal
Description. A clear, colourless or faintly straw-coloured chloride, calculated as Cl.
solution. For calcium chloride To 50.0 ml add 5.0 ml of
0.01 M magnesium sulphate and 5 ml of ammonia buffer pH
Identification
10.9 and titrate with 0.01 M disodium edetate using eriochrome
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution; black T mixture as indicator. From the volume of
the solution remains blue and clear. Heat to boiling, a copious 0.01 M disodium edetate required subtract the volume of
red precipitate is fonned. 0.01 M magnesium sulphate added.
B. When warmed with potassium permanganate gives 1 ml ofthe remainder of O. 01 M disodium edetate is equivalent
acetaldehyde, recognisable by its odour. to 0.00147 gofCaClz,2HzO.
C. The residue on evaporation, when moistened with For lactate - Determine by liquid chromatography (2.4.14).
hydrochloric acid and introduced on a platinum wire into a
flame imparts a yellow colour to the flame. When viewed Test solution. The preparation under examination.
through a suitable blue glass, the flame is tinged reddish Reference solution. A 0.28 per cent w/v solution of lithium
purple. lactate RS in the mobile phase.
D. Gives reaction C ofcalcium salts and reaction A ofchlorides Chromatographic system
(2.3.1). - a stainless steel column 20 cm x 4.6 rom, packed with
octadecylsilane bonded to porous silica (5 /lm),
Tests
mobile phase: a mixture of 90 volumes of water and
pH (2.4.24). 4.0 to 6.5. 10 volumes of a 2 per cent v/v solution of ocfylamine in

2127

www.webofpharma.com
MODIFIED COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION IP 2010

acetonitrile, the pH of which is adjusted to 7.0 with a Category. Fluid and electrolyte replenisher.
10 per cent v/v solution of phosphoric acid,
Description. A clear, colourless or faintly. straw-coloured
flow rate. 2 m1 per minute,
solution.
spectrophotometer set at 210 nm,
injection volume. 10 Ill.
Identification
Inject separately the test solution and the reference solution
and measure the responses for the major peak. A. To 1 ml add 0.05 ml ofpotassium cupri-tartrate solutiori;
the solution remains blue and clear. Heat to boiling, a copious
Calculate the content of C3H60 3 in the injection.
red precipitate is formed.
For dextrose - To an accurately measured volume containing
B. When warmed with potassium permanganate gives
2 g to 5 g ofDextrose, add 0.2 ml of5 M ammonia and sufficient
acetaldehyde, recognisable by its odour.
water to produce 100.0 ml. Mix well, allow to stand for
30 minutes and measure the optical rotation in a 2-dm tube C. The residue on evaporation, when moistened with
(2.4.22). The observed rotation in degrees multiplied by 0.9477 hydrochloric acid and introduced on a platinum wire into a
represents the weight, in g, ofdextrose, C6H 120 6, in the volume flame imparts a yellow colour to the flame. When viewed
taken for assay. through a suitable blue glass, the flame is tinged reddish
Storage. Store in single dose containers of glass or plastic at purple.
a temperature not exceeding 30°. On keeping, small particles
D. Gives reaction C ofcalcium salts and reaction A ofchlorides
may separate from the solution in glass containers.
(2.3.1).
Labelling. The label states (1) that the injection contains, in
millimoles per litre, the following approximate amounts ofthe Tests
ions. sodium, 65.5, potassium, 2.5, calcium, 1, bicarbonate
(as lactate), 14.5, and chloride, 55.5; (2) the total osmolar pH (2.4.24). 4.0 to 6.5.
concentration in mOsmol per litre; (3) that the injection should 5-Hydroxymethylfurfural and Related substances. Dilute a
not be used if it contains visible particles. volume containing 1.0 g of Dextrose to 500.0 ml with water
and measure the absorbance of the resulting solution at the
maximum at about 284 nm; absorbance at about 284 nm (2.4.7),
Modified Compound Sodium Lactate not more than 0.25.

and Dextrose Injection Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
perml.
Modified Compound Sodium Lactate with Dextrose
Other tests. Complies with the tests stated under Parenteral
Intravenous Infusion; Modified Lactated Ringer's and
Preparations (Injections).
Dextrose Injection.
Assay. For sodium - Dilute suitably with water and determine
Modified Compound Sodium Lactate and Dextrose Injection
by Method A for flame photometry (2.4.4), or by Method A for
is a sterile solution containing 0.048 per cent v/v of Lactic
atomic absorption spectrophotometry (2.4.2), measuring at
Acid (equivalent to 0.064 per cent w/v ofsodium lactate) with
589 nm and using sodium solution FP or sodium solution
0.12 per cent w/v of Sodium Chloride, 0.008 per cent w/v of
AAS respectively, suitably diluted with water for the standard
Potassium Chloride, 0.0054 per cent w/v ofCalcium Chloride
solutions.
and 5 per cent w/v of Dextrose in Water for Injections.
For potassium Dilute suitably with water and determine
Modified Compound Sodium Lactate and Dextrose Injection
by Method A for flame photometry (2.4.4), or by Method A for
contains not less than 0.054 percent and not more than
atomic absorption spectrophotometry (2.4.2), measuring at
0.064 per cent w/v ofsodium, Na, not less than 0.0038 per cent
767 nm and using potassium solution FP or potassium
and not more than 0.0044 per cent w/v of potassium, K, not
solution AAS respectively, suitably diluted with water for the
less than 0.074 per cent and not more than 0.084 per cent w/v
standard solutions.
oftotal chloride, Cl, not less than 0.005 per cent and not more
than 0.0058 per cent w/v ofcalcium chloride, CaCIz,2H20, and For total chlorides - To 20.0 ml add 30 m1 ofwater, 50.0 mlof
not less than 0.046 per cent and not more than 0.056 per cent 0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the
w/v oflactate, calculated as C3H60 3, and not less than 4.5 per precipitate with water slightly acidified with nitric acid and
cent and not more than 5.25 per cent wIv ofdextrose, C6H 1206. titrate the excess of silver nitrate with 0.1 M ammonium
It contains no antimicrobial agent. thiocyanate using ferriC ammonium sulphate solution as

2128

www.webofpharma.com
IP 2010 COMPOUND SODIUM LACTATE INJECTION

indicator until a reddish yellow colour is produced. Carry out Compound Sodium Lactate Injection
a blank titration.
Compound Sodium Lactate IntrFlvenous Infusion; Ringer-
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g oftotal
chloride, calculated as Cl.
Lactate Solution for Injection; Hartrnann's Solution for
Injection
For calcium chloride - Evaporate 100.0 ml on a water-bath
Compound Sodium Lactate Injection is a sterile solution
to 50 ml, add to this solution 5.0 ml of 0.01 M magnesium
containing 0.24 per cent v/v of Lactic Acid (equivalent to
sulphate and 5 ml of ammonia bufferpH 10.9 and titrate with
0.32 per cent w/v of sodium lactate) with 0.6 per cent w/v of
0.01 M disodium edetate using eriochrome black T mixture
Sodium Chloride, 0.04 per cent w/v ofPotassium Chloride and
as indicator. From the volume of 0.01 M disodium edetate
0.027 per cent w/v ofCalcium Chloride in Water for Injections.
required subtract the volume of 0.01 M magnesium sulphate
added. Compound Sodium LaCtate Injection contains not less than
0.27 per cent and not more than 0.32 per cent w/v of sodium,
1 ml ofthe remainder of O. DIMdisodium edetate is equivalent
Na, not less than 0.019 per cent and not more than 0.022 per
to 0.00147 gofCaCI2,2H20.
cent w/v of potassium, K, not less than 0.37 per cent and not
For lactate - Determine by liquid chromatography (2.4.14). more than 0.42 per cent w/v oftotal chloride, CI, not less than
0.025 per cent and not more than 0.029 per cent wIv ofcalcium
Test solution. The preparation under examination. chloride, CaCh,2H20, and not less than 0.23 per cent and not
Reference solution. A 0.28 per cent w/v solution of lithium more than 0.28 per cent w/v oflactate, calculated as C 3H 60 3 •
lactate RS in the mobile phase. . Category. Fluid and electrolyte replenisher.
Chromatographic system Description. A clear, colourless solution.
a stainless steel column 20 cm x 4.6 rom, packed with
octadecylsilane bonded to porous silica (5 f.Lm), Identification
mobile phase: a mixture of 90 volumes of water and
10 volumes of a 2 per cent v/v solution of octylamine in A. When warmed with potassium permanganate gives
acetonitrile, the pH of which is adjusted to 7.0 with a acetaldehyde, recognisable by its odour.
10 per cent v/v solution of phosphoric acid, B. The residue on evaporation, when moistened with
flow rate. 2 ml per minute, hydrochloric acid and introduced on a platinum wire into a
spectrophotometer set at 210 nm, flame, imparts a yellow colour to the flame. When viewed
injection volume. 10 f.Ll. through a suitable blue glass, the flame is tinged with reddish
purple.
Inject separately the test solution and the reference solution
and measure the responses for the major peak. C. Gives reaction C ofcalcium salts (2.3.1).

Calculate the content of C3H 60 3 in the injection. Tests


For dextrose - To an accurately measured volume containing pH (2.4.24). 5.0to 7.0.
2 g to 5 g ofDextrose, add 0.2 ml of5 M ammonia and sufficient
water to produce 100.0 ml. Mix well, allow to stand for Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin
30 minutes and determine the optical rotation in a 2-dm tube Unit per ml.
(2.4.22). The observed rotation in degrees multiplied by Other tests. Complies with the tests stated under Parenteral
0.9477 represents the weight, in g, ofdextrose, C6H 120 6 , in the Preparations (Intravenous Infusions).
volume taken for assay.
Assay. For sodium - Dilute suitably with water and determine
Storage. Store in single dose containers of glass or plastic at by Method A for flame photometry (2.4.4), or by atomic
a temperature not exceeding 30°. On keeping, small particles absorption spectrophotometry (2.4.2), measuring at 589 nm
may separate from the solution in glass containers. and using sodium solution FP or sodium solution AAS,
suitably diluted for the standard solutions.
Labelling. The label states (1) that the injection contains, in
millimoles per litre, the following approximate amounts ofthe For potassium - Dilute suitably with water and determine
ions. sodium, 26.2, potassium, 1, calcium, 0.4, bicarbonate (as by Method A for flame photometry (2.4.4), or by atomic
lactate), 5.8, and chloride, 22.2; (2) the total osmolar absorption spectrophotometry (2.4.2), measuring at 767 nm
concentration in mOsmol per litre; (3) that the injection should and using potassium solution FP or potassium solution AAS,
not be used if it contains visible particles. suitably diluted for the standard solutions.

2129

www.webofpharma.com
COMPOUND SODIUM LACTATE INJECTION IP 2010

For total chlorides- To 20 ml add 30 mlofwater, 50.0 ml of than 0.022 percentw/v ofpotassium, K, not less than 0.37 per
0.1 M silver nitrate and 2 ml of nitric acid, filter, wash the cent and not more than 0.42 per cent w/v oftotal chloride, Cl,
precipitate with water slightly acidified with nitric acid and not less than 0.025 per cent and not more than 0.029 per cent
titrate the excess of silver nitrate with 0.1 Mammonium w/v of calcium chloride, CaClz,2HzO, and not less than
thiocyanate using ferric ammonium sulphate solution as 0.23 per cent and not more than 0.28 per cent w/v oflactate,
indicator. Carry out a blank titration. calculated as C3H60 3 • It contains no antimicrobial agent.
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g oftotal Category. Irrigation fluid.
chloride, calculated as Cl. Description. A clear, colourless solution.
For calcium chloride- To 50 ml add 5.0 ml ofO. 01 M magnesium
sulphate and 5 ml ofammonia bufferpH 10.9 and titrate with Identification
0.01 M disodium edetate using mordant black 11 mixture as A. When warmed with potassium permanganate gives
indicator. From the volume of 0.01 M disodium edetate acetaldehyde, recognisable by its odour.
required subtract the volume of 0.01 M magnesium sulphate
added. B. The residue on evaporation, when moistened with
hydrochloric acid and introduced on a platinum wire into a
1 ml ofthe remainder of 0.01 M disodium edetate is equivalent flame imparts a yellow colour to the flame. When viewed
to 0.00147 g ofCaCh, 2HzO. through a suitable blue glass, the flame is tinged reddish
For lactate, calculated as C3H 60 3 - Evaporate 50 ml to purple.
dryness in a platinum dish and ignite very gently until C. Gives reaction C ofcalcium salts and reaction A ofchlorides
completely carbonised. Boil the residue with 25.0 ml of (2.3.1).
0.05 M sulphuric acid, filter, and wash thoroughly with hot
water.Titrate the excess of acid in the combined filtrate and Tests
washings with 0.1 M sodium hydroxide using methyl orange
pH (2.4.24). 5.0to 7.0.
solution as indicator.
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
1 ml of O. 05 M sulphuric acid is equivalent to 0.009008 g of
perml.
C3H60 3•
Other tests. Complies with the tests stated under Parenteral
Storage. Store in single dose containers of glass or plastic.
Preparations (Injections).
On keeping, small solid particles may separate from the solution
in glass containers. Assay. For sodium - Dilute suitably with water and determine
by Method A for flame photometry (2.4.4), or by Method A for
Labelling. The label states (1) that the injection contains, in
atomic absorption spectrophotometry (2.4.2), measuring at
millimoles per litre, the following approximate amounts ofthe
589 nm and using sodium solution FP or sodium solution
ions. sodium, 131, potassium, 5, calcium, 2, bicarbonate
AAS respectively, suitably diluted with water for the standard
(as lactate), 29 and chloride, Ill; (2) that the injection should
solutions.
not be used if the solution contains visible solid particles.
For potassium - Dilute suitably with water and determine
by Method A for flame photometry (2.4.4), or by Method A for
atomic absorption spectrophotometry (2.4.2), measuring at
Compound Sodium Lactate Solution 767 nm and using potassium solution FP or potassium
solution AAS respectively, suitably diluted with water for the
for Irrigation standard solutions.
Ringer-Lactate Solution for Irrigation; Hartmann's For calcium chloride - To 50.0 ml add 5.0 ml of
Solution for Irrigation O.O} M magnesium sulphate and 5 ml of ammonia buffer
Compound Sodium Lactate Solution for Irrigation is a sterile pH 10.9 and titrate with 0.01 M disodium edetate using
solution containing 0.24 per cent v/v ofLactic Acid (equivalent eriochrome black T mixture as indicator. From the volume of
to 0.32 per cent wIv ofsodium lactate) with 0.6 per cent wIv of 0.01 M disodium edetate required subtract the volume of
Sodium Chloride, 0.04 per cent w/v ofPotassium Chloride and 0.01 M magnesium sulphate added.
0.027 per cent w/v ofCalcium Chloride in Water for Injections. 1 ml ofthe remainder of O. 01 M disodium edetate is equivalent
Compound Sodium Lactate Solution for Irrigation contains to 0.00147 g ofCaCh, 2HzO.
not less than 0.27 per cent and not more than 0.32 per cent For total chlorides - To 20.0 ml add 30 ml of water, 50.0 ml of
w/v of sodium, Na, not less than 0.019 per cent and not more 0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash· the

2130

www.webofpharma.com
IP 2010 SODIUM LAURYL SULPHATE

precipitate with water slightly acidified with nitric acid and Identification
titrate the excess of silver nitrate with 0.1 M ammonium
thiocyanate using ferric ammonium sulphate solution as A. A 1 per cent w/v solution, when shaken, produces plenty
indicator until a reddish yellow colour is produced. Carry out offoam.
a blank titration. B. Mix 0.1 mlofa 1percentw/v solution with 0.1 mlofaO.1 per
1 ml of0.1 M silver nitrate is equivalent to 0.003545 g oftotal cent w/v solution ofmethylene blue and 2 ml of 1 M sulphuric
chloride, calculated as Cl. acid, add 2 ml of dichloromethane and shake; the
dichloromethane layer is intensely blue.
For lactate - Determine by liquid chromatography (2.4.14).
C. Mix about 10 mg with 10 ml of ethanol and heat to boiling
Test solution. The preparation under examination. on a water-bath, shaking frequently. Filter immediately and
Reference solution. A 0.28 per cent w/v solution of lithium evaporate the ethanol. Dissolve the residue in 8 ml of water,
lactate RS in the mobile phase. add 3 ml of 2 M hydrochloric acid, evaporate the solution to
half its volume and cool. Filter and to the filtrate add 1 ml of
Chromatographic system
barium chloride solution; a white, crystalline precipitate is
a stainless steel column 20 em x 4.6 mm, packed with
produced.
octadecylsilane bonded to porous silica (5 /lm),
mobile phase: a mixture of 90 volumes of water and D. Gives reaction B ofsodium salts (2.3.1).
10 volumes ofa 2 per cent v/v solution ofoctylamine in
acetonitrile, the pH of which is adjusted to 7.0 with a Tests
10 per cent v/v solution ofphosphoric acid,
- flow rate. 2 ml per minute, Alkalinity. Dissolve 1.0 g in 100 ml of carbon dioxide-free
- spectrophotometer set at 210 urn, water and add 0.1 ml ofphenol red solution. Not more than
- injection volume. 10 /ll. 0.5 ml of 0.1 M hydrochloric acid is required to change the
colour of the solution.
Inject separately the test solution and the reference solution
and measure the responses for the major peak. Non-esterified alcohols. Not more than 4 per cent, detelmined
by the following method. Dissolve 10.0 g in 100 ml bfwater,
Calculate the content of C3H60 3, in the injection. add 100 ml of ethanol (95 per cent) and extract the solution
Storage. Store in single dose containers of glass or plastic. with three quantities, each of 50 ml, of n-pentane, adding
On keeping, small solid particles may separate from the solution sodium chloride, if necessary, to promote separation of the
in glass containers. The container may be designed to empty two layers. Wash the combined organic layers with three
rapidly and may contain a volume ofmore than one litre. quantities, each of 50 ml, of water. Dry the organic solution
over anhydrous sodium sulphate, filter and evaporate on a
Labelling. The label states (1) that the irrigation solution water-bath until the odour ofpentane is no longer detectable.
contains, in millimoles per litre, the following approximate Heat the residue at 105° for 15 minutes, cool and weigh.
amounts of the ions. sodium, 131, potassium, 5, calcium,
2, bicarbonate (as lactate), 29, and chloride, 111; (2) that the Sodium Chloride and Sodium Sulphate. Not more than a total
solution should not be used ifit contains visible particles; (3) of8.0 per cent, determined by the following methods.
'For irrigation only' and 'Not for injection';-(4) that once the For sodium chloride-Dissolve 5.0 gin 50 ml of water, add
container is opened, the unused portion should be discarded. 2 M nitric acid dropwise until the solution is neutral to litmus
paper, add 2 ml of potassium chromate solution and titrate
with 0.1 M silver nitrate.
1 ml of0.1 M silver nitrate is equivalent to 0.005844 g ofNaCl.
Sodium Lauryl Sulphate For sodium sulphate - Dissolve 0.5 g in 20 ml of water,
Sodium Lauryl Sulphate is a mixture ofsodium alkyl sulphates warming gently if necessary, and add 1 ml of a 0.05 per cent
consisting mainly of sodium dodecyl sulphate, w/v solution of dithizone in acetone. If the solution is red,
CH3 [CH z]'oCHzOS03Na. add 1 M nitric acid, dropwise, until it becomes bluish green.
Add 2 ml ofdichloroacetic acidsolution and 80 ml ofacetone
Sodium Lauryl Sulphate contains not less than 85.0 per cent
and titrate with 0.01 M lead nitrate until a permanent orange-
ofsodium alkyl sulphates, calculated as ClzHzsNa04S, red colour is obtained.
Category. Pharmaceutical aid (anionic emulsifying agent).
1 ml of 0.01 M lead nitrate is equivalent to 0.001420 g of
Description. A white or pale yellow powder or crystals. NazS04'

2131

www.webofpharma.com
SODIUM METABISULPHITE IP 2010

Assay. Weigh accurately about 1.15 g, dissolve in sufficient hydrochloric acid and 20 ml ofwater and add a few drops of
water to produce 1000.0 ml, warming ifnecessary. To 20.0 ml bromine solution, cool, dilute with water to 25 mI, then add
add 15 ml of chloroform, 10 ml of dilute sulphuric acid and 50 mg of ammonium persulphate and 5 ml of ammonium
1 ml of dimethyl yellow-oracet blue B solution and titrate thiocyanate solution. Any colour produced is not more intense
with 0.004 M benzethonium chloride, shaking vigorously than that obtained by adding 5 ml of ammonium thiocyanate
and allowing the layers to separate after each addition, until solution to a mixture of iron standard solution (20 ppm Fe),
the chloroform layer acquires a permanent clear green colour. 2 ml of hydrochloric acid, 50 mg of ammonium persulphate
1 ml of 0.004 M benzethonium chloride is equivalent to and sufficient water to produce 25 ml (40 ppm).
0.001154 g of sodium alkyl sulphates, calculated as Thiosulphate. Dissolve 1.0 g in 10 ml of 2 M hydrochloric
ClzHzsNa04S, acid and heat on a water-bath for 10 minutes; not more than a
Storage. Store protected from moisture. faint opalescence is produced.
Assay. Weigh accurately about 0.1 g and dissolve in 50.0 ml
0.05 M iodine, add 1 ml of hydrochloric acid and titrate the
excess of iodine with 0.1 M sodium thiosulphate using starch
Sodium Metabisulphite solution, added towards the end of the titration, as indicator.
Sodium Pyrosulphite; Sodium Disulphite 1 ml of 0.05 Miodine is equivalent to 0.004753 g ofNazSzOs.
NazSzOs Mol. Wt. 190.1 Storage. Store protected from light and moisture. On exposure
to air and moisture it is slowly oxidised to sulphate with
Sodium Metabisulphite may be prepared by saturating a
disintegration of the crystals.
solution of Sodium Hydroxide with sulphur dioxide and
allowing crystallisation to occur.
Sodium Metabisulphite contains not less than 95.0 per cent
and not more than 100.5 per cent ofNazSzOs.
Sodium Methylparaben
Category. Pharmaceutical aid (antioxidant).
Sodium Methyl Hydroxybenzoate
Description. Colourless, prismatic crystals or a white or creamy
white powder; odour, sulphurous.

Identification
A. Gives the reactions of sodium salts (2.3.1).
B. A solution decolorises iodinatedpotassium iodide solution
and the resulting solution gives the reactions of sulphates CsH7Na03 Mol. Wt. 174.1
(2.3.1).
Sodium Methylparaben is the sodium salt of methyl
Tests 4-hydroxybenzoate.
Sodium Methylparaben contains not less than 99.0 per cent
Acidity. A solution is acidic to phenol red solution.
and not more than 102.0 per cent ofCsH7Na03' calculated on
Arsenic (2.3.10). Mix 2.5 g in a porcelain dish with 10 ml of the anhydrous basis.
water, 1.25 g ofpotassium chlorate and 16 ml ofhydrochloric
Category. Pharmaceutical aid (antimicrobial preservative).
acid and heat to expel chlorine, remove the last traces of
chlorine with a few drops of stannous chloride solution AsT Description. A white, crystalline powder; odourless or almost
and add 35 ml of water. The resulting solution complies with odourless; hygroscopic.
the limit test for arsenic (4 ppm).
Identification
Heavy metals (2.3.13). Dissolve 1.0 g in 10 mi ofwater, add 5 ml
of hydrochloric acid and evaporate to dryness on a water- A. Dissolve 0.5 g in 5 ml ofwater and acidify to litmus paper
bath. Dissolve the residue in 25 ml ofwater containing 2 ml of with hydrochloric acid; a white precipitate is produced. Wash
dilute acetic acid. The solution complies with the limit test the precipitate with water and dry.
for heavy metals, Method A ( 20 ppm).
Determine by infrared absorption spectrophotometry (2.4.6).
Iron. To 0.5 g add 2 ml ofhydrochloric acid and evaporate on Compare the spectrum with that obtained with methylparaben
a water-bath to dryness. Dissolve the residue in 2 ml of RS.

2132

www.webofpharma.com
IP 2010 SODIUM PHOSPHATE

B. The residue on ignition gives the reactions of sodium salts Sodium Phosphate
(2.3.1).
Disodium Hydrogen Phosphate; Disodium Hydrogen
Tests Phosphate Dodecahydrate
Appearance of solution. Al 0.0 per cent w/v solution in water NalIP04, 12HzO Mol. Wt. 358.1
is clear (2.4.1).
Sodium Phosphate contains not less than 98.0 per cent and
pH (2.4.24). 9.5 to 1O.5,determined in a 0.1 per cent w/v solution. not more than 101.0 per cent ofNazHP0 4, calculated on the
Related substances. Determine by thin-layer chromatography anhydrous basis.
(2.4.17), coating the plate with silica gel GF254. Category. Cathartic; pharmaceutical aid (buffering agent).
Mobile phase. A mixture of 1 volume ofglacial acetic acid, 30 Description. Colourless, transparent crystals; very
volumes of water and 70 volumes of methanol. efflorescent.
Test solution. Dissolve 0.1 g of the substance under
examination in 10 ml of water. Immediately add 2 ml of Identification
hydrochloric acid and shake with 50 illl of ether. Evaporate
A 10.0 per cent w/v solution in distilled water (solution A)
the upper layer to dryness and take up the residue with 10 ml
gives the reactions of sodium salts and ofphosphates (2.3.1).
of acetone.
Reference solution (a). Dissolve 34.3 mg of 4-hydroxybenzoic Tests
acid (sodium propylparaben impurity A) in 100.0 of acetone.
Appearance of solution. Solution A is clear (2.4.1), and
Reference solution (b). Dilute 0.5 ml of test solution (a) to colourless (2.4.1).
100.0 ml with acetone.
Arsenic (2.3.10). Dissolve 5.0 gin50mlofwaterandadd IOrnl
Reference solution (c). Dissolve 10 mg of ethyl of stannated hydrochloric acid AsT. The resulting solution '
parahydroxybenzoate RS in 1 ml oftest solution (a) and dilute complies with the limit test for arsenic (2 ppm).
to 10.0 ml with acetone.
Heavy metals (2.3.13). Dissolve 2.0 g in 10 rnl of water, adding
Apply 5 IJ.I ofeach solution. Allow the mobile phase to rise 15 4 ml of 1 M acetic acid and diluting to 25 ml with water. The
cm. Dry the plate in air and examine in ultraviolet light at 254 solution complies with the limit test for heavy metals, Method
nm. In the chromatogram obtained with the test solution, any A(lOppm).
spot due to 4- hydroxybenzoic acid is not more intense than
the corresponding spot in the chromatogram obtained with Iron (2.3.14). 20 ml ofsolutionA complies with the limittest for,
reference solution(a) (4.0 per cent) and any other secondary iron (20 ppm).
spot is not more intense than the spot in the chromatogram Chlorides (2.3.12). To 10 ml of solution A add 10 ml of
obtained with reference solution (b) (0.5 per cent). The test is 2 M nitric acid and dilute to 20 ml with water. The resulting
not valid unless the chromatogram obtained with reference solution complies with the limit test for chlorides (250 ppm).
solution (c) shows two clearly separated principal spots.
Sulphates (2.3.17). To 2.5 ml of solution A add 2 ml of
Chlorides (2.3.12). Dissolve 1.0 g in 20 ml of water, add 0.2 ml 2 M hydrochloric acid and dilute to 15 ml with distilled water.
of nitric acid and filter. 15 ml ofthe filtrate complies with the The resulting solution complies with the limit test for sulphates
limit test for chlorides (330 ppm). (600 ppm).
Sulphates (2.3.17). Dissolve 0.5 gin 40 ml of water, add 3.5 ml Reducing substances. To 5 ml of solution A add 5 ml of
of 2 M hydrochloric acid, dilute to 60 ml with water and filter. 1 M sulphuric acid and 0.25 ml of 0.02 M potassium
15 ml of the filtrate complies with the limit test for sulphates permanganate and heat on a water-bath for 5 minutes; the red
(0.12 per cent). colour is not completely discharged. '
Water (2.3.43). Not more tban 5.0 percent, determined on 1.0 g. Sodium dihydrogen phosphate. The value ofthe expression
Assay. Dissolve 0.15 gin 50 ml of anhydrous acetic acid. (nr 25)/(25 - nl),
Titrate with 0.1 M perchloric acid, determining the end-point where nl and n2 are the titres of 1 M sodium hydrOXide obtained
potentiometrically (2.4.25). Carry out a blank titration. in the Assay, does not exceed 0.025.
1 ml of 0.1 Mperchloric acid is equivalent to 0.01741 g of
Water (2.3.43). 57.0 to 61.0 per cent, determined on 0.1 g
CgH7Na03' dissolved in a mixture of 10 volumes of methanol and 40
Storage. Store protected from moisture. volumes of dimethylformamide.

2133

www.webofpharma.com
SODIUM PROPYLPARABEN IP 2010

Assay. Weigh accurately about 4.0 g (w), dissolve in 25 ml of Mobile phase. A mixture of 1 volume ofglacial acetic acid, 30
water, add 25.0 ml of i M hydrochloric acid and titrate volumes of water and 70 volumes of methanol.
potentiometrically with 1 M sodium hydroxide to the first Test solution. Dissolve 0.1 g of the substance under
inflection ofthe pH curve (nl mI). Continue the titration until examination in 10 ml of water. Immediately add 2 ml of
the second inflection ofthe curve is reached; the total volume hydrochloric acid and shake with 50 m1 of ether. Evaporate
of sodium hydroxide required is n2 ml. the upper layer to dryness and take up the residue with 10 ml
Calculate the content of NazHP0 4 from the expression of acetone.
1420(25 - n l)/W (100 - d), Reference solution (a). Dissolve 34.3 mg of4-hydroxybenzoic
where d is the percentage water content. acid (sodium propylparaben impurity A) in 100.0 of acetone.
Storage. Store protected from moisture. Reference solution (b). Dilute 0.5 ml oftest solution (a) to 100
ml with acetone.
Reference solution (c). Dissolve 10 mg of ethyl
parahydroxybenzoate RS in 1 m1 oftest solution (a) and dilute
Sodium Propylparaben to 10.0 m1 with acetone.
Sodium Propyl Hydroxybenzoate Apply 5 f-LI ofeach solution. Allow the mobile phase to rise 15
cm. Dry the plate in air and examine in ultraviolet light at 254
nm. In the chromatogram obtained with the test solution, any
spot due to 4- hydroxybenzoic acid is not more intense than
the corresponding spot in the chromatogram obtained with
reference solution (a) (4.0 per cent) and any other secondary
spot is not more intense than the spot in the chromatogram
Mol. Wt. 202.2 obtained with reference solution (b) (0.5 per cent). The test is
not valid unless the chromatogram obtained with reference
Sodium Propylparaben is the sodium salt of propyl
solution (c) shows two clearly separated principal spots.
4- hydroxybenzoate.
Chlorides (2.3.12). Dissolve 1.0 g in 20 ml ofwater, add 0.2 ml
Sodium Propylparaben contains not less than 99.0 per cent of nitric acid and filter. 15 ml ofthe filtrate complies with the
and not more than 102.0 per cent ofCIOHIINa03, calculated on limit test for chlorides (330 ppm).
the anhydrous basis.
Sulphates (2.3.17). Dissolve 0.5 g in 40 ml ofwater; add 3.5 ml
Category. Pharmaceutical aid (antimicrobial preservative). of 2 M hydrochloric acid, dilute to 60 ml with water and filter.
Description. A white, crystalline powder; odourless or almost 15 ml ofthe filtrate complies with the limit test for sulphates
odourless; hygroscopic. (0.12 percent).
Water (2.3.43). Not more than 5.0 per cen, determined on 1.0 g.
Identification
Assay. Dissolve 0.15 g in 50 ml of anhydrous acetic acid.
A. Dissolve 0.5 gin 5 ml of water and acidify to litmus paper Titrate with 0.1 M perchloric acid, determining the end-point
with hydrochloric acid; a white precipitate is produced. Wash potentiometrically (2.4.25). Carry out the blank titration.
the precipitate with water and dry. 1 ml of 0.1 M perchloric acid is equivalent to 0.02022 g of
Determine by infrared absorption spectrophotometry (2.4.6). CIOHIINa03'
Compare the spectrum with that obtained withpropylparaben Storage. Store protected from moisture.
RS.
B. The residue on ignition gives the reactions ofsodium salts
(2.3.1). Sodium Salicylate
Tests COONa
Appearance ofsolution. A 10.0 per cent w/v solution in water ~OH
is clear (2.4.1).
pH (2A24); 9;5 to 105, determined in a 0.1 per cent w/v solution.
V
C7HsNa03 Mol. Wt. 160.1
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Sodium Salicylate is sodium 2-hydroxybenzoate.

2134

www.webofpharma.com
IP 2010 SODIUM STARCH GLYCOLLATE

Sodium Salicylate contains not less than 99.0 per cent and not Assay. Weigh accurately about 0.15 g, dissolve in 30 ml of
more than 101.0 per cent ofC7HsNa03' calculated on the dried anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
basis. acid, determining the end-point potentiometrically (2.4.25).
Cany out a blank titration.
Category. Anti-inflammatory; analgesic.
1 ml of 0.1 M perchloric acid is equivalent to 0.01601 g of
Dose. 500 mg to 2 g, with food; in the treatment of acute
rheumatism, 5 to 109 daily, in divided doses. C7HsNa03.
Storage. Store protected from light and moisture.
Description. Colourless, small crystals or shiny flakes or a
white, crystalline powder.

Identification
Sodium Starch Glycollate
Test A may be omitted if tests Band C are carried out. Test B
may be omitted if tests A and C are carried out. Sodium Carboxymethyl Starch
A. Detennine by infrared absorption spectrophotometry (2.4.6). Sodium Starch Glycollate is the sodium salt of a poly-a-
Compare the spectrum with that obtained with sodium glucopyranose in which some of the hydroxyl groups are in
salicylate RS or with the reference spectrum of sodium the form ofcarboxymethyl ether.
salicylate.
Sodium Starch Glycollate contains not less than 2.8 per cent
B. A 10.0 per cent w/v solution in carbon dioxide-free water and not more than 4.5 per cent of sodium', Na, calculated on
prepared from distilled water (solution A) gives the reactions the material washed with Ethanol (95 per cent) and dried as
ofsalicylates (2.3.1). described under Assay.
C. Gives reaction B ofsodium salts (2.3.1). Category. Phannaceutical aid (tablet disintegrant).
Description. A very fine, white or off-white, free-flowing
Tests
powder; odourless or almost odourless.
Appearance of solution. Solution A is clear (2.4.1), and not
more intensely coloured than reference solution BYS6 (2.4.1). Identification
Acidity. To 20 ml ofsolutionA add 0.1 ml ofphenol redsolution; A. Detennine by infrared absorption spectrophotometry (2.4.6).
the solution is yellow. Titrate with 0.01 M sodium hydroxide Compare the spectrum with that obtained with sodium starch
to a reddish violet colour; not more than 2.0 ml of glycol/ate RS or with the reference spectrum ofsodium starch
0.01 M sodium hydroxide is required. glycollate.
Arsenic (2.3 .10). Mix 5.0 g with 10 ml of bromine solution and B. to 5 ml ofa 2 per cent w/v dispersion in water add 0.05 ml
evaporate to dryness on a water-bath. Ignite gently, cool, of 0.005 M iodine; a dark blue colour is produced.
dissolve the residue, ignoring any carbon, in 50 ml of water
and 14 ml of brominated hydrochloric acid AsT and remove C. The solution obtained in the test for Heavy metals gives
the excess ofbromine with 2 ml of stannous chloride solution the reactions of sodium salts (2.3.1).
AsT. The resulting solution complies with the limit test for
arsenic (2 ppm). Tests
Heavy metals (2.3.13). Dissolve 2.0 g in 46 ml of water, add pH (2.4.24).5.5 to 7.5, detennined in a2.0 percentw/v dispersion
with constant stirring 4 ml of dilute hydrochloric acid, filtering in carbon dioxide-free water.
and using 25 ml ofthe filtrate. The solution complies with the Heavy metals (2.3.13). To 4.0 g in a silica or platinum dish add
limit test for heavy metals, Method A ( 20ppm). 2 ml of a 50 per cent w/v solution of sulphuric acid, heat in a
Chlorides (2.3 .12). To 25 ml of solution A add 15 ml of water water-bath and then cautiously over a flame at about 600°.
and 10 ml of 2 M nitric acid and filter. 25 ml of the filtrate Continue heating until all black particles have disappeared,
complies with ~he limit test for chlorides (200 ppm). allow to cool, add 0.1 rol of 1 M sulphuric acid, heat to ignition
once again and allow to cool. Add 0.1 ml of 2 M ammonium
Sulphates (2.3 .17). 2.5 ml of solution A diluted to 15 ml with
carbonate, evaporate to dryness and cautiously ignite. To
distilled water complies with the limit test for sulphates
the residue add 5 ml of hydrochloric acid, evaporate to
(600 ppm).
dryness on a water-bath and dissolve the residue in 100 ml of
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined water. 25 ml ofa solution complies with the limit test for heavy
on 1.0 g by drying in an oven at 105°. metals, Method A (20 ppm).

2135

www.webofpharma.com
SODIUM STARCH GLYCOLLATE IP 2010

Iron (2.3.14). 50 ml of the solution obtained in the test for Storage. Store protected from light and moisture.
Heavy metals complies with the limit test for iron (20 ppm).
Sodium Chloride. Not more than 10.0 per cent, determined by
the following method. To 1.0 g add 20.0 ml of 0.1 M silver Sodium Stibogluconate
nitrate and 30 ml ofnitric acidand boil carefully for 30 minutes.
Cool and add a sufficient volume of a saturated solution of SodiumAntimony Gluconate
potassium permanganate to change the colour ofthe solution
to red. Discharge the colour by the dropwise addition of
OH
hydrogen peroxide solution (10 vol), add 3 m1 of dibutyl HO
phthalate and titrate with 0.1 M ammonium thiocyanate using
ferric ammonium sulphate solution as indicator, shaking OH
HO HO\/0
vigorously after each addition of titrant. Carry out a blank 0\ O-Sb-O
titration. o-stf \0
/\ ONa
1 ml of0.1 M silver nitrate is equivalent to 0.005844 g ofNaCI. o OH
NaO
Sodium glycollate. To 0.2 g add 5 m1 of glacial acetic acid, o
mix well and add 5 ml of water, stirring occasionally until o
solution is complete. Slowly add 50 ml ofacetone with stirring
and then add 1 g of sodium chloride. Filter, wash the residue Sodium Stibogluconate is mainly the disodium salt of
with acetone and dilute the filtrate to 100 m1 with acetone. ~-oxy-bis[gluconato(3)_02,()3 ,()4-hydroxoantimony].
Transfer 2 ml ofthis solution to an open flask, heat on a water- The method of manufacture is such as to ensure consistently
bath for exactly 20 minutes, cool, add 5 ml ofnaphthalenediol controlled reaction stoichiometry in order to yield sodium
reagent and mix thoroughly. Add a further 15 ml ofthe same stibogluconate that is satisfactory with regard to intrinsic
reagent, mix, cover the flask with aluminium foil and heat on a toxicity.
water-bath for 20 minutes. Cool and dilute to 25 ml with
sulphuric acid. The absorbance (2.4.7) ofthe resulting solution Sodium Stibogluconate contains not less than 30.0 per cent
at the maximum at about 540 om using water as the blank, is and not more than 34.0 per cent of pentavalent antimony,
not more than that of a solution prepared in the following calculated on the dried and methanol-free basis.
manner. To 5 ml of a 0.062 per cent w/v solution ofglycollic Category. Antiprotozoal.
acid, previously dried at a pressure not exceeding 2 kPa for
Dose. By intramuscular or intravenous injection, 600 mg to 2 g
16 hours, add 5 ml ofglacial.acetic acid, dilute to 100 ml with
daily, for 10 to 30 days.
acetone and complete the procedure described above
beginning at the words "Transfer 2 ml...." (2.0 per cent). Description. A colourless, mostly amorphous powder;
odourless or almost odourless.
Microbial Contamination (2.2.9). 1.0 g is free from Escherichia
coli and Salmonellae. Identification
Loss on drying (2.4.19). Not more than 10.0 per cent, A. An aqueous solution is dextro-rotatDly.
determined on 0.5 g by drying in an oven at 105°.
B. Pass hydrogen sulphide into a 5 percent w/v solution for
Assay. Weigh accurately about 4.0 g, add 350 ml ofa mixture several minutes; an orange precipitate is produced.
of4 volumes of ethanol (95 per cent) and 1 volume of water,
add 0.25 ml of phenolphthalein solution and mix. Add C. When heated, it chars without melting, emitting an odour
1 M sodium hydroxide dropwise until the colour of the ofburnt sugar and leaving a residue which gives the reactions
suspension becomes faintly pink, shake for 30 minutes and of antimony compounds and the reactions of sodium salts
decant through a sintered glass crucible. Repeat the extraction (2.3.1). .
three times, or until a test for chloride ions is negative. Transfer
Tests
the bulk of the residue to the crucible, wash the residue with
ethanol (95 per cent) and dry at 110° to constant weight. pH (2.4.24),5.0 to 5.6, determined in the solution obtained in
Weigh accurately 0.5 g ofthe residue, add 80 ml ofanhydrous the test for Stability of solution.
glacial acetic acid, heat under a reflux condenser for 2 hours,
cool. Titrate with 0.1 Mperchloric acid, determining the end-
StabiliiYor solution. Heat a solution contaming 10.0 per cent
wlv ofpentavalent antimony in an autoclave at 115.5° arid at a
point potentiometrically (2.4.25).
pressure of 70 kPa for 30 minutes. The resulting solution is
1 ml of 0.1 M perchloric acid is equivalent to 0.0023 g ofNa. colourless or almost colourless.

2136

www.webofpharma.com
IP 2010 SODIUM STIBOGLUCONATE INJECTION

Trivalent antimony. Dissolve 2.0 g in 30 ml ofwater, add 15 ml Sodium Stibogluconate Injection


of hydi-ochloric acid and titrate with 0.00833 M potassium
bromate using methyl orange solution as indicator. Not more Sodium Antimony Gluconate Injection
than 1.3 ml of 0.00833 M potassium bromate is required. Sodium Stibogluconate Injection is a sterile solution ofSodium
Chlorides. Dissolve 2.5 g in 50 ml of water and add 2 ml of Stibog1uconate in Water for Injections. Injection in multiple
2 M nitric acid and 75 ml ofacetate bufferpH 5.0. Titrate with dose containers must not contain phenol as preservative.
0.1 M silver nitrate, detennining the end-point potentio-
Sodium Stibogluconate Injection contains not less than
metrically (2.4.25). Not more than 3.0 ml of0.1 M silver nitrate
9.5 per cent w/v and not more than 10.5 per cent w/v of
is required.
pentavalent antimony, Sb.
Methanol. Not more than 2.0 per cent w/w, determined by gas
Usual strength. The equivalent of 100 mg of pentavalent
chromatography (2.4.13).
antimony per m!.
Test solution. Add 5 ml of water to 0.5 g of the substance
Description. A clear, colourless or faintly straw-coloured
under examination and mix with the aid ofultrasound until the
solution.
solution is complete.
Reference solution (a). Add 5 ml ofa 0.2 per centv/v solution Identification
of ethanol (internal standard) to 0.5 g of the substance under
examination and mix with the aid ofultrasound Until the solution A. It is dextro-rotatOlY.
is complete. B. Dilute 2 ml to 10 ml with water and pass hydrogen sulphide
Reference solution (b). Add 1 ml ofa 1.0 per centv/v solution through the solution; no immediate precipitate is produced.
ofmethanol to 5 ml ofa 0.2 per cent v/v solution ofthe internal Continue to pass hydrogen sulphide through the solution for
standard. several minutes; an orange precipitate is produced.
Chromatographic system C. The residue obtained by evaporation to dryness, after
- a glass column 1.5 m x 4 mm, packed with porous polymer incineration, gives the reactions of antimony compounds and
beads (80 to 100 mesh), the reactions of sodium salts (2.3.1).
- temperature: column. 130°,
Tests
Calculate the percentage w/w ofmethanol taking 0.792 g as its
weight per ml at 20°. pH (2.4.24).5.0 to 5.6.
Undue toxicity. Dissolve a suitable quantity ofthe substance Undue toxicity. Dilute a suitable volume ofthe injection under
under examination in water for injections to give a solution examination with sufficient water for injections to give a
containing 28 mg of pentavalent antimony per ml. Inject solution containing the equivalent of 28 mg of pentavalent
intravenously 0.3 ml ofthe solution into each of 10 mice that antimony per m!. Inject intravenously 0.3 ml of the solution
have been deprived of food for not less than 17 hours. After into each of 10 mice that have been deprived of food for not
injections allow the mice access to food and water. None of less than 17 hours. After injections allow the mice access to
the mice dies within 24 hours. If one of the mice dies within food and water. None of the mice dies within 24 hours. If one
24 hours, repeat the test. None of the second group of mice of the mice dies within 24 hours, repeat the test. None of the
dies within 24 hours. second group of mice dies within 24 hours.
Loss on drying (2.4.19). Not more than 15.0 percent,
Other tests. Complies with the tests stated under Parenteral
determined on 0.25 g by drying in an oven at 130° at a pressure
Preparations (Injections).
not exceeding 0.7 kPa.
Assay. Weigh accurately about 0.16 g, dissolve in 30 ml of Assay. To 1.0 ml, accurately measured, add 30 ml of
hydrochloric acid, add 70 ml of phosphoric acid and stir hydrochloric acid and 70 ml of phosphoric acid and mix.
carefully until completely mixed. Titrate with 0.05 Mferrous Titrate with 0.05 Mferric ammonium sulphate prepared using
ammonium sulphate prepared using sulphuric acid (1 per sulphuric acid (l per cent), determining the end-point
cent), determining the end-point potentiometrically (2.4.25), potentiometrically (2.4.25), using a platinum electrode and a
using a platinum electrode and a silver-silver chloride reference silver-silver chloride reference electrode.
electrode. 1 ml of 0.05 M ferric ammonium sulphate is equivalent to
1 ml of 0.05 Mferric ammonium sulphateis equivalent to 0.003044 g ofpentavalent antimony.
0.003044 g ofpentavalent antimony. Labelling. The label states the strength in terms of the
Storage. Store protected from moisture. equivalent amount of pentavalent antimony per ml.

2137

www.webofpharma.com
SODIUM THIOSULPHATE IP 2010

Sodium Thiosulphate Sulphides. To 10 ml of solution A add 0.05 ml of a "!'reshly


prepared 5 per cent w/v solution of sodium nitroprusside; the
Sodium Hyposulphite solution does not become violet.
Na2S203,5H20 Mol. Wt. 248.2 Sulphates and sulphites (2.3.17). Dilute 2.5 ml ofsolutionA to
Sodium Thiosulphate contains not less than 99.0 per cent 10 ml with distilled water. To 3 ml ofthis solution add 2 ml of
and not more than 101.0 per cent ofNa2S203,5H20. iodine solution and gradually add more iodine solution and
dilute to 15 ml with distilled water. The resulting solution
Category. Antidote to cyanide poisoning complies with the limittest for sulphates (0.2 per cent).
Dose. By intravenous or intramuscular injection, 300 mg to 1 g. Assay. Weigh accurately about 0.5 g, dissolve in 20 ml of
Description. Colourless large crystals or a coarse, crystalline water and titrate with 0.05 M iodine using starch solution,
powder; odourless; deliquescent in moist air and effloresces added towards the end of the titration, as indicator.
in dry air at temperature above 33°. It dissolves in its water of 1 ml of 0.05 M iodine is equivalent to 0.02482 g of
crystallisation at about 49°. Na2S203,5H20.
Identification Storage. Store protected from moisture.

A. To 0.5 ml ofa 10.0 per cent w/v solution in carbon dioxide-


free water (solution A) add 0.5 ml of water and 2 ml of
0.1 M silver nitrate; a white precipitate is produced which Sodium Thiosulphate Injection
quicldy becomes yellowish and [mally black.
Sodium Hyposulphite Injection
B. To a portion of solution A add a few drops of iodine
Sodium Thiosulphate Injection is a sterile solution of Sodium
solution; the colour is discharged.
Thiosulphate in Water for Injections.
C. Dilute 2.5 mlofsolutionAto 5 ml with water and add 1 ml of
Sodium Thiosulphate Injection contains not less than
hydrochloric acid; a gas is evolved which turns starch-iodate
95.0 per cent and not more than 105.0 per cent of the stated
paper blue and a precipitate of sulphur is produced.
amount of sodium thiosulphate, Na2S203,5H20.
D. 1 ml of solution A gives reaction Aof sodium salts (2.3.1).
Usual strength. 100 mg per ml.
Tests Description. A clear, colourless solution.

Appearance of solution. Solution A is clear (2.4.1), and Identification


colourless (2.4.1).
A. To 0.5 ml ofa 10.0 per cent w/v solution in carbon dioxide-
pH (2.4.24).6.0 to 8.4, determined in solution A. fi-ee water (solution A) add 0.5 ml of water and 2 ml of
Arsenic (2.3.1 0). Dissolve 5.0 g in 50 ml of water and add 10 ml 0.1 M silver nitrate; a white precipitate is produced which
of stannated hydrochloric acid AsT. The resulting solution quickly becomes yellowish and finally black.
complies with the limit test for arsenic (2 ppm). B. To a portion of solution A add a few drops of iodine
Heavy metals (2.3.13). Dissolve 1.0 g in 10 ml ofwater. Slowly solution; the colour is discharged.
add 5 ml of dilute hydrochloric acid and evaporate the mixture C. Dilute 2.5 ml ofsolution A to 5ml with water and add 1 mlof
to dryness on a water-bath. Gently boil the residue with 15 ml hydrochloric acid; a gas is evolved which turns starch-iodate
of water for 2 minutes and filter. Heat the filtrate to boiling, paper blue and a precipitate of sulphur is produced.
add sufficient bromine solution to the hot filtrate to produce
D. 1 ml of solution A gives reaction A of sodium salts (2.3.1).
a clear solution and add a slight excess of bromine solution.
Boil the solution to expel the bromine completely, cool to room Tests
temperature and add a drop of phenolphthalein solution and
sodium hydroxide solution until a slight pink colour is pH (2.4.24). 7.0t09.0.
produced. Add 2 ml of dilute acetic acid and dilute with water Other tests. Complies with the tests stated under Parenteral
to 25 ml. The solution complies with the limit test for heavy Preparations (Injections).
metals;MethodA (20 ppm);
Assay. To an accurately measured volume containing about
Chlorides (2.3.12). To 12.Sri:il orsoliitioiiA add IS fir of 0.5 g of Sodium Thiosulphate add about 20 ml of water and
2 M nitric acid, boil gently for 3 to 4 minutes, cool and filter. titrate with 0.05 M iodine, using 3 ml of starch solution, added
The filtrate complies with the limit test for chlorides (200 ppm). towards the end of the titration, as indicator.

2138

www.webofpharma.com
IP 2010 SODIUM VALPROATE

1 ml of 0.05 M iodine is equivalent to 0.02482 g of Test solution (a). Add 5 ml of 1 Msulphuric acid to 10 ml ofa
NaZSZ03,5RzO. 0.04 per cent w/v solution ofthe substance under examination
and shake with three quantities, each of20 ml, of ether. Add
Storage. Store in single dose containers.
10 ml of a 0.02 per cent w/v solution of butyric acid (internal
standard) in ether to the combined ether extracts, shake with
Sodium Valproate anhydrous sodium sulphate, filter and evaporate the filtrate
to a volume of about 10 ml at a temperaturt;l not exceeding 30°
Divalproex Sodium using a rotary evaporator.
Test solution (b). Add 0.5 ml of 1 M sulphuric acidto 10 ml of
a 0.04 per centw/v solution ofthe substance under examination
and shake with three quantities, each of 5 ml, of ether. Shake
the combined ether extracts with anhydrous sodium sulphate,
filter and evaporate the filtrate to a volume ofabout 10 ml at a
Mol. Wt. 166.2 temperature not exceeding 30° using a rotary evaporator.
Sodium Valproate is sodium 2-propylpentanoate. Test solution (c). Dissolve 0.5 g of the substance under
Sodium Valproate contains not less than 98.5 per cent and not . examination in 10 ml ofwater, add 5 ml of 1 M sulphuric acid
more than 101.0 per cent ofCaR,sNaOz, calculated on the dried and treat as described for test solution (a) beginning at the
basis. words "shake with three...".
Category. Anticonvulsant. Reference solution. Prepare in the same manner as test solution
Dose. 600 mg to 1.6 g daily, in divided doses. (b) but using sodium valproate RS in place of the substance
under examination.
Description. A white or almost white, crystalline powder;
hygroscopic. Chromatographic system
- a glass column 2.6 m x 2 rom, paclced with silariised
Identification diatomaceous support (125 to 180 mesh) impregnated
with 5 per cent w/w of polyethylene glycol 20,000
A. Determine by infrared absorption spectrophotometry (2.4.6). 2-nitroterephthalate and 1 per cent w/w ofphosphoric
Compare the spectrum with that obtained with sodium acid,
valproate RS or with the reference spectrum of sodium - temperature. colurnn.150° to 170° to obtain a retention
valproate. time ofabout 12 minutes for valproic acid [the principal
B. In the test for Related substances, the principal peak in the peak in test solution (b)],
chromatogram obtained with test solution (b) corresponds to inlet port at 200° and detector at 300°,
the peak in the chromatogram obtained with the reference - flow rate. 20 ml per minute ofthe carrier gas.
solution.
Allow the chromatography to proceed for 2.5 times the
C. Dissolve 1.25 g in 20 ml of distilled water in a separating retention time of valproic acid. Adjust the sensitivity so that
funnel, add 5 ml of2 M nitric acid, shake and allow the mixture the height of the peak due to the internal standard in the
t6 stand for 12 hours; the lower layer (solution A) gives chromatogram obtained with test solution (a) is not less than
reactions of sodium salts (2.3.1). 70 per cent of the full-scale deflection on the recorder. In the
chromatogram obtained with test solution (c), the sum of the
Tests areas of any secondary peaks is not greater than the area of
Appearance of solution. A 20.0 per cent w/v solution is not the peak due to the internal standard. Ignore any peaks the
more opalescent than opalescence standard OS2 (2.4.1), and area ofwhich is less than I per cent ofthe area ofthe peak due
is not more intensely coloured than reference solution to the internal standard. The test is not valid unless the
, YS6(2.4.l). resolution between the peak due to the internal standard and
the principal peak in the chromatogram obtained with test
Acidity or alkalinity. To 10 ml ofa 10.0 per cent w/v solution
solution (a) is at least 12.
in carbon dioxide-free water add 0.1 ml of dilute
phenolphthalein solution; not more than 0.75 ml of either Chlorides (2.3.12). Dissolve 1.25 gin 10 ml of water. The
0.1 M sodium hydroxide or 0.1 M hydrochloric acid is resulting solution complies with the limit test for chlorides
required to change the colour of the solution. (200 ppm).
Related substances. Determine by gas chromatography Sulphates (2.3.17). Solution A complies with the limit test for
(2.3.13). sulphates (200 ppm).

2139

www.webofpharma.com
SODIUM VALPROATE ORAL SOLUTION IP 2010

Heavy metals (2.3.13). 1.0 g complies with the limit test for Test solution (a). Mix a quantity ofthe oral solution containing
heavy metals, Method B. Use 2 ml of lead standard solution 0.50 g of Sodium Valproate with 10 ml of water, acidify with
(10 ppm) to prepare the standard (20 ppm). 2 M sulphuric acid and shake with three quantities, each of
20 ml, of dichloromethane. Wash the combined
Loss on drying (2.4.19). Not more than 2.0 per cent, determined
dichloromethane extracts with 10 ml of water, shake with
on 1.0 g by drying in an oven at 105°.
anhydrous sodium sulphate, filter and evaporate the filtrate
Assay. Weigh accurately about 0.15 g and dissolve in 25 ml of to a volume ofabout 10 ml at a temperature not exceeding 30°
anhydrous glacial acetic acid Titrate with 0.1 M perchloric using a rotary evaporator.
acid, determining the end-point potentiometrically (2.4.25). Test solution (b). Mix a quantity ofthe oral solution containing
Carry out a blank titration. 0.5 g of Sodium Valproate with 10 ml of a 0.02 per cent w/v
I ml of 0.1 M perchloric acid is equivalent to 0.01662 g of solution of octanoic acid (internal standard) in 0.1 M sodium
CSH1SNaOz. hydroxide and continue as described for test solution (a)
beginning at the words "acidify with 2 M sulphuric acid...".
Storage. Store protected from moisture.
Reference solution. A 0.02 per cent w/v solution ofthe internal
standard in dichloromethane.
Chromatographic system
Sodium Valproate Oral Solution - a glass column 1.5 m x 4 mm. packed with acid-washed,
silanised diatomaceous support (80 to 180' mesh)
Sodium Valproate Elixir
coated with 15 per cent w/w of free fatty acid phase
Sodium Valproate Oral Solution is a solution of Sodium (such as Supelco FFAP 2-1063) and 1 per cent w/w of
Valproate in a suitable flavoured vehicle. phosphoric acid,
Sodium Valproate Oral Solution contains not less than - temperature. column. 170°.
95.0 per cent and not more than 105.0 per cent of the stated In the chromatogram obtained with test solution (b) the sum
amount ofsodium valproate, CSH1SNaOz. of the areas of any secondary peaks is not greater than the
area of the peak due to the internal standard.
Usual strength. 200 mg in 5 ml.
Other tests. Complies with the tests stated under Oral Liquids.
Identification Assay. Weigh accurately a quantity containing about 0.15 g
ofSodium Valproate, add 50 ml ofwater, mix thoroughly and
A. Shake a quantity containing about 0.25 g of Sodium
add 10 ml of saturated solution of sodium chloride, 10 ml of
Valproate with two quantities, each of 25 ml, of chloroform
2 M hydrochloric acid and 40 ml ofa mixture of2 volumes of
and discard the chloroform extracts. Add 10 ml of a saturated
ether and 1 volume of light petroleum (40° to 60°), shake,
solution of sodium chloride and 10 ml of 2 M hydrochloric
allow to separate and reserve the ether layer. Shake the
acid, mix and shake with 25 ml of chloroform. Wash the
aqueous layer with a further 40 ml ofthe ether-lightpetroleum
chloroform layer with 5 ml of water, shake with anhydrous
mixture and discard the aqueous layer. Shake each of the
sodium sulphate, filter and evaporate to dryness.
ether extracts with the same three quantities, each of 10 ml, of
On the residue determine by infrared absorption a saturated solution ofsodium chloride, combine the extracts
spectrophotometry (2.4.6). Compare the spectrum with that and evaporate to a volume of about 1 ml at a temperature not
obtained with valproic acidRS or with the reference spectrum exceeding 30°. Add 50 ml ofethanol (95 per cent), previously
ofvalproic acid. neutralised with 0.01 M sodium hydroxide using dilute
phenolphthalein solution as indicator, and titrate with
B. Shake a quantity containing about 0.25 g of Sodium
0.1 M sodium hydroxide.
Valproate with a mixture of 10 ml of a saturated solution of
sodium chloride, 10 ml of2 M hydrochloric acid and 25 ml of 1 ml of 0.1 M sodium hydroxide is equivalent to 0.01662 g of
chloroform. Evaporate the chloroform layer to dryness, CSH1SNaOz.
dissolve the residue in 2 ml of water, make just alkaline with Determine the weight per ml of the preparation (2.4.29) and
2 M sodium hydroXide and add 0.5 ml of a 10 per cent w/v calculate the content ofCsHlsNaOz weight in volume.
solution of cobalt nitrate; a purple precipitate is produced
which is soluble in dichloromethane.
Sodium Valproate Tablets
Tests
Sodium Valproate Tablets contain not less than 95.0 per cent
Related substances. Determine by gas chromatography and not more than 105.0 per cent of the stated amount of
(2.4.13). sodium valproate, CSHlSNaOz.

2140

www.webofpharma.com
IP 2010 SORBIC ACID

Usual strengths. 100 mg; 200 mg. Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.3 g of Sodium
Identification Valproate, add 70 ml of water, shake for 10 minutes, dilute to
A. Shake a quantity of the powdered tablets containing about 100.0 ml with water and filter. To 50.0 ml ofthe filtrate add 10 ml
0.5 g ofSodium Valproate with 10 ml of water and centrifuge. of a 30 per cent w/v solution of sodium chloride and 10 ml of
AcidifY 5 ml of the supernatant liquid with 2 M sulphuric 2 M hydrochloric acid. Extract with 40 ml of a mixture of
acid, shake with 25 ml of chloroform and wash the chloroform 2 volumes of ether and 1 volume of light petroleum (boiling
layer with 5 ml of water. Dry by shaking with anhydrous range 40° to 60°) and allow to separate. Shake the aqueous
sodium sulphate, filter and evaporate the chloroform. layer with a further 40 ml ofthe ether-light petroleum mixture.
Shake each ofthe ether extracts with the same three quantities,
On the residue determine by infrared absorption each of 10 ml, of a saturated solution of sodium chloride,
spectrophotometry (2.4.6). Compare the spectrum with that combine the ether extracts and evaporate to a volume ofabout
obtained with valproic acidRS or with the reference spectrum 1 m1 at a temperature not exceeding 30°. Add 50 ml of ethanol
ofvalproic acid. (95 per cent), previously neutralised with 0.01 M sodium
B. Shake a quantity of the powdered tablets containing about hydroxide using dilute phenolphthalein solution as indicator,
0.25 g ofSodium Valproate with 3 ml of water and centrifuge. and titrate with 0.1 M sodium hydroxide.
To 2 ml of the supernatant liquid add 0.5 ml of a 10 per cent
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01662 g of
w/v solution of cobalt nitrate; a purple precipitate is produced
CSHISNa02.
which is soluble in dichloromethane.

Tests

Related substances. Determine by gas chromatography


Sorbic Acid
(2.4.13).
Test .solution (a). Shake a quantity of the powdered tablets
containing 0.50 g of Sodium Valproate with 10 ml of water,
acidifY with 2 M sulphuric acid and shake with three quantities,
each of 20 ml, of dichloromethane. Wash the combined MoL Wt. 112.1
dichloromethane extracts with 10 ml of water, shake with
Sorbic Acid is (2E,4E)-hexa-2,4-dienoic acid.
anhydrous sodium sulphate, filter and evaporate the filtrate
to a volume ofabout 10 ml at a temperature not exceeding 30° Sorbic Acid contains not less than 99.0 per cent and not more.
using a rotary evaporator. than 101.0 per cent of C 6Hs0 2 , calculated on the anhydrous
basis.
Test solution (b). Shake a quantity of the powdered tablets
containing 0.50 g ofSodiurn Valproate with 10 ml ofa 0.020 per Category. Pharmaceutical aid (antimicrobial preservative).
cent w/v solution of octanoic acid (internal standard) in Description. A white or almost white, crystalline powder.
0.1 M sodium hydroxide and continue as described for test
solution (a) beginning at the words "acidifY with 2 M sulphuric Identification
acid...".
Test A may be omitted if tests Band C are carried out. Tests B
Reference solution. A 0.02 per cent w/v ofthe internal standard
and C may be omitted if test A is carried out.
in dichloromethane.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Chromatographic system
Compare the spectrum with that obtained with sorbic acid RS
a glass column 1.5 m x 4 mm, packed with acid-washed,
or with the reference spectrum of sorbic acid.
silanised diatomaceous support (80 to 180 mesh) coated
with 15 per cent w/w of free fatty acid phase (such as B. Dissolve 50 mg in sufficient water to produce 250 ml and
Supelco FFAP 2-1063) and 1 per cent w/w ofphosphoric dilute 2 ml ofthis solution to 200 ml with 0.1 M hydrochloric
acid, acid.
- temperature. column. 170°, When examined in the range 230 urn to 360 urn (2.4.7), the
In the chromatogram obtained with test solution (b) the sum resulting solution shows an absorption maximum at about
of the areas of any secondary peaks is not greater than the 264 urn; absorbance at about 264 urn, 0.43 to 0.51.
area of the peak due to the internal standard.
C. Dissolve 0.2 g in 2 ml of ethanol (95 per cent) and add
Other tests. Comply with the tests stated under Tablets. 0.2 ml of bromine water; the solution is decolorised.

2141

www.webofpharma.com
SORBIC ACID IP 2010

Tests B. Complies with the test for iodine value (2.3.28).

Appearance ofsolution. A 5.0 per cent w/v'solution in ethanol C. Complies with the test for composition offatty acids.
(95 per cent) is clear (2.4.1), and colourless (2.4.1). D. Margaric acid. Not more than 0.2 per cent for oleic acid of
Heavy metals (2.3.13). 2.0 g complies with the limit test for vegetable origin and not more than 4.0 per cent for oleic acid
heavy metals, Method A (1 0 ppm). ofanimal origin.

Aldehydes. Not more than 0.15 per cent, determined by the Tests
following method. Dissolve 1.0 g in a mixture of 50 ml of
2-propanol and 30 ml of water, adjust the pH of the solution Relative density (2.4.29). About 0.99.
to 4.0 with 0.1 M hydrochloric acid or 0.1 M sodium Acid value (2.3.23). Not more than 8.0, determined on 5.0 g.
hydroxide and dilute to 100 ml with water. To 10 ml of the
Hydroxyl value (2.3.27). 190 to 210.
resulting solution add 1 ml of decolorised filchsin solution
and allow to stand for 30 minutes. Any colour produced is not Saponification value (2.3.37). 145 to 160. Carry out the
more intense than that obtained in a solution prepared saponification for 1 hour.
simultaneously by adding 1 ml of decolorisedfilchsin solution
Iodine value (2.3.28).62 to 76.
to a mixture of 1.5 ml of acetaldehyde standard solution
(100 ppm C2H 40), 4 ml of 2-propanol and 4.5 mlofwater. Peroxide value (2.3.35). Not more than 10.0.
Sulphated ash (2.3.18). Not more than 0.2 per cent. Composition offatty acids. Determine by gas chromatography
(2.4.13).
Water (2.3.43). Not more than 1.0 per cent, determined on
2.0g. Composition of the fatty acid fraction of the substance:
Assay. Weigh accurately about 0.2 g, dissolve in 20 ml of - myristic acid: Not more than 5.0 per cent;
ethanol (95 per cent) and titrate with 0.1 M sodium hydroxide, - palmitic acid: Not more than 16.0 per cent;
using 0.2 ml of phenolphthalein solution as indicator, until a -palmitoleic acid: Not more than 8.0 per cent;
pink colour is produced.
stearic acid: Not more than 6.0 per cent;
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01121 g of - oleic acid: 65.0 per cent to 88.0 per cent;
C6HsOz.
-linoleic acid: Not more than 18.0 per cent;
Storage. Store protected from light and moisture.
-linolenic acid: Not more than 4.0 per cent;
- fatty acids with chain length greater than C 18: Not more
than 4.0 per cent.
Sorbitan Oleate Heavy metals (2.3.13). In a silica crucible, mix thoroughly 2.0 g
of the substance under examination with 0.5 g of magnesium
oxide. Ignite to dull redness until a homogeneous white or
greyish-white mass is obtained.
Ifafter 30 min ofignition the mixture remains coloured, allow
to cool, mix using a fine glass rod and repeat the ignition. If
necessary repeat the operation. Heat at 8000 for about 1 hour.
Mol. Wt. 428.6 Take up the residue in 2 quantities, each of5 mI, of a mixture of
equal volumes of hydrochloric acid and water. Add 0.1 mlof
Sorbitan Oleate is mixture usually obtained by esterification
phenolphthalein solution and then concentrated ammonia
of 1 mole ofsorbitol and its mono-and di- anhydrides per mole
until a pink colour is obtained. Cool, add glacial acetic acid
of oleic (cis-9-octadecenoic) acid. A suitable antioxidant may
until the solution is decolourised and add 0.5 ml in excess.
be added.
Filter if necessary and wash the filter. Dilute to 20 ml with
NOTE-When sorbitan mono-oleate is demanded, Sorbitan wate/: 12 ml of resultant solution complies with the limit test
Oleate shall be supplied. for heavy metals, Method D (10 ppm).
Description;Abrownish;yellow, viscous liquid. Total ash (2.3.19). Not mOre than OSper cent, detefifiified ofi
1.5 g.
Identification
Water (2.3.43). Not more than 1.5 per cent, determined on
A. Complies with the test for hydroxyl value (2.3 .27). LOg.

2142

www.webofpharma.com
IP 2010 SORBITOL

Storage. Store protected from light. 50 ml (solution A). To 10 ml ofsolution A add 10 ml of carbon
dioxide-fi-ee water. To 10 ml of the resulting solution add
Labelling. The label states the origin of the oleic acid used
0.05 ml of phenolphthalein solution; not more than 0.2 ml of
(animal or vegetable).
0.01 M sodium hydroxide is required to change the colour of
the solution to pink. To a further 10 ml of the solution add
0.05 ml of methyl redsolution; Not more than 0.3 ml of 0.01 M
Sorbitol hydrochloric acid is required to change the colour of the
solution to red.
D-Glucitol
Specific optical rotation (2.4.22). +4.0° to +7.0°, determined in
a solution prepared in the following manner. Dissolve a mixture
CHzOH
I of5.0 g ofthe substance under examination and 6.4 g of borax
H-C-OH
I in 40 ml of water, allow to stand for 1 hour, shaking occasionally,
HO-C-H
I
dilute to 50 ml with water and filter, if necessary.
H-C-OH
I
Related substances. Determine by liquid chromatography
H-C-OH
I
(2.4.14).
CHzOH Test solution. Dissolve 5.0 g of the substance under
examination in 100.0 ml of water.
Mol. Wt. 182.2
Reference solution (a). Dissolve 0.5 g of sorbitol RS in 10.0
Sorbitol is o-glucitol, a hexahydric alcohol related to mlofwatel:
glucose. Reference solution (b). Dilute 2.0 ml of the test solution to
Sorbitol contains not less than 98.0 per cent and not more 100.0 ml with water.
than 101.0 per cent ofC 6H 140 6, calculated on the anhydrous Reference solution (c). Dilute 5.0 ml ofreference solution (b)
basis. to 100.0 ml with water.
Category. Nutrient (for parenteral administration); Reference solution (d). Dissolve 0.5 g each of sorbitol and
pharmaceutical aid (sweetening agent; excipient). mannitol (sorbitol impurity A) in 10.0 ml of water.
Description. A white, crystalline powder; odourless. Chromatographic system
a stainless steel column 30 cm x 7.8 mm, packed with
Identification strong cation exchange resin (calcium form) (9 Jlm),
A. In the Assay, the principal peak in the chromatogram column temperature. 85°,
obtained with the test solution corresponds to the principal mobile phase: water,
peak in the chromatogram obtained with reference solution flow rate. 0.5 ml per minute,
(a). refractometer at a constant temperature,
injection volume. 20 Jll.
B. Dissolve 50 mg in 3 ml of water, add 3 ml of catecholsolution
Inject reference solution (d). The test is not valid unless the
and pour the mixture into 6 ml ofsulphuric acid; a pink colour
resolution between the peaks due to sorbitol and sorbitol
is produced.
impurity A is not less than 2.0. The relative retention time with
C. Dissolve 5 g in 3 ml of water with the aid of gentle heat, reference to sorbitol for maltitol (sorbitol impurity C) is about
cool, add 7 ml of methanol, 1 ml of benzaldehyde and 1 ml of 0.6, for mannitol (sorbitol impurity A) is about 0,8 and for iditol
hydrochloric acid, mix and shake continuously for 2 hours. (sorbitol impurity B) is about 1.1.
Filter, dissolve the crystals in 20 ml of boiling sodium
Inject the test solution, reference solution (b), (c) and (d). Run
bicarbonate solution and allow to crystallise. The residue,
the chromatogram 3 times the retention time of the principal
after washing rapidly with 5 ml ofa mixture ofequal volumes
peale. In the chromatogram obtained with the test solution the
of methanol and water and drying in a current of air, melts at
area of any secondary peak is not more than the area of the
about 175°(2.4.21).
principal peak in the chromatogram obtained with reference
Tests solution (b) (2.0 per cent) and the sum of aJI the secondary
peaks is not more than 1.5 times the area ofthe principal peak
Appearance of solution. Solution A is clear (2.4.1), and in the chromatogram obtained with reference solution (b) (3.0
colourless (2.4.1). per cent). Ignore any peak with an area less than the area of
Acidity or alkalinity. Dissolve 5.0 g in sufficient carbon the principal peak in the chromatogram obtained with reference
dioxide-free water prepared from distilled water to produce solution (c) (0.1 per cent).

2143

www.webofpharma.com
SORBITOL IP 2010

Arsenic (2.3.1 0). Dissolve 5.0 g in 50 ml ofwater and add 10 ml Labelling. The label states whether or not the contents are
of stannated hydrochloric acid AsT. The resulting solution intended for use in the manufacture ofparenteral preparations.
complies with the limit test for arsenic (2 ppm).
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method A (10 ppm).
Chlorides (2.3.12). 5.0 g complies with the limit test for chlorides
Sorbitol Solution (70 Per cent)
(50 ppm). (Crystallising)
Nickel. Dissolve 10.0 g in sufficient water to produce 20 ml, Sorbitol (70 Per cent) (Crystallising)
add 3 ml of bromine water and 2 ml of a 20 per cent w/v
Sorbitol Solution (70 per cent) (Crystallising) is an aqueous
solution of citric acid, mix and add 10 ml of 6 M ammonia and
solution of hexitoIs.
1 rn1 ofa 1per cent w/v solution of dimethylglyoxime in ethanol
(95 per cent). Mix, dilute to 50 ml with water and allow to Sorbitol Solution ( 70 per cent) (Crystallising) contains not
stand for 5 minutes; any colour produced is not more intense less than 68.0 per cent w/w and not more than 72.0 per cent
than that produced by treating in the same manner and at the w/w ofhexit0 Is, calculated as D-glucitol (C 6H I40 6).
same time 1.0 ml of nickel standard solution (10 ppm Ni) Category. Pharmaceutical aid (sweetening vehicle); humectant.
diluted to 20 ml with water (1 ppm).
Description. A clear, colourless, syrupy liquid.
Sulphates (2.3.17). 12 ml ofsolution A complies with the limit
test for sulphates (125 ppm).
Identification
Reducing sugars. Dissolve 5.0 g in 3 ml of water with the aid
of gentle heat, cool, add 20 ml of cupri-citric solution and a A. Dilute 7.0 gwith40 ml of water, add 6.4 gofborax, allow to
few glass beads, heat in such a manner that the solution boils stand for 1 hour, shaking occasionally, and dilute to 50.0 ml
in 4 minutes and continue boiling for a further 3 minutes. Cool with water. Filter if necessary. The optical rotation of the
rapidly and add 100 ml ofa 2.4 per cent v/v solution ofglacial resulting solution is 0° to +1,50 (2.4.22).
acetic acid followed by 20.0 ml of 0.025 M iodine. Add, B. Dry 1 g over phosphorus pentoxide at a pressure of 1.5 to
shaking continuously, 25 ml of a 6 per cent v/v solution of 2.5 kPa. Heat 0.5 g of the residue with a mixture of 5 ml of
hydrochloric acid and, when any precipitate has redissolved, acetic anhydride and 0.5 ml pyridine with the aid ofheat and
titrate the excess iodine with 0.05 M sodium thiosulphate allow to stand for 10 minutes. Pour the mixture into 25 ml of
using 1 ml of starch solution, added towards the end of the water, allow to stand in ice for 2 hours and filter. The precipitate,
titration, as indicator. Not less than 12.8 ml of 0.05 M sodium after recrystallisation from a small volume of ethanol (95 per
thiosulphate is required. cent) and drying over phosphorus pentoxide at a pressure of
Sulphated ash (2.3.18). Not more than 0.1 per cent. 1.5 to 2.5 kPa, melts at about 100° (2.4.21).

Water (2.3.43). Not more than 1.5 per cent, determined on C. Determine by thin-layer chromatography (2.4.17), coating
LOg. the plate with a uniform 0.75-mm thick layer ofthe following
mixture. Mix 0.1 g of cm'bomer with 110 ml of water and allow
Assay. Determine by liquid chromatography (2.4.14) as to stand, with gentle stirring, for 1 hour. Adjust to pH 7.0 by
described in the test for Related substances with the following the gradual addition, with continuous shaking, of 2 M sodium
modification. hydroxide and add 30 g of silica gel H. Heat the plate at 110°
Inject the test solution and reference solution (a). for 1 hour, allow to cool and use immediately.
Calculate the content ofC6H 140 6 • Mobile phase. A mixture of 85 volumes of 2-propanol and
Sorbitol intended for use in the manufacture ofparenteral 15 volumes of a 0.2 per cent w/v solution of boric acid.
preparations without a further appropriate procedure for Test solution. Dissolve 0.35 g of the substance under
removal ofbacterial endotoxins complies with the following examination in 100 ml of ethanol (95 per cent).
additional requirement.
Reference solution. A 0.25 per cent w/v solution of sorbitol
Bacterial endotoxins (2.2.3). Not more than 4.0 Endotoxin Units RS in ethanol (95 per cent).
per g for parenteral preparations having a concentration of
Apply to the plate 2 !J.l of each solution. After development,
less than 10 per cent WIY 6fS6rbit61 and not more than 2.5
dry the plate at 100° to 105° for 15 minutes, allow to cool, spray
Endotoxin Units per g for parenteral preparations containing
with a 0.5 per cent w/v solution of potassium permanganafe
10 per cent w/v or more of sorbitol.
in 1 M sodium hydrOXide and heat at 100° for 2 minutes. The
Storage. Store protected from moisture. principal spot in the chromatogram obtained with the test

2144

www.webofpharma.com
IP 2010 SORBITOL SOLUTION ( 70 PER CENT) ( NON-CRYSTALLISING)

solution corresponds to that in the chromatogram obtained Test solution. Dissolve 1.0 g of the substance under
with the reference solution. examination in 50.0 ml of wate!:

Tests
Reference solution (a). Dissolve 65 mg of sorbitol RS in 5.0
mlof wate!:
Appearance ofsolution. A 14.0 per cent w/v solution in carbon
Reference solution (b). Dissolve 65 mg each of mannitol and
dioxide-free water (solution A) is clear (2.4.1), and colourless sorbitol in 5.0 ml of wate!:
(2.4.1).
Chromatographic system
Acidity or alkalinity. To 10 ml of solution A add 0.05 ml of
- a stainless steel column 30 cm x 7.8 mm, packed with
phenolphthalein solution; not more than 0.2 ml of 0.01 M strong cation exchange resin (calcium form) (9 /lm),
sodium hydroxide is required to change the colour of the - column temperature. 85°,
solution to pink. To a further 10 ml ofthe solution add 0.05 ml
- mobile phase: water,
of methyl red solution. Not more than 0.3 ml of 0.01 M
- flow rate. 0.5 ml per minute,
hydrochloric acid is required to change the colour of the - refractometer at a constant temperature,
solution to red.
- injection volume. 20 Ill.
Refractive index (2.4.27). 1.457 to 1.462.
Inject reference solution (b). The test is not valid unless the
Relative density (2.4.29). Not less than 1.290. resolution between the peaks due to sorbitol and mannitol
(sorbitol impurity A) is not less than 2.0. The relative retention
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
time with reference to sorbitol for mannitol (sorbitol impurity
of stannated hydrochloric acid AsT. The resulting solution
C) is about 0.8.
complies with the limit test for arsenic (2 ppm).
Inject the test solution and reference solution (a).
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method A (1 0 ppm). Calculate the content of D-sorbitol, C 6H 140 6 .
CWorides (2.3.12). 5.0 g complies with the limit test for chlorides Storage. Store protected from moisture.
(50 ppm).
Nickel. Dissolve 10.0 g in sufficient water to produce 20 ml,
add 3 rn1 of bromine water and 2 ml of a 20 per cent w/v Sorbitol Solution (70 Per cent)
solution of citric acid, mix and add 10 ml of 6 M ammonia and
1 mlofa 1 percentw/v solution ofdimethylglyoxime in ethanol (N on-Crystallising)
(95 per cent). Mix, dilute to 50 ml with water and allow to Sorbitol (70 per cent) (Non-crystallising)
stand for 5 minutes; any colour produced is not more intense
than that produced by treating in the same manner and at the Sorbitol Solution (70 per cent) (Non-crystallising) is an aqueous
same time 1.0 m1 of nickel standard solution (10 ppm Ni) solution of hydrogenated, partly hydrolysed starch.
diluted to 20 ml with water (1 ppm). Sorbitol Solution (70 per cent) (Non-crystallising) contains
Sulphates (2.3 .17). 12 ml ofsolution A complies with the limit not less than 68.0 per cent w/w and not more than 72.0 per
test for sulphates (125 ppm). cent w/w ofsolid matter and not less than 62.0 per cent w/w of
polyols expressed as D-glucitol (C 6H I4 0 6).
Reducing sugars. Dissolve 5.0 g in 3 ml of water with the aid
of gentle heat, cool, add 20 ml of cupri~citric solution and a Category. Pharmaceutical aid (sweetening vehicle); humectant.
few glass beads, heat in such a manner that the solution boils Description. A clear, colourless or faintly yellow, syrupy liquid;
in 4 minutes and continue boiling for a further 3 minutes. Cool odourless.
rapidly and add 100 ml ofa 2.4 per cent v/v solution ofglacial
acetic acid followed by 20.0 ml of 0.025 M iodine. Add, Identification
shaking continuously, 25 ml of a 6 per cent v/v solution of
A. Dilute 7.0 g with sufficient carbon dioxide-free water to
hydrochloric acid and, when any precipitate has redissolved,
produce 50 ml (solution A). To 3 ml ofa freshly prepared 10 per
titrate the excess iodine with 0.05 M sodium thiosulphate
cent w/v solution of catechol add 6 ml of sulphuric acid while
using 1 ml of starch solution, added towards the end of the
cooling in ice. To 3 ml ofthe mixture add 0.3 ml of solution A
titration, as indicator. Not less than 12.8 ml of 0.05 M sodium
and heat gently over a naked flame for 30 seconds; a pink
thiosulphate is required. colour is produced which becomes deep brownish red.
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
R Dry I g over phosphorus pentoxide at 80° at a pressure of
Assay. Determine by liquid chromatography (2.4.14). 1.5 to 2.5 kPa. Dissolve 0.5 g ofthe residue in a mixture of5 mi

2145

www.webofpharma.com
SORBITOL SOLUTION ( 70 PER CENT) ( NON-CRYSTALLISING) IP 2010

ofacetic anhydride and 0.5 ml ofpyridine with the aid ofheat Chlorides (2.3 .12). 5.0 g cotDplies with the limit test for cWorides
and allow to stand for 10 minutes: Pour the mixtureinto 25 ml (50 ppm).
ofwater, allow to stand in ice for 2 hours and filter. The residue, Nickel. Dissolve 10.0 g in sufficient water to produce 20 ml,
after recrystallisation from a small volume of ethanol (95 per add 3 ml of bromine water and 2 ml of a 20 per cent w/v
cent) and drying over phosphorus pentoxide at a pressure of solution ofcitric acid, mix and add 10 ml of 6 M ammonia and
1.5 to 2.5 kPa, melts at about 100° (2.4.21). 1 ml ofa 1per cent w/v solution ofdimethylglyoxime in ethanol
C. Determine by thin-layer chromatography (2.4.17), coating (95 per cent). Mix, dilute to 50 ml with water and allow to
the plate with a uniform 0.75-mm thick layer ofthe following stand for 5 minutes; any colour produced is not more intense
mixture. Mix 0.1 g ofcarbomer with 110 ml ofwater and allow than that produced by treating in the same manner and at the
to stand, with gentle stirring, for 1 hour. Adjust to pH 7.0 by same time 1.0 ml of nickel standard solution (10 ppm Ni)
the gradual addition, with continuous shaking, of 2 M sodium diluted to 20 ml with water (1 ppm).
hydroxide and add 30 g ofsilica gel H. Heat the plate at 110° Sulphates (2.3.17). 12 ml ofsolution A complies with the limit
for 1 hour, allow to cool and use immediately. test for sulphates (125 ppm).
Mobile phase. A mixture of 85 volumes of 2-propanol and Reducing sugars. Dissolve 5.0 g in 3 ml ofwater with the aid
15 volumes of a 0.2 per cent w/v solution of boric acid. of gentle heat, cool, add 20 ml of cupri-citric solution and a
Test solution. Dissolve 0.35 g of the substance under few glass beads; heat in such a manner that the solution boils
examination in 100 ml of ethanol (95 per cent). in 4 minutes and continue boiling for a further 3 minutes. Cool
Reference solution. A 0.25 per cent w/v solution of sorbitol rapidly and add 100 ml ofa 2.4 per cent v/v solution ofglacial
RS in ethanol (95 per cent). acetic acid followed by 20.0 ml of 0.025 M iodine. Add,
shaking continuously, 25 ml of a 6 per cent v/v solution of
Apply to the plate 2 f!l of each solution. After development, hydrochloric acid and, when any precipitate has redissolved,
dry the plate at 100° to 105° for 15 minutes, allow to cool, spray titrate the excess iodine with 0.05 M sodium thiosulphate
with a 0.5 per cent w/v solution ofpotassium permanganate using 1 ml of starch solution, added towards the· end of the
in 1 M sodium hydroxide and heat at 100° for 2 minutes. The titration, as indicator. Not less than 12.8 ml of0.05 M sodium
principal spot in the chromatogram obtained with the test thiosulphate is required.
solution corresponds to that in the chromatogram obtained
with the reference solution. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Assay. Determine by liquid chromatography (2.4.14).
Tests
Test solution. Dissolve 1.0 g of the substance under
Appearance of solution. Solution A is clear (2.4.1), and examination in 50.0 ml ofwater.
colourless (2.4.1).
Reference solution (a). Dissolve 55 mg of sorbitol RS in 5.0
Acidity or alkalinity. To 10 ml of solution A add 10 ml of mlof water.
carbon dioxide-free water. To 10 ml of the resulting solution Reference solution (b). Dissolve 65 mg of mannitol and 55
add 0.05 ml ofphenolphthalein solution; not more than 0.2 ml mg of sorbitol in 5.0 ml of water.
of 0.01 M sodium hydroxide is required to change the colour
Chromatographic system
ofthe solution to pink. To a further 10 ml of the solution add
- a stainless steel column 30 cm x 7.8 mm, packed with
0.05 ml of methyl red solution; Not more than 0.3 ml of
strong cation exchange resin (calcium form) (9 f!m),
0.01 M hydrochloric acid is required to change the colour of
- column temperature. 85°,
the solution to red.
- mobile phase: water,
Optical rotation (2.4.22). +1.5° to + 3.so, determined in a solution - flow rate. 0.5 rnl per minute,
prepared in the following manner. Dilute 7.0 g with 40 ml of - refractometer at a constant temperature,
water, add 6.4 g of borax, allow to stand for 1 hour, shaking - injection volume. 20 f!l.
occasionally, dilute to 50 ml with water and filter, ifnecessary.
Inject reference solution (b). The test is not valid unless the
Refractive index (2.4.27). 1.455 to 1.465. resolution between the peaks due to sorbitol and mannitol
Relative density (2.4.29). Not less than 1.285. (sorbitol impurity A) is not less than 2.0. The relative retention
time with reference to sorbitol for mannitol (sorbitol impurity
Arsenic (2.3.1 0). Dissolve 5.0 g in 50 rnl ofwater and add 10 rnl .
C) is about 0.8.
oCsfarznated hydrochloric aCid AsT. The resulting solution
complies with the limit test for arsenic (2 ppm). Inject the test solution and reference solution (a).

Heavy metals (2.3.13). 2.0 g complies with the limit test for Calculate the content of D-sorbitol, C6H 140 6. .
heavy metals, MethodA(lO ppm). Storage. Store protected from moisture.

2146

www.webofpharma.com
IP 2010 SPIRONALACTONE

Spironolactone top, remove the plate from the tank and allow the solvent to
evaporate. Use within 2 hours, with the flow of the mobile
phase in the direction in which the aforementioned treatment
was done.
Apply to the plate 2 fll of each solution. Allow the mobile
phase to rise 12 cm. Dry the plate in a current ofwarm air, allow
the solvent to evaporate, heat at 120° for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120° for a further 10 minutes, allow to cool and examine
o in d'aylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with
reference solution (a). The principal spot in the chromatogram
obtained with reference solution (b) appears as a single,
Mol. Wt. 416.6
compact spot.
Spironolactone is 7a-acetylthio-3-oxo-17a-pregn-4-ene-
21,17~-carbolactone.
C. Shake about 10 mg with 2 ml of sulphuric acid
(50 per cent); an orange solution with an intense yellowish
Spironolactone contains not less than 97.0 per cent and not fluorescence is produced. Heat the solution gently; the colour
more than 102.0 per cent ofC 24 H320 4S, calculated on the dried becomes deep red and hydrogen sulphide is evolved which
basis. turns lead acetate paper black. Add the solution to 10 ml of
Category. Diuretic. water; a greenish yellow solution is produced which shows
opalescence or a precipitate.
Dose. 100 to 200 mg daily, in divided doses.
Description. A yellowish white to buff coloured powder; Tests
odourless or with a slight odour ofthioacetic acid. It exhibits
Specific optical rotation (2.4.22). -33.00 to-37.0°, determined
polymorphism.
in a 1.0 per cent w/v solution in chloroform.

Identification Related substances. Determine by liquid chromatography


(2.4.14).
Test A may be omitted if tests Band C are carried out. Test B
Test solution. Dissolve 62.5 mg of the substance under
may be omitted if tests A and C are carried out.
examination in 2.5 ml oftetrahydrojUran and dilute to 25.0 rnl
A. Determine by infrared absorption spectrophotometry (2.4.6). with the mobile phase.
Compare the spectrum with that obtained with spironolactone
Reference solution (a). Dilute 1.0 ml of the test solution to
RS or with the reference spectrum ofspironolactone. Examine
100.0 ml with the mobile phase.
the substances as 5 per cent w/v solutions in chloroform.
Reference solution (b). Dissolve 25 mg of canrenone RS in
B. Determine by thin-layer chromatography (2.4.17), coating
1.0 ml of tetrahydrojUran and dilute to 10.0 ml with the mobile
the plate with silica gel G.
phase.
Solvent mixture. A mixture of 90 volumes of acetone and
Reference solution (c). Dilute 1.0 rnl ofreference solution (b)
10 volumes of 1,2-propanediol.
to 100.0 ml with the mobile phase.
Mobile phase. A mixture of 40 volumes of cyclohexane and
Reference solution (d). Dilute 1.0 ml ofthe test solution and 1
10 volumes of toluene.
ml ofreference solution (b) to 100.0 ml with the mobile phase.
Test solution. Dissolve 25 mg of the substance under
Reference solution (e). Dilute 0.5 ml ofreference solution (a)
examination in 10 m1 ofthe solvent mixture.
to 10.0 ml with the mobile phase.
Reference solution (a). Dissolve 25 mg of spironolactone RS
in 10 ml ofthe solvent mixture. Chromatographic system
- a stainless steel column 15 cm x 4.6 mm, packed with
Reference solution (b). Mix equal volumes ofthe test solution octylsilane bonded to porous silica (5 flm),
and reference solution (a). - mobile phase: a mixture of8 volumes of acetonitrile, 18
Place the dry plate in a tank containing a shallow layer of the volumes of tetrahydrofitran and 74 volumes of water,
solvent mixture, allow the solvent mixture to ascend to the - flow rate. 1.8 ml per minute,

2147

www.webofpharma.com
SPIRONALACTONE IP 2010

- spectrophotometer set at 254 urn and283 nm, Spironolactone Tablets


- injection volume. 20 l.tI.
Spironolactone Tablets contain not less than 95.0 per cent
Inject reference solution (d). The test is not valid unless the
and not more than 105.0 per cent of the stated amount of
resolution between the peaks due to canrenone and
spironolactone, Cz4H32 0 4S. The tablets may contain added
spironolactone is not less than 1.4 and the signal-to-noise
flavouring agents.
ratio of the principal peak is not less than 6.
Usual strengths. 25 mg; 50 mg; 100 mg.
Inject the test solution, reference solution (a), (c), (d) and (e).
At 254 urn, run the chromatogram twice the retention time of Identification
the principal peale. In the chromatogram obtained with the test
solution, the sum ofthe areas ofall the secondary peaks other A. Extract a quantity of the powdered tablets containing
than canrenone, is not more than the area ofthe principal peak 0.125 g of Spironolactone with two quantities, each oflO ml,
in the chromatogram obtained with reference solution (a) (1.0 of chloroform, filter, evaporate the combined filtrates to
per cent). Ignore any peak with an area less than the area of dryness and dissolve the residue in 2.5 ml of chloroform.
the principal peale in the chromatogram obtained with reference On the resulting solution determine by infrared absorption
solution (e). At 283 rim, the area ofthe peak due to canrenone spectrophotometry (2.4.6). Compare the spectrum with that
is not more than the area of the corresponding peak in the obtained with spironolactone RS or with the reference
chromatogram obtained with reference solution (c) (1.0 per spectrum of spironolactone.
cent). Calculate the content of canrenone at 283 urn and the
content ofthe other impurities at 254 nm. The sum ofareas of B. Determine by thin-layer chromatography (2.4.17), coating
all the peaks is not more than 1.0 per cent. the plate with silica gel G.

Chromium. Mix 0.2 g with 1 g of potassium carbonate and Solvent mixture. A mixture of 90 volumes of acetone and
0.3 g of potassium nitrate in a platinum crucible, heat gently 10 volumes of 1,2-propanediol.
until fused and ignite at 6000 to 6500 until the carbon is removed. Mobile phase. A mixture of 40 volumes of cyclohexane and
Cool, dissolve the residue as completely as possible in 10 ml 10 volumes of toluene.
of water with the aid of gentle heat, filter and dilute to 20 ml
with water. To 10 ml ofthe solution add 0.5 g of urea and just Test solution. Extract a quantity of the powdered tablets
acidify with sulphuric acid (14 per cent). When effervescence containing 50 mg ofSpironolactone with 10 ml of chloroform,
ceases add a further 1 ml of sulphuric acid (14 per cent), filter and evaporate the filtrate to dryness. Dissolve 25 mg the
residue in 10 ml ofthe solvent mixture.
dilute to 20 ml with water and add 0.5 ml of diphenylcarbazide
solution. Any colour produced is not more intense than that Reference solution (a). Dissolve 25 mg of spironolactone RS
obtained by adding 1 ml of sulphuric acid (14 per cent) to in 10 ml ofthe solvent mixture.
0.5 ml of a freshly prepared 0.00283 per cent w/v solution of
Reference solution (b). Mix equal volumes ofthe test solution
potassium dichromate, diluting to 20 ml with water and adding
and reference solution (a).
0.5 ml of diphenylcarbazide solution (50 ppm).
Place the dry plate in a tank containing a shallow layer ofthe
Free mercapto compounds. Shake 2.0 g with 20 ml ofwater for
solvent mixture, allow the solvent mixture to ascend to the
1 minute and filter. To 10 ml of the filtrate add 0.05 ml of
top, remove the plate from the tank and allow the solvent to
0.01 M iodine and 0.1 ml of starch solution and mix; a blue
evaporate. Use within 2 hours, with the flow of the mobile
colour develops.
phase in the direction in which the aforementioned treatment
Sulphated ash (2.3.18). Not more than 0.1 percent. was done.

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Apply to the plate 2 III of each solution. Allow the mobile
on 1.0 g by drying in an oven at 105 0 for 3 hours, phase to rise 12 em. Dry the plate in a current ofwarm air, allow
the solvent to evaporate, heat at 1200 for 15 minutes and spray
Assay. Weigh accurately about 50 mg, dissolve in sufficient the hot plate with ethanolic sulphuric acid (20 per cent v/v).
methanol to produce 250.0 ml, dilute 5.0 ml to 100.0 ml with Heat at 1200 for a further 10 minutes, allow to cool and examine
methanol and measure the absorbance of the resulting in daylight and in ultraviolet light at 365 nm. The principal
solution at the maximum at about 238 urn (2.4.7). spot in the chrOlnatogram obtained \Vith the test solution
cOl:responds to that in -the chroIIl-atogram obtained with
Calculate the content ofC z4H 32 0 4S talcing 470 as the specific
absorbance at 238 urn. reference solution (a). The pnnCipalspofintlie Clrromatogram
obtained with reference solution (b) appears as a single,
Storage. Store protected from light and moisture. compact spot.

2148

www.webofpharma.com
IP 2010 STAVUDINE

C. Shake about 10 mg of the residue obtained in test B with - flow rate. 1.8 ml per minute,
2 ml of sulphuric acid (50 per cent); an orange solution with - spectrophotometer set at 254 nm and 283 nm,
an intense yellowish fluorescence is produced. Heat the - injection volume. 20 f..ll.
solution gently; the colour becomes deep red and hydrogen
Inject reference solution (d). The test is not valid unless the
sulphide is evolved which turns lead acetate paper black.
resolution between the peaks due to canrenone and
Add the solution to 10 ml ofwater;'a greenish yellow solution
spironolactone is not less than 1.4 and the signal-to-noise
is produced which shows opalescence or a precipitate.
ratio of the principal peak is not less than 6.
Tests Inject the test solution, reference solution (a), (c), (d) and (e).
Dissolution (2.5.2). At 254 nm, run the chromatogram twice the retention time of
principal peak. In the chromatogram obtained with the test
Apparatus. No.1,
solution, the sum ofthe areas ofall the secondary peaks other
Medium. 1000 ml of 0.1 M hydrochloric acid containing than the canrenone, is not more than the area of the principal
0.1 per cent w/v of sodium dodecyl sulphate, peak in the chromatogram obtained with reference solution
Speed and time. 75 rpm and 60 minutes. (a) (1.0 per cent). Ignore any peak with an area less than the
Withdraw a suitable volume of the medium, filter through a area ofthe principal peak in the chromatogram obtained with
membrane filter disc with an average pore diameter not greater reference solution (e). At 283 nm, the area of the peak due to
than 1.0 f..lm. Measure the absorbance of the filtrate, suitably canrenone is not more than the area of the corresponding
diluted ifnecessary, at the maximum at about 242 nm (2.4.7). peak in the chromatogram obtained with reference solution
Calculate the content of Cz4H 32 0 4S in the medium taking 445 (c) (1.0 per cent). Calculate the content ofcanrenone at 283 nm
as the specific absorbance at 242 nm. and the content ofthe other impurities at 254 nm. The sum of
areas of all the peaks is not more than 1.0 per cent.
D. Not less than 70.0 per cent ofthe stated amount ofCz4 H32 0 4S.,
Other tests. Comply with the tests stated under Tablets.
Related substances. Determine by liquid chromatography
(2.4.14). Assay. Weigh and powder 20 tablets. Weigh accurately a
Test solution. Disperse a quantity of the powdered tablets quantity of the powder containing about 25 mg of Spirono-
containing about 62.5 mg of Spironolactone with 25 ml of lactone, add 100 ml of methanol and heat just to boiling, with
chloroform, sonicate for 15 minutes, centrifuge and filter the swirling. Cool, add sufficient methanol to produce 250.0 ml,
supernatant. Repeat the procedure on the residue with a dilute 10.0 ml to 100.0 ml with methanol and measure the
further 25 ml of chloroform. Combine the chloroform extracts absorbance ofthe resulting solution at the maximum at about
and evaporate to dryness using a rotary evaporator. Add 2.5 238 nm (2.4.7).
ml of tetrahydrojUran and 22.5 ml of the mobile phase to the Calculate the content ofC z4 H32 0 4S taking 470 as the specific
residue. absorbance at 238 nm.
Reference solution (a). Dilute 1.0 ml of the test solution to
100.0 ml with the mobile phase.
Storage. Store protected from light and moisture.

Reference solution (b). Dissolve 25 mg of canrenone RS in


1.0 ml oftetrahydrojUran and dilute to 10.0 ml with the mobile
phase. Stavudine
Reference solution (c). Dilute 1.0 ml ofreference solution (b)
to 100.0 ml with the mobile phase.
Reference solution (d). Dilute 1.0 ml of the test solution and
1.0 ml of reference solution (b) to 100.0 ml with the mobile
phase.
Reference solution (e). Dilute 0.5 ml ofreference solution (a)
to 10.0 ml with the mobile phase.
Chromatographic system Mol. Wt. 224.2
- a stainless steel column 15 cm x 4.6 rom, packed with
Stavudine is 2',3'-didehydro-3'-deoxythymidine.
endcapped octylsilane bonded to porous silica (5 f..lm)
(such as Spherisorb C8), Stavudine contains not less than 98.0 per cent and not more
- mobile phase: a mixture of8 volumes of acetonitrile, 18 than 102.0 per cent of C IOH 12N z0 4, calculated on the dried
volumes of tetrahydrojUran and 74 volumes of water, basis.

2149

www.webofpharma.com
STAVUDINE IP 2010

Category. Antiretroviral. Reference solution (c). A 0.05 per cent w/v solution of
Dose. 30 to 40 mg twice daily depending on body weight of stavudine RS in the mobile phase.
the patient. Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
Description. A white or almost white powder.
octadecylsilane bonded to porous silica (5 !-Lm),
- column temperature 50°,
Identification
- mobile phase: a mixture of20 volumes of methanol and
A. Determine by infrared absorption spectrophotometry (2.4.6). 80 volumes of water,
Compare the spectrum with that obtained with stavudine RS - flow rate. 1 ml per minute,
or with the reference spectrum of stavudine. - spectrophotometer set at 265 rrm,
- injection volume. 20 !-Ll.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak due Inject separately reference solutions (a) and (c). The order of
to stavudine in the chromatogram obtained with reference elution in the chromatogram obtained with reference solution
solution (c). (a) is thymine, a-thymidine and stavudine with relative
retention times of about 0.5, 0.7 and 1.0 respectively. The test
C. Melts at about 164° (2.4.21).
is not valid unless the column efficiency determined from the
stavudine peak in the chromatogram obtained with reference
Tests
solution (a) is not less than 3000 theoretical plates, the tailing
Specific optical rotation (2.4.22). -39.0° to-46.0°, determined factor is not more than 1.5 and the relative standard deviation
in a 0.7 per cent w/v solution in water. for replicate injections of reference solution (c) is not more
than 2.0 per cent.
Related substances. Determine by liquid chromatography
(2.4.14), as described in the Assay. Inject separately the test solution and reference solution (c)
and measure the responses of the principal peak.
Separately inject the test solution, reference solutions (a) and
(b) and record the chromatograms for at least three times the Calculate the content OfCIOHlzNz04.
retention time of the principal peak. In the chromatogram Storage. Store protected from light and moisture.
obtained with the test solution, the area of any peak
corresponding to thymine is not greater than the area of the
peak in the chromatogram obtained with reference solution
(b) (1 per cent); the area of any peak corresponding to
Stavudine Capsules
~-thymidine is not greater than the area of the corresponding Stavudine Capsules contain not less than 90.0 per cent and
peak in the chromatogram obtained with reference solution not more than 110.0 per cent ofthe stated amount ofstavudine,
(a) (1 per cent); the area ofany individual impurity peak other CIOHIZNz04.
than thymine and ~-thymidine is not more than 0.5 per cent Usual strengths. 30 mg; 40 mg.
and the sum of the areas of all the impurity peaks other than
thymine and ~-thymidine is not more than 1.0 per cent. Identification
Heavy metals (2.3.13). 1.0 g complies with the limit test for A.Shake a quantity of the mixed contents of the capsules
heavy metals, Method B (20 ppm). containing about 50 mg of Stavudine with 80 ml of water for
Sulphated ash (2.3.18). Not more than 0.3 percent. 10 minutes. Add sufficient water to produce 100 ml, mix and
filter. Dilute 10 ml ofthe filtrate to 100 ml with water.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
When examined in the range 200 nm to 300 nm (2.4.7), the
on 1.0 g by drying at 105° for 3 hours.
resulting solution shows an absorption maximum at about
Assay. Determine by liquid chromatography (2.4.14). 265rrm.
Test solution. Dissolve 50.0 mg of the substance under B. In the Assay, the principal peak in the chromatogram
examination in 100.0 ml ofthe mobile phase. obtained with the test solution corresponds to the peak due
to stavudine in the chromatogram obtained with the reference
Reference solution (a). A solution containing 0.05 per cent
solution.
w/v of stavudineRS;--O;Ol 'percent-w/v of thymine and
0.0005 per cent w/v of a-thymidine in the mobile phase; Tests
Reference solution (b). A 0.0005 per cent w/v solution of Related substances. Determine by liquid chromatography
thymine in the mobile phase. (2.4.14).

2150

www.webofpharma.com
IP 2010 STAVUDINE CAPSULES

Test solution. Mix well the contents of20 capsules and transfer Test solution. The filtrate obtained as given above.
an accurately weighed quantity containing about 50 mg of
Reference solution. In the case of capsules containing 30 mg
Stavudine to a 100-ml volumetric flask. Add about 60 ml of
of Stavudine, weigh accurately about 30 mg of stavudine RS
water, mix with the aid ofultrasound for 10 minutes, dilute to
and transfer to a 1OO-ml volumetric flask. Dissolve and dilute
volume with water, mix and filter.
to volume with 0.01 M hydrochloric acid. Dilute 5.0 ml ofthis
Reference solution. Weigh 5 mg each of stavudine RS and solution to 50.0 ml with 0.01 M hydrochloric acid; in the case
thymine RS and transfer to a 25-ml volumetric flask. Add 5 ml of capsules containing 40 mg of stavudine, weigh accurately
of methanol, mix with the aid of ultrasound to dissolve and about 40 mg ofstavudine RS and transfer to a 1OO-ml volumetric
dilute to volume with water. flask. Dissolve and dilute to volume with 0.01 Mhydrochloric
acid. Dilute 5.0 ml of this solution to 50.0 ml with
Chromatographic system
0.01 M hydrochloric acid.
a stainless steel column 25 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 11m), Chromatographic system
mobile phase: gradient mixtures of acetonitrile and a stainless steel column 15 cm x 4.6 mm, packed with
0.1 M ammonium acetate, octadecylsilane bonded to porous silica (5 11m),
flow rate. I ml per minute, - mobile phase: a mixture of 85 volumes of water and
a linear gradient programme using the conditions given 15 volumes of methanol,
below, - flow rate. 1.5 ml per minute,
spectrophotometer set at 270 nm, - spectrophotometer set at 266 nm,
injection volume. 20 Ill. injection volume. 20 Ill.
Time 0.1 M Ammonium acetate Acetonitrile Inject the reference solution and record the chromatograms
(in min.) (per cent v/v) (per cent v/v) for twice the retention time of stavudine. The test is not valid
o 95 5 unless the column efficiency detelmined from the stavudine
5 95 5 peak is not less than 3000 theoretical plates, the tailing factor
25 20 80 is not more than 1.5 and the relative standard deviation for
replicate injections is not more than 1.5 per cent
30 20 80
31 95 5 Inject separately the test solution and the reference solution
and measure the responses for the major peale
35 95 5
Inject the reference solution. The test is not valid unless the Calculate the content ofCIOHI2N204 in the medium.
column efficiency determined from the peaks of stavudine D. Not less than 70 per cent of the stated amount of
and thymine is not less than 7000 theoretical plates and the CIOH12N204.
tailing factor is not more than 2.0.
Other tests. Comply with the tests stated under Capsules.
Inject separately water and the test solution. Examine the
water chromatogram for any extraneous peaks and ignore the Assay. Detennine by liquid chromatography (2.4.14).
corresponding peaks observed in the chromatogram obtained Test solution. Weigh accurately a quantity of the mixed
with the test solution. contents of20 capsules containing about 50 mg ofStavudine
Any secondary peak observed in the chromatogram obtained and transfer to a 1OO-ml volumetric flask. Add about 60 ml of
with the test solution corresponding to thymine is not more water, mix with the aid ofultrasound for 10 minutes, dilute to
than 3.0 per cent. Any other secondary. peak observed in the volume with water, mix and filter. Further dilute 10.0 ml ofthe
chromatogram obtained with the test solution is not more filtrate to 25.0 ml with water.
than 0.5 per cent and the sum ofthe areas of all the secondary Reference solution. Weigh accmately about 50 mg ofstavudine
peaks is not more than 4.0 per cent when calculated by. RS and transfer to a 1OO-ml volumetric flask. Add about 60 ml
percentage area normalisation. of water, mix with the aid ofultrasound to dissolve and dilute
Dissolution (2.5.2). to volume with water. Dilute 10.0 ml ofthis solution to 25.0 ml
Apparatus No.1, with water.
Medium. 900 ml of 0.01 M hydrochloric acid, Chromatographic system
Speed and time. 75 rpm and 45 minutes. a stainless steel column 25 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 11m),
Withdraw asuitable volume ofthe medium and filter.
mobile phase: a mixture of 5 volumes of acetonitrile
Determine by liquid chromatography (2.4.14). and 95 volumes of 0.1 M ammonium acetate,

2151

www.webofpharma.com
STAVUDINE ORAL SOLUTION IP 2010

flow rate. 1 ml per minute, Reference.solution (a). A 0.05 per cent w/v solution of
- spectrophotometer set at 270 nm, stavudine RS in water.
- injection volume. 20 III
Reference solution (b). Dilute 1ml ofreference solution (a) to
Inject the reference solution. The test is not valid unless the 100 ml with water.
column efficiency determined from the stavudine peak is not
Reference solution (c). A solution containing 0.0125 per cent
less than 3000 theoretical plates, the tailing factor is not more
w/v each ofthymine and thymidine in water. Dilute 2 ml ofthe
than 2.0 and the relative standard deviation for replicate
solution to 100 ml of water.
injections is not more than 2.0 per cent.
Use the chromatographic system described under Assay.
Inject separately the test solution and the reference solution
and measure the responses for the major peak. Iriject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
Calculate the content of ClOHIZNz04 in the capsules.
not less than 2000 theoretical plates.
Storage. Store protected from moisture.
Inject reference solution (c). The test is not valid unless the
resolution between thymine and thymidine peak is not less
than 8.4.
Inject the test solution and reference solution (b). Run the
Stavudine Oral Solution chromatogram for 4 times the retention time (about 9 minutes)
Stavudine Oral Solution is a mixture consisting of Stavudine of the principal peak. In the chromatogram obtained with the
test solution, the area of any secondary peak is not more than
with buffering agents and other excipients. It contains a
suitable flavouring agent. It is filled in a sealed container. 1.5 times the area of the peak in the chromatogram obtained
with the reference solution (b) (1.5 per cent) and the sum of
The oral solution is constituted by dispersing the contents of areas of all the secondary peaks is not more than 3 times the
the sealed container in the specified volume of water just area of the peak in the chromatogram obtained with the
before issue. reference solution (b) (3.0 per cent).
Stavudine Oral Solution contains not less than 90.0 per cent Other tests. Complies with the tests stated under Oral Liquids.
and not more than 110.0 per cent of the stated amount of
Water (2.3.43). Not more than 2.0 per cent, determined on
stavudine CIOHIZNz04.
0.5g.
Storage. Store the constituted solution in a refrigerator
Assay. Determine by liquid chromatography (2.4.14).
(2° to 8°). Discard any unused portion after 30 days of
reconstitution.. NOTE - Prepare the solutions immediately before use.
The contents of the sealed container comply with the . Test solution. Weigh accurately a quantity ofthe reconstituted
requirements stated under Oral Liquids and with the solution containing 10 mg of Stavudine, disperse in sufficient
following requirements. water and diluted to 100.0 ml with water.
Reference solution (a). A 0.01 per cent w/v solution of
Identification stavudine RS in water.
In the Assay, the principal peak in the chromatogram obtained Reference solution (b). A solution containing 0.0125 per cent
with the test solution corresponds to the peak in the w/v each ofthymine and thymidine in water. Dilute 2 mlofthe
chromatogram obtained with reference solution (a). solution to 100 ml with water.
Chromatographic system
Tests a stainless steel column 3.3 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 /lm),
pH (2.4.24).5.0 to 7.0, determined in the reconstituted solution.
mobile phase: A. 95 volumes of 25 mM ammonium
Related substances. Determine by liquid chromatography acetate and 5 volumes of methanol,
(2.4.14). B. 50 volumes of 25 mM ammonium
acetate and 50 volumes of methanol.
NOTE - Prepare the solutions immediately before use.
flow rate. ml per minute,
Test solution. Weigh accurately a quantity ofthe reconstituted - a linear gradient programme using the conditions given
solution containing 10 mg of stavudine and dissolve in 20 ml below,
of water. spectrophotometer set at 268 nm,

2152

www.webofpharma.com
IP 2010 STAVUDlNE AND LAMIVUDINE TABLETS

injection volume. 20 /ll. 25-ml volumetric flask. Add 5 ml of methanol, sonicate to


Time mobile phase A mobile'phase B dissolve, dilute to volume with water and mix.
(in min.) (per cent v/v) (per cent v/v) Reference solution (b). Weigh accurately about 100 mg of
o 100 o lamivudine RS, transfer to a 200 ml volumetric flask, add about
60 100 o 100 ml ofwater and mix with the aid ofultrasound to dissolve.
Add 10 ml ofreference solution (a) to this solution and dilute
120 0 100
to volume with water and mix.
155 100 o
Chromatographic system
Inject reference solution (b). The test is not valid unless the a stainless steel column 25 cm x 4.6 rom, packed with
resolution between thymine and thymidine is not less than octylsilane bonded to porous silica (5 /lm),
8.4. mobile phase: gradient mixtures of acetonitrile and
Inject reference solution (a). The test is not valid unless the 0.1 M ammonium acetate,
tailing factor is not more than 2.0, the column efficiency in not flow rate. 1 ml per minute,
less than 2000 theoretical plates and the relative standard - a linear gradient programme using the conditions given
deviation for replicate injections is not more than 2.0 per cent. below,
spectrophotometer set at 270 urn,
Inject the test solution and reference solution (a).
injection volume. 20 /ll.
Calculate the content ofC IOH 12N 20 4 in the oral solution. Time 0.1 M Ammonium acetate Acetonitrile
Storage. Store protected from moisture, at a temperature not (in min.) (per cent v/v) (per cent v/v)
exceeding 30°. o 95 5
5 95 5
25 20 80
,Stavudine and Lamivudine Tablets 30 20 80
Stavudine and Lamivudine Tablets contain not less than 31 95 5
90.0 per cent and not more than 110.0 per cent of the stated 35 95 5
amounts of stavudine, CIOHI2N204 and lamivudine, Inject separately reference solutions (b) and (c). The test is
CSHIIN303S, not valid unless the column efficiency determined from the
Usual strengths. Stavudine, 30 mg and Lamivudine, 150 mg; thymine, stavudine and lamivudine peaks is not less than
Stavudine, 40 mg and Lamivudine, 150 mg. 3000 theoretical plates and the tailing factor for the same peaks
is not more than 2.0.
Identification
Inject separately water and the test solution. Examine the
In the Assay, the two principal peaks in the chromatogram chromatogram obtained with water for any extraneous peaks
obtained with the test solution correspond to the peaks due and ignore the corresponding peaks observed in the
to stavudine and lamivudine in the chromatogram obtained chromatogram obtained with the test solution.
with the reference solution.
Any secondary peak observed in the chromatogram obtained
Tests with the test solution corresponding to thymine is not more
than 3.0 per cent. Any other secondary peak observed in the
Related substances. Determine by liquid chromatography chromatogram obtained with the test solution is not more
(2.4.14). than 0.5 per cent and the sum ofthe areas ofall the secondary
Test solution. Weigh accurately a quantity of the powdered peaks is not more than 4.0 per cent when calculated by
tablets containing about 100 mg oflamivudine and transfer to percentage area normalisation.
a 200-ml volumetric flask. Add about 100 ml ofwater, mix with Dissolution (2.5.2).
the aid of ultrasound for 10 minutes with occasional shaldng
Apparatus No.1,
to obtain a uniform dispersion, cool to room temperature, dilute
Medium. 900 ml of 0.01 M hydrochloric acid,
to volume with water and mix. Filter through a membrane filter
disc with an average pore diameter not greater than Speed and time. 50 rpm and 45 minutes.
1.0 /lm, rejecting the first few ml ofthe filtrate. Withdraw a suitable volume ofthe medium and filter.
Reference solution (a). Weigh accurately about 65 mg of Determine by liquid chromatography (2.4.14).
stavudine RS and 6.5 mg of thymine RS, and transfer to a Test solution. The filtrate obtained as given above.

2153

www.webofpharma.com
STAVUDINE AND LAMIVUDINE TABLETS IP 2010

Reference solution. In the case of tablets containing 30 mg of than 1.0 11m, rejecting the first few ml of the filtrate. Dilute
stavudine, weigh accurately about 33 mg of stavudine RS and suitably with the same solvent mixture to obtain a solution
167 mg of lamivudine RS and transfer to a 100 ml volumetric with a concentration of0.15 mg per ml oflamivudiIie.
flask. Add about 20 ml of methanol, sonicate to dissolve and
Reference solution. A similarly prepared solution containing
dilute to volume with a solvent mixture of equal volumes of
0.015 per cent w/v of lamivudine RS and a concentration of
methanol and water (diluent). Dilute 5.0 ml of this solution to
stavudine RS similar to that ofthe concentration ofstavudine
50.0 ml with 0.01 M hydrochloric acid; in the case oftablets
in test solution.
containing 40 mg ofstavudine, weigh accurately about 44 mg
of stavudine RS and 167 mg of lamivudine RS and transfer to Use the chromatographic system described under Dissolution.
a 100-ml volumetric flask. Add about 20 ml of methanol, mix Inject the reference solution. The test is not valid unless the
with the aid of ultrasound to dissolve and dilute to volume column efficiency determined from the lamivudine peak is not
with the diluent. Dilute 5.0 ml ofthis solution to 50.0 ml with less than 2000 theoretical plates, the tailing factor for the
0.01 M hydrochloric acid. individual peaks due to lamivudine and stavudine is not more
Chromatographic system than 1.5 and the relative standard deviation for replicate
a stainless steel column 15 cm x 4.6 mm, packed with injections for each of the peaks corresponding to stavudine
octadecylsilane bonded to porous silica (5 11m), and lamivudine is not more than 2.0 per cent.
mobile phase: a mixture of35 volumes of methanol and Inject separately the test solution and the reference solution
65 volumes of a buffer prepared by dissolving 0.68 g of and measure the responses for the major peaks.
potassium dihydrogen phosphate and 1.0 g of sodium
octanesulphonate in 1000.0 ml of water to which 1 mlof Calculate the contents of CIOHIZNz04 and CgHIIN303S in the
triethylamine is added and the pH of which is adjusted tablets.
to 2.5 with phosphoric acid. Storage. Store protected from moisture.
flow rate. 1 ml per minute,
spectrophotometer set at 266 nm,
- injection volume. 10 Ill.
Inject the reference solution. The test is not valid unless the Stearic Acid
column efficiency determined from the lamivudine peak is not
less than 2000 theoretical plates, the tailing factor for the COOH
individual stavudine and lamivudine peaks is not more than
1.5 and the relative standard deviation for replicate injections
Stearic Acid consists ofa mixture offatty acids, chiefly stearic
for each of the peaks corresponding to stavudine and
acid (ClgH360Z) and palmitic acid (C I6H 32 0 Z). It may contain a
lamivudine is not more than 1.0 per cent.
suitable antioxidant.
Inject separately the test solution and the reference solution
Stearic Acid contains not less than 40.0 per cent of CIgH360Z
and measure the responses for the major peaks due to
and the sum of the contents of CIgH360Z and C I6H 320 Zis not
stavudine and lamivudine.
less than 90.0 per cent.
Calculate the contents ofCIOHlzNz04and CgHIIN303S in the
Category. Pharmaceutical aid (tablet lubricant; emulsion
medium.
adjunct).
D. Not less than 70 per cent of the stated amounts of
Description. A white powder or white, greasy, flaky crystals
CIOH 1zNZ0 4and CgHIIN303S.
or hard masses showing signs of crystallisation.
Other tests. Comply with the tests stated under Tablets.
Assay. Determine by liquid chromatography (2.4.14). Identification
Test solution. Weigh and powder 20 tablets. Transfer an In the Assay, the chromatogram obtained with the test solution
accurately weighed quantity of the powder containing about shows two principal peaks which correspond to the two
150 mg ofLamivudine to a 100-ml volumetric flask, add about principal peaks in the chromatogram obtained with reference
20 ml of methanol and 50 ml ofa mixture ofequal volumes of solution (c).
water and methanol,!!li;x,,,,,i!h _!h~lliQor1l1tra.~()_1l1!.c! J()!
5 minutes with occasional shaking to obtain a uniform Tests
dispersion:·CooltoroomtemperaiiifeanddHiite to volume Congealing temperature (2.4.1 0). Not lower than 54°.
with the same solvent mixture. Filter the solution through a
membrane filter disc with an average pore diameter not greater Acid value (2.3.23). 200 to 212, determined on 1.0 g.

2154

www.webofpharma.com
IP 2010 STEARYL ALCOHOL

Iodine value (2.3.28). Not more than 4.0 (iodine monochloride Description. A white, unctuous mass or almost white flakes or
method). granules; odour, faint and characteristic.
Mineral acid. Shake 5 g ofthe melted substancewith an equal
volume of hot water for 2 minutes, cool and filter. To the Tests
filtrate add 0.05 ml of methyl orange solution; no red colour is
Appearance of solution. Dissolve 0.5 g in 20 ml of ethanol
produced.
(95 per cent) by heating to boiling and allow to cool. The
Heavy metals (2.3.13). 1.0 g complies with the limit test for solution is clear (2.4.1), and not more intensely coloured than
heavy metals, Method B (20 ppm). reference solution BS6 (2.4.1).
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined
Melting range (2.4.21). 55°to 60°.
on2.0 g.
Acid value (2.3.23). Not more than 2.0.
Assay. Determine by gas chromatography (2.4.13).
Test solution. Add 5 ml of boron trifluoride solution to 0.1 g Hydroxyl value (2.3.27). 195 to 220.
of the substance under examination and heat under a reflux Iodine value (2.3.28). Not more than 2.0 (iodine bromide
condenser for 15 minutes, cool and transfer to a separating method), determined on 2.0 g dissolved in 25 ml of chloroform,
funnel with the aid of 10 ml of hexane. Add 10 ml ofa saturated warming if necessary to effect solution.
solution of sodium chloride and 10 ml of water, shake, discard
the lower aqueous layer and dry the upper layer over Saponification value (2.3.37). Not more than 2.0, determined
anhydrous sodium sulphate. on 10.0 g.

Reference solution (a). Add 5 ml of a 1 per cent w/v solution Assay. Determine by gas chromatography (2.4.13).
of nonadecanoic acid (internal standard) in toluene to a
Test solution. Dissolve 0.1 g 'of the substance under
mixture of50 mg ofstearic acidRS and 50 mg ofpalmitic acid
examination in 10.0 ml.of ethanol (95 per cent).
RS and evaporate to dryness. Add 5 m1 of boron trifluoride
solution and complete the procedure described for the test Reference solution (a). Dissolve 50 mg of cetyl alcohol in
solution beginning at the words "heat under a reflux 10.0 ml of ethanol (95 per cent).
condenser...." ..
Reference solution (b). Dissolve 50 mg of stealyl alcohol RS
Reference solution (b). Add 5 ml of the internal standard in 5.0 ml of ethanol (95 per cent).
solution to 0.1 g ofthe substance under examination, evaporate
Reference solution (c). To 1.0 ml ofreference solution (a), add
to dryness, add 5 ml of boron trifluoride solution and complete
1.0 ml of reference solution (b) and dilute to 10.0 ml with .
the procedure described for the test solution beginning at the
ethanol (95 per cent).
words "heat under a reflux condenser......".
Reference solution (c). Prepare in the same manner as reference Chromatographic system
solution (a) but omitting the internal standard. a stainless steel column 30 m x 0.32 rom, packed with
poly(dimethyl)siloxane (lllm),
Chromatographic system
temperature:
a glass column 1.5 m x 4 rom, packed with acid-washed,
Time Temperature
silanised diatomaceous support (100 to 120 mesh) coated
(min) (0)
with 15 per cent w/w of diethylene glycol succinate
polyester, Column 0-20 150-250
temperature: column 170°, 20-40 250
inlet port and detector at 240 0 , injector port and detector at 250 0 ,
- flow rate. 30 ml per minute ofthe carrier gas. flame ionization detector,
Calculate the content OfClSH360z, and C 16H 32 0 Z• flow rate. 1 ml per minute using nitrogen as the carrier
gas.
Storage. Store protected from moisture.
Inject reference solution (c). The test is not valid unless the
resolution between the peaks due to cetyl alcohol and stearyl
alcohol is not less than 5.0.
Stearyl Alcohol
Inject 1 III of the test solution, reference solution (b) and (c).
StearylAlcohol is a mixture ofsolid alcohols consisting chiefly
of l-octadecanol, (C 1sH380). Calculate the content ofC 1s H38 0.
Category. Pharmaceutical aid (stiffening agent). Storage. Store protected from moisture.

2155

www.webofpharma.com
STILBOESTROL IP 2010

Stilboestrol Mobile phase. A mixture of 90 volumes of toluene and


10 volumes of diethylamine.
Diethylstilboestrol
Test solution. Dissolve 0.5 g of the substance under
examination in 100 ml of ethanol (95 per cent).
OH
Reference solution (a). A 0.5 per cent w/v solution of
stilboestrol RS in ethanol (95 per cent).
Reference solution (b). A 0.05 per cent w/v solution of
HO stilboestrol monomethyl ether RS in ethanol (95 per cent).

Mol. Wt. 268.4 Reference solution (c). A 0.05 per cent w/v solution of
stilboestrol dimethyl ether RS in ethanol (95 per cent).
Stilboestrol is (E)-a,~-diethylstilbene-4,4'-diol.
Reference solution (d). A solution containing 0.25 per cent
Stilboestrol contains not less than 97.0 per cent and not more w/v each of dienoestrol RS and stilboestrol RS
than 101.0 per cent ofClsHzoOz, calculated on the dried basis.
Apply to the plate 1 III of each solution. After development,
Category. Oestrogen. dry the plate in air, spray with ethanolic sulphuric acid
Dose. In the treatment of menopausal symptoms, 100 Ilg to (20 per cent) and heat at 120° for 10 minutes. Any secondary
I mg daily; for the suppression oflactation, 5 mg thrice daily spots in the chromatogram obtained with the test solution
for 3 days, followed by 5 mg daily for 6 days; in the treatment corresponding to the mono-and di-methyl ethers of
ofcarcinoma ofthe prostate and mammary carcinoma, 10 to 20 stilboestrol are not more intense than the spots in the
mgdaily. chromatograms obtained with reference solutions (b) and (c)
respectively. Stilboestrol sometimes produces two spots. The
Description. A white or almost white, crystalline powder;
test is not valid unless the chromatogram obtained with
odourless.
reference solution (d) shows two clearly separated spots of
approximately the same intensity.
Identification
Sulphated ash (2.3.18). Not more than 0.1 percent.
Test A may be omitted iftests B, C andD are carried out. Tests
Band C may be omitted if tests A and D are carried out. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105°.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with stilboestrol RS Assay. Weigh accurately about 20 mg, dissolve in sufficient
or with the reference spectrum of stilboestrol. ethanol to produce 100.0 ml and dilute 10.0 ml ofthis solution
to 100.0 ml with ethanol. To 25.0 ml of the resulting solution
B. When examined in the range 230 nm to 450 nm (2.4.7), the add 25.0 ml of a solution prepared by dissolving 1 g of
irradiated solution prepared as directed in the Assay shows dipotassium hydrogen phosphate in 55 ml of water, transfer a
absorption maxima at about 292 nm and 418 nm. portion of the mixture to a l-cm closed quartz cell, place the
C. In the test for Mono- and di-methyl ethers, the principal cell 10 cm from a 15-watt short-wave, ultraviolet light ofmercury
spot in the chromatogram obtained with the test solution lamp and irradiate for 10 minutes. Measure the absorbance of
corresponds to that in the chromatogram obtained with the irradiated solution at the maximum at about 418 nm (2.4.7).
reference solution (a).
Calculate the content of ClsHzoOz from the absorbance
D. Dissolve 0.5 mg in 0.2 ml ofglacial acetic acid, add I mlof obtained by repeating the operation using stilboestrol RS in
phosphoric acid and heat in a water-bath for 3 minutes; a place of the substance under examination.
deep yellow colour is produced which almost disappears on
Storage. Store protected from light and moisture.
dilution with 3 ml of glacial acetic acid (distinction from
dienoestrol).

Tests
Stilboestrol Tablets
4,4'-Dihydroxystilben and related ethers. Absorbance of a
1.0 per cent w/v solution in ethanol at about 325 run, not more Diethylstilboestrol Tablets
than 0.50 (2.4.7).
Stilboestrol Tablets contain not less than 90.0 per cent and
Mono- and di-methyl ethers. Determine by thin-layer not more than 110.0 per cent of the stated amount of
chromatography (2.4.1 7), coating the plate with silica gel. stilboestrol, ClsHzoOz.

2156

www.webofpharma.com
IP 2010 STREPTOKINASE

Usual strengths. 500 Ilg; 1 mg; 5 mg; 25 mg. Streptokinase


Identification Streptokinase is a preparation ofa protein obtained from culture
filtrat",s ofcertain strains of Streptococcus haemolyticus group
A. When examined in the range 230 nm to 450 nm (2.4.7), the C. It has the property of combining with human plasminogen
irradiated solution prepared as directed in the Assay shows to form plasminogen activator and is purified to contain not
absorption maxima at about 292 nm and 418 nm. less than 600 Units ofStreptokinase activity per Ilg ofnitrogen
before addition of any stabiliser or carrier. It usually contains
B. Extract a quantity ofthe powdered tablets containing 3 mg
a buffer and may be stabilised by the addition of suitable
of Stilboestrol with ether, filter and evaporate the filtrate to
substances such as Human Albumin.
dryness. Dissolve 0.5 mg of the residue in 0.2 ml of glacial
acetic acid, add 1 ml of phosphoric acid and heat in a water- Category. Fibrinolytic enzyme.
bath for 3 minutes; a deep yellow colour is produced which Dose. By intravenous infusion, 250,000 to 600,000 Units over
almost disappears on dilution with 3 ml of glacial acetic acid 30 minutes followed· by 100,000 Units every hour for upto 1
(distinction from dienoestrol). week.

Tests Description. A white powder or a. white, friable solid;


hygroscopic.
Uniformity of content. For tablets containing 10 mg or less
- Comply with the test stated under Tablets. Identification
Finely crush one tablet, add 10 ml of ethanol, shake for A. Place 0.5 ml ofcitrated human, canine or rabbit plasma in a
30 minutes, add sufficient ethanol to produce 25.0 ml and haemolysis tube maintained in a water-bath at 37°. Add 0.1 ml
centrifuge. Pipette an aliquot of the supernatant liquid of a solution of the substance under examination containing
containing 0.5 mg of Stilboestrol add 25.0 ml of a solution 10,000 Units per ml in citro-phosphate buffer pH 7.2 and
prepared by dissolving 1 g of dipotassium hydrogen 0.1 ml ofa solution of thrombin containing 20 Units per ml in
phosphate in 55 ml of water, transfer a portion ofthe mixture
°
to a l-cm closed quartz cell, place the cellI cm from a 15-watt
short-wave, ultraviolet light ofmercury lamp and irradiate for
citro-phosphate buffer pH 7.2 and shake immediately; a clot
forms and lyses within 30 minutes. Repeat the procedure using
citrated bovine plasma; lysis does not occur within 1 hour.
10 minutes. Measure the absorbance ofthe iITadiated solution
at the maximum at about 418 nm (2.4.7). B. Dissolve 0.6 g of agar in 50.0 ml of mixed barbitone buffer
pH 8.6, heating until a clear solution is obtained. Place glass
Calculate the content of ClsHzoOz in the tablet from the plates (50 mm x 50 mm) that are free from traces ofgrease on a
absorbance obtained by repeating the operation using level surface. Apply to each plate 4 ml ofthe agar solution and
stilboestrol RS in place of the substance under examination. allow to cool until set. Bore a hole 6 mm in diameter in the
centre of the agar and an appropriate number of holes (not
Other tests. Comply with the tests stated under Tablets.
exceeding six) at distances of 11 mm from the central hole
Assay. Weigh and powder 20 tablets. Weigh accurately a removing the residual agar by means of a cannula connected
quantity ofthe powder containing about 5 mg of Stilboestrol, to a vacuum pump. Place a quantity of 80 III of goat or rabbit
add 5 ml of ethanol, shake for 15 minutes, add sufficient anti streptokinase serum containing 10,000 Units of
ethanol to produce 100.0 ml and centrifuge. Dilute 20.0 ml of antistreptokinase activity per ml in the central hole and 80 III
the clear, supernatant liquid to 50.0 ml with ethanol and mix. of a solution of the substance under examination containing
To 25.0 ml ofthe resulting solution, add 25.0 ml of a solution 125,000 Units of streptokinase activity per ml of each of the
prepared by dissolving 1 g of dipotassium hydrogen sUITounding holes. Place the plates in a humidified tame for
phosphate in 55 ml of water, transfer a portion ofthe mixture 24 hours.
to a l-cm closed quartz cell, place the cellI 0 cm from a 15-watt
Only one precipitation arc is produced which is well-defined
short-wave, ultraviolet light ofmercury lamp and iITadiate for
and localised between the application point of the serum and
10 minutes. Measure the absorbance ofthe irradiated solution
each hole containing the solution of the substance under
at418nm(2.4.7).
examination.
Calculate the content of ClsHzoOz in the tablet from the
absorbance obtained by repeating the operation using Tests
stilboestrol RS in place of the substance under examination.
pH (2.4.24). 6.8 t07.5, determined on a solution prepared in
Storage. Store protected from light and moisture. freshly boiled and cooled water containing 5000 Units per m!.

2157

www.webofpharma.com
STREPTOKINASE IP 2010

Streptodornase. Introduce 0.5 ml ofa 0.1 per cent wIv solution Assay. The potency of streptokinase is determined by
of sodium deoxyribonucleate in imidazole buffer pH 6.5 into comparing its ability to activate human plasminogen to form
each of eight centrifuge tubes. To each of the first two tubes plasmin with that of the Standard Preparation. The plasmin
add 0.25 ml of imidazole bufferpH 6.5 and 0.25 ml ofsolution generated is detefll1ined by measurement of the time taken to
of the substance under examination in imidazole bz!ffer lyse a fibrin clot under the conditions of a suitable method of
pH 6.5 containing 150,000 Units per ml (solution A) followed· assay.
immediately by 3.0 ml of 0.25 M perchloric acid. Mix the
contents of each tube, centrifuge for 5 minutes at 3000 rpm Standard Preparation
and measure the absorbance ofeach ofthe supernatant liquids
at about 260 nm (2.4.7), using as the blank a mixture of 1.0 ml of The Standard Preparation is the 2nd International Standard
imidazole bufferpH 6.5 and 3.0 ml of 0.25 Mperchloric acid. for Streptokinase, established in 1989, consisting of freeze-
Calculate the sum ofthe two absorbances (AI)' To each of the dried streptokinase (supplied in ampoules containing
remaining six tubes add, respectively, 0.25, 0.25, 0.125, 0.125, 0 700 Units of streptokinase activity), or another suitable
and 0 ml of imidazole buffer pH 6.5 followed by 0.25 ml of preparation the activity of which has been detefll1ined in
solution A and finally 0, 0, 0.125, 0.125, 0.25 and 0.25 ml relation to the International reference preparation.
respectively of a solution of the Standard Preparation
containing 20 Units of streptodornase activity per ml in Method
imidazole buffer pH 6.5. [The Standard Preparation is the Use citro-phosphate buffer pH 7.2 containing 3 per cent w/v
1st International Standard Preparation for Streptodornase, of bovine serum albumin for the preparation of solutions and
established in 1964, consisting of a freeze-dried mixture of dilutions.
streptodornase and streptokinase with lactose (supplied in
ampoules containing 2400 Units of streptodornase activity), Prepare a solution of the Standard Preparation to contain
or another suitable preparation the activity of which has been 1000 Units of streptokinase activity per ml and prepare a
determined in relation to the International Standard]. Mix the solution of the preparation under examination expected to
contents of each tube, incubate at 37° for 15 minutes and add have the same concentration; keep the solutions in ice and
to each tube 3.0 ml of 0.25 M perchloric acid. Mix the contents use within 6 hours. Prepare three 1.5-fold serial dilutions of
of each tube, centrifuge and measure the absorbance of each the solution of the Standard Preparation so that the longest
ofthe supernatant liquids at about 260 nm (2.4.7), using as the' clot-lysis time is less than 20 minutes and prepare three similar
blank the mixture specified above. Ifthe sum ofthe absorbances dilutions ofthe solution ofthe preparation under examination.
of the liquids in the third and fourth tubes is A], that of the Keep the solutions in ice and use within 1 hour. Using
liquids in the fifth and six tubes is A 3 , and that ofthe liquids in 24 tubes, 8 mm in diameter, label the tubes S" S2, S3 for the
the seventh and eighth tubes is (A 4 ), (AI-A]) is less than dilutions of the Standard Preparation and T 1, Ti, T3 for the
0.5(A 3 +A 4)-A]. dilutions ofthe preparation under examination, allocating four
tubes to each dilution. Place the tubes in ice. Into each tube
Streptolysin. Dissolve a quantity of the substance under
introduce 0.2 ml of the appropriate dilution, 0.2 ml of citro-
examination containing 500,000 Units in 0.5 ml ofa mixture of
phosphate buffer pH 7.2 containing 3 per cent w/v of bovine
90 volumes of saline solution and 10 volumes of citro-
serum albumin and 0.1 ml ofa solution containing 20 Units of
phosphate bufferpH 7.2 in a haemolysis tube. Add 0.4 ml ofa
thrombin per ml. Place the tubes in a water-bath at 37° and
2.3 per cent w/v solution of sodium thioglycollate and
allow to stand for 2 minutes to attain temperature equilibrium.
incubate in a water-bath at 37° for 10 minutes. Add 0.1 mlofa
Using an automatic pipette, introduce into the bottom of the
solution ofa reference preparation of human antistreptolysin
first tube 0.5 ml of a 1 per cent w/v solution to human
o containing 5 Units per ml and incubate at 37° for 5 minutes. euglobulins ensuring mixing. At 5-second intervals introduce
Add 1 ml of rabbit elythrocyte suspension, continue the
successively into the remaining tubes 0.5 ml of a 1 per cent
incubation for 30 minutes and centrifuge at about 1000 rpm.
w/v solution of human euglobulins. Using a stop-watch,
The absorbance of the supernatant liquid at about 550 nm
measure for each tube the time in seconds that elapses between
(2.4.7), is not more than 1.5 times the absorbance obtained
the addition of the euglobulin and the lysis of the clot.
by repeating the above procedure using 0.5 ml of the mixture
of saline solution and citro-phosphate buffer pH 7.2 in Using the logarithms ofthe lysis times, calculate the result of
place of the solution containing the substance under the assay by standard statistical methods.
examination,
The estimated potency is not less than 90 per cent and not
Loss Oil dryillg (2.4.19). Not more thal14.0 per cent, deterrriiIled iiiore tha.n III per cent of the sta.ted potency. The fiducial
on 1.0 g by drying over phosphorus pentoxide at a pressure limits of error are not less than 80 per cent and not more than
not exceeding 2.7 kPa for 24 hours. 125 per cent of the stated potency.

2158

www.webofpharma.com
IP 2010 STREPTOKINASE INJECTION

Streptokinase intended for use in the manufacture ofparenteral Identification


preparations complies with the following additional
requirements. A. Place 0.5 ml ofcitrated human, canine or rabbit plasma in a
haemolysis tube maintained in a water-bath at 37°. Add 0.1 ml
Abnormal toxicity (2.2.1). Determine by Method A, using a of a solution of the contents of the container containing
solution containing 50,000 Units in 0.5 ml of water for 10,000 Units per ml in citro-phosphate buffer pH 7.2 and
injections administered in 15 to 20 seconds. 0.1 ml ofa solution of thrombin containing 20 Units per ml in
Bacterial endotoxins (2.2.3). Dissolve the contents of the citro-phosphate buffer pH 7.2 and shake immediately; a clot
sealed container in water BET to give a solution containing forms and lyses within 30 minutes. Repeat the procedure using
10,000 Units ofStreptokinase per ml. Carry out the test on the citrated bovine plasma; lysis does not occur within 1 hour.
resulting solution; the maximum allowable endotoxin B. Dissolve 0.6 g of agar in 50.0 ml of mixed barbitone buffer
concentration of the solution is 23.33 Units of endotoxin per pH 8.6, heating until a clear solution is obtained. Place glass
ml. Carry out the test using the maximum valid dilution ofthe plates (50 mm x 50 mm) that are free from traces ofgrease ona
prepared solution calculated from the declared sensitively of level surface. Apply to each plate 4 ml ofthe agar solution and
the lysate used in the test. allow to cool until set. Bore a hole 6 mm in diameter in the
Sterility (2.2.11). Complies with the test for sterility. centre of the agar and an appropriate number of holes
(not exceeding six) at distances of 11 mm from the central hole
Storage. Store in sealed containers, protected from light. The removing the residual agar by means of a cannula connected
containers should be sterile; tamper-evident and sealed so as to a vacuum pump. Place a quantity of 80 III of goat or rabbit
to exclude micro-organisms. Under these conditions the anti streptokinase serum containing 10,000 Units of
contents may be expected to retain their potency for 2 years. antistreptokinase activity per ml in the central hole and 80 III
Labelling. The label states (1) the number of Units of of a solution of the contents of the container containing
streptokinase activity in the container; (2) the number ofUnits 125,000 Units of streptokinase activity per ml of each ofthe
of streptokinase activity per mg, calculated with reference to surrounding holes. Place the-plates in a humidified tanle for
the dried preparation; (3) the name and quantity of any 24 hours.
added substances; (4) the storage conditions; (5) whether or Only one precipitation arc is produced which is well-defined
not it is intended for use in the manufacture of parenteral and localised between the application point of the serum and
preparations. each hole containing the solution of the substance under
examination.

Tests
Streptokinase Injection pH (2.4.24). 6.8 to 7.5, detennined on a freshly constituted
Streptokinase Injection is a sterile material consisting of injection containing 5000 Units per ml.
Streptokinase with or without auxiliary agents. It is filled in a
Streptodornase. Introduce 0.5 ml ofa 0.1 per centw/v solution
sealed container. '
of sodium deoxyribonucleate in imidazole buffer pH 6.5 into
The injection is constituted by dissolving the contents of the each of eight centrifuge tubes. To each of the first two tubes
sealed container in the requisite amount of sterile Water for add 0.25 ml of imidazole bufferpH 6.5 and 0.25 ml ofsolution
Injections, immediately before use. of the contents of the container with the substance under
examination in imidazole buffer pH 6.5 containing
The constituted solution complies with the requirements for
150,000 Units per ml (solution A) followed immediately by
Clarity of solution and Particulate matter stated under
3.0 ml of 0.25 M perchloric acid. Mix the contents of each
Parenteral Preparations (Injections).
tube, centrifuge for 5 minutes at 3000 rpm and measure the
Storage. The constituted solution should be used immediately absorbance of each of the supernatant liquids at about
after preparation but, in any case, within the period 260 mn (2.4.7), using as the blank a mixture of 1.0 ml ofimidazole
recommended by the manufacturer. bufferpH 6.5 and 3.0 ml of 0.25 M perchloric acid. Calculate
the sum ofthe two absorbances (AI)' To each ofthe remaining
The contents of the sealed container comply with the
six tubes add, respectively, 0.25, 0.25, 0.125, 0.125, 0 and 0 ml
requirements stated under Parenteral Preparations (Powders
of imidazole btiffer pH 6.5 followed by 0.25 ml of solution A
for Injection) and with the following requirements.
and finally 0, 0, 0.125, 0.125, 0.25 and 0.25 ml respectively ofa
Usual strengths. 50,000 Units; 100,000 Units; 750,000 Units; solution of the Standard Preparation containing 20 Units of
1,500,000 Units. streptodornase activity per ml in imidazole btiffer pH 6.5.

2159

www.webofpharma.com
STREPTOKINASE INJECTION IP 2010

[The Standard Preparation is the 1st International Standard Standard Preparation


Preparation for Streptodornase, established in 1964, consisting
The Standard Preparation is the 2nd' International. Standard
of a freeze-dried mixture of streptodornase and streptokinase
for Streptokinase, established in 1989, consisting of freeze-
with lactose (supplied in ampoules containing 2400 Units of
dried streptokinase (supplied in ampoules containing
streptodornase activity), or another suitable preparation the
700 Units of streptokinase activity), or another suitable
activity of which has been determined in relation to the
preparation the activity of which has been determined in
International Standard]. Mix the contents of each tube,
relation to the International reference preparation.
incubate at 37° for 15 minutes and add to each tube 3.0 ml of
0.25 M perchloric acid. Mix the contents of each tube, Method
centrifuge and measure the absorbance of each of the
supernatant liquids at about 260 nm (2.4.7), using as the blank Use citro-phosphate buffer pH 7.2 containing 3 per cent w/v
the mixture specified above. Ifthe sum of the absorbances of of bovine serum albumin for the preparation of solutions and
the liquids in the third and fourth tubes is A 2 , that of the dilutions.
liquids in the fifth and six tubes is A 3 , and that ofthe liquids in Prepare a solution of the Standard Preparation to contain
the seventh and eighth tubes is (A 4), (A I-A 2) is less than 1000 Units of streptokinase activity per ml and prepare a
0.5(A 3 +A,J-A 2 • solution ofthe contents ofthe container expected to have the
same concentration; keep the solutions in ice and use within
Streptolysin. Dissolve a quantity of the contents of the 6 hours. Prepare three 1.5-fold serial dilutions ofthe solution
container containing 500,000 Units in 0.5 ml of a mixture of of the Standard Preparation so that the longest clot-lysis time
90 volumes of sali;1e solution and 10 volumes of citro- is less than 20 minutes and prepare three similar dilutions of
phosphate bufferpH 7.2 in a haemolysis tube. Add 0.4 ml ofa the solution of the preparation under examination. Keep the
2.3 per cent w/v solution of sodium thioglycollate and solutions in ice and use within 1 hour. Using 24 tubes, 8 mm in
incubate in a water-bath at 37° for 10 minutes. Add 0.1 mlofa diameter, label the tubes S], S2, S3 for the dilutions of the
solution of a reference preparation of human antistreptolysin Standard Preparation and T], T2 , T3 for the dilutions of the
o containing 5 Units per ml and incubate at 37° for 5 minutes. preparation under examination, allocating four tubes to each
Add 1 ml of rabbit elythrocyte suspension, continue the dilution. Place the tubes inice. Into each tube introduce 0.2 ml
incubation for 30 minutes and centrifuge at about 1000 rpm. of the appropriate dilution, 0.2 ml of citro-phosphate buffer
The absorbance of the supernatant liquid at about 550 nm pH 7.2 containing 3 per cent w/v of bovine serum albumin
(2.4.7), is not more than 1.5 times the absorbance obtained and 0.1 ml of a solution containing 20 Units of thrombin per
by repeating the above procedure using 0.5 ml ofthe mixture ml. Place the tubes in a water-bath at 37° and allow to stand for
of saline solution and citro-phosphate buffer pH 7.2 in 2 minutes to attain temperature equilibrium. Using an automatic
place of the solution containing the substance under pipette, introduce into the bottom of the first tube 0.5 ml of a
examination. 1 per cent w/v solution to human euglobulins ensuring mixing.
At 5-second intervals introduce successively into the
Bacterial endotoxins (2.2.3). Dissolve the contents of the
remaining tubes 0.5 ml of a 1 per cent w/v solution of human
sealed container in water BET to give a solution containing
euglobulins. Using a stop-watch, measure for each tube the
10,000 Units ofStreptokinase per ml. Can)' out the test on the
time in seconds that elapses between the addition of the
resulting solution; the maximum allowable endotoxin
euglobulin and the lysis of the clot.
concentration of the solution is 23.33 Units of endotoxin per
ml. Carry out the test using the maximum valid dilution ofthe Using the logarithms of the lysis times, calculate the result of
prepared solution calculated from the declared sensitively of the assay by standard statistical methods.
the lysate used in the test.
The estimated potency is not less than 90 per cent and not
Other tests. Complies with the tests stated under Parenteral more than 111 per cent of the stated potency. The fiducial
Preparations (Injections). limits of error are not less than 80 per cent and not more than
125 per cent of the stated potency.
Assay. Determine on the mixed .contents often containers. Storage. Store in sealed containers, protected from light in a
refrigerator (2° to 8°). The containers should be sterile and
The potency of streptokinase is determined by comparing
sealed so as to exclude micro-organisms. Under these
its ability. to activate human plasiUinogen to fOrm plaslnin
co~ditions -the contents· may -be expected to retain their
with that of the Standard Preparation. The plasmin
potency for 2 years.
generated is determined by measurement ()fthe time taken to
lyse a fibrin clot under the conditions of a suitable method of Labelling. The label states the total number of Units of
assay. streptokinase activity contained in it.

2160

www.webofpharma.com
IP 2010 STREPTOMYCIN SULPHATE

Streptomycin Sulphate Test solution. Dissolve 0.1 g of the substance under


examination in 100 ml of water.
Reference solution (a). A 0.1 per cent w/v of streptomycin
sulphate RS in water.
Reference solution (b). A solution containing 0.1 per cent
w/v of streptomycin sulphate RS, 0.1 per cent w/v of neomycin
sulphate RS and 0.1 per cent w/v of kanamycin monosulphate
RSin water.
Apply to the plate 10 fll of each solution. Allow the mobile
phase to rise 12 cm. Dry the plate in warm air, spray with a
mixture of equal volumes of a 0.2 per cent w/v solution of
naphthalene-l,3-diol in ethanol (95 per cent) and sulphuric
acid (45 per cent) and heat at 150° for 5 to 10 minutes. The
principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
with reference solution (a). The test is not valid unless the
chromatogram obtained with reference solution (b) shows
three clearly separated spots.
2
B. Dissolve 5 to 10 mg in 4 ml of water and add 1 ml of
1 M sodium hydroxide. Heat in a water-bath for 4 minutes. Add
a slight excess of2 Mhydrochloric acid and 0.1 ml ofa 10 per
(CzIH39N7012)z,3HzS04 Mol. Wt. 1457.4
cent w/v solution ofjerric chloride; a violet colour is produced.
Streptomycin Sulphate is the sulphate of 0-2-deoxy-
C. Dissolve 0.1 g in 2 ml of water and add 1 ml of dilute
2-methylamino-a-L-glucopyranosyl-( 1-72)-O-5-deoxy-
I-naphthol solution and 2 ml ofa mixture ofequal volumes of
3-C-formyl-a-L-lyxofuranosyl-(I-4)-NJ,N3-diamidino-
dilute sodium hypochlorite solution and water; a red colour
D-streptamine, a substance produced by the growth of
is produced.
certain strains of Streptomyces griseus or obtained by any
other means. D. Dissolve 10 mg in 5 ml ofwater and add 1 ml of1 Mhydro-
chloric acid. Heat in a water-bath for 2 minutes. Add
Streptomycin Sulphate has a potency equivalent to not less
2 ml ofa 0.5 percentw/v solution of I-naphthol in 1 Msodium
than 700 flg and not more than 850 flg ofstreptomycin per mg.
hydroxide and heat in a water-bath for 1 minute; a faint yellow
It contains not less than 90.0 per cent of the stated amount of
colour is produced.
streptomycin, C21H39N7012, calculated on the dried basis.
E. Gives the reactions of sulphates (2.3.1).
Category. Antitubercular.
Dose. By intramuscular injection, the equivalent of 500 mg to Tests
1 g of streptomycin daily, or at longer intervals.
Appearance ofsolution. A25.0 per cent w/v solution in carbon
Description. A white or almost white powder; odourless or dioxide-ji'ee water is not more intensely coloured than degree
with slight odour; hygroscopic. 3 of the appropriate range of reference solutions (2.4.1). The
solution, after standing protected from light at a temperature
Identification of about 20° for 24 hours, is not more opalescent than
A. Determine by thin-layer chromatography (2.4.17). Prepare opalescence standard OS2 (2.4.1).
the plate by mixing 0.3 g of carbomer with 240 ml of water, pH (2.4.24). 4.5 to 7.0, determined in a25.0percentw/v solution.
allowing to stand with moderate stirring for 1 hour, adjusting
Sulphates. 18.0 to 21.5 per cent, calculated on the dried basis,
the pH to 7.0 by the gradual addition with constant shaking of
when determined by the following method. Dissolve 0.25 g in
2 M sodium hydroxide and adding 30 g of silica gel H. Spread
100 ml of water, adjust the pH to 11 with strong ammonia
a uniform layer of the resulting suspension 0.75 mm thick.
solution and add 10.0 ml of 0.1 Mbarium chloride and 0.5 mg
Heat the plate at 110° for 1 hour, allow to cool and use
of metalphthalein. Titrate the excess of barium chloride with
immediately.
0.1 M disodium edetate, adding 50 ml of ethanol (95 per
Mobile phase. A 7 percent w/v solution of potassium cent) when the colour of the solution begins to change and
dihydrogen phosphate. continuing the titration until the violet-blue colour disappears.

2161

www.webofpharma.com
STREPTOMYCIN SULPHATE IP 2010

1 ml of 0.1 M barium chloride is equivalent to 0.009606 g of Chromatographic system


sulphate, S04. - a glass column 1.5 to 2.0 m x 2 to 4 mm, packed with
ethylvinylbenzene-divinylbenzene copolymer (150 to
Colorimetric test. Dissolve 0.1 g in sufficient water to produce
180 Ilm) porous polymer beads (such as Porapak Q),
100 ml. To 5 ml, add 5 ml of 0.2 M sodium hydroxide and heat
- temperature: column 120° to 140°,
in a water-bath for exactly 10 minutes. Cool in ice for exactly
inlet port and detector at least 50° higher than that of
5 minutes, add 3 ml of a 1.5 per cent w/v solution of ferric
the column,
ammonium sulphate in 0.25 M sulphuric acid and sufficient
flow rate. 30 to 40 ml per minute ofthe carrier gas.
water to produce 25 ml and mix. Exactly 20 minutes after the
addition of the ferric ammonium sulphate solution, measure The area of any peak corresponding to methanol in the
the absorbance ofa 2-cm layer ofthe solution atthe maximum chromatogram obtained with the test solution is not greater
at about 525 nm (2.4.7), using as the blank a solution prepared than that of the peak in the chromatogram obtained with
in the same manner but omitting the substance under reference solution (0.3 per cent).
examination. The absorbance is not less than 90.0 per cent of Sulphated ash (2.3.18). Not more than 1.0 per cent.
that obtained by carrying out the procedure at the same time
and in the same manner using streptomycin sulphate RS in Loss on drying (2.4.19). Not more than 7.0 per cent, determined
place of the substance under examination, each absorbance on 1.0 g by drying over phosphorus pentoxide at 60° at a
being calculated on the dried basis. pressure not exceeding 0.1 kPa for 24 hours.
Assay. Determine by the microbiological assay ofantibiotics,
Streptomycin B. Determine by thin-layer chromatography
Method A or B (2.2.10), and express the results in Ilg of
(2.4.17), coating the plate with silica gel G
streptomycin per mg.
Mobile phase. A mixture of50 volumes of toluene, 25 volumes
Streptomycin Sulphate intended for use in the manufacture
of glacial acetic acid and 25 volumes of methanol.
ofparenteral preparations without a further appropriate
Test solution. Dissolve 0.2 g of the substance under procedurefor removal ofbacterial endotoxins complies with
examination in 5 ml ofa freshly prepared mixture of97 volumes the following additional requirement.
of methanol and 3 volumes of sulphuric acid, heat under a Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
reflux condenser for I hour, cool, wash down the condenser Unit per mg ofstreptomycin.
with methanol and add sufficient methanol to produce 20 m!.
Streptomycin Sulphate intended for use in the manufacture
Reference solution. Dissolve 36 mg of D-mannose in 5 ml ofa of parenteral preparations without a further appropriate
freshly prepared mixture of 97 volumes of methanol and sterilisation procedure complies with the following
3 volumes of sulphuric acid, heat under a reflux condenser additional requirement.
for 1 hour, cool, wash down the condenser with methanol and
add sufficient methanol to produce 50 m!. Dilute 5 rnl of the Sterility (2.2.11). Complies with the test for sterility.
resulting solution to 50 ml with methanol; this solution Storage. Store protected from light and moisture. If it is
contains the equivalent of0.03 per cent w/v ofstreptomycin B intended for use in the manufacture ofparenteral preparations
(1 mg of D-mannose is equivalent to 4.13 mg of strepto- the container should be sterile and sealed so as to exclude
mycinB). micro-organisms.
Apply to the plate 10 III of each solution. Allow the mobile Labelling. The label states (1) the equivalent weight of
phase to rise 13 to 15 cm. Dry the plate in air, and spray with a streptomycin contained in it; (2) whether or not the contents
freshly prepared mixture of equal volumes of a 0.2 per cent are intended for use in the manufacture of parenteral
w/v solution ofnaphthalene-1,3-diol in ethanol (95 per cent) preparations; (3) the name and quantity ofany added stabiliser.
and a 20 per cent v/v solution of sulphuric acid and heat at
110° for 5 minutes. Any spot in the chromatogram obtained
with the test solution is not more intense than the Streptomycin Injection
corresponding spot in the chromatogram with the reference
solution. Streptomycin Sulphate Injection

Methanol. Determine by gas chromatography (2.4.13). Streptomycin Injection is a sterile material consisting of
Streptomycin Sulphate with or without auxiliary agents. It is
Test solution. A 4 per cent w/v solution of the substance filredin-a-seah:~a-container~----'-- --- '-'"--'-"-'--'"--'-'._.__._._.,-
under examination in water.
The injection is constituted by dissolving the contents of the
Reference solution. A 0.012 per cent wlV solution of methanol sealed container in the requisite amount of sterile Water for
in water. Injections, immediately before use.

2162

www.webofpharma.com
IP 2010 STREPTOMYCIN TABLETS

The constituted solution complies with the requirements for B. Dissolve 5 to 10 mg in 4 ml ofwater and add 1 ml of1Msodium
Clarity of solution and Particulate matter stated under hydroxide, Heat in a water-bath for 4 minutes. Add a slight
Parenteral Preparations (Injections). excess of 2 M hydrochloric acid and 0.1 ml of a 10 per cent
w/v solution offerric chloride; a violet colour is produced.
Storage. The constituted solution'should be used immediately
after preparation but, in any case, within the period C. Dissolve 0.1 g in 2 ml of water and add 1 ml of dilute
recommended by the manufacturer. I-naphthol solution and 2 ml ofa mixture ofequal volumes of
dilute sodium hypochlorite solution and water; a red colour
Streptomycin Injection contains not less than 90.0 per cent
is produced.
and not more than 115.0 per cent of the stated amount of
streptomycin, CZIH39N70Iz. D. Dissolve 10mgin.5 mlofwaterandadd 1 mlof I Mhydro-
chloric acid. Heat in a water-bath for 2 minutes. Add
The contents of the sealed container comply with the 2 ml ofa 0.5 per cent w/v solution of I-naphthol in I M sodium
requirements stated under Parenteral Preparations hydroxide and heat in a water-bath for 1 minute; a faint yellow
(Powders for Injection) and with the following requirements. colour is produced.
Usual strengths. The equivalent of 750 mg and 1 g of Tests
streptomycin.
Description. A white or almost white powder which yields a pH (2.4.24). 4.5 to 7.0, determined in a25.0 per cent w/v solution.
clear, colourless or faintly yellow coloured solution when Loss on drying (2.4.19). Not more than 7.0 per cent, determined
dissolved in water. on 1.0 g by drying in an oven at 60° over phosphoruspentoxide
at a pressure not exceeding 0.1 kPa for 24 hours.
Identification Other tests. Complies with the tests stated under Parenteral
A, Determine by thin-layer chromatography (2.4.17). Prepare Preparations (Injections).
the plate by mixing 0.3 g of carbomer with 240 ml of water, Assay. Determine on the mixed contents often containers by
allowing to stand with moderate stirring for 1 hour, adjusting the microbiological assay ofantibiotics, MethodA or B (2.2.10).
the pH to 7.0 by the gradual addition with constant shaking of
Bacterial endotoxins (2.2.3): Not more than 0.25 Endotoxin
2 M sodium hydroxide and adding 30 g of silica gel H. Spread
Unit per mg ofstreptomycin.
a uniform layer of the resulting suspension 0.75 rom thiclc.
Heat the plate at 110° for 1 hour, allow to cool and use Storage. Store protected from moisture.
immediately. Labelling. The label states the strength in terms of the
Mobile phase. A 7 per cent w/v solution of potassium equivalent amount of streptomycin.
dihydrogen phosphate.
Test solution. Dissolve 0.1 g of the substance under
examination in 100 ml of water.
Streptomycin Tablets
Reference solution (a). A 0.1 per cent w/v of streptomycin
sulphate RS in water. Streptomycin Sulphate Tablets

Reference solution (b). A solution containing 0.1 per cent Streptomycin Tablets contain not less than 90.0 per cent and
w/v of streptomycin sulphate RS, 0.1 per cent w/v of neomycin not more than 110.0 per cent of the stated amount of
sulphate RS and 0.1 per cent w/v of kanamycin monosulphate streptomycin, CZIH39N7012.
RSin water. Usual strength. The equivalent of500 mg ofstreptomycin.
Apply to the plate 10 III of each solution. Allow the mobile Identification
phase to rise 12 cm. Dry the plate in warm air, spray with a
mixture of equal volumes of a 0.2 per cent w/v solution of A. Triturate a quantity of the powdered tablets containing
naphthalene-I,3-diol in ethanol (95 per cent) and sulphuric 0.2 g of streptomycin in a mixture of 2 ml of methanol and
acid (45 per cent) and heat at 150° for 5 to 10 minutes. The 0.1 ml of sulphuric acid, filter, ifnecessary, and allow to stand
principal spot in the chromatogram obtained with the test at about 25°; crystals of streptidine sulphate separate in the
solution corresponds to that in the chromatogram obtained course of 2 to 3 days. Dissolve the crystals in a solution of
with reference solution (a). The test is not valid unless the 0.1 g of picric acid in 10 m1 of hot water and cool; the
chromatogram obtained with reference solution (b) shows precipitate, after recrystallisation from hot water, melts at about
three clearly separated spots. 283° (2.4.21).

2163

www.webofpharma.com
SUCCINYLCHOLINE CHLORIDE IP 2010

B. Boil a small quantity of the powdered tablets containing B. Dissolve about 25 mg in 1 ml of water, add 0.1 ml ofal per
0.1 g ofstreptomycin with 5 ml of1 M sodium hydroxide for a cent w/v solution of cobalt chloride and 0.1 ml of potassium
few minutes, add a slight excess of 2 M hydrochloric acid ferrocyanide solution; a green colour is produced.
and 0.15 ml of a 10 per cent w/v solution offerric chloride; a
C. Dissolve 1 g in sufficient carbon dioxide-.free water to
brilliant violet colour is produced.
produce 20 ml. To 1 ml ofthis solution add 9 ml of water, 10 ml
Tests of 1 M sulphuric acid and 30 ml of ammonium reineckate
solution; a pink precipitate is produced. Allow to stand for
Other tests. Comply with the tests stated under Tablets. 30 minutes, filter and wash with water, then with ethanol
(95 per cent) and finally with ether. The residue, after drying
Assay. Weigh and powder 20 tablets. Weigh accurately a
at 80° melts at l800 to 185°(2.4.21).
quantity ofthe powder containing about 0.3 g ofstreptomycin
and, triturate with 20 ml of buffer solution pH 8.0. Dilute to D. Gives the reactions of chlorides (2.3.1).
100.0 ml with water.
Tests
Determine by the microbiological assay ofantibiotics, Method
A or B (2.2.10). Appearance of-solution. A 5.0 per cent w/v solution in carbon
dioxide-free water (solution A) is clear (2.4.1).4 ml ofsolution
Storage. Store protected from moisture.
A diluted to 10 ml with water is colourless (2.4.1).
Labelling. The label states the strength in terms of the
pH (2,4,24). 4.0 to 5.0, determined in a 0.5 per cent w/v solution.
equivalent amount of streptomycin.
Choline chloride. Determine by thin-layer chromatography
(2.4.17), coating the plate with microclystalline cellulose.
Mobile phase. A mixture of 50 volumes of I-butanol,
Succinylcholine Chloride 40 volumes of water and 10 volumes of anhydrous formic
Suxamethonium Chloride acid, shake for 10 minutes, allow to separate. Use the upper
layer as the mobile phase.
Test solution. Dissolve 0.4 g of the substance under
examination in 10 ml of methanol.
Reference solution. A solution containing 4 per cent w/v of
succinylcholine chloride RS and 0.02 per cent w/v of choline
chloride in methanol.
Mol. Wt. 397.3
Apply to the plate 5 III of each solution. After development,
Succinylcholine Chloride is 2,2'-succinyldioxy-
dry the plate in air, spray with potassium iodobismuthate
bis(ethyltrimethylammonium) dichloride dihydrate.
solution. Any secondary spot in the chromatogram obtained
Succinylcholine Chloride contains not less than 98.0 per cent with the test solution is not more intense than the spot due to
and not more than 101.0 per cent ofC14H30ChN204, calculated choline chloride in the chromatogram obtained with the
on the anhydrous basis. reference solution. The test is not valid unless the
chromatogram obtained with the reference solution shows
Category. Depolarising skeletal muscle relaxant.
two clearly separated spots.
Dose. By intravenous injection, 20 to 100 mg, according to the
Sulphated ash (2.3.18). Not more than 0.1 per cent.
needs of the patient.
Water (2.3.43). 8.0 to 10.0 percent, determined on 0.3 g.
Description. A white or almost white, crystalline powder;
almost odourless; hygroscopic. Assay. Weigh accurately about 0.3 g, dissolve in 30 ml of
anhydrous glacial acetic acid, add 30 ml of acetic anhydride
Identification and 10 ml of mercuric acetate solution. Titrate with
0.1 M perchloric acid, using crystal violet solution as
Test A may be omitted iftests B, C andD are carried out. Tests
indicator, until a bluish green colour is produced. Carry out a
Band C may be omittediftests A and D are carried out.
blank titration.
A. Determine by infrared absorption spectrophotometry (2.4.6).
1 ml of 0.1 Mperchloric acid is equivalent to 0.01807 g of
Compare the spectrum with that obtained with succinylcholine
C14H30ChN204.
chloride RS or with the reference spectrum ofsuccinylcholine
chloride. Storage. Store protected from light and moisture.

2164

www.webofpharma.com
IP 2010 SUCRALOSE

Succinylcholine Injection Sucralose


Suxamethonium Chloride Injection

~
Succinylcholine Injection is a sterile solution of
Succinylcholine Chloride in Water for Injections. O: CI
OHo~CI
Succinylcholine Injection contains not less than 90.0 per cent HO 0
and not more than 107.5 per cent of the stated amount of
succinylcholine chloride, C,4H30CIzN204,2H20.
Usual strength. 50 mg per m!. OH

Identification Mol. Wt. 397.6

Dilute a volume containing 20 mg ofSuccinylcholine Chloride Sucralose is 1,6-dichloro-l ,6-dideoxy-~-o-fructofuranosyl-4­


to 50 ml with water. To 0.5 ml add 2 ml of chloroform, 2 ml ofa chloro-4-deoxy-0'.-o-galactopyranoside.
solution containing 0.16 per cent w/v of citric acid and 6.6 per Sucralose contains not less than 98.0 per cent and not more
cent w/v of disodium hydrogen phosphate and 0.1 ml of a than 102.0 per cent ofC12H19ChOs, calculated on the anhydrous
solution containing 0.15 per cent w/v ofeach of bromothymol basis.
blue and anhydrous sodium carbonate. Shake for 2 minutes
and allow to separate. The chloroform layer is yellow. Category. Pharmaceutical aid (sweetening vehicle).

Tests Identification

pH (2.4.28).3.0 to 5.0. A. Detennine by infrared absorption spectrophotometry (2.4.6).


Compare the spectrum with that obtained with sucralose RS
Hydrolysis products. The volume of 0.1 M sodium hydroxide or with the reference spectrum of sucralose.
required for the preliminary neutralisation in the Assay is not
B. In the Assay, the principal peak in the chromatogram
more than one tenth of the total volume of 0.1 M sodium
obtained with the test solution corresponds to the peak in the
hydroxide required for the preliminary neutralisation and the
chromatogram obtained with the reference solution.
hydrolysis.
Other tests. Complies with the tests stated under Parenteral Tests
Preparations· (Injections).
Specific optical rotation (2.4.22). +84.0° to +87.5°, determined'
Assay. To an accurately measured volume containing about at 20° in a 1.0 per cent w/v solution in wate!:
0.25 g ofSuccinylcholine Chloride add 30 ml of carbon dioxide-
fi-ee water and shake with five quantities, each of 25 ml, of Related substances. Determine by thin-layer chromatography
ether. Wash the combined ether solutions with two quantities, (2.4.17), coating the plate with silica gel.
each of 10 ml, of water and discard the ether. Shake the Mobile phase. A mixture of 70 volumes of 5 per cent w/v
combined washings with two quantities, each of 10 ml, of solution of sodium chloride and 30 volumes of acetonitrile.
ether, add the washings to the original aqueous solution and
neutralise with 0.1 M sodium hydroxide using bromothymol Test solution. Dissolve about 1.0 g of the substance under
blue solution as indicator. Add 25.0 ml of 0.1 M sodium examination in 10 ml of methanol.
hydroxide, heat under a reflux condenser for 40 minutes, allow Reference solution (a). A 1.0 per cent w/v solution ofsucralose
to cool and titrate the excess ofallcali with 0.1 M hydrochloric RS in methanol.
acid using bromothymol blue solution as indicator. Repeat
Reference solution (b). Dilute 5.0 ml ofreference solution (a)
the operation using 40 rol of carbon dioxide-free water
to 100.0 ml with methanol.
beginning at the words "Add 25.0 ml of 0.1 M sodium
hydroxide .....". The difference between the titrations Apply to the plate 5 III of each solution. After development,
represents the amount of sodium hydroxide required. dry the plate in air and spray the plate with 15 per cent w/v
solution of sulphuric acid in methanol. Heat the plate at 125°
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01987 g of
for 10 minutes. The Rf value of the principal spot obtained
CI~30CIzN204,2H20.
with the test solution corresponds to the spot in the
Storage. Store protected from light, at a temperature between chromatogram obtained with reference solution (a). Any
2° to 8°. The injection should not be allowed to freeze. secondary spot in the chromatogram obtained with the test

2165

www.webofpharma.com
SUCRALOSE IP 2010

solution is not more intense than the spot in the chromatogram Sucrose
obtained with reference solution (b) (0.5 per cent).
Refilled Sugar
Hydrolysis products. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel.
HO
NOTE-This test does not require a developing solvent.
.)---0
Test solution. Dissolve about 2.5 g of the substance under
examination in 10 mtofmethanol. OH
Reference solution (a). A 10.0 per cent w/v solution of OH 0
mannitol in water. H°l.----O~
Reference solution (b). A solution containing 0.04 per cent
w/v offructose and 10 per cent w/v of mannitol in water. ~l
OH OH
Apply to the plate 5 J.ll of each solution. Spray with a solution
containing 1.23 g ofp-anisidine and 1.66 g ofphthalic acid in ClzHn011 Mol. Wt. 342.3
100 rn1 ofmethanol, heatthe plate at 105° for 15 minutes. Ifthe
Sucrose is ~-D-fructofuranosyl a-D-glucopyranoside.
spot from reference solution (a) has darkened, repeat the test,
heating for a shorter period oftime. Immediately after heating, Category. Pharmaceutical aid (sweetening agent; tablet
view the plate against a dark background: the color of the excipient).
spot obtained from the test solution is not more intense than Description. An almost white or colourless crystals, dry
that obtained from reference solution (b) (0.1 per cent). crystalline powder; odourless; taste, sweet.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method B (10 ppm). Identification

Sulphated ash (2.3.18). Not more than 0.7 per cent. Dissolve 150.0 g in sufficient carbon dioxide-free water
prepared from distilled water to produce 300 ml (solution A).
Water (2.3.43). Not more than 2.0 per cent, determined on Dilute 1 ml of solution A to 100 ml with water. To 5 ml of the
LOg. solution add 2 ml of freshly prepared 2 M sodium hydroxide
Assay. Determine by liquid chromatography (2.4.14). and 0.15 ml of a freshly prepared copper sulphate solution;
the solution is clear and blue and remains so on boiling. To
Test solution. Dissolve about 25 mg of the substance under the hot solution add 4 ml of 2 M hydrochloric acid, heat to
examination in 25 ml ofthe mobile phase. boiling and add 4 ml of 2 M sodium hydroxide; an orange
precipitate is produced immediately.
Reference solution. A 0.1 per cent w/v solution of sucralose
RS in the mobile phase. Tests
Chromatographic system
Acidity or alkalinity. To 10 ml of solution A add 0.3 ml of
a stainless steel column 10 cm x 8.0 mm, packed with
phenolphthalein solution. The solution is colourless and not
octadecylsilane bonded to porous silica (5 J.lm),
more than 0.6 m1 of 0.01 M sodium hydroxide is required to
mobile phase: a mixture of 17 volumes of water and 3
change the colour of the solution to pink.
volumes of acetonitrile,
flow rate. 1.5 ml per minute, Specific optical rotation (2.4.22). +65.9° to +67.0°, determined
refractive index detector in a 10 per cent w/v solution.
injection volume. 20 J.ll. Barium. To 10 ml ofsolution A add I ml of 1 M sulphuric acid.
Inject the reference solution. The test is not valid unless the When examined immediately and after 1 hour any opalescence
relative standard deviation for replicate injections is not more is not more intense than that of a mixture of 1 nil of distilled
than 2.0 percent. water and 10 ml of solution A.

test solution and the reference solution. Calcium. To 1 ml of solution A add 9 ml ofwater and 1 mlof
Cfrffrff6f11Uftfoxalate-S6Itttiol1;-tlre-so1utionremains clear-·for at
Calculate the cOntent ofClzH19ChOso least 1 minute.
Storage. Store protected from moisture, at a temperature not Heavy metals (2.3.13). Add 0.1 ml ofdilute hydrochloric acid
exceeding 21°. to 4 ml ofsolution A and dilute with sufficient water to produce

2166

www.webofpharma.com
IP 2010 SULPHACETAMIDE SODIUM

25 ml. The solution complies with the limit test for heavy metals, Sulphacetamide Sodium is the monohydrate ofthe sodium
Method A (10 ppm). salt ofNI-acetylsulphanilamide.
Sulphites. To 4 ml ofsolution A add sufficient water to produce Sulphacetamide Sodium contains not less than 99.0 per cent
20 ml, add 0.05 ml of 0.1 M iodine and 0.05 ml ofstarch solution; and not more than 101.0 per cent ofCsH9N2Na03S, calculated
a blue colour develops. on the anhydrous basis.
Dextrins. To 2 ml of solution A add 8 ml of water, 0.05 ml of Category. Antibacterial.
2 M hydrochloric acid and 0.05 ml of 0.05 M iodine; the
Description. A white or yellowish white, crystalline powder;
solution remains yellow or becomes faint bluish green.
odourless.
Glucose and invert sugar. Dissolve 20 g in sufficient water to
make 100 ml and filter if necessary. Place 50 ml of the clear Identification
solution in a 250-ml beaker, add 50 ml of alkaline cupric
Test A may be omitted iftests B, C, D, E and F are carried out.
tartarate solution, cover the beaker with a watch glass, heat
Tests B, C, D and E may be omitted iftests A andF are carried
the mixture at such a rate that it comes to a boil in approximately
out.
4 minutes and continue boiling for exactly 2 minutes. Add
immediately 100 ml of recently boiled and cooled water and A. Detennine by infrared absorption spectrophotometry (2.4.6).
collect the precipitated cuprous oxide on a tared sintered.- Compare the spectrum with that obtained with sulphacetamide
glass crucible. Wash the residue with the hot water, then with sodium RS or with the reference spectrum of sulphacetamide
10 ml of ethanol (95 per cent) and fmally with 10 ml of ether. sodium.
Dry at 105° for 1 hour; the weight of the cuprous oxide is not B. In the test for Related substances, the principal peak in the
more than 112 mg. chromatogram obtained with the test solution corresponds to
Colouring matter. A. To 100 mLof solution A in a ground- the principal peak in the chromatogram obtained with the
glass~stoppered tube add 1 ml of dilute hypophosphorous reference solution.
acid and allow to stand for 1 hour; no unpleasant odour is C. Dissolve 1 g in 10 ml of water, add 6 ml of2 M acetic acid
detectable. and filter. Wash the precipitate with a small volume of water
B. Examine solution A in ultraviolet light at 365 nm. Any and dry at 105° for 4 hours. The melting range ofthe precipitate
fluorescence is not more intense than that of a solution is 181°to 185° (2.4.21).
containing 0.4 Ilg per ml of quinine sulphate in 0.005 M D. Dissolve 0.1 g ofthe precipitate obtained in test C in 5 mlof
sulphuric acid. . ethanol (95 per cent), add 0.2 ml of sulphuric acid and heat;
Sulphated ash (2.3.18). Not more than 0.1 per cent detennined ethyl acetate, recognizable by its odour, is produced.
by dissolving 5.0 g in 5 ml of water, adding 2 ml of sulphuric E. Dissolve 1 mg ofthe precipitate obtained in test C in 5 mlof
acid, evaporating to dryness and igniting to constant weight. water with the aid of heat. The solution gives the reaction of
Sucrose intended for use in the manufacture ofparenteral primary aromatic amines (2.3.1), producing an orange-red
preparations without a fitrther appropriate procedure for precipitate.
the removal of bacterial endotoxins complies with the F. A 5 per cent w/v solution gives the reactions of sodium
following additional requirement. salts (2.3.1).
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
unit per mg of Sucrose. Tests
Storage. Store protected from light and moisture. Appearance ofsolution. A 5.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution GYS4 (2.4.1).
SuIphacetamide Sodium pH (2.4.28).8.0 to 9.5, detennined in a 5.0 per centw/v solution.
Related substances. Detennine by liquid chromatography
(2.4.14).
Note-Prepare the solutions immediately before use and
protected from light.
Test solution. Dissolve 0.2 g of the substance under
Mol. Wt. 254.2 examination in 10.0 ml ofthe mobile phase.

2167

www.webofpharma.com
SULPHACETAMIDE SODIUM IP 2010

Reference solution (a). Dissolve 5 mg each of sulphacetamide Sulphacetamide Eye Drops


sodium RS and sulphanilamide (sulphacetamide sodium
impurity A) in 1.0 m1 ofthe mobile phase. Sulphacetamide Sodium Eye Drops
Reference solution (b). Dilute 1.0 m1 of the test solution to Sulphacetamide Eye Drops are a sterile solution of
100.0 ml with the mobile phase. Further dilute 1.0 ml ofthis Su1phacetamide Sodium in Purified Water. It may contain a
solution to 10.0 ml with the mobile phase. suitable antimicrobial agent.

Chromatographic system Sulphacetamide Eye Drops contain not less than 95.0 per cent
- a stainless steel column 12.5 cm x 4 mm, packed with and not more than 105.0 per cent of the stated amount of
endcapped octadecylsilane bonded to porous silica (5 sulphacetamide sodium, CSH9NzNa03S,HzO.
Jill1) Usual strengths. 10 per cent w/v; 20 per cent w/v; 30 per cent
- mobile phase: a mixture of I volumes of glacial acetic w/v.
acid, 10 volumes of methanol and 89 volumes of water,
- flow rate. 0.8m1perminute, Identification
- spectrophotometer set at 254 nm, To a volume containing 0.5 g of Suiphacetamide Sodium add
- injection volume. 10 fll. 6 m1 of 5 M acetic acid, stirring constantly. Filter the precipitate,
Inject reference solution (a). The test is not valid unless the wash with water and dry at 105° for 4 hours. The residue
resolution between the peaks due to sulphacetamide impurity complies with the following tests.
A and sulphacetamide is not less than 5.0. The relative retention A. Determine by infrared absorption spectrophotometry (2.4.6).
time with reference to sulphacetamide for sulphacetamide Compare the spectrum with that obtained with sulphacetamide
impurity A is about 0.5. RS or with the reference spectrum of sulphacetamide.
Inject the test solution and reference solution (b). Run the B. In the test for Related substances, the principal peak in the
chromatogram 7 times the retention time ofthe principal peak. chromatogram obtained with the test solution corresponds to
In the chromatogram obtained with the test solution the area the principal peak in the chromatogram obtained with the
ofthe peak due to sulphacetamide impurity A is not more than reference solution.
twice the area of the principal peak in the chromatogram C. Dissolve 10 mg in 2 ml of 2 M hydrochloric acid. The
obtained with reference solution (b) (0.2 per cent), the area of solution gives the reaction ofprimary aromatic amines (2.3.1).
any secondary peak is not more than the area of the principal
peak in the chromatogram obtained with reference solution Tests
(b) (0.1 per cent). The sum of all the secondary peaks is not Appearance of solution. Dilute the eye drops, ifnecessary, to
more than 5 times the area of the principal peak in the contain 10.0 per cent w/v of Sulphacetamide Sodium. The
chromatogram obtained with reference solution (b) (0.5 per solution is not more intensely coloured than reference solution
cent). Ignore any peak with an area less than 0.5 times the area BYS4(2.4.1).
of the principal peak in the chromatogram obtained with
pH (2.4.24).6.6 to 8.6.
reference solution (b) (0.05 per cent).
Related substances. Determine by liquid chromatography
Heavy metals (2.3.13). 1.0 g complies with the limit test for (2.4.14).
heavy metals, Method B (20 ppm).
Note-Prepare the solutions immediately before use and
Sulphates (2.3 .17). Dissolve 1.5 g in sufficient distilled water protected from light.
to produce 25ml, add 25 ml of 2 M acetic acid, shake for Test solution. Dissolve a volume of solution containing about
30 minutes and filter. 25 m1 ofthe filtrate complies with the limit 0.2 g ofSuiphacetamide Sodium in 10.0 ml ofthe mobile phase.
test for sulphates (200 ppm).
Reference solution (a). Dissolve 5 mg each of sulphacetamide
Water (2.3.43).6.0 to 8.0 per cent, determined on 0.2 g. sodium RS and sulphanilamide (sulphacetamide sodium
impurity A) in 1.0 ml ofthe mobile phase.
Assay. Weigh accurately about 0.25 g, dissolve in a mixture of
50 ml of water and 20 ml of2 M hydrochloric acid, add 3 g of Reference solution (b). Dilute 1.0 ml of the test solution to .
potassium bromide, cool in ice and carry out the nitrite.titration 100.0 ml with the mobile phase. Further dilute 1.0 m1 of this
(4,JJl). solution to 10.0 ml with the mobile phase.
Chromatographic system
1 ml of 0.1 M sodium nitrite is equivalent to 0:02362 g of
- a stainless steel column 12.5 cm x 4 rom, packed with
CSH9NzNa03S,
endcapped octadecylsilane bonded to porous silica
Storage. Store protected from light and moisture. (5 flm)

2168

www.webofpharma.com
IP 2010 SULPHADIAZINE

- mobile phase: a mixture of I volumes of glacial acetic Sulphadiazine contains not less than 99.0 per cent and not
acid, 10 volumes ofmethanol and 89 volumes ofwater, more than 101.0 per cent of C IO H ION 40 2S, calculated on the
- flow rate. 0.8 ml per minute, dried basis.
- spectrophotometer set at 254 nm,
Category. Antibacterial.
- injection volume. 10 Ill.
Dose. Initial dose, 3 g; subsequent doses, upto 4 g daily, in
Inject reference solution (a). The test is not valid unless the
divided doses..
resolution between the peaks due to sulphacetamide impurity
A and sulphacetamide is not less than 5.0. The relative retention Description. White, yellowish white or pinkish white crystals
time with reference to sulphacetamide for sulphacetamide or crystalline powder; almost odourless.
impurity Ais about 0.5.
Inject the test solution and reference solution (b). Run the Identification
chromatogram 7 times the retention time ofthe principal peak.
Test A may be omitted iftests B, C andD are carried out. Tests
In the chromatogram obtained with the test solution the area
C and D may be omitted if tests A and B are carried out.
ofthe peak due to sulphacetamide impurity A is not more than
twice the area of the principal peak in the chromatogram A. Determine by infrared absorption spectrophotometry (2.4.6).
obtained with reference solution (b) (0.2 per cent), the area of Compare the spectrum with that obtained with sulphadiazine
any secondary peak is not more than the area ofthe principal RS or with the reference spectrum of sulphadiazine.
peak in the chromatogram obtained with reference solution B. In the test for Related substances, the principal spot in the
(b) (0.1 per cent). The sum of all the secondary peaks is not chromatogram obtained with test solution (b) corresponds to
more than 5 times the area of the principal peak in the that in the chromatogram obtained with the reference solution.
chromatogram obtained with reference solution (b) (0.5 per
cent). Ignore any peak with an area less than 0.5 times the area C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
of the principal peak in the chromatogram obtained with dilute 1 ml of this solution to 10 ml with water. The solution,
reference solution (b) (0.05 per cent). without further acidification, gives the reaction of primary
aromatic amines (2.3.1).
Other tests. Comply with the tests stated under Eye Drops.
D. Heat 3 g in a test-tube inclined at an angle of 45° with the
Assay. To an accurately measured volume containing about
lower part immersed in a silicone oil-bath at about 270°. It
0.5 g ofSuiphacetamide Sodium add 75 ml ofwater and 10 ml
decomposes and a white or yellowish white sublimate is
of hydrochloric acid. Add 3 g ofpotassium bromide, cool in
produced. The sublimate, after recrystallisation from·toluene
ice and carry out the nitrite titration (2.3.31).
and drying at 100° melts at 123° to 127°(2.4.21).
1 ml of 0.1 M sodium nitrite is equivalent to 0.02542 g of
CgH9N2Na03S,H20. Tests
Storage. Store protected from light and moisture. The Eye
Drops should not be allowed to freeze. Appearance of solution. Dissolve 0.8 gin 10 ml of1 M sodium
hydroxide. The solution is not more intensely coloured than
Labelling. The label states (1) the name and concentration of reference solution YS5, BYS5 or GYS5 (2.4.1).
any antimicrobial agent used; (2) that it is not meant for
injection; (3) that the solution should be used within one Acidity. Heat 1.25 g ofthe finely powdered substance at about
month of opening the container; (4) that the solution should 70° with 25 ml of carbon dioxide-free water for 5 minutes.
not be used ifit is dark brown in colour; (5) that it should not Cool for about 15 minutes in ice and filter. To 20 ml of the
be allowed to freeze. filtrate add 0.1 ml of bromothymol blue solution. Not more
than 0.2 ml of 0.1 M sodium hydroxide is required to change
the colour of the solution.
Sulphadiazine Related substances (2.3.7). Complies with test C.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
q~ IIo N:)
I I heavy metals, Method B (20 ppm).
r(YS'N~N Sulphated ash (2.3.18). Not more than 0.1 per cent.

H2 N
M H Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105°.
C IOH ION40 2S Mol. Wt. 250.3 Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
Sulphadiazine is N -(pyrimidin-2-yl)sulphanilamide. 20 ml of2 M hydrochloric acid and 50 ml ofwater. Add 3 gof

2169

www.webofpharma.com
SULPHADIAZINE TABLETS IP 2010

potassium bromide, cool in ice and carry out the nitrite titration Medium. 900 ml of 0.1 M hydrochloric acid,
(2.3.31). Speed and time. 100 rpm and 60 minutes.
1 ml of 0.1 M sodium nitrite is equivalent to 0.02503 g of Withdraw a suitable volume ofthe sample and filter promptly
C IOH ION 40 ZS. through a membrane filter disc with an average pore diameter
Storage. Store protected from light and moisture. not greater than 1.0 mIll. Rej ect the first few ml ofthe filtrate
and dilute a suitable volume of the filtrate with the same
solvent. Dilute suitably with 0,01 M sodium hydroxide.
Measure the absorbances of the resulting solution and of a
Sulphadiazine Tablets standard solution of sulphadiazine RS ofsimilar concentration
in the same medium atthe maximum at about 254 urn (2.4.7).
Sulphadiazine Tablets contain not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of Calculate the content ofC IOH lON 40 ZS in the medium.
sulphadiazine, C IOH ION 40 ZS. D. Not less than 70.0 per cent of the stated amount of
Usual strength. 500 mg. C IOH ION 40 ZS.
Other tests. Comply with the tests stated under Tablets.
Identification
Assay. Weigh and powder 20 tablets. Weigh accurately a
A. Triturate a quantity of the powdered tablets containing quantity ofthe powder containing about 0.5 g ofSulphadiazine
0.5 g Sulphadiazine with two successive quantities, each of and dissolve as completely as possible in amixture of50 ml of
5 ml, of chloroform and reject the chloroform. Triturate the water and 10 ml of hydrochloric acid. Carry out the nitrite
residue with 10 ml of dilute ammonia solution for 5 minutes, titration (2.3.31).
add 10 ml of water and filter. Warm the filtrate until most ofthe 1 ml of 0.1 M sodium nitrite is equivalent to 0.02503 g of
ammonia has been expelled, cool and acidifY with acetic acid. C IOH ION 40 ZS.
Collect the precipitate, wash with water and dry at about 100°;
the residue melts at about 256°, with decomposition (2.4.21). Storage. Store protected from light and moisture.

B. On the residue obtained in test A determine by infrared


absorption spectrophotometry (2.4.6). Compare the spectrum
with that obtained with sulphadiazine RS or with the reference Sulphadoxine
spectrum of sulphadiazine. Sulphonnethoxine; Sulphoethomidine
C. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with the reference solution.

Tests
Related substances (2.3.7). Complies with test C, but using
the following solutions.
Test solution (a). Extract a quantity of the powdered tablets Mol. Wt. 310.3
containing 0.5 g of Sulphadiazine with 25 ml ofa mixture of
Sulphadoxine is NI-( 5,6-dimethoxypyrirnidin-
90 volumes of methanol and 10 volumes of strong ammonia
4-yl)sulphanilamide.
solution by shaking for 10 minutes, filter and use the filtrate.
Sulphadoxine contains not less than 99.0 per cent and not
Test solution (b). Dilute I volume of test solution (a) to
more than 101.0 per cent ofClzH14N404S, calculated on the
5 volumes with a mixture of 24 volumes of methanol and
dried basis.
I volume of strong ammonia solution.
Category. Antibacterial; antimalarial in combination with
Test solution (c). Dilute I volume of test solution (a) to
pyrimethamine.
200 volumes with the same solvent mixture.
Dose. Orally, initially, 2 g; subsequent doses, I to 1.5 g weekly.
Reference solution. A 0.4 per cent w/v solution of
sulphadiazine RS in the same solvent mixture.
By deep intramuscUlar of slow intravenous injeCtion, 2.5 g
initially, followed by 1.5 g after four days.
Dissolution (2.5.2). Description. White or yellowish white crystals or crystalline
Apparatus. No. I, powder.

2170

www.webofpharma.com
IP 2010 SULPHAMETHIZOLE

Identification 1 ml of 0.1 M sodium nitrite is equivalent to 0.03103 g of


CI2HI4N404S.
Test A may be omitted iftests B, C andD are carried out. Tests
Band D may be omitted if tests A and C are carried out. Storage. Store protected from light and moisture.

A. Determine by infrared absorption spectrophotometry (2.4.6).


Compare the spectrum with that obtained with sulphadoxine
RS or with the reference spectrum of sulphadoxine. Sulphamethizole
B. In the test for Related substances, the principal spot in the
chromatogram obtained with reference solution (a)
corresponds to that in the chromatogram obtained with the
reference solution (c).
C. Dissolve 0.5 g in 1 ml of sulphuric .acid (40 per cent),
heating gently to effect solution, and continue heating until a
crystalline precipitate is produced. Allow to cool, add 10 ml of Mol. Wt. 270.3
2 M sodium hydroxide, cool again, add 25 ml of ether and Sulphamethizole is Nl-(5-methyl-l ,3,4-thiadiazol-
shake for 5 minutes. Dry the upper layer over anhydrous 2-yl)sulphanilamide.
sodium sulphate, filter and evaporate the solvent by heating
on a water-bath. The residue melts either at 80° to 82° or at 90° Sulphamethizole contains not less than 99.0 per cent and not
to 92°(2.4.21). more than 101.0 per cent of C9HION402S2, calculated on the
dried basis.
D. Dissolve about 5 mg in 10 ml of1 M hydrochloric acid and
dilute 1 ml to 10 ml with water. The resulting solution, without Category. Antibacterial.
further acidification, gives the reaction of primary aromatic Dose. 100 to 200 mg every four to six hours.
amines (2.3.1 ).
Description. White or yellowish white crystals or crystalline
powder; odourless.
Tests
Appearance ofsolution. Dissolve 0.8 gin 10 ml of1 Msodium Identification
hydroxide. The solution is not more intensely coloured than
Test A may be omitted iftests B, C andD are carried out. Tests
reference solution YS5, BYS5 or GYS5 (2.4.1).
C and D may be omitted if tests A and B are carried out.
Acidity. Heat 1.25 g ofthe finely powdered substance at about
A. Determine by infrared absorption spectrophotometry (2.4.6).
70° with 25 ml of carbon dioxide-free water for 5 minutes.
Compare the spectrum with that obtained with sulphamethizole
Cool for about 15 minutes in ice and filter. To 20 ml of the
RS or with the reference spectrum of sulphamethizole.
filtrate add 0.1 ml of bromothymol blue solution. Not more
than 0.2 ml of 0.1 M sodium hydroxide is required to change B. In the test for Related substances, the principal spot in the
the colour of the solution. chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution
Related substances. Complies with the test for related
(b).
substances in sulphonamides (2.3.7), Method C, but using
the following solution. C. Dissolve 50 mg in 4 ml of methanol and add 0.2 ml of a
4.0 per cent w/v solution of cupric acetate; a flocculent,
Test solution. A 2 per cent w/v solution of the substance
yellowish green precipitate is produced which becomes dark
under examination in a mixture of24 volumes of methanol and
green.
1 volume of strong ammonia solution.
D. Dissolve about 5 mg in 10 ml of1 M hydrochloric acid and
Heavy metals (2.3.13).1.0 g complies with the limit test for
dilute 1 ml ofthis solution to 10 ml with water. The solution,
heavy metals, Method B (20 ppm).
without further acidification, gives the reaction of primary
Sulphated ash (2.3.18). Not more than 0.1 per cent. aromatic amines (2.3.1).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105°. Tests
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of Appearance ofsolution. Dissolve 0.8 gin 10 ml of1 M sodium
2 M hydrochloric acid, add 3 g ofpotassium bromide, cool in hydroxide. The solution is not more intensely coloured than
ice and carry out the nitrite titration (2.3.31). reference solution YS5, BYS5 or GYS5 (2.4.1).

2171

www.webofpharma.com
SULPHAMETHIZOLE IP 2010

Acidity. Heat 1.25 g ofthe finely powdered substance at about Sulphamethoxazole contains not less than 99.0 per cent and
70° with 25 ml of carbon dioxide:free water for 5 minutes. not more than 101.0 per cent ofC IOH"N30 3S, calculated on the
Cool for about 15 minutes in ice and filter. To 20 ml of the dried basis.
filtrate add 0.1 ml of bromothymol blue solution. Not more
Category. Antibacterial.
than 0.2 ml of 0.1 M sodium hydroxide is required to change
the colour of the solution. Dose. Initial dose, 2 g; subsequent doses, 1 g two or three
times daily.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Description. A white or almost white, crystalline powder;
almost odourless.
Mobile phase. A mixture of 80 volumes of chloroform and
15 volumes of methanol.
Identification
Test solution (a). Dissolve 0.3 g of the substance under
examination in 10 ml of acetone. Test A may be omitted iftests B, C and D are carried out. Tests
Band C may be omitted if tests A and D are carried out.
Test solution (b). Dissolve 0.3 g of the substance under
examination in 100 ml of acetone. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with sulpha-
Reference solution (a). Dissolve 15 mg ofthe substance under
methoxazole RS or with the reference spectrum of sulpha-
examination in 100 ml of acetone.
methoxazole.
Reference solution (b). A 0.3 per cent w/v solution of
B. In the test for Related substances, the principal spot in the
sulphamethizole RS in acetone.
chromatogram obtained with reference solution (a)
Apply to the plate 2 f..Ll of each solution. After development, corresponds to that in the chromatogram obtained with the
dry the plate at 105° and examine in ultraviolet light at 254 nm. reference solution (c).
Any secondary spot in the chromatogram obtained with test
solution (a) is not more intense than the spot in the C. Dissolve about 5 mg in 10 ml of1 M hydrochloric acid and
chromatogram obtained with reference solution (a). dilute 1 ml to 10 ml with water. The resulting solution, without
further acidification, gives the reaction of primary aromatic
Heavy metals (2.3.13). 1.0 g complies with the limit test for amines (2.3.1).
heavy metals, Method B (20 ppm).
D. Melting range (2.4.21). 169° to 172°.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Tests
on 1.0 g by drying in an oven at 105°.
Appearance ofsolution. Dissolve 0.8 gin 10 ml of1 M sodium
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of2 M
hydroxide. The solution is not more intensely coloured than
hydrochloric acid, add 3 g of potassium bromide, cool in ice
reference solution YS5, BYS5 or GYS5 (2.4.1).
and carry out the nitrite titration (2.3.31).
Acidity. Heat 1.25 g of the finely powdered substance with
1 ml of 0.1 M sodium nitrite is equivalent to 0.02703 g of
25 ml of carbon dioxide:free water at 70° for 5 minutes. Cool
C9HION40zSz.
for about 15 minutes in ice and filter. To 20 ml ofthe filtrate add
Storage. Store protected from light and moisture. 0.1 ml of bromothymol blue solution. Notmore than 0.3 ml of
0.1 M sodium hydroxide is required to change the colour of
the solution.
Sulphamethoxazole Related substances. Complies with the test for Related
Substances in Sulphonamides (2.3.7), Method C, but using
the following solution.
Test solution. Dissolve 0.1 g of the substance under
examination in sufficient of a mixture of 1 volume of strong
ammonia solution and 24 volumes of methanol to produce
5ml.
C IOH lI N 30 3S Mol. Wt. 253.3 Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Sulphamethoxazole is Nl-(5-methylisoxazol-
3-yl)sulphanilarnide. Sulphated ash (2.3 .18). Not more than 0.1 per cent.

2172

www.webofpharma.com
IP 2010 SUMATRIPTAN

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Chromatographic system
on 1.0 g by drying in an oven at 105°. - a stainless steel column 25 cm x 4.6 rom, packed with
silica gel (5 j.Lm) (such as Spherisorb silica S5W),
Assay. Weigh accurately about 0.2 g, dissolve in 50 ml of
- mobile phase: a mixture of 10 volumes of 10 M
2 M hydrochloric acid, add 3 g ofpotassium bromide, cool in
ice and carry out the nitrite titration (2.3.31).
ammonium acetate and 90 volumes of methanol,
- flow rate. 2 ml per minute,
I ml of 0.1 M sodium nitrite is equivalent to 0.02533 g of - spectrophotometer set at 282 nm,
CIQH 11 N30 3S. - injection volume. 20 j.Ll.
Storage. Store protected from light and moisture. Inject the reference solution. Run the chromatogram 5 times
the retention time of the principal peak. The test is not valid
unless the resolution between [3-[2-(dimethylamino)ethyl]-
2-[[3-[2-(dimethylamino)ethyl]-IH-indol-5-yl]methyl]-IH-
Sumatriptan indol-5-yl]-N- methylmethanesulphonamide (sumatriptan
impurity A) and sumatriptan is not less than 1.5.
Inject test solution (a) and (b). In the chromatogram obtained
with test solution (a) the area of any peak corresponding to
sumatriptan impurity A is not more than the area ofthe principal
peak in the chromatogram obtained with test solution (b) (0.6
per cent, taking into account the correction factor of 0.6 for
impurity A) and the area of any peak due to [3-[2-
(dimethylamino)ethyl]-I-[[3-[2-(dimethylamino)ethyl]-IH-
CIJ1ZIN30ZS Mol. Wt. 295.4 indol-5-yl]methyl]-IH-indol-5-yl]-N- methylmethanesulphona-
mide (sumatriptan impurity H) is not more than 0.3 times the
Sumatriptan is 3-(2-dimethylaminoethyl)indol-5-yl-N-
area of the principal peak in the chromatogram obtained with
methylmethanesulphonamide.
test solution (b) (0.3 per cent).
Sumatriptan contains not less than 97.5 per cent and not more
Related substances. Determine by liquid chromatography
than 102.0 per cent of CI4HzIN30ZS, calculated on the
(2.4.14).
anhydrous basis.
Solvent mixture. 25 volumes of acetonitrile and 75 volumes
Category. Antimigraine.
of 0.025 M sodium dihydrogen orthophosphate, adjusted to
Description. A white to pale yellow powder. pH6.5.

Identification Test solution (a). A 0.2 per cent w/v solution of the substance
under examination in the solvent mixture.
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with sumatriptan Test solution (b). Dilute I ml oftest solution (a) to 100 ml with
RS. the solvent mixture and further dilute I ml of the resulting
solution to 10 ml with the solvent mixture.
Tests
Reference solution. Dilute the contents ofa vial of sumatriptan
Impurities A and H. Determine by liquid chromatography impurity mixture RSto I ml with 0.1 M hydrochloric acid.
(2.4.14).
Chromatographic system
Solvent mixture. I volume of acetonitrile and 3 volumes of - a stainless steel column 25 cm x 4.6 mm, packed with
0.025 M sodium dihydrogen orthophosphate, adjusted to octadecylsilane bonded to porous silica (5 j.Lm) ( such
pH 6.5. as Spherisorb ODS I),
Test solution (a). A 0.2 per cent w/v solution of the substance mobile phase: a mixture of 75 volumes of a solution
under examination in the solvent mixture. containing 0.97 g of dibutylamine, 0.735 g of sodium
orthophosphoric acid and 2.93 g of sodium dihydrogen
Test solution (b). Dilute I ml oftest solution (a) to 100 inl with orthophosphate in 750 ml water; adjusted to pH 6.5
the solvent mixture. with 10M sodium hydroxide and diluted to 1000 ml
Reference solution. Dissolve the contents of a vial of with water;
sumatriptan for system suitability RS to I ml with 1 M flow rate. 1.5 ml per minute,
hydrochloric acid. spectrophotometer set at 282 urn,

2173

www.webofpharma.com
SUMATRIPTAN IP 2010

- injection volume. 20 ,.ti. Sumatriptan Injection


Inject the reference solution. The test is not valid unless the Sumatriptan Injection is a sterile isotonic solution of
resolution between the peaks due to [3-[2-(dimethylamino)
Sumatriptan Succinate in Water for Injections.
ethyl]-I-(hydroxymethyl)-IH-indol-5-yl]-N- methylniethane-
sulphonamide (sumatriptan impurity C) and sumatriptan is Sumatriptan Injection contains not less than 95.0 per cent and
not less than 1.5. not more than 105.0 per cent of the stated amount of
sumatriptan succinate, CI4H2IN302S,C4H604.
Inject test solution (a) and (b). In the chromatogram obtained
Usual strength. 1 mg per ml.
with test solution (a) the areas of any peaks corresponding to
N-methyl[3- [2-(methylamino )ethyl] -lH-indol-5- Description. A clear, colourless to pale yellow solution.
yl]methanesulphonamide (sumatriptan impurity B), [3-[2-
(dimethylamino)ethyl]-I-(hydroxymethyl)-lH-indol-5-yl]-N- Identification
methylmethanesulphonamide (sumatriptan impurity C) and
To a volume ofthe injection containing 40 mg of sumatriptan
N ,N-dimethyl-2-[5-[(methyIsulphamoyl)methyl]-IH-indol-3-
succinate add 1 ml of saturated sodium chloride solution and
yl]ethanamine N-oxide (sumatriptan impurity D) is not more
1ml of saturated sodium carbonate solution. Shake
than 5 times the area ofthe principal peak in the chromatogram
vigorously for 30 seconds, add two quantities of2 rn1 ofpropan-
obtained with test solution (b) (0.5 per cent); the area of any
peak corresponding to [3-(2-aminoethyl)-lH-indol-5-yl]-N-
2-01, shake, allow to separate (this may take up to 24 hours)
and discard the aqueous layer. Evaporate under a stream of
methylmethanesulphonamide (sumatriptan impurity E) is not
nitrogen and dry at 100°. On the residue, determine by infrared
more than the principal peak in the chromatogram obtained
absorption spectrophotometry (2.4.6). Compare the spectrum
with test solution (b) (0.1 per cent); the area of any other
with that obtained with sumatriptan succinate RS.
secondary peak is not more than the principal peak in the
chromatogram obtained with test solution (b) (0.1 per cent).
The total impurity content in the test for Impurities A and H Tests
and in the test for Related substances is not more than 1.5 per pH (2.4.24). 4.2 to 5.3.
cent. Ignore any peak with an area less than 0.5 times the area
of the peak in the chromatogram obtained with test solution For impurities A and H. Determine by liquid chromatography
(b) (0.05 per cent). (2.4.14).
Solvent mixture. 75 volumes of 0.025 M sodium dihydrogen
Water (2.3.43). Not more than 1.0 per cent, determined on orthophosphate, adjusted to pH 6.5 and 25 volumes of
LOg. acetonitrile.
Assay. Determine by liquid chromatography (2.4.14) as Test solution. Dilute a volume ofinjection containing 30 mg of
described under Related substances, using the following Sumatriptan Succinate to 10.0 ml with the solvent mixture.
solutions.
Reference solution (a). Dilute 1 ml ofthe test solution to 100
Test solution. Dissolve 10 mg of the substance under ml with the solvent mixture.
examination in 100.0 ml ofthe
I
solvent mixture. Reference solution (b). Dilute the contents of a vial of
sumatriptan for system suitability RS (containing impurities
Reference solution (a). A 0.014 per cent w/v solution of A ([3-[2-(dimethylamino)ethyl]-2-[[3-[2-(dimethylamino)ethyl]-
sumatriptan succinate RS in the solvent mixture. 1H- indol-5-yl]methyl]-1 H-indol-5- yl]-N-methy Imethane
sulphonamide) and H ([3-[2-(dimethylamino)ethyl]-I-[[3-[2-
Reference solution (b). Dilute the contents of a vial of
(dimethylamino)ethyl]-lH-indol-5-yl]methyl]-lH-indol-5-yl]-
sumatriptan impurity mixture RS to 1 ml with 0.1 M
N-methylmethanesulphonamide) to 1 ml with 1 Mhydrochloric
hydrochloric acid.
acid.
Inject reference solution (b). The test is not valid unless the Chromatographic system
resolution between the peaks due to sumatriptan and a stainless steel column 25 cm x 4.6 mm, packed with
sumatriptan impurity C is not less than 1.5. silica (such as Spherisorb silica S5W),
mobile phase: a mixture of 10 ~()lu_IIl~~~iJO)\i
Inject reference solution (a) and the test solutioR
ammonium acetate and 90 volumes of methanol,
flow rate. 2 ml per minute,
- spectrophotometer set at 282 nm,
Storage. Store protected from light. injection volume. 20 Ill.

2174

www.webofpharma.com
IP 2010 SUMATRIPTAN INJECTION

Inject reference solution (b). The test is not valid unless the - injection volume. 20 fll.
resolution between the peaks due to sumatriptan and Inject reference solution (a). The test is not valid unless the
sumatriptan impurity A is not less than 1.5. resolution between the peaks due to sumatriptan impurity C
Inject the test solution and reference solution (a). Run the and sumatriptan is not less than 1.5.
chromatogram 5 times the retention time ofthe principal peak. Inject test solution (a), (b) and reference solution (b). In the
In the chromatogram obtained with the test solution the area chromatogram obtained with test solution (a) the area of any
of any peak corresponding to sumatriptan impurity A is not peak corresponding to (3a-hydroxy,l, I-dimethyl-5-
more than 1.5 times the area of the principal peak in the [(methylamino )sulfonyl]methyl-l ,2,3,3 a,8,8 a-
chromatogram obtained with reference solution (a) (1.5 per hexahydropyrrolo[2,3-b ]indol-l-ium trifuoroacetate)
cent) and the area of secondary peak corresponding to (sumatriptan impurity A) is not more than 1.5 times the area of
sumatriptan impurity H is not more than the area ofthe principal the principal peak in the chromatogram obtained with test
peak in the chromatogram obtained with reference solution solution (b) (1.5 per cent), the area ofany peak corresponding
(a) (1.0 per cent). to 2 (1-3-[2-(dimethylamino)ethyl]-3-hydroxy-2-oxo-2,3-
Related substances. Determine by liquid chromatography dihydro-l H-indol-5-y I-N-methy Imethanesulfonamide)
(2.4.14). (sumatriptan impurity H) is not more than the area of the
principal peak in the chromatogram obtained with test solution
Solvent mixture. 75 volumes of 0.025 M sodium dihydrogen
(b) (1.0 per cent). The area of any other secondary peak is not
orthophosphate, the pH of which has been adjusted to 6.5
more than the area of the principal peak in the chromatogram
and 25 volumes acetonitrile.
obtained with test solution (b) (1.0 per cent) and sum of areas
Test solution (a). Dilute a volume of injection containing 30 ofthe peaks due to sumatriptan impurity A and H is not more
mg ofSumatriptan Succinate in 10 ml ofthe solvent mixture. than 4 times the area ofthe principal peak in the chromatogram
Test solution (b). Dilute 1.0 ml oftest solution (a) to 100.0 ml obtained with test solution (b) (4.0 per cent).
with the solvent mixture. Other tests. Complies with the tests stated under Parenteral
Reference solution (a). Dilute the contents of a vial of Preparations (Injections).
sumatriptan impurity mixture RS (contains sumatriptan Bact,erial endotoxins (2.2.3). Notmore than 350.0 Endotoxin
impurity B (N-methyl[3-[2-(methylamino)ethyl]-IH-indol-5- Units per ml of sumatriptan succinate.
yl]methanesulphonamide), C ([3-[2-(dimethylamino)ethyl]-l-
(hydroxymethyl)- iH- indol-5-yl]-N-methylmethanesulphona- Assay. Determine by liquid chromatography (2.4.14).
mide), D (.N,N-dimethyl-2-[5-[(methylsulphamoyl)methyl]-IH- Test solution. Dilute the volume of injection containing 3 mg
indol-3-yl]ethanamine N- oxide), and E ([3-(2-aminoethyl)-IH- ofSumatriptan succi~ate in 100 ml ofthe solvent mixture.
indol-5-yl]-N-methylmethanesulphonamide) to 1 ml with 0.1
M hydrochloric acid. Reference solution (a). A 0.003 per cent w/v solution of
sumatriptan succinate RS in the solvent mixture.
Reference Solution (b). A 0.3 per cent w/v solution of
sumatriptan impurity standard RS in the solvent mixture. Reference solution (b). Dilute the contents of a vial of
sumatriptan impurity mixture RS (contains impurities B, C, D
Chromatographic system
and E) to 1 ml with 0.1 M hydrochloric acid.
- a stainless steel column 25 cm x 4.6 rom, packed with
octadecylsilane bonded to porous silica (5 flm) (such as Use chromatographic system as described under Related
SpherisorbODS 1), substances.
mobile phase: a mixture of 25 volumes of acetonitrile
Inject reference solution (b). The test is not valid unless
and 75 volumes of a solution containing 0.97 g of
resolution between the peaks due to sumatriptan succinate
dibutylamine, 0.735 g of orthophosphoric acid and 2.93
and sumatriptan succinate impurityC is not less than 1.5.
g of sodium dihydrogen orthophosphate, adjust the
pH to 7.5 with 10M sodium hydroxide and diluted to Inject reference solution (a) and the test solution.
1000 ml with water,
Calculate the content ofC14Hz1N30zS,C4H604in the injection.
flow rate. 1.5 ml per minute,
spectrophotometer set at 282 nm, Storage. Store protected from light.

2175

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

T
Talc 2181
Tamoxifen Citrate 2181
TamoxifenTablets 2183
Tamsulosin Hydrochloride 2184
Tartaric Acid 2185
Telmisartan 2186
TelmisartanTablets 2187
Tenofovir Disoproxil Fumarate 2188
Tenofovir Disoproxil Fumarate Tablets 2190
Tenofovir and Emtricitabine Tablets 2191
Temozolomide 2193
Temozolomide Capsules 2193
Terazosin Hydfochloride Dihydrate 2194
Terbutaline Sulphate 2195
Terbutaline Inhalation 2197
Terbutaline Injection 2197
Terbutaline Tablets 2198
Testosterone Propionate 2199
Testosterone Propionate Injection 2200
Tetracycline 2201
Tetracycline Hydrochloride 2202
Tetracycline Capsules 2203
Tetracycline Ointment 2204
Theophylline 2205
Theophylline Injection 2206
Thiabendazole 2206
Thiabendazole Tablets 2207
Thiacetazone 2208
Thiacetazone and Isoniazid Tablets 2208
Thiamine Hydrochloride 2209

2177

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Thiamine Injection 2210


Thiamine Tablets 2211
Thiamine Mononitrate 2212
Thiocolchicoside 2213
Thiocolchicoside Capsules 2214
Thiomersal 2215
Thiopentone Sodium 2216
Thiopentone Injection 2217
Thiotepa 2218
ThiotepaInjection 2219
Thymol 2220
Thyroxine Sodium 2221
ThyroxineTablets 2222
Ticarcillin and ClavulanicAcidInjection 2223
Timolol Maleate 2224
Timolol Eye Drops 2224
Timolol Tablets 2225
Tinidazole 2226
Tinidazole Tablets 2227
Tiotropium Bromide Monohydrate 2227
Tiotropium Powder for Inhalation 2228
TitaniumDioxide 2229
Tizanidine Hydrochloride 2230
TizanidineTablets 2231
Tobramycin ·2232
Tobramycin Injection 2233
Tocopheryl Acetate 2234
Tolazarnide 2235
Tolazarnide Tablets 2235
Tolbutamide 2236
Tolbutamide Tablets 2237
Tolnaftate 2238
Tolnaftate Cream 2239

2178

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Tolnaftate Gel 2239


Tolnaftate Topical Powder 2239
Tolnaftate Topical Solution 2240
Tolterodine Tartrate 2240
Topiramate 2242
Topiramate Tablets 2243
Topotecan Hydrochloride 2243
Topotecan Injection 2245
Tramadol Hyrochloride 2245
Tramadol Capsules 2246
Trandolapril 2247
Trandolapril Tablets 2248
Travoprost 2250
Travoprost Eye Drops 2251
Triamcinolone 2252
Triamcinolone Tablets 2252
TriamcinoloneAcetonide 2253
TriamcinoloneAcetonide Injection 2255
Triamterene 2256
.Triamterene Capsules 2256
Tributyl Citrate 2257
Trichloromonofluoromethane 2257
Triethyl Citrate 2258
Trifluoperazine Hydrochloride 2259
Trifluoperazine Injection 2260
Trifluoperazine Tablets 2260
TriflupromazineHydrochloride 2261
Triflupromazine Injection 2262
Triflupromazine Tablets 2262
Trimetazidine Hydrochloride 2263
Trimethoprim 2264
Trimethoprim and Sulphamethoxazole Oral Suspension 2265
Trimethoprim and Sulphamethoxazole Tablets 2266

2179

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

TrimethoprimTablets 2267
Triprolidine Hydrochloride 2268
Triprolidine Tablets 2268
Trisodium Edetate Concentrate for Injection 2269
Tropicamide 2270
Tropicamide Eye Drops 2270
Troxidone 2271
Troxidone Capsules 2272
Tubocurarine Chloride 2272
Tubocurarine Injection 2273

2180

www.webofpharma.com
IP 2010 TAMOXIFEN CITRATE

Talc C. The residue obtained in identification test B gives the


reaction ofsilicates (2.3.1).
Purified Talc; Talcum
Tests
Talc is a powdered, selected natural hydrated magnesium
silicate. It may contain varying amounts of aluminium and Acidity or allmUnity. Shake 5.0 g with 25 ml of carbon dioxide-
iron in forms insoluble in 1 M sulphuric acid. free water for 1 minute, filter and add to the filtrate 0.5 ml of
bromothymol blue solution; the solution is not acidic and
Category. Pharmaceutical aid (dusting powder).
requires not more than 0.3 ml of 0.1 M hydrochloric acid to
Description. A white or almost white powder, free from make it acidic.
grittiness; readily adheres to the skin; unctuous to the touch; Iron (2.3.14). Boil 4.0 g with 25 ml of water for 30 minutes,
odourless. replacing the water lost by evaporation, and filter. The filtrate,
Production after the addition of5 ml of nitric acid, complies with the limit
test for iron (10 ppm).
Talc derived from deposits that are known to contain associated
Acid-soluble substances. Not more than 2.0 per cent,
asbestos is not suitable for pharmaceutical use. The
determined by the following method. Digest 2.0 g with 40 ml of
manufacturer is responsible for demonstrating by the test for
dilute hydrochloric acid for 15 minutes, filter, evaporate the
amphiboles and serpentines that the product is free from
filtrate; to the residue add 0.1 ml of sulphuric acid and ignite
asbestos. The presence of amphiboles and of serpentines is
to constant weight.
revealed by infrared spectrophotometry. If detected, the
specific morphological criteria of asbestos are investigated Water-soluble substances. Shake 5.0 g with 25 ml of water for
by a suitable method of optical microscopy to determine 1 minute, filter, evaporate the filtrate and dry to constant weight;
whether tremolite asbestos or chrysotile is present, as the residue weighs not more than 10 mg.
described below. Carbonates. To 1 g add 20 ml of dilute hydrochloric acid; no
Determine by infrared absorption spectrophotometry (2.4.6). effervesc~nce is
produced.
In the range 740 em-I to 760 em-I using scale expansion, any Chlorides (2.3.12). Suspend 2.0 gin 10 mlofwater, add lOml
absorption band at 758 cm- I may indicate the presence of of2 M nitric acid, shake for 15 minutes and filter. 10 ml ofthe
tremolite or of chlorite. If the absorption band remains after filtrate complies with the limit test for chlorides (250 ppm).
ignition ofthe substance under examination at 850 ± 500 for at Organic compounds. The residue obtained in the test for Loss
least 30 minutes, it indicates the presence ofthe tremolite. In on drying is not more than slightly yellow or grey.
the range 600 em-I to 650 cm- 1 using scale expansion, any
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
absorption band or shoulder may indicate the presence of
on 1.0 g by drying in an oven at 1800 for 1 hour.
serpentines.
Microbial contamination (2.2.9).
Identification
If intended for topical administration, the total viable aerobic
TestA may be omitted if tests B, Care carried out. Tests Band count is not more than a total of 102 bacteria and fungi per gram.
C may be omitted if test A is carried out. If intended for oral administration, the total viable aerobic
A. Determine by infrared absorption spectrophotometry (2.4.6), count is not more than 103 bacteria and not more than 102 fungi
shows absorption bands at 3677 em-I, 1018 em-I and 669 em-I. per gram.
B. In a platinum crucible, melt a mixture of0.2 g of anhydrous Storage. Store protected from moisture.
sodium carbonate and 2.0 g of potassium carbonate. To the
melted mass add 0.1 g of the substance under examination
and heat until the mixture is completely melted. Allow to cool
and transfer the melted mass into an evaporating dish with 50 Tamoxifen Citrate
mlofhot water. Add hydrochloric acid until effervescence
ceases. Add 10 ml of hydrochloric acid and evaporate to
dryness on a water-bath. Allow to cool. Add 20 ml of water, HO COOH
heat to boiling and filter (the residue is used for identification
test C). To 5 ml ofthe filtrate add 1 ml of ammonia and 1 mlof
, HOOC~COOH
ammonium chloride solution and filter. To the filtrate add 1
ml of· disodium hydrogen phosphate solution. A white,
crystalline precipitate is formed. Mol. Wt. 563.7

2181

www.webofpharma.com
TAMOXIFEN CITRATE IP 2010

Tamoxifen Citrate is (Z)-2-[4-(1,2-diphenylbut-l-enyl)- Reference solution (b). A 0.25 per cent w/v solution of
1- phenoxy]ethyldimethylamine citrate. tamoxifen citrate impurity standard RS in the same solvent
Tamoxifen Citrate contains not less than 99.0 per cent and not mixture.
more than 101.0 percent ofC26H 29NO, C 6H s0 7, calculated on Chromatographic system
the dried basis. a stainless steel column 20 cm x 5 rom, packed with
.Category. Antioestrogen. octadecylsilane bonded to porous silica (5 Ilm),
mobile phase: a mixture of 60 volumes of acetonitrile,
Dose: The equivalent of 20 to 40 mg of tamoxifen daily as a 25 volumes of water, 15 volumes of tetrahydrojitran
single dose or in 2 divided doses. and 0.4 volume of strong ammonia solution,
Description. A white or almost white, crystalline powder. flow rate. 1.5 ml per minute,
spectrophotometer set at 240 nm,
Identification injection volume. 20 Ill.
Test A may be omitted iftests B, C andD are carried out. Tests In the chromatogram obtained with reference solution (b) a
C and D may be omitted iftests A and B are carried out. peak due to E-isomer appears immediately following the peak
due to Z-tamoxifen. Adjust the sensitivity of the instrument
A. Determine by infrared absorption spectrophotometry (2.4.6).
so that the height ofthe peak due to E-isomer is about 15 per
Compare the spectrum with that obtained with tamoxifen
cent ofthe full-scale deflection on the recorder. Measure the
citrate RS or with the reference spectrum oftamoxifen citrate.
height ofthe peak due to E-isomer by dropping a perpendicular
B. When examined in the rang~ 220 nm to 360 nm (2.4.7), a from the top of the peak to a line drawn tangentially between
0.002 per cent w/v solution in methanol shows absorption the troughs on each side of the E-isomer peak or the trough
maxima at about 237 nm and 275 nin. between the E- and Z-isomer peaks and the baseline, whichever
is appropriate.
C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254. The test is not valid unless the height ofthe trough separating
Mobile phase. A mixture of 90 volumes of toluene and the peaks due to E- and Z-tamoxifen in the chromatogram
10 volumes of triethylamine. obtained with reference solution (b) is less than 7 per cent of
full-scale deflection on the recorder and the retention time of
Test solution. Dissolve 0.1 g of the substance under the principal peak is less than 30 minutes. (The retention time
examination in 10 ml of methanol. decreases with increasing concentration of ammonia in the
Reference solution. A 1.0 per cent w/v solution of tamoxifen mobile phase).
citrate RS in methanol. The content of E-isomer in the substance under examination
Apply to the plate 5 I.d of each solution. After development, is not more than 1.0 per cent calculated from the declared
dry the plate in air and examine in ultraviolet light at 254 nm. content of E-isomer in tamoxifen citrate impurity standard
The principal spot in the chromatogram obtained with the test RS. The area of any secondary peak in the chromatogram
solution corresponds to that in the chromatogram obtained obtained with the test solution, other than a peak due to E-
with the reference solution. isomer, is not greater than halfthat ofthe peak due to tamoxifen
citrate in the chromatogram obtained with reference solution
D. To 10 mg add 4 ml ofpyridine and 2 ml of acetic anhydride
(a) and the sum of the areas of all such peaks is not greater
and shake; a yellow colour is produced. Heat in a water-bath
than the area ofthe peak due to tamoxifen in the chromatogram
for 2 minutes; a pink to red colour is produced.
obtained with reference solution (a). Ignore any peak with a
retention time less than 2.5 minutes and any peak with an area
Tests
less than 0.05 times the area of the principal peak in the
E-Isomer and related substances. Determine by liquid chromatogram obtained with reference solution (a).
chromatography (2.4.14).
Heavy metals (2.3.13). 2.0 g complies with the limit test for
Test solution. Dissolve 0.25 g of the substance under heavy metals, Method B (10 ppm).
examination in 100 ml ofa mixture of60 volumes ofacetonitrile,
Sulphated ash (2.3.18). Not more than 0.2 per cent.
25 volumes of water and 15 volumes of tetrahydrofilran.
Loss on drying(2.4; 19);Notmore than05percent;determined
Reference solution (a). A 0.0025 per cent w/v solution of the on 1.0 g by drying in an oven at 105°.
substance under examination in a mixture of 60 volumes of
acetonitrile, 25 volumes of water and 15 volumes of Assay. Weigh accurately about 1.0 g, dissolve in 150 ml of
tetrahydrofuran. anhydrous glacial acetic acid. Titrate with 0.1 M perchloric

2182

www.webofpharma.com
IP 2010 TAMOXIFEN TABLETS

acid, using 1-naphtholbenzein solution as indicator. Carry solution corresponds to that in the chromatogram obtained
out a blank titration. with the reference solution.
1 ml of 0.1 M perchloric acid is equivalent to 0.05636 g of D. Dissolve 10 mg of the residue obtained in the preparation
C26H29NO, C6Hs0 7• of the test solution in test C in 4 ml of pyridine, add 2 mlof
acetic anhydride and heat on a water-bath for 2 minutes; a
Storage. Store protected from light and moisture.
pink to red colour is produced.

Tests
Tamoxifen Tablets E-Isomer and Related substances. Determine by liquid
chromatography (2.4.14).
Tamoxifen Citrate Tablets
Test solution. Mix a quantity of the powdered tablets
Tamoxifen Tablets contain not less than 90.0 per cent and not
containing 60 mg of tamoxifen with 60 ml of a mixture of
more than 110.0 per cent of the stated amount oftamoxifen,
60 volumes of acetonitrile, 25 volumes of water and
C26H29NO.
15 volumes oftetrahydroJuran with the aid of ultrasound for
Usual strengths. The equivalent of 10 mg, 20 mg and 40 mg of 5 minutes, dilute to 100 ml with the same solvent mixture and
tamoxifen. filter through Whatrnan No.1 filter paper.
Reference solution (a). Dilute 1.0 ml of the test solution to
Identification
100 ml with the same solvent mixture.
A. To a quantity of the powdered tablets containing 0.1 g of Reference solution (b). A 0.1 per cent w/v solution of
tamoxifen add 20 ml of water, warm, add 2 ml of 5 M sodium tamoxifen citrate impurity standard RS in the same solvent
hydroxide and cool. Extract with two quantities, each of 10 ml, mixture.
of ether, filtering after each extraction. Combine the ether
extracts and evaporate to dryness in a stream of nitrogen at Chromatographic system
room temperature. Dry the residue at a pressure not exceeding a stainless steel column 20 em x 5 rom, packed with
0.7 kPa for 30 minutes. octadecylsilane bonded to porous silica (5/Lm),
- mobile phase: a mixture of 60 volumes of acetonitrile,
On the residue, determine by infrared absorption 25 volumes of water, 15 volumes of tetrahydrofilran
spectrophotometry (2.4.6). Compare the spectrum with that and 0.4 volume of strong ammonia solution,
obtained with tamoxifen citrate RS or with the reference flow rate. 1.5 rnl per minute,
spectrum oftamoxifen citrate. spectrophotometer set at 240 nm,
B. When examined in the range 220 om to 360 nm (2.4.7), a injection volume. 20 ILL
0.002 per cent w/v solution in methanol ofthe residue obtained In the chromatogram obtained with reference solution (b) a
in test A shows absorption maxima at about 237 nm and peak due to E-isomer appears immediately following the peak
275nm. due to Z-tamoxifen. Adjust the sensitivity of the instrument
C. Determine by thin-layer chromatography (2.4.17), coating so that the height of the peak due to E-isomer is about 15 per
the plate with silica gel GF254. cent offull-scale deflection on the recorder. Measure the height
ofthe peak due to E-isomer by dropping a perpendicular from
Mobile phase. A mixture of 100 volumes of ethyl acetate, the top of the peak to'a line drawn tangentially between the
10 volumes of methanol and 1 volume of strong ammonia troughs on each side of the E-isomer peak or the trough
solution. between the E- and Z-isomer peaks and the baseline, whichever
Test solution. Shake a quantity or the powdered tablets is appropriate.
containing 0.1 g tamoxifen with 10 ml of methanol, filter, The test is not valid unless the height ofthe trough separating
evaporate the filtrate to dryness on a water-bath, dry the the peaks due to E- and Z-tamoxifen in the chromatogram
residue at 100° for 30 minutes and dissolve 10 mg ofthe residue obtained with reference solution (b) is less than 7 per cent of
in 10 ml of methanol. the full-scale deflection on the recorder and the retention time
Reference solution. A 0.1 per cent w/v solution of tamoxifen of the principal peak is less than 30 minutes. (The retention
citrate RS in methdnol. time decreases with increasing concentration of ammonia in
the mobile phase).
Apply to the plate 10 ILl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. The content of E-isomer in the substance under examination
The principal spot in the chromatogram obtained with the test is not more than 1 per cent calculated from the declared content

2183

www.webofpharma.com
TAMOXIFEN TABLETS IP 2010

of E-isomer in tamoxifen citrate impurity standard RS. The Tamsulosin Hydrochloride contains not less than 98.0 per
area of any secondary peak in the chromatogram obtained cent and not more than 102.0 per cent of CzoHz8NzOsS,HCI,
with the test solution, other than a peak due to E-isomer, is calculated on the dried basis.
not greater than half that of the peak due to tamoxifen citrate Description. A white to offwhite powder.
in the chromatogram obtained with reference solution (a) and
the sum of the areas of all such peaks is not greater than the Dose. 400 to 800 Jlg daily.
area ofthe peak due to tamoxifen in the chromatogram obtained
with reference solution (a). Ignore any peak with a retention Identification
time less than 2.5 minutes and any peak with an area less than A. Determine by infrared absorption spectrophotometry (2.4.6).
0.05 times the area ofthe principal peak in the chromatogram Compare the spectrum with that obtained with tamsulosin
obtained with reference solution (a). hydrochloride RS or with the reference spectrum of
Uniformity of content. (For tablets containing 10 mg or less) tamsulosin hydrochloride.
- Comply with the test stated under Tablets. B. When examined in the range 200 nm to 350 urn (2.4.7), a
0.001 per cent w/v solution in methanol shows absorption
Crush one tablet and transfer to a 1OO-ml volumetric flask with
maxima at about 279 and 225 urn.
the aid of 75 ml of methanol. Shake well for 5 minutes, add
sufficient methanol to produce 100.0 ml and filter. Dilute C. Dissolve with heating 0.75 g in water and dilute to 100 ml
10.0 ml ofthe filtrate to 100.0 ml with methanol. Measure the with water. To 5 ml of the solution, cooled in an ice-bath add
absorbance ofthe resulting solution at the maximum at about 3 ml of dilute nitric acid and shake. Allow to stand at room
275 urn (2.4.7). Calculate the content ofC z6H z9NO in the tablet temperature for 30 minutes and filter. The filtrate gives reaction
taking 325 as the specific absorbance at 275 urn. A ofchlorides (2.3.1).

Other tests. Comply with the tests stated under Tablets. Tests
Assay. Weigh and powder 20 tablets. Weigh accurately a Specific optical rotation (2.4.22). -17.0° to-21.0°, determined
quantity of the powder containing about 25 mg oftamoxifen, in a 0.75 per cent w/v solution.
shake with 100 ml of methanol for 15 minutes, add sufficient Related substances. Determine by liquid chromatography
methanol to produce 250.0 ml and filter. Dilute 10.0 ml ofthe (2.4.14).
filtrate to 100.0 ml with methanol and measure the absorbance
A. Test solution. Dissolve 50.0 mg of the substance under
ofthe resulting solution at the maximum at about 275 run (2.4.7).
examination in 10 ml ofthe mobile phase.
Calculate the content of Cz6H z9NO taking 325 as the specific Reference solution(a). Dilute 1.0 ml of the test solution to
absorbance at 275 nm. 100.0 ml with the mobile phase. Dilute 1.0 ml ofthis solution to
Storage. Store protected from light and moisture. 10.0 ml with the mobile phase.
Labelling. The label states the strength in terms of the Reference solution (b). Dissolve 4 mg of 2-methoxy-5-[(2R)-
equivalent amount oftamoxifen. 2-[(2-(2-methoxyphenoxy)ethyljaminoJpropyljbenzenesul-
phonamide RS (tamsulosin impurity A RS) and 4 mg of the
substance under examination in the mobile phase and dilute
to 20.0 ml with the mobile phase. Dilute 2.0 ml ofthis solution
to 20.0 ml with the mobile phase.
Tamsulosin Hydrochloride Reference solution (c). Dissolve 4 mg of (2R)-N- [2-(2-
ethoxyphenoxy)ethyIJ-l-(4-methoxyphenyl)propan-2-amine
RS (tamsulosin impurity B RS) and 4 mg of the substance
under examination in the mobile phase and dilute to 20.0 ml
with the mobile phase. Dilute 2.0 ml ofthis solution to 20.0 ml
, Hel
with the mobile phase
Chromatographic system
- a stainless steel column 15 cm x 4.6 rum, packed with
octadecylsilane bonded to porous silica (5 !!m),
CzOHZ8NzOsS,HCL_.. _ .. _._ _MoLWt.A45.0. (il'liiinn"temp'eiilfiiTe'.···40d~· . -.-.-.--.-.,----"--,-",','-,-',',-~,,',',"-,,---,-'_ . _._-
Tamsulosin Hydrochloride is (R)..5..(2 ([2"(0"ethoxyphenoxy) mobile· phase: a mixture of 600 ml of acetiiizifrile arid
ethyl]aminopropyl)-2-methoxybenzenesulfonamide 1400 ml of a solution prepared by dissolving 3.0 g of
hydrochloride. sodium hydroxide in a mixture of 8.7 ml of perchloric

2184

www.webofpharma.com
IP 2010 TARTARIC ACID

acid and 1900 ml of water, adjusting the pH to 2.0 with Reference solution (b). Dissolve 5.0 mg of tamsulosin
0.5 M sodium hydroxide and diluting to 2000 ml with racemate RS in methanol and dilute to 25.0 ml with methanol.
water, Dilute 2.0 ml ofthis solution to 10.0 ml with methanol.
- flow rate. 1.3 ml per minute, Chromatographic system
- spectrophotometer set at 225 nm, . astainless steel column 25 cm x.4.6 mm packed with
- injection volume. 10 Ill. silica gel OD for chiral separations (Such as Chiralpak
Inject the test solution and reference solutions (a) and (b) and AD-H) (5 Ilm),
record the chromatograms for 1.5 times the retention time of - column temperature. 40°,
tamsulosin (retention time = about 6 minutes). In the - mobile phase: a mixture of 1 volume of diethylamine,
chromatogram obtained with reference solution (b), the 150 volumes of methanol, 200 volumes of anhydrous
resolution between the peaks due to tamsu10sin impurity A ethanol and 650 volumes of hexane,
and tamsulosin is not less than 6. - flow rate. 0.5 mlperminute,
- spectrophotometer set at 225 nm,
In the chromatogram obtained with the test solution, for each
- injection volume. 10 Ill.
unspecified impurity eluting before tamsuIosin, the area of
the peak is not more than the area of the principal peak in the Inject reference solution (b). Record the chromatogram for
chromatogram obtained with reference solution (a) (0.1 per about 20 minutes. The resolution between tamsulosin and
cent). Ignore any peak with an area less than 0.5 times the area impurity C (5-[(2S)-2-[[2-(2-ethoxyphenoxy) ethyl]amino]
of the principal peak in the chromatogram obtained with propyl]-2-methoxybenzenesulphonamide) is not less than 2.
reference solution (a) (0.05 per cent). The relative retention with reference to tamsulosin (retention
time = about 14 minutes) oftamsulosin impurity C is about 0.8.
B. Repeat the chromatographic procedure with the following
modifications. Inject the test solution and reference solution (a). In the
- mobile phase: dissolve 3.0 g of sodium hydroxide in a chromatogram obtained, the area ofthe peak obtained due to
mixture of8.7 ml ofperch10ric acidand 900 ml ofwater, tamsulosin impurity C is not more than the area ofthe principal
adjust the pH to 2.0 with 0.5 M sodium hydroxide and peak in the chromatogram obtained with reference solution
dilute to 2000 ml with water; add 2000 ml ofacetonitrile, (a) (0.1 percent).
- flow rate. 1 ml per minute, Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Inject the test solution and reference solutions (a) and (c).
Run the chromatograms for 5 times the retention time of Sulphated ash (2.3.18). Not more than 0.1 per cent.
tamsulosin (retention time = 2.5 minutes). In the chromatogram Loss on drying (2.4.19). Not more than 0.5 per cent, determined
obtained with reference solution (c), the resolution between on 1.0 g by drying in an oven at 105° for 2 hours.
the peaks due to tamsulosin and tamsuIosin impurity B is not
Assay. Weigh accurately about 0.35 g and dissolve in 5.0 ml of
less than 2. .
anhydrousformic acid, add 75 ml ofa mixture of2vo1umes of
In the chromatogram obtained with the test solution, for each acetic anhydride and 3 volumes ofglacial acetic acid Titrate
unspecified impurity eluting after tamsulosin, the area of any immediately with 0.1 M perchloric acid, determining the end-
peak is not more than the area of the principal peak in the point potentiometrically (2.4.25). Carry out a blank titration.
chromatogram obtained with reference solution (a) (0.1 per
1 ml of 0.1 M perchloric acid is equivalent to 0.0445 g of
cent). The sum of the areas of the impurities eluting before
CzoHzsNzOsS,HCl.
tamsulosin in test A and after tamsulosin in test B is not more
than twice the area ofthe principal peak in the chromatogram Storage. Store protected from light.
obtained with reference solution (a) (0.2 per cent). Ignore any
peak with an area less than 0.5 times the area of the principal
peak in the chromatogram obtained with reference solution Tartaric Acid
(a) (0.05 per cent).
L- Tartaric Acid
S-isomer. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve 0.05 g of the substance under
examination in methanol and add sufficient methanol to
H
,,'OH
HOOC/\ /COOH.
produce 25.0 ml.
H;<'OH
Reference solution (aj. Dilute 1.0 ml of the test solution to
100.0 ml with methanol. Dilute 1.0 m1 ofthis solution to 10.0 ml C4H60 6 Mol. Wt. 150.1
with methanol. Tartaric Acid is (2R,3R)-2,3-dihydroxybutanedioic acid.

2185

www.webofpharma.com
TARTARIC ACID IP 2010

Tartaric Acid contains not less than 99.5 per cent and not Telmisartan
more than 101.0 per cent ofC"H60 6, calculated on the dried
basis.
Category. Pharmaceutical aid (buffering agent).
Description. Colourless crystals or a white or almost white
crystalline powder.

Identification
A. Ignite a few mg; it gradually decomposes giving off an
odour resembling that of burnt sugar (distinction from citric
acid). .
Mol. Wt. 514.6
B. A 10 per centw/v solution in distilled water (solution A) is
strongly acidic. Telmisartan is 4' - ([4-methyl-6-(1-methyl-lH-benzimidazol-2-
yl)-2-propyl-lH-benzimidazol-l-yl]methyl}-2-
C. Gives reactions A and B oftartrates (2.3.1). biphenylcarboxylic acid.
Telmisartan contains not less than 98.0 per cent and not more
Tests than 102.0 per cent of C33H30N40z, calculated on the dried
basis.
Appearance of solution. Solution A is clear (2.4.1), and not
more intensely coloured than reference solution YS6 (2.4.1). Category. Antihypertensive.
Dose. 40 mg once a day.
Specific optical rotation (2.4.22). +12.0° to +12.8°, determined
in a 20.0 per cent w/v solution. Description. A white to off-white crystalline powder.

Arsenic (2.3.1 0). Dissolve 5.0 g in 50 ml ofwater and add 10 ml Identification


of stannated hydrochloric acid AsT. The resulting solution Detennine by infrared absorption spectrophotometry (2.4.6).
complies with the limit test for arsenic (2 ppm). Compare the spectrum with that obtained with telmisartan RS
or with the reference spectrum oftelmisartan.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method A (10 ppm). Tests
Chlorides (2.3.12). 20 ml ofsolution A complies with the limit Appearance of solution. A 5 p~r cent w/v solution in 1 M
test for chlorides (125 ppm). sodium hydroxide is not more intensely coloured than
reference solution YS4(2.4.1).
Sulphates (2.3.17). 10 ml ofsolution A complies with the limit
Related substances. Detennine by liquid chromatography
test for sulphates (150 ppm).
(2.4.14).
Oxalate. Neutralise 10 ml of solution A with dilute ammonia Test solution. Dissolve about 25 mg of the substance under
solution, add 0.1 ml of dilute acetic acid and 10 ml of calcium examination in 5 ml of methanol, add 100 III ofa 4 per cent w/v
sulphate solution; no opalescence is produced within solution of sodium hydroxide and dilute to 50.0 ml with
20 minutes. methanol.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Reference solution. Dilute 1.0 ml ofthe test solution to 10.0 ml
with methanol. Dilute 1.0 ml ofthis solution to 100.0 ml with
Loss on drying (2.4.19). Not more than 0.2 per cent, determined methanol.
on 1.0 g by drying in an oven at 105°. Chromatographic system
Assay. Weigh accurately about 1.0 g, dissolve in 25 ml of - a stainless steel column 12.5 cm x 4.0 mm, packed with
water and titrate with 1 M sodium hydroxide using 0.5 ml of octadecylsilane bonded to porous silica (5 11m), (Such
phenolphthalein solution. as indicator. as Thennoquest),
colurnntemperature. 40°,
1 ml of 1 M sodium hydroxide is equivalent to 0.07505 g of mobile phase: A. dissolve 2.0 g ofpotassium dihydrogen
CJI60 6. phosphate and 3.8 g of sodium pentanesulphonate
monohydrate in water, adjusted to pH 3.0 with
Storage. Store protected from moisture. orthophosphoric acid and dilute to 1000 ml with water,

2186

www.webofpharma.com
IP 2010 TELMISARTAN TABLETS

B. a mixture of 20 volumes of methanol and adjust the pH to 2.4 with orthophosphoric acid
and 80 volumes of acetonitrile, and 40 volumes of acetonitrile,
a linear gradient programme using the conditions given flow rate. 1 ml per minute,
below, spectrophotometer set at 298 nm,
flow rate. 1 ml per minute, injection volume. 20 /-ll.
- spectrophotometer set at 230 nID,
Inject the reference solution. The test is not valid unless the
injection volume. 10 /-ll.
theoretical plates is not less than 3000, the,tailing factor is not
Time Mobile phase A Mobile phase B more than 2.0 and the relative standard deviation for replicate
(min.) (per cent v/v) (per cent v/v) injections is not more than 2.0 per cent.
0-3 70 30 Inject the test solution and the reference solution.
3-28 70-+20 30-+80 Calculate the content ofC33H30N40z.
30 70 30
Storage. Store protected from light and moisture.
Inject the reference solution. The test is not valid unless the
theoretical plates is not less than 2000, tailing factor is not
more than 2.0 and the relative standard deviation for replicate
injections is not more than 2.0 per cent. Telmisartan Tablets
Inject the test solution and the reference solution. In the
Telmisartan Tablets contain not less than 90.0 per cent and
chromatogram obtained with the test solution, the area of any
not more than 11 0.0 per cent of the stated amount of
secondary peak is not more than 2 times the area ofthe principal
telmisartan, C33H30N40z.
peak in the chromatogram obtained with the reference solution
(0.2 per cent). The sum of areas of all the secondary peaks is Usual strengths. 20 mg; 40 mg; 80 mg.
not more than 10 times the area of the principal peak in the
chromatogram obtained with the reference solution (1.0 per Identification
cent). Ignore any peak with an area less than 0.5 times thearea In the Assay, the retention time of the principal peak in the
of the principal peak in the chromatogram obtained with the chromatogram obtained with the test solution corresponds to
reference solution (0.05 per cent). the peak in the chromatogram obtained with the reference
Heavy metals (2.3.13). 1.0 g complies with the limit test for solution.
heavy metals, Method B (20 ppm).
Tests
Sulphated ash (2.3.18). Not more than 0.1 percent.
Dissolution (2.5.2).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1 g by drying in oven at 105°. Apparatus No.1,
Medium. 900 ml of 0.1 M hydrochloric acid,
Assay. Determine by liquid chromatography (2.4.14).
Speed and time. 75 rpm and 60 minutes.
Solvent mixture. 80 volumes of buffer solution prepared by Determine by liquid chromatography (2.4.14), as described
diluting 5.0 ml of triethylamine to 2000 ml with water and 20 under Assay using the following solutions;
volumes of methanol.
Solvent mixture. 80 volumes of buffer solution prepared by
Test solution. Dissolve 40 mg ofsubstance under examination diluting 5.0 ml of triethylamine to 2000 ml with water and 20
in 100.0 ml ofthe solvent mixture. Dilute 5.0 ml ofthe solution volumes of methanol.
to 50.0 ml with the solvent mixture.
Test solution. Use the filtrate, dilute if necessary with the
Reference solution. A 0.004 per cent w/v solution of dissolution medium.
tebnisartan RS in the solvent mixture.
Reference solution. A 0.044 per cent w/v solution of
Chromatographic system telmisartan RS in the solvent mixture. Dilute 5,0 ml of this
a stainless steel column 15 cm x 4.6 mm, packed with solution to 100.0 ml with the dissolution medium.
octadecylsilane bonded to porous silica (5 /-lm) (such
as Inertsil ODS-3), D. Not less than 70 per cent of the stated amount of
- mobile phase: a mixture of60 volumes ofbuffer solution C33H30N40Z.
prepared by dissolving 2.72 g ofpotassium dihydrogen Related substances. Determine by liquid chromatography
phosphate in 1000 ml ofwater; add 2 ml of triethylamine (2.4.14).

2187

www.webofpharma.com
TELMISARTAN TABLETS IP 2010

Solvent mixture. 80 volumes of buffer solution prepared by Solvent mixture. 80 volumes of buffer solution prepared by
diluting 5.0 ml of triethylamine to 2000ml with water and 20 diluting 5.0 ml of triethylamine to 2000 ml with water and 20
volumes of methanol. volumes of methanol.
Test solution. Weigh and powder 20 tablets. Disperse a quantity Test solution. Shake 5 intact tablets with 100 ml ofthe solvent
ofpowder containing about 100 mg ofTelmisartan in 100.0 ml mixture and sonicate for 45 minutes. Dilute to 250 ml with the
ofsolvent mixture, sonicate for 45 minutes and filter. solvent mixture and filter. Further dilute to obtain 0.004 per
cent w/v solution with the solvent mixture.
Reference solution. A 0.0005 per cent w/v solution of
telmisartan RS in the solvent mixture. lleference solution. A 0.004 per cent w/v solution of
telmisartan RS in the solvent mixture.
Chromatographic system
a stainless steel column l5cm x 4.6 mm, packed with Chromatographic system
octadecylsilane bonded to porous silica (5 /lm) (suc4 as - a stainless steel column 15 cm x 4.6 mm, packed with
InertsilODS-3), octadecylsilane bonded to porous silica (5 /lm) (such
mobile phase: A. dissolve 0.5 g of potassium as Inertsil ODS-3),
dihydrogen phosphate in 1000 ml of water; add 2 ml of - mobile phase: a mixture of60 volumes ofbuffer solution
triethylamine, adjusted to pH 3.2 with orthophosphoric prepared by dissolving 2.72 g ofpotassium dihydrogen
acid, phosphate in 1000 ml of water; add 2 ml of triethylamine
B. acetonitrile, and adjust the pH to 2.4 with orthophosphoric acid
a linear gradient programme using the conditions given and 40 volumes of acetonitrile,
below, flow rate. 1 ml per minute,
flow rate. 1.8 ml per minute, - spectrophotometer set at 298 nIn,
spectrophotometer set at 298 nIn, - injection volume. 20 Ill.
injection volume. 20 Ill. Inject the reference solution. The test is not valid unless the
Time Mobile phase A Mobile phase B theoretical plates is not less than 3000, the tailing factor is not
(min.) (per cent v/v) (per cent v/v) more than 2.0 and the relative standard deviation for replicate
o 78 22 injections is not more than 2.0 percent.

6 80 20 Inject the test solution and the reference solution.


7 70 30 Calculate the content of C33H30N40z in the tablets.
15 60 40
25 60 40 Tenofovir Disoproxil Fumarate
26 40 60
35 20 80
40 78 22
Inject the reference solution. The test is not valid unless the
HXGaaH
theoretical plates is not less than 3000, tailing factor is not , I
more than 2.0 and relative standard deviation for replicate HaaG H
injections is not more than 5.0 per cent.
Inject the test solution and the reference solution. In the
chromatogram obtained with the test solution the area of any Mol. Wt. 635.5
secondary peak is not more than the area ofthe principal peak Tenofovir Disoproxil Fumarate salt ofbis(isopropyloxy-
in the chromatogram obtained with the reference solution (0.5 carbonyloxymethyl ester of (R)-9-(2-phosphonomethoxy-
per cent), the sum ofthe area of all the secondary peaks is not propyl)adenine with fumaric acid.
more than 4 times the area of the principal peak in the
Tenofovir Disoproxil Fumarate contain not less than 97.0 per
chromatogram obtained with reference solution (2.0 per cent).
Ignore any peak with an area less than 0.1 times the area ofthe cent and not more than 102. 0 per cent ofCI9H30N501OP,C4~04,
c!ll~lll_at~~ on tl1eanb:ydr()us basis.
-principal peakinthe chromatogram obtained with the reference
solution (0.05 per cent). Category. Antiviral.
Other tests. Comply with the tests stated under Tablets. Dose. 300 mg daily.
Assay. Determine by liquid chromatography (2.4.14). Description. A white to off- white crystalline powder.

2188

www.webofpharma.com
IP 2010 TENOFOVIR DISOPROXIL FUMARATE

Identification Inject the test solution, reference solution (b) and reference
solution (c). In the chromatogram obtained with the test
A. Detennine by infrared absorption spectrophotometry (2.4.6). solution, the area of any secondary peak is not more than
Compare the spectrum with that obtained with tenofovir the area of the peak in the chromatogram obtained with the
disoproxil jitmarate. RS or with the reference spectrum of reference solution (b) (1.0 per cent) and the sum of all the
tenofovir disoproxil fumarate. secondary peaks is not more than 2.5 times the' area of the
B. In the Assay, the principal peak in the chromatogram peak in the chromatogram obtained with the reference solution
obtained with the test solution corresponds to the peak in the (b) (2.5 per cent). Ignore any peak corresponding to the peak
chromatogram obtained with the reference solution. obtained in the chromatogram in the reference solution (c)
and any peak having area less than 0.02 times the area of the
Tests principal peak in the chromatogram obtained with reference
solution (b) (0.02 per cent).
Related substances. Detennine by liquid chromatography
(2.4.14). Fumaric Acid. 17.5 per cent to 19.0 percent.

NOTE - Prepare the solutions immediately before use. Detennine by liquid chromatography (2.4.14).

Test solution. Dissolve 100 mg of the substance under Test solution. Dissolve 25 mg of the substance under
examination in 50 ml of methanol. examination in 100 ml ofthe mobile phase.

Reference solution (a). A 0.2 per cent w/v solution of tenofovir Reference solution. Dissolve 25 mg of the jitmaric acid in
disoproxil jitmarate RS in methanol. 50 ml of the mobile phase. Dilute 10 ml of the solution to
100 ml with the mobile phase.
Reference solution (b). Dilute 1.0 ml ofreference solution (a)
to 100.0 ml with methanol. Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with
Reference solution (c). Dissolve 10 mg ofthejUmaric acid in octadecylsilane bonded to porous silica (5 Ilm),
50 ml of methanol. - mobile phase: a mixture of 40 volumes of acetonitrile
Chromatographic system and 60 volumes ofa buffer solution prepared by adding
a stainless steel column 25 cm x 4.6 mm, packed with 0.1 per cent v/v of triethylamine in D.D5M sodium
octadecylsilane bonded to porous silica (5 Ilm) (Such dihydrogen phosphate with the pH adjusted to 2.3 with
asODS3V), orthophosphoric acid imd filtered,
column temperature. 30°, flow rate. 1 ml per minute,
mobile phase: A. D.1 M ammonium acetate solution spectrophotometer set at 210 nm,
with the pH adjusted to 3.8 with glacial acetic acid, injection volume. 10 Ill.
B. a mixture of 70 volumes of methanol Inject the reference solution. The test is not valid unless the
and 30 volumes of acetonitrile, tailing factor is not more than 2.0, the column efficiency in not
flow rate. 1.5 ml per minute, less than 2000 theoretical plates and the relative standard
a linear gradient programme using the conditions given deviation for replicate injections is not more than 2.0 per cent.
below,
spectrophotometer set at 260 nm, Inject the test solution and the reference solution.
injection volume. 10 Ill. Calculate the content of fumaric acid.
Time mobile phase A mobile phase B
Water (2.3.43). Not more than 1.0 per cent, detennined on
(in min.) (per cent v/v) (per cent v/v)
1.0g.
o 95 5
Assay. Detennine by liquid chromatography (2.4.14).
10 50 50
NOTE - Prepare the solutions immediately before use and
25 50 50
carry out the test protectedfrom light.
50 20 80
Test solution. Dissolve 50 mg of the substance under
60 95 5
examination in 50.0 ml ofthe mobile phase. Dilute 5.0 ml ofthe
65 95 5 solution to 50.0 ml with the mobile phase.
Inject reference solution (a). The test is not valid unless the Reference solution. A 0.1 per cent w/v solution of tenofovir
tailing factor is not more than 2.0 and the column efficiency in disoproxiljillnarate RS in the mobile phase. Dilute 5.0 ml of
not less than 2000 theoretical plates. the solution to 50.0 ml with the mobile phase.

2189

www.webofpharma.com
TENOFOVIR DISOPROXIL FUMARATE TABLETS IF 2010

Chromatographic system Related substances. Determine by liquid chromatography


a stainless steel column 25 cm x 4.6 mm, packed with (2.4.14).
octadecylsilane bonded to porous silica (5 /lm),
NOTE - Prepare the solutions immediately before use.
mobile phase: a mixture of 40 volumes of acetonitrile
and 60 volumes ofa buffer solution prepared by adding Test solution. Weigh and powder 20 tablets. Weigh accurately
1 ml trieti1ylamine to 0.05M sodium dihydrogen a quantity of the powder containing 100 mg of Tenofovir
phosphate with the pH adjusted to 2.3 with ortho- Disoproxil Fumarate and disperse in 50 ml.of methanol and
phosphoric acid and :filtered, filter.
- flow rate. 1 ml per minute, Reference solution (a). A solution of tenofovir disoproxil
spectrophotometer set at 260 nm, fumarate RS containing 0.2 per cent of tenofovir disoproxil in
injection volume. 20 /ll. methanol.
Inject the reference solution. The test is not valid unless the
Reference solution (b). Dilute 1 ml ofreference solution (a) to
tailing factor is not more than 2.0, the column efficiency in not
100 ml with methanol.
less than 2000 theoretical plates and the relative standard
deviation for replicate injections is not more than 2.0 per cent. Reference solution (c). Dissolve 10 mg of fumaric acid in
50 ml of methanol.
Inject the test solution and the reference solution.
Chromatographic system
Calculate the content ofCI9H30NsOIOP,C4H404. a stainless steel column 25 cm x 4.6 mm, packed with
Storage. Store protected from light in a refrigerator (2° to 8°). octadecylsilane bonded to porous silica (5 /lm),
mobile phase: A. dissolve 1.9 g of ammonium acetate
in 1000 ml of water and adjust the pH to 3.8 with glacial
acetic acid,
Tenofovir Disoproxil Fumarate Tablets B. methanol,
Tenofovir Disoproxil Fumarate Tablets contain not less than - flow rate. 1.5 ml per minute,
90.0 per cent and not more than 110.0 per cent of the stated a linear gradient programme using the conditions given
below,
amount oftenofovir disoproxil fumarate, CI9H30NsOIOP,C4H404.
spectrophotometer set at 260 nm,
Usualstrength. 300 mg. injection volume. 10 /ll.

Identification Time mobile phase A mobile phase B


(in min.) (per cent v/v) (per cent v/v)
A. In the Assay, the principal peak in the chromatogram o 95 5
obtained with the test solution corresponds to the peak in the 10 50 50
chromatogram obtained with the reference solution.
25 50 50
B. When examined in the range 200 nm to 400 nm (2.4.7), a 50 20 80
0.001 per cent w/v solution in methanol shows an absorption
60 95 5
maximum at the same wavelength as the reference solution.
Inject reference solution (a). The test is not valid unless the
Tests tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates.
Dissolution (2.5.2).
Inject the test solution, reference solutions (b) and (c). In the
Apparatus No.1,
chromatogram obtained with the test solution, the area of any
Medium. 900 ml of 0.1 M hydrochloric acid, secondary peak is not more than 3.5 times the area ofthe peak
Speed and time. 50 rpm and 45 minutes in the chromatogram obtained with the reference solution (b)
Withdraw a suitable volume ofthe medium and :filter promptly. (3.5 per cent) and the sum of areas of all the secondary peaks
Dilute the filtrate, ifnecessary, with the same solvent. Measure is not more than 6 times the area of the peak in the
the absorbance (2.4.7) ofthe resulting solution at the maximum chromatogram obtained with the reference solution (b)
at about 260.nm. Calculate the content ofC19H30NsOIOP,C4:I-404 (6.0 per cent). Ignore the peak corresponding to the peak in
in the mediiim from the absorbance obtained fromasoliition the chromatogram obtained in the reference solution (c).
oflmown concentration of tefzofovir dis6pt6xilfutJ1atate RS. Other tests. Comply with the tests stated under Tablets.
D. Not less than 80 per cent of the stated amount of Water (2.3.43). Not more than 5.0 per cent, determined on
CI9H30NsOIOP,C4:I-404' 0.5g.

2190

www.webofpharma.com
IP 2010 TENOFOVIR AND EMTRICITABINE TABLETS

Assay. Detennine by liquid chromatography (2.4.14). Tests


NOTE - Prepare the solutions immediately before use. Dissolution (2.5.2).
Solvent mixture. Equal volumes of water and methanol. Apparatus No.1
Test solution. Weigh and powder 20 tablets. Weigh accurately Medium. 900 ml of 0.1 M hydrochloric acid,
a quantity of the powder containing 300 mg of Tenofovir Speed and time. 50 rpm and 45 minutes.
Disoproxil Fumarate, dissolve in 100 ml ofsolvent mixture and
Withdraw a suitable volume ofthe medium and filter.
filter. Dilute 5.0 ml of the filtrate to 50.0 ml with the mobile
phase. Determine by liquid chromatography (2.4.14)
Reference solution. A 0.120 per cent w/v solution of tenofovir Test solution. The filtrate obtained as given above. Dilute the
disoproxilfitmarate RS in the solvent mixture. Dilute 5.0 ml of filtrate if necessary, with the dissolution medium.
the solution to 20.0 ml with the mobile phase.
Reference solution. Asolution containing 0.32 per cent w/v of
Chromatographic system tenofovir disoproxil fillnarate RS and 0.24 per cent w/v of
a stainless steel column 15 cm x 4.6 mm, packed with emtricitabine RS in methanol. Dilute 5 ml of the solution to
octadecylsilane bonded to porous silica (5 Ilm) (Such 50 ml with the dissolution medium.
as Inertsil ODS 3V),
Use the chromatographic system given under Assay.
mobile phase: a mixture of60 volumes ofa buffer solution
prepared by dissolving 7.8 g of sodium dihydrogen Inject the test solution and the reference solution.
orthophosphate in 1000 ml of water, adding 1 ml of
Calculate the contents of CI9H30NsOIOP,C4H404 and
triethylamine and adjusting the pH to 2.3 with
CgH IOFN30 3S.
orthophosphoric acid (1 0 per cent v/v), and 40 volumes
of acetonitrile, D. Not less than 75 per cent of the stated amounts of
flow rate. 1 m1 per minute, C19H30NsOIOP.C~04and CgH IOFN30 3S.
spectrophotometer set at 260 llill,
Related substances. Detennine by liquid chromatography
injection volume. 10 Ill.
(2.4.14).
Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0, the column efficiency in not For Emtricitabine
less than 2000 theoretical plates and the relative standard Test solution. Weigh and powder 20 tablets. Weigh accurately
deviation for replicate injections is not more than 2.0 per cent. a quantity of the powder containing 50 mg of Emtricitabine,
Inject the test solution and the reference solution. disperse in 5 ml of methanol, dilute to 50 ml with mobile phase
A and filter.
Calculate the content OfC19H30NsOIOP,C4H404in the tablets.
Reference solution (a). A 0.1 per cent w/v solution of
Storage. Store protected from moisture, at a temperature not emtricitabine RS in mobile phase A.
exceeding 30°.
Reference solution (b). Dilute 1 ml ofreference solution (a) to
100 ml with mobile phase A.
Chromatographic system,
Tenofovir and Emtricitabine Tablets a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5Ilm),
Tenofovir Disoproxil Fumarate and Emtricitabine Tablets column temperature. 35°,
Tenofovir and Emtricitabine Tablets contain not less than mobile phase: A. a mixture of 95 volumes of a buffer
90.0 per cent and not more than 110.0 per cent of the stated solution prepared by dissolving 1.9 g of ammonium
amounts oftenofovir disoproxil fumarate, CI9H30NsOIOP,C4~04 acetate in 1000 ml of water, adjusted the pH to 3.8 with
and emtricitabine, CgHIO FN30 3S. glacial acetic acid, and 5 volumes of methanol,
B. a filtered mixture oBO volumes ofthe
Usual strength. Tenofovir 300 mg and Emtricitabine 200 mg.
buffer solution and 70 volumes of methanol,
Identification - flow rate. 1 ml per minute,
a linear gradient programme using the conditions given
In the Assay, the principal peak in the chromatogram obtained below,
with the test solution corresponds to the peak in the spectrophotometer set at 277 nm,
chromatogram obtained with the reference solution. injection volume. 20 Ill.

2191

www.webofpharma.com
TENOFOVIR AND EMTRICITABINE TABLETS IP 2010

Time Mobile phase A Mobile phase B Inject reference solution (a). The test is not valid unless the
(in min.) (per cent v/v) (per cent v/v) column efficiency is not less than 2000 theoretical plates and
o 100 o the tailing factor is not more than 2.0.
30 100 o Inject the test solution, reference solutions (b) and (c). In the
35 o 100 chromatogram obtained with the test solution, the area of any
55 o 100 secondary peak is not more than 3.5 times the area ofthe peak
60 100 o in the chromatogram obtained with the reference solution (b)
75 100 o (3.5 per cent) and the sum of areas of all the secondary peaks
is not more than 6 times the area of the peak in the
Inject reference solution (a). The test is not valid unless the
chromatogram obtained with the reference solution (b)
column efficiency is not less than 500 theoretical plates and
(6.0 per cent). Ignore the peak corresponding to the peak in
the tailing factor is not more than 2.0.
the chromatogram obtained in the reference solution (c).
Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any Other tests. Comply with the tests stated under the Tablets.
secondary peak is not more than the area of the peak in the Assay. Determine by liquid chromatography (2.4.14).
chromatogram obtained with the reference solution (b)
(1.0 per cent) and the sum of areas of all the secondary peaks Solvent mixture. 30 volumes of water and 70 volumes of
is not more than 3 times the area of the peak in the methanol.
chromatogram obtained with the reference solution (b) Test solution. Weigh and powder 20 tablets. Weigh accurately
(3.0 per cent). a quantity of the powder containing 100 mg of Tenofovir
For Tenofovir Disoproxil Fumarate Disoproxil Fumarate, disperse in 10 ml of water and dilute to
Test solution. Weigh and powder 20 tablets. Weigh accurately 250.0 ml with methanol. Dilute 5.0 ml ofthe solution to 25.0 ml
a quantity of the powder containing 50 mg of Tenofovir with the solvent mixture.
Disoproxil Fumarate and disperse in 50 ml of methanol. Reference solution. A solution containing 0.025 per cent w/v
Reference solution (a). A 0.1 per cent w/v solution of tenofovir of emtricitabine RS and 0.04 per cent w/v of tenofovir
disoproxil fumarate RS in methanol. disoproxil ji/marate RS in methanol. Dilute 5.0 ml of the
Reference solution (b). Dilute 1 ml ofreference solution (a) to solution to 25.0 rill with the solvent mixture.
100 ml with methanol. Chromatographic system
Reference solution (c). Dissolve 10 mg of ji/maric acid in 50 - a stainless steel column 4.6 mm ~ 5 cm packed with
ml of methanol. octylsilane bonded to porous silica (3 /lm),
Chromatographic system column temperature 40°,
a stainless steel column 25 cm x 4.6 mm packed with mobile phase: A. a buffer solution prepared by
octadecylsilane bonded to porous silica (5/lm), dissolving 1.35 g of monobasic potassium phosphate
mobile phase: A. a buffer solution prepared by in 1000 ml of water and adjusting the pH to 3.0 with
dissolving 1.9 g of ammonium acetate in 1000 ml of orthophosphoric acid,
water and adjusting the pH to 3.8 with glacial acetic B. a mixture of20 volumes ofthe buffer
acid, solution and 80 volumes of acetonitrile,
B. methanol, flow rate. 1.5 ml per minute,
flow rate. 1.5 ml per minute, a linear gradient programme using the conditions given
- a linesr gradient programme using the conditions given below,
below, spectrophotometer set at 260 nm,
spectrophotometer set at 260 nm, injection volume. 10 Ill.
injection volume. 20 Ill. Time Mobile phase A Mobile phase B
Time Mobile phase A Mobile phase B (in mins) (per cent) (per cent)
(in mins) (per cent) (per cent) o 94 6
o 95 5 3 94 6
5 50 -- -- -50--~------
25 50. 50
8 35 65
50 20 80
60 95 5 9 94 6
75 95 5 12 94 6

2192

www.webofpharma.com
IP 2010 TEMOZOLOMIDE CAPSULES

Inject the reference solution. The test is not valid unless the Chromatographic system
tailing factor is not more than 2.0, the column efficiency in not a stainless steel column 25 cm x 4.6 mm, packed with
less than 2000 theoretical plates for the peak due to tenofovir octadecylsilane bonded to porous silica (5 Ilm), (Such
disoproxil, 500 theoretical plates for the peak due to as Hypersil BDS),
emtricitabine and the relative standard deviation for replicate - mobile phase: a mixhJre of90 volumes of 0.5 per cent
injections is not more than 2.0 per cent for each component. w/v solution of acetic acid and 10 volumes of
Inject the test solution and the reference solution.
acetronitrile,
flow rate. 1 ml per minute,
Calculate the contents of C19H30NsOIOP,C4H404 and - spectrophotometer set at 329 nm,
CgHIQFN30 3S in the tablets. - injection volume. 20 Ill.
Storage. Store protected from moisture, at a temperahlre not Inject the reference solution. The test is not valid unless the
exceeding 30°. theoritical plates is not less than 2000 and the tailing factor is
not more than 2.0.
Temozolomide Inject the test solution and the reference solution. In the
chromatogram obtained with the test solution, area of any
secondary peak is not more than 0.5 times the area of the
principal peak in the chromatogram obtained with the reference
solution (0.5 per cent). The sum of areas of all the secondary
peaks is not more than the area of the principal peak in the
chromatogram obtained with the reference solution (1.0 per
cent). Ignore any peak with an area less than 0.1 times the
area of the peak in the chromatogram obtained with the
reference solution (0.1 per cent).
Mol. Wt. 194.2
Heavy metals (2.3.13). 1.0 gm complies with the limit test for
Temozolornide is 3,4-dihydro-3-methyl-4-oxoimidazo[5, I-d]- heavy metals, Method A (20 ppm).
1,2,3,5-tetrazine-8-carboxarnide.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Temozolomide contains not less than 98.0 per cent and not
more than 102.0 per cent ofC 6H 6N 60 2, calculated on the dried Loss on drying (2.4.19). Not more than 0.5 per cent, determined
basis. on 1.0 gm by drying in an oven at 105° for 3 hours.
Category. Anticancer. Assay. Determine by liquid chromatography (2.4.14).
Dose. 75 mg daily. Test solution. Dissolve 20 mg of the substance under
examination in 100.0 ml ofthe mobile phase.
Description. A white to almost white powder.
Reference solution. A 0.02 per cent w/v solution of
CA UTION- Temozolomide is cytotoxic; extra care required
temozolomide RS in the mobile phase.
to prevent inhaling particles and exposing the skin to it.
Use the chromatographic system as described under Related
Identification substances.
A. Determine by infrared absorption spectrophotometry (2.4.6). Inject the reference solution. The test is not valid unless the
Compare the spectrum with that obtained with temozolomide relative standard deviation for replicate injections is not more
RS or with the reference spectrum oftemozolomide. than 2.0 per cent.
B. In the Assay, the principal peak in the chromatogram Inject the test solution and the reference solution.
obtained with test solution (b) corresponds to the peak in the
Calculate the content ofC6H 6N 60 2 •
chromatogram obtained with reference solution (a).
Storage. Store protected from light and moisture, at a
Tests temperature not exceeding 30°.
Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Dissolve 100 mg of the substance under Temozolomide Capsules
examination in 100.0 ml ofthe mobile phase. Temozolomide Capsules contain not less than 90.0 per cent
Reference solution. Dilute 1.0 ml of the test solution to and not more than 110.0 per cent of the stated amount of
100.0 ml with the mobile phase. temozolornide, C6H6N 60 Z .

2193

www.webofpharma.com
TEMOZOLOMIDE CAPSULES IP 2010

CAUTION -Temozolomide is cytotoxic; extra care required Terazosin Hydrochloride contains not less than 99.0 per cent
to prevent inhaling particles and exposing the sIan to it. and not more than 101.0 per cent ofCJ9H2sNs04,HCI, calculated
on the anhydrous basis.
Usual strengths. 20 mg; 100 mg; 250 mg.
Category. Antihypertensive.
Identification
Description. A white or slightly yellow, crystalline powder.
In the Assay, the principal peak in the chromatogram obtained
Dose. 1 mg daily.
with the test solution corresponds to the principal peak in the
chromatogram obtained with reference solution. Identification
Other tests. Complies with tests stated under capsules. A. Determine by infrared absorption spectrophotometry (2.4.6).
Assay. Determine by liquid chromatography (2.4.14). Compare the spectrum with that obtained with terazosin
hydrochloride RS or with the reference spectrum ofterazosin
Test solution. Weigh accurately a quantity of mixed contents hydrochloride.
of20 capsules containing 20 mg ofTemozolomide in 100.0 ml
of mobile phase.
B. Gives reaction A ofchlorides (2.3.1).

Reference solution. A 0.02 per cent w/v solution of Tests


temozolomide RS in the mobile phase. Appearance ofsolution. A 1.0 per cent w/v solution in carbon-
Chromatographic system dioxide free water is clear (2.4.1) and not more intensely
a stainless steel column 25 cm x 4.6 mm, packed with coloured than reference solution YS7 (2.4.1).
octadecylsilane bonded to porous silica (5 ).tm), (such pH (2.4.24). 3.0 to 5.0, determined in 2.0 per cent w/v solution
as Hypersil BDS), in carbon-dioxide free water.
- mobile phase: a mixture of 90 volumes of 0.5 per cent
Impurities Nand O. Determine by liquid chromatography
v/v acetic acid and 10 ml volumes of acetonitrile,
(2.4.14).
flow rate. 1 ml per minute,
spectrophotometer set at 329 nm, Solvent mixture. 20 volumes of acetonitrile and 80 volumes
injection volume. 20 ).tl. of water.
Test solution. Dissolve 50 mg of the substance under
Inject the reference solution. The test is not valid unless the
examination in 50 ml ofthe solvent mixture.
tailing factor is not more than 2.0, the column efficiency is not
less than 2000 theoretical plates and the relative standard Reference solution (a). Dissolve 5 mg each of 2-chloro-6, 7-
deviation for replicate injections is not more than 2.0 per cent. dimethoxyquinazolin-4-amine RS (terazosin impurity A RS)
and 1-[[(2RS)-tetrahydrojilran-2-yl]carbonyl]piperazine RS
Inject the test solution and the reference solution. (terazosin impurity N RS) in acetonitrile, add 5 ml of the test
Calculate the content of C6H6N602 in the capsules. solution and dilute to 50 ml with acetonitrile. Dilute 10 mlof
this solution to 100 ml with the solvent mixture.
Storage. Store protected from light and moisture.
Reference solution (b). Dilute 10 ml of reference solution (a)
to 100 ml with the solvent mixture.

Terazosin Hydrochloride Chromatographic system


a stainless steel column 25 cm x 4.0 mm, packed with
octadecylsilane bonded to porous silica (5 ).tm),
- mobile phase: dissolve 2.8 g of sodium laurylsulphate
in 1000 mlof water and add 11 ml ofa solution containing
20.24 per cent w/v of triethylamine and 23.0 per cent
w/v of orthophosphoric acid and adjust the pH to 2.5
with orthophosphoric acid; mix 60 volumes of this
solution with 40 volumes of acetonitrile,
flow rate. 1.0 ml per minute,
sp~ctrophotometer set at 210 nIll,
- -----·-Morwc459~9
injection volume. 20 ).tl.
Terazosin Hydrochloride is (RS)-6, 7-dimethoxy-2-[4- Inject reference solution (b). The relative retention time with
(tetrahydrofuran-2-carbonyl)piperazin-l-yl} quinazolin-4- reference to terazosin for I,4-bis[(tetrahydrofuran-2-
ylamine hydrochloride dihydrate. yl)carbonyl]piperazine (terazosin impurity 0) is about 0.2, for

2194

www.webofpharma.com
IP 2010 TERBUTALINE SULPHATE

terazosin impurity N is about 0.3 and for terazosin impurity A Inject the test solution, reference solution (a) and (c). Run the
is about 0.4. The test is not valid unless the resolution between chromatogram 4 times the retention time ofthe principal peak.
the peaks due to terazosin impurity A and N is not less than In the chromatogram obtained with the test solution the area
1.5. of secondary peak corresponding to terazosin impurity A, C,
Inject the test solution and reference solution (b). Run the E and K is not more than 5 times the area ofthe principal peak
chromatogram 4 times the retention time ofthe principal peak. in the chromatogram obtained with reference solution (a) (0.5
In the chromatogram obtained with the test solution the area per cent). The area of secondary peak corresponding to
of each peak corresponding to terazosin impurity Nand terazosin impurity L is not more than the area of the
terazosin impurity 0 is not more than the area ofthe peak due corresponding peak in the chromatogram obtained with
to terazosin in the chromatogram obtained with reference reference solution (c) (0.1 per cent). The area of secondary
solution (b) (0.1 per cent). peak corresponding to terazosin impurity B, J, M is not more
than the area of the principal peak in the chromatogram
Related substances. Determine by liquid chromatography obtained with reference solution (a) (0.1 per cent). Multiply
(2.4.14).
the peak areas of the impuirites by the correction factor for
Test solution. Dissolve 50 mg of the substance under calculating the contents, for impuirty C is 0.7 and for impuirty
examination in 100 ml ofthe mobile phase. M is 1.6. The area of any other secondary peak is not more
Reference solution (a). Dilute 2.0 ml ofthe test solution to 100 than the area of the principal peak in the chromatogram
ml with the mobile phase. Dilute 5.0 ml ofthissolution to 100 obtained with reference solution (a) (0.1 per cent); The sum of
ml with the mobile phase. the areas ofall other secondary peaks is not more than 5 times
the area of the principal peak in the chromatogram obtained
Reference solution (b). Dissolve the contents of a vial of with reference solution (a) (0.5 per cent). Ignore any peak with
terazosin for system suitability RS (containing terazosin an area less than 0.5 times the area ofthe principal peak in the
impurity A, terazosin impurity B (1-(4-hydroxy-6, 7- chromatogram obtained with reference solution (a) (0.05 per
dimethoxyquinazolin-2-yl)-4-[[(2RS)- tetrahydrofuran-2- cent).
yl]carbonyl]piperazine), terazosin impurity C (6,7-dimethoxy-
2-(piperazin-l-yl)quinazolin-4-amine), terazosin impurity J (1- Heavy metals (2.3.13). 1.0 g complies with the limit test for
(4-amino-6, 7-dimethoxyquinazolin-2-yl)-4-[(2RS)-2- heavy metals, Method B (20 ppm).
hydroxypentanoyl]piperazine), terazosin impurity K (prazosin) Sulphated ash (2.3.18). Not more than 0.1 per cent.
and terazosin impurity M (1,4-bis(furan-2"ylcarbonyl) Water (2.3.43).7.8 to 8.6 per cent, determined on 0.2 g.
piperazine) in 10 ml ofthe mobile phase.
Assay. Weigh accurately about 0.3 g , dissolve in a mixture of
Reference solution (c). Dissolve 5 mg of 1-(furan-2- 5 ml of 0.01 M hydrochloric acid and 50 ml of methanol.
ylcarbonyl)piperazine RS (terazosin impurity L RS) in 100 Titrate with 0.1 M sodium hydroxide, determining the end-
ml ofthe mobile phase. Dilute 1.0 ml ofthis solution to 100 ml point potentiometrically (2.4.25). Carry out a blank titration.
with the mobile phase.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.04239 g of
Reference solution (d). To 5 mg of 2,2-(piperazine-1,4-
CI9Hz6ClNS04.
diyl)bis(6,7-dimethoxyquinazolin-4-amine) RS (terazosin
Storage. Store protected from light.
impurity E RS ) add 70 ml of methanol and 30 ml of water.
Allow to stand for at least 1 hour to dissolve the substance.
Chrmatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with Terbutaline Sulphate
octylsilane bonded to porous silica (5 /lm),
- column temperature. 30°,
mobile phase: mix 2 volumes of triethylamine, 350
volumes of acetonitrile, and 1650 volumes ofa solution
containing 0.6 per cent w/v of sodium citrate and 1.425
per cent w/v of anhydrous citric acid,
- flow rate. 1 ml per minute,
- spectrophotometer set at 245 nm,
2
- injection volume. 20 Ill.
Inject reference solution (b). The test is not valid unless the (ClzHI9N03)z,HzS04 Mol. Wt. 548.7
resolution between the peaks due to terazosin impurity Band Terbutaline Sulphate is (RS)-2-(tert-butylamino)-
terazosin impurity J is not less than 1.5. 1-(3,5-dihydroxyphenyl)ethanol sulphate.

2195

www.webofpharma.com
TERBUTALINE SULPHATE IP 2010

Terbutaline Sulphate contains not less than 98.0 per cent and RS (terbutaline impurity C RS) and 22.5 mg of terbutaline
not more than 101.0 per cent of(C 12H I9N03h H 2S04 , calculated sulphate RS in 50.0 ml of the mobile phase. Dilute 1.0 ml of
on the dried basis. this solution to 100.0 ml with the mobile phase.
Category. Beta-adrenoceptor agonist. Reference solution (b). Dilute 1.0 ml of the test solution to'
50.0 ml with the mobile phase. Dilute 2.0 ml ofthis solution to
Dose. Orally, upto 15 mg daily, in divided doses; by inhalation,
20.0 ml with the mobile phase.
for an adult, one to two inhalations, each of 250 )..Lg, 3 or 4
times a day up to a maximum of24 inhalations in 24 hours; for Chromatographic system
a child, one to two inhalations, each of250 )..Lg, 2 or 3 times a - a stainless steel column 15 cm x 4.6 mm, packed with
day up to a maximum of 10 inhalations in 24 hours; by endcapped octadecylsilane bonded to porous silica (5
subcutaneous injection, 250 to 500 )..Lg, 4 times a day. )..Lm),
- mobile phase. dissolve 4.23 g of sodium
Description. A white or almost white, clystalline powder;
hexanesulphonate in 770 ml of 0.05 M ammonium
odourless or almost odourless.
formate solution prepared by dissolving 3.15 g of
Identification ammonium formate in about 980 ml of water, adjusted
to pH 3.0 by anhydrousformic acid and dilute to 1000
Test A may be omitted iftests B, C and D are carried out. Tests
ml with water and add 230 ml of methanol,
Band C may be omitted if tests A and D are carried out.
- flow rate. 1 ml per minute,
A. Determine by infrared absorption spectrophotometry (2.4.6). spectrophotometer set at 276 nm,
Compare the spectrum with that obtained with terbutaline - injection volume. 20 )..Ll.
sulphate RS or with the reference spectrum of terbutaline
Inject reference solution (a). The test is not valid unless the
sulphate.
resolution between the peaks due to terbutaline impurity C
B. When examined in the range 230 nrn to 360 nrn (2.4.7), a and terbutaline is not less than 2.0.
0.007 per cent w/v solution in 0.1 M hydrochloric acid shows Inject the test solution, reference solution (a) and (b). Run the
absorption maxima at about 276 nm and 280 nrn, which may be
chromatogram 6 times the retention time ofthe principal peak.
fused; absorbance at both 276 nrn and 280 nrn, 0.46 to 0.49.
In the chromatogram obtained with the test solution, the area
C. In the test for Related substances, the principal spot in the of the peak due to terbutaline impurity C is not more than
chromatogram obtained with test solution (b) corresponds to twice area of the corresponding peak in the chromatogram
that in the chromatogram obtained with reference solution (b). obtained with reference solution (a) (0.2 per cent), the area of
each peak dueto 3,5-dihydroxybenzoic acid (a-resorcylic acid)
D. A 2.0 per cent w/v solution in carbon dioxide-free water
(terbutaline impurity A), (4RS)-2-(l, I-dimethylethyl)-1,2,3,4-
gives reaction A of sulphates (2.3.1).
tetrahydroisoquinoline-4,6,8-triol (terbutaline impurity B) and
Tests 2-[benzyl-(l, I-dimethylethyl)amino]-I-(3,5-dihydroxyphenyl)
ethanone (terbutaline impurity D) is not more than area ofthe
Appearance of solution. A2.0 per cent w/v solution in carbon principal peak in the chromatogram obtained with reference
dioxide-fi-ee water is clear (2.4.1); absorbance ofa 2-cm layer
solution (b) (0.2 per cent). The sum ofall the secondary peaks
at 400 nm, not more than 0.11 (2.4.7). other than terbutaline impurity C is not more than twice the
Acidity. Dissolve 0.2 g in 10 ml of carbon dioxide-free water area ofthe principal peak in the chromatogram obtained with
and titrate with 0.01 M sodium hydroxide, using methyl red reference solution (b) (0.4 per cent). Ignore any peak with an
solution as indicator. Not more than 1.2 ml of 0.01 M sodium area less than 0.1 times the area of the principal peak in the
hydroxide is required to change the colour of the solution to chromatogram obtained with reference solution (b) (0.02 per
yellow. cent).
tert-Butylamino-3,5-dihydroxyacetophenone. Absorbance of Heavy metals (2.3.13). Mix 1.6 g with 0.6 g ofanhydrous sodium
a 2 per cent w/v solution in 0.01 M hydrochloric acid at sulphate and ignite without melting the sodium sulphate. Cool,
about 330 nm, not more than 0.50 (2.4.7). add 3 ml of 2 Mhydrochloric acid, boil and dilute to 50 ml
Related substances. Determine by liquid chromatography with water. Cool and filter, 25 ml ofthe filtrate complies with
(2.4.14). the limit test for heavy metals, Method A (25 ppm).
Tesisoliliion. Dissol've 75 illg of tIie sl.ibstance ·l.inder Lossondrying·(2.4.19);Not more than O.5percent,-determined
examination 50.0 ml ofthe mobile phase. on 1.0 g by drying in an oven at 105° for 3 hours.
Reference solution (a). Dissolve 7.5 mg of 1-(3,5- Assay. Weigh accurately about 0.5 g, dissolve in 60 ml of
dihydroxyphenyl)-2 -[(1, 1~dimethylethyl)amino] ethanone anhydrous glacial acetic acid with the aid of heat. Titrate

2196

www.webofpharma.com
IP 2010 TERBUTALINE INJECTION

with 0.1 M perchloric acid, determining the end-point Use 35 ml of water and finally dilute to 50.0 ml with water.
potentiometrically (2.4.25). Carry out a blank titration. Dilute a suitable volume ofthis solution with water to produce
a solution containing 50 Ilg ofTerbutaline Sulphate per m!. To
I ml of 0.1 M perchloric acid is equivalent to 0.05486 g of
10.0 ml ofthis solution in a 50-ml volumetric flask add 35 ml of
(CI2HI9N03)2, H2S04,
buffer solution pH 9.5 and 1.0 ml of 4-aminoantipyrine
Storage. Store protected from light and moisture. solution. Mix, add 1.0 ml of potassium jerricyanide solution
with vigorous swirlingofthe flask, dilute to volume with water
and mix. Exactly 75 seconds after the addition ofthe potassium
jerricyanide solution measure the absorbance ofthe resulting
Terbutaline Inhalation solution at the maximum at about 550 nm (2.4.7), using as the
Terbutaline Sulphate Inhalation; Terbutaline Inhalation blank a solution prepared in the same manner using 10.0 ml of
Aerosol; Terbutaline Sulphate Inhalation Aerosol water in place of the solution of the substance under
examination.
Terbutaline Inhalation is a suspension ofTerbutaline Sulphate,
as a superfine powder, in a suitable liquid in a suitable Calculate the content of (CI2HI9N03)z,H2S04 in the solution
pressurised container. It may contain suitable pharmaceutical from the absorbance obtained by repeating the operation using
aids such as surfactants, stabilising agents, etc. a solution containing 100 Ilg of terbutaline sulphate RS in
place of the solution of the substance under examination.
Terbutaline Inhalation delivers not less than 75.0 per cent and
not more than 125.0 per cent ofthe stated amount ofterbutaline Calculate the amount of (CI2HI9N03)2,H2S04 delivered per
sulphate (CI2HI9N03)2,H2S04, per inhalation, by actuation of actuation of the valve.
the valve. Determine the content of active ingredient a second and third
Usual strength. 250 Ilg in each metered dose. time by repeating the procedure on the middle ten and on the
last ten successive combined actuations of the valve. For
Identification each of the three determinations the average content of
Determine by thin-layer chromatography (2.4.17), coating the (C12H19N03)2,H2S04 delivered per actuation ofthe valve meets
plate with silica gel GF254. the requirements.

Mobile phase. A mixture of 65 volumes of 2-propanol, Storage. Store protected from moisture at a temperature not
25 volumes of cyclohexane and 5 volumes ofjormic acid. exceeding 30°.

Test solution. Remove the actuator from the pressurised Labelling. The label states the amount of active ingredient
container, shake the container for about 30 seconds and place delivered per inhalation.
it in an inverted position in a small beaker containing 5 ml of
water. Discharge a sufficient number of deliveries containing
5 mg ofTerbutaline Sulphate, under the surface ofthe solvent.
Rejerence solution. A 0.1 per cent w/v solution ofterbutaline Terbutaline Injection
sulphate RS in water. Terbutaline Sulphate Injection
Apply to the plate 2 III of each solution. After development,
Terbutaline Injection is a sterile solution of Terbutaline
dry the plate in air, spray with a 2 per cent wiv solution of
Sulphate in Water for Injections.
4-aminoantipyrine in methanol. Examine in daylight and in
ultraviolet light at 365 nm. The principal spot in the Terbutaline Injection contains not less than 90.0 per cent and
chromatogram obtained with the test solution corresponds to not more than 110.0 per cent ofthe stated amount ofterbutaline
the chromatogram obtained with reference solution. sulphate (CI2HI9N03)2, H 2S04•
Usual strength. 500 Ilg per m!.
Tests
Other tests. Complies with the tests stated under Inhalation Identification
Preparations (Pressurised metered-dose Preparations).
Determine by thin-layer chromatography (2.4. I7), coating the
Follow the procedure described under Assay wherever the
plate with silica gel G
amount of active substance is to be determined in any test.
Mobile phase. A mixture of 65 volumes of 2-propanol,
Assay. Carry out the test for Content of active ingredient
25 volumes of cyclohexane and 5 volumes ofjormic acid.
delivered per actuation stated under Inhalation Preparations
(Pressurised metered-dose Preparations). Test solution. Use the injection.

2197

www.webofpharma.com
TERBUTALINE INJECTION IP 2010

Reference solution. A 0.1 per cent w/v solution of terbutaline for 10 minutes, dilute to 100 ml with 0.1 M sodium hydroxide
sulphate RS in saline solution. and filter. Dilute 20 ml ofthe filtrate to 50 ml with 0.1 M sodium
Apply to the plate 2 III of each solution. After development, hydroxide.
dry the plate in air, spray with a 2 per cent w/v solution of When examined in the range 230 urn to 360 nm (2.4.7), the
4-aminoantipyrine in methanol. Dry the plate in air and spray resulting solution shows an absorption maximum at about
with a freshly prepared 8.0 per cent w/v solution ofpotassium 296 urn.
ferricyanide in a mixture of 4 volumes of strong ammonia B. Determine by thin-layer chromatography (2.4.17), coating
solution and 1 volume of water. The principal spot in the
the plate with silica gel G
chromatogram obtained with the test solution corresponds to
that in the chromatogram obtained with the reference solution. Mobile phase. A mixture of 65 volumes of 2-propanol,
25 volumes of cyclohexane and 5 volumes offormic acid.
Tests
Test solution. Shake a quantity of the powdered tablets
pH (2.4.24). 3.0 to 5.0. containing 10 mg ofTerbutaline Sulphate with 4 ml ofa mixture
of equal volumes of ethanol (95 per cent) and water for
Other tests. Complies with the tests stated under Parenteral
10 minutes, centrifuge and use the clear supernatant liquid.
preparations (Injections).
Reference solution (a). A 0.25 per cent w/v solution of
Assay. Measure accurately a volume containing about 5 mg
terbutaline sulphate RS in water.
of Terbutaline Sulphate and add sufficient water to produce
50.0 m!. To 5.0 ml add 35 ml of a buffer solution prepared by Reference solution (b). A mixture of equal volumes ofthe test
dissolving 36.3 g of tris (hydroxymethyl) aminomethane in solution and reference solution (a).
900 ml of water, adjusting the pH to between 9.4 and 9.6 and Apply to the plate 2 III of each solution. After development,
adding sufficient water to produce 1000 m!. Add 1.0 ml of a dry the plate in air, allow to stand for a few minutes in an
freshly prepared 2.0 per cent w/v solution of atmosphere saturated with diethylamine and spray with
4-aminoantipyrine, mix and add 1.0 ml of a freshly prepared diazotised nitroaniline solution. The principal spot in the
8.0 per cent w/v solution of potassium ferricyanide with chromatogram obtained with the test solution corresponds to
vigorous swirling and sufficient of the buffer solution to that in the chromatogram obtained with reference solution (a)
produce 50.0 m!. Exactly 75 seconds after the addition ofthe and the principal spot in the chromatogram obtained with
potassium ferricyanide solution measure the absorbance of reference solution (b) appears as a single compact spot.
the resulting solution at the maximum at about 550 nm (2.4.7),
using water as the blank. Tests
Calculate the content of (CI2HI9N03)2, H 2S04 from the Dissolution (2.5.2).
absorbance obtained by repeating the operation using a 0.01 Apparatus No.2,
per cent w/v solution of terbutaline sulphate RS and Medium. 900 ml of water,
beginning at the words "To 5.0 ml add 35 ml... ...". Speed and time. 100 rpm and 45 minutes.
Storage. Store protected from light, in single dose containers. Withdraw a suitable volume ofthe medium and filter. Measure
Labelling. The label states that the injection should not be the absorbance of the filtered solution, suitably diluted with
used" if the solution is discoloured. the medium if necessary, at the maximum at about 550 urn
(2.4.7). Calculate the content of (ClzHI9N03)z.HzS04 in the
medium from the absorbance obtained from a solution of
known concentration of terbutaline RS in the same medium.
Terbutaline Tablets
D. Not less than 70 per cent of the stated amount of
Terbutaline Sulphate Tablets (CI2HI9N03)2.HZS04.
Terbutaline Tablets contain not less than 90.0 per cent and Uniformity of content. Comply with the test stated under
not more than 110.0 per cent ofthe stated amount ofterbutaline Tablets.
sulphate (CI2HI9N03)z, H2S04.
Powder one tablet, transfer to a 25-ml volumetric flask, add
Usual strengths. 2.5 mg; 5 mg. 1~.lTIl of 0, OJ Ml1ydrQcl}lorJc gciciand ~Qak:e f()r lQ l11illlJtes,
Dilute to volume with 0.01 M hydrochloric acid and filter,
Identification
rejecting the first 5 ml of the filtrate. Dilute, if necessary, a
A. Shake a quantity ofthe powdered tablets containing 20 mg suitable volume of the filtrate with 0.01 M hydrochloric acid
ofTerbutaline Sulphate with 50 ml of 0.1 M sodium hydroxide to produce a solution containing 0.01 per cent w/v solution of

2198

www.webofpharma.com
IP20l0 TESTOSTERONE PROPIONATE

Terbutaline Sulphate. Carry out the method as described under Description. A white or almost white powder or colourless
Assay beginning at the words "To 5.0 ml add 35 ml ofa buffer crystals; odourless.
solution....". Calculate the content of (CI2HI9N03h H2S04 in
the tablet from the absorbance obtained by carrying out the Identification
Assay simultaneously using terbutaline sulphate RS.
TestA may be omitted iftests Band Care carried out. Tests B
Other tests. Comply with the tests stated under Tablets. and C may be omitted if test A is carried out.
Assay. Weigh and powder 20 tablets. Weigh accurately a A. Determine by infrared absorption spectrophotometry (2.4.6).
quantity ofthe powder containing about 5 mg ofTerbutaline Compare the spectrum with that obtained with testosterone
Sulphate, transfer to a 50-ml volumetric flask, add 30 ml of propionate RS or with the reference spectrum of testosterone
0.01 M hydrochloric acid and shake for 10 minutes. Dilute to propionate.
volume with 0.01 M hydrochloric acid and filter, rejecting the
first 5 ml ofthe filtrate. To 5.0 mladd35 ml ofa buffer solution B. In the test for Related substances the principal peak in the
prepared by dissolving 36.3 g of tris (hydroxymethyl) chromatogram obtained with the test solution corresponds to
aminomethane in 900 ml ofwater, adjusting the pH to between the peak due to testosterone propionate in the chromatogram
9.4 and 9.6 and adding sufficient water to produce 1000 ml. obtained with reference solution (a).
Add 1.0 ml of a freshly prepared 2.0 per cent w/v solution of C. Melting range. 119°to 123° (2.4.21).
4-aminoantipyrine, mix and add 1.0 ml of a freshly prepared
8.0 per cent w/v solution of potassium ferricyanide with Tests
vigorous swirling and sufficient of the buffer solution to
produce 50.0 ml. Exactly 75 seconds after the addition of the Specific optical rotation (2.4.22). +83.0° to +90.0°, determined
potassium ferricyanide solution measure the absorbance of in a 1.0 per cent w/v solution in ethanol.
the resulting solution at the maximum at about 550 urn (2.4.7), Related substances. Detelmine by. liquid chromatography
using water as the blank. (2.4.14).
Calculate the content of (C 12H I9N0 3h, H 2S0 4 from the Test solution. Dissolve 50 mg of the substance under
absorbance obtained by carrying out the Assay simultaneously examination in methanol and dilute to 50 ml with the same
using terbutaline sulphate RS. solvent.
Storage. Store protected from light and moisture. Reference solution (a). Dissolve 2 mg of the substance under
examination and 2 mg oftestosterone acetate RS in methanol
and dilute to 50 ml with the same solvent.
Testosterone Propionate Reference solution (b). Dilute 1 ml of the test solution to
100 m1 with methanol.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 !lm),
- mobile phase: a mixture of 20 volumes of water and
80 volumes of methanol,
flow rate. 1.5 m1 per minute,
spectrophotometer set at 254 nm,
o injection volume. 20 !ll.
~2H3203 Mol. Wt. 344.5 Inject the test solution and the reference solutions. Continue
the chromatography for twice the retention time for testo-
Testosterone Propionate is 3-oxoandrost-4-en-17~-yl
sterone propionate.
propionate.
The relative retention time of about four impurities with
Testosterone Propionate contains not less than 97.0 per cent
reference to testosterone propionate (retention time, about
and not more than 103.0. per cent ofC 22 H320 3, calculated on
9 minutes) range from 0.5 to about 1.4.
the dried basis.
The test is not valid unless in the chromatogram obtained
Category. Androgen; anabolic steroid.
with reference solution (a) the resolution between the peaks
Dose. By intramuscular injection, 5 to 25 mg, once or twice due to testosterone and testosterone acetate is not less than
weeldy. 4.0.

2199

www.webofpharma.com
TESTOSTERONE PROPIONATE INJECTION. IP 2010

In the chromatogram obtained with the test solution the area Solvent mixture. A mixture of 90 volumes of light petroleum
of any peak other than the principal peak is not more than (40° to 60°) and 10 volumes of liquid paraffin.
0.5 times the area of the principal peak in the chromatogram
obtained with reference solution (b) (0.5 per cent); the sum of Mobile phase. A mixture of 30 volumes of water and
the areas of any secondary peaks is not greater than the area 20 volumes of glacial acetic acid.
of the principal peak in the chromatogram obtained with
Test solution. Dissolve 25 mg of the residue in 10 ml of the
reference solution (b) (1.0 per cent). Ignore any peak with an
solvent mixture.
area less than 0.05 times the area ofthe principal peak in the
chromatogram obtained with reference solution (b) (0.05 per Reference solution (a). A 0.25 per cent w/v solution of
cent). testosterone propionate RS in the solvent mixture.
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
Reference solution (b). Mix equal volumes ofthe test solution
Loss on drying (2.4.19). Not more than 0.5 per cent, determined and reference solution (a).
on 1.0 g by drying in an oven at 105° for 2 hours.
Place the dry plate in a tame containing a shallow layer of the
Assay. Weigh accurately about 25 mg, dissolve in sufficient solvent mixture, allow the solvent mixture to ascend to the
ethanol to produce 250.0 ml, mix, dilute 5.0 ml to 50.0 ml with top, remove the plate from the tank and allow the solvent to
the same solvent and measure the absorbance ofthe resulting evaporate. Use within 2 hours, with the flow of the mobile
solution at the maximum at about 241 urn (2.4.7). phase in the direction in which the aforementioned treatment
Calculate the content of C22 H 320 3 taking 490 as the specific was done.
absorbance at 241 nm.
Apply to the plate 2 J!l of each solution. Allow the mobile
Storage. Store protected from light and moisture. phase to rise 12 cm. Dry the plate in a current ofwarm air, allow
the solvent to evaporate, heat at 1200 for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 1200 for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
Testosterone Propionate Injection spot in the chromatogram obtained with the test solution
Testosterone Propionate Injection is a sterile solution of corresponds to that in the chromatogram obtained with
Testosterone Propionate in Ethyl Oleate or any other suitable reference solution (a). The principal spot in the chromatogram
ester, in a suitable fixed oil or in any mixture ofthese. obtained with reference solution (b) appears as a single,
compact spot A.
Testosterone Propionate Injection contains not less than
92.5 per cent and not more than 107.5 per cent of the stated
amount of testosterone propionate, C22H 32 0 3 • Tests
Usual strength. 25 mgper ml; 50 mgper ml. Other tests. Complies with the tests stated under Parenteral
preparations (Injections).
Identification
Assay. To an accurately measured volume containing about
Dissolve a volume containing 50 mg ofTestosterone Propionate 0.1 g ofTestosterone Propionate add sufficient chloroform to
in 8 ml of light petroleum (40° to 60°) and extract with three produce 100.0 ml and mix. Dilute 3.0 ml to 50.0 ml with
quantities, each of 8 ml, of a mixture of 7 volumes.of glacial chloroform and to 5.0 ml ofthe solution add lOml of isoniazid
acetic acid and 3 volumes of water. Wash the combined solution and sufficient methanol to produce 20.0 ml. Allow to
extracts with 10 ml of lightpetroleum (40° to 60°), dilute with stand for 45 minutes and measure the absorbance of the
water until the solution becomes turbid, allow to stand for resulting solution at the maximum at about 380 urn (2.4.7),
2 hours in ice and filter. The precipitate, after washing with using as the blank a solution prepared by treating 5 ml of
water and drying at 105°, complies with the following tests. chloroform in the same manner.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Calculate the content of C22H3203 from the absorbance
Compare the spectrum with that obtained with testosterone
obta.ined by repeating the operation usingaQ.@_6.p_eccent
propionate RS or with the reference spectrum oftestosterone
propionate. w/v solution of testosterone propionate RS in chloroform
and beginning at the words "to 5.0 ml of the solution.....".
B. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G. Storage. Store protected from light.

2200

www.webofpharma.com
IP 2010 TETRACYCLINE

Tetracycline Reference solution (b). A solution containing 0.0025 per cent


w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent
w/v of tetracycline hydrochloride RS in the diluting solvent.
OH 0 o
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 to 10 /lm),
mobile phase: a mixture of 680 ml of 0.1 M ammonium
oxalate, 270 ml of dimethylformamide and 50 ml of
0.2 M dibasic ammonium phosphate, the pH of the
Mol. Wt. 444.5 mixture being adjusted, if necessary, to 7.6 to 7.7 with
3 M ammonia or 3 M phosphoric acid,
Tetracycline is (4S,4aS,5aS,6S,12aS)-4-dimethylamino- - flow rate. 2 ml per minute,
1,4,4a,5,5a,6,11,12a-octahydro-3,6,10,12,12a-pentahydroxy- - spectrophotometer set at 280 nm,
6-methyl-1, 11-dioxonaphthacene-2-carboxamide. It contains injection volume. 20 /ll.
a variable quantity of water.
The test is not valid unless the resolution between the two
Tetracycline contains not less than 88.0 per cent ofC2zHz4NzOs, principal peaks in the chromatogram obtained with reference
calculated on the dried basis. solution (b) is not less than 1.5. In the chromatogram obtained
Category. Antibacterial. with the test solution, the area of any peak corresponding to
4-epitetracycline is not greater than the area of the principal
Dose. Orally, 250 mg every 6 hours, increased in severe peak in the chromatogram obtained with reference solution
infections to 500 mg every 6 to 8 hours. (a) and the total area of all the peaks other than the principal
Description. A yellow, crystalline powder. peak is not greater than 5.0 per cent.
Heavy metals (2.3.13). 0.5 g complies with the limit test for
Identification
heavy metals, Method B (50 ppm). Use 2.5 rnl ofleadstandard
A. In the test for Assay, the principal peak in the chromatogram solution (l0 ppm Pb) to prepare the standard.
obtained with the test solution corresponds to the peak due Sulphated ash (2.3.18). Not more than 0.5 per cent.
to tetracycline hydrochloride in the chromatogram obtained
with reference solution (a). Loss on drying (2.4.19). Not more than 13.0 per cent,
determined on 0.5 g by drying in an oven at 105°.
B. To about 2 mg add 5 ml of sulphuric acid; a reddish violet
colour develops. Add the solution to 2.5 ml of water; the Assay. Determine by liquid chromatography (2.4.14).
colour changes to yellow. Test solution. Weigh accl,lfately about 50 mg ofthe substance
C. Dissolve about 10 mg in a mixture of 1 ml of 2 M nitric acid under examination and dissolve in 100.0 ml ofa mixture of
and 5 ml of water, shake well and add 1 ml of silver nitrate 68 ml of 0.1 M ammonium oxalate and 27 m1 of
solution. Any opalescence produced is not more intense than dimethylformamide (diluting solvent).
that in a solution prepared in the same manner omitting the Reference solution (a). A 0.05 per cent w/v solution of
substance under examination. tetracycline hydrochloride RS in the diluting solvent.

Tests Reference solution (b). A solution containing 0.0025 per cent


w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent
pH (2.4.24). 3.5 to 6.0, determined in a 1.0 per cent w/v w/v of tetracycline hydrochloride RS inthe diluting solvent.
suspension in carbon dioxide-free water.
Chromatographic system
Specific optical rotation (2.4.22).-260° to -280°, determined at - a stainless steel column 25 cm x 4.6 rom, packed with
20° in a 1.0 per cent w/v solution in 0.1 M hydrochloric acid. octylsilane bonded to porous silica (5 to 10 /lm),
Related substances. Determine by liquid chromatography - mobile phase: a mixture of 680 ml of 0.1 M ammonium
(2.4.14) oxalate, 270 ml of dimethylformamide and 50 ml of
0.2 M dibasic ammonium phosphate, the pH of the
Test solution. Dissolve 0.1 g of the substance under mixture being adjusted, if necessary, to 7.6 to 7.7 with
examination in 100 rnl ofa mixture of68 rnl of 0.1 M ammonium 3 M ammonia or 3 M phosphoric acid,
oxalate and 27 ml of dimethylformamide (diluting solvent). - flow rate. 1.5 ml per minute,
Reference solution (a). A 0.0025 per cent w/v solution of - spectrophotometer set at 280 nm,
4-epitetracycline hydrochloride RS in the diluting solvent. - injection volume. 20 /ll.

2201

www.webofpharma.com
TETRACYCLINE HYDROCHLORIDE IF 2010

The test is not valid unless the resolution between the two Specific optical rotation (2.4.22). -239° to -255°, detennined
principal peaks in the chromatogram obtained with reference at20° in a 1.0 per centw/v solution in 0.1 Mhydrochloric acid.
solution (b) is not less than 1.5.
Related substances. Determine by liquid chromatography
Calculate the content ofCzzHz4NzOs. (2.4.14).
1mg ofC22Hz~zOs,HCI is equivalent to 0.92 mg ofCzzHz4NzOs. Test solution. Dissolve 0.1 g of the substance under
examination in 100 ml ofa mixture of68 ml of 0.1 M ammonium
Storage. Store protected fi'om light and moistUTe.
oxalate and 27 ml of dimethylformamide (diluting solvent).
Reference solution (a). A 0.0025 per cent Mv solution of
4-epitetracycline hydrochloride RS in the diluting solvent.
Tetracycline Hydrochloride Reference solution (b). A solution containing 0.0025 per cent
w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent
w/v of tetracycline hydrochloi'ide RS in the diluting solvent.
Chromatographic system
, Hel - a stainless steel column 25 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 to 10 /lm),
mobile phase: 680 ml of 0.1 M ammonium oxalate,
270 ml of dimethylformamide and 50 ml of 0.2 M dibasic
ammonium phosphate, the pH of the mixtUTe being
Mol. Wt. 480.9 adjusted, ifnecessary, to 7.6 to 7.7 with 3 M ammonia or
Tetracycline Hydrochloride is (4S,4aS,5aS,6S, 12aS)- 3 M phosphoric acid,
4- dimethylamino-1 ,4,4a,5,5a,6,11,12a-octahydro- - flow rate. 2 ml per minute,
3,6,10,12,12a-pentahydroxy-6-methyl-1,11-dioxo- - spectrophotometer set at 280 nm,
naphthacene-2- carboxamide hydrochloride. - injection volume. 20 Ill.

Tetracycline Hydrochloride contains not less than 95.0 per The test is not valid unless the resolution between the two
cent and not more than 100.5 per cent of CzzHz4NzOs, HCI, principal peaks in the chromatogram obtained with reference
calculated on the dried basis. solution (b) is not less than 1.5. In the chromatogram obtained
with the test solution the area of any peak corresponding to
Category. Antibacterial. 4-epitetracycline is not greater than the area of the principal
Dose. Orally, 250 mg every 6 hOUTS, increased in severe peak in the chromatogram obtained with reference solution
infections to 500 mg every 6 to 8 hOUTS; by intramuscular (a) and the total area of all peaks other than the principal peak
injection, 100 mg every 8 to 12 hOUTS, increased in severe is not greater than 5.0 per cent.
infections to every 4 to 6 hOUTS; by intravenous infusion, 500 Heavy metals (2.3.13). 0.5 g complies with the limit test for
mg every 12 hOUTS; maximum 2 g daily. heavy metals, Method B (50 ppm). Use 2.5 ml ofleadstandard
Description. A yellow, crystalline powder. solution (l0 ppm Pb) to prepare the standard.
Sulphated ash (2.3.18). Not more than 0.5 per cent.
Identification
Loss on drying (2.4.19). Not more than 2.0 per cent, detennined
A. In the test for Assay, the principal peak in the chromatogram on 1.0 g by drying in an oven at 60° over phosphorus pentoxide
obtained with the test solution corresponds to the peak due at a pressure not exceeding 0.7 kPa for 3 hOUTS.
to tetracycline hydrochloride in the chromatogram obtained Assay. Determine by liquid chromatography (2.4.14).
with reference solution (a).
Test solution. Weigh accUTately about 50 mg ofthe substance
B. To about 2 mg add 5 ml of sulphuric acid; a reddish violet under examination and dissolve in about 50 ml ofthe diluting
colOUT develops. Add the solution to 2.5 ml of water; the solvent described in the test for related substances and further
COIOUT changes to yellow. add sufficient diluting solvent to produce 100.0 ml.
C. Gives reaction A ofchlorides (2.3.1). Reference solution (a). A 0.05 per cent w/v solution of

Tests
Reference solution (b). A solution containing 0.0025 per cent
pH (2.4.24). 1.8 to 3.0, determined in a LOper cent w/v w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent
suspension in carbon dioxide-ji-ee water. w/v of tetracycline hydrochloride RS in the diluting solvent.

2202

www.webofpharma.com
IP 2010 TETRACYCLINE CAPSULES

Chromatographic system B. To a quantity of the mixed contents of 20 capsules


a stainless steel column 25 cm x 4.6 mm, packed with containing about 10 mg of Tetracycline Hydrochloride, add
octylsilane bonded to porous silica (5 to 10 /.un), 20 ml of warm ethanol (95 per cent), allow to stand for
mobile phase: a mixture of 680 volumes of 0.1 M 20 minutes, filter and evaporate the filtrate to dryness on a
ammonium oxalate, 270 volumes of dimethylformamide water-bath. To 0.5 mg of the residue add 2 ml of sulphuric
and 50 volumes of 0.2 M dibasic ammonium phosphate, acid; a reddish violet colour develops. Add the solution to 2.5
the pH ofthe mixture being adjusted, ifn:ecessary, to 7.6 ml of water; the colour changes to yellow.
to 7.7 with 3 M ammonia or 3 M phosphoric acid,
C. The residue obtained in test B gives reaction A of chlorides
flow rate. 1.5 ml per minute,
(2.3.1).
- spectrophotometer set at 280 nm,
- injection volume. 20 Ill.
Tests
The test is not valid unless the resolution between the two
principal peaks in the chromatogram obtained with reference Related substances. Determine by liquid chromatography
solution (b) is not less than 1.5. (2.4.14).

Calculate the content ofC22H24N20s, HCl. Test solution. Shake a quantity of the mixed contents of
20 capsules containing about 25 mg of Tetracycline
Tetracycline Hydrochloride intended for use in the
Hydrochloride with 80 ml of 0.01 M methanolic hydrochloric
manufacture ofparenteral preparations without a fitrther
acid for 10 minutes, dilute to 100 ml with the same solvent, mix
appropriate procedure for removal of bacterial endotoxins
and filter if necessary.
complies with the following additional requirement.
Reference solution (a). A 0.002 per cent w/v solution of
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
4-epitetracycline hydrochloride RS in 0.01 M methanolic
per mg.
hydrochloric acid.
Tetracycline Hydrochloride intended for use in the
Reference solution (b). A solution containing 0.0015 per cent
manufacture ofparenteral preparations without a fitrther
w/v each of 4-epitetracycline hydrochloride RS and
appropriate sterilisation procedure complies with the
tetracycline hydrochloride RS in 0.01 M methanolic
following additional requirement.
hydrochloric acid.
Sterility (2.2.11). Complies with the test for sterility.
Chromatographic system
Storage. Store protected from light and moisture. If it is a stainless steel column 25 cm x 4.6 mm, packed with
intended for use in the manufacture ofparenteral preparations, octadecylsilane bonded to porous silica (10 Ilm),
the container should be sterile, tamper-evident and sealed so - column. temperature 40°,
as to exclude microorganisms. - mobile phase: a mixture of 5 volumes of dimethyl-
Labelling. The label states whether or not the material is formamide and 95 volumes of 0.1 M oxalic acid, the
intended for use in the manufacture ofparenteral preparations. pH of the mixture being adjusted to 3.9 with
triethylamine,
flow rate. 2 ml per minute,
spectrophotometer set at 280 nm,
injection volume. 20 Ill.
Tetracycline Capsules
The test is not valid unless the resolution between the two
Tetracycline Hydrochloride Capsules principal peaks in the chromatogram obtained with reference
Tetracycline Capsules contain not less than 90.0 per cent and solution (b) is not less than 2.0. In the chromatogram obtained
not more than 110.0 per cent of the stated amount of with the test solution the area of any peak corresponding to
tetracycline hydrochloride, C22H24N20s, HCl. 4-epitetracycline hydrochloride is not greater than the area of
the principal peak in the chromatogram obtained with reference
Usual strength. 250 mg; 500 mg. solution (a) and the total area of all the peaks other than the
principal peak is not greater than 10.0 per cent.
Identification
Dissolution (2.5.2).
A. In the test for Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak due Apparatus No.1,
to tetracycline hydrochloride in the chromatogram obtained Medium. 900 ml of water freshly prepared by distillation,
with reference solution (a). Speed and time. 75 rpm and 60 minutes.

2203

www.webofpharma.com
TETRACYCLINE CAPSULES IP 2010

Withdraw a suitable volume ofthe medium and filter through Tetracycline Ointment
a membrane filter disc having an average pore diameter not
greater than 1.0 /lm, rejecting the first few ml of the filtrate. Tetracycline Hydrochloride Eye Ointment
Measure the absorbance of the resulting solution, suitably Tetracycline Ointment contains not less than 90.0 per cent
diluted ifnecessary, at the maximum at about 276 nm (2.4.7). and not more than 115.0 per cent of the stated amount of
Calculate the content ofC22H24N20g, HCl in the medium from tetracycline hydrochloride, C22H24N20g, HCl.
the absorbance obtained from a solution of known
concentration of tetracycline hydrochloride RS. Usual strengths: 0.5 per cent w/w; 1 per cent w/w.

D. Not less than 70 per cent of the stated amount of Identification


C22H24N20g, HCl. .
In the test for Assay, the principal peak in the chromatogram
Loss on drying (2.4.19). Not more than 3.0 per cent, determined obtained with the test solution corresponds to the peak due
on 1.0 g of the contents of the capsules by drying in an oven to tetracycline hydrochloride in the chromatogram obtained
at 60° at a pressure not exceeding 0.7 kPa for 3 hours. with the reference solution.
Other tests. Comply with the tests stated under Capsules.
Tests
Assay. Determine by liquid chromatography (2.4.14). Water (2.3.43). Not more than 0.5 per cent, determined on
Test solution. Weigh accurately a quantity of the mixed 2.0 g dissolved in a mixture of 2 volumes of carbon
contents of 20 capsules containing about 50 mg of tetrachloride, 2 volumes of chloroform and 1 volume of
Tetracycline Hydrochloride, shake with about 50 ml of methanol.
0.01. M methanolic hydrochloric acid and dilute with the Other tests. Complies with the tests stated under Eye
same solvent to produce 100.0 ml, mix and filter. Discard the Ointments.
first few ml ofthe filtrate.
Assay. Determine by liquid chromatography (2.4.14).
Reference solution (a). A 0.05 per cent w/v solution of Test solution. Weigh accurately a quantity containing about
tetracycline hydrochloride RS in 0.01 M methanolic 25 mg of Tetracycline Hydrochloride and transfer to a
hydrochloric acid. separating funnel with the aid of 15 ml of cyclohexane. Add
Reference solution (b). A solution containing 0.0025 per cent 15 ml ofa mixture of68 volumes of 0.1 M ammonium oxalate
w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent and 27 volumes of dimethylformamide (diluting solvent) and
w/v of tetracycline hydrochloride RS in 0.01 M methanolic shake well. Collect the lower layer in a 50-ml volumetric flask.
hydrochloric acid. Repeat the extraction with two further quantities, each of
15 ml, of the diluting solvent, combining the extracts in the
Chromatographic system same 50-ml volumetric flask. Add sufficient diluting solvent to
a stainless steel column 25 cm x 4.6 mm, packed with produce 50.0 ml, mix and filter.
octylsilane bonded to porous silica (5 tolO /lm), Reference solution. A 0.05 per cent w/v solution of tetracycline
mobile phase: a mixture of 68 volumes of 0.1 M hydrochloride RS in the diluting solvent. .
ammonium oxalate and 27 volumes of dimethyl-
formamide and 5 volumes of 0.2 M dibasic ammonium Chromatographic system
phosphate, the pH of the mixture being adjusted, a stainless steel column 25 cm x 4.6 mm, packed with
if necessary, to 7.6 to 7.7 with 3 M ammonia or octylsilane bonded to porous silica (5 tolO /lm),
3 M phosphoric acid, - mobile phase: a mixture of 6~ volumes of 0.1 M
ammonium oxalate and 27 volumes of dimethyl-
flow rate. 1.5 ml per minute,
formamide and 5 volumes of 0.2 M dibasic ammonium
spectrophotometer set at 280 nm,
phosphate, the pH of the mixture being adjusted, if
injection volume. 20 /ll. necessary, to 7.6 to 7.7 with 3 M ammonia or
The test is not valid unless the resolution between the two 3 M phosphoric acid,
principal peaks in the chromatogram obtained with reference - flow rate. 1.5 ml per minute,
solution (b) is not less than 1.5. spectrophotometer set at 280 nm,
injection volume. 20 /ll.
Calculate the content ofC22H24N20g, HCl in the capsules.
Calculate the content ofC22H24N20g, HCI in the eye ointment.
Storage. Store protected from light and moisture. Storage. Store protected from moisture.

2204

www.webofpharma.com
IP 2010 THEOPHYLLINE

Theophylline Test solution. Dissolve 40 mg of the substance under


examination in 20.0 ml ofthe mobile phase.
Reference solution (a). Dilute 1.0 ml of the test solution to
100.0 ml with the mobile phase. Dilute 1.0 ml ofthis solution to
10.0 ml with the mobile phase. .
Reference solution (b). Dissolve 10 mg of theobromine in the
mobile phase, add 5 ml ofthe test solution and dilute to 100 ml
with the mobile phase. Dilute 5 ml of this solution to 50.0 ml
with the mobile phase.
C7HsN40 2 Mol. Wt. 180.2 (anhydrous)
C7HsN402,H20 Mol. Wt. 198.2 (hydrate) Chromatographic system
- a stainless steel column 25 cm x 4 mm, packed with
Theophylline is 1,3-dimethyl-3,7-dihydro-1H-purine-2, octadecylsilane bonded to porous silica (7 /-Lm),
6-dione. It contains one molecule of water or is anhydrous. mobile phase: a mixture of 7 volumes of acetonitrile
Theophylline contains not less than 98.0 per cent and not and 93 volumes of0.14 per cent w/v solution of sodium
more than 101.0 per cent ofC7H sN 40 2, calculated on the dried acetate containing 0.5 per cent v/v solution of glacial
basis. acetic acid,
- flow rate. 2 ml per minute,
Category. Xanthine bronchodilator.
- spectrophotometer set at 272 nm,
Dose. By intravenous infusion, 2.5 to 5.0 mg of anhydrous - injection volume. 20 /-Ll.
theophylline per kg body weight over a period of20 minutes.
Inject reference solution (b). The test is not valid unless the
Description. Awhite, crystalline powder; odourless. resolution between the peaks. due to theobromine and
theophylline is not less than 2.0. The relative retention time
Identification with reference to theophylline for theophylline impurity C is
about 0.3, for theophylline impurity B is about 0.4, for
Test A may be omitted iftests B, C andD are carried out. Tests theophylline impurity D is about 0.5 and for theophylline
Band D may be omitted if tests A and C are carried out. impurity Ais about 2.5.
A. Determine by infrared absorption spectrophotometry (2.4.6). Inject the test solution and reference solution (a). Run the
Compare the spectrum with that obtained with theophylline chromatogram 3.5 times the retention time of the principal
RS or with the reference spectrum oftheophylline. peak. In the chromatogram obtained with the test solution,
B. Dissolve about 10 mg in 10 ml of water, add 0.5 ml ofa 5 per the area of any secondary peak is not more than the area of
cent w/v solution of mercuric acetate and allow to stand; a the principal peak in the chromatogram obtained with reference
white, crystalline precipitate is produced. solution (aHO.1 per cent). The sum ofall the secondary peaks
is not more than 5 times the area of the principal peak in the
C. The melting range, after drying at 105°,270° to 274° (2.4.21).
chromatogram obtained with reference solution (a) (0.5 per
D. Gives the reaction ofxanthines (2.3.1 ). cent) Ignore any peak with an area less than 0.5 times the area
of the principal peak in the chromatogram obtained with
Tests reference solution (a) (0.05 per cent).
Appearance of solution. Dissolve 0.5 gin 75 ml of carbon Heavy metals (2.3.13).1.0 g complies with the limit test for
dioxide-free water with heating; the resulting solution heavy metals, Method B (20 ppm).
(solution A), is clear (2.4.1), and colourless (2.4.1).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Acidity. To 50 ml ofsolution Aadd 0.1 rnl ofmethyl redsolution.
Loss on drying (2.4.19). Not more than 0.5 per cent (for the
The solution is red and not more than 1.0 ml of 0.01 Msodium
anhydrous form) and 8.0 per cent to 9.5 per cent (for the
hydroxide is required to change the colour of the solution to
hydrated form), determined on 1.0 g by drying in an oven at
yellow.
105°.
Light absorption (2.4.7). Absorbance ofa 0.001 per cent w/v
Assay. Weigh accurately about 0.25 g, add 50 ml of water and
solution in 0.1 M hydrochloric acid at the maximum at about
gently warm the mixture on a water-bath until complete solution
270 nm, not less than 0.53.
is effected. Cool, add 20.0 ml of 0.1 M silver nitrate and 1.0 ml
Related substances. Determine by liquid chromatography of bromothymol solution and titrate with 0.1 M sodium
(2.4.14). hydroxide until a blue colour is obtained..

2205

www.webofpharma.com
THEOPHYLLINE INJECTION IP 2010

1 ml of 0.1 M sodium hydroxide is equivalent to 0.01802 g of Assay. For theophylline - Dilute a suitable volume of the
C7H gN 40 Z' injection with sufficient 0.1 M sodium hydroxide to produce a
solution containing 0.008 per cent w/v of anhydrous
Storage. Store protected from moisture.
theophylline. Measure the absorbance of the resulting
solution at the maximum at about 274 nm (2.4.7), using as the
blank a solution prepared in the same manner omitting the
Theophylline Injection substance under examination.

Theophylline in Dextrose Injection Calculate the content of C 7H gN 4 0 Z from the absorbance


obtained by repeating the operation using theophylline RS in
Theophylline Injection is a sterile solution of Theophylline place of the substance under examination.
and Dextrose in Water for Injections.
For dextrose - Transfer an accurately measured volume of
Theophylline Injection contains not less than 92.5 per cent the injection containing about 5 mg of Dextrose to a 100-ml
and not more than 107.5 per cent of the stated amount of volumetric flask, add 0.2 ml of 6 M ammonia, dilute to volume
anhydrous theophylline, C 7H gN 40 Z, and not less than 95.0 per with water and mix. Determine the optical rotation of the
cent and not more than 105.0 per cent ofthe stated amount of resulting solution in a suitable polarimeter tube at 25° (2.4.22).
dextrose, C6H 1Z0 6, HzO. The observed rotation, in degrees multiplied by 1.042571,
Usual strengths. Anhydrous theophylline, 0.4, 0.8, 1.6,2,3.2, represents the weight, in g, of C6HIZ06,HzO in the volume of
4 mg per ml in 5 per cent w/v Dextrose. the injection taken for the assay, where A is the ratio 200 divided
by the length, in mm, ofthe polarimeter tube employed.
Identification
Storage. Store protected from light, in single dose containers.
A. When examined in the range 230 nm to 360 nm (2.4.7), the
Labelling. The label states the strength in terms ofthe amounts
solution obtained in the Assay for theophylline shows an
of anhydrous theophylline and Dextrose.
absorption maximum at about 274 nm.
B. Add 0.2 ml of the injection to 5 ml of potassium cupri-
tartrate solution and heat to boiling; a red to orange precipitate
is formed. Thiabendazole
Tests Tiabendazole

pH (2.4.24). 3.5 to 6.5.


5-Hydroxymethylfurfural and Related substances. Use a glass
chromatographic column (66 cm x 11 mm) with a sealed-in,
coarse-porosity sintered disc or a glass wool plug and fitted
with a stopcock. Mix 8 g of a 20- to 50-mesh styrenedivinyl~ C IOH7N3S Mol. Wt. 201.3
benzene anion-exchange resin in the hydroxide form with
Thiabendazole is 2-(1 ,3-thiazol-4-yl)-IH-benzimidazole.
25 ml of water, allow to settle and decant the supernatant
liquid until a slurry of resin remains. Pour the slurry into the Thiabendazole contains not less than 98.0 per cent and not
column and allow to settle as a homogeneous bed having a more than 101.0 per cent of C IOH 7N 3S, calculated on the
bed volume ofabout 15 ml. Wash the resin bed at a flow rate of anhydrous basis.
about 3 rn1 per minute with 100 ml ofa5.7 percentw/v solution Category. Anthelmintic.
of ammonium carbonate followed by washing with water
until the eluate has a pH of 7. Dose. In the treatment of nematode infestation, 1.5 g twice
daily for three days.
Dilute an accurately measured volume, of the injection
containing 1.0 g of Dextrose, C6H I2 0 6,HzO, to 250.0 ml with Description. A white or almost white, crystalline powder.
water. Pass this solution through the resin bed in the column Identification
at a flow rate of about 3.5 ml per minute, discarding the first
50 ml of the eluate. Measure the absorbance of the eluate at Test A may be omitted iftests B, C andD are carried out. Tests
284'nm; usingwGteYas'theblafiK;'tne-absorbanceisnotfuofe B;CwidflmajJ oeomittecliftesrATscafriea out:'
__
..... .. ..... - -_ .. ... ".". __ ... ... -
than 0.25 (2.4.7).
~ ~ ~

A. Determine by infrared absorption spectrophotometry (2.4.6).


Other tests. Complies with the tests stated under Parenteral Compare the spectrum with that obtained with thiabendazole
Preparations (Injections). RS or with the reference spectrum of thiabendazole.

2206

www.webofpharma.com
IP 2010 THIABENDAZOLE TABLETS

B. When examined in the range 230 nm to 360 nm (2.4.7), a Water (2.3.43). Not more than 0.5 per cent, determined on
0.0005 per cent w/v solution in 0.1 Mhydrochloric acid shows 1.0 g.
absorption maxima at about 243 nm and 302 urn;ratio ofthe
Assay. Weigh accurately about 0.15 g, dissolve in 30 ml of
absorbance at the maximum at about 302 nm to that at about
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
243 nm, 1.8 to 2.1.
acid, determining the end-point potentiometrically (2.4.25).
C. In the test for Related substances, the principal spot in the Carry out a blank titration.
chromatogram obtained with the test solution (b) corresponds 1 ml of 0.1 M perchloric acid is equivalent to 0.02013 g of
to that in the chromatogram obtained with reference solution CIOH7N3S.
(c).
Storage. Store protected from light and moisture.
D. Dissolve 5 mg in 5 ml of 0.1 M hydrochloric acid, add 3 mg
of 4-phenylenediamine dihydrochloride and shake until
dissolved. Add 0.1 g of zinc powder, mix, allow to stand for
2 minutes and add 10 ml offen-ic ammonium sulphate solution;
a deep blue or bluish violet colour is produced. Thiabendazole Tablets
Tiabendazole Tablets
Tests
Thiabendazole Tablets contain not less than 95.0 per cent and
Appearance of solution. Add 5 ml of methanol to 0.5 g in a not more than 105.0 per cent of the stated amount of
flask fitted with a ground-glass stopper, stir for 5 minutes, thiabendazole, C IO H7N3 S.
with a magnetic stirrer and filter through a sintered-glass filter
Usual strength. 500 mg.
(1.6 Ilm to 4 Ilm). The solution is not more intensely coloured
than reference solution BS6 (2.4.1). Identification
Related substances. Determine by thin-layer chromatography
A. When examined in the range 230 urn to 360 nm (2.4.7), the
(2.4.17), coating the plate with silica gel HF254.
final solution obtained in the Assay shows an absorption
Mobile phase. A mixture of 62.5 volumes of toluene, maximum at about 302 nm.
25 volumes of glacial acetic acid, 10 volumes of acetone and B. Dissolve a quantity of the powdered tablets containing
2.5 volumes of water.
30 mg of Thiabendazole in 5 ml of 0.1 M hydrochloric acid,
Test solution (a). Dissolve 0.1 g of the substance under add 3 mg of 4-phenylenediamine dihydrochloride and shake
examination in 10 ml of methanol. until dissolved. Add 0.1 g of zinc powder, mix, allow to stand
for 2 minutes and add 10 ml of ferric ammonium sulphate
Test solution (b). Dilute 2 ml oftest solution (a) to 20 ml with solution; a deep bluish violet colour is produced.
methanol.
Reference solution (a). Dilute 1 ml oftest solution (b) to 10 ml Tests
with methanol. Disintegration. The test does not apply.
Reference solution (b). Dilute 1 ml oftest solution (b) to 25 ml Other tests. Comply with the tests stated under Tablets.
with methanol.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Reference solution (c). Dissolve 20 mg of thiabendazole RS quantity of the powder containing about 0.1 g of
in 20 ml of methanol. Thiabendazole, add 75 ml of 0.1 M hydrochloric acid, warm
Apply to the plate 20 III of each solution. After development, on a water-bath for 15 minutes, shaking occasionally, cool,
dry the plate in air and examine in ultraviolet light at 254 urn. dilute to 100.0 ml with 0.1 M hydrochloric acid and filter.
Any secondary spot in the chromatogram obtained with test Dilute 5.0 ml ofthe filtrate to 1000.0 ml with 0.1 Mhydrochloric
solution (a) is not more intense than the spot in the acid and measure the absorbance of the resulting solution at
chromatogram obtained with reference solution (a) and not the maximum at about 302 urn (2.4.7).
more than one such spot is more intense than the spot in the Calculate the content ofC IOH 7N 3S taking 1230 as the specific
chromatogram obtained with reference solution (b) absorbance at 302 nm.
Heavy metals (2.3.13). 2.0 g complies with the limit test for Storage. Store protected from light and moisture.
heavy metals, Method B (10 ppm).
Labelling. The label states that the tablets should be chewed
Sulphated ash (2.3.18). Not more than 0.2 per cent. before swallowing.

2207

www.webofpharma.com
THIACETAZONE IP 2010

Thiacetazone Mobile phase. Ethyl acetate.

Test solution. Dissolve 0.4 g of the substance under


examination in 100 ml of methanol.
Reference solution. Dissolve with the aid of heat 0.016 g of
4-acetamidobenzalazine RS in 150 ml of methanol, cool and
dilute to 200 ml with methanol and further dilute 5 ml ofthis
solution to 50 ml with methanol.
Apply to the plate 5 III of each solution. After development,
C IOH I2N40S Mol. Wt. 236.3
dry the plate in air, spray with 2 M nitric acid and within
Thiacetazone is 4-acetamidobenzaldehyde 2 minutes examine in ultraviolet light at 254 nIn. Any secondary
thiosemicarbazone. spot in the chromatogram obtained with the test solution is
Thiacetazone contains not less than 98.0 per cent and not not more intense thl:ln the spot in the chromatogram obtained
more than 102.0 per cent of C IOH I2N 40S, calculated on the with the reference solution.
dried basis. Heavy metals (2.3.13). 2.0 g complies with the limit test for
Category. Antitubercular. heavy metals, Method B (l0 ppm).
Dose. 150 mg daily. Sulphated ash (2.3.18). Not more than 0.2 per cent.
Description. Pale yellow crystals or a crystalline powder; Loss on drying (2.4.19). Not more than 0.5 per cent, determined
almost odourless. on 1.0 g by drying in an oven at 105°.

Identification Assay. Weigh accurately about 0.1 g, dissolve in 60 ml of


methanol by heating at 60° in a water-bath, add slowly 20 ml
Test A may be omitted if tests Band C are carried out. Tests B ofhot methanolic silver nitrate solution, maintain the solution
and C may be omitted if test A is carried out. at 60° until the precipitate coagulates and leaves a clear
A. Determine by infrared absorption spectrophotometry (2.4.6). supematant liquid. Cool, filter through a sintered-glass crucible
Compare the spectrum with that obtained with thiacetazone (porosity No.4), wash the residue with methanol until the
RS or with the reference spectrum of thiacetazone. washings are free from silver nitrate and dry to constant weight
at 105°.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.0003 per cent w/v solution in ethanol (95 per cent) shows I g ofresidue is equivalent to 0.4606 g ofC IOH I2N 40S.
an absorption maximum at about 328 nIn absorbance at about
328 nIn, about 0.55. Storage. Store protected from light and moisture.

C. Boil I0 mg with 5 ml of 1 M hydrochloric acid for 3 minutes,


cool and add sufficient water to produce 200 ml. Mix 5 ml of
this solution with 0.25 ml of sodium nitrite solution and add
the mixture to 0.5 ml of 2-naphthol solution; a red colour is
produced. Thiacetazone and Isoniazid Tablets
Tests Thiacetazone and Isoniazid Tablets contain one part by weight
of Thiacetazone and two parts by weight ofIsoniazid.
Thiosemicarbazide. To 2.0 g, finely powdered, add sufficient
water to produce 50 ml, shake, allow to stand for I hour with Thiacetazone and Isoniazid Tablets contain not less than
occasional shaking and filter, discarding the first few ml ofthe 92.5 per cent and not more than 107.5 per cent of the stated
filtrate. Acidify 25 ml ofthe clear filtrate with dilute sulphuric amounts ofthiacetazone, C IOH I2N 40S, and isoniazid, C6H7N 30 2•
acid, add 0.1 ml of o-phenanthroline-ferrous complex solution Category. Antitubercular.
and titrate with 0.1 M eerie ammonium sulphate to a blue
end-pointwhich persists for I minute; not more than 0.8 ml is Dose. Thiacetazone, 150 mg and Isoniazid, 300 mg daily, in
required. ----- --_._-- -,-,-,.. -----.~-,,--,,--'''--,~'"
divided doses.
4-acetamidobenzalazine. Determine by thin-layer Usual strengths. Thiacetazone, 37.5 mg and Isoniazid, 75 mg;
chromatography (2.4.17), coating the plate with silica gel Thiacetazone, 75 mg and Isoniazid, 150 mg; Thiacetazone, 150
GF254. mg and Isoniazid, 300 mg.

2208

www.webofpharma.com
IP 2010 THIAMINE HYDROCHLORIDE

Identification Thiamine Hydrochloride


A. Extract a quantity ofthe powdered tablets containing about Aneurine Hydrochloride; Vitamin B 1
30 mg of Thiacetazone with 70 ml of ethanol (95 per cent)
with the aid of heat for I5-minutes with occasional shaking.
Cool, dilute to 100 ml with ethanol (95 per cent) and filter.
-
Dilute 1 ml to 100 ml with ethanol (95 per cent). The solution CI, HCI
complies with the following test.
When examined in the range 230 nm to 360 nm (2.4.7), a
0.0005 per cent w/v solution in 0.1 M hydrochloric acid shows
absorption maxima at about 243 nm and 302 nm; ratio of the C 1zH 17ClN40S, HCl Mol. Wt. 337.3
absorbance at the maximum at about 302 nm to that at about
Thiamine Hydrochloride is 3-[(4-amino-2-methylpyrimidin-
243 run, 1.8 to 2.1.
5-yl)methyl]-5-(2-hydroxyethyl)-4-methylthiazolium chloride
B. Shake a quantity of the powdered tablets containing 1 mg hydrochloride.
ofIsoniazid with 50 ml of ethanol (95 per cent) and filter. To
Thiamine Hydrochloride contains not less than 98.5 per cent
5 ml ofthe filtrate add 0.1 g of borax and 5 ml of a 5 per cent
and not more than 101.5 per cent of C 12H 17CIN4 0S, HCl,
w/v solution of l-chloro-2,4-dinitrobenzene in ethanol
calculated on the dried basis.
(95 per cent), evaporate to dryness on a water-bath and
continue heating for a further 10 minutes. To the residue add Category. B-group vitamin.
10 ml of methanol and mix; a reddish purple colour is produced. Dose. Prophylactic, orally, 2 to 5 mg once daily; therapeutic,
orally or by subcutaneous or intramuscular injection, 25 to
Tests 100 mg daily. In multivitamin preparations, prophylactic, orally,
I to 2 mg daily; therapeutic, orally, 4.5 to 10 mg daily.
Disintegration (2.5.1). Not more than 30 minutes.
Description. A white or almost white, crystalline powder or
Other tests. Comply with the tests stated under Tablets. small colourless crystals; odour, slight and characteristic.
Assay. For thiacetazone - Weigh and finely powder
20 tablets. Weigh accurately a quantity of the powder Identification
containing about 30 mg ofThiacetazone, add 70 ml of ethanol Test A may be omitted if tests Band C are carried out. Test B
(95 per cent) and heat on a water-bath for 15 minutes with may be omitted if tests A and C are carried out.
intermittent shaking. Cool, add sufficient ethanol (95 per cent)
to produce 100.0 ml and filter. To 1.0 ml of the filtrate add A. Determine by infrared absorption spectrophotometry
sufficient ethanol (95 per cent) to produce 100.0 ml and (2.4.6). Compare the spectrum with that obtained with thiamine
measure the absorbance of the resulting solution at the hydrochloride RS or with the reference spectrum of thiamine
maximum at about 328 nm (2.4.7), using ethanol (95 per cent) hydrochloride.
as the blank. B. Dissolve about 20 mg in 10 ml of water, add 1 ml of
Calculate the content of C IO H 1ZN 40S from the absorbance
2 M acetic acid and 1.6 ml of 1 M sodium hydroxide, heat on
a water-bath for 30 minutes and allow to cool. Add 5 ml of
obtained by repeating the operation using thiacetazone RS in
2 M sodium hydroxide, 10 ml of potassium ferricyanide
place of the tablets under examination.
solution and 10 ml of I-butanol and shake vigorously for
For isoniazid- Weigh accurately a quantity ofthe powdered 2 minutes. The upper layer exhibits an intense light blue
tablets containing about 0.2 g of Isoniazid, dissolve as fluorescence, particularly in ultraviolet light at 365 run. Repeat
completely as possible in 100 ml of water and filter. Wash the the test but adding 0.9 ml of 1 M sodium hydroxide and 0.2 g
residue with water, combine the filtrate and washings and of sodium sulphite in place of the 1.6 ml of 1 M sodium
dilute to 250.0 ml with water. To 50.0 rn1 ofthe resulting solution hydroxide; practically no fluorescence is produced.
add 50 ml of water, 20 ml of hydrochloric acid and 0.2 g of
C. Gives reaction A ofchlorides (2.3.1).
potassium bromide. Titrate with 0.0167 M potassium bromate,
determining the end-point potentiometrically (2.4.25).
Tests
I ml of 0.0167 M potassium bromate is equivalent to
Appearance of solution. A 5.0 per cent w/v solution is dear
0.003429 g ofC6H7N 30 z.
(2.4.1), and not more intensely coloured than reference solution
Storage. Store protected from light and moisture. YS70rGYS6(2.4.1).

2209

www.webofpharma.com
THIAMINE HYDROCHLORIDE IP 2010

pH (2.4.24). 2.7 to 3.3, determined in a 2.5 per cent w/v solution. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method A (20 ppm).
Related substances. Determine by liquid chromatography
(2.4.14). Nitrates. To 2 ml of a 2.0 per cent w/v solution add 2 ml of
sulphuric acid, cool and superimpose 2 ml offerrous sulphate
Solvent mixture. 5 volumes of glacial acetic acid and
solution; no brown ring is produced at the junction ofthe two
95 volumes ofwater.
layers.
Test solution. Dissolve 0.35 g of the substance under
Sulphates (2.3.17). 0.5 g complies with the limit test for
examination in 15.0 ml of the solution mixture and dilute to
sulphates (300 ppm).
100.0 ml with water.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Reference solution (a). Dissolve 5 mg each ofthe substance
under examination and thioxothiamine RS (thiamine impurity Loss on drying (2.4.19). Not more than 5.0 per cent, determined
A RS) in 4 ml ofthe solvent mixture and dilute to 25.0 ml with on 1.0 g by drying in an oven at 105°.
water. Dilute 5.0 ml ofthe solution to 25.0 ml with water. Assay. Weigh accurately about 0.15 g, dissolve in 5 ml of
Reference solution (b). Dilute 1.0 ml of the test solution to anhydrousformic acid, add 65 ml ofanhydrous glacial acetic
50.0 ml with water. Dilute 5.0 ml ofthis solution to 25.0 ml with acid and 10 ml of mercuric acetate solution, with stirring.
water. Titrate with 0.1 M perchloric acid, determining the end-point
potentiometrically (2.4.25). Carry out a blank titration.
Chromatographic system
- a stainless steel column 25 cm x 4 mm, packed with 1 ml of 0.1 M perchloric acid is equivalent to 0.01686 g of
endcapped octadecylsilane bonded to porous silica C1zH 17CIN40S, HCl.
(7Ilm), Storage. Store protected from light and moisture, non-metallic
column temperature. 45°, containers.
mobile phase: A. 0.38 per cent w/v solution of sodium
hexanesulphonate, adjusted to pH 3.1 with
orthophosphoric acid,
B. methanol, Thiamine Injection
a linear gradient programme using the conditions given
below, Thiamine Hydrochloride Injection; Aneurine Hydro-
flow rate. 1 ml per minute, chloride Injection; Vitamin B 1 Injection
spectrophotometer set at 248 nm,
Thiamine Injection is a sterile solution of Thiamine Hydro-
injection volume. 25 Ill. chloride in Water for Injection.
Time Mobile phase A Mobile phase B
Thiamine Injection contains not less than 95.0 per cent and
(in min.) (per cent v/v) (per cent v/v)
not more than 110.0 per cent ofthe stated amount of thiamine
0-25 90 -"170 10 -"130 hydrochloride, C 1zH 17CIN40S, HCl.
25 - 33 70 -"150 30 -"150
Usualstrength.100mgpermi.
33 -40 50 50
40-45 50 -"190 50 -"110 Identification
Inject reference solution (a). The test is not valid unless the A. To a volume containing 20 mg ofThiamine Hydrochloride
resolution between the peaks due to thiamine impurity A and in 10 ml of water, add 1 ml of 2 M acetic acid and 1.6 ml of
thiamine is not less than 1.6. 1 M sodium hydroxide, heat on a water-bath for 30 minutes
and allow to cool. Add 5 ml of2 M sodium hydroxide, 10 ml of
Inject the test solution and reference solution (b). In the
potassium ferricyanide solution and 10 ml of I-butanol and
chromatogram obtained with the test solution, the area of any
shake vigorously for 2 minutes. The upper layer exhibits an
secondary peak is not more than the area ofthe principal peak
intense light blue fluorescence, particularly in ultraviolet light
in the chromatogram obtained with reference solution (b)
at 365 nm. Repeat the test but adding 0.9 m1 of 1 M sodium
(0.4 per cent). The sum of all the secondary peaks is not more
hydroxide and 0.2 g ofsodium sulphite in place of the 1.6 ml
than2.5times the area ofthe principalpeak inthe chromatogram
obtained with reference solution (b) (1.0 per cent). Ignore any
of 1 MsoClTlini liyCl,;oxICle; practically no flu()res~cence is
peak with an area less than 0.125 times the area ofthe principal
prOduced.·
peak in the chromatogram obtained with reference B. To a mixture of0.1 ml ofnitrobenzene and 0.2 ml ofsulphuric
solution (b) (0.05 per cent). acidadd a volume containing 5 mg ofThiamine Hydrochloride.

2210

www.webofpharma.com
IP 2010 THIAMINE TABLETS

Allow to stand for 10 minutes, cool in ice and add slowly with 2 minutes. The upper layer exhibits an intense light blue
stirring 5 ml of water followed by 5 ml of 10M sodium fluorescence, particularly in ultraviolet light at 365 nm. Repeat
hydroxide. Add 5 ml of acetone and allow to stand; no violet the test but adding 0.9 ml of 1 M sodium hydroxide and 0.2 g
colour is produced in the upper layer. of sodium sulphite in place of the 1.6 ml of 1 M sodium
hydroxide; practically no fluorescence is produced.
Tests
B. To a mixture of0.1 ml of nitrobenzene and 0.2 ml ofsulphuric
pH (2.4.24).2.5 to 4.5. acid add the powdered tablets containing 5 mg of Thiamine
Other tests. Complies with the tests stated under Parenteral Hydrochloride. Allow to stand for 10 minutes, cool in ice and
Preparations (Injections). add slowly with stirring 5 ml of water followed by 5 ml of
10M sodium hydroxide. Add 5 ml of acetone and allow to
Assay. Determine by liquid chromatography (2.4.14). stand; no violet colour is produced in the upper layer.
Test solution. Dilute a volume ofthe injection containing about C. The powdered tablets give the reactions ofchlorides (2.3.1).
0.1 g of Thiamine Hydrochloride to 100.0 ml with
0.1 M hydrochloric acid and further dilute 5.0 ml to 100.0 ml Tests
with water.
Uniformity of content. (For tablets containing 10 mg or less)
Reference solution. A 0.005 per cent w/v solution of thiamine - Comply with the test stated under Tablets.
mononitrate RS in 0.005 M hydrochloric acid.
Finely crush one tablet, add 20 ml of ethanol (95 per cent),
Chromatographic system stir the mixture for 30 minutes and centrifuge. Repeat the
- a stainless steel column 10 cm x 4.6 mm, packed with extraction with three further quantities, each of 15 ml, of
octadecylsilane bonded to porous silica (5 /lm), ethanol (95 per cent). Combine the extracts, add sufficient
- mobile phase: a solution prepared by dissolving 1 g of ethanol (95 per cent) to produce 100.0 ml and mix; Dilute a
sodium heptanesulphonate in a mixture of 180 ml of suitable volume of the resulting solution containing 1 mg of
methanol and. 10 ml of triethylamine, diluting to 1000 ml Thiamine Hydrochloride with sufficient ethanol (95 per cent)
with water and adjusting the pH to 3.2 with to produce 100.0 m!. Measure the absorbance ofthe resulting
orthophosphoric acid, solution at the maximum at about 233 nm (2.4.7).
- flow rate. 2 ml per minute,
Calculate the content ofC 1zH 17C1N40S, HCI in the tablet taking
- spectrophotometer set at 244 nm,
380 as the specific absorbance at 233 nm.
- injection volume. 20 Ill.
Other tests. Comply with the tests stated under Tablets.
Calculate the content ofC ,zH 17ClN40S.
Assay. Determine by liquid chromatography (2.4.14).
1 mg ofC 12H 17Ns0 4S is equivalent to 1.030 mg ofC 12H 17C1N40S,
HCl. Test solution. For tablets containing less than 10 mg of
Thiamine Hydrochloride, add 5 ml of 0.1 Mhydrochloric acid
Storage. Store protected from light.
and 50 ml of water to a quantity of the powdered tablets
containing 6 mg of Thiamine Hydrochloride, shake for
20 minutes, dilute to 100.0 ml with water and filter. For tablets
Thiamine Tablets containing 10 mg or more of Thiamine Hydrochloride, add
50 ml of 0.1 M hydrochloric acid and 500 ml of water to a
Thiamine Hydrochloride Tablets; Anemine Hydrochloride quantity ofthe powdered tablets containing 60 mg ofThiamine
Tablets; Vitamin ~1 Tablets Hydrochloride, shake for 20 minutes, dilute to 1000.0 ml with
Thiamine Tablets contain not less than 92.5 per cent and not water and filter.
more than 110.0 per cent of the stated amount of thiamine Reference solution. A 0.006 per cent w/v solution of thiamine
hydrochloride, C 1zH 17CIN40S, HCl. mononitrate RS in 0.005 M hydrochloric acid.
Usual strengths. 5 mg; 10 mg; 50 mg. Chromatographic system
- a stainless steel column 10 cm x 4.6 mm, packed with
Identification
octadecylsilane bonded to porous silica (5 /lm),
A. Dissolve a quantity of the powdered tablets containing - mobile phase: a solution prepared by dissolving 1 g of
20 mg ofThiamine Hydrochloride as completely as possible in sodium heptanesulphonate in a mixture of 180 ml of
10 ml of water and 2 ml of 1 M acetic acid and filter. Add 5 ml methanol and 10 ml of triethylamine, diluting to 1000 ml
of 2 M sodium hydroxide, 10 ml of potassium ferricyanide with water and adjusting the pH to 3.2 with
solution and 10 ml of 1-butanol and shake vigorously for orthophosphoric acid,

2211

www.webofpharma.com
THIAMINE MONONITRATE IP20l0

flow rate. 2 ml per minute, 0.2 g of sodium sulphite in place ofthe 1.6 ml of I M sodium
spectrophotometer set at 244 nm, hydroxide; practically no fluorescence is produced.
- injection volume. 20 f..tl.
C. Gives reaction A ofnitrates (2.3.1).
Calculate the content ofC 1zH 17ClNPS in the tablets.
Tests
1mg OfCIZH17Ns04S is equivalent to 1.030 mg ofC 1zH 17ClN40S,
HCI. Appearance of solution. A2.0 per cent w/v solution in carbon
Storage. Store protected from light and moisture in non-metallic dioxide-free water is clear (2.4.1), and not more intensely
containers. coloured than reference solution YS7 (2.4.1).
pH (2.4.24). 6.8 to 7.6, determined in a 2.0 per cent w/v solution.
Related substances. Determine by liquid chromatography
Thiamine Mononitrate (2.4.14).

Thiamine Nitrate Solvent mixture. 5 volumes of glacial acetic acid and 95


volumes of water.
Tes.t solution. Dissolve 0.35 g of the substance under
examination in 15.0 ml of the solution mixture and dilute to
100.0 ml with water.
Reference solution (a). Dissolve 5 mg each of the substance
under examination and thioxothiamine RS (thiamine impurity
A RS) in 4 ml ofthe solvent mixture and dilute to 25.0 ml with
ClzH17Ns04S Mol. Wt. 327.4 water. Dilute 5.0 ml ofthe solution to 25.0 ml with water.
Thiamine Mononitrate is 3-[(4-amino-2-methylpyrimidin- Reference solution (b). Dilute 1.0 ml of the test solution to
5-yl)methyl]-5-(2-hydroxyethyl)-4-methylthiazolium nitrate. 100.0 ml with water.
Thiamine Mononitrate contains not less than 98.0 per cent Chromatographic system
and not more than 102.0 per cent of ClzHI7Ns04S, calculated - a stainless steel column 25 cm x 4 mm, packed with
on the dried basis. endcapped octadecylsilane bonded to porous silica
Category. B-group vitamin. (5f..tm),
column temperature. 45°,
Dose. Prophylactic, 2 to 5 mg daily; therapeutic, 25 to 100 mg mobile phase: A. 0.38 per cent w/v solution of sodium
daily. In multivitamin oral preparations, prophylactic, hexanesulphonate, adjusted to pH 3.1 with
1 to 2 mg daily; therapeutic, 4.5 to 10 mg daily. orthophosphoric acid,
Description. A white or almost white, crystalline powder or B. methanol,
small, colourless crystals. a linear gradient programme using the conditions given
below,
Identification flow rate. 1 ml per minute, .
Test A may be omitted if tests Band C are carried out. Test B spectrophotometer set at 248 nm,
may be omitted if tests A and C are carried out. injection volume. 25 pI.
Time Mobile phase A Mobile phase B
A. Determine by infrared absorption spectrophotometry (2.4.6).
(min.) (per cent v/v) (per cent v/v)
Compare the spectrum with that obtained with thiamine
mononitrate RS or with the reference spectrum of thiamine 0-25 90 ~70 10 ~30
mononitrate. 25-33 70 ~50 30 ~50
B. Dissolve about 20 mg in 10 ml of water, add 1 ml of 33-40 50 . 50
2 M acetic acid and 1.6 ml of I M sodium hydroxide, heat on 40-45 50 ~90 50 ~IO
a water-bath for 30 minutes ~nd allow to cool. Add 5 ml of
Inject reference solution (a). The test·is not valid unless the
2 M sodium hydroxide, 10mi of potassium,ferxicyanide
res61Uti6iibet:We'en tliepeiik:fdue t6tliiiimiiie impuritY Aiiiid
solution and 10 ml of I-butanol and shake vigorously for
thiamine is not less than 1.6.
2 minutes. The upper layer exhibits an intense light blue
fluorescence, particularly in ultraviolet light at 365 nm. Repeat Inject the test solution and reference solution (b). In the
the test but adding 0.9 ml of I M sodium hydroxide and chromatogram obtained with the test solution, the area ofany

2212

www.webofpharma.com
IP 2010 THIOCOLCHICOSIDE

secondary peak is not more than the area ofthe principal peak Identification
in the chromatogram obtained with reference solution (b)
A. Determine by infrared absorption spectrophotometry (2.4.6).
(1.0 per cent). The sum ofall the secondary peaks is not more
Compare the. spectrum with that obtained with
than 1.5 times the area ofthe principal peak in the chromatogram
thiocolchicoside RS or with the reference spectrum of
obtained with reference solution (b) (1.5 per cent). Ignore any
thiocolchicoside.
peak with an area less than 0.05 times the area ofthe principal
peak in the chromatogram obtained with reference solution B. When examined in the range of220 run to 440 nm (2.4.7), a
(b) (0.05 per cent). 0.0015 per cent w/v solution in water shows absorption maxima
at the same wavelength as obtained with the reference
Heavy metals (2.3.13). 1.0 g complies with limit test for heavy solution.
metals, Method A (20 ppm).
Tests
Chlorides (2.3.12). 1.0 g complies with the limit test for chlorides
(250 ppm). pH (2.4.24).6.0 to 7.5, determined in a 0.5 per cent w/v solution
in carbon-dioxide free water.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Specific optical rotation (2.4.22). - 550° to - 580° at 23°,
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
determined on 0.5 per cent w/v solution in water.
on 1.0 g by drying in an oven at 105°.
Related substances. Determine by liquid chromatography
Assay. Weigh accurately about 0.15 g, dissolve in 5 ml of (2.4.14).
anhydrousformic acid, add 70 ml of acetic anhydride. Titrate
Test solution. Dissolve about 50 mg of the substance under
with 0.1 M perchloric acid, determining the end-point
examination in 50.0 ml of methanol.
potentiometrically (2.4.25). Carry out a blank titration.
Reference solution (a). A 0.05 per cent w/v solution of
1 ml of 0.1 M perchloric acid is equivalent to 0.01637 g of
colchicoside RS in methanol.
ClzHI7Ns04S.
Reference solution (b). A 0.05 per cent w/v solution of
Storage. Store protected from light and moisture, non-metallic thiocolchicoside RS in methanol.
containers.
Reference solution (c). Dilute 1.0 m1 each ofreference solution
(a) and (b) to 100.0 m1 with methanol.
Chromatographic system
- a stainless steel column 25 cm x 4.6 rom, packed with
Thiocolchicoside
silica (5 flm),
mobile phase: A. n- heptane containing 2 per cent acetic
OH acid,

~.
0 HsCO ?"
B. chloroform,
HO
HO ~
I C. methanol,
- a linear gradient programme using the conditions given
OH 0 below,
HsCO flow rate. 0.8 ml per minute,
o spectrophotometer set at 360 run,
- injection volume. 20 fll.
SCH s
Time Mobile Mobile Mobile
phase A phase B phase C
Mol.Wt. 563.5 (in min.) (per cent v/v) (per cent v/v) (per cent v/v)
Thiocolchicoside is 2-demethoxy-2-g1ucosidoxythio- o 86 10 4
colchicine. 50 o 65 35
Thiocolchicoside contains not less than 98.0 per cent and not 51 86 10 4
more than 102.0 per cent of CZ7H33NOIOS, calculated on the 60 86 10 4
dried basis.
Inject reference solution (c). The test is not valid unless the
Description. A yellow crystalline powder. theoretical plates is not less than 80000, the tailing factor is
Category. Muscle relaxant. not more than 1.5 and the relative standard deviation for
replicate injections is not more than 2.0 per cent for the peak
Dose. 4 mg to 8 mg daily. due to thiocolchicoside.

2213

www.webofpharma.com
THIOCOLCHICOSIDE IP 2010

The relative retention time with reference to thiocolchicoside Inject the reference solution. The test is not valid unless the
for colchicine is about 0.55, for N~ deacetyl N- formyl relative standard deviation for replicate injections is not more
thiocolchicoside is about 1.05 and for colchicoside is than 2.0 per cent. .
about 1.10.
Inject the test solution and the reference solution.
Inject the test solution and reference solution (c). In the
chromatogram obtained with the test solution the area ofeach Calculate content ofC27H33NOIOS.
peak corresponding to N-deacetyl N-formyl thiocolchicoside Storage: Store protected from light and at a temperature not
and colchicoside is not more than the area of corresponding exceeding 30°.
peak in the chromatogram obtained with reference solution
(c) (0.5 per cent) and the sum ofthe areas ofall other secondary
peaks is not more than the area ofpeak due to thiocolchicoside Thiocolchicoside Capsules
in the chromatogram obtained with reference solution (c)
(0.5 per cent). Thiocolchicoside Capsules contain not less than 90.0 per cent
and not more than 110.0 per cent of the stated amount of
Sulphated ash (2.3.18). Not more than 0.1 per cent.
thiocolchicoside, C27H33NOIOS.
Loss on drying (2.4.19). Not more than 4.0 per cent, determined
on 1.0 g by drying in oven at 105° for 3 hours. Dose. 4 mg; 8 mg.

Microbial contamination (2.2.9). Total microbial count not more Identification


than 1000 CFU per g, total yeast and mould count not more
than 100 CFU per g, 1 g is free from Escherichia coli, A. In the Assay, the principal peak in the chromatogram
Salmonella, Staphylococcus aureus, Pseudomonas obtained with the test solution corresponds to the principal
aeruginosa and Enterobacteriaceae. peak in the chromatogram obtained with the reference solution.
Bacterial endotoxins (2.2.3). Not more than 87.5 Endotoxin B. Examine by thin-layer chromatography (2.4.17), coating the
Units per mg ofThiocolchicoside. plate with silica gel G.
Assay. Determine by liquid chromatography (2.4.14). Mobile phase. A mixture of 70 volumes of ethyl acetate, 20
Test solution. Dissolve 0.1 g of substance under examination volumes of ethanol and 10 volumes of water.
in 100.0 ml of water. Dilute 10.0 ml ofthis solution to 100.0 ml Test solution. Dissolve 50 mg of the substance under
with watel: examination in 5.0 ml of methanol.
Reference solution. A 0.1 per cent w/v solution of Reference solution. A 1.0 per cent w/v solution of
thiocolchicoside RS in water. Dilute 10.0 ml ofthis solution to thiocolchicoside RS in methanol.
100.0ml with watel:
Apply to the plate 10 ml of each solution. After development,
Chromatographic system dry the plate in a current ofair and examine in ultraviolet light
a stainless steel column 25 cm x 4.6 mm, packed with at 254 nm. The principal spot in the chromatogram obtained
octylsilane bonded to porous silica (5 Ilm), with the test solution corresponds to the chromatogram
mobile phase: A. watel; obtained with the reference solution.
B. acetonitrile,
a linear gradient programme using the conditions given Tests
below,
spectrophotometer set at 370 nm, Dissolution (2.5.2).
- injection volume. 20 Ill. Apparatus No.1,
Time Flow Mobile Phase A Mobile Phase B Medium. 900 ml ofphosphate bufferpH 7.5,
(in min.) (rnlImin.) (per cent lv/v) (per cent v/v) Speed and time. 100 rpm and 45 minutes.
o 1.0 90 10 Withdraw a suitable volume ofthe medium and filter.
10 1.0 90 10 Detelmine by liquid chromatography (2.4.14).
11 1.0 75 25
U 1~ ~ 25 Test solution. Use the filtrate.
___. 12_ J--,-~._ _72 _ ------~­ Reference solution, A 0,-001 per cent w/v solution of
17 1.8 70 30 thiocolchicoside RS in water.
18 1.8 70 30
20 1.8 90 10 Use the chromatographic condition as described under Assay.
22 1.0 90 10 Calculate the content ofC27H33NOIOS.

2214

www.webofpharma.com
IP 2010 THIOMERSAL

D. Not less than 80 per cent of the stated amount of Other tests. Comply with the tests stated under Capsules.
C:17H33NO lOS.
Assay. Determine by liquidchromatography (2.4.14).
Related substances. Detennine by liquid chromatography
Test solution. Shake a quantity of the content of 20 capsules
(2.4.14).
containing about 10 mg ofThocolchicoside with 100.0 nil of
Test solution. Disperse the content of the capsules containing water.
about 50 mg ofThiocolchicoside in 50.0 ml of methanol.
Reference solution. A 0.01 per cent w/v solution of
Reference solution (a). A 0.05 per cent w/v solution of thiocolchicoside RS in water.
colchicoside RS in methanol.
Chromatographic system
Reference solution (b). A 0.05 per cent w/v solution of - a stainless steel column 25 cm x 4.6 mm, packed with
thiocolchicoside RS in methanol. octylsilane bonded to porous silica (5 /-tm),
Reference solution (c). Dilute 1.0 ml each ofreference solution mobile phase: A. water,
(a) and (b) to 100.0 ml with methanol. B. acetonitrile,
- a linear gradient programme using the conditions given
Chromatographic system
below,
- a stainless steel column 25 cm x 4.6 mm, packed with
- spectrophotometer set at 370 nm,
silica (5 /-tm),
- injection volume. 20 /-tl.
- mobile phase: A. n- heptane containing 2 per cent acetic
acid, Time Flow Mobile Phase A Mobile Phase B
B. chloroform, (in min.) (ml/min.) (per cent/v/v) (per cent v/v)
C. methanol, om 1.0 90 10
- a linear gradient programme using the conditions given 10 Lo 90 10
below,
flow rate. 0.8 ml per minute,
11 1.0 75 25
spectrophotometer set at 360 nm, 12 1.8 75 25
- injection volume. 20 /-tl. 15 1.8 75 25
Time Mobile Mobile Mobile 17 1.8 70 30
phase A phase B phase C 18 1.8 70 30
(in min.) (per cent v/v) (per cent v/v) (per cent v/v)
20 1.8 90 . 10
o 86 10 4
22 1.0 90 10
50 o 65 35
51 86 10 4 Inject the reference solution. The test is not valid unless the
relative standard deVIation for replicate injections is not more
60 86 10 4
than 2.0 per cent.
Inject reference solution (c). The test is not valid unless the
theoretical plate is not less than 80000, the tailing factor is not Inject the test solution and the reference solution.
more than 1.5 and the relative standard deviation for replicate Calculate the content ofC 27H 33NO IO S in the capsules.
injections is not more than 2.0 per cent for the peak due to
thiocolchicoside.
The relative retention time with reference to thiocolchicoside
for colchicine is about 0.55, for N- deacetyl N- formyl Thiomersal
thiocolchicoside is about 1.05 and for colchicoside is about
1.10. Thimerosal

Inject the test solution and reference solution (c). In the


SHgCH 2CH 3
chromatogram obtained with the test solution the area ofeach
peak corresponding to N-deacetyl N-fonnyl thiocolchicoside ~COONa
and colchicoside is not more than the area of corresponding
peak in the chromatogram obtained' with reference solution
(c) (0.5 percent) and the sum ofthe areas ofall other secondary
V
C9H9HgNa02S Mol. Wt. 404.8
peaks is not more than the area ofpeak due to thiocolchicoside
in the chromatogram obtained with reference solution (c) (0.5 Thiomersal is the sodium salt of [(2-carboxyphenyl)thio]
per cent). ethylmercury.

2215

www.webofpharma.com
THIOMERSAL IF 2010

Thiomersal contains not less than 97.0 per cent and not more Assay. Weigh accurately about 0.5 g, transfer to a 100-mllong-
than 101.0 per cent ofC9H9HgNaOzS, calculated on the dried necked flask, add 5 ml of sulphuric acid and heat gently until
basis. charring occurs; continue to heat and add hydrogen peroxide
solution (l00 vol) dropwise, until the mixture is colourless.
Category. Antiseptic; pharmaceutical aid (antimicrobial
Dilute with water, evaporate until slight fuming occurs, dilute
preservative).
to 10 ml with water, cool and titrate with 0.1 M ammonium
Description. A light cream, crystalline powder; odour, slight thiocyanate using ferric ammonium sulphate solution as
and characteristic. indicator.
1 ml of 0.1 M ammonium thiocyanate is equivalent to
Identification 0.02024 g ofC;H9HgNaOzS.
A. Dissolve 0.1 g in 10 ml of water and add 2 ml of silver Storage. Store protected from light and moisture.
nitrate solution; a pale yellow precipitate is produced.
B. Dissolve 0.5 g in 10 ml of water and add 2 ml of2 Mhydro-
chloric acid; a white precipitate is produced 'which, after
washing with water and drying over phosphorus pentoxide Thiopentone Sodium
at a pressure not exceeding 0.7 kPa, melts at about 110°
(2.4.21). Thiopental Sodium

Tests
pH (2.4.24). 6.0 to 8.0, determined in a 1.0 per cent w/v solution.
Ether-soluble matter. Shake 0.5 g with 20 ml of ether for
10 minutes, filter and evaporate to dryness. The residue, after
drying over phosphoruspentoxide at a pressure not exceeding
0.7 lePa for 24 hours, weighs not more thEm 3 mg.
Mol. Wt. 264.3
Inorganic mercury compounds. Not more than 0.7 per cent,
determined by the following method. Thiopentone Sodium is a mixture ofsodium (RS)-5-ethyl-
5-(1-methylbutyl)-2-thiobarbiturate and anhydrous sodium
NOTE-Protect the solutions from light.
carbonate.
Label five 10-ml volumetric flasks A, B, C, D and E. Place 5 rn1
Thiopentone Sodium contains not less than 84.0 per cent and
of a 0.1 per cent w/v solution of the substance under
not more than 87.0 per cent ofCjjHjsNzOzS and not less than
examination in each offlasks A, B, C and D. To each offlasks
10.2 per cent and not more than 11.2 per cent of Na, both
C and D add 0.5 ml ofa 0.0095 per cent w/v solution of mercuric
calculated on the dried basis.
chloride. Add sufficient water to flasks A and C to produce
10.0 ml and add sufficient of a freshly prepared 33.2 per cent Category. General anaesthetic.
w/v solution ofpotassium iodide to flasks Band D to produce Dose. By intravenous injection, initially, 100 to 500 mg, repeated
10.0 ml. Place 5 ml ofthe potassium iodide solution in flask E if necessary after 20 to 30 seconds; maximum dose, 2 g.'
and add sufficient water to produce 10.0 ml. Measure the
absorbances of each of the solutions (Aa, Ab, Ac, Ad, Ae) at Description. A yellowish white powder; odour, faintly
about 323 nm (2.4.7), using water as the blank. resembling garlic; hygroscopic.

Calculate the content of inorganic mercury compounds, Identification


expressed as Hg, in the substance under examination from the
expression 0.7019 (Ab -Aa -Ae)/(Aa + Ad -Ab -Ac), where Test A may be omitted if tests B, C, D and E are carried out.
the figure 0.7019 is a constant obtained from the formula Tests Band D may be omitted if tests A, C and E are carried
out.
atomic weight of mercury concentration of HgCl z A. AcidifY 10 rn1 ofa 10 per cent w/v solution in carbon dioxide-
-----=-------=--x --------=--=-
molecular weight of HgCl z 100 free water with 2 M hydrochloric acid; the solution
effervesces. Shake the solution with 20 ml of ether, separate
Loss on drying (2.4.19). Not more than 0.5 per cent, determined the ether layer, wash with 10 ml of water and dry over
on 1.0 g by drying over phosphorus pentoxide at a pressure anhydrous sodium sulphate. Filter, evaporate the filtrate to
not exceeding 0.7 kPa for 24 hours. dryness and dry the residue at 105°.

2216

www.webofpharma.com
IP 2010 THIOPENTONE INJECTION

On the residue, determine by infrared absorption 1 ml of 0.1 M lithium methoxide is equivalent to 0.02423 g of
spectrophotometry (2.4.6). Compare the spectrum with that CIIHISN202S.
obtained with thiopentone RS or with the reference spectrum
For sodium - Weigh accurately about 0.4 g, dissolve in 30 ml
of thiopentone.
of water, add 1 drop of methyl red solution and titrate with
B. Complies with the test for identification of barbiturates 0.05 M sulphuric acid until the yellow colour changes to red.
(2.3.2), but using the following solutions. Boil gently for 2 minutes, cool and, ifnecessary, continue the
titration with 0.05 M sulphuric acid until the red colour is
Test solution. A 0.1 per cent w/v solution of the substance
restored.
under examination in water.
1 ml of 0.05 M sulphuric acid is equivalent to 0.002299 g of
Reference solution. Dissolve 85 mg of thiopentone RS in
Na.
10 ml of2 M sodium hydroxide and dilute to 100 ml with water.
Storage. Store protected from light and moisture.
C. Acidify 5 ml of a 5 per cent w/v solution with dilute acetic
acid and filter. Wash the precipitate with water, recrystallise
from water and dry at 700; the crystals melt at about 1600
(2.4.21).
D. Dissolve 1 mg of the crystals obtained in test A in 1 ml of Thiopentone Injection
0.1 M sodium hydroxide. Add about 1 mg of sodium Thiopentone Sodium Injection; Thiopental Injection
nitroprusside and, after 15 minutes, 1 ml of dilute hydrochloric
acid; a reddish violet colour is produced. Thiopentone Injection is a sterile material consisting of
Thiopentone Sodium with or without auxiliary agents. It is
E. Gives the reactions of sodium salts (2.3.1).
filled in a sealed container.
Tests The injection is constituted by dissolving the contents of the
Appearance ofsolution. A 10.0 per cent w/v solution in carbon sealed container in the requisite amount of sterile Water for
dioxide-free water (solution A)is clear (2.4.1), and not more Injections, immediately before use.
intensely coloured than reference solution GYS3 (2.4.1). The constituted solution complies with the requirements for
Related substances (2.3.4). Complies with the test, but using Clarity of solution and Particulate matter stated under
water as the solvent for the test solution and the reference Parenteral Preparations (Injections).
solution. After development, examine the plate in ultraviolet Storage. The constituted solution should be used immediately
light at 254 om but do not spray it with the diphenylcarbazone- after preparation but, in any case, within the period
mercury reagent. recommended by the manufacturer.
Chlorides (2.3.12). To 10 ml ofsolutionAadd 30 ml of water Thiopentone Injection contains thiopentone, CIIHlSN202S that
and 10 ml of 2 M nitric acid. Shake successively with three is not less than 77.0 per cent and not more than 92.0 per cent
quantities, each of25 ml, of ether, discard the ether lay~rs and and sodium, Na that is not less than 9.4 per cent and not more
heat the aqueous solution on water-bath to remove any than 11.8 per cent ofthe stated amount ofthiopentone sodium.
residual ether. 30 ml of the aqueous layer complies with the
limit test for chlorides (330 ppm). The contents of the sealed container comply with the
requirements stated under Parenteral Preparations
Loss on drying (2.4.19). Not more than 2.5 per cent, determined (Powders for Injection) and with the following requirements.
on 0.5 g by drying in an oven at 1000 at a pressure of 1.5 to
2.5 kPa for 4 hours. Usual strengths. 250mg; 500 mg; 1 g; 5 g.

Assay. For thiopentone - Weigh accurately about 0.15 g, Description. A yellowish white powder; hygroscopic.
dissolve in 5 ml of water, add 2 ml of 1 M sulphuric acid and
extract with four quantities, each of 10 ml, of chloroform. Filter Identification
the combined chloroform extracts, evaporate the filtrate to
Test A may be omitted if tests B, C, D and E are carried out.
dryness on a water-bath and dissolve the residue in 30 ml of
Tests Band D may be omitted if tests A, C and E are carried
dimethylformamide, previously neutralised with 0.1 M lithium
out.
methoxide. Titrate immediately with 0.1 Mlithium methoxide,
using 1 drop of a 0.2 per cent w/v solution of thymol blue in A. Acidify 10 ml ofa 10 per cent w/v solution in carbon dioxide-
methanol as indicator, until a blue colour is obtained. Protect free water with 2 M hydrochloric acid; the solution
the solution from absorption of carbon dioxide during the effervesces. Shake the solution with 20 ml of ether, separate
titration. the ether layer, wash with 10 ml of water and dry over

2217

www.webofpharma.com
THIOPENTONE INJECTION IP 2010

anhydrous sodium sulphate. Filter, evaporate the filtrate to For sodium - Mixed the contents of 10 containers and weigh
dryness and dry the residue at 105°. accurately about 0.4 g of the contents, dissolve in 30 ml of
water, add 1 drop of methyl red solution and titrate with
On the residue, determine by infrared absorption
0.05 M sulphuric acid until the yellow colour changes to red.
spectrophotometry (2.4.6). Compare the spectrum with that
Boil gently for 2 minutes, cool and, if necessary, continue the
obtained with thiopentone RS or with the reference spectrum
titration with 0.05 M sulphuric acid until the red colour is
of thiopentone.
restored
B. .Complies with the test for identification of barbiturates
1 ml of 0.05 M sulphuric acid is equivalent to 0.002299 g of
(2.3.2), but using the following solutions.
Na.
Test solution. 0.1 per cent w/v solution of the substance
Storage. Store in single dose containers.
under examination in water.
Labelling. The label states the amount of active ingredient in
Reference solution. Dissolve 85 mg of thiopentone RS in
terms of Thiopentone Sodium.
10 ml of2 M sodium hydroxide and dilute to 100 ml with water.
C. Acidify 5 ml ofa 5 per cent w/v solution with dilute acetic
acid and filter. Wash the precipitate with water, recrystallise
from water and dry at 70°; the crystals melt at about 160° Thiotepa
(2.4.21).
D. Dissolve 1 mg of the crystals obtained in test A in 1 mlof s
0.1 M sodium hydroxide. Add about 1 mg of sodium [:::N-~-N::J
I
nitroprusside and, after 15 minutes, 1 ml of dilute hydrochloric
acid; a reddish violet colour is produced.
N
D
E. Gives the reactions of sodium salts (2.3.1).
C6H I2N3PS Mol. Wt. 189.2
Tests Thiotepa is tris(l-aziridinyl)phosphine sulphide.

Appearance ofsolution. A 10.0 per cent w/v solution in carbon Thiotepa contains not less than 97.0 per cent and not more
dioxide-free water (solution A) is clear (2.4.1), and not more than 102.0 per cent ofC 6H I2N 3PS, calculated on the anhydrous
intensely coloured than reference solution GYS3 (2.4.1). basis.
Related substances (2.3.4). Complies with the test, but using Category. Anticancer.
water as the solvent for the test solution and the reference Dose. By injection, 300 to 400 ~g per kg of body weight at
solution. After development, examine the plate in ultraviolet intervals of 1 to 4 weeks.
light at 254 urn but do not spray it with the diphenylcarbazone-
mercUlY reagent. Description. Fine, white, crystalline flakes; odourless or almost
odourless.
Loss on drying (2.4.19). Not more than 2.5 per cent, determined
on 0.5g by drying in an oven at 100° at a pressure of 1.5 to
Identification
2.5 kPa for 4 hours.
Assay. For thiopentone - Mixed the contents of! 0 containers A. Determine by infrared absorption spectrophotometry (2.4.6).
and weigh accurately about 0.15 g, of the contents, dissolve Compare the spectrum with that obtained with thiotepa RS or
in 5 ml of water, add 2 ml of 1 M sulphuric acid and extract with the reference spectrum of thiotepa.
with four quantities, each of 10 ml, of chloroform. Filter the B. Determine on 20 mg by the oxygen-flask method (2.3.34),
combined chloroform extracts, evaporate the filtrate to dryness using 5 ml of 1.25 M sodium hydroxide as the absorbing
on a water-bath and dissolve the residue in 30 mlof liquid. When the process is complete, dilute to 25 ml with
dimethylformamide, previously neutralised with 0.1 M lithium water (solution A). To 5 ml ofsolutionAadd 0.1 mlofhydrogen
methoxide. Titrate immediately with 0.1 M lithium methoxide, peroxide solution (100 vol) and 1 ml of 1 M hydrochloric
using 1 drop of a 0.2 per cent w/v solution of thymol blue in acid, mix and add 0.05 ml of barium chloride solution; a
methanol as indicator, until a blue colour is obtained. Protect turbidity is produced.
the solution from of carbon dioxide dl.ifiiig:the
' __ __"__'''.' __.,._ _'.' ',_..' ..'.'_'_.. __"._,.·, __._, ,_. _., ,.". '''_, __

abso~rptioii
'
_
~
_
"
~
'
_
~
'
,
.
_
.
,
~
,
_
·
,
~
,
_
.
M
.
~
_
.
,
_
,
_
,
_
.
_
,
_
,
'
'
'
'
,
~
,
C. To 2ml ofsolutionAadd40 mlofwater and 4 ml ofammonium
titrati611.
molybdate-sulphuric acid solution, mix, add 0.1 g of
1 ml of 0.1 M lithium methoxide is equivalent to 0.02423 g of L-ascorbic acid and boil for 1 minute; a blue colour is
CIIHISN202S. produced.

2218

www.webofpharma.com
IP 2010 THIOTEPA INJECTION

Tests and titrate immediately with 0.1 M hydrochloric acid, using


methyl orange solution as indicator, until a faint red colour
Appearance of solution. A 2.0 per cent w/v solution is clear persists for 10 seconds. Stopper the flask, aUow to stand for
(2.4.1). 30 minutes and titrate with 0.1 M sodium hydroxide using
Related substances. Determine by liquid chromatography phenolphthalein solution as indicator. Subtract the volume
(2.4.14). of 0.1 M sodium hydroxide used from the volume of 0.1 M
hydrochloric acid used. Repeat the operation without the
Test solution. Dissolve 0.35 g of the substance under
substance under examination. The difference between the
examination 100.0 ml ofthe water.
titrations represents the amount of hydrochloric acid required.
Reference solution (a). Dissolve 0.35 mg of the substance
1 ml of 0.1 M hydrochloric acid is equivalent to 0.006307 g of
under examination in 100.0 ml ofthe water.
C6H I2N3PS.
Reference solution (b). Dissolve 10 mg of the substance
Storage. Store protected from light and moisture. At higher
under examination in 2 ml of methanol in a ground-glass-
temperatures it tends to polymerise with loss of activity.
stoppered tube, add 50 III of a 0.1 per cent v/v solution of
orthophosphoric acid, stopper the tube and heat in a water
bath at.65° for 50 seconds (generation of methoxythiotepa).
Allow the solution to cool and add 1 ml of methanol. . Thiotepa Injection
Reference solution (c). Dissolve 15 mg of the substance Thiotepa Injection is a sterile material consisting of Thiotepa
under examination in 10 ml of water, add I g of sodium with or without auxiliary agents. It is filledin a sealed container.
chloride, boil in a water bath for 10 minutes and cool
The injection is constituted by dissolving the contents of the
(generation of chloro- adduct).
sealed container in the requisite amount of sterile Water for
Chromatographic system Injections, immediately before use.
a stainless steel column 15 cm x 4.6 mm, packed with
The constituted solution complies with the requirements for
endcapped octadecylsilane bonded to porous silica
Clarity of solution and Particulate matter stated under
(5 11m) (Such as Nucleosil CI8),
Parenteral Preparations (Injections).
- mobile phase: a mixture of IS volumes of acetonitrile.
and 85 volumes of 0.1 M phosphate buffer pH 7.0, Storage. The constituted solution should be used immediately
- flow rate. 1 mlperminute, . after preparation but, in any case, within the period
- spectrophotometer set at 215 nm, recommended by the manufacturer.
- injection volume. 20 Ill. Thiotepa Injection contains not less than 90.0 per cent and
Inject reference solution (b) and (c). The test is not valid unless not more than 110.0 per cent ofthe stated amount ofthiotepa,
the resolution between the two principal peaks is not less C6HI2N3PS.
than 3. The relative retention time with reference to thiotepa The contents of the sealed container comply with the
for methoxythiotepa is about 1.3 and for chloro-adduct requirements stated under Parenteral Preparations
(thiotepa impurity A) is about 3.75. (Powders for Injection) and with the following requirements.
Inject the test solution and reference solution (a). Run the Usual strength. 15 mg.
chromatogram 4 times the retention time ofthe principal peak.
Description. A white powder.
In the chromatogram obtained with the test solution, the area
of any peak corresponding to the chioro-adduct is not more Identification
than 1.5 times the area of the principal peak in the
chromatogram obtained with reference solution (a.) (0.15 per A. Determine on 20 mg by the oxygen-flask method (2.3.34),
cent). The area of any other secondary peak is not more than using 5 ml of 1.25 M sodium hydroxide as the absorbing
the area of the principal peak in the chromatogram obtained liquid. When the process is complete, dilute to 25 ml with
with reference solution (a) (0.1 per cent) and the sum of the water (solution A). To 5 rnl ofsolution A add 0.1 ml of hydrogen
areas of all the secondary peaks is not more than twice the peroxide solution (l00 vol) and 1 ml of 1 M hydrochloric
area ofthe principal peak in the chromatogram obtained with acid, mix and add 0.05 ml of barium chloride solution; a
reference solution (a) (0.2 per cent). turbidity is produced.
B. To 2 ml ofsolutionAadd40rnl ofwaterand4mlofammonium
Water (2.3.43). Not more than 2.0 percent, determined on 1.2 g.
molybdate-sulphuric acid solution, mix, add 0.1 g of
Assay. Weigh accurately about 0.2 g into a stoppered flask, L~ascorbic acid and boil for 1 minute; a blue colour is
add 50 ml ofa 20 per cent wlv solution of sodium thiosulphate produced.

2219

www.webofpharma.com
THYMOL IP 2010

Tests A. Determine by infrared absorption spectrophotometry (2.4.6).


Compare the spectrum with that obtained with thymol RS or
Appearance ofsolution. Dissolve a quantity containing 15 mg with the reference spectrum ofthymol.
ofThiotepa in 4 ml ofwater; the solution is clear (2.4.1).
B. Dissolve 0.2 g with heating in 2 ml of2 M sodium hydroxide,
pH (2.4.24).5.5 to 7.5, determined in a 1.0 per centw/v solution
add 0.2 ml of chloroform and heat on a water-bath; a violet
in carbon dioxide-free water.
colour develops.
Bacterial endotoxins (2.2.3). Not more than 6.25 Endotoxin
C. Dissolve about 2 mg in 1 ml ofanhydrous acetic acid, add
Units per mg of Thiotepa.
0.15 ml ofsulphuric acid and 0.05 ml of nitric acid; a bluish
Assay. Determine by liquid chromatography (2.4.14). green colour develops.
Test solution. Dilute a suitable quantity of the injection D. Melting range (2.4.21).48° to 52°.
containing 0.15 g ofThiotepa in 100 ml ofwater.
Tests
Reference solution. A 0.15 per cent w/v solution of thiotepa
RS in water. Appearance ofsolution. Dissolve 1.0 g in 10 ml of 2 M sodium
Chromatographic system hydroxide. The solution is not more opalescent than
- a stainless steel column 20 cm x 4.6 mm, packed with opalescence standard OS4 (2.4.1), and not more intensely
octadecylsilane bonded to porous silica (5 ~m), coloured than reference solution RS6 (2.4.1).
- mobile phase: a mixture of 70 volumes of water and Acidity. To 1.0 g in a glass-stoppered flask add 20 ml ofwater,
30 volumes of acetonitrile, boil until dissolution is complete, cool, stopper the flask and
- flow rate. 1 ml per minute, shake vigorously for 1 minute. Add a few crystals of the
- spectrophotometer set at 215 nm, substance under examination to induce crystallisation, shake
- injection volume. 20 ~l. vigorously for 1 minute and filter. To 5 ml of the filtrate add
Calculate the content ofC 6H 12N 3PS in the injection. 0.05 ml of methyl red solution and 0.05 ml of 0.01 M sodium
hydroxide; the solution is yellow.
Storage. Store protected from light. If solid matter separates
from the constituted injection, the solution should not be Related substances. Determine by gas chromatography
used. (2.4.13).
Test solution. Dissolve 0.1 g in sufficient ethanol (95 per
cent) to produce 10 ml.
Reference solution (a). Dilute 1 ml of the test solution to
Thymol 100 ml with ethanol (95 percent).
Reference solution (b). Dilute 1 ml ofreference solution (a) to
10 ml with ethanol (95 per cent).
Reference solution (c). Dilute 5 ml ofreference solution (b) to
10 ml with ethanol (95 per cent).
Chromatographic system
- a glass column 4 m x 2 mm, packed with diatomaceous
Mol. Wt. 150.2 support (125 to 180 mesh) impregnated with a mixture
Thymol is 2-isopropyl-5-methylphenol. suitable for the separation of free fatty acids (such as
FFAP),
Thymol contains not less than 99.0 per cent and not more - temperature:
than 101.0 per cent ofC IOH I4 0. column 80° for 2 minutes, increase @ 8° to 240° stand for
Category. Pharmaceutical aid (antimicrobial preservative; 15 minutes,
antiseptic). inlet port at 250° and detector at 300°,
- flow rate. 30 ml per minute ofthe carrier gas.
Description. Colourless crystals; odour, characteristic.
Inject test solution, reference solution (a),'{b) and (c):
Identification
Intbe chiomatogramobta.ined with tbetest solutIon the sum
Test A may be omitted iftests B, C andD are carried out. Tests of the areas of any secondary peaks is not more than the area
B, C and D may be omitted if test A is carried out. of the principal peak in the chromatogram obtained with

2220

www.webofpharma.com
IP 2010 THYROXINE SODIUM

reference solution (a). Ignore any peak with an area less than an absorption maximum at about 325 nm; absorbance at about
that of the principal peak in the chromatogram obtained with 325 nm, 0.73 to 0.79.
reference solution (c).
B. In the test for Liothyronine, the principal spot in the
Residue on evaporation. Evaporate 2.0 g on a water-bath and chromatogram obtained with the test solution corresponds to
heat at 105° for I hour. The residue weighs not more than that in the chromatogram obtained with reference solution
1.0 mg (0.05 percent). (b).
Assay. Weigh accurately about 0.1 g, transfer to an iodine C. To about 50 mg in a porcelain dish add a few drops of
flask and dissolve in 25 ml of 1 M sodium hydroxide. Add sulphuric acid (96 per cent w/w); violet vapors are evolved.
20 ml of hot dilute hydrochloric acid and immediately titrate
D. To 20 mg add 2 ml of 1 M sulphuric acid. Heat on a water-
with 0.05 M bromine to within 1 to 2 ml ofthe calculated end-
bath followed by heating carefully over a naked flame,
point. Warm the solution to about 75°, add 0.1 m1 of methyl
increasing the temperature to about 600°. Continue ignition
orange solution and shake vigorously. If the solution is red,
until most of the black particles have disappeared. Dissolve
continue the titration, dropwise and with shaking until the red
the residue in 2 ml of water; the solution gives reaction A of
colour is discharged. Repeat the alternate addition of 0.05 M sodium salts (2.3.1).
bromine and methyl orange solution until the red colour is
discharged after the addition of the methyl orange solution.
Tests
1 n:l of 0.05 Mbromine is equivalentto 0.003755 gofC IO H I4 0.
Appearance of solution. Dissolve 0.5 g in 23 ml of a gently
Storage. Store protected from light and moisture. boiling mixture of 4 volumes of ethanol (95 per cent) and
1 volume of 1 M hydrochloric acid. Cool and dilute to 25 ml
with the same mixture of solvents (solution A). The freshly
prepared solution is not more intensely coloured than reference
Thyroxine Sodium solution BYS3 (2.4.1).
Levothyroxine Sodium; L-Thyroxine Sodium Specific optical rotation (2.4.22).+16.0° to +20.0°, determined
in solution A.
Liothyronine. Determine by thin-layer chromatography
(2.4.17), coating the plate with a slurry oBO g ofsilica gel H in
60 ml of a 0.75 per cent w/v solution of soluble starch.
Solvent mixture. 70 volumes of methanol and 5 volumes of
strong ammonia solution.
Mobile phase. A mixture of 55 volumes of ethyl acetate,
35 volumes of 2-propanol and 20 volumes of strong ammonia
solution.
ClsHIO!4NNa04,xH20 Mol. Wt. 798.9 (anhydrous)
Test solution. Dissolve 0.1 g of the substance under
Thyroxine Sodium is sodium Q4-(4-hydroxy-3,5-diiodo- examination in 10 ml ofthe solvent mixture.
phenyl)-3,5-diiodo-L-tyrosinate and contains a variable
quantity of water of crystallisation. Reference solution (a). A solution containing 1.0 per cent
w/v of the substance under examination and 0.01 per cent
Thyroxine Sodium contains not less than 97.0 per cent and w/v of the liothyronine RS in the solvent mixture..
not more than 101.0 per cent ofClsHIOI4NNa04, calculated on
the dried basis. Reference solution (b). A 1.0 per cent w/v solution of
levothyroxine sodium RS in the solvent mixture.
Category. Thyroid hormone.
Reference solution (c). A 0.01 per cent w/v solution of
Dose. 50 to 300 p.g daily. liothyronine RS in the solvent mixture.
Description. A white or slightly brownish yellow powder, Apply to the plate 5 III of each solution. After development,
slightly coloured, crystalline powder. dry the plate in air, spray lightly with ferric chloride-
ferricyanide-arsenite solution. Any spot corresponding to
Identification
liothyronine in the chromatogram obtained with the test
A. When examined in the range 230 nm to 360 nm (2.4.7), a solution is not more intense than the spot in the chromatogram
0.01 per cent w/v solution in 0.1 M sodium hydroxide shows obtained with reference solution (c). The test is not valid unless

2221

www.webofpharma.com
THYROXINE SODIUM IP 2010

the chromatogram obtained with reference solution (a) shows of sodium nitrite and boil; a yellow colour is produced. Cool
two clearly separated spots. and make alkaline with 5 M ammonia; the solution becomes
orange.
Loss on drying (2.4.19). 6.0to 12.0 per cent, determined on
0.5 g by drying in an oven at 105°. R In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak due
Assay. Determine by liquid chromatography (2.4.14).
to thyroxine sodium in the chromatogram obtained with the
Test solution. Dissolve 10 mg of the substance under reference solution.
examination in 40 ml of 0.1 M sodium hydroxide. Shake for
about 30 minutes, add 40 ml of methanol, cool, mix well and Tests
dilute to 100 ml with a mixture of equal volumes of 0.1 M
Uniformity of content. Comply with the test stated under
sodium hydroxide and methanol. Dilute 5.0 ml ofthe solution
Tablets. using the following solutions.
to 50.0 ml with the same solvent.
Test solution. To one tablet, add 10 ml of 0.1 M sodium
Reference solution. A 0.001 per cent w/v solution of
hydroxide and shake for about 30 minutes. Add 10 ml of
levothyroxine sodium RS in a mixture of equal volumes of
methanol, cool and dilute with sufficient ofa mixture ofequal
0.1 M sodium hydroxide and methanol.
volumes of 0.1 M sodium hydroxide and methanol to produce
Chromatographic system 25.0 ml, mix well and filter. Dilute, ifnecessary, with the same
a stainless steel column 25 cm x 4.6 rom, packed with a solvent mixture to produce a solution containing 0.0002 per
stationary phase consisting of porous silica particles cent w/v ofThyroxine Sodium.
(5 to 10 jlm) to which nitrile groups are chemically
Reference solution. A 0.0002 per cent w/v solution of
bonded,
levothyroxine sodium RS in a mixture of equal volumes of
mobile phase: a mixture of65 volumes of water, 1 volume
0.1 M sodium hydroxide and methanol.
of orthophosphoric acid and 35 volumes of acetonitrile,
flow rate. 1 ml per minute, Follow the procedure described under Assay.
spectrophotometer set at 225 urn, Calculate the content ofC IsH IOI4NNa04 in the tablet.
injection volume. 20 jll.
Other tests. Comply with the tests stated under Tablets.
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more Assay. Determine by liquid chromatography (2.4.14).
than 2.0 per cent. Test solution. Weigh and powder 20 or more tablets. Weigh
Inject the test solution and the reference solution. accurately a quantity of the powder containing about 1 mg of
Thyroxine Sodium, add 40 ml of 0.1 M sodium hydroxide and
Calculate the content OfC1SHIOI4NNa04. shake for about 30 minutes. Add 40 ml of methanol, cool, mix
Storage. Store protected from light and moisture. well and dilute with sufficient of a mixture ofequal volumes of
0.1 M sodium hydroxide and methanol to produce 100.0 ml,
mix well and filter.
Reference solution. A 0.001 per cent w/v solution of
Thyroxine Tablets levothyroxine sodium RS in a mixture of equal volumes of
Thyroxine Sodium Tablets; Levothyroxine Tablets; 0.1 M sodium hydroxide and methanol.
Levothyroxine Sodium Tablets; L-Thyroxine Sodium Chromatographic system
Tablets a stainless steel column 25 cm x 4.6 mm, packed with a
stationary phase consisting of porous· silica particles
Thyroxine Tablets contain not less than 90.0 per cent and not
more than 110.0 per cent of the stated amount of anhydrous (5 to 10 jlm) to which nitrile groups are chemically
bonded,
thyroxine sodium, ClsHIOI4NNa04.
mobile phase: a mixture of65 volumes ofwatel; 1 volume
Usual strength. 50 jlg. of orthophosphoric acid and 35 volumes of acetonitrile,
flow rate. 1 ml per minute,
Identification spectrophotometer set at 225 urn,
A.. To aquantity ot'the powdered tablets cOJ:ltalJ:liIigS66 ~gof injection volume: 20 ill.
cllihydroL/s ihyl'oxil1e sodium a.dd a.roix.tureof3 rol of ethanol Inject the reference solution. The test is not valid unless the
(50 per cent) and 0.2 ml of hydrochloric acid, boil gently for relative standard deviation for replicate injections is not more
30 seconds, cool, filter, add 0.1 mlofa 10 per cent w/v solution than 2.0 per cent.

2222

www.webofpharma.com
IP 2010 TlCARClLLIN AND CLAVULANlC ACID INJECTION

Inject the test solution and the reference solution. Assay. Determine by liquid chromatography (2.4.14).
Calculate the content of CisHlOI4NNa04 in the tablets. Sodium phosphate bufferpH4.3. Dissolve 13.8 g ofmonobasic
Storage. Store protected from light and moisture. sodium phosphate in 900 ml ofwater, adjust the pH to 4.3 with
orthophosphoric acid or 10M sodium hydroxide, dilute to
Labelling. The label states the strength in terms of the
1000 ml with water.
equivalent amount of anhydrous thyroxine sodium.
Sodium phosphate bufferpH 6.4. Dissolve 6.9 g ofmonobasic
sodium phosphate in 900 ml ofwater, adjust the pH to 6.4 with
10Msodium hydroxide, dilute 1000 ml with water.
Ticarcillin and Clavulanic Acid
Clavulanate lithium stock solution. A 0.06 per cent w/v
Injection solution ofclavulanate lithium RS in sodium phosphate buffer
Clavulanic Acid and Ticarcillin Injection pH 6.4.

Ticarcillin and Clavulanic Acid Injection is a sterile iso-osmotic Test solution. Dissolve a quantity ofpowder in sufficient water
solution ofTicarcillin Monosodium and Clavulanate Potassium and dilute with sodium phosphate btiffer pH 6.4 to obtain a
in Water for Injections. It contains one or more suitable 0.09 per cent w/v solution ofTicarcillin.
buffering agents and a tonicity adjusting agent.
Reference solution. Transfer about 100 mg of ticarcillin
Ticarcillin and Clavulanic Acid Injection contains not less monosodium monohydrate RS to a 100-ml volumetric flask
than 90.0 per cent and not more than 115.0 per cent of the and add 150/J ml of clavulanate lithium stock solution (J is
stated amount of ticarcillin, CIsHl6N206S2 and not less than the ratio ofthe stated amount ofticarcillin to the stated amount
85.0 per cent and not more than 120.0 per cent of the stated of clavulanic acid in the powder) and dilute to 100.0 ml with
amount ofclavulanic acid, CSH9NO s. sodium phosphate btiffer pH 6.4.

Ticarcillin and Clavulanic Acid for Injection Chromatographic system


- a stainless steel column 30 cm x 4 mm, packed with
Ticarcillin and Clavulanic acid for Injection is a sterile, dry octadecylsilane chemically bonded to porous silica (5
mixture ofTicarcillin Monosodium and Clavulanate Potassium. 11m),
The constituted solution complies with the requirements for mobile phase: a mixture of 95 volumes of sodium
clarity of solution and particulate matter stated under phosphate btifferpH 4.3 and 5 volumes of acetonitrile,
Parenteral Preparations (Injections). - flow rate. 2 ml per minute,
- spectrophotometer set at 220 nm,
Usual Strength. Ticarcillin 3 g and Clavullanic acid 100 mg.
- injection volume. 20 Ill.
Storage. The constituted solution should be used immediately
after preparation but, in any case, within the period Inject the reference solution. The test is not valid unless the
recommended by the manufacturer. column efficiency of the principal peaks is not less than 1000
theoretical plates; the tailing factor for the principal peaks are
Identification not more than 2.0; the resolution between the peaks due to
ticarcillin and clavulanic acid is not less than 5.0; and the
In the Assay, the principal peak due to ticarcillin in the relative standard deviation for replicate injections is not more
chromatogram obtained with the test solution corresponds to than 2.0 per cent. The relative retention time with reference to
that in the chromatogram obtained with the reference solution. ticarcillin for clavulanic acid is about 0.2.
Tests Inject the test solution and the reference solution.
pH (2.4.24).5.5 to 7.5. Calculate the content of ClsHl6N206S2 and CSH9NO s in the
Bacterial endotoxins (2.2.3). Not more than 0.07 Endotoxin injection.
Unit per mg ofticarcillin.
Storage. Store in single dose or multiple dose containers.
Sterility (2.2.11). Complies with the test for sterility.
Labelling. The label states (l) that the injection is to be thawed
Water (2.3.43). Not more than 4.2 per cent. just prior to use; (2) conditions for proper storage of the
Other tests. Complies with the tests stated under Parenteral resultant solution; (3) directs that the solution is not to be
Preparations (Powders for Injections). refrozen.

2223

www.webofpharma.com
TIMOLOL MALEATE IP 2010

Timolol Maleate Tests

Appearance ofsolution. A 2.0 per cent w/v solution in carbon


dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution BS8 (2.4.1).
(eOOH
'~ pH (2.4.24). 3.8 to 4.3, determined in a 2.0 per cent w/v solution.
eOOH Specific optical rotation (2.4.22). -5.7° to -6.20, determined in
a 10.0 per cent w/v solution in 1 M hydrochloric aeid.
Related substances. Determine by thin-layer chromatography
Mol. Wt. 432.5 (2.4.17), coating the plate with silica gel GF254.
Timolol Maleate is (S)-I-tert-butylamino-3-( 4-morpholino- Mobile phase. A mixture of 80 volumes of dichloromethane,
1,2,5-thiadiazol-3-yloxy)propan-2-01 hydrogen maleate. 20 volumes of methanol and 1 volume of strong ammonia
solution.
Timolol Maleate contains not less than 98.5 per cent and not
more than 101.0 per cent ofC13H24N403S,C4H404, calculated Test solution (a). Dissolve 0.5 g of the substance under
on the dried basis. examination in 10 ml of methanol.
Category. Beta-adrenoceptor antagonist (Antiglaucoma). Test solution (b). Dissolve 0.1 g of the substance under
examination in 100 ml of methanol.
Dose. In hypertension, initially 5 mg twice daily or 10 mg once
daily; maximum 60 mg daily. In angina, initially 5 mg 2 to 3 Reference solution (a). A 0.02 per cent w/v solution of the
times daily; maintenance dose, 15 to 45 mg daily. For substance under examination in methanol.
prophylaxis after infarction, initially 5 mg twice daily, started 1
Reference solution (b). A 0.1 per cent w/v solution of timolol
to 4 weeks after infarction. For migraine prophylaxis, 10 to 20
maleate RS in methanol.
mgdaily.
Apply to the plate 10 III of each solution. After development,
Description. A white or almost white, crystalline powder or
dry the plate in air and examine in ultraviolet light at 254 nm
colourless crystals.
and then e)(:.pose to iodine vapour for 2 hours and examine in
daylight. By both methods of visualisation, any secondary
Identification spot in the chromatogram obtained with test solution (a) is
Test A may be omitted iftests Band C are carried out. Tests B not more intense than the spot in the chromatogram obtained
and C may be omitted if test A is carried out. with reference solution (a).

A. Determine by infrared absorption spectrophotometrY (2.4.6). Sulphated ash (2.3.18). Not more than 0.1 per cent.
Compare the spectrum with that obtained with timolol maleate Loss on drying (2.4.19). Not more than 0.5 per cent, determined
RS or with the reference spectrum oftimolol maleate. on 1.0 g by drying in an oven at 105°.
B. In the test for Related substances, after exposure to iodine Assay. Weigh accurately about 0.35 g, dissolve in 60 ml of
vapour, the principal spot in the chromatogram obtained with anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
test solution (b) corresponds to that in the chromatogram acid, determining the end-point potentiometrically (2.4.25).
obtained with reference solution (b). Carry out a blank titration.
C. Triturate 0.1 g with a mixture of! ml of2 Msodium hydroxide 1 ml of 0.1 M perchloric acid is equivalent to 0.04325 g of
and 3 ml of water and shake with three quantities, each of5 ml, CI3H24N403S, C~04.
of ether. To 0.1 ml ofthe aqueous layer add a solution of 10 mg
Storage. Store protected from light and moisture.
of resorcinol in 3 ml of sulphuric acid and heat on a water-
bath for 15 minutes; no violet-red colour is produced. Neutralise
the remainder of the aqueous layer with 1 M sulphuric acid,
add 1 mlofbrominewater, heat on a water-bath for 15 minutes, Timolol Eye Drops
then heat to boiling and cool. To 0.2 m1 ofthis solution add a
Timolol Maleate Eye Drops
solution of 10 mg of resorcinol in 3 ml of sulphuric acid and
heat on a water-bath for 15 minutes; a violet-red colour is Timolol Eye Drops are a sterile solution ofTimolol Maleate;: in
produced. Add 0.2 ml ofa 10 per cent w/v solution ofpotassium Purified Water. They may contain suitable antimicrobial
bromide and heat on a water-bath for 5 minutes; the colour preservatives, buffering agents, stabilisers and suitable
changes to violet-blue. substances to increase the viscosity of the solution.

2224

www.webofpharma.com
IP 2010 TIMOLOL TABLETS

Timolol Eye Drops contain not less than 90.0 per cent and not peak, other than the peak corresponding to maleic acid, is not
more than 110.0 per cent of the stated amount of timolol, greater than the area ofthe peak in the chromatogram obtained
C13H24N403S. with reference solution (a) and not more than two such peaks
have an area greater than that of the peak obtained with
Usual strengths. The equivalent of 0.25 per cent w/v and 0.5
per cent w/v oftimolol. refer.ence solution (b).
Other tests. Comply with the tests stated under Eye Drops.
Identification
Assay. To a volume containing about 25 mg of timolol add
A. Add a volume containing 50 mg of timolol to an equal water to produce 50.0 ml and mix. To 5.0 ml add 15 ml of
volume of carbonate buffer pH 9.7 and extract with two carbonate buffer pH 9.7 and extract with three quantities,
quantities, each of 40 ml, of dichloromethane. Reserve the each of20 ml, and one quantity of I 0 ml of toluene. Wash each
aqueous layer for test B. Dry the combined dichloromethane extract successively with the same I O-ml volume of carbonate
extracts with anhydrous sodium sulphate and evaporate to bufjer pH 9. 7. Combine the toluene extracts and extract with
dryness. Dry the residue at 60° at a pressure of 2 kPa for four quantities, each of 20 ml, of 0.05 M sulphuric acid.
15 minutes. Combine the extracts, dilute to 100.0 ml, filter and measure the
absorbance of the filtrate at the maximum at about 295 nm
On the residue, determine by infrared absorption
(2.4.7), using as the blank a solution prepared by treating
spectrophotometry (2.4.6). Compare the spectrum with that
5.0 ml of water in the same manner beginning at the words
obtained with timolol RS or with the reference spectrum of
"add 15 ml of carbonate bufferpH 9.7...".
timolol.
Calculate the content ofCI3H24N403S taking 279 as the specific
B. Filter the aqueous layer reserved in test A and evaporate to
absorbance at 295 nm.
about I ml. Add I ml of bromine solution, heat in a water-bath
for 10 minutes, boil, cool and add 0.1 ml of the solution to a Storage. Store protected from light and moisture.
solution of I 0 mg resorcinol in 3 ml ofsulphuric acid; a bluish
Labelling. The label states (1) the strength in terms of the
black colour is produced on heating in a water-bath for
equivalent amount oftimolol; (2) the names and concentration
15 minutes.
of any added antimicrobial preservatives.
Tests
pH (2.4.24).6.5 to 7.5.
Related substances. Determine by liquid chromatography Timolol Tablets
(2.4.14). Timolol Maleate Tablets
Test solution. Use the undiluted eye drops. Tirnolol Tablets contain not less than 90.0 percent and not
Reference solution (a). Dilute I volume of the eye drops to more than 110.0 per cent ofthe stated amount oftimolol maleate,
250 volumes with the mobile phase. CI3H24N403S,C~404'

Reference solution (b). Dilute I volume of the eye drops to Usual strength. 10 mg.
500 volumes with the mobile phase.
Identification
Reference solution (c). A 0.3 per cent w/v solution of maleic
acid in the mobile phase. A.To a quantity of the powdered tablets containing 70 mg of
Chromatographic system Timolol Maleate add 20 ml of carbonate buffer pH 9. 7 and
a stainless steel column 20 cm x 4 mm, packed with extract with two quantities, each of40 ml, of dichloromethane.
octadecylsilane bonded to porous silica (10 flm), Reserve the aqueous layer for test B. Dry the extracts with
mobile phase: a mixture of 57.5 volumes of methanol anhydrous sodium sulphate, evaporate to dryness using a
and 42.5 volumes of 0.02 M sodium octanesulphonate, rotary evaporator, dry the residue at 60° at a pressure of2 kPa
adjusted to pH 3.0 with glacial acetic acid, for 15 minutes.
- flow rate. 2 ml per minute, On the residue, determine by infrared absorption
spectrophotometer set at 295 nm, spectrophotometry (2.4.6). Compare the spectrum with that
injection volume. 20 fll. obtained with timolol RS or with the reference spectrum of
Record the chromatogram of the test solution for 4 times the timolol.
retention time of the principal peak. In the chromatogram B. Filter the aqueous layer reserved in test A and evaporate to
obtained with the test solution the area of any secondary about I ml. Add I ml of bromine water, heat on a water-bath

2225

www.webofpharma.com
TIMOLOL TABLETS IP 2010

for 15 minutes, then heat to boiling and cool. Add 0.1 ml ofthis maximum at about 295 nm (2.4.7), using as the blank a solution
solution to a solution of 10 mg of resorcinol in 3 ml ofsulphuric prepared by treating a mixture of 5 ml of water and 15 ml of
acid and heat on a water-bath for 15 minutes; a bluish black carbonate buffer pH 9. 7 in the same manner beginning at the
colour is produced. words "and extract with three quantities,..."

Tests Calculate the content of CI3H24N403S,C4H404 taking 204 as


the specific absorbance at 295 nm.
Related substances. Detennine by liquid chromatography
Storage. Store protected from moisture.
(2.4.14).
Test solution. Shake a quantity of the powdered tablets
containing 0.1 g ofTimolol Maleate with 10 ml of the mobile
phase for 10 minutes and filter. Tinidazole
Reference solution (a). Dilute 1 volume ofthe test solution to
250 volumes with the mobile phase.
Reference solution (b). Dilute 1 volume ofthe test solution to
500 volumes with the mobile phase.
Chromatographic system
- a stainless steel column 20 cm x 4 mm, packed with
Mol. Wt. 247.3
octadecylsilane bonded to porous silica (l0 /lm),
mobile phase: a mixture of 57.5 volumes of methanol Tinidazole is 1-[2-(ethylsulphonyl)ethyl]-2-methyl-
and 42.5 volumes of 0.02 M sodium octanesulphonate, 5-nitroimidazole.
adjusted to pH 3.0 with glacial acetic acid, Tinidazole contains not less than 98.0 per cent and not more
flow rate. 2 ml per minute, than 100.5 per cent of CSHI3N304S, calculated on the dried
spectrophotometer set at 295 nm, basis.
injection volume. 20 /ll.
Category. Antiprotozoal.
Run the chromatogram of the test solution for 4 times the
retention time of the principal peak. In the chromatogram Dose. For trichomoniasis, 2 g as a single dose and repeat after
obtained with the test solution the area of any secondary 3 to 5 days rest, ifnecessary. For giardiasis, 150mgtwice daily
peak, other than the peak corresponding to maleic acid, is not for seven days or as a single dose of 2 g. For intestinal
more than the area ofthe peak in the chromatogram obtained amoebiasis, 600 mg as a single dose daily for 2 to 6 days. For
with reference solution (a) and not more than two such peaks extraintestinal amoebiais, 2 g as a single daily dose for 3 days.
have an area more than that ofthe peak obtained with reference Description. Pale yellow crystals or a crystalline powder;
solution (b). odour, slight and characteristic.
Uniformity of content. Comply with the test stated under
Identification
Tablets.
Test A may be omitted if tests Band C are carried out. Tests B
To one tablet, add 25.0 ml of 0.05 M sulphuric acid, shake for
and C may be omitted if test A is carried out.
20 minutes and complete the Assay beginning at the words
"and centrifuge....". A. Detennine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with tinidazole RS
Other tests. Comply with the tests stated under Tablets.
or with the reference spectrum oftinidazole.
Assay. Weigh and powder 20 tablets. Weigh accurately a
B. When examined in the range 230 nm to 360 nm (2.4.7), a
quantity of the powder containing about 15 mg of Timolol
0.00 I per cent w/v solution in methanol shows an absorption
Maleate, add 25.0 ml of 0.05 M sulphuric acid, shake for
maximum at about 31 0 nm; absorbance at about 310 nm, about
20 minutes and centrifilge until clear. Add 5.0 ml ofthe resulting
0.35.
supernatant liquid to 15 ml of carbonate bujfer pH 9.7 and
extract with three quantities, each of 20 ml, and one quantity C. To about 5 mg, add 5 ml of 0.1 M hydrochloric acid, 50 mg
of 1 0 ml of toluene. Wash each extract successively with.the of zinc powder, 4 ml ofhydrochlQric.JJcidandal1QwJQ s.tand
same 10-ml volume of carbonate bujferpH 9. 7, combiue the for 30 minutes. Add 4 ml of a I per cent w/v solution of
toluene extracts and extract with four quantities, each of20 ml, vanillin, heat on a boiling water-bath for 20 minutes, allow to
of 0.05 M sulphuric acid. Combine the acid extracts, dilute to cool to room temperature and dilute to 20 ml with water; a
100.0 ml, filter and measure the absorbance ofthe filtrate at the greenish yellow colour is produced.

2226

www.webofpharma.com
IP 2010 TIOTROPIUM BROMIDE MONOHYDRATE

Tests room temperature and dilute to 20 ml with water; a greenish


yellow colour is produced.
Related substances. Detennine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. C. Melting range (2.4.21). 125° to 128°.

Mobile phase. A mixture of 95 volumes of ethyl acetate, Tests


5 volumes of methanol and 5 volumes of diethylamine.
Other tests. Comply with the tests stated under Tablets.
Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml ofa mixture ofequal volumes ofchlorofonl1 Assay. Weigh and powder 20 tablets. Weigh accurately a
and methanol. quantity ofthe powder containing about 0.15 g ofTinidazole,
add 20 ml of methanol, shake well and add sufficient methanol
Reference solution. A 0.02 per cent w/v solution of the to produce 100.0 ml. Mix well and filter. Dilute 10.0 ml ofthe
substance under examination in a mixture ofequal volumes of solution to 100.0 ml with methanol and further dilute 10.0 ml of
chloroform and methanol. this solution to 100.0 ml with methanol. Measure the
Apply to the plate 10 III of each solution. After development, absorbance of the resulting solution at the maximum at about
dry the plate in air and examine in ultraviolet light at 254 nm. 310nm(2.4.7).
Any secondary spot in the chromatogram obtained with the Calculate the content ofCgHl3N304S taking 356 as the specific
test solution is not more intense than the spot in the absorbance at 310 nm.
chromatogram obtained with the reference solution:
Storage. Store protected from light and moisture.
Sulphated ash (2.3.18). Not more than 0.2 per cent.
Loss on drying (2.4.19). Not more than 2.0 per cent, detennined
on 1.0 g by drying in an oven at 105° for 4 hours.
Tiotropium Bromide Monohydrate
Assay. Weigh accurately about 0.5 g, dissolve in 30 ml of
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric

:~f~3k
acid, using 0.15 ml of nile blue A solution as indicator. Carry
out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.02473 g of
CgH 13N 30 4S. ~1 ~s
Storage. Store protected from light and moisture.
o s
o OH ~ ~
Tinidazole Tablets
CI9H22BrN04S2, H20 Mol. Wt. 490.2
Tinidazote Tablets contain not less than 95.0 per cent and not
Tiotropium Bromide Monohydrate is 6/3,7{3-epoxy-3{3-hydroxy-
more than 105.0 per cent of the stated amount oftinidazole,
8-methyl-1 cxH-5cxH-tropanium bromide monohydrate.
CgH 13N30 4 S.
Tiotropium Bromide Monohydrate contains not less than
Usual strengths. 150 mg; 300 mg; 500 mg. 98.0 per cent and not more than 102.0 per cent oftiotropium
Identification bromide, CI9H22N04S2Br, calculated on the anhydrous basis.
Category. Bronchodilator.
A. When examined in the range 230 nm to 360 nm (2.4.7), the
final solution obtained in the Assay shows an absorption Description. A white to off-white powder.
maximum at about 31 0 nm.
Identification
Extract a quantity of the powdered tablets containing 0.1 g of
Tinidazole with i 0 ml of methanol, filter and evaporate the A. Detennine by infrared absorption spectrophotometry
filtrate to dryness. The residue complies with the following (2.4.6). Compare the spectrum with that obtained with
tests. tiotropium bromide monohydrate RS or with the reference
spectrum oftiotropium bromide monohydrate.
B. To about 5 mg add 5 ml of 0.1 M hydrochloric acid, 50 mg
of zinc powder, 4 ml of hydrochloric acid and allow to stand B. In the Assay, the principal peak in the chromatogram
for 30 minutes. Add 4 ml ofa 1per cent w/v solution of vanillin, obtained with test solution corresponds to the principal peak
heat on a boiling water-bath for 20 minutes, allow to cool to in the chromatogram obtained with the reference solution.

2227

www.webofpharma.com
TIOTROPIUM BROMIDE MONOHYDRATE IP 2010

Tests Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0, the column efficiency in not
Related substances. Determine by liquid chromatography less than 3000 theoretical plates and the relative standard
(2.4.14). deviation for replicate injections is not more than 2.0 per cent.
Test solution. Dissolve 10 mg of the substance under Inject the test solution and the reference solution.
examination in 100 ml ofthe mobile phase.
Calculate the content ofCI9HzzN04Sz.Br.
Reference solution. A 0.002 per cent w/v solution oftiotropium
bromide monohydrate RS in the mobile phase. Storage. Store protected from light and moisture.

Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 ,"un),
mobile phase: a mixture of45 volumes of methanol and Tiotropium Bromide Powder for
55 volumes of a buffer solution prepared by dissolving Inhalation
3.85 g of ammonium acetate in 1000 ml of water and
adjusting the pH to 5.5 with dilute acetic acid Tiotropium Powder for Inhalation consists of Tiotropium
(10 per cent vlv), Bromide in microfine powder either alone or admixed with
- flow rate. I ml per minute, Lactose in a pre-metered unit for use in a suitable powder
inhaler.
- spectrophotometer set at 235 run,
- injection volume. 20 J..L1. Tiotropium Powder for Inhalation contains not less than
90.0 per cent and not more than 125.0 per cent of the stated
Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency is amount oftiotropium, CI9H22N04Sz per unit dose.
not less than 3000 theoretical plates.
Inject the test solution. Any individual impurity is not more
Identification
than 0.5 per cent and the sum of all the impurities is not more In the Assay, the principal peak in the chromatogram obtained
than 1.0 per cent. with the test solution corresponds to the peak in the
Heavy metals (2.3.13). 1.0 g complies with the limit test for chromatogram obtained with the reference solution.
heavy metals, Method B (20 ppm).
Tests
Sulphated ash (2.3.18). Not more than 0.3 percent.
Other tests. Complies with the tests stated under Inhalation
Water (2.3.43). Not more than 4.5 per cent, determined on 0.5 g. Preparations (Powders for Inhalation).
Assay. Determine by liquid chromatography (2.4.14). Follow the procedure described under Assay with suitable
Test solution. Dissolve 20 mg of the substance under dilution of the reference solution wherever the amount of
examination in 100.0 ml ofthe mobile phase. Dilute 5.0 ml of active substance is to be determined in any test.
the resulting solution to 50.0 ml with the mobile phase. Assay. Determine by liquid chromatography (2.4.14).
Reference solution. A 0.002 per cent w/v solution oftiotropium Solvent mixture. 90 volumes of 0.05 per cent v/v ortho-
bromide monohydrate RS in the mobile phase. phosphoric acid and 10 volumes of acetonitrile.
Chromatographic system
Test solution. Dissolve a quantity of the mixed contents of
a stainless steel column 25 cm x 4.6 mm, packed with 20 capsules in sufficient ofthe solvent mixture to get a solution
octadecylsilane bonded to porous silica (5 J..Lm), containing 1.8 J..Lg per ml ofTiotropium.
- mobile phase: a mixture of45 volumes of methanol and
55 volumes ofa buffer solution prepared by dissolving Reference solution. A solution of tiotropium· bromide RS
3.85 g of ammonium acetate in 1000 ml of water and equivalent to tiotropium 1.8 J..Lg per ml in the solvent mixture.
adjusting the pH to 55 with dilute acetic acid ( 10 per

a stainless steel column 15 cm x 4.6 lnm, packed with


flow rate. 1 nil per minute,
octadecylsilane bonded to porous silica (5 J..Lm),
- spectrophotometer set at 235 urn, mobile phase: a mixture of80 volumes ofa buffer solution
injection volume. 20 J..L1. prepared by dissolving 2 ml triethylamine in 1000 ml of

2228

www.webofpharma.com
IP 2010 TITANIUM DIOXIDE

water and adjusting the pH to 2.5 with orthophosphoric Water-soluble substances. Not more than 0.5 per cent,
acid, and 20 volumes of acetonitrile, determined by the following method. Boil 10.0 g for 5 minutes
- flow rate. 2 ml per minute, with 150 ml ofwater containing 0.5 g ofammonium sulphate.
- spectrophotometer set at 237 nm, Cool, dilute to 200 ml with water and filter until a clear solution
- inject 200 fll. is obtained. Evaporate 100 ml of the filtrate to dryness ignite
and weigh.
Inject the reference solution. The test is not valid unless the
column efficiency is not less than 4500 theoretical plates, the Arsenic (2.3.10). To 0.2 g in a 100 ml Kjeldahl flask, add 2 g of
tailing factor is not more than 1.7 and the relative standard anhydrous sodium sulphate, 7 ml of sulphuric acid and 5 ml
deviation for replicate injections is not more than 2.0 per cent. of nitric acid. Heat gently until a clear solution is obtained,
cool, add 10 ml of water, cool again and add 5 g of hydrazine
Inject the test solution and the reference solution.
reducing mixture and 10 ml ofhydrochloric acid. Immediately
Calculate the content of C19H22N04Sz per unit. attach an air condenser and distil into 15 ml of cooled water
Storage. Store protected from moisture, at temperature not until a total volume 000 ml is obtained. Rinse the condenser
exceeding 30°. with 5 ml of water and dilute the combined distillate and
rinsings to 40 ml with water. 20 ml of the resulting solution
Labelling. The label states the quantity of active ingredient complies with the limit test for arsenic. Use a mixture of0.5 ml
per pre-metered .unit. of arsenic standardsolution (l ppm As) and 24.5 ml ofwater
to prepare the standard (5 ppm).
Barium. Shake 20.0 g with 30 ml of hydrochloric acid, add
Titanium Dioxide 100 ml of distilled water and boil. Filter while hot through a
hardened filter paper until a clear filtrate is obtained. Wash the
Mol. Wt. 79.9 filter. with 60 ml of distilled water and dilute the combined
Titanium Dioxide contains not less than 98.0 per cent and not filtrate and washings to 200 ml with distilled water. To 10 ml of
more than 100.5 per cent ofTiOz. this solution add 1 ml of 1 M sulphuric acid. After 30'minutes
any opalescence is not more intense than that of a mixture of
Category. Pharmaceutical aid and topical protectant. 10 ml of the test solution and 1 ml of distilled water.
Description. A white or almost white, infusible powder; Heavy metals (2.3.13). Dilute 10 ml ofthe solution prepared in
odourless. the test for Barium to 20 ml with water. 12 ml ofthe solution
complies with the limit test for heavy metals, Method D
Identification
(20 ppm).
A. When strongly heated it becomes pale yellow; the colour Iron. To 8 ml of solution A, add 4 ml of water, mix and add
is discharged on cooling. 0.05 ml ofbromine water, allow to stand for 5 minutes, remove
B. To 0.5 g, add 5 g ofanhydrous sodium sulphate and 10 ml the excess of bromine with a current of air and add 3 ml of
ofwater and mix. Add 10 ml ofsulphuric acid and boil gently potassium thiocyanate solution. Any colour in the solution
until clear; cool, add slowly 30 ml ofa 25 per cent v/v solution is not more intense than that in a standard prepared at the
ofsulphuric acid and dilute with water to 100 ml (solution A). same time and in the same manner using a mixture of4 ml of
To 5 ml of solution A add 0.1 ml ofstrong hydrogen peroxide iron standard solution (2 ppm Fe) and 8 ml of a 20 per cent
solution; an orange-red colour is produced. w/v solution ofsulphuric acid (200 ppm).
C. To 5 ml of solution A add 0.5 g of zinc, in granules; after Assay. Weigh accurately about 0.5 g, transfer to a 300 ml
45 minutes a violet-blue colour is produced. Kjeldahl flask, add 5 g of anhydrous sodium sulphate and
10 ml ofwater, mix and add 10 ml ofsulphuric acid. Boil gently
Tests until clear, cool, add slowly 40 ml of cooled sulphuric acid
(25 per cent), cool again and dilute with water to 100.0 ml
Appearance of solution. Solution A is not more opalescent
(solution B). To 300 g ofzinc, in granules, add 300 ml ofa2 per
than opalescence standard OS2 (1.4.1), and colourless (2.4.1).
cent w/v solution of mercuric nitrate and 2 ml of nitric acid,
Acidity or alkalinity. Shake 5.0 g with 50 ml ofcarbon dioxide- shake for 10 minutes and wash with water. Pack the zinc
free water for 5 minutes and centrifuge until a clear solution is amalgam into a glass tube'(400 mm x 20 mm) fitted with a tap
obtained. To 10 ml ofthe solution add 0.1 ml of bromothymol and a filter plate. Pass through the column, at a rate of about
blue solution. Not more than 1.0 ml of either 0.01 M hydro- 3 ml per minute, 100 ml of 1 M sulphuric acid followed by
chloric acid or 0.01 M sodium hydroxide is required to change 100 ml of water, ensuring that the amalgam is covered with
the colour of the solution. liquid throughout. Pass slowly through the column, at a rate

2229

www.webofpharma.com
TIZANIDINE HYDROCHLORIDE IP 2010

of about 3 ml per minute, 200 mlof 0.5 M sulphuric acid Tests


followed by 100 ml of water. Collect the combined eluates in a
500-ml conical flask containing 50 ml of a 15 per cent w/v pH (2.4.24). 3.5 to 5.3, determined on 5.0 per cent w/v solution
solution of ferric ammonium sulphate in sulphuric acid in water.
(25 per cent) and titrate immediately with 0.1 M eerie Related substances. Determine by liquid chromatography
ammonium nitrate usingferroin solution as indicator until a (2.4.14).
greenish colour is obtained (nl ml). Pass slowly through the
column 100 ml of 0.5 Msulphuric acid followed by 20.0 ml of Test solution. Dissolve 50 mg of the substance under
solution B, wash with 100 ml of 0.5 M sulphuric acid followed examination in 50.0 ml ofthe mobile phase.
by 100 ml of water. Collect the combined eluates in a 500-ml Reference solution (a). A 0.1 per cent w/v solution of
conical flask containing 50 ml ofa 15 percentw/v solution of tizanidine hydrochloride RS in the mobile phase.
ferric ammonium sulphate in sulphuric acid (25 per cent),
rinse the lower end of the column with water and titrate Reference solution (b). Dilute I ml ofreference solution (a) to
immediately with 0.1 M eerie ammonium nitrate usingferroin 100 ml with mobile phase.
solution as indicator until a greenish colour is obtained Chromatographic system
(n2 ml). Calculate the percentage content of Ti0 2 from the
a stainless steel column 25 cm x 4.6 mm packed with
expression 3.99(n2 - nl)/w
octadecylsilane bonded to porous silica (5 11m),
Where, w is the weight, in g, of the substance under exami- column temperature 50 0 ,
nation used in the preparation of solution A. mobile phase: a mixture of 80 volumes of buffer solution
Storage. Store protected from moisture; Avoid contact with prepared by dissolving 3.5 g of sodium-I-pentane
aluminium. sulphonate in 1000 ml of water, adjust the pH to 3.0 with
12 per cent orthophosphoric acid solution or 1 M
sodium hydroxide and 20 volumes of acetonitrile,
flow rate. I ml per minute,
Tizanidine Hydrochloride spectrophotometer set at 230 nm,
injection volume. 10 Ill.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency is
not less than 5000 theoretical plates.
,Hel
Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any
secondary peak is not more than 0.5 times the area ofthe peak
Mol. Wt. 290.2
in the chromatogram obtained with reference solution (b) (0.5
Tizanidine Hydrochloride is 5-chloro-N-(2-imidazolin-2-yl)- per cent) and the sum of areas of all the secondary peaks is
2,1 ,3-benzothiadiazol-4-yl amine. not more than the area of the peak in the cl:rromatogram
Tizanidine Hydrochloride contains not less than 98.0 per cent obtained with the reference solution (b) (1.0 per cent).
and not more than 102.0 per cent ofCgHgClNsS.HCI, calculated Heavy metals (2.3.13). Dissolve 1.0 g in 20 rnl ofwater. 12 rnl of
on the dried basis. this solution complies with limit test for heavy metals, Method
Category. a-adrenergic agonist. D(20ppm).
Dose. 2 mg three times a day.
Total Chloride. 11.9 per centto 12.5 per cent.
Description. A white to yellowish white, crystalline powder.
Weigh accurately about 0.5 g and dissolve in 50 ml of water.
Identification Titrate with 0.1 M silver nitrate. Determine the end point
A. Determine by infrared absorption spectrophotometry (2.4.6). potentiometrically (2.4.25). Carry out a blank titration.
Compare the spectrum with that obtained with tizanidine I ml of 0.1 M silver nitrate is equivalent to 0.003545 g of
hydrochloride RS.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the Sulphated ash (2.3.18). Not more than 0.1 percent.
chromatogram obtained with the reference solution. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
C. Gives reaction A for chlorides (2.3.1). on I g by drying in an oven at 105 0 for 3 hours.

2230

www.webofpharma.com
IP 2010 TlZANIDINE TABLETS

Assay. Determine by liquid chromatography (2.4.14). Calculate the content of C9HsClNsS in the medium from the
absorbance obtained from a solution of known concentration
Test solution. Dissolve 25 mg of the substance under
of tizanidine Hydrochloride RS in the same medium.
examination in 50.0 ml ofthe mobile phase. Dilute 10.0 ml of
the solution to 50.0 ml with the same solvent. D. Not less than 70 per cent of the stated amount of
Reference solution. A 0.01 per cent w/v solution of tizanidine C9HsClNsS.
hydrochloride RS in the mobile phase.
Uniformity of content. Comply with the tests stated under
Chromatographic system Tablets.
- a stainless steel column 15 cm x 4.6 mm packed with
Determine by liquid chromatography (2.4.14), as described
octylsilane bonded to porous silica (5 Ilm),
mobile phase: a mixture of 50 volumes ofa buffer solution under Assay.
prepared by dissolving 6.8 g of monobasicpotassium Test solution. Cmsh one tablet and disperse in 50 ml phosphate
phosphate in 1000 ml of water adjusting the pH to 7.5 bufferpH 6. 6, dilute to 100.0 ml with acetonitrile and filter.
with 5.3 M potassium hydroxide, and 50 volumes of
acetonitrile, Reference solution. Weigh accurately 10 mg of tizanidine
flow rate. 1 ml per minute, hydrochloride RS, dissolve in 25 mlphosphate bufferpH 6.6
spectrophotometer set at 230 urn, and dilute to 50.0 ml with acetonitrile. Dilute 5.0 ml of this
injection volume. 10 Ill. solution to 50.0 ml with acetonitrile.
Inject the reference solution. The test is not valid unless the Calculate the content ofC 9H sClNsS.
tailing factor is not more than 2.0 and the relative standard
deviation for replicate injections is not more than 2.0 per cent. Other tests. Comply with the tests stated underTablets.
Inject the test solution and the reference solution. Assay. Determine by liquid chromatography (2.4.14).
Calculate the content ofC 9H sClNsS.HCl. Test solution. Weigh'and powder 20tablets. Weigh accurately
Storage. Store protected from light. a quantity of the powdered tabl.et containing about 20 mg of
Tizanidine, disperse in 50 ml phosphate buffer pH 6.6 and
dilute to 100.0 ml with acetonitrile and filter.

Tizanidine Tablets Reference solution. Weigh accurately '10 mg of


tizanidine hydrochloride RS, dissolve in 25 ml phosphate
Tizanidine Hydrochloride Tablets buffer pH 6. 6 and dilute to 50.0 ml with acetonitrile.
Tizanidine Tablets contain not less than 90.0 per cent and not
Chromatographic system
more than 110.0 per cent of the stated amount oftizanidine,
C9HsClNsS. - a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 Ilm),
Dose. 2 mg; 4 mg.
mobile phase: a mixture of 80 volumes of phosphate
buffer pH 6.6 and 20 volumes of acetonitrile,
Identification
- flow rate. 1 ml per minute,
In the Assay, the principal peak in the chromatogram obtained - spectrophotometer set at 320 nm,
with the test solution corresponds to the peak in the - injection volume. 20 Ill.
chromatogram obtained with the reference solution.
Inject the reference solution. The test is not valid unless the
Tests relative standard deviation for replicate injections is not more
than 2.0 per cent.
Dissolution (2.5.2).
Apparatus No.1, Inject the test solution and the reference solution.
Medium: 900 ml of 0.1 M hydrochloric acid, Calculate the content ofC 9HsClNsS.
Speed and time. 50 rpm and 45 minutes.
Storage. Store protected from light and moisture.
Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance ofthe filtrate, suitably diluted with dissolution Labelling. The label states the strength in, terms of the
medium ifnecessary, at the maximum at about 320 nm (2.4.7). equivalent amount ofTizanidine.

2231

www.webofpharma.com
TOBRAMYCIN IP 2010

Tobramycin cent) and heat at 105° for 10 minutes. The principal spot in the
chromatogram obtained with the test solution corresponds to
that in the chromatogram obtainedwith reference solution (a).
OH The test is not valid unless the chromatogram obtained with
reference solution (b) shows three clearly separated principal
spots.
NH 2
B. Dissolve about 5 mg in 5 ml of water, add 5 ml ofa 0.1 per
cent w/v solution of ninhydrin in ethanol (95 per cent) and
heat in a water-bath for 3 minutes; a violet-blue colour
HO develops.

NH 2 Tests
NH 2
pH (2.4.24). 9.0 to 11.0, determined in a 10.0 per cent w/v
solution.
Specific optical rotation (2.4.22). +138° to +148°, determined
Mol. Wt. 467.3 in a 4.0 per cent w/v solution.
Tobramycin is 6-0-(3-amino-3-deoxy-a-D-glucopyranosyl)- 2-Methyl-l-propanol. Not more than 1.0 per cent w/w,
2-deoxy-4-0-(2,6-diamino-2,3,6-trideoxy-a -D-ribo- determined by gas chromatography (2.4.13).
hexopyranosyl)-D-streptamine, an antimicrobial substance
Test solution (a). A 10 per cent w/v solution ofthe substance
produced by Streptomyces tenebrarius or by any other
under examination in wate!:
means.
Test solution (b). A solution containing 10 per cent w/v of the
Tobramycin has potency not less than 930 Units per mg,
substance under examination and 0.2 per cent v/v of
calculated on the anhydrous and 2-methyl-l-propanol-free
2-propanol (internal standard).
basis.
Reference solution. A solution containing 0.1 per cent w/v of
Category. Antibacterial.
2-methyl-l-propanol and 0.2 per cent v/v of the internal
Dose. By intramuscular or intravenous injection, 3 to 5 mg per standard.
kg daily, in divided doses.
Chromatographic system
Description. A white or almost white powder. a glass column 1.5 m x 4 mm, packed with porous polymer
beads (80 to 100 mesh) (such as Porapak Q),
Identification - temperature. column 165°,
A. Determine by thin-layer chromatography (2.4.17), coating Calculate the percentage w/w of 2-methyl-l-propanol.
the plate with silica gel H.
Related substances. Determine by thin-layer chromatography
Mobile phase. A mixture of 60 volumes of methanol, (2.4.17), coating the plate with silica gel H.
40 volumes of strong ammonia solution and 20 volumes of
chloroform. Mobile phase. A mixture of equal volumes of strong ammonia
solution, 2-butanone and ethanol (95 per cent).
Test solution. Dissolve 0.4 g of the substance under
examination in 100 ml of water. Test solution. Dissolve 0.8 g of the substance under
examination in 100 ml of 0.02 M ammonia.
Reference solution (a). A 0.4 per cent w/v solution of
tobramycin RS in water. Reference solution. A 0.008 per cent w/v solution of the
substance under examination in 0.02 M ammonia.
Reference solution (b). A solution containing 0.4 per cent
Apply to the plate 5 I!l of each solution. After development,
w/v each of kanamycin sulphate RS, neomycin sulphate RS
dry the plate in warm air, heat at 110° for 10 minutes and spray
and tobramycin RS in water.
t~e hotplate with a so!~i~!J:J~r_eEl3:r~_~_~~_~i~!~IY_~~X()r~_~~e
Apply to the plate 5 I!l of each solution. After development, by diluting sodium hypochlorite solution (3 per cent CI)
dry the plateiilwaim arr,spiay witha mlxtllieofequal volumes fo
with water cOiltainO.5 percent Vi/v of available chlorine.
of a 46 per cent w/v solution of sulphuric acid and a 0.2 per Dry in a current of cold air until a sprayed area of the plate
cent w/v solution of 1,3-naphthalenediol in ethanol (95 per below the line of application gives at most a very faint blue

2232

www.webofpharma.com
IP 2010 TOBRAMYCIN INJECTION

colour with a drop of potassium iodide and starch solution; Mobile phase. A mixture of 60 volumes of methanol,
avoid prolonged exposure to cold air. Spray the plate with 40 volumes of strong ammonia solution and 20 volumes of
potassium iodide and starch solution. Any secondary spot chloroform.
in the chromatogram obtained with the test solution is not
Test solution. Dilute a suitable volume of the injection with
more intense than the spot in the chromatogram obtained
water to produce a solution containing 0.4 per cent w/v
with the reference solution.
solution oftobramycin.
Sulphated ash (2.3.18). Not more than OJ per cent.
Reference solution (a). A 0.4 per cent w/v solution of
Water (2.3.43). Not more than 8.0 per cent, determined on tobramycin RS in water.
OJ g. Reference solution (b). A solution containing 0.4 per cent
Assay. Determine by the microbiological assay ofantibiotics, w/v each of kanamycin sulphate RS, neomycin sulphate RS
Method B (2.2.10). and tobramycin RS in water.

Tobramycin intendedfor use in the manufacture ofparenteral Apply to the plate 5 III of each solution. After development,
preparations without a further appropriate procedure for dry the plate in wann air, spray with a mixture ofequal volumes
the removal of bacterial endotoxins complies with the of a 46 per cent w/v solution of sulphuric acid and a 0.2 per
following additional requirement. cent w/v solution of 1,3-naphthalenediol in ethanol (95 per
cent) and heat at 105° for 10 minutes. The principal spot in the
Bacterial endotoxins (2.2.3). Not more than 2.0 Endotoxin Units
chromatogram obtained with the test solution corresponds to
per mg oftobramycin. that in the chromatogram obtained with reference solution (a).
Tobramycin intendedfor use in the manufacture ofparenteral The test is not valid unless the chromatogram obtained with
preparations or eye drops without a further appropriate reference solution (b) shows three clearly separated principal
sterilisation procedure complies with the following spots.
additional requirements. B. Gives the reactions ofsulphates (2.3.1).
Sterility (2.2.11). Complies with the test for sterility.
Tests
Storage. Store protected from moisture. Ifthe material is sterile,
the container should be sterile, tamper-evident and sealed so pH (2.4.24). 3.5 to 6.0.
as to exclude micro-organisms.
Related substances. Determine by thin-layer chromatography
Labelling. The label states (1) the number ofUnits per ri:J.g; (2) (2.4.17), coating the plate with silica gel H .
where applicable, that it is sterile; (3) where applicable, that it
is free from bacterial endotoxins and depressor substances. Mobile phase. A mixture ofequal volumes ofstrong ammonia
solution, 2-butanone and ethanol (95 per cent).
Test solution. Dilute a suitable volume of the injection with
0.01 M ammonia to obtain a solution containing 40 mg of
Tobramycin in 4 ml. Shake with 10 ml of ether and use the
Tobramycin Injection aqueous layer.
Tobramycin Sulphate Injection Reference solution. A 0.008 per cent w/v solution of the
Tobramycin Injection is a sterile solution of Tobramycin in substance under examination in 0.02 M ammonia.
Water for Injections containing sufficient Sulphuric Acid to Apply to the plate 5 III of each solution. After development,
adjust the pH to 3.5 to 6.0. dry the plate in warm air, heat at 110° for 10 minutes and spray
Tobramycin Injection contains not less than 90.0 per cent and the hot plate with a solution prepared immediately before use
not more than 120.0 per cent of the stated amount of by diluting sodium hypochlorite solution (3 per cent CI)
tobramycin, ClsH37NsOg. with water to contain 0.5 per cent w/v of available chlorine.
Dry in a current of cold air until a sprayed area of the plate
Usual strengths. 10 mg per ml; 40 mg per ml. below the line of application gives at most a very faint blue
Description. A colourless solution. colour with a drop of potassium iodide and starch solution;
avoid prolonged exposure to cold air. Spray the plate with
Identification potassium iodide and starch solution. Any secondary spot
in the chromatogram obtained with the test solution is not
A. Determine by thin-layer chromatography (2.4.17), coating more intense than the spot in the chromatogram obtained
the plate with silica gel H with the reference solution.

2233

www.webofpharma.com
TOCOPHERYLACETATE IP 2010

Bacterial endotoxins (2.23). Not more than 2.0 Endotoxin C. Determine by thin-layer chromatography (2.4.17), coating
Units per mg oftobramycin. the plate with silica gel HF254.
Other tests. Complies with the tests stated under Parenteral Mobile phase. A mixture of 80 volumes of cyclohexane and
Preparations (Injections). 20 volumes of ether.
Assay. Determine by the microbiological assay ofantibiotics, Test solution (a). Dissolve 0.5 g of the substance under
Method B (2.2.10). examination in 100 ml of cyclohexane.
Calculate the content of tobramycin in the injection, taking Test solution (b). Dissolve 10 mg of the substance under
each 1000 Units found to be equivalentto 1 mg oftobnimycin. examination in 2 ml of 5 M ethanolic sulphuric acid, heat on
The upper fiducial limits of error is not less than 97.0 per cent a water-bath for 5 minutes, cool, add 2 ml of water and 2 ml of
and not more than 110.0 per cent of the stated potency. cyclohexane and shake for 1 minute; use the upper layer.
Reference solution (a). A 0.5 per cent w/v solution of
CMocopheryl acetate RS in' cyclohexane.
Reference solution (b). Prepare in the same maimer as test
Tocopheryl Acetate. solution (b) but using 10 mg of a-tocopheryl acetate RS in
place of the substance under examination.
(X- Tocopheryl Acetate; (X-Tocopherol Acetate; Vitamin

E Acetate Apply to the plate 10 III of each solution. After development,


dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with test
solution (a) correspondsto that in the chromatogram obtained
withTeference solution (a). There are two spots in each ofthe
chromatograms obtained with test solution (b) and reference
solution (b). The spots of higher R f value are due to
a-tocopheryl acetate and correspond to that in the
chromatogram obtained with reference solution (a). The spots
oflower Rf value are due to a-tocopheryl. Spray the plate with
Mol. Wt. 472.8 a mixture of 1 volume of hydrochloric acid, 4 volumes of a
Tocopheryl Acetate is (2RS,4'RS,8'RS)-6-acetoxy- 0.25 per cent w/v solution of ferric chloride in ethanol
2,5,7,8- tetramethyl-2-(4', 8', 12'-trimethyltri- (95 per cent) and 4 volumes of a 1.0 per cent w/v solution of
decyl)chroman(all-rac-a-tocopherol acetate). 1,10-phenanthroline hydrochloride in ethanol (95 per cent).
In the' chromatograms obtained with test solution (b) and
Tocopheryl Acetate contains not less than 96.0 per cent and
reference solution (b) the spot oflower R f value a-tocopheryl
not more than 102.0 per cent ofC31Hs203.
is orange.
Category. Vitamin E supplement.
Dose. Prophylactic, 5 to 10 mg; therapeutic, to be determined Tests
by the physician according to the needs of the patient.
Refractive index (2.4.27). 1.494 to 1.498, determined at 20°.
Description. A clear, slightly greenish yellow, viscous, oily
Acid value (2.3.23). Not more than 2.0, determined on 2.0 g.
liquid.
Free tocopherol. Not more than 2.0 per cent, determined by
Identification the following method. Weigh accurately about 0.5 g, dissolve
in 100 ml of 0.25 M ethanolic sulphuric acid, add 20 ml of
Test A may be omitted iftests Band C are carried out. Tests B water and 0.1 ml of a 0.25 per cent w/v solution of
arid C may be omitted if test A is carried out. diphenylamine in sulphuric acid and titrate with 0.01 M eerie
A. Determine by infrared absorption spectrophotometry (2.4.6). ammonium nitrate until a bluecolour is produced that persists
Compare the spectrum with that obtained with a-tocopheryl for at least 5 seconds. Repeat the operation without the
acetate RS. substance under examination. The difference between the
B.W1;J.en examinSldin the range 230 om to 360 om (2.4.7), I:!
required.
0.01 per cent w/v solution in ethanol exhibits a maximum at
about 284 nm, a shoulder at about278 nm and a minimum at 1 ml of 0.01 M eerie ammonium nitrate is equivalent to
about 254 nm. 0.002154 g oftocopherol.

2234

www.webofpharma.com
IP 2010 TOLAZAMIDE TABLETS

Heavy metals (2.3.13). 1.0 g complies with the limit test for absorbances at the maxima are about 0.78, aboutO.83 and
heavy metals, Method B (20 ppm). about 0.62 respectively.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Tests
Assay. Weigh accurately about 0.3 g, dissolve in 25 ml of
Related substances. Determine by thin-layer chromatography
ethanol (95 per cent), add 20 ml of 2.5 M ethanolic sulphuric
(2.4.17), coating the plate with silica gel G
acid and heat on a water-bath under a reflux condenser for
3 hours. Cool, transfer the solution quantitatively to a 200-ml Mobile phase.A mixture of200 volumes of chloroform, 100
volumetric flask, rinse the apparatus with ethanol (95 per volumes of methanol, 60 volumes of cyclohexane and 23
cent) and add the rinsings to the flask. Make up to volume volumes of 13.5 M ammonia.
with ethanol (95 per cent) and mix. To 25.0 ml ofthe resulting
Test solution. Dissolve 1.0 g of the substance under
solution in a flask add 25 ml of 0.25 M ethanolic sulphuric
examination in 50 ml of acetone.
acid, 10 ml of water and titrate with.0.01 M eerie ammonium
nitrate using 0.1 ml of diphenylamine as indicator, until a blue Reference solution. A 0.01 per cent w/vsolution of toluene-
colour persisting for at least 5 seconds is obtained. Repeat p-sulphonamide in acetone.
the operation without the substance under examination. The Apply to the plate 10 III ofeach solution. After removal ofthe
difference between the titrations represents the amount of plate, dry in a current of cold air, heat at 110° for 10 minutes,
eerie ammonium nitrate required. place the hot plate in a tank of chlorine gas prepared by the
1 ml of 0.01 M eerie ammonium nitrate is equivalent to addition of hydrochloric acid to a 5 per cent w/v solution of
0.002364 g ofC31H5203. potassium permanganate contained in a beaker placed in the
tank and allow to stand for 2 minutes. Dry it in a CillTent of
Storage. Store protected from light and moisture.
cold air until an area ofthe plate below the line of application
gives at most a very faint blue colour with a 0.5 per cent w/v
solution of potassium iodide in starch mucilage; avoid
prolonged exposure to cold air. Spray the plate with a 0.5 per
Tolazamide cent w/v solution of potassium iodide in starch mucilage.
Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Heavy metals (2.3.13).1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Sulphated ash (2.3.18). Not more than 0.2 per cent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Ct.#2tN303S Mol. Wt. 311.4 on 1.0 g by drying in an oven at 60° at a pressure not exceeding
Tolazamide is l-perhydroazepin-l-yl-3-tolyl-p- 0.7kPa.
sulphonylurea Assay. Weigh accurately about 0.5 g, dissolve in 20 ml of
Tolazamide contains not less than 98.0 per cent and not more butan-2-one with the aid of gentle heat. Allow to cool, add 30
than 101.0 per cent of C14H2tN303S, calculated on the dried ml of ethanol (95 per cent) and titrate with 0.1 M sodium
basis. hydroxide using phenolphthalein solution as indicator.

Category. Hypoglycaemic. 1 ml of 0.1 M sodium hydroxide is equivalent to 0.03114 g of


Ct.#21 N 30 3S.
Description. A white or almost white, crystalline powder.

Identification
A. Determine by infrared absorption spectrophotometry (2.4.6). Tolazamide Tablets
Compare the spectrum with that obtained with tolazamide RS
or with the reference spectrum oftolazamide. Tolazamide Tablets contain not less than 95.0 per cent and not
more than 105.0 per cent ofthe stated amount oftolazamide,
B. When examined in the range 230 to 350 nm (2.4.7), a 0.04 per
Ct.#2t NP3 S,
cent w/v solution in ethanol (95 per cent) exhibits maxima at
256 mu, 263 nm and 275 nm and a shoulder at 268 nm. The Usual strengths. 250 mg; 500 mg.

2235

www.webofpharma.com
TOLAZAMIDE TABLETS lP 2010

Identification Tolbutamide
Triturate a quantity of the powdered tablets containing about
0.25 g ofTolazamide with 50 m1 of acetone and filter. Evaporate
the filtrate to dryness and dry the residue at 60 0 at a pressure
of 2 kPa for 3 hours. On the residue, determine by infrared
absorption spectrophotometry (2.4.6). Compare the spectrum
with that obtained with tolazamide RS or with the reference
spectrum oftolazamide.
Mol. Wt. 270.4
Tests
Tolbutamide is 3-butyl-1-[(4-methylphenyl)sulphonyl]urea.
Related substances. Determine by tliin-layer chromatography
Tolbutamide contains not less than 99.0 per cent and not more
(2.4.17), coating the plate with silica gel G.
than 101.0 per cent ofCI2HlsNz03S, calculated on the dried
Mobile phase. A mixture of 23 volumes of 13.5 M ammonia, basis.
60 volumes of cyclohexane , 100 volumes of methanol and Category. Hypoglycaemic.
200 volumes of chloroform.
Dose. 500 mg to 1.5 g daily as a single dose with breakfast or
Test solution. Shake a quantity of the powdered tablets in divided doses.
containing about 0.2 g ofTolazamide with 10 ml of acetone Description. A white, crystalline powder; almost odourless.
and filter.
Identification
Reference solution. A 0.0 1 per cent w/v solution of toluene-p-
sulphonamide in acetone. Test A may be omitted iftests B, C and D are carried out. Tests
B, C and D may be omitted if test A is carried out.
Apply to the plate 10 III of each solution. After development,
dry the plate in a current of cold air, heat at 1100 for 10 minutes, A. Determine by infrared absorption spectrophotometry (2.4.6).
place the hot plate in a tank of chlorine gas, prepared by the Compare the spectrum with that obtained with tolbutamide
addition of hydrochloric acid to a 5 per cent w/v solution of RS or with the reference spectrum oftolbutamide.
potassium permanganate contained in a beaker placed in the B. Dissolve 25 mg in sufficient methanol to produce 100.0 ml.
tanle and allow to stand for 2 minutes. Dry the plate in a
When examined in the range 245 nm to 360 nm (2.4.7), the
current of cold air until an area of the plate below the line of
resulting solution shows absorption maxima at about 258 nm,
application gives at most a very faint blue colour with a 0.5 per
263 nm and 275 nm and a shoulder at about 268 nm. Dilute the
cent w/v solution of potassium iodide in starch mucilage
solution with methanol to produce a 0.001 per cent w/v
avoid prolonged exposure to cold air. Spray the plate with a
solution. When examined in the range 220 nm to 235 nm, the
0.5 per cent w/v solution of potassium iodide in starch
resulting solution shows an absorption maximum at about
mucilage. Any secondary spot in the chromatogram obtained
228 nm; absorbance at about 228 nm, about 0.50.
with the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution (0.5 per C. Boil 0.1 gwith 8 ml ofa 50 per centv/v solution ofsulphuric
cent). acid under a reflux condenser for 30 minutes. Make the solution
strongly allcaline with sodium hydroxide solution and steam
Other tests. Comply with the tests stated under Tablets. distil for 30 minutes, receiving the distillate in 20 ml of 0.1 M
hydrochloric acid. To 1 ml of the solution containing the
Assay. Weigh and powder 20 tablets. Shake a quantity of the
distillate add 0.1 g of sodium acetate and 10 mlof buffer
powdered tablets containing about 0.5 g of Tolazamide with
solution pH 9.4. Cool in an ice-bath for 10 mmutes, add 1 mlof
50 ml of chloroform, filter, wash the residue with chloroform
diazotised nitroaniline solution, set aside for 20 minutes and
and evaporate the combined filtrate and washings to dryness.
add dropwise 1 ml of sodium hydroxide solution; an orange-
Dissolve the residue in 20 ml of butan-2-one with the aid of
red colour is produced.
gentle heat. Allow to cool, add 30 ml of ethanol (95 per cent)
and titrate the resulting solution with 0;1 Msodiumhydroxide Do Boil 0.1 gwith 8 ml ofa 50 per centv/vsolution of sulphuric
using phenolphthalein solution as indicator. acid under a reflux condenser for 30 minutes. Cool in an ice-
bath; a crystalline precipitate of 4-toluenesulphonylamide is
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03114 g of formed, which after recrystallisation from hot water and dry'ing
CIJIzIN303S, at105°melts at 135° to 140° (2.4.21).

2236

www.webofpharma.com
IP 2010 TOLBUTAMIDE TABLETS

Tests 1 ml of 0.1 M sodium hydroxide is equivalent to 0.02704 g of


C12HlsN203S.
Appearance of solution. A 2.0 per cent w/v solution in 1 M
sodium hydroxide is clear (2.4.1), and colourless (2.4.1). Storage. Store protected from moisture.

pH (2.4.24). 4.5 to 5.5, determined in a solution prepared by


dissolving 2.0 g in 50 ml of carbon dioxide-fi-ee water by
Tolbutamide Tablets
heating at 70° for 5 minutes, cooling rapidly and filtering.
Tolbutamide Tablets contain not less than 95.0 per cent and
Non-sulphonyl urea. Dissolve 0.5 gin 1 ml of dilute ammonia
not more than 105.0 per cent of the stated amount of
solution and 9 ml of water; not more than a faint opalescence
tolbutamide, C12HlsN203S.
is produced.
Usual strength. 500 mg.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G Identification
Mobile phase. A mixture of 90 volumes of chloroform, Extract a quantity of the powdered tablets containing 1 g of
8 volumes of methanol and 2 volumes of anhydrous formic Tolbutamide with 10 ml of chloroform, filter, evaporate the
acid. filtrate to dryness, scratching the sides of the vessel, if
Test solution. Dissolve 0.5 g of the substance under necessary, to induce crystallisation, and dry the residue at
examination in 10 ml of acetone. 105° for 30 minutes. The residue complies with the following
tests.
Reference solution (a). A 0.015 per cent w/v solution of
4-toluenesulphonamide in acetone. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with tolbutamide
Reference solution (b). A mixture of equal volumes of the RS or with the reference spectrum of tolbutamide.
test solution and reference solution (a).
B. Boil 0.1 g ofresidue with 8 ml of a 50 per cent v/v solution
Apply to the plate 5 III of each of test solution and reference of sulphuric acid under a reflux condenser for 30 minutes.
solution (a) and 10 III of reference solution (b). After Cool in an ice-bath; a crystalline precipitate of
development, dry the plate in a current ofwarm air and heat at 4-toluenesulphonylamide is formed, which after
110° for 10 minutes. While still hot, place the plate in a recrystallisation from hot water and drying at 105° melts at
chromatographic tame with an evaporating dish containing a 135° to 140°.
5 per cent w/v solution of potassium permanganate, add an
equal volume of hydrochloric acid and close the tank. Leave
Tests
the plate in the tank for 2 minutes, then place it in a current of Related substances. Determine by thin-layer chromatography
cold air until the excess of chlorine is removed and an area of (2.4.17), coating the plate with silica gel G
coating below the line of application gives only a very faint
Mobile phase. A mixture of 90 volumes of chloroform,
blue colour with potassium iodide and starch solution; avoid
8 volumes of methanol and 2 volumes of anhydrous formic
prolonged exposure to cold air. Spray the plate with potassium acid. .
iodide and starch solution and allow to stand for 5 minutes.
Any secondary spot in the chromatogram obtained with the Test solution. Shake a quantity of the powdered tablets
test solution is not more intense than the spot in the containing 0.5 g of Tolbutamide with 10 ml of acetone and
chromatogram obtained with reference solution (a). The filter.
chromatogram obtained with reference solution (b) shows two Reference solution (aJ. A 0.015 per cent w/v solution of
clearly separated spots. 4ctoluenesulphonamide in acetone.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Reference solution (b). A mixture of equal volumes of the
heavy metals, Method B (20 ppm). test solution and reference solution (a).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Apply to the plate 5 III of each of test solution and reference
solution (a) and 10 III of reference solution (b). After
.Loss on drying (2.4.19). Not more than 0.5 per cent, determined
development, dry the plate in a current ofwarm air and heat at
on 1.0 g by drying in an oven at 105° for 3 hours.
110° for 10 minutes. While still hot, place the plate in a
Assay. Weigh accurately about 0.5 g and dissolve in a mixture chromatographic tame with an evaporating dish containing a
of 40 ml of ethanol (95 per cent) and 20 ml of water. Titrate 5 per cent w/v solution of potassium permanganate, add an
with 0.1 M sodium hydroxide usingphenolphthalein solution equal volume of hydrochloric acid and close the tame. Leave
as indicator. the plate in the tame for 2 minutes, then place it in a current of

2237

www.webofpharma.com
TOLBUTAMIDE TABLETS IP 2010

cold air until the excess of chlorine is removed and an area of Description. A white or yellowish white powder.
coating below the line of application gives only a very faint
blue colour with potassium iodide and starch solution; avoid Identification
prolonged exposure to cold air. Spray the plate with potassium Test A may be omitted iftests B, C andD are carried out. Tests
iodide and starch solution and allow to stand for 5 minutes. B, C and D may be omitted if test A is carried out.
Any secondary spot in the chromatogram obtained with the
A. Determine by infrared absorption spectrophotometry (2.4.6).
test solution is not more intense than the spot in the
Compare the spectrum with that obtained with tolnaftate RS
chromatogram obtained with reference solution (a). The
or with the reference spectrum of toInaftate.
chromatogram obtained with reference solution (b) shows two
clearly separated spots. B. Determine by thin-layer chromatography (2.4.17), as
described under Related substances.
Dissolution (2.5.2).
The principal spot in the chromatogram obtained with test
Apparatus No.1,
solution (b) corresponds to that in the chromatogram obtained
Medium. 900 ml of a solution containing 2.04 per cent wlv of
with reference solution (a).
disodium hydrogen phosphate and 0.135 per cent wlv of
potassium dihydrogen phosphate, C. Mix about 1mg with 0.5 mlofsulphuric acid. Add 0.05 ml of
Speed and time. 50 rpm and 30 minutes. formaldehyde solution. A greenish-blue colour develops.
D. Melting range (2.4.21).109° to 112°.
Withdraw a suitable volume ofthe medium and filter. Measure
the absorbance of the filtrate suitably diluted if necessary, at Tests
the maximum at about 228 nm (2.4.7). Calculate the content of
CizHISNz03S in the medium taking 417 as the specific Appearance ofsolution. A 5.0 per cent wIv solution in acetone
absorbance at 228 nm. is clear (2.4.1) and not more intensely coloured than reference
solution YS6 (2.4.1).
D. Not less than 70 per cent of the stated amount of
ClzHISNz03S, Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Other tests. Comply with the tests stated under Tablets.
Test solution (a). Dissolve 0.1 g of the substance under
Assay. Weigh and powder 20 tablets. Weigh accurately a examination in 2 ml ofacetone.
quantity ofthe powder containing about 0.5 g ofTolbutamide,
add 50 ml of ethanol (95 per cent), previously neutralised to Testsolution (b). Dilute 0.5 ml oftest soh,ltion (a) to 10 ml with
phenolphthalein solution, warm to dissolve, add 25 ml of acetone.
water and titrate with 0.1 M sodium hydroxide using Reference solution (a). Dissolve 25 mg of toInaftate RS in 10
phenolphthalein solution as indicator. ml of acetone.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02704 g of Reference solution (b). Dilute 1 ml oftest solution (b) to 10 ml
CI2HISNz03S, with acetone.
Storage. Store protected from moisture. Reference solution (c). Dissolve 50 mg of{3-naphthol in 1 ml
of test solution (a) and dilute to 10 ml with acetone.
Apply to the plate 5 III of each solution. Allow the mobile
Tolnaftate phase to rise 12 em. Dry the plate in current ofair and examine

vJtro
under ultraviolet light at 254 nm. Any secondary spot in the
chromatogram obtained with test solution (a) is not more
intense than the spot in the chromatogram obtained with
H3C ~ 0 reference solution (b) (0.5 per cent). The test is not valid unless
the chromatogram obtained with reference solution (c) shows
CH3
two clearly separated spots.
C I9H 17NOS Mol.Wt. 307.4 Sulphated ash (2.3.18). Not more than 0.1 percent.
Tolnaftate is O-naphthalen-2-yl methyl(3-methylphenyl)- Loss ondrying (2.4.19). Not more than 0.5 per cent, determined
thiocarbamate. ()Ill ,0 g by drying in vacuUll1 at 60° aJ a pressure. not exceeding
TOlllliftlite. fOIltainsIl()t le.s§tJ:1aIl 9T·0 IJe.r cent aIld ll()tIl1o~e. 0.7 kPa for 3 hours.
than 103.0 per cent of C I9H 17NOS, calculated on the dried Assay. Dissolve 50 mg in 250 rn1 ofmethanol. Dilute 2 rn1 ofthe
basis. solution to 50 ml with methanol. Measure the absorbance at
Category. Antifungal. the maximum at 257 nm (2.4.7).

2238

www.webofpharma.com
IP 2010 TOLNAFTATE TOPICAL POWDER

Calculate the content ofC 19H I7NOS taking 720 as the specific Usualstrength. 1 per cent w/w.
absorbance at 257 run.
Identification
Storage. Store protected from light.
Determine by thin-layer chromatography (2.4.17), coating the
plate with silica GF254.

Tolnaftate Cream Mobile phase. Toluene.

Tolnaftate Cream contains not less than 90.0 per cent and not Test solution. Evaporate 10 ml ofthe SolutionAon steam bath
more than 110.0 per cent of the stated amount of tolnaftate, to dryness and dissolve the residue in 1.0 ml of ethanol (95
C I9H17NOS. per cent).
Reference solution. A 0.1 per cent w/v solution of tolnaftate
Identification RS in ethanol (95 per cent).
Determine by thin-layer chromatography (2.4.17), coating the Apply to the plate 10 III of each solution. Allow the mobile
plate with silica gel. phase to rise 8 cm. Dry the plate in air and examine in ultraviolet
Mobile phase. Toluene. light at 254 nm. The principal spot in the chromatogram
obtained with the test solution corresponds to that in the
Test solution. Evaporate 10 ml ofthe SolutionAon steam bath chromatogram obtained with the reference solution.
to dryness and dissolve the residue in 1.0 ml of ethanol (95
per cent). Tests
Reference solution. A 0,1 per cent w/v solution of tolnaftate Other tests. Complies with the tests stated under Gels.
RS in ethanol (95 per cent).
Assay. Transfer an accurately weighed quantity of the gel
Apply to the plate 10 III of each solution. Allow the mobile containing about 10 mg ofToInaftate to a separator containing
phase to rise 8 cm. Dry the plate in air and examine in ultraviolet 75 ml of chloroform. Wash the chloroform solution
light at 254 nm. The principal spot in the chromatogram successively with two 25 ml portions of 0.1 M hydrochloric
obtained with the test solution corresponds to that in the acid and 25 ml of water. Dilute to 100 ml with chloroform and
chromatogram obtained with the reference solution. mix (Solution A). Dilute 5.0 ml ofthe solution with chloroform
to 50 ml and mix. Measure the absorbance of the resulting
Tests solution at the maximum at about 258 run (2.4.7), using 0.001
Other tests. Complies with the tests stated under Creams. per cent w/v solution of tolnaftate RS in chloroform.

Assay. Transfer an accurately weighed quantity of the cream Calculate the content OfC I9H17NOS inthe gel.
containing about 10 mg ofToInaftate to a separator containing Storage. Store protected from moisture.
75 ml of chloroform. Wash the chloroform solution
successively with two 25 ml portions of 0.1 M hydrochloric
acid and 25 ml of water. Filter the chloroform layer through a
chloroform-washed cotton pledget into a 100-ml volumetric Tolnaftate Topical Powder
flask. Add chloroform to volume and mix (Solution A). Dilute
5.0 ml of the solution with chloroform to 50 ml and mix. Tolnaftate Topical Powder contains not .less than 90.0 per
Measure the absorbance of the resulting solution at the cent and not more than 110.0 per cent ofthe stated amount of
maximum at about 258 run (2.4.7), using 0.001 per cent w/v C I9H 17NOS.
solution of tolnaftate RS in chloroform.
Identification
Calculate the content ofC ,9H 17NOS in the cream.
Determine by thin-layer chromatography (2.4.17), coating the
Storage. Store protected from moisture. plate with silica gel GF254.
Mobile phase. Toluene.
Test solution. Evaporate the 5.0 ml portion of the methanol
Tolnaftate Gel solution reserved in the Assay, on a steam bath just to dryness
Tolnaftate Gel contains not less than 90.0 per cent and not and dissolve the residue in 1.0 ml of alcohol.
more than 11 0.0 per cent of the stated amount of tolnaftate, Reference solution. A 0.1 per cent w/v solution of tolnaftate
C I9H 17NOS. RS in ethanol (95 per cent).

2239

www.webofpharma.com
TOLNAFTATE TOPICAL POWDER IP 2010

Apply to the plate 10 III of each solution. Allow the mobile Identification
phase to rise 8 cm. Dry the plate in air and examine in ultraviolet
light at 254 nm. The principal spot in the chromatogram Determine by thin-layer chromatography (2.4.17), coating the
obtained with the test solution corresponds to that in the plate with silica gel GF25i
chromatogram obtained with the reference solution. Mobile phase. Toluene.
Test solution. Evaporate 25 ml of solution A on steam bath to
Tests dryness and dissolve the residue in 1.0 ml of the ethanol (95
Assay. Determine by liquid chromatography (2.4.14).
per cent).
Reference solution. A 0.1 per cent w/v solution of tolnaftate
Internal standard solution. A 0.1 per cent w/v solution of
RS in ethanol (95 per cent).
progesterone in methanol.
Apply to the plate 10 III of each solution. Allow the mobile
Test solution. Transfer an accurately weighed quantity of the
phase to rise 8 cm. Dry the plate in air and examine in ultraviolet
topical powder containing about 5.0 mg oftolnaftate to a screw-
light at 254 nm. The principal spot in the chromatogram
capped, 50 ml centrifuge tube. Add 25 ml of methanol, place
obtained with the test solution corresponds to that in the
the cap on the tube and rotate on a rotating device for 10
chromatogram obtained with the reference solution.
minutes, and centrifuge at about 2000 rpm for 5 minutes, filter
the supernatant and transfer 20 ml of the filtrate to 50-ml Tests
volumetric flask, retaining the remaining portion ofthe filtrate
for identification test. Add 5.0 ml ofinternal standard solution Assay. Take an accurately measured volume containing about
to the flask and dilute to volume with methanol and mix. 10 mg ofToInaftate, add 50 ml of chloroform and extract with
50 ml of 0.1 M sodium hydroxide. Filter the chloroform layer
Reference solution. A 0.02 per cent w/v solution of tolnaftate through a chloroform-washed cotton pledget into a 250- ml
RS in methanol. To 20 ml ofthis solution add 5.0 ml ofinternal volumetric flask and extract the aqueous layer with two 45 ml
standard solution and dilute to 50.0 ml with methanol. portions of chloroform, filtering each into the flask. Add
Chromatographic system chloroform to volume and mix (Solution A). Dilute 25 mlof
- a stainless steel column 30 cm x 4 mm, packed with this solution with chloroform to 100 ml and mix. Measure the
octadecylsilane bonded to pbrous silica (10 Ilm), absorbance ofthe resulting solution at the maximum at about
258 urn (2.4.7), using 0.001 per cent w/v solution of tolnaftate
- mobile phase: a mixture of 2 volumes of acetonitrile
RS in chloroform.
and 1 volume of water,
- flow rate. 1 ml per minute, Calculate the content ofC I9H 17NOS in topical solution.
- spectrophotometer set at 254 urn, Storage. Store protected from moisture.
- injection volume. 10 Ill.
Inject the reference solution. The test is not valid unless the
resolution between the tolnaftate and internal standard peak
is not less than 3.0 and the relative standard deviation for Tolterodine Tartrate
replicate injections is not more than 3.0 per cent. The relative
retention time of progesterone and tolnaftate are about 0.7 0HH H3 CCH3
minute and 1.0 minute respectively. y • HOXCOOH
~~NyCH3
Inject the test solution and the reference solution.
HOOC OH
Calculate the content ofC I9H 17NOS in the powder. ~ CH3

Storage. Store protected from moisture.

Mol Wt. 475.6


Tolterodine Tartrate is 2-[(1R)-3-[bis(l-methylethyl)amino]-1-
phenylpropyl]-4-methylphenol tartrate.
Tolnaftate Topical Solution Tolterodine Tartrate contains not less than 98.0 per cent and
Tolnaftate Topical Solution contains not less than 90.0 per not more than 102.0 per cent ofC22H3INO,C4H606,calculated
cent and not more than 115.0 per cent ofthe stated amount of on the dried basis.
tolnaftate, C I9 H 17NOS. Description. A white to off-white crystalline powder.

2240

www.webofpharma.com
IP 2010 TOLTERODINE TARTRATE

Identification Inject the test solution, reference solution (a) and (b). In the
chromatogram obtained with the test solution the area of any
A. Determine by infrared absorption spectrophotometry (2.4.6). secondary peak is not more than the area ofthe principal peak
Compare the spectrum with that obtained with tolterodine in the chromatogram obtained with reference solution (a) (0.5
tartrate RS or with the reference spectrum of t6/terodine per cent) and the sum of the areas of all the secondary peaks
tartrate. is not more than twice the area of the principal peak in the
B. In the Assay, the principal peak in the chromatogram chromatogram obtained with reference solution (a) (1.0 per
obtained with the test solution corresponds to the peak in the cent). Ignore the peak due to tartaric acid cOlTesponding to
chromatogram obtained with the reference solution. the principal peak in the chromatogram obtained with reference
solution (b).
Tests
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Specific optical rotation (2.4.22). +33° to +38°, detennined in heavy metals, Method B (20 ppm).
1.0 per cent w/v solution in methanol.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Related substances. Determine by liquid chromatography
(2.4.14). Loss on drying (2.4.1 9). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105°.
Solvent mixture. 10 volumes ofmobile phase A and 10 volumes
ofmobile phase B. Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve about 50 mg of the substance under Solvent mixture. 10 volumes ofmobile pqase A and 10 volumes
examination in 10 ml of water and dilute to 50.0 ml with the ofmobile phase B.
solvent mixture.
Test solution. Dissolve about 50 mg of the substance under
Reference solution (a). Dissolve about 10 mg of tolterodine examination in 20 ml of water, sonicate and dilute to 100.0 ml
tartrate RS in 20 ml of water and dilute to 100.0 ml with the with the solvent mixture. Dilute 5.0 ml ofthis solution to 50.0
solvent mixture. Dilute 5.0 ml ofthis solution to 100.0 ml with ml with the solvent mixture.
the solvent mixture.
Reference solution (a). Dissolve about 50 mg of tolterodine
Reference solution (b). A 0.03 per cent w/v solution oftartaric
tartrate RS in 20.0 ml of water, sonicate and dilute to 100.0 ml
acid in the solvent mixture.
with the solvent mixture. Dilute 5.0 ml ofthis solution to 50.0
Chromatographic system ml with the solvent mixture.
- a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 Jlm) (Such Reference solution (b). A 0.03per cent w/v solution oftartaric
as Inertsil), acid in the solvent mixture.
mobile phase: A. a 0.05 M potassium dihydrogen Chromatographic system
orthophosphate, adjusted to pH 3.5 with - a stainless steel column 25 cm x 4.6 mm, packed with
orthophosphoric acid, octadecylsilane bonded to porous silica (5 Jlm) (Such
B. acetonitrile, as Inertsil),
a linear gradient programme using the conditions given mobile phase: A. a 0.05 M potassium dihydrogen
below, orthophosphate, adjusted to pH 3.5 with
flow rate. 1 ml per minute, orthophosphoric acid,
spectrophotometer set at 215 nm, B. acetonitrile,
injection volume. 20 Jll. a linear gradient programme using the conditions given
Time Mobile Phase A Mobile Phase B below,
(in min) (per cent v/v) (per cent v/v) flow rate. 1 ml per minute, .
0-5 65 35 spectrophotometer set at 215 urn,
injection volume. 20 Jll.
5-20 65~50 35 ~50
Time Mobile Phase A Mobile Phase B
20-40 50~30 50 ~70
(in min) (per cent v/v) (per cent v/v)
40-41 30~65 70~35
0-5 65 35
41-50 65 35
5-20 65 ~50 35~50
Inject reference solution (a). The test is not valid unless the
20-21 50~65 50~35
relative standard deviation for replicate injections is not more
than 5.0 per cent. 21-25 65 35

2241

www.webofpharma.com
TOPlRAMATE IP 2010

Inject reference solution (a) and (b). The test is not valid Chromatographic system
unless the theoretical plates of the principal peak is not less a stainless steel column 25 cm x 4.6 mm, packed with
than 2000 and the tailing factor is not more than 2.0. The octadecylsilane bonded to porous silica (5 pm),
relative standard deviation for replicate injections is not more column temperature. 40°,
than 2.0 per cent. mobile phase: a mixture of 50 volumes of water and 50
volumes of acetonitrile,
Inject the test solution and the reference solution.
flow rate. 1 ml per minute,
Calculate the content ofCzzH31NO,C4H606. refractive index detector,
Storage. Store protected from moisture. detector cell temperature 50°,
injection volume. 50 Ill.
Inject the reference solution. Run the chromatogram for 40
Topiramate minutes. The retention times of topiramate impurity A and
impurity B are about 2.9 and 4.5 minutes and the relative
retention times with respect to topiramate are 0.54 and 0.84
respectively. The test is not valid unless the resolution between
topiramate impurity A and topiramate impurity B is not less
than 3.
Inject the test solution and the reference solution. For the
calculation ofcontent, multiply the peak areas ofthe following
impurities by the corresponding correction factor: topiramate
Mol. Wt. 339.4 impurity A = 1.09; topiramate impurity B = 0.94 .
Topiramate is 2,3 :4,5-di-O-l-isopropylidene-~-D­ Not more than 0.15 percent each ofimpurity Aand impurity
fructopyranose sulphamate. B, and not more than 0.1 per cent of any other impurity is
Topiramate contains not less than 98.0 per cent and not more found.
than 102.0 per cent ofClzHzlNOsS, calculated on the anhydrous Heavy metals (2.3.13). 1.0 g complies with the limit test for
basis. heavy metals, Method B (20 ppm).
Category. Anti-convulsant. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Dose. 25 mg daily.
Water (2.3.43). Not more than LOper cent, deterrninedon 1.0 g.
Description. A white to off-white powder. Assay. Detennine by liquid chromatography (2.4.14).
Identification Test solution. Weigh accurately about 0.1 g of the substance
under examination and dissolve in 10 ml ofthe mobile phase.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution. Weigh accurately ;1bout 0.1 g of topiramate
Compare the spectrum with that obtained with topiramate RS
RS and dissolve in 10.0 ml of the mobile phase.
or with the reference spectrum of topiramate.
B. Melting range (2.4.21). 120° to 130°. Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with
Tests octadecylsilane bonded to porous silica (5 Ilm),
Specific optical rotation (2.4.22). -28.0° to -36.0°, detennined column temperature. 50°
in a 0.4 per cent w/v solution in methanol at 20°. mobile phase: a mixture of 50 volumes of water and 50
volumes of acetonitrile,
Related substances. Detennine by liquid chromatography
flow rate. 0.6 ml per minute,
(2.4.14). .
refractive index detector (Detector cell temperature: 50°)
Test solution. Weigh accurately about 0.4 g of the substance injection volume. 50 Ill.
under examination and dissolve in 10 ml of a mixture of 50
volumes of water and 50 volumes of acetonitrile. Inject the reference solution. The relative standard deviation
for replicate injections is not more than 2.0 per cent.
Reference solution. Weigh accurately about 10 mg each of
(2;3;'4; 5-Bis-O-(l-methylethylidene)-beta-D-fructopyranose Inject the test solution and the reference solution.
RS (topiramate impurity A . RS) and2,3-0-(l- Ca.lculate the coIiteIit ofClzHzlNOsS
methylethylidene)-beta-D-fructopyranose sulphamate RS
(topiramate impurity B RS) and dissolve in sufficient mobile Storage. Store protected from light, at a temperature not
phase to produce 10.0 ml. exceeding 30°.

2242

www.webofpharma.com
IP 2010 TOPOTECAN HYDROCHLORIDE

Topiramate Tablets Medium. 500 ml of water,


Speed and time. 50 rpm and 45 minutes.
Topiramate Tablets contain not less than 90.0 per cent and
Withdraw a suitable volume ofthe medium and filter.
not more than 110.0 per cent of the stated amount of
topiramate, C12H21NOsS. Determine by liquid chromatography (2.4.14), using the
chromatographic conditions described in the test for Related
Usual strengths. 25 mg; 50 mg.
substances.
Identification Test solution. Dilute the filtrate, if necessary, with the
dissolution medium.
A. Weigh a quantity ofthe powdered tablets containing 1 g of
Topiramate, disperse in 30 ml of dichloromethane, centrifuge Reference solution. Dissolve an accurately weighed quantity
at 2000 rpm for 10 minutes and filter. Evaporate the filtrate to of topiramate RS in the dissolution medium and dilute with
dryness. The residue complies with the following test. the dissolution medium to obtain a solution having a lchown
concentration similar to the expected concentration of the
Detennine by infrared absorption spectrophotometry (2.4.6).
test solution.
Compare the spectrum with that obtained with topiramate RS
or with the reference spectrum oftopiramate. D. Not less than 70 per cent of the stated amount of
B. In the Assay, the principal peak in the chromatogram C12H21NOsS.
obtained with the test solution corresponds to the peak in the Other tests. Comply with the tests stated under Tablets.
chromatogram obtained with the reference solution.
Assay. Determine by liquid chromatography (2.4.14) as
Tests described in the test for Related substances.

Related substances. Determine by liquid chromatography Testsolution. Weigh and powder 20 tablets. Weigh accurately
(2.4.14). . . a quantity of the powder containing. about! OOmg of
Topiramate, disperse in 100.0 ml with the mobile phase and
Test solution. To a quantity ofthe powdered tablets containing centrifuge. Use the supernatant liquid.
100 mg ofTopiramate, add sufficient mobile phase to produce
100 ml, mix and centrifuge. Use the supernatant liquid. Reference solution. A 0.1 per cent w/v solution of topiramate
RS in the mobile phase.
Reference solution. A 0.1 per cent w/v solution of topiramine
RS in the mobile phase. Inject the test solution and the reference solution.

Chromatographic system Calculate the content OfC12H21NOsS in the tablets.


- a stainless steel column 25 cm x 4.6 mm packed with Storage. Store protected from moisture, at a temperature not
octadecylsilane bonded to porous silica (5 Ilm) (Such exceeding 30°.
as Inertsil ODS 3),
- column temperature. 50°,
- mobile phase: a mixture of 50 volumes of acetonitrile
and 50 volumes of water, . Topotecan Hydrochloride
- flow rate. 1 ml perminute,
- refractive index detector (Detector cell temperature 50°),
- injection volume. 20 Ill.
Inject the reference solution. The relative standard deviation
for the replicate injections is not more than 2.0 per cent. The HO
retention time oftopiramate is about 5 minutes. The test is not
valid unless the column efficiency is not less than 3000 , Hel
theoretical plates and the tailing factor is notmore than 2.0
Inject the test solution. Determine the amount of related
substances by the area normalization method. The content of o
any individual impurity is not more than 1.0 per cent and the
sum of all impurities is not more than 2.0 per cent. C23H23N30s.HCI Mol.Wt. 457.9
Topotecan Hydrochloride is (48)-10-[(dimethylamino) methyl]-
Dissolution (2.5.2).
4-ethyl-4,9-dihydroxy-lH-pyrano[3' ,4' :6,7]indolizino[1 ,2-
Apparatus No. 1, b]quinoline-3,14(4H, 12H)-dione hydrochloride.

2243

www.webofpharma.com
TOPOTECAN HYDROCHLORIDE IP 2010

Topotecan Hydrochloride contains not less than 98.0 per cent camptothecin is about 1.87 and 2.62 with respect to topotecan
and not more than 102.0 per cent ofC23H23N30s,HCl, calculated hydrochloride and the relative response factors for 10-hydroxy
on the anhydrous basis. camptothecin and camptothecin are about 0.79 and 0.85 with
respect to topotecan hydrochloride.
Category. Anticancer.
Description. A light yellow to greenish yellow powder. Inject the test solution and reference solution (b). In the
chromatogram obtained with the t~st solution, the area of any
CAUTION - Topotecan Hydrochloride is cytotoxic; extra secondary peak is not more than 0.15 times the area of the
care is required to prevent inhaling particles and exposing peak in the chromatogram obtained with reference solution
the skin to it. (b) (0.15 per cent) and the sum ofthe areas ofall the secondary
peaks is not more than the area ofthe peak in the chromatogram
Identification
obtained with reference solution (b) (1.0 per cent).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Heavy metals (2.3.13).1.0 g complies with the limit test for
Compare the spectrum with that obtained with topotecan heavy metals, Method B (20 ppm).
hydrochloride RS or with the reference spectrum oftopotecan
hydrochloride. Sulphated ash (2.3.18). Not more than 0.1 per cent.

B. In the Assay, the principal peak in the chromatogram Water (2.3.43). Not more than 13.0 per cent, detennined on 0.1 g.
obtained with the test solution corresponds to the peak in the Bacterial endotoxins (2.2.3). Not more than 16 Endotoxin Units
chromatogram obtained with the reference solution. per mg of topotecan hydrochloride.
Tests Microbial contamination (2.2.9). Total viable aerobic count,
not more than 100 cfu per g. It also meets the requirements of
pH (2.4.24).3.5 to 4.5, determined in a 1.0 per cent w/v solution. the tests for the absence of Staphylococcus aureus,
Specific optical rotation (2.4.22). + 30.0° to + 38.0°, detennined Pseudomonas aeruginosa, Salmonella species, and
in a 1.0 per cent w/v solution in methanol. Escherichia coli.
Total chloride. 7.6 per cent to 8.1 per cent. Assay. Determine by liquid chromatography (2.4.14).
Weigh accurately about 0.5 g, dissolve in 10 ml of methanol, Solvent mixtur~. A mixture of30 volumes of acetonitrile and
add 20 ml of water and 20 ml of glacial acetic acid. Titrate 70 volumes of a buffer solution, prepared by diluting 1 ml of
with 0.1 M silver nitrate solution using eosin yellow solution trifluoroacetic acid to 1000 ml with water.
as indicator. Colour changes from orange to dark pink.
Test solution. Dissolve 10 mg of the substance under
1 ml of 0.1 Msilver nitrate is equivalent to 0.003545 g ofCI. examination in 25.0 ml ofthe solvent mixture.
Related substances. Determine by liquid chromatography Referencesolution. A 0.04 per cent w/v solution of topatecan
(2.4.14). hydrochloride RS in the solvent mixture.
Solvent mixture. A mixture of 30 volumes of acetonitrile and Chromatographic system
70 volumes of a buffer solution prepared by diluting 1 ml of - a stainless steel column 25 cm x 4.6 mm, packed with
trifluoroacetic acid in 1000 ml of water. octadecylsilane bonded to porous silica (5 /lm),
Test solution. Dissolve 40 mg of the substance under mobile phase: A. a solution of 1 ml of trifluoroacetic
examination in 100 ml ofthe solvent mixture. acid in 1000 ml of water,
B. acetonitrile,
Reference solution (a). A 0.04 per cent w/v solution of
flow rate. 1.2 ml per minute,
topotecan hydrochloride RS in the solvent mixture.
a linear gradient programme using the conditions given
Reference solution (b). Dilute 1 ml of the reference solution below,
(a) to 100 ml with the solvent mixture. spectrophotometer set at 260 urn,
Reference solution (c). A solution containing 0.006 per cent injection volume. 10 Ill.
w/v each of 10-hydroxy comptothecin RS and camptothecin Time Mobile phase A Mobile phase B
RS inN,N'-dimethylformamide. Add 1.0 ml ofthis solution to (in min.) (per cent v/v) (per cent v/v)
a 0.04 per cent w/v solution of topotecan hydrochloride RS o 85 15
-in the solvent mixture. 28 70 30
Use the chromatographic system described under Assay. 38 70 30
Inject reference solution (c). The test is not valid unless the 43 85 15
relative retention time for the 10-hydroxy camptothecin and 48 85 15

2244

www.webofpharma.com
IP 2010 TRAMADOL HYDROCHLORIDE

Inject the reference solution. The test is not valid unless the - mobile phase: a mixture of 78 volumes of water,
relative standard deviation for replicate injections is not more 22 volumes of acetonitrile and 1 volume of
than 2.0 per cent. 1 M hydrochloric acid,
- flow rate. 0.7 ml per minute,
Inject the test solution and the reference solution.
- spectrophotometer set at 254 nm,
Calculate the content ofC23H23N30s,HCI. - injection volume. 20 Ill.
Storage. Store protected from light, at a temperature between Inject the reference solution. The test is not valid unless the
2° to 8°. relative standard deviation is not more than 2.0 per cent.
Inject the test solution and the reference solution.
Calculate the content ofC23H23N30s.
Topotecan Injection Storage. Store protected from light, at a temperature not
Topotecan Hydrochloride Injection exceeding 2° to 8°.

Topotecan Injection is a sterile, stabilised solution of


Topotecan Hydrochloride in Water for Injection.
Topotecan Injection contains not less than 90.0 per cent and Tramadol Hydrochloride
not more than 110.0 per cent of the stated amount of the
topotecan, C23H23N30S.
Usual strength. 1 mg per mi.
Description. A clear, light yellow solution.
• Hel
Identification
A. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution.
B. It gives the reaction of chlorides (2.3.1). Mol. Wt. 299.8
Tests Tramadol Hyrochloride is (lRS,2RS)-2-[(dimethylamino)-
methyl]-I-(3-methoxyphenyl)cyclohexanol hydrochloride.
pH (2.4.24). 2.5 to 3.5.
Other tests. Complies with the tests stated under Parenteral Tramadol Hydrochloride contains not less than 99.0 per cent
Preparations (Injections). and not more than 101.0 per cent of C16H26ClN02, calculated
on the anhydrous basis.
Bacterial endotoxins (2.2.3). Not more than 64.9 Endotoxin
Unit per mg of topotecan. Category. Analgesic.
Sterility (2.2.11). Complies with the test for sterility. Description. A white or almost white, crystalline powder.
Assay. Determine by liquid chromatography (2.4.14). Identification
Test solution. Accurately measure the volume of injection
Test B may be omitted if tests A and C are carried out. Test A
containing 2 mg ofTopotecan, dilute to 50.0 ml with mobile
may be omitted if tests Band C are carried out.
phase.
Reference solution. Dissolve 10 mg of topotecan A. Determine by infrared absorption spectrophotometry (2.4.6).
hydrochloride RS in 5 ml of water and dilute to 25.0 ml with Compare the spectrum with that obtained with tramadol
mobile phase. Dilute 5.0 ml of the solution to 50.0 ml with hydrochloride RS or with the reference spectrum oftramadoI
mobile phase. hydrochloride.

Chromatographic system B. In the test for impurity E, the principal spot in the
- a stainless steel column 25 cm x 4.6 mm, packed with chromatogram obtained with test solution (b) corresponds to
octadecylsilane bonded to poro.us silica the principal spot in the chromatogram obtained with reference
(5 11m), solution (a).

2245

www.webofpharma.com
TRAMADOL HYDROCHLORIDE IP 2010

C. Gives reaction (a) ofchlorides (2.3.1). Reference solution (b). Dissolve 5 mg of tramadol impurity A
RS in 4.0 m1 ofthe test solution and dilute to 100.0 ml with the
Tests mobile phase.
Appearance of solution. A 5 per cent w/v solutionis clear Chromatographic system
(2.4.1) and colourless (2.4.1). - a stainless steel column 25 cm ?I- 4.0 rmn packed with
Acidity. To 10 ml of 5 per cent w/v solution, add 0.2 ml of endcapped octylsilane bonded to porous silica (5 J.tm),
methyl red solution and 0.2 ml of 0.01 M hydrochloric acid. - mobile phase: a mixture of29.5 volumes of acetonitrile
The solution is red. Not more than O.4ml of 0.01 M sodium and 70.5 volumes ofa mixture of0.2 m1 of trifluoroacetic
hydroxide is required to change the colour of the indicator to acid and 100 ml of water,
yellow. - flow rate. 1 m1 per minute,
- spectrophotometer set at 270 nm,
Specific optical rotation (2.4.22). - 0.1 00 to + 0.1 00 , determined
- injection volume. 20 J.tl.
on 5.0 per cent w/v solution.
Inject reference solution (b). The test is not valid unless the
Impurity E. Determine by thin-layer chromatography (2.4.17),
resolution between the peaks due to tramadol impurity A and
coating the plate with silica gel GF254.
tramadol is not less than 2.0. The relative retention time with
Mobile phase. A mixture of 1 volume of concentrated reference to tramadol fortramadol impurity Ais about 0.85.
ammonia, 19 volumes of 2.,.propanol and 80 volu!iles \If
IyUect the test solution and reference solution (a). Run the
toluene.
chromatogram 4 times the retention time ofthe principal peak.
Test solution (a). Dissolve 0.1 g of the substances under In the chromatogram obtained with the test solution, the area
examination in 2 ml of methanol. of the peak due to tramadol impurity A is not more than the
Test solution (b). Dilute 1 ml oftest solution (a) to 10 ml with area ofthe principal peak in the chromatogram obtained with
methanol. reference solution (a) (0.2 per cent), the area of any other
secondary peak is not more than 0.5 times the area of the
Reference solution (a). A 0.5 per cent w/v solution of
principal peak in the chromatogram obtained with reference
tramadol hydrochloride RS in methanol.
solution (a) (0.1 per cent). The sum of all the secondary peaks
Reference solution (b). A 0.1 per cent w/v solution of (2RS)- is not more than twice the area of the principal peak in the
2-[(dimethylamino)methyl]cyclohexanone RS (tramadol chromatogram obtained with reference solution (a) (0.4 per
impurity E RS) in methanol. Dilute 1 ml ofthis solution to 10 cent). Ignore any peak with an area less than 0.1 times the area
ml with methanol. of the principal peak in the chromatogram obtaiiled with
Reference solution (c). A 0.5 per cent w/v solution of reference solution (a) (0.02 per cent).
(1 RS,2SR)-2-[(dimethylamino)methyl]-1-(3-methoxyphenyl) Heavy metals (2.3.13). 2.0 g complies with the limit test for
cyclohexanol RS (tramadol impurity A RS) in reference heavy metals, Method D (20 ppm).
solution (a). Sulphated ash (2.3.18). Not more than 0.1 percent.
Saturate the plate for 20 minutes with concentrated ammonia. Water (2.3.43). Not more than 0.5 per cent, determined on 1.0 g.
Apply to the plate 10 J.tl of each solution. Allow the mobile
phase to rise 15 cm. Dry the plate in air, expose the plate to Assay. Dissolve 0.18 gin 25 ml of anhydrous acetic acid and
iodine vapour for 1 hour, and examine in ultraviolet light at 254 add 10 ml of acetic anhydride. Titrate with 0.1 M perchloric
mn. The chromatogram obtained with reference solution (c) acid, determining the end-point potentiometrically (2.4.25).
shows 2 clearly separated spots. In the chromatogram obtained Carry out a blank titration.
with test solution (a), any secondary spot corresponding to 1 ml of 0.1 M perchloric acid is equivalent to 0.02998 g of
tramadol impurity E is not more intense than the spot in the C16Hz6ClNOz.
chromatogram obtained with reference solution (b) (0.2 per Storage. Store protected from light.
cent).
Related substances. Determine by liquid chromatography
(2.4.14).
Tramadol Capsules
Test solution. Dissolve 0.15 g of the substance under
examination in 1OO.Omlofthe mobile phase. Tramadol Hydrochloride Capsules
Reference solution (a). Dilute 2.0ml of the test solution to Tramadol Capsules contain not less than 95.0 per cent and
10.0 m1 with the mobile phase. Dilute 1.0 m1 ofthis solution to not more than 105.0 per cent ofthe stated amount oftramado1
100.0 ml with the mobile phase. hydrochloride, CI6HzsNOz,HCl.

2246

www.webofpharma.com
IP 2010 TRANDOLAPRlL

Identification Inject reference solution (c). The test is not valid unless the
resolution between the peaks due to tramadol impurity A and
A. Detennine by thin-layer chromatography (2.4.17), coating
tramadol hydrochloride is not less than 3.0.
the plate with silica gel GF254.
Inject the test solution, reference solution (a) and (b). Run the
Mobile phase. A mixture of 1 volume of anhydrous formic chromatogram four times the retention time of the principal
acid, 50 volumes of acetone and 50 volumes of methanol. peak. In the chromatogram obtained with the test solution the
Test solution. Disperse the contents of capsules containing area of peak corresponding to tramadol impurity Ais not more
about 50 mg ofTramadol Hydrochloride in 25 ml of methanol than the area of the principal peak in the chromatogram
and filter through a glass fiber filter (Such as Whatmann OF/ obtained with reference solution (b) (0.3 per cent), the area of
A). secondary peak other than tramadol impurity A is not more
than the area of the principal peak in the chromatogram
Reference solution. A 0.2 per cent w/v solution of tramadol
obtained with reference solution (a) (0.2 per cent) and the sum
hydrochloride RS in methanol.
of the areas of all other secondary peaks is not more than 5
Apply to the plate 10 III of each solution. After development, times the area of the principal peak in the chromatogram
dry the plate in air and expose to iodine vapour until spots .obtained with reference solution (a) (1.0 per cent).
appear and examine in daylight. The principal spot in the
Other tests. Comply with the tests stated under Capsules.
chromatogram obtained with the test solution corresponds to
spot obtained with the reference solution. Assay. Determine by liquid chromatography (2.4.14).
B. In the Assay, the principal peak in the chromatogram Test solution. Dissolve a quantity of the mixed contents of20
obtained with the test solution corresponds to the peak in the capsules containing about 50 mg ofTramadol Hydrochloride
chromatogram obtained with the reference solution. in 150 ml of the mobile phase with the aid of ultrasound and
dilute to 200 ml with the mobile phase, filter.
Tests Reference solution (a). A 0.025 per cent w/v solution of
Related substances. Determine by liquid chromatography tramadol hydrochloride RS in the mobile phase.
(2.4.14). Reference solution (b). A solution containing 0.0015 per cent
w/v each of tramadol hydrochloride RS and tramadol
Test solution. Disperse the contents of capsules containing
impurity A RS in the mobile phase.
about 0.5 g ofTramadol Hydrochloride in 80 ml ofthe mobile
phase with the aid ofultrasound and dilute to 100 ml with the Use the chromatographic system as described under Related
mobile phase, filter through a glass fiber filter (Such as substances.
Whatmann OF/A).
Inject reference solution (b). The test is not valid unless the
Reference solution (a). Dilute 2 ml ofthe test solution to 100 resolution between the peaks due to tramadol impurity A and
ml with the mobile phase. Dilute 1 ml ofthis solution to 10 ml tramadol hydrochloride is not less than 3.0.
with the mobile phase. Inject the test solution and reference solution (a).
Reference solution (b). A 0.0015 per cent w/v solution of Calculate the content ofC,6H2sN02,HCl in the Capsules.
(1RS, 2SR)- 2-[(dimethylamino)methyl]-1-(3 -methoxyphenyl)
cyclohexanol RS ( tramadol impurity A RS) in the mobile
phase.
Reference solution (c). A solution containing 0.0015 per cent
Trandolapril
w/v each of tramadol hydrochloride RS and tramadol
impurity A RS in the mobile phase. .
Chromatographic system
- a stainless steel column 25 cm x 4.6 mrn packed with
octadecylsilane bonded to porous silica (5 Ilm),
- mobile phase: a mixture of I volume of trifluoroacetic
acid, 30 volumes of acetonitrile and 69 volumes of
water, C24H34N20s Mol. Wt. 430.5
- flow rate. 1ml per minute, Trandolapril is (2S,3aR,7as)-1 [(5)-2-[[(8)-1-(ethoxycarbonyl)-
- spectrophotometer set at 271 11ill, 3-phenylpropyl]amino]-1-oxo-propyl]octahydro-1H-indole-2-
injection volume. 20 Ill. carboxylic acid.

2247

www.webofpharma.com
TRANDOLAPRlL IP 2010

Trandolapril contains not less than 98.5 per cent and not more Loss on drying (2.4.19). Not more than 0.5 per cent, determined
than 101.5 per cent ofC24H34N20s, calculated on the dried basis. on 1.0 g by drying at 50° for 3 hours under vacuum.
Category. Antihypertensive. Assay. Determine by liquid chromatography (2.4.14).
Description. A white to offwhite crystalline powder. Test solution. Dissolve 10 mg of the substance under
examination in 100.0 ml ofthe mobile phase.
Identification Reference solution. A 0.01 per cent w/v solution of
A. Determine by infrared absorption spectrophotometry (2.4.6). trandolapril RS in the mobile phase.
Compare the spectrum with that obtained with trandolapril Chromatographic system
RS or with the reference spectrum oftrandolapril. a stainless steel column 15 cm x 4.6 mm, packed with
B. In the Assay, the principal peak in the chromatogram octadecylsilane bonded to porous silica (5 !-lm) (Such
obtained with the test solution corresponds to the peak in the as Inertsil CI8),
chromatogram obtained with the reference solution. mobile phase: a mixture of52 volumes ofbuffer solution
prepared by dissolving 6.71 g of monobasic potassium
Tests orthophosphate in 1000 ml of water, 43 volumes of
acetonitrile and 5 volumes of methanol, adjusted to
Specific optical rotation (2.4.22). -16.so to -18.5°, determined pH 4.0 with orthophosphoric acid,
on 2.0 per cent w/v solution in ethanol (95 per cent). - flow rate. 1 ml per minute,
Related substances. Determine by liquid chromatography spectrophotometer set at 210 nm,
(2.4.14). injection volume. 20 !-ll.
Test solution. Dissolve 100 mg of the substance under Inject the reference solution. The test is not valid unless the
examination in 100 ml ofthe mobile phase. tailing factor is not more than 2.0 and the relative standard
Reference solution. A 0.0005 per cent w/v solution of deviation for replicate injections is not more than 2.0 per cent.
trandolapril RS in the mobile phase. Inject the test solution and the reference solution.
Chromatographic system Calculate the content ofCz4H34NzOs.
- a stainless steel column 25 cm x 4.6 mID, packed with
Storage. Store protected from light and moisture.
octylsilane bonded to porous silica (5 !-lm),
- mobile phase: a mixture of60 volumes ofbuffer solution
prepared by dissolving 6.71 g of monobasic potassium
orthophosphate in 1000 ml of water, 35 volumes of Trandolapril Tablets
acetonitrile and 5 volumes of methanol, adjusted to Trandolapril Tablets contain not less than 90.0 per cent and
pH 4.0 with orthophosphoric acid, not more than 110.0 per cent of the stated amounts of
flow rate. I ml per minute, trandolapril, C24H34N20s.
- spectrophotometer set at 210 nm, Usual strengths. 1 mg; 2 mg.
injection volume. 20 !-ll.
Identification
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicates injections is not more A. In the Assay, the principal peak in the chromatogram
than 5.0 per cent. obtained with the test solution corresponds to the peak in the
Inject the reference solution and the test solution. In the chromatogram obtained with the reference solution.
chromatogram obtained with the test solution, the area of any B. When examined in the range of200 nm to 400 nm (2.4.7), a
secondary peak is not more than the area ofthe principal peak 0.05 per cent w/v solution in 0.1 M hydrochloric acid shows
in the chromatogram obtained with the reference solution (0.5 an absorption maximum as obtained with trandolapril RS of
per cent) and the sum of the areas of all the secondary peaks the same concentration.
is not more than twice the area of the principal peak in the
chromatogram obtained with the reference solution (1.0 per Tests

Heavy metals (2.3.13).1.0 g complies with the limit test for


Apparatus No.1,
heavy metals, Method B(20 ppm).
Medium. 500 ml of water,
Sulphated ash (2.3.18). Not more than 0.1 percent. Speed and time. 50 rpm and 45 minutes.

2248

www.webofpharma.com
IP 2010 TRANDOLAPRIL TABLETS

Withdraw a suitable volume ofthe medium and filter. the chromatogram obtained with the reference solution (2.0
Determine by liquid chromatography (2.4.14). per cent).

Test solution. Dilute the filtrate, if necessary, with the Uniformity of content. Comply with the test stated under
dissolution medium. Tablets.

Reference solution. Dissolve an accurately weighed quantity Detennine by liquid chromatography (2.4.14), as described
of trandolapril RS in the mobile phase and dilute with under Assay with the following solutions.
dissolution medium to obtain a solution having a known Test solution. Disperse one tablet in the mobile phase, dilute
concentration similar to the test solution. to obtain a solution containing 0.005 per cent w/v oftrandolapril
Use chromatographic system as described under Assay using in the mobile phase and filter.
100 III injection volume.
Reference solution. Dissolve an accurately weighed quantity
Inject the reference solution. The test is not valid unless the of trandolapril RS in the mobile phase and dilute to obtain a
relative standard deviation for replicate injections is not more solution having a known concentration similar to the test
than 2.0 per cent. solution.
Inject the test solution and the reference solution. Calculate the content of CZ4H34NzOs in the tablet.
Calculate the content of CZ4H34NzOs. Water (2.3.43). Not more than 7.0 per cent, determined on
D. Not less than 80 per cent of the stated amount of 0.5 g.
CZ4H34NzOs. Other tests. Comply with the tests stated under Tablets.
Related substances. Determine by liquid chromatography Assay. Determine by liquid chromatography (2.4.14).
(2.4.14).
Test solution. Weigh and disperse 10 intact tablets in 100.0 ml
NOTE-Use fi-eshly prepared solutions. with the mobile phase, filter. Dilute 5.0 ml of this solution to
Test solution. Disperse a quantity of powdered tablets 10.0 ml with the mobile phase.
containing about 10 mg ofTrandolapril in 10.0 ml ofthe mobile
Reference solution. A 0.005 per cent w/v solution of
phase.
trandolapril RS in the mobile phase.
Reference solution. A 0.001 per cent w/v solution of
Chromatographic system
trandolapril RS in the mobile phase.
a stainless steel column 15 cm x 4.6 mm, packed with
Chromatographic system octadecylsilane bonded to porous silica (5 11m) (Such
a stainless steel column 25 cm x 4.6 mm, packed with as Inertsil ODS~3),
octylsilane bonded to porous silica (5 11m) (Such as mobile phase: a mixture of52 volumes ofbuffer solution
ACEC8), prepared by dissolving 6.71 g of monobasic potassium
mobile phase: a mixture of60 volumes ofbuffer solution orthophosphate anhydrous in 1000 ml of water, 43
prepared by dissolving 6.71 g of monobasic potassium volumes of acetonitrile and 5 volumes of methanol,
orthophosphate anhydrous in 1000 ml of wate]; 35 adjusted to pH 4.0 with orthophosphoric acid,
volumes of acetonitrile and 5 volumes of methanol, flow rate. 1.2 ml per minute,
adjusted to pH 4.0 with orthophosphoric acid,
spectrophotometer set at 210 nm,
- flow rate. 1 ml per minute,
injection volume. 20 Ill.
spectrophotometer set at 210 nm,
injection volume. 20 Ill. Inject the reference solution. The test is not valid unless the
theoretical plates of the principal peak is not less than 2000,
Inject the reference solution. The relative standard deviation
the tailing factor is not more than 2.0 and the relative standard
for replicate injections is not more than 5.0 per cent. .
deviation for replicate injections is not more than 2.0 per cent.
Inject the test solution and the reference solution. Run the
chromatogram 5 times the retention time ofthe principal peak. Inject the test solution and the reference solution.
In the chromatogram obtained with the test solution the area Calculate the content of CZ4H34NzOs in the tablet.
ofany secondary peak is not more than the area ofthe principal
peak in the chromatogram obtained with the reference solution Storage. Store at a temperature not exceeding 30°.
(1.0 per cent) and the sum of the areas of all the secondary Labelling. The label states the strength in terms ofthe amount
peaks is not more than twice the area ofthe principal peak in of Trandolapril.

2249

www.webofpharma.com
TRAVOPROST IP 2010

Travoprost secondary peak is not more than the area ofthe principal peak
in the chromatogram obtained with the reference solution
(0.5 per cellt). The sum ofthe areas of all the secondary peaks
is not more than 12 times the area ofthe principal peak in the
chromatogram obtained with the reference solution (6.0 per
cent). "
Water (2.3.43). Not more than LO per cent, determined on
0.1 g.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve 100 mg of the substance under
Mol. Wt. 500.6
examination in acetonitrile and dilute to 100.0 m1 with water.
Travoprost is (52)-7-[(lR,2R,3R,5S)- 3,5-dihydroxy-2-[(lE,3R)-
Reference solution. Dissolve 10 mg of travoprost RS in
3-hydroxy-4-[3-(trifluromethyl)phenoxy]-1-buteny1]-
acetonitrile and dilute to 10.0 m1 with watel:
cyclopentyl]-5-heptenoic acid 1-methylethyl ester.
Chromatographic system
Travoprost contains not less than 96.0 per cent and not more a stainless steel column 25 cm x 4.6 mm, packed with
than 102.0 per cent ofC26H3sF306, calculated on the anhydrous octadecylsilane bonded to porous silica (5 Ilm), (Such
basis. as Hypersil ODS),
Category. Antiglaucoma. - column temperature. 50°,
Description. A colourless to yellowish oil.· mobile phase: A. a mixture of 66 volumes of buffer
solution prepared by diluting 1 ml of orthophosphoric
Identification acidto 1000 m1 with water, adjusted to pH 5.0 with 1 M
sodium hydroxide, 30 volumes of acetonitrile and 4
A. Determine by infrared absorption spectrophotometry (2.4.6).
volumes of propanol-2-ol,
Compare the spectrum with that obtained with travoprost RS
B. a mixture of80 volumes of acetonitrile,
or with the reference spectrum of travoprost.
4 volumes ofpropanol-2-o1 and 16 volumes ofwatel;
B. In the Assay, the principal peak in the chromatogram a linear gradient programme using the conditions given
obtained with the test solution corresponds to the peak in the below,
chromatogram obtained with the reference solution. flow rate. 1 ml per minute,
spectrophotometer set at 220 nm,
Tests injection volume. 50 Ill.
Appearance of solution. A 1.0 per cent w/v solution in ethanol Time Mobile phase A Mobile phase B
(95 per cent) is not more intensely coloured than reference (in min) (per cent v/v) (per cent V N)
solution YS7 (2.4.1). o 100 o
Specific optical rotation (2.4.22). +52° to +58°, determined on 5-50 80 20
2.0 per cent. w/v solution in ethanol (95 per cent) at20°. 50-70 80--70 20--7100
Related substances. Not more than 3.5 per cent of5,6-trans- . 70-80 0 100
travoprost. 80-82 0--7100 100--70
Determine by liquid chromatography (2.4.14). 82-90 100 o
Test solution. Dissolve 100 mg of the substance under Inject the reference solution. The test is not valid unless the
examination in acetonitrile and dilute to 100.0 ml with water. resolution between the peaks due to travoprost and 5-trans-
Reference solution. Dissolve 10 mg of travoprost RS in travoprost is not less than 1.5, the theoretical plates is not
acetonitrile and dilute to 100.0 rn1 with watel: Dilute 1.0 ml of less than 1500, the tailing factor is not more than 2.0 and the
the solution to 20.0 ml with watel: relative standard deviation for replicate injections is not more
Use chromatographic system as described under Assay. than 2.0 per cent.

Inject thereferences6Iiiti6n. The reliitiveretenli6iifiilleWith Inject the test solution and the reference solution.
reference t6 travopr6st f6r 5;6;trans:trav6pmstis ab6ut L06: Calculate the content ofC26H3sF306"
Inject the test solution and the reference solution. In the Storage. Store protected from moisture, at a temperature
chromatogram obtained with the test solution, the area of any between 2° and 8°.

2250

www.webofpharma.com
IP 2010 TRAVOPROST EYE DROPS

Travoprost Eye drops peak in the chromatogram obtained with the reference solution
(4.0 per cent) and the sum of the areas of all the secondary
Travoprost Eye Drops contains not less than 90.0 per cent peaks is not more than 6 times the area ofthe principal peak in
and not more than 110.0 per cent of the stated amount of the chromatogram obtained with the reference solution (6.0
travoprost, C26H3sF306. per cent).

Identification Other tests. Complies with the tests stated under Eye
Drops.
A. In the Assay, the principal peak in the chromatogram
Assay. Determine by liquid chromatography (2.4.14).
obtained with the test solution corresponds to the principal
peak inthe chromatogram obtained with the reference solution. Solvent mixture. Equal volumes of water and acetonitrile.
B. Determine by thin-layer chromatography (2.4.17), coating Test solution. Dilute a suitable volume of eye drops containing
the plate with silica gel G. 0.004 per cent w/v oftravoprost in the solvent mixture.
Mobile phase. A mixture of 50 volumes of benzene and 40 Reference solution. Dissolve 20 mg of travoprost RS with
volumes of dioxane. 10.0 ml of acetonitrile, sonicate and dilute to 20.0 ml with
Test solution. Extract eye drops containing about 0.8 mg of water. Dilute 1.0 ml ofthis solution to 25.0 ml with water.
Travoprost with 20.0 ml of chloroform in a separating funnel.
Chromatographic system
Collect the chloroform layer in a 250 ml beaker. Give three
- a stainless steel column 25 cm x 4.6 mm, packed with
washings with chloroform. Evaporate the chloroform layer to
octadecy1silane bonded to porous silica (5 !J.m) (Such
dryness and add 3.0 ml of methanol.
as Hypersil ODS),
Reference solution. Dissolve 20 mg of travoprost RS in 10 ml column temperature. 50°,
of acetonitrile, sonicate and dilute to 20.0 ml with water. Dilute sample temperature. 5°,
5.0 ml ofthis solution to 20.0 ml with methanol. mobile phase: A. a mixture oBO volumes of acetonitrile,
Apply to the plate 20 !J.l of each solution. Allow the mobile 4 volumes of propan-2-o1 and 66 volumes of buffer
phase to rise 8 cm. Dry the plate and spray with 10 per cent solution prepared by diluting 1.0 ml of orthophosphoric
w/v solution of phosphomolybdic acid in ethanol, heat the acid to 1000.0 ml with water, adjusted to pH 5.0 with
plate at 1l0° for 15 minutes and examine immediately. The 1 M sodium hydroxide and filter,
principal spot in the chromatogram obtained with the test B. a mixture of80 volmnes of acetonitrile,
solution corresponds to the spot obtained with the reference 4 volumes ofpropan-2-o1 and 16 volumes of water,
solution. flow rate. Iml per minute,
spectrophotometer set at 220 nm,
Tests injection volume. 100 !J.l.
Time Mobile phase A Mobile phase B
pH (2.4.24). 5.7 to 6.3.
(in mill) (per cent v/v) (per cent v/v)
Related substances. Determine by liquid chromatography 0- 5 100-780 0-720
(2.4.14).
5-50 80 20
Test solution. Dilute the eye drops to obtain a solution
50-70 80-70 20-7100
containing 0.004 per cent w/v oftravoprost.
70-80 0 100
Reference solution. Dissolve 20 mg of travoprost RS in 10.0
ml of acetonitrile, sonicate and dilute to 20.0 ml with water. 80 -85 0-7100 100-70
Dilute 1.0 m1 of this solution to 25.0 m1 with water. Further,
Inject the reference solution. The test is not valid unless the
dilute 1.0 ml ofthis solution to 100.0 ml with water.
theoretical plates is not less than 1500, the tailing factor is not
Use chromatographic system as described under Assay. more than 2.0 and the relative standard deviation for replicate
injections is not more than 2.0 per cent.
Inject the reference solution. The relative standard deviation
for replicate injections is not more than 5.0 per cent. Inject the test solution and the reference solution.
Inject the test solution and the reference solution. In the Calculate the content ofC26H3sF306.
chromatogram obtained with the test solution the area of any
.secondary peak is not more than 4 times the area ofthe principal Storage. Store at a temperature not exceeding 30°.

2251

www.webofpharma.com
TRIAMCINOLONE IP 2010

Triamcinolone base-deactivated end-capped octadecylsilane bonded


to porous silica (5 /-lm),
- mobile· phase: a mixture prepared by mixing 525 ml of
methanol with 400 ml of water, allowing to equilibrate,
adjusting the volume to 1000.0 ml with water and mixing
again,
- flow rate. 1 ml per minute,
- spectrophotometer set at 238 nrn,
o - injection volume. 20 /-ll.

Mol. Wt. 394.4 Inject the test solution and the reference solution. Continue
the chromatography for 4.5 times the retention time of
Triamcinolone is 9cx-fluoro-ll {3, 16~ 17 ~21-tetrahydroxy­ triamcinolone (about 11 minutes).
pregna-l,4-diene-3,20-dione.
In the chromatogram obtained with the test solution the area
Triamcinolone contains not less than 97.0 per cent and not of any peak other than the principal peak is not more than the
more than 103.0 per cent ofCz1Hz7F06, calculated on the dried area ofthe principal peak in the chromatogram obtained with
basis. the reference solution (l per cent); not more than two such
Category. Corticosteroid. peaks have an area greater than half the area of the principal
Dose. 2 to 24 mg daily. peak in the chromatogram obtained with the reference solution
(0.5 per cent); the sum of the areas of all the peaks other than
Description. A white or almost white, crystalline powder; the principal peak is not more than twice the area ofthe principal
slightly hygroscopic. peak in the chromatogram obtained with the reference solution
Identification (2 per cent). Ignore any peak with an area less than 0.05 times
the area of the principal peak in the chromatogram obtained
A. Determine by infrared absorption spectrophotometry (2.4.6). with the reference solution (0.05 per cent).
Compare the spectrum with that obtained with triamcinolone
Heavy metals (2.3.13). 0.8 g complies with the limit test for
RS or with the reference spectrum of triamcinolone.
heavy metals, Method B (25 ppm).
B. When examined in the range 220 nm to 360 nrn (2.4.7), a
Sulphated ash (2.3.18). Not more than 0.2 per cent.
0.002 per cent w/v solution in methanol shows an absorption
maximum at about 238 mn; absorbance at about 238 nrn, about Loss on drying (2.4.19). Not more than 2.0 per cent, determined
0.76. on 1.0 g by drying in an oven at 60° at a pressure not exceeding
0.7 kPa for 3 hours.
C. Dissolve 1 mg in 6 ml of ethanol (95 per cent), add 5 ml of
a 1 per cent w/v solution of butylated hydroxytoluene in Assay. Weigh accurately about 25 mg, dissolve in sufficient
ethanol (95 per cent) and 5 ml of 1 M sodium hydroxide and ethanol (95 per cent) to produce 100.0 ml and mix. Dilute
heat on a water-bath under a reflux condenser for 20 minutes; 2.0 ml to 50.0 ml with ethanol (95 per cent) and measure the
a pinkish lavender colour is produced. absorbance ofthe resulting solution at the maximum at about
238 nrn (2.4.7).
Tests
Calculate the content ofCzIHz7F06 taking 380 as the specific
Specific optical rotation (2.4.22). +65.0° to +72.0°, determined absorbance at 238 nm.
in a 1.0 per cent w/v solution in dimethylformamide. Storage. Store protected from light and moisture.
Related substances. Detennine by liquid chromatography
(2.4.14).
Prepare the following solutions immediately before use and Triamcinolone Tablets
protect fi'OIn light.
Triamcinolone Tablets contain not less than 90.0 per cent and
Test solution. Dissolve 25 mg of the substance under not more than 110.0 per cent of the stated amount of
examination in a mixture of equal volumes of methanol and triamcinolone, CZIHz7F06.
water and dilute to 10 ml with the same solvent mixture.
Q§!!ahtrengths.. 2_mg;A_mg;BJJJ.g•.__ . . . _
Reje,;ence solutio'i. DilliteTinCoffhefesfs-ollifion fo~rOO iiil
with a mixture of equal voluIlles of methanol and water. Identification
Chromatographic system A. Dissolve 1 mg ofthe residue obtained in the test for Related
- a stainless steel column 25 cm x 4.6mm, packed with substances in 6 ml of ethanol (95 percent), add 5 ml ofa 1 per

2252

www.webofpharma.com
IP 2010 TRIAMCINOLONE ACETONIDE

cent w/v solution of butylated hydroxytoluene in ethanol Uniformity of content. Comply with the test stated under
(95 per cent) and 5 ml of 1 M sodium hydroxide and heat on Tablets.
a water-bath under a reflux condenser for 20 minutes; a pinkish Crush one tablet to a fine powder, add 100 ml of ethanol
lavender colour is produced. (95 per cent) and shake for 10 minutes. Filter, dilute 2.0 ml of
B. In the Assay, the chromatogram obtained with the principal the filtrate with sufficient ethanol (95 per cent) to produce a
peak obtained with the test solution corresponds to the peak solution containing 0.002 per cent w/v of Triamcinolone and
due to h'iamcinolone in the chromatogram obtained with measure the absorbance of the resulting solution at the
reference solution (a). maximum at about 238 urn (2.4.7).
Calculate the content of CZIHz7F06 in the tablet taking 380 as
Tests the specific absorbance at 238 nm.
Related substances. Detennine by liquid chromatography Other tests. Comply with the tests stated under Tablets.
(2.4.14). Assay. Detennine by liquid chromatography (2.4.14).
Test solution. Shake a quantity of the powdered tablets Test solution. Weigh and powder 20 tablets. Weigh accurately
containing 15 mg of Triamcinolone with 15 ml of ethanol for a quantity ofthe powder containing 2.5 mg ofTriamcinolone,
15 minutes, filter under reduced pressure through a fine filter shake with 5 ml of methanol and 20 ml ofa mixture of5 volumes
paper (such as Whahnan No 42) and evaporate the filtrate to of methanol and 3 volumes of water. Shake for 15 minutes, mix
dryness using a rotary evaporator. Reserve 1 mg ofthe residue with the aid of ulh'asound for 10 minutes, centrifuge and use
for Identification testA. Dissolve the remainder ofthe residue the supernatant liquid.
in 15 ml of methanol. Reference solution (a). Prepare in the same manner as the test
Reference solution (a). Dilute 4 ml of the test solution to solution but add 5 ml of a 0.06 per cent w/v solution of
100 ml with methanol. testosterone (internal standard) in methanol (solution A) in
place of the 5 mlofmethanol.
Reference splution (b). Dilute 5 ml ofthe test solution to 50 ml
Reference solution (b). Add 5 ml of solution A to 5 ml of a
with methanol. Dilute 5 ml of the resulting solution to 50 ml
0.08 per cent w/v solution of triamcinolone RS in methanol
with the same solvent.
and add 15 ml of methanol (50 per cent).
Chromatographic system Chromatographic system
- a stainless steel column 20 cm x 4.6 mm, packed with a stainless steel column 20 cm x 4 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm), octadecylsilane bonded to porous silica (10 Ilm),
mobile phase: a mixture of methanol and water, adjusted mobile phase: a mixture of 70 volumes of methanol,
so that the retention time of triamcinolone is about 30 volumes of water and 0.1 volume of glacial acetic
5 minutes (approximately equal volumes of methanol acid,
and water), flow rate. 2 ml per minute,
- flow rate. 2 ml per minute, spectrophotometer set at 238 urn,
spectrophotometer set at 238 urn, injection volume. 20 Ill.
- injection volume. 20 Ill.
Calculate the content ofCzIHz7F06 in the tablets.
Inject reference solution (b) and record the chromatogram for
Storage. Store protected from moisture.
4 times the retention time of the triamcinolone peak.
The test is not valid unless the column efficiency detennined
from the principal peak in the chromatogram obtained with Triamcinolone Acetonide
reference solution (a) is at least 10,000 theoretical plates per
metre.
In the chromatogram obtained with the test solution the area
of any secondary peak is not more than twice the area of the
principal peak in the chromatogram obtained with reference
solution (b) (2 per cent) and not more than one such peak has
an area more than that ofthe principal peak in the chromatogram o
obtained with reference solution (b) (1 per cent). The sum of
the areas of any such peaks is not more than the area of the CZJf31F06 Mol. Wt. 434.5
principal peak in the chromatogram obtained with reference Triamcinolone Acetonide is 9a- fluoro-I1 ~,21-dihydroxy­
solution (a) (4 per cent). 16a,17a-isopropylidenedioxy-l ,4-pregnadiene-3,20-dione.

2253

www.webofpharma.com
TRIAMCINOLONE ACETONIDE IF 2010

Triamcinolone Acetonide contains not less than 96.0 per cent examination and again heat in a naked flame until white fumes
and not more than 104.0 per cent ofCz4H3IF06, calculated on appear; the solution does not wet the sides of the tube and
the anhydrous basis. does not pour easily from the tube.
Category. Corticosteroid. Tests
Dose. By deep intramuscular injection, 40 mg to 100 mg.
Light absorption (2.4.7). When examined in the range 230 run
Description. A white or almost white, crystalline powder; to 360 mn, a 0.001 per cent w/v solution in ethanol (95 per
odourless or almost odourless. cent) shows an absorption maximum at about 239 nm;
absorbance at about 239 nm, 0.34 to 0.37.
Identification
Specific optical rotation (2,4,22). +100° to +107°, determined
Test A may be omitted if tests Band C are carried out. Test C in a 1.0 per cent w/v solution in dioxan.
may be omitted if tests A and B are carried out.
Related substances. Determine by liquid chromatography
A. Determine by infrared absorption spectrophotometry (2.4.6). (2.4.14).
Compare the spectrum with that obtained with triamcinolone
NOTE - CarlY out the test protectedii"om light.
acetonide RS.
Test solution. Dissolve 25 mg of the substance under
B. Determine by thin-layer chromatography (2.4.17), coating
examination in 7 ml of methanol and dilute to 10 ml with water.
the plate with silica gel G.
Reference solution (a). Dissolve 2 mg of triamcinolone
Solvent mixture. A mixture of 90 volumes of acetone and
acetonide RS and 2 mg of triamcinolone in the mobile phase
10 volumes offormamide.
and dilute to 100 ml with the mobile phase.
Mobile phase. A mixture of 115 volumes of cyclohexane,
Reference solution (b). Dilute 1 ml of the test solution to
56 volumes of chlor(}form and 29 volumes of toluene.
100 ml with the mobile phase.
Test solution. Dissolve 25 mg of the substance under
Chromatographic system
examination in 10 ml ofthe solvent mixture.
a stainless steel column 25 cm x 4.6 mm, packed with
Reference solution (a). Dissolve 25 mg of triamcinolone octadecylsilane bonded to porous silica (5 /-lm),
acetonide RS in 10 ml ofthe solvent mixture. mobile phase: a mixture prepared by mixing 525 ml of
Reference solution (b). Mix equal volumes ofthe test solution methanol with 400 ml of water, allowing to equilibrate,
adjusting the volume to 1000.0 ml with water and mixing
and reference solution (a).
again,
Place the dry plate in a tan1e containing a shallow layer of the flow rate. 1.5 ml per minute,
solvent mixture, allow the solvent mixture to ascend to the spectrophotometer set at 254 nm,
top, remove the plate from the tan1e and allow the solvent to injection volume. 20 /-l1.
evaporate. Use within 2 hours, with the flow of the mobile
Equilibrate the column with the mobile phase for about
phase in the direction in which the aforementioned treatment
was done. 10 minutes.
Inject reference solution (b). Adjust the sensitivity of the
Apply to the plate 5 /-ll of each solution. Allow the mobile
system so that the height of the principal peak is at least
phase to rise 12 cm. Dry the plate in a current ofwarm air, allow
the solvent to evaporate, heat at 120° for 15 minutes and spray 50 per cent ofthe full scale of the recorder.
the hot plate with ethanolic sulphuric acid (20 per cent vlv). Inject reference solution (a). The retention times are;
Heat at 120° for a further 10 minutes, allow to cool and examine triamcinolone, about 5 minutes and triamcinolone acetonide
in daylight and in ultraviolet light at 365 mn. The principal about 17 minutes. The test is not valid unless the resolution
spot in the chromatogram obtained with the test solution between the peaks corresponding to triamcinolone and
corresponds to that in the chromatogram obtained with triamcinolone acetonide is at least 15; if necessary, adjust the
reference solution (a). The principal spot in the chromatogram concentration of methanol in the mobile phase.
obtained with reference solution (b) appears as a single,
Inject the test solution and reference solution (b). Continue
compact spot.
tbe~bromJ:ltogmpby for 3.5 timesJhe retentiontimeofthe
e. Heat 0.5 ml of chromic-sulphuric acid in a test-tube (5 cm x principal peak in the chromatogram obtained with the test
about 6 mm) in a naked flame until white fumes are evolved; solution. In the chromatogram obtained with the test solution
the solution wets the sides of the tube readily and there is no the area of any peak other than the principal peak is not more
greasiness. Add about 2 mg of the substance under than 0.25 times the area of the principal peak in the

2254

www.webofpharma.com
IP 20ID TRIAMCINOLONE ACETONIDE INJECTION

chromatogram obtained with the reference solution (b) Test solution. Dissolve 25 mg of the substance under
(0.25 per cent); the sum ofthe areas ofall the peaks other than examination in 10 ml ofthe solvent mixture.
the principal peak is not greater than half the area of the
Reference solution (a). Dissolve 25 mg of triamcinolone
principal peak in the chromatogram obtained with reference
acetonide RS in 10 ml ofthe solvent mixture.
solution (b) (0.5 per cent). Ignore any peak due to the solvent
and any peak with an area less than 0.05 times the area ofthe Reference solution (b). Mix equal volumes ofthe test solution
principal peak in the chromatogram obtained with reference and reference solution (a).
solution (b).
Place the dry plate in a tanle containing a shallow layer of the
Sulphated ash (2.3.18). Not more than 0.2 percent. solvent mixture, allow the solvent mixture to ascend to the
Water (2.3.43). Not more than 2.0 per cent, detennined on 0.2 g. top, remove the plate from the tank and allow the solvent to
evaporate. Use within 2 hours, with the flow of the mobile
Assay. Weigh accurately about 25 mg and dissolve in sufficient phase in the direction in which the aforementioned treatment
ethanol to produce 100.0 ml and mix. Dilute 5.0 ml to 100.0 ml was done.
with ethanol (95 per cent) and measure the absorbance of
the resulting solution at the maximum at about 239 nm (2.4.7). Apply to the plate 5 III of each solution. Allow the mobile
phase to lise 12 cm. Dry the plate in a current ofwarm air, allow
Calculate the content ofC24H31F06 taking 354 as the specific the solvent to evaporate, heat at 120° for 15 minutes and spray
absorbance at 239 urn. the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Storage. Store protected from light and moisture. Heat at 120° for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with
Triamcinolone Acetonide Injection reference solution (a). The principal spot in the chromatogram
Triamcinolone Acetonide Injection is a sterile suspension of obtained with reference solution (b) appears as a single,
Triamcinolone Acetonide in very fine particles in Water for compact spot.
Injections containing suitable dispersing agents. e. Heat 0.5 ml of chromic-sulphuric acid in a test-tube (5 cm x
Triamcinolone Acetonide Injection contains not less than about 6 mm) in a naked flame until white fumes are evolved;
90.0 per cent and not more than 110.0 per cent of the stated the solution wets the sides of the tube readily and there is no
amount oftriamcinolone acetonide, C24H31F06. greasiness. Add about 2 mg of the substance under
examination and again heat in a naked flame until white fumes
Usual strength. 40 mg per ml. appear; the solution does not wet the sides of the tube and
Identification does not pour easily from the tube.

Extract a volume containing about 50 mg of Triamcinolone


Tests
Acetonide with two quantities, each of 10 ml, ofperoxide-free
ether and discard the ether extracts. Filter the aqueous layer pH (2.4.24).5.0 to 7.5.
through a sintered-glass filter, wash the residue with· four
Other tests. Complies with the tests stated under Parenteral
quantities, each of5 ml ofwater and dry at 105° for 1 hour. The
Preparations (Injections).
residue complies with the following tests.
Test A may be omitted if tests Band C are carried out. Test C Assay. Detennine by liquid chromatography (2.4.14).
may be omitted if tests A and B are carried out. Test solution. Mix an accurately measured volume of the
A. Determine by infrared absorption spectrophotometry (2.4.6). injection, diluted with methanol, with sufficient of a solution
Compare the spectnim with that obtained with triamcinolone ofprednisolone RS (internal standard) with methanol to obtain
acetonide RS or with the reference spectrum oftriamcinolone a final concentration of 0.02 per cent w/v of triamcinolone
acetonide. acetonide and 0.01 per cent w/v of prednisolone, centrifuge
B. Determine by thin-layer chromatography (2.4.17), coating and use the clear supernatant liquid.
the plate with silica gel G. Reference solution (a). A solution containing 0.02 per cent
Solvent mixture. A mixture of 90 volumes of acetone and w/v of triamcinolone acetonide RS and 0.01 per cent w/v of
10 volumes offormamide. prednisolone RS in methanol.
Mobile phase. A mixture of 115 volumes of cyclohexane, Reference solution (b). Prepare in the same manner as test
56 volumes of chloroform and 29 volumes of toluene. solution but omitting the internal standard.

2255

www.webofpharma.com
TRIAMTERENE IP 2010

Chromatographic system sodium hydroxide is required to change the colour of the


- a stainless steel column 25 cm x 4 mm, packed with solution.
octadecylsilane bonded to porous silica (10 Ilm),
Related substances. Determine by thin-layer chromatography
- mobile phase: a mixture of56 volumes of methanol and
(2.4.17), coating the plate with silica gel G
44 volumes of water,
- flow rate. 1.4 ml per minute, Mobile phase. A mixture of 90 volumes of ethyl acetate,
- spectrophotometer set at 254 urn, 10 volumes of 18 M ammonia and 10 volumes of methanol.
- injection volume. 20 Ill. Test solution. Dissolve 0.1 g of the substance under exami-
Adjust the flow rate of the mobile phase such that the nation in 20 ml of dimethyl sulphoxide and dilute 2 ml of the
separation ofthe triamcinolone acetonide and internal standard resulting solution to 50 ml with methanol.
is optimised with a retention time of about 15 minutes for Reference solution. Dilute 1 volume of the test solution to
triamcinolone acetonide. 200 volumes with methanol.
Calculate the content of CZ4H31F06 in the injection. Apply to the plate 5 III of each solution. After development,
Storage. Store protected from light. dry the plate in air until the odour of solvent is no longer
detectable and examine in ultraviolet light at 365 urn. Any
secondary spot in the chromatogram obtained with the test
solution is not more intense than the spot in the chromatogram
Triamterene obtained with the reference solution.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105°.
Assay. Weigh accurately about 0.15 g, dissolve in 5 ml of
anhydrous formic acid, add 100 ml of anhydrous glacial
Mol. Wt. 253.3
acetic acid. Titrate with 0.1 M perchloric acid, determining
the end-point potentiometrically (2.4.25). Carry out a blank
Triamterene is 2,4,7-triamino-6-phenylpteridine. titration.
Triamterene contains not less than 99.0 per cent and not more 1 ml of 0.1 M perchloric acid is equivalent to 0.02533 g of
than 101.0 per cent ofC 1ZH lI N 7, calculated on the dried basis. ClzHIIN7.
Category. Potassium sparing diuretic. Storage. Store protected from light and moisture.
Dose. 150 to 250 mg daily, in divided doses.
Description. A yellow, crystalline powder; odourless.
Triamterene Capsules
Identification Triamterene Capsules contain not less than 95.0 per cent and
A. When examined in the range 250 mn to 380 nm (2.4.6), a not more than 105.0 per cent of the stated amount of
0.001 per cent w/v solution in a mixture of9 volumes of ethanol triamterene, ClzHIIN7'
(95 per cent) and 1 volume of 1 M hydrochloric acid shows Usual strength. 50 mg.
absorption maxima at about 262 urn and 360 urn, and a shoulder
at about 285 nm. Identification
B. A 0.1 per cent w/v solution in anhydrousformic acid, when The final solution obtained in the Assay has a bluish
examined in ultraviolet light at 365 nm shows an intense blue fluorescence and when examined in the range 250 nm to
fluorescence. Solutions in other acids also exhibit a blue 380 nm (2.4.7), shows an absorption maximum at about
fluorescence. 360nm.

Tests Tests

Acidity. Boil 1.0 g with 20 ml of water for 5 minutes, cool, filter


Related substances. Deteiil'iine by thin~layeichromatograpliy
(2.4.17), coating the plate with silica gel G
and wash the filter with three quantities, each of 10 ml, of
water. Combine the filtrate and washings and add 0.3 ml of Mobile phase. A mixture of 90 volumes of ethyl acetate,
phenolphthalein solution. Not more than 1.5 ml of 0.01 M 10 volumes of 18 M ammonia and 10 volumes of methanol.

2256

www.webofpharma.com
IP 2010 TRICHLOROMONOFLUOROMETHANE

Test solution. Dissolve a quantity of the contents of the Tests


capsules containing 0.1 g ofTriamterene in sufficient dimethyl
sulphoxide to produce 20 ml and dilute 2 ml to 50 ml with Weight per ml (2.4.29). 1.037 to 1.045.
methanol. Refractive index (2.4.27). 1.443 to 1.445.
Reference solution. Dilute 1 volume of the test solution to Acidity. Dissolve 32.0 g in 30 ml of ethanol (95 per cent),
200 volumes with methanol. previously neutralised to bromothymol blue, add bromothymol
Apply to the plate 5 III of each solution. After development, blue solution and titrate with 0.1 M sodium hydroxide to a
dry the plate in air until the odour of solvent is no longer faint blue endpoint; not more than 1.0 ml is required.
detectable and examine in ultraviolet light at 365 nm. Any Water (2.3.43). Not more than 0.2 percent.
secondary spot in the chromatogram obtained with the test
solution is not more intense than the spot in the chromatogram Heavy metals (2.3.13). 2.0 g complies with the limit test for
obtained with the reference solution. heavy metals, Method B (10 ppm).

Other tests. Comply with the tests stated under Capsules. Assay. Determine by gas chromatography (2.4.13).
Assay. Weigh accurately a quantity of the contents of Test solution. Dissolve about 300 mg of substance under
20 capsules containing about 0.1 g ofTriamterene, dissolve in examination in 10 ml of toluene.
50 ml ofa mixture ofequal volumes ofglacial acetic acid and Reference solution. A solution containing 3.0 per cent w/v of
water with the aid of gentle heat, cool and add sufficient tributyl citrate RS and acetyltributyl citrate RS in toluene.
water to produce 500.0 ml. Dilute 5.0 ml of this solution to
100.0 ml with 1 M acetic acid and measure the absorbance of Chromatographic system
the resulting solution at the maximum at about 360 urn (2.4.7). - a glass or stainless steel column 30 m x 0.32 mm, packed
with bonded with a 0.5-llm layer ofphase 042,
Calculate the content of C l2 H Il N 7 from the absorbance - temperature:
obtained by repeating the operation using triamterene RS in column. 80° to 220° from 0.5 to 10 minutes,
place of the contents of the capsules. inlet port. 85° to 250° from 0.5 to 10 minutes (increased
Storage. Store protected from moisture. @ 20° per minute) and detector 275°,
- a flame ionisation detector,
- flow rate. 2.3 ml per minute ofthe nitrogen as carrier gas.
Tributyl Citrate Inject 1 III ofthe reference solution. The test is not valid unless
the relative retention times with reference to acetyltributyl
citrate for tributyl citrate is about 0.9, the resolution between
the peaks corresponding to tributyl citrate and acetyltributyl
citrate is not less than 1.5 and the relative standard deviation
for replicate injections is not more than 2.0 per cent.
Inject 1 III ofthe test solution and the reference solution.
Calculate the content C 1s H32 0 7•
Storage. Store protected from moisture.

Mol. Wt. 360.5


Tributyl Citrate contains not less than 99.0 per cent of Trichloromonofluoromethane
C 1sH 32 0 7, calculated on the anhydrous basis.
Category. Excipient. CI
I
CI-C-F
Identification I
CI
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with tributyl citrate CC~F Mol. Wt. 137.4
RS or with the reference spectrum oftributyl citrate. Trichloromonofluoromethane contains not less than 99.6 per
B. In the Assay, the principal peak in the chromatogram cent and not more than 100.0 per cent ofCChF, calculated on
obtained with the test solution corresponds to the peak in the the anhydrous basis.
chromatogram obtained with the reference solution. Category. Pharmaceutical aid (Excipient).

2257

www.webofpharma.com
TRICHLOROMONOFLUOROMETHANE IP 2010

Identification temperature:
column 70° to 170° @10 0 per minute and 170° for 5
Determine by infrared absorption spectrophotometry (2.4.6),
minutes,
compare the spectrum with the obtained with
trichloromonofluromethane RS. inlet port at 110° and detector at 200°,
a flame ionisation detector,
Tests flow rate. 20 ml per minute ofthe nitrogen as carrier gas
Boiling temperature. Transfer a 100 ml sample at about 24° to Head-space conditions which may be used:
a tared, pear-shaped, 100-ml centrifuge tube containing a few bath temperature 100°,
boiling stones, and weigh. Suspend a thermometer in the liquid, valve/loop temperature 105°,
and place the tube in a medium maintained at a temperature of
sampling time is 3 seconds.
32° above the expected boiling temperature. When the
thennometer reading becomes constant, record as the boiling The relative retention time with reference to
temperature the thelmometer reading after at least 5per cent of trichloromonofluoromethane for dichlorodifluoromethaneis
the substance under ex,amination has distilled. Retain the about 0.5 and for dichlorotetrafluoroethane is about 0.8. The
remainder of the substance for the determination of High- sum ofthese two peak areas is not more than 0.2 per cent of
Boiling Residues. the total of all peak areas and the sum ofthe areas of all peaks
Water (2.3.43). Not more than 0.001 per cent (Method 3) with other than that for trichloromonofluoromethane is not more
the following modifications (a) provide the closed-system than 0.4 per cent of the total of all peak areas.
titrating vessel with an opening through which passes a Inject the test solution and reference solution. The test is not
coarse-porosity gas dispersion tube connected to a sample valid unless the resolution between the peaks due
cylinder; (b) dilute the reagent with anhydrous methanol to dichlorotetrafluoroethane and trichloromonofluoromethane is
give a water equivalence factor ofbetween 0.2 and 1.0 mg per not less than 2.0.
ml, age this diluted solution for not less than 16 hours before
sanitation; (c) obtain a 100 g sample as directed under inhalation Calculate the content of CCI3F.
preparation, and introduce the same into the titration vessel Storage. Store protected from moisture and prevent exposure
through the gas dispersion tube at a rate of about 100 ml of to excessive heat.
gas per minute.
High-boiling residues. Not more than 0.01 per cent, Allow 85
ml ofthe sample to distill as directed in the test for Approximate
Boiling Temperature, and transfer the centrifuge tube Triethyl Citrate
containing the remaining 15 ml of substance to a medium
maintained at a temperature 10° above the boiling temperature.
After 30 minutes, remove the tube from the water-bath, blot
dry, and weigh. Calculate the weight ofthe residue.
Inorganic chlorides. Place 5 ml of anhydrous methanol in a
test tube, add 3 drops of a saturated solution of silver nitrate
in anhydrous methanol, shake, and add 7 g of
Trichloromonofluoromethane; no opalescence or turbidity is
produced.
Assay. Determine by gas chromatography (2.4.13). Mol. Wt.276.3
Test solution. Introduce the liquid phase of Trichloromono- Triethyl Citrate is triethyl 2-hydroxypropane-l, 2, 3-
fluoromethane into an evacuated headspace vial tricarboxylate.
Reference solution. Introduce a liquid-phase mixture of Triethyl Citrate contains not less than 98.5 per cent and not
dichlorodifluoromethane, dichlorotetrafluoroethane, and more than 101.0 per cent of ClzHzo07, calculated on the
trichloromonofluoromethane into an evacuated headspace yjal. anhydrous basis.
Chromatographic system Category. Excipient.
- a steel column 1.8 m x 2 mm, packed with support S12, .Description. A clear, viscous, colourless or almost colourless,
containing 1 per cent phase G25, hygroscopic liquid.

2258

www.webofpharma.com
IP 2010 TRIFLUOPERAZINE HYDROCHLORIDE

Identification principal peak in the chromatogram obtained with the reference


solution. Ignore anypeak with an area less than 0.04 per cent
Test A may be omitted if tests Band C are carried out. Tests B the area of the principal peak in the chromatogram obtained
and C may be omitted if test A is carried out. with the reference solution.
A. Detennine by infrared absorption spectrophotometry (2.4.6). Heavy metals (2.3.13). Dissolve 4.0 g in 8 ml of ethanol (95 per
Compare the spectrum with that obtained with triethyl citrate cent) and dilute to 20 ml with water. 12 ml of the resulting
RS or with the reference spectrum oftriethyl citrate. solution complies with the limit test for the heavy metals,
B. To 0.5 ml add 5 ml of ethanol (95 per cent) and 4 ml of method D (5 ppm). Prepare the standard solution using lead
dilute sodium hydroxide solution. Boil under reflux for about standard solution (l ppm Pb) obtained by diluting lead
10 minutes, 2 ml ofthe solution gives the reactions of citrates standard solution (l00 ppm Pb) with a mixture of equal
(2.3.1). volumes of ethanol (95 per cent) and wate/:

C. It gives the reactions of esters (2.3.1). Sulphated ash (2.3.18). Not more than 0.1 per cent.
Water (2.4.19). Not more than 0.25 per cent, detennined on
Tests LOg.
Appearance of solution. The substance under examination is Assay. To 1.5 g, add 25 ml of 2-propanol, 50 rnl of water, 25.0
clear (2.4.1) and not more intensely coloured than reference ml of 1 M sodium hydroxide and a few glass beads into a 250-
solution BYS6 (2.4.1). ml borosilicate-glass flask fitted with a reflux condenser. Heat
under a reflux condenser for 1 hour and allow to cool. Titrate
Acidity. Dilute 109 with 10 rnl ofpreviously neutralized ethanol
(95 per cent). Add 0.5 ml of bromothymol blue solution. Not with 1 M hydrochloric acid, using 1 ml of phenolphthalein
more than 0.3 ml of 0.1 M sodium hydroxide is required to
solution as indicator. Carry out a blank titration.
change the color of the indicator to blue. 1 ml of 1 M sodium hydroxide is equivalent to 0.0921 g of
Refractive index (2.4.27). 1.440 to 1.446. ClzHzo07.

Related substances. Determine by gas chromatography Storage. Store protected from moisture.
(2.4.13).
Test solution. Disperse 1.0 ml of the substance under
examination in 50.0 ml of dichloromethane. Trifluoperazine Hydrochloride
Reference solution. Disperse 1.0 ml of the substance under
~
sn
examination and 0.5 ml of methyl tridecanoate in 50.0 ml of
dichloromethane.
Y"N~Nl
Chromatographic system
- a fused silica column 30 m x 0.32 mrn, packed with poly I ~N'CH
" 3
,2HCI

(dimethyl) siloxane (5 /lm), ~ CF 3


temperature:
column.200°, CzIHz~3N3S,2HCl Mol. Wt. 480.4
injection port and detector at 220°,
Trifluoperazine hydrochloride is 10-[3-(4-methylpiperazin-1-
a flame ionization detector,
yl)propyl]-2-trifluoromethylphenothiazine dihydrochloride.
linear velocity about 26 cm per second using nitrogen
as carrier gas Trifluoperazine Hydrochloride contains not less than 99.0 per
cent and not more than 101.0 per cent ofCzIHz4F3N3S, 2HCl,
Inject 1 III ofthe reference solution. The test is notvalid unless
calculated on the dried basis.
the resolution between the peaks due to triethyl citrate and
methyl tridecanoate is not less than 1.5. Category. Antipsychotic; antiemetic.
Inject 1 /ll each ofthe test solution and the reference solution. Dose. As antipsychotic, orally, the equivalent of2 to 30 mg of
Run the chromatogram twice the retention time ofthe prinCipal trifluoperazine daily, in divided doses; by intramuscular
peak. In the chromatogram obtained with the test solution, injection, the equivalent ofl to 3 mg oftrifluoperazine daily, in
the area of"any secondary peak is not more than 0.2 per cent divided doses. As antiemetic, orally, the equivalent of 2 to 4
the area of the principal peak in the chromatogram obtained mg of trifluoperazine daily; by intramuscular injection, the
with the reference solution. The sum of the areas of all the equivalent of 1 to 3 mg of trifluoperazine daily, in divided
secondary peaks is not more than 0.5 per centthe area of the doses.

2259

www.webofpharma.com
TRIFLUOPERAZINE HYDROCHLORIDE IP 2010

Description. A white to pale yellow, crystalline powder; Trifluoperazine Injection


odourless or almost odourless; slightly hygroscopic.
Trifluoperazine Hydrochloride Injection
NOTE - In the following procedures, the test and standard
specimens and the solutions containing them should be Trifluoperazine Injection is a sterile solution ofTrifluoperazine
protected by carrying out the tests without delay and in Hydrochloride in Water for Injections.
subdued light. Trifluoperazine Injection contains not less than 90.0 per cent
Identification and not more than 110.0 per cent of the stated amount of
trifluoperazine, C2IH2<tF3N3S,
A. Dissolve 20 mg in 10 ml ofwater, make the solution alkaline
Usual strength. The equivalent of 1mg oftrifluoperazine perml.,
to litmus paper with 5 M sodium hydroxide and extract with
two quantities, each of20 ml, of light petroleum (60° to 80°). Description. A clear, colourless solution.
Combine the extracts, wash with 10 ml of water, shake with 5 g
NOTE - In the following procedures, the test and standard
of anhydrous sodium sulphate, filter and evaporate the filtrate
specimens and the solutions containing them should be
carefully to dryness.
protected by carrying out the tests without delay and in
On the residue, determine by infrared absorption subdued light.
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with trifluoperazine hydrochloride RS or with the Identification
reference spectrum oftrifluoperazine hydrochloride. When examined in the range 230 nm to 360 nm (2.4.7), the final
B. Complies with the test for identification ofphenothiazines solution obtained in Assay shows an absorption maximum at
(2.3.3), using as reference solution a 0.2 per cent w/v solution about 256 nm.
of trifluoperazine hydrochloride RS in chloroform.
Tests
C. When examined in the range 280 nm to 360 nm (2.4.7), a
0.01 per cent w/v solution in 0.1 M hydrochloric acid pH (2.4.24). 4.0 to 5.0.
measured immediately after preparation shows an absorption Other tests. Complies with the tests stated under Parenteral
maximum at about 305 nm. Dilute 10 ml of the solution to preparations (Injections).
100 ml with 0.1 M hydrochloric acid. When examined in the
Assay. To an accurately measured volume of the injection
range 230 nm to 280 nm, the resulting solution shows an
containing about 50 mg of trifluoperazine add 10 ml of 2 M
absorption maximum at about 256 nm; absorbance at about
256 nm, about 0.65.
sulphuric acid and extract with three quantities, each of25 ml,
of carbon tetrachloride. Discard the carbon tetrachloride
D. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand extract after each extraction. Add 10 ml of strong ammonia
for 5 minutes; an orange colour is produced. solution and extract with five quantities, each of 50 ml, of
E. A 10 per cent w/v solution gives the reactions of chlorides cyclohexane. Extract the combined cyclohexane extracts with
(2.3.1). five quantities, each of 50 ml, of 0.1 M hydrochloric acid.
Dilute the combined acid extracts to 500.0 ml and mix. Measure
Tests the absorbance of the resulting solution at the maximum at
pH (2.4.24). 1.7 to 2.6, determined in a 10.0 per cent w/v solution. about 256 nm (2.4.7).
Related substances. Complies with the test for related Calculate the content ofC21H24F3N3S taking 743 as the specific
substances in phenothiazines (2.3.5), using mobile phase A. absorbance at 256 11)11.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Storage. Store protected from light.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined Labelling. The label states the strength in terms of the
on 1.0 g by drying in an oven at 105° at a pressure not exceeding equivalent amount oftrifluoperazine per ml.
0.7kPa.
Assay. Weigh accurately about 0.4 g, dissolve in 50 ml of
anhydrous glacial acetic acid add 10 ml of mercuric acetate Trifluoperazine Tablets
solution. Titrate with 0.1 M perchloric acid, using crystal
vialelsalm/on as~indic~ator.~Cany~ouCablaiilnitfation.~~-~-~~ TrifluoperazineHydrochloride~Tablets'
1 .tril of 0.1 Mperchl()ric~ acid is equivalent to 0.02402 g of Trifluoperazine Tablets contain not less than 92.5 per cent
C2IH2<tF3N3S,2HCl. and not more than 107.5 per cent of trifluoperazine,
Storage. Store protected from light and moisture. C21H24F3N3S, The tablets are coated.

2260

www.webofpharma.com
IP 2010 TRIFLUPROMAZINE HYDROCHLORIDE

Usual strengths. The equivalent of 1 mg and 5 mg of Triflupromazine Hydrochloride


trifluoperazine.
Flupromazine Hydrochloride
NOTE - In the following procedures, the test and standard
specimens and the solutions containing them should be
protected by canying out the tests without delay and in
subdued light.

Identification I Hel

A. Shake a quantity ofthe powdered tablets containing 20 mg


of trifluoperazine with 30 ml of I M hydrochloric acid for
10 minutes, filter, make the filtrate alkaline to litmus paper with
5 M sodium hydroxide and extract with two quantities, each MoL Wt. 388.9
of20 ml, of lightpetroleum (60° to 80°). Combine the extracts,
Triflupromazine Hydrochloride is 10-[3-(dimethylamino)-
wash with 10 ml of water, shake with 5 g of anhydrous sodium
propyl)]-2-trifluoromethylphenothiazine hydrochloride.
sulphate, filter and evaporate the filtrate carefully to cJ.r:Yness.
On the residue, determine by infrared absorption Triflupromazine Hydrochloride contains not less than
spectrophotometry (2.4.6). Compare the spectrum with that 97.0percent and not more than 103.0 per centofClsHl9F3N2S,
obtained with trifluoperazine hydrochloride RS or with the HCI, calculated on the dried basis.
reference spectrum oftrifluoperazine hydrochloride. Category. Antipsychotic; antiemetic.
B. Extract a quantity ofthe powdered tablets containing 5 mg Dose. As antipsychotic, orally, 30 to 150 mg oftriflupromazine
oftrifluoperazine with 5 ml ofacetone, filter and evaporate the daily, in divided doses; by intramuscular injection, 10 mg
filtrate to dryness. Add 2 ml of sulphuric acid to the residue repeated 4-hourly, ifnecessary; by intravenous injection, 1 to
and allow to stand for 5 minutes; an orange colour is produced. 3 mg repeated 4-hourly, if necessary. As antiemetic, 20 to 30
mg oftriflupromazine daily; by intramuscular injection 1 to 3
Tests mg.
Uniformity of content. Comply with the test stated under Description. A white to pale yellowish brown, crystalline
Tablets. powder; odour slight and characteristic.
Place one tablet in a 100-ml volumetric flask, add 50 ml of a NOTE - In the following procedures, the test and standard
mixture of 19 volumes of water and 1 volume of hydrochloric specimens and the solutions containing them should be
acid, shake until the tablet has completely disintegrated, dilute protected by carrying out the tests without delay and in
to volume with the same mixture, mix and filter, rejecting the subdued light.
first few ml ofthe filtrate. Dilute suitably, ifnecessary with the
same solvent mixture and measure the absorbance of the Identification
resulting solution at the maximum at about 256 nm (2.4.7).
Calculate the content ofC21H24F3N3S taking 743 as the specific A. Dissolve 20 mg in 10 ml of water, make the solution alkaline
absorbance at 256 nm. to litmus paper with 5 M sodium hydroxide and extract with
two quantities, each of20 ml, of light petroleum (60° to 80°).
Other tests. Comply with the tests stated under Tablets. Combine the extracts, wash with 10 ml of water, shake with 5 g
Assay. Weigh and powder 20 tablets. Weigh accurately a of anhydrous sodium sulphate, filter and evaporate the filtrate
quantity ofthe powder containing about 5 mg oftrifluoperazine, carefully to dryness.
shake for 15 minutes with 400 ml ofa m~ture of 19 volumes of The residue complies with the following test. Determine by
water and 1 volume of hydrochloric acid, dilute to 500.0 ml
infrared absorption spectrophotometry (2.4.6). Compare the
with the same solvent mixture, mix and filter. Measure the spectrum with that obtained with trijlupromazine hydro-
absorbance of the filtrate at the maximum at about 256 nm chloride RS or with the reference spectrum oftriflupromazine
(2.4.7).
hydrochloride.
Calculate the content ofC21H24F3N3S taking 743 as the specific
B. Complies with the test for identification ofphenothiazines
absorbance at 256 nm.
(2.3.3), using as reference solution a 0.2 per cent w/v solution
Storage. Store protected £i'om light and moisture. of triflupromazine hydrochloride RS in chloroform.
Labelling. The label states the strength in terms of the C. When examined in the range 230 nm to 360 nm (2.4.7), a
equivalent amount of trifluoperazine. 0.00 I per cent w/v solution in 0.1 M hydrochloric acid shows

2261

www.webofpharma.com
TRIFLUPROMAZINE INJECTION IP 2010

an absorption maximum atabout 256 nm and a less well-defined Tests


maximum at about 305 nm.
pH (2.4.24).3.5 to 5.2.
D. A 10 per cent wlv solution gives the reactions of chlorides
Other tests. Complies with the tests stated under Parenteral
(2.3.1).
Preparations (Injections).
Tests Assay. To an accurately measured volume of the injection
Related substances. Complies with the test for Related containing about 20 mg ofTriflupromazine Hydrochloride add
substances in phenothiazines (2.3.5), using mobile phase A. sufficient ofa mixture ofl9 volumes of water and I volume of
hydrochloric acid to produce 100.0 ml and mix. Dilute 5.0 ml
Sulphated ash (2.3.18). Not more than 0.1 per cent. of the solution to 100.0 ml with the same mixture and mix.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Measure the absorbance of the resulting solution at the
on 1.0 g by drying in an oven at 105 0 for 2 hours. maximmn at about 256 nrtJ. (2.4.7).

Assay. Weigh accurately about 0.8 g, dissolve in 50 ml of Calculate the content of ClsHI9F3N2S, HCI taking 700 as the
anhydrous glacial acetic acid add 10 ml of mercuric acetate specific absorbance at 256 nm.
solution. Titrate with 0.1 M perchloric acid, using clystal Storage. Store protected from light.
violet solution as indicator. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.03889 g of
ClsHI9F3N2S, HCI. Triflupromazine Tablets
Storage. Store protected from light and moisture.
Triflupromazine Hydrochloride Tablets; Flupromazine
Hydrochloride Tablets; Flupromazine Tablets

Triflupromazine Injection Triflupromazine Tablets contain not less than 90.0 per cent
and not more than 110.0 per cent of the stated amount of
Triflupromazine Hydrochloride Injection; Flupromazine triflupromazine hydrocWoride, ClsHI9F3N2S, HCI.
Hydrochloride Injection; Flupromazine Injection Usual strengths. 10 mg; 25 mg.
Triflupromazine Injection is a sterile solution ofTriflupromazine NOTE - In the following procedures, the test and standard
Hydrochloride in Water for Injections. . specimens and the solutions containing them should be
Triflupromazine Injection contains not less than 90.0 per cent protected by canying out the tests without delay and in
and not more than 110.0 per cent of the stated amount of subdued light.
triflupromazine hydrochloride, ClsHI9F3N2S, HCI.
Identification
Usual strength. 10 mg per mI.
A. Shake a quantity ofthe powdered tablets containing 20 mg
Description. A clear, almost colourless solution. of Triflupromazine Hydrochloride with 30 ml of 1 M
NOTE - In the following procedures, the test and standard hydrochloric acid for 10 minutes, filter, malce the filtrate al1caline
specimens and the solutions containing them should be to litmus paper with 5 M sodium hydroxide and extract with
protected by carrying out the tests without delay and in two quantities, each of 20 ml, of light petroleum (boiling
subdued light. range 600 to 800) . Combine the extracts, wash with 10 ml of
water, shake with 5 g of anhydrous sodium sulphate, filter and
Identification evaporate the filtrate carefully to dryness.
A. To an appropriate quantity of the injection add sufficient On the residue, determine by infrared absorption
0.1 M hydrochloric acid to produce a solution containing spectrophotometry (2.4.6). Compare the spectrum with that
0.001 per cent wIv solution ofTriflupromazine Hydrochloride. obtained with trijlupromazine hydrochloride RS or with the
When examined in the range 230 nm to 360 nm (2.4.7), the reference spectrum oftriflupromazine hydrochloride.
resulting solution shows an absorption maximum at about B. Extract a quantity of the powdered tablets containing 5 mg
256nm. ofTriflupromazine Hydrochloride with 5 ml of acetone, filter
and evaporate the filtrate to dryness. . - - _. --
B. To a volume containing about 100 mg of TrifluprOluazine
Hydrochloride add 5 ml of 8 M nitric acid and mix; a pink to When examIned in the range 230 nm to 360 11m (2A.7), a
amber colour develops which quicldy turns dark brown and 0.00 I per cent wlv solution in 0.1 M hydrochloric acid shows
then changes to a clear solution having a yellow tint. an absorption maximum at about 256 nm.

2262

www.webofpharma.com
IP 2010 TRIMETAZIDINE HYDROCHLORIDE

Tests Tests
Uniformity of content. (For tablets containing 10 mg or less. Appearance of solution. A 10 per cent w/v solution is clear
Comply with the test stated under Tablets. (2.4.1) and not more intensely coloured than reference solution
BYS6(2.4.l).
Crush one tablet and transfer to a 1OO-ml volumetric flask with
Related substances. Determine by liquid chromatography
the aid of a mixture of 19 volumes of water and 1 volume of
(2.4.14).
hydrochloric acid. Shake well, dilute to volume withthe same
mixture and complete the procedure described under the Assay Test solution. Dissolve 0.2 g of the substance under
beginning at the words "mix and filter, ...". examination in 50 ml of water.
Calculate the content ofClsH19F3NzS, HCl in the tablet. Reference solution. Dilute 2.0 ml of the test solution to 100.0
Other tests. Comply with the tests stated under Tablets.
ml with water. Dilute 5.0 ml of this solution to 100.0 ml with
water.
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 50 mg of Chromatographic system
Triflupromazine Hydrochloride and shake for 15 minutes with - a stainless steel columnl5 cm x 4.6 mm packed with
200 ml of a mixture of 19 volumes of water and 1 volume of octadecylsilane bonded to porous silica (5 /lm),
hydrochloric acid. Dilute to 500.0 ml with the same solvent
mobile phase: A. a mixture of35.7 volumes of methanol
mixture, mix and filter, rejecting the first few ml ofthe filtrate. and 64.3 volumes of a 0.29 per cent w/v solution of
sodium heptanesulphonate, adjusted to pH 3.0 with
Dilute 10.0 ml to 100.0 ml with the same solvent mixture.
orthophosphoric acid,
Measure the absorbance ofthe filtrate at the maximum at about
256 urn (2.4.7). B. methanol,
a linear gradient programme using the conditions given
Calculate the content ofClsH19F3NzS, HCl taking 700as the below,
specific absorbance at 256 urn. flow rate. 1.2 ml per minute,
Storage. Store protected from light and moisture. spectrophotometer set at 240 urn,
injection volume. 10 Ill.
Time Mobile phase A Mobile phase B
(min.) (per cent v/v) (per cent v/v)
Trimetazidine Hydrochloride 0-50 95~75 5~25

50-52 75~95 25~5

Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
than 2.0 per cent. The relative retention time with reference to
trimetazidine for (2,3,4-trimethoxyphenyl)methanol
CIJfzzNz03,2HCl Mol. Wt. 339.3 (trimetazidine impurity D) is about 0.2, for 2,3,4-
trimethoxybenzaldehyde (trimetazidine impurity C) is about
Trimetazidine Hydrochloride is 1-[(2,3,4-trimethoxyphenyl)-
0.4, for ethyl 4-(2,3,4-trimethoxybenzyl)piperazine-l-
methyl]piperazine dihydrochloride.
carboxylate (trimetazidine impurity H) is about 0.6, for 1-(3,4,5-
Trimetazidine Hydrochloride contains not less than 98.5 per trimethoxybenzyl)piperazine (trimetazidine impurity A) and
cent and not more than 101.5 per cent, calculated on the dried N-methyltrimetazidine (trimetazidine impurity I) is about 0.9,
basis. for 1-(2,4,5-trimethoxybenzyl)piperazine (trimetazidine impurity
Category. Antiischemic metabolic agent. E) is about 0.95, for 1-(2,4,6-trimethoxybenzyl) piperazine
(trimetazidine impurity F) is about 1.4 and for 1,4-bis(2,3,4-
Description. A white or almost white, crystalline powder. trimethoxybenzyl) piperazine (trimetazidine impurity B) is about
1.8.
Identification
Inject the test solution and the reference solution. In the
A. Determine by infrared absorption spectrophotometry (2.4.6). chromatogram obtained with the test solution, the area of
Compare the spectrum with that obtained with trimetazidine each peak due to trimetazidine impurity A, B, C, D, E, F, H, I is
dihydrochloride RS or with the reference spectrum of not more than the area of the principal peak in the
trimetazidine dihydrochloride. chromatogram obtained with the reference solution (0.1 per
B. Gives reaction (a) for chlorides (2.3.1). cent), the area of any other secondary peak is not more than

2263

www.webofpharma.com
TRIFLUPROMAZINE HYDROCHLORIDE IP 2010

the area of the principal peak in the chromatogram obtained Trimethoprim contains not less than 98.5 per cent and not
with the reference solution (0.1 per cent). The sum ofareas of more than 101.0 per cent of CJ4HJ8N403, calculated on the
all the secondary peaks is not more than twice the area ofthe dried basis.
principal peak in the chromatogram obtained with the reference
Category. Antibacterial.
solution (0.2 per cent). Ignore any peak with an area less than
0.5 times the area ofthe principal peak in the chromatogram Dose. 200 mg twice daily.
obtained with the reference solution (0.05 per cent). Description. A white or yellowish white powder; odourless or
Impurity G. Determine by thin-layer chromatography (2.4.17), almost odourless.
coating the plate with silica gel G.
Identification
Mobile phase. A mixture of 20 volumes of concentrated
ammonia and 80 volumes of ethanol (95 per cent). Test A may be omitted iftests Band C are carried out. Tests B
. and C may be omitted if test A is carried out.
Test solution. Dissolve 0.1 g of the substance under
examination in 10.0 ml of methanol. A. Determine by infrared absorption spectrophotometry (2.4 .6).
Compare the spectrum with that obtained with trimethoprim
Reference solution. Dissolve 22.6 mg of piperazine hydrate
RS or with the reference spectrum oftrimethoprim.
(impurity G) in 100 m1 of methanol. Dilute 10 m1 ofthis solution
to 100 ml with methanol. B. Dissolve 25 mg in 25 ml of ethanol (95 per cent) and dilute
2.0 ml to 100 ml with 0.1 M sodium hydroxide.
Apply to the plate 10 f.l.l of each solution. Allow the mobile
phase to rise 8 cm. Dry the plate at 105 0 for 30 minutes and When examined in the range 230 urn to 360 nm (2.4.7), the
spray with iodoplatinate reagent. Any secondary spot resulting solution shows an absorption maximum at about
corresponding to trimetazidine impurity G is not more intense 287 urn; absorbance at about 287 urn, about 0.49.
than the spot in the chromatogram obtained with the reference C. Dissolve about 25 mg in 5 ml of 0.005 M sulphuric acid,
solution (0.1 per cent). with heating ifnecessary, add 2 ml ofa 1.6 per cent wlv solution
Heavy metals (2.3.13). 1.0 g complies with the limit test for ofpotassium permanganate in 0.1 M sodium hydroxide. Heat
heavy metals, Method B (20 ppm). to boiling and to the hot solution add 0.4 ml offormaldehyde
solution. Mix, add I ml of 0.5 M sulphuric acid, mix and heat
Sulphated ash (2.3 .18). Not more than 0.1 per cent.
to boiling. Cool and filter. Add 2 ml of chloroform to the filtrate
Loss on drying (2.4.19). Not more than 2.5 per cent, determined and shake vigorously. The chloroform layer exhibits a green
on 1.0 g by drying in an oven at 105 0 over diphosphorus fluorescence when examined in ultraviolet light at 365 urn.
pentoxide at a pressure not exceeding 15 kPa.
Tests
Assay. Dissolve 0.12 g in 50.0 ml of water. Add 1 mlofnitric
acid and titrate with 0.1 M silver nitrate, determining the end- Appearance ofsolution. A 5.0 per cent wIv solution in a mixture
pointpotentiometrically (2.4.25). Carry out a blank titration. of 10 volumes of chloroform, 9 volumes of methanol and
I ml of 0.1 M silver nitrate is equivalent to 0.01696 g of 2 volumes of water is not more intensely coloured than
ClJIz4ClzNz03' reference solution BYS7 (2.4.1).

Storage. Store protected from moisture. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 85 volumes of ethyl acetate,
10 volumes of methanol, 5 volumes of water and 2 volumes of
Trimethoprim anhydrous formic acid.
Test solution. Dissolve 0.4 g of the substance under
examination in 10 m1 ofa mixture of 10 volumes of chloroform,
9 volumes of methanol and 2 volumes of water.
Reference solution. A 0.008 per cent wlv solution of the
substance under examination in a mixture of 10 volumes of
ch7orojoriii;9 v61urnes6fmelhCiiWland2v61iiiiies6f water:
Mol. Wt. 290.3
Apply to the plate 5 f.l.l of each solution. Allow the mobile
Trimethoprim is 5-(3,4,5-trimethoxybenzyl)pyrimidine- phase to rise 17 cm. Dry the plate in a current of cold air for
2,4-diamine. 5 minutes and examine in ultraviolet light at 254 urn. Place an

2264

www.webofpharma.com
IP 20ID TRIMETHOPRIM AND SULPHAMETHOXAZOLE ORAL SUSPENSION

evaporating dish containing a mixture of 2 volumes of a Usual strengths. Trimethoprim 40 mg and Sulpha- methoxazole
1.5 per cent w/v solution of potassium permanganate, 200 mg in 5 ml; Trimethoprim 80 mg and Sulphamethoxazole
1 volume of 7 M hydrochloric acid and 1 volume of water at 400 mg in 5 ml.
the bottom of a closed tank and allow to stand for 15 minutes.
Place the dried plate in the closed tank and expose to the Identification
chlorine vapour for 5 minutes. Remove the plate from the tank
and remove the chlorine in a current of cold air until an area Determine by thin-layer chromatography (2.4.17), coating the
below the line of application does not give a blue colour on plate with silica gel G .
the addition of 0.05 ml of potassium iodide and starch Mobile phase. A mixture of 20 volumes of chloroform,
solution. Spray the plate with potassium iodide and starch 2 volumes of methanol and 1 volume of dimethylformamide.
solution and examine in daylight. By both methods of
visualisation any secondary spot in the chromatogram Test solution. Add 20 ml of methanol (or a suitable volume of
obtained with the test solution is not more intense than the methanol to yield 0.4 per cent w/v solution ofTrimethoprim)
spot in the chromatogram obtained with the reference solution. to 5 ml of the suspension, mix, shake with 10 g of anhydrous
sodium sulphate, centrifuge and use the supernatant liquid.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm). Reference solution (a). A 2.0 per cent w/v solution of
sulphamethoxazole RS in methanol.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Reference solution (b). A 0.4 per cent w/v solution of
Loss on drying (2.4.19). Notmorethan 1.0 percent, determined trimethoprim RS in methanol.
on 1.0 g by drying in an oven at 105°.
Apply to the plate 5 Jll of each solution. After development,
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of dry the plate in air, spray with dilute potassium
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric iodobismuthate solution. One of the principal spots in the
acid, determining the end-point potentiometrically (2.425). chromatogram obtained with the test solution corresponds to
Carry out a blank titration. that in the chromatogram obtained with reference solution (a)
1 ml of 0.1 M perchloric acid is equivalent to 0.02903 g of and the other corresponds to· that in the chromatogram
CIJIISN403. obtained with reference solution (b)
Storage. Store protected from light.
Tests
pH (2.4.24). 5.0 to 6.5.
Trimethoprim and Sulphamethoxazole Other tests. Complies with the tests stated under Oral liquids.
Oral Suspension Assay. For trimethoprim Weigh accurately about 4 g, add
30 ml of 0.1 M sodium hydroxide, shake and extract with four
Sulphamethoxazole and Trimethoprim Oral Suspension; quantities, each of50 ml, of chloroform,washing each extract
Co-trimoxazole Oral Suspension; Co-trimoxazole Mixture with the same two quantities, each of 10 ml, of 0.1 M sodium
Trimethoprim and Sulphamethoxazole Oral Suspension is a hydroxide. Reserve the combined aqueous solution and
suspension of Trimethoprim and Sulphamethoxazole in a washings for the Assay for sulphamethoxazole. Extract the
suitable flavoured vel1icle. It contains 5 parts of combined chloroform extracts with four quantities, each of
Sulphamethoxazole for 1 part, by weight, ofTrimethoprim. 50 ml, of 1 M acetic acid. Wash the combined extracts with
. 5 ml of chloroform and dilute the aqueous extracts to 250.0 ml
Trimethoprim and Sulphamethoxazole Oral Suspension
contains not less than 90.0 per cent and not more than with 1 M acetic acid. To 10.0 ml ofthis solution add 10mi of
110.0 per cent of the stated amounts of trimethoprim, 1 M acetic acid and sufficient water to produce 100.0 ml, mix
C14HISN403, and sulphamethoxazole, CIOHIIN303S. and measure the absorbance of the resulting solution at the
maxirninn at about 271 nm(2.4.7).
Category. Antibacterial.
Calculate the content ofC,4H,sN403 taking 204 as the specific
Dose. For children upto 1 year, Trimethoprim, 40 mg and absorbance at 271 nm. Detennine the weight per ml of the
Sulphamethoxazole, 200 mg daily, in divided doses. For children suspension (2.4.29), and calculate the content ofC,4H,sN403,
1 to 5 years of age, Trimethoprim, 80 mg and Sulpha-
weight in volume.
methoxazole, 400 mg daily, in divided doses. For adults,
Trimethoprim, 160 to 480 mg and Sulphamethoxazole, 800 mg For sulphamethoxazole - Carry out thefollowing procedure
to 2.4 g daily, in divided doses. protected fi'om light.

2265

www.webofpharma.com
TRIMETHOPRIM AND SULPHAMETHOXAZOLE ORAL SUSPENSION IP 2010

Dilute the combined aqueous solution reserved in the Assay Trimethoprim, 160 to 480 mg and Sulphamethoxazole, 800 mg
for ~ethoprim to 250.0 m1 with water, filter and dilute 5.0 m1 to 2.4 g daily, in divided doses.
ofthe filtrate to 200.0 ml with water (solution A). To 2.0 ml of Usual strengths. Trimethoprim 20 mg and Sulpha- methoxazole
solution A add 0.5 ml of 4 M hydrochloric acid and 1 ml of a 100 mg; Trimethoprim 80 mg and Sulphamethoxazole 400 mg;
0.1 per cent w/v solution ofsodium nitrite and allow to stand Trimethoprim 160 mg and Sulphamethoxazole 800 mg.
for 2 minutes. Add 1 ml of a 0.5 per cent w/v solution of
ammonium sulphamate and allow to stand for 3 minutes. Add Identification
1 ml of a 0.1 per cent w/v solution of N- (l-napthyl)
ethylenediamine dihydrochloride and allow to stand for A. To a quantity of the powdered tablets containing 50 mg of
10 minutes. Dilute the solution to 25.0 ml with water and Trimethoprim add 30 ml of0.1 M sodium hydroxide and extract
measure the absorbance of the resulting solution at about with two quantities, each of 50ml, of chloroform. Wash the
538 Dill (2.4.7), using as the blank a solution prepared in the combined chloroform extracts with two quantities, each of
same manner but using 2 ml of water in place of solution A. 10 ml, of 0.1 M sodium hydroxide and then with 10 ml of
Weigh accurately about 0.25 g of sulphamethoxazole RS, water. Combine the aqueous extract and washings (solution
dissolve in 50 ml of 0.1 M sodium hydroxide and dilute to A) and reserve for test B. Shake with 5 g ofanhydrous sodium
250.0 ml with water. Dilute 5.0 ml ofthe resulting solution to sulphate, filter and evaporate to dryness using a rotary
200.0 ml with water (solution B). Repeat the procedure using evaporator.
2.0 ml ofsolution B and beginning atthe words "add 0.5 mlof The residue complies with the following test. Determine by
4 M hydrochloric acid......". infrared absorption spectrophotometry (2.4.6). Compare the
Calculate the content ofCJOHIIN303S from the values of the spectrum with that obtained with trimethoprim RS or with the
absorbances obtained. Calculate the content ofC JOH lI N 30 3S, reference spectrum oftrimethoprim.
weight in volume from the weight per m1 determined in the B. Filter solution A, add, dropwise, sufficient 2 M hydro-
Assay for trimethoprim. chloric acid to the filtrate to make it just acidic and extract
with 50 ml of ether. Wash the ether layer with 10 ml ofwater,
Storage. Store protected from light and moisture. The
shake with 5 g of anhydrous sodium sulphate, filter and
suspension should not be allowed to freeze.
evaporate the filtrate to dryness using a rotary evaporator.
Labelling. The label states (1) the content of Trimethoprim Dissolve the residue in the minimum volume of a 5 per cent
and ofSulphamethoxazole in each 5 ml ofthe suspension; (2) w/v solution of sodium carbonate, add 1 M hydrochloric
that the contents should be shaken before use; (3) that a acid dropwise until precipitation is complete and filter. Wash
suspension containing 40 mg ofTrimethoprim and 200 mg of the residue sparingly with water and dry at 105°.
Sulphamethoxazole in 5 ml is meant for paediatric use.
The residue complies with the following test. Determine by
infrared absorption spectrophotometry (2.4.6). Compare the
spectrum with that obtained with sulphamethoxazole RS or
with the reference spectrum ofsulphamethoxazole.
Trimethoprim and Sulphamethoxazole
C. Determine by thin-layer chromatography (2.4.17), coating
Tablets the plate with silica gel G
Sulphamethoxazole and Trimethoprim Tablets; Mobile phase. A mixture of 20 volumes of chloroform,
Co-trimoxazole Tablets 2 volumes of methanol and 1 volume of dimethylformamide.
Trimethoprim and Sulphamethoxazole Tablets contain 5 parts Test solution. Shake a quantity of the powdered tablets
ofSulphamethoxazole for 1 part, by weight, ofTrimethoprim. containing 0.4 g ofSulphamethoxazole with 20 m1 ofmethanol
and filter.
Trimethoprim and Sulphamethoxazole Tablets contain not less
than 92.5 per cent and not more than 107.5 per cent of the Reference solution (a). A 2.0 per cent w/v solution of
stated amounts of trimethoprim, C14HISN403, and sulpha- sulphamethoxazole RS in methanol.
methoxazole, CJOHIIN303S. .
Reference solution (b). A 0.4 per cent w/v solution of
Category. Antibacterial. trimethoprim RS in methanol.
Dose. For children upto 1 year, Trimethoprirn,40 rngand Apply to the plate 5 lliof {Jach solution. After development,
Sulphamethoxazole, 200 mg daily, in divided doses. For children dry the plate in air, spray with dilute potassium
1 to 5 years of age, Trimethoprim, 80 mg and Sulpha- iodobismuthate solution. One of the principal spots in the
methoxazole, 400 mg daily, in divided doses. For adults, chromatogram obtained with the test solution corresponds to

2266

www.webofpharma.com
IP 2010 TRIMETHOPRIM TABLETS

that in the chromatogram obtained with reference solution (a) B. Shake a quantity of the powdered tablets containing 0.1 g
and the other corresponds to that in the chromatogram of Trimethoprim with 60 ml of 0.1 M hydrochloric acid for
obtained with reference solution (b) 20 minutes, add sufficient 0.1 Mhydrochloric acidto produce
100 ml, filter and dilute 5 ml of the filtrate to 250 ml with
Tests 0.1 M sodium hydroxide.
Other tests. Comply with the tests stated under Tablets. When examined in the range 230 nm to 360 nm (2.4.7), the
Assay. For trimethoprzm - Weigh accurately a quantity of resulting solution shows an absorption maximum at about
the powdered tablets containing about 50 mg ofTrimethoprim, 287nm.
add 30 ml of 0.1 M sodium hydroxide and extract with four
quantities, each of50 ml, of chloroform, washing each extract Tests
with the same two quantities, each of 10 ml, of 0.1 M sodium
hydroxide. Combine the chloroform extracts and extract with Related substances. Determine by liquid chromatography
four quantities, each of 50 ml, of 1 M acetic acid. Wash the (2.4.14).
combined extracts with 5 ml of chloroform and dilute the Test solution. Shake a quantity of the powdered tablets
aqueous extracts to 250.0 ml with 1 M acetic acid. To 10.0 ml containing 0.2 g of Trimethoprim with 50 ml of the mobile
of the solution add 10 ml of 1 M acetic acid and sufficient phase, filter and use the filtrate.
water to produce 100.0 ml, mix and measure the absorbance of
the resulting solution at the maximum at about 271 nm (2.4.7). Reference solution. Dilute 1 volume of the test solution to
Calculate the content OfCl4HlSN403 taking 204 as the specific 100 volumes with the mobile phase.
absorbance at 271 nm. Chromatographic system
For sulphamethoxazole - Weigh and powder 20 tablets. - a stainless steel column 20 cm x 5 rom, packed with
Weigh accurately a quantity of the powder containing about octadecylsilane bonded to porous silica (5 11m),
0.2 g ofSulphamethoxazole, dissolve as completely as possible - mobile phase: 0.14 per cent w/v solution of sodium
in 60 ml of water and 10 ml of hydrochloric acid. Add 3 g of perchlorate in methanol (60 per cent) adjusted to
potassium bromide, cool in ice and carry out the nitrite titration pH 3.1 with 0.1 M hydrochloric acid,
(2.3.31). - flow rate. 1.3 ml per minute,
1 ml of 0.1 M sodium nitrite is equivalent to 0.02533 g of - spectrophotometer set at 280 nm,
CIOHIIN303S. - injection volume. 20 Ill.
Storage. Store protected from light and moisture. The column efficiency, determined using the peak due to
trimethoprim in the chromatogram obtained with reference
Labelling. The label states the quantities of Trimethoprim
solution, should be at least 8,000 theoretical plates per
and of Sulphamethoxazole in each tablet.
metre.
In the chromatogram obtained with the test solution the sum
of the areas of any secondary peaks is not greater than the
Trimethoprim Tablets area ofthe principal peak in the chromatogram obtained with
reference solution.
Trimethoprim Tablets contain not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of· Other tests. Comply with the tests stated under Tablets.
trirnethoprim, Cl~ISN403'
Assay. Weigh and powder 20 tablets. Weigh accurately a
Usualstrengths. 100 mg; 200 mg. quantity ofthe powder containing about 0.1 g ofTrimethoprim,
add 100 ml ofglacial acetic acid, shake for 20 minutes, dilute
Identification
to 200.0 ml with glacial acetic acid and filter. To 5.0 ml ofthe
A. Shake a quantity of the powdered tablets containing 0.1 g filtrate add 15 ml of glacial acetic acid and dilute to 100.0 ml
ofTrimethoprim with 10 ml of chloroform, filter and evaporate with water. Measure the absorbance ofthe resulting solution
the filtrate to dryness. atthe maximum at about 271 nm (2.4.7).
On the residue, determine by infrared absorption Calculate the content OfCl4HlSN403 taking 204 as the specific
spectrophotometry (2.4.6). Compare the spectrum with that absorbance at 271 nm.
obtained with trimethoprim RS or with the reference spectrum
oftrirnethoprim. Storage. Store protected from moisture.

2267

www.webofpharma.com
TRlPROLIDINE HYDROCHLORIDE IP 2010

Triprolidine Hydrochloride Reference solution (b). A 0.02 per cent w/v solution of
Z-triprolidine RS in methanol.
Apply to the plate 5 III of each solution. After development,
H
Nf) dry the plate in air and examine in ultraviolet light at 254 nm. In
the chromatogram obtained with the test solution any spot
y
N
corresponding to Z-triprolidine is not more intense than the
spot in the chromatogram obtained with reference solution
(b) and any other secondary spot is not more intense than the
spot in the chromatogram obtained with reference solution (a).
CI9HzzNz,HCl,HzO Mol. Wt. 332.9
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Triprolidine Hydrochloride is (E)-2-(3-pyrrolidin-l-yl-
heavy metals, Method B (20 ppm).
1-(p-tolyl)prop-l-enyl)pyridine hydrochloride monohydrate.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Triprolidine Hydrochloride contains not less than 98.0 per
cent and not more than 101.0 per cent of C19HzzNz, HCl, Water (2.3.43).4.0 to 6.0 per cent, determined on 0.4 g.
calculated on the anhydrous basis. Assay. Weigh accurately about 0.25 g and dissolve in a mixture
Category. Histamine HI-receptor antagonist. of 50 ml of anhydrous glacial acetic acid and 0.5 ml of acetic
anhydride. Titrate with 0.1 M perchloric acid, using crystal
Dose. 10 to 20 mg daily, in divided doses.
violet solution as indicator. Carry out a blank titration.
Description. A white, crystalline powder; almost odourless.
1 ml of 0.1 M perchloric acid is equivalent to 0~01574 g of
Identification CI9HzzNz,HCl.
Storage. Store protected from light and moisture.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with triprolidine
hydrochloride RS or with the reference spectrum oftriprolidine
hydrochloride. Triprolidine Tablets
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Triprolidine Hydrochloride Tablets
0.001 per cent wIv solution shows absorption maxima at about
230 nm and 276 nm; absorbance at about 230 nm, about Triprolidine Tablets contain not less than 90.0 per cent and
0.50 and at about 276 nm, about 0.25. not more than 110.0 per cent ofthe stated amount oftriprolidine
hydrochloride, CI9HzzNz,HCl,HzO.
C. When examined in the range 230 nm to 360 nm (2.4.7), a
0.002 per cent w/v solution in 0.05 M sulphuric acid shows Usual strength. 10 mg.
an absorption maximum at about 290 nm; absorbance at about
290 nm, about 0.6. Identification
D. Dissolve 0.1 g in 2 ml of 2 M hydrochloric acid and add A. Extract a quantity of the powdered tablets containing
0.5 ml of potassium mercuri-iodide solution; a pale yellow 10 mg ofTriprolidine Hydrochloride with ether, filter, discard
precipitate is produced. the ether extract and dry the residue. Extract the residue with
E. A 5 per cent w/v solution gives the reactions of chlorides chloroform, filter and evaporate the filtrate to dryness. Add
(2.3.1). 0.1 ml of ether, stir and allow to evaporate.
On the residue, determine by infrared absorption
Tests spectrophotometry (2.4.6). Compare the spectrum with that
obtained with triprolidine hydrochloride RS or with the
Related substances. Determine by thin-layer chromatography
reference spectrum oftriprolidine hydrochloride.
(2.4.17), coating the plate with silica gel HF254.
B. In the test for Related substances, the principal spot in the
Mobile phase. A mixture of equal volumes of 2-butanone and
chromatogram obtained with the test solution corresponds to
dimethylformamide.
that in the chromatogram obtained with reference solution (b).
Testsolution. Dissolve 0.1 g of the substance underexami~
nation in 10 ml of methanol. Tests
Reference solution (a). Dissolve 10 mg ofthe substance under Related substances. Determine by thin-layer chromatography
examination in 100 ml of methanol. (2.4.17), coating the plate with silica gel HF254.

2268

www.webofpharma.com
IP 2010 TRISODIUM EDETATE CONCENTRATE FOR INJECTION

Mobile phase. A mixture of equal volumes of 2-butanone and Trisodium Edetate Concentrate for
dimethylformamide.
Injection
Test solution. Extract a quantity ofpowdered tablets containing
Trisodium Edetate Concentrate for Injection is a sterile
10 mg of Triprolidine Hydrochloride with methanol, filter,
solution in Water for Injections containing 20 per cent w/v
evaporate to dryness and dissolve the residue in 1 ml of
solution of trisodium edetate prepared by the interaction of
methanol.
Disodium Edetate and Sodium Hydroxide. It should be diluted
Reference solution (a). A 0.02 per cent w/v solution of with a suitable diluent in accordance with the manufacturer's
Z-triprolidine RS in methanol. instructions.
Trisodium Edetate Concentrate for Injection contains not less
Reference solution (b). A 1.0 per cent w/v solution of
triprolidine hydrochloride RS in methanol. than 19.0 per cent w/v and not more than 21.0 per cent w/v
solution oftrisodium edetate, CIOHI3N2Na30g.
Reference solution (c). A 0.01 per cent w/v solution of Category. Used in the treatment ofhypercalcaemia.
triprolidine hydrochloride RS in methanol.
Dose. By slow intravenous infusion, upto 70 mg per kg daily
Apply to the plate 5 JlI of each solution. After development, over 2 to 3 hours. For topical use in the eye, dilute 1 ml to 50 ml
dry the plate in air and examine in ultraviolet light at 254 nm. In . with Water for InjeGtion.
the chromatogram obtained with the test solution any spot
Description. A colourless solution.
corresponding to Z-triprolidine is not more intense than the
spot in the chromatogram obtained with reference solution (a) Identification
and any other secondary spot is not more intense than the
spot in the chromatogram obtained with reference solution A. Dissolve 2 g in 25 ml of water, add 6 ml of lead nitrate
(c). solution, shake and add 3 ml of potassium iodide solution;
no yellow precipitate is produced. Make alkaline to red litmus
Uniformity of content. (For tablets containing 10 mg or less). paper with 2 M ammonia and add 5 ml of ammonium oxalate
Comply with the test stated under Tablets. solution; no precipitate is produced.
Powder one tablet, weigh accurately a quantity ofthe powder B. Dissolve 0.5 gin 10 ml of water, add 0.5 ml ofa 10 percent
containing 7.5 mg ofTriprolidine Hydrochloride and carry out w/v solution of calcium chloride, make alkaline to red litmus
the Assay beginning at the words "add 15 ml of water....". paper with 2 M ammonia and add 3 ml of ammonium oxalate
solution; no precipitate is produced.
Calculate the content ofC,9H22N2,HCl,H20 in the tablet.
C. Evaporate to dryness and ignite. The residue gives the
Other tests. Comply with the tests stated under Tablets.
reactions of sodium salts (2.3.1).
Assay. Weigh and. powder 20 tablets. Weigh accurately a
Tests
quantity ofthe powder containing about 7.5 mg ofTriprolidine
Hydrochloride, add 15 ml of water and 1 g of sodium chloride, pH (2.4.24). 7.0 to 8.0.
shake for 2 to 3 minutes and add sufficient 5 M sodium
Pyrogens (2.2.8). Complies with the test, using per kg of the
hydroxide to make it alkaline. Extract with four quantities,
rabbit's weight 5 ml of a solution prepared in the following
each of 20 ml, of ether and wash the combined extracts with
manner. To 1 volume ofthe injection add 2.5 volumes of calcium
two quantities, each of5 ml, ofa mixture ofequal volumes ofa
gluconate solution. Dilute the resulting solution with
saturated solution of sodium chloride and water. Extract the
sufficient water for injection to give a final concentration of
ether solution with 20 ml of 0.1 M hydrochloric acid, wash
the ether with two quantities, each of 5 ml, of water and add 5.0 per cent w/v solution of trisodium edetate.
the washings to the acid extract. Heat on a water-bath for· Other tests. Complies with the tests stated under Parenteral
30 minutes, cool and add sufficient water to produce 50.0 ml. Pryparations (Concentrated Solutions for Injection)..
Dilute 10.0 ml to 100.0 nil with 0.1 M hydrochloric acid.
Assay. Dilute 10.0 ml to 100.0 ml with water and use this
Measure the absorbance of the resulting solution at the
solution to titrate a mixture of25.0 ml of 0.05 M magnesium
maximum at about 290 nm (2.4.7).
sulphate and 10 ml of ammonia bufferpH 1 0.9 using mordant
Calculate the content ofC19H22N2, HCl, H 20 taking 290 as the black II mixed triturate as indicator.
specific absorbance at 290 nm.
1 ml of 0.05 Mmagnesium sulphate is equivalentto 0.01791 g
Storage. Store protected from light and moisture. ofC,oHI3N2Na30g.

2269

www.webofpharma.com
TROPICAMIDE IP 2010

Storage. Store in hermetically-sealed, lead-free glass Reference solution (a). A 0.005 per cent w/v solution of the
containers. substance under examination in chloroform.
Labelling. The label states (1) the strength in terms of Reference solution (b).·A 0.002 per cent w/v solution of the
anhydrous trisodium edetate contained in a suitable dose- substance under examination in chloroform.
volume; (2) 'Trisodium Edetate Concentrate for Injection'; (3) Apply to the plate 20 Jll of each solution. After development,
that the solution must be dilutedwith either Sodium Chloride dry the plate in air and examine in ultraviolet light at 254 nm.
Intravenous Infusion or Dextrose Intravenous Infusion before Any secondary spot in the chromatogram obtained with the
administration. test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a) and not
more than one such spot is more intense than the spot in the
Tropicamide chromatogram obtained with reference solution (b).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 80° at a pressure not exceeding
0.7 kPa for 4 hours.
Assay. Weigh accurately about 0.5 g, dissolve in 80 ml of
anhydrous glacial acetic acid Titrate with 0.1 M perchloric
Mol. Wt. 284.4 acid, using 1-naphtholbenzein solution as indicator. Carry
Tropicamide is (RS)-N-ethyl-3-hydroxy-2-phenyl-N-(pyrid- out a blank titration.
4-ylmethyl)propionamide 1 ml of 0.1 M perchloric acid is equivalent to 0.02844 g of
Tropicamide contains not less than 99.0 per cent and not more C17HzoNzOz·
than 101.0 per cent of C17HzoNzOz, calculated on the dried Storage. Store protected from light and moisture.
basis.
Category. Mydriatic; cycloplegic.
Description. A white or almost white, crystalline powder; Tropicamide Eye Drops
odourless or almost odourless. Tropicamide Eye Drops are a sterile solution of Tropicamide
in Purified Water. They may contain stabilisers, suitable
Identification
antimicrobial agents and suitable substances to increase the
A. Determine by infrared absorption spectrophotometry (2.4.6). viscosity of the solution.
Compare the spectrum with that obtained with tropicamide
Tropicamide Eye Drops contain not less than 95.0 per cent
RS or with the reference spectrum of tropicamide.
and not more than 110.0 per cent of the stated amount of
B. When examined in the range 230 nm to 360 nm (2.4.7), a tropicamide, C17HzoNzOz.
0.003 per cent w/v solution in 0.1 M hydrochloric acid shows
Usual strengths. 0.5 per cent w/v; 1.0 per cent w/v.
an absorption maximum only at about 254 nm; absorbance at
about 254 nm, about 0.54. Identification
C. Dissolve 5 mg in 3 ml ofa mixture of9 ml ofacetic anhydride, A. Shake a volume containing 20 mg of Tropicamide with
I ml of 6 M acetic acid and 0.1 g of citric acid and heat on a 10 ml of chloroform, dry the chloroform layer over anhydrous
water-bath for 5 to 10 minutes; a reddish yellow colour is sodium sulphate, filter and evaporate the filtrate to dryness.
produced. Dissolve the residue in minimum quantity of chloroform, add
Tests dropwise to finely powdered potassium bromide JR, mix and
dry at 60°.
Related substances. Determine by thin-layer chromatography
On the residue determine by infrared absorption
(2.4.17), coating the plate with silica gel GF254.
spectrophotometry (2.4.6). Compare the spectrum with that
Mobile phase. A mixture of 95 volumes of chloroform, obtained with tropicamide RS or with the reference SPf~ctrum
S volumesofmethiiiiol-ana-O:YvolUiiie--ofstFiTiTg-ammonfa 6ftropiCamide.
solution.
B. When examined in the range 230nm to 360 nm (2.4.7), the
Test solution. Dissolve 0.1 g of the substance under exami- final solution obtained in the Assay shows an absorption
nation in 10 ml of chloroform. maximum at about 254 nm.

2270

www.webofpharma.com
IP 2010 TROXIDONE

Tests Troxidone contains not less than 98.0 per cent and not more
than 102.0 per cent ofC6H9N0 3, calculated on the dried basis.
pH (2.4.24). 4.0 to 5.8.
Category. Anticonvulsant.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Dose. For a child, 250 mg to 500 mg daily, in divided doses; for
an adult, I to 2 g daily, in divided doses.
Mobile phase. A mixture of 95 volumes of chloroform,
5 volumes of methanol and 0.5 volume of strong ammonia Description. Colourless or almost colourless crystals; odour,
solution. slightly camphoraceous.
Test solution. A volume ofthe eye drops containing 0.2 mg of
Identification
Tropicamide.
Reference solution (a). Dilute I volume of the eye drops to Test A may be omitted if tests Band C are carried out. Tests B
200 volumes with chloroform. and C may be omitted if test A is carried out.
Reference solution (b). Dilute I volume of the eye drops to A. Determine by infrared absorption spectrophotometry (2.4.6).
500 volumes with chloroform. Compare the spectrum with that obtained with troxidone RS.
Apply to the plate 20 III of each solution. After development, B. To 2 ml ofa 5.0 per cent w/v solution in carbon dioxide-free
dry the plate in air and examine in ultraviolet light at 254 nm. water (solution A) add I ml of barium hydroxide solution; a
Any secondary spot in the chromatogram obtained with the white precipitate is produced which dissolves on the addition
test solution is not more intense than the spot in the of 1 ml of 2 M hydrochloric acid.
chromatogram obtained with reference solution (a) and not
more than one such spot is more intense than the spot in the C. Dissolve 0.3 g in a mixture of 5 ml of ethanolic potassium
chromatogram obtained with reference solution (b). hydroxide solution and 5 ml of ethanol (95 per cent) and
allow to stand for 10 minutes. Add 0.05 rn1 ofphenolphthalein
Other tests. Comply with the tests stated under Eye Drops. solution and carefully add hydrochloric acid until the solution
Assay. To a volume containing about 30 mg ofTropicamide is neutral. Evaporate to dryness on a water-bath, shake the
add sufficient water to produce 100.0 ml. To 10.0 ml of the residue with four quantities, each of 5 ml, of ether, filter the
resulting solution add 2 ml of a 10 per cent w/v solution of combined ether extracts and evaporate the filtrate to dryness.
anhydrous sodium carbonate and extract with four quantities, The residue, aft.er recrystallisation from 5 ml of toluene and
each of20 ml, of chloroform. Wash the combined chloroform drying, melts at about 80° (2.4.21).
extracts with 25 ml of phosphate buffer pH 6.5. Wash the
aqueous layer with 10 ml of chloroform, combine the Tests
chloroform extracts and shake with four quantities, each of
Appearance of solution. Solution A is clear (2.4.1), and
20 ml, of 0.5 M sulphuric acid. Combine the acid extracts,
colourless (2.4.1).
dilute to 100.0 ml with 0.5 M sulphuric acid and measure the
absorbance of the resulting solution at the maximum at about Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of
254 nm (2.4.7). methyl red solution. Not more than 0.1 ml of 0.01 M
Calculate the content ofC17H2oN202 taking 172 as the specific hydrochloric acid or 0.01 M sodium hydroxide is required to
absorbance at 254 nm. change the colour of the solution.
Storage. Store iJ;l a refrigerator (8° to 15°). It should not be Heavy metals (2.3.13). 1.0 g complies with the limit test for
allowed to freeze. heavy metals, Method B (20 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.

Troxidone Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying over anhydrous silica gel for 6 hours.
Trimethadione Assay. Determine by gas chromatography(2.4.13).
H3 C ° Test solution. Weigh accurately 0.1 g of the substance under
H3 C}:T
o
O

CH 3
examination and dissolve in sufficient solutions prepared by
dissolving 0.125 g of 1-decanol (internal standard) in sufficient
ethanol to produce 25.0 ml (solution B).
CJf9N03 Mol. Wt. 143.1 Reference solution. Dissolve 0.1 g of troxidone RS in sufficient
Troxidone is 3,5,5-trimethyloxazolidine-2,4-dione. solution B to produce 10.0 rnl.

2271

www.webofpharma.com
TROXIDONE CAPSULES IP 2010

Chromatographic system Tests


- a stainless steel column 0.75 m x 3 mm, packed with
porous polymer beads (120 to 150 /lm), Other tests. Comply with the tests stated under Capsules.
- temperature: Assay. Determine by gas chromatography (2.4.13).
column 210°, Test solution. To a quantity of the contents of the capsules
inlet port at 240° and detector at 270°, containing about LOg ofTroxidone add 25 ml ofa 0.5 per cent
- flow rate. 30 ml per minute ofthe carrier gas. w/v solution of l-decanol (internal standard) in ethanol, shake
Inject 1 /ll of the test solution and the reference solution. for 30 minutes, add sufficient of internal standard solution to
produce 100.0 ml, mix and centrifuge. Use the supernatant
Calculate the content ofC6HgN0 3.
liquid.
Storage. Store protected from light and moisture. Reference solution. Dissolve 0.1 g of troxidone RS in sufficient
solution B to produce 10.0 ml.
Chromatographic system
a stainless steel column 0.75 m x 3 mm, packed with
Troxidone Capsules porous polymer beads (120 to 150 /lm),
- temperature:
Trimethadione Capsules column21Q0 ,
Troxidone Capsules contain not less than 95.0 per cent and inlet port at 240° and detector at 270° ,
not more than 105.0 per cent ofthe stated amount oftroxidone, - flow rate. 30 ml per minute oftIle carrier gas.
C6HgN03• Inject 1 /ll of the test solution and the reference solution.
Usual strength. 250 mg. Calculate the content of C6HgN0 3in the capsules.

Identification Storage. Store protected from moisture.

To a quantity ofthe contents of the capsules containing 1 g of


Troxidone add 25 mlof ether, set aside in a stoppered flask for
20 minutes, decant the ether through a filter, and ifan insoluble Tubocurarine Chloride
residue remains, digest it with another 10-ml portion of ether
as before, and filter into the first ether filtrate. Evaporate the
ether extracts to dryness with the aid of a current of air and
dry the residue at a pressure of2 kPa for 2 hours. The residue
complies with the following tests.
Test A may be omitted if tests Band C are carried out. Tests B
and C may be omitted if test A is carried out.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with .that obtained with troxidone RS
or with the reference spectrum oftroxidone.
B. To 2 ml ofa 5.0 per cent w/v solution in carbon dioxide-free
water (solution A) add 1 ml of barium hydroxide solution; a
white precipitate is produced which dissolves bnthe addition C37IillClNz06,HCl,5HzO Mol. Wt. 771.7
of 1 ml of 2 M hydrochloric acid. Tubocurarine Chloride is 7' ,12' -dihydroxy-6,6' -dimethoxy-
C. Dissolve 0.3 g in a mixture of 5 ml of ethanolic potassium 2,2' ,2' -trimethyltubocuraranium chloride hydrochloride
hydroxide solution and 5 ml of ethanol (95 pel~ cent) and pentahydrate.
allow to stand for 19 minutes. Add 0.05 ml ofphenolphthalein Tubocurarine Chloride contains riot less than 98.0 per cent
solution mid carefully add hydrochloric acid untilihe solution and not more than 102.0 per cent of C37H41CINz06,HCl,
is~neiltrar. . EvaliorateJQ ~dryIless •. ollJI. :Wflt(.)r::b.!lth,.sllake 1M calculated on the anhydrous-basis.
residue with four quantities, each Of 5. 1111, ()fet~el;, filter tl1e
combined ether extracts and evaporate the filtrate to dryness. Category. General anaesthetic.
The residue, after recrystallisationfrorn 5 ml of toluene and Dose. By intramuscular or intravenous injection, 100 to 300 /lg
drying, melts at about 80° (2.4.21). per kg body weight, total dose not exceeding 40 mg.

2272

www.webofpharma.com
IP 2010 TUBOCURARINE INJECTION

Description. A white or yellowish white, crystalline powder. Mobile phase. The lower layer of a mixture of equal volumes
of chloroform, methanol and a 12.5 pei cent w/v solution of
Identification trichloroacetic acid.
Test A may be omitted if tests B, C, D and E are carried out. Test solution. Dissolve 0.25 g of the substance under
Tests B, C and D may be omitted if tests A and E are carried examination in 10 ml ofwater.
out.
Reference solution (a). A 0.0375 per cent w/v solution ofthe
A. Determine by infrared absorption spectrophotometry (2.4.6). substance under examination in water.
Compare the spectrum with that obtained with tubocurarine
Reference solution (b). AO.01875 per cent w/v solution ofthe
chloride RS or with the reference spectrum of tubocurarine
substance under examination in water.
chloride.
Apply to the plate 5 J..lI of each solution. After development,
B. When examined in the range 230 nm to 360 nm (2.4.7), a
dry the plate in a current ofcold air and spray with a mixture of
0.005 per cent w/v solution shows an absorption maximum at
1 volume of potassium ferricyanide solution, 1 volume of
about 280 nm and a minimum at about 255 run, 0.56 to 0.62,
water and 2 volumes of ferric chloride solution, prepared
calculated on the anhydrous basis.
immediate,ly before use. Any secondary spot in the
C. To I ml of a 2.5 per cent w/v solution add 0.2 ml offerric chromatogram obtained with the test solution is not more
chloride solution and heat in a water-bath for I minute; a intense than the spot in the chromatogram obtained with
green colour is produced; 1 ml of water treated in the same reference solution (a) and not more than one such spot is
manner gives a brown colour. more intense than the spot in the chromatogram obtained
with reference solution (b).
D. To 20 ml of a 0.05 per cent w/v solution add 0.2 ml of
sulphuric acid and 2 ml of a I per cent w/v solution of Sulphated ash (2.3.18). Not more than 0.25 per cent, determined
potassium iodate, mix and warm on a water-bath for 30 minutes; onO.2g.
a yellow colour is produced.
Water (2.3.43).9.0 to 12.0 per cent, determined on 0.3 g.
E. Gives reaction A ofchlorides .and the reactions of allcaloids
Assay. Weigh accurately about 25 mg, dissolve in sufficient
(2.3.1).
water to produce 500.0 ml and measure the absorbance ofthe
resulting solution at the maximum at about 280 nm (2.4.7).
Tests
Calculate the content ofC37H4 1CIN20 6 ,HCI from the absorbance
Appearance of solution. Dissolve 0.5 g in sufficient carbon obtained by repeating the operation using 25 mg, accurately
dioxide-free water to produce 50.0 ml (solution A). Solution A weighed, oftubocurarine chloride RS in place ofthe substance
is clear (2.4.1), and not more intensely coloured than reference under examination.
solution YS6 (2.4.1).
Storage. Store protected from moisture.
pH (2.4.24). 4.0 to 6.0, determined in Solution A.
Specific optical rotation (2.4.22). +210° to +222°, determined
in solution A 3 hours after preparation. Tubocurarine Injection
Chloroform-soluble substances. Not more than 2 per cent,
Tubocurarine Chloride Injection
determined by the following method. Dissolve 0.25 gin 150 ml
of water contained in a separating funnel with a grease-free Tubocurarine Injection is a sterile solution of Tubocurarine
stopcock. Add 5 ml of a saturated solution of sodium Chloride in Water for Injections. It may contain suitable
bicarbonate and extract with three quantities, each of 20 ml, buffering agents. .
of chloroform. Wash the combined chloroform extracts with Tubocurarine Injection contains not less than 95.0 per cent
10 ml ofwater, filter the chloroform solution into a tared beaker and not more than 105.0 per cent of the stated amount of
and wash the filter with two successive quantities, each of tubocurarine chloride, C37~1 CIN20 6 ,HCI,5H20.
5 ml, of chloroform. Add the washings to the filtrate. Remove
the chloroform on a water-bath, dry the residue at 105° for Usual strength. 10 mg per ml.
I hour, cool and weigh. The residue does not dissolve in 10 ml Description. A colourless or faintly coloured solut\on.
of water but dissolves on the addition of 1 ml of 2 M
hydrochloric acid. Identification
Related substances. Determine by thin-layer chromatography A. Mix a volume containing 15 mg ofTubocurarine Chloride
(2.4.17), coating the plate with silica gel G. with 5 ml of acetone, evaporate the liquid at a pressure of

2273

www.webofpharma.com
TUBOCURARINE INJECTION IP 2010

2 kPa and add successive quantities of 2 ml of acetone, Reference solution (a). Dilute· 3 volumes of test solution to
evaporating each quantity at a pressure of 2 kPa until a dry 200 volumes with water.
residue is obtained.
Reference solution (b). Dilute I volume of reference solution
On the residue, determine by infrared absorption (a) to 2 volumes with water.
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with tubocurarine chloride RS or with the reference Apply to the plate 10 III of each solution. After development,
spectrum of tubocurarine chloride. dry the plate in a current ofcold air and spray with a mixture of
1 volume of potassium ferricyanide solution, 1 volume of
B. Dilute I ml to 30 ml with water. To 1 ml of the resulting
water and 2 volumes of ferric chloride solution, prepared
solution add 0.5 m1 of mercuric nitrate solution; a cherry-red
immediately before use. Any secondary spot in the
colour slowly develops.
chromatogram obtained with the test solution is not more
Tests intense than the spot in the chromatogram obtained with
reference solution (a) and not more than one such spot is
pH (2.424). 4.0 to 6.0. more intense than the spot in the chromatogram obtained
Optical rotation (2.422).+0.172° to +0.206° for each mg of with reference solution (b).
tubocurarine chloride, C37H41CIN206,HCI,5H20 per ml stated Other tests. Complies with the tests stated under Parenteral
on the label. Preparations (Injections).
Related substances. Determine by thin-layer chromatography
Assay. Dilute a volume containing about 50 mg ofTubocurarine
(2.4.17), coating the plate with silica gel G.
Chloride to 1000.0 m1 with water and measure the absorbance
Mobile phase. The lower layer of a mixture of equal volumes ofthe resulting solution at the maximum at about 280 nm (2.4.7).
of chloroform, methanol and a 12.5 per cent w/v solution of
trichloroacetic acid. Calculate the content OfC37~1 ClN20 6,HCI,5H20 taking 105 as
the specific absorbance at 280 nm.
Test solution. A volume of the injection containing 10 mg of
Tubocurarine Chloride diluted to 1 ml. Storage. Store protected from moisture.

2274

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

u
Undecenoic Acid 2277
Urea 2277
Urea Cream 2278
Urokinase 2279

2275

www.webofpharma.com
www.webofpharma.com
IP 2010 UREA

Undecenoic Acid Assay. Weigh accurately about 0.75 g, dissolve in lO ml of


ethanol (95 per cent) and titrate with 0.5 M sodium hydroxide
Undecylenic Acid using 0.1 ml of dilute phenolphthalein solution as indicator.
1 ml of 0.5 M sodium hydroxide is equivalent to 0.09214 g of
H
Cll H200 2.
H2C~COOH Storage. Store protected from light and moisture.

C,JI200 2 Mol. Wt. 184.3


Undecenoic Acid is 10-undecenoic acid.
Urea
Undecenoic Acid contains not less than 97.0 per cent and not
more than 102.0 per cent ofC ll H200 2 •
Category. Antifungal (topical).
Description. A white or very pale yellow, crystalline mass or
colourless or pale yellow liquid; odour, characteristic.
CHiN20 Mol. Wt. 60.1
Identification Urea is the diamide ofcarbonic acid.
A. Dissolve 0.1 g in a mixture of2 ml of 1 M sulphuric acid and Urea contains not less than 99.0 per cent and not more than
5 ml of glacial acetic acid and add dropwise 0.25 mlof 101.0 per cent ofCH4N 20, calculated on the dried basis.
potassium permanganate solution; the colour of the Category. Keratolytic.
permanganate solution is discharged.
Dose. 5 to 15 g.
B. Boil 2 g under a reflux condenser with 2 ml offreshly distilled
aniline for 10 minutes, allow to cool, add 30 ml of ether and Description. A white, crystalline powder or transparent
extract with three quantities, each of20 ml, of2 Mhydrochloric crystals; odourless or almost odourless, but may gradually
acid and then with 20 ml of water. Evaporate the organic layer develop a slight odour of ammonia upon long standing;
to dryness on a water-bath. The residue, after recrystallising slightly hygroscopic.
twice from ethanol (70 per cent) and drying over phosphorus
pentoxide at a pressure of 1.5 to 2.5 kPa for 3 hours melts at Identification
66° to 68°. Test A may be omitted iftests Band C are carried out. Tests B
and C may be omitted if test A is carried out.
Tests
A. Determine by infrared absorption spectrophotometry (2.4.6):
Congealing range (2.4.10). 21°to 24°. Compare the spectrum with that obtained with urea RS or
Refractive index (2.4.27). 1.447 to 1.450. with the reference spectrum of urea.

Peroxide value (2.3.35). Not more than 10. B. Heat 0.5 g in a test-tube; itliquefies, and ammonia is evolved
which is recognised by its characteristic odour. Heat further
Iodine value (2.3.28). 131 to 140. until the liquid is turbid, cool and dissolve in 10 ml of water.
Fixed and mineral oils. Boil 1.0 g with 25 ml ofwater and 5 ml Add 1 ml of a 10 per cent w/v solution of sodium hydroxide
of sodium carbonate solution for 3 minutes.· The hot solution and 0.05 ml of coppersulphate solution; a reddish violet colour
is not more opalescent than opalescence standard OS2 (2.4.1). is produced.
Water-soluble acids. Shake 2.0 g with 20 ml ofwarm water, C. Dissolve 0.1 gin 1 ml of water and add 1 ml of nitric acid;
allow to separate and filter .the aqueous layer through a a white, crystalline precipitate is produced.
moistened filter paper. To 5 ml of the filtrate add 0.01 mlof
dilute phenolphthalein solution. Not more than 0.1 mlof Tests
0.1 M sodium hydroxide is required to change the colour of
Appearance of solution. Solution A is clear (2.4.1), and
the solution.
colourless (2.4.1).
Heavy metals (2.3.13). 2.0 g complies with the limit test for
Alkalinity. To 10 ml ofa 5.0 per cent w/v solution (solution A)
heavy metals, Method B (l0 ppm).
add 0.1 ml of methyl red solution and 0.4 ml of 0.01 M
Sulphated ash (2.3.18). Not more than 0.15 percent. hydrochloric acid; the resulting solution is red to orange.

2277

www.webofpharma.com
UREA IP 2010

Biuret. Not more than 0.1 per cent, determined by the following a mixture of 99 volumes of ethanol and 1 volume of strong
method. To 10 rnl of a 20 per cent w/v solution add 5 ml of ammonia solution.
water, 0.5 m1 ofa 0.5 per cent w/v solution of copper sulphate Test solution. Disperse with heating a quantity of the cream
and 0.5 ml of 10Msodium hydroxide and allow to stand for containing 50 mg of Urea in 1 m1 of water, cool, add 4 ml of
5 minutes. Any reddish violet colour obtained is not more acetone, mix and filter.
intense than that in a standard prepared at the same time and
in the same manner using lOrnl ofaO.02 percentw/v solution Referenc'e solution (a). Dissolve 50 mg of urea RS in 1 ml of
of biuret in place of the substance under examination. water and add 4 ml of acetone.

Ethanol-insoluble matter. Not more than 0.04 per cent, Reference solution (b). A mixture of equal volumes ofthe test
solution and reference solution (a).
determined by the following method. Dissolve 5.0 g in 50 ml of
warm ethanol (95 per cent), filter through a tared filter, wash Apply to the plate 10 ILl of each solution. After development,
the filter with 20 ml ofwarm ethanol (95 per cent) and dry at dry the plate in air, spray with a solution containing 0.5 per
105° for 1 hour. cent w/v solution of 4-dimethylaminobenzaldehyde and
0.5 per cent v/v of sulphuric acid in ethanol. The principal
Heavy metals (2.3.13). Dissolve 1.0 g in 20 rnl ofwater and 5 rnl
spot in the chromatogram obtained with the test solution
of 0.1 M hydrQchloric acid. The solution complies with the
corresponds to that in the chromatogram obtained with
limit test for heavy metals, Method A (20 ppm).
reference solution (a). The chromatogram obtained with
Sulphated ash (2.3.18). Not more than 0.1 percent. reference solution (b) shows a single, compact spot.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined B. To a quantity containing 0.1 g of Urea add 50 ml of water
on 1.0 g by drying in an oven at 105° for 1 hour. and heat until dispersed, cool in ice and filter through glass
Assay. Weigh accurately about 0.5 g, dissolve in sufficient of wool. Adjust the pH to 6.0 to 7.0 using 0.1 M hydrochloric
a 10 per cent v/v solution of sulphuric acid to produce acid or 0.1 M sodium hydroxide as necessary. To 5 ml add
100.0 ml and mix. Place 5.0 ml of the resulting solution in a 5 ml ofa 0.1 per cent w/v suspension of urease-active meal
long-necked flask, add 10 rnl ofsulphuric acid and heat gently and allow to stand for 30 minutes in a stoppered flask at 37°.
until evolution ofgas ceases. Boil gently for 10 minutes, cool, When the resulting solution is heated in a water-bath a vapour
cautiously add 40 ml of water, cool again and place in a steam- is produced that turns moist red litmus paper blue.
distillation apparatus. Add 50 ml of 10Msodium hydroxide
Tests
and distil immediately by passing steam through the mixture.
Continue the distillation for 1 hour, collecting the distillate in Other tests. Complies with the tests stated under Creams.
40 ml of a 4 per cent w/v solution of boric acid. Titrate with Assay. Weigh accurately a quantity containing about 42 mg
0.1 M hydrochloric acid, using 0.25 ml of methyl red- ofUrea, shake with 150 rnl ofhot water for 20 minutes, allow to
methylene blue solution as indicator. Carry out a blank cool and dilute to 500.0 ml with water. Filter through a fine
titration. glass microfibrefilter paper or by any other means, transfer
1 ml of 0.1 Mhydrochloric acid is equivalent to 0.003003 g of 1.0 ml ofthe filtrate to a 100-rnl volumetric flask, add 2 rnl ofa
c:H4NzO. 0.1 per cent w/v suspension of urease-active meal, stopper
the flask and allow to stand for 15 minutes at 37°. Immediately
Storage. Store protected from moisture.
add 25 ml of a solution containing 12 g of sodium salicylate
and 0.24 g of sodium nitroprusside in 200 ml and 25 ml of a
solution prepared by diluting a volume of sodium hypochlorite
Urea Cream solution containing 0.66 g of available chlorine with 0.2 M
Urea Cream contains Urea in a suitable basis. sodium hydroxide to 1000 ml. Mix well, allow to stand at 37°
for 5 minutes and dilute to 100.0 m1 with water. Measure the
Urea Cream contains not less than 90.0 per cent and not more absorbance of the resulting solution at the maximum at about
than 110.0 per cent ofthe stated amount of urea, CH4N zO. 665 nm (2.4.17), using as the blank a solution prepared in the
Usualstrength. 10 per cent w/w. same manner but using 1~ 0 rnl of water in place ofl.0 ml ofthe
filtrate.
Identification
Calculate the content ofCH4N zO from the absorbance obtained
A. Deferrn.ille by tllill-layer C:1l.I'ornatograplly (2.4:17), coatlng by llsiJ.ig 42 IIlg of urea RS in place of the sllbstan-ce-Ui.ii:lef
the plate with silica gel G exa.mina.tion.
Mobile phase. For the first development use 2,2,4-trimethyl- Storage. Store in accordance with the instructions of the
pentane. Dry the plate in air. For the second development use manufacturer.

2278

www.webofpharma.com
IP 2010 UROKINASE

Urokinase Apply 1 ml ofthe test solution to the column, rinse twice with
. O.5-ml portions of the buffer and then carry out the elution.
Urokinase is an enzyme, obtained from human urine that The eluate may be collected in I-ml :Ii-actions. Plot the individual
activates plasminogen. It consists ofa mixture oflow molecular absorbances on a graph. Draw perpendicular lines towards
weight and high molecular weight forms, the high molecular the axis of the abscissae from the minima before the high
weight form being predominant. The molecular weights ofthe molecular weight peak, between the high and the low molecular
low and high molecular weight forms are 33,000 and 54,000 weight peaks, and after the low molecular weight peak.
respectively. Combine separately the high and low molecular weight
It is prepared in conditions designed to minimise microbial fractions and for each combined fraction determine the activity
and viral contamination. In particular, adequate measures, such by the method described under Assay. The ratio ofthe activity
as heating the substance in solution at 60° for 10 hours, are in the combined high molecular weight fraction to that in the
taken to inactivate viruses. combined low molecular weight fraction is not less than 2.0.
Total protein. Determine by Method C for the determination
Urokinase contains not less than 70,000 Units of urokinase
of nitrogen (2.3.30), using 10 mg of the substance under
activity per mg of protein.
examination and multiplying the result by 6.25 to obtain the
Category. Fibrinolytic enzyme. content of protein.
Dose. By instillation into arteriovenous shunt and for Hepatitis-B surface antigen. Examine by a suitably sensitive
intraocular administration, 5000 to 37,500 Units in 2 to 3 ml of method such as radio-immunoassay; hepatitis-B surface
Sodium Chloride Injection. antigen is not detected.
Description. A white or almost white, amorphous powder. Abnormal toxicity (2.2.1). Complies with the test, using a
solution containing 2,500 Units in 0.5 ml of saline solution.
Identification Thromboplastic contaminants. Dissolve suitable quantities
of the substance under examination in barbitone bziffer
A. Place 0.5 ml ofcitrated human plasma and 0.5 ml ofcitrated pH 7.4 to obtain solutions containing 5000, 2500, 1250, 625
bovine plasma in two separate haemolysis tubes maintained and 312 Units per ml. Into each of six haemolysis tubes, 1 cm
in a water-bath at 37°. To each tube add 0.1 ml ofa solution of in internal diameter, place 0.1 ml ofcitrated rabbit plasma. Add
the substance under examination containing 1000 Units per 0.1 ml of one of each of the solutions of the substance under
ml in phosphate buffer pH 7.4 and 0.1 ml of a solution of examination to each offive ofthe tubes and 0.1 ml of barbitone
thrombin containing 20 Units per ml in phosphate buffer bziffer pH 7.4 to the sixth (control). Incubate the six tubes at
pH 7.4 and shake immediately; in both tubes, a clot forms and 25°± O,SOfor 5 minutes and then add 0.1 ml ofa 0.3675 percent
lyses within 30 minutes. w/v solution of calcium chloride. Using a stop-watch,
B. Carry out a suitable immunodiffusion test. measure the clotting time for each solution and the control.
Plot the shortening ofthe recalcification time (control clotting
Tests time minus clotting time for each solution) against log
concentration. Extrapolate the best-fitting straight line through
Appearance ofsolution.AO.1 percentw/v solution in water is the five points until it reaches the log concentration axis. The
clear (2.4.1), and colourless (2.4.1). urokinase activity at the intersection point represents the limit
concentration for clotting activity (zero clotting activity). The
Molecular fractions. Determine by size-exclusion
zero clotting activity is not less than 150 Units per ml.
chromatography (2.4.16).
Vasoactive substances. Anesthetise a rabbit by intraperitoneal
Test solution. Dissolve 0.1 g of the substance under exami-
injection of 0.15 g of phenobarbitone sodium per kg of body
nation in 100 ml of 0.02 M phosphate buffer pH 8.0.
weight. Dissolve in normal saline solution a sufficient
Chromatographic system quantity ofthe substance under examination to give a solution
a column 90 cm x 16 mm, packed with a cross-linked containing 40,000 Units per m!. Administer by intravenous
dextran suitable for fractionation ofproteins in the range infusion at a rate of 1 ml per minute a sufficient volume ofthe
of molecular weight from 4000 to 150,000 (such as solution of the substance under examination such that the
Sephadex 0-100), at 5°, dose is 20,000 Units per kg ofbody weight. Measure the arterial
- mobile phase: 1.75 per cent w/v solution of sodium pressure and heart rate at intervals of 15 minutes for 5 hours
chloride in 0.02 M phosphate bziffer pH 8.0, after the infusion. No significant and lasting alterations in
- flow rate. 6 ml per hour, arterial pressure or heart rate are produced, except those arising
- spectrophotometer set at 280 nm. from the effects of the anaesthetic.

2279

www.webofpharma.com
UROKJNASE IP 2010

Assay. The potency ofurokinase is determined by comparing containing 3 per cent w/v solution of bovine albumin and
its ability to activate human plasminogen to form plasmin with 0.1 ml of a solution containing 20 Units of thrombin per ml.
that of the Standard Preparation. The plasmin generated is Place the tubes in a water-bath at 37° and allow to stand for
determined by measurement of the time taken to lyse a fibrin 2 minutes to attain temperature equilibrium. Using an automatic
clot under the conditions of a suitable method of Assay. pipette, introduce into the bottom ofthe first tube 0.5 ml of a
1.0 per cent w/v solution of bovine euglobulin fraction
Standard Preparation
ensuring mixing. At 5-second intervals introduce successively
The Standard Preparation is the 1st International Reference into the remaining tubes 0.5 ml ofa 1.0 per cent w/v solution of
Preparation for Urokinase, human, established in 1968, bovine euglobulin fraction. Using a stop-watch, measure for
consisting of partially purified freeze-dried urokinase from each tube the time in seconds that elapses between the
human urine with lactose (supplied in ampoules containing addition of the euglobulin and the lysis of the clot.
4800 Units ofurokinase activity) or another suitable preparation
Using the logarithms of the lysis times, calculate the result of
the activity of which has been determined in relation to the
the assay by standard statistical methods.
International Reference Preparation.
The estimated potency is not less than 90.0 per cent and not
Method more than 111.0 per cent of the stated potency.
Unless otherwise prescribed, use phosphate btiffer pH 7.4 The fiducial limits oferror are not less than 80 per cent and not
containing 3 per cent w/v solution of bovine albumin for the more than 125 per cent of the stated potency.
preparation of solutions and dilutions.
Urokinase intendedfor use in the manufacture ofparenteral
Prepare a solution of the Standard Preparation containing preparations or ophthalmic preparations complies with the
1000 Units ofurokinase activity per ml and prepare a solution following additional requirements.
of the preparation under examination expected to have the
Bacterial endotoxins (2.2.3). Not more than 0.002 Endotoxin
same concentration; keep the solutions in ice and use within
Unit per Unit ofurokinase activity.
6 hours. Prepare three 1.5-fold serial dilutions ofthe solution
ofthe Standard Preparation so that the longest clot-lysis time Sterility (2.2.11). Complies with the test for sterility.
is less than 20 minutes and the shortest clot-lysis time is greater Storage. Store protected from light in a refrigerator (2° to 8°).
than 3 minutes. Prepare three similar dilutions ofthe solution The containers should be sterile, tamper-evident and sealed
of the preparation under examination. Keep the solutions in
so as to exclude micro-organisms.
ice and use within 1 hour. Using 24 tubes 8 mm in diameter,
label the tubes SJ, S2, S3 for the dilutions of the Standard Labelling. The label states (1) the number ofUnits ofuroldnase
Preparation and T I, T2, T3for the dilutions of the preparation activity in the container; (2) the number ofUnits ofuroldnase
urider examination, allocating four tubes to each dilution. Place activity per mg of protein; (3) the storage conditions; (4)
the tubes in ice. Into each tube introduce 0.2 ml of the whether or not it is intended for use in the manufacture of
appropriate dilution, 0.2 ml of phosphate btiffer pH 7.4 parenteral preparations or ophthalmic preparations.

2280

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

v
Valproate Injection 2283
Valproic acid 2283
Valproic Acid Capsules 2284
ValproicAcid Oral Solution 2285
Valsartan 2286
Valsartan Tablets 2287
Valsmian and Hydrochlorothiazide Tablets 2287
Vancomycin Hydrochloride 2289
Vancomycin Capsules 2291
Vancomycin Intravenous Infusion 2291
Vancomycin Oral Solution 2292
Vanillin 2294
Vasopressin Injection 2294
Verapamil Hydrochloride 2295
Verapamil Injection 2296
Verapamil Tablets 2297
Vmblastine Sulphate 2298
Vmblastine Injection 2299
Vmcristine Sulphate 2300
Vmcristine Injection 2302
Vmorelbine Tartrate 2303
Vmorelbine Injection 2304
Vitamin A Concentrate Oil 2305
Vitamin A Concentrate Powder 2306
Water-Miscible Vitamin AConcentrate 2306
Vitamins A and D Capsules 2307
ConcentratedVitamin D Solution 2308
Concentrated Vitamins A and D Solution 2308

2281

www.webofpharma.com
www.webofpharma.com
IP 2010 VALPROIC ACID

Valproate Injection Inject 2111 ofreference solution (b). The test in not valid unless
the resolution between the peaks due to valproic acid and
Valproate Sodium Injection biphenyl peaks is not less than 3.0 and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
Valproate Injection is a sterile solution of sodium valproate in
Water for Injections with one or more suitable buffering or Inject 2 III of the test solution and reference solution (a).
sequestering agents.
Calculate the content of C SH I6 0 2 in the injection.
Valproate Injection contains not less than 90.0 per cent and
Storage. Store at a temperature not exceeding 30° and in single
not more than 110.0 per cent ofthe stated amount ofvalproic
dose containers, preferably of Type 1 glass.
acid, C SH I6 0 2•
Labelling. The label states the amount of any buffering or
Usual strength. 20 mg per ml.
sequestering agents used.

Identification
A. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to that in the
Valproic Acid
chromatogram obtained with the reference solution.
B. Gives the reaction of sodium salts (2.3.1).

Tests
pH (2.4.24).7.0 to 9.0. Mol. Wt. 144.2
Bacterial endotoxins (2.2.3). Not more than 23.0 Endotoxin Valproic acid is 2-propylpentanoic acid.
Units per ml.
Valproic acid contains not less than 99.0 per cent and not
Sterility (2.2.11). Complies with the test for sterility. more than 101.0 percent of C SH I6 0 2 •
Other tests. Complies with the tests stated under Parenteral Category. Anticonvulsant.
Preparations (Injections).
Description. A colourless or very slightly yellow liquid, slightly
Assay. Determine by gas chromatography (2.4.13). viscous.
Internal standard solution. A 0.5 per cent w/v solution of
biphenyl in dichloromethane. Identification
Test solution. Dilute a volume ofthe injection containing about Test A may be omitted if tests Band C are carried out. Tests B
400 mg ofvalproic acid with 20 ml of5 per cent v/v solution of and C may be omitted if test A is carried out.
hydrochloric acid and add 50 ml ofinternal standard solution. A. Determine by infrared absorption spectrophotometry (2.4.6).
Shake for 1 hour, ifemulsion fonus, stir with a glass rod. Take Compare the spectrum with that obtained with valproic acid
5 ml of the organic layer and dilute to 50 ml with RS or with the reference spectrum ofva1proic acid.
dichloromethane.
B. Detenuine by thin-layer chromatography (2.4.17), coating
Reference solution (a). A 0.8 per cent w/v solution ofvalproic the plate with silica gel G.
acid RS in internal standard solution.
Mobile phase. A mixture of50 volumes of ether and 50 volumes
Reference solution (b). Dilute 5.0 ml ofreference solution (a) of dichloromethane.
to 50 m1 with dichloromethane.
Test solution. A 1.0 per cent w/v solution of the substance
Chromatographic system
under examination in methanol.
- a glass column 1.8 m x 2 mm, packed with 10 per cent
phase G34 on 80 to 100 mesh support S lA, Reference solution. A 1.0 per cent w/v solution of valproic
temperature: acid RS in methanol.
column. 155°, Apply 2 III ofeach solution. Allow the mobile phase to rise 15
injection port at 275° and detector at 300°, cm. Dry the plate in air and spray with bromocresol green
- a flame ionization detector, solution. The principal spot in the chromatogram obtained
- flow rate. 20 ml per minute using nitrogen as the carrier with the test solution corresponds to that in the chromatogram
gas. obtained with the reference solution.

2283

www.webofpharma.com
VALPROIC ACID IP 2010

C. To 1 ml add 3.0 m1 ofdilute sodium hydroxide solution. Add Sulphated ash (2.3.18). Not more than 0.1 per cent.
3.0 ml of water and 1.0 m1 of a 10 per cent w/v solution of
Assay. Weigh accurately about O.lg and dissolve in 25 m! of
cobalt nitrate. A violet precipitate is formed, filter it. The
ethanol (95 per cent). Add 2 ml of water. Titrate with 0.1 M
precipitate dissolves in di~hloromethane.
sodium hydroxide, determining the end-point
Tests potentiometrically (2.4.25). Carry out a blank titration.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01442 g of
Appearance of solution. A20.0 per cent w/v solution in dilute
CgH I60 2 •
sodium hydroxide is clear (2.4.1) and not more intensely
coloured than reference solution YS5 (2.4.1). Storage. Store protected from moisture.

Refractive index (2.4.27). 1.422 to 1.425.


Related substances. Determine by gas chromatography
(2.4.13).
Valproic Acid Capsules
Valproic Acid Capsules contain not less than 90.0 per cent
Internal standard solution. A 0.005 per cent w/v solution of
and not more than 110.0 per cent of the stated amount of
butyric acid in heptane.
valproic acid, CgH I6 0 2 •
Test solution. A 5.0 per cent w/v solution of the substance
Usual strength. 250 mg.
under examination in the internal standard solution. Dilute 1.0
ml ofthis solution to 10 ml with heptane. Identification
Reference solution. Dissolve 20 mg of the substance under
A. In the Assay, the retention time ratio of the principal peak
examination and 20 mg of 2-(l-methylethyl)pentanoic acid
to the internal standard peak in the chromatogram obtained
RS (valproic acid impurity C RS) in 10 ml of heptane. Dilute
with the test solution and reference solution, are not more
1.0 ml ofthis solution to 10 ml with heptane.
than 2.0 per cent.
Chromatographic system
B. Disperse a quantity of the content of capsules containing
- a fused-silica capillary column 30 m x 0.53 mm, coated
about 250 mg of valproic acid, add 20 ml of 1 M sodium
with wide-bore rnacrogol20000 2-nitroterephthalate (0.5
hydroxide, shake and allow the layers to separate. Filter the
11m),
aqueous layer, add 4 ml of hydrochloric acid, mix and extract
- temperature.
with 40 ml of n-heptane. Filter the n-heptane layer through
column 130° from 0-10 minutes and 130° - 190° from 10-
glass wool and evaporate the solvent completely on a steam
30 minutes
bath with the aid of a current of air. Transfer 2 drops of the
- inlet port and detector 220°,
residue to a test tube containing 0.5 ml each of2 per cent v/v
- a flame ionisation detector,
ofpotassiunm iodide solution and 4 per cent v/v ofpotassium
- flow rate. 8 ml per minute ofthe carrier gas (helium).
iodate solution and mix, a yellow colour is produced.
Inject I III ofthe reference solution. The test is not valid unless
the resolution between the peaks due to valproic acid impurity Tests
C and valproic acid is not less than 3.0.
Dissolution (2.5.2).
Inject I III of the test solution and the reference solution. In Apparatus No. I,
the chromatogram obtained with the test solution the area of Medium. 900 ml ofa 0.5 per cent w/v solution ofsodium lauryl
peak due to any impurity, for each impurity is not more than sulphate in simulated intestinal fluid prepared by dissolving
0.1 times the area ofthe peak due to the internal standard (0.1 6.8 g of monobasic sodium phosphate in 250 ml water, mix and
per cent). The sum of areas of all the secondary peaks is not 77 ml of 0.2 M sodium hydroxide, add 500 ml of water. Adjust
more than 0.3 times the area of the peak due to the internal the pH to 6.8 with 0.2 M sodium hydroxide or 0.2 M
standard (0.3 per cent). Ignore any peak with an area less than hydrochloric acid dilute to 1000 ml with water adjust to pH
0.01 times the area of the peak due to the internal standard 7.5 with 5 M sodium hydroxide,
(0.01 percent).
Speed and time. 50 rpm and 60 minutes.
Heavy metals (2.3.13). Dissolve 2.0 g in 20 ml ethanol (80per
Withdraw a suitable volume ofthe medium and filter.
c::enJL!!'DJ1.J!~e.J 2.ml of.Jhes.olutioD...J'he.f.e.s.uhings.olution
complies with the limit test for heavy metals, Method D (20 DeterrlJ.ille by gas cll1:omat()grapl1y (2.4.13) as described under
ppm). Prepare the standard using lead standard solution (2 Assay, using the following solutions.
ppm Pb) obtained by diluting lead standard solution (100 Test solution. To 10 ml ofthe filtrate, add about 3.0 g ofsodium
ppm Pb) with ethanol (80 per cent). chloride, and mix on a vortex mixer for 5 minutes. Add about

2284

www.webofpharma.com
IP 2010 VALPROIC ACID ORAL SOLUTION

1.0 ml of6 M hydrochloric acid and 5.0 ml ofintemal standard Valproic Acid Oral Solution
solution, shake and allow to separate, remove the n-heptane
layer and filter. Discard the aqueous layer. Valproic Acid Oral Solution contains not less than 90.0 per
cent and not more than 110.0 per cent of the stated amount of
Reference solution. Weigh accurately a quantity of valproic
valproic acid, CgH I6 0 2 •
acid RS and dilute with the dissolution medium to obtain a
solution having a concentration similar to that of the test Usual strengths. 40 mg per ml; 50 mg per ml.
solution. Take 10 ml of this solution and proceed as in test
solution beginning with the words "add about 3.0 g........". Identification
Calculate the content ofC gH I6 0 2 in the capsule. A. In the Assay, the retention time ratio of the principal peak
D. Not less than 85 per cent of the stated amount ofC gH 160 2• to the internal standard peak in the chromatogram obtained
with the test solution and reference solution is not more than
Other tests. Comply with the tests stated under Capsules.
2.0 per cent.
Assay. Detennine by gas chromatography (2.4.13).
B. Dissolve a volume of solution containing about 250 mg of .
Test solution. Take 20 capsules in a container and add 150 ml valproic acid in 40 ml of water and 2.0 ml of hydrochloric
of dichloromethane, cool in a solid carbon dioxide-acetone acid. Mix and extract with 40 ml of n-heptane. Filter the n-
mixture until the contents have solidified. If necessary, heptane layer through glass wool into a beaker, and evaporate
transfer the mixture of capsules and dichloromethane to a the solvent completely on a steam bath with the aid ofa current
blender jar and blend until all the solids are reduced to fine of air. Transfer 2 drops ofthe residue to a test tube containing
particles. Transfer the mixture to a 500-ml flask, add n-heptane 0.5 ml each 2 per cent v/v ofpotassium iodide solution and 4
to volume, mix and allow the solids to settle. Transfer an per cent v/v solution ofpotassium iodate solution and mix; a
accurately measured volume ofthis solution, containing about yellow colour is produced.
250 mg ofvalproic acid, to a 100 ml flask, dilute with n-heptane
to volume and mix. Transfer 5.0 ml to a container equipped Tests
with a closure. Add 2.0 ml of internal standard solution, close
the container and mix. pH (2.4.24).7.0 to 8.0.
Reference solution. A 0.25 per cent w/v solution of valproic Other tests. Complies with the tests stated under Oral Liquids.
acid RS in n-heptane. Transfer 5.0 ml to a container equipped
Assay. Detennine by gas chromatography (2.4.13).
with a closure. Add 2.0 ml of internal standard solution, close
the container and mix. Test solution. Dissolve a volume of solution containing about
250 mg of valproic acid in 40 ml of water, add 2.0 ml of
Internal standard solution. A 0.5 per cent w/v solution of
hydrochloric acid and extract with 80 ml of n-heptane until
biphenyl in n-heptane.
the aqueous layer is clear. Filter the n-heptane layer through
Chromatographic system glass wool with small portions of n-heptane, add the rinsings
a glass column 1.8 rn x 2mm, packed with 10 per cent to the flask, dilute to 100.0 ml with n-heptane and mix.. Transfer
phase 034 on 80 to 100 mesh support SlA, 5.0 ml to a container equipped with a closure. Add 2.0 ml ofthe
temperature: internal standard solution, close the container and mix.
column. 150°,
injection port and detector. 250°, Reference solution. A 0.25 per cent w/v solution of valproic
a flame ionization detector, acid RS in n-heptane. Transfer 5.0 ml to a container equipped
flow rate. 40 ml per minute using nitrogen as carrier gas. with a closure. Add 2.0 ml of internal standard solution and
mix.
Inject 2 f.Ll ofthe reference solution. The test is not valid unless
the resolution between the peak due to valproic acid and Internal standard solution. A 0.5 per cent w/v solution of
biphenyl is not less than 3.0 and the relative standard deviation biphenyl in n-heptane.
for replicate injections is not more than 2.0 per cent. The Chromatographic system
relative retention time with reference to valproic acid for glass column 1.8 m x 2 mrn, packed with 10 per cent
biphenyl is about 2.0. phase 034 on 80 to 1010 mesh support SlA,
Inject 2 f.Ll of the test solution and the reference solution. temperature:
column. 150°,
Calculate the content of C gH I6 0 2 in the capsules. injection port and detector at 250°,
Storage. Store protect from moisture, at a temperature not a flame ionization detector,
exceeding 30°. flow rate. 40 ml per minute using nitrogen as carrier gas.

2285

www.webofpharma.com
VALSARTAN IP 2010

Inject 2 III ofthe reference solution. The test is not valid unless Test solution. Dissolve 50 mg of the substance under
the resolution between valproic acid and biphenyl is not less examination in 100.0 ml ofthe mobile phase.
than 3.0 and the relative standard deviation for replicate
Reference solution (a). A 0.05 per cent w/v solution of
injections is not more than 2.0 per cent. The relative retention
valsartan RS in the mobile phase.
time with reference to valproic acid for biphenyl is about 2.0.
Reterence solution (b). Dilute 1 ml ofreference solution (a) to
Inject 2 III each ofthe test solution and the reference solution.
100 ml with the mobile phase.
Calculate the content of C SH I6 0 2in oral solution.
Chromatographic system
Storage. Store protected from moisture. a stainless steel column 12.5 cm x 3.0 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm) (Such
as Nucleosil),
Valsartan mobile phase: a mixture of 50 volumes of water, 50
volumes of acetonitrile and 0.1 volume of glacial
acetic acid,
. H3 C CH 3

H3C~NXCOOH f'J=tj
flow rate. 0.4 ml per minute,
spectrophotometer set at 225 nm,
i~ection volume. 10 Ill.

~ HN ..-;N Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0.
Inject the test solution and reference solution (b). Run the
chromatogram three times the retention time of the principal
peak. In the chromatogram obtained with the test solution,
Mol. Wt. 435.5 the area of any secondary peak is not more than 0.2 times the
area ofthe peak in the chromatogram obtained with reference
Valsartan is N-pentanonyl-N-[2' -(lH-tetrazol-5-yl)biphenyl- solution (b) (0.2 per cent) and the sum of areas of all the
4-ylmethyl]-L-valine. secondary peaks is not more than 0.5 times the area of the
Valsartan contains not less than 98.0 per cent and not more peak in the chromatogram obtained with reference solution
than 102.0 per cent ofC24H29Ns03, calculated on the anhydrous (b)(0.5 per cent).
basis.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Category. Antihypertensive. heavy metals, Method B (20 ppm).
Dose. 80-160 mg daily.
Sulphated ash (2.3.18). Not more than 0.1 percent.
Description. A white to almost white powder.
Water (2.3.43). Not more than 2.0 per cent, detennined on 1 g.
Identification
Assay. Determine by liquid chromatography (2.4.14).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Test solution. Dissolve 25 mg of the substance under
Compare the spectrum with that obtained with valsartan RS
examination in 50.0 ml ofthe mobile phase. Dilute 5.0 ml ofthe
or with the reference spectrum of vaIsartan.
solution to 50.0 ml with the mobile phase.
B. In the Assay, the principal peak in the chromatogram
Reference solution. A 0.005 per cent w/v solution ofvalsal'tan
obtained with the test solution corresponds to the peak in the
RS in the mobile phase.
chromatogram obtained with the reference solution.
C. When examined in the range 200 nm to 300 nm (2.4.7), a Chromatographic system
0.001 percentw/v solution in 0.1 Mhydrochloricacidexhibits a stainless steel column 12.5 cm x 3.0 mm, packed with
a maximum at about 248 nm. octadecylsilane bonded to porous silica (5 Ilm) (Such
as Purospher 18e),
Tests mobile phase: a mixture of 50 volumes of water, 50
_~~Y9Jume~_Q.fQ..~{Qlll([ileJ!J1dj1Lv_olum5).Qf glClI:1CllClf:.rgLc.
Specfiic optical rotation (2.4.22). ---- 60° to ----~67°;deterliimea ill acid,
1.0 pet cent 'ii/v sohition in 111ethanol. flow rate. 0.4 ml per minute,
Related substances. Determine by liquid chromatography spectrophotometer set at 273 nm,
(2.4.14). . injection volume. 10 Ill.

2286

www.webofpharma.com
IP 2010 VALSARTAN AND HYDROCHLOROTHIAZIDE TABLETS

Inject the reference solution. The test is not valid unless the Reference solution (a). A 0.1 per cent w/v solution of
tailing factor is not more than 2.0 and the relative standard valsartan RS in the mobile phase.
deviation for replicate injections is not more than 2.0 per cent. Reference solution (b). Dilute 1.0 ml of reference solution (a)
Inject the test solution and the reference solution. to 100.0 ml with the mobile phase.
Calculate the content ofCz4Hz9Ns03. Chromatographic system as described under Assay.

Storage. Store protected from light and moisture, at a Inject reference solution (a). The test is not valid unless the
temperature not exceeding 30·. tailing factor is not more than 2.0.
Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any
secondary peak is not more than 0.5 times the area of the
Valsartan Tablets principal peak in the chromatogram obtained with reference
solution (b) (0.5 per cent) and the sum of areas of all the
Valsartan Tablets contain not less than 90.0 per cent and not secondary peaks is not more than the area of the principal
more than 110.0 per cent of the stated amount of valsartan, peak in the chromatogram obtained with reference solution
CZ4Hz9Ns03. . (b) (1.0 per cent).
Usual strengths. 40 mg; 80 mg; 160 mg; 320 mg. Other tests. Comply with the tests stated under Tablets.
Identification Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh and powder 20 tablets. Weigh accurately
A. Shake a quantity of the powdered tablets containing 0.1 g
a quantity ofpowder containing 50 mg ofValsartan, disperse
ofValsartan with 40 ml of methanol, filter and evaporate the
in 25 ml ofthe mobile phase and dilute to 100.0 ml with the
filtrate to dryness. On the residue, determine by infrared
mobile phase and filter. Dilute 5.0 ml ofthis solution to 50.0 ml
absorption spectrophotometry (2.4.6). Compare the spectrum
with the mobile phase.
with that obtained from valsartan RS or with the reference
spectrum of valsartan. Reference solution. A 0.005 per cent w/v solution of vaIsartan
RS in the mobile phase.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the Chromatographic system
chromatogram obtained with the reference solution. - a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm),
Tests - mobile phase: a mixture of 50 volumes of water, 50
volumes of acetonitrile and 0.1 volume ofglacial acetic
Dissolution (2.5.2). acid,
Apparatus No.1, flow rate. 1 ml per minute,
Medium. 900 ml ofphosphate bufferpH 6.8, spectrophotometer set at 273 nm,
Speed and time. 50 rpm and 45 minutes. - injection volume. 10 Ill.

Withdraw a suitable volume ofthe medium and filter. Measure Inject the reference solution. The test is not valid unless the
the absorbance of the filtered solution, suitably diluted with tailing factor is not more than 2.0 and the relative standard
the medium if necessary, at the maximum at about 250 nm deviation for replicate injections is not more than 2.0 per cent.
(2.4.7). Calculate the content of CZ4Hz9Ns03 in the medium Inject the test solution and the reference solution.
from the absorbance obtained from a solution of known Calculate the content ofCz4Hz9Ns03 in the tablets.
concentration of valsartan RS prepared by dissolving in
minimum amount of methanol and diluted with the dissolution Storage. Store protected from light and moisture.
medium.
D. Not less than 70 per cent of the stated amount of
Cz~z9Ns03' Valsartan and Hydrochlorothiazide
Related Substances. Determine by liquid chromatography Tablets
(2.4.14).
Valsartan and Hydrochlorothiazide Tablets contain not less
Test solution. Shake a quantity of the powdered tablets than 90.0 per cent and not more than 110.0 per cent of the
containing 25 mg ofValsartan with 15 Jill of mobile phase and stated amount of valsartan, CZ4Hz9Ns03 and
dilute to 25.0 ml with the mobile phase, filter. hydrochlorothiazide, C7HsCIN304SZ.

2287

www.webofpharma.com
VALSARTAN AND HYDROCHLOROTHIAZIDE TABLETS IP 2010

Usual Streugth . Valsartan 80 mg and Hydrochlorothiazide Al percent Vm,llll = absorptivity (l per cent, 0.2 cm, 272 urn)
12.5 mg; Valsartan 60 mg and Hydrochlorothiazide 12.5 mg. ofvaIsartan in medium,
Al per cent V250 11111 = absorptivity (1 per cent, 0.2 cm, 250 urn)
Identification ofvalsartan in medium,
A. Determine by thin-layer chromatography (2.4.17), coating Al per cent H mlllll = absorptivity (l percent, 0.2 cm, 272 urn)
the plate with silica gel GF254. ofhydrochlorothiazide in medium,
Al per cent H 250 11/11 = absorptivity (l per cent, 0.2 cm, 250 urn)
Mobile phase. A mixture of 80 volumes of ethyl acetate, 20 ofhydrochlorothiazide in medium,
volumes of dehydrated alcohol and 10 volumes of a 25 per
cent w/v solution of ammonium hydroxide. D. Not less than 80 per cent ofthe stated amount ofCz4Hz9Ns03
and C7HgCIN304SZ'
Test solution. Disperse a quantity of powdered tablets
containing about 20 mg of valsartan in 100 ml of acetone, Related substances. Determine by liquid chromatography
sonicate for 15 minutes and filter. (2.4.14), as described under Assay using the following
solutions.
Reference solution. A solution containing 0.02 per cent w/v
each of valsal'tan RS and hydrochlorothiazide RS in acetone. Solvent mixture. Equal volumes of acetonitrile and water.
Apply to the plate 2 j..ll of each solution. Allow the mobile Test solution. Disperse a quantity of powdered tablets
phase to rise 7 cm. Dry the plate in a current of warm air and containing about 62.5 mg ofHydrochlorothiazide with 5.0 ml
examine in ultraviolet light at 254 nm. The principal spot in the of water and 100 ml ofsolvent mixture, sonicate for 15 minutes
chromatogram obtained with the test solution corresponds to and shake for 30 minutes. Dilute to 250 ml with the solvent
that in the chromatogram obtained with the reference solution. mixture, centrifuge and dilute 25.0 ml ofthe supematantto200
ml with the solvent mixture.
B. In the Assay, the retention time ofthe principal peak in the
chromatogram obtained with the test solution corresponds to Reference solution (a). A solution containing 0.003 per cent
that in the chromatogram obtained with the reference solution. w/v of benzothiadiazine impurity A RS, 0.006 per cent w/v of
hydrochlorothiazide RS, 0.008 per cent w/v of valsartan RS
Tests and 0.02 per cent w/v of valsartan impurity B RS in the
Dissolution (2.5.2). solvent mixture.

Apparatus No.1, Reference solution (b). Dilute 5.0 ml ofreference solution (a)
Medium. 1000 ml ofphosphate bufferpH 6.8, to 100 ml with the solvent mixture.
Speed and time. 50 rpm and 30 minutes. Reference solution (c). Dilute 10 ml of reference solution (b)
Withdraw a suitable volume ofthe medium and filter. Measure to 100 ml with the solvent mixture.
the absorbance of the filtered solution, suitably diluted with Inject reference solution (b) and (c). The test is not valid unless
the medium ifnecessary, at the maximum at about 250 nm for in the chromatogram obtained with reference solution (b), the
valsartan and 272 urn for hydrochlorothiazide (2.4.7). Calculate resolution between va1sartan impurity Band valsartan and
the content of CZ4Hz9Ns03 and C7HgCIN304SZ in the medium between benzothiadiazine impurity A and hydrochlorothiazide
from the absorbance obtained from a solution of known is not less than 1.4. In the chromatogram obtained with
concentration of valsartan RS and hydrochlorothiazide RS reference solution (c) the relative standard deviation for
in the same medium. replicate injections is not more than 10 per cent.
Calculate the content of valsartan dissolved by the formula: Inject the test solution, reference solution (a), (b) and (c). The
area of the peak due to benzothiadiazine impurity A is not
(ATZx AI per cellI H"'"m) - (AT! x AI percent H",,,,,,) xlZ,SOO
(AI percelltV2sUIlnl X Alpe1'ce11l H 272l1m ) -(AI percentV2721lnl X Al percent H 2SO /lm )
more than 1.0 per cent; the area of any secondary peak other
than valsartan impurity A is not more than 0.2 per cent; and
Calculate the content of hydrochlorothiazide dissolved by the sum of areas of all the secondary peaks other than
the formula: valsartan impurity A is not more than 1.3 per cent. (Valsartan
impurity A is the enantiomer of valsartan and coelutes with
(AT! x Alpercell/V"'"m) -(AT2x AlpercentV"'"m) xgO,OOO
(AI percent H 272nm X Al percent V2S0mn) - (AI percent H2S0nnl X AI percent V21211m ) valsartan in this test.)

where, ATI = absorbance of the test solution at 272 Unifofinitj of content. Comply with the tests shitedTiiider
urn, Tablets.
AT2 = absorbance of the test solution at 250 Determine by liquid chromatography (2.4.14), as described
lllll, under Assay using the following solution as the test solution.

2288

www.webofpharma.com
IP 2010 VANCOMYCIN HYDROCHLORIDE

Solvent mi.;r:ture. Equal volumes of acetonitrile and water. Inject the reference solution. The test is not valid unless the
Test solution. Disperse 1 tablet with 5.0 ml of water and about relative standard deviation for replicate injections is not more
100 ml ofsolvent mixture, sonicate for 15 minutes and dilute to than 2.0 per cent.
200 ml with the solvent mixture. Centrifuge a portion of this Inject the test solution and the reference solution.
solution and dilute a volume of the clear supernatant with the
Calculate the content ofC24H29Ns03 and C7HgCIN304S2 in the
solvent mixture to obtain a 0.02 per cent w/v solution of
tablet.
valsartan.
Storage. Store protected from moisture.
Calculate the content ofC24H29Ns03 and C7HgCIN304S2 in the
tablet.
Other tests. Comply with the tests stated under Tablets.
Assay. Determine by liquid chromatography (2.4.14). Vancomycin Hydrochloride
Solvent mixture. Equal volumes of acetonitrile and water.
Test solution. Disperse a quantity of powdered tablets
containing about 62.5 mg ofHydrochlorothiazide with 5.0 ml
of water and 100 ml ofsolvent mixture, sonicate for 15 minutes
and shake for 30 minutes. Dilute to 250 ml with the solvent
mixture, centrifuge and dilute 25.0 ml ofthe supernatantto 200
ml with the solvent mixture. Dilute a suitable volume of this
solution with the solvent mixture to obtain a 0.02 per cent w/v
solution of VaIsartan.
Reference solution. Transfer about 12.5 mg of
hydrochlorothiazide RS to a 200 ml volumetric flask and add
about 12.5 J mg of vaIsartan RS, J being the ratio ofthe stated
, Hel
amount, in mg, of valsartan to the stated amount, in mg, of
hydrochlorothiazide per tablet. Add about 100 ml of solvent
mixture, sonicate for 15 minutes, dilute to volume and mix.
Transfer 25 ml of this solution to a 50 ml flask, dilute with
solvent mixture to volume and mix. Dilute a suitable volume of
this solution with the solvent mixture to obtain a 0.02 per cent
w/v solution of valsartan RS.
Chromatographic system
- a stainless steel column 12.5 cm x 3.0 mm, packed with
octadecylsilane bonded to porous silica
(5Ilm),
- mobile phase: A. a mixture of 90 volumes of water,
10 volumes of acetonitrile and 0.1 volume of
Mol.Wt. 1485.7
trifluroacetic acid,
B. a mixture of90 volumes of acetonitrile,
Vancomycin Hydrochloride is a mixture ofrelated
10 volumes of water and 0.1 volume of trifluroacetic
glycopeptides, consisting principally of the
acid,
monohydrochloride of(3S,6R,7R,22R,23S,26S,30aS,36R,
- a linear gradient programme using the conditions given
38aR)-3-(2-amino-2-oxoethyl)-44-[[2-0-(3-amino-2,3,6-
below,
trideoxy-3-C-methyl-{3-L-lyxo-hexopyranosyl)-{3-D-
- flow rate. 0.4 ml per minute,
glucopyranosyl]oxy]-10,19-dichloro-7,22,28,30,32-
- spectrophotometer set at 265 nm,
pentahydroxy-6-[[(2R)-4-methyl-2-
- injection volume. 10 Ill.
(methylamino)pentanoyl]amino]-2,5,24,38,39-pentaoxo-
Time Mobile phase A Mobile phase B
2,3,4,5,6,7,23,24,25,26,36,37,38,38a-tetradecahydro-22H-
(in min.) (per cent v/v) (per cent v/v)
8,11: 18,21-dietheno-23,36-(iminomethano)-13,16:31,35-
0-25 90 -710 10-790 dimetheno-lH, 13H-[1 ,6,9]oxadiazacyclohexadecinol[4,5-m]
25-27 10-790 90-710 [10,2,16]benzoxadiazacyclotetracosine-26-carboxylic acid
27-40 90 10 (vancomycin B).

2289

www.webofpharma.com
VANCOMYCIN HYDROCHLORIDE IP 2010

It is produced by certain strains of Amycolatopsis orientalis Time Mobile phase A Mobile phase B
or obtained by any other means. (in min.) (per cent v/v) (per cent v/v)
Vancomycin Hydrochloride contains not less than 1050 ill 0-13 100 o
per mg ofC66H7sClzNg024, RCI, calculated on the anhydrous 13-22 100-70 0-7100
basis and not less than 93.0 per cent of vancomycin B. 22-26 o 100
Category. Antibacterial. 26-27 100 o
Description. A white or almost white hygroscopic powder. Inject test solution (b), (c) and the reference solution. The test
is not valid unless the resolution between the 2 principal peaks
Identification in the chromatogram obtained with the reference solution is
A. In the test for Vancomycin B, the principal peak in the not less than 5.0, signa1-to-noise ratio for the principal peak in
chromatogram obtained with test solution (a) corresponds to the chromatogram obtained with test solution (c) is not less
that in the chromatogram obtained with the reference solution. than 5.0 and tailing factor for the peak due to vancomycin in
the chromatogram obtained with the test solution (b) is not
B. Gives reaction A ofchlorides (2.3.1).
more than 1.6.
Tests Inject test solution (b) and the reference solution.

Appearance ofsolution. Al 0.0 per cent w/v solution in water Calculate the content of vancomycin B hydrochloride.
is clear (2.4.1), absorbance ofthe solution at 450 TIm (2.4.7) is Related substances. Determine by liquid chromatography
not more than 0.1. (2.4.14) as described under Vancomycin B with the following
pH (2.4.24).2.5 to 4.5, determined in a 5.0 per cent w/v solution modifications.
in carbon-dioxide ji-ee water. Inject test solution (a), (b) and (c). In the chromatogram
Vancomycin B. Determine by liquid chromatography (2.4.14). obtained with the test solution the area of any secondary
peak due to N-demethylvancomycin B (Vancomycin
NOTE-Use freshly prepared solutions. Hydrochloride impurity A), (4S, 7R,8R,23R,24S,27 S,
Test solution (a). Dissolve 0.2 g of the substance under 31aSa,37R,39aR)-45-[[2-0-(3-amino-2,3,6-trideoxy-3-C-methyl-
examination in 100 m1 ofthe mobile phase A. a-L-Iyxo-hexopyranosyl)-~ -D-glucopyranosyl]oxy]-11,20-
dichloro-8,23,29,31 ,33-pentahydroxy-7-[[(2R)-4-methyl-2-
Test solution (b). Dilute 2.0 ml oftest solution (a) to 50 m1 with
(methylamino)pentanoyl]amino]-2,6,25,39,40-pentaoxo-
mobile phase A.
1,2,3,4,5,6,7,8,24,25,26,27,37,38,39,39a-hexadecahydro-23H-
Test solution (c). Dilute 0.5 ml oftest solution (b) to 20 ml with 9,12: 19,22-dietheno-24,37-(iminomethano)-14,17:32,36-
mobile phase A. dimetheno-14H-[ 1,6,1 O]oxadiazacycloheptadecino[4,5-
Reference solution. A 0.5 per cent w/v solution of vancomycin m][10,2,16]benzoxadiazacyclotetracosine-4,27-dicarboxylic
hydrochloride RS in water. Heat at 65° for 24 hours, allow to acid ([ ~ ASp3] vancomycin B) (Vancomycin Hydrochloride
cool. impurity B), aglucovancomycin B (Vancomycin Hydrochloride
impurity C) and desvancosaminylvancomycin B (Vancomycin
Chromatographic system Hydrochloride impurity D) is not more than 4.0 per cent. The
- a stainless steel column 25 cm x 4.6 mm, packed with sum ofarea ofall the secondary peaks is not more than 7.0 per
octadecylsilane bonded to porous silica (5 11m), cent. Ignore any peak with an area less than the area of the
- mobile phase: A. a lllixture of 1 volume of principal peak in the chromatogram obtained with test solution
tetrahydrofilran, 7 volumes of acetonitrile and 92 (cHO.1 percent).
volumes ofbuffer solution prepared by diluting I ml of
triethylamine to 499 ml with watel; adjust the pH to 3.2 Heavy metals (2.3.13). 1.0 g complies with the limit test for
with orthophosphoric acid, heavy metals, Method B (30 ppm).
B. a mixture of I volume of Sulphated ash (2.3.18). Not more than 1.0 per cent.
tetrahydrofilran, 29 volumes of acetonitrile and
70 volumes ofbuffer solution, Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
. . . JdiI!~atgmdientpmgm111J1le_usiI!gJhe_c_oI!ditioI!sgi_v_eI! Unit per mg.
bel()~, Vancomycin Hydrochloride intended for use in the
- flow rate. 1 ml per minute, manufacture ofparenteral preparations without a filrther
- spectrophotometer set at 280 nm, appropriate sterilization procedure complies with the
- injection volume. 20 Ill. following additional requirement.

2290

www.webofpharma.com
IP 2010 VANCOMYCIN INTRAVENOUS INFUSION

Sterility (2.2.11). Complies with the test for sterility. D. Not less than 85 per cent of the stated amount of
Water (2.3.43). Not more than 5.0 per cent, determined on C66H7sClzN9024.
0.5 g. Water (2.3.43). Not more than 8.0 percent.
Assay. Determine by the microbiological assay ofantibiotics, Other tests. Comply with the tests stated under Capsules.
Method B (2.2.10) using vancomycin hydrochloride RS.
Assay. Determine by the microbiological assay ofantibiotics,
Storage. Store protected from light and moisture. Method A (2.2.10) on a solution prepared in the following
manner.
If it is intended for use in the manufacture of parenteral
preparations, the container should be sterile, tamper-proof Place not less than 5 capsules in a glass blender jar and blend
and sealed so as to exclude micro-organisms. at high speed for 3 to 5 minutes with a sufficient volume of
Buffer No.3 to yield a stock solution having a convenient
concentration of vancomycin. Dilute an accurately measured
volume'ofthis stock solution with Buffer No.3 to obtain a test
Vancomycin Capsules dilution having a concentration assumed to be equal to the
Vancomycin Hydrochloride Capsules median dose level of the standard.
Storage. Store protected from moisture.
Vancomycin Capsules contain not less than 90.0 per cent and
not more than 115.0 per Ctlnt of the stated amount of
vancomycin, C66H7sC12N9024.
Usual strength. 125 mg. Vancomycin Intravenous Infusion
Identification Vancomycin Intravenous Infusion is a sterile solution of
Vancomycin Hydrochloride in Water for Injections. It is
Determine by paper chromatography (2.4.15), using a suitable prepared by dissolving Vancomycin Hydrochloride for
sheet of chromatographic filter paper. Intravenous Infusion in Water for Injections and then diluting
Mobile phase. A mixture of 6 volumes of butyl alcohol, 4 with the requisite volume of a suitable diluent in accordance
voli.nnes of water and 3 volumes of pyridine. with the manufacturer's instructions.
Test solution. Disperse the content of capsules containing The intravenous infilsion complies with the tests stated under
about 100 mg ofvancomycin in 100.0 ml ofwater. Parenteral Preparations.
Reference solution. A 0.1 per cent w/v solution of vancomycin Storage. Vancomycin Intravenous Infusion should be used
hydrochloride RS in water. immediately after preparation but, in any case, within the period
recommended by the manufacturer when prepared and stored
Apply to the plate 5 l.d of each solution. Develop by
strictly in accordance with the manufacturer's instructions.
descending chromatography for about 7 hours. After
development, dry the paper and place it on an inoculated agar Usual strengths. 500 mg; 1 g.
surface of sufficient area to accommodate the paper and
prepared for vancomycin assay described in microbiological Vancomycin Hydrochloride for Intravenous
assay ofantibiotics (2.2.10) except to use Medium B. Remove Infusion
the paper from the agar surface after 30 minutes and incubate
the agar medium at 37° for 18 hours; clear zones of inhibition Vancomycin Hydrochloride for Intravenous Infusion is a sterile
are produced at corresponding positions on the two material consisting of Vancomycin Hydrochloride with or
chromatograms. without excipients. It is filled in a sealed container.
The contents of the sealed container comply with the
Tests requirements stated under Parenteral Preparations
Dissolution (2.5.2). (Powders for Injection) and with thefollowing requirements.

Apparatus No.2, Vancomycin Hydrochloride for Intravenous Infusion contains


not less than 88.0 per cent ofVancomycin B.
Medium. 900 ml of water,
Speed and time. 100 rpm and 45 minutes. Identification
Withdraw a suitable volume ofthe medium and filter.
A. In the test for vancomycin B, the retention time of the
Carry out the method as described under Assay. principal peak in the chromatogram obtained with the test

2291

www.webofpharma.com
VANCOMYCIN INTRAVENOUS INFUSION IP 2010

solution corresponds to that in the chromatogram obtained solution (c), signal-to- noise ratio of the principal peak is not
with the reference solution. less than 5; the tailing factor of the vancomycin peak in the
chromatogram obtained with test solution (b) is not more than
B. Gives reaction A ofchlorides (2.3.1).
1.6 and the resolution between the two principal peaks in the
Tests chromatogram obtained with the reference solution is not less
than 5.0.
pH (2.4.24).2.5 to 4.5, determined on a 5.0 per centw/v solution
Related substances. Determine by liquid chromatography
ofVancomycin Hydrochloride.
(2.4.14), using test solutions (a), (b) and (c) as described under
Appearance of solution. A 10.0 per cent w/v solution of Vancomycin B.
Vancomycin Hydrochloride is clear (2.4.1) and the absorbance
Chromatographic system as described under Vancomycin B.
ofthe solution at 450 urn (2.4.7) is not more than 0.1.
In the chromatogram obtained with test solution (a) calculate
Vancomycin B. Determine by liquid chromatography (2.4.14).
the content of each impurity.
NOTE-Use the solutions within 4 hours ofpreparation.
The content of any impurity is not more than 4.0 per cent and
Test solution (a). Dissolve a quantity of the substance under the sum of the contents of any such impurities is not more
examination in sufficient mobile phase A to produce a solution than 12.0 per cent. Ignore any peak with an area less than that
containing 2,000 ill ofvancomycin per ml. of the principal peak in the chromatogram obtained with test
Test solution (b). Dilute I ml oftest solution (a) to 25 ml with solution (c) (0.1 per cent).
mobile phase A. Water (2.3.43). Not more than 5.0 per cent, determined on
Test solution (c). Dilute 1 ml oftest solution (b) to 40 ml with 0.5g.
mobile phase A. Bacterial endotoxins (2.2.3). Dissolve the contents of the
Reference solution. A 0.05 per cent w/v solution of sealed container in tris-chloride bufferpH 7.4 prepared using
vancomycin hydrochloride RS in water. Heat at 65° for water BET to give a solution containing 9000 Units of
24 hours and allow to cool. vancomycin per ml. Carry out the test on the resulting solution;
the maximum allowable endotoxin concentration ofthe solution
Chromatographic system is 2.5 Units of endotoxins per ml. Carry out the test using the
- a stainless steel column 25 cm x 4.6 mm, packed with maximum valid dilution of the prepared solution calculated
endcapped octadecylsilane bonded to porous silica from the declared sensitivity of the lysate used in the test.
(5 Ilm) (Such as Hypersil ODS),
mobile phase: A. a mixture of 1 volumes of Assay. Determine the weight ofthe contents often containers
tetrahydrofuran, 7 volumes of acetonitrile and as described in the test for Uniformity of weight under
92 volumes ofbuffer solution prepared by diluting I ml Parenteral Preparations (Powders for Injection).
of triethylamine to 500 ml with water, adjust the pH to Determine on the mixed contents of ten containers by the
3.2 with orthophosphoric acid, microbiological assay ofantibiotics, Method B (2.2.10).
B. a mixture of I volume of
The upper fiducial limit of error is not less than 95.0 per cent
tetrahydrofuran, 29 volumes of acetonitrile and
and the lower fiducial limit oferror is not more than 115.0 per
70 volumes of buffer solution,
cent of the stated number of Units.
flow rate. 1 ml per minute,
a linear gradient programme using the conditions given Labelling. The label states (1) the total number of Units of
below, vancomycin in the container and (2) the number of Units of
spectrophotometer set at 280 urn, vancomycin per mg.
injection volume. 20 Ill.
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent v/v)
Vancomycin Oral Solution
0-13 100 o
Vancomycin Oral Solution is a solution of Vancomycin
13-21 100-*> 0-400
Hydrochloride in a suitable flavoured vehicle. It is prepared
21-25 0 100 by dissolvingVancofiiyciii·HydrochlofidefofOfalSolutioiiifi
25-35 100 o the requisite amount of a suitable diluent.
Inject the reference solution, test solution (b) and (c). The test The oral solution complies with the tests stated under Oral
is not valid unless in the chromatogram obtained with test Liquids.

2292

www.webofpharma.com
IP 2010 VANCOMYCIN ORAL SOLUTION

Storage. Vancomycin Oral Solution should be used - mobile phase: A. a mixture of4 volumes oftriethylamine
immediately after preparation but, in any case, within the period diluted with 1996 volumes of water, adjusted to pH 3.2
recommended by the manufacturer when prepared and stored with orthophosphoric acid (solution A), 10 volumes
strictly in accordance with the manufacturer's instructions. of tetrahydrofitran and 70 volumes of acetonitrile to
920 volumes of solution A,
Usual strength. 50 mg per ml.
B. a mixture of 10 volumes of
Vancomycin Hydrochloride for Oral Solution tetrahydrojitran, 290 volumes of acetonitrile and 700
volumes of solution A,
Vancomycin Hydrochloride for Oral Solution is a dry powder - a linear gradient programme using the conditions given
consisting of Vancomycin Hydrochloride with or without below,
excipients. It is filled in a sealed container. flow rate. I ml per minute,
The contents of the sealed container comply with the spectrophotometer set at 280 urn,
requirements stated under Powders and Granules for Oral injection volume. 20 /ll.
Solutions and Oral Suspensions stated under Oral Liquids Time Mobile phase A Mobile phase B
and with the following requirements. (in min.) (per cent v/v) (per cent v/v)
Vancomycin Hydrochloride for Oral Solution contains not less 0-13 100 o
than 88.0 per cent ofVancomycin B. 13 -21 100-70 0-7100
21-25 0 100
Identification 25-35 100 o
A. In the test for Vancomycin B, the retention time of the Inject each solution. The test is not valid unless in the
principal peak in the chromatogram obtained with the test chromatogram obtained with test solution (c) signal-to- noise
solution corresponds to that in the chromatogram obtained ratio of the principal peak is not less than 5.0. The tailing
with the reference solution. factor of the principal peak in the chromatogram obtained
B. Gives reaction A ofchlorides (2.3.1). with test solution (b) is not more than 1.6; and the resolution
between the two principal peaks in the chromatogram obtained
Tests with the reference solution is not less than 5.0.

pH (2.4.24).2.5 to 4.5, determined on a 5.0 per cent w/v solution Related substances. Determine by liquid chromatography
ofVancomycin HydrocWoride. (2.4.14), using test solution (a), (b) and (c) as described under
Vancomycin B.
Appearance of solution. A 10.0 per cent w/v solution is clear
(2.4.1) and the absorbance at 450 urn (2.4.7) is not more than Use chromatographic system as described under Vancomycin
0.1. B.

Vancomycin B. Determine by liquid chromatography (2.4.14). Inject test solution (c) and the reference solution. The area of
any secondary peak is not more than 4.0 per cent and the sum
NOTE-Use the solutions within 4 hours ofpreparation. of the areas of all the secondary peaks is not more than 12.0
Test solution (a). Dissolve a quantity ofthe powder in mobile per cent. Ignore any peak with an area less than the area ofthe
phase A to produce a solution containing 2,000 IU of principal peak in the chromatogram obtained with the test
vancomycin per ml. solution (c)(0.1 per cent).

Test solution (b). Dilute 1 ml oftest solution (a) to 25 ml with Water (2.3.43). Not more than 5.0 per cent, determined on
mobile phase A. 0.5g.
Test solution (c). Dilute 1 ml oftest solution (b) to 40 ml with Assay. Determine on the mixed contents often containers by
mobile phase A. the microbiological assay ofantibiotics, Method A or Method
B(2.2.10).
Reference solution. A 0.05 per cent w/v solution of vancomycin
hydrochloride RS in water. Heat at 65° for 24 hours and allow The upper fiducial limit of error is not less than 95.0 per cent
to cool. and the lower fiducial limit oferror is not more than 115.0 per
cent of the stated number of Units.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with Labelling. The label states (1) the total number of Units of
end-capped octadecylsilane bonded to porous silica (5 vancomycin in the container and (2) the number of Units of
/lm) (Such as HypersiIODS), vancomycin per mg.

2293

www.webofpharma.com
VANILLIN IP 2010

Vanillin Reference solution (b). A 0.2 per cent w/v solution of vanillin
RS in methanol.
Use an unsaturated tame and allow the mobile phase to rise
CHO

o
yOCH 3
10 cm. Apply to the plate 5 III of each solution. After
development, dry the plate in cold air and examine in ultraviolet
light at 254 urn. Any secondary spot in the chromatogram
obtained with test solution (a) is not more intense than the
spot in the chromatogram obtained with reference solution
OH
(a). Spray with dinitrophenylhydrazine-aceto-hydrochloric
solution and examine in daylight. Any secondary spot in the
CSHS0 3 Mol. Wt. 152.2
chromatogram obtained with test solution (a) is not more
Vanillin is 4-hydroxy-3-methoxybenzaldehyde. intense than the spot in the chromatogram obtained with
Vanillin contains not less than 99.0 percent and not more than reference solution (a).
10 1.0 per cent of CSH S0 3, calculated on the dried basis. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Category. Pharmaceutical aid (flavouring agent). Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Description. A white or slightly yellow powder or crystalline on 1.0 g by drying over phosphorus pentoxide for 4 hours.
needles; odour, characteristic of vanilla. Assay. Weigh accurately about 0.12 g, dissolve in 20 ml of
ethanol (95 per cent), add 60 ml ofcarbon dioxide-Fee water.
Identification Titrate with 0.1 M sodium hydroxide, determining the end-
Test A may be omitted iftests B, C andD are carried out. Tests point potentiometrically(2.4.25).
B, C and D may be omitted if test A is carried out. I ml of 0.1 M sodium hydroxide is equivalent to 0.01521 g of
A. Determine by infrared absorption spectrophotometry (2.4.6). CSH S0 3•
Compare the spectrum with that obtained with vanillin RS. Storage. Store protected from light and moisture.
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution Vasopressin Injection
(b). Examine the chromatograms in daylight after spraying.
Vasopressin Injection is a sterile aqueous solution containing
C. To 5 ml ofa saturated solution add 0.2 ml offerric chloride
the water soluble pressor principle obtained from the posterior
solution; a blue colour is produced. Heat to 800 ; the solution
lobe of the pituitary of healthy oxen or other mammals or by
becomes brown and a white precipitate is produced on cooling.
synthesis.
D. Melting range (2.4.21). 81° to 84°.
Vasopressin Injection contains a pressor activity equivalent
Tests to not less than 90.0 per cent and not more than 110.0 per cent
of the stated number of Units.
Appearance ofsolution. A 5.0 per cent w/v solution in ethanol
Usual strength. 20 Units per ml.
(95 per cent) is clear (2.4.1) and not more intensely coloured
than reference solution BS6{2.4.1). Description. A clear, colourless or practically colourless liquid;
odour, faint and characteristic.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Identification
Mobile phase. A mixture of98.5 volumes ofdichloromethane,
A. In the Assay, the principal peak in the chromatogram
1 volume of methanol and 0.5 volume of anhydrous acetic
obtained with test solution corresponds to the peak in the
acid.
chromatogram obtained with the reference solution.
Test solution (a). Dissolve 0.2 g of the substance under
B. Inject into the vein ofa mammal anaesthetised by a general
examination in 10 ml ofmethanol.
anaesthetic or by destruction of the brain; it causes a rise of
Test solidioli (b). Dissolve O:Z g of the substance-iiiider blood pressure.
exatl1inatiol1 i111 00 illl ofmethdl1ol.
Tests
Reference solution (a). Dissolve 10 mg ofthe substance under
examination in 100 ml ofmethanol. pH (2.4.24). 2.5 to 4.5.

2294

www.webofpharma.com
IP 2010 VERAPAMIL HYDROCHLORIDE

Bacterial endotoxins (2.2.3). Less than 17.0 Endotoxin Units mobile phase: a mixture of87 volumes of0.66 per cent
per Unit of vasopressin. w/v solution of dibasic ammonium phosphate, adjusted
to pH 3.0 with orthophosphoric acid and 13 volumes of
Other tests. Complies with the tests stated under Parenteral
acetonitrile, filter through 0.45 /lm nylon membrane,
Preparations (Injections).
flow rate. 1 ml per minute,
Vasopressin injection containing Vasopressin ofnatural origin - spectrophometer set at 220 nm,
obtained by extraction and purification complies with the - injection volume. 20 /ll.
following additional requirement.
Equilibrate the column at least for one hour. Run the
Oxytocin impurity. Not more than 1.2 Units per ml. chromatogram minimum of60 minutes.
Detennine by liquid chromatography (2.4.14). Inject the reference solution. The test is not valid unless the
resolution between vasopressin and the adjacent peak is not
Test solution. Use the injection under examination. less than 1.5 and relative standard deviation for replicate
Reference solution. Dissolve the contents of one vial of injections is not more than 2.0 per cent.
oxytocin RS in a 1.65 per cent w/v solution of sodium Inject the reference solution and the test solution.
dihydrogen orthophosphate to produce a solution containing
the same concentration in /lg ofoxytocin as that stated on the Storage. Store at the temperature between 2° to 8°.
label ofthe injection. Labelling. The label states (1) the number of Units of the
vasopressor activityper ml; (2) either the animal source ofthe
Chromatographic system
vasopressin or that it is synthetic.
- a stainless steel column 10 Cln x 4.6 mm, packed with
endcapped octadecylsilane bonded to porous silica
(5 /lm) (Such as Nucleosil C18),
- column temperature. 40°, Verapamil Hydrochloride
- mobile phase: a mixture of85 volumes ofa 0.2 per cent
v/v solution of orthophosphoric acid and 15 volumes Verapamil Chloride; Iproveratril Hydrochloride
of acetonitrile,
- flow rate. 1 ml per minute,
H3CO~ ~OCH3
- spectrophotometer set at 220 nm,
- injection volume. 200 /ll.
~ ~I , HCI
H3CO N HC OCH 3
Inject the reference solution. The test is not valid unless the
H
3
C 3 . CN
CH 3
theoretical plates is not less than 50,000.
Inject the test solution and the reference solution.
Mol. Wt. 491.1
Calculate the content of C13 H66N 12012S2 in the injection. Verapamil Hydrochloride is (RS)-2-(3,4-dimethoxyphenyl)-5-
Assay. Detennine by liquid chromatography (2.4.14). [[2-(3,4-dimethoxyphenyl)ethyl](methyl)amino]-2-(1-
methylethyl)pentanenitrile hydrochloride.
Solvent mixture. Dissolve 5.0 g of chlorobutanol in 5.0 ml of
glacial acetic acid, add 1.1 g of sodium acetate, 5.0 g of Verapamil Hydrochloride contains not less than 99.0 per cent
ethanol, and dilute to 1000 ml with water and mix. and not more than 101.0 per cent of~7H38N204, HCl, calculated
on the dried basis.
Test solution. Dilute 2.0 ml of injection under examination to
25 ml with 0.25 per cent w/v of glacial acetic acid and mix. Category. Calcium channel blocker.
Dose. Orally, as antiarrhythmic, 40 to 120 mg thrice daily; as
Reference solution. Dissolve the contents of one vial of
antianginal, 80 to 120 mg thrice daily; as antihypertensive, 240
vasopressin RS in a known volume of solvent mixture. If
to 480 mg daily, in 2 to 3 divided doses; by slow intravenous
necessary dilute the prepared solution to a working
injection, 5 to 10 mg.
concentration range.
Description. A white, crystalline powder.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with Identification
endcapped octadecylsilane bonded to porous silica (5
/lm), Test A may be omitted iftests B, C andD are carried out. Tests
column temperature. 30°, Band C may be omitted if tests A and D are carried out.

2295

www.webofpharma.com
VERAPAMIL HYDROCHLORIDE IP 2010

A. Determine by infrared absorption spectrophotometry (2.4.6). Time Mobile phase A Mobile phase B
Compare the spectrum with that obtained with verapamil (min.) (per cent v/v) (per cent v/v)
hydrochloride RS or with the reference spectrum ofverapamil 0-22 63 37
hydrochloride.
22-27 63-735 37 -765
B. When examined in the range 220 nm to 360 nm (2.4.7), a
. 27 -35 35 65
0.002 per cent w Iv solution in 0.01 M hydrochloric acid shows
absorption maxima at about 229 nm and 278 nm and there may 35-36 35-763 65-737
be a shoulder at about 282 nm. The ratio ofthe absorbance at 36-50 63 37
the maximum at about 278 nm to that at the maximum at about
229 nm is 0.35 to 0.39. Inject reference solution (a). The test is not valid unless the
resolution between the peaks due to verapamil and verapamil
C. In test A for Related substances, the principal spot in the
impurity A is not less than 5.0.
chromatogram obtained with test solution (b) conesponds to
that in the chromatogram obtained with reference solution (a). Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution the area of any
D. Gives reaction B ofchlorides (2.3.1).
secondary peak is not more than the area ofthe principal peak
Tests in the chromatogram obtained with reference solution (b) (0.1
per cent). The sum ofall the secondary peaks is not more than
Appearance of solution. A5.0 per cent w/v solution in carbon 3 times the area of the principal peak in the chromatogram
dioxide-free water prepared with the aid of gentle heat is clear obtained with reference solution (b) (0.3 per cent). Ignore any
(2.4.1), and colourless (2.4.1). peak with an area less than 0.1 times the area of the principal
peak in the chromatogram obtained with reference solution
pH (2.4.24).4.5 to 6.0, detennined in a 5.0 per cent w/v solution
prepared with the aid of gentle heat.
(b) (0.01 per cent).

Related substances. Detennine by liquid chromatography Sulphated ash (2.3.18). Not more than 0.1 per cent.
(2.4.14). Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Solvent mixture. 63 volmnes ofmobile phase A and 37 volumes on 1.0 g by drying in an oven at 105°.
ofmobile phase B. Assay. Weigh accurately about 0.4 g, dissolve in 40 ml of
Test solution. Dissolve 25 mg of the substance under anhydrous glacial acetic acid, add 6 ml of mercuric acetate
examination in 10.0 ml ofthe solvent mixture. solution. Titrate with 0.1 M perchloric acid, detennining
the end-point potentiometrically (2.4.25). Carry out a blank
Reference solution (a). Dissolve 5 mg each of of verapamil titration.
hydrochloride RS and (2RS)-2-(3,4-dimethoxyphenyl)-2-[2-
[[2-(3, 4-dimethoxy-phenyl)ethyl](methyl)aminoJethyl] -3- 1 ml of 0.1 M perchloric acid is equivalent to 0.04911 g of
methylbutanenitrile RS (verapamil impurity A RS) in 20.0 ml CnH38N204, HCl.
ofthe solvent mixture. Dilute 1 ml ofthis solution to 10 ml with Storage. Store protected from light and moisture.
the solvent mixture.
Reference solution (b). Dilute 1.0 ml of the test solution to
100.0 ml with the solvent mixture. Dilute 1.0 ml ofthis solution
to 10.0 ml with the solvent mixture.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
Verapamil Injection
palmitamidopropylsilane bonded to porous silica (5 /-lm), Verapamil Hydrochloride Injection; Verapamil Chloride
mobile phase: A. a 0.7 per cent w/v solution of
Injection; Iproveratril Hydrochloride Injection
dipotassium hydrogen phosphate adjusted to pH 7.2
with orthophosphoric acid, Verapamil Injection is a sterile solution ofVel'apami1
B. acetonitrile, Hydrochloride in Water for Injections.
a linear gradientprogrammeusingthe conditions given
Verapamil Injection contains not less than 90;0 percent and
below,
not more than 110.0 per cent ofthe stated amount ofverapamil
flow rate. 1.5 ml per minute,
hydrochloride, C 27H 38N 20 4, HCl.
spectrophotometer set at 278 nm,
injection volume. 10 /-ll. Usual strength. 2.5 mg per ml.

2296

www.webofpharma.com
IP 2010 VERAPAMIL TABLETS

Identification B. Carry out test A but using a mixture of 85 volumes of


cyclohexane and 15 volumes of diethylamine as the mobile
A. Dilute a volume containing 10 mg of Verapamil
phase and applying separately to the plate 30 Itl of each 6fthe
Hydrochloride to 5 ml with 0.1 M hydrochloric acid, extract
test solution and the reference solution.
with 5 ml of ether, discard the ether extract and make the
aqueous layer just alkaline with 2 M potassium carbonate. Other tests. Complies with the tests stated under Parenteral
Extract with 5 ml of ether, filter the ether layer through Preparations (Injections).
anhydrous sodium sulphate and evaporate to dryness. The Assay. Dilute an accurately measured volume containing 5 mg
residue complies with the following test. of Verapamil Hydrochloride to 100.0 ml with 0.01 M
Determine by infrared absorption spectrophotometry (2.4.6). hydrochloric acid and measure the absorbance ofthe resulting
Compare the spectrum with that obtained with verapamil solution at the maximum at about 278 nm (2.4.7). Calculate the
hydrochloride RS treated in the same manner or with the content of CZ7H3sNz04, HCl taking 118 as the specific
reference spectrum ofverapamil. absorbance at 278 urn.
Storage. Store protected from light.
B. To a volume conta~ning 5 mg of Vel'apami1Hydrochloride
add 0.2 rnl ofa 5 per cent w/v solution of mercuric chloride; a
white precipitate is produced.

C. To a volume containing 5 mg ofVerapamil Hydrochloride


add 0.5 ml of 3 M sulphuric acid and 0.2 ml of dilute potassium Verapamil Tablets
permanganate solution; a violet precipitate is produced which Verapamil Hydrochloride Tablets; Verapamil CWoride
quickly dissolves to produce a very pale yellow solution.
Tablets; Iproveratril Hydrochloride Tablets

Tests Verapamil Tablets contain not less than 92.5 per cent and not
more than 107.5 per cent of the stated amount of verapamil
pH (2.4.24).4.5 to 6.0. hydrochloride, CZ7H3sNz04' HCl. The tablets may be coated.
Related substances. A. Determine by thin-layer Usual strengths.40 mg; 80 mg; 120 mg; 160 mg.
chromatography (2.4.17), coating the plate with silica gel G
Identification
Mobile phase. A mixture of70 volumes of toluene, 20 volumes
of methanol, 5 volumes of glacial acetic acid and 5 volumes A. Shake a quantity of the powdered tablets containing 0.1 g
of acetone. ofVerapamil Hydrochloride with 25 ml of 0.1 Mhydrochloric
acid, filter, extract the filtrate with 25 ml of ether, discard the
Test solution. Evaporate a volume containing 5 mg ofVerapamil
ether extract and make the aqueous layer just allmline with
Hydrochloride carefully to dryness on a water-bath in a current
2 M potassium carbonate. Extract with 25 ml of ether, filter the
of nitrogen and dissolve the residue as completely as possible
ether layer through anhydrous sodium sulphate and
in 0.25 ml of chloroform.
evaporate to dryness. The residue complies with the following
Reference solution. Dilute 1 volume of the test solution to tests.
100 volumes with chlorojorm and dilute 1 volume of the
Determine by infrared absorption spectrophotometry (2.4.6).
resulting solution to 10 volumes with chloroform.
Compare the spectrum with that obtained with verapamil
Apply to the plate 30 Itl of each solution. After development, hydrochloride RS treated in the same manner or with the
dry the plate in air for 10 minutes and repeat the development. reference spectrum ofverapamil.
Dry the plate at 110° for 30 minutes and allow to stand until the
B. Shake a quantity ofthe powdered tablets containing 0.1 g
odour ofsolvent is no longer detectable. Spray with a solution
ofVerapamil Hydrochloride with 10 ml of dichloromethane,
prepared by dissolving 5 g of ferric chloride hexahydrate
filter, evaporate the filtrate to dryness and dissolve the residue
and 2 g of iodine in sufficient ofa mixture ofequal volumes of
in 10 ml of water (solution A). To 2 ml ofthe resulting solution
acetone and a 20 per cent w/v solution of (+ )-tartaric acid to
add 0.2 ml ofa 5 per cent w/v solution of mercuric chloride; a
produce 100 ml, applying a total of 15 to 20 ml ofthe reagent
white precipitate is produced.
for a plate (20 cm x 20 cm), and examine immediately. Any
secondary spot in the chromatogram obtained with the test C. To 2 ml of solution A obtained in test B add 0.5 ml of 3 M
solution is not more intense than the principal spot in the sulphuric acid and 0.2 ml of dilute potassium permanganate
chromatogram obtained with the reference solution. Ignore solution; a violet precipitate is produced which quickly
. any spot remaining on the line of application. dissolves to produce a very pale yellow solution.

2297

www.webofpharma.com
VERAPAMIL TABLETS IP 2010

Tests Vinblastine Sulphate


Related substances. Detennine by liquid chromatography
(2.4.14).
Test solution. Shake a quantity of the powdered tablets
containing about 0.24 g ofVerapamil Hydrochloride with 100.0
ml ofthe mobile phase.
Reference solution (a). Dilute 1 ml ofthe test solution to 100.0
ml with the mobile phase. Further dilute 1.0 ml of this solution
to 10.0 ml with the mobile phase.
Reference solution (b). A solution containing 0.005 per cent
w/v each of verapamil hydrochloride RS and (2RS)-2-(3,4-
di methoxyphenyl) -2- [2 - [[2 - (3, 4 -dime thoxy- Mol. Wt. 909.1
phenyl)ethylj(methyl)aminoJethylj-3-methylbutanenitrile
RS (verapamil impurity A RS) in the mobile phase. Vinblastine Sulphate is methyl (3aR,4R,5S,5aR, IObR,13aR)-
4-acetoxy-3a-ethyl-9-[(5S,7R,9S)-5-ethyl-5-hydroxy-
Chromatographic system 9-methoxycarbonyl-l,4,5,6,7,8,9,10-octahydro-2H-3,
- a stainless steel column 12.5 cm x 4 mm, packed with 7-methanoazacycloundecino[(5,4-b)indol-9-yl]-
octadecylsilane bonded to porous silica (3 /lm) (Such 5-hydroxy-8-methoxy-6-methyl-3a,4,5,5a,6, 11,12,13a-
as Hypersil ODS), octahydro-lH-il1dolizino[8,I-cd]carbazole-5-carboxylate
- mobile phase: a mixture of 1 volume of n-heptylamine, sulphate.
4.7 volumes of glacial acetic acid, 58 volumes of Vinblastine Sulphate contains not less than 95.0 per cent and
acetonitrile and 137 volumes of0.01 Msodiumacetate, not more than 104.0 per cent ofC46HssN409, H2S04, calculated
- flow rate. 0.85 ml per minute, on the dried basis.
- spectrophotometer set at 278 nm,
Category. Anticancer.
injection volume. 10 /ll.
Dose. By intravenous injection, according to the needs ofthe
Inject reference solution (b). The test is not valid unless the patient.
resolution between the peaks due to verapamil and verapamil
impurity A is not less than 2.0. Description. A white or yellowish, amorphous or crystalline
powder; very hygroscopic.
Inject the test solution and reference solution (a). Run the
CAUTION~ Handle Vinblastine Sulphate with great care
chromatogram 4 times the retention time ofthe principal peak.
since it is a potent cytotoxic agent. Avoid contact with eyes;
In the chromatogram obtained with the test solution the area
irritant to tissues.
ofany secondary peak is not more than the area ofthe principal
peak in the chromatogram obtained with reference solution Identification
(a) (0.1 per cent) and the sum ofthe areas of all the secondary
peaks is not more than three times the area of the principal A. Detennine by infrared absorption spectrophotometry (2.4.6).
peak in the chromatogram obtained with reference solution Compare the spectrum with that obtained with vinblastine
(a) (0.3 per cent). Ignore any peak with an area less than 0.5 sulphate RS.
times the area of the principal peak in the chromatogram B. In the Assay, the principal peak in the chromatogram
obtained with reference solution (a) (0.05 per cent). obtained with the test solution corresponds to the peak due
Other tests. Comply with the tests stated under Tablets. to vinblastine sulphate in the chromatogram obtained with
reference solution (b).
Assay. Weigh and powder 20 tablets. Weigh accurately a
C. A 10 per cent w/v solution gives the reaction ofsulphates
quantity of the powder containing 0.1 g of Verapamil
(2.3.1).
Hydrochloride, shake with 150 ml of 0.1 M hydrochloric acid
for 10 minutes, add sufficient 0.1 M hydrochloric acid to Tests
~~produce 200.0ml and filter. Dilute 10.OmLoLthe filtrate to
100.0 ml with water and measure the absorbance of the Appearance of solution. A 0.5 per cent w/v solution in carbon
resulting solution at the maximum at about 278 nm (2.4.7). dioxide-free water is clear (2.4.1), and not more intensely
Calculate the content of C27H3sN204, HCl taking 118 as the coloured than reference solution YS7 (2.4.1).
specific absorbance at 278 nm. pH (2.4.24).3.5 to 5.0, determined in a 0.15 percentw/v solution.

2298

www.webofpharma.com
IP 2010 VINBLASTINE INJECTION

Related substances. In the Assay, the area of any secondary to-noise ratio of the peak in the chromatogram obtained with
peak in the chromatogram obtained with the test solution is reference solution (d) is at least 5.
not greater than that ofthe principal peak in the chromatogram
Calculate the percentage content of C46H5sN409, H 2S04,
obtained with reference solution (c) and the sum of the areas
of any such peaks is not greater than 2.5 times the area of the Vinblastine Sulphate intendedfor use in the manufacture of
principal peak in the chromatogram obtained with reference parenteral preparations without a further appropriate
solution (c). Ignore any peak with an area less than that procedure for the removal ofbacterial endotoxins complies
of the peak in the chromatogram obtained with reference with the following additional requirement.
solution (d).
Bacterial endotoxins (2.2.3). Not more than 10.0 Endotoxin
Loss on drying (2.4.19). Not more than 15.0 per cent, Units per mg.
determined by Method B, on an appropriately calibrated
Vinblastine Sulphate intended for use in the manufacture of
instrument using about 3.0 mg, accurately weighed. Heat the
parenteral preparations without a fitrther appropriate
substance under examination at the rate of 5° per minute
sterilisation procedure complies with the following
between ambient temperature and 200° in a current ofnitrogen
additional requirement.
for chromatography with a flow rate of40 ml per minute. From
the thermogram, determine the accumulated loss in weight Sterility (2.2.11). Complies with the test for sterility.
between ambient temperature and a point on the plateau before
Storage. Store protected from light and moisture in a deep
decomposition is indicated (at about 160°).
freezer (Below -18°). If the material is intended for use in the
Assay. Determine by liquid chromatography (2.4.14). manufacture ofparenteral preparations, it should be stored in
Test solution. Dissolve 0.1 g of the substance under sterile, tamper-evident glass containers and sealed so as to
examination in 100 ml of water. exclude micro-organisms.

Reference solution (a). A solution containing 0.1 per cent Labelling. The label states whether or not the contents are
w/v each of vinblastine sulphate RS and vincristine sulphate suitable for use in the manufacture ofparenteral preparations.
RSin watel:
Reference solution (b). A 0.1 per cent w/v solution of
vinblastine sulphate RS in watel:
Reference solution (c). A 0.002 per cent w/v solution of Vinblastine Injection
vinblastine sulphate RS in watel: Vinblastine Sulphate Injection
Reference solution (d). A 0.0001 per cent w/v solution of Vinblastine Injection is a sterile material consisting of
vinblastine sulphate RS in water. Vinblastine Sulphate with or without auxiliary substances. It
Chromatographic system is filled in a sealed container.
a stainless steel column 25 cm x 4.6 mm, packed with The injection is constituted by dissolving the contents of the
octylsilane bonded to porous silica (5 /lm), (b) a guard sealed container in the requisite amount of sterile 0.9 per cent
column packed with a suitable silica gel placed between w/v solution ofSodium Chloride, immediately before use.
the pump and the injection device,
mobile phase: a mixture of 50 volumes of methanol, The constituted solution complies with the requirements for
38 volumes ofa 1.5 per cent v/v solution of diethylamine Clarity of solution and Particulate matter stated under
adjusted to pH 7.5 with phosphoric acid and 12 volumes Parenteral Preparations (Injections).
of acetonitrile, Storage. The constituted solution should be used immediately
flow rate. 1 ml per minute, after preparation but, in any case, within the period
- spectrophotometer set at 262 om, recommended by the manufacturer.
injection volume. 10 Ill.
Vinblastine Injection contains not less than 90.0 per cent and
NOTE - Store all solutions in ice before use. not more than 11 0.0 per cent of the stated amount of
vinblastine sulphate, C46H5sN409,H2S04.
Inject each solution and record the chromatograms for 3 times
the retention time of the peak due to vinblastine. Usual strength. 10 mg.
The assay is not valid unless the resolution between the peaks The contents of the sealed container comply with the
due to vincristine and vinblastine in the chromatogram requirements stated under Parenteral Preparations
obtained with ref~rence solution (a) is at least 4 and the signal- (Powders for Injection) and with the following requirements.

2299

www.webofpharma.com
VINBLASTINE INJECTION IP 2010

Identification Labelling. The label states (1) the strength in terms of the
weight of dried vinblastine sulphate contained in it; (2) the
CAUTION - Handle Vinblastine Injection with great care names ofauxiliary substances, if any; (3) that the contents are
since it is a potent cytotoxic agent. Avoid contact with eyes; to be used by intravenous injection only; (4) the storage
irritant to tissues. conditions.
A. When examined in the range 230 nm to 360 nm (2.4.7), the
solution obtained in the Assay shows an absorption maximum
at about 267 nm.
Vincristine Sulphate
B. In the test for Related substances, the principal spot in the
chromatogram obtained with the test solution corresponds to
that in the chromatogram obtained with reference solution (a).
C. To 1 ml add 0.2 ml of a freshly prepared 1 per cent w/v
solution of vanillin in hydrochloric acid; a pink colour is
produced in about 1 minute (distinction from Vincristine
Sulphate).

Tests
pH (2.4.24).3.5 to 5.0, deterrnined in a 0.15 per cent w/v solution
of the dried contents.
Mol. Wt. 923.1
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Vinblastine Sulphate is methyl (3aR,4R,5S,5aR, 1ObR, 13aR)
4-acetoxy-3a-ethyl-9-[(5S,7R,9S)- 5-ethyl-5-hydroxy-
Mobile phase. A mixture of80 volumes of toluene, 40 volumes 9-methoxycarbonyl-l,4,5,6,7,8,9,10-octahydro-2H-3,
of chloroform and 6 volumes of diethylamine. 7- methanoazacycloundecino[(5,4-b)indol-9-yl]-6-formyl-5-
Test solution. Dissolve a quantity ofthe contents ofa container hydroxy- 8-methoxy-3a,4,5,5a,6,11,12,13a-octahydro-
in sufficient methanol to produce a solution containing the IH- indolizino[8,I-cd]carbazole-5-carboxylate sulphate.
equivalent of 1 per cent w/v of dried vinblastine sulphate. Vincristine Sulphate contains not less than 95.0 per cent and
Reference solution (a). A 1 per cent w/v solution of vinblastine not more than 104.0 per cent ofC46Hs6N401O,HzS04, calculated
.
sulphate RS in methanol. on the dried basis.

Reference solution (b). A 0.02 per cent w/v solution of Category. Anticancer.
vincristine sulphate RS in methanol. Dose. By intravenous injection, according to the needs of the
Apply to the plate 5 III of each solution. After development, patient.
dry the plate in air and examine in ultraviolet light at 254 nm. Description. A white to slightly yellowish, amorphous or
Any secondary spot in the chromatogram obtained with the crystalline powder; very hygroscopic.
test solution is not more intense than the spot in the
CAUTION- Handle Vincristine Sulphate with great care
chromatogram obtained with reference solution (b).
since it is a potent cytotoxic agent. Avoid contact with eyes;
Bacterial endotoxins (2.2.3). Not more than 10.0 Endotoxin irritant to tissues.
Units per mg of the dried contents.
Identification
Assay. Weigh the contents of 20 sealed containers. Weigh
accurately a quantity of the mixed contents containing about A. Determine by infrared absorption spectrophotometry (2.4.6).
20 mg ofdried vinblastine sulphate and dissolve it in 100.0 ml Compare the spectrum with that obtained with vincristine
of methanol. Dilute 10.0 ml to 100.0 ml with methanol and sulphate RS.
measure the absorbance of the resulting solution at the B. In the Assay, the principal peak in the chromatogram
maximum at about 267 nm (2.4.7). Calculate the content of obtained with the test solution corresponds to the peak due
C<ji;HssN<j09,HzSO<j taking 185· as· thespecificaosoroance -at fo viriciistirie- stiTphafe-iilfhe-chfomafogram-obtafnecfwitIi
267nm. reference solution (b).
Storage. Store in sealed containers in a deep freezer (Below C. A 10 per cent w/v solution gives the reaction for sulphates
-18°). (2.3.1).

2300

www.webofpharma.com
IP 2010 VINCRISTINE SULPHATE

Tests cent). Ignore any peak with an area less than 0.1 per cent the
area ofthe principal peak in the chromatogram obtained with
Appearance ofsolution. A 0.5 per cent w/v in carbon dioxide- reference solution (d).
free water is clear (2.4.1), and not more intensely coloured
than reference solution YS7 (2.4.1). Loss on drying (2.4.19). Not more than 12.0 per cent,
determined by Method B, on an appropriately calibrated
pH (2.4.24).3.5 to 4.5. Determined in a 0.1 per centw/v solution. instrument using about 3.0 mg, accurately weighed. Heat the
Related substances. Determine by liquid chromatography substance under examination at the rate of 50 per minute
(2.4.14). between ambient temperature and 2000 in current of nitrogen
NOTE- Keep the solutions in iced water before use. for chromatography with a flow rate of40 ml per minute.

Test solution. Dissolve about 50.0 mg of the substance under Assay. Determine by liquid chromatography (2.4.14).
examination in 10.0 ml of carbon dioxide-free water. Dilute 1.0 Test solution. Dissolve 0.1 g of the substance under exami-
ml ofthis solution to 5.0 ml with water. nation in 100 ml of water.
Reference solution (a). A 0.1 per cent w/v solution of Reference solution (a). A solution containing 0.1 per cent
vincristine sulphate RS in water. w/v each of vinblastine sulphate RS and vincristine sulphate
Reference solution (b). A 0.1 per cent w/v solution of RSin wate1:
vinblastine sulphate RS in reference solution (a). Reference solution (b). A 0.1 per cent w/v solution of
Reference solution (c). Dilute 1.0 ml of the test solution to vincristine sulphate RS in wate1:
50.0 ml with water. Reference solution (c). A 0.002 per cent w/v solution of
Reference solution (d). Dilute 1.0 ml of reference solution (c) vincristine sulphate RS in water.
to 20.0 ml with water. Reference solution (d). A 0.0001 per cent w/v solution of
Chromatographic system vincristine sulphate RS in wate1:
a stainless steel column 25 cm x 4.6 mm packed with
Chromatographic system
octylsilane bonded to porous silica (5 Ilm),
- a stainless steel column 25 cm x 4.6mm, packed with
mobile phase: A. a 1.5 per cent v/v solution of
octylsilane bonded to porous silica (5 Ilm), (b) a guard
diethylamine, adjusted to pH 7.5 Witl;l orthophosphoric
column packed with a suitable silica gel placed between
acid the pump and the injection device,
B. methanol, - mobile phase: a mixture of70 volumes of methanol and
- a linear gradient programme using the condition given
30 volumes ofa 1.5 per cent v/v solution of diethylamine
below,
adjusted to pH 7.5 with phosphoric acid,
- flow rate. 2 ml per minute,
flow rate. 1 ml per minute,
spectrophotometer set at 297 urn,
spectrophotometer set at 297 urn,
injection volume. 20 Ill.
injection volume. 10 Ill.
Time Mobile phase A Mobile phase B
NOTE - Store all solutions in ice before use.
(min.) (per cent v/v) (per cent v/v)
0-12 38 62 Inject each solution and record the chromatograms for 3 times
the retention time of the peak due to vincristine.
12-27 38-8 62-92
27-29 8-38 92-62 The assay is not valid unless the resolution between the peaks
due to vincristine and vinblastine in the chromatogram
29-34 38 62
obtained with reference solution (a) is at least 4 and the signal-
Inject reference solution (b). The test is not valid unless the to-noise ratio in the peak in the chromatogram obtained with
resolution between the peaks due to vincristine and vinblastine reference solution (d) is at least 5.
is not less than 4.
Calculate the content ofC46Hs6N401O,H2S04'
Inject the test solution and reference solution (c). In the
Vincristine Sulphate intended for use in the manufacture of
chromatogram obtained with the test solution, the area of any
parenteral preparations without a further appropriate
secondary peak is not more than the area ofthe principal peak
procedure for the removal of bacterial endotoxins complies
in the chromatogram obtained with reference solution (c) (2.0
with the following additional requirement.
per cent) and the sum of areas of all the secondary peaks is
.not more than 2.5 times the area of the principal peak in the Bacterial endotoxins (2.2.3). Not more than 62.5 Endotoxin
chromatogram obtained with reference solution (c) (5.0 per Units per mg.

2301

www.webofpharma.com
VINCRISTINE INJECTION IP 2010

Vincristine Sulphate intended for use in the mamifacture of the combined chloroform solutions to dryness at 40°. Add
parenteral preparations without a further appropriate 0.2 ml ofa freshly prepared 1 per cent w/v solution of vanillin
sterilisation procedure complies with the following in hydrochloriC acid to the residue; an orange colour is
additional requirement. produced in about 1 minute (distinction from vinblastine
sulphate).
Sterility (2.2.11). Complies with the test for sterility.
Storage. Store protected from light in a deep freezer (Below Tests
-18°). If the material is intended for use in the manufacture of Appearance of solution. A solution prepared by dissolving
parenteral preparations, it should be stored in sterile, tamper- the contents of a sealed container in 10 ml of carbon dioxide-
evident glass containers and sealed so as to exclude micro- free water is clear (2.4.1).
organisms.
pH (2.4.24). 3.5 to 5.0, determined in a solution containing
Labelling. The label states whether or not the contents are 0.15 per cent w/v solution of the dried contents.
suitable for use in the manufacture ofparenteral preparations.
Related substances. Deteremine by liquid chromatography
(2.4.14).

Vincristine Injection NOTE-Keep the solutions in ice before use.


Test solution. Dissolve the contents of a sealed container in
Vincristine Sulphate Injection
water to produce a solution containing about 0.1 per cent wi
Vrncristine Injection is a sterile material consisting ofVmcristine v of anhydrous vincristine sulphate.
Sulphate with or without auxiliary substances. It is filled in a Reference solution (a). A solution containing 0.1 per cent wi
sealed container. veach of vincristine sulphate RS and vinblastine sulphate
The injection is constituted by dissolving the contents of the RSin water.
sealed container in the requisite amount of sterile 0.9 per cent Reference solution (b). A 0.1 per cent w/v solution of
w/v solution ofSodium Chloride, immediately before use. vincristine sulphate RS in water.
The constituted solution complies with the requirements for Reference solution (c). A 0.002 per cent w/v solution of
Clarity of solution and Particulate matter stated under vincristine sulphate RS in water.
Parenteral Preparations (Injections).
Reference solution (d). A 0.0001 per cent w/v solution of
Storage. The constituted solution should be used immediately vincristine sulphate RS in water.
after preparation but, in any case, within the period recom-
mended by the manufacturer. Chromatographic system
a stainless steel colunrn 25 em x 4.6 mm packed with
Vincristine Injection contains not less than 90;0 per cent and endcapped octylsilane bonded to porous silica (5 /lm),
not more than 110.0 per cent ofthe stated amount ofvincristine (Such as Zorbax C8),
sulphate, C46Hs6N401O,HzS04' mobile phase: a mixture oBO volumes of a 1.5 per cent
Usual strengths. The equivalent of 1 mg, 2 mg and 5 mg of v/v solution of diethylamine adjusted to pH 7.5 with
dried vincristine sulphate. orthophosphoric acid and 70 volumes of methanol,
- flow rate. 1.2 ml per minute,
The contents of the sealed container comply with the
- spectrophotometer set at 297 nm,
requirements stated under Parenteral Preparations
- injection volume. 10 /ll.
(Powders for Injection) and with thefollowing requirements.
Inject reference solution (a). Run the chromatogram 3 times
Identification the retention time of the principal peak.
CAUTION - Handle Vincristine Injection with great care The test is not valid unless the resolution between the peaks
since it is a potent cytotoxic agent. Avoid contact with eyes; due to vincristine and vinblastine in the chromatogram
irritant to tissues. obtained with reference solution (a) is not less than 4 and
A. In the test for Related substances, the principal peak in the signal-to-noise ratio in the principal peak in the chromatogram
chromatogram obtained with the test solution corresponds to obtained with reference solution (d) is not less than 5.
-1:I1arffitl:fecnromatograrh oommedWitnreference solUtion (or
~-,~-"",,-,,~-"."' .. '.'"~~'''''--''''''''''-~''''''~''''' " ,,' "" ~"~'~",,,,,,,-,,,-,,~~~,,,,~.,,,,-,,-,,-,-,,,-,~._,,,,,,,._~,,,,,-,,,,._,,,,'"~~".,''-''''~-'''''

Inject the test solution, reference solution (c) and (d). In the
B. Shake a quantlt,Ycontalning Tmg o{Cifledvincristlne chromatogram obtained with the test solution the area of any
sulphate with 3 ml of chloroform, filter and wash the filter with secondary peak is not more than the ania ofthe principal peak
2 ml of chloroform. Reserve the residue for test D. Evaporate in the chromatogram obtained with reference solution (c)

2302

www.webofpharma.com
IP 2010 VINORELBINE TARTRATE

(2.0 per cent) and th~ sum of the areas of all the secondary Vinorelbine Tartrate contains not less than 98.0 per cent and
peaks is not more than 2.5 times the area ofthe principal peak not more than 102.0 per cent ofC4sHs4N40s.2C4H606, calculated
in the chromatogram obtained with reference solution (c) on the anhydrous basis.
(5.0 percent). Ignore any peak with an area less than the ofthe
Category. Anticancer.
principal peak in the chromatogram obtained with reference
solution (d) (0.1 per cent). Description. A white to yellow or light brown amorphous
powder.
Uniformity of content. The content of anhydrous vincristine
sulphate in each of 10 individual containers as determined in CAUTION - Vinorelbine Tartrate is cytotoxic; extra care
the Assay is not less than 90.0 per cent and not more than required to prevent inhaling particles and exposing the skin
110.0 per cent ofthe average except that in one container the to it.
content may be not less than 80.0 per cent and not more than
120.0 per cent ofthe average.. Identification
Bacterial endotoxins (2.2.3). Not more than 62.5 Endotoxin A. Dissolve 10 mgin 5 ml of water, add 0.5 ml of5 Msodium
Units per mg of dried vincristine sulphate. hydroxide, and extract with 5 ml of methylene chloride. Filter
Assay. Dissolve the contents ofa sealed container in a suitable the organic layer through anhydrous sodium sulphate, and
volume of methanol to produce a solution containing about evaporate to dryness.
0.005 per cent w/v ofanhydrous vincristine sulphate. Measure
Determine by Infrared absorption spectrophotometry (2.4.6).
the absorbance of the resulting solution at the maximum at
Compare the spectrum with that obtained with vinorelbine
297 nm (2.4.7). Calculate the content of C46Hs6N401O,H2S04
tartrate RS treated in the same manner.
taking 177 as the specific absorbance at 297 urn.
B. In the test for Related substances, the principal peakin the
Repeat the procedure with a further nine containers.
chromatogram of the test solution corresponds to that due to
Calculate the average content of C46Hs6N401O,H2S04 in the vinorelbine tartarate in the chromatogram obtained with the
sealed container. refrence solution (a).
Storage. Store in sealed containers in a deep freezer (Below C. It gives the reactions for tartrate (2.3.1).
-18°).
Labelling. The label states (1) the strength in terms of the Tests
weight of dried vincristine sulphate contained in it; (2) the
Appearance of solution. A 1.0 per cent w/v solution is clear
names of auxilary substances, if any; (3) that the contents are
(2.4.1).
to be used by intravenous injection only; (4) the storage
conditions. Light absorption (2.4.7). The absorbance of 1.0 per cent w/v
solution, at about 420 urn is not more than 0.03.
pH (2.4.24).3.3 to 3.8, determined on a 1.0 per cent w/v solution.
Vinorelbine Tartrate Related substances. Detennine by liquid chromatography
(2.4.14).
Test solution. Dissolve 35 mg of the substance under
examination in 25 ml ofthe mobile phase.
Reference solution (a). A 0.14 per cent w/v solution of
H ,oH . vinorelbine tartrate RS in the mobile phase.
COOH
,2HOOCXyOH
H Reference solution (b). Dilute 1 ml of the reference solution
(a) to 100 ml with the mobile phase.
Chromatographic system as described under Assay.
C4sHs4N40S, (C~606)2 Mol Wt. 1079.1 Inject reference solution (a). The test is not valid unless the
Vinorelbine Tartrate is 3' ,4' -didehydro-4' -deoxy-C' - column efficiency is not less than 2000 theoretical plates and
norvincaleukoblastine ditartrate. the tailing factor is not more than 2.0.
Vinorelbine Tartrate is the ditartrate salt ofvinorelbine,a Inject the test solution and reference solution (b). In the
semisynthetic Vinca alkaloid; structurally relate to chromatogram obtained with the test solution, the area ofany
vinblastine. secondary peak is not more than 0.3 times the area ofthe peak

2303

www.webofpharma.com
VINORELBINE TARTRATE IP 2010

in the chromatogram obtained with reference solution (b) Vinorelbine Injection contains not less than 90.0 per cent and
(0.3 per cent) and the sum of areas of all the secondary not more than 110.0 per cent of the stated amount of
peaks is not more than the area of the peak in the vinorelbine, C4sHs4N40g.
chromatogram obtained with reference solution (b)
Usual strength. 10 mg per ml.
(1.0 per cent). Ignore any secondary peak having area less
than 0.1 per cent. Description. A clear, colourless to pale yellow solution.

Sulphated ash (2.3 .18). Not more than 0.1 per cent. Identification
Water (2.3.43). Not more than 4.0 per cent, determined on 0.1 g.
A. In the Assay, the principal peak in the chromatogram
Bacterial endotoxins (2.2.3). Not more than 1.5 Endotoxin Unit obtained with the test solution corresponds to the peak in the
per mg of vinorelbine base. chromatogram obtained with the reference solution.
Microbial contamination (2.2.9). Total viable aerobic count, B. When examined in the range 220 nm to 380 nm (2.4.7), a
not more than 100 cfu per g, total combined molds and yeast solution containing 0.01 per cent w/v ofVinorelbine Tartrate,
does not exceed 50 cfu per g. It also meets the requirement of exhibits the maxima at about 267 nm.
the tests for the absence of Staphylococcus aureus,
Pseudomonas aeruginosa, Salmonella species, Escherichia Tests
coli, Enterobacteria and Closteridia. pH (2.4.24).3.0 to 3.8.
Assay. Determine by liquid chromatography (2.4.14). Light absorption. The absorbance of the injection at about
Test solution. Dissolve 35 mg of the substance under 420 nm (2.4.7), is not more than 0.06.
examination in 25.0 ml ofmobile phase. Related substances. Determine by liquid chromatography
Reference solution. A 0.14 per cent w/v solution ofvinorelbine (2.4.14).
tartrate RS in mobile phase. Test solution. Accurately measured volume of injection
containing 10 mg ofVrnorelbine, dilute to 10 ml with the mobile
Chromatographic system
phase.
a stainless steel column 15 cm x 3.9 mm, packed with
octadecylsilane bonded to porous silica (5 /lm), Reference solution (a). A 0.14 per cent w/v solution of
column temperature 40°, vinorelbine tartrate RS in the mobile phase.
mobile phase: 62 volumes of methanol containing Reference solution (b). Dilute 1 m1 of the reference solution
1.22 g of sodium I-decane sulphonate and 38 volumes (a) to 100 ml with the mobile phase.
of phosphate buffer solution, prepared by dissolving
6.9 g of monobasic sodiumphosphate in 900 ml of water, Chromatographic system as described under Assay.
adjust the pH to 4.2 with orthophosphoric acid and Inject reference solution (a). The test is not valid unless the
dilute to 1000 ml with water, column efficiency is not less than 2000 theoretical plates and
flow rate. 1 ml per minute, the tailing factor is not more than 2.0.
- spectrophotometer set at 267 nm,
- injection volume. 20 Ill. Inject the test solution and reference solution (b). Run the
chromatograms three times the retention time ofthe peak due
Inject the reference solution. The test is not valid unless the to vinorelbine. In the chromatogram obtained with the test
relative standard deviation for replicate injections is not more solution, the area of any secondary peak is not more than
than 2.0 per cent. 0.2 times the area of the peak in the chromatogram obtained
Inject the test solution and reference solution. with reference solution (b) (0.2 per cent) and the sum ofareas
Calculate the content ofC4sHs4N40g.2C4H606. of all the secondary peaks is not more than twice the area of
the peak in the chromatogram obtained with reference solution
Storage. Store protected from light, in a deep freezer (below - (b) (2.0 per cent).
18°).
Other tests. Complies with the tests stated under Parenteral
Preparation (Injections).
Yiuordbixte Illle_cJion . Bacterialendotoxins(2.2.3). Notmore than 3.0 EndotoxinDnit
per mg ofvinorelbinetart.rate.
Vinorelbine Tartrate Injection
Sterility (2.2.11). Complies with the test for sterility.
Vinorelbine Injection is a sterile solution ofvinorelbine tartrate
in Water for Injection. Assay. Determine by liquid chromatography (2.4.14).

2304

www.webofpharma.com
IP 2010 VITAMIN A CONCENTRATE OIL

Test solution. Accurately measured volume of injection B. Determine by thin-layer chromatography (2.4.17), coating
containing 10 mg ofVinorelbine, diluted to 10.0 ml with water. the plate with silica gel G
Reference solution. A 0.14 per cent w/v solution ofvinorelbine Mobile phase. A mixture of 80 volumes of cyclohexane and
tartrate RS in water. 20 volumes of ether.
Chromatographic system Test solution. Prepare a solution containing 5 Units per fll of
- a stainless steel column 15 cm x 3.9 mm, packed with the substance under examination in cyclohexane.
octadecylsilane bonded to porous silica (5 flm),
column temperature 40°, Reference solution (a). Prepare a solution containing
mobile phase: a mixture of 62 volumes of methanol 5 Units per fll of retinyl acetate RS in cyclohexane.
containing 1.22 g of sodiuml-decane sulphonate and Reference solution (b). Prepare a solution containing
38 volumes of phosphate buffer solution prepared by 5 Units per fll of retinyl propionate RS in cyclohexane.
dissolving 6.9 g of monobasic sodium phosphate in
900 ml of watel; adjusted the pH to 4.2 with ortho- Reference solution (c). Prepare a solution containing
phosphoric acid and dilute to 1000 ml with water, 5 Units per fll of retinyl palmitate RS in cyclohexane.
flow rate. 1 ml per minute, Apply to the plate 2 fll of each solution. After development,
spectrophotometer set at 267 TIm, dry the plate in air, spray with antimony trichloride solution.
injection volume. 20 fll. The principal spot or spots in the chromatogram obtained
Inject the reference solution. The test is not valid unless the with the test solution correspond to one or more of the spots
relative standard deviation for replicate injection is not more in the chromatograms obtained with reference solutions (a),
than 2.0 per cent. (b) and (c).

Inject the test solution and the reference solution. C. Dissolve a quantity containing 10 to 15 Units in 1 mlof
chloroform and add 5 ml of antimony trichloride solution; a
Calculate the content ofC4sHs4N40g. transient bright blue colour is produced immediately.
Storage. Store protected from light, in a single-dose container,
in a refrigerator (2° to 8°) ; do not freeze. Tests
Acid value (2.3.23). Not more than 2.0, determined on 2.0 g.
Vitamin A Concentrate Oil Peroxides. Add 0.3 g to 25.0 ml ofa mixture of6 volumes of
Synthetic Vitamin A Concentrate (Oily Form); Synthetic toluene and 4: volumes of methanol (solution A). Add in a
Retinol Concentrate (Oily Form). test-tube, in the following order, mixing after each addition,
0.3 ml ofa 1.8 percent w/v solution of ammonium thiocyanate,
Vitamin A Concentrate Oil consists of an ester or a mixture of 10.0 ml of methanol, 0.3 ml offerrous sulphate solution and
esters ofretinol (as acetate, propionate or palmitate) prepared 15.0 ml of toluene and add 1.0 ml of solution A. The colour
by synthesis. It may be diluted with a suitable vegetable oil. It produced after 5 minutes is not more intense than that obtained
may contain suitable stabilising agents such as antioxidants. in a solution prepared at the same time and in the same manner
Vitamin A Concentrate Oil contains not less than 500,000 Units but using a solution prepared in the following manner in place
of Vitamin A per g, and not less than 95.0 per cent and not of solution A. Add 1.0 ml of a 27.0 per cent w/v solution of
more than 110.0 per cent of the stated number of Units of ferriC chloride hexahydrate to 99 ml ofa mixture of6 volumes
Vitamin A per g. of toluene and 4 volumes of methanol and dilute 2.0 ml to
100.0 ml with the same solvent mixture.
Category. Antixerophthalmic vitamin.
Dose. Prophylactic, 5000 Units ofVitamin A, daily; therapeutic, Assay. Carry out the assay ofvitamin A, Method A (2.3.41).
10,000 to 200,000 Units ofVitamin A, daily. Storage. Store protected from light, in well-filled containers.
Description. A yellow to brownish yellow, oily liquid; odour, Once the container has been opened, its contents should be
faint and characteristic. used as soon as possible; any part ofthe contents not used at
once should be protected by an atmosphere of an inert gas.
Identification
Labelling. The label states (1) the number ofUnits ofVitamin
A. When examined in the range 230 TIm to 360 TIm (2.4.7), a A per g; (2) the name ofthe ester or esters; (3) the name(s) of
solution in 2-propanol containing 10 to 15 Units per ml shows any added stabilising agent(s); (4) the method of restoring
an absorption maximum at about 325 TIm to 327 nm. the solution if partial crystallisation has occurred.

2305

www.webofpharma.com
VITAMIN A CONCENTRATE POWDER IP 2010

Vitamin A Concentrate Powder Reference solution (c). Prepare a solution containing 5 Units
per III of retinyl palmitate RS in cyclohexane.
Synthetic Vitamin A Concentrate (Powder Form);
Apply to the plate 2 III of each solution. After development,
Synthetic Retinol Concentrate (Powder Form).
dry the plate in air, spray with antimony trichloride solution.
Vitamin A Concentrate Powder consists ofan ester or a mixture The principal spot or spots in the chromatogram obtained
ofesters ofretinol (as acetate, propionate or palmitate) prepared with the test solution correspond to one or more of the spots
by synthesis and dispersed in a matrix of Gelatin, Acacia or in the chromatograms obtained with reference solutions (a),
any other suitable material. It may contain suitable stabilising (b) and (c).
agents such as antioxidants.
C. Dilute 2 ml of sblution A to 50 ml with n-pentane and
Vitamin A Concentrate Powder contains not less than evaporate 1 ml of the solution to dryness in a current of
250,000 Units of Vitamin A per g, and not less than 95.0 per nitrogen. Dissolve the residue in 1 ml of chloroform and add
cent and not more than 115.0 per cent ofthe stated number of 5 ml of antimony trichloride solution; a transient bright blue
Units ofVitaminA per g. colour is produced irilrnediately.
Category. Antixerophthalmic vitamin.
Tests
Dose. Prophylactic, 5000 Units ofVitamin A, daily; therapeutic,
10,000 to 200,000 Units ofVitamin A, daily. In multi-vitamin Related substances and degradation products. Using the
oral preparations, prophylactic, 1600 to 2500 Units ofVitamin relative absorbances obtained in the Assay, the ratio
A; therapeutic, 5000 to 10,000 Units ofVitamin A. A 30o/Am is not more than 0.612 and the sum of the ratios
A 30o /A m and A 350 /A m is not more than 1.054, where
Description. A yellowish powder usually in the form ofpellets A 300 , Am and A 350 are the absorbances measured at about
or particles ofalmost uniform size. 300 nm, 325 urn and 350 urn respectively.

Identification Assay. Carry out the assay of vitamin A, Method B (2.3.41),


adding 5 ml of water to the mixture for saponification, using
To a quantity containing 50,000 Units ofVitamin A, add 1.5 ml 2-propanol as the blank and taking 0.612 as the maximum
of 2 M ammonia previously heated to 60° and heat in a water- value of the ratio A 300/ Am.
bath at 60°, shaking occasionally. After 10 minutes add 40 ml Storage. Store protected from light. Once the container has
of ethanol, dilute to 200 ml with ether and shake. Allow to been opened, its contents should be used as soon as possible;
stand for a few minutes and use the supernatant liquid any part of the contents not used at once should be protected
(solution A) for the following tests. by an atmosphere of an inert gas.
Certain samples may not react sufficiently during the course Labelling. The label states (1) the number ofUnits ofVitamin
of the above treatment In such cases the volume of solution A per g; (2) the name ofthe ester or esters; (3) the name ofthe
A used in the following tests should be increased which may principal excipient or excipients used; (4) the name of any
be as much as lO-fold. added stabilising agents.
A. When examined in the range 230 urn to 360 urn (2.4.7), a
solution in2-propanol containing 10 to 15 Units perml shows
an absorption maximum at about 325 urn to 327 urn.
B. Determine by thin-layer chromatography (2.4.17), coating Water-Miscible Vitamin A Concentrate
the plate with silica gel G.
Synthetic Vitamin A Concentrate (Water-dispersible
Mobile phase. A mixtu+e of 80 volumes of cyclohexane and
20 volumes of ether.
Form); Synthetic Retinol Concentrate (Water-dispersible
Form).
Testsolution. Evaporate 10 ml of solution A to dryness in a
Water-miscible Vitamin AConcentrate consists of an ester or
current of nitrogen and dissolve the residue in 0.5 ml of
a mixture ofesters ofretinol (as acetate, propionate or palmitate)
cyclohexane.
prepared by synthesis to which suitable solubilisers have
Reference solution (aJ .Prepareasolution-containingSUnits lJeenadded: lfifiaycolltainSuitaole-stalJilising· agentssuch
per III of retinyl acetate RS in cyclohexane. as antimicrobial preservatives and antioxidants.
Reference solution (b). Prepare 5 Units per III of retinyl Water-miscible Vitamin A Concentrate contains not less than
propionate RS in cyclohexane. 100,000 Units of Vitamin A per g, and not less than 95.0 per

2306

www.webofpharma.com
IP 20ID VITAMINS A AND D CAPSULES

cent and not more than 115.0 per cent of the stated number of Tests
Units ofVitamin A per g.
Water miscibility. Mix about 1 g with 10 ml of water previously
Category. Antixerophthalmic vitamin. heated to 50° and cool to 20°. Immediately after cooling, a
Dose. Prophylactic, 5000 Units ofVitamin A, daily; therapeutic, uniform, slightly opalescent and slightly yellow dispersion is
10,000 to 200,000 Units ofVitamin A, daily.. In multi-vitamin obtained.
oral preparations, prophylactic, 1600 to 2500 Units ofVitamin Related substances and degradation products. Using the
A; therapeutic, 5000 to 10,000 Units ofVitamin A. relative absorbances obtained in the Assay, the ratio
Description. A yellow or yellowish liquid of variable A 3001A325 is not more than 0.618 and the sum of the ratios
opalescence and viscosity; odour, characteristic. Highly A 3001A 325 and A 3soIA325 is not more than 1.060, where A300, A325
concentrated solutions may become cloudy at low and A 3S0 are the absorbances measured at about 300 urn, 325
temperatures or gel at room temperature. urn and 350 nm respectively.
Assay. Carry out the assay of vitamin A, Method B (2.3.41),
Identification using 2-propanol as the blank and taking 0.618 as the maximum
value of the ratio A 30o /A 325 •
To a quantity containing about 10,000 Units ofVitamin A, add
5 ml of water and homogenise. Add 5 ml of ethanol (95 per Storage. Store protected from light at the temperature stated
cent) and 20 ml of n-pentane and shake vigorously for on the label. Once the container has been opened, its contents
30 seconds. Allow to stand for a few minutes and use the should be used as soon as possible; any part of the contents
supernatant liquid (solution A) for the f?llowing tests. not used at once should be protected by an atmosphere of an
inert gas.
A. Dilute solution A with sufficient 2-propanol so that the
absorbance at the wavelength of maximum absorption is Labelling. The label states (1 )the number ofUnits ofVitamin
0.3 to 0.7 (2.4.7). A per g; (2) the name ofthe ester or esters; (3) the name ofthe
principal excipient or excipientsused; (4) the temperature at
When examined in the range 230 run to 360 urn (2.4.7), the which it should be stored.
solution shows an absorption maximum at 325 to 327 nm.
B. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G. Vitamins A.and D Capsules
Mobile phase. A mixture of 80 volumes of cyclohexane and Vitamins A and D Capsules contain Vitamin A Oil and a source
20 volumes of ether. of vitamin D such as Cholecalciferol or Ergocalciferblin an
Test solution. Evaporate 10 ml of solution A to dryness in a edible vegetable oil.
current of nitrogen and dissolve the residue in 0.5 ml of Vitamins A and D Capsules contain not less than 90.0 per cent
cyclohexane. of the stated number of Units of vitamin A and vitamin D.
Reference solution (a). Prepare a solution containing 5 Units Category. Antixerophthalmic and antirachitic vitamins.
per III of retinyl acetate RS in cyclohexane.
Dose. Prophylactic, 1600 to 2500 Units ofVitamin A and 100 to
Reference solution (b). Prepare a solution containing 5 Units 200 Units ofvitamin D once a day; therapeutic, 5000 to 10,000
per III of retinyl propionate RS in cyclohexane. Units ofVitamin A and 400 to 1000 Units ofvitamin D once a
day.
Reference solution (c). Prepare a solution containing 5 Units
per III of retinyl palmitate RS in cyclohexane. Tests
Apply to the plate 2 III of each solution. After development, Other tests. Comply with the tests stated under Capsules.
dry the plate in air, spray with antimony trichloride solution.
The principal spot or spots in the chromatogram obtained Assay. For vitamin A - Weigh accurately a portion of the
with the test solution correspond to one or more of the spots mixed contents of20 capsules containing about 500 Units of
in the chromatograms obtained with reference solutions (a), Vitamin A and carry out the assay of vitamin A, Method A
(b) and (c). (2.3.41). .

C. Evaporate 0.1 ml of solution A to dryness in a current of For vitamin D - Weigh accurately a portion of the mixed
nitrogen, dissolve the residue in 1 ml of chloroform and add contents of20 capsules containing about 5000 Units ofvitamin
5 ml of antimony trichloride solution; a transient bright blue D and carry out the assay ofvitamin D (2.3.42).
colour is produced immediately. Storage. Store protected from light and moisture.

2307

www.webofpharma.com
CONCENTRATED VITAMIN D SOLUTION IP 2010

Labelling. The label states the number of Units of vitamin A Labelling. The label states (1) the number ofUnits ofvitamin
and vitamin D per capsule. D per g; (2) the storage conditions; (3) the nature and
concentration of any stabilising agent added.

Concentrated Vitamin D Solution


Concentrated Vitamin D Solution is a solution of Concentrated Vitamins A and D
Cholecalciferol or Ergocalciferol in an edible vegetable oil. It Solution
may contain suitable stabilising agents such as antioxidants.
Concentrated Vitamins A and D Solution is a solution of
Concentrated Vitamin D Solution contains not less than 10,000 Vitamin A Oil and a source ofvitamin D such as Cholecalciferol
Units ofvitamin D and not less than 90.0 per cent ofthe stated or Ergocalciferol in an edible vegetable oil. It may contain
number ofUnits ofvitamin D. suitable stabilising agents such as antioxidants.
Category. Antirachitic vitamin. Concentrated Vitamins A and D Solution contains not less
Dose. Prophylactic, 400 to 1000 Units daily; therapeutic, 5000 than 50,000 Units ofvitamin A and 5000 Units ofvitamin D per
to 50,000 Units daily. g, and not less than 90.0 per cent and not more than 110.0 per
cent ofthe stated number ofUnits ofVitamin A and vitamin D
Description. A pale yellow to yellow, oily liquid; odour, faint perg.
but not rancid.
Category. Antixerophthalmic and antirachitic vitamins.
Identification Dose. 2500 to 25,000 Units ofVitamin A and 250 to 2500 Units
ofvitamin D daily.
Dissolve a quantity containing about 1000 Units ofvitamin D
in I ml of chloroform and add 10 ml of antimony trichloride Tests
solution; a pinkish red colour appears at once.
Acid value (2.3.23). Not more than 2.5, determined on 2.0 g.
Tests Assay. For vitamin A ~ Carry out the assay of vitamin A,
Acid value (2.3.23). Not more than 2.5, determined on 2.0 g. MethodA (2.3.41).

Assay. Carry out the assay ofvitamin D (2.3.42). For vitamin D - Carry out the assay ofvitamin D (2.3.42).

Storage. Store protected from light, in well-filled containers at Storage. Store protected from light and moisture.
a temperature of 6° to 15°. The contents ofan opened container Labelling. The label states (1) the number ofUnits ofvitamin
should be used as soon as possible. A and vitamin D per gram; (2) the storage conditions.

2308

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

w
Warfarin Sodimn 2311
Warfarin Sodimn Clathrate 2312
Warfarin Tablets 2313
Purified Water 2314
Water for Injections 2315
Water for Injections in Bulle 2315
Sterile Water for Injections 2316
Wool Fat 2317
Hydrous Wool Fat 2318

2309

www.webofpharma.com
www.webofpharma.com
IP 2010 WARFARIN SODIUM

Warfarin Sodium Phenolic ketones. Absorbance ofa 12.5 per cent w/v solution
in a 5.0 per cent w/v solution of sodium hydroxide at the
maximum at about 385 urn, measured within 15 minutes of
preparation, is not more than 0.20 (2.4.7).
Related substances. Determine by liquid chromatography
(2.4.14).
Solvent mixture. 25 volumes of methanol and 75 volumes of
o wate/:

Mol. Wt. 330.3 Test solution. Dissolve 40 mg of the substance under


examination in 50.0 ml ofthe solvent mixture.
Warfarin Sodium is sodium 2-oxo-3-[(1RS)-3-oxo-l-phenyl-
butyl]-2H-l-benzopyran-4-0Iate. Reference solution (a). Dissolve 2 mg each of of 4-
hydroxycoumarin (warfarin impurity B) and benzalacetone
Warfarin Sodium contains not less than 98.0 per cent and not (warfarin impurity C) in 25 ml of methanol and dilute to 100 ml
more than 102.0 per cent of CI9HlSNa04, calculated on the with wate/:
anhydrous basis.
Reference solution (b). Dilute 1.0 ml of the test solution to
Category. Anticoagulant. 100.0 rnl with the solvent mixture. Dilute 1.0 ml ofthis solution
Dose. 10 to 15 mg. to 10.0 ml with the solvent mixture.
Description. A white powder; hygroscopic. Reference solution (c). A 0.08 per cent w/v solution of wa/1arin
sodium RS in the solvent mixture.
Identification
Chromatographic system
Test A may be omitted if tests B, C, D and E are carried out. - a stainless steel COIUlllil 25 cm x 4.0 mm, packed with
Tests Band D may be omitted if tests A, C and E are carried spherical nitrile silica gel (5 /lm),
out. mobile phase: a mixture of 1 volume of glacial acetic
acid, 25 volumes of acetonitrile and 75 volumes of water,
A. Dissolve 1 gin 25 ml of water, add 2 ml of 2 M hydrochloric
flow rate. 1.5 ml per minute,
acid and filter. Wash the residue with water and dry over
spectrophotometer set at 260 urn,
phosphorus pentoxide.
- injection volume. 20 Ill.
On the residue, determine by infrared absorption
Inject reference solution (a). The test is not valid unless the
spectrophotometry (2.4.6). Compare the spectrum with that
resolution between the peaks due to warfarin impurity Band
obtained with warfarin sodium RS or with the reference
warfarin impurity C is not less than 2.0. The relative retention
spectrum ofwarfarin sodium.
time with reference to warfarin for warfarin impurity B is about
B. In the test for Related substances, the principal peak in the 0.4 and for warfarin impurity C is about 0.6.
chromatogram obtained with the test solution corresponds to
Inject the test solution and reference solution (b). Run the
the principal peale in the chromatogram obtained with reference
chromatogram twice the retention time ofthe principal peak.
solution (c).
In the chromatogram obtained with the test solution the area
C. Dissolve 1 g in 10 ml of water, add 5 ml of nitric acid and ofthe peak due to warfarin impurity B and warfarin impurity C
filter. To the filtrate add 2 ml ofpotassium dichromate solution, is not more than the area of the principal peak in the
shake for 5 minutes and allow to stand for 20 minutes; the chromatogram obtained with reference solution (b) (0.1 per
solution i.s not greenish blue when compared with a blank. cent) and the area of any other secondary peak is not more
D. The residue obtained in test A, after washing with water than the area of the principal peak in the chromatogram
and drying at 105°, melts at 159 0 to 163 0 (2.4.21). obtained with reference solution (b) (0.1 per cent). The sum of
all the secondary peaks is not more than 3 times the area ofthe
E. The filtrate obtained in testA gives the reactions of sodium principal peak in the chromatogram obtained with reference
salts (2.3.1). solution (b) (0.3 per cent). Ignore any peak with an area less
Tests than 0.5 times the area of the principal peak in the
chromatogram obtained with reference solution (b) (0.05 per
Appearance of solution. A 5.0 per cent w/v solution is clear cent).
(2.4.1), and colourless (2.4.1).
Water (2.3.43). Not more than 4.0 per cent, determined on
pH (2.4.24). 7.6 to 8.6, determined ina 1.0 per centw/v solution. 0.75g.

2311

www.webofpharma.com
WARFARIN SODIUM CLATHRATE IP 2010

Assay. Weigh accurately about 0.1 g and dissolve in sufficient Tests


0.01 M sodium hydroxide to produce 100.0 m1. Dilute 10.0 ml
Appearance of solution. A 5.0 per cent w/v solution is clear
to 100.Oml with 0.01 Msodium hydroxide and dilute 5.0 ml to
(2.4.1), and colourless (2.4.1).
50.0 ml with 0.01 M sodium hydroxide. Measure the
absorbance of the resulting solution at the maximum at about pH (2.4.24).7.2 to 8.3, determined in a 1.0 per cent w/v solution.
308 run (2.4.7). Calculate the content ofCI9HIsNa04 taking 431
Phenolic ketones. Absorbance ofa 12.5 per cent w/v solution
as the specific absorbance at 308 nm.
in a 5.0 per cent w/v solution of sodium hydroxide at the
Storage. Store protected from light and moisture. maximum at about 385 nm, measured within 15 minutes of
preparation, is not more than 0.20 (2.4.7).
Related substances. Determine by liquid chromatography
(2.4.14).
Warfarin Sodium Clathrate Solvent mixture. 25 volumes of methanol and 75 volumes of
Warfarin Sodium Clathrate is a clathrate form of Warfarin wate/:
Sodium consisting principally of Warfarin Sodium and Test solution. Dissolve 40 mg of the substance under
Isopropyl Alcohol in a 2: 1 molecular ratio. examination in 50.0 ml ofthe solvent mixture.
Warfarin Sodium Clathrate contains not less than 97.0 per Reference solution (a). Dissolve 2 mg each of 4-
cent and more than 102.0 per cent ofCI9HIsNa04 calculated on hydroxycoumarin (warfarin impurity B) and benzalacetone
the anhydrous, isopropyl alcohol-free basis and not less than (warfarin impurity C) in 25 ml of methanol and dilute to 100 ml
8.0 per cent and not more than 8.5 per cent ofisopropy alcohol. with water.
Category. Anticoagulant. Reference solution (b). Dilute 1.0 ml ofthe test solution to
Dose. 10 to 15 mg. 100.0 ml with the solvent mixture. Dilute 1.0 ml ofthis solution
to 10.0 ml with the solvent mixture.
Description. A white crystalline powder; hygroscopic.
Rejerencesolution (c). AO.08 percentw/v solution ofwaifarin
sodium RS in the solvent mixture.
Identification
Chromatographic system
Test A may be omitted if test B, C, D and E are carried out. - a stainless steel column 25 cm x 4.0 mm, packed with
Tests B ai7d D may be omitted if tests A, C and E are carried spherical nitrile silica gel (5 ).un),
out. - mobile phase: a mixture of 1 volume of glacial acetic
A. Dissolve about 1 g in 25 ml of water, add 2 ml of 2 M acid, 25 volumes of acetonitrile and 75 volumes of water,
- flow rate. 1.5 ml per minute,
hydrochloric acid and filter. Wash the precipitate 5 to 6 times
with water. Dry the residue over phosphorus pentoxide. - spectrophotometer set at 260 nm,
injection volume. 20 Ill.
On the residue, determine by infrared absorption
Inject reference solution (a). The test is not valid unless the
spectrophotometry (2.4.6). Compare the spectrum with that
resolution between the peaks due to warfarin impurity Band
obtained with waifarin sodium RS or with the reference
warfarin impurity C is not less than 2.0. The relative retention
spectrum ofwarfarin sodium.
time with reference to warfarin for warfarin impurity B is about
B. In the test for Related substances, the principal peak in the 0.4 and for warfarin sodium impurity C is about 0.6.
chromatogram obtained with the test solution corresponds to
Inject the test solution and reference solution (b). Run the
the principal peak in the chromatogram obtained with reference
chromatogram twice the retention time ofthe principal peak.
solution (c).
In the chromatogram obtained with the test solution the area
C. Dissolve 1 g in 10 ml of water, add 5ml of nitric acid and ofthe peak due to warfarin impurity B and warfarin impurity C
filter. To the filtrate, add 2 ml ofpotassium dichromate solution, is not more than the area of the principal peak in the
shake for 5 minutes and allow to stand for 20 minutes; the chromatogram obtained with reference solution (b) (0.1 per
solution is not greenish-blue when compared with a blank. cent) and the area of any other secondary peak is not more
...... ~D: Tlfefesidue-615tainedinlestA, aftefwasningwithwater .. thl:!nthe.l:!rl:1!Qf tl1l:principl:!l Peak in tl1e CJ:JIQ!1!!ltograJ;!1
obta-ined \\lith. referencesolutio.n(~) (O.lpl;lr cl;lnt). The sulIlo.f
and drying at105° melts at 159° to 163°.
all the secondary peaks is not more than 3 times the area ofthe
E. The filtrate obtained in testA gives the reactions of sodium principal peak in the chromatogram obtained with reference
salts (2.3.1). solution (b) (0.3 per cent). Ignore any peak with an area less

2312

www.webofpharma.com
IP 2010 WARFARIN TABLETS

than 0.5 times the area ofthe principal peak in the chromatogram Identification
obtained with reference solution (b) (0.05 per cent).
A. Extract a quantity ofthe powdered tablets containing 0.1 g
Water (2.3.43). Not more than 4.0 per cent, determined on of Warfarin Sodium with 30 ml of water, add 0.1 ml of 2 M
0.75g. hydrochloric acid, filter, wash the precipitate with water and
Isopropyl alcohol. Detennine by gas chromatography (2.4.13). dry. Wann the residue gently with 3 ml of ethanol (95 per
cent), filter and add the filtrate to 25 .ml of water containing
Test solution. Dissolve 0.5 g of the substance under 0.1 ml of 2 M hydrochloric acid. Filter, wash the precipitate
examination in sufficient water to produce 10 ml. with water and dry it at 105°.
Reference solution (a). A solution containing 5.0 per cent On the residue, determine by infrared absorption
w/v of the substance under examination and 0.5 per cent v/v spectrophotometry (2.4.6). Compare the spectrum with that
of propan-1-o1 (internal standard). obtained with waljarin sodium RS or with the reference
spectrum ofwarfarin sodium.
Reference solution (b). A solution containing 0.5 per cent v/v
each of propan-2-o1 and the internal standard. B. The final residue obtained in test A melts at about 159°
(2.4.21).
Chromatographic system
a glass column 1.5 m X 4 mm, packed with porous polymer
beads (125 to 150 mm) (Such as Porapak Q), Tests
temperature: Related substances. Detennine by thin-layer chromatography
column 150°, (2.4.17), coating the plate with silica gel GF254.
inlet port at 180° and detector at 200 0 ,
- flow rate. 40 Jill per minute ofthe carrier gas. Mobile phase. A mixture of 50 volumes of chloroform,
50 volumes of cyclohexane and 20 volumes of glacial acetic
The column temperature may be varied so that the resolution, acid.
R, between propan-l-ol and propanol-2 01 is not less than 2.0,
the tailing factor. T, for the propan-2-01 is not less than 2.0 the Test solution. Shake a quantity of the powdered tablets
containing 40 mg ofWarfarin Sodium with 30 ml of water for
tailing factor T, for the propan-2-01 peak is not more than 1.5
15 minutes, add 0.1 ml of hydrochloric acid and extract with
and the relative standard deviation ofthe ratio ofthe area due
three quantities, each of 10 ml, of chloroform, drying each
to the peak ofproptl1101-2-01 to that due to propan-l-ol for five
extract with anhydrous sodium sulphate. Evaporate the
replicate injections of reference solution (b) is not more than
combined extracts at a temperature not exceeding 40° and
2.0 per cent.
dissolve the residue in 2 ml of acetone.
Calculate the content of isopropyl alcohol.
.Reference solution (a). Dilute I volume of the test solution to
Assay. Weigh accurately about 0.1 g and dissolve in sufficient 200 volumes with acetone.
0.01 M sodium hydroxide to produce 100.0 ml. Dilute 10.0 ml Reference solution (b). A 0.002 per cent w/v solution of
to 100.0 ml with 0.01 M sodium hydroxide and dilute 5.0 ml to (E)-4-phenylbut-3-en-2-one in acetone.
50.0 ml with 0.01 M sodium hydroXide. Measure the
absorbance ofthe resulting solution at the maximum at about Reference solution (c). A 0.02 per cent w/v solution of
308 urn (2.4.7). Calculate the content ofCI9HIsNa04 taking 431 4-hydroxycoumarin in acetone.
as the specific absorbance at 308 nm. Apply to the plate 20 III of each solution. After development,
Storage. Store protected from light and moisture. dry the plate in air and examine immediately in visible light
noting the position ofany coloured spots and then examine in
ultraviolet light at 254 urn, ignoring any spot that was noted in
visible light. Any spots corresponding to (E)-4-phenylbut-
3-en-2-one and 4-hydroxycoumarin in the chromatogram
~arfarin 1rablets obtained with the test solution are not more intense than the
spots in the chromatograms obtained with reference
Warfarin Sodium Tablets solutions (b) and (c) respectively and any other secondary
Warfarin Tablets contain not less than 92.5 per cent and not spot is not more intense than the spot in the chromatogram
more than 107.5 per cent of the stated amount of warfarin obtained with reference solution (a).
sodium, C,9H,sNa04.
Uniformity of content. Comply with the test stated under
Usual strengths. I mg; 3 mg; 5 mg; 10 mg. Tablets.

2313

www.webofpharma.com
WARFARIN TABLETS IP 2010

Detennine by liquid chromatography (2.4.14). Purified Water


Testsolution. Shake one tablet with 10 ml of 0.01 M sodium
Mol. Wt. 18.0
hydroxide for 15 minutes, add 10 ml ofa 2 per cent v/v solution
ofglacial acetic acid in acetonitrile, centrifuge for lOminutes Purified Water is prepared by distillation, by means of ion
and use the clear supernatant liquid. exchange or by any other appropriate means from suitable
potable water that complies with all relevant statutory
Chromatographic system
regulations.
a stainless steel column 10 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 Ilm), During production and subsequent storage, it is recommended
mobile phase: a mixture of 55 volumes of acetonitrile that adequate measures are taken to ensure that the microbial
45 volumes of water and 1 volume of glacial aceti~ quality is controlled and monitored. Appropriate alert and
acid, action limits are set so as to detect adverse trends. Under
controlled conditions, an appropriate action limit is a total
- flow rate. 2 ml per minute,
viable count (2.2.9) of 100 micro-organisms per ml, determined
spectrophotometer set at 283 11m,
by membrane filtration. In addition, the test for oxidisable
- injection volume. 20 Ill.
substances (given below) is carried out. The adequacy of
Detennine the content ofC I9H 1sNa04 in the tablet. these measures may be detennined by carrying out the test
for conductivity (2.4.9) off-line or on-line.
Dissolution (2.5.2).
Category. Phannaceutical aid (solvent).
Apparatus No.1,
Medium. 900 ml of a 0.68 per cent w/v solution ofpotassium Description. A clear, colourless and odourless liquid.
dihydrogen phosphate with the pH adjusted to 6.8 by the
addition of 1 M sodium hydroxide, Tests
Speed and time. 100 rpm and 45 minutes. Acidity or alkalinity. To 10 ml, freshly boiled and cooled in a
For tablets containing 2 mg or less of warfarin sodium, use borosilicate glass flask, add 0.05 ml of methyl red solution;
three tablets for each test; for tablets containing more than the resulting solution is not red. To 10 ml add 0.1 ml of
2 mg of warfarin sodium, use a single tablet for each test. bromothymol blue solution; the resulting solution is not blue.
Ammonium. To 20 ml add 1 ml ofalkalinepotassium mercuri-
Withdraw a suitable volume ofthe medium and filter. Measure
iodide solution, and allow to stand for 5 minutes. When
the absorbance of a layer of suitable thickness of the filtrate,
viewed vertically the solution is not more intensely coloured
suitably diluted if necessary, at the maxima at about 307 nm
than a solution prepared at the same time by adding 1 ml of
and 360 nm (2.4.7), and calculate the difference between the
alkaline potassium mercuri-iodide solution to a mixture of
two absorbances (DA). Calculate the total content ofwarfarin
4.0 ml of ammonium standard solution (l ppm NH4) and
sodium, CI9HlsNa04, in the medium taking 428 as the specific
16.0 ml ofammonia-free water (0.2 ppm).
absorbance value of DA.
Calcium and magnesium. To.l 00 ml add 2 ml of ammonia
D. Not less than 70 per cent of the stated amount of bufferpH 10.0, 50 mg of mordant black II mixture and 0.5 ml
CI9H,sNa04. of 0.01 M disodium edetate; a pure blue colour is produced.
Other tests. Comply with the tests stated under Tablets. Heavy metals (2.3.13). Evaporate 150 ml to 15 ml on a water-
Assay. Weigh and powder 20 tablets. Weigh accurately a bath; 12 ml of the solution complies with the limit test for
quantity of the powder containing about 20 mg of Warfarin heavy metals, Method D (0.1 ppm). Use leadstandardsolution
Sodium ~n,d shake with 250.0 ml of 0.01 M sodium hydroxide (l ppm Pb) to prepare the standard.
for 15 rrunutes and filter. To 20.0 ml ofthe filtrate add 0.15 ml of Chlorides. To 10 ml add 1 ml of 2 M nitric acid and
hydrochloric acid and extract with three quantities, each of 0.2 ml of 0.1 M silver nitrate; the appearance of the solution
15 ml, ofchloroform. Extract the combined chlorofonn layers does not change for at least 15 minutes.
with three quantities, each of20 ml, of0.01 M sodium hydroxide.
Dilute the combined aqueous layers to 100.0 ml with Nitrates. To 5 ml in a test-tube immersed in ice add 0.4 ml ofa
0.01 M sodium hydroxide, filter and measure the absorbance 10 per cent w/v solution of potassium chloride, 0.1 ml of
oftnetesaltingsolationattnelffaximartfatabout307tuu (2.4:7)~ diphenylamine solutionand, . dropwisewithshaking,5_mLof
Calculate the content ofC1ijHisNaO" taking 431 as the specific sJA./phur}c:a,cid•. 'TIans{erthe ttlbc:: tq 11. Wl1tc::r::batb at 50° and
absorbance at 307 nm. allow to stand for 15 minutes. Any blue colour in th~~~luti~~
is not more intense than that in a solution prepared at the
Storage. Store protected from light. same time and in the same manner using a mixture of4.5 ml of

2314

www.webofpharma.com
IP 2010 WATER FOR INJECTIONS IN BULK

nitrate-fi"ee water and 0.5 ml of nitrate standard solution stored in conditions designed to prevent the growth ofmicro-
(2 ppm N0 3) (0.2 ppm). organisms and to avoid any other contamination.
Sulphates. To 10 ml add 0.1 ml of 2 M hydrochloric acid and During production and subsequent storage, it is recommended
0.1 ml of barium chloride solution. The appearance of the that adequate measures are taken to ensure that the microbial
solution does not change for at least 1 hour. quality is controlled and monitored. Appropriate action limit
of total viable count (2.2.9) of 10 micro-organisms per ml,
Oxidisable substances. To 100 ml add 10 ml of 1 M sulphuric
detennined by membrane filtration using at least 200 ml of
acid and 0.1 ml of 0.02 M potassium permanganate and boil
water for injection in bulle. Appropriate alert and action limits
for 5 minutes; the solution remains faintly pink.
are set so as to detect adverse trends. The adequacy of these
Residue on evaporation. Evaporate 100 ml to dryness on a measures is determined by the following tests that may be
water-bath and dry to constant weight at 105°. The residue done off-line or on-line.
weighs not more than I mg (0.001 per cent).
Total organic carbon (2.4.30). Not more than 0.5 mg per litre.
Purified Water intendedfor use in the manufacture ofdialysis
Conductivity (2.4.9). Meets the requirements ofthe test.
solutions and also without a fitrther procedure for the
removal ofbacterial endotoxins complies with the following Description. A clear, colourless and o"dourless liquid.
additional requirements. l!l
Tests
Aluminium (2.3.8). Not more than 10 ppb, determined using
the following solutions. Acidity or alkalinity. To 10 ml, freshly boiled and cooled in a
borosilicate glass flask, add 0.05 ml of methyl red solution;
Test solution. To 400 ml of the water under examination, add the resulting solution is not red. To 10 ml add 0.1 ml of
10 ml of acetate buffer solution pH 6. aand 100 ml of ~istilled bromothymol blue solution; the resulting solution is not blue.
water.
Ammonium. To 20 ml, add 1ml of alkalinepotassium mercuri-
Reference solution. Mix 2 ml of aluminium standardsolution iodide solution and allow to stand for 5 minutes. When viewed
a
(2 ppm AI), 10 ml of acetate buffer solution pH 6. and 98 ml vertically the solution is not more intensely coloured than a
of distilled water. solution prepared at the same time by adding 1 ml of alkaline
Blank solution. Mix 10 ml of acetate btifJer solution pH 6.0 potassium mercuri-iodide solution to a mixture of 4.0 ml of
and 100 ml of distilled water. ammonium standard solution (1 ppm NH4) and 16.0 ml of
ammonia-free water (0.2 ppm).
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
Unit per ml. Calcium and magnesium. To 100 ml, add 2 ml of ammonia
btifJerpH 10.0,50 mg ofmordant black II mixture and 0.5 ml of
0.01 M disodium edetate; a pure blue colour is produced.

Water for Injections Heavy metals (2.3 .13). Evaporate 150 ml to 15 ml on a water-
bath. 12 ml of the solution complies with the limit test for
H20 Mol. Wt. 18.0 heavy metals, Method D (0.1 ppm). Use leadstandardsolution
(1 ppm Pb) to prepare the standard.
Water for Injections is water intended for use in the
preparations of medicines for parenteral administration when Chlorides. To 10 ml, add 1 ml of 2 M nitric acid and
water is used as a vehicle (Water for Injections in bulk) and for 0.2 ml of 0.1 M silver nitrate; the appearance of the solution
dissolving or diluting substances or preparations for injectable does not change for at least 15 minutes.
preparations (Sterile Water for Injections). Nitrates. To 5 ml in a test-tube immersed in ice add 0.4 ml ofa
10 per cent w/v solution of potassium chloride, 0.1 ml of
diphenylamine solution and, dropwise with shaking, 5 ml of
Water for Injections in Bulk sulphuric acid. Transfer the tube to a water-bath at 50° and
allow to stand for 15 minutes. Any blue colour in the solution
Production is not more intense than that in a solution prepared at the
same time and in the same manner using a mixture of4.5 mlof
Water for Injections in bulk is obtained by distilling potable
nitrate-free water and 0.5 ml of nitrate standard solution
water or Purified Water from a neutral glass, quartz or suitable
metal still fitted with an effective device for preventing the (2 ppm N0 3) (0.2 ppm).
entrainment of droplets; the still must be suitably maintained Sulphates. To 10 ml, add 0.1 ml of2 M hydrochloric acid and
to ensure the production ofapyrogenic water. The first portion 0.1 ml of barium chloride solution. The appearance of the
ofthe distillate is discarded and the remainder is collected and solution does not change for at least 1 hour.

2315

www.webofpharma.com
WATER FOR INJECTIONS IN BULK IP 2010

Aluminium (2.3.8) For waterfor injections intendedfor use Calcium and magnesium. To·l 00 ml, add 2 ml of ammonia
in the manufacture of dialysis solutions. bufferpH 10.0,50 mg of mordant blackII mixture and 0.5 ml
of 0.01 M disodium edetate; a pure blue colour is produced.
Not more than 10 ppb, determined using the following
solutions. Heavy metals (2.3.13). Evaporate 150 ml to 15 ml on a water-
Test solution. To 400 ml of the water under examination add bath. 12 ml of the solution complies with the limit test for
10 m1 of acetate buffer solution pH 6. 0 and 100 ml of distilled heavy metals, Method D (0.1 ppm). Use leadstandardsolution·
water. (1 ppm Pb) to prepare the standard.

Reference solution. Mix 2 rnl of aluminium standardsolution Chlorides.To 10 ml, add 1 ml of 2 Mnitric acid and 0.2 ml of
(2 ppm Al), 10 ml of acetate buffer solution pH 6.0 and 98 ml 0.1 ivJ silver nitrate; the appearance of the solution does not
of distilled water. change for at least 15 minutes.

Blank solution. Mix 10 ml of acetate buffer solution pH 6.0 For containers with a nominal volume oflOO ml or less, 15 ml
and 100 ml of distilled water. complies with the limit test for chlorides (2.3.12) (0.5 ppm),
using a standard solution prepared by mixing 1.5 ml of chloride
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin standard solution (5 ppm Cl) and 13.5 ml of water.
Unitperml.
Nitrates. To 5 ml in a test-tube immersed in ice add 0.4 ml ofa
Storage. Store in containers designed to prevent the growth 10 per cent w/v solution of potassium chloride, 0.1 ml of
ofmicro-organisms. diphenylamine solution and, dropwise with shaking, 5 ml of
Labelling. The label on the container in which the bulk has sulphuric acid. Transfer the tube to a water-bath at 50° and
been distributed states that the contents haVeliot been allow to stand for 15 minutes. Any blue colour in the solution
sterilised. is not more intense than that in a solutioIl prepared at the
same time and in the same manner using a mixture of4.5 mlof
nitrate-free water and 0.5 ml of nitrate standard solution
(2 ppm N03) (0.2 ppm).
Sulphates. To 10 ml, add 0.1 ml of2 Mhydrochloric acid and
Sterile Water for Injections
0.1 ml of barium chloride solution. The appearance of the
Sterile Water for Injections is Water for Injections in bulle that solution does not change for at least 1 hour.
has been distributed in suitable containers of glass or any
Oxidisable substances. Boil 100 rnl with 10 rnl of1 M sulphuric
other material, sealed and sterilised by heat under conditions
acid, add 0.4 ml of 0.02 M potassium permanganate (for Sterile
that ensure that the water complies with the test for bacterial
Water for Injection in containers with fill volume ofless than
endotoxins. It is free from any added substances. Each
50 ml) or 0.2 ml of 0.02 Mpotassiumpermanganate (for Sterile
container contains a sufficient quantity ofWater for Injections
Water for Injection in containers with fill volume of 50 ml or
to permit the withdrawal ofthe nominal volume.
more) and boil for 5 minutes. Ifa precipitate fonus, cool in an
Description. A clear, colourless liquid; odourless. ice-bath to room temperature and filter through a sintered
glass filter (porosity No.3). The pink colour of the solution
Tests does not disappear completely.
Appearance ofsolution. When examined in suitable conditions Residue on evaporation. Evaporate 100 ml to dryness on a
ofvisibility, it is clear (2.4.1) colourless (2.4.1) and practically water-bath and dry the residue to constant weight at 105°. For
free from suspended particles. containers with a nominal volume of 10 ml or less, the residue
weighs not more than 4 mg (0.004 per cent) and for containers
Acidity or alkalinity. To 20 ml, add 0.05 ml of phenol red
with a nominal volume greater than 10 ml, the residue weighs
solution. Ifthe solution is yellow, it becomes red on the addition
not more than 3 mg (0.003 per cent).
of0.1 rnl of O. 01 M sodium hydroxide; ifred, it becomes yellow
on the addition of 0.15 ml of 0.01 M hydrochloric acid. Particulate contamination (2.5.9). Complies with the
Ammonium. To 20 m1, add 1 rnl ofalkalinepotassium mercuri- requirements of Method 1 or Method 2.
iodide solution and allow to stand for 5 minutes. When viewed Bacterial endotoxills (2.2.3). Not more than 0.25 Endotoxin
.vertically the. solution is. not mcireintenselycolouredthana -Unitper ml.--------------------------------- . c

soilltion prepared at the same time by adding 1. ml of alkaline


Sterility (2.2;11 ).Complies with the test for sterility;
potassium mercuri-iodide solution to a mixture of 4.0 ml of
ammonium standard solution (l ppm NH,J and 16.0 ml of Storage. Store in single dose containers ofnot larger than one
ammonia-free water (0.2 ppm). litre size.

2316

www.webofpharma.com
IP 2010 WOOL FAT

Wool Fat hydroxide and boil; the vapours do not turn red litmus paper
blue.
Anhydrous Lanolin
Chlorides. Boil 1.0 g with 20 ml of ethanol (90 per cent) under
Wool Fat is purified, anhydrous, waxy material obtained from a reflux condenser for 5 minutes, cool, add 40 ml of water and
the wool of sheep. It may contain Butylated Hydroxytoluene 0.5 ml of nitric acid and filter. To the filtrate add 0.15 ml of a
as an antioxidant. I per cent w/v solution of silver nitrate in ethanol (90 per
Category. Pharmaceutical aid (absorbent ointment base). cent). After 5 minutes, protected from light; any opalescence
produced is not more intense than that obtained by adding
Description. A pale yellow, unctuous substance; odour, 0.15 ml ofa I per cent w/v solution of silver nitrate in ethanol
characteristic. (90 per cent) to a mixture of 0.2 ml of 0.02 M hydrochloric
acid, 20 ml of ethanol (90 per cent), 40 ml of water and 0.5 ml
Identification
of nitric acid (150 ppm).
A. To a solution of 0.5 g in 5 ml of chloroform add I ml of
Paraffins. Prepare an alumina column 23 cm x 2 cm by adding
acetic anhydride and 0.1 ml of sulphuric acid; a green colour
a slurry of anhydrous aluminium oxide and light petroleum
develops.
(40° to 60°) to a glass tube fitted with a tap and containing
B. To a solution of 50 mg in 5 ml of chloroform add 5 ml of the light petroleum; the tap and absorbent cotton plugs should
sulphuric acid and shake; a red colour is produced and a be free from grease. Allow to settle and reduce the depth of
strong fluorescence appears in the lower layer. the solvent above the column to about 4 cm. Dissolve 3.0 g of
the substance under examination in 50 ml of warm light
Tests
petroleum (40° to 60°), cool, pass the solution through the
Melting range (2.4.21). 34° to 44°, determined by Method IV. column at a rate on ml per minute and wash with 250 ml ofthe
To fill the metal cup, melt the substance under examination on light petroleum. Distil the combined eluate and washings to
a water-bath, cool to about 50°, pour into the cup and allow to low bulle, evaporate to dryness on a water-bath and heat the
stand at 15° to 20° for 24 hours. residue at 105° for periods of 10 minutes until the difference
between two successive weighings is not greater than 1 mg;
Acid value (2.3.23). Not more than 1.0, determined on 5.0 g
the residue weighs not more than 30 mg.
dissolved in 25 ml ofthe prescribed mixture of solvents.
Butylated hydroxytoluene (ifpresent). Not more than 200 ppm,
Peroxide value (2.3.35). Not more than 20.
detennined by gas chromatography (2.4.13).
Saponification value. (2.3.37). 90 to 105. Heat for 4 hours.
Test solution (a). A 10 per cent w/v solution of the substance
Water-absorption capacity. Weigh 10.0 g into a mortar. Add under examination in carbon disulphide.
water in quantities of 0.2 to 0.5 ml from a burette and stir
vigorously, incorporating all the water before proceeding to
Test solution (b). A solution containing 10 per cent w/v ofthe
substance under examination and 0.002 per cent w/v of methyl
the next addition. The end-point is reached when visible
n-decanoate (internal standard) in carbon disulphide.
droplets r~main that cannot be incorporated; not less than
20 ml ofwater is absorbed. Reference solution. A solution containing 0.002 per cent w/v
Water-soluble acidic or alkaline substances. Shake each of butylated hydroxytoluene and the internal standard.
vigorously 5.0 g, previously melted on a water-bath, for Chromatographic system
2 minutes with 75 ml of water previously heated to 90° to 95°. a glass column 1.5 m x 4 mm, packed with silanised
Allow to cool and filter through filter paper previously washed diatomaceous support (80 to 100 mesh) (such as
with water. To 60 ml ofthe filtrate, which may not be clear, add Diatomite) impregnated with 10 percent w/w ofsilicone
0.25 ml of bromothymol blue solution. Not more than 0.2 ml of gum rubber (methyl) (such as SE-30),
0.02 M hydrochloric acid or 0.15 ml of 0.02 M sodium temperature:
hydroxide is required to change the colour of the solution. column 150°,
Water-solubleoxidisable substances. To 10 ml ofthe filtrate inlet port at 180° and detector at 300°,
obtained in the test for Water-soluble acidic or alkaline - flow rate. 40 ml per minute ofthe caITier gas.
substances add 1 ml of 1 M sulphuric acid and 0.1 ml of
Calculate the content of butylated hydroxytoluene in the
0.02 M potassium permanganate; the solution is not substance under examination from the heights or areas of the
completely decolorised within 10 minutes.
peaks due to butylated hydroxytoluene and the internal
Ammonia. To 10 ml ofthe filtrate obtained in the test for Water- standard in the chromatograms obtained with test solution
soluble acidic or alkaline substances, add 1 ml of 1 M sodium (b) and the reference solution.

2317

www.webofpharma.com
HYDROUS WOOL FAT IP 2010

Sulphated ash (2.3.18). Not more than 0.15 per cent. residue at 105° for periods of 10 minutes until the difference
between two successive weighings is not greater than 1 mg;
Loss on drying (2.4.19). Not more than 0.5 per cent, detennined
the residue weighs not more than 30 mg.
on 1.0 g by drying in an oven at 105° for 1 hour.
Storage. Store protected from moisture. Peroxide value (2.3.35). Not more than 15.

Labelling. The label states the proportion of any butylated Saponification value (2.3.37). 67 to 79. Heat for 4 hours.
hydroxytoluene present. Water-absorption capacity. Weigh 10.0 g of the residue
obtained in the test for Wool fat content into a mortar. Add
water in quantities of 0.2 to 0.5 ml from a burette and stir
Hydrous Wool Fat vigorously, incorporating all the water before proceeding to
the next addition. The end-point is reached when visible
Hydrous Wool Fat is a mixture of75 per cent w/w ofWool Fat droplets remain that cannot be incorporated; not less than
and 25 per cent w/w ofPurified Water. 20 ml ofwater is absorbed.
Category. Phannaceutical aid (water-in-oil emulsion ointment Water-soluble acidic or alkaline substances. Shake
base). vigorously 6.7 g, previously melted on a water-bath,
Description. A pale yellow, unctuous substance; odour, faint for 2 minutes with 75 ml of water previously heated to 90° to
and characteristic. On heating, it separates at first into two 95°, Allow to cool and filter through filter paper previously
layers; with continued heating with stirring, water is driven washed with water. To 60 ml ofthe filtrate, which may not be
off and the residue which is transparent while wann, cools to clear, add 0.25 ml of bromothymol blue solution. Not more
fonn a yellowish, tenacious, soft mass. than 0.2 ml of 0.02 M hydrochloric acid or 0.15 ml of
0.02 M sodium hydroxide is required to change the colour of
Identification the solution.
A. To a solution of 0.5 g in 5 ml of chloroform add 1 ml of Water-soluble oxidisable substances. 10 ml of the filtrate
acetic anhydride and 0.1 ml of sulphuric acid; a green colour obtained in the test for Water-soluble acidic or alkaline
develops. substances, add 1 ml of 1 M sulphuric acid and 0.1 ml of
0.02 M potassium permanganate; the solution is not
B. To a solution of 50 mg in 5 ml of chloroform add 5 ml of
completely decolorised within 10 minutes.
sulphuric acid and shake; a red colour is produced and a
strong fluorescence appears in the lower layer. Ammonia. To 10 ml ofthe filtrate obtained in the test for Water-
soluble acidic or alkaline substances add 1 ml of 1 M sodium
Tests hydroxide and boil; the vapours do not turn red litmus paper
Melting range (2.4.21). 34° to 44°, detennined by Method IV blue.
To fill the metal cup, melt the substance under examination on Chlorides. Boil 1.0 g with 20 ml of ethanol (90 per cent) under
a water-bath, cool to about 50°, pour into the cup and allow to a reflux condenser for 5 minutes, cool, add 40 ml of water and
stand at 15° to 20° for 24 hours. 0.5 ml of nitric acid and filter. To the filtrate, add 0.15 ml of
Acid value (2.3.23). Not more than 1.0, detennined on 5.0 g 1.0 per cent w/v solution of silver nitrate in ethanol (90 per
dissolved in 25 ml ofthe prescribed mixture of solvents. cent). After 5 minutes, protected from light, any opalescence
produced is not more intense than that obtained by adding
Paraffins. Prepare an alumina column 23 cm x 2 cm by adding
0.15 ml ofa 1 per cent w/v solution of silver nitrate in ethanol
a slurry of anhydrous aluminium oxide and light petroleum
(90 per cent) to a mixture of 0.2 ml of 0.02 M hydrochloric
(40° to 60°) to a glass tube fitted with a tap and containing
acid, 20 ml of ethanol (90 per cent), 40 ml of water and 0.5 ml
the light petroleum; the tap and absorbent cotton plugs should
of nitric acid (150 ppm).
be free from grease. Allow to settle and reduce the depth of
the solvent above the column to about 4 cm. Dissolve 3 g of Wool fat content. 72.5 to 77.5 per cent.
the substance under examination in 50 ml of warm light
Weigh accurately about 30 g in a tared porcelain dish
petroleum (40° to 60°); cool, pass the solution through the
containing a glass rod, heat on a water-bath with continuous
column at a rate 00 ml per minute and wash with 250 ml ofthe
stirring to constant weight and weigh the residue.
lightpetroleum.Distil the combined eluate and washings to
low bulk, evaporate to dryness on a water~bath and heat the Storage. Store protected from light and moisture.

2318

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

x
XanthanGum 2321
Xylometazoline Hydrochloride 2322
Xylometazoline Nasal Drops 2323
Xylose 2324

2319

www.webofpharma.com
www.webofpharma.com
IP 2010 XANTHAN GUM

Xanthan Gum 2-Propanol. Not more than 750 ppm


Determine by gas chromatography (2.4.13).
Xanthan Gum is high-molecular-mass anionic polysaccharide
produced by fermentation ofcarbohydrates withXanthomonas Internal standard solution. Dilute 0.50 g of 2-methyl-2-
campestris. It consists ofa principal chain ofb(1-lJ1.)-linked propanol to 500 ml with water.
o-glucose units with trisaccharide side chains, on alternating
Test solution. To 200 ml of water in a IOOO-mi round bottomed
anhydroglucose units, consisting of 1 glucuronic acid unit
flask, add 5.0 g of the substance under examination and I ml
included between 2 mannose units. Most ofthe terminal units
ofa 1.0 per cent w/v emulsion of dimeticone in liqzdd paraffin,
contain a pyruvate moiety and the mannose unit adjacent to
stopper the flask and shake for I hour. Distil about 90.0 ml, mix
the principal chain may be acetylated at C-6.
the distillate with 4.0 ml ofthe internal standard solution and
Xanthan Gum has a relative molecular mass ofapproximately dilute to 100.0 ml with water.
1 x 106 . It exists as the sodium, potassium or calcium salt.
Reference solution. Dilute a suitable quantity of 2-propanol,
Xanthan Gum contains not less than 1.5 per cent ofpyruvoyl accurately weighed, with water to obtain a solution having a
groups, (C 3H30 2 ; Mol. Wt. 71.1), calculate on the dried basis. Imown concentration of 2-propanol of about I mg per ml. To
Category. Pharmaceutical aid (Excipient). 4.0 ml of this solution add 4.0 ml of the internal standard
Description. A white or yellowish-white, free-flowing powder. solution and dilute to 100.0 ml with water.

Identification Chromatographic system


- a glass or stainless steel column 1.8 m x 4 mm, packed
A. Suspend 1 g in 15 ml of 0.1 M hydrochloric acid in a flask, with styrene-divinylbenzene copolymer (149-177 /lm),
close the flask with a fermentation bulb containing barium - temperahlre:
hydroxide solution and heat carefully for 5 minutes. The column. 165°,
barium hydroxide solution shows a white turbidity. injection port and detector 200°,
B. To.300 m1 of water, previously heated to 80° and stirred - a flame ionisation detector,
rapidly with a mechanical stirrer in a 400 ml beaker, add, at the - flow rate. 30 ml per minute ofnitrogen as carrier gas.
point ofmaximum agitation, a dry blend of 1.5 g of carob bean Inject 5 /ll of the reference solution. The test is not valid
gum and 1.5 g of the substance under examination. Stir until unless the relative retention times with reference to 2-propanol
the mixture forms a solution, and then continue stirring for 30 for 2-methyl-2-propanol is about 1.5.
minutes or longer. Do not allow the water temperature to drop
Inject 5 /ll of the test solution and the reference solution.
below 60° during stirring. Discontinue stirring and allow the
mixture to stand for at least 2 hours. A firm rubbery gel forms Other polysaccharides. Determine by thin-layer
after the temperature drops below 40° but no such gel forms in chromatography (2.4.17), coating the plate with silica gel.
a I per cent control solution of the sample prepared in· the
Mobile phase. A mixture of 10 volumes of 1.6 per cent w/v
same manner but omitting the carob bean gum.
solution of sodium dihydrogen phosphate, 40 volumes of
Tests butanol and 50 volumes of acetone.
pH (2.4.24). 6.0 to 8.0 determined in I per cent w/v solution. Test solution. To 10 mg ofthe substance under examination in
Viscosity (2.4.28). Not less than 600 mPas. a thick-walled centrifuge test tube add 2ml of a 23 per cent
w/v solution of trifluoroacetic acid, shake vigorously to
Add 3.0 g within 45-90 seconds into 250 ml of a 1.2 per cent
dissolve the forming gel, stopper the test tube, and heat the
w/v solution ofpotassium chloride in a 500 ml beaker stirring
mixture at 120° for 1 hour. Centrifuge the hydrolysate, transfer
with a low-pitch propeller-type stirrer rotating at 800 rpm.
the clear supernatant liquid carefully into a 50 ml flask, add
When adding the substance take care that agglomerates are
10 ml of water and evaporate the solution to dryness under
destroyed. Add an additional quantity of 44 ml of water, to
reduced pressure. Take up the residue thus obtained in 10 ml
rinse any adhering residue from the walls of the beaker. Stir
of water and evaporate to dryness under reduced pressure.
the preparation at 800 rpm for 2 hours whilst maintaining the
Wash 3 times with 20 ml of methanol and evaporate under
temperature at 24 ± 1°. Determine the viscosity within 15 min
reduced pressure. To the resulting clear film which has no
at 24 ± I° using a rotating viscosimeter set at 60 rpm and
a
equipped with rotating spindle 12.7 rom in diameter and
odour ofacetic acid, add 0.1 ml of water and 1 m10fmethanol.
Centrifuge to separate the amorphous precipitate. Dilute the
1.6 rom high which is attached to a shaft 3.2 mm in diameter.
supernatant liquid, if necessary, to 1 ml with methanol.
The distance from the top of the cylinder to the lower tip of
the shaft being 25.4 mm, and the immersion depth being Reference solution. Dissolve 10 mg of glucose and 10 mg of
50.0mm. mannose in 2 ml of water and dilute to 10 ml with methanol.

2321

www.webofpharma.com
XANTHAN GUM IP 2010

Apply to the plate 5 JlI of each solution as bands 10 mm by 2 Xylometazoline Hydrochloride


nun. Allow the mobile phase to rise 15 cm. Dry the plate in air
and spray with 2 per cent w/v solution of diphenylamine in
methanol to which added 0.5 ml of aniline and 2.5 ml of
orthophosphoric acid. Heat the plate at 1200 for 5 minutes
and examine in day light. The principal spot in the
chromatogram obtained with the test solution shows 2 spots
cOlTesponding to the spots due to glucose and mannose in
the chromatogram obtained with the reference solution. In
addition, 1 weak reddish and 2 faint bluish-grey bands may be
CI6H2~2,HCI Mol. Wt. 280.8
visible just above the starting line. 1 or 2 bluish-grey bands
may also be seen in the upper quarter of the chromatogram. Xylometazoline Hydrochloride is 2-(4-tert-butyl-
No other bands are visible. 2,6-dimethylbenzyl)-2-inlidazoline hydrochloride.

The test is not valid unless the chromatogram obtained with Xylometazoline Hydrochloride contains not less than 99.0 per
the reference solution shows two clearly separated greyish- cent and not more than 101.0 per cent of C16H24N2' HCl,
brown spots due to glucose and mannose in the middle third. calculated on the dried basis.
Category. Sympathomimetic amine.
Total ash (2.3.19). Not less than 6.5 and not more than 16.0per
cent, determined on 1.0 g. Description. A white or almost white, crystalline powder;
odourless or almost odourless.
Loss on drying (2.4.19). Not more than 15.0 per cent,
determined on 1.0 g by drying in an oven at 105 0 for 2.5 hours. Identification
Microbial contamination (2.2.9). Total microbial count is not A. Determine by infrared absorption spectrophotometry (2.4.6).
more than 103 bacteria and 102fungi per gram, determined by Compare the spectrum with that obtained with xylometC17oline
plate count. 1 g is free from Escherichia coli. hydrochloride RS or with the reference spectrum of
xylometazoline hydrochloride.
Assay.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Test solution. Dissolve a quantity of the substance under 0.05 per cent w/v solution in 0.1 M hydrochloric acid shows
examination containing 120 mg ofthe dried substance in 20 ml an absorption maximum at about 265 urn and a minimum at
of water. about 257 nm with two inflections at about 270 nm and
275 nm; absorbance at about 265 nm, about 0.5.
Reference solution. Dissolve 45 mg ofpyruvic acid in 500 ml
of water. C. To 1 ml ofa 0.05 per cent w/v solution, add 0.2 ml ofa 5 per
cent w/v solution of sodium nitropnlsside and 0.1 ml of 5 M
Place 10.0 ml of the test solution in a 50 ml round-bottomed sodium hydroxide, allow to stand for 10 minutes and add 2 ml
flask, add 20.0 ml of 0.1 M hydrochloric acid and weigh. Boil of sodium bicarbonate solution; a violet colour is produced.
on a water-bath under a reflux condenser for 3 hours. Weigh
D. Gives the reactions ofchlorides (2.3.1).
and adjust to the initial mass with water. In a separating funnel
mix 2.0 ml ofthe solution with 1.0 ml ofdinitrophenylhydrazine- Tests
hydrochloric solution. Allow to stand for 5 minutes and add
5.0 ml of ethyl acetate. Shake and allow the solids to settle. pH (2.4.24).5.0 to 6.6, determined in a 5.0 per centw/vsolution.
Collect the upper layer and shake with three quantities, each N-(2-Aminoethyl)-4-tert-butyl-2, 6-xylylacetamide. Determine
of 5.0 ml, of sodium carbonate solution. Combine the aqueous by thin-layer chromatography (2.4.17), coating the plate with
layers and dilute to 50.0 ml with sodium carbonate solution, silica gel HF254.
mix. Treat 10.0 ml of the reference solution at the same time
and in the same manner as for the test solution. Mobile phase. A mixture of 200 volumes of methanol and
3 volumes of strong ammonia solution.
Immediately measure the absorbance of the two solutions at
Test solution. Dissolve 0.2 g of the substance under
the maximumat about375nm (2.4.7), using sodium carbonate exaiiiiiiafioniiiTOillI6f mi!lhanoZ:-~----------- - - - --,------- -
solution asthe COmpensation liquid. The absorbance of the
test solution is not less than that of the reference solution, Reference solution .. A 0.01 per cent w/v solution of
which corresponds to a content of pyruvic acid of not less N-(2-aminoethyl)-4-tert-butyl-2,6-xylyl- acetamide RS in
than 1.5 per cent. methanol.

2322

www.webofpharma.com
IP 2010 XYLOMETAZOLINE NASAL DROPS

Apply to the plate 5 III of each solution. After development, Inject the test solution, reference solution (a) and (c). In the
dry the plate in air, spray with a solution containing 0.3 g of chromatogram obtained with the test solution the area of the
ninhydrin in a mixture of 100 ml of 1-butanol and 3 ml of peak due to xylometazoline impurity A is not more than the
glacial acetic acid. Heat at 1000 for 10 minutes, allow to cool, area ofthe corresponding peak in the chromatogram obtained
and spray with dilute potassium iodobismuthate solution. with reference solution (c) (0.2 per cent) and the area of any
Any spot corresponding to N-(2-aminoethyl)-4-tert-butyl- other secondary peak is not more than the area ofthe principal
2,6-xylyl-acetamide in the chromatogram obtained with the peak in the chromatogram obtained with reference solution
test solution is not more intense than the spot in the (a) (0.1 per cent). The sum of all the secondary peaks is not
chromatogram obtained with the reference solution. more than 5 times the area of the principal peak in the
Related substances. Determine by liquid chromatography chromatogram obtained with reference solution (a) (0.5 per
(2.4.14). cent). Ignore any peak with an area less than 0.5 times the area
of the principal peak in the chromatogram obtained with
Test solution. Dissolve 50 mg of the substance under reference solution (a) (0.05 per cent).
examination in 50.0 ml of watel: Allow to stand for 1 hour
before injection. Iron (2.4.14). Moisten the residue obtained in the test for
Sulphated ash with 5 ml of hydrochloric acid, evaporate to
Reference solution (a). Dilute 5.0 ml of the test solution to dryness and dissolve in sufficient water to produce 50 ml.
100.0 ml with water. Dilute 2.0 ml ofthis solution to 100.0 ml 10 ml of the resulting solution complies with the limit test for
with water. iron (50 ppm).
Reference solution (b). Dissolve 5 mg each of N-(2- Sulphates (2.3.17). O. 75 g complies with the limit test for
aminoethyl)-2-[4-(1, 1-dimethylethyl)-2, 6- dimethylphenyl} sulphates (200 ppm).
acetamide RS (xylometazoline impurity A RS) and the
substance under examination in 50.0 of watel: Dilute 10.0 ml of Sulphated ash (2.3.18). Not more than 0.1 percent.
this solution to 50.0 ml with water. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Reference solution (c). Dilute 5.0 ml ofreference solution (b) on 1.0 g by drying in an oven at 105 0 •
to 50.0 ml with watel: Assay. Weigh accurately about 0.5 g, dissolve in 50 ml of
Chromatographic system anhydrous glacial acetic acid, add 10 ml of mercuric acetate
- a stainless steel column 25 cm x 4.6 mm, packed with solution. Titrate with 0.1 M perchloric acid, using
endcapped octadecylsilane bonded to porous silica 1-naphtholbenzein solution as indicator. Carry out a blank
(5Ilm), titration.
mobile phase: A. a 0.14 per cent w/v solution of I ml of 0.1 M perchloric acid is equivalent to 0.02808 g of
potassium dihydrogen phosphate adjusted to pH 3.0 C 16Hz4Nz, HCl.
with orthophosphoric acid,
B. acetonitrile, Storage. Store protected from light and moisture.
a linear gradient programme using the conditions given
below,
flow rate. 1 ml per minute, Xylometazoline Nasal Drops
spectrophotometer set at 220 nm,
injection volume. 10 Ill. Xylometazoline Hydrochloride Nasal Drops
Time Mobile phase A Mobile phase B Xylometazoline Nasal Drops are a solution ofXylometazoline
(min.) (per cent v/v) (per cent v/v) Hydrochloride in Purified Water.
0-5 70 30
Xylometazoline Nasal Drops contain not less than 90.0 per
5-20 70 ~15 30~85 cent and not more than 110.0 per cent ofthe stated amount of
20-35 15 85 xylometazoline hydrochloride, C16Hz4Nz, HCl.
35-37 15 ~70 85~30 Usual strengths. 0.05 per cent w/v; 0.1 per cent w/v.
37-47 70 30
Identification
Inject reference solution (b). The test is not valid unless the
resolution between the peaks due to xylometazpline and A. To a volume containing 50 mg of Xylometazoline
xylometazoline impurity A is not less than 2.5. The relative Hydrochloride add 5 ml of 1 M sodium hydroxide, extract with
retention time with reference to xylometazoline for 10 ml of dichloromethane, evaporate to dryness and dissolve
xylometazoline impurity A is about 0.79. the residue in 0.5 ml of dichloromethane.

2323

www.webofpharma.com
XYLOMETAZOLINE NASAL DROPS IP 2010

Determine by infrared absorption spectrophotometry (2.4.6). the residue in 10.0 ml of 0.01 Mhydrochloric acid. To 2.0 ml
Compare the spectrum with that obtained with xylometazoline of this solution add 3 ml of water, 2.5 ml of 1 M sodium
hydrochloride RS treated in the same manner or with the hydroxide and 2.5 ml of a 5 per cent w/v solution of sodium
reference spectrum ofxylometazoline. nitroprusside, mix and allow to stand protected from light for
10 minutes. Add 10 ml of a freshly prepared 8.3 per cent w/v
B. To a volume containing 0.5 mg of Xylometazoline
solution of sodium bicarbonate, dilute to 100.0 ml with water,
HydrocWoride add 0.2 m1 ofa 5 per cent w/v solution ofsodium
allow to stand protected from light for 10 minutes and measure
nitroprusside and 0.1 ml of 5 M sodium hydroxide, allow to
the absorbance of the resulting solution at the maximum at
stand for 10 minutes and add 1 inl of sodium bicarbonate
about 560 nm (2.4.7), using as blank a solution prepared by
solution; a violet colour is produced.
treating 5 ml of water and 2.5 ml of 1 M sodium hydroxide in
the same manner beginning at the words "and 2.5 m1 ofa 5 per
Tests
cent w/v solution of sodium nitroprusside,.....".
pH (2.4.24).5.6 to 6.6.
Calculate the content of C16H24N2, HCl from the absorbance
N-(2..,Aminoethyl)-4-tert-butyl-2,6-xylylacetamide. Determine obtained by repeating the operation using a 0.1 per cent w/v
by thin-layer chromatography (2.4.17), coating the plate with solution of xylometazoline hydrochloride RS in place of the
silica gel HF254. nasal drops.
Mobile phase. A mixture of 200 volumes of methanol and Storage. Store protected from light and moisture.
3 volumes of strong ammonia solution.
Test solution. Add a volume containing 10 mg of
Xylometazoline Hydrochloride to 30 ml of water, add 5 mlof
5 M sodium hydroxide, mix, extract with three quantities, each Xylose
of20 ml, of dichloromethane, evaporate the combined extracts D-Xylose; D-Xylopyranose
to dryness and dissolve the residue in 1 ml of dichloro-
methane.
Reference solution. A 0.03 per cent vi/v solution of
N-(2-aminoethyl)-4-tert-butyl-2,6-xylylacetamide RS in
~O~OH
dichloromethane. H6~
Apply to the plate 5 III of each solution. After development,
OH
illy the plate in air, spray with a solution containing 0.3 g of
Mol. Wt. 150.1
ninhydrin in a mixture of 100 ml of I-butanol and 3 ml of
glacial acetic acid. Heat at 100° for 10 minutes, allow to cool, Xylose contains not less than 98.0 per cent and not more
and spray with dilute potassium iodobismuthate solution. than 102.0 per cent of CSHIOOS, calculated on the dried basis.
Any spot corresponding to N-(2-aminoethylj::4-tert-butyl- Category. Diagnostic aid in intestinal malfunction.
2,6-xylyl- acetamide in the chromatogram obtained with test
solution is not more intense than the spot in the chromatogram Description. Colourless needles or a white, crystalline powder.
obtained with reference solution.
Identification
Other tests. Comply with the tests stated under Nasal
Preparations. A. Determine by thin-layer chromatography (2.4.17), coating
the plate with a suspension of silica gel G in a 0.3 per cent
Assay. To a, volume containing 10 mg of Xylometazoline w/v solution of sodium acetate to fonn a uniform layer 0.5 mm
Hydrochloride add 5 ml of water, 10 ml of 2 M hydrochloric thiclc
acid and 10 ml of dichloromethane and shake for 1 minute.
Discard the dichloromethane layer and repeat the extraction Mobile phase. A mixture ono volumes ofglacial acetic acid,
with two further quantities, each of 10 ml, of dichloromethane. 60 volumes of chloroform and 10 volumes of water.
Add to the aqueous extract 10 ml of 5 M sodium hydroxide Test solution. Dissolve 0.5 g of the substance under
and 10 ml of dichloromethane, shake for 1 minute and allow to examination in 10 m1 of water.
.~t~.I2fI.I!lt~,. f il!{l! tl1~Qi(;111QrQ1!1{ltl1ll.11_{l~x:trll.~t1lrQl1gl1.g!ll.~i3.~Q9!
Referencesolution{a):·k5:0per··centw/vsolutionufxylose
and repeat the extraction with four further quantities, each of
RS in water.
10 ml, of dichloromethane. Evaporate the combined
dichloromethane extracts almost to dryness on a water-bath, Reference solution (b). A mixture of equal volumes ofthe test
remove the final traces ofsolvent in a current ofair and dissolve solution and reference solution (a).

2324

www.webofpharma.com
IP 2010 XYLOSE

Apply to the plate 2 III of each solution. Develop the plate in Iron (2.3.14). A solution of 4.0 g in 25 ml of water complies
a continuous elution tank for about 4 hours. Dry the plate in with the limit test for iron (l0 ppm).
warm air, spray with a solution in acetone containing 1 per
Chlorides (2.3.12). Dissolve 3.0 g in 20 ml of water. 5 mlofthe
cent w/vsolution of diphenylamine, 1 per cent v/v of aniline
resulting solution complies with the limit test for chlorides
and 1 per cent v/v ofphosphoric acid and heat for 10 minutes
(330 ppm).
at 1300. The principal spot in the chromatogram obtained with
the test solution corresponds to that in the chromatogram Sulphated ash (2.3.18). Not more than 0.1 per cent.
obtained with reference solution (a). The principal spot in the Loss on drying (2.4.19). Not more than 0.5 per cent, determined
chromatogram obtained with reference solution (b) appears on 1.0 g by drying in an oven at 1000at a pressure not exceeding
as a single, compact spot. 0.7kPa.
B. When heated with potassium cupri-tartrate solution it Assay. Carry out the follOWing procedure keeping strict
produces a copious precipitate of cuprous oxide. control oftime between steps.
Weigh accurately about 1.0 g of the substance under
Tests
examination, dissolve in a saturated solution of benzoic acid
Appearance ofsolution. A 10.0 per cent w Iv solution in carbon in a 1OO-ml volumetric flask and dilute to volume with the same
dioxide-ji-ee water is clear (2.4.1), and colourless (2.4.1). solvent. Dilute 1.0 ml ofthis solution to 100.0 rn1 with the same
solvent. To 1.0 ml of the resulting solution, in two different
Acidity. Dissolve 5.0 g in 50 ml of carbon dioxide-ji-ee water.
test-tubes, add 5 ml of 4-bromoaniline solution into each
Not more than 0.2 ml of 0.1 M sodium hydroxide is required to
tube and mix. Loosely stopper one tube, place in a water-bath
neutralise the solution using dilute phenolphthalein solution
maintained at 700for 10 minutes, remove, cool rapidly to room
as indicator.
temperature and mix. Keep the tube in the dark for 70 minutes
Specific optical rotation (2.4.22). +l8S to +19S, determined and measure the absorbance at the maximum at about 520 urn
at 200 in a 10.0 per cent w/v solution containing 0.4 per cent (2.4.7), using the untreated solution in the second test tube as
v/v of 5 M ammonia. the blank. Simultaneously, carry out the operation using
1.0 ml ofa 0.01 per centw/v solution ofxylose RS ill a saturated
Arsenic (2.3.10). Dissolve 10.0 g ill 50 ml of water and add
solution of benzoic acid beginning at the words "add 5 ml of
10 ml of stannated hydrochloric acid AsT. The resulting
4-bromoaniline solution.....".
solution complies with the limit test for arsenic (l ppm).
Calculate the content ofCsHIQOs.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method A (20 ppm). Storage. Store protected from moisture.

2325

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

z
Zidovudine 2329
Zidovudine Capsules 2330
Zidovudine fujection 2331
Zidovudine Oral Solution 2332
Zidovudine Tablets 2333
Zidovudine, Lamivudine and Nevirapine Tablets 2334
Zinc Chloride 2335
Zinc Oxide 2336
Zinc Oxide Cream 2336
Zinc Stearate 2337
Zinc Sulphate 2337
Zinc Sulphate Eye Drops 2338
Zinc Undecenoate 2338
Zinc Undecenoate Ointment 2339
Zoledronic Acid 2339
Zoledronic Acid fujection 2340
Zolpidem Tartrate 2341
Zolpidem Tablets 2342

2327

www.webofpharma.com
www.webofpharma.com
IP 20ID ZIDOVUDINE

Zidovudine Dry the plate in air and examine in ultraviolet light at 254 om.
Any secondary spots observed in the chromatogram obtained
with the test solution correspond to those of the principal
spots in the chromatogram obtained with the reference
solution. No secondary spot in the chromatogram obtained
with the test solution is more intense than the principal spot
in the chromatogram obtained with the reference solution
(0.5 per cent).
Spray the plate with a mixture of0.5 g of carbazole in 95 m10f
ethanol (95 per cent) and 5 ml of sulphuric acid, heat for
10 minutes at 120° and compare the intensities ofany secondary
spots observed in the chromatogram obtained with the test
solution with those ofthe principal spots in the chromatogram
Mol. Wt. 267.2
obtained with the reference solution. No spot corresponding
Zidovudine is 1-(3-azido-2,3-dideoxy-{3-o-ribofuranosyl)- to triphenylmethanol (Rr value about 2.3 relative to the Rr of
5-methylpyrimidine-2,4(lH,3H)-dione. zidovudine) is more intense than the corresponding spot in
Zidovudine contains not less than 97.0 per cent and not more the chromatogram obtained with the reference solution
than 102.0 per cent ofC IOH 13Ns0 4, calculated on the anhydrous (0.5 per cent). No secondary spot in the chromatogram
basis. obtained with the test solution is more intense than the
principal spot in the chromatogram obtained with the reference
Category. Antiretroviral.
solution.
Dose. 300 mg twice daily.
B. Deterinine by liquid chromatography (2.4.14).
Description. A white or almost white powder.
Test solution. Dissolve 0.1 g of the substance under exami-
nation in 100 ml of methanol.
Identification
Reference solution (a). A 0.001 per cent w/v solution of
A. Determine by infrared absorption spectrophotometry (2.4.6). zidovudilJe RS in methanol.
Compare the spectrum with that obtained with zidovudine RS
or with the reference spectrum ofzidovudine. Reference solution (b). A solution containing 0.1 per cent
w/v of zidovudine RS and 0.001 per cent w/v each of
B. In the Assay, the principal peak in the chromatogram zidovudine-related compound B RS and zidovudine- related
obtained with the test solution corresponds to the peak due compound C RS in methanol.
to zidovudine in the chromatogram obtained with reference
solution (a). Chromatographic system
a stainless steel column 15 cm x 4.6 mm, packed with
C. Melting range (2.4.21). 122° to 125°, octadecylsilane' bonded to porous silica (5 /lm),
mobile phase: A. water
Tests B. methanol,
Specific optical rotation (2.4.22). +60.so to +63.0°, determined flow rate. 1 ml per minute,
in a 1.0 per cent w/v solution in ethanol (95 per cent). a linear gradient programme using the conditions given
below,
Related substances. A. Determine by thin-layer
chromatography (2.4.17), coating the plate with silica gel spectrophotometer set at 265 om,'
GF254. injection volume. 10 /ll.
Time Mobile phase A Mobile phase B
Mobile phase. A mixture of 90 volumes of dichloromethane
(in min.) (per cent v/v) (per cent v/v)
and 10 volumes of methanol.
Test solution. Dissolve 0.2 g of the substance under
o 90 10
examination in 10 ml of methanol. 15 35 65
30 35 65
Reference solution. A solution containing 0.01 per cent w/v
each of zidovudine RS and triphenylmethanol in methanol. 35 5 95
Apply to the plate 10 /ll of each solution. Allow the mobile 40 5 95
phase to rise to about three-fourths of the height of the plate. 45 90 10

2329

www.webofpharma.com
ZIDOVUDINE IP 2010

Inject reference solution (b). The test is not valid unless the Zidovudine Capsules
resolution between the peaks due to zidovudine and
zidovudine-related compound B is not less than 1.5 and the Zidovudine Capsules contain not less than 90.0 per cent and
tailing factor for zidovudine is not more than 1.5. not more than 110.0 per cent of the stated amount of
zidovudine, C IOH J3N s0 4•
Separately inject the test solution and reference solution (a).
The area of the peak corresponding to zidovudine- related Usual strength. 100 mg.
compound B is not greater than 1.0 per cent and of that to
zidovudine-related compound C is not greater than 2.0 per Identification
cent.
A. Transfer a quantity of the mixed contents of the capsules
The sum of the percentages of related substances by tests A containing about 15 mg ofZidovudine to a 1OO-ml volumetric
and B is not greater than 3.0 per cent. flask. Add about 80 ml ofa mixture of75 volumes of methanol
Heavy metals (2.3.13). 1.0 g complies with limit test for heavy and 25 volumes of water, shake for 10 minutes and dilute to
metals, Method B (20 ppm). volume with the same solvent mixture, mix and filter. Dilute
10 ml ofthe filtrate to 100 ml with the same solvent mixture.
Sulphated ash (2.3.18). Not more than 0.25 percent.
When examined in the range 200 nm to 300 nm (2.4.7), the
Water (2.3.43). Not more than 1.0 per cent, determined on 0.5 g. resulting solution shows an absorption maximum at about
Assay. Determine by liquid chromatography (2.4.14). 265nm.
Test solution. Weigh accurately about 100 mg ofthe substance B. In the Assay, the principal peak in the chromatogram
under examination, dissolve in a suitable quantity of methanoI obtained with the test solution corresponds to the peak due
in a 50-ml volumetric flask and make up to volume with the to zidovudine in the chromatogram obtained with reference
mobile phase. Dilute 5.0 ml ofthis solution to 50.0 ml with the solution (c).
mobile phase.
Reference solution (a). Weigh accurately about 100 mg of Tests
zidovudine RS, dissolve in a suitable quantity of methanol in Related substances. Determine by liquid chromatography
a 50-ml volumetric flask and make up to volume with the mobile (2.4.14).
phase (solution A). Dilute 5.0 ml ofsolutionAto 50.0 ml with
the mobile phase. Test solution. Weigh accurately a quantity of the mixed
contents of20 capsules containing about 100 mg ofZidovudine
Reference solution (b). Transfer 2.0 ml ofa 0.005 percentw/v and transfer to a 100-ml volumetric flask. Add about 60 ml of
solution of zidovudine-related compound B RS in the mobile the mobile phase, mix with the aid ofultrasound for 10 minutes,
phase to a 50-ml volumetric flask, add 5.0 ml ofsolution A and dilute to volume with the mobile phase, mix and filter. Dilute
make up to volume with the mobile phase. 5.0 ml ofthe filtrate to 50.0 ml with the mobile phase.
Chromatographic system
Reference solution (a). Weigh accurately about 100 mg of
a stainless steel column 15 cm x 4.6 mm, packed with
zidovudine RS and transfer to a 100-ml volumetric flask.
octadecylsilane bonded to porous silica (5 Ilm),
Dissolve in about 60 ml of the mobile phase and dilute to
- mobile phase: a mixture of 70 volumes of water and
volume with the mobile phase.
30 volumes of methanol,
- flow rate. 1 ml per minute, Reference solution (b). Weigh accurately about 20 mg of
- spectrophotometer set at 265 nm, thymine and transfer to a 200-ml volumetric flask, dissolve
- injection volume. 20 Ill. and make up to volume with methanol.
Inject reference solution (b). The test is not valid unless the Reference solution (c). Transfer 10.0 ml of the reference
resolution between the peaks due to zidovudine and solution (a) and 2.0 ml of reference solution (b) to a 100-ml
zidovudine-related compound B is not less than 2.0. volumetric flask and make up to the volume with the mobile
Inject reference solution (a). The relative standard deviation phase.
for replicate injections is not more than 2.0 per cent. Chromatographic system
.......... Separately-inject ·the test solution-andreferencesolution(a} . a.stainless.. steeLcolumn 25_cm.~ . 4.6.mm,pJtc.kedJYith
and measure the responses for the principal peak. Qctadecylsilal1e b()nded t() porous.sili(;fl. (5 IllTI)'
mobile phase: a mixture of20 volumes of methanol and
Calculate the content ofC IOHJ3N s0 4• 80 volumes of water,
Storage. Store protected from light. flow rate. 1 ml per minute,

2330

www.webofpharma.com
IP 2010 ZIDOVUDINE INJECTION

- spectrophotometer set at 265 nm, solution and reference solution (c) and measure the responses
- injection volume. 10 fll. for the principal peak.
Inject reference solution (c). The test is not valid unless the Calculate the content ofCIOHI3Ns04 in the capsules.
relative retention times are about 0.2 for thymine and 1.0 for Storage. Store protected from moisture.
zidovudine, the resolution between zidovudine and thymine
is not less than 5.0, the tailing factor is not more than 2.0 and
the relative standard deviation for replicate injections is not
more than 2.0 per cent. Zidovudine Injection
Inject separately the test solution and reference solution (c) Zidovudine Injection is a sterile solution of Zidovudine in
and measure the responses for the thymine peak. The content Water for Injections. .'
of thymine in the capsules should not be more than 3.0 per
Zidovudine Injection contains not less than 90.0 per cent and
cent.
not more than 110.0 per cent of the stated amount of
Dissolution (2.5.2). CIOH 13N s0 4•
Apparatus No.1, Usual strengths. 10 mg per ml.
Medium. 900 ml ofwate]; Description. A clear, colourless solution.
Speed and time. 50 rpm and 45 minutes.
Identification
Withdraw a suitable volume ofthe medium and filter promptly
through a membrane filter disc having an average pore diameter A. When examined in the range 220 nm to 360 nm (2.4.7), a
not greater than 1.0 flm, rejecting the first few ml ofthe filtrate 0.0015 per cent w/v solution in a mixture of75 volumes of
and dilute a suitable volume of the filtrate, if necessary with methanol and 25 volumes ofwater shows absorption maxima
water. similar to those obtained with a solution of zidovudine RS of
the same concentration.
Determine by liquid chromatography (2.4.14).
B. In the Assay, the principal peak in the chromatogram
Test solution. The filtrate obtained as given above.
obtained with the test solution corresponds to that in the
Reference solution. A known quantity of zidovudine RS is chromatogram obtained with the reference solution.
dissolved in l.ml ofmethanol and suitably diluted with water
to obtain a solution having a similar concentration as that of Tests
the test solution. pH (2.4.24). 3.5 to 7.0, in a mixture containing a volume of
Chromatographic system injection containing 150 mg ofzidovudine and 5 ml of0.12 M
- a stainless steel column 25 cm x 4.6 rom, packed with potassium chloride.
octadecylsilane bonded to porous silica (5 flm), Related substances. Determine by liquid chromatography
- mobile phase: a mixture of20 volumes ofmethanol and (2.4.14).
80 volumes of wate];
- flow rate. 1 ml per minute, Test solution. Dilute an accurately measured volume of the
- spectrophotometer set at 265 run, injection containing 25 mg of Zidovudine, to 25 ml with
injection volume. 10 fll. methanol.

Inject the reference solution. The tailing factor is not more Reference solution (a). A 0.1 per cent w/v solution of
than 2.0 for zidovudine peak and the relative standard deviation zidovudine RS in methanol.
for replicate injections is not more than 2.0 per cent. Reference solution (b). A solution containing 0.1 per cent
Inject the test solution and the reference solution and measure w/v of zidovudine RS and 0.001 per cent w/v of zidovudine
the peak responses of the major peak. related compound C (thymine).
Chromatographic system
Calculate the content ofCIOHI3Ns04 in the medium.
- a stainless steel column 25 cm x 4.0 mm, packed with
D. Not less than 75 per cent of the stated amount of octadecylsilane bonded to porous silica ( 5 flm),
C IOH 13N s0 4. - mobile phase: a mixture of 80 volumes of water and 20
volumes of methanol,
Other tests. Comply with the tests stated under Capsules.
flow rate. 1 ml per minute,
Assay. Determine by liquid chromatography (2.4.14), as - spectrophotometer set at 265 nm,
described under Related substances. Inject separately the test injection volume. 10 fll.

2331

www.webofpharma.com
ZIDOVUDlNE ORAL SOLUTION IP 2010

Inject reference solution (b). The test is not valid unless the B. In the Assay, the principal peak in the chromatogram
resolution between zidovudine and thymine is not less than obtained with the test solution corresponds to the peak due
4.0, the tailing factor is not more than 2.0 and the relative to zidovudine in the chromatogram obtained with reference
standard deviation for replicate injections is not more than solution (c).
2.0 per cent.
Inject the test solution and reference solution (b). In the Tests
chromatogram obtained with the test solution the area of any pH (2.4.24). 3.0 to 4.0, determined in a mixture containing a
peak corresponding to thymine is not greater than twice the volume of the preparation under examination containing
area ofthe corresponding peak in the chromatogram obtained 150 mg ofzidovudine and 5 ml of 0.i2 M potassium chloride.
with reference solution (b) (2.0 per cent).
Related substances. Determine by liquid chromatography
Other tests. Complies with the tests stated under Parenteral (2.4.14).
Preparations (Injections).
Test solution. Transfer an accurately measured volume ofthe
Bacterial endotoxins (2.2.3). Not more than 1.0 Endotoxin
preparation under examination containing about 100 mg of
Unit per mg ofzidovudine.
zidovudine to a 100-ml volumetric flask, dissolve and dilute to
Assay. Detennine by liquid chromatography (2.4.14), as given volume with the mobile phase and mix. Dilute 5.0 m1 of this
under the test for Related substances. solution to 50.0 ml with the mobile phase.
Inject alternately the test solution and reference solution (a). Reference solution (a). Weigh accurately about 100 mg of
Calculate the content ofC IOH 13N s0 4 in the injection. zidovudine RS and transfer to a 100-ml volumetric flask, add
about 50 ml ofthe mobi1ephase, mix with the aid ofultrasound
Storage. Store protected from light and moisture. to dissolve, dilute to volume with the mobile phase and mix.
Reference solution (b). Weigh accurately about 20 mg of
thymine RS and transfer to a 200-ml volumetric flask, add about
Zidovudine Oral Solution 150 ml of the mobile phase, mix with the aid of ultrasound to
dissolve, dilute to volume with the mobile phase and mix.
Zidovudine Oral Solution is a solution of Zidovudine in a
suitable flavoured vehicle. Reference solution (c). Transfer 10.0 ml of the reference
solution (a) and 2.0 ml of reference solution (b) to a lOO-ml
Zidovudine Oral Solution contains not less than 90.0 per cent
volumetric flask and make up to the volume with the mobile
and not more than 110.0 per cent of the stated amount of
phase.
zidovudine, C IOH 13N s0 4•
Chromatographic system
Usual strength. 50 mg in 5 ml.
a stainless steel column 12.5 cm x 4.0 mm, packed with
Identification octadecylsilanebonded to porous silica (5 /-lm),
mobile phase: a mixture of90 volumes of 0.04 M sodium
A. Determine by thin-layer chromatography (2.4.17), coating acetate, 9 volumes of methanol, 1 volume of acetonitrile
the plate with silica gel GF254. and 0.2 volume of glacial acetic acid,
Mobile phase. A mixture of 40 volumes of i-butanol, flow rate. 1 ml per minute,
30 volumes of heptane, 30 volumes of acetone and 10 volumes spectrophotometer set at 240 nm,
of strong ammonia solution. injection volume. 10 /-ll.
Test solution. Dilute the preparation under examination with Inject reference solution (c). The test is not valid unless the
methanol to obtain a solution containing 5 mg of zidovudine relative retention times are about 0.12 for thymine and 1.0 for
perml. zidovudine, the resolution between zidovudine and thymine
is not less than 4.0, the taiiing factor is not more than 2.0 and
Reference solution. A 0.5 per cent w/v solution of zidovudine
the relative standard deviation for replicate injections is not
RS in a mixture of75 volumes of methanol and 25 volumes of
more than 2.0 per cent.
water.
Inject separately the test solution and reference solution (c)
:6jJPJytQ!!l~pI~teJ .. l!.l()f.t:l.a:~!ls()I!1!i()Il,Af1:~r_Q~y~I().PJll~l1t,
dry the plate in air and examine in ultraviolet light at 254 nm. atRlm:easure the responses for the thymifiep-eak The-content
The principal spot in the chromatogram obtained with the test of thymine in the capsules should not be more than 3:0 per
solution corresponds to that in the chromatogram obtained cent.
with the reference solution. Other tests. Complies with the tests stated under Oral Liquids.

2332

www.webofpharma.com
IP 2010 ZlDOVUDINE TABLETS

Assay. Determine by liquid chromatography (2.4.14), as mobile phase: a mixture of90 ml of methanol and 40 ml
described under Related substances. Inject the test solution of acetonitrile and a buffer prepared by dissolving
and reference solution (c) and measure the responses for the 3.0 g of sodium acetate and 3.0 g of sodium
major peak. l-octanesulphonate in 900 ml of water and adjusting
the pH to 5.3 with glacial acetic acid,
Calculate the content of CIOHI3Ns04 in the preparation under
flow rate. 1.3 ml per minute,
examination.
spectrophotometer set at 265 nm,
Storage. Store protected from light and moisture. injection volume. 10 fll.
Inject the reference solution. The tailing factor is not more
than 2.0 for zidovudine peak and the relative standard deviation
Zidovudine Tablets for replicate injections is not more than 2.0 per cent.

Zidovudine Tablets contain not less than 90.0 per cent and Inject the test solution and the reference solution and measure
not more than 110.0 per cent of the stated amount of the peak responses of the major peak.
zidovudine, C IOH 13 N s0 4. Calculate the content ofC IO H 13 N s0 4 in the medium.
Usual strengths. 100 mg; 300 mg. D. Not less than 80 per cent of the stated amount of
C IOH 13N s0 4 •
Identification
Related substances. Determine by liquid dU'omatography
A. In the Assay, the principal peak in the chromatogram
(2.4.14).
obtained with the test solution corresponds to the peak due
to Zidovudine in the chromatogram obtained with reference Test solution. Disperse the powdered tablets containing about
solution (a). 1500 mg ofZidovudine in 50 ml of water, shake for 30 minutes.
Add about 150 ml of methanol, sonicate for 10 minutes and
B. Remove the coating from a few tablets and crush them in a
dilute to 500.0 ml with wate/: Dilute 4.0 ml ofthis solution to
mortar so that no large pieces remain.
100.0 ml with water, filter.
On the powder determine by infrared absorption
Reference solution (a). A 0.01 per cent w/v solution of 3'-
spectrophotometry (2.4.6). Compare the spectrum with that
chloro-3 '-deoxythymidine (zidovudine impurity B RS) in
obtained with zidovudine RS or with the reference spectrum
of zidovudine.
methanol.
Reference solution (b). A 0.02 per cent w/v solution of thymine
Tests RS (zidovudine impurity C RS) in methanol.
Dissolution (2.5.2).
Reference solution (c). Dissolve about 30 mg of zidovudine
Apparatus No.1, RS in 3.0 ml of methanol, add 2.5 ml ofreference solution (a),
Medium. 900 ml of water, 5.0 ml of reference solution (b) and dilute to 250.0 ml with
Speed and time. 50 rpm and 30 minutes. water.
Withdraw a suitable volume ofthe medium and filter promptly Chromatographic system
through a membrane filter disc having an average pore diameter a stainless steel column 15 cm x 4.6 rom, packed with
not greater than 1.0 flm, rejecting the first few ml ofthe filtrate octadecylsilane bonded to porous silica (5 flm),
and dilute a suitable volume of the filtrate, if necessary with mobile phase: dissolve 3.0 g of sodium acetate and
water. 1.3 g of sodium l-octanesulphonate in 900 ml of water.
Detennine by liquid chromatography (2.4.14). Add 90 ml of methanol and 40 ml of acetonitrile,
adjusted to pH 5.3 with glacial acetic acid, filter,
Test solution. The filtrate obtained as given above. flow rate. 1.3 ml per minute,
Reference solution. A known quantity of zidovudine RS is - spectrophotometer set at 265 nm,
dissolved in 1 ml of methanol and suitably diluted with water injection volume. 20 fll.
to obtain a solution having a similar concentration as that of Inject reference solution (c). The test is not valid unless the
the test solution. resolution between the peaks due to zidovudine and
Chromatographic system zidovudine impurity B is'not less than 2.5, the tailing factor of
a stainless steel column 15 cm x 4.6 rum, packed with the principal peak is not more than 2.0 and the relative standard
octadecylsilane bonded to porous silica (5 flm) (Such deviation for replicate injections is not more than 2.0 per cent.
as Hypersil BDS C 18), The relative retention time with reference to zidovudine for

2333

www.webofpharma.com
ZIDOVUDINE, LAMIVUDINE AND NEVIRAPINE TABLETS IP 2010

zidovudine impurity C is about 0.17 and for zidovudine impurity Chromatographic system
B is about 1.2. a stainless steel column 25 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 /lm) (Such as
Inject reference solution (c) and the test solution. In the
Hypersil C8),
chromatogram obtained the test solution, the area ofthe peak
mobile phase: A. 0.1 M ammonium acetate,
due to zidovudine impurity C is not more than 1.5 per cent,
B. acetonitrile,
multiplying with response factor of 1.7 and the area of other
flow rate. 1 ml per minute,
secondary peaks is not more than 0.2 per cent. The sum of
a linear gradient programme using the conditions given
areas of all the secondary peaks is not more than 2.0 per cent.
below,
Other tests. Comply with the tests stated under Tablets. spectrophotometer set at 270 nm,
Assay. Determine by liquid chromatography (2.4.14), as injection volume. 20 /ll.
described under Related substances. Separately inject the test Time 0.1 M ammonium acetate acetonitrile
solution and reference solution (c) and measure the peak (in min.) (per cent v/v) (per cent v/v)
responses for the major peak. o 95 5
Calculate the content OfClOHl3Ns04 in the tablets. 5 95 5
Storage. Store protected from light and moisture. 25 20 80
30 20 80
31 95 5
35 95 5
Inject the reference solution. The test is not valid unless the
Zidovudine, Lamivudine and column efficiency determined from the zidovudine, lamivudine
Nevirapine Tablets and nevirapine peaks is not less than 3000 theoretical plates
and the tailing factor for the same peaks is not more than 2.0.
Zidovudine, Lamivudine and Nevirapine Tablets contain not
less than 90.0 percent and not more than 110.0 per cent ofthe Inject separately water and the test solution. Examine the
stated amounts of zidovudine, CloH13Ns04, lamivudine, chromatogram obtained with water for any extraneous peaks
CSHIIN303S andnevirapine, C 1sH 14N 40. and ignore the corresponding peaks observed in the
chromatogram obtained with the test solution.
Usual strength. Zidovudine, 300 mg, Lamivudine, 150 mg and
Nevirapine , 200 mg. Any secondary peak observed in the chromatogram obtained
with the test solution corresponding to a relative retention
Identification time of 0.35 should not be more than 1.0 per cent. Any other
secondary peak observed in the chromatogram obtained with
A. In the Assay, the three principal peaks in the chromatogram the test solution should not be more than 0.5 per cent and the
obtained with the test solution correspond to the peaks due sum of the areas of all the secondary peaks should not be
to zidovudine, lamivudine and nevirapine in the chromatogram more than 2.5 per cent when calculated by percentage area
obtained with the reference solution. normalisation.

Tests Dissolution (2.5.2).


Apparatus No.1,
Related substances. Determine by liquid chromatography Medium. 900 ml of 0.1 M hydrochloric acid,
(2.4.14).
Speed and time. 50 rpm and 30 minutes.
Test solution. Weigh accurately a quantity of the powdered
Withdraw a suitable volume ofthe medium and filter promptly
tablets containing about 100 mg of Zidovudine and transfer
through a membrane filter disc having an average pore diameter
to a 200-ml volumetric flask. Add about 150 ml of water, mix
not greater than 1.0 /lm, rejecting the fIrst few ml ofthe filtrate
with the aid of ultrasound for 10 minutes, dilute to volume
and dilute a suitable volume of the filtrate, if necessary with
with water, mix and filter.
water.
Reier.en~~_sQ1J-'JigJ1,Wejgb_!l_cJ<ltratelY_j~12Q1tLlJ1! Lmg-9J Hetermine-byliquidchromatography(2;4;14);
zirjovu(jin!} R.§, 50 mg oflamivudine R.§ and§5 IIlg ofnevirapine
RS and transfer to a 200-ml volumetric flask. Add about 20 ml Test solution. The filtrate obtained as given above.
ofmethanol, mix with the aid ofultrasound to dissolve, dilute Reference solution. Weigh accurately about 300 mg of
to volume with water and mix. zidovudine RS, 150 mg of lamivudine RS and 200 mg of

2334

www.webofpharma.com
IP 2010 ZINC CHLORIDE

nevirapine RS and transfer to a 100 ml volumetric flask. Add replicate injections of all the analyte peaks is not more than
about 20 ml of methanol, mix with the aid of ultrasound to 2.0 per cent.
dissolve and dilute to volume with a solvent mixture of equal Inject the test solution and the reference solution and measure
volumes of methanol and water. Dilute 5.0 Ji11 ofthis solution
the responses for the major peaks.
to 50.0 ml with 0.1 M hydrochloric acid.
Calculate the contents of C IOH 13N s0 4, C SH 1\N 30 3 S and
Chromatographic system
C\SH I4N 40 in the tablets.
- a stainless steel column 15 cm X 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 /lm) (Such Storage. Store protected from moisture.
as Hypersil BDS C18),
- mobile phase: a mixture of35 volumes of methanol and
65 volumes of a buffer solution prepared by dissolving
0.68 g ofpotassium dihydrogen phosphate and 1.0 g of
Zinc Chloride
sodium octanesulphonate in 1000.0 ml of water to which ZnCh Mol. Wt. 136.3
1 ml of triethylamine is added and the pH of which is
Zinc Chloride contains not less than 95.0 per cent and not
adjusted to 2.5 with phosphoric acid,
more than 100.5 per cent ofZnCh.
- flow rate. 1 ml per minute,
- spectrophotometer set at 266 run, Category. Pharmaceutical aid.
- injection volume. 10 Ill. Descl"iption. A white or practically white, crystalline powder;
Inject the reference solution. The tailing factor for the individual odourless; very deliquescent.
zidovudine, lamivudine and nevirapine peaks is not more than
2.0 and the relative standard deviation for replicate injections Identification
of all the analyte peaks is not more than 2.0 per cent. A. To 2 g add 38 ml of carbon dioxide-free water prepared
Inject the test solution and the reference solution and measure from distilled water and add 2 M hydrochloric acid dropwise
the peak responses of the major peaks due to zidovudine, until solution is complete and dilute to 40 ml with carbon
lamivudine and nevirapine. dioxide-free water prepared from distilled water (solution
A). Solution A gives the reaction of zinc salts (2.3.1).
Calculate the contents of C IOH 13N s0 4, C sH\\N 30 3 S and
C\SH\4N40 in the medium. B. A 5 per cent w/v solution in 2 M nitric acid gives the
reactions ofchlorides (2.3.1).
D. Not less than 70 per cent of the stated amounts of
C IOH 13Ns0 4, CsH\\N30 3S and C\SH\4N40. Tests
Other tests. Comply with the tests stated under Tablets. pH (2.4.24). 4.6 to 6.0, determined in a solution prepared by
dissolving 1.0 g in 9 ml of freshly boiled and cooled water,
Assay. Determine by liquid chromatography (2.4.14).
ignoring any slight turbidity.
Test solution. Weigh and powder 20 tablets. Weigh accurately
Aluminium, calcium, heavy metals, iron and magnesium. To
a quantity of the powder containing about 100 mg of
8 ml of solution A add 2 ml of strong ammonia solution and
Zidovudine and transfer to a 200-ml volumetric flask. Add
shake; the solution is clear (2.4.1), and colourless (2.4.1). Add
about 20 ml of methanol, mix with the aid of ultrasound for
1 ml of a 9 per cent w/v solution of disodium hydrogen
10 minutes, dilute to volume with water, mix and filter. Further
phosphate; the resulting solution remains clear for at least
dilute 10.0 ml ofthe filtrate to 25.0 ml with water.
5 minutes. Add 0.2 ml of sodium sulphide solution; a white
Reference solution. Weigh accurately about 100 mg of precipitate is produced and the supernatant liquid remains
zidovudine RS, 50 mg of lamivudine RS and 65 mg of nevirapine colourless.
RS and transfer to a 200-ml volumetric flask. Add about 20 ml
Ammonium salts. To 5 ml of a 10 per cent w/v solution add
of methanol, mix with the aid ofultrasound to dissolve, dilute
1 M sodium hydroxide until the precipitate fIrst formed is
to volume with water and mix. Further dilute 10.0 ml of this
redissolved and then warm the solution; no odour ofammonia
solution to 25.0 ml with water.
is perceptible.
Follow the chromatographic procedure described under
Oxychlorides. Dissolve 1.5 gin 1.5 ml of carbon dioxide-free
Dissolution.
water; the solution is not more opalescent than opalescence
Inject the reference solution. The tailing factor for the standard OS2 (2.4.1). Add 7.5 ml of ethanol (95 percent); the
individual peaks due to zidovudine, lamivudine and nevirapine solution may become cloudy within 10 minutes but becomes
is not more than 2.0 and the relative standard deviation for clear on the addition of 0.2 ml of 2 M hydrochloric acid.

2335

www.webofpharma.com
ZINC OXIDE IP 2010

Sulphates (2.3.17).15 ml ofsolution A complies with the limit solution AsT. The resulting solution complies with the limit
test for su1phates (225 ppm). test for arsenic (5 ppm).
Assay. Weigh accurately about 3.0 g, dissolve in 125 mlof Iron (2.3.14). Dissolve 0.1 gin a mixture of5 ml ofwater and
water, add 3 g ofammonium chloride and add sufficient water 1 ml ofhydrochloric acid and dilute to 40 ml with water. The
to produce 250.0 ml. To 25.0 ml ofthe resulting solution add resulting solution complies with the limit test for iron
100 ml of water and 10 ml of strong ammonia-ammonium (400 ppm). Use 0.5 ml of thioglycollic acid in the test.
chloride solution. Titrate with 0.1 M disodium edetate, using Lead. Dissolve 2 g in a mixture of 20 ml water and 5 ml of
eriochrome black T solution as indicator until a deep blue glacial acetic acid and add 0.25 ml of potassium chromate
colour is obtained. solution; the solution remains clear.
1 ml of 0.1 M disodium edetate is equivalent to 0.01363 g of Loss on ignition (2.4.20). Not more than 1.0 per cent, determined
ZnCh. on 2.0 g by igniting at 500°.
Storage. Store protected from moisture, in non-metallic Assay. Dissolve 0.15 gin 10 ml of2 M acetic acid and dilute to
containers. 50 ml with water. To the resulting solution add about 50 mg of
xylenol orange triturate and sufficient hexamine to produce
violet-pink colour. Add a further 2 g of hexamine and titrate
with 0.1 M disodium edetate until the solution becomes yellow.
1 ml of 0.1 M disodium edetate is equivalent to 0.008138 g of
Zinc Oxide ZnO.
ZnO Mol. Wt. 81.4 Storage. Store protected from moisture.
Zinc Oxide contains not less than 99.0 per cent and not
more than 100.5 per cent of ZnO, calculated on the ignited
basis.
Category. Mild astringent; topical protectant. Zinc Oxide Cream
Description. A soft, white or faintly yellowish white Zinc Cream
amorphous powder, free from grittiness. It gradually absorbs
Zinc Oxide Cream contains 32 per cent w/v ofZinc Oxide in a
carbon dioxide from air.
suitable water-in-oil emulsified base.
Identification Zinc Oxide Cream contains not less than 30.0 per cent and not
more than 34.0 per cent w/w ofzinc oxide, ZnO.
A. It becomes yellow when strongly heated; the yellow colour
disappears on cooling. Identification
B. Dissolve 0.1 gin 1.5 ml of2 M hydrochloric acid and dilute The residue obtained in the Assay is yellow when hot and
to 5 ml with water. The solution gives the reaction ofzinc salts white when cool.
(2.3.1).
Tests
Tests
Other tests. Complies with the tests stated under Creams.
Alkalinity. Shake 1.0 g with 10 ml ofboiling water, add 0.1 ml
Assay. Weigh accurately about 0.5 g in a porcelain dish, heat
ofphenolphthalein solution and filter. Ifthe filtrate is red, not
gently over a small flame until the base is completely volatilised
more than 0.3 ml of 0.1 M hydrochloric acid is required to
or charred. Increase the heat until all the carbon is removed.
discharge the colour.
Dissolve the residue in 10 ml of 2 M acetic acid and add
Carbonate and substances insoluble in acids. Dissolve 1.0 g sufficient water to produce 50 ml. To the resulting solution
in 15 ml of 2 M hydrochloric acid; no effervescence is add about 50 mg of xylenol orange triturate and sufficient
produced and the solution is not more opalescent than hexamine to produce violet-pink colour. Add a further 2 g of
. . ._.~
olfalescef1cestaf1dataOS2-(2.4-·n;~af1dc~ol(jatless-(2.4;1):~ nexamineand titrate with O:IM disodiilmedeti:tte~untilthe
solution becomes yellow.
Arsenic (2.3.10). Dissolve 2.0 g in 15 ml of brominated
hydrochloric acid AsT and 45 ml of water and remove the 1 ml of 0.1 M disodiumedetate is equivalent to 0.008138 g of
excess of bromine with a few drops of stannous chloride ZnO.

2336

www.webofpharma.com
IP 2010 ZINC SULPHATE

Zinc Stearate AsT. The resulting solution complies with the limit test for
arsenic (2 ppm).
Mol. Wt. 632.3
Heavy metals (2.3 .13). Heat 5.0 g with 40 ml of2 M acetic acid
Zinc Stearate consists mainly of zinc stearate but many and allow to cool. Filter, wash the residue with two successive
contain variable proportions ofzinc palmitate (C 1sH 3I COO)Z quantities, each of5 ml, ofwarm water and dilute the.combined
Zn, and zinc oleate (C I7 H33 COO)ZZn. filtrate and washings to 100.0 rnl with water. 12 rnl ofthe solution
Zinc Stearate contains not less than 10.0 per cent and not complies with the limit test for heavy metals, Method D
more than 12.0 per cent ofzinc, Zn. (20 ppm). Use 1.0 ml oflead standardsolution (10 ppm Pb) to
prepare the standard.
Category. Pharmaceutical aid (dusting powder).
Chlorides (2.3.12).10 ml ofsolution A complies with the limit
Description. A [me, white, bulky, amorphous powder, free from test for chlorides (250 ppm).
grittiness; odour, faint and characteristic.
Sulphates (2.3.17). 2.5 ml ofsolution A complies with the limit
Identification test for sulphates (0.6 per cent).

A. To 5.0 g add 50 ml of ether and 40 ml of a 7.5 per cent v/v Assay. Weigh accurately about 1.0 g and boil with 50 ml of
solution of nitric acid in distilled water and heat under a 2 M acetic acid until the fatty acid layer which separates is
reflux condenser until dissolution is complete. Allow to cool, clear, adding more water ifnecessary to maintain the original
separate the aqueous layer and shake the ether layer with two volume. Cool, filter and wash the filter and the flask thoroughly
quantities, each of 4 ml, of distilled water. Combine the with water until the last washing is not acidic to blue litmus
washings with the aqueous layer, wash with 15 ml of ether paper. To the combined filtrate and washings add about
and heat on a water-bath until ether is completely eliminated. 50 mg of xylenol orange triturate and sufficient hexamine to
Allow to cool and dilute to 50.0 ml with distilled water (solution produce violet-pink colour. Add a further 2 g ofhexamine and
A). Evaporate the ether layer to dryness and dry the residue titrate with 0.1 M disodium edetate until the colour changes
at 105°. The freezing point ofthe residue is notlower than 53° to yellow.
(2.4.11). 1 ml of 0.1 M disodium edetate is equivalent to 0.00654 g of
B. Neutralise 5 ml ofsolution A to red litmus paper with 10 M Zn.
sodium hydroxide. The solution gives the reactions of zinc
salts (2.3.1).

Tests Zinc Sulphate


Appearance of solution. Solution A is not more intensely ZnS04,7HzO Mol. Wt. 287.5
coloured than reference solution YS6 (2.4.1). Zinc Sulphate contains not less than 99.0 per cent and not
Acidity or alkalinity. Shake 1.0 g with 5 ml ofethanol (95 per more than 104.0 per cent ofZnS04, 7H zO.
cent) and add 20 ml ofcarbon dioxide-free water and 0.1 mlof Category. Astringent.
phenol red solution. Not more than 0.3 ml of 0.1 M
Description. Colourless, transparent crystals or a white,
hydrochloric acid or 0.1 ml of 0.1 M sodium hydroxide is
crystalline powder; odourless; efflorescent.
required to change the colour of the solution.
Alkalis and alkaline earths. Add 1.0 g to a mixture of25 ml of Identification
water and 5 ml of hydrochloric acid, boil, filter immediately Dissolve 2.5 g in sufficient carbon dioxide-free water to
and wash with 25 ml of hot water. Add dilute ammonia produce 50 ml (solution A). Solution A gives the reactions of
solution to make the filtrate just alkaline and then add zinc salts and sulphates (2.3.1).
ammonium sulphide solution in excess to precipitate the zinc
as zinc sulphide completely. Filter, add 0.5 ml of sulphuric Tests
acid to the filtrate, evaporate to dryness and ignite to constant
weight; the residue weighs not more than 20 mg. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Arsenic (2.3.1 0). Mix 5.0 g with 10 ml ofbromine solution and
evaporate to dryness on a water-bath. Ignite gently, dissolve pH (2.4.24). 4.4 to 5.6, determined in solution A.
the cooled residue, ignoring any carbon, in 50 ml ofwater and Arsenic (2.3.1 0). Dissolve 1.0 g in 50 nil ofwater and add 10 ml
14 ml of brominated hydrochloric acid AsT and remove the of stannated hydrochloric acid AsT. The resulting solution
excess of bromine with 2 ml of stannous chloride solution complies with the limit test for arsenic (10 ppm).

2337

www.webofpharma.com
ZINC SULPHATE EYE DROPS IP 2010

Iron (2.3.14). Dissolve 0.4 g in 20 m1 of water. The resulting Zinc Undecenoate contains not less than 98.0 per cent and
solution complies with the limit test for iron (100 ppm). not more than 102.0 per cent ofCzzH3s04Zn, calculated on the
Chlorides (2.3.12). 20 ml ofsolution A complies with the limit dried basis.
test for chlorides (250 ppm). Category. Antifungal (topical).
Assay. Weigh accurately about 0.5 g and dissolve in 5 ml of Description. A white or almost white, fine powder.
2 M acetic acid and dilute to 50 ml with water. To the resulting
solution add about 50 mg of xylenol orange triturate and Identification
sufficient hexamine to produce violet-pink colour. Further,
add 2 g of hexamine and titTate with 0.1 M disodium edetate A. To 2.5 g add 10 ml of water and 10 ml of1 M sulphuric acid
until the colour changes to yellow. and extract with two quantities, each of 10 ml, ofether. Reserve
the aqueous layer for test C. Wash the combined ether extracts
1 ml 0/0.1 M disodium edetate is equivalent to 0.02875 g of with water and evaporate to dryness. To the residue add 2 ml
ZnS04,7HzO. of freshly distilled aniline and boil under a reflux condenser
Storage. Store protected from moisture, in non-metallic for 10 minutes, cool and add 30 ml of ether. Extract with three
containers. quantities, each of 20 ml, of 2 M hydrochloric acid and then
with 20 ml of water. Evaporate the ether extract to dryness on
a water-bath. The residue, after recrystallising twice from
Zinc Sulphate Eye Drops ethanol (70 per cent) and drying at a pressure not exceeding
2 kPa for 3 hours, melts at about 67° (2.4.21).
Zinc Sulphate Eye Drops are a sterile solution containing
0.25 pet cent \>.iN of Zinc Sulphate ifiPtirified Water. B. Dissolve 0.1 g in a mixture of2 mLof1 Msulphuric acidand
5 ml of glacial acetic acid and add, dropwise, 0.25 ml of
Zinc Sulphate Eye Drops contain not less than 0.22 per cent potassium permanganate solution; the pink colour of
and not more than 0.28 per cent w/v of zinc sulphate, ZnS04, permanganate is discharged.
7HzO.
C. A mixture of 1 ml ofthe aqueous layer reserved in test A and
Usual strenghts. 200 mg; 500 mg; 1 g. 4 ml of water gives the reaction of zinc salts (2.3.1).
Description. A clear, colourless solution. D. Melting range (2.4.21). 115° to 121°.

Identification Tests
Give the reactions of zinc salts and of sulphates (2.3.1). Alkalinity. Mix 1.0 g with 5 ml of ethanol (95 per cent) and
0.5 ml of phenol red solution, add 50 ml of carbon dioxide-
Tests
free water and examine immediately; no reddish colour is
Other tests. Comply with the tests stated under Eye Drops. produced.
Assay. To 5.0 rnl add 50 ml of water and 5 ml ofammonia bliffer Allmlis and alkaline earths. Add 1.0 g to a mixture of25 mlof
pH 10.9 and titrate with 0.01 M disodium edetate using water and 5 ml of hydrochloric acid, boil, filter immediately
mordant black II solution as indicator. and wash with 25 ml of hot water. Add dilute ammonia
1 ml of 0.01 M disodium edetate is equivalent to 0.002875 g of solution to make the filtrate just alkaline and then add
ZnS0 4,7HzO. ammonium sulphate solution in excess to precipitate the zinc
as zinc sulphide completely. Filter, add 0.5 ml of sulphuric
Storage. Store in containers ofglass or any other non-metallic acid to the filtrate, evaporate to dryness and ignite to constant
material and sealed so as to exclude micro-organisms. weight; the residue weighs not more than 20 mg.
Sulphates (2.3.17). To 0.25 g add a mixture ofl 0 rnl of distilled
water and 5 ml of 2 M hydrochloric acid. Cool, filter and
Zinc Undecenoate dilute to 20 ml with distilled water. The resulting solution
complies with the limit test for sulphates (600 ppm).
Degree of unsaturation. Dissolve 0.1 g in a mixture of5 mlof
2Mhydrochloricacid and 30 ml of glCic.:ial aceticacid and
titrate
wTth~0~05Mbro/;ZineusiniO.05ml ofethoXychrysoidine
hydrochloride solution, added towards the end ClfthetihitiClll,
CZZH3S04Zn Mol. Wt. 431.9 as indicator. Not less than 9.1 ml and not more than 9.4 ml of
Zinc Undecenoate is zinc di(undec-l O-enoate). 0.05 M bromine is required to discharge the red colour.

2338

www.webofpharma.com
IP 2010 ZOLEDRONIC ACID

Loss on drying (2.4.19). Not more than 1.5 per cent, determined small separator and drain the carbon tetrachloride layer into
on 0.5 g by drying in an oven at 105°. a 100-ml volumetric flask. Rinse the beaker with three
quantities, each of 5 ml, of carbon tetrachloride and transfer
Assay. Weigh accurately about 0.35 g, add 25 m1 of2 M acetic
the rinsings to the volumetric flask, dilute to volume with
acid, heat to boiling, cool and dilute to 50 ml with water. Add
carbon tetrachloride and mix. Evaporate 50.0 ml ofthe resulting
about 50 mg of xylenol orange triturate and sufficient
solution to about 5 ml, add 100 ml of ethanol (95per cent),
hexamine to produce a violet-pink colour. Add a further 2 g of
previously neutralised, and 0.15 ml of phenolphthalein
hexamine and titrate with 0.1 M disodium edetate until the
colour changes to yellow.
solution and titrate the total undecenoic acid with
0.1 M sodium hydroxide.
1 ml of 0.1 M disodium edetate is equivalent to 0.04319 g of
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01843 g of
C22H3S04Zn.
CIIH2002.
Storage. Store protected from light and moisture..
Calculate the content of free undecenoic acid from the
difference between the total undecenoic acid and the
undecenoic acid equivalent to the determined zinc
undecenoate (the content of zinc undecenoate multiplied by
Zinc Undecenoate Ointment 0.8533 gives the equivalent ofundecenoic acid).
Zinc Undecylenate Ointment Storage. Store protected from light and moisture.
Zinc Undecenoate Ointment contains 20 per cent w/v of Zinc
Undecenoate in a suitable ointment basis.
Zinc Undecenoate Ointment contains not less than 18.0 per Zoledronic acid
cent and not more than 22.0 per cent of zinc undecenoate,
C22H3S04Zn, and not less than 4.5 per cent and not more than
5,5 per cent of free undecenoic acid, CIIH2002.

Tests
Other tests. Complies with the tests stated under Ointments.
Assay. For zinc undecenoate - Weigh accurately about
2.0 g, add 20 ml of dilute hydrochloric acid and boil under a Mol. Wt. 272.1
reflux condenser for at least 20 minutes or until the fatty layer
is clear. Filter while hot and wash the residue with hot water. ZoledronicAcid is [1-hydroxy-2-(lH-imidazol-l-
Cool the combined filtrate and washings, neutralise to litmus yl)ethylidene]diphosphonic acid.
paper with dilute ammonia solution, add 3 ml of dilute Zoledronic Acid contains not less than 98.0 per cent and not
hydrochloric acid and 5 g of hexamine and titrate with more than 102.0 per cent of CsHIQN20 7P2, calculated on the
0.05 M disodium edetate using xylenol orange solution as dried basis.
indicator, until the colour changes to yellow.
Category. Bone resorption inhibitor.
1 mlof 0.05 M disodium edetate is equivalent to 0.02160 g of
Description. A white to almost white crystalline powder.
C22H3S04Zn.
For ji-ee undecenoic acid Weigh accurately about 5.0 g, Identification
add 100 ml of dilute hydrochloric acid and heat to 70° with
constant stirring. Cool and transfer to a separator with the aid A. Determine by infrared absorption spectrophotometry (2.4.6).
of four quantities, each of 25 ml of ether and add the rinsings Compare the spectmm with that obtained with zoledronic
to the mixture in the separator. Dilute the aqueous phase to acidRS or with the reference spectrum ofzoledronic acid.
300 ml, saturate it with sodium chloride and shake the mixture. B. In the assay, the principal peak in the chromatogram
Transfer the aqueous layer to a second separator and extract obtained with the test solution corresponds to the peak in the
with another 100 ml of ether. Wash the combined ether extracts chromatogram obtained with the reference solution.
with successive quantities, each of 10 ml, of water until the
washings are free from chloride. Transfer the ether solution to Tests
a beaker and evaporate on a water-bath to about 5 ml. Add
20 ml of carbon tetrachloride, mix, transfer the mixture to a pH (2.4.24). 2.0 to 4.0, determined in a 0.02 per cent w/v solution.

2339

www.webofpharma.com
ZOLEDRONIC ACID IP 2010

Related substances. Determine by liquid chromatography Zoledronic Acid Injection


(2.4.14).
Zoledronic Acid Injection is prepared by dissolving the
Test solution. Dissolve 50 mg of the substance uJider contents of a sealed container containing Zoledronic Acid
examination in 50.0 m1 ofthe mobile phase. with or without auxiliary substances in the requisite a.mount
Reference solution. Dilute 1 ml of the test solution to 100 ml of water for injection as directed on the label.
with the mobile phase. The constituted solution complies with the requirements for
Chromatographic system Clarity of solution and Particulate matter stated under
a stainless steel column 25 em x 4 mm, packed with Parenteral Preparations (Injections).
octadecylsilane bonded to porous silica (5 /lm) (Such Usual strength. 4 mg per ml.
as Hypersil BDS),
- mobile phase: mix 1 ml of triethylamine with 500 ml of Storage. The constituted solution should be used immediately
water, adjust the pH to 3.2 with orthophosphoric acid, after preparation but, in any case, within the period
- flow rate. 1 ml per minute, recommended by the manufacturer.
spectrophotometer set at 215 nm, Zoledronic Acid Injection contains not less than 90.0 per cent
injection volume. 20 Ill. and not more than 110.0 per cent ofstated amount ofzoledronic
Inject the reference solution. The test is not valid unless the acid, CSHION207P2.
column efficiency is not less than 2000 theoretical plates and The contents of the sealed container comply with the
the tailil1g factor is not more than 2.0. requirements stated under Parenteral Preparations
Inject the test solution and the reference solution. Run the (Powders for Injection) and with thefollowing requirements:
chromatogram 3 times the retention time ofthe principal peak.
In the chromatogram obtained with the test solution, area of Identification
any secondary peale is not more than the area ofthe principal
In the Assay, the principal peak in the chromatogram obtained
peak in the chromatogram obtained with the reference solution
with the test solution corresponds to the peak in the
(1.0 per cent). The sum of areas of all the secondary peaks is
chromatogram obtained with the reference solution.
not more than twice the area of the principal peak in the
chromatogram obtained with the reference solution (2.0 per Tests
cent). Ignore any peak with an area less than 0.1 times the
area of the peak in the chromatogram obtained with the pH (2.4.24).4.5 to 7.0, determined in a solution constituted as
reference solution (0.1 per cent). directed on the label, in Water for Injections.
Loss on drying (2.4.19).5.0 to 8.0 per cent, determined on Other tests. Complies with the tests stated under Parental
1.0 g by drying in an oven at 105° for 6 hours. Preparations (Injections).
Assay. Determine by liquid chromatography (2.4.14). Bacterial endotoxins (2.2.3). Not more than 17.0 Endotoxin
Test solution. Dissolve 50 mg of the substance under Units per mg of Zoledronic acid.
examination in 50.0 ml ofthe mobile phase. Dilute 20 ml ofthe Sterility (2.2.11). Complies with the test for sterility.
solution to 100 ml with the mobile phase.
Assay. Determine by liquid chromatography (2.4.14).
Reference solution. A 0.02 per cent w/v solution ofzoledronic
acid RS in the mobile phase. Test solution. Dissolve the contents of sealed container in
sufficient mobile phase to give a concentration 0.16 mg per m!.
Use the chromatographic system as described under Related
Reference solution. A 0.0 16 per cent w/v solution ofzoledronic
substances.
acid RS in the mobile phase.
Inject the reference solution. The test is not valid unless the
Chromatographic system
relative standard deviation for replicate injections is not more
than 2.0 per cent. a stainless steel column 25 em x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 J.lm) (Such
Inject the test solution and the reference solution. as Hypersil BDS),
Calculate-the-content-ofCsHloN2Q7P2.--------------------------- rnobilephase:.. .mix..l..ml_oftr.iethylaminewith..5_0.0mlo f
water, adjusted to pH: 3.2 with orthophosphoric acid,
Storage. Store protected froIIllight a.nd IIloisture. flow rate. 1 ml per minute,
Labelling. The label states whether or not the material· is spectrophotometer set at 215 urn,
intended for use in the manufacture ofparenteral preparations. injection volume. 20 Ill.

2340

www.webofpharma.com
IP 2010 ZOLPIDEM TARTRATE

Inject the reference solution. The test is not valid unless the Reference solution (a). A 0.01 per cent w/v solution of N,N-
relative standard deviation for replicate injections is not more dimethyl- 2-[7-methyl-2-(4-methylphenyl) imidazo[1 ,2-aJ
than 2.0 per cent. pyridin-3-yl acetamide RS (zolpidem impurity A RS) in the
mobile phase.
Inject the test solution and the reference solution.
Reference solution (b). A 0.05 per cent w/v solution of
Calculate the content ofCsHIONz07Pz in the Injection.
zolpidem tartrate RS in the mobile phase.
Storage. Store the sealed container at a temperature not
Reference solution (c). Dilute 1.0 ml ofthe reference solution
exceeding 30°. Use the prepared solution within the period
(b) to 100.0 ml with the mobile phase.
recommended by the manufacturer and stored strictly in
accordance with the instructions of the manufacturer. Reference solution (d). To 2.0 ml of reference solution (b),
add 10.0 ml ofreference solution (a).
Chromatographic system
Zolpidem Tartrate - a stainless steel column 15 cm x 3.9 rom, packed with
octadecylsilane bonded to porous silica (4 J.tm),
- mobile phase: 18 volumes of acetonitrile, 23 volumes
of methanol and 59 volumes of 0.56 per cent w/v
solution of orthophosphoric acid adjusted to pH 5.5
with triethylamine,
- flow rate. 1.5 ml per minute,
- spectrophotometer set at 254 nm,
- injection volume. 20 J.tl.
Inject reference solution (d). Adjust the sensitivity of the
system so that the height ofthe peak due to zolpidem impurity
Mol. Wt. 765.0 A is at least 50 per cent of the full scale of the recorder. The
test is not valid unless the resolution between the peaks due
Zolpidem Tartrate is bis[N,N-dimethyl-2-[6-methyl-2-(4- to zolpidem impurity A and zolpidem tartrate is at least 2.0.
methylphenyl)imidazo[1,2-a]pyridine-3-yl]acetamide] tartrate.
.Inject the test solution and reference solution (c). In the
Zolpidem Tartrate contains not less than 98.5 per cent and not
chromatogram obtained with the test solution, the area of any
more than 101.0 percent Of(CI9HzIN30)Z' C 4H 60 6, calculated secondary peak is not more than 0.2 times the area ofthe peak
on the anhydrous basis.
in the chromatogram obtained with reference solution (c)
Category. Sedatives. (0.2 per cent) and the sum of the areas of all the secondary
peaks is not more than the area ofthe peak in the chromatogram
Description. A white or almost white crystalline powder.
obtained with reference solution (c) (1.0 per cent). Ignore any
Identification peak with a relative retention time of0.16 to the zolpidem peak,
corresponding to tartaric acid and any secondary peak
A. Detenmne by infi:ared absorption spectrophotometry (2.4.6). with an area less than 0.05 times the area of the peak in
Compare the spectrum with that obtained with zolpidem the chromatogram obtained with reference solution (c)
tartrate RS or with the reference spectrum ofzolpidem tartrate. (0.05 per cent).
B. Al 0 per cent w/v solution in methanol gives reaction (c) of Sulphated ash (2.3.18). Not more than 0.1 percent.
tartrates (2.3.1).
Water (2.3.43). Not more than 3.0 per cent, detennined on
Tests 0.5g.

Appearance ofsolution. Dissolve 1.0 g ofthe substance under Assay. Weigh accurately about 0.3 g, dissolve in a mixture of
examination in 100 ml ofa 0.5 per centw/v solution of tartaric 20 ml of anhydrous acetic acid and 20 ml of acetic anhydride.
acid. The solution is clear (2.4.1), and not more intensely Titrate with 0.1 M perchloric acid, detennining the end point
coloured than reference solution YS6 or BYS6 (2.4.1). potentiometrically (2.4.25). Carry out a blank titration

Related substances. Detennine by liquid chromatography 1 ml of 0.1 M perchloric acid is equivalent to 0.03824 g of
(2.4.14). (CI9HzIN30)Z, C~606'
Test solution. Dissolve 25 mg of the substance under Storage. Store protected from light and moisture, at a
examination in 50.0 ml ofthe mobile phase. temperature not exceeding 30°.

2341

www.webofpharma.com
ZOLPIDEM TABLETS IP 2010

Zolpidem Tablets Test solution. Disperse a quantity of the powdered tablets


containing 25 mg ofZolpidem Tartrate with 25 ml ofthe mobile
Zolpidem Tartrate Tablets phase and dilute to 50.0 ml with the same solvent and filter.
Zolpidem Tablets contain not less than 90.0 per cent and not Reference solution (a). A 0.01 per cent w/v solution of N,N-
more than 110.0 per cent of the stated amount of zolpidem dimethyl- 2-[7-methyl-2-(4-methylphenyl) imidazo[1,2-a]
tartrate, C42RtsN60S' pyridin-3-yl acetamide RS (zolpidem impurity A RS) in the
mobile phase.
Usual strengths. 5 mg; 10 mg.
Reference solution (b). A 0.05 per cent w/v solution of
Identification zolpidem tartrate RS in the mobile phase.
Reference solution (c). Dilute 1.0 ml of the reference solution
Shake a quantity ofthe powdered tablets containing 0.1 g of (b) to 100.0 ml with the mobile phase.
Zolpidem Tartrate with 40 m1 of methanol, filter and evaporate
the filtrate to dryness. The residue complies with the following Reference solution (d). To 2.0 ml of reference solution (b),
test. add 10.0 ml ofreference solution (a).
Chromatographic system
A. Determine by infrared absorption spectrophotometry (2.4.6).
a stainless steel column 15 cm x 3.9 mm, packed with
Compare the spectrum with that obtained with zolpidem
octadecylsilane bonded to porous silica (4 J!m),
tartrate RS or with the reference spectrum ofzolpidem tartrate.
- mobile phase: 18 volumes of acetonitrile, 23 volumes
B. In the Assay, the principal peak in the chromatogram of methanol and 59 volumes of0.56 per cent w/v solution
obtained with the test~olutiQ!1 QOITesPQIlclS to the R~al.<: in the of orthophosphoric acid adjusted to pH 5.5 with
chromatogram obtained with the reference solution. triethylamine,
flow rate. 1.5 ml per minute,
Tests spectrophotometer set at 254 nm,
- injection volume. 20 J!l.
Dissolution (2.5.2).
Inject reference solution (d). Adjust the sensitivity of the
Apparatus No.1, system so that the height ofthe peak-due to zolpidem impurity
Medium. 900 ml of 0.01 Mhydrochloric acid, A is at least 50 per cent of the full scale of the recorder. The
Speed and time. 50 rpm and 30 minutes. test is not valid unless the resolution between the peaks due
Withdraw a suitable volume ofthe medium and filter. to zolpidem impurity A and zolpidem tartrate is at least 2.0.
Inject the test solution and reference solution (c). In the
Determine by liquid chromatography (2.4.14).
chromatogram obtained with the test solution, the area of any
Test solution. Dilute the filtrate, if necessary, with the secondary peak is not more than 0.5 times the area ofthe peak
dissolution medium. in the chromatogram obtained with reference solution (c)
(0.5 per cent) and the sum of the areas of all the secondary
Reference solution. Dissolve 0.01 g'of zolpidem tartrate RS
peaks is not more than twice the area of the peak in the
in sufficient methanol to produce 100 ml. Dilute the resulting
chromatogram obtained with reference solution (c) (2.0 per
solution with the dissolution medium to obtain a solution
cent). Ignore any peak with a relative retention time of0.16 to
containing 0.0005 per cent w/v ofzolpidem tartrate.
the zolpidem peak, corresponding to tartaric acid and any
Use the chromatographic system described in the test for secondary peak with an area less than 0.05 times the area of
Related substances. the peak in the chromatogram obtained with reference solution
(c) (0.05 per cent).
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more Uniformity of content. Comply with the test stated under
than 2.0 per cent. Tablets.

Inject the reference solution and the test solution. Determine by liquid chromatography (2.4.14), as described in
the Assay, using the following solution as the test solution.
Calculate the content ofC42H4sN60s.
Test solution. Disperse one tablet in the mobile phase, mix and
D;-Not-lessthan-7-5-per ·cent-of-the--statedamount-of diluteto····obtain··a-solution-containing-0;005per-cent-w/vof
C42RtsN60S- zolpidem tartrate and filter.
Related substances. Determine by liquid chromatography Other tests. Comply with the tests stated under Tablets.
(2.4.14). Assay. Determine by liquid chromatography (2.4.14).

2342

www.webofpharma.com
IP 2010 ZOLPIDEM TABLETS

Test solution. Weigh and powder 20 tablets. Weigh accurately Inject the reference solution. The test is not valid unless the
a quantity ofpowder containing 25 mg ofZolpidem Tartrate, tailing factor is not more than 2.0 and the relative standard
disperse in 25 ml ofthe mobile phase and dilute to 50.0 ml with deviation for replicate injections is not more than 2.0 per cent.
same solvent and filter. Dilute 5.0 ml ofthe solution to 50.0 ml
with the mobile phase. Inject the test solution and the reference solution.
Reference solution. A 0.005 per cent w/v solution ofzolpidem Calculate the content of C42H4SN60S in the tablets.
tartrateRS in the mobile phase.
Use the chromatographic system described in the test for Storage. Store protected from light and moisture, at a
Related substances. temperature not exceeding 30°.

2343

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

VACCINES AND IMMUNOSERAFORHUMAN USE

General Requirements
Vaccines 2347
Antisera 2349
Monographs
Adsorbed Diphtheria, Tetanus and Hepatitis B (rDNA) Vaccine 2350
Adsorbed Diphtheria, Tetanus, Pertussis (Acellular Component) and
Haemophilus Type b Conjugate Vaecine 2351
Adsorbed Diphtheria, Tetanus, Pertussis (Acellular Component) and
Hepatitis B (rDNA) Vaccine 2354
Adsorbed Diphtheria, Tetanus, Pertussis (Acellular Component),
Inactivated Poliomyelitis Vaccine and Haemophilus Type b Conjugate Vaccine 2356
Adsorbed Diphtheria, Tetanus, Pertussis (Acellular Component) and
Inactivated Poliomyelitis Vaccine 2359
Adsorbed Diphtheria, Tetanus, Pertussis and Poliomyelitis (Inactivated) Vaccine 2361
Adsorbed Diphtheria, Tetanus, Pertussis, Poliomyelitis (Inactivated) and
Haemophilus Type b Conjugate Vaccine 2364
Adsorbed Pertussis Vaccine (Acellular Component) 2366
Adsorbed Pertussis Vaccine (Acellular, Co-purified) 2369
Bacillus Calmette-Guerin Vaccine (Freeze-Dried) 2371
DiphtheriaAntitoxin 2374
Diphtheria and Tetanus Vaccine (Adsorbed) 2375
Diphtheria and Tetanus Vaccine (Adsorbed) for Adults andAdolescents 2376
Diphtheria, Tetanus and Pertussis Vaccine (Adsorbed) 2378
Diphtheria, Tetanus, Pertussis (Whole Cell), Hepatitis B (rDNA) and
Haemophilus Type b Conjugate Vaccine (Adsorbed) 2379
Diphtheria, Tetanus, Pertussis (Whole Cell) Hepatitis B (rDNA)
Vaccine (Adsorbed) 2382
Diphtheria, Tetanus, Pertussis (Whole Cell) and Haemophilus Type b
Conjugate Vaccine (Adsorbed) 2384
Diphtheria Vaccine (Adsorbed) 2387
Gas-Gangrene Antitoxin (Oedematiens) 2391
Gas-Gangrene Antitoxin (perfringens) 2392
Gas-Gangrene Antitoxin (Septicum) 2393

2345

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Mixed Gas-Gangrene Antitoxin 2394


Haemophilus Type b Conjugate Vaccine 2395
Hepatitis A (Inactivated) and Hepatitis B (rDNA) Vaccine (Adsorbed) 2398
Hepatitis B Vaccine (rDNA) 2399
Inactivated Hepatitis AVaccine (Adsorbed) 2402
InactivatedHepatitis B Vaccine 2404
Inactivated Influenza Vaccine (SplitVIrion) 2406
Inactivated Influenza Vaccine (Surface Antigen) 2408
Inactivated InfluenzaVaccine (Whole VIrion) 2410
Japanese Encephalitis Vaccine (Human) 2411
Measles and Rubella Vaccine (Live) 2414
Measles Vaccine (Live) 2415
Measles, Mumps and Rubella Vaccine (Live) 2416
Meningococcal Polysaccharide Vaccine 2417
Mumps Vaccine (Live) 2420
Pertussis Vaccine 2421
Plague Vaccine 2423
Pneumococcal Polysaccharide Vaccine 2424
Poliomyelitis Vaccine (Inactivated) 2426
Poliomyelitis Vaccine, Live (Oral) 2430
RabiesAntiserum 2435
Rabies Vaccine, Human 2436
Rubella Vaccine (Live) 2439
Snake VenomAntiserum 2441
TetanusAntitoxin 2441
Tetanus Vaccine (Adsorbed) 2443
Tick-borne Encephalitis Vaccine (Inactivated) 2447
Tuberculin Purified Protein Derivative 2449
Typhoid (Strain Ty 21 a) Vaccine, Live (Oral) 2451
Typhoid Polysaccharide Vaccine 2452
Typhoid ParatyphoidAVaccine 2454
Typhoid Vaccine 2455
Typhoid Vaccine (Freeze Dried) 2455
Iypbm; Ya,GGiJ:l~ 2455
.VaricellaVaccine (Live) 2456
Viper Venom 2458
Yellow Fever Vaccine (Live) 2458

2346

www.webofpharma.com
IP 2010 VACCINES: GENERAL REQUIREMENTS

Vaccines : General Requirements be freeze-dried so that the water content is not more than 3.0
per cent w/w unless otherwise stated in the individual
Vaccines are preparations of antigenic substances that are monograph. They may be standardized in tenns ofinteropacity
administered for the purpose of inducing in the recipient a units or, where appropriate, by numbers of living or killed
specific and active immunity against the infective agent or bacteria detennined by direct cell count or by viable count.
toxin produced by it.
Bacterial toxoids. Bacterial toxoids are toxins or material
Vaccines may contain living micro-organisms suitably treated derived therefrom, the toxicity ofwhich has been reduced to a
to attenuate their virulence but retain their immunogenicity or very low level or completely eliminated by,chemical or physical
they may consist of pathogenic organisms which have been means without destroying their immunizing potency. The toxins
killed or inactivated. Some vaccines consist of antigenic are obtained from selected strains ofspecific micro-organisms,
fractions or substances produced by the same pathogenic grown in media free from ingredients known to cause toxic,
organisms but rendered harmless whilst retaining their allergic or other undesirable immunological reactions in
immunogenicity. Vaccines may b~ prepared from one species humans. Toxoids produced by the action offonnaldehyde are
only or from a mixture oftwo or more species. lmown as formol toxoids.
Vaccines may be prepared by the method described in the Bacterial toxoids may be liquid or may be prepared by adsorbing
individuill monographs or by the general methods given below on mineral carriers such as aluminium phosphate, aluminium
or in any other manner provided the identity ofthe antigens is hydroxide or any other suitable adsorbent; the adsorbed
maintained and the vaccines are free from microbial product may be separated, washed and suspended in a saline
contamination and extraneous agents. Suitable adjuvants may or other appropriate solution isotonic with blood.
be added during the preparation but streptomycin, penicillin
or other p-Iactam antibiotics may not be added at any stage of Bacterial toxoids are clear or slightly opalescent liquids,
manufacture or in the final vaccine. A suita.ble microbicide colourless or slightly yellow. Adsorbed toxoids may be white
may be added to sterile and inactivated vaccines. The final or greyish white suspensions or pale-yellow liquids with a
products are distributed aseptically into sterile containers sediment at the bottom of the container. Freeze-dried
which are then sealed to exclude extraneous micro-organisms. preparations are greyish white or yellowish white powders or
Unless otherwise indicated in the monograph, thefinal vaccine pellets.
may be filled into single dose or multiple dose containers but Viral and rickettsial vaccines. Viral and rickettsial vaccines
vaccines in multiple dose containers must invariably contain are suspensions of viruses or rickettsiae and are prepared
a microbicide. from infected tissues or blood obtained from artificially infected
animals, from cultures in fertile eggs, or from cell or tissue
Bacterial Vaccines. Bacterial vaccines are either sterile
culture. Viral vaccines may be live or killed and'they may be
suspensions of live or killed bacteria or sterile extracts of
derivatives ofbacteria. They may be simple vaccines prepared freeze-dried. Live vaccines are usually prepared using
from one species or may be mixed vaccines prepared by attenuated strains of the specific organisms. Killed vaccines
may be inactivated by suitable chemical or physical means.
blending two or more simple vaccines from different species
or strains. Mixed Vaccines. Mixed vaccines are mixtures oftwo or more
Bacterial vaccines may be prepared from cultures grown on vaccines. A suitable antibacterial substance may be added to
suitable solid or liquid media. The whole culture or parts of it inactivated or live viral and rickettsial vaccines provided that
may be used in preparing the vaccine. The identity, antigenic it has no action against the specific organisms.
potency and purity ofeach bacterial culture must be carefully
Production
controlled.
Vaccines containing killed organisms may be prepared by General provisions. Requirements for production including
killing the organisms by chemical or physical means provided in-process testing are included in individual monographs.
the immunogenicity of the vaccine is preserved. Vaccines Where justified and authorized, certain tests may be omitted
containing living bacteria may be prepared from str~ins which where it can be demonstrated, for example by validation
are avirulent for humans but can stimulate the production of studies, that the production process consistently ensures
antibodies active against pathogenic strains of the same compliance with the test.
species. The final vaccines must be free from any substance Unless otherwise justified and authorized, vaccines are
known to cause toxic, allergic or other undesirable produced using a seed-lot system. The methods ofpreparation
immunological reactions in humans. are designed to maintain adequate immunogenic properties,
Bacterial vaccines are suspensions of varying degrees of to render the preparation harmless and to prevent
opacity in colourless or slightly coloured liquids or they may contamination with extraneous agents.

2347

www.webofpharma.com
VACCINES: GENERAL REQUIREMENTS IP 2010

Unless otherwise justified and authorized, in the production Control eggs. For live vaccines produced in SPF eggs, control
of a final lot ofvaccine, the number ofpassages of a virus, or eggs are incubated and tested as prescribed in the monograph.
the number of subcultures of a bacterium, from the master
Purification. Where applicable, validated purification
seed lot shall not exceed that used for production ofthe vaccine
procedures may be applied.
shown in clinic~.l studies to be satisfactory with respect to
safety and efficacy. Inactivation. Inactivated vaccines are produced using a
validated inactivation process whose effectiveness and
Vaccines are as far as possible free from ingredients Imown to consistency have been demonstrated. Where there are
cause toxic, allergic or other undesirable reactions in man. recognised potential contaminants of a harvest, for example
Suitable additives, including stabilizers and adjuvants may be in vaccines produced in eggs from healthy, non-SPF flocks,
incorporated. Penicillin and streptomycin are neither used at . the inactivation process is also validated with respect to the
any stage of production nor added to the final product; potential contaminants. A test for inactivation is carried out
however, master seed lots prepared with media containing as soon as possible after the inactivation process, unless
penicillin Or streptomycin may, where justified and authorized, otherwise justified and authorised.
be used for production.
Intermediates. Where applicable, the stability ofintermediates
Substrates for propagation. Substrates for propagation in given storage conditions shall be evaluated and a period of
comply with the relevant requirements ofthe Pharmacopoeia validity .established.
or in the absence of such requirements with those of the
competent authority. Processing of cell banks and subsequent Final bulk. The final bulk is prepared by aseptically blending
cell cultures is done under aseptic conditions in an area where the ingredients of the vaccine.
no other cells are being handled. Serum and trypsin used in Adsorbents. Vaccines rtlay be adsorbed on aluminium
the preparation of cell suspensions shall be shown to be fi:ee hydroxide, aluminium phosphate, calcium phosphate or other
from extraneous agents. suitable adsorbent; the adsorbents are prepared in special
Seed lot. The strain of bacterium or virus used in a master conditions which confer the appropriate physical form and
seed lot is identified by historical records that include adsorptive properties.
information on the origin of the strain and its subsequent Antimicrobial preservative. A suitable antimicrobial
manipulation. No micro-organism other than the seed strain preservative may be included in sterile and inactivated
shall be present in a seed lot. vaccines and is invariably added if these preparations are
Culture media. Culture media are as far as possible free from issued in multidose containers, unless otherwise stated. If an
ingredients Imown to cause toxic, allergic or other undesirable antimicrobial preservative is used, it shall be shown that it
reactions in man; ifinclusion of such ingredients is necessary, does not impair the safety or efficacy of the vaccine and its
it shall be demonstrated that the amount present in the final effectiveness throughout the period of validity shall be
lot is reduced to such a level as to render the product safe. demonstrated.
Approved animal (but not human) serum may be used in the Final lot. For vaccines for parenteral administration, the final
growth medium for cell cultures but the medium used for lot is prepared by aseptically distributing the final bulle into
maintaining cell growth during virus multiplication shall not sterile tamper-proofcontainers which, after freeze-drying where
contain serum, unless otherwise stated. Cell culture media applicable, are closed so as to exclude contamination. For
may contain a pH indicator such as phenol red and approved vaccines for administration by a non-parenteral route, the final
antibiotics at the lowest effective concentration although it is lot is prepared by distributing the final bulle under suitable
preferable to have a medium free from antibiotics during conditions into sterile, tamper-proof containers.
production.
Stability. Maintenance of potency ofthe final lot throughout
Propagation and harvest. The seed cultures are propagated the period of validity shall be demonstrated by validation
and harvested under defined conditions. The purity of the studies; the loss of potency in the recommended storage
harvest is verified by suitable tests as defined in the conditions is assessed and excessive loss even within the
monograph. limits of acceptable potency may indicate that the vaccine is
unacceptable.
Control cells. For vaccines produced in cell cultures, control
cells. are maintained and tested as prescribed. In order to Degree of adsorption. During development of an adsorbed
provIde av~aHdcontror these ce1fsmustbemalntalnedin vaccIne; the-degreeof adsorptlon!seva1uatedaspartoftiie
conditions that are rigorously identical with those used· for consistency testing. A release specification for the degree of
the production cell cultures, including use ofthe same batches adsorption is established in the light of results found for
of media and media changes. batches used in clinical testing. From the stability data

2348

www.webofpharma.com
IP 2010 ANTISERA

generated for the vaccine it must be shown that at the end of Antisera
the period ofvalidity the degree of adsorption will not be less
than for batches used in clinical testing. Immunosera
Antisera are native (unconcentrated) sera or preparations from
Tests
native sera containing specific immunoglobulins that have
Vaccines, reconstituted where necessary, comply with the prophylactic or therapeutic action when injected into persons
following requirements unless otherwise stated in the exposed to or suffering from a disease caused by a specific
individual monograph. micro-organism.
Phenol (Ifpresent) (2.3.36). Not more than 0.25 per cent w/v.
Antisera are prepared by injecting antigens which are
Thiomersal (Ifpresent) (2.3.48). Between 0.005 per cent w/v preparations of cultures of the specific organisms or venoms
and 0.02 per cent w/v. or their products into healthy humans or animals such as
Free formaldehyde (Ifpresent) (2.3.20). Maximum 0.2 gil. horses so as to produce in them antibodies which are normally
Aluminium (Ifpresent) (2.3.9). Not more than 1.25 mg per associated with the globulin fraction of serum. Antigens
dose. commonly used for this purpose are toxins, toxoids, venoms,
bacterial and viral vaccines. Non-lethal amounts of the toxin
Sterility (2.2.11). Unless otherwise stated all vaccines comply or the corresponding toxoid/vaccine are injected in gradually
with tests for sterility, except that for living bacterial vaccines, increasing doses into animals. Specific antitoxins are formed
growth of the organism from which the vaccine was prepared in the serum and the animals become actively immune. During
is permitted (sterility means absence of bacterial and fungal the process of immunisation, the animals should not be treated
contaminants except where specified in the individual with penicillin. When a satisfactory degree ofthe immunity is
monograph). produced, larger volumes of blood are withdrawn from the
Abnormal toxicity (2.2.1). Unless otherwise stated, all vaccines animals and the plasma or serum is processed to :produce
comply with the test for abnormal toxicity, Method B. In specificantjsera. The globulins may be obtained from the
vaccines containing phenol as preservative, the test in mice immune serum by enzyme treatment and fractional precipitation
may be inappropriate. or by other physical or chemical methods.

NOTE - The statements given in this general chapter is Antiviral sera with the exception of Rabies Antiserum are
intended to be read in conjunction with the monographs on usually obtained from the plasma or serum ofhuman patients
the individual vaccine in. this Pharmacop0tf,ia which refer to who have recovered from the viral disease or who have been
preparations for human use; they do not necessarily apply artificially immunised. Rabies Antiserum is obtained from
to vaccines for use in veterinmy practice. animals by injecting gradually increasing doses of a rabies
vaccine, a killed vaccine being used first and when some
Storage. Liquid vaccines must be stored at a temperature
immunity is established living virus being used as an antigen.
between 20 and 80 and should not be allowed to freeze unless
When· sufficient virus-neutralising titre is reached the blood
otherwise specified in the individual monograph. Freeze~dried
is collected and processed.
preparations must be stored at temperatures between 20 and
80 and for long term storage a temperature of -200 protected. Antisera in liquid form are distributed under aseptic conditions
At higher temperatures vaccines deteriorate rapidly. into sterile containers which are then sealed so as to exclude
micro-organisms. A suitable antibacterial substance may be
Labelling. The label states (1) for liquid vaccines, the total
added and is invariably added when the final product is filled
number of ml in the container and, for freeze dried vaccines,
in multiple dose containers. The produCt may be freeze-dried
the number of doses in the container; (2) unless otherwise
by a procedure which reduces the water content of the final
indicated the minimum number ofUnits per dose or perml or,
product to less than 1.0 per cent w/w. Antisera are almost
for viral vaccines, the minimum viral titre; (3) the dose and
colourless or very faintly yellow liquids free from turbidity.
route of administration; (4) the name and proportion of any
Freeze-dried antisera consist ofwhite or pale-yellow powders
antibacterial preservative or other auxiliary substances added
or friable masses which dissolve in water to form colourless or
to the vaccine; (5) the date after which the vaccine is not
pale yellow liquids having the same characteristics as the
intended to be used; (6) the conditions under which it should
corresponding liquid preparations.
be stored; (7) for freeze dried vaccines, the liquid to be used
for reconstitution and its volume; (8) that the vaccine should Tests
be used immediately after reconstitution; (9) unless otherwise
directed, that the vaccine should be shaken well before use; The following tests refer to liquid antisera and to the
(10) any contraindication to the use of the vaccine. reconstituted freeze-dried preparations.

2349

www.webofpharma.com
ADSORBED DIPHTHERIA, TETANUS AND HEPATITIS B (rDNA) VACCINE IP 2010

pH (2.4.24).6.0 to 7.0. inject subcutaneously 5 times the single human dose stated
on the label into each of5 healthy guinea-pigs, each weighing
Total protein. Not more than 17.0 per cent w/v, determined by
between 250 and 350 g, that have not previously been treated
carrying out the determination ofnitrogen, Method C (2.3.30)
and multiplying the result by 6.25. with any material that will interfere with the test. Ifwithin 42
days ofthe injection any ofthe animals shows signs ofor dies
Foreign proteins. When examined by precipitation reactions from diphtheria toxaemia or tetanus, the vaccine does not
with specific antisera, they are·shown to consist exclusively comply with the test. If more than 1 animal dies from non-
of protein of the decla.red animal species. specific causes, repeat the test once; if more than 1 animal
Phenol (2.3.36) (ifpresent). Not more than 0.25 per cent w/v. dies in the second test, the vaccine does not comply with the
test.
Sterility (2.2.11). Comply with the tests for sterility.
The content of bacterial endotoxins in the bulle purified
Abnormal toxicity (2.2.1). Comply with the test for abnormal diphtheria toxoid and tetanus toxoid is determined to monitor
toxicity. In antisera containing phenol as preservative, the the purification procedure and to limit the amount in the final
test in mice may be inappropriate. vaccine. For each .component, the content of bacterial
NOTE - The statements given in this general monograph endotoxin is less than the limit approved for the particular
are intended to be read in cOlyunction with the monographs vaccine and in any case the contents are such that the final
on the individual antiserum in the Pharmacopoeia which vaccine contains less than 100 ill per single human dose.
refer to preparations for human use; they do not necessarily Reference vaccine(s)
apply to antisera for use in veterinary practice.
Provided valid assays can be performed, monocomponent
Storage. Antisera should be stored at a temperature between reference vaccines may be used for the aSsays on the combined
2° and 8° and should not be allowed to freeze unless otherwise vaccine. Ifthis is not possible because of interaction between
stated in the individual monograph. the components of the combined vaccine or because of the
Labelling. The label states the name and quantity of difference in composition between monocomponent reference
antibacterial substance added, if any. vaccine and the test vaccine, a batch of combined vaccine
shown to be effective in clinical trials or a batch representative
thereof is used as a reference vaccine. For the preparation of
a representative batch, strict adherence to the production
Adsorbed Diphtheria, Tetanus and process used for the batch tested in clinical trials is necessary.
Hepatitis B (rDNA) Vaccine The reference vaccine may be stabilised by a method that has
been shown to have no effect on the assay procedure.
Diphtheria, Tetanus and Hepatitis B (rDNA) Vaccine
(Adsorbed) is a combined vaccine composed of: diphtheria Production of the components
formol toxoid; tetanus formol toxoid; hepatitis B surface
The production of the components complies with the
antigen (HBsAg); a mineral adsorbent such as aluminium
requirements of the monographs on Diphtheria Vaccine
hydroxide or hydrated aluminium phosphate.
(Adsorbed), Tetanus Vaccine (Adsorbed) and Hepatitis B
The formol toxoids are prepared from the. toxins produced by Vaccine (rDNA).
the growth of COlynebacterium diphtheriae and Clostridium
tetani, respectively. . FINAL BULK VACCINE

HBsAg is a component protein ofhepatitis B virus; the antigen The final bulk vaccine is prepared by adsorption, separately
is obtained by recombinant DNA technology. or together, of suitable quantities of bulle purified diphtheria
toxoid, tetanus toxoid and HBsAg onto a mineral carrier such
Production as aluminium hydroxide or hydrated aluminium phosphate.
Suitable antimicrobial preservatives may be added.
General provisions
Only a final bulle vaccine that complies with the following
The production method shall have been shown to yield requirements may be used in the preparation ofthe final lot.
consistently 'vaccines comparable with the vaccine ofproven Antimicrobial preservative. Where applicaqle, determine the
clinical efficacy and safety in man; !l.!U~-':lI1_t ()r!lnctil1?-icro~_~!l1 I'~~erv_a~~~~:t!l.slli~!l~l~<::_~~I1?-~<::!ll
The production method is validated to demonstrate that the method. The content is not less than 85.0 per cent and not
product, if tested, would comply with the test for abnormal greater thanll5.0per cent of the intended amount.
toxicity for antisera and vaccines, and with the following test Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk
for specific toxicity ofthe diphtheria and tetanus components: for each sterility medium.

2350

www.webofpharma.com
IP 2010 ADTP (ACELLULAR COMPONENT) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE

FINAL LOT Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
the equivalent of 1 human dose into each rabbit.
Only a final lot that is satisfactory with respect to the test for
osmolality and with respect to each ofthe requirements given Assay
below under Identification, Tests and Assay may be released Diphtheria component
for use.
Carry out one of the prescribed methods for the assay as
Provided the test for antimicrobial preservative and the assays stated under Diphtheria Vaccine (Adsorbed).
for the diphtheria and tetanus components have been carried
The lower confidence limit (P = 0.95) ofthe estimated potency
out with satisfactory results on the final bulle vaccine, they
is not less than 30 ill per single human dose.
may be omitted on the final lot.
Provided the content of free formaldehyde has been Tetanus component
detennined on the bulk purified antigens or on the final bulle Cany out one of the prescribed methods for the assay as
and it has been shown that the content in the final lot will not stated under Tetanus Vaccine (Adsorbed).
exceed 0.2 gil, the test for free fonnaldehyde may be omitted
The lower confidence limit (P = 0.95) ofthe estimated potency
on the final lot.
is not less than 40 ill per single human dose.
If an in vivo assay is used for the hepatitis B component,
provided it has been carried out with satisfactory results on Hepatitis B component
the final bulk vaccine, it may be omitted on the final lot. It complies with the assay of Hepatitis B Vaccine.
Osmolality (2.4.23). The osmolality of the vaccine is within Labelling. The label states (1) the minimum number of
the limits approved for the particular preparation. International Units of diphthetia and tetanus toxoid per single
human dose; (2) the amount ofHBsAg per single human dose;
Identification (3) the type of cells used for production of the HBsAg
component; (4) where applicable, that the vaccine is intended
A. Diphtheria toxoid is identified by a suitable irnmunochemical
for primary vaccination of children and is not necessarily
method (2.2.14). The following method, applicable to certain
suitable for reinforcing doses or for administration to adults;
vaccines, is given as an example. Dissolve in the vaccine under
(5) the name and the amount of the adsorbent; (6) that the
examination sufficient sodium citrate to give a 10 per cent
vaccine must be shaken before use; (7) that the vaccine is not
w/v solution. Maintain at 37° for about 16 hours and centrifuge
to be frozen.
until a clear supernatant is obtained. The clear supernatant
liquid reacts with a suitable diphtheria antitoxin, giving a
precipitate. Adsorbed Diphtheria, Tetanus,
B. Tetanus toxoid is identified by a suitable immunochemical Pertussis (Acellular Component) and
method (2.2.14). The following method, applicable to certain Haemophilus Type b Conjugate
vaccines, is given as an example. The clear supernatant liquid
obtained during identification test A reacts with a suitable Vaccine
tetanus antitoxin, giving a precipitate. Diphtheria, Tetanus, Pertussis (Acellular Component) and
C. The assay or, where applicable, the electrophoretic profile, Haemophilus Type b Conjugate Vaccine (Adsorbed) is a
serves also to identifY the hepatitis B component of the combined vaccine composed of: diphtheria formol toxoid;
vaccine. tetanus formol toxoid; individually purified antigenic
components of Bordetella pertussis; polyribosylribitol
Tests phosphate (PRP) covalently bound to a carrier protein; a
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, mineral absorbent such as aluminium hydroxide or hydrated
if aluminium hydroxide or hydrated aluminium phosphate is aluminium phosphate. The product may be presented with
used as the adsorbent. the haemophilus type b component in a separate container,
the contents of which are mixed with the,ofh~Lcomponents
Free formaldehyde (2.3.20). Maximum 0.2 gil.
immediately before use.
Antimicrobial preservative. Where applicable, detennine the The fOlmol toxoids are prepared from the toxins producedoy
amount of antimicrobial preservative by a suitable chemical the growth of Corynebacterium diphtheriae and Clostridium
method. The content is not less than 85.0 per cent and not tetani respectively.
greater than 115.0 per cent ofthe intended amount.
The vaccine contains either pertussis toxoid or a pertussis-
Sterility (2.2.11). Complies with the test for sterility. toxin-like protein free from toxic properties produced by

2351

www.webofpharma.com
ADTP (ACELLULAR COMPONENT) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE IP 2010

expression ofa genetically modified form ofthe corresponding Reference vaccine


gene. Pertussis toxoid is prepared from pertussis toxin by a Provided valid assays can be performed, monocomponent
method that renders the toxin harmless while maintaining reference vaccines may be used for the assays on the combined
adequate immunogenic properties and avoiding reversion to vaccine. If this is not possible because of interaction between
toxin. The acellular pertussis component may also contain the components ofthe combined vaccine or because of the
filamentous haemagglutinin, pertactin (a 69 lilia outer- difference in composition between mOriocomponent reference
membrane protein) and other defined components' of vaccine and the test vaccine, a batch of combined vaccine
B. pertussis such as fimbrial-2 and fimbrial-3 antigens. The
shown to be effective in clinical trials or a batch representative
latter two antigens may be copurified. The antigenic thereof is used as a reference vaccine. For the preparation of
composition and characteristics are based on evidence of a representative batch, strict adherence to the production
protection and freedom from unexpected reactions in the target
process used for the batch tested in clinical trials is necessary.
group for which the vaccine is intended. The reference vaccine may be stabilised by a method that has
PRP is a linear copolymer composed of repeated units of been shown to have no effect on the assay procedure.
3 -[3- D-ri bofurano sy 1-( 1 ~ 1)-ri b i tol- 5 -phosphate
[(CIOHI9 0 ,l),J, with a defined molecular size and derived from Production of the components
a suitable strain of Haemophilus injluenzae type b. The carrier
protein, when conjugated to PRP, is capable of inducing a The production of the components complies with the tests of
T-cell-dependent B-cell immune response to the the monographs on Diphtheria Vaccine (Adsorbed), Tetanus
polysaccharide. Vaccine (Adsorbed), Pertussis Vaccine (Acellular Component,
Adsorbed) and Haemophilus Type b Conjugate Vaccine.
Production
FINAL BULK VACCINE
General provisions
Different methods of preparation may be used: a fmal bulle
The production method shall have been shown to yield vaccine may be prepared by adsorption, separately or together,
consistently the vaccines comparable with the vaccine of ofsuitable quantities ofbulle purified diphtheria toxoid, tetanus
proven clinical efficacy and safety in man. toxoid, acellular pertussis components and PRP conjugate
If the vaccine is presented with the haemophilus component onto a mineral carrier such as aluminium hydroxide or hydrated
in a separate vial, as part of consistency studies the assays of aluminium phosphate; or 2 final bulles may be prepared and
the diphtheria, tetanus and pertussis components are carried filled separately, one containing the diphtheria, tetanus and
out on a suitable number of batches of vaccine reconstituted pertussis components, the other containing the haemophilus
for use. For subsequent routine control, the assays of these component, which may be freeze-dried. Suitable antimicrobial
components may be carried out without mixing with the preservatives may be added.
haemophilus component. Only a final bulle vaccine that complies with the following
The content ofbacterial endotoxins in bulle purified diphtheria requirements may be used in thepreparation ofthe final lot.
toxoid, tetanus toxoid, pertussis components and PRP Antimicrobial preservative. Where applicable, determine the
conjugate is determined to monitor the purification procedure amount of antimicrobial preservative by a suitable chemical
and to limit the amount in the final vaccine. For each method. The content is not less than 85.0 per cent and not
component, the content of bacterial endotoxins is less than greater than 115.0 per cent ofthe intended amount.
the limit approved for the particular vaccine; ifthe vaccine is
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle
presented with the haemophilus component in a separate
for each sterility medium.
container, the contents ofthe diphtheria, tetanus and pertussis
antigens are in any case such that the final vial for these
FINAL LOT
components contains less than 100 IV per single human dose.
Only a final lot that is satisfactory with respect to the test for
The production method is validated to demonstrate that the osmolality shown below and with respect to each of the
product, if tested, would comply with the test for abnormal requirements given below under Identification, Tests and
toxicity for antisera and vaccines. Assay may be released for use.
During de'vel,opIIlelllt t~sts.f()r, !l~sellS:e. ()f..re:s.i~ll,aLpe.t:J:lls.sis.t(»)(il1?
necessary, it shall be demonstrated by tests in animals that irreversibility ofpertussis toxoid and antimicrobial preservative
the vaccine induces a T-cell dependent B-cell immune response and the assays have been carried out with satisfactory results
toPRP. on the final bulk vaccine, they may be omitted on the final lot.

2352

www.webofpharma.com
IP 2010 ADTP (ACELLULAR COMPONENT) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE

Provided the free fonnaldehyde content has been detennined Ifthe haemophilus component is freeze-dried, some tests may
on the bulle purified antigens or the final bulk and it has been be carried out on the freeze-dried product rather than on the
shown that the content in the final lot will not exceed 0.2 gil, bulle conjugate where the freeze-drying process may affect
the test for free fonnaldehyde may be omitted on the final lot. the component under test.
Osmolality (2.4.23). The osmolality of the vaccine,
Absence ofresidual pertussis toxin and irreversibility
reconstituted where applicable, is within the limits approved
of pertussis toxoid
for the particular preparation.
This test is not necessaJyfor the product obtained by genetic
pH (2.4.24). The pH ofthe vaccine, reconstituted ifnecessary,
modification. Use 3 groups each of not less than 5 histamine-
is within the range approved for the particular product.
sensitive mice. Inject intraperitoneally into each mouse ofthe
Free PRP. Unbound PRP is determined after removal of the first group twice the single human dose ofthe vaccine stored
conjugate, for example by anion-exchange, size-exclusion or at 2° to 8°. Inject intraperitoneally into each mouse of the
hydrophobic chromatography (2.4.16), ultrafiltration or other second group twice the single human dose of the vaccine
validated methods. The amount offree PRP is not greater than incubated at 37° for 4 weeks. Inject diluent into the third group
that approved for the particular product. ofmice. After 5 days, inject into each mouse 2 mg ofhistamine
Identification base intraperitoneally in a volume not exceeding 0.5 ml and
observe for 24 hours. The test is invalid if 1 or more control
If the vaccine is presented with the haemophilus component mice die following histamine challenge. The vaccine complies
in a separate vial: identification tests A, Band C are carried with the test if no animal in the first or second group dies
out using the vial containing the diphtheria, tetanus and following histamine challenge. If 1 mouse dies in either or
pertussis components; identification test D is carTied out on both of the first and second groups, the test may be repeated
the vial containing the haemophilus components. with the same number ofmice or with a greater number and the
A. Diphtheria toxoid is identified by a suitable imrnunochemical results ofvalid tests combined; the vaccine complies with the
method (2.2.14). The following method, applicable to certain test if, in both ofthe groups given the vaccine, not more than
vaccines, is given as an example. Dissolve in the vaccine under 5.0 per cent ofthe total number ofmice die following histamine
examination sufficient sodium citrate to give a 10 per cent challenge.
wlv solution. Maintain at 37° for about 16 hours and centrifuge
The histamine sensitivity ofthe strain ofmice used is verified
until a clear supernatant is obtained. The clear supernatant
at suitable intervals as follows: inject intravenously three-
reacts with a suitable diphtheria antitoxin, giving a precipitate.
fold dilutions of a reference pertussis toxin preparation in
B. Tetanus toxoid is identified by a suitable immunochemical phosphate-buffered saline solution containing 0.2 per cent
method (2.2.14). The following method, applicable to certain wlv of gelatin and challenge with histamine as above; the
vaccines, is given as an example. The clear superna tan t strain is suitable if more than 50 per cent of the animals are
obtained as described in Identification test A reacts with a sensitised by 50 ng of pertussis toxin and none of the control
suitable tetanus antitoxin, giving a precipitate. animals injected with only diluent and challenged similarly
C. The pertussis components are identified by a suitable with histamine shows symptoms of sensitisation.
immunochemical method (2.2.14). The following method,
PRP. Minimum 80.0 per cent ofthe amount ofPRP stated on
applicable to certain vaccines, is given as an example. The
the label. PRP is detennined either by assay ofribose (2.7.1) or
clear supernatant obtained as described in Identification
phosphorus (2.7.1), by an immunochemical method (2.2.14) or
test A reacts with a specific antisera to the pertussis
by anion-exchange liquid chromatography with pulsed-
components of the vaccine.
amperometric detection.
D. The haemophilus component is identified by a suitable
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
imrntinochemical method (2.2.14) for PRP.
if aluminium hydroxide or hydrated aluminium phosphate is
Tests used as the adsorbent.
If the product is presented with the haemophilus component Free formaldehyde (2.3.20). Maximwn 0.2 gil.
in a separate contairter: the tests for absence of residual
Antimicrobial preservative. Where applicable, detennine the
pertussis toxin, irreversibility ofpertussis toxoid, aluminium,
amount of antimicrobial preservative by a suitable chemical
free fonnaldehyde, antimicrobial preservative and sterility are
method. The content is not less than 85.0 per cent and not
carried out on the container with the diphtheria, tetanus and
greater than 115.0 per cent of the intended amount.
pertussis components; the tests for PRP content, water (where
applicable), sterility and pyrogens are carried out on the Water (2.3.43). Maximum 3.0 per cent for the freeze-dried
container with the haemophilus component. haemophilus component.

2353

www.webofpharma.com
ADTP (ACELLULAR COMPONENT) AND HAEMOPHILUS TYPE b CONWGATE VACCINE IP 2010

Sterility (2.2.11). Complies with the test for sterility. The formol toxoids are prepared from the toxins produced by
the growth of Corynebacterium diphtheriae and Clostridium
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
tetani, respectively.
per kg ofthe rabbit's mass a quantity ofthe vaccine equivalent
to: 1 mg ofPRP for a vaccine with diphtheria toxoid or CRM The vaccine contains either pertussis toxoid or a pertussis-
197 diphtheria protein as carrier; 0.1 mg ofPRP for a vaccine toxin-like protein free from toxic properties, produced by
with tetanus toxoid as carrier; 0.025 mg ofPRP for a vaccine expression ofa genetically modified form ofthe corresponding
with OMP as carrier. gene. Pertussis toxoid is prepared from pertussis toxin by a
method that renders the latter harmless while maintaining
Assay
adequate immunogenic propelties and avoiding reversion to
Diphtheria component toxin. The vaccine may also contain filamentous
Carry out one of the prescribed methods for the assay as haemagglutinin, peltactin (a 691cDa outer-membrane protein)
stated under Diphtheria Vaccine (Adsorbed). and other defined components ofB. pertussis such as fimbrial-
2 and fimbrial-3 antigens. The latter 2 antigens may be
The lower confidence limit (P = 0.95) ofthe estimated potency
copurified. The antigenic composition and characteristics are
is not less than the minimum potency stated on the label.
based on evidence ofprotection and freedom from unexpected
Unless otherwise justified and authorised, the minimum reactions in the target group for which the vaccine is intended.
potency stated on the label is 30 IU per single human dose.
Hepatitis B surface antigen is a component protein ofhepatitis
Tetanus component B virus; the antigen is obtained by recombinant DNA
Carry out one of the prescribed methods for the assay as technology.
stated under Tetanus Vaccine (Adsorbed).
Production
The lower confidence limit (P = 0.95) ofthe estimated potency
is not less than 40 IU per single human dose. ._ General provisions
Pertussis component The production method shall have been shown to yield
The vaccine complies with the assay as the stated Adsorbed consistently vaccines comparable with the vaccine of proven
Pertussis Vaccine (Acellular Component). clinical efficacy and safety in man.
Labelling. The label states (1) the minimum number of The content of bacterial endotoxins in the bulk purified
International Units of diphtheria and tetanus toxoid per single diphtheria toxoid, tetanus toxoid and pertussis components
human dose; (2) the names and amounts of the pertussis is determined to monitor the purification procedure and to
components per single human dose; (3) the number of limit the amount in the final vaccine. For each component, the
micrograms ofPRP per single human dose; (4) the type and content ofbacterial endotoxins is less than the limit approved
nominal amount of carrier protein per single human dose; (5) for the particular vaccine.
where applicable, that the vaccine is intended for primary
vaccination of children and is not necessarily suitable for Reference vaccine(s)
reinforcing doses or for administration to adults; (6) the name
and the amount ofthe adsorbent; (7) that the vaccine must be Provided valid assays can be performed, monocomponent
shaken before use; (8) that the vaccine is not to be frozen; reference vaccines may be used for the assays on the combined
vaccine. If this is not possible because of interaction between
(9) where applicable, that the vaccine contains a pertussis
toxin-like protein produced by genetic modification. the components of the combined vaccine or because of the
difference in composition between monocomponent reference
vaccine and the test vaccine, a batch of combined vaccine
Adsorbed Diphtheria, Tetanus, shown to be effective in clinical trials or a batch representative
thereof is used as a reference vaccine. For the preparation of
Pertussis (Acellular Component) and a representative batch, strict adherence to the production
Hepatitis B (rDNA) Vaccine process used for the batch tested in clinical trials is necessary.
The reference vaccine may be stabilised by a method that has
Diphtheria, Tetanus; Pertussis (Acellular Component) and
been shown to have no effect on the assay procedure.
Hepatitis B (rDNA) Vaccine (Adsorbed) is a combined vaccine
composed_of:diphtheriaforrnoLtoxoid; teJanus_formolJoxoid; ProductionoHbecomponents
individuaUy PllriJ'ii::<:ll!l1t.igi::l1icc()lllPQlleI1ts .of 13grq?tejla.
pertussis; hepatitis B surface antigen; a mineral adsorbent The production of the components complies with the
such as aluminium. hydroxide or hydrated aluminium requirements of the monographs on Diphtheria Vaccine
phosphate. (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine

2354

www.webofpharma.com
IP 2010 ADTP (ACELLULAR COMPONENT) AND HEPATITIS B (rDNA) VACCINE

(Acellular Component, Adsorbed) and Hepatitis B Vaccine vaccines, is given as an example. The clear supernatant
(rDNA). obtained as described in identification test A reacts with a
suitable tetanus antitoxin, giving a precipitate.
FINAL BULK VACCINE C. The pertussis components are identified by a suitable
immunochemical method (2.2.14). The following method;
The final bull( vaccine is prepared by adsorption, separately
applicable to certain vaccines, is given as an example. The
or together, of suitable quantities of bulk purified diphtheria
clear supernatant obtained as described in identification test
toxoid, tetanus toxoid, acellular pertussis components and
A reacts with a specific antisera to the pertussis components
hepatitis B surface antigen onto a mineral carrier such as
of the vaccine.
aluminium hydroxide or hydrated aluminium phosphate.
Suitable antimicrobial preservatives may be added. D. The assay or, where applicable, the electrophoretic profile,
serves also to identify the hepatitis B component of the
Only a final bulk vaccine that complies with the following
vaccine.
requirements may be used in the preparation of the final lot.
Antimicrobial preservative. Where applicable, determine the Tests
amount of antimicrobial preservative by a suitable chemical
Absence of residual pertussis toxin and iueversibility
method. The content is not less than 85.0 per cent and not
of pertussis toxoid
greater than 115.0 per cent of the intended amount.
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbull( This test is not necessmyfor the product obtained by genetic
modification. Use 3 groups each ofnot less than 5 histamine-
for each sterility medium.
sensitive mice. Inject intraperitoneally into each mouse ofthe
FINAL LOT first group twice the single human dose of the vaccine stored
at 2° to 8°. Inject intraperitoneally into each mouse of the
Only a final lot that is satisfactory with respect to the test for second group twice the single human dose of the vaccine
osmolality and with respect to each ofthe requirements given incubated at 37° for 4 weeks. Inject diluent into the third group
below under Identification, Tests and Assay may be released ofmice. After 5 days, inject into each mouse 2 mg ofhistamine
for use. base intraperitoneally in a volume not exceeding 0.5 ml and
Provided the tests for absence of residual pertussis toxin, observe for 24 hours. The test is invalid if 1 or more control
irrever:sibility ofpertussis toxoid and antimicrobial preservative mice die following histamine challenge. The vaccine complies
and the assays for the diphtheria, tetanus and pertussis with the test if no animal in the first or second group dies
components have been carried out with satisfactory results following histamine challenge. If 1 mouse dies in either or
on the final bull( vaccine, they may be omitted on the final lot. both of the first and second groups, the test may be repeated
Provided the content of free formaldehyde has been with the same number ofmice or with a greater number and the
determined on the bull( purified antigens or on the final bull< results ofvalid tests combined; the vaccine complies with the
and it has been shown that the content in the final lot will not test if, in both of the groups given the vaccine, not more than
exceed 0.2 gil, the test for free formaldehyde may be omitted 5 per cent ofthe total number ofmice die following histamine
on the final lot. . challenge.
If an in vivo assay is used for the hepatitis B component, The histamine sensitivity ofthe strain ofmice used is verified
provided it has been carried out with satisfactory results on at suitable intervals as follows: inject intravenously threefold
the final bull( vaccine, it may be omitted on the final lot. dilutions of a reference pertussis toxin preparation in
phosphate-buffered saline solution containing 0.2 per cent
Osmolality (2.2.23). The osmolality of the vaccine is within
wlv of gelatin and challenge with histamine as above; the
the limits approved for the particular preparation.
strain is suitable if more than 50.0 per cent of the animals are
Identification sensitised by 50 ng of pertussis toxin and none ofthe control
A. Diphtheria toxoid is identified by a suitable irnmunochemical animals injected with only diluent and challenged similarly
method (2.2.14). The following method, applicable to certain with histamine shows symptoms of sensitisation.
vaccines, is given as an example. Dissolve in the vaccine under Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
examination sufficient sodium citrate to give a 10 per cent if aluminium hydroxide or hydrated aluminium phosphate is
wlv solution. Maintain at 37° for about 16 hours and centrifuge used as the adsorbent.
until a clear supernatant is obtained. The clear supernatant Free formaldehyde (2.3.20). Maximum 0.2 gil.
reacts with a suitable diphtheria antitoxin, giving a precipitate.
Antimicrobial preservative. Where applicable, determine the
B. Tetanus toxoid is identified by a suitable immunochemical amount of antimicrobial preservative by a suitable chemical
method (2.2.14). The following method, applicable to certain method. The content is not less than 85.0 per cent and not

2355

www.webofpharma.com
ADTP (ACELLULAR COMPONENT) AND HEPATITIS B (rDNA) VACCINE IP 2010

greater than 115.0 per cent ofthe intended amount. composed of: diphtheria fonnol toxoid; tetanus fonnol toxoid;
Sterility ( 2.2.11). Complies with the test for sterility. individually purified antigenic components of Bordetella
pertussis; suitable strain of human polioviruses 1,2, and 3
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
grown in suitable cell cultures and inactivated by validated
the equivalent of 1 human dose into each rabbit.
method; polyribosylribitol phosphate (PRP) covalently bound
Assay to a carrier protein; a mineral absorbent such as alumipium
hydroxide or hydrated aluminium phosphate. The product may
Diphtheria component
be presented with the haemophilus type b component in a
Carry out one of the prescribed methods for the assay as separate container, the contents of which are mixed with the
stated under Diphtheria Vaccine (Adsorbed). other components immediately before use.
The lower confidence limit (P = 0.95) ofthe estimated potency The formol toxoids are prepared from the toxins produced by
is not less than the minimum potency stated on the label. the growth of COIynebacterium diphtheriae and Clostridium
Unless otherwise justified and authorised, the minimum tetani respectively.
potency stated on the label is 30 IV per single human dose. The vaccine contains either pertussis toxoid or a pertussis-
toxin-like protein free from toxic properties produced by
Tetanus component
expression ofa genetically modified fonn ofthe corresponding
Carry out one of the prescribed methods for the assay as gene. Pertussis toxoid is prepared from pertussis toxin by a
stated under Tetanus Vaccine (Adsorbed). method that renders the toxin harmless while maintaining
The lower confidence limit (P = 0.95) ofthe estimated potency adequate immunogenic properties and avoiding reversion to
is not less than 40 IV per single human dose. toxin. The acellular pertussis component may also contain
filamentous haemagglutinin, pertactin (a 69 lcDa outer-
Pertussis component membrane protein) and other defined components of B.
The vaccine complies with the assay as stated under Adsorbed pertussis such as fimbrial-2 and fimbrial-3 antigens. The latter
Pertussis Vaccine (Acellular Component). 2 antigens may be co-purified. The antigenic composition and
characteristics are based on evidence of protection and
Hepatitis B component freedom from unexpected reactions in the target group for
The vaccine complies with the assay as stated under Hepatitis which the vaccine is intended.
B Vaccine (rDNA). PRP is a linear copolymer composed of repeated units of
Labelling. The label states (1) the minimum number of 3-13-D-ri bofuranosy 1-( 1-71 )-ri bito 1- 5 -phosphate
International Units ofdiphtheria and tetanus toxoid per single [(CIOHI9 0 IZP\], with a defined molecular size and derived from
human dose; (2) the names and amounts of the pertussis a suitable strain ofHaemophilus injluenzae type b. The carrier
components per single human dose; (3) the amount ofHBsAg protein, when conjugated to PRP, is capable of inducing a
per single human dose; (4) the type ofcells used for production T-cell-dependent B-cell immune response to the
of the hepatitis B component; (5) where applicable, that the polysaccharide.
vaccine is intended for primary vaccination of children and is
not necessarily suitable for reinforcing doses or for Production
administration to adults; (6) the name and the amount of the General provisions
adsorbent; (7) that the vaccine must be shaken before use;
The production method shall have been shown to yield
(8) that the vaccine is not to be frozen (9) where applicable,
consistently vaccines comparable with the vaccine of proven
that the vaccine contains a pertussis toxin-like protein
clinical efficacy and safety in man.
produced by genetic modification.
The content ofbacterial endotoxins in bulk purified diphtheria
Adsorbed Diphtheria, Tetanus, toxoid, tetanus toxoid, pertussis components, purified,
inactivated monovalent poliovirus harvests and bulk PRP
Pertussis (Acellular Component), conjugate is determined to monitor the purification procedure
Inactivated Poliomyelitis Vaccine and and to limit
I
the amount in the final vaccine. For each
Haemophilus Type b Conjugate component, the content of bacterial endotoxins is less than
th-«limitapprov-«d forJhe particular vaccine and, ih any case,
Vaccine the contents aresuch that the finalvact;:inepontains less than
Diphtheria, Tetanus, Pertussis (Acellular Component), 100 IV per single human dose.
Inactivated Poliomyelitis Vaccine and Haemophilus Type b The production method is validated to demonstrate that the
Conjugate Vaccine· (Adsorbed) is a combined vaccine product, iftested, would comply with the following test. Inject

2356

www.webofpharma.com
IP 2010 ADTP (ACELL. COMP.), INACTIVATED POLIOMYELITIS VACCINE AND HAEMOPHILUS TYPE b CONJUGATE VACCINE

subcutaneously 5 times the single human dose stated on the of a trivalent pool of such monovalent harvests. Suitable
label into each of 5 healthy guinea-pigs, each weighing antimicrobial preservatives may be added.
between 250 and 350 g, that have not previously been treated
The final bulle of the haemophilus component is prepared by
with any material that will interfere with the test. If within
dilution ofthe bulk conjugate to the final concentration with a
42 days of the injection any of the animals shows signs of or
suitable diluent. A stabiliser may be added.
dies from diphtheria, toxaemia or tetanus, the vaccine does
not comply with the test. Ifmore than 1 animal dies from non- Only a final bulle vaccine that complies with the following
specific causes, repeat the test once; if more than I animal requirements may be used in the preparation of the final lot.
dies in the second test, the vaccine does not comply with the
Bovine serum albumin. Determine on the poliomyelitis
test.
components by a suitable immunochemical method (2.2.14)
During development studies and wherever revalidation is during preparation of the final bulle vaccine, before addition
necessary, it shall be demonstrated by tests in animals that of the adsorbent, the amount ofbovine serum albumin is such
the vaccine induces a T-cell dependent B-cell immune response that the content in the final vaccine will not be more than 50
toPRP. ng per single human dose.
As part of consistency studies the assays of the diphtheria, Antimicrobial preservative. Where applicable, determine the
tetanus, pertussis and poliomyelitis components are carried amount of antimicrobial preservative by a suitable chemical
out on a suitable number of batches of vaccine reconstituted method. The content is not less than 85.0 per cent and not
for use. For subsequent routine control, the assays of these greater than 115.0 per cent of the intended amount.
components may be carried out without mixing with the
haemophilus component. ' Sterility (2.2.11). Carry out test for sterility using 10 ml ofbullc
for each sterility medium.
Refe,~ence vaccine(s)
Provided valid assays can be performed, monocomponent FINAL LOT
reference vaccines may be used for the assays on the combined The final bulk of the haemophilus component is fi:eeze-dried.
vaccine. Ifthis is not possible because of interaction between Only a final lot that is satisfactory with respect to the test for
the components of the combined vaccine or because of the osmolality shown below and with respect to each of the
difference in composition between monocomponent reference requirements given below under Identification, Tests and
vaccine and the test vaccine, a batch of combined vaccine Assay may be released for use.
shown to be effective in clinical trials or a batch representative
thereof is used as a reference vaccine. For the preparation of Provided that the test for absence of residual pertussis toxin
a representative batch, strict adherence to the production and irreversibility ofpertussis toxoid, the test for antimicrobial
process used for the batch tested in clinical trials is necessary. preservative and the assay have been carried out with
The reference vaccine may be stabilised by a method that has satisfactOly results on the final bulle vaccine, they may be
been shown to have no effect on the assay procedure. omitted on the final lot.
Provided that the free formaldehyde content has been
Production ofthe components determined on the bulk purified antigens and the purified
The production of the components complies with the monovalent harvests or the trivalent pool of polioviruses or
requirements of the monographs on Diphtheria Vaccine the final bulle and it has been shown that the content in the
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine final lot will not exceed 0.2 gil, the test for free formaldehyde
(Acellular Component, Adsorbed), Poliomyelitis Vaccine may be omitted on the final lot.
(Inactivated) and Haemophilus Type b Conjugate Vaccine. Provided that the in vivo assay for the poliomyelitis component
has been carried out with satisfactory results on the final bulle
FINALBULKVACClNE vaccine, it may be omitted on the final lot.
The final bulk of the diphtheria, tetanus, pertussis and Osmolality (2.4.23). The osmolality of the vaccine,
poliomyelitis components is prepared by adsorption, reconstituted where applicable, is within the limits approved
separately or together, of suitable quantities of bulk purified for the particular preparation.
diphtheria toxoid, bulle purified tetanus toxoid and bulle purified
acellular pertussis components onto a mineral carrier such as Free PRP
aluminium hydroxide or hydrated aluminium phosphate and
admixture of suitable quantities of purified, monovalent Unbound PRP is determined on the haemophilus component
harvests ofhuman polioviruses 1,2 and 3 or a suitable quantity after removal ofthe conjugate, for example by anion-exchange,

2357

www.webofpharma.com
ADTP (ACELL. COMP.), INACTIVATED POLIOMYELITIS VACCINE AND HAEMOPHILUS TYPE b CONJUGATE VACCINE IP 2010

size-exclusion or hydrophobic chromatography (2.4.16), sensitive mice. Inject intraperitoneally into each mouse ofthe
ultrafiltration or other validated methods. The amount offree .first group twice the single human dose ofthe vaccine stored
PRP is not greater than that approved for the particular product. at 2° to 8°. Inject intraperitoneally into each mouse of the
second group twice the single human dose of the vaccine
Identification incubated at 37° for 4 weeks. Inject diluent into the third group
ofmice. After 5 days, inject into each mouse 2 mg Ofhistamine
Identification tests A, B, C and D are carried out using the vial
base intraperitoneally in a volume not exceeding 0.5 ml and
containing the diphtheria, tetanus, pertussis and poliomyelitis
observe for 24 hours. The test is invalid if 1 or more control
components; identification test E is carried out on the vial
mice die following histamine challenge. The vaccine complies
containing the haemophilus component.
with the test if no animal in the first or second group dies
A. Diphtheria toxoid is identified by a suitable immunochemical following histamine challenge. If 1 mouse dies in either or
method (2.2.14). The following method, applicable to certain both of the first and second groups, the test may be repeated
vaccines, is given as an example. Dissolve in the vaccine under with the same number ofmice or with a greater number and the
examination sufficient sodiuin citrate to give a 10 per cent results ofvalid tests combined; the vaccine complies with the
w/v solution. Maintain at 37° for about 16 hours and centrifuge test if, in both ofthe groups given the vaccine, not more than
until a clear supernatant is obtained. The clear supernatant 5 per cent ofthe total number ofmice die following histamine
reacts with a suitable diphtheria antitoxin, giving a precipitate. challenge.
B. Tetanus toxoid is identified by a suitable immunochemical
The histamine sensitivity ofthe strain ofmice used is verified
method (2.2.14). The following method, applicable to certain
at suitable intervals as follows: inject intravenously threefold
vaccines, is given as an example. The clear supernatant
dilutions of a reference pertussis toxin preparation in
obtained during identification test A reacts with a suitable
phosphate-buffered saline solution containing 0.2 per cent
tetanus antitoxin, giving a precipitate.
w/v of gelatin and challenge with histamine as above; the
C. The pertussis components are identified by suitable strain is suitable if more than 50.0 per cent of the animals are
immunochernical methods (2.2.14). The following method, sensitised by 50 ng of pertussis toxin and none ofthe control
applicable to certain vaccines, is given as an example. The animals injected with only diluent and challenged similarly
clear supernatant obtained during identification test A reacts with histamine show symptoms of sensitisation.
with specific antisera to the pertussis components of the
vaccine. PRP. Minimum 80.0 per cent ofthe amount ofPRP stated on
D. The vaccine is shown to contain human polioviruses 1,2 the label. PRP is detennined either by assay of ribose ( 2.7.1)
and 3 by a suitable immunochemical method (2.2.14), such as or phosphorus (2.7.1), by an immunochemical method (2.2.14)
detennination ofD-antigen by enzyme-linkedimmunosorbent or by anion-exchange liquid chromatography with pulsed-
assay (ELISA). amperometric detection.

E. The haemophilus component is identified by a suitable Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
immunochemical method (2.2.14) for PRP. if aluminium hydroxide or hydrated aluminium phosphate is
used as the adsorbent.
Tests
The testsfor absence ofresidualpertussis toxin, irreversibility Free formaldehyde (2.3.20). Maximum 0.2 gil.
of pertussis toxoid, aluminium, free formaldehyde, Antimicrobial preservative. Where applicable, detennine the
antimicrobial preservative and sterility are carried out on amount of antimicrobial preservative by a suitable chemical
the container with the diphtheria, tetanus, pertussis and method. The content is not less than 85.0 per cent and not
poliomyelitis components; the tests for PRP content, wate]~ greater than 115.0 per cent of the intended amount.
sterility and pyrogens are carried out on the container with
the haemophilus component. Water (2.3.43). Maximum 3.0 per cent for the haemophilus
Some tests for the haemophilus component may be carried component.
out on the fi-eeze-dried product rather than on the bulk Sterility (2.2.11 ). Complies with the test for sterility.
conjugate where the fi-eeze-drying process may affect the
component under test. J,lyrogens (2.2.8). Complies with the test for pyrogens. Inject
per leg. Qftll~l'fl1:>1:>it 'sl11assa qllfllltitYQftlle vflcci!l.~t::q1Ji"fll~ll.t
Absence of resldual pertuSSiS toxin and irreverSibility to: 1 mg ofPRP for a vaccine with diphtheriatoxoid orCRM:
of pertussis toxoid
197 diphtheria protein as canier; 0.1 mg ofPRP for a vaccine
This test is not necessaryfor the product obtained by genetic with tetanus toxoid as carrier; 0.025 mg ofPRP for a vaccine
modification. Use 3 groups each ofnot fewer than 5 histamine- with OMP as a carrier.

2358

www.webofpharma.com
IP 2010 ADTP (ACELLULAR COMPONENT) AND INACTIVATED POLIOMYELITIS VACCINE

Assay Adsorbed Diphtheria, Tetanus,


Diphtheria component Pertussis (Acellular Component) and
Carry out one of the prescribed methods for the assay as Inactivated Poliomyelitis Vaccine
stated under Diphtheria Vaccine (Adsorbed).
Diphtheria, Tetanus, Pertussis (Acellular Component) and
Unless otherwise justified and authorised, the lower
Poliomyelitis (Inactivated) Vaccine is a combined vaccine
confidence limit (P = 0.95) ofthe estimated potency is not less
containing: diphtheria formol toxoid; tetanus formol toxoid;
than 30 IU per single human dose.
individually purified antigenic components of Bordetella
Tetanus component pertussis; suitable strains of human polioviruses 1, 2 and 3
Carry out one of the prescribed methods for the assay as grown in suitable cell cultures and inactivated by a validated
stated under Tetanus Vaccine (Adsorbed). method; a mineral adsorbent such as aluminium hydroxide or
hydrated aluminium phosphate.
The lower confidence limit (P = 0.95) ofthe estimated potency
is not less than 40 IU per single human dose. The formol toxoids are prepared from the toxins produced by
the growth of COIynebacterium diphtheriae and Clostridium
Pertussis component tetani respectively.
It complies with the assay as stated under Adsorbed Pertussis The vaccine contains either pertussis toxoid or a pertussis-
Vaccine (Acellular Component). toxin-like protein free from toxic properties produced by
expression ofa genetically modified form ofthe corresponding
Poliomyelitis component
gene. Pertussis toxoid is prepared from pertussis toxin by a
D-antigen content method that renders the toxin harmless while maintaining
adequate immunogenic properties and avoiding reversion to
As a measure of consistency of production, determine the toxin. The vaccine may also contain filamentous
D-antigen content for human polioviruses 1, 2 and 3 by a haemagglutinin, 'pertactin (a 69lcDa outer-membrane protein)
suitable immunochemical method (2.2.14) using a reference and other defined components ofB. pertussis such as fimbrial-
preparation calibrated in units of D-antigen. For each type, 2 and fimbrial-3 antigens. The latter 2 antigens may be
the content, expressed with reference to the amount of copurified. The antigenic composition and characteristics are
D-antigen stated on the label, is within the limits approved for based on evidence ofprotection and freedom from unexpected
the particular product. Poliomyelitis vaccine (inactivated) reactions in the target group for which the vaccine is intended.
reference preparation is calibrated in Units and intended for
use in the assay ofD-antigen. The Unit and the International Production
Unit are equivalent.
General provisions
In vivo test The production method shall have been shown to yield
The vaccine complies with the in vivo assay as stated under consistently vaccines comparable with the vaccine ofproven
Inactivated Poliomyelitis Vaccine. clinical efficacy and safety in man.

Labelling. The label states (1) the minimum number of The production method is validated to demonstrate that the
International Units of diphtheria and tetanus toxoid per single product, if tested, would comply with the test for abnormal
human dose (2) the names and amounts of the pertussis toxicity for antisera and vaccines.
components per single human dose; (3) the nominal amount The content ofbacterial endotoxins in bulk purified diphtheria
of poliovirus of each type (1, 2 and 3), expressed in units of toxoid, tetanus toxoid, pertussis components and purified,
D-antigen per single human dose; (4) the type of cells used inactivated monovalent poliovirus harvests is determined to
for production ofthe poliomyelitis component; (5) the number monitor the purification procedure and to limit the amount in
ofmicrograms ofPRP per single human dose; (6) the type and the final vaccine. For each component, the content ofbacterial
nominal amount of carrier protein per single human dose; endotoxins is less than the limit approved for the particular
(7) where applicable, that the vaccine is intended for primary vaccine and, in any case, the contents are such that the final
vaccination of children and is not necessarily suitable for vaccine contains less than 100 IU per single human dose,
reinforcing doses or for administration to adults; (8) the name
Reference vaccine(s)
and the amount of the adsorbent; (9) that the vaccine must be
shaken before use; (10) that the vaccine is not to be frozen; Provided valid assays can be performed, monocomponent
(11) where applicable, that the vaccine contains a pertussis reference vaccines may be used for the assays on the combined
toxin-like protein produced by genetic modification. vaccine. Ifthis is not possible because of interaction between

2359

www.webofpharma.com
ADTP (ACELLULAR COMPONENT) AND INACTIVATED POLIOMYELITIS VACCINE IP 2010

the components of the combined vaccine ,or because of the components have been carried out with satisfactory results
difference in composition between monocomponent reference on the final bulle vaccine, they may be omitted on the final lot.
vaccine and the test vacCine, a batch of combined vaccine
Provided the free formaldehyde content has been determined
shown to be effective in clinical trials or a batch representative
on the bulk purified antigens or on the final bulle and it has
thereof is used as a reference vaccine. For the preparation of
been shown that the content in the final lot will not exceed
a representative batch, strict adherence to the production
0.2 gil, the test for free formaldehyde may be omitted on the
process used for the batch tested in cliniCal trials is necessary.
final lot.
The reference vaccine may be stabilised by a method that has
been shown to have no effect on the assay procedure. Provided that the determination ofD-antigen content has been
carried out with satisfactory results during preparation ofthe
Production ofthe components final bulk before addition ofthe adsorbent, it may be omitted
on the final lot.
The production of the components complies with the
requirements of the monographs on Diphtheria Vaccine Provided that the in vivo assay for the poliomyelitis component
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine has been carried out with satisfactory results on the final bulk
(Acellular Component, Adsorbed) and Poliomyelitis Vaccine vaccine, it may be omitted on the final lot.
(Inactivated). Osmolality (2.4.23). The osmolality of the vaccine is within
the limits approved for the particular preparation.
FINAL BULK VACCINE
The final bulk vaccine is prepared by adsorption onto a mineral Identification
carrier such as aluminium hydroxide or hydrated aluminium
A. Diphtheria toxoid is identified by a suitable immunochemical
phosphate, separately or together, of suitable quantities of
method (2.2.14). The following method, applicable to certain
bulk purified diphtheria toxoid, tetanus toxoid, acellular
vaccines, is given as an example. Dissolve in the vaccine under
pertussis components and admixture of suitable quantities of
examination sufficient sodium citrate to give a 10 per cent
purified monovalent harvests of human polioviruses 1,2 and
wlv solution. Maintain at 37° for about 16 h and centrifuge
3 or a suitable quantity of a trivalent pool of such purified
until a clear supernatant is obtained. The clear supernatant
monovalent harvests. Suitable antimicrobial preservatives may
reacts with a suitable diphtheria antitoxin, giving a precipitate.
be added.
B. Tetanus toxoid is identified by a suitable immunochemical
Only a final bulle vaccine that complies with the following
method (2.2.14). The following method, applicable to certain
requirements may be used in the preparation of the final lot.
vaccines, is given as an example. The clear supernatant
Bovine serum albumin. Determine on the poliomyelitis obtained as described in Identification test A reacts with a
components by a suitable immunochemical method (2.2.14) suitable tetanus antitoxin, giving a precipitate.
after virus harvest and before addition ofthe adsorbent in the
C. The pertussis components are identified by a suitable
preparation of the final bulle vaccine, the amount of bovine
immunochemical method (2.2.14). The following method,
serum albumin is such that the content in the final vaccine will
applicable to certain vaccines, is given as an example. The
be not more than 50 ng per single human dose.
clear supernatant obtained as described in Identification test
Antimicrobial preservative, Where applicable, determine the A reacts with a specific antisera to the pertussis components
amount of antimicrobial preservative by a suitable chemical of the vaccine.
method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent ofthe intended amount. D. The vaccine is shown to contain human polioviruses 1,2
and 3 by a suitable immunochemical method (2.2.14) such as
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle the determination of D-antigen by enzyme-linked immuno-
for each sterility medium. sorbent assay (ELISA).

FINAL LOT Tests


Only a final lot that is satisfactory with respect to the test for
osmolality and with respect to each ofthe requirements given Absence of residual pertussis toxin and irreversibility
below under Identification, Tests and Assay may be released of pertussis toxoid
This test is not necessaryfor theproc/uct obtained by genetic
Provided the tests for absence of residual pertussis toxin, modification. Use 3 groups each of not less than 5 histamine-
irreversibility ofpertussis toxoid and antimicrobial preservative sensitive mice. Inject intraperitoneally into each mouse ofthe
and the assays for the diphtheria, tetanus and pertussis first group twiCe the single human dose ofthe vaccine stored

2360

www.webofpharma.com
IP 2010 ADSOR)3ED DIPHTHERIA, TETANUS, PERTUSSIS AND POLIOMYELITIS (INACTIVATED) VACCINE

at 2° to 8°. Inject intraperitoneally into each mouse of the Pertussis component


second group twice the single human dose of the vaccine
The vaccine complies with the assay as stated under Adsorbed
incubated at 37° for 4 weeks. Inject diluent into the third group
Pertussis Vaccine (Acellular Component).
ofmice. After 5 days, inject into each mouse 2 mg ofhistamine
base intraperitoneally in a volume not exceeding 0.5 ml and Poliomyelitis component
observe for 24 hours. The test is invalid if 1 or more control
D-antigen content
mice die following histamine challenge. The vaccine complies
with the test if no animal in the first or second group dies As a measure of consistency of production, determine the D-
following histamine challenge. If 1 mouse dies in either or antigen content for human polioviruses 1,2 and 3 by a suitable
both of the first and second groups, the test may be repeated immunochemical method (2.2.14) following desorption using
with the same number ofmice or with a greater number and the a reference preparation calibrated in units of D-antigen. For
results ofvalid tests combined; the vaccine complies with the each type, the content, expressed with reference to the amount
test if, in both of the groups given the vaccine, not more than ofD-antigen stated on the label; is within the limits approved
5.0 per cent ofthe total number ofmice die following histamine for the particular product. Poliomyelitis vaccine (inactivated)
challenge. reference preparation is calibrated in Units and intended for
use in the assay ofD-antigen. The Unit and the International
The histamine sensitivity ofthe strain ofmice used is verified Unit are equivalent.
at suitable intervals as follows: inject intravenously three-
fold dilutions of a reference pertussis toxin preparation in In vivo test
phosphate-buffered saline solution containing 0.2 per cent The vaccine complies with the in vivo assay as stated under
w/v of gelatin and challenge with histamine as above; the Inactivated Poliomyelitis Vaccine.
strain is suitable ifmore than 50.0 per cent of the animals are
sensitised by 50 ng ofpertussis toxin and none ofthe control Labelling. The label complies with the requirements stated
animals injected with only diluent and challenged similarly under Vaccine and also states (1) the minimum number of
with histamine shows symptoms of sensitisation. International Units ofdiphtheria and tetanus toxoid per single
human dose; (2) the names and amounts of the pertussis
Aluminium (2.3.9). Maximum 1.25 mg per single human dose components per single human dose; (3) the nominal amount
if aluminium hydroxide or hydrated aluminium phosphate is of poliovirus of each type (1, 2 and 3), expressed in units of
used as the adsorbent. D-antigen per single human dose; (4) the type of cells used
Free formaldehyde (2.3.20). Maximum 0.2 gil. for production ofthe poliomyelitis component; (5) the number
ofmicrograms ofPRP per single human dose; (6) the type and
Antimicrobial preservative. Where applicable, determine the
nominal amount of carrier protein per single human dose;
amount of antimicrobial preservative by a suitable chemical
(7) where applicable, that the vaccine is intended for primary
method. The content is not less than 85.0 per cent and not
vaccination of children and is not necessarily suitable for
greater than 115.0 per cent ofthe intended amount.
reinforcing doses or for administration to adults; (8) the name
Sterility (2.2.11). Complies with the test for sterility. and the amount ofthe adsorbent; (9) that the vaccine must be
shaken before use; (10) that the vaccine is not to be frozen;
Assay (11) where applicable, that the vaccine contains a pertussis
toxin-like protein produced by genetic modification.
Diphtheria component
Carry out one of the prescribed methods for the assay as
stated under Diphtheria Vaccine (Adsorbed).
The lower confidence limit (P = 0.95) ofthe estimated potency
Adsorbed Diphtheria, Tetanus,
is not less than the minimum potency stated on the label. Pertussis and Poliomyelitis
Unless otherwise justified and authorised, the minimum (Inactivated) Vaccine
potency stated on the label is 30 ill per single human dose.
Diphtheria, Tetanus, Pertussis and Poliomyelitis (Inactivated)
Tetanus component Vaccine (Adsorbed) is a combined vaccine containing:
diphtheria formol toxoid; tetanus formol toxoid; an inactivated
Carry out one of the prescribed methods for the assay as
suspension of Bordetella pertussis; suitable strains of human
stated under Tetanus Vaccine (Adsorbed).
polioviruses 1, 2 and 3 grown in suitable cell cultures and
The lower confidence limit (P = 0.95) ofthe estimated potency a
inactivated by a validated method; mineral adsorbent such
is not less than 40 ill per single human dose. as aluminium hydroxide or hydrated aluminium phosphate.

2361

www.webofpharma.com
ADSORBED DIPHTHERIA, TETANUS, PERTUSSIS AND POLIOMYELITIS (INACTIVATED) VACCINE IP 2010

The formol toxoids are prepared from the toxins produced by suspension of B. pertussis and purified monovalent harvests
the growth of COlynebacterium diphtheriae and qostridium of human polioviruses 1,2 and 3 or a suitable quantity of a
tetani respectively. ' trivalent pool of such purified monovalent harvests. Suitable
antimicrobial preservatives may be added.
Production
Only a final bulle vaccine that complies with the following
General provisions requirements may be used in the preparation of the final lot.

The production method shall have been shown to yield Specific toxicity
consistently the vaccines comparable with the vaccine of
Use not less than 5 healthy mice each weighing between
proven clinical efficacy and safety in man.
14 and 16 g for the vaccine group and for the saline control.
The production method is validated to demonstrate that the Use mice ofthe same sex or distribute males and females equally
product, if tested, would comply with the test for abnormal between the groups. Allow the animals access to food and
toxicity for antisera and vaccines, and with the following test water for at least 2 hours before injection and durIng the test.
for specific toxicity ofthe diphtheria and tetanus components Inject each mouse ofthe vaccine group intraperitoneally with
: inject subcutaneously 5 times the single human dose stated 0.5 ml, containing a quantity of the vaccine equivalent to not
on the label into each of5 healthy guinea-pigs, each weighing less than halfthe single human dose. Inject each mouse ofthe
between 250 and 350 g, that have not previously been treated control group with 0.5 ml of a 0.9 per cent sterile solution of
with any material that will interfere with the test. If within 42 sodium chloride, preferably containing the same amount of
days ofthe injection any ofthe animals shows signs ofor dies antimicrobial preservative as that injected with the vaccine.
from diphtheria toxaemia or tetanus, the vaccine does not Weigh the groups of mice immediately before the injection
comply with the test. If more than 1 animal dies from non- and 72 hours and 7 days after the injection. The vaccine
specific causes, repeat the test once; if more than 1 animal complies with the test if: (a) at the end of 72 hours the total
dies in the second test, the vaccine does not comply with the mass of the group of vaccinated mice is not less than that
test. preceding the injection; (b) at the end of 7 days the average
increase in mass per vaccinated mouse' is not less than
Reference vaccine(s) 60 per cent of that per control mouse; and (c) not more than
5 per cent of the vaccinated mice die during the test. The test
Provided valid assays can be performed, monocomponent
may be repeated and the results of the tests combined.
reference vaccines may be used for the assays on the combined
vaccine. If this is not possible because of interaction between Bovine serum albumin. Determine on the poliomyelitis
the components of the combined vaccine or because of the components by a suitable immunochemical method (2.2.14)
difference in composition between monocomponent reference during preparation of the final bulle vaccine; before addition
vaccine and the test vaccine, a batch of combined vaccine ofthe adsorbent, the amount ofbovine serum albumin is such
shown to be effective in clinical trials or a batch representative that the content in the final vaccine will be not more than 50
thereof is used as a reference vaccine. For the preparation of ng per single human dose.
a representative batch, strict adherence to the production Antimicrobial preservative. Where applicable, determine the
process used for the batch tested in clinical trials is necessary. amount of antimicrobial preservative by a suitable chemical
The reference vaccine may be stabilised by a method that has method. The content is not less than 85.0 per cent and not
been shown to have no effect on the assay procedure. greater than 115.0 per cent ofthe intended amount.
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle
Production ofthe components
for each sterility medium.
The production of the components complies with the
requirements of the monographs on Diphtheria Vaccine FINAL LOT
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine Only a final lot that is satisfactory with respect to the test for
(Adsorbed) and Poliomyelitis Vaccine (Inactivated). osmolality and with respect to each ofthe requirements given
below under Identification, Tests and Assay may be released
FINAL BULK VACCINE for use.
ThefinaLbulkvaccinejsprepared.by adsorption onto. amineral Provided thatthe tests for specifictoxicity and antimicrobial
Qarril:lr. such as ah,uninium hydroxide or hyclnl.ted aluminium preservative, and the assays focthediphtheria,Jeta.l1usa.l1cl
phosphate, separately or together, of suitable quantities of pertussis components have been carried out with satisfactory
bulle purified diphtheria toxoid and bulle purified tetanus toxoid results on the final bulle vaccine, they may be omitted on the
and admixture of suitable quantities of an inactivated final lot.

2362

www.webofpharma.com
IP 2010 ADSORBED DIPHTHERIA, TETANUS, PERTUSSIS AND POLIOMYELITIS (INACTIVATED) VACCINE

Provided that the free formaldehyde content has been Assay


determined on the bulk purified antigens, the inactivated
B. pertussis suspension and the purified monov~lent harvests Diphtheria component
or the trivalent pool ofpolioviruses or on the final bulle and it Carry out one of the prescribed methods for the assay as
has been shown that the content in the final lot will not exceed stated under Diphtheria Vaccine (Adsorbed).
0.2 gil, the test for free formaldehyde may be omitted on the The lower confidence limit (P = 0.95) ofthe estimated potency
final lot. is not less than 30 IU per single human dose.
Provided that the in vivo assay for the poliomyelitis component Tetanus component
has been carried out with satisfactory ~esults on the final bulle
vaccine, it may be omitted on the final lot. Carry out one of the prescribed methods for the assay as
stated under Tetanus Vaccine (Adsorbed).
Osmolality (2.4.23). The osmolality of the vaccine is within
If the test is carried out in guinea pigs, the lower confidence
the limits approved for the particular preparation.
limit (P = 0.95) ofthe estimated potency is not less than 40 IU
per single human dose; if the test is carried out in mice, the
Identification lower confidence limit (P = 0.95) ofthe estimated potency is
A. Diphtheria toxoid is identified by a suitable immunochemical not less than 60 IU per single human dose.
method (2.2.14). The following method, applicable to certain
vaccines, is given as an example. Dissolve in the vaccine under Pertussis component
examination sufficient sodium citrate to give aiD per cent Carry out the assay as stated under Pertussis Vaccine.
wIv solution. Maintain at 37° for about 16 hours and centrifuge The estimated potency is not less than 4 IU per single human
until a clear supernatant is obtained. The clear supernatant dose and the lower confidence limit (P = 0.95) ofthe estimated
reacts with a suitable diphtheria antitoxin, giving a precipitate. potency is not less than 2 IU per single human dose.
B. Tetanus toxoid is identified by a suitable immunochemical Poliomyelitis component
method (2.2.14). The following method, applicable to certain
vaccines, is given as an example. The clear supernatant D-antigen content
obtained during identification test A reacts with a suitable
tetanus antitoxin, giving a precipitate. .As a measure of consistency of production, determine the D-
antigen content for human polioviruses 1,2 and 3 by a suitable
C. The centrifugation residue obtained in identification A may immunochemical method (2.2.14) using a reference preparation
be used. Other suitable methods for separating the bacteria calibrated in Units ofD-antigen. For each type, the content,
from the adsorbent may also be used. Identify pertussis expressed with reference to the amount of D-antigen stated
vaccine by agglutination ofthe bacteria from the resuspended on the label, is within the limits approved for the particular
precipitate by antisera specific to B. pertussis or by the assay product. Poliomyelitis vaccine (inactivated) refei-ence
of the pertussis component prescribed under Assay. preparation is calibrated in Units and is intended for use in
the assay ofD-antigen. The Unit and the IU are equivalent.
D. The vaccine is shown to contain humanpolioviruses 1,2
and 3 by a suitable immunochemical method (2.2.14) such as In vivo test
the determination ofD-antigen by enzyme-linked immuno-
sorbent assay (ELISA). The vaccine complies with the in vivo assay as stated under
Poliomyelitis Vaccine (Inactivated).
Tests Labelling. The label states (1) the minimum number of
International Units ofdiphtheria and tetanus toxoid per single
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
human dose; (2) the minimum number ofInternational Units
if aluminium hydroxide or hydrated aluminium phosphate is
of pertussis vaccine per single human dose; (3) the nominal
used as the adsorbent.
amount of poliovirus of each type (1, 2 and 3), expressed in
Free formaldehyde (2.3.20). Maximum 0.2 gil. units ofD-antigen per single human dose; (4) the type ofcells
used for production ofthe poliomyelitis component; (5) where
Antimicrohial preservative. Where applicable, determine the
applicable, that the vaccine is intended for primary vaccination
amount of antimicrobial preservative by a suitable chemical
'ofchildren and is not necessarily suitable for reinforcing doses
method. The content is not less than 85.0 per cent and not
or for administration to adults; (6) the name and the amount of
greater than 115.0 per cent of the intended amount.
the adsorbent; (7) that the vaccine must be shaken before
Sterility (2.2.11). Complies with the test for sterility. use; (8) that the vaccine is not to be frozen.

2363

www.webofpharma.com
ADTP, POLIOMYELITIS (INACTIVATED) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE IF 2010

Adsorbed Diphtheria, Tetanus, As part of consistency studies the assays of the diphtheria,
tetanus, pertussis and poliomyelitis components are carried
Pertussis, Poliomyelitis (Inactivated) out on a suitable number of batches of vaccine reconstituted
and Haemophilus Type b Conjugate for use. For subsequent routine control, the assays of these
components may be carried out without mixing with the
Vaccine haemophilus component.
Diphtheria, Tetanus, Pertussis, Poliomyelitis (Inactivated) and
Reference vaccine(s)
Haemophilus Type b Conjugate Vaccine (Adsorbed) is a
combined vaccine composed of: diphtheria formol toxoid; Provided valid assays can be performed, monocomponent
tetanus formol toxoid; an inactivated suspension ofBordetella reference vaccines may be used for the assays on the combined
pertussis; suitable strains of human polioviruses 1, 2 and 3 vaccine. If this is not possible because of interaction between
grown in suitable cell cultures and inactivated by a suitable the components of the combined vaccine or because of the
method; polyribosylribitol phosphate (PRP) covalently bound difference in composition between monocomponent reference
to a carrier protein; a mineral adsorbent such as aluminium vaccine and the test vaccine, a batch of combined vaccine
hydroxide or hydrated aluminium phosphate.· The product is shown to be effective in clinical trials or a batch representative
presented with the haemophilus component in a separate thereof is used as a reference vaccine. For the preparation of
container, the contents of which are mixed with the other a representative batch, strict adherence to the production
components immediately before use. process used for the batch tested in clinical trials is necessary.
The reference vaccine may be stabilised by a method that has
The formo1 toxoids are prepared from the toxins produced by been shown to have no effect on the assay procedure.
the growth of Corynebacterium diphtheriae and Clostridium
tetani respectively. Production of components
PRP is a linear copolymer composed of repeated units of The production of the components complies with the
3 - ~-D-ri bofuranosy 1- (1 ~ 1)-ri bi to 1- 5 -phosphate requirements of the monographs on Diphtheria Vaccine
[(CIOH1Pll)n]' with a defined molecular size and derived from (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
a suitable strain of Haemophilus injluenzae type b. The carrier (Adsorbed), Poliomyelitis Vaccine (Inactivated) and
protein, when conjugated to PRP, is capable of inducing a T- Haemophilus Type b Conjugate Vaccine.
cell-dependent B-ce1l immune response to the polysaccharide.
FINAL BULK VACCINE
Production
The final bulk of the diphtheria, tetanus, pertussis and
General provisions poliomyelitis components is prepared by adsorption,
The production method shall have been shown to yield separately or together, of suitable quantities of bulk purified
consistently vaccines comparable with the vaccine of proven diphtheria toxoid, and bulk purified tetanus toxoid onto a
clinical efficacy and safety in man. mineral carrier such as aluminium hydroxide or hydrated
aluminium phosphate and admixture of suitable quantities of
The production method is validated to demonstrate that the
an inactivated suspension of B. pertussis and of purified,
product, if tested, would comply with the test for abnormal
monovalent harvests of human polioviruses 1, 2 and 3 or a
toxicity for antisera and vaccines, and with the following test
suitable quantity of a trivalent pool of such monovalent
for specific toxicity ofthe diphtheria and tetanus components:
harvests. Suitable antimicrobial preservatives may be added.
inject subcutaneously 5 times the single human dose stated
on the label into each of 5 healthy guinea-pigs, each weighing The final bulk ofthe haemophilus component is prepared by
between 250 and 350 g, that have not previously been treated dilution ofthe bulle conjugate to the final concentration with a
with any material that will interfere with the test. Ifwithin 42 suitable diluent. A stabiliser may be added.
days ofthe injection any ofthe animals shows signs ofor dies Only final bulk that complies with the following requirements
from diphtheria toxaemia or tetanus, the vaccine does not may be used in the preparation of the final lot.
comply with the test. If more than 1 animal dies from non-
specific causes, repeat the test once; if more than 1 animal Specific toxicity
dies in the second test, the vaccine does not comply with the Use not less than 5 healthy mice each weighing between 14
test. andJ 6-i~, for the-.Yaccine grQ!JP andJ.QLthe salin(;t~QIlJ:rol.JJse
During development studies and wherever revalidation is Illice()f t~~saIlle s~x ()~.<ii.str:i1:>llte I118cles al1clfeIll~les .eqll~llY
necessary, it shall be demonstrated by tests in animals that between the groups. Allow the animals access to food and
the vaccine induces a T-cell dependent B-cell immune response water for at least 2 hours before injection and during the test.
toPRP. Inject each mouse ofthe vaccine group intraperitoneally with

2364

www.webofpharma.com
IP 2010 ADTP, POLIOMYELITIS (INACTIVATED) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE

0.5 ml, containing a quantity of the vaccine equivalent to not FreePRP


less than halfthe single human dose. Inject each mouse ofthe
Unbound PRP is determined on the haemophilus component
control group with 0.5 ml of a 0.9 per cent sterile solution of
after removal ofthe conjugate, for example by anion-exchange,
sodium chloride, preferably containing the same amount of
size-exclusion or hydrophobic chromatography (2.4.16),
antimicrobial preservative as that injected with the vaccine.
ultrafiltration or other validated methods. The amount offree
Weigh the groups of mice immediately before the injection
PRP is not greater than that approved for the particular product.
and 72 hours and 7 days after the injection. The vaccine
complies with the test if (a) at the end of 72 hours the total Identification
mass of the group of vaccinated mice is not less than that
preceding the injection; (b) at the end of 7 days the average Identification tests A, B, C and D are carried out using the
increase in mass per vaccinated mouse is not less than vial containing the diphtheria, tetanus, pertussis and
60 per cent of that per control mouse; and (c) not more than poliomyelitis components; identification test E is carried
5 per cent of the vaccinated mice die during the test. The test out on the vial containing the haemophilus component.
may be repeated and the results of the tests combined. A. Diphtheria toxoid is identified by a suitable immunochernical
Bovine serum albumin. Determine on the poliomyelitis method (2.2.14). The following method, applicable to certain
components by a suitable immunochemical method (2.2.14) vaccines, is given as an example. Dissolve in the vaccine under
during preparation of the final bulle vaccine, before addition examination sufficient sodium citrate to give a 10 per cent
ofthe adsorbent, the amount ofbovine serum albumin is such wlv solution. Maintain at 37° for about 16 hours and centrifuge
that the content in the final vaccine will not be more than until a clear supernatant is obtained. The clear supernatant
50 ng per single human dose. reacts with a suitable diphtheria antitoxin, giving a precipitate.
Antimicrobial preservative. Where applicable, determine the B. Tetanus toxoid is identified by a suitable immunochemical
amount of antimicrobial preservative by a suitable chemical method (2.2.14). The following method, applicable to certain
method. The content is not less than 85.0 per cent and not vaccines, is given as an example. The clear sup ern a tan t
greater than 115.0 per cent of the intended amount. obtained during identification test A reacts with a suitable
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle tetanus antitoxin, giving a precipitate.
for each sterility medium. C. The centrifugationresidue obtained in identification A may
be used. Other suitable methods for separating the bacteria
FINAL LOT from the adsorbent may also be used. Identify pertussis
vaccine by agglutination ofthe bacteria from the resuspended
The final bulle ofthe haemophilus component is freeze-dried.
precipitate by antisera specific to B. pertussis or by the assay
Only a final lot that is satisfactory with respect to the test for
of the pertussis component prescribed under Assay.
osmolality shown below and with respect to each of the
requirements given below under Identification, Tests and D. The vaccine is shown to contain human polioviruses 1,2
Assay may be released for use. and 3 by a suitable immunochemical method (2.2.14), such as
determination ofD-antigenby enzyme linked immunosorbent
Provided that the tests for specific toxicity and antimicrobial
assay (ELISA).
preservative, and the assays for the diphtheria, tetanus and
pertussis components have been carried out with satisfactory E. The haemophilus component· is identified by a suitable
results on the final bulle vaccine, they may be omitted on the immunochemical method (2.2.14) for PRP.
final lot. Tests
Provided that the free formaldehyde content has been
The tests for speCific toxicity, aluminium, free formaldehyde,
determined on the bulle purified antigens, the inactivated B.
antimicrobial preservative and sterility are carried out on
pertussis suspension and. the purified monovalent harvests
the container with diphtheria, tetanus, pertussis and
or the trivalent pool ofpolioviruses or on the final bulle and it
poliomyelitis components; the tests for PRP content, wate/;
has been shown that the content in the final lot will not exceed
sterility and pyrogens are carried out on the container with
0.2 gil, the test for free formaldehyde may be omitted on the
the haemophilus component.
final lot.
Provided that the in vivo assay for the poliomyelitis component Some tests for the haemophilus component may be carried
has been carried out with satisfactory results on the final bulle out on the freeze-dried product rather than on the· bulk
vaccine, it may be omitted on the final lot. conjugate where theji-eeze-drying process may affect the
component under test.
Osmolality (2.4.23). The osmolality of the vaccine,
reconstituted where applicable, is within the limits approved PRP. Minimum 80.0 per cent ofthe amount ofPRP stated on
for the particular preparation. the label. PRP is determined either by assay ofribose (2.7.1) or

2365

www.webofpharma.com
ADTP, POLIOMYELITIS (INACTIVATED) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE9 IP 2010

phosphorus (2.7.1), by an immunochemical method (2.2.14)or expressed with reference to the amount of D-antigenstated
by anion-exchange liquid chromatography with pulsed- on the label, is within the limits approved for the particular
amperometric detection. product. Poliomyelitis vaccine (inactivated) reference
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, preparation is calibrated in Units and intended for use in the
if aluminiUlnhydroxide or hydrated aluminiumphosphate is assay of D-antigen. The Unit and the ill are equivalent.
used as the adsorbent. .
In vivo test
Free formaldehyde (2.3.20). Maximum 0.2 gil.
The vaccine complies with the in vivo assay as stated under
Antimicrobial preservative. Where applicable, determine the Poliomyelitis Vaccine (Inactivated).
amount of antimicrobial preservative by a suitable chemical
Labelling. The label states (1) the minimum number of
method. The content is not less than 85.0 per cent and not
International Units ofdiphtheria and tetanus toxoid per single
greater than 115.0 per cent of the intended amount.
human dose; (2) the minimum number ofIntemational Units
Water (2.3.43). Maximum 3.0 per cent for the haemophilus of pertussis vaccine per single human dose; (3) the nominal
component. amount of poliovirus of each type (1, 2 and 3), expressed in
Sterility (2.2. n). Complies with the test for sterility. UnitsofD-antigen per single human dose; (4) the type ofcells
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject used for production of the poliomyelitis component; (5) the
per kg ofthe rabbit's mass a quantity ofthe vaccine equivalent number ofmicrograms ofPRP per single human dose; (6) the
to 1 mg ofPRP for a vaccine with diphtheria toxoid or CRM type and nominal amount of carrier protein per single human
197 diphtheria protein as carrier; 0.1 mg ofPRP for a vaccine dose; (7) where applicable, that the vaccine is intended for
with tetanus toxoid as carrier; 0.025 mg ofPRP for a vaccine primary vaccination ofchildren and is not necessarily suitable
with OMP as carrier. for reinforcing doses or for administration to adults; (8) the
name and the amount of the adsorbent; (9) that the vaccine
Assay must be shaken before use; (10) that the vaccine is not to be
Diphtheria component frozen.

Carry out one of the prescribed methods for the assay as


stated under Diphtheria Vaccine (Adsorbed). Adsorbed Pertu.ssis Vaccine (Acellu.lar
The lower confidence limit (P = 0.95) ofthe estimated potency Component)
is not less than 30 IU per single human dose.
Pertussis Vaccine (Acellular Component, Adsorbed) is a
Tetanus component preparation of individually prepared and purified antigenic
components of Bordetella pertussis adsorbed on a mineral
Carry out one of the prescribed methods for the assay as carrier such as aluminium hydroxide or hydrated aluminium
stated under Tetanus. Vaccine (Adsorbed). phosphate.
If the test is carried out in guinea-pigs, the lower confidence The vaccine contains either pertussis toxoid or a pertussis
limit (P = 0.95) ofthe estimated potency is not less than 40 ill toxin, like protein free from toxic properties, produced by
per single human dose; if the test is carried out in mice, the expression ofa genetically modified form ofthe corresponding
lower confidence limit (P = 0.95) ofthe estimated potency is gene. Pertussis toxoid is prepared from pertussis toxin by a
not less than 60 ill per single human dose. method that renders the latter harmless while maintaining
adequate immunogenic properties and avoiding reversion to
Pertussis component
toxin. The vaccine may also contain filamentous
Carry o.ut the assay as stated under Pertussis Vaccine. haemagglutinin, pertactin (a 691cDa outer-membrane protein)
The estimated potency is not less than 4 ill per single human and other defined components ofB. pertussis such as fimbrial-
dose and the lower confidence limit (P = 0.95) ofthe estimated 2 and fimbrial-3 antigens. The latter 2 antigens may be
potency is not less than 2 IU per single human dose. copurified. The antigenic composition and characteristics are
based on evidence ofprotection and freedom from unexpected
Poliomyelitis component reactions in the target group for which the vaccine is intended.

D-antigen content Production


As a Illeasure of consistency of productio.n,deterIl1in~t~eD-
antigen content for human polioviruses 1,2 and 3 by a suitable The production method shall have been shown to yield
immunochemical method (2.2..1 4) using a reference preparation consistently the vaccines comparable with the vaccine of
calibrated in Units of D-antigen. For each type, the content, proven clinical efficacy and safety in man.

2366

www.webofpharma.com
IP 2010 ADSORBED PERTUSSIS VACCINE (ACELLULAR COMPONENT)

Reference vaccine PURIFIED COMPONENTS


A batch ofvaccine shown to be effective in clinical trials or a Production of each component is based on a seed-lot system.
batch representative thereof is used as a reference vaccine. The seed cultures from which toxin is prepared are managed
For the preparation of a representative batch, strict adherence to conserve or where necessary restore toxinogenicity by
to the production process used for the batch tested in clinical deliberate selection.
trials is necessary. The reference vaccine is preferably None of the media used at any stage contains blood or blood
stabilised by a method that has been shown to have no products of human origin. Media used for the preparation of
significant effect on the assay procedure when the stabilised seed lots and inocula may contain blood or blood products of
and non-stabilised batches are compared. animal origin.
Pertussis toxin and, where applicable, filamentous
CHARACTERISATION OF COMPONENTS haemagglutinin and pertactin are purified and, after appropriate
During development of the vaccine, the production process characterisation, detoxified using suitable chemical reagents,
shall be validated to demonstrate that it yields consistently by a method that avoids reversion of the toxoid to toxin,
individual components that comply with the following particularly on storage or exposure to heat. Other components
requirements; after demonstration of consistency, the tests such as fimbrial-2 and fimbrial-3 antigens are purified either
need not be applied routinely to each batch. separately or together, characterised and shown to be free
from toxic substances. The purification procedure is validated
Adenylate cyclase. Not more than 500 ng in the equivalent of to demonstrate appropriate clearance of substances used
1 dose ofthe final vaccine, determined by immunoblot analysis during culture or purification.
or another suitable method.
The content of bacterial endotoxins is determined to monitor
Tracheal cytotoxin. Not more than 2 pmol in the equivalent of the purification procedure and to limit the amount in the final
1 dose of the final vaccine, determined by a suitable method vaccine. The limits applied for the individual components are
such as a biological assay or liquid chromatography (2.4.14). such that the final vaccine contains less than 100 IU per single
human dose.
Absence ofresidual dermonecrotic toxin. Inject intradennally Before detoxification, the purity of the components is
into each of 3 unweaned mice, in a volume of 0.1 ml, the determined by a suitable method such as polyacrylamide gel
amount of component or antigenic fraction equivalent to 1 electrophoresis (pAGE) or liquid chromatography. SDS-PAGE
dose of the final vaccine. Observe for 48 hours. No or irnmunoblot analysis with specific monoclonal or polyclonal
dermonecrotic reaction is demonstrable. antibodies may be used to characterise subunits. Requirements
Specific properties. The components of the vaccine are are established for each individual product.
analysed by one or more of the methods shown below in Only purified components that comply with the following
order to detelmine their identity and specific properties (activity requirements may be used in the preparation of the final bulle
per unit amount of protein) in comparison with reference vaccine.
preparations. Sterility (2.2.11). Carry out the test for sterility using for each
medium a quantity of purified component equivalent to not
Pertussis toxin less than 100 doses.
Chinese hamster ovary (CHO) cell-clustering effect and Absence of residual pertussis toxin
haemagglutination as in vitro methods; lymphocytosis-
promoting activity, histamine-sensitising activity and insulin This test is not necessmyfor the product obtained by genetic
secretory activity as in vivo methods. The toxin shows ADP- modification. Use a group of not fewer than 5 histamine-
ribosyl transferase activity using transducin as the acceptor. se;nsitive mice each weighing between 18 and 26 g. Inject into
each mouse the equivalent of 1 human dose intravenously or
Filamentous haemalfglunnin twice the human dose intraperitoneally, diluted to not more
than 0.5 ml with phosphate-bufferedsaline solution containing
Haemagglutination and inhibition by specific antibody. 0.2 per cent w/v of gelatin. Inject diluent into a second group
Pertactin, fimbrial-2 and fimbrial-3 antigens. Reactivity with of control mice. After 5 days, inject 2 mg of histamine base
specific antibody. intraperitoneally in a volume not exceeding 0.5 ml and observe
for 24 hours. Ifno animal dies, the preparation complies with
Pertussis toxoid the test.
The toxoid induces in animals production of antibodies The histamine sensitivity ofthe strain ofmice used is verified
capable of inhibiting all the properties of pertussis toxin. at suitable intervals as follows: inject three-fold dilutions ofa

2367

www.webofpharma.com
ADSORBED PERTUSSIS VACCINE (ACELLULAR COMPONENT) IP 2010

reference pertussis toxin preparation in phosphate-buffered sufficient sodium citrate to give a 10 per cent w/v solution;
saline solution containing 0.2 per cent w/v of gelatin and maintain at 37° for about 16 hours and centrifuge until a clear
challenge with histamine as above; the strain is suitable if supernatant liquid is obtained. Examined by a suitabl,e
more than 50 per cent ofthe animals are sensitised by 50 ng of immunochemical method (2.2.14), the clear supematant liquid
pertussis toxin and none of the control animals injected with reacts with specific antisera to the components stated on the
only diluent and challenged similarly with histamine show label.
symptoms of sensitisation.
Tests
A validated test based on the clustering effect ofthe toxin for
Chinese hamster ovary (CHO) cells may be used instead of Absence of residual pertussis toxin and irreversibility of
the test on mice. pertussis toxoid

Residual detoxifying agents and other reagents This test is not necessaryfor the product obtained by genetic
modification. Use 3 groups each ofnot fewer than 5 histamine-
, The content ofresidual detoxifying agents and other reagents sensitive mice. Inject intraperitoneally into the first group twice
is determined and shown to be below approved limits unless the single human dose ofthe vaccine stored at 2° to 8°. Inject
validation of the process has demonstrated acceptable intraperitoneally into the second group twice the single human
clearance. dose ofthe vaccine incubated at 37° for 4 weeks. Inject diluent
Antigen content into the third group of mice. After 5 days, inject into each
mouse 2 mg of histamine base intraperitoneally in a volume
Determine the antigen content by a suitable immunochemical not exceeding 0.5 ml and observe for 24 hours. The test is
method (2.2.14) and protein nitrogen by sulphuric acid invalid if 1 or more control mice die following histamine
digestion (2.2.30) or another suitable method. The ratio of challenge. The vaccine complies with the test ifno animal in
antigen content to protein nitrogen is within the limits the first or second group dies following histamine challenge.
established for the product. If 1 m.ouse dies in either or both ofthe first and second groups,
the test may be repeated with the same number ofmice or with
FINAL BULK VACCINE
a greater number and the results of valid tests combined; the
The vaccine is prepared by adsorption of suitable quantities vaccine complies with the test if, in both of the groups given
ofpurified components, separately or together, onto aluminium the vaccine, not more than 5.0 per cent ofthe total number of
hydroxide or hydrated aluminium phosphate. A suitable mice die following histamine challenge.
antimicrobial preservative may be added. The histamine sensitivity ofthe strain ofmice used is verified
Only a final bulk vaccine that complies with the following at suitable intervals as follows: inject intravenously threefold
requirements may be used in the preparation of the final lot. dilutions of a reference pertussis toxin preparation in
phosphate-buffered saline solution c'ontaining 0.2 per cent
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical w/v of gelatin and challenge with histamine as above; the
method. The content is not less than 85.0 per cent and not strain is suitable if more than 50.0 per cent ofthe animals are
greater than 115.0 per cent of the intended amount. sensitised by 50 ng of pertussis toxin and none of the control
animals injected with only diluent and challenged similarly
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk with histamine show symptoms of sensitisation.
for each sterility medium.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
FINAL LOT if aluminium hydroxide or hydrated aluminium phosphate is
used as the adsorbent.
Only a final lot that is satisfactory with respect to each of the
requirements given below under Identification, Tests and Free formaldehyde (2.3.20). Maximum 0.2 gil.
Assay may be released for use. Provided that the tests for Antimicrobial preservative. Where applicable, determine the
absence of residual pertussis toxin and irreversibility of amount of antimicrobial preservative by a suitable chemical
pertussis toxoid, antimicrobial preservative, free formaldehyde method. The content is not less than 85.0 per cent and not
and the assay have been carried out with satisfactory results greater than 115.0 per cent of the intended amount.
on the final bulk vaccine, these tests may be omitted on the
Sterility (2.2.11). Complies with the test for sterility.

Identification Assay

Subject the vaccine to a suitable desorption procedure such The capacity ofthe vaccine to induce the formation ofspecific
as the following: dissolve in the vaccine under examination antibodies is compared with the same capacity of a reference

2368

www.webofpharma.com
IP 2010 ADSORBED PERTUSSIS VACCINE (ACELLULAR, CO-PURIFIED)

preparation examined in parallel; antibodies are determined The test is not valid if (a) the value found for the control
using suitable immunochemical methods (2.2.14) such as serum differs by more than 2 standard deviations from the
enzyme-linked immunosorbent assay (ELISA). The test on assigned value; (b) the confidence interval of the potency
mice shown below uses a three-point model but, after estimate is greater than 50.0 per cent to 200.0 per cent.
validation, for routine testing a single-dilution method may be
Calculation
used.
The antibody titres in the sera of mice immunised with
Requirement reference and test vaccines are calculated and from the values
obtained the potency of the test vaccine in relation· to the
The capacity to induce antibodies is not significantly
reference vaccine is calculated by the usual statistical methods.
(P = 0.95) less than that of the reference vaccine.
LabeUing. The label states (1) the names and amounts of the
The following test model is given as an example of a method components present in the vaccine; (2) where applicable, that
that has been found to be satisfactory. the vaccine contains a pertussis toxin-like protein produced
Selection and distribution of test animals by genetic modification; (3) the name and amount of the
adsorbent; (4) that the vaccine must be shaken before use;
Use in the test healthy mice (for example, CDI strain) of the
(5) that the vaccine is not to be frozen.
same stock 4 to 8 weeks old. Distribute the animals in 6 groups
of a number appropriate to the requirements ofthe assay. Use
3 dilutions ofthe vaccine under examination and 3 dilutions of
a reference preparation and attribute each dilution to a group Adsorbed Pertussis Vaccine (Acellular,
of mice. Inject intraperitoneally or subcutaneously into each Co-Purified)
mouse 0.5 ml of the dilution attributed to its group.
Pertussis Vaccine (Acellular, Co-Purified, Adsorbed) is a
Collection of serum samples preparation of antigenic components of Bordetella pertussis
4 to 5 weeks after vaccination, bleed the mice individually adsorbed on a mineral carrier such as aluminium hydroxide or
under anaesthesia. Store the sera at -20° until tested for hydrated aluminium phosphate.
antibody content. The vaccine contains an antigenic fraction purified without
separation ofthe individual components. The antigenic fraction
Antibody determination
is treated by a method that transfonns pertussis toxin to toxoid,
Assay the individual sera for content of specific antibodies to rendering it hannless while maintaining adequate immunogenic
each component using a validated method such as the ELISA properties of all the components and avoiding reversion to
test shown below. toxin. The antigenic fraction is composed ofpertussis toxoid,
filamentous haemagglutinin, pertactin (a 69 kDa outer-
ELISA
membrane protein) and other defined components of
Microtitre plates (polyvinyl chloride or polystyrene as B. pertussis such as fimbrial-2 and fimbrial-3 antigens. It may
appropriate for the specific antigen) are coated with the purified contain residual pertussis toxin up to a maximum level
antigen at a concentration of 100 ng per well. After washing, approved by the competent authority. The antigenic
unreacted sites are blocked by incubating with a solution of composition and characteristics are based on evidence of
bovine serum albumin and then washed. Two-fold dilutions protection and freedom from unexpected reactions in the target
of sera from mice immunised with test or reference vaccines group for which the vaccine is intended.
are made on the plates. After incubation at 22° to 25° for
1 hour, the plates are washed. A suitable solution of anti- Production
mouse IgG enzyme conjugate is added to each well and General provisions
incubated at 22° to 25° for 1· hour. After washing, a substrate is The production method shall have been shown to yield
added from which the bound enzyme conjugate liberates a consistently vaccines comparable with the vaccine ofproven
chromophore which can be quantified by measurement of clinical efficacy and safety in man.
absorbance. The test conditions are designed to obtain a linear Reference vaccine. A batch of vaccine shown to be effective
response for absorbance with respect to antibody content in clinical trials or a batch representative thereof is used as a
over the range of measurement used and absorbance values reference vaccine. For the prepar~tion of a representative
within the range 0.1 to 2.0. batch, strict adherence to the production process usedfor the
A reference antiserum of assigned potency is used in the test batch tested in clinical trials is necessary. The reference vaccine
and serves as the basis for calculation of the antibody levels is preferably stabilised, by a method that has been shown to
in test sera. A standardised control serum is also included in have no significant effect on the assay procedure when the
the test. stabilised and non-stabilised batches are compared.

2369

www.webofpharma.com
ADSORBED PERTUSSIS VACCINE (ACELLULAR, CO-PURIFIED) IP 2010

CHARACTERISATION OF COMPONENTS The antigenic fraction is purified and, after appropriate


characterisation, detoxified using suitable reagents by a
During development of the vaccine, the production process method that ensures minimal reversion of toxoid to toxin,
shall be validated to demonstrate that it yields consistently particularly on or exposure to heat. The purification procedure
an antigenic fraction that complies with the following is validated to demonstrate appropriate clearance of
requirements; after demonstration of consistency, the tests substances used during culture or purification.
need not be applied routinely to each batch.
The content of bacterial endotoxins is determined to monitor
Adenylate cyclase. Not more than 500 ng in the equivalent of the purification procedure and to limit the amount in the final
I dose ofthefinal vaccine, determined by immunoblot analysis vaccine. The limits applied are such· that the final vaccine
or another suitable method. contains not more than 100 ill per single human dose.
Tracheal cytotoxin. Not more than 2 pmol in the equivalent of Before detoxification, the purity of the antigenic fraction is
1 dose of the final vaccine, determined by a suitable method determined by a suitable method such as polyacrylamide gel
such as a biological assay or liquid chromatography (2.4,14). electrophoresis (PAGE) (2.4.12) or liquid chromatography
(2.4.14). SDS-PAGE or immunoblot analysis with specific
Absence of residual dermonecrotic toxin. Inject intradermally monoclonal or polyclonal antibodies may be used to
into each of3 unweaned mice, in a volumeofO.1 ml, the amount characterise subunits. Requirements are established for each
of antigenic fraction equivalentto I dose ofthe final vaccine. individual product.
Observe for 48 hours. No dermonecrotic reaction is
Only a purified antigenic fraction that complies with the
demonstrable.
following requirements may be used in the preparation ofthe
Specific properties. The antigenic fraction is analyzed by one final bulk vaccine.
or more ofthe methods shown below in order to determine the Sterility (2.2.11). Carry out the test for sterility using for each
. identity and specific properties (activity per unit amount of medium a quantity ofpurified antigenic fraction equivalent to
protein) of its components in comparison with reference not less than 100 doses of the final vaccine.
preparations.
Test for residual pertussis toxin. Use 3 groups of not fewer
Pertussis toxin
than 5 histamine-sensitive mice each weighing between 18
and 26 g. Using phosphate-buffered saline containing 0.2 per
Chinese hamster ovary (CRO) cell-clustering effect and cent of gelatin, prepare a series of dilutions of the purified
haemagglutination as in vitro methods; lymphocytosis- antigenic fraction that have been shown to yield a graded
promoting activity, histamine-sensitising activity and insulin response and attribute each dilution to a separate group of
secretory activity as in vivo methods. The toxin shows mice. Inject intraperitoneally into each mouse the dilution
ADP-ribosyl transferase activity using transducin as the attributed to its group. Inject diluent into a fourth group of
acceptor. control mice. After 5 days, inject intraperitoneally into each
mouse 1 mg of histamine base in a volume not exceeding
Filamentous haemagglutinin
0.5 mL Record the number ofanimals that die within 24 hours
Raemagglutination and inhibition by specific antibody. of histamine challenge. Calculate the weight or volume of a
Pertactin, fimbrial-2 and fimbrial-3 antigens. Reactivity with preparation that sensitises 50.0 per cent ofthe mice injected
specific antibody. using a suitable statistical method such as probit analysis.
The residual activity of pertussis toxin does not exceed that
Pertussis toxoid of batches shown to be safe in clinical studies.
The toxoid induces in animals the production of antibodies The histamine sensitivity ofthe strain ofmice used is verified
capable of inhibiting all the properties of pertussis toxin. at suitable intervals as follows: inject threefold dilutions of a
reference pertussis toxin preparation in phosphate-buffered
PURIFIED ANTIGENIC FRACTION saline solution containing 0.2 per cent w/v of gelatin and
challenge with histamine as described above; the strain is
Production of the antigenic fraction is based on a seed-lot suitable if more than 50 per cent of the animals are sensitised
system. The seed cultures are managed to conserve or, where by 50 ng of pertussis toxin and none of the control animals
necessary, restore toxinogenicity by deliberate selection. injected with only diluent and challenged similarly with
~_._., ."_'~-'._.'."._~-,-_.'- ",.,',-,'-,,"--- ' . " ... '_'--'~-_'_---'_'_~'_ .. _._"._,.~._-,-,."._,._,._.'.-_ .. ,._---~-,. __ __ .,----_ ,.,-_._._ _, _._.... .. __.. .....__..
. .. ... .•. _-, -

None of the lTIedia used atally stage contains blood or blood histalJlil1e show·symptoills of sellsitisation.
products of human origin.· Media used for the preparation of A validated test based on the clustering effect ofthe toxin for
seed batches and inocula may contain blood or blood products Chinese hamster ovary (CRO) cells may be used instead of
ofanimal origin. the test on mice.

2370

www.webofpharma.com
IP 2010 BACILLUS CALMETTE-GUERIN VACCINE (FREEZE-DRIED)

Residual detoxifying agents and other reagents. The content saline containing 0.2 per cent w/v of gelatin, prepare a series
ofresidual detoxifYing agents and other reagents is determined of dilutions of the vaccine under examination that have been
and shown to be below approved limits unless validation of shown to yield a graded response and attribute each dilution
the process has demonstrated acceptable clearance. to a separate group of mice. Inject intraperitoneally into each
mouse the dilution attributed to its group. Inject diluent into
Antigen content. Determine the complete quantitative antigen
a fourth group of control mice. After 5 days, inject
composition of the antigenic fraction by suitable
intraperitoneally into each mouse 1 mg of histamine base in a
immunochemical methods (2.2.14) and protein nitrogen by
volume not exceeding 0.5 ml. Note the number ofanimals that
sulphuric acid digestion or another suitable method. The ratio
die within 24 hours ofhistamine challenge. Calculate the weight
oftotal antigen content to protein nitrogen is within the limits
or volume of a preparation that sensitises 50 per cent of the
established for the product.
mice injected using a suitable statistical method such as probit
analysis. The residual activity of pertussis toxin does not
FINAL BULK VACCINE
exceed that of batches shown to be safe in clinical studies.
The vaccine is prepared by adsorption of a suitable quantity
Reversibility oftoxoid. Carry out the testfor residual pertussis
ofthe antigenic fraction onto aluminium hydroxide or hydrated
toxin described above using the vaccine incubated at 37°for
aluminium phosphate. A suitable antimicrobial preservative
4 weeks in parallel with a sample stored at 2° to 8°. The degree
may be added.
of reversibility does not exceed that of batches shown to be
Only a final bulk vaccine that complies with the following safe in clinical studies.
requirements may be used in the preparation of the final lot.
Antimicrobial preservative. Where applicable, determine the
Antimicrobial preservative. Where applicable, determine the amount of antimicrobial preservative by a suitable chemical
amount of antimicrobial preservative by a suitable chemical method. The content is not less than 85.0 per cent andnot
method. The content is not less than 85.0 per cent and not greater than 115.0 per cent of the intended amount.
greater than 115.0 per cent ofthe intended amount. Aluminium (2.3 .9). Maximum 1.25 mg per single human dose,
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulle if aluminium hydroxide or hydrated aluminium phosphate is
for each sterility medium. used as the adsorbent.
Free formaldehyde (2.3.20). Maximum 0.2 gil.
FINAL LOT
Sterility (2.2.11). Complies with the test for sterility.
Only a final lot that is satisfactory with respect to each of the
requirements given below under Identification, Tests and Assay
Assay may be released for use. The vaccine complies with the assay as stated underAdsorbed
Provided that the tests for residual pertussis toxin, reversibility Pertussis Vaccine (Acellular Component).
of toxoid, antimicrobial preservative, free formaldehyde and Labelling. The label states (1) the names and amounts of the
the assay have been carried out with satisfactory results on antigenic components present in the vaccine; (2) the maximum
the final bulle vaccine, these tests may be omitted on the final amount of residual pertussis toxin present in the vaccine;
lot. (3) the maximum degree ofreversion oftoxoid to toxin during
the period of validity; (4) the name and amount of the
Identification adsorbent; (5) that the vaccine must be shaken before use; (6)
that the vaccine is not to be frozen.
Subject the vaccine to a suitable desorption procedure such
as the following: dissolve in the vaccine under examination
sufficient sodium citrate to give a 10 per cent w/v solution;
maintain at 37° for about 16 hours and centrifuge until a clear
Bacillus Calmette-Guerin Vaccine
supernatant is obtained. Examine by a suitable immunochemical
method (2.2.14), the clear supernatant reacts with specific (Freeze-Dried)
antisera to the components in the vaccine. Freeze-dried BCG Vaccine is a preparation of live bacteria
derived from a culture of the bacillus ofCalmette and Guerin
Tests (Mycobacterium bovis BCG) capacity of which to protect
Test for residual pertussis toxin. Use 3 groups of not fewer against tUberculosis has been established.
than 5 histamine-sensitive mice (see under Production) each Vaccine complies with the requirements stated under Vaccines
weighing between 18 and 26 g. Using phosphate-buffered with the following modifications.

2371

www.webofpharma.com
BACILLUS CALMETTE-GUERIN VACCINE (FREEZE-DRIED) IP 2010

Production Identification
General provisions The bacteria in the working seed lot are identified as
Mycobacterium bovis BCG using microbiological techniques,
The production method is validated to demonstrate that the
which may be supplemented by molecular biology techniques
product, if tested, would comply with the tests for safety and
(for example, nucleic acid amplification and restriction-
efficacy.
fragment-length polymorphism).
BCG vaccine shall be produced by a staffconsisting ofhealthy Sterility (2.2.11). Complies with the test for sterility, carried
persons who do not work with other infectious agents; in out using 10 ml for each medium. The working seed lot
particular they shall not work with virulent strains of complies with the test for sterility exceptfor the presence of
Mycobacterium tuberculosis, during the course ofproduction mycobacteria.
cycle nor shall they be exposed to a known risk oftuberculosis
infection. BCG vaccine is susceptible to sunlight: the Virulent mycobacteria
procedures for the preparation of the vaccine shall be so Examine the working seed lot as prescribed under Tests, using
designed that all cultures and vaccines are protected from 10 guinea pigs.
direct sunlight and from ultraviolet light at all stages of
manufacture, testing and storage. PROPAGATION AND HARVEST

Production of the vaccine is based on a seed-lot system. The The bacteria are grown in a suitable medium for not more than
production method shall have been shown to yield 21 days by surface or submerged culture. The culture medium
consistently B.CG vaccines that induce adequate sensitivity shall contain no substances known to cause toxic or allergic
to tuberculin in man, that have acceptable protective potency reactions in human beings or to cause the bacteria to becol11e
in animals and are safe. The vaccine is prepared from cultures virulent for guinea-pigs. The culture is harvested and
which are derived from the master seed lot by as few suspended in a sterile liquid medium that protects the viability
subcultures as possible and in any case not more than 12 of the vaccine as determined by a suitable method of viable
subcultures e.g. Ifthe secondary seed lot is 4 culture passages count.
removed from the primmy seed lot, the number of passages Test for purity. Purity is checked by acid fast staining.
from the secondary seed lot must not exceed 8.
FINAL BULK VACCINE
The capacity of the working seed lot to induce sensitivity to
tuberculin in guinea-pigs is demonstrated. The final bulle vaccine is prepared from a single harvest or by
pooling a number of single harvests. A stabiliser may be
Ifa bioluminescence test or other biochemical method is used added; if the stabiliser interferes with the determination of
instead of viable count, the method is validated against the bacterial concentration on the final bulk vaccine, the
viable count for each stage of the process at which it is used. determination is carried out before addition of the stabiliser.

SEED LOT Only final bulk vaccine that complies with the following
requirements may be used in the preparation of the final lot.
The strain used to establish the master seed lot is chosen for
and maintained to preserve its stability, its capacity to sensitise Virulent mycobacteria. Examine as prescribed under Tests.
man and guinea-pigs to tuberculin and to protect animals Sterility (2.2.11). Complies with the test for sterility using
against tuberculosis, and its relative absence ofpathogenicity 10 ml for each medium except for the presence ofmycobacteria.
for man and laboratory animals. The strain used shall be
identified by historical records that include information on its Count of viable units
origin and subsequent manipulation.
Determine the number of viable units per ml by viable count
A suitable batch ofvaccine is prepared from the first working on solid medium using a method suitable for the vaccine under
seed lot and is reserved for use as the comparison! in-house examination or by determination of adenosine triphosphate
reference vaccine. When a new working seed lot is by a bioluminescence reaction. Carry out the test in parallel
established, a suitable test for delayed hypersensitivity in on a reference preparation of the same strain.
guinea-pigs is carried out on a batch ofvaccine prepared from
thenewwofk.ingseea·lot; th€-vac-cin€·is snown fo -be not Bacterial concentration
significantly different in activity from. the com.parison vaccine.
Determine the total bacterial concentration by a suitable
Only a working seed lot that complies with the following method, either directly by determining the mass ofthe micro-
requirements may be used for propagation. organisms, or indirectly by an opacity method that has been

2372

www.webofpharma.com
IP 2010 BACILLUS CALMETTE-GUERIN VACCINE (FREEZE-DRIED)

calibrated in relation to the mass of the organisms; if the Inject subcutaneously or intramuscularly into each of6 guinea-
bacterial concentration is determined before addition of a pigs,each weighing between 250 and 400 g and having received
stabiliser, the concentration in the final bulk vaccine is no treatment likely to interfere with the test, a quantity of
established by calculation. The total bacterial concentration vaccine equivalent to at least 50 human doses. Observe the
is within the limits approved for the particular product by animals for at least 42 days. At the end of this period, kill the
National Regulatory Authority. guinea-pigs and examine by autopsy for signs of infection
with tuberculosis, ignoring any minor reactions at the site of
The ratio of the count of viable units to the total bacterial injection. Animals that die during the observation period are
concentration is not less than that approved for the particular also examined for signs oftuberculosis. The vaccine complies
product by National Regulatory Authority. with the test if none of the guinea-pigs shows signs of
tuberculosis and if not more than one animal dies during the
FINAL LOT observation period. If 2 animals die during this period and
autopsy does not reveal signs of tuberculo~is repeat the test
The final bulle vaccine is distributed into sterile containers
on 6 other guinea-pigs. The vaccine complies with the test if
and freeze-dried to a moisture content favourable to the
not more than one animal dies during the 42 days following
stability ofthe vaccine; the containers are closed either under
the injection and autopsy does not reveal any sign of
vacuum or under a gas that is not deleterious to the vaccine.
tuberculosis.
Except where the filled and closed containers are stored at a Sterility (2.2.11). The reconstituted vaccine complies with the
temperature of_20 0 or lower, the expiry date is not later than 4 test for sterility except for the presence of mycobacteria.
years from the date of harvest.
Excessive dermal reacti~ity
Only a final lot that complies with the following requirement
for count of viable units and with each of the requirements Use 6 healthy white or pale-coloured guinea-pigs, each
given below under Identification, Tests and Assay may be weighing not less than 250 g and having received no treatment
released for use. Provided the test for virulent mycobacteria likely to interfere with the test. Inject intradennally into each
has been carried out with satisfactory results on the final bulle guinea-pig, according to a randomised plan, 0.1 ml of the
vaccine, it may be omitted on the final lot. Provided the test reconstituted vaccine and of 2 tenfold serial dilutions of the
for excessive dermal reactivity has been· carried out with vaccine and identical doses of the comparison vaccine.
satisfactory results on the working seed lot and on 5 Observe the lesions formed at the site of the injection for 4
consecutive final lots produced from it, the test may be omitted weeks. The vaccine complies with the test if the reaction it
on the final lot. produces is not markedly different froin that produced by the
comparison vaccine.
Count of viable units
Temperature stability
Determine the number of viable units per ml of the
reconstituted vaccine by viable count on solid medium using Maintain samples ofthe freeze-dried vaccine at 37 0 for 4 weeks.
a method suitable for the vaccine under examination or by Determine the number of viable units in the heated vaccine
determination ofadenosine triphosphate by a bioluminescence and in unheated vaccine as described below. The number of
reaction. The ratio of the count of viable units after freeze- viable units in the heated vaccine is not less than 20.0 per cent
drying to that before is not less than that approved for the of that in unheated vaccine.
particular product. Water (2.3.43). Not more than 3.0 per cent, determined by the
Identification semi-micro determination ofwater.

BCGvaccine is identified by microscopic examination ofthe Assay


bacilli in stained smears demonstrating their acid-fast property
Determine the number of viable units in the reconstituted
and by the characteristic appearance of colonies grown on
vaccine by viable count on solid medium or using a suitable
solid medium. Alternatively, molecular biology techniques (like
validated biochemical method for the vaccine under
nucleic acid amplification) may be used.
examination. The number is within the range stated on the
Tests label. Determine the number ofviable units in the comparison
vaccine in parallel.
Virulent mycobacteria
Labelling. The label states (l) the minimum and maximum
Ifthis test is satisfactory at final bulle stage it can be omitted at number of viable units per ml in the reconstituted vaccine;
the final lot. (2) that the vaccine must be protected from direct sunlight;

2373

www.webofpharma.com
IP 2010
DIPHTHERIA ANTITOXIN

(3) that the vaccine is to be used immediately after broaching Selection of test toxin.. In selecting a toxin for use as the test
the container; (4) the age group for which the vaccine is toxin detennine the following.
intended; (5) the dose for each age group; (6) follow
instructions as mentioned in the product insert/leaflet. Lr/l00 dose -This is the smallest quantity ofthe toxin which,
when mixed with 0.01 Unit of antitoxin and injected
intracutaneously into guinea-pigs or rabbits causes a
Diphtheria Antitoxin characteristic reaction at the site of the injection within 48
hours.
Diphtheria Antitoxin is a preparation containing the specific
antitoxic globulins or their derivatives obtained by purification Minimal reacting dose - This is the smallest quantity of
of hyperimmune serum or plasma of healthy horses or other toxin which, when injected intracutaneously into guinea-pigs
suitable animals and having the specific activity ofneutralising or rabbits, causes a characteristic reaction at the site of
the toxin fonned by Corynebacterium diphtheriae. The liquid injection within 48 hours.
preparation may contain a suitable antimicrobial preservative.
A suitable toxin is one which contains at least 200 minimal
Diphtheria Antitoxin has a potency ofnot less than 1000 Units reacting doses in the Lrll 00 dose. The test toxin is allowed to
per ml when obtained from horse serum and not less than 500 stand for some months before being used for the assay of
Units per ml when obtained from other animals. samples ofantitoxin. During this time its toxicity declines and
Description. A clear, colourless or pale yellow liquid or a freeze- the Lrll 00 dose may be slightly increased. When experiment
dried, cream-coloured powder or pellet. shows that the Lrll 00 dose is constant, the test toxin is ready
for use and may be used for a long period. Determine the
Identification minimal reacting dose and the Lrll 00 dose at frequent intervals:
Specifically neutralises and renders the toxin fonned by C. Store the test toxin in the dark at a temperature between 0° and
diphtheriae hannless to susceptible animals or by any other 50 . Maintain its sterility by the addition of toluene or other
suitable in-vitro test. antimicrobial preservative which does not cause a rapid decline
in specific toxicity.
Tests
Determination oftest dose oftoxin [Lr/l00 dose). Prepare a
Potency. Carry out the biological assay ofdiphtheria antitoxin solution ofthe Standard Preparation with saline solution such
described below. that 1 rnl contains 0.1 Unit. Prepare mixtures such that 2.0 ml of
each mixture contains 1.0 ml of the dilution of the Standard
Biological Assay of Diphtheria Antitoxin
Preparation (0.1 Unit) and one of a series of graded volumes
The potency of diphtheria an.titoxin is determined by of the test toxin. Dilute each mixture with saline solution to
comparing the dose necessary to protect guinea-pigs or the same final volume (2.0 ml). Allow the mixtures to stand at
rabbits against the erythrogenic effects of a fixed dose of the room temperature, protected from light, for 15 to 60 minutes
Standard Preparation ofdiphtheria antitoxin necessary to give and inject intracutaneously 0.2 ml of each mixture at suitably
the same protection. For this purpose, a suitable preparation spaced sites into the shaven or depilated flanks oftwo animals.
of diphtheria toxin is required to be used as a test toxin. The Observe the animals for 48 hours.
test dose ofthe toxin is detennined in relation to the Standard
Preparation. The potency ofthe preparation under examination The test dose (Lrll 00) ofthe toxin is the amount present in 0.2
is then detennined in relation to the Standard Preparation using ml ofthat mixture which causes at the site ofinjection a small,
the test toxin. characteristic reaction in the skin of the guinea-pig or rabbit.
Mixtures containing larger amounts of toxin cause larger
Standard Preparation reaction and necrosis and mixtures containing smaller amount
of toxin cause no reaction.
The Standard Preparation is the 1st International Standard for
Diphtheria antitoxin, equine, established in 1934, consisting Determination ofpotency ofthe antitoxin. Dilute the test toxin
ofthe dried hyperimmune horse serum and glycerin, or another with saline solution so that 1.0 ml contains 10 times the test
suitable preparation the potency ofwhich has been detennined dose. Prepare mixtures such that 2.0 ml ofeach mixture contains
in relation to the International Standard. 1.0 ml ofthe dilution ofthe toxin and one ofa series ofgraded
volumes ofthe preparation under examination. PrePase:futi1:ler
Suggested Method mixtures such that 2.0 rnl ofeach contains 1.0 rnl ofthe solution
Test toxin. Prepare diphtheria toxin by filtering through ofthe test toxin and 0.1 Unit of antitoxin. Dilute each mixture
bacteria-prooffilter the medium in which a toxigenic strain of with saline solution to the same final volume (2.0 ml). Allow
C. diphtheriae has grown. Store at a temperature of 2° and 8°. the mixtures to stand at room temperature, protected from

2374

www.webofpharma.com
IP 2010 DIPHTHERIA AND TETANUS VACCINE (ADSORBED)

light, for 15 to 60 minutes. Inject a dose of 0.2 ml of each adsorbent. Animals that die shall be autopsied and examined
mixture into the animals under the conditions described in the for symptoms of diphtheria intoxication (red adrenals). The
determination ofthe Lr/lOO dose ofthe toxin. bulle purified toxoid shall pass the test ifno guinea-pig shows
The mixture ofthe preparation under examination that contains symptoms ofspecific intoxication within six weeks ofinjection
0.01 Unit ofantitoxin in 0.2 ml is the mixture that produces the and ifat least 80 per cent ofthe animals survive the test period.
same degree oflocal reaction as that produced by the injection The guinea-pigs shall not have been used previously for
into the same animals ofthe mixture ofthe Standard Preparation experimental purposes.
that contains in 0.2 ml the test dose (Lr/l 00) of the toxin and Alternatively, a cell-culture test system may be used; in this
0.01 Unit ofantitoxin. case, the sensitivity of the test shall have been demonstrated
When at least four distinct tests are carried out by this method, to be not less than that of the guinea-pig test, and the test
the limits of error have been estimated to be between 90 per procedures shall be approved by the National Regulatory
cent and III per cent. Authority.
Other tests. Complies with the tests stated under Antisera. Each bulk purified toxoid shall be tested to ensure that
Storage. As stated under Antisera. reversion to toxicity cannot take place on storage. The bulle
purified toxoid shall be diluted in order to obtain the same
Labelling. The label states (1) the number ofUnits per ml; (2)
concentration and chemical environment as that present in
the species of animal from which the preparation has been
the final bulle vaccine, except for the presence of adjuvant.
made; (3) the recommended dose (if space is inadequate, it
may be stated in the instruction leaflet); (4) the name and To determine whether reversion has occurred, diluted toxoids
proportion ofany added preservative; (5) that the preparation, that have been stored at 37° for six weeks shall be tested. The
if liquid, should not be allowed to freeze; (6) that the test employed shall be approved by the National Regulatory
preparation, if dried, should be used immediately after Authority and should be sufficiently sensitive to detect very
reconstitution in the stated quantity of the diluent supplied small amounts oftoxin. No toxicity shall be detected.
by the manufacturer. Intradermal tests in guinea-pigs and cell-culture tests both
are considered to be suitable.

Antigenic purity
Diphtheria and Tetanus Vaccine
Not less than 1500 Lfper mg ofprotein nitrogen for diphtheria
(Adsorbed) toxoid and not less than 1000 Lflmg of protein nitrogen for
Diphtheria and Tetanus Vaccine (Adsorbed) is a preparation tetanus toxoid.
ofdiphtheria formol toxoid and tetanus formol toxoid adsorbed
FINAL BULK VACICNE
on mineral carrier. The formol toxoids are prepared from the
toxins produced by the growth of Corynebacterium The final bulk vaccine is prepared by adsorption of suitable
diphtheriae and Clostridium tetani, respectively. quantities ofbulk purified diphtheria toxoid and tetanus toxoid
The specification for individual component used in formulation onto mineral carrier such as hydrated aluminium phosphate,
is referred in the text of individual monograph. aluminium hydroxide; the resulting mixture is approximately
isotonic with blood. Suitable antimicrobial preservatives may
Production be added. Antimicrobial preservatives of the phenolic type
must not be used.
General provisions
Only final bulk vaccine that complies with the following
Bulk purified diphtheria and tetanus toxoids requirements may be used in the preparation ofthe final lot.
The bulk purified diphtheria and tetanus toxoids are prepared Identification
as described in the monographs on Diphtheria vaccine
(adsorbed) and Tetanus vaccine (adsorbed) and comply with A. Dissolve sufficient sodium citrate in the vaccine under
the requirements prescribed therein. examination to give a 10 per cent wIv concentration. Maintain
Sterility (2.2.11). Carry out test for sterility using 10 ml of at 37° for about 16 hours and centrifuge. The clear supernatant
bulle for each sterility medium. reacts with a suitable diphtheria antitoxin and yields a
precipitate.
Absence of toxin and irreversibility of toxoid
B. The clear supernatant obtained in testA reacts with a suitable
Inject subcutaneously into each of5 guinea-pigs at least 500 Lf
tetanus antitoxin and yields a precipitate.
ofthe non-incubated bulle purified toxoid in a volume of 1 ml,
using the same buffer solution as for the final vaccine, without pH (2.4.24).6.0 to 7.0.

2375

www.webofpharma.com
DIPHTHERIA AND TETANUS VACCINE (ADSORBED) IP 2010

Specific toxicity. Use 5 normal, healthy guinea-pigs weighing The clear supernatant liquid obtained during test A reacts
between 250 and 350 g which have been maintained for at with a suitable tetanus antitoxin, giving a precipitate or visible
least 1 week on a uniform, unrestricted diet, and have not floccules.
been previously treated with any material that will interfere
with the test. Weigh the animals separately and record their Tests
weights. Inject subcutaneously into each animal 5 times the Sterility (2.2.11). Complies with the test for sterility.
dose stated on the labe1. Weigh all the animals at weekly
intervals for 6 weeks. None of the animals shows any Abnormal toxicity (2.2.1). Complies with the test for abnormal
toxicity
symptoms of diphtheria or tetanus toxaemia or dies from
diphtheria within 42 days or loses weight at the end of the Aluminium (2.3 .9). Not more than 1.25 mg per single human
test. Ifmore than one animal diys from non-specific causes or dose when hydrated aluminium phosphate or aluminium
loses weight, repeat the test. If an animal dies or loses weight hydroxide is used as the adsorbent.
in the second test, the vaccine fails the test.
pH (2.4.24). The pH ofthe vaccine is within the range approved
Assay for the product (6.0 to 7.0).

Diphtheria toxoid Free formaldehyde (2.3.20). Maximum 0.2 gil.

Complies with the test as stated under Diphtheria Vaccine Antimicrobial preservative. Where applicable, determine the
(Adsorbed). amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not
Tetanus toxoid greater than 115.0 per cent ofthe quantity stated on the label.
Complies with the test as stated under Tetanus Vaccine Assay
(Adsorbed).
Antimicrobial preservative. Where applicable, determine the Diphtheria component
amount of antimicrobial preservative by a suitable chemical Carry out one of the described methods for the assay of
method. The amount is not less than 85.0 per cent and not Diphtheria Vaccine (Adsorbed).
greater than 115.0 per cent of the intended amount.
Viz a) Intradermal challenge method, b) Lethal challenge
Free formaldehyde (2.3.20): Maximum 0.2 gil method, c) Antibody induction method, d) Validated serological
Sterility (2.2.11). Carry out test for sterility using 10 ml of assay in guinea pigs or mice as approved by National
bulle for each sterility medium. Regulatory Authority.
FINAL LOT Tetanus component
The final bulle vaccine is filled and stored aseptically into Carry out one of the described methods for the assay of
sterile containers. The containers are closed so as to prevent Tetanus Vaccine (Adsorbed) Viz a) Antibody induction
contamination. method; b) Challenge method in guinea pigs/mice; c) Validated
Only a final lot that is satisfactory with respect to each ofthe serological assay in guinea pigs or mice as approved by
requirements given below under Identification, Tests and National Regulatory Authority.
Assay may be released for use. Provided the tests for specific Labelling. The label states (1) the human dose; (2) the minimum
toxicity, free formaldehyde and antimicrobial preservative and Lfunits per single human dose or the minimum International
the assay have been carried out with satisfactory results on Units per single human dose ifpotency test done by challenge
the final bulle vaccine, they may be omitted on the final lot. method; (3) the name and the amount of the adsorbent and
Identification preservative; (4) that the vaccine must be shaken before use;
(5) that the vaccine is not to be frozen.
A. Diphtheria toxoid is identified by a suitable immuno-
chemical method (2.2.14).
Dissolve in the vaccine under examination by adding sufficient
sodium citrate to give a 10 per cent w/v solution. Maintain at
Diphtheria and Tetanus Vaccine
37° for about 16 hours and centrifuge until a clear supernatant (Adsorbed) for Adults and Adolescents
liqUid is obtained. Thecleaf siipefriatanffea6ts with a suitable Diphtheria and Tetanus Vaccine (Adsorbed) for Adults and
diphtheria antitoxin, giving a precipitate or visible floccules. Adolescents is a preparation of diphtheria formol toxoid and
B. Tetanus toxoid is identified by a suitable immuno-chemical tetanus formo1 toxoid adsorbed ona mineral carrier. The formol
method (2.2.14). toxoids are prepared from the toxins produced by the growth

2376

www.webofpharma.com
IP 2010 DIPHTHERIA AND TETANUS VACCINE (ADSORBED) FOR ADULTS AND ADOLESCENTS

of Corynebacterium diphtheriae and Clostridium tetani, Free formaldehyde (2.3.20). Maximum 0.2 gil.
respectively.
Sterility (2.2.11). Carry out the test for sterility using 10 ml for
Production each medium.

General provisions FINAL LOT

Bulk purified diphtheria and tetanus toxoids The final bulle vaccine is distributed aseptically into sterile,
tamper-proof containers. The containers are closed so as to
The bulle purified diphtheria and tetanus toxoids are prepared prevent contamination.
as described in the monographs on Diphtheria vaccine
(adsorbed) and Tetanus vaccine (adsorbed) and comply with Only a final lot that is satisfactory with respect to each of the
the requirements prescribed therein. requirements given below under Identification, Tests and
Assay may be released for use. Provided the tests for free
FINAL BULK VACCINE formaldehyde and antimicrobial preservative and the assay
The vaccine is prepared by adsorption of suitable quantities have been carried out with satisfactory results on the final
of bulle purified diphtheria toxoid and tetanus toxoid onto a bulle vaccine, they may be omitted on the final lot.
mineral carrier such as hydrated aluminium phosphate or
aluminimn hydroxide. Suitable antimicrobial preservatives may
Identification
be added. Certain antimicrobial preservatives, particularly A. Diphtheria toxoid is identified by a suitable immunochemical
those of the phenolic type, adversely affect the antigenic method (2.2.14). The following method, applicable to certain
activity and must not be used. vaccines, is given as an example. Dissolve in the vaccine under
Only final bulle vaccine that complies with the following examination sufficient sodium citrate to give a 10 per cent
requirements may be used in the preparation ofthe final lot. w/v solution. Maintain at 37° for about 16 hours and centrifuge
until a clear supernatant is obtained. The clear supernatant
Identification reacts with a suitable diphtheria antitoxin, giving a precipitate.
A. Dissolve sufficient sodium citrate in the vaccine under Ifa satisfactory result is not obtained with a vaccine adsorbed
examination to give a 10 per cent w/v concentration. Maintain on aluminium hydroxide, carry out the test as follows.
at 37° for about 16 hours and centrifuge. The clear supernatant Centrifuge 15 ml ofthe vaccine under examination and suspend
reacts with a suitable diphtheria antitoxin and yields a the residue in 5 m1 ofa freshly prepared mixture of 1 volume of
precipitate. a 56 gil solution of sodium edetate and 49 volumes of a 90 gl
B. The clear supernatant obtained in testAreacts with a suitable I solution of disodium hydrogen phosphate. Maintain at 37°
tetanus antitoxin and yields a precipitate. for not less than 6 hours and centrifuge. The clear supernatant
reacts with a suitable diphtheria antitoxin, giving a precipitate.
pH (2.4.24 ). 6.0 to 7.0.
B. Tetanus toxoid is identified by a suitable immunochemica1
Specific toxicity method (2.2.14). The following method, applicable to certain
Inject subcutaneously 5 times the single human dose stated vaccines, is given as an example. The clear supernatant
on the label into each of5 healthy guinea-pigs, each weighing obtained during identification test A reacts with a suitable
between 250 and 350 g, that have not previously been treated tetanus' antitoxin, giving a precipitate.
with any material that will interfere with the test. Weigh the
animals separately and record their weights. Inject Tests
subcutaneously into each animal 5 times the dose stated on Aluminium (2.3.9). Maximum 1.25 mg per single human dose.
the label. Weigh all the animals at weekly intervals for 6 weeks.
None of the animals shows any symptoms of diphtheria or Free formaldehyde (2.3.20). Maximum 0.2 gil.
tetanus toxaemia or dies from diphtheria within 42 days or Antimicrobial preservative. Where applicable, determine the
loses weight at the end of the test. If more than one animal amount of antimicrobial preservative by a suitable chemical
dies from non-specific causes or loses weight, repeat the test. method. The amount is not less than 85.0 per cent and not
Ifan animal dies or loses weight in the second test, the vaccine greater than 115.0 per cent of the intended amount.
fails the test.
Sterility (2.2.11). Complies with the test for sterility.
Antimicrobial preservative. Where applicable, determine the
pH (2.4.24).6.0 to 7.0.
amount of antimicrobial preservative by a suitable chemical
method. The amount is not less than 85.0 per cent and not Abnormal toxicity (2.2.1). Complies with the test for abnormal
greater than 115.0 per cent ofthe intended amount. toxicity.

2377

www.webofpharma.com
DIPHTHERIA, TETANUS AND PERTUSSIS VACCINE (ADSORBED) IP 2010

Assay FINAL BULK VACCINE


The fmal bulk vaccine is prepared by adsorption of suitable
Diphtheria component
quantities ofbulk purified diphtheria toxoid and tetanus toxoid
Carry out the prescribed method for assay of Diphtheria' onto hydrated aluminium phosphate or aluminium hydroxide
Vaccine by lethal challenge method described in the assay of and admixture of an appropriate quantity of a suspension of
Diphtheria Vaccine (Adsorbed). inactivated B. pertussis. The B. pertussis concentration ofthe
The lower confidence limit (P = 0.95) ofthe estimated potency final bulle vaccine does not exceed that corresponding to an
is not less than 2 ill per single human dose. opacity of20 IOU per single human dose. Iftwo or more strains
of B. pertussis are used, the composition of consecutive lots
Tetanus component of the final bulle vaccine shall be consistent with respect to
the proportion of each strain as measured in opacity units.
Carry out one of the prescribed methods for the assay as
Suitable antimicrobial preservatives may be added to the bulk
stated under Tetanus Vaccine (Adsorbed).
vaccine. Antimicrobial preservatives particularly those of
The lower confidence limit (P = 0.95) ofthe estimated potency phenolic type which affect the antigenic activity must not be
is not less than 20 ill per single human dose. used.
Labelling. The label states (l) the human dose; (2) the minimum Only final bulk vaccine that complies with the following
number of International Units of each component per single requirements may be used in the preparation of the final lot.
human dose, ifpotency determined by challenge method; (3) Antimicrobial preservative. Where applicable, determine the
the name and the amount of the adsorbent and preservative; amount of antimicrobial preservative by a suitable chemical
(4) that the vaccine must be shaken before use; (5) that the method. The content is not less than 85.0 percent and not
vaccine is not to be frozen. more thanl15.0 per cent of the intended amount.
Sterility (2.2.11). Carry out test for sterility using 10 ml of
bulle for each sterility medium.

Diphtheria, Tetanus and Pertussis FINAL LOT


Vaccine (Adsorbed) The final bulk vaccirie is filled and stored aseptically into
sterile containers. The containers are closed so as to prevent
Diphtheria, Tetanus and Pertussis Vaccine (Adsorbed) is a contamination.
preparation ofdiphtheria formol toxoid, tetanus formol toxoid
Only a final lot that is satisfactory with respect to each ofthe
adsorbed on mineral carrier and a suspension of killed
requirements given below under Identification, Tests and
Bordetella pertussis organisms. The formal toxoids are
Assay may be released for use. Provided the tests for specific
prepared from the toxins produced by the growth of
toxicity of diphtheria, tetanus and pertussis components, free
Corynebacterium diphtheriae and Clostridium tetani,
formaldehyde, antimicrobial preservative and the Assay have
respectively. The Bordetella pertussis suspension is prepared
been carried out with satisfactory results on the final bulk
by growth of suitable strains in an appropriate medium, under
vaccine, they may be omitted on the final lot.
controlled conditions.
Identification
The specification for individual component used in formulation
is referred in the text of individual monograph. A. Diphtheria toxoid is identified by a suitable immuno-
chemical method (2.2.14).
Production Dissolve in the vaccine under examination by adding sufficient
sodium citrate to give a 10 per cent w/v solution. Maintain at
General Provisions
37° for about 16 hours and centrifuge until a clear supernatant
The production method must be validated to demonstrate is obtained; reserve the precipitate for identification test C.
that the product if tested, would comply with the tests for The clear supernatant reacts with a suitable diphtheria
safety as described under monographs ofDiphtheria Vaccine, antitoxin, giving a precipitate.
Tetanus Vaccine (Adsorbed) and Pertussis Vaccine. B. Tetanus toxoid is identified by a suitable immunochemical
The hullcpmified_diphtheria_ancLtetanus.. . toxoids.-and ~l::'~1?~ (2.2.14).
inactivated B. pertussis suspension are preparl'l<:l as <:lesGribl'ld The clear supernatant obtained during identification test A
in the monograph on Diphtheria Vaccine (Adsorbed), Tetanus reacts with a suitable tetanus antitoxin, giving a precipitate.
Vaccine (Adsorbed) and Pertussis Vaccine respectively and C. The pertussis component is identified by agglutination of
comply with the respective requirements. the bacteria from the resuspended centrifugation residue (see

2378

www.webofpharma.com
IP 2010 DTP (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE (ADSORBED)

identification test A; other suitable methods for separating Aluminium (2.3 .9). Not more than 1.25 mg per single human
the bacteria from the adsorbent may also be used) by antisera dose, when hydrated aluminium phosphate or aluminium
specific to B. pertussis or by the assay of the pertussis hydroxide is used as the adsorbent.
component. pH (2.4.24).6.0 to 7.0.
Tests Free formaldehyde (2.3.20). Maximum 0.2 gil.
Sterility (2.2.11). Complies with the test for sterility. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Abnormal toxicity (2.2.1). Each final lot shall be tested for
method. The content is not less than 85.0 per cent and not
abnormal toxicity by injecting intraperitoneally one human
more than 115.0 per cent of the intended amount.
dose, but not more than 0.25 ml into each of the five mice
weighing between 17 to 22 g and at least one human dose but Assay
not more than 1.0 ml into each ofthe two guinea pigs weighing Diphtheria component
between 250 and 350 g. The preparation passes the test if
none of the animals dies or shows signs of ill health in 7 days Carry out one of the methods for the assay as stated under
following the injection. If one ofthe animal dies or shows the Diphtheria Vaccine (Adsorbed).
signs of ill health, repeat the test. The preparation passes the
Tetanus component
test if none of the animals in the second group dies or shows
signs of ill health in the time interval specified. Carry out one of the methods for the assay as stated under
Tetanus Vaccine (Adsorbed).
Specific toxicity
Iftest is carried out in Guinea pigs, the lower confidence limit
Diphtheria and tetanus components. Inject subcutaneously (P=0.95) of estimated potency is not less than 40 IUISingle
five times the single human dose stated on the label into each human dose; ifthe test is carried in mice, the lower confidence
of five healthy guinea-pigs, each weighing between 250 and limit (P=0.95) of estimated potency is not less than 60 lUI
350 g, that have not previously been treated with any material Single human dose.
that will interfere with the test. The animals should be weighted
every week and observations be made. None of the animals Pertussis component
shows any symptoms of diphtheria or tetanus toxaemia or Carry out the assay as stated under Pertussis Vaccine.
dies from diphtheria within 42 days or loses weight at the end Labelling. The label states (l) in case done by challenge
of the test. If more than one animal dies from non-specific method the minimum number of International Units; if units
cause or loses weight, repeat the test. If an animal dies or (as applicable for each component) per single human dose;
loses weight in the second test, the vaccine fails the test. (2) In case done by antibody induction method the minimum
Pertussis component. Use not less than 10 healthy mice each number of International Units per single human dose of
weighing between 14 and 16 g for the vaccine group and for pertussis component and minimum number ofLfofdiphtheria
the saline control. Use mice ofthe same sex or distribute males toxoid and tetanus toxoid; (3) the name and the amount ofthe
and females equally between the groups. Allow the animals adsorbent and preservative; (4) that the vaccine must be
access to food and water for at least 2 hours before injection shaken before use; (5) that the vaccine is not to be frozen;
and during the test. Inject each mouse of the vaccine group (6) for vaccine contained in single-dose containers where the
intraperitoneally with 0.5 ml, containing a quantity of the space is too small to accommodate the full name ofthe vaccine,
vaccine equivalent to not less than half the single human the abbreviation 'DTP' may be used in the label and the
dose. Inject each mouse of the control group with 0.5 ml of a container provided that the same code is also stated in the
0.9 per cent sterile solution of sodium chloride, preferably label on the package.
containing the same amount of antimicrobial preservative as
that injected with the vaccine. Weigh the mice groups
immediately before the injection and 72 hours and 7 days after Diphtheria, Tetanus, Pertussis (Whole
the injection. The vaccine complies with the test if: (a) at the
end of 72 hours the total mass ofthe group ofvaccinated mice Cell), Hepatitis B (rDNA) and
is not less than that preceding the injection; (b) at the end of Haemophilus Type b Conjugate
7 days the average increase in mass per vaccinated mouse is
not less than 60 per cent ofthat per control mouse; and (c) not
Vaccine (Adsorbed)
more than 5 per cent ofvaccinated mice should die during the Diphtheria, Tetanus, Pertussis (Whole cell), Hepatitis B (rDNA)
test. The test may be repeated and the results of the tests and Haemophilus Type b Conjugate Vaccine (Adsorbed) is a
combined. combined vaccine composed of diphtheria formol toxoid

2379

www.webofpharma.com
DTP (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE (ADSORBED) IP20lO

containing not less than 1,500 Lf, (2.2.16) per mg of protein inject subcutaneously 5 times the single human ,dose. stated
nitrogen, purified tetanus formol toxoid containing not less on the label into each of 5 healthy guinea pigs, each weighing
than 1,000 Lf, (2.2.16), per mg ofprotein nitrogen, hepatitis B between 250 and 350 g, that have not previously been treated
surface antigen and haemophilus type b conjugated to suitable with any material that will interfere with the test. Ifwithin 42
protein with a mineral adsorbent to which a suspension of days ofthe injection any ofthe animals shows signs of or dies
killed Bordetellapertussis has been added. Mineral adsorbent from diphtheria, toxemia or tetanus, the vaccine does not
is a suspension ofhydrated aluminium hydroxide,aluminium comply with the test. If more than 1 animal dies from non-
phosphate or calcium. phosphate, in saline solution or other specific causes, repeat the test once; if more than 1 animal
appropriate solution isotonic with blood. shows signs of or dies in the second test, the vaccine does
not comply with the test.
The formol toxoids are prepared from the toxin produced by
the growth of Corynebacterium diphtheriae and Clostridium The stability of the final lot and the relevant intennediates is
tetani, respectively, in suitable media. The toxins are converted evaluated using one or more indicator tests. For the
to toxoids by treatment with formaldehyde solution by haemophilus component, such tests may include determination
methods which avoid reversibility of the toxoids. ofrnolecular size, determinatiol1offree PRP in the conjugate
and kinetics ofdepolymerisation. Taking accountofthe results
Hepatitis B surface antigen is a component protein ofhepatitis
of the stability testing, release requirements are set for these
B virus; the antigen is obtained by recombinant DNA
indicator tests to ensure that the vaccine will be satisfactory
technology.
at the end of the period of validity.
The polysaccharide, polyribosyl ribitol phosphate, PRP is a
lillear cop()lYlller c.Olnp()s~~ of repeated units of 3-~-D­ Reference vaccine(s)
ribofuranosyl-(1-.71 )-ribitol-5-phosphate [(CIOH,P,l)n]' with Provided valid assays can be performed, monocomponent
a defined molecular size and derived from a suitable strain of reference vaccines may be used for the assays on the combined
Haemophilus injluenzae type b. The canier protei'n, when vaccine. If this is not possible because of interaction between
conjugated to PRP, is capable of inducing a T-cell dependent the components of the combined vaccine or because of the
B-cell immune response to the polysaccharide. difference in composition between monocomponent reference
vaccine and the test vaccine, a batch of combined vaccine
The product may be presented with the haemophilus
shown to be effective in clinical trials or a batch representative
component in a separate container, the contents of which are
thereof is used as a reference vaccine. For the preparation of
mixed with the other components immediately before or during
a representative batch, strict adherence to the production
use.
process used for the batch tested in clinical trials is necessary.
The final product contains a suitable antimicrobial The reference vaccine may be stabilized by a method that has
preservative. The antigenic properties of the vaccine are been shown to have no effect on the assay procedure.
adversely affected by the presence of certain antimicrobial
preservatives particularly those ofthe phenolic type and some Production of the components
of the quaternary ammonium type and must not be used. The production of the components complies with the
requirements of the monographs on Diphtheria Vaccine
Production (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine,
Hepatitis B Vaccine (rDNA) and Haemophilus Type b
General provisions
Conjugate Vaccine.
The production method shall have been shown to yield
consistently the vaccines comparable with the vaccine of FINAL BULK VACCINE
proven clinical efficacy and safety in man. Vaccine with all components in the same container
If the vaccine is presented with the haemophilus component
The final bulk is prepared by adsorption, separately or
in a separate vial, as part of consistency studies the assays of
together, of suitable quantities of bulle purified diphtheria
the diphtheria, tetanus, pertussis and hepatitis Bare canied
toxoid, bulle purified tetanus toxoid, bulle purified hepatitis B
out on a suitable number of batches of vaccine reconstituted
surface antigen onto a mineral canier such as aluminium
as for use. For subsequent routine control, the assays ofthese
hydroxide or hydrated aluminium phosphate, admixture ofan
components may becanied out without mixing with the
liaemophiliis" comiioneiil~ _._~ . _"._-_._-"',"-"'-, .,._-" .-,-,'",-".. . --.-.. .-.-.-.- appmpriate__ quantity_oLa_ suspensioll-oLinactiYated
B, p?,r(ussis component and admixture ofa sJlitaple quantity
The production method is validated to demonstrate that the of PRP conjugate; the resulting mixture is approximately
product, if tested, would comply with the following test for isotonic with blood. The B. pertussis concentration of the
specific toxicity of the diphtheria and tetanus component: final bulle vaccine does not exceed that conesponding to an

2380

www.webofpharma.com
IP 2010 DTP (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE (ADSORBED)

opacity of20 ill per single human dose. If2 or more strains of FreePRP
B. pertussis are used, the composition of consecutive lots of
Unbound PRP is determined after removal of the conjugate,
the final bulle vaccine shall be consistent with respect to the
for example by anion exchange, size exclusion or hydrophobic
proportion ofeach strain as measured in opacity units. Suitable
chromatography (2.4.16), ultrafiltration or other validated
antimicrobial preservatives may be added.
methods. The amount of free PRP is not greater than that
Vaccine with the haemophilus component in a separate approved for the particular product.
container
Osmolality (2.4.23). The osmolality ofthe vaccine is within
The final bulk is prepared by adsorption, separately or the limits approved for the particular preparation.
together, of suitable quantities of bulk purified diphtheria
toxoid, bulk purified tetanus toxoid, bulk purified hepatitis B pH (2.4.24). 6.0 to 7.0.
surface antigen onto a mineral carrier such as aluminium Description. Whitish turbid liquid in which the mineral carrier
hydroxide or hydrated aluminium phosphate, admixture ofan tends to settle down slowly on keeping.
appropriate quantity of a suspension of inactivated B.
pertussis component and admixture of a suitable quantity of Identification
PRP conjugate; the resulting mixture is approximately isotonic
with blood. The B. pertussis concentration of the final bulle Tests A, B, C, D and E may be omitted if test F is carried out.
vaccine does not exceed that corresponding to an opacity of Test F may be omitted iftests A, B, C, D and E are carried out.
20 ill per single human dose. If2 or more strains ofB. pertussis A. Diphtheria toxoid. Dissolve sufficient sodium citrate in
are used, the composition of consecutive lots ofthe final bulle the vaccine under examination to give a 10 per cent w/v
vaccine shall be consistent with respect to the proportion of concentration. Maintain at 37° for about 16 hours and
each strain as measured in opacity units. The final bulle is centrifuge. Reserve the residue for test C. The clear
filled separately. Suitable antimicrobial preservatives may be supernatant reacts with a suitable diphtheria antitoxin and
added. The final bulk of the haemophilus component is yields a precipitate.
prepared by dilution of the bulk conjugate to the final
B. Tetanus toxoid. Tetanus toxoid is identified by suitable
concentration with a suitable diluent. A stabilizer may be added.
immunochemical method. The clear supernatant obtained as
Only a final bulle vaccine that complies with the following described in identification testA reacts with a suitable tetanus
requirements may be used in the preparation ofthe final lot. antitoxin to give a positive reaction, when tested by a suitable
Antimicrobial preservative. Where applicable, determine the validated immunological method.
amount of antimicrobial preservative by a suitable chemical C. Pertussis component. To a suspension of the residue
method. The amount is not less than 85.0 per cent and not obtained in test A in saline solution add a suitable Bordetella
greater than 115.0 per cent ofthe intended content. pertussis antiserum; agglutination indicates presence of
Sterility (2.2.11). Carry out test for sterility using 10 ml of pertussis component.
bulle for each sterility medium. D. Hepatitis B swface antigen. The suspension ofthe residue
obtained in test A gives a positive reactions when tested by
FINAL LOT
suitable in-vitro assay.
Onlya final lot that is satisfactory with respect to the test for E. PRP. The suspension of the residue obtained in the test A
osmolality and with respect to eachofthe requirements given gives a positive reaction when tested by a suitable
below under Identification, Tests and Assay may be released immunochemical method for PRP.
for use. Provided the tests for specific toxicity of diphtheria
toxoid, tetanus toxoid and pertussis component and F. The vaccine confers an active immunity in mjce and
antimicrobial preservative and the assays for the diphtheria, bguinea-pigs when administered as directed in the test for
tetanus and pertussis components have been carried out with Assay.
satisfactory results on the final bulle vaccine, they may be Tests
omitted on the final lot. Provided the content of free
formaldehyde has been determined on the bulk purified If the product is presented with the haemophilus component
antigens or on the final bulle and ithas been shown that the in a separate container; the tests for specific toxicity of
content in the final lot will not exceed 0.2 gil, the test for free diphtheria toxoid, tetanus toxoid and pertussis component,
formaldehyde may be omitted on the final lot. If an in vivo aluminium, free formaldehyde, antimicrobial preservative
assay is used for the hepatitis B component, provided it has and sterility are carried out on the container with the
been carried out with satisfactory results on the final bulle diphtheria, tetanus, pertussis and hepatitis B components;
vaccine, it may be omitted on the final lot. the tests for PRP content, water (where applicable), sterility

2381

www.webofpharma.com
DTP (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE (ADSORBED) IF 2010

Assay

and pyrogens are carried out on the container with the Diphtheria toxoid (adsorbed)
haemophilus component. Complies with the test as stated under Diphtheria and Tetanus
Ifthe haemophilus component isfreeze-dried, some tests may Vaccine (Adsorbed).
be carried out on the freeze-dried product rather than on the Tetanus toxoid (adsorbed)
bulk conjugate where the freeze-drying process may affect
the component under test. Complies with the test as stated under assay ofTetanus Vaccine
(Adsorbed).
PRP. Not less than 80.0 per cent of the amount ofPRP stated
on the label. PRP is determined either by assay ofribose (2.7.1), Pertussis vaccine
or phosphorus (2.7.1), by an immunochemical method (2.2.14)
Complies with the test as stated under Diphtheria, Tetanus
or by anion exchange liquid chromatography with pulsed
and Pertussis Vaccine (Adsorbed).
amperometric detection.
Aluminium (2.3.9). Not more than 1.25 mg per single human Hepatitis B suiface antigen (adsorbed)
dose, ifaluminium hydroxide or hydrated aluminium phosphate Complies with the test as stated under Hepatitis B Vaccine
is used as the adsorbent. (Adsorbed).
Free formaldehyde (2.3.20). Maximum 0.2 gil. Storage. When stored under the prescribed conditions the
Antimicrobial preservative. Where applicable, determine the vaccine may be expected to retain potency for not less than 2
amount of antimicrobial preservative by a suitable chemical years from the date on which the potency test forthe pertussis
method. The content is not less than 85 per cent and is not component was started.
greater than 115 per cent of the quantity stated on the label. Labelling. The label states (l) the human dose; (2) Diphtheria
Sterility (2.2.11). Complies with the test for sterility. and Tetanus components; (a) in case done by challenge
method, the minimum number of International Units (as
Abnormal toxicity (2.2.1). Each final lot shall be tested for applicable for each component) per single human dose; (b) in
abnormal toxicity by injecting intraperitoneally 0.25 ml ofthe case done by antibody induction, the minimum Lf units per
vaccine into each offive mice weighing between 17 and 22 g single human dose; (3) pertussis component - ill or IOU per
and at least one human dose but not more than 1.0 ml into single human dose; (4) hepatitis B component - J.lg HBsAg
each oftwo guinea pigs weighing between 250 and 350 g. The per single human dose; (5) haemophilus conjugate component
preparation passes the test if none of the animals dies or - mg PRP per single human dose; (6) the type and nominal
shows signs of ill health in seven days following the injection. amount ofcarrier protein per single human dose; (7) the name
If one of the animals dies or shows signs of ill health, repeat and amount of adsorbent and added preservative; (8) that the
the test. The preparation passes the test if none of the animals vaccine must be shaken before use; (9) that the vaccine is not
in the second group dies or shows signs of ill health in the to be frozen.
time interval specified.
Pyrogens (2.2.8). This test is carried out for Haemophilus
injluenzae type b vaccine only if Haemophilus injluenzae
type b vaccine is presented as separate lyophilized vial. The Diphtheria, Tetanus, Pertussis (Whole
vaccine complies with the test for pyrogens. Inject per kg of Cell) and Hepatitis B (rDNA) Vaccine
the rabbit's mass a quantity ofthe vaccine equivalent to: I mg
of PRP for a vaccine with diphtheria toxoid or CRM 197
(Adsorbed)
diphtheria toxoid as carrier; 0.1 mg ofPRP for a vaccine with Diphtheria, Tetanus, Pertussis and Hepatitis B (rDNA) Vaccine
tetanus toxoid as carrier protein; 0.025 mg ofPRP for vaccine (Adsorbed) is a combined vaccine composed of diphtheria
with aMP as carrier. formol toxoid containing not less than 1,500 Lf (2.2.16), per
mg of protein nitrogen, purified tetanus formol toxoid
Specific toxicity containing not less than 1,000 Lf ( 2.2.16), per mg of protein
Diphtheria and tetanus components nitrogen and hepatitis B surface antigen with a mineral
~'_"_·_'~_' _ _'_"_""'_······_·_'· __•· __.__ ,,_.·._.,m_.,,_
adsorbent-towhich-asuspension-ofkilledBordetellapertussis-
.•_ _ ,_ _. ,._"_...•... ., ._ _•_ _ •_ _ "._ _ ~ ,__,.__._._.

_c;~lIlPli~s",ith the t~sta~stat~dul1d.er piphtheriaand 'Tetanus has been added. MineraLadsorbenLis a suspension of
Vaccine (Adsorbed). hydrated aluminium hydroxide, aluminium phosphate or
Pertussis component calcium phosphate in saline solution or other appropriate
solution isotonic with blood.
Complies with the test as stated under Diphtheria, Tetanus
and Pertussis Vaccine (Adsorbed):
2382

www.webofpharma.com
IP 2010 DIPHTHERIA, TETANUS, PERTUSSIS (WHOLE CELL), HEPATITIS B (rDNA) VACCINE (ADSORBED)

The formol toxoids are prepared from the toxin produced by (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
the growth of Corynebacterium diphtheriae and Clostridium and Hepatitis B Vaccine (rDNA).
tetani, respectively, in suitable media. The toxins are converted
to toxoids by treatment with formaldehyde solution by FINAL BULK VACCINE
methods, which avoid reversibility of the toxoids.
The final bulk vaccine is prepared by adsorption, separately
Hepatitis B surface antigen is a component protein ofhepatitis or together, of suitable quantities of bulle purified diphtheria
B virus; the antigen is obtained by recombinant DNA toxoid, tetanus toxoid, pertussis whole cell suspension and
technology. hepatitis B surface antigen onto a mineral carrier such as
The final product contains a suitable antimicrobial aluminium hydroxide or hydrated aluminium pqosphate.
preservative. The antigenic properties of the vaccine are Suitable antimicrobial preservatives may be added. Only a
adversely affected by the presence of certain antimicrobial final bulle vaccine that complies with the following requirements
preservatives particularly those ofthe phenolic type and some may be used in the preparation ofthe final lot.
of the quaternary ammonium type and must not be used. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Production method. The amount is not less than 85.0 per cent and not
General provisions greater than 115.0 per cent of the intended content.

The production method shall have been shown to yield pH (2.4.24). 6.0 to 7.0.
consistently the vaccines comparable with the vaccine of Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk
proven clinical efficacy and safety in man. for each sterility medium.
The production method is validated to demonstrate that the
product, if tested, would comply with the following test for FINAL LOT
specific toxicity of the diphtheria and tetanus component: Only a final lot that is satisfactory with respect to the test for
inject subcutaneously 5 times the single human dose stated osmolality and with respect to each ofthe requirements given
on the label into each of 5 healthy guinea pigs, each weighing below under Identification, Tests and Assay may be released
between 250 and 350 g, that have not previously been treated for use. Provided the tests for specific toxicity of diphtheria
with any material that will interfere with the test. If within toxoid, tetanus toxoid and pertussis component and
42 days of the injection any of the animals shows signs of or antimicrobial preservative and the assays for the diphtheria,
dies from diphtheria, toxaemia or tetanus, the vaccine does tetanus and pertussis components have been carried out with
not comply with the test. Ifmore than 1 animal dies from non- satisfactory results on the final bulle vaccine, they may be
specific causes, repeat the test once; if more than 1 animal omitted on the final lot. Provided the content of free
shows signs of or dies in the second test, the vaccine does formaldehyde has been determined ori the bulk purified
not comply with the test. antigens or on the final bulle and it has been shown that the
The stability of the final lot and the relevant intermediates is content in the final lot will not exceed 0.2g/l, the test for free
evaluated using one or more indicator tests. Taking account formaldehyde may be omitted on the final lot. If an in vivo
of the results of the stability testing, release requirements are assay is used for the hepatitis B component, provided it has
set for these indicator tests to ensure that the vaccine will be been carried out with satisfactory results on the final bulle
satisfactory at the end of the period of validity. vaccine, it may be omitted on the final lot.
Reference vaccine(s) Osmolality (2.4.23). The osmolality of the vaccine is within
Provided valid assays can be performed, monocomponent the limits approved for the particular preparation.
reference vaccines may be used for the assays on the combined Description. Whitish turbid liquid in which the mineral carrier
vaccine. Ifthis is not possible because of interaction between tends to settle down slowly on keeping.
the components of the combined vaccine or because of the
difference in composition between monocomponent reference Identification
vaccine and the test vaccine, a batch of combined vaccine
shown to be effective in clinical trials or a batch representative Tests A, B, C andD may be omitted iftest E is carried out. Test
thereof is used as a reference vaccine. E may be omitted if tests A, B C and D are carried out.

Production ofthe components A. Diphtheria toxoid. Dissolve sufficient sodium citrate in


the vaccine under examination to give a 10 per cent w/v
The production of the components complies with the concentration. Maintain at 37° for about 16 hours and
requirements of the monographs on Diphtheria Vaccine centrifuge. Reserve the residue for test C. The clear

2383

www.webofpharma.com
DIPHTHERIA, TETANUS, PERTUSSIS (WHOLE CELL), HEPATITIS B (rDNA) VACCINE (ADSORBED) IP 2010

supernatant reacts with a suitable diphtheria antitoxin and Tetanus toxoid (adsorbed)
yields a precipitate.
Complies with the test as stated under assay for Tetanus
B. Tetanus toxoid. The clear supernatant obtained in test A Vaccine (Adsorbed).
reacts with a suitable tetanus antitoxin and yields a precipitate.
Pertussis vaccine
C. Pertussis component. To a suspension of the residue
obtained in testA in saline solution add a suitable B. pertussis Complies with the test as stated under Pertussis Vaccine
antiserum; agglutination indicates presence of pertussis (Adsorbed).
component. Hepatitis B sw1ace antigen (adsorbed)
D. HepafJitis B surface antigen. The suspension ofthe residue Complies with the test as stated under Hepatitis B Vaccine
obtained in test A gives a positive reactions when tested by (Adsorbed).
suitable in-vitro assay.
Storage. When stored under the prescribed conditions the
E. The vaccine confers an active immunity in mice and guinea- vaccine may be expected to retain potency for not less than 2
pigs when administered as directed under Assay. years from the date on which the potency test for the pertussis
component was started.
Tests
Labelling. The label states (1) the human dose (ml); (2)
pH (2.4.24).6.0 to 7.0. diphtheria and tetanus components, (a) in case done by
Aluminium (2.3.9). Not more than 1.25 mg per single human challenge method, the minimum number ofInternational Units
dose, ifaluminium hydroxide or hydrated aluminium phosphate (as applicable for each component) per single human dose
is used as the adsorbent. and (b) in case done by antibody induction, the minimum Lf
Free formaldehyde (2.3.20). Maximum 0.2 gil. units per single human dose; (3) pertussis component - ill or
Antimicrobial preservative. Where applicable, determine the IOU per single human dose; (4) hepatitis B component -!J.g
amount of antimicrobial preservative by a suitable chemical HBsAg per single human dose; (5) the name and amount of
method. The content is not less than 85.0 per cent and not adsorbent and added preservative; (6) that the vaccine must
greater than 115.0 per cent ofthe intended amount. be shaken before use; (7) that the vaccine is not to be frozen.
Sterility (2.2.11). Complies with the test for sterility.
Abnormal toxicity (2.2.1). Each final lot shall be tested for
abnormal toxicity by injecting intraperitoneally one human Diphtheria, Tetanus, Pertussis (Whole
dose but not more than 0.25 m1 into each offive mice weighing Cell) and Haemophilus Type b
between 17 and 22 g and at least one human dose but not
more than 1.0 ml into each of two guinea pigs weighing
Conjugate Vaccine (Adsorbed)
between 250 and 350 g. The preparation passes the test if Diphtheria, Tetanus, Pertussis and Haemophilus type b
none of the animals dies or shows signs of ill health in seven Conjugate Vaccine (Adsorbed) is a combined vaccine
days following the injection. If one of the animals dies or composed ofdiphtheria formol toxoid containing not less than
shows signs of ill health, repeat the test. The preparation 1,500 Lf, (2.2.16) per mg ofprotein nitrogen, purified tetanus
passes the test ifnone ofthe animals in the second group dies formol toxoid containing not less than 1,000 Lf, (2.2.16), per
or shows signs of ill health in the time interval specified. mg of protein nitrogen, and Haemophilus type b conjugated
Specific toxicity to suitable protein with a mineral adsorbent to which a
suspension of killed Bordetella pertussis has been added.
Diphtheria and tetanus components The mineral adsorbent is a suspension ofhydrated aluminium
Complies with the test as stated under Diphtheria and Tetanus hydroxide, aluminium phosphate or calcium phosphate, in
Vaccine (Adsorbed). saline solution or other appropriate solution isotonic with
blood.
Pertussis component
Complies with the test as stated under Pertussis Vaccine The fonnol toxoids are prepared from the toxin produced by
(Adsorbed). the growth of COIynebacterium diphtheriae and Clostridium
tetani, respectively in suitable media. The toxins are converted
Assay to toxoids by treatment with formaldehyde solution by
Diphtheria toxoid (adsorbed) methods which avoid reversibility ofthe toxoids;· .
Complies with the test as stated under Diphtheria Vaccine The polysaccharide, polyribosyl ribitol phosphate, PRP is a
(Adsorbed). linear copolymer composed of repeated units of

2384

www.webofpharma.com
IP 2010 DTP (WHOLE CELL) AND HAEMOPHILUS TYPE b CONJUGATE VACCINE (ADSORBED)

3 - ~ - D-ri b ofurano s y 1- (1-71) -ri b i to 1- 5 -phosp hate Reference vaccine(s)


[(CIQH'Pll)n]' with a defined molecular size and derived from ' Provided valid assays can be performed, monocomponent
a suitable strain ofHaemophilus injluenzae type b. The carrier reference vaccines may be used for the assays on the combined
protein, when conjugated to PRP, is capable of inducing a T- vaccine. If this is not possible because of interaction between
cell dependent B-cell immune response to the polysaccharide. the components of the combined vaccine or because of the

The product may be presented with the haemophilus difference in composition between monocomponent reference
component in a separate container, the contents of which are vaccine and the test vaccine, a batch of combined vaccine
mixed with the other components immediately before or during shown to be effective in clinical trials or a batch representative
use. thereof is used as a reference vaccine. For the preparation of
a representative batch, strict adherence to the production
The final product contains a suitable antimicrobial process used for the batch tested in clinical trials is necessary.
preservative. The antigenic properties of the vaccine are The reference vaccine may be stabilized by a method that has
adversely affected by the presence of certain antimicrobial been shown to have no effect on the assay procedure.
preservatives particularly those ofthe phenolic type and some
of the quaternary ammonium type must not be used. Production of the components
The production of the components complies with the
Production requirements of the monographs on Diphtheria Vaccine
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
General provisions (Whole Cell) and Haemophilus injluenzae Type b Conjugate
The production method shall have been shown to yield Vaccine.
consistently the vaccines comparable with the vaccine of
FINAL BULK VACCINE
proven clinical efficacy and safety in man.
Vaccine with all components in the same container
If the vaccine is presented with the haemophilus component
in a separate vial, as part of consistency studies, the assays The final bulk is prepared by adsorption, separately or
of the diphtheria, tetanus and pertussis are carried out on a together, of suitable quantities of bulk purified diphtheria
suitable number of batches of vaccine reconstituted for use. toxoid, bulle pmi.fied tetanus toxoid, onto a mineral carrier such
For subsequent routine control, the assays of these as aluminium hydroxide or hydrated aluminium phosphate,
components may be carried out without mixing with the admixture of an appropriate quantity of a suspension of
haemophilus component. inactivated B. pertussis component and admixture ofa suitable
quantity of PRP conjugate; the resulting mixture is
The production method is validated to demonstrate that the approximately isotonic with blood. The B. pertussis
product, if tested, would comply with the following test for concentration of the final bulle vaccine does not exceed that
specific toxicity of the diphtheria and tetanus component: corresponding to an opacity of 20 ill per single human dose.
inject subcutaneously 5 times the single human dose stated If2 or more strains ofB. pertussis are used, the compositionof
on the label into each of5 healthy guinea pigs, each weighing consecutive lots of the final bulle vaccine shall be consistent
between 250 and 350 g, that have not previously been treated with respect to the proportion of each strain as measured in
with any material that will interfere with the test. If within 42 opacity units. Suitable antimicrobial preservatives may be
days ofthe injection any ofthe animals shows signs of or dies
added.
from diphtheria, toxemia or tetanus, the vaccine does not
comply with the test. If more than 1 animal dies from non- Vaccine with thehaemophilus component in a separate
specific causes, repeat the test once; if more than 1 animal container
shows signs of or dies in the second test, the vaccine does
not comply with the test. The final bulk is prepared by adsorption, separately or
together, of suitable quantities of bulk purified diphtheria
The stability of the final lot and the relevant intermediates is toxoid, bulle purified tetanus toxoid onto a mineral carrier such
evaluated using one or more indicator tests. For the as aluminium hydroxide or hydrated aluminium phosphate,
haemophilus component, such tests may include determination admixture of an appropriate quantity of a suspension of
of molecular size, determination offree PRP in the conjugate inactivated B. pertussis component and admixture ofa suitable
and kinetics ofdepolymerisation. Taldng account ofthe results quantity of PRP conjugate; the resulting mixture is
of the stability testing, release requirements are set for these approximately isotonic with blood. The B. pertussis
indicator tests to ensure that the vaccine will be satisfactory concentration of the final bulle vaccine does not exceed that
at the end of the period of validity. corresponding to opacity of20 ill per single human dose. If 2

2385

www.webofpharma.com
DTP (WHOLE CELL) AND HAEMOPillLUS TYPE b CONJUGATE VACCINE (ADSORBED) IP 2010

or more strains of B. pertussis are used, the composition of A. Diphtheria toxoid. Dissolve sufficient sodium citrate in
consecutive lots of the final bulk vaccine shall be consistent the vaccine under examination to give a 10 per cent w/v
with respect to the proportion of each strain as measured in concentration. Maintain at 37° for about 16 hours and
opacity units. The final bulle is filled separately. Suitable centrifuge. Reserve the residue for test C. The clear
antimicrobial preservatives may be added. The final bulle of supernatant reacts with a suitable diphtheria antitoxin and
the haemophilus component is prepared by dilution of the yields a precipitate.
bulle conjugate to the final concentration with a suitable diluent.
B. Tetanus toxoid. The clear supernatant obtained in test A
A stabilizer may be added.
reacts with a suitable tetanus antitoxin and yields a precipitate.
Only a final bulk vaccine that complies with the following C. Pertussis component. To a suspension of the residue
requirements may be used in the preparation of the final lot. obtained in test A in saline solution add a suitable Bordetella
Antimicrobial preservative. Where applicable, determine the pertussis antiserum; agglutination indicates presence of
amount of antimicrobial preservative by a suitable chemical pertussis component.
method. The amount is not less than 85.0 per cent and not D. PRP. The suspension of the residue obtained in test A
greater than 115.0 percent of the intended content. gives a positive reaction when tested by a suitable
Sterility (2.2.11). Carry out test for sterility using 10 ml of immunochemical method for PRP.
bulk for each sterility medium. E. The vaccine confers an active immunity in mice and guinea
pigs when administered as directed in the test for Potency.
FINAL LOT
Tests
Where the haemophilus component is in a separate container,
the final bulk of the haemophilus component is freeze-dried.
If the product is presented with the haemophilus component
in a separate container; the tests for specific toxicity of
Only a final lot that is satisfactory with respect to the test for
diphtheria toxoid, tetanus toxoid and pertussis component,
osmolality and with respect to each ofthe requirements given
aluminium, free formaldehyde, antimicrobial preservative
below under Identification, Tests and Assay may be released
and sterility are carried out on the container with the
for use. Provided the tests specific toxicity ofdiphtheria toxoid,
diphtheria, tetanus and pertussis components; the tests for
tetanus toxoid and pertussis component and antimicrobial
PRP content, water (where applicable), sterility and
preservative and the assays for the diphtheria, tetanus and
pyrogens are carried out on the container with the
pertussis components have been calTied out with satisfactory
haemophilus component.
results on the final bulk vaccine, they may be omitted on the
final lot. Provided the content of free formaldehyde has been Ifthe haemophilus component isfreeze-dried, some tests may
detennined on the bulle purified antigens or on the final bulle be carried out on the jioeeze-driedproduct rather than on the
and it has been shown that the content in the final lot will not bulk conjugate where the freeze-d,ying process may affect
exceed 0.2 gil, the test for free formaldehyde may be omitted the component under test.
on the final lot. PRP. Not less than 80.0 per cent ofthe amount ofPRP stated
Free PRP. Unbound PRP is determined after removal of the on the label. PRP is determined either by as~ay ofribose (2.7.1 )
conjugate, for example by anion exchange, size exclusion or or phosphorus (2.7.1), by an immunochemical method (2.2.14)
hydrophobic chromatography (2.4.16), ultrafiltration or other or by anion exchange liquid chromatography with pulsed
validated methods. The amount offree PRP is not greater than amperometric detection.
approved for the particular product. Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
Osmolality (2.4.23). The osmolality of the vaccine is within if aluminium hydroxide or hydrated aluminium phosphate is
the limits approved for the particular preparation. used as the adsorbent.

pH (2.4.24). 6.0 to 7.0. Free formaldehyde (2.3.20). Maximum 0.2 gil.

Description. Whitish turbid liquid in which the mineral carrier Antimicrobial preservative. Where applicable, determine the
tends to settle down slowly on keeping. amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not
Production greater than 115.0 per cent of the intended amount.
test
Identification
Abnormal toxicity (2.2.1). Each final lot shall be tested for
Tests A, B, C andD may be omitted iftest E is carried out. Test abnormal toxicity by injecting intraperitoneally a maximum of
E may be omitted if tests A, B, C and D are carried out. one human dose, but not more than 0.25 rol into each of the

2386

www.webofpharma.com
IP 2010 DIPHTHERIA VACCINE (ADSORBED)

five mice weighing between 17 to 22 g and at least one human nominal amount of carrier protein per single human dose; (6)
dose but not more than 1.0 ml into each ofthe two guinea pigs the name and amount· of adsorbent and added preservative;
weighing between 250 and 350 g. The preparation passes the (7) that the vaccine must be shaken before use; (9) that the
test if none ofthe animals dies or shows signs of ill health in vaccine is not to be frozen.
7 days following the injection . If one of the animal dies or
shows the signs of ill health, repeat the test. The preparation
passes the test ifnone of the animals in the second group dies Diphtheria Vaccine (Adsorbed)
or shows signs of ill health in the time interval specified.
Diphtheria Vaccine (Adsorbed) is a preparation of diphtheria
Pyrogens (2.2.8). This test is carried out for Haemophilus
formol toxoid with a mineral adsorbent. The formol toxoid is
injluenzae type b vaccine only if Haemophilus injluenzae
prepared from the toxin produced by the growth of
type b vaccine is presented as separate lyophilized vial. The
COIynebacterium diphtheriae.
vaccine complies with the test for pyrogens. Inject per kg of
the rabbit's mass a quantity ofthe vaccine equivalent to: 1 mg Production
of PRP for a vaccine with diphtheria toxoid or CRM 197
diphtheria toxoid as carrier; 0.1 mg ofPRP for a vaccine with General provisions
.tetanus toxoid as carrier protein; 0.025 mg ofPRP for vaccine
with aMP as carrier. The maximum number ofLfunits per single human dose
of diphtheria vaccine (adsorbed) is 30.
Specific toxicity
Diphtheria and tetanus components Bulk purified toxoid
Complies with the test as stated under Diphtheria and Tetanus For the production of diphtheria toxin, from which toxoid is
Vaccine (Adsorbed). prepared, seed cultures are managed in a defined seed-lot
Pertussis component system in which toxinogenicity is conserved and, where
necessary, restored by deliberate reselection. A highly
Complies with the test as stated under Diphtheria, Tetanus toxinogenic strain of Corynebacterium diphtheriae with
and Pertussis Vaccine (Adsorbed). lmown origin and history is grown in a suitable liquid medium.
Assay At the end of cultivation, the purity of each culture is tested
and contaminated cultures are discarded. Toxin-containing
Diphtheria toxoid (adsorbed) culture medium is separated aseptically from the bacterial mass
Complies with the test as stated under Diphtheria Vaccine as soon as possible. The toxin content (Lf per ml) is checked
(Adsorbed). to monitor consistency ofproduction. Single harvests may be
pooled to prepare the bulle purified toxoid. The toxin is purified
Tetanus toxoid (adsorbed) to remove components likely to cause adverse reactions in
Complies with the test as stated under assay of Tetanus humans. The purified toxin is detoxified with formaldehyde by
Vaccine (Adsorbed). a method that avoids destruction ofthe immunogenic potency
ofthe toxoid and reversion ofthe toxoid to toxin, particularly
Pertussis vaccine on exposure to heat. Alternatively, purification may be carried
Complies with the test as stated under Pertussis Vaccine out after detoxification.
(Adsorbed). Only bulle purified toxoid that complies with the following
Storage. When stored under the prescribed conditions the requirements may be used in the preparation ofthe final bulle
vaccine may be expected to retain potency for not less than 2 vaccine.
years from the date on which the potency test for the pertussis Sterility (2.2.11). Carry out test for sterility using 10 ml of
component was started. bulle for each sterility medium.
Labelling. The label states (1) the human dose (ml); (2)
Absence of toxin and irreversibility of toxoid
Diphtheria and Tetanus components (a) in case done by
challenge method the minimum number ofintemational units Inject subcutaneously into each of5 guinea-pigs at least 500 Lf
(as applicable for each component) per single human dose; of the non-incubated bulk purified toxoid in a volume of
(b) in case done by antibody induction method, the minimum I ml, using the same buffer solution as for the final vaccine,
Lfunits per single human dose; (3) pertussis component - ill without adsorbent. Animals that die shall be autopsied and
or IOU per single human dose; (4) haemophilus conjugate examined for symptoms of diphtheria intoxication (red
component - mg PRP per single human dose; (5) the type and adrenals). The bulle purified toxoid shall passthe test if no

2387

www.webofpharma.com
DIPHTHERIA VACClNE (ADSORBED) IP 2010

guinea-pig shows symptoms of specific intoxication within plates and incubate at 37° for 5 to 6 days. Cytotoxic effect is
six weeks ofinjection and ifat least 80 per cent ofthe animals judged to be present where there is complete metabolic
survive the test period. The guinea-pigs shall not have been inhibition of the Vero cells, indicated by the pH indicator of
used previously for experimental purposes. the medium. Confirm cytopathic effect by microscopic
examination or suitable staining such as MIT dye. The test is
Altematively, a cell-culture test system may be used; in this invalid if5 x 10-5Lfperml ofreference diphtheria toxin in 100
case, the sensitivity of the test shall have been demonstrated Lfper m1 toxoid has no cytotoxic effect on Vero cells or ifthe
to be not less than that of the guinea-pig test, and the test cytotoxic effect of this amount of toxin is not" neutralised in
procedures shall be approved by the National Regulatory the wells containing diphtheria antitoxin. The bulk purified
Authority. toxoid complies with the test if no toxicity neutralisable by
Each bulk purified toxoid shall be tested to ensure that antitoxin is found in either sample.
reversion to toxicity cannot take place on storage. The bulk Antigenic purity. Not less than 1,500 Lf per mg of protein
purified toxoid shall be diluted in order to obtain the same nitrogen.
concentration and chemical enviromnent as those present in
the final bulk vaccine, except for the presence of adjuvant.
FINAL BULK VAc:CINE
To determine whether reversion has occurred, diluted toxoids
The final bulk vaccine is prepared by adsorption of a suitable
that have been stored at 37° for six weeks shall be tested: The
quantity ofbulk purified toxoid onto a mineral carrier such as
test employed shall be approved by the National Regulatory
hydrated aluminium phosphate or aluminium hydroxide; the
.Authority and should be sufficiently sensitive to detect very
resulting mixture is approximately isotonic with blood. Suitable
smaJI amounts oftoxin. No toxicity shall be detected.
antimicrobial preservatives may be added. Certain
Intradermal tests in guinea-pigs and cell-culture tests both antimicrobial preservatives, particularly those ofthe phenolic
are considered to be suitable. type, adversely affect the antigenic activity and must not be
used.
Cell culture method
Using the same buffer solution as for the final vaccine, without Only a final bulk vaccine that complies with the following
adsorbent, prepare a solution ofbulk purified toxoid at 100 Lf requirements may be used in the preparation of the final lot.
per m!. Divide the solution into 2 equal parts. Maintain I part
at 5° ± 3° and the other at 37° for 6 weeks. Carry out a test in Identification
Vero cells for active diphtheria toxin using 50 III per well of
Dissolve sufficient sodium citrate in the vaccine under
both samples. The sample should not contain antimicrobial
examination to give a 10 per cent w/v concentration. Maintain
preservatives and detoxifying agents should be detennined
at 37° for about 16 hours and centrifuge. The clear supematant
to be below the concentration toxic to Vera cells. Non-specific
reacts with a suitable diphtheria antitoxin and yields a
toxicity may be eliminated by dialysis.
precipitate.
Use freshly trypsinised Vero cells at a suitable concentration,
for example 2.5 x 105per ml and a reference diphtheria toxin pH (2.4.24). 6.0 to 7.0.
diluted in 100 Lfper ml diphtheria toxoid. A suitable reference Specific toxicity. Use 5 normal, healthy guinea-pigs weighing
diphtheria toxin will contain either not less than 100 LD5iml between 250 and 350 g, which have been maintained for at
or 67 to 133 h"/100 in I Lfand 25,000 to 50,000 minimal reacting least 1 week on a uniform, unrestricted diet, and have not
doses for guinea-pig skin in I Lf (diphtheria toxin RP is been previously treated with any material that will interfere
suitable for use as the reference toxin). Dilute the toxin in 100 with the test. Weigh the animals separately and record their
Lflml diphtheria toxoid to a suitable concentration, for example weights. Inject subcutaneously into each animal 5 times the
2 x 10-4 Lfperml. Prepare serial twofold dilutions ofthe diluted dose stated on the label. Weigh all the animals at weekly
diphtheria toxin and use undiluted test samples (50 III per intervals for 6 weeks. None of the animals show signs of or
well). Distribute them in the wells of a sterile tissue culture dies from diphtheria toxaemia within 42 days or loses weight
plate containing a medium suitable for Vera cells. To ascertain at the end of the test. Ifmore than one animal dies from non-
that any cytotoxic effect noted is specific to diphtheria toxin, specific causes or loses weight, repeat the test. If an animal
prepare.inparalleLdilutions.where.the toxin.isneutralisedbya dies or loses weight in-the second test, the vaccine fails the
suitable concentration of diphtheria antitoxin, .for example test.
100 IU/m!. Include control wells without toxoid or toxin and
with non-toxic toxoid at 100 Lfper ml on each plate to verify Potency. Determine by any ofthe methods ofbiological assay
normal cell growth. Add cell suspension to each well, seal the of Adsorbed Diphtheria Vaccine described.

2388

www.webofpharma.com
IP 2010 DIPHTHERIA VACCINE (ADSORBED)

Biological assay of adsorbed diphtheria vaccine ml of the challenge toxin solution and with 0.2 ml ofeach of
the five dilutions thereof in such a way as to minimize
(a) Intradermal challenge method
interference between adjacent sites. If necessaty, inject the
The potency of adsorbed diphtheria vaccine is detennined by unvaccinated control guinea-pigs with dilutions containing
comparing the dose necessary to protect guinea-pigs against 8x10-5 , 4x10-5, 2x 10-5 , Ix10-5 and 5xlO-6 Lfofthe challenge toxin.
the erythrogenic effects ofa range ofintradermal injections of Examine all the injection sites 48 hours after injection of the
diphtheria toxin with the dose of the Standard preparation of challenge toxin and record the incidence ofspecific diphtheria
adsorbed diphtheria toxoid necessary to give the same elythema. Record also the number of sites free from such
protection. For this comparison, the Standard preparation of reactions as the intradermal challenge score. Tabulate the
adsorbed diphtheria toxoid and a suitable preparation of intradermal challenge scores for all the animals receiving the
diphtheria toxin, for use as a challenge toxin, are required. same dilution of vaccine and use those data with a suitable
transfonnation, such as (score? or arcsin [(score/6)2], to obtain
Standard preparation
an estimate of the relative potency for each of the test
The Standard preparation is International standard of preparations by parallel-line quantitative analysis.
Diphtheria toxoid, adsorbed, or another suitable preparation The test is not valid unless (a) for both the preparation under
the potency of which has been detennined in relation to the examination and the Standard preparation, therriean score
International Standard. obtained at the lowest dose level is more than three; (b) if
applicable, the toxin dilution that contains 4x10·5 Lf gives a
Suggested method
positive erythema in at least 80.0 per cent ofthe control guinea-
Test animals. Use white guinea-pigs, weighing between 250 pigs and the dilution that contains 2x I0-5 Lf gives no reaction
and 350 g, from the same stock. Distribute the guinea-pigs in at least 80 per cent of the guinea-pigs (if these criteria are
into no fewer than six equal groups; use groups containing a not met a different toxin has to be selected); (c) the fiducial
number of animals sufficient to obtain results that fulfill the limits of the assay fall between 50.0 and 200.0 per cent ofthe
requirements for a valid Assay prescribed below. The guinea- estimated potency; (d) the statistical analysis shows no
pigs are all of the same sex or the males and females are deviation from linearity and parallelism. The test may be
distributed equally among the groups. If the challenge toxin repeated but when more than one test is perfonned the results
to be used has not been shown to be stable or has not been ofall valid tests must be combined in the estimate ofpotency.
adequately standardized, include five guinea-pigs as
The lower fiducial limit oferror ofthe estimated potency is not
unvaccinated controls.
less than 30 Units per dose.
Selection of the challenge toxin. Select a preparation of
diphtheria toxin containing 67 to 133 Limes reactionis/I 00 (Lr/
(b) Lethal challenge method
100) in Limes flocculationis (Lf) and 25,000 to 50,000 minimal
reacting doses for guinea-pig skin in I Lf. If the challenge Test animals. Use healthy, white or light-coloured guinea-
toxin preparation has been shown to be stable, it is not pigs from the same stock, weighing between 250 and 350 g.
necessary to verif'y the activity for every assay. Distribute them into six groups ofsixteen; and four groups of
four. The guinea-pigs should all be of the same sex or the
Preparation ofthe challenge toxin solutions. Immediately prior
males and females should be distributed equally between the
to use, dilute the challenge toxin with a suitable diluent to
six groups of sixteen.
obtain a challenge toxin solution containing about 512xI04 Lf
in 0.2 m!. Dilute a portion of this challenge toxin solution to Challenge toxin. Select a preparation of diphthe~ia toxin
give a series of five 4-fold dilutions. containing not less than 100 LD50 in 1.0 m!.
Determination of potency of the vaccine. Prepare in saline Preparation ofthe challenge toxin solutions. Immediately prior
solution dilutions of the vaccine under examination and of to use, prepare from the challenge toxin by dilution in
the Standard preparation such that, for each, the dilutions phosphate buffiredsalinepH 7.4, or normal saline a challenge
fonn a series differing by not more than 2.5 fold steps and in toxin containing approximately 100 LD 50 in 1.0 m!. Dilute
which the dilutions of intermediate concentration, when portions of this challenge toxin solution to 2LD50' I LD 50 and
injected subcutaneously in 1.0 ml volumes into guinea-pigs, Yz LD 50 in the same solution
result in an intradennal score ofapproximately three when the Determination of potency of the vaccine. Prepare in saline
animals are challenged. Allocate the dilutions, one to each of solution three dilutions of the vaccine under examination and
the groups of guinea-pigs, and inject subcutaneously 1.0 ml three dilutions ofthe Standard preparation such that for each,
of each dilution into each guinea-pig in the group to which the dilutions form a series differing by not more than 2.5 fold
that dilution is allocated. After 28 days shave both flanks of steps and in which the dilutions ofintermediate concentration,
each guinea-pig and inject each animal intradennally with 0.2 when injected subcutaneously in 1.0 ml volumes into guinea-

2389

www.webofpharma.com
DIPHTHERIA VACCINE (ADSORBED IP 2010

pigs, protect approximately 50 per cent of the animals from Antimicrobial preservative. Where applicable, determine the
the lethal effects ofthe subcutaneous injection ofthe quantity amount of antimicrobial preservative by a suitable chemical
of diphtheria toxin prescribed for this test. Allocate the six method. The amount is not less than 85.0 per cent and not
dilutions, one to each of the six groups of sixteen guinea- greater than 115.0 per cent of the intended amount.
pigs, and inject subcutaneously 1.0 ml of each dilution into
Sterility (2.2.11). Carry out test for sterility using 10 ml of
each guinea-pig in the groups to which that dilution is
bulk for each sterility medium.
allocated. After 28 days inject subcutaneously into each animal
in the six groups of sixteen, 1.0 ml of the challenge toxin
FINAL LOT
solution. Allocate the challenge toxin solution and the three
dilutions made from it, one to eachofthe four groups of four The final bulk vaccine is distributed aseptically into sterile,
guinea-pigs and inject subcutaneously 1.0 ml of each toxin tamper-proof containers. The containers are closed so as to
solution into each guinea-pig in the group to which that prevent contamination.
solution is allocated. Examine the guinea pigs twice in a day,
Only a final lot that is satisfactory with respect to each ofthe
remove dead animals and loll the animals showing definite
requirements given below under, Identification, Tests and
signs of diphtheria. Count the number ofsurviving animals 5
Assay may be released for use. Provided the tests for specific
days later and calculate the potency of the vaccine under
toxicity, free formaldehyde and antimicrobial preservative and
examination relative to the potency of the Standard
the assay have been carried out with satisfactory results on
preparation on the basis ofthe number ofanimals that survive
the final bulle vaccine, they may be omitted on the final lot.
in each ofthe six groups ofsixteen, using appropriate statistical
methods. Identification
The test is not valid unless (a) for the vaccine under
examination and the Standard preparation the 50.0 per cent Diphtheria toxoid is identified by a suitable immunochemical
protective doses lie between the largest and smallest doses method. The following method, applicable to certain vaccines,
ofthe preparations given to the guinea-pigs; (b) the survivors is given as an example. Dissolve in the vaccine under
among the four groups of guinea-pigs injected with the examination sufficient sodium citrate to give a 10 per cent
challenge toxin and its dilutions indicate that the challenge w/v solution. Maintain at 37° for about 16 hours and centrifuge
was approximately 100 LD50 and; (c) statistical analysis shows until a clear supernatant liquid is obtained. The clear
parallelism, linearity and a significant slope of the dose- supernatant liquid reacts with a suitable diphtheria antitoxin,
response lines. The test may be repeated any number oftimes giving a precipitate.
but when more than one test is performed the results of all
valid tests must be combined in the estimat~ of potency. Tests
Estimated potency should not be less than 30 ill per single Aluminium (2.3.9). Maximum 1.25 mgper single human dose,
human dose. If the lower limit of 95.0 per cent confidence if aluminium hydroxide or hydrated aluminium phosphate is
interval of estimated potency is less than 30 ill per single used as the absorbent.
human dose then the limits of the 95.0 per cent confidence
Free formaldehyde (2.3.20). Maximum 0.2 gil.
interval should be within 50 to 200 ofthe estimated potency.
Antimicrobial preservative. Where applicable, determine the
(c) Antibody induction method amount of antimicrobial preservative by a suitable chemical
Inject subcutaneously on each of two occasions separated method. The content is not less than 85.0 per cent and not
by an interval of not more than 4 weeks, one-fiftieth of the greater than 115.0 per cent of the intended amount.
stated human dose diluted to 1 ml with saline solution, into
Sterility (2.2.11). Complies with the test for sterility.
each of 10 normal, healthy guinea-pigs weighing between 250
and 350 g. Not earlier than 2 weeks and not later than 3 weeks Abnormal toxicity (2.2.1). Complies with the test for abnormal
after the second injection, collect the serum from each animal toxicity.
and determine the antitoxin content of the serum of each
pH (2.4.24). 6.0 to 7.0.
animal, as described under Diphtheria Antitoxin or any other
method approved by National Regulatory Authority. The Assay
geometric mean of the antitoxin contents shall be not less
than2.0 ITiilis permfw-lfureferel1cetofueDiphfuenaantitoXiii Carry -ouCone-ofllw- prescri15ecl metlfods-foY-tl:nnrssay-of
stannard. --- Diphtheria Vaccine (Adsorbed).
(d) Any other validated serological assay in guinea pigs or The lower confidence limit (P = 0.95) ofthe estimated potency
mice as approved by National Regulatory Authority is not less than 30 IV per single human dose.

2390

www.webofpharma.com
IP 2010 GAS-GANGRENE ANTITOXIN (OEDEMATIENS)

Labelling. The label states (I) the human dose; (2) the minimum Preparation and the potency of the preparation under
Lfunits per single human dose or the minimum International examination is then determined in relation to the Standard
Units per single human dose ifpotency test done by challenge preparation using the test toxin.
method; (3) the name and the amount of the adsorbent and
preservative; (4) that the vaccine must be shaken before use; Standard Preparation
(5) that the vaccine is not to be frozen.
The Standard Preparation is a dried hyperimmune horse serum
or another suitable preparation of gas-gangrene antitoxin
(oedematiens), the potency of which has been determined in
Gas-Gangrene Antitoxin relation to the International Standard. The Unit is the specific
(Oedematiens) neutralising activity for gas-gangrene toxin (oedematiens)
contained in such an amount of the Standard Preparation as
Gas-gangrene Antitoxin (Novyi); Anti-gas-gangrene the Ministry of Health and Family Welfare, Government of
(Oedematiens) Serum India may from time to time indicate as the quantity exactly
Gas-gangrene Antitoxin (Oedematiens) is a preparation equivalent to the Unit accepted for international use.
containing the specific antitoxic globulins obtained by
Suggested Method
purification ofhyperimmune serum ofhorses or other suitable
animals and having the specific activity of neutralising the Test animals. Use healthy mice having body weights such
alpha toxin formed by Clostric/ium oedematiens (c. novyi). that the difference between the lightest and heaviest is not
Gas-gangrene Antitoxin (Oedematiens) reconstituted where more than 5 g.
necessary as stated on the label complies with the following Test toxin. Prepare gas-gangrene toxin (oedematiens) by
tests. growing C. oedematiens in a liquid culture medium for about
Gas-Gangrene Antitoxin (Oedematiens) has a potency of not 5 days, filtering aseptically and precipitating with ammonium
less than 3750 Units per ml. sulphate. The resulting precipitate, which contains the toxin,
is collected, dried over phosphorus pentoxide at a pressure of
Description. A clear colourless or pale yellow liquid free from 1.5 to 2.5 kPa, powdered and kept my.
suspended particles ora freeze-dried, cream coloured powder
or pellet for reconstitution with the diluent supplied by the Selection oftest toxin. Select toxin for use as the test toxin by
manufacturer. determining the following quantities.
L+ dose - This is the smallest quantity of the toxin which,
Identification
when mixed with I Unit ofantitoxin and injected intramuscularly
Specifically neutralises and renders the alpha toxin formed by into mice, causes the death of the animals within 72 hours.
Cl. oedematiens harmless to susceptible animals or may be LDso - This is the smallest quantity of toxin which, when
identified by any other suitable in-vitro test. injected intramuscularly into mice, causes the death of half
the animals within 72 hours.
Tests
A suitable toxin is one which has an L+ dose in 0.5 mg or less
Potency. Carry out the biological assay of gas-gangrene
and contains not less than 25 LD so in an L+ dose.
antitoxin (oedematiens).
Determination of test dose of toxin (L+ dose). Prepare a
Biological Assay of Gas-Gangrene Antitoxin solution ofthe Standard Preparation with saline solution such
(Oedematiens) that 1.0 ml contains 12.5 Units. Weigh accurately a quantity of
the dried toxin and dissolve in saline solution so that 1.0 ml
The potency of the gas-gangrene antitoxin (oedematiens) is contains a precise known amount such as 10 mg. Prepare
determined by comparing the dose necessary to protect mice mixtures so that 2.0 ml of each mixture contains 0.8 ml ofthe
or other suitable animals against the lethal effects of gas- solution of the Standard Preparation (l0 Units) and one of a
gangrene toxin (oedematiens) with the dose of the Standard series of graded volumes of the solution of the test toxin.
Preparation of gas-gangrene antitoxin (C. oedematiens) Dilute each mixture with saline solution to the same final
necessary to give the same protection. For this purpose the volume (2.0 ml). Allow the mixtures to stand at room
Standard Preparation of gas-gangrene toxin (c. oedematiens) temperature, protected from light, for 60 minutes and, using
and a suitable preparation of gas-gangrene toxin six mice for each mixture, inject intramuscularly into each mouse
(oedematien),for use as a test toxin, are required. The test a dose of 0.2 ml of each mixture. Observe the mice thereafter
dose of the toxin is determined in the relation to the Standard for 72 hours.

2391

www.webofpharma.com
GAS-GANGRENE ANTITOXIN (OEDEMATIENS) IP 2010

The test dose of toxin is the amount present in 0.2 ml of that Description. A clear, colourless or pale yellow liquid free from
mixture which contains the smallest amount oftoxin sufficient suspended particles or a freeze- dried, cream coloured powder
to cause the death within 72 hours of all six mice injected or pellet for reconstitution with the diluent supplied by the
with it. manufacturer.
Determination ofpotency ofthe antitoxin. Dilute the test toxin
with saline solution so that 1.0 ml contains 12.5 times the test
Identification
dose. Prepare mixtures such that 2.0 rn1 ofeach mixture contains Specifically neutralises and renders the alpha toxin formed by
0.8 ml ofthe dilution ofthe toxin and one ofa series ofgraded G. perfringens harmless to susceptible animals or may be
volumes ofthe preparation under examination. Prepare further identified by any other suitable in-vitro test.
mixtures such that 2.0 ml ofeach contains 0.8 ml ofthe dilution
of the toxin and one of a series of graded volumes, of the Tests
Standard Preparation diluted with saline solution so that the
Potency. Carry out the biological assay of gas-gangrene
central graded volume contains 10 Units. Dilute each mixture
antitoxin (perfringens).
with saline solution to the same final volume (2.0 ml). Allow
the mixtures to stand at room temperature, protected from Biological Assay of Gas-Gangrene Antitoxin
light, for 60 minutes and then inject intramuscularly a dose of (Perfringens)
0.2 rn1 ofeach mixture into each ofsix mice under the conditions
described for determination ofthe test dose oftoxin. Observe The potency of the gas-gangrene antitoxin (perfringens) is
the mice for 72 hours. Ifnone ofthe mice is lulled, 0.2 ml ofthe determined by comparing the dose necessary to protect mice
mixture contains more than 1 Unit ofantitoxin. Ifall the mice or other suitable animals against the lethal effects of fixed
are lulled, 0.2 ml of the mixture contains less than 1 Unit of dose of the Standard Preparation of gas-gangrene toxin
antitoxin. (perfringens, type A) with the dose ofthe Standard Preparation
The mixture that contains the largest volume ofthe preparation ofgas-gangrene antitoxin (perfringens) (G. perfringens alpha
under examination that fails to protect the mice from death antitoxin) necessary to give the same protection. For this
contains 10 Units. The test is not valid unless all the mice purpose the Standard Preparation of gas-gangrene antitoxin
injected with mixtures containing 0.8 ml or less ofthe solution (G. perfringens alpha antitoxin) and a suitable preparation of
of the Standard Preparation die and all those injected with gas-gangrene toxin (perfringens, j:ype A),for use as a test toxin,
mixtures containing more survive. are required. The test dose of the toxin is determined in the
relation to the Standard Preparation and the potency of the
Other tests. Complies with the tests stated under Antisera.
preparation under examination is then determined in relation
Storage. Store protected from light at a temperature between to the Standard preparation using the test toxin.
2° to 8°. Liquid preparations should not be allowed to freeze.
Labelling. The label states (1) the number of Units per ml, Standard Preparation
where appropriate; (2) the animal species from which the
The· Standard Preparation is a dried 1:3 dilution of
preparation has been made; (3) the recommended dose; (4)
hyperimmune horse serum or another suitable preparation of
the name and concentration ofany antimicrobial preservative
gas-gangrene antitoxin (perfringens), the potency of which
added; (5) that in case of a liquid preparation, it should not to
has been determined in relation to the International Standard.
be allowed to freeze.
The Unit is the specific neutralising activity for gas-gangrene
toxin (perfringens) contained in such an amount of the
Gas-Gangrene Antitoxin (Perfringens) Standard Preparation as the Ministry of Health and Family
Welfare, Government ofIndia may from time to time indicate
Anti-gas-gangrene (perfringens) Serum as the quantity exactly equivalent to the Unit accepted for
Gas-gangrene Antitoxin (Perfringens) is a preparation international use.
containing the specific antitoxic globulins obtained by
purification ofhyperimmune serum ofhorses or other suitable Suggested Method
animals and having the specific activity of neutralising the
Test animals. Use healthy mice having body weights such
alpha toxin formed by Clostridium perfringens.
that the difference between the lightest and heaviest is not
Qal':g al1gr l:ll1 e .Antito)(il1(.(>l:lrfril1gl:l.l1s1rl:lC:Q}!~ti1:l11:l:lcl.""hl:lre more than Sg.
necessary as stated on the label complies with the following
tests. Test toxin. Prepare gas-gangrene toxin (perfringens) by
Gas-Gangrene Antitoxin (perfringens) has a potency of not growing G. perfringens, j:ype A, in a liquid culture medium for
less than 1500 Units per ml. about 5 days, filtering aseptically and precipitating with

2392

www.webofpharma.com
IP 2010 GAS-GANGRENE ANTITOXIN (SEPTICUM)

ammonium sulphate. The resulting precipitate, which contains injected with mixtui'es containing 2.0 ml or less ofthe solution
the toxin, is collected, dried over phosphoruspentoxide at a of the Standard Preparation die and all those injected with
pressure of 1.5 to 2.5 kPa,powdered and kept dry. mixtures containing more survive.
Selection oftest toxin. Select toxin for use as the test toxin by Other tests. Complies with the tests stated under Antisera.
detennining the following quantities.
Storage. Store protected from light at a temperature between
L+ dose - This is the smallest quantity of the toxin which, 2° to 8°. Liquid preparations should not be allowed to freeze.
when mixed with 1 Unit ofantitoxin and injected intravenously
Labelling. The label states (1) the number of Units per ml,
into mice, causes the death of the animals within 48 hours.
where appropriate; (2) the animal species from which the
LD jO - This is the smallest quantity of toxin which, when preparation has been made; (3) the recommended dose; (4)
injected intravenously into mice, causes the death of half the the name and concentration ofany antimicrobial preservative
animals within 48 hours. added; (5) that in case ofa liquid preparation, it should not be
allowed to freeze.
A suitable toxin is one which has an L+ dose in 4 mg or less,
and contains not less than 20 LD so in an L+ dose.
Determination of test dose of toxin (L+dose). Prepare a
solution ofthe Standard Preparation with saline solution such Gas-Gangrene Antitoxin (Septicum)
that 1.0 ml contains 5 Units. Weigh accurately a quantity of
the dried toxin and dissolve it in saline solution so that 1.0 ml Anti-gas-gangrene (Septicum) Serum
contains a precise known amount such as 10 mg. Prepare Gas-gangrene Antitoxin (Septicum) is a preparation containing
mixtures such that 5.0 ml ofeach mixture contains 2.0 ml ofthe the specific antitoxic globulins obtained by purification of
solution of the Standard Preparation (10 Units) and one of a hyperimmune serum of horses or other suitable animals and
series of graded volumes of the solution of the toxin. Dilute having the specific activity of neutralising the alpha toxin
each mixture with saline solution to the same final volume (5.0 fonned by Clostridium septicum.
mI). Allow the mixtures to stand at room temperature, protected Gas-gangrene Antitoxin (Septicum) reconstituted where
from light, for 60 minutes and, using six mice for each mixture, necessary as stated on the label complies with the following
inject intravenously into each mouse a dose of 0.5 ml of each tests.
mixture. Observe the mice thereafter for48 hours.
Gas-gangrene Antitoxin (Septicum) has a potency of not less
The test dose of toxin is the amount present in 0.5 mlofthat than 1500 Units perml.
mixture which contains the smallest amount oftoxin sufficient
Description. A clear, colourless or pale yellow liquid free from
to cause the death within 48 hours of all six mice injected
suspended particles or a freeze-dried, cream coloured powder
. with it.
or pellet for reconstitution with the diluent supplied by the
Determination ofpotency ofthe antitoxin. Dilute the test toxin manufacturer.
with saline solution so that 1.0 ml contains five times the test
dose. Prepare mixtures such that 5.0 ml ofeach mixture contains Identification
2.0 ml ofthe dilution ofthe toxin and one ofa series ofgraded Specifically neutralises and renders the alpha toxin fonned by
volumes ofthe preparation under examination. Prepare further C. septicum harmless to susceptible animals or may be
mixtures such that 5.0 ml ofeach contains 2.0 ml ofthe dilution identified by any other suitable in-vitro test.
of the toxin and one of a series of graded volumes of the
Standard Preparation diluted with saline solution so that the Tests
central graded volume contains 10 Units. Dilute each mixture
with saline solution to the same final volume (5.0 ml). Allow Potency. Carry out the biological assay of gas-gangrene
the mixtures to stand at room temperature, protected from antitoxin (septicum).
light, for 60 minutes and then inject intravenously a dose of
0.5 ml ofeach mixture into each ofsix mice under the conditions
Biological Assay of Gas-Gangrene Antitoxin
described for detennination ofthe test dose oftoxin. Observe (Septicum)
the mice for 48 hours. Ifnone ofthe mice is killed, 0.5 ml ofthe The potency of the gas-gangrene antitoxin (septicum) is
mixture contains more than 1 Unit of antitoxin. Ifall mice are determined by comparing the dose necessary to protect mice
killed, 0.5 ml ofthe mixture contains less than 1 Unit ofantitoxin. or other suitable animals against the lethal effects of gas-
The mixture that contains the largest volume ofthe preparation gangrene toxin (septicum) with the dose of the Standard
under examination that fails to protect the mice from death Preparation of gas-gangrene antitoxin (c. septicum)
contains 10 Units. The test is not valid unless all the mice necessary to give the same protection. For this purpose the

2393

www.webofpharma.com
GAS-GANGRENE ANTITOXIN (SEPTICUM) IP 2010

Standard Preparation of gas-gangrene antitoxin (c. septicum) from light, for 60 minutes and, using six mice for each mixture,
and a suitable preparation of gas-gangrene toxin (septicum), inject intravenously intoeach mouse a dose of 0.5 ml of each
for use as a test toxin, are required. The test dose ofthe toxin mixture. Observe the mice thereafter for 72 hoUrs.
is determined in the relation to the Standard Preparation and
The test dose of toxin is the amount present in 0.5 ml of that
the potency of the preparation under examination is then
mixture which contains the smallest amount oftoxin sufficient
determined in relation to the Standard preparation using the
to cause the death within 72 hours of all six mice injected
test toxin.
with it.
Standard Preparation Determination ofpotency ofthe antitoxin. Dilute the test toxin
The Standard Preparation is a dried 1: 3 dilution of with saline solution so that 1.0 ml contains five times the test
hyperimmune horse serum of gas-gangrene antitoxin dose. Prepare mixtures such that 5.0 ml ofeach mixture contains
(septicum) in phosphate-buffered saline or another suitable 2.0 ml ofthe dilution ofthe toxin and one ofa series ofgraded
preparation the potency of which has been determined in volumes ofthe preparation under examination. Prepare further
relation to the International Standard. The Unit is the specific mixtures such that 5.0 ml ofeach contains 2.0 ml ofthe dilution
neutralising activity for gas-gangrene toxin (septicum) of the toxin and one of a series of graded volumes of the
contained in such an amount of the Standard Preparation as Standard Preparation diluted with,saline solution so that the
the Ministry of Health and Family Welfare, Government of central graded volume contains 10 Units. Dilute each mixture
India may from time to time indicate as the quantity exactly with saline solution to the same final volume (5.0 mI). Allow
equivalent to the Unit accepted for international use. the mixtures to stand at room temperature, protected from
light, for 60 minutes and then inject intravenously a dose of
Suggested Method 0.5 ml ofeach mixture into each ofsix mice under the conditions
described for determination ofthe test dose of toxin. Observe
Test animals. Use healthy mice having body weights such themice for 48 hours. Ifnone ofthe mice is killed, 0.5 mlofthe
that the difference between the lightest and heaviest is not mixture contains more than 1 Unit of antitoxin. If all the mice
more than 5 g. are killed, 0.5 ml of the mixture contains less than 1 Unit of
Test toxin. Prepare gas-gangrene toxin (septicum) by growing antitoxin.
C. septicum (Vibrion septique) in a liquid culture medium for The mixture that contains the largest volume ofthe preparation
about 5 days, filtering aseptically and precipitating with under examination that fails to protect the mice from death
ammonium sulphate. The resulting precipitate, which contains contains 10 Units. The test is not valid unless all the mice
the toxin, is collected, dried over phosphorus pentoxide at a injected with mixtures containing 2.0 ml or lessofthe solution
pressure of 1.5 to 2.5 kPa, powdered and kept dry. of the Standard Preparation die and all those injected with
Selection oftest toxin. Select toxin for use as the test toxin by mixtures containing more survive.
detennining the following quantities. Other tests. Complies with the tests stated under Antisera.
L+ dose - This is the smallest quantity oftoxin which, when Storage. Store protected from light at a temperature between
mixed with 1 Unit ofantitoxin and injected intravenously into 2° to 8°. Liquid preparations should not be allowed to freeze.
mice, causes the death of the animals within 72 hours.
Labelling. The label states (1) the number of Units per ml,
LDso - This is the smallest quantity of the toxin which, when where appropriate; (2) the animal species from which the
injected intravenously into mice, causes the death of half the preparation has been made; (3) the recommended dose; (4)
animals within 72 hours. the name and concentration of any antimicrobial preservative
A suitable toxin is one which has an L+ dose in 0.5 mg or less added; (5) that in case of a liquid preparation, it should not to
and contains not less than 25 LD so in an L+ dose. be allowed to freeze.

Determination of test dose of toxin (L+dose). Prepare a


solution of the Standard Preparation in saline solution such
that 1.0 ml contains 5 Units. Weigh accurately a quantity of Mixed Gas-Gangrene Antitoxin
the dried toxin and dissolve in saline solution so that 1.0 ml
contains a precise known amount such as 20 mg. Prepare Mixed Gas-gangrene Antitoxin is prepared by mixing Gas-
mixtures suchthat-5.0-ml-ofeachmixturecontains 2.0-mlofthe gangrene Antitoxin (Oedematiens), Gas-gangrene Anti-toxin
solution ofthe Standard Preparation (1 0 Units) and one of a (Perfringens) and Gas=gangrene AntItoxin·· (Septicl.lrnfin
series of graded volumes of the solution of the toxin. Dilute appropriate quantities.
each mixture with saline solution to the same final volume (5.0 Mixed Gas-gangrene Antitoxin has a potency of not less than
rnl). Allowthe mixtures to stand at room temperature, protected 1000 Units per ml ofGas-gangrene Antitoxin (Oedematiens),

2394

www.webofpharma.com
IP 2010 HAEMOPHILUS TYPE b CONJUGATE VACCINE

not less than 1000 Units per ml of Gas-gangrene Antitoxin the stability testing, release requirements are set for these
(Perfringens) and not less than 500 Units per ml of Gas- indicator tests to ensure that the vaccine will be satisfactory
gangrene Antitoxin (Septicum). at the end of the period of validity.
Description. An almost colourless or very faintly yellow liquid,
free from turbidity. SEED LOT
The strain of H. inflllenzae type b used in preparing
Identification Haemophilus type b conjugate vaccine shall be identified by
Specifically neutralises and renders the alpha-toxins formed a record of its history, including the source from which it was
by C. oedematiens, C. perfringens and C. septiclim harmless obtained and the tests made to determine the characteristics
to susceptible animals. of the strain. The strain shall have been shown to be capable
of producing Type b polysaccharide.
Tests
The production of PRP and of the canier protein is based on
Potency. Carry out the biological assay for each component defined seed lot systems. Master seed lot and working seed
as described in the relevant individual monographs. lot shall be properly characterized and defined. Cultures
Other tests. Complies with the tests stated under Antisera. derived from the working seed shall have the same
characteristics as ofthe master seed lot. The sample ofculture
Storage. Store protected from light at a temperature 2° to 8°. It of single harvests taken before killing shall be tested for
should not be allowed to freeze. Liquid preparations should contamination by examination ofGram-stained smears and by
not be allowed to freeze. inoculation on suitable media.
Labelling. The label states the number of Units of each
component per m!. H. Injlllenzae Type b Polysaccharide (pRP)
H. inflllenzae Type b is grown in a liquid medium that does
not contain high-molecular-weight polysaccharides; if any
ingredient of the medium contains blood-group substances,
Haemophilus Type b Conjugate the process shall be validated to demonstrate that after the
Vaccine purification step they are no longer detectable. The culture
may be inactivated. PRP is separated from the culture liquid
Haemophilus Type b Conjugate Vaccine is a liquid or freeze-
and purified by a suitable method. Volatile matter, including
dried preparation ofa polysaccharide, derived from a suitable
water, In the purified polysaccharide is determined by methods
strain of Haemophillis inflllenzae type b, covalently bound
such as thermogravimetry, Karl Fischer or any other suitable
to a carrier protein. The polysaccharide, polyribosylribitol
method. All chemical analysis shall be based on the dry weight
phosphate, referred to as PRP, is a linear copolymer composed
of the polysaccharide, in its salt form.
of repeated units of 3-~-D-ribofuronosyl-(l-71)-ribitol-5-
phosphate [(C IOH tPtl)Il]' with a defined molecular size. The Only those pools of PRP that comply with the following
carrier protein, when conjugated to PRP, is capable ofinducing requirements may be used in the preparation ofthe conjugate.
a T-cell-dependent B-cell immune response to the The partially purified PRP shall be stored frozen at or
polysaccharide. below -20°.

Production Identification
General provisions The PRP is identified by an immunochemical method (2.2,14)
or other suitable method (e.g. IH or 13C NMR spectroscopy).
The production method shall have been shown to yield
consistently H. inflllenzae tYpe b conjugate vaccines of Molecular size. The percentage ofPRP eluted before a given
adequate safety and immunogenicity in humans. The K o value or within a range of K o values, is determined by gel
production method is validated to demonstrate that the product, filtration or high performance size-exclusion chromatography
iftested, would comply with the test for safety and efficacy of (HPSEC) (2.4.16), either alone or in combination with light
Vaccines. The stability of the final lot and relevant scattering and refractive index detectors (e.g. multiple angle
intermediates is evaluated using one or more indicator tests. laser light scattering i.e. MALLS) or any other suitable method.
Such tests may include determination of molecular size, An acceptable value is established for the particular product
determination of free PRP in the conjugate and the and each batch of PRP must be shown to comply with this
immunogenicity test on mice. Taking account ofthe results of limit. Limits for currently approved products, using the

2395

www.webofpharma.com
HAEMOPHILUS TYPE b CONJUGATE VACCINE IP 2010

Table 1 - Specifications for different components ofHaemophilus Type b ConjugateVaccine.

Polysaccharide Carrier Protein Conjugation


TypeofPRP Nominal Type Purity Nominal Coupling Procedure
amount amount method
per dose per dose
Polysaccharide 25 flg Diphtheria >1500 Lfpermg 18 flg Cyanogen Activated diphtheria
(size reduced) toxoid of protein nitrogen bromide toxoid (D-AH+),
K o 60;0 per cent: activation cyanogen bromide
0.6-0.7 ofPRP activated PRP
Polysaccharide: 10flg Tetanus >1500 Lfpermg 20flg Carbodiimide- ADH-activated PRP
PRP 2': 50.0 per toxoid of protein nitrogen mediated (pRP-cov.-AH) +
cent :::Ko: 0.30 coupling tetanus toxoid +
EDAC
Polysaccharide 10 flg CRM197 >90.0 per cent 25flg Reductive Direct coupling of
(size reduced) diphtheria diphtheria amination one- PRP to CRM 197
Dp=15-35 or 10-35 protein protein (step method) or (cyanoboro-hydride
N-hydroxy~ activated)
succinimide
activation
Polysaccharide 15 flg Meningococcal Outer membrane 125 or Thioether PRP activation by
(size-reduced) group B outer protein vesicles 250 flg bond CDIPRP-IM+
K o°.3-0.6 membrane <8.0 per cent of BuA2+ BrAc=
protein (OMP) lipopolysaccharide PRP-BuA2-BrAc+
OMP

Abbreviations:
ADH adipic acid dihydrazide Dp degree ofpolymerization
BrAc bromoacetyl chloride EDAC l-ethyl-3-(3-dimethylaminopropyl) carbodimide
BuA2 butane-l, 4-diarnide 1M imidazoliuill
CDl carbonyldi-imidazole Mw weight-average molecular weight.

Table 2 - Requirements on bulle conjugate

Test Protein Carrier


SpecificatiOlis Diphtheria toxoid Tetanus toxoid CRM 197 OMP
Free Polysaccharide (PRP) <37.0 per cent <20.0 per cent <25.0 per cent <15.0 per cent
Unbound protein <5.0 per cent, <1.0 per cent, <1.0 per cent or Not applicable
where applicable where applicable <2.0 per cent,
depending oh the
coupling method
PRP to protein ratio 1.25-1.75 0.30-0.55 0.3-0.7 0.05-0.1
Molecular size (Ko): Cross- 95.0 per cent <0.75 60.0 per cent <0.2 50.0 per cent 85.0 per cent < 0.25

chromatography R
Cross-linked agarose for 0.6-0.7 85.0 per cent <0.5
chromatography Rl

2396

www.webofpharma.com
IP 2010 HAEMOPHILUS TYPE b CONJUGATE VACCINE

indicated stationary phases, are shown for information in Diphtheria toxoid. Diphtheria toxoid is produced as stated
Tables 1 and 2. Where applicable, the molecular-size under Diphtheria Vaccine (Adsorbed) and complies with the
distribution is also determined after chemical modification of requirementsprescribed there for bulk purified toxoid.
the polysaccharide. Tetanus toxoid. Tetanus toxoid is produced as stated under
A validated determination of the degree ofpolymerization or Tetanus Vaccine (Adsorbed) and complies with the
ofthe weight-average molecular weight and the dispersion of requirements prescribed there for bulle purified toxoid except
molecular masses may be used instead ofthe determination of that the antigenic purity is not less than 1500 Lf per mg of
molecular size distribution. protein nitrogen.
Ribose (2.7.1). Not less than 32.0 per cent, calculated with Diphtheria protein CRM 197. Suitable tests are carried out,
reference to the dried substance, as estimated by Bial reaction for validation or routinely, to demonstrate that the product is
for pentose, using D-ribose as a standard or any other suitable non-toxic. The protein obtained contains not less than 90.0
assay. per cent of diphtheria CRM 197 protein, when prepared by
Phosphorus (2.7.1). 6.8 per cent to 9.0 per cent, calculated liquid chromatography (2.4.14) or any other suitable method.
with reference to the dried substance. The carrier protein shall be characterized by a suitable chemical
or physicochemical method like SDS-PAGE, HPLC, isoelectric
Protein (2.3.49). Not more than 1.0 per cent, calculated with focusing, amino acid sequencing, circular dichroism,
reference to the dried substance. Use sufficient PRP to allow fluorescence spectroscopy, peptide mapping or mass
detection of 1 per cent ofprotein (e.g. a minimum of 1 mg of spectroscopy, as appropriate.
PRP).
Nucleic acid (2.7.1). Not more than 1.0 per cent, calculated OMP (Meningococcal group B outer membrane protein
with reference to the dried substance by spectroscopy or any complex)
other suitable method.
aMP complex of Neisseria meningitidis complies with the
Bacterial endotoxins (2.2.3 ). Not more than 25 ill ofendotoxin following requirements for lipopolysaccharide and pyrogens.
per microgram ofPRP.
Lipopolysaccharide. Not more than 8.0 per cent of
Residual reagents. Where applicable, tests are carried out to lipopolysaccharide, determined by a suitable method.
determine residues of reagents used during inactivation and
Pyrogens (2.2.8). Inject into each rabbit 0.25 jJ.g ofaMP per kg
purification. An acceptable value for each reagent is
body weight, for determining the pyrogenic effect.
established for the particular product and each batch of PRP
must be shown to comply with this limit. Where validation Bulk conjugate
studies have demonstrated removal of a residual reagent, the
test on PRP may be omitted. PRP is chemically.modified to enable conjugation; it is usually
partly depolymerised either before or during this procedure.
Carrier protein Reactive functional groups or spacers may be introduced into
the carrier protein or PRP prior to conjugation. The conjugate
The carrier protein is chosen in a way so that when the PRP is is obtained by the covalent binding ofPRP and carrier protein.
conjugated it is able to induce a T-cell-dependent immune Where applicable, unreacted but potentially reactogenic
response. Currently approved carrier proteins and coupling functional groups are made unreactive by means of capping
methods are listed for information in Table 1. The carrier agents; the conjugate is purified to remove reagents. Where
proteins are produced by culture of suitable microorganisms; validation studies have demonstrated removal of a residual
the bacterial purity of the culture is verified; the culture may reagent (eg. CN, Br etc.), the test on bulle conjugate may be
be inactivated; the carrier protein is purified by a suitable omitted.
method.
Only a bulk conjugate that complies with the following
Only a carrier protein that complies with the following requirements may be used in preparation of the final bulk
requirements may be used in preparation of the conjugate. vaccine. For each test and for each particular product, limits
of acceptance are established and each batch of conjugate
Identification
must be shown to comply with these limits. Limits applied to
The carrier protein is identified by a suitable immunochemical cunently approved products for some of these tests are listed
method (2.2.14).. for information in Table 2.
Sterility (2.2.11). Carry out the test for sterility using for each PRP. The PRP content is determined by assay ofphosphorus
medium 10 ml or the equivalent of one hundred doses, (2.7.1) or by assay ofribose (2.7.1) or by an immunochemical
whichever is less. method (2.2.14) or by any suitable method.

2397

www.webofpharma.com
HAEMOPHJLUS TYPE b CONJUGATE VACCINE IP 2010

Protein (2.7.1). The protein content is determined by a suitable Identification


chemical method.
The vapcine is identified by a suitable irnmunochernical method
PRP to protein ratio. Determine the ratio by calculation. (2.2.14) for PRP.
Molecular size. Molecular-size distribution is determined by Tests
gel filtration or size-exclusion chromatography (2.4.16), using
a gel matrix, appropriate to the expected size ofthe conjugate. Sterility (2.2.11). Complies with the test for sterility.
Free PRP. Unbound PRP is determined after removal of the Abnormal toxicity (2.2.1). Complies with the test for abnormal
conjugate, for example by size-exclusion or hydrophobic toxicity.
chromatography (2.4.16), ultra-filtration or other validated Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
methods. per kg ofthe.rabbit's mass a quantity ofthe vaccine equivalent
Free carrier protein. Free carrier protein is determined by a to 1 Ilg ofPRP for a vaccine with diphtheria toxoid or CRM 197
suitable method (which may include deriving the content by diphtheria protein as carrier; O.lllg ofPRP for a vaccine with
calculation from the results of other tests). The amount is tetanus toxoid as carrier; 0.025 Ilg ofPRP for a vaccine with
within the limits approved for the particular product. OMP as carrier.
Unreacted functional groups. No unreacted functional groups pH (2.4.24). The pH ofthe vaccine, reconstituted ifnecessary,
are detectable in the bulk conjugate unless process validation is within the range approved for the product.
has shown that unreacted functional groups detectable at Aluminium (2.3.9). Not more than 1.25 mg per single human
this stage are removed during the subsequent manufacturing dose. When aluminium hydroxide or hydrated aluminium
process (for example, owing to short half"life); phosphate is tised as the -adsorbent;
Residual reagents. Removal of residual reagents such as Antimicrobial preservative. Where applicable, determine the
cyanide, EDAC (ethyldimethylaminopropylcarbodimide) and amount of antimicrobial preservative by a suitable chemical
phenol is confirmed by suitable tests or by validation of the method. The content is not less than 85 per cent and is not
purification process. greater than 115.0 per cent of that stated on the label.
Sterility (2.2.11). Carry out the tests for sterility using for Water (2.3.43). Not more than 3.0 per cent.
each medium 10 ml or the equivalent of one hundred doses, PRP. Not less than 80.0 per cent and not greater than 120.0
whichever is less. per cent of the amount of PRP stated on the label as
determined by ribose assay (2.7.1) or by phosphorus assay
FINAL BULK VACCINE (2.7.1) or by an immunochemical method (2.2.14) or by any
other. suitable method like colorimetery or by anion exchange
An adjuvant, an antimicrobial preservative and a stabilizer
liquid chromatography (2.4.14) with pulsed amperometric
may be added to the bulle conjugate before dilution to the final
detection.
concentration with a suitable diluent.
Free PRP. Unbound PRP is determined after removal of the
Only a final bulk vaccine that complies with the following
conjugate for example by size-exclusion or hydrophobic
requirements may be used in preparation ofthe final lot.
chromatography (2.4.16), ultra filtration or other validated
Antimicrobial preservative. Where applicable, determine the methods.
amount of antimicrobial preservative by a suitable chemical
Labelling. The label states (l) the number of micrograms of
method. The content is not less than 85.0 per cent and not
PRP per human dose; (2) the type·and nominal amount of
greater than 115.0 per cent ofthe intended amount.
carrier protein per single human dose; (3) for vaccine contained
Sterility (2.2.11). Carry out test for sterility using 10 ml of in single-dose containers where the space is too small to
bulle for each sterility medium. accommodate the full name of the vaccine the abbreviation
'Hib' may be used in the label on the container provided that
FINAL LOT the same code is also stated in the label on the package.
The final lots are filled in suitable containers, under stringent
aseptic conditions. Hepatitis A (Inactivated) and Hepatitis
Qply a filla11Qt tl1fl.t i!i !ill,ti!ifll,c:tQry witl1.!~!ill.el.c:t tO~fl.c:l!.Qft!lt;: B (rDNA) Vaccine (Adsorbed)
requirements given under Identification, Tests and Assay may
be released for use. Provided the test for antimicrobial Hepatitis A (Inactivated) and Hepatitis B (rDNA) Vaccine
preservative has been carried out on the final bulk vaccine, it (Adsorbed) is a suspension consisting of a suitable strain of
may be omitted on the final lot. hepatitis A virus, grown in cell cultures and inactivated by a

2398

www.webofpharma.com
IP 2010 HEPATITIS B VACCINE (rDNA)

validated method, and ofhepatitis B surface antigen (HBsAg), carried out with satisfactory results on the final bulle vaccine,
a component protein of hepatitis B virus obtained by it may be omitted on the final lot.
recombinant DNA technology; the antigens are adsorbed on
Identification
a mineral carrier such as aluminium hydroxide or hydrated
aluminium phosphate. The vaccine is shown to contain hepatitis A virus antigen and
hepatitis B surface antigen by suitable immunochemical
Production methods (2.2.14), using specific antibodies or by the mouse
General provisions immunogenicity tests described under assay.

The two components are prepared as described in the Tests


monographs on Hepatitis A Vaccine (Inactivated, Adsorbed)
Aluminium (2.3 .9). Maximum 1.25 mg per single human dose
and Hepatitis B Vaccine (rDNA) and comply with the
if aluminium hydroxide or hydrated aluminium phosphate is
requirements prescribed therein. The production method is
used as the adsorbent.
validated to demonstrate that the product, if tested, would
comply with the test for abnormal toxicity for antisera and Free formaldehyde (2.3.20). Maximum 0.2 gil.
vaccines. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Reference preparation
method. The content is not less than 85.0 per cent and not
The reference preparation is part of a representative batch greater than 115.0 per cent of the intended amount.
shown to be at least as immunogenic in animals as a batch
that, in clinical studies in young, healthy adults, produces not Sterility (2.2.11). Complies with the test for sterility.
less than 95.0 per cent seroconversion, corresponding to a Bacterial endotoxins (2.2.3). Less than 2 IV per human dose.
level of neutralizing antibody recognized to be protective,
after a full-course primary immunization. For hepatitis A, an
Assay
antibody level not less than 20 mIU/ml determined by enzyme- Hepatitis A component
linked immunosorbent assay is recognized as being protective.
For hepatitis B, antibody level not less than 10 mill/ml against Complies with the assay as stated under Inactivated Hepatitis
HBsAg is recognized as being protective. A Vaccine (Adsorbed).

Hepatitis B component
FINAL BULK VACCINE
Complies with the-assay as stated under Hepatitis B Vaccine
The final bulk vaccine is prepared from one or more inactivated
(rDNA).
harvests of hepatitis A virus and one or more batches of
purified antigen ofHepatitis B (rDNA). Labelling.The label states (1) the amount ofhepatitis A virus
antigen and hepatitis B surface antigen per container; (2) the
Only a final bulk vaccine that complies with the following type of cells used for production of the vaccine; (3) the name
requirements may be used in the preparation ofthe final lot. and amount of the adsorbent used; (4) that the vaccine must
Antimicrobial preservative. Where applicable, determine the be shaken before use; (5) that the vaccine must not be frozen.
amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount. Hepatitis B Vaccine (rDNA)
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk Hepatitis B Vaccine (rDNA) is a non-infectious preparation
for each sterility medium. containing the purified major surface antigen of Hepatitis B
FINAL LOT virus (HBsAg). This preparation is white or almost white
translucent liquid in which the mineral carrier tends to settle
Only a final lot that complies with each of the requirements down slowly on keeping but is free from foreign' particles/
given below under Identification, Tests and Assay may be floccules.
released for use. Provided that the tests for free formaldehyde
(where applicable) and antimicrobial preservative content Production
(where applicable) have been carried out on the final bulle General provisions
vaccine with satisfactory results, they may be omitted on the
final lot. If the assay of the hepatitis A and/or the hepatitis B The antigen is manufactured by recombinant DNA technology
component is carried out in vivo, then provided it has been by culturing genetically engineered yeast cells or other suitable

2399

www.webofpharma.com
HEPATITIS B VACCINE (rDNA) IP 2010

cell lines which carry the gene that codes for major surface Total protein (2.3 .49). The total protein is determined by a
antigen ofthe Hepatitis-B virus as approved by the competent validated method. The content is within the limits approved
authority. Several physico-chemical steps are employed to for the specific product.
purify the Hepatitis-B surface antigen (HBsAg). The vaccine Antigen content and identification. The quantity and
may contain the product of the S gene (major protein), a specificity of HBsAg is determined in comparison with the
combination of the S gene and pre-S2 gene products (middle International standard for HBsAg subtype ad or an in-house
protein) or a combination ofS gene, the pre-S2 gene, and pre- reference, by a suitable immunochemical method such as
SI gene products (large protein). The purity of the antigen is radioimmunoassay (RIA), enzyme-linked immunosorbent
determined by comparison with a reference preparation using assay (ELISA), immunoblot (preferably using a monoclonal
liquid chromatography or other suitable methods such as SDS- antibody directed against a protective epitope) or single radial
PAGE with any suitable staining method. The purified antigen diffusion. The antigen/protein ratio is within the limits approved
is finally adsorbed on aluminium hydroxide or aluminium for the specific product.
phosphate.
The molecular weight of the major band in a sodium dodecyl
The method used for production of the vaccine must have sulphate polyacrylamide gel electrophoresis (SDS-PAGE)
been shown to yield a product consistently complying with under reduced conditions corresponds to the value expected
the requirements for immunogenicity and safety. It must also from the gene sequence and possible glycosylation.
have been shown to induce specific, protective antibodies in
Antigenic purity. The purity of the antigen is determined by
human beings.
comparison with a refereIice preparation using liquid
The production method must be validated to demonstrate chromatography or other suitable methods such as SDS-PAGE
that the product if tested; would· comply with the tests for with staining. Asuitable method is sensitive enough to detect
safety and efficacy. a potential contaminant at a concentration of 1.0 per cent of
Reference preparation. A part of a batch shown to be at least total protein. Not less than 95.0 per cent of the total protein
as immunogenic as a batch that was used in clinical studies consists of hepatitis B surface antigen.
and approved by National Regulatory Authority and Composition. The content of proteins, lipids, nucleic acids
detennined by any suitable method. For hepatitis B, antibody and carbohydrates is detelmined.
level not less than 10 mIDIml against HBsAg is recognized as Host-cell and vector-derived DNA (2.2.15). Ifmammalian cells
being protective.. are used for production, not more than 10 pg of DNA in the
quantity ofpurified antigen equivalent to a single human dose
Characterisation ofthe substance of vaccine.
Development studies are carried out to characterize the Caesium. If a caesium salt is used during production, a test
antigen. The complete protein, lipid and carbohydrate structure for residual caesium is carried out on the purified antigen. The
ofthe antigen is established. The morphological characteristics content is within the limits approved for the specific product.
ofthe antigen particles are established by electron microscopy.
Sterility (2.2.11). The purified antigen complies with the test
The buoyant density oftheantigen particles is determined by
for sterility, carried out using 10 ml for each medium.
a physico-chemical method, for example gradient
centrifugation. The antigenic epitopes are characterized. The Additional tests on the purified antigen may be required
protein fraction ofthe antigen is characterized in terms ofthe depending on the production method used: for example, a test
primary stmcture (for example, by detennination ofthe amino- for residual animal semm where mammalian cells are used for
acid composition, by partial amino-acid sequence analysis). production or tests for residual chemicals used during
extraction and purification.
PROPAGATION AND HARVEST
FINAL BULK VACCINE
Identity, microbial purity, plasmid retention and consistency
of yield are determined at suitable production stages. If An antimicrobial preservative and an adjuvant may be
mammalian cells are used, tests for extraneous agents and included in the vaccine.
mycoplasmas are performed in accordance with tests for Only a final bulle vaccine that complies with the following
extraneous agents in viral vaccines for human use. requirements may be used in the preparation of the final lot.
Antimicrobial preservative. Where applicable, detennine the
PURIFIED ANTIGEN
amount of antimicrobial preservative by a suitable chemical
Only a purified antigen that complies with the following method. The content is not less than 85.0 per cent and not
requirements may be used in the preparation ofthe final bulle. greater than 115.0 per cent. of the intended amount.

2400

www.webofpharma.com
IP 2010 HEPATITIS B VACCINE (rDNA)

Sterility (2.2.11). Carty out test for sterility using 10 ml ofbulk old) that have not been previously treated with any material
for each sterility medium. that will interfere with the test will also be suitable for the test.
Use animals ofthe same sex in the test. Inject similar groups of
FINAL LOT
animals with the reference preparation ofHepatitis-B vaccine
Only a final lot that complies with each of the requirements (r DNA). One group of control animals remains unvaccinated
given below under Identification, Tests and Assay may be but is injected intraperitoneally with the same volume of the
released for use. Provided that the tests for free formaldehyde, diluent alone. Anaesthetize and bleed the animals 28 to 42
antimicrobial preservative content and the assay in animals, days later, keeping the individual sera separate. Assay the
where applicable, have been carried out on the final bulle individual sera for specific HBsAg antibody concentration
vaccine with satisfactory results, they may be omitted on the by a suitable immunochemical method such as ELISA or RIA.
final lot. The estimate ofpotency shall be expected in terms of Calculate the result of the assay by standard statistical
Ilg/ml i.e not less than 20 Ilg/ml. methods (5.7). From the distribution of reaction levels
Identification measured on all the sera in the unvaccinated (control group),
the maximum reaction level that can be expected to occur in an
The assay or, where applicable, the electrophoretic profile, unvaccinated animal for that particular assay is determined.
also serves to identify the vaccine. Any response in the vaccinated animals that exceeds this
level is by definition seroconversion. The percentage of
Tests
animals showing seroconversion in each group is transformed
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, (for example, probit transformation) and a parallel line model,
if aluminium hydroxide or hydrated aluminium phosphate is using the log dose response curve, is applied to the data. The
used as the adsorbant. potency of the preparation under examination relative to the
Antimicrobial preservative. Where applicable, determine the reference preparation is thus established. The test is not valid
amount of antimicrobial preservative by a suitable chemical unless (a) for both the test and reference vaccine, the EDsolies
method. The content is not less than the minimum amount between the smallest and the largest doses given to animals ;
shown to be effective and is not greater than 115.0 per cent of (b) the statistical analysis shows no deviation from linearity
that stated on the label. or parallelism; (c) the fiducial limits of the estimated relative
potency fall between 33.0 and 300.0 per cent ofthe estimated
Sterility (2.2.11). Complies with the test for sterility. potency.
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
Method B(ln vitro)
the equivalent of one human dose into each rabbit.
The potency of the vaccine under examination is determined
A validated test for bacterial endotoxins may be used instead
by an in vitro method that has been validated against the
of the test for pyrogens.
biological test.
Assay. The vaccine complies with the assay of Hepatitis-B Enzyme Linked Immunosorbent Assay (ELISA) using
Vaccine (rDNA) described below. monoclonal antibodies specific for protection inducing
Potency. The upper fiducial limit (P = 0.95) ofthe estimated epitopes ofHBsAg have been shown to be suitable. Adequate
relative potency is not less than 1.0. The estimate of potency number of dilutions and replicates of the vaccine under
shall be expressed in terms of Ilg/ml i.e, not less than 20 Ilg/ml. examination and the reference standard are employed in the
Determine the potency either in animals (Method A) or by a assay. The data obtained is analyzed by a parallel-line model
validated in vitro procedure (Method B) described below: and may be suitably transformed for statistical evaluation.
Commercially available kits for measuring HBsAg in vitro may
Method A (Biological) be used provided they are validated to produce equally precise
The potency of the vaccine under examination is determined and accurate results.
in animals by comparing in given conditions its capacity to The test is not valid unless (a) the st\ltistical analysis shows
induce specific anti-HBsAg antibodies in mice or guinea-pigs no deviation from linearity or parallelism; (b) the fiducial limits
with the same capacity as with the reference standard. ofthe estimated relative potency fall between 80.0 and 125.0
per cent of the estimated potency.
Inject intraperitoneally not less than three suitable dilutions
of the vaccine under examination diluted with adjuvant used The acceptance criteria are approved for a given reference
in the vaccine into groups ofa suitable strain ofmice, weighing preparation by the National Regulatory Authority in the light
between 15 and 20 g (about 5 weeks old), of either sex of the validation data.
distributed randomly into several groups of mice. Healthy Labelling. The label states (a) the amount ofHBsAg per dose;
guinea pigs weighing between 300 and 350 g (about 7 weeks (b) the type ofcells used for production ofthe vaccine; (c) the

2401

www.webofpharma.com
INACTIVATED HEPATITIS A VACCINE (ADSORBED) IP 2010

name and amount of the adjuvant; (d) that the vaccine must Virus concentration. The virus concentration ofeach master
be shaken before use; (e) that the vaccine mustnot be frozen. and working seed lot is determined to monitor consistency of
production.
Extraneous agents (2.7.3). The master and working seed lots
Inactivated Hepatitis A Vaccine comply with the requirements for seed lots for virus vaccines.
(Adsorbed) In addition, if primary monkey cells have been used for
isolation of the strain, measures are taken to ensure that the
Hepatitis A Vaccine (Inactivated, Adsorbed) is a liquid
strain is not contaminated with simian viruses such as simian
preparation of a suitable strain of hepatitis A virus grown in
immunodeficiency virus and filoviruses.
cell cultures, inactivated by a validated method and adsorbed
on a mineral carrier. The vaccine is an opalescent suspension.
PROPAGATION AND HARVEST
The vaccine complies with the monograph on Vaccines.
All processing ofthe cell bank and subsequent cell cultures is
Production done under aseptic conditions in an area where no other cells
Production of the vaccine is based on a virus seed-lot system are being handled. Animal serum (but not human serum) may
and a cell-bank system. The production method shall have be used in the cell culture media. Serum and trypsin used in
been shown to yield consistently vaccines that comply with the preparation of cell suspensions and media are shown to
the requirements for immunogenicity, safety and stability. be free from extraneous agents. The cell culture media may
The production method is validated to demonstrate that the contain a pH indicator, such as phenol red and approved
product, if tested, would comply with the test for safety and antibiotics at the lowest effective concentration. Not less than
efficacy. 500 ml ofthe cell cultures employed for vaccine production is
set aside as uninfected cell cultures (control cells). Multiple
Unless otherwise justified and authorised, the virus in the harvests from the same production cell culture may be pooled
final vaccine shall not have undergone more passages from and considered as a single harvest.
the master seed lot than were used to prepare the vaccine
shown in clinical studies to be satisfactory with respect to Only a single harvest that complies with the following
safety and efficacy. requirements may be used in the preparation of the vaccine.
Reference preparation. A part of a batch shown to be at least When the determination of the ratio of virus concentration to
as immunogenic as a batch that, in clinical studies in young antigen content has been carried out on a suitable number of
healthy adults, produced not less than 95.0 per cent single harvests to demonstrate consistency, it may
seroconversion, corresponding to a level of neutralising subsequently be omitted as a routine test.
antibody accepted to be protective, after a full-course of
primary immunisation is used as a reference preparation. An Identification
antibody level of 20 mIU Iml determined by enzyme-linked
The test for antigen content also serves to identify the single
immunosorbent assay is recognised as being protective.
harvest.
Substrate for virus propagation Sterility (2.2.11). The single harvest complies with the test for
The virus is propagated in a human diploid cell line or in a sterility, carried out using 10 ml for each medium.
continuous cell line approved by the competent authority. Mycoplasmas (2.7.4). The single harvest complies with the
SEED LOT test for mycoplasmas carried out using Iml for each medium.
The strain ofhepatitis A virus used to prepare the master seed Control cells. The control cells ofthe production cell culture
lot shall be identified by historical records that include comply with a test for identity and the requirements for
information on the origin of the strain and its subsequent extraneous agents.
manipulation. Antigen content. Determine the hepatitis A antigen content
Only a seed lot that complies with the following requirements by a suitable immunochemical method (2.2.14) to monitor
may be used for virus propagation. production consistency; the content is within the limits
approved for the particular product.
Description. A._-clear, colourless or light coloured liquid.
_.~,~ - Ratio--oLvh:us_concentration_toantigencontent.The
Identification consistency of the ratio of the .concentration of infectious
virus, as determined by a suitable cell culture method, to
Each master and working seed lot is identified as hepatitis A antigen content is established by validation on a suitable
virus using specific antibodies. number of single harvests.

2402

www.webofpharma.com
IP 2010 INACTIVATED HEPATITIS A VACCINE (ADSORBED)

PURIFICATION AND PURIFIED HARVEST Inactivation. Carry out an amplification test for residual
infectious hepatitis A virus by inoculating a quantity of the
The harvest, which may be a pool of several single harvests,
inactivated harvest equivalent to 5 per cent of the batch or if
is purified by validated methods. If continuous cell lines are
the harvest contains the equivalent of 30,000 doses or mort<,
used for production, the purification process shall have been
not less than 1,500 doses of vaccine into cell cultures of the
shown to reduce consistently the level ofhost-cell DNA. Only
same type as those used for production of the vaccine and
a purified harvest that complies with the following requirements
incubating the cells for at least 28 days. Make two passages
may be used in the preparation of the inactivated harvest.
and at the end of incubation carry out a test of suitable
Virus concentration. The concentration of infective virus in sensitivity for residual infectious virus. No evidence of
the purified harvest is determined by a suitable cell culture hepatitis A virus multiplication is found in the samples taken
method to monitor production consistency and as a starting at the end of the inactivation process. Use infective virus
point for monitoring the inactivation curve. inocula concurrently as positive controls to demonstrate
Antigen total protein ratio. Determine the hepatitis A virus cellular susceptibility and absence of interference.
antigen content by a suitable immunochemical method (2.2.14). Sterility (2.2.11). The inactivated viral harvest complies with
Determine the total protein by a validated method. The ratio the test for sterility, carried out using i 0 ml for each medium.
of hepatitis A virus antigen content to total protein content is Bacterial endotoxins (2.2.3). Not more than 2 ill ofendotoxin
within the limits approved for the particular product.
in the equivalent of a single human dose.
Bovine serum albumin. Not more than 50 ng in the equivalent Antigen content. Determine the hepatitis A virus antigen
of a single human dose, determined by a suitable content by a suitable immunochemical method (2.2.14).
imrnunochemical method (2.2.14). Where appropriate in view
of the manufacturing process, other suitable protein markers Residual chemicals. As stated under Purification and Purified
may be used to demonstrate effective purification. Harvest.

Residual host-cell DNA (2.2.15). If a continuous celllineis FINAL BULK VACCINE


used for virus propagation, the content of residual host-cell
The final bulle vaccine is prepared from one or more inactivated
DNA, determined using a suitable method is not greater than
harvests. Approved adjuvants, stabilisers and antimicrobial
10 ng per single human dose.
preservatives may be added.
Residual chemicals. If chemical substances are used during
Only a final bulle vaccine that complies with the following
the purification process, tests for these substances are carried
requirements may be used in the preparation of the final lot.
out on the purified harvest (or on the inactivated harvest),
unless validation of the process has demonstrated total Antimicrobial preservative. Where applicable, determine the
clearance. The concentration must not exceed the limits amount of antimicrobial preservative by a suitable chemical
approved for the particular product. method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent ofthe intended amount.
INACTIVATION AND INACTIVATED HARVEST
Sterility (2.2.11). Carry out test for sterility using 10 ml of
Several purified harvests may be pooled before inactivation. bulle for each sterility medium.
In order to avoid interference with the inactivation process,
virus aggregation must be prevented or aggregates must be FINAL LOT
removed immediately before and/or during the inactivation
process. The virus suspension is inactivated by a validated The final bulle vaccine is distributed aseptically into sterile
method; the method shall have been shown to be consistently containers. The containers are then closed so as to avoid
capable of inactivating hepatitis A virus without destroying contamination.
the antigenic and immunogenic activity; as part of the Only a final lot that complies with each of the requirements
validation studies, an inactivation curve is plotted representing given below under Identification, Tests and Assay may be
residual live virus concentration measured on at least three released for use. Provided that the tests for free formaldehyde
occasions (for example, on days 0, 1 and 2 ofthe inactivation (where applicable) and antimicrobial preservative content
process). Iffonnaldehyde is used for inactivation, the presence (where applicable) and the assay have been carried out on the
of excess free formaldehyde is verified at the end of the final bulk vaccine with satisfactory results, these tests may be
inactivation process. omitted on the final lot. If the assay is carried out using mice
Only an inactivated harvest that complies with the following or other animals, then provided it has been carried with
requirements may be used in the preparation ofthe final bulle satisfactory results on the final bulle vaccine, it may be omitted
vaccine. on the final lot.

2403

www.webofpharma.com
INACTNATED HEPATITIS A VACCINE (ADSORBED) IP 2010

Identification Calculations. Cany out the calculations by the usual statistical


methods (5.7) for an assay with a quantal response.
The vaccine is shown to contain hepatitis A virus antigen by
a suitable immunochemical method using specific antibodies From the distribution of reaction levels measured on all the
or by the mouse immunogenicity test described under Assay. sera in the unvaccinated group, determine the maximum
reaction level that can be expected to occur in an unvaccinated
Tests animal for that particular assay. Any response in vaccinated
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, animals that exceeds this level is by definition a seroconversion.
if hydrated aluminium phosphate or aluminium hydroxide is Make a suitable transformation of the percentage of animals
used as the adsorbent. showing seroconversion in each group (for example, a probit
Free formaldehyde (2.3.20). When formaldehyde has been transformation) and analyse the data according to a parallel-
used for inactivation, the vaccine complies with the test line log dose-response model. Detennine the potency of the
prescribed in Vaccines. test preparation relative to the reference preparation.
Antimicrobial preservative. Where applicable, determine the Validity conditions. The test is not valid unless (a) for both
amount of antimicrobial preservative by a suitable chemical the test and the reference vaccine, the ED so lies between the
method. The content is not less than 85.0 per cent and not smallest and the largest doses given to the animals; (b) the
greater than 115.0 per cent ofthe intended amount. statistical analysis shows no significant deviation from
Sterility (2.2.11). Complies with the test for sterility. linearity or parallelism; (c) the fiducial limits ofthe estimated
Abnormal toxicity (2.2.1). Complies with the test for abnormal relative potency fall between 33.0 and 300.0 per cent of the
toxicity. estimated potency.
Potency. The upper fiducial limit (P = 0.95) of the estimated
Assay relative potency is not less than 1.0.
The vaccine complies with the assay of Hepatitis A vaccine.
In vitro assay
The assay is carried out either in vivo, by comparing in given
condition the capacity to induce specific antibodies in mice Carry out an immunochemical determination ofantigen content
with the same capacity of a reference preparation or in vitro (2.2.14) with acceptance criteria validated against the in vivo
by an immunochemical determination of antigen content test. The acceptance criteria are approved for a given reference
(2.2.14). preparation by the National Regulatory Authority in the light
of the validation data.
In vivo assay
Labelling. The label states (1) the biological origin ofthe cells
The test on mice shown below is given as an example of a and; (2) the adjuvant used for the preparation of the vaccine.
method that has been found suitable for a given vaccine;
other validated methods may also be used.
Selection and distribution ofthe test animals. Healthy mice
from the same stock, about 5 weeks old and from a strain
Inactivated Hepatitis B Vaccine
shown to be suitable should be used in the test. Use animals Inactivated Hepatitis B Vaccine is a non-infectious inactivated
ofthe same sex. Distribute the animals in at least seven equal liquid preparation derived from the surface antigen ofHepatitis
groups of a number suitable for the requirements of the assay. B virus (HbsAg). This preparation is white or almost white
Determination ofpotency ofthe vaccine under examination. translucent liquid in which the mineral carrier tends to settle
Using a 0.9 per cent w/v solution of sodium chloride containing down slowly on keeping but is free from foreign particles /
the aluminium adjuvant used for the vaccine, prepare at least floccules.
three dilutions ofthe vaccine under examination and matching
Production
dilutions of the reference preparation. Allocate the dilutions
one to each of the groups of animals and inject The antigen is harvested and purified from the plasma ofhmnan
subcutaneously not more than 0.5 ml of each dilution into carriers of Hepatitis B virus. The surface antigen contains all
each animal in the group to which that dilution is allocated. the three antigen species (S, Pre-S 1, Pre-S2). The individual
Maintain a group of unvaccinated controls, injected donor plasma is shown by sensitive tests to be seronegative
··subGutane0usly-with·thesame-v01ume-0fdiluent~After-28-toformv~landHJ:V,,2. andforHCV..Theplasmapoolistestedfor
32 days, anaesthetise and bleed all animals, keeping the freedom from adventitious viruses and blood borne
individual sera separate. Assay the individual sera for specific transmissible pathogens by appropriate methods. The purified
antibodies against hepatitis A virus· by a suitable antigen is further inactivated by a validated method, usually
inununochemical method (2.2.14). with formalin or any other inactivating agent, to render the

2404

www.webofpharma.com
lP 2010 INACTIVATED HEPATITIS B VACCINE

hepatitis B virus harmless. The preparation is also tested for The molecular weight of the major band in a sodium dodecyl
the residual HBV DNA using a sensitive test approved by the sulphate polyacrylamide gel electrophoresis (SDS-PAGE)
competent authority and the level is shown to be less than 1 under reduced conditions corresponds to the value expected
pg HBV DNA per 50 doses. from the gene sequence and possible glycosylation.
The method used for production of the vaccine must have Antigenic purity. The purity of the antigen is determined by
been shown to yield a product consistently complying with comparison with a reference preparation using liquid
the requirements ofimmunogenicity, safety and stability. The chromatography or other suitable methods such as SDS-PAGE
production method must also be validated to demonstrate with staining. A suitable method is sensitive enough to detect
that the product, if tested, would comply with the tests for a potential contaminant at a concentration of 1.0 per cent of
safety and efficacy. total protein. Not less than 95.0 per cent of the total protein
consists of hepatitis B surface antigen.
Reference preparation. A part of a batch shown to be at least
as immunogenic as a batch that produced in clinical studies in Composition. The content of proteins, lipids, nucleic acids
young healthy adults not less than 95.0 per cent and carbohydrates is determined.
seroconversion, corresponding to a level of neutralizing
Sterility (2.2.11). The purified antigen complies with the test
antibody accepted to be protective (HbsAg antibody titre not
for sterility carried out using 10 ml for each medium.
less than 10 mIU/rnl after a full course ofprimary immunization
determined by enzyme-linked immunosorbent assay (ELISA) Additional tests on the purified antigen may be required
is used as a reference preparation. depending on the production method used.

Characterisation of the substance FINAL BULK VACCINE


Development studies are carried out to characterize the An antimicrobial preservative and an adjuvant may be
antigen. The complete protein, lipid and carbohydrate structure included in the vaccine.
ofthe antigen is established. The morphological characteristics Only a final bulle vaccine that complies with the following
ofthe antigen particles are established by electron microscopy. requirements may be used in the preparation of the final lot.
The buoyant density ofthe antigen particles is determined by
a physico-chemical method (2.4.29), for example gradient Antimicrobial preservative. Where applicable, determine the
centrifugation. The antigenic epitopes are characterized. The amount ofantimicrobial preservative by a suitable chemical or
protein fraction ofthe antigen is characterized in terms of the physico-chemical method. The amount is not less than the
primary structure (for example, by determination ofthe amino- 85.0 per cent and not greater than 115.0 per cent ofthat stated
acid composition, by partial amino-acid sequence analysis). on the label.
Sterility (2.2.11). Carry out test for sterility using J 0 ml of
PROPAGATION AND HARVEST bulle for each sterility medium.
Identity, microbial purity, plasmid retention and consistency FINAL LOT
of yield are determined at suitable production stages.
Only a final lot that complies with each of the requirenlents
PURIFIED ANTIGEN given below under Identification, Tests and Assay may be
released for use. Provided that the tests for free formaldehyde,
Only a purified antigen that complies with the following antimicrobial preservative content and the assay in animals,
requirements may be used in the preparation ofthe final bulle where applicable, have been carried out on the final bulle
Total protein (2.3.49). The total protein is determined by a vaccine with satisfactory results, they may be omitted on the
validated method. The content is within the limits approved final lot.
for the specific product.
Identification
Antigen content and identification. The quantity and
specificity of HBsAg is determined in comparison with the The assay or, where applicable, the electrophoretic profile,
International standard for HBsAg subtype ad or an in-house also serves to identify the vaccine.
reference, by a suitable immunochemical method such as radio
Tests
immunoassay (RIA), enzyme-linked immunosorbent assay
(ELISA), immunoblot (preferably using a monoclonal antibody Aluminium (2.3.9). When hydrated aluminium phosphate or
directed against a protective epitope) or single radial diffusion. aluminium hydroxide is used as the adsorbent, the vaccine
The antigen/protein ratio is within the limits approved for the complies with the test prescribed in the monograph on
specific product. Vaccines.

2405

www.webofpharma.com
INACTIVATED INFLUENZA VACCINE (SPLIT VIRION) IP 2010

Test for inactivating agent. The concentration of any The origin and passage history of virus strains shall be
inactivating agent remaining in the final vaccine shall be approved by the National Regulatory Authority.
determined by methods approved by the competent authority.
The concentration shall not exceed a specified upper limit. Substrate for virus propagation

Antimicrobial preservative. Where applicable, determine the Influenza virus seed to be used in the production ofvaccine is
amount of antimicrobial preservative by a suitable chemical propagated in fertilised eggs from chicken flocks free from
method. The content is not less than 85.0 per cent and not specified pathogens or in. suitable cell cultures, such as chick-
greater than 115.0 per cent of the intended amount. embryo fibroblasts or chick kidney cells obtained from chicken
flocks free from specified pathogens. For production, the virus
Sterility (2.2.11). Complies with the test for sterility.
of each strain is grown in the allantoic cavity of eggs derived
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject from specific pathogen free flocks.
the equivalent of one human dose into each rabbit.
A validated test for bacterial endotoxins (2.2.3) may be used SEED LOT
instead of the test for pyrogens.
The production of vaccine is based on a seed-lot system.
Assay Working seed lots represent not more than fifteen passages
from the approved reassorted virus or the approved virus
The upper fiducial limit (P = 0.95) of the estimated relative isolate. The final vaccine represents one passage from the
potency is not less than 1.0. working seed lot. The haemagglutinin and neuraminidase
Determine the potency by method A (I3iological)as ciescribed antigens ofeach seed lot are identified as originating from the
under Hepatitis-B vaccine (rDNA). correct strain of influeni8.viriisbY suitable methods..

Labelling. The label states (1) the amount ofHBsAg per dose; Only a working virus seed lot that complies with the following
(2) the name and amount of inactivating agent; (3) the name requirements may.be used in the preparation ofthe monovalent
and amount of the adjuvant; (4) that the vaccine must be pooled harvest.
shaken before use; (5) that the vaccine must not be frozen. Sterility (2.2.11). Carry out the test for sterility using 10 ml for
each medimn.
Mycoplasmas (2.7.4). Carry out the test for mycoplasmas using
Inactivated Influenza Vaccine (Split lOml.
Virion) PROPAGATION AND HARVEST
Influenza Vaccine (Split Virion, Inactivated) is a sterile, aqueous
An antimicrobial agent may be added to the inoculum. After
suspension of a strain or strains of influenza virus, type A or
incubation at a controlled temperature, the allantoic fluids are
B, or a mixture of strains ofthe two types grown individually
harvested and combined to form a monovalent pooled harvest.
in eggs derived from specific pathogen free flock or cell
An antimicrobial agent may be added at the time of harvest.
cultures, inactivated and treated so that the integrity of the
At no stage in the production, penicillin or streptomycin is
virus particles has been disrupted without diminishing the
used.
antigenic properties ofthe haemagglutinin and n,euraminidase
antigens. The stated amount of haemagglutinin antigen for MONOVALENT POOLED HARVEST
each strain present in the vaccine is 15 /-lg per dose, unless
clinical evidence supports the use of a different amount. To limit the possibility ofcontamination, inactivation is initiated
as soon as possible after preparation. The virus is inactivated
by a method that has been demonstrated on three consecutive
Production batches to be consistently effective for the manufacturer. The
The production method is validated to demonstrate that the inactivation process shall have been shown to be capable of
product, if tested, would comply with the test for safety and inactivating the influenza virus without destroying its
efficacy. antigenicity; the process should cause minimum alteration of
the haemagglutinin and neuraminidase antigens. The
Choice of vaccine strain
inactivation.processshallalsohavebeenshownJobecapable
The World Health. Organisation reviews the world of inactivating avian leucosis viruses and mycoplasmas. If
epidemiological situation annually and if necessary the monovalent pooled harvest is stored after inactivation, it
recommends new strains corresponding to prevailing is held at a temperature of5 ± 30. Ifformaldehyde solution is
epidemiological evidence. used, the concentration does not exceed 0.2g/l of

2406

www.webofpharma.com
IP 2010 INACTIVATED INFLUENZA VACCINE (SPLIT VIRION)

formaldehyde at any time during inactivation;· if FINAL LOT


betapropiolactone is used, the concentration does not exceed
The final bulk vaccine is distributed aseptically into sterile,
0.1 per cent v/v at any time during inactivation.
tamper-proof containers. The containers are closed so as to
Before or after the inactivation procedure, the monovalent prevent contamination.
pooled harvest is concentrated and purified by high-speed
Only a final lot that is satisfactory with respect to each of the
centrifugation or other suitable method and the virus particles
requirements given below under Tests and Assay may be
are disrupted into component subunits by the use of approved
released for use. Provided that the test for viral inactivation
procedures. For each new strain, a validation test is carried
has been performed with satisfactory results on each
out to show that the monovalent bulk consists predominantly
monovalent pooled harvest and that the tests for free
of disrupted virus particles.
formaldehyde, ovalbumin and total protein have been
Only a monovalent pooled harvest that complies with the performed with satisfactory results on the final bulk vaccine,
following requirements may be used in the preparation ofthe they may be omitted on the final lot.
final bulle vaccine.
Description. The vaccine is a slightly opalescent liquid.
Haemagglutinin antigen. Determine the content of
haemagglutinin antigen by an immunodiffusion test (2.2.14), Identification
by comparison with a haemagglutinin antigen reference The assay serves to confinn the antigenic specificity of the
preparation or with an antigen preparation calibrated against vaccine.
it. Carry out the test at 20° to 25°.
For some vaccines, the physical form of the haemagglutinin Tests
particles prevents quantitative determination by Viral inactivation. Inoculate 0.2 ml of the vaccine into the
immunodiffusion after inactivation of the virus. For these allantoic cavity of each of ten fertilised eggs and incubate at
vaccines, a determination of haemagglutinin antigen is made 33° to 37° for 3 days. The test is not valid unless at least eight
on the monovalent pooled harvest before inactivation. The ofthe ten embryos survive. Harvest 0.5 ml of the allantoic
production process is validated to demonstrate suitable fluid from each surviving embryo and pool the fluids. Inoculate
conservation of haemagglutinin antigen and a suitable tracer 0.2 ml ofthe pooled fluid into a further ten fertilised eggs and
is used for formulation, for example, protein content. incubate at 33° to 37° for 3 days. The test is not valid unless at
Neuraminidase antigen. The presence and type of least eight of the ten embryoes survive. Harvest about 0; 1 ml
neuraminidase antigen are confirmed by suitable enzymatic or ofthe allantoic fluid from each surviving embryo and examine
immunological methods (2.2.14) on the first three monovalent each individual harvest for live virus by a haemagglutination
pooled harvests from each working seed lot. test. Ifhaemagglutination is found for any ofthe fluids, carry
out for that fluid a further passage in eggs and test for
Sterility (2.2.11). Carry out the test for sterility using 10 ml for
haemagglutination; no haemagglutination occurs.
each medium.
Total protein (2.3.49). Not more than six times the total
Viral inactivation. Carry out as described below under Tests.
haemagglutinin content of the vaccine as determined in the
Chemicals used for disruption. Tests are carried out on the assay, but in any case, not more than 100 f!g of protein per
monovalent pooled harvest for the chemicals used for virus strain per human dose and not more than a total of 300
disruption, the limits being approved by the National f!g of protein per human dose.
Regulatory Authority.
Ovalbumin. Not more than 1 f!g ofovalbumin per human dose,
FINAL BULK VACCINE determined by a suitable technique using a suitable reference
Appropriate quantities of the monovalent pooled harvests preparation of ovalbumin.
are blended to make the final bulk vaccine. Antimicrobial preservative. Where applicable, determine the
Only a final bulk vaccine that complies with the following amount of antimicrobial preservative by a suitable chemical
requirements may be used in the preparation of the final lot. method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical Sterility (2.2.11). Complies with the test for sterility.
method. The content is not less than 85.0 per cent and not Abnormal toxicity (2.2.1). Complies with the test for abnormal
greater than 115.0 per cent ofthe intended amount. toxicity.
Sterility (2.2.11). Carry out the test for sterility using 10 ml for Bacterial endotoxins (2.2.3). Not more than 100 ill ofendotoxin
each sterility medium. per human dose.

2407

www.webofpharma.com
INACTIVATED INFLUENZA VACCINE (SURFACE ANTIGEN) IP 2010

Assay specified pathogens or in suitable cell cultures, such as chick-


Determine the content of haemagglutinin antigen by an embryo fibroblasts or chick kidney cells obtained from chicken
immunodiffusion test (2.2.14), by comparison with an flocks free from specified pathogens. For production, the virus
appropriate haemagglutinin antigen reference preparation. of each strain is grown in the allantoic cavity of eggs derived
Carry out the test at 20 to 25°. The confidence interval (P = from SPF flocks.
0.95) ofthe assay is not greater than 80.0 per cent to 125.0 per SEED LOT
cent ofthe estimated content. The lower confidence limit (P =
The production of vaccine is based on a seed-lot system.
0.95) of the estimate ofhaemagglutinin antigen content is not
Working seed lots represent not more than fifteen passages
less than 80.0 per cent of the amount stated on the label for
from the approved reassorted virus or the approved virus
each strain.
isolate. The final vaccine represents one passage from the
For some vaccines, quantitative determination of working seed lot. The haemagglutinin and neuraminidase
haemagglutinin antigen with respect to available reference antigens ofeach seed lot are identified as originating from the
preparations is not possible. An immunological identification correct strain of influenza virus by suitable methods.
of the haemagglutinin antigen and a semi-quantitative
Only a working virus seed lot that complies with the following
detetmination are carried out instead by suitable methods.
requirements may be used in the preparation ofthe monovalent
Labelling. The label complies with the requirements stated pooled harvest.
under Vaccines and also states (a) that the vaccine has been
Sterility (2.2.11). Carry out the test for sterility using 10 ml for
prepared on eggs; (b) the strain or strains of influenza virus
each medium.
used to prepare the vaccine; (c) the method of inactivation;
Mycoplasmas (2.7.4). Carry out the test for mycoplasmas using
(d) the haemagglutinin content in fJ.g per virus strain per dose;
(e) the season during which the vaccine is intended to protect. lOmi.
PROPAGATION AND HARVEST
Inactivated Influenza Vaccine (Surface An antimicrobial agent may be added to the inoculum. After
incubation at a controlled temperature, the allantoic fluids are
Antigen) harvested and combined to form a monovalent pooled harvest.
Influenza Vaccine (Surface Antigen, Inactivated) is a sterile, An antimicrobial agent may be added at the time of harvest.
aqueous suspension of a strain or strains of influenza virus, At no stage in the production penicillin or streptomycin is
type A or B, or a mixture of strains of the two types grown used.
individually in eggs derived from specific pathogen free flocks MONOVALENT POOLED HARVEST
or cell cultures, inactivated and treated so that the preparation
To limit the possibility ofcontamination, inactivation is initiated
consists predominantly ofhaemagglutinin and neuraminidase
as soon as possible after preparation. The virus is inactivated
antigens, without diminishing the antigenic properties ofthese
by a method that has been demonstrated on three consecutive
antigens. The stated amount of haemagglutinin antigen for
batches to be consistently effective for the manufacturer. The
each strain present in the vaccine is 15 fJ.g per dose, unless
inactivation process shall have been shown to be capable of
clinical evidence supports the use of a different amount.
inactivating the influenza virus without destroying its
Production antigenicity; the process should cause minimum alteration of
The production method is validated to demonstrate that the the haemagglutinin and neuraminidase antigens. The
product, if tested, would comply with the test for safety and inactivation process shall also have been shown to be capable
efficacy. of inactivating avian leucosis viruses and mycoplasmas. If
the monovalent pooled harvest is stored after inactivation, it
Choice of vaccine strain is held at a temperature of5±3°. If formaldehyde solution is
The World Health Organisation reviews the world used, the concentration does not exceed 0.2 gil of
epidemiological situation annually and if necessary formaldehyde at any time during inactivation; if
recommends new strains corresponding to prevailing betapropiolactone is used, the concentration does not exceed
epidemiological evidence. 0.1 per cent vlv at any time during inactivation.
The origin and passage history of virus strains shall be Before or after the inactivation process, the wonovalent
approve_db~Jhe competentauthority.__ _ p_o_oled_hanrest.is._concentrate&andpurifiedbyhigh~speed
c~ntrifugati()l1.gr. otlle! . sl!itll1:>le.l11etl1()Q, Yil:us. pr:lJiiclesare
Substrate for virus propagatioll disrupted into component subunits by approved procedures
Influenza virus seed to be used in the production ofvaccine is and further purified so that the monovalent bulk consists
propagated in fertilised eggs from chicken flocks free from mainly of haemagglutinin and neuraminidase antigens.

2408

www.webofpharma.com
IP 2010 INACTIVATED INFLUENZA VACCINE (SURFACE ANTIGEN)

Only a monovalent pooled harvest that complies with the Description. The vaccine is a clear liquid.
following requirements may be used in the preparation ofthe
final bulle vaccine.
Identification
Haemagglutinin antigen. Determine the content of The assay serves to confinn the antigenic specificity of the
haemagglutinin antigen by an immunodiffusion test (2.2.14), vaccine.
by comparison with a haemagglutinin antigen reference Tests
preparation or with an antigen preparation calibrated against
it. Carry out the test at 20 0 to 25 0 • .
Viral inactivation. Inoculate 0.2 ml of the vaccine into the
allantoic cavity of each of ten fertilised eggs and incubate at
Neuraminidase antigen. The presence and type of 33 0 to 37 0 for 3 days. The test is not valid unless at least eight
neuraminidase antigen are confinued by suitable enzymatic or of the ten embryos survive. Harvest 0.5 ml of the allantoic
immunological methods (2.2.14) on the first three monovalent fluid from each surviving embryo and pool the fluids. Inoculate
pooled harvests from each working seed lot. 0.2 ml of the pooled fluid into a further ten fertilised eggs and
Sterility (2.2.11). Carry out the test for sterility, using incubate at 33 0 to 37 0 for 3 days. The test is not valid unless at
10 ml for each medium. least eight ofthe ten embryos survive. Harvest about O.lml of
the allantoic fluid from each surviving embryo and examine
Viral inactivation. Carry out the test described below under each indiviclual harvest for live virus by a haemagglutination
Tests. test. Ifhaemagglutination is found for any of the fluids, carry
Purity. The purity of the monovalent pooled harvest is out for that fluid a further passage in eggs and test for
examined by polyacrylamide gel electrophoresis or by other haemagglutination; no haemagglutination occurs.
approved techniques. Mainly haemagglutinin and
Total protein (2.3.49). Not more than 40 Ilg of protein other
neuraminidase antigens shall be present.
than haemagglutinin per virus strain per human dose and not
Chemicals used for disruption and purification. Tests are more than a total of120 Ilg ofprotein other than haemagglutinin
carried out on themonovalent pooled harvest for the chemicals per human dose.
used for disruption and purification, the limits being approved
Ovalbumin. Not more than 11lg ofovalbumin per human dose,
by the competent authority.
detennined by a suitable technique using a suitable reference
FINAL BULK VACCINE preparation of ovalbumin.
Appropriate quantities of the monovalent pooled harvests Free formaldehyde (2.3.20). Complies with the test for free
are blended to make the final bulk vaccine. fonnaldehyde as stated under General Requirements for
Vaccines for Human Use.
Only a final bulle vaccine that complies with the following
requirements may be used in the preparation of the final lot. Antimicrobial preservative. Where applicable, determine the
Antimicrobial preservative. Where applicable, detennine the amount of antimicrobial preservative by a suitable chemical
amount of antimicrobial preservative by a suitable chemical method. The content is not less than 85.0 per cent and not
method. The content is not less than 85.0 per cent and not greater than 115.0 per cent of the intended amount.
greater than 115.0 per cent ofthe intended amount. Abnormal toxicity (2.2.1). Complies with the test for abnonnal
Sterility (2.2.11). Carry out the test for sterility using 10 ml for toxicity.
each medium. Sterility (2.2.11). Complies with the test for sterility.

FINAL LOT Bacterial endotoxins (2.2.3). Not more than 100 ill ofendotoxin
per human dose.
The final bulk vaccine is distributed aseptically into sterile,
tamper-proof containers. The containers are closed so as to Assay
prevent contamination. Determine the content haemagglutinin antigen by an
Only a final lot that is satisfactory with respect to each of the immunodiffusion test (2.2.14), by comparison with an
requirements given below under Identification, Tests and appropriate haemagglutinin antigen reference preparation.
Assay may be released for use. Provided that the test for viral Carry out the test at 20 0 to 25 0 • The confidence interval
inactivation has been perfonned with satisfactory results on (P = 0.95) ofthe assay is not greater than 80.0 per centto 125.0
each monovalent pooled harvest and that the tests for free per cent of the estimated content. The lower confidence limit
formaldehyde, ovalbumin and total protein have been (P = 0.95) ofthe estimate ofhaemagglutinin antigen content is
perfonned with satisfactory results on the final bulle vaccine, not less than 80.0 per cent of the amount stated on the label
they may be omitted on the final lot. for each strain.

2409

www.webofpharma.com
INACTIVATED INFLUENZA VACCINE (WHOLE VIRION) IP 2010

Labelling. The label states (1) that the vaccine has been antigens ofeach seed lot are identified as originating from the
prepared on eggs; (2) the strain or strains of influenza virus correct strain of influenza virus by suitable methods.
used to prepare the vaccine; (3) the method of inactivation; Only a working virus seed lot that complies with the following
(4) the haemagglutinin content in micrograms per virus strain requirements may be used in the preparation ofthe monovalent
per dose; (5) the season during which the vaccine is intended pooled harvest.
to protect.
Sterility (2.2.11). Carry out the test for sterility using 10 ml for
each medium.
Mycoplasmas (2.7.4). Carry out the test for mycoplasmas using
Inactivated Influenza Vaccine (Whole IOml.
Virion)
PROPAGATION AND HARVEST
Influenza Vaccine (Whole Virion, Inactivated) is a sterile,
aqueous suspension of a strain or strains of influenza virus, An antimicrobial agent may be added to the inoculum. After
type A or B, or a mixture of strains of the two types grown incubation at a controlled temperature, the allantoic fluids are
individually in eggs derived from specific pathogen free flocks harvested and combined to fonn a monovalent pooled harvest.
or cell culture and inactivated in such a manner that their An antimicrobial agent may be added at the time of harvest.
antigenic properties are retained. The stated amount of At no stage in the production is penicillin or streptomycin
haemagglutinin antigen for each strain present in the vaccine used.
is 15 J.l.g per dose, unless clinical evidence supports the use of
a different amount. MONOVALENT POOLED HARVEST
To limit the possibility ofcontamination, inactivation is initiated
Production as soon as possible after preparation. The virus is inactivated
by a method that has been demonstrated on three consecutive
The production method is validated to demonstrate that the batches to be consistently effective for the manufacturer. The
product, if tested, would comply with the test for safety and inactivation process shall have been shown to be capable of
efficacy. inactivating the influenza virus without destroying its
antigenicity; the process should cause minimum alteration of
Choice of vaccine strain the haemagglutinin and neuraminidase antigens. The
inactivation process shall also have been shown to be capable
The· World Health Organisation reviews the world
of inactivating avian leucosis viruses and mycoplasmas. If
epidemiological situation annually and, if necessary,
the monovalent pooled harvest is stored after inactivation, it
recommends new strains corresponding to prevailing
is held at a temperature of 5±3 0. Iffonnaldehyde solution is
epidemiological evidence.
used, the concentration does not exceed 0.2 gil of
The origin and passage history of virus strains shall be formaldehyde at any time during inactivation; if
approved by the competent authority. betapropiolactone is used, the concentration does not exceed
0.1 per cent vlv at any time during inactivation.
Substrate for virus propagation Before or after the inactivation process, the monovalent
Influenza virus seed to be used in the production ofvaccine is pooled harvest is concentrated and purified by high-speed
propagated in fertilised eggs from chicken flocks free from centrifugation or other suitable method.
specified pathogens or in suitable cell cultures, such as chick- Only a monovalent pooled harvest that complies with the
embryo fibroblasts or chick kidney cells obtained from chicken following requirements may be used in the preparation ofthe
flocks free from specified pathogens. For production, the virus final bulk vaccine.
of each strain is grown in the allantoic cavity of eggs derived Haemagglutinin antigen. Determine the content of
from specific pathogen free flocks. haemagglutinin antigen by an immunodiffusion test (2.2.14),
by comparison with a haemagglutinin antigen reference
SEED LOT preparation or with an antigen preparation calibrated against
The_productiolLoLvaccine_is_basecLolLa__see<hloLsystem. it. CafI}'_()l:!t th.e test at?.2°_t()}5°_:.. _
Working Seed lots represent not more thanfifteell passages Neununinid_ase_antigen._ ThepJ'es_enG.eaod type oJ
from the approved reassorted virus or the approved virus neuraminidase antigen are confinned by suitable enzymatic or
isolate. The final vaccine represents one passage from the immunological methods (2.2.14) on the first three monovalent
working seed lot. The haemagglutinin and neuraminidase pooled harvests from each working seed lot.

2410

www.webofpharma.com
IP 2010 JAPANESE ENCEPHALITIS VACCINE (HUMAN)

Sterility (2.2.11). Carry out the test for sterility using 10 ml for assay, but in any case, not more than 100 /lg of protein per
each medium. virus strain per human dose and not more than a total of
Viral inactivation. Carry out the test described below under 300 /lg ofprotein per human dose.
Tests. Ovalbumin. Not more than l/lg ofovalbumin per human dose,
FINAL BULK VACCINE determined by a suitable technique using a suitable reference
preparation of ovalbumin.
Appropriate quantities of the monovalent pooled harvests
Free formaldehyde (2.3.20). Maximum 0.2 gil.
are blended to make the final bulk vaccine.
Antimicrobial preservative. Where applicable, determine the
Only a final bulle vaccine that complies with the following
amount of antimicrobial preservative by a suitable chemical
requirements may be used in the preparation of the final lot.
method. The content is not less than 85 per cent and is not
Antimicrobial preservative. Where applicable, determine the greater than 115.0 per cent of the quantity stated on the label.
amount of antimicrobial preservative by a suitable chemical
Abnormal toxicity (2.2.1). Complies with the test for abnormal
method. The content is not less than 85.0 per cent and not
toxicity.
greater than 115.0 per cent of the intended amount.
Sterility (2.2.11). Carry out test for sterility using 10 ml of Sterility (2.2.11). Complies with the test for sterility.
bulle for each sterility medium. Bacterial endotoxins (2.2.3). Not more than 100 IU ofendotoxlll
FINAL LOT per human dose.

The final bulk vaccine is distributed aseptically into sterile, Assay


tamper-proof containers. The containers are closed so as to Determine the content of haemagglutinin antigen by an
prevent contamination. immunodiffusion test (2.2.14), by comparison with an
Only a final lot that is satisfactory with respect to each ofthe appropriate haemagglutinin antigen reference preparation.
requirements given below under Tests and Assay may be Carry out the test at 20° to 25°. The confidence interval
released for use. Provided that the test for viral inactivation (P = 0.95) oftheassay is not greater than 80.0 per centto 125.0
has been performed with satisfactory results on each' per cent of the estimated content. The lower confidence limit
monovalent pooled harvest and that the tests for free (P = 0.95) ofthe estimate ofhaemagglutinin antigen content is
formaldehyde, ovalbumin and total protein have been not less than 80.0 per cent of the amount stated on the label
performed with satisfactory results on the final bulle vaccine, for each strain.
they may be omitted on the final lot. Labelling. The label states (1) that the vaccine has been
Description. The vaccine is a slightly opalescent liquid. prepared on eggs; (2) the strain or strains of influenza virus
used to prepare the vaccine; (3) the method of inactivation;
Identification (4) the haemagglutinin content in micrograms per virus strain
The assay serves to confirm the antigenic specificity of the per dose; (5) the season during which the vaccine is intended
vaccine. to protect.

Tests
Viral inactivation. Inoculate 0.2 ml of the vaccine into the Japanese Encephalitis Vaccine
allantoic cavity of each of ten fertilised eggs and incubate at (Human)
33° to 37° for 3 days. The test is not valid unless at least eight
of the ten embryos survive. Harvest 0.5 ml of the allantoic Japanese Encephalitis Vaccine for human use'is a liquid or
fluid from each surviving embryo and pool the fluids. Inoculate freeze dried preparation ofJapanese encephalitis virus grown
0.2 ml ofthe pooled fluid into a further ten fertilised eggs and in approved substrate and inactivated by a validated method.
incubate at 33° to 37° for 3 days. The test is not valid unless at
least eight ofthe ten embryos survive. Harvest about 0.1 mlof PrOduction
the allantoic fluid from each surviving embryo and examine General provisions
each individual harvest for live virus by a haemagglutination The vaccine is produced on the basis of virus seed lot system
test. Ifhaemagglutination is found for any ofthe fluids, carry The production method shall have been shown to yield
out for that fluid a further passage in eggs and test for consistently the vaccines that comply with the tests for
haemagglutination; no haemagglutination occurs. immunogenicity, safety and stability. The production method
Total protein (2.3.49). Not more than six times the total is validated to demonstrate that the product, if tested, would
haemagglutinin content of the vaccine as determined in, the comply with the test for safety and efficacy.

2411

www.webofpharma.com
JAPANESE ENCEPHALITIS VACCINE (~ IP 2010

Substrate for virus propagation Virus harvests that comply with the tests given under
Identification and Virus concentration are pooled in the
The virus is propagated in an approved cell substrate like a
preparation of the inactivated viral harvest.
Vero cell line.
Control cells. The control cells ofthe production cell culture
SEED LOT from which the single harvest is derived should comply with
The strain of Japanese encephalitis virus used shall be the test for identification and with the tests for extraneous
identified by historical records that include information on agents (2.7.3).
the origin of the strain and its subsequent manipulation.
Purification and inactivation
The National Regulatory Authority shall determine the
acceptable number ofpassages from the master virus seed lot The virus harvest may be concentrated and/or purified by
to produce working virus seed lots. suitable methods; the virus harvest is inactivated by a validated
Only a working seed lot that complies with the following tests method at a fixed, well defined stage ofthe process which may
may be used for virus propagation. be before, during or after any concentration or purification.
The method shall have been shown to be capable of
Identification inactivating Japanese encephalitis virus without destruction
Each working seed lot is identified as Japanese encephalitis of the immunogenic activity. If formalin is used, the
virus using specific antibodies by an approved method. concentration shall at no time exceed 1:2000.
Virus concentration. The virus concentration ofeach working Only an inactivated viral suspension that complies with the
seed lot is determined by a cell culture method using following tests may. be .used in the preparation. of the fmal
immunofluorescence or any other approved method. bulle vaccine.
Extraneous agents (2.7.3). The working seed lot complies with
the tests for the virus seed lots. Inactivation. Inactivation is confirmed by carrying out an
amplification test for residual infectious Japanese encephalitis
virus. Inoculate a quantity of inactivated viral suspension
PROPAGATION AND HARVEST
equivalent to not less than 25 doses into cell cultures of the
a) Mouse brain vaccine same type as those used for production of the vaccine. Make
a passage after 7 days. Maintain the cultures for a further
The vaccine is prepared by using a seed-lot system. An
period of 14 days and then examine the cell cultures for
approved strain ofvirus is grown by inoculating intracerebrally
Japanese encephalitis virus using an immunofluorescence test.
into healthy mice. Virus harvests are pooled, concentrated
No Japanese encephalitis virus is detected. Alternatively,S ml
and inactivated by addition of formalin or any other suitable
ofeach culture fluid is pooled on day 14 and 21 and 0.03 ml is
inactivating agent. It may contain a suitable preservative. The
inoculated intracerebrally into each of the 10 mice weighing
vaccine may be issued in single or multidose containers.
between 12 and 15 g. The mice are observed for 14 days for
symptoms caused by Japanese encephalitis virus, and mice
b) Cell culture vaccine showing symptoms of Japanese encephalitis virus are
sacrificed and virus presence is confirmed by
All processing of the cell bank and subsequent cell cultures
immunofluorescence test. No Japanese encephalitis virus shall
are done under aseptic conditions in an area where no other
be detected.
cells are handled. Approved animal (but not human) serum
may be used in the media, but the final medium for maintaining Residual host-cell DNA (2.2.15). The content ofresidual host-
cell growth during virus multiplication does not contain animal cell DNA, determined using a suitable method, should not be
serum; the media may contain human albumin. Serum proteins, greater than lOng per single human dose if cells are used in
ifpresent are reduced to an acceptable level by suitable method the production.
of purification. Serum and trypsin used in the preparation of
cell suspension and media are shown to be free from infectious FINAL BULK VACCINE
extraneous agents. The cell culture media may contain a pH
indicator such as phenol red. Not less than 500 ml of the cell The final bulle vaccine is prepared from one or more inactivated
viral suspensions. An approved stabilizer may be added to
c;l,ltt':!!:fl se_lIlPlQYflcl fQr V3,c;c;illfl. prQclllctiQIl 3,!fJ. SfJt afjiclfJ .3,S
uninfected cell cultures (control cells). The virus suspension llla:inta:in-t1re activity of the product Thioff:fersa:tca:nl:5(~msed
as preservative;
is harvested on one or more occasions during incubation.
Multiple harvests from the same production cell culture may Only a final bullc vaccine that complies with the following
be pooled and considered as a single harvest. requirements may be used in the preparation of the final lot.

2412

www.webofpharma.com
IP 2010 JAPANESE ENCEPHALITIS VACCINE (HUMAN)

Formaldehyde (2.3.20). Not more than 0.01 per centw/v. immunized mice by plaque reduction method or serum
neutralization test (SNT) using appropriate cell culture.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical Standard preparation
method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount. The Standard preparation is a freeze-dried Japanese
encephalitis virus vaccine the potency of which has been
Sterility (2.2.11). Carry out test for sterility using 10 ml of detennined in relation to the Japanese encephalitis reference
bulle for each sterility medium. vaccine obtained from the National Institute ofHealth, Tokyo,
Japan.
FINAL LOT
Suggested method
The final bulle vaccine is distributed aseptically into sterile
containers. The containers are then sealed so as to prevent Preparation of challenge virus suspension
contamination.
The approved challenge virus strain is stored in freeze-dried
Only a final lot that complies with each of the tests given form or in liquid form stored below -70°. Prepare a working
under Identification, Tests and Assay may be released for pool ofthe challenge virus strain by inoculating intracerebrally
use. Provided that the test for inactivation has been carried 0.03 ml of 100 fold dilution of the standard strain in Hanks'
out with satisfactory results on the inactivated virus balanced salt solution containing 5 per cent calf serum into a
suspension and the test for bovine serum albumin has been suitable number of 2-day old suckling mice. Sacrifice the
carried out with satisfactory results on the final bulle vaccine, animals after they show characteristic symptoms of
these tests may be omitted on the final lot. encephalitis and become moribund. Harvest their brains
aseptically, wash them in chilled sterile saline solution to
Identification
remove blood clots. Homogenize the brains with Hanks'
The vaccine is shown to contain Japanese encephalitis virus balanced salt solution containing 5 per cent calf serum to'
antigen by a suitable immunochemical method using specific make a 10 per cent emulsion. Centrifuge the emulsion at
antibodies, alternatively, the Assay also serves to identify the 2000 g for 30 minutes. Dilute the supernatant with Hanks'
vaccine. balanced salt solution containing 5 per cent calf serum so as
to contain about 200 Plaque-Forming Units (PFU) ofthe virus
Tests per OA rnI.
Complies with the test for Inactivation under final Purification Determination of potency
and Inactivation. Prepare appropriate dilutions ofthe vaccine under examination
Sterility (2.2.11). Complies with the test for sterility. and of the Standard Preparation in a suitable medium. Inject
intraperitoneally in two doses of 0.5 ml each at 7-day interval
Bacterial endotoxins (2.2.3). Less than 25 ill per single human
dose. . into at least 20 mice of 4 weeks of age. Bleed each mouse 7
days after the second injection, pool the separated serum
Abnormal toxicity (2.2.1). Complies with the test for abnormal from each group and inactivate the sera by heating at 56° for
toxicity. 30 minutes. The inactivated sera may be stored at -20°, if
Bovine serum albumin (for cell culture vaccine). Not more necessary.
than 50 ng per single human dose, determined by a suitable Dilute the sera appropriately, e.g. 1:40. 1: 160, 1:640 etc., mix
immunochernical method (2.2.14). with an equal volume of the challenge virus suspension and
Residual host-cell DNA (for continuous cell line vaccines) incubate at 37° for 90 minutes for neutralization. Inoculate the
(2.2.15). Not more than 10 ng per single human dose. mixture into cell cultures and overlay the infected cells with 1
per cent agar. After incubation for an appropriate time (about
Free formaldehyde (2.3.20). Not more than 0.01 per cent w/v.
48 hours), stain the cells and count the number of plaques
Antimicrobial preservative. Determine the amount of formed on the cultures to obtain the plaque reduction rates
.antiniicrobial preservative by a suitable chemical method. The for the vaccine under examination and the Standard
content is not less than 85 per cent and is not greater than preparation. Calculate the neutralizing antibody titres for each
115.0 per cent of that stated on the label. group using standard statistical methods (5.7). The test is not
valid unless (a) the mean number ofplaques obtained with the
Biological assay Standard preparation is between 100 and 150 per dish and (b)
Potency of Japanese encephalitis virus vaccine is determined the potency of the vaccine under examination is not less than
by titrating the neutralizing antibodies produced in the that of the Standard preparation.

2413

www.webofpharma.com
MEASLES AND RUBELLA VACCINE (LIVE) IP 2010

Labelling. The label states (1) the biological origin ofthe cells Identification
used for the preparation of the vaccine; (2) the strain ofvirus
When the vaccine reconstituted as stated on the label is mixed
used.
with antibodies specific for measles virus and rubella virus, it
is no longer able to infect cell cultures susceptible to these
Measles and Rubella Vaccine (Live) viruses. When the vaccine reconstituted as stated on the
label is mixed with quantities of specific antibodies sufficient
Measles and Rubella Vaccine (Live) is a freeze-dried preparation
to neutralize anyone viral components, the second viral
of suitable attenuated strains of measles virus and rubella
component infects susceptible cell cultures.
virus grown in suitable cell cultures.
The vaccine is reconstituted immediately before use to give a Tests
clear liquid that may be coloured owing to the presence of a
Water (2.3.43). Not more than 3.0 per cent, determined by Karl
pH indicator.
Fischer, semi-micro determination ofwater or by any suitable
Production validated method.

General provisions Sterility (2.2.11). The reconstituted vaccine complies with the
test for sterility.
The two components are prepared as described in the
Abnormal toxicity (2.2.1). Complies with the test for abnormal
monographs on Measles vaccine (live) and Rubella vaccine
toxicity.
(live) and comply with the tests prescribed therein.
The production method is validated to demonstrate that the Bovine serum albumin. Not more than 50 ng per single human
product~iftested;would-coiJjplywith the tesrforsafety and dose, determined by .a suitable immunochemicalmethod
efficacy. (2.2.14).

'FINAL BULK VACCINE Assay


Virus harvests for each component are pooled and clarified to A. Mix the vaccine with a sufficient quantity of antibodies
remove cells. A suitable stabilizer may be added and the pooled specific for rubella virus.Titrate the vaccine for infective
harvests diluted as appropriate. Suitable quantities of the measles virus at least in triplicate, using at least eight cell
pooled harvest for each component are mixed. cultures for each dilution 0.5 10glO step or by a method of
Only a final bulk vaccine that complies with the following equal precision. Use an appropriate virus reference preparation
requirements may be used in the preparation ofthe final lot. to validate each assay. The estimated measles virus
Sterility (2.2.11). Carry out test for sterility using 10 ml of concentration is not less than that stated on the label; the
bulk for each sterility medium. minimum measles virus concentration stated on the label is
not less than 1x 103 CCID50 per single human dose. The assay
FlNALLOT is not valid ifthe confidence limits (P = 0.95) ofthe logarithm
of the virus concentration are greater than ± 0.3.
For each component, a minimum virus concentration for
release of the product is established such as to ensure, in the Measles vaccine (Live) RS is suitable for use as a reference
light of stability data, that the minimum concentration stated preparation.
on the label will be present at the end ofthe period ofvalidity. B. Titrate the vaccine for infective rubella virus at least in
Only a final lot that complies with the tests for minimum virus triplicate, using at least eight cell cultures for each 0.5 10glO
concentration of each component for release, with the dilution step or by a method of equal precision. Use an
following test for thermal stability and with each of the tests appropriate virus reference preparation to validate each assay.
given below under Identification and Tests and Assay may be The estimated rubella virus concentration is not less than that
released for use. Provided that the test for bovine serum albumin stated on the label; the minimum rubella virus concentration
has been carried out with satisfactory results on the final bulk stated on the label are not less than 1xl03 CCID50 per single
vaccine, it may be omitted on the final lot. human dose. The assay is not valid if the confidence limits .
(P = 0.95) ofthe logarithm ofthe virus concentration are greater,
Thermal stability. Maintain samples ofthe final lot offreeze-
than±O.3.
dried vaccine in the dry state at 37° for 7 days. Determine the
..virus.. concentration.as.described_underAssayJn.parallelfor 1i1!!!~II{l~{lccine (Live) RS issllitab1e for use asa reference
the vaccine held at 37° for 7 days and for vaccine stored at 2° preparation.
to 8°. For each component, the virus concentration of the Labelling. The label states (1) the strains of virus used in the
heated vaccine is not more than 1.0 loglo lower than that ofthe preparation ofthe vaccine; (2) the type and origin ofthe cells
unheated vaccine. used for the preparation ofthe vaccine; (3) the minimum virus

2414

www.webofpharma.com
IP 20ID MEASLES VACCINE (LIVE)

concentration for each component ofthe vaccine; (4) the time Extraneous agents (2.7.3). The working seed lot complies with
within which the vaccine must be used after reconstitution; the tests for seed lots.
(5) that the vaccine must not be given to a pregnant woman
Neurovirulence (2.7.5). The master/working seed lot complies
and that a woman should not become pregnant within two
with the test for neurovirulence oflive virus vaccines. Macaca
months after having the vaccine.
and Cercopithecus monkeys susceptible to measles virus are
suitable for the test.

Measles Vaccine (Live) PROPAGATION AND HARVEST


Measles Vaccine (Live) is a freeze-dried preparation of a All processing ofthe cell banle and subsequent cell cultures is
suitable attenuated strain of measles virus. The vaccine is done under aseptic conditions in an area where no other cells
reconstituted immediately before use, as stated on the label, are handled. Suitable animal (but not human) serum may be
to give a clear liquid that may be coloured owing to the used in the growth mediLUTI, but the final medilUTI for maintaining
presence of a pH indicator. cell growth during virus multiplication does not contain animal
serum. Serum and trypsin used in the preparation of cell
Production suspensions and culture media are shown to be free from
General provisions extraneous agents. The cell culture medium may contain a pH
indicator such as phenol red and suitable antibiotics at the
The production ofvaccine is based on a virus seed-lot system lowest effective concentration. It is preferable to have a
and, if the virus is propagated in human diploid cells, a cell- substrate free from antibiotics during production. Not less
bank system. Unless otherwise justified and authorized, the than 500 ml of the production cell culture is set aside as
virus in the final vaccine shall have undergone no more uninfected cell cultures (control cells). The viral suspensions
passages from the master seed lot than were used to prepare are harvested at a time appropriate to the strain ofvirus being
the vaccine shown in clinical studies to be satisfactory with used.
respect to abnormal toxicity and efficacy; even with authorized
exceptions, the number ofpassages beyond the level used for Only a single harvest that complies with the following tests
clinical studies shall not exceed five. may be used in the preparation of the final bulle vaccine.

The production method is validated to demonstrate that the Identification


product, if tested, would comply with the tests for abnormal
toxicity and efficacy. The single harvest contains virus that is identified as measles
virus by serum neutralisation in cell culture, using specific
Substrate for virus propagation antibody.
The virus is propagated in human diploid cells or in cultures Virus concentration. The virus concentration in the single
of chick embryo cells derived from a chicken flock free from harvest is determined as prescribed under Assay to monitor
specified pathogens. consistency ofproduction and to determine the dilution to be
SEED LOT used for the final bulle vaccine.

The strain of measles virus used in the production of measles Extraneous agents (2.7.3). Complies with the test for extraneous
vaccine shall be identified by historical records that include agents.
information on the origin of the strain and its subsequent
Control cells. Ifhuman diploid cells are used for production,
.manipulation. Virus seed lots are prepared in large quantities
the control cells comply with the test for identification and
and stored at temperatures below -20 0 iffreeze-dried, or below
extraneous agents.
-60 0 ifnot freeze-dried.
Only a seed lot that complies with the following tests may be
FINAL BULK VACCINE
used for virus propagation.
Virus harvests that comply with the above tests are pooled
Identification
and clarified to remove cells. A suitable stabilizer may be added
The master and working seed lots are identified as measles and the pooled harvests diluted as appropriate.
virus by serum neutralization in cell culture, using specific
Only a final bulle vaccine that complies with the following
antibodies.
requirements may be used in the preparation of the final lot.
Virus concentration. The virus concentration of the master
and working seed lots is determined to monitor consistency Sterility (2.2.11). The final bulle vaccine complies with the test
of production. for sterility carried out using 10 ml for each medium.

2415

www.webofpharma.com
· MEASLES VACCINE (LIVE) IP 2010

FINAL LOT concentration; (4) the time within which the vaccine must be
used after reconstitution.
A minimum virus concentration for release of the product is
established so as to ensure, in the light of stability data, that
the minimum concentration stated on the label will be present
at the end ofthe period of validity. Measles, Mumps and Rubella Vaccine
Only a final lot that complies with the tests for minimum virus (Live)
concentration for release, with the following requirement for Measles, Mumps and Rubella Vaccine (Live) is a freeze-dried
thermal stability and with each ofthe requirements given below preparation of suitabie attenuated strains of measles virus,
under Identification, Tests and Assay may be released for mumps virus and rubella virus grown in suitable cell cultures.
use. Provided that the test for bovine serum albumin has been
carried out with satisfactory results on the final bulk vaccine, The vaccine is reconstituted immediately before use to give a
it may be omitted on the final lot. clear liquid that may be coloured owing to the presence of a
pH indicator.
Thermal stability. Maintain samples ofthe final lot offreeze-
dried vaccine in the dry state at 37° for 7 days. Determine the Production
virus concentration as described under Assay in parallel for
the vaccine held at 37° for 7 days and for vaccine stored at 2° General provisions
to 8°. The virus concentration of the heated vaccine is not The three components are prepared as described in the
more than 1.0 10glO lower than that of the unheated vaccine. monographs on Measles Vaccine (Live), Mumps Vaccine (Live)
and Rubella Vaccine (Live) and comply with the tests
Identification prescribed therein.
When the vaccine reconstituted as stated on the label is mixed
with specific measles antibodies, it is no longer able to infect The production method is validated to demonstrate that the
susceptible cell cultures. product, if tested, would comply with the test for safety and
efficacy.
Tests
Water (2.3.43). Not more than 3.0 per cent, determined by Karl FINAL BULK VACCINE
Fischer, semi-micro determination ofwater or by any suitable Virus harvests for each component are pooled and clarified to
validated method. remove cells. A suitable stabilizer may be added and the pooled
Sterility ( 2.2.11). The reconstituted vaccine complies with harvests diluted as appropriate. Suitable quantities of the
the test for sterility. pooled harvest for each component are mixed.
Abnormal toxicity (2.2.1). Complies with the test for abnormal
Only a final bulk vaccine that complies with the following
toxicity.
requirements may be used in the preparation of the final lot.
Bovine serum albumin. Not more than 50 ng per single human
dose, determined by a suitable immunochemical method Sterility (2.2.11). Carry out the test for sterility using 10 ml for
(2.2.14). each medium.
Assay FINAL LOT
Titrate the vaccine for infective virus at least in triplicate,
For each component, a minimum virus concentration for
using at least five cell cultures for each 0.5 log,o dilution step
release of the product is established such as to ensure, in the
or by a method of equal precision. Use an appropriate virus
light of stability data, that the minimum concentration stated
reference preparation to validate each assay. The estimated
on the label will be present at the end ofthe period ofvalidity.
virus concentration is not less than that stated on the label;
the minimum virus concentration stated on the label is not Only a final lot that complies with the tests for minimum virus
less than 1 x 103 CCID50 per human dose. The assay is not concentration of each component for release, with the
valid ifthe confidence limits (P =:= 0.95) ofthe logarithm ofthe following requirement for thermal stability and with each of
virus concentration is greater than ± 0.3. the requirements given below under Identification and Tests
!lfeq§[?§.yq(::J:}I1?(LiJl?)RS. iil §Jl1tl:ipl~ IQr.1J§J:l . .l:iil.. l:i I~lfl.r~!1<::fl may be released for use. Provided that the test for bovine
preparation. serumalbumiIilias been -carrIed out WIth .satIsfactoryfesu1ts
on the final bulle va.ccine, itmay be omitted on the final lot.
Labelling. The label states (1) the strain of virus used for the
preparation ofthe vaccine; (2) the type and origin ofthe cells Thermal stability. Maintain samples ofthe final lot offreeze-
used for the preparation ofthe vaccine; (3) the minimum virus dried vaccine in the dry state at 37° for 7 days. Determine the

2416

www.webofpharma.com
IP 2010 MENINGOCOCCAL POLYSACCHARIDE VACCINE

virus concentration as described under Assay in parallel for not valid ifthe confidence limits (P = 0.95) ofthe logarithm of
the vaccine held at 37° for 7 days and for vaccine stored at 2° the virus concentration are greater than ± 0.3.
to 8°. The virus concentration of the heated vaccine is not
Mumps vaccine (Live) RS is suitable for use as a reference
more than 1.0 loglo lower than that of the unheated vaccine.
preparation.
Identification C. Mix the vaccine with a sufficient quantity of antibodies
specific for mumps virus and measles virus. Titrate the vaccine
When the vaccine reconstituted as stated on the label is mixed
for infective rubella virus at least in triplicate, using at least
with antibodies specific for measles virus, mumps virus and
eight cell cultures for each dilution 0.510g lO step or by a method
rubella virus, it is no longer able to infect cell cultures
of equal precision. Use an appropriate virus reference
susceptible to these viruses. When the vaccine reconstituted
preparation to validate each assay. The estimated rubella virus
as stated on the label is mixed with quantities of specific
concentration is not less than that stated on the label; the
antibodies sufficient to neutralize any two viral components,
minimum rubella virus concentration stated on the label is not
the third viral component infects susceptible cell cultures.
less than 1x103 CCIDso per single human dose. The assay is
Tests not valid ifthe confidence limits (P = 0.95) ofthe logarithm of
the virus concentration are greater than ± 0.3.
Water (2.3.43). Not more than 3.0 per cent, determined by Karl
Rubella vaccine (Live) RS is suitable for use as a reference
Fischer, semi-micro determination ofwater or by any suitable
preparation.
validated method.
Labelling. The label states (1) the strains ofvirus used in the
Sterility (2.2.11). The reconstituted vaccine complies with the
preparation of the vaccine; (2) the type and origin of the cells
test for sterility.
used for the preparation ofthe vaccine; (3) the minimum virus
Abnormal toxicity (2.2.1). Complies with the test for abnormal concentration for each component ofthe vaccine; (4) the time
toxicity. within which the vaccine must be used after reconstitution;
(5) that the vaccine must not be given to a pregnant woman
Bovine serum albumin. Not more than 50 ng per single human
and that a woman should not become pregnant within two
dose, determined by a suitable immunochemical method
months after having the vaccine.
(2.2.14).

Assay
A. Mix the vaccine with a sufficient quantity of antibodies Meningococcal Polysaccharide
specific for mumps virus and rubella virus.Titrate the vaccine Vaccine
for infective measles virus at least in triplicate, using at least
eight cell cultures for each dilution 05 10glO step or by a method Meningococcal Polysaccharide Vaccine is a freeze-dried
of equal precision. Use an appropriate virus reference preparation of one or more purified capsular polysaccharides
preparation to validate each assay. The estimated measles obtained from one or more suitable strains of Neisseria
virus concentration is not less than that stated on the label; meningitidis group A, group C, group Y and group W135 that
the minimum measles virus concentration stated on the label are capable ofconsistently producing polysaccharides known
is not less than 1xl0 3 CCIDso per single human dose. The to be safe and effective in man.
assay is not valid if the confidence limits (P = 0.95) of the N. meningitidis group A polysaccharide consists of partly
logarithm ofthe virus concentration are greater than ± 0.3. O-acetylated repeating units ofN-acetylmannosamine, linked
Measles vaccine (Live) RS is suitable for use as a reference with 1&n6 phosphodiester bonds.
preparation.
N. meningitidis group C polysaccharide consists of partly
B. Mix the vaccine with a sufficient quantity of antibodies O-acetylated repeating units of sialic acid, linked with 2&n9
specific for measles virus and rubella virus. Titrate the vaccine glycosidic bonds.
for infective mumps virus at least in triplicate, using at least
N. meningitidis group Y polysaccharide consists of partly
eight cell cultures for each dilution 0.510g 10 step or by a method
O-acetylated alternating units of sialic acid and D-glucose,
of equal precision. Use an appropriate virus reference
linked. with 2&n6 and 1&n4 glycosidic bonds.
preparation to validate each assay. The estimated mumps virus
concentration is not less than that stated on the label; the N. meningitidis group W135 polysaccharide consists ofpartly
minimum mumps virus concentration stated on the'label is not O-acetylated alternating units of sialic acid and D-galactose,
less than 5 x 103 CCIDso per single human dose. The assay is linked with 2&n6 and 1&n4 glycosidic bonds.

2417

www.webofpharma.com
MENINGOCOCCAL POLYSACCHARIDE VACCINE IF 2010

Production proteins and lipopolysaccharides. The purification step


consists of ethanol precipitation of the polysaccharides or
General provisions purification with chloroform and n-butanol or by cold phenol
Production of the meningococcal polysaccharides is based treatment, which are then dried and stored at or below -20°.
on a well defined seed-lot system. The method of production The loss on drying is detennined by thennogravimetry, Karl
shall have been shown to yield consistently meningococcal Fischer or any other suitable method and the value is used to
polysaccharide vaccines of satisfactory immunogenicity and calculate the results ofthe other chemical tests with reference
safety for man. . to the dried substance.
The production method is validated to demonstrate that the Only purified polysaccharides that tested comply with the
product, if tested, would comply with the test of abnonnal following requirements may be used in the preparation ofthe
toxicity for antisera and vaccines. final bulle vaccine.
SEED LOT Protein (2.7.1). Not more than 10 mg ofprotein per gram of
purified polysaccharide for group A and C organisms and less
The strains of N. meningitidis used for the master seed lots than 50 mg ofprotein per gram ofpolysaccharide for group Y
shall be identified by historical records that include infonnation and W135 calculated using bovine plasma albumin as a
on their origin and by their biochemical, serological, reference or other methods approved by National Regulatory
physicochemical or molecular characteristics. Cultures from Authority.
the working seed lot shall have the same characteristics as the
strain that was used to prepare the master seed lot. The strains Nucleic acids (2.7.1). Not more than 10 mg ofnucleic acids per
have the following characteristics: gram ofpurified polysaccharide, calculated with reference to
the dried substance.
a) Colonies obtained from a culture are round, unifonn in
shape and smooth with a mucous, opalescent, greyish O-Acetyl groups (2.7.1). Not less than 2 mmol ofO-acetyl
appearance. groups per gram of purified polysaccharide for group A, not
b) Gram staining reveals characteristic Gram-negative less than 1.5 mmol per gram of polysaccharide for group C,
diplococci in 'coffee-bean' arrangement. not less than 0.3 romol per gram ofpolysaccharide for groups
Y and W135, all calculated with reference to the dried
c) The oxidase test is positive.
substance.
d) The culture utilizes glucose and maltose.
Phosphorus (2.7.1). Not less than 80 mg of phosphorus per
e) Suspensions of the culture agglutinate with specific
gram of group A purified polysaccharide, calculated with
antisera oflmown titre.
reference to the dried substance.
PROPAGATION AND HARVEST Sialic acid (2.7.1). Not less than 800 mg ofsialic acid per gram
The working seed lots are cultured on solid media that do not of group C polysaccharide and not less than 560 mg of sialic·
contain blood-group substances or ingredients of mammalian acid per gram of purified polysaccharide for groups Y and
origin. The inoculum may undergo one or more subcultures in W135, all calculated with reference to the dried substance.
liquid medium before being used for inoculating the final Use the following reference solutions:
medium. The liquid media used and the final medium are Group C polysaccharide. A 150 mg/l solution of N-
semisynthetic and free from substances precipitated by acetylneuraminic acid.
cetrimonium bromide (hexadecyltrimethylammonium
Group Ypolysaccharide. A solution containing 95 mg/l of N-
bromide) and do not contain blood-group substances or high-
acetylneuraminic acid and 55 mg/l of glucose.
molecular-mass polysaccharides. The bacterial purity of the
culture is verified by microscopic examination ofGram-stained Group W135 polysaccharide. A solution containing 95 mg/l
smears and by inoculation into appropriate media. The cultures of N-acetylneuraminic acid and 55 mg/l of galactose.
are centrifuged and the polysaccharides precipitated from the Calcium. If a calcium salt is used during purification,
supernatant by addition of cetrimonium bromide. The determination of calcium is carried out on the purified
precipitate obtained is harvested and may be stored at or polysaccharide by a suitable method; the content is within
below -20° awaiting further purification. the limits approved for the product.

PURIFIEDPOINSACCHARIDES - Molecular size. Examinebygel.filtrationor.highp_erfonnance


size-ex_clusion. chrOInatography (HPSEC) (2.4.16), using
The polysaccharides are purified, after dissociation of the agarose for chromatography or cross-linked agarose for
complex of polysaccharide and cetrimonium bromide, using chromatography either alone or in combination with light
suitable procedures to remove successively nucleic acids, scattering and refractive index detector (e.g. multiple angle

2418

www.webofpharma.com
IP 2010 MENINGOCOCCAL POLYSACCHARIDE VACCINE

LASER light scattering, MALLS) or any other suitable method. Tests


Use a column 0.9 m x 15 mm equilibrated with a solvent having
an ionic strength of0.2 mollkg and a pH of7.0 to 7.5. Apply to Sterility (2.2.11). Complies with the test for sterility.
the column about 2.5 mg of polysaccharide in a volume of Abnormal toxicity (2.2.1). Complies with the test for abnonnal
about 1.5 ml and elute at about 20 mllh. Collect fractions of toxicity.
about 2.5 ml and detennine the content of polysaccharide by
a suitable method. Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
each of the rabbit with lml per kg body weight of solution
At least 65.0 per cent ofgroup A polysaccharide, 75.0 per cent containing
of group C polysaccharide, 80.0 per cent of group Y
a) 0.025 Ilg ofpolysaccharide for a monovalent vaccine,
polysaccharide and 80.0 per cent of group W135
polysaccharide is eluted before a distribution coefficient (Ko) b) 0.050 Ilg ofpolysaccharide for a bivalent vaccine,
of 0.50 is reached. In addition, the percentages eluted before c) 0.075 Ilg ofpolysaccharide for a trivalent vaccine,
this distribution coefficient are within the limits approved for
the particular product. d) 0.100 Ilg ofpolysaccharide for a tetravalent vaccine.
Water (2.3.43). Not more than 3.0 per cent, ofmoisture content
Identification and serological specificity by thennogravimetery, Karl Fischer or any other suitable
The identity and serological specificity are detennined by a method.
suitable immunochemical method (2.2.14). Identity and purity Molecular size. Examine by gel filtration or size-exclusion
of each polysaccharide shall be confinned; it shall be shown chromatography (2.4.16). Use a column about 0.9 m long and
that there is not more than LO per cent mlm of group- 16 mm in internal diameter equilibrated with a solvent having
heterologous N. meningitidis polysaccharide. an ionic strength of0.2 mollkg and a pH of7.0 to 7.5. Apply to
the column about 2.5 mg of each polysaccharide in a volume
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
ofabout 1.5 ml and elute at about 20 ml/h. Collect fractions of
each of the rabbit with 1ml per kg body weight of solution
about 2.5 ml and detennine the content of polysaccharide by
containing
a suitable method.
a) 0.025 Ilg ofpolysaccharide for a monovalent vaccine,
Use cross-linked agarose for chromatography and apply a
b) 0.050 Ilg ofpolysaccharide for a bivalent vaccine, suitable immunochemical method (2.2.14) to establish the
c) 0.075 Ilg ofpolysaccharide for a trivalent vaccine, elution pattern ofthe different polysaccharide(s). The vaccine
complies with the test if:
d) 0.100 Ilg ofpolysaccharide for a tetravalent vaccine.
a) 65.0 per cent of Group A polysaccharide is eluted before
FINAL BULK VACCINE KoofO.50,
One or more purified polysaccharides of one or more N. b) 75.0 per cent of Group C polysaccharide is eluted before
meningitidis groups are dissolved in a suitable solvent that ~of0.50,
may contain a stabilizer. c) 80.0 per cent ofGroup Y & W135 polysaccharide is eluted
Only a final bull( vaccine that complies with the following before Ko ofO.50.
requirement may be used in the preparation of the final lot. For a tetravalent vaccine (group A + group C + group Yand
group W135), use cross linked agarose for chromatography
Sterility (2.2.11). Carry out test for sterility using 10 ml of
R1 and apply a suitable immunochemical method (2.2.14) to
bulk for each sterility medium.
establish the elution pattern of the different polysaccharides.
FINAL LOT The vaccine complies with the test ifKo for the principal peak
is
The final bull( vaccine is distributed aseptically into sterile
a) not greater than 0.70 for group A and group C
containers. The containers are then closed so as to avoid
polysaccharide,
contamination. Only a final lot that is satisfactory with respect
to each of the requirements prescribed below under b) not greater than 0.57 for group Y polysaccharide,
Identification, Tests and Assay may be released for use. c) not greater than 0.68 for group W135 polysaccharide.

Identification Assay
Carry out an identification test for each polysaccharide present Carry out an assay as stated under each polysaccharide
in the vaccine by a suitable immunochemical method (2.2.14). present in the vaccine.

2419

www.webofpharma.com
MUMPS VACCINE (LIVE) - IP 2010

For a divalent vaccine (group A + group C), use measurement and its subsequent manipulation. To avoid unnecessary use
of phosphorus (2.7.1) to determine the content of ofmonkeys in the test for neurovirulence, Virus seed lots are
polysaccharide A and measurement of sialic acid (2.7.1) to prepared in large quantities and stored attemperatures below
detennine the content ofpolysaccharide C. To detennine sialic -20 0 iffreeze-dried, or below -60 0 ifnot freeze-dried.
acid, use as reference solution a 150 mg/l solution of N-
Only a seed lot that complies with the following tests may be
acetylneuraminic acid. used for virus propagation.
For a tetravalent yaccine (group A + group C + group Y +
group W135) a suitable immunochemical method (2.2.14) is Identification
used with a reference preparation of purified polysaccharide
for each group. The master and working seed lots are identified as mumps
virus by serum neutralisation in cell culture, using specific
The vaccine contains not less than 70.0 per cent and not more
antibodies. .
than 130.0 per cent of the quantity of each polysaccharide
stated on the label. Virus concentration. The virus concentration of the master
Labelling. The label states (1) the group or groups of and working seed lots is detennined to ensure consistency of
polysaccharides (A, C, Y or W135) present in the vaccine; production.
(2) the number of Ilg of polysaccharide per human dose. Extraneous agents (2.7.3). The working seed lot complies with
the tests for seed lots.
Neurovirulence (2.7.5). The master/working seed lot complies
Mumps Vaccine (Live) with the test for neurovirulence oflive virus vaccines. Macaca
Mumps Vaccine (Live) is a freeze-dried preparation ofa suitable and Cercopithecus monkeys are suitable for the test.
attenuated strain ofmumps virus. The vaccine is reconstituted
immediately before use to give a clear liquid that may be PROPAGATION AND HARVEST
coloured owing to the presence of a pH indicator.
All processing ofthe cell bank and subsequent cell cultures is
done under aseptic conditions in an area where no other cells
Production
are handled. Suitable animal (but not human) serum may be
General provisions used in the growth media. Serum and trypsin used in the
The production ofvaGcine is based on a virus seed-lot system preparation of cell suspensions and culture media are shown
and, if the virus is propagated in human diploid cells, a cell- to be free from extraneous agents. The cell culture medium
bank system. The production method shall have been shown may contain a pH indicator such as phenol red and suitable
to yield consistently live mumps vaccines of adequate antibiotics at the lowest effective concentration. It is preferable
immunogenicity and safety in man. to have a substrate free from antibiotics during production.
Not less than 500 ml ofthe production cell culture is set aside
Unless otherwise justified and authorised, the virus in the
as uninfected cell culture (control cells). The viral suspensions
final vaccine shall have undergone no more passages from
are harvested at a time appropriate to the strain ofvirus being
the master seed lot than were used to prepare the vaccine
shown in clinical studies to be satisfactory with respect to used.
safety and efficacy even with authorized exceptions, the Only a single harvest that complies with the following
number ofpassages beyond the level used for clinical studies requirements may be used in the preparation of the final bulk
shall not exceed five. vaccine.
The production method is validated to demonstrate that the
product, if tested, would comply with the tests for safety and Identification
efficacy.
The single harvest contains virus that is identified as mumps
Substrate for virus propagation virus by serum neutralization in cell culture, using specific
antibodies.
The virus is propagated in human diploid cells or in primary
cultures of chick embryo cells derived from a chicken flock Virus concentration. The virus concentration in the single
J!eeJ!0Il1 spe.cifi~~path.0gens. harvest is detennined as prescribed under Assay to monitor
conslstencyofprocfiiCiiOn-ancrfodeteriiilneThediliition fO-be
SEED LOT used fof the fmal bulk vaccine.
The strain ofmumps virus used shall be identified by historical Sterility (2.2.11). Single harvest complies with sterility test
records that include infonnation on the origin of the strain should be processed further.

2420

www.webofpharma.com
IP 2010 PERTUSSIS VACCINE

Control cells. The control cells comply with a test for or by a method of equal precision. Use an appropriate virus
extraneous agents (2.7.3). reference preparation to validate each assay. The estimated
virus concentration is not less than that stated on the label;
FINAL BULK VACCINE the minimum virus concentration stated on the label is not
Single harvests that comply with the above tests are pooled less than 5 x 103 CCIDso per human dose. The assay is not
and clarified to remove cells. A suitable stabilisermay be added valid ifthe confidence limits (P = 0.95) ofthe logarithm ofthe
and the pooled harvests diluted as appropriate. virus concentration is greater than ± 0.3.

Only a final bulle vaccine that complies with the following Mumps vaccine (Live) RS is suitable for use as a reference
requirements may be used in the preparation ofthe final lot. preparation.
Sterility (2.2.11). The final bulle vaccine complies with the test Labelling. The label states (1) the strain of virus used for the
for sterility, carTied out using 10 ml for each medium. preparation of the vaccine; (2) the type and origin ofthe cells
FINAL LOT used for the preparation ofthe vaccine; (3) the minimum virus
concentration and; (4) the time within which the vaccine must
A minimum virus concentration for release of the product is be used after reconstitution.
established such as to ensure, in the light of stability data,
that the minimum concentration stated on the label will be
present at the end of the period of validity.
, Only a fma110t that complies with the tests for minimum virus Pertussis Vaccine
concentration for release, with the following requirement for
thermal stability and with each ofthe requirements given below Pertussis Vaccine is a sterile saline suspension of inactivated
under Identification and Tests and Assay may be released for whole cells of one or more strains of Bordetella pertussis.
use. Provided that the test for bovine serum albumin has been
carried out with satisfactory results on the final bulle vaccine, Production
it may be omitted on the fmallot. General provisions
Thermal stability. Maintain samples ofthe final lot offreeze-
dried vaccine in the dry state at 37° for 7 days. Determine the Inactivated B. pertllssis suspension
virus concentration as described under Assay in parallel for
the vaccine held at 37° for 7 days and for vaccine stored at 2° Production is based on a seed-lot system. One or more strains
to go. The virus concentration of the vaccine exposed to 37° ofB. pertussis with Imown origin and history are used. Strains,
for 7 days is not more than 1.0 10glO lower than that of the culture medium and cultivation method are chosen in such a
unheated vaccine. way that agglutinogens 1, 2 and 3 are present in the final
vaccine. Each strain is grown for 24 to 72 hours in a liquid
Identification medium or on a solid medium; the liquid medium used in the
final cultivation stage does not contain blood or blood
When the vaccine is reconstituted as stated on the label is
mixed with specific mumps antibodies, it is no longerable to products. Human blood or blood products are not used in any
infect susceptible cell cultures. culture media. The bacteria are harvested, washed to remove
substances derived from the medium and suspended in a
Tests 0.9 per cent w/v solution of sodium chloride or other suitable
isotonic solution. The opacity ofthe suspension is deterrriined
Water (2.3.43). Not more than 3.0 percent, determined by Karl
not later than 2 weeks after harvest by comparison with the
Fischer, semi-micro determination ofwater or by any suitable
reference preparation of Opacity and used as the basis of
validated·method.
calculation for subsequent stages in vaccine preparation.
Sterility ( 2.2.11). Complies with the test for sterility. Single harvests are not used for the final bulle vaccine unless
Abnormal toxicity (2.2.1). Complies with the test for abnormal they have been shown to contain B. pertussis cells with the
toxicity. same characteristics with regard to growth and agglutinogens,
Bovine serum albumin. Not more than 50 ng per single human as the parent strain and to be free from contaminating bacteria
dose, determined by a suitable immunochemical method and fungi. The bacteria are lalled and detoxified in controlled
(2.2.14). conditions by means ofa suitable chemical agent or by heating
or by a combination of these methods. Freedom from live B.
Assay
pertussis is tested using a suitable culture medium. The
Titrate the vaccine for infective virus at least in triplicate, suspension is maintained at 5 ± 3° for a suitable period to
using at least five cel! cultures for each 0.5 10glO dilution step diminish its toxicity.

2421

www.webofpharma.com
PERTUSSIS VACCINE IP 2010

FINAL BULK VACCINE (b) at the end of 7 days the average increase in mass per
vaccinated mouse is not less than 60.0 per cent of that per
Suitable quantities ofthe inactivated single harvests are pooled
control mouse; and (c) not more than 5.0 per cent of the
to prepare the final bulk vaccine. Suitable antimicrobial
vaccinated mice die during the test. The test may be repeated
preservatives may be added. The bacterial concentration of
and the results of the tests combined.
the final bulle vaccine does not exceed that con-esponding to
an ~pacity of20 ill per single human dose. If2 or more strains Free formaldehyde (2.3.20). Maximum 0.2 gil.
of B. pertussis are used, the composition of consecutive lots Antimicrobial preservative. Where applicable, determine the
of the final bulle vaccine shall be consistent with respect to amount of antimicrobial preservative by a suitable chemical
the proportion of each strain as measured in opacity units. method. The content is not less than 85.0 per cent and not
Only a final bulle vaccine that complies with the following more than 115.0 per cent of the intended amount.
requirements may be used in the preparation of the fmallot. Sterility (2.2.11). Complies with the test for sterility.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical Assay
method. The amount is not less than 85.0 per cent and not Carry out the assay of Pertussis Vaccine as described below:
greater than 115.0 per cent of the intended amount.
Biological assay of pertussis vaccine
Sterility (2.2.11). Carry out test for sterility using 10 ml of
bulle for each sterility medium. The potency of PertussisVaccine is detennined by comparison
of the dose necessary to protect mice against the effects of a
FINAL LOT lethal dose of B. pertussis challenge culture, administered
The final bulle vaccine is distributed aseptically into sterile, intracerebrally, with the dose of a reference preparation,
tamper-proof containers. The containers are closed so as to calibrated in Internatiqnal Units, required to give the same
prevent contamination. level of protection. For this comparison, the Standard
preparation of Pertussis vaccine & a suitable strain of B.
Only a final lot that is satisfactory with respect to each of the pertussis (e.g.18323, to be used as challenge strain), are
requirements given below under Identification, Tests and required.
Assay may be released for use. Provided the tests for specific
toxicity, free formaldehyde and antimicrobial preservative and Reference preparation
the assay have been can-ied out with satisfactory results on
The reference preparation is an International standard of
the final bulle vaccine, they may be omitted on the final lot.
Pertussis vaccine, consisting of a freeze dried vaccine or
Identification another suitable preparation, calibrated in comparison to
International standard, from time to time. The International
Identify pertussis vaccine by agglutination of the bacteria in Unit is the activity contained in a stated amount of the
the vaccine by antisera specific to B. pertussis. International standard, which consists of a quantity of dried
Tests pertussis vaccine. The equivalence in International Units of
the International Standard is stated by the World Health
Specific toxicity Organization.
Use not less than 10 healthy mice each weighing between 14 Use healthy mice of a suitable strain, weighing between 13
to 16 g for the vaccine group and for the saline control. Use and 16 g from the same stock. Distribute the mice randomly, in
mice of the same sex or distribute males and females equally six to eight groups of not less than 16 and not more than 24
between the groups. Allow the animals access to food and and four groups of 10. The mice should all be ofthe same sex
water for at least 2 hours before injection and during the test. or the males and females should be distributeq equally between
Inject each mouse ofthe vaccine group intraperitoneally with the groups. Half of the groups of 16 to 24 should receive the
0.5 ml, containing a quantity of the vaccine equivalent to not reference preparation and the other half should receive the
less than halfthe single human dose. Inject each mouse ofthe vaccine under examination. The four groups of 10 each should
control group with 0.5 ml ofa 0.9 per cent w/v sterile solution be used for the LD 50 titration of challenge suspension.
of sodium chloride, preferably containing the same amount
ofantimicrobial-preseFVativeas-that-injected-with-the-vaccine. Us_eat le.ast, threedill!tiOl1s.QftheI.ef.eLell~ Yflf(;il}.eJ:t!1<:l§i@~I
Weigh the groups ofmice-immediately before the injection, dilutions of the vaccine under examination. In each case the
72 hours and 7 days after the injection. The vaccine complies dilutions are so selected that the dilution protecting 50 per
with the test if(a) at the end of 72 h the total mass_ofthe group cent ofthe mice (ED 50) is as near as possible to middle ofthe
ofvaccinated mice is not less than that preceding the injection; dilution range. For example, suggested ~ilutions are (1/8,1/40

2422

www.webofpharma.com
IP 2010 PLAGUE VACCINE

and 1/200 ofthe human dose ofthe vaccine under examination) than 4.0 ru per single human dose and the lower fiducial limit
and (0.5 IV, 0.1 IV and 0.02 IV or any other suitable (P = 0.95) of estimated potency is not less than 2.0 ru.
standardized dilutions, ofthe reference preparation), each dose The test may be repeated once, but when more than one test
being contained in a volume, not exceeding 0.5 ml. For each is performed the results ofall valid tests must be combined, in
dilution use 16 to 24 mice and inject intraperitoneally, into the estimate of potency.
each mouse one dose of the dilution.
Labelling. The label states (1) the minimum number of
Select a suitable strain ofB. pertussis (e.g. 18323), capable of International Units per single human dose; (2) that the vaccine
causing the death of mice within 14 days of intracerebral must be shaken before use; (3) that the vaccine is not to be
injection. Make two subcultures after reviving the strain on a frozen.
suitable medium (e.g. E.G. medium) and suspend the harvested
growth in a solution containing 1 per cent w/v of casein
hydrolysate (e.g. casamino acid) and 0.85 per cent w/v of
sodium chloride and having a pH of 7.0 to 7.2 or in another
Plague Vaccine
suitable solution. Determine the opacity ofthe suspension by Fonnolised Plague Vaccine
using 5th International reference preparation for opacity (10
aU) and/or spectrophotometrically. Alternatively, aliquots of Plague Vaccine is a sterile suspension of killed plague bacilli,
challenge suspension frozen in liquid nitrogen with a suitable Yersinia pestis, ofthe 195/P strain grown in a suitable enriched
preservative like 10 per cent DMSO may be used, to avoid medium such as acid hydrolysate of casein, harvested and
heterogenicity. After 14 to 17 days of immunization, Inject killed by the addition offormaldehyde. It may contain a suitable
intracerebrally, a dose of 0.02 to 0.03 ml of the challenge preservative.
dilution randomly, into each immunized mouse. The challenge Plague Vaccine contains in each ml of human dose not less
should contain, approximately 1,00,000 organisms and 100 to than 250 mouse median effective irnmunising doses (250 ED50)'
1000 LD50 per dose, in a volume ofnot more than 0.03 ml. In the Description. A turbid, golden yellow or brownish liquid with
same way, inject 4 groups of 10 control mice each, for LD 50 or without flakes or clumps.
titration of challenge preparation, prepared by a series of
dilutions, from the dilution selected for challenge. The Tests
challenge should be completed within 2 to 2.5 hours of
pH (2.4.24). 6.8 to 7.4.
preparation. Exclude any mouse from consideration, that dies
within 3 days ofchallenge. Count the number ofmice surviving Sterility (2.2.11). Complies with the tests for sterility.
in each ofthe groups, after 14 days. On the basis ofthe numbers Abnormal toxicity (2.2.1). Complies with the test for abnormal
of animals surviving in each of the groups of 16 to 24 mice, toxicity but injecting into each mouse 1.0 ml and into each
calculate the potency of vaccine under examination, against guinea-pig 3.0 ml.
the potency of reference preparation. Seed a suitable highest
Potency. Carry out the biological assay of plague vaccine
dilution of the challenge suspension, into each of two B.G
described below.
medium plates, before and after challenge. Incubate the plates
at 37° for 48 to 72 hours and calculate the number of colony Biological Assay of Plague Vaccine
forming units (CFUs).
The potency of plague vaccine is estimated by determining
Calculate the potency of the vaccine by Probit analysis and
the dose necessary to protect mice against a lethal dose of a
, LD 50 of the challenge suspension by Reed and Munch
virulent strain of Yersinia pestis.
Method.
Test animals. Use white mice, 6 to 7 weeks old, each weighing
The test is not valid unless: a) for both the vaccine under
between 20 and 28 g and of a strain susceptible to plague
examination and the reference preparation, the ED 50 (protective
infection. The selected strain of mice should be such that an
dose), lies between the largest and the smallest doses given
infective dose of 6 to 12 organisms of a virulent strain of Y.
to the mice; b) the number of animals, which die in the four
pestis per mouse given subcutaneously produces a mortality
groups of 10 injected with the challenge suspension and its
of not less than 80 per cent of the animals used. The animals
dilutions indicate that the challenge dose contains 100 to 1000
should be healthy and free from intercurrent infection with
LD 50 and 1 LD 50 contains not more than 300 colony forming
organisms such as Salmonella.
units; c) and the statistical analysis shows no deviation from
linearity or parallelism, in terms ofsignificance ofthe scope of Suggested Method
dose response curve; d) the vaccine passes the requirements
for potency, if the test results of a statistically valid assay Selection of suitable virulent strain. A freeze-dried virulent
show that the estimated potency of the vaccine is not less culture of Y. pestis, established to be suitable for challenge, is

2423

www.webofpharma.com
PLAGUE VACCINE IP 2010

revived by subculturing 0.5 ml in 9.5 ml of nutrient broth levels of specific antibodies in man. It contains upto 23
contained in another test-tube and incubating at 28° for exactly immunochemically different capsular polysaccharides listed
48 hours. Such a culture should contain 300 to 600 million in the Table 1.
organisms per ml. Make lO-fold dilutions in nutrient broth
and test for virulence. Use a 10-7 dilution containing 6 to 12 Production
organisms in 0.2 ml as the test infective dose per animal. Those
strains which produce a mortality of 80 per cent among the General provisions
animals tested with this dose are considered to be virulent. Production ofthe vaccine is based on a well defined seed-lot
Standard challenge dose. FresWy reconstitute the freeze-dried system for each type. The production method shall have been
culture and dilute with nutrient broth to a strength such that shown to yield consistently pneumococcal polysaccharide
0.2 ml contains 60 to 120 organisms. vaccines of acceptable immunogenicity and safety in man.
Measurement of protective power. Prepare a series of five The production method is validated to demonstrate that the
graded doses of the preparation under examination arranged product, if tested, would comply with the tests for abnormal
in such a manner that the 50 per cent protective dose (ED so) toxicity of vaccines for human use, modified as follows for
lies about the middle of the selected series. For each dose a the test on guinea-pig; inject 10 human dose into each guinea-
batch of 16 mice is used. Inject subcutaneously the selected pig and observe for 12 days.
dose in two equal parts with an interval of 7 days between
them. Seven days after the second half of the dose, inject Monovalent bulk polysaccharides
subcutaneously into each group ofmice the standard challenge
The bacteria are grown in a suitable liquid medium that does
dose. At the same time inject into 10 control mice, 8 to 9 weeks
not contain blood-group substances or high-molecular-mass
old and weighing between 28 and 30 g, the standard challenge
polysaccharides. The bacterial purity ofthe culture is verified
dose.
and the culture is inactivated with phenol. Impurities are
Observe the animals for 15 days and record the number of removed by such techniques as fractional precipitation,
deaths in each group. Carry out a post-mortem on the dead enzymatic digestion and ultrafiltration. The polysaccharide is
animals and look for signs of plague in them. If plague obtained by fractional precipitation, washed, and dried in a
organisms are not seen, such deaths are excluded from the vacuum to a residual moisture content shown to be favourable
calculation. The test is not valid unless the number of such to the stability of the polysaccharide. The residual moisture
deaths is not more than 1. At the end of the period of content is determined by drying under reduced pressure over
observation kill all the surviving animals and examine for signs diphosphorus pentoxide or by thermogravimetric analysis and
of plague. Calculate the median effective immunising dose, the value obtained is used to calculate the results of the tests
ED so ' by standard statistical methods. The vaccine passes the shown below with reference to the dried substance. The
test ifit has an ED so of 0.004 ml or less per mouse. monovalent bulk polysaccharide is stored at a suitable
Storage. Store at a temperature between 2° and 8° . The vaccine temperature in conditions that avoid the uptake of moisture.
should not be allowed to freeze. When stored under the Only a monovalent bulk polysaccharide that complies with
prescribed conditions the vaccine is expected to retain its the following requirements may be used in the preparation of
potency for not less than 3 years from the date of initiation of the final bulk vaccine. Percentage contents of components,
the potency test. determined by the methods prescribed below, are shown in
Labelling. The label states (1) ED so unitage per dose; (2) the the Table 1.
name and proportion of any preservative added; (3) that the The purified polysaccharides comply with the following tests
contents should be shaken well before use; (4) that the vaccine as applicable:
should not be allowed to freeze. Protein (2.7.1). Comply with the test for protein.
Nucleic acids (2.7.1). Comply with the test for nucleic acids.
Pneumococcal Polysaccharide Vaccine Total nitrogen (2.3.30). Comply with the test for total nitrogen.

Pneumococcal Polysaccharide Vaccine consists of a mixture Phosphorus (2.7.1). Comply with the test for phosphorus.
Qfequal.. partsofpJU'ifiedcaps.ular polysaccharide ofvarious Uronicacids.(2.7. L).Comply.with.the.test-for.uronic.acids..--
serotype antigens prepared from suitableJ')Clt}:1()g~I!ic_§.tl'llil1§
Hexosamine (2.7.1). Comply with the test for hexosamine.
of Streptococcus pneumoniae in different desired
combinations whose capsules have been shown to be made Methylpentoses (2.7.1). Comply with the test for
up ofpolysaccharides that are capable ofinducing satisfactory methylpentoses.

2424

www.webofpharma.com
IP 2010 PNEUMOCOCCAL POLYSACCHARIDE VACCINE

Table 1- Specifications on monovalent bulk polysaccharides (per cent contents):


Molecular Proteins Nucleic Total Phosphorus Molecular size Ko Uronic Hexo- Methyl- O-acetyl
Type* acids Nitrogen CL-4B** CL-2B*** acids sammes pentoses Groups
1 :::;2 :::;2 3.5-6 0-1.5 :::;0.15 ;;::45 ;;:: 1.8
2 :::;2 :::;2 0-1 0-1.0 :::;0.15 ;;:: 15 ;;::38
3 :::;5 :::;2 0-1 0-1.0 :::;0.15 ;;::40
4 :::;3 :::;2 4-6 0-1.5 :::;0.15 ;;::40
5 :::;7.5 :::;2 2.5-6.0 :::;2 :::;0.60 ;;::12 ;;::20
6B :::;2 :::;2 0-2 2.5-5.0 :::;0.50 ;;:: 15
7F :::;5 :::;2 1.5-4.0 0-1.0 :::;0.20 ;;::13
8 :::;2 :::;2 0-1 0-1.0 :::;0.15 ;;::25
9N :::;2 :::;1 2.2-4 0-1.0 :::;0.20 320 ;;::28
W :::;2 :::;2 0.5-3 0-1.0 :::;0.45 315 ;;::13
lOA :::;7 :::;2 0.5-3.5 1.5-3.5 :::;0.65 ;;::12
llA :::;3 :::;2 0-2.5 2.0-5.0 :::;0.40 ;;::9
l2F :::;3 :::;2 3-5 0-1.0 :::;0.25 ;;::25
14 :::;5 :::;2 1.5-4 0-1.0 :::;0.30 ;;::20
l5B :::;3 :::;2 1-3 2.0-4.5 :::;0.55 ;;:: 15
l7Aor17F :::;2 :::;2 0-1.5 0-3.5 :::;0.45 ;;::20
l8C :::;3 :::;2 0-1 2.4-4.9 :::;0.15 ;;:: 14
19A :::;2 :::;2 0.6-3.5 3.0-7.0 :::;0.45 ;;::12 ;;::20
19F :::;3 :::;2 1.4-3.5 3.0-5.5 :::;0.20 ;;::12.5 ;;::20
20 :::;2 :::;2 0.5-2.5 1.5-4.0 :::;0.60 ;;::12
22F :::;2 :::;2 0-2 0-1.0 :::;0.55 ;;:: 15 ;;::25
23F :::;2 :::;2 0-1 3.0-4.5 :::;0.15 ;;::37
33F :::;2.5 :::;2 0-2 0-1.0 :::;0.50
* The different types are indicated using the Danish nomenclature
** Cross linked agarose for chromatography R
*** Cross linked agarose for chromatography Rl

O-Acetyl groups (2.7.1). Comply with the test for O-acetyl Specificity
groups.
For establishing the specificity, no reaction should occur,
Sterility (2.2.11). Comply with the test for sterility. when the antigens are tested against all the antisera specific
Molecular size. Molecular size is determined by gel filtration for the other polysaccharides of the vaccine, including factor
or high performance size-exclusion chromatography (HPSEC) sera for distinguishing types within groups. The
(2.4.16) using cross linkedAgarose for chromatography R or polysaccharides are tested at a concentration of50 Ilg/ml using
chromatography Agarose for chromatograph Rl, either alone a method capable ofdetecting 0.5 Ilg/ml.
or Multiple angle light laser scattering (MALLS) or any other
suitable method. FINAL BULK VACCINE
The final bulle vaccine is obtained by aseptically mixing the
Identification different polysaccharide powders. The uniform mixture is
Confirm the identity of the monovalent bulle polysaccharide aseptically dissolved in a suitable isotonic solution so that
by immunochemical method (2.2.14)(except for polysaccharides one human dose of 0.5 ml contains 25 Ilg of each
7F,14and33F). polysaccharide. An antimicrobial preservative may be added.

2425

www.webofpharma.com
PNEUMOCOCCAL POLYSACCHARIDE VACCINE IP 2010

The solution is sterilized by filtration through a bacteria- (2.2.14), using antisera specific for each polysaccharide
retentive filter. contained in the vaccine, including factor sera for types within
Only a final bulk vaccine that complies with the following groups, and purified polysaccharides of each type as
requirements may be used in the preparation of the final lot. standards.
The vaccine contains not less than 70.0 per cent and not more
Antimicrobial preservative. Where applicable, determine the
than 130.0 per cent ofthe quantity stated on the label for each
amount of antimicrobial preservative by a suitable chemical
polysaccharide. The confidence interval (P = 0.95) ofthe assay
method. The content is not less than 85.0 per cent and not
is not less than 80.0 and not more than 120.0 per cent of the
greater than 115.0 per cent ofthe intended amount.
estimated content.
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk Labelling. The label states (1) the number of Ilg of each
for each sterility medium. polysaccharide per human dose; (2) the total amount of
polysaccharide in the container.
FINAL LOT
The final bulk vaccine is distributed and filled aseptically into
sterile containers (vials or ampoules). Only a final lot that is Poliomyelitis Vaccine (Inactivated)
satisfactory with respect to each of the requirements given Poliomyelitis Vaccine (Inactivated) is a liquid preparation of
below under Identification, Tests and Assay may be released suitable strains of human polioviruses 1, 2 and 3 grown in
for use. Provided that the tests for phenol and for antimicrobial suitable cell cultures and inactivated by a validated method.
preservative have been carried out with satisfactory results
on the final bulle vaccine, they may be omitted on the final lot.
ProductiOl1.
When consistency of production has been established on a
suitable number of consecutive batches, the assay may be General provisions
replaced by a qualitative test that identifies each The production method should consistently yield vaccines
polysaccharide, provided that an assay has been performed of acceptable safety and immunogenicity in man.
on each monovalent bulk polysaccharide used in the
Production of the vaccine is based on a virus seed-lot system.
preparation ofthe final lot.
Cell lines are used according to a cell-bank system. Ifprimary,
secondary or tertiary monkey kidney cells are used, production
Identification
complies with the requirements indicated below.
The assay also serves to identify the vaccine. Unless otherwise justified and authorised, the virus in the
final vaccine shall not have undergone more passages from
Tests the master seed lot than was used to prepare the vaccine
Sterility (2.2.11). Complies with the test for sterility. shown in clinical studies to be satisfactory with respect to
safety and efficacy.
Abnormal toxicity (2.2.1). Complies with the test for abnormal
toxicity with the following modifications. The production method is validated to demonstrate that the
product, if tested, would comply with the test for safety and
Inject 10 human doses each in two guinea pigs weighing
efficacy.
between 250 and 350 g by intraperitoneal route and observe
for 12 days, Substrate for virus propagation
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject The virus is propagated in a human diploid cell line (2.7.2), in
each of the rabbit with 1 ml of a dilution of the vaccine a continuous cell line (2.7.2) or in primary, secondary or tertiary
containing 2.5 Ilgfml of each polysaccharide. monkey kidney cells.
Antimicrobial preservative. Where applicable, determine the Primary, secondmy or tertiary monkey kidney cells. The
amount of antimicrobial preservative by a suitable chemical following special requirements for the substrate for virus
method. The content is not less than 85.0 per cent and not propagation apply to primary, secondary or tertiary monkey
greater than 115.0 per cent ofthe intended amount. kidney cells.
Phenol (2.3.36). Not more than 2.5 gil. Monkeys used in the preparation of kidney cell cultures for
llU(2:4:24). 45to-7.4: - production andcontrolofthe vaccine. The animals-used are
of a species approved by the competent authority, in good
Assay
health and, unless otherwise justified and authorised, have
Determine the content of each polysaccharide by a suitable not been previously employed for experimental purposes.
biochemical, physicochemical or immunochemical method Kidney cells used for vaccine production and control are

2426

www.webofpharma.com
IP 2010 POLIOMYELITIS VACCINE (INACTIVATED)

derived from monitored, closed colonies of monkeys bred in Only a working seed lot that complies with the following
captivity, not from animals caught in the wild; a previously requirements may be used for virus propagation.
approved seed lot prepared using virus passaged in cells from
wild monkeys may, subject to approval by the competent Identification
authority, be used for vaccine production if historical data on
Each working seed lot is identified as human poliovirus 1,2 or
safety justify this.
3 by virus neutralisation in cell culture using specific
Monitored, closed colonies of monkeys. The monkeys are antibodies.
kept in groups in cages. Freedom from extraneous agents is
achieved by the use of animals maintained in closed colonies Virus concentration. The virus concentration of each
that are subject to continuous and systematic veterinary and working seed lot is determined to define the quantity
laboratory monitoring for the presence of infectious agents. of virus to be used for inoculation of production cell
The supplier ofanimals is certified by the competent authority. cultures.
Each monkey is tested serologically at regular intervals during Extraneous agents (2.7.3). The working seed lot complies with
a quarantine period of not less than 6 weeks imposed before the requirements for seed lots for virus vaccines. In addition,
entering the colony and then during its stay in the colony. if primary, secondary or tertiary monkey Iddney cells have
The monkeys used are shown to be tuberculin-negative been used for isolation of the strain, measures are taken to
and free from antibodies to simian virus 40 (SV40) and ensure that the strain is not contaminated with simian
simian immunodeficiency virus. If Macaca spp. monkeys are viruses such as simian immunodeficiency virus, simian virus
used for production, the monkeys are also shown to 40, filoviruses and herpesvirus B (Cercopithecine
be free from antibodies to herpesvirus B (Cercopithecine helpesvirus 1). A working seed lot produced in primary,
helpesvirus 1) infection. Human herpesvirus 1 has been used secondary or tertiary monkey Iddney cells complies with the
as an indicator for freedom from herpesvirus B antibodies on requirements given below under Virus Propagation and
account of the danger of handling herpesvirus B Harvest for single harvests produced in such cells.
(Cercopithecine herpesvirus 1).
PROPAGATION AND HARVEST
Monkeys from which kidneys are to be removed are thoroughly
examined, particularly for evidence of tuberculosis and All processing ofthe cell banle and subsequent cell cultures is
herpesvirus B (Cercopithecine herpesvirus 1) infection. If a done under aseptic conditions in an area where no other cells
monkey shows any pathological lesion relevant to the use of or viruses are being handled. Approved animal serum (but not
its kidneys in the preparation of a seed lot or vaccine, it is not human serum) may be used in the cell culture media. Serum
to be used nor are any of the remaining monkeys of the group and trypsin used in the preparation of cell suspensions and
concerned unless it is evident that their use will not impair the media are shown to be free from extraneous agents. The cell
safety of the product. culture media may contain a pH indicator such as phenol red
and approved antibiotics at the lowest effective concentration.
All the operations described in this section are conducted
Not less than 500 ml ofthe cell cultures employed for vaccine
outside the area where the vaccine is produced.
production is set aside as uninfected cell cultures (control
Monkey cell cultures for vaccine production. Kidneys that cells); where continuous cell lines in a fermenter are used for
show no pathological signs are used for preparing cell cultures. production, 200 x 106 cells are set aside to prepare control
Each group ofcell cultures derived from a single monkey forms cells; where primary, secondary or tertiary monleey Iddney
a separate production cell culture giving rise to a separate cells are used for production, a cell sample equivalent to at
single harvest. least 500 ml of the cell suspension, at the concentration
The primary monkey kidney cell suspension complies with employed for vaccine production, is taken to prepare control
• the test for mycobacteria ; disrupt the cells before carrying cell cultures.
out the test. Only a single harvest that complies with the following
If secondary or tertiary cells are used, it shall be demonstrated requirements may be used in the preparation of the vaccine.
by suitable validation tests that cell cultures beyond the The tests for Identification and Sterility may be carried out
passage levelused for production are free from tumorigenicity. instead on the purified, pooled monovalent harvest. After
demonstration of consistency of production at the stage of
SEED LOT the single harvest, the test for virus concentration may be
carried out instead on the purified, pooled monovalent harvest.
Each ofthe three strains ofpoliovirus used shall be identified
by historical records that include information on the origin of Control cells. The control cells of the production cell culture
the strain and its subsequent manipulation. comply with a test for Identification (if a cell-banle system is

2427

www.webofpharma.com
POLIOMYELITIS VACCINE (INACTIVATED) IP 2010

used for production) and with the requirements for extraneous demonstrated to be at least as susceptible for SV40. Incubate
agents, where primary, secondary or tertiary monkey kidney the cultures at 37° and observe for 14 days. At the end ofthis
cells are used, the tests in cell cultures are carried out as period, make at least one subculture of fluid in the same cell
shown below under Test in Rabbit Kidney Cell Cultures and culture system and observe both primary cultures and
Test in Cercopithecus Kidney Cell Cultures). subcultures for an additional 14 days.
Test in rabbit kidney cell cultures. Test a sample of at least
PURIFICATION AND PURIFIED MONOVALENT
10 ml ofthe pooled supernatant fluid from the control cultures
HARVEST
for the absence of herpesvirus B (Cercopithecine herpesvirus 1)
and other viruses in rabbit kidney cell cultures. The dilution Several single harvest of the same type may be pooled and
of supernatant in the nutrient medium is not greater than 1:4 may be concentrated. The monovalent harvest or pooled
and the area ofthe cell layer is at least 3 cm2 per m1 ofinoculum. monovalent harvest is purified by validated methods. If
Set aside one or more containers of each batch of cells with continuous cell lines are used for production, the purification
the same medium as non-inoculated control cells. Incubate process shall have been shown to reduce consistently the
the cultures at 37° and observe for at least 2 weeks. The test is content of substrate-cell DNA to not more than 500 pg per
not valid if more than 20 per cent of the control cells are single human dose.
discarded for non-specific, accidental reasons. Only a purified monovalent harvest that complies with the
Test in Cercopithecus kidney cell cultures. Test a sample of following requirements may be used for the preparation ofthe
at least 10 ml ofthe pooled supernatant fluid from the control inactivated monovalent harvest.
cultures for the absence of SV40 virus and other extraneous
agents by inoculation onto cell cultures prepared from the Identification
kidneys of cercopithecus monkeys, or other cells shown to be The virus is identified by virus neutralisation in cell cultures
at least as sensitive for SV40, by the method described under using specific antibodies or by determination ofD-antigen.
Test in Rabbit Kidney Cell Cultures. The test is not valid if
more than 20 per cent ofthe control cell cultures are discarded Virus concentration. The virus concentration is determined
for non-specific, accidental reasons. by titration of infectious virus.
Specific activity. The ratio ofthe virus concentration or the D-
Identification antigen content, determined by a suitable immunochemical
method (2.2.14) to the total protein content (specific activity)
The single harvest is identified as containing human poliovirus
ofthe purified monovalent harvest is within the limits approved
1, 2 or 3 by virus neutralisation in cell cultures using specific
for the particular product.
antibodies.
Virus concentration. The virus concentration of each single INACTIVATION AND INACTIVATED MONOVALENT
harvest is determined by titration of infectious virus in cell HARVEST
cultures.
Several purified monovalent harvests of the same type may
Sterility (2.2.11). The single harvest complies with the test for be mixed before inactivation. To avoid failures in inactivation
sterility, carried out using 10 ml for each medium. caused by the presence ofvirus aggregates, fil1Tation is carried
out before and during inactivation; inactivation is started
Mycoplasmas (2.7.4). The single harvest complies with the
within a suitable period, preferably not more than 24 h and in
test for mycoplasmas, carried out using 10 ml.
any case not more than 72 h, ofthe prior filtration. The virus
Test in rabbit kidney cell cultures. Where primary, secondary suspension is inactivated by a validated method that has been
or tertiary monkey kidney cells are used for production, test a shown to inactivate poliovirus without destruction of
sample ofat least 10 ml ofthe single harvest for the absence of immunogenicity; during validation studies, an inactivation
herpesvirus B (Cercopithecine herpesvirus 1) and other viruses curve with at least four points (for example, time 0, 24, 48, and
in rabbit kidney cell cultures as described for the control cells. 96 h) is established showing the decrease in concentration of
Test in Cercopithecus Iddney cell cultures. Where primary, live virus with time. Ifformaldehyde is used for inactivation,
secondary or tertiary monkey kidney cells are used for the presence of an excess of formaldehyde at the end of the
inactivation period is verified.
E~~~~~ti~ll,_~~!a.~a.1!1J>It::()fa.tl~s!}Q
!Jl1of~~~siI!gl~.~~rye~t _. __ _ _-,_._ __.-
..•.....• ..... ..

for the absence of SV40 virus and other extraneous agents. Only an inactivated monovalent harvest that complies with
Neutralise fhe sample· by -il high::iftreaiitiserl.lmagaiiisfthe the following requirements may be used in the preparation of
specific type of poliovirus. Test the sample in primary a trivalent pool of inactivated monovalent harvests or a final
cercopithecus kidney cell cultures or cells that have been bulle vaccine.

2428

www.webofpharma.com
IP 2010 POLIOMYELITIS VACCINE (INACTIVATED)

Test for effective inactivation. After neutralisation of the for inactivation is carried out on that pool rather than on the
formaldehyde with sodium bisulphite (where applicable), verifY final bulk vaccine.
the absence of residual live poliovirus by inoculation on
FINAL LOT
suitable cell cultures of two samples of each inactivated
monovalent harvest, corresponding to at least 1500 human Only a final lot that complies with each of the requirements
doses. Take one sample not later than three-quarters of the given below under Identification, Tests and Assay may be
way through the inactivation period and the other at the end. released for use. Provided that the tests for free formaldehyde
Inoculate the samples in cell cultures such that the dilution of and antimicrobial preservative and the in vivo assay have
vaccine in the nutrient medium is not greater then Y4 and the been performed with satisfactory results on the final bulle
area of the cell layer is at least 3 cm2 per ml of inoculum. Set vaccine, they may be omitted on the final lot. Provided that
aside one or more containers with the same medium as non- the test for bovine serum albumin has been performed with
inoculated control cells. Observe the cell cultures for at least satisfactory results on the trivalent pool of inactivated
3 weeks. Make not fewer than two passages from each monovalent harvests or on the final bulk vaccine, it may be
container, one at the end of the observation period and the omitted on the final lot.
other 1 week before; for the passages, use cell culture
supernatant and inoculate as for the initial sample. Observe Identification
the subcultures for at least 2 weeks. No sign of poliovirus The vaccine is shown to contain human polioviruses 1, 2 and
multiplication is present in the cell cultures. At the end of the 3 by a suitable immunochemical method such as the
observation period, test the susceptibility of the cell culture detennination of D-antigen by enzyme-linked immunosorbent
used by inoculation oflive poliovirus ofthe same type as that assay (ELISA).
present in the inactivated monovalent harvest.
Tests
Sterility (2.2.11). The inactivated monovalent harvest complies
with the test for sterility, carried out using 10 ml for each Free formaldehyde (2.3.20). Maximum 0.2 gil.
medium. Antimicrobial preservative. Where applicable, determine the
D-antigen content. The content of D-antigen determined by a amount of antimicrobial preservative by a suitable chemical
suitable immunochemical method (2.2.14) is within the limits method. The content is not less than 85.0 per cent and not
approved for the particular preparation. greater than 115.0 per cent of the intended amount.
Protein content (2.3.49). Not more than 10 Ilg of protein
FINAL BULK VACCINE
nitrogen per human dose.
The final bulle vaccine is prepared directly from the inactivated Bovine serum albumin. Not more than 50 ng per single human
monovalent harvests ofhuman polioviruses 1,2 and 3 or from dose, determined by a suitable immunochemical method
a trivalent pool of inactivated monovalent harvests. If a (2.2.14).
trivalent pool of inactivated monovalent harvests is used, a
test for effective inactivation is carried out on this pool instead Sterility (2.2.11). Complies with the test for sterility.
of on the final bulk vaccine. A stabiliser and an antimicrobial Bacterial endotoxins (2.2.3). Not more than 5 ill per human
preservative may be added. dose.
Only a final bulk vaccine that complies with the following Assay
requirements may be used in the preparation of the final lot.
D-antigen content. As a measure ofconsistency ofproduction,
Antimicrobial preservative. Where applicable, determine the determine the D-antigen content for human polioviruses 1, 2
amount of antimicrobial preservative. by a suitable chemical and 3 by a suitable immunochemical method (2.2.14) using an
method. The content is not less than 85.0 per cent and not appropriate reference preparation calibrated in D-antigen units.
greater than 115.0 per cent of the intended amount. For each type, the content, expressed with reference to the
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle amount of D-antigen stated on the label, is within the limits
for each sterility medium. approved for the particular product.

Inactivation. Before addition ofany antimicrobial preservative, In vivo test. The capacity ofthe vaccine toinduce the formation
a sample ofat least 1500 ml or, for a purified and concentrated of neutralizing antibodies is detennined in-vivo by one ofthe
vaccine, the equivalent of 1500 doses is tested for residual following methods:
live poliovirus in cell cultures, as described for the inactivated Test in chicks or guinea-pigs. Prepare a suitable series of at
monovalent harvest. Ifthe final bulk vaccine is prepared from least three dilutions ofthe vaccine under examination using a
a trivalent pool of inactivated monovalent harvests, the test suitable bufferedsaline solution. Inject 0.5 ml ofthe dilutions

2429

www.webofpharma.com
POLIOMYELITIS VACCINE (INACTIVATED) IP 2010

intramuscularly into groups of ten 3-week-old chickens or geometric mean titres ofthe series on or more tests is used as
groups of ten guinea-pigs, each weighing between 250 and the cut-off value. For each of the 3 poliovirus types, the
350 g, using a separate group for each dilution of vaccine. potency of the vaccine is not significantly less than that of
Bleed the animals on the fifth or sixth day after the injection the reference preparation. The test is not valid unless (1) for
and separate the sera. Examine the sera for the presence of both the test and reference vaccines the ED so lies between the
neutralising antibody, at a dilution of 1 in 4, to each of the smallest and the largest doses given to the animals; (2) the
human polioviruses 1,2 and 3. Mix 100 CCID so ofvirus with statistical analysis shows no significant deviation from
the dilution ofserum and incubate at 37° for 4 h 30 min to 6 h. linearity or parallelism; (3) the fiducial limits ofthe estimated
Keep at 5 ± 3° for 12 to 18 h. Inoculate the mixtures into cell relative potency fall between 25.0 per cent and 400.0 per cent
cultures for the detection of unneutralised virus and read the of the estimated potency.
results up to 7 days after inoculation. For each group ofanimals,
Labelling. The label states (1) the types of poliovirus
note the number of sera which have neutralising antibody
contained in the vaccine; (2) the nominal amount of virus of
and calculate the dilution of the vaccine giving an antibody
each type (1, 2 and 3), expressed in units of D-antigen per
response in 50.0 per cent ofthe animals. Carry out in parallel a
single human dose; (3) the cell substrate used to prepare the
control test using a suitable reference preparation.
vaccine.
The vaccine complies with the test if a dilution of 1 in 100 or
more produces an antibody response for each of the three
types of virus in 50.0 per cent of the animals.
Poliomyelitis Vaccine, Live (Oral)
Test in rats. A suitable in vivo assay method consists of
intramuscular injection into the hind limb(s) ofnot fewer than Oral PoliomyelitisyacciIle.is a preparation ()fappr()v(;:dstraill.s
3 dilutions of the vaccine under examination and a reference of live attenuated poliovirus type 1, 2 or 3 grown in in vitro
cultures of approved cells, containing anyone type or any
vaccine, using for each dilution a group of 10 specific
combination ofthe three types of Sabin strains, prepared in a
pathogen-free rats of the suitable strain. Use of 4 dilutions is
fonn suitable for oral administration. The vaccine is a clear
often necessary to obtain valid results for all 3 serotypes. The
liquid that may be coloured owing to the presence of a pH
number of animals per group must be sufficient to obtain
indicator.
results that meet the validity criteria; groups of 10 rats are
usually sufficient although valid results may be obtained with Production
fewer animals per group. The weight ofindividual animal must
not vary by more than 10.0 per cent from the group mean. An
.General provisions
inoculum of 0.5 ml per rat is used. The dose range is chosen The vaccine strains and the production method should
such that a dose response to all 3 poliovirus types is obtained. consistently yield vaccines that are both immunogenic and
Bleed the animals after 20 to 22 days. Neutralising titres against safe inman.
all 3 polivirus types are measured separately using 100 CCID50
The production ofvaccine is based on a virus seed-lot system.
of the Sabin strains as challenge viruses. Vero or Hep2 as
Cell lines are used according to a cell-bank system. Ifprimary
indicator cells, and neutralization conditions of 3 h at 35° to
37° followed by 18 h at 2° to 8°. Results are read monl<ey kidney cells are used, production complies with the
microscopically following fixation and staining after 7 days of requirements indicated below. Unless otherwise justified and
incubation at 35°. For a valid antibody assay, the titre of each authorised, the virus in the final vaccine shall not have
challenge virus must be shown to be within the range of 10 to undergone more than two passages from the master seed lot.
1000 CCID so and the neutralizing antibody titre of a control Substrate for virus propagation
serum must be within 2 twofold dilutions of the geometric
The virus is propagated in human diploid cells (2.7.2) or in
mean titre of the serum. The potency is calculated by
continuous cell lines (2.7.2) or in primary monkey kidney cells
comparison of the preparation of responders for the vaccine
(including serially passaged cells from primary monkey kidney
under examination and the reference vaccine by the probit
cells). Continuous cell lines are approved by the competent
method or, after validation, using a parallel-line model. For the
authority.
probit method it is necessary to establish a cut-offneutralising
antibody titre for each poliovirus type to define a responder. Primary monkey kidney cells. The following special
J:)ue JQ!Ilt~IaJ?Ql"CltQryYCll"iClti~n,.jJi~IlQtl?QssilJl(;:JQ_~flll~ requirements for the substrate for virus propagation apply
cut-offvalues that could be applied by all laboratories. Rather, toprlma,y··monkeY7craiiey··cells~
the cut-off values are detennined for each laboratory based Monkeys usedfor preparation ofIddney cell cultures andfor
on a minimum series on tests with the reference vaccine. The testing ofvirus. If the vaccine is prepared in monkey lodney
mid-point on a log 2 scale of the minimum and maximum cell cultures, animals of a species approved by the competent

2430

www.webofpharma.com
IP 2010 POLIOMYELITIS VACCINE, LNE (ORAL)

authority, in good health, and not previously employed for propagated in series. Vims for the preparation of vaccine is
experimental purposes shall be used. grown by aseptic methods in such cultures. Ifanimal semm is
used in the propagation ofthe cells, the maintenance medium
The monkeys shall be kept in well-constructed and adequately
ventilated animal rooms in cages spaced as far apart as after vims inoculation shall contain no added serum.
possible. Adequate precautions shall be taken to prevent Each group of cell cultures derived from a single monkey or
cross-infection between cages. Not more than two monkeys from fetuses from no more than ten near-term monkeys is
shall be housed per cage and cage-mates shall not be prepared and tested as an individual group.
interchanged. The monkeys shall be kept in the country of
manufacture of the vaccine in quarantine groups for a period SEED LOT
of not less than 6 weeks before use. A quarantine group is a
colony of selected, healthy monkeys kept in one room, with The strains ofpoliovirus used shall be identified by historical
separate feeding and cleaning facilities, and having no contact records that include information on the origin and subsequent
with other monkeys during the quarantine period. If at any manipulation of the strains.
time dming the quarantine period the overall death rate of a Working seed lots are prepared by a single passage from a
shipment consisting of one or more groups reaches 5 per cent master seed lot and at an approved passage level from the
(excluding deaths from accidents or where the cause was original Sabin vims. Vims seed lots are prepared in large
specifically determined not to be an infectious disease), quantities and stored at a temperature below -60°.
monkeys from that entire shipment shall continue in quarantine
from that time for a minimum of 6 weeks. The groups shall be Only a virus seed lot that complies with the following
kept continuously in isolation, as in quarantine, even after requirements may be used for vims propagation.
completion of the quarantine period, until the monkeys are
used. After the last monkey of a group has been taken, the Identification
room that housed the group shall be thoroughly cleaned and
Each working seed lot is identified as poliovirus of the given
decontaminated before being used for a fresh group. Ifkidneys
type, using specific antibodies.
from near-term monkeys are used, the mother is quarantined
for the term ofpregnancy. Virus concentration. Determined by the method described
below, the virus concentration is the basis for the quantity of
Monkeys from which kidneys are to be removed shall be
virus used in the nemovirulence test.
anaesthetised and thoroughly examined, particularly for
evidence of tuberculosis and cercopithecid herpesvirus 1 (B Extraneous agents (2.7.3). Ifthe working seed lot is produced
vims) infection. in human diploid cells (2.7.2) or in continuous cell lines (2.7.2)
it complies with the requirements for seed lots for virus
Ifa monkey shows any pathological lesion relevant to the use
vaccines. If the working seed lot is produced in primary
of its kidneys in the preparation of a seed lot or vaccine, it
monkey cells, it complies with the requirements given below
shall not be used, nor shall any of the remaining monkeys of
under Propagation and Harvest and Monovalent Pooled
the quarantine group concerned be used unless it is evident
Harvest and with the tests in adult mice, suclding mice and
that their use will not impair the safety ofthe product.
guinea-pigs given under Tests for extraneous agents in viral
All the operations described in this section shall be conducted vaccines for human use.
outside the areas where the vaccine is produced.
Working seed lot shall be free from detectable DNA sequences
The monkeys used shall be shown to be free from antibodies from simian virus 40 (SV40).
to simian virus 40 (SV40) and simian immunodeficiency virus.
Neurovirulence (2.7.6). Each master and working seed lot
If Macaca spp. are used for production, the monkeys shall
complies with the test for neurovimlence of poliomyelitis
also be shown to be free from antibodies to cercopithecid
vaccine (oral) in monkeys. Furthermore, the seed lot shall cease
herpesvims 1 (B virus). Human herpesvirus has been used as
to be used in vaccine production ifthe frequency offailme of
an indicator for freedom from B virus antibodies on account
the monovalent pooled harvests produced from it is greater
ofthe danger ofhandling cercopithecid herpesvirus 1 (B vims).
than predicted statistically. This statistical prediction is
Monkey ladney cell culturesfor vaccine production. Kidneys calculated after each test on the basis of all the monovalent
that show no pathological signs are used for preparing cell pooled harvests tested; it is equal to the probability of false
cultures. If the monkeys are from a colony maintained for rejection on the occasion of a first test (i.e. 1 per cent), the
vaccine production, serially passaged monkey kidney cell probability offalse rejection on retest being negligible. Ifthe
cultmes from primary monkey kidney cells may be used for test is carried out only by the manufacturer, the test slides are
vims propagation, otherwise the monkey kidney cells are not provided to the control authority for assessment.

2431

www.webofpharma.com
POLIOMYELITIS VACCINE, LIVE (ORAL) IP 2010

Genetic markers. Each working seed lot is tested for its fetuses from not more than ten near-term monkeys is divided
replicating properties at temperatures ranging from 36° to 40° into two equal portions. One portion of the pooled fluid is
as described under Monovalent Pooled Harvest. tested in monkey Iddney cell cultures prepared from the same
species, but not the same animal, as that used for vaccine
PROPAGATION AND HARVEST production. The other portion of the pooled fluid is, where
All processing ofthe cell-banks and subsequent cell-cultures necessary, tested in monkey Iddney cell cultures from another
is done under aseptic conditions in an area where no other species so that tests on the pooled fluids are done in cell
cells are handled. Approved animal (but not human) serum cultures from at least one species known to be sensitive to
may be used in the media, but the final medium for maintaining SV40. The pooled fluid is inoculated into bottles ofthese cell
cell growth during virus multiplication does not contain animal cultures in such a way that the dilution of the pooled fluid in
serum. Serum and trypsin used in the preparation of cell the nutrient medium does not exceed 1 in 4. The area ofthe cell
suspensions and media are shown to be free from live sheet is at least 3 cm2 per ml ofpooled fluid. At least one bottle
extraneous agents. The cell-culture medium may contain a pH ofeach kind of cell culture remains uninoculated to serve as a
indicator such as phenol red and approved antibiotics at the control. If the monkey species used for vaccine production is
lowest effective concentration. It is preferable to have a known to be sensitive to SV40, a test in a second species is
substrate free from antibiotics during production. Not less not required. Animal serum may be used in the propagation of
than 5 per cent and not more than 1000 ml of the cell cultures the cells, provided that it does not contain SV40 antibody, but
employed for vaccine production are set aside as uninfected the maintenance medium after inoculation of test material
cell cultures (control cells); special requirements, given below, contains no added serum except as described below.
apply to control cells when the vaccine is produced in primary The cultures are incubated at a temperature of 35° to 37° and
monkey cells The vilUs suspension is harvested not later than are observed for a total period of at least 4 weeks. During this
4 days after vilUs inoculation. After inoculation of the observation period and after not less than 2 weeks incubation,
production cell culture with the vilUs working seed lot, at least one subculture of fluid is made from each of these
inoculated cells are maintained at a fixed temperature, shown cultures in the same cell culture system. The subcultures are
to be suitable, within the range 33° to 35°; the temperature is also observed for at least 2 weeks.
maintained constant to ±0.5°; control cell cultures are Serum may be added to the original culture at the time of
maintained at 33° to 35° for the relevant incubation periods. subculturing, provided that the serum does not contain SV40
Only a single virus harvest that complies with the following antibody.
requirements may be used in the preparation ofthe monovalent
Fluorescent-antibody techniques may be useful for detecting
pooled harvest.
SV40 virus and other viruses in the cells.
Virus concentration. The vilUs concentration of vilUs
A further sample of at least 10 ml ofthe pooled fluid is tested
harvests is determined as prescribed under Assay to monitor
for cercopithecid herpesviIus I (B virus) and other viruses in
consistency ofproduction and to determine the dilution to be
rabbit lddney cell cultures. Serum used in the nutrient medium
used for the final bulk vaccine.
of these cultures shall have been shown to be free from
Extraneous agents (2.7.3 ). Complies with tests for extraneous inhibitors ofB virus. Human herpesvirus has been used as an
agents. indicator for freedom from B virus inhibitors on account ofthe
Control cells. The control cells ofthe production cell culture danger ofhandling cercopithecid herpesvirus I (B virus). The
from which the vilUs harvest is derived comply with a test for sample is inoculated into bottles ofthese cell cultures in such
identity and with the requirements for extraneous agents or, a way that the dilution of the pooled fluid in the nutrient
where primmy monkey cells are used, as shown below. medium does not exceed 1 in 4. The area ofthe cell sheet is at
Primary monkey kidney cells. The following special least 3 cm2 per ml ofpooled fluid. At least one bottle ofthe cell
requirements apply to virus propagation and harvest in cultures remains uninoculated to serve as a control.
primmy monkey Iddney cells. The cultures are incubated at a temperature of 35° to 37°
Cell cultures. On the day of inoculation with virus seed, each and observed for at least 2 weeks.
cell culture is examined for degeneration caused by an infective A further sample of I0 rnl ofthe pooled fluid removed from the
agent. If, in this examination, evidence is found ofthe presence cell cultures on the day of inoculation with the seed lot virus
il1 .a cell c;ulture oLal1y eXtran~()lls agent, the entire group of i~te~e4J()E tl1.e prese llce ()f e)(tr8:Ileoll~ ageIlts]:>y inoculation
cultures concerned shall be rejected. into human cell cultures sensitive to measles virus.
On the day of inoculation with the vilUs worldng seed lot, a The tests are not valid if more than 20 per cent ofthe culture
sample of at least 30 ml ofthe pooled fluid removed from the vessels have been discarded for non-specific accidental
cell cultures of the kidneys of each single monkey or from reasons by the end of the respective test periods.

2432

www.webofpharma.com
IP 2010 POLIOMYELITIS VACCINE, LIVE (ORAL)

If, in these tests, evidence is found of the presence of an If the presence of Cercopithecid herpesvirus J (B virus) is
extraneous agent, the single harvest from the whole group of demonstrated, the production cell cultures shall not be used
cell cultures concerned is rejected. and the measures concerning vaccine production described
If the presence of Cercopithecid helpesvirus J (B virus) is above must be undertaken.
demonstrated, the manufacture of oral poliomyelitis vaccine The fluids collected fi'om the control cell cultures at the time
shall be discontinued and the competent authority shall be ofvims harvest and at the end of the observation period may
informed. Manufacturing shall not be resumed until a thorough be pooled before testing for extraneous agents. A sample of
investigation has been completed and precautions have been 2 per cent of the pooled fluid is tested in each of the cell
taken against any reappearance of the infection, and then culture systems specified.
only with the approval of the competent authority.
Single harvests
Ifthese tests are not done immediately, the samples ofpooled
cell-culture fluid shall be kept at a temperature of -60 0 or below, Tests jor neutralised single harvests in monkey kidney cell
with the exception ofthe sample for the test for B virus, which cultures. A sample of at least 10 ml of each single harvest is
may be held at 4 0 , provided that the test is done not more than neutralised by a type-specific poliomyelitis antiserum prepared
7 days after it has been taken. in animals other than monkeys. In preparing antisera for this
purpose, the immunising antigens used shall be prepared in
Control cell cultures. On the day of inoculation with the virus
non-simian cells.
working seed lot 25 per cent (but not more than 2500 ml) ofthe
cell suspension obtained from the kidneys of each single Half of the neutralised suspension (corresponding to at least
monkey or from not more than ten near-term monkeys is taken 5 ml ofsingle harvest) is tested in monkey kidney cell cultures
to prepare uninoculated control cell cultures. These control prepared from the same species, but not the same animal, as
cell cultures are incubated in the same conditions as the that used for vaccine production. The other half of the
inoculated cultures for at least 2 weeks and are examined during neutralised suspension is tested, if necessary, in monkey
this period for evidence of cytopathic changes. The tests are kidney cell cultures from another species so that the tests on
not valid if more than 20 per cent of the control cell cultures the neutralised suspension are done in cell cultures from at
have been discarded for non-specific, accidental reasons. At least one species known to be sensitive to SV40.
the end ofthe observation period, the control cell cultures are The neutralised suspensions are inoculated into bottles of
examined for degeneration caused by an infectious agent. If these cell cultures in such a way that the dilution of the
this examination or any of the tests required in this section suspension in the nutrient medium does not exceed 1 in 4. The
shows evidence of the presence in a control culture of any area of the cell sheet is at least 3 cm2 per ml of neutralised
extraneous agent, the poliovirus grown in the corresponding suspension. At least one bottle of each type of cell culture
inoculated cultures from the same group shall be rejected. remains uninoculated to serve as a control and is maintained
Tests for haemadsorbing viruses. At the time of harvest or by nutrient medium containing the same concentration ofthe
within 4 days of inoculation of the production cultures with specific antiserum used for neutralisation.
the virus working seed lot, a sample of4 per cent ofthe control Animal serum may be used in the propagation of the cells,
cell cultures is taken and tested for haemadsorbing viruses. provided that it does not contain SV40 antibody, but the
At the end of the observation period, the remaining control maintenance medium, after the inoculation ofthe test material,
cell cultures are similarly tested. The tests are made as described contains no added serum other than the poliovirus neutralising
in (2.7.3), Tests for extraneous agents in viral vaccines for antiserum, except as described below.
human use. The cultures are incubated at a temperature of35° to 37 0 and
Tests for other extraneous agents. At the time of harvest, or observed for a total period of at least 4 weeks. During this
within 7 days of the day of inoculation of the production observation period and after not less than 2 weeks incubation,
cultures with the working seed lot, a sample ofat least 20 m1 of at least one subculture of fluid is made from each of these
the pooled fluid fi:om each group of control cultures is taken cultures in the same cell-culture system. The subcultures are
and tested in two kinds of monkey kidney cell culture, as also observed for at least 2 weeks.
described above. Serum may be added to the original cultures at the time of
At the end of the observation period for the original control subculturing, provided that the serum does not contain SV40
antibody.
cell cultures, similar samples ofthe pooled fluid are taken and
the tests referred to in this section in the two kinds ofmonkey Additional tests are made for extraneous agents on a further
kidney cell culture and in the rabbit cell cultures are repeated, sample of the neutralised single harvests by inoculation of
as described above under Cell cultures. lO ml into human cell cultures sensitive to measles virus.

2433

www.webofpharma.com
POLIOMYELITIS VACCINE, LIVE (ORAL) IP 2010

Fluorescent-antibody techniques may be useful for detecting Neurovirulence (2.7.6). Each monovalent pooled harvest
SV40 virus and other viruses in the cells. complies with the test for neurovirulence of poliomyelitis
The tests are not valid if more than 20 per cent of the culture vaccine (oral). Ifthe test is carried out only by the manufacturer,
vessels have been discarded for non-specific accidental the test slides are provided to the competent authority for
reasons by the end of the respective test periods. assessment. The TgPVR21 transgenic mouse model provides
a suitable alternative to the monkey neurovirulence test for
If any cytopathic chauges occur in any of the cultures, the neurovirulence testing of types 1, 2 or 3 vaccines once a
causes of these change are investigated. If the cytopathic laboratory qualifies as being competent to perform the test
changes are shown to be due to unneutralised poliovirus, the and the experience gained is to the satisfaction of the
test is repeated. Ifthere is evidence ofthe presence ofSV40 or competent authority. The test is carried out using a standard
other extraneous agents attributable to the single harvest, operating procedure approved by the competent authority. A
that single harvest is rejected. suitable procedure (Neurovintlence test oftype 1,2 or 3 live
poliomyelitis vaccines (oral) in transgenic mice susceptible
MONOVALENT POOLED HARVEST to poliovirus) is available from WHO, Quality and Safety of
Monovalent pooled harvests are prepared by pooling a number Biologicals, Geneva.
of satisfactory single harvests of the same virus type. Primary monkey kidney cells. The following special
Monovalent pooled harvests from continuous cell lines may requirements apply to monovalent pooled harvests derived
be purified. Each monovalent pooled harvest is filtered through from primmy monkey kidney cells.
a bacteria-retentive filter. Retroviruses. The monovalent pooled harvest is examined
Only a monovalent pooled harvest that complies with the using a reverse transcriptase assay. No indication of the
following requirements may be used in the preparation ofthe presence of retrovirus is found.
final bulk vaccine. Test on rabbits. A sample ofthe monovalent pooled harvest is
tested for cercopithecid herpesvirus 1 (B virus) and other
Identification viruses by injection ofnot less than 100 ml into not fewer than
Each monovalent pooled harvest is identified as poliovirus of 10 healthy rabbits each weighing between 1.5 and 2.5 kg. Each
the given type, using specific antiserum. rabbit receives not less than 10 ml and not more than 20 ml, of
which 1 ml is given intradermally at multiple sites, and the
Virus concentration remainder subcutaneously. The rabbits are observed for at
least 3 weeks for death or signs of illness.
The virus concentration is determined by the method
All rabbits that die after the first 24 h of the test and those
described below and serves as the basis for calculating the
showing signs of illness are examined by autopsy, and the
dilutions for preparation of the final bulle, for the quantity of
brain and organs removed for detailed examination to establish
virus used in the neurovirulence test and to establish and
the cause of death.
monitor production consistency.
The test is not valid ifmore than 20.0 per cent ofthe inoculated
Genetic markers. A ratio of the replication capacities of the rabbits show signs of intercurrent infection during the
virus in the monovalent pooled harvest is obtained over a observation period. The monovalent pooled harvest passes
temperature range between 36° and 40° in comparison with the test if none of the rabbits shows evidence of infection
the seed lot or a reference preparation for the marker tests and with B virus or with other extraneous agents or lesions of any
with appropriate rct/40- and rct/40+ strains of poliovirus of kind attributable to the bulk suspension.
the same type. The incubation temperatures used in this test
If the presence of B virus is demonstrated, the measures
are controlled to within ±0.1 0. The monovalent pooled harvest
concerning vaccine production described above under Cell
passes the test if, for both the virus in the harvest and the
cultures are taken.
appropriate reference material, the titre determined at 36° is at
least 5.0 log greater than that detennined at 40°. If growth at Test on guinea pigs. Administer to not less than five guinea-
40° is so low that a valid comparison cannot be established, a pigs, each weighing between 350 and 450 g, 0.1 ml of the
temperature in the region of39.0° to 39.5° is used, at which monovalent pooled harvest by intracerebral injection and
temperature the reduction in titre of the reference material 0.5 ml by intraperitoneal injection. Measure the rectal
must hI:: jnlh.t:: nlligl:: 3,tLJo5,Q IQgQLi~ Yl:ih!Sl.flt 36°~~11J: temperature of each animal on each working day for 6 weeks.
acceptable mininmm reduction is determined for each virus Afi1ie~end ofihe observatIon perrodcarry~outaufopsyoii
strain at a given temperature. If the titres obtained for one or each animal.
more of the reference viruses are not concordant with the In addition, administer to not fewer than five guinea-pigs
expected values, the test must be repeated. 0.5 ml by intraperitoneal injection and observe as described

2434

www.webofpharma.com
IP 2010 RABIES ANTISERUM

above for 2 to 3 weeks. At the end of the observation period, more than one poliovirus type, titrate each type separately,
carry out a passage from these animals to not fewer than five using appropriate type-specific antiserum (or preferably a
guinea pigs using blood and a suspension of liver or spleen monoclonal antibody) to neutralise each of the other types
tissue. Measure the rectal temperature of the latter guinea present.
pigs for 2 to 3 weeks. Examine by autopsy all animals that,
For a trivalent vaccine, the estimated mean virus titres must
after the first day of the test, die or are lolled because they
be: not less than 1 x 106.0 infectious virus units (CCID so ) per
show disease or show for three consecutive days a body
single human dose for type I; not less than 1 x IO s.oinfectious
temperature higher than 39°; carry out histological examination
virus units (CCID so) for type 2; and not less than I x 10s.8
to detect infection with Marburg virus; in addition, inject a
infectious virus 'units (CCID so ) for type 3.
suspension of liver or spleen tissue or of blood
intraperitoneally into not fewer than three guinea pigs. If any For monovalent or divalent vaccine, the minimum virus titres
signs ofinfection with Marburg virus are noted, confilwatory are decided by the competent authority.
serological tests are carried out on the blood of the affected Method. Groups of eight to twelve flat-bottomed wells in a
animals. The monovalent pooled harvest complies with the microtitre plate are inoculated with 0.05 ml of each of the
test if not fewer than 80.0 per cent ofthe guinea pigs survive selected dilutions of virus followed by a suitable cell
to the end ofthe observation period and remain in good health suspension of the Hep-2 (Cincinnati) line. The plates are
and no animal shows signs of infection with filoviruses virus. incubated at a suitable temperature. Examine the cultures on
FINAL BULK VACCINE days 7 to 9.
The assay is not valid if(a) the confidence interval (P = 0.95)
The final bulk vaccine is prepared from one or more
of the logarithm of the virus concentration is greater than
satisfactory monovalent pooled harvests and may contain
±0.3; (b) the virus concentration of the reference preparation
more than one virus type. Suitable flavouring substances and
differs by more than 0.5 log CCIDso from the assigned value.
stabilisers may be added.
Only a final bulk vaccine that complies with the following Labelling. The- label states (l) the types of poliovirus
requirement may be used in the preparation of the final lot. contained in the vaccine; (2) the minimum amount ofvirus of
each type contained in one single human dose; (3) the cell
Sterility (2.2.11). Complies with the test for sterility.
substrate used for the preparation of the vaccine; (4) that the
vaccine is not to be injected.
FINAL LOT

Only a final lot that complies with the following requirement


for thennal stability and is satisfactory with respect to each of
the requirements given below under Identification, Tests and Rabies Antiserum
Assay may be released for use.
Antirabies Serum; Antirabic Serum
Thermal stability. Expose samples of the final lot at 37° for
Rabies Antiserum is a preparation containing the specific
48 hours. Determine the total virus concentration as described
globulin or its derivatives obtained by purification of
under Assay in parallel for the heated vaccine and for unheated
hyperimmune serum or plasma of healthy horses or other
vaccine. The estimated difference between the total virus
suitable animals having the specific activity of neutralising
concentration of the unheated and heated vaccines is not
the rabies virus. It may contain a suitable preservative.
greater than 0.5 loglo infectious virus units (CCIDso) per single
human dose. Rabies Antiserum contains not less than 300 Units per m!.
Description. A clear, colourless or pale yellow liquid free from
Identification suspended particles or cream-coloured powder or pellet to be
The vaccine is shown to contain poliovirus ofeach type stated reconstituted with diluent supplied by the manufacturer.
on the label, using specific antibodies.
Identification
Tests Specifically neutralizes the standard strain of rabies virus
Sterility (2.2.11). Complies with the test for sterility. rendering it harmless to susceptible animals or by any other
suitable in vitro method.
Assay
Titrate for infectious virus at least in triplicate using the method Tests
described below. Use an appropriate virus reference Potency. Carry out the biological assay of rabies antiserum
preparation to validate each assay. If the vaccine contains described below.

2435

www.webofpharma.com
RABIES ANTISERUM IP 2010

Biological Assay of Rabies Antiserum Storage. As stated under Antisera.


Labelling. The label states (1) the number of Units per ml in
The potency of rabies antiserum is determined by comparing
case ofliquid preparation; (2) the total volume in the container;
the dose necessary to protect mice against a lethal intracerebral
(3) the nameand volume of the diluent to be used for
dose ofrabies virus with the dose ofthe Standard Preparation
reconstituting the freeze-dried preparation; (4) the
of rabies antiserum necessary to give the same protection.
recommended dose; (5) the name and concentration of any
Standard Preparation added preservative; (6) the animal species in which the
antiserum has been raised; (7) that the liquid preparation
The standard preparation is a dried serum or other suitable should not be allowed to freeze.
preparation the potency of which has been detennined in
relation to the International Standard.
Rabies Vaccine, Human
Suggested Method
Rabies Vaccine for Human use is a freeze-dried or liquid
Test animals. Use healthy mice of either sex from the same (adsorbed) preparation ofa suitable approved, strain of fixed
stock weighing between 10 and 14 g, but animals ofthe same rabies virus grown in an approved cell culture/ embryos of
sex should be used in any given test. duck/chicken and inactivated by a validated method.
Test virus. Any suitable strain ofrabies virus ofknown potency The freeze-dried vaccine is reconstituted immediately before
such as the CVS strain may be used. use as stated on the label to give a clear or slightly opalescent
solution/suspension. It may be coloured owing tothe presence
Test dose of virus. The test dose of virus is such that each of a pH indicator.
mouse receives by intracerebralinjection between 20 and 100()
LD so preferably about 100 LD so ' To determine the number of The vaccine complies with the General Requirements of
LD so ofvirus used, mix the virus dilution used in the test with Vaccines for Human Use.
an equal quantity of a 2 per cent v/v solution of heat- Production
inactivated normal horse serum in water and maintain the
mixture at 37° for 1 hour. Prepare 10-fold dilutions in a 2 per General provisions
cent v/v solution of heat-inactivated normal horse serum The vaccine is produced on the basis of virus seed lot system
and inject them into mice. Carry out this test at the same time and if a cell line is used for virus propagation, a cell-bank
as the test for determining the potency ofthe rabies antiserum. system shall be followed. The production method shall have
The test is not valid unless the quantity of virus used lies been shown to yield consistently vaccines that comply with
between 20 and 1000 LDso' the requirements for immunogenicity, safety and stability.
Determination ofpotency ofthe rabies antiserum. Prepare a Unless otherwise justified and authorized, the virus in the
series of six 2-fold dilutions of the Standard Preparation and final vaccine shall not have undergone more passages from
ofthe preparation under examination with water containing 2 the master seed lot than was used to prepare the vaccine
per cent v/v of heat inactivated normal horse serum. To each shown in clinical studies to be satisfactory with respect to
dilution add a quantity of a suspension of the test virus safety and efficacy.
containing the test dose and keep the mixtures at 37° for 1 The production method is validated to demonstrate that the
hour. Inject intracerebrally 0.03 ml ofeach mixture into 10 mice. product, if tested, would comply with the test for safety and
Observe the mice for 14 days after the injection. Mice dying efficacy.
before the fifth day after inoculation with the virus are
Substrate for virus propagation
eliminated from the test; all the mice dying between the fifth
and fourteenth days after showing signs of rabies are The virus is propagated in any suitable approved cell substrate
considered to have died of rabies; mice living upto the like a human diploid cell line (2.7.2), a continuous cell line, or
fourteenth day but showing signs of rabies are also counted in duck embryos or in cultures of chicken embryos derived
as having died from rabies. Calculate the result ofthe test by from a flock certified as free from specified pathogens(2.7.7).
standard statistical methods. The preparation under
SEED LOT
examination passes the test if, in a single comparative assay,
His f()lllld t() hav<l~O or lllore lJnits p<lrllll~ Ifitis fOlilld tohaye '[he Straill of r~~iesvirtl~ll~eds~all bei<ientifie4 ~y ~is~ori~~
less than 80 Units per ml, two more similar assays may be records that include information on the origin of the strain
carried out; the preparation passes the test if, in hoth these and its subsequent manipulation.
additional assays, it is found to have 80 or more Units per ml. Working seed lots are prepared by not more than five passages
Other tests. Complies with the tests stated under Antisera. from the master seed lot.

2436

www.webofpharma.com
IP 2010 RABIES VACCINE, HUMAN

Only a working seed lot that complies with the following test for identification and with the requirements for extraneous
requirements may be used for virus propagation. agents (2.7.3).

Identification Control eggs. Control eggs shall be tested for freedom from
haemagglutinating agents, and other extraneous agents.
Each working seed lot is identified as rabies virus using specific
antibodies by an approved method. PURIFICATION AND INACTIVATION
Virus concentration. The virus concentration ofeach working The virus harvests may be concentrated and/or purified by
seed lot is determined by a cell culture meth9d using suitable methods; the virus harvest is inactivated by a validated
immunofluoresence or any other approved method. method at a fixed, well defined stage ofthe process which may
Extraneous agents (2.7.3). The working seed lot complies with be before, during or after any concentration or purification.
the requirements for the virus seed lots. The method shall have been shown to be capable of
inactivating rabies virus without destruction of the
PROPAGATION AND HARVEST immunogenic activity. If betapropiolactone is used, the
concentration shall at no time exceed 1:3500.
All processing of the cell banle and subsequent cell cultures
are done under aseptic conditions in an area where no other Cell culture vaccines
cells are handled. Approved animal (but not human) serum
may be used in the media, but the final medium for maintaining Only an inactivated viral suspension that complies with the
cell growth during virus multiplication does not contain animal following requirements may be used in the preparation of the
serum; the media may contain human albumin. Serum proteins, final bulle vaccine.
if present are reduced to an acceptable level by a suitable Inactivation. Inactivation is confirmed by carrying out an
method of purification. Serum and trypsin used in the amplification test for residual infectious rabies virus, not more
preparation ofcell suspension and media are shown to be free than 4 days after inactivation or on the sample frozen after
from infectious extraneous agents. The cell culture media may inactivation and stored at -70°. Inoculate a quantity of
contain a pH indicator such as phenol red and approved inactivated viral suspension equivalent to. not less than 25
antibiotics at the lowest effective concentration. Not less than vaccine doses into cell cultures of the same type as those
500 ml of the cell cultures employed for vaccine production used for production of the vaccine. Make a passage after 7
are set aside as uninfected cell cultures (control cells). The days. Maintain the cultures for a further period of14 days and
virus suspension is harvested on one or more occasions then examine the cell cultures for rabies virus using an immune
during incubation. Multiple harvests from the same production fluorescence test. No rabies virus is detected. Alternatively, 5
cell culture may be pooled and considered as a single harvest. m1 ofeach culture fluid is pooled on days 14and21 and 0.03 m1
When vaccine is prepared in embryonated eggs, the egg is inoculated intracerebrally into each ofthe. I0 mice weighing
proteins are minimized by an appropriate method ofpurification. 12 to 15 g. The mice are observed for 14 days for symptoms
The eggs are inoculated with virus seed by the yolle sac route. caused by rabies virus, and mice showing symptoms ofrabies
The infected sterile living embryos are harvested, minced and are sacrificed and virus presence is confirmed by
emulsified in suitable diluent, and stabilizer with aseptic immunofluorescence test or tested for live virus in cell culture
precautions. Emulsions are centrifuged, supernatants are by immunofluorescence test or method of equal sensitivity.
collected and stored as raw virus harvest at a suitable No rabies virus should be detected.
temperature. Residual host-cell DNA (2.2.15). If a continuous cell line is
Viral harvests that comply with the following requirements are used for virus propagation, the content of residual host-cell
pooled in the preparation of the inactivated viral harvest. DNA, detennined using a suitable method, should not be
greater than lOng per single human dose.
Identification
Embryonated egg vaccine
The single harvest contains virus that is identified as rabies
Only concentrated viral suspensions that comply with the
virus using specific antibodies by an approved method.
test for sterility, antigen content and endotoxin requirements
Virus concentration. Titrate for infective virus in cell cultures may be used for preparation of bulle for inactivation.
or by any other approved method. The titre is used to monitor
Inactivation is confirmed by carrying out Mice Inoculation
consistency of production.
Test for residual infectious rabies virus, not more than four
Control cells. The control cells ofthe production cell culture days after inactivation or on the sample frozen4 days after
from which the single harvest is derived should comply with a inactivation are stored at-70°. Inoculate intracerebrally 0.03

2437

www.webofpharma.com
RABIES VACCINE, HUMAN IP 2010

ml into each of 20 mice weighing between 12 and 15 g. The test is already performed on inactivated virus used for final
mice are observed for 14 days for symptoms caused by rabies lot, it may be omitted from the test on final lot.
virus and mice showing symptoms ofrabies are sacrificed and
Sterility (2.2.11). Complies with the test for sterility.
virus presence is confirmed by an immunofluorescence test
or tested for live virus in cell culture by immunofluorescence Bacterial endotoxins (2.2.3). Less than 25 IU per single human
test or method of equal sensitivity. No rabies virus should be dose.
detected. Pyrogens (2.2.8). Complies with the test for pyrogens. Unless
otherwise justified and authorized, inject into each rabbit a
FINAL BULK VACCINE single human dose of the vaccine diluted to ten times its
The final bulk vaccine is prepared from one or more inactivated volume.
viral suspensions. An approved stabilizer may be added to Water (2.3.43). Not more than 3.0 per cent determined by an
maintain the activity of the product during and after freeze- approved method.
drying. Thiomersal can be used as preservative.
Abnormal toxicity (2.2.1). Complies with the test for abnormal
Only a final bulle vaccine that complies with the following toxicity.
requirements may be used in the preparation ofthe final lot.
Accelerated degradation. The potency determined by method
Antigen content. Determine the antigen content by a suitable described under "Assay" ofa sample ofthe preparation under
approved in vitro or in vivo method. The content should be examination after storage at 37° for 4 weeks is not less than 2.5
within the limits approved for the particular product. units per single human dose. This may not be mandatory for
Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle lot release, once the consistency of the product is approved
for each sterility medium. by National Regulatory Authority.
Bovine serum albumin (for cell culture vaccine). Not more
FINAL LOT than 50 ng per single human dose, determined by a suitable
The final bulle vaccine is distributed aseptically into sterile immunochemical method (2.2.14).
containers and can be freeze-dried in case of lyophilized Aluminium content (for gel absorbed vaccine) (2.3.9). Not
products. The containers are then sealed so as to prevent more than 1.25 mg per single human dose.
contamination and the introduction of moisture.
Ovalbumin (for egg based vaccines). Not more than Illg of
Only a final lot that complies with each of the requirements ovalbumin per human dose, determined by a suitable technique
given below under Identification, Tests and Assay may be using a suitable reference preparation of ovalbumin.
released for use. Provided that the test for inactivation has
Residual host-cell DNA (for continuous cell line vaccines)
been carried out with satisfactory results on the inactivated
(2.2.15). Should not be greater than 10 ng per single human
virus suspension and the test for bovine serum albumin has
dose.
been carried out with satisfactory results on the final bulk
vaccine, these tests may be omitted on the final lot. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Identification method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount.
The vaccine is shown to contain rabies virus antigen by a
suitable immunochemical method using specific antibodies, Assay
alternatively, the Assay also serves to identify the vaccine.
The Standard preparation is the international standard or
Tests another suitable preparation, the potency of which has been
determined in relation to the International standard. The
Inactivation. Inoculate a quantity equivalent to not less than
25 human doses of vaccine into cell cultures ofthe same type potency of rabies vaccine is determined by comparing the
as those used for production of the vaccine. Make a passage dose necessary to protect mice against a lethal intracerebral
after 7 days. Maintain the culture for a further 14 days and dose of rabies vaccine necessary to provide the same
then examine the cell culture for rabies virus using an protection. For this comparison, a reference preparation of
. immunofluorescence test. No rabies virus is detected. rabies vaccine, calibrated in international units, and a suitable
Alternatively, injectO.03mlofthe vaccine intracerebrallyintoprep~aratioIiofrablesvrrus~foJ:·useastlieclial1engepreparatlon
each of the 10 mice weighing between 12 and 15g. Neither are necessary.
symptoms of disease in the central nervous system nor death The international unit is the activity contained in a stated
occurs in any ofthe animal within 14 days. Ifthe inactivation quantity of the international standard. The equivalence in

2438

www.webofpharma.com
IP 2010 RUBELLA VACCINE (LIVE)

international units of the international standard is stated by Determination of potency of the vaccine. Reconstitute the
the World Health Organization. standard preparation with a suitable diluent. Prepare at least
three 5-fold serial dilutions of the solution of the standard
The test described below uses a parallel-line model with at
preparation lJ,nd three 5-fold serial dilutions of the vaccine
least three points for the vaccine under examination and the
under examination. For both, the standard preparation and
reference preparation.
the preparation under examination, the serial dilutions should
Test animals. Use mice ofa suitable strain, drawn from a uniform be prepared in such a way that the lowest dilution protects
stock three to four weeks old, weighing between 11 and 15 g. more than 50 per cent ofthe injected mice. Allocate one dilution
Distribute the mice into six groups ofat least 16 mice each and to each of the six groups of 16 mice each. Inject
four groups of 10 mice each and must be ofthe same sex or the intraperitoneally each mouse in each group with dilutions of
sexes must be equally distributed among the groups. the vaccine and reference preparation and repeat the injections.
Throughout the test all mice that die before the fifth day after After 7 days, prepare identical dilutions of the vaccine and
challenge are excluded from the test and all mice that die with reference preparation and repeat the injections.
signs of rabies between the fifth and fourteenth day after After a further 7 days, inject each vaccinated mouse
challenge are counted as failing to resist the challenge. intracerebrally with 0.03 ml of the standard challenge virus
The strain of mice suitable for the test is such that when suspension such that on the basis of preliminary titration,
0.03 ml containing 5 to 50 LD so of the challenge virus 0.03 rnl contains between 5 to 50 LD so ' Observe the mice for 14
suspension is injected intracerebrally per mouse there is 100 days and record the number of mice surviving the challenge
per cent mortality. in each group. Calculate the potency ofthe preparation under
Standard challenge virus suspension. A working pool ofthe examination by standard statistical methods.
challenge virus strain is prepared by injecting intracerebrally The vaccine complies with the test ifthe estimated potency is
0.03ml ofa 10 fold dilution ofthe CVS strain ofrabies virus in not less than 2.5 ill per single human dose.
2 per cent v/v sterile inactivated normal horse serum in water
The test is not valid unless (a) for both the preparation under
for injection or another suitable diluent approved by the
examination and the standard preparation, the 50 per cent
competent authority into a suitable number of test animals.
protective dose lies between the largest and smallest doses
The animals when moribund after showing characteristic signs
given to the mice; (b) there is not deviation from linearity or
of rabies are sacrificed and their brains harvested aseptically.
parallelism of the dose response lines, the confidence limit
They are then washed in chilled saline solution to remove
(P = 0.95) are not less than 25.0 per cent and not more than 400
blood clots. A 10 per cent suspension ofthe brains is prepared
per cent ofthe estimated potency; (c) the titre ofthe challenge
in a suitable diluent approved by the competent authority and
virus suspension lies between 5 to 50 LD so '
thoroughly homogenised. After centrifuging lightly, the
supernatant liquid is distributed into sterile vials and freeze Labelling. The label states the biological origin of the cells
dried. The sealed and freeze-dried supernatant liquid containing used for the preparation of the vaccine.
vials are stored at -20°. When stored under prescribed
conditions the virus titre of the freeze-dried preparation may
be expected to be maintained for not less than 3 years.
Alternatively, the washed brains are homogenised in a suitable
Rubella Vaccine (Live)
diluent approved by the competent authority to give 10 per Rubella Vaccine (Live) is a freeze-dried preparation ofa suitable
cent suspension. It is then centrifuged lightly, distributed into attenuated strain ofrubella virus. The vaccine is reconstituted
sterile ampoules or sterile plastic vials and sealed. The sealed immediately before use to give a clear liquid or may be coloured
ampoules or plastic vials can be stored at -60° or below. When owing to the presence of a pH indicator.
stored under prescribed conditions, the virus titre may be
expected to be maintained for not less than one year. Storage Production
time needs to be validated by the manufacturer.
General provisions
Virus titre of the challenge virus. Prepare ten fold serial
dilutions of the standard challenge virus suspension. Using The production ofvaccine is based on a virus seed-lot system
the four groups of 10 mice each, inject 0.03 ml of the virus and a cell-bank system. The production method shall have
suspension intracerebrally into each mouse, using a different been shown to yield consistently live rubella vaccines of
group for each suspension. Observe the mice for 14 days. adequate immunogenicity and safety in man. Unless otherwise
Calculate the virus titre of the standard challenge virus justified and authorised, the virus in the final vaccine shall
suspension in LD so per dose of 0.03 ml by standard statistical have undergone no more passages from the master seed lot
methods. than were used to prepare the vaccine shown in clinical studies

2439

www.webofpharma.com
RUBELLA VACCINE (LIVE) IP 2010

to be satisfactory with respect to safety and efficacy. production cell culture may be pooled and considered as a
The production method is validated to demonstrate that the single harvest.
product, if tested, would comply with the test for safety and Only a single harvest that complies with the following
efficacy. requirements may be used in the preparation ofthe final bulk
vaccine.
Substrate for virus propagation
Identification
The virus is propagated in human diploid cells (2.7.2).
The single harvest contains virus that is identified as rubella
SEED LOT virus by serum neutralization in cell culture, using specific
The strain of rubella virus used in the production of rubella antibodies.
vaccine shall be identified by historical records that include Virus concentration. The virus concentration in the single
information on the origin of the strain and its subsequent harvest is detennined as prescribed under Assay to monitor
manipulation. consistency ofproduction and to determine the dilution to be
used for the final bulle vaccine.
To avoid unnecessary use of monkeys in the test for
neurovirulence virus seed lots are prepared in large quantities Extraneous agents (2.7.3). The single harvest complies with
and stored at temperatures below -20° iffreeze-dried, or below the tests for extraneous agents.
-60° ifnot freeze-dried. Control cells. The control cells comply with a test for
identification and with the tests for extraneous agents (2.7.3).
Only a seed lot that complies with the following tests may be
used for virus propagation. FINAL BULK VACCINE
Single harvests that comply with the above tests are pooled
Identification
and clarified to remove cells. A suitable stabilizer may be added
The master and working seed lots are identified as rubella and the pooled harvests diluted as appropriate.
virus by serum neutralisation in cell culture, using specific Only a final bulk vaccine that complies with the following
antibodies. requirements may be used in the preparation of the final lot.
Virus concentration. The virus concentration of the master Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk
and working seed lots is determined to ensure consistency of for each sterility medium.
production.
FINAL LOT
Extraneous agents (2.7.3). The working seed lot complies with
the tests for seed lots. A minimum virus concentration for release of the product is
established such as to ensure, in the light of stability data,
Neurovirulence (2.7.5). The master/working seed lot complies
that the minimum concentration stated on the label will be
with the test for neurovirulence oflive virus vaccines. Macaca
present at the end of the period of validity.
and Cercopithecus monkeys are suitable for the test.
Only a final lot that complies with the tests for minimum virus
PROPAGATION AND HARVEST concentration for release, with the following requirement for
thermal stability and with each ofthe requirements given below
All processing of the cell bank and subsequent cell cultures is
under Identification and Tests may be released for use.
done under aseptic conditions in an area where no other cells
Provided that the test for bovine serum albumin has been
are handled. Suitable animal (but not human) serum may be
carried out with satisfactory results on the final bulk vaccine,
used in the growth medium, but the final medium for maintaining
it may be omitted on the final lot.
cell growth during virus multiplication does not contain animal
serum. Serum and trypsin used in the preparation of cell Thermal stability. Maintain samples ofthe final lot offreeze-
suspensions and culture media are shown to be free from dried vaccine in the dry state at 37° for 7 days. Determine the
extraneous agents. The cell culture medium may contain a pH virus concentration as described under Assay in parallel for
indicator such as phenol red and suitable antibiotics at the the vaccine held at 37° for 7 days and for vaccine stored at 2°
lowest effective concentration. It is preferable to have a to 8°. The virus concentration of the heated vaccine is not
substrate free from antibiotics during production. Not less more than 1.0 10glO lower than that of the unheated vaccine.
than5_QQ_ml._oLthe_produ.ctionc_ell_culture is set aside as Identification- -------------- ------------- -------------
_uninfected cell culture (control celIs). The temperatur.e of
incubation is controlled during the growth of the virus. The When the vaccine reconstituted as stated. 011 tIle label is mixed
virus suspension is harvested, on one or more occasions, with specific rubella antibodies, it is no longer able to infect
within 28 days ofinoculation. Multiple harvests from the same susceptible cell cultures.

2440

www.webofpharma.com
IP 2010 TETANUS ANTITOXIN

Tests Identification
Sterility (2.2.11). The reconstituted vaccine complies with the Specifically renders the corresponding venom or venoms
test for sterility. harmless to susceptible animals. Itmay also be identified by
Water (2.3.43). Not more than 3.0 per cent, detennined by Karl any other alternate suitable in vitro method.
Fischer, semi-micro detennination ofwater or by any suitable
validated method. Tests
Bovine serum albumin. Not more than 50 ng per single human Potency. In view of the numerous toxic fractions in venoms
dose, determined by a suitable immunochemical method and varying requirements of different localities, no standard
(2.2.14). for potency is prescribed.
Abnormal toxicity (2.2.1). Complies with the test for abnormal The potency of the snake venom antiserum is detennined by
toxicity. estimating the ability ofthe antiserum to protect mice or other
suitable animals against the lethal effect of a fixed dose of a
Assay
reference preparation of snake venom ofthe relevant species
Titrate the vaccine for infective virus at least in triplicate, or by comparing its ability to do so with that of a reference
using at least five cell cultures for each 0.5 lag lo dilution step preparation of antiserum of established potency at two or
or by a method of equal precision. Use an appropriate virus more dose levels ofthe venom.
reference preparation to validate each assay. The estimated Other tests. Complies with the tests stated under Antisera.
virus concentration is not less than that stated on the label;
the minimum virus concentration stated on the label is not Storage. Store the freeze-dried preparation in a cool, dark place
less than 1 x 103 CCID so per human dose. The assay is not and avoid exposure to excessive heat. Store the liquid
valid ifthe confidence limits (P=0.95) of the logarithm ofthe preparation at a temperature between 2° and 8°. It should not
virus concentration is greater than ± 0.3. be allowed to freeze.

Rubella vaccine (Live) RS is suitable for use as a reference Labelling. The label states (1) the volume ofthe contents and
preparation. in case of freeze-dried preparation, the directions for
reconstitution; (2) the species of snake against whose venom
Labelling. The label states (1) the strain ofvirus used for the
the antiserum is effective; (3) the animal species from which
preparation ofthe vaccine; (2) the type and origin ofthe cells
the antiserum has been obtained; (4) the name and proportion
used for the preparation ofthe vaccine; (3) the minimum virus
of any preservative added; (5) that in case of a liquid
concentration; (4) the time within which thevaccine must be
preparation, it should not be allowed to freeze.
used after reconstitution; (5) that the vaccine must not be
given to a pregnant woman and that a woman must not become
pregnant within two months.

Tetanus Antitoxin
Snake Venom Antiserum Tetanus Antitoxin is a preparation containing the specific
Snake Antivenin; Snake Antivenom Serum; Snake antitoxic globulins or their derivatives obtained by purification
Venom Antitoxin of hyperimmune serum of horses or other suitable animals
and have the specific activity ofneutralising the toxin fonned
Snake Venom Antiserum is a sterile preparation containing by Clostridium tetani. The liquid preparation may contain a
antitoxic globulins or their derivatives that have the power of suitable antimicrobial preservative.
specifically neutralising the venom of one or more species of
Tetanus Antitoxin has a potency of not less than 1000 Units
snakes. It may be obtained from the serum or plasma ofhealthy
equines or other suitable animals immunised against venoms per ml when intended for prophylactic use and not less than
of specific species usually elapine and viperine. It may be a 3000 Units per ml when intended for therapeutic use.
purified liquid preparation or a freeze-dried product. It may Description. A clear, colourless or pale yellow liquid or a freeze-
contain a suitable preservative. dried, cream-coloured powder or pellet.
Description. A clear to slightly opalescent, colourless or pale Identification
yellow liquid, free from suspended particles or cream-coloured
powder or pellet which when reconstituted with the diluent Specifically neutralises and renders the toxin formed by
supplied by the manufacturer with the freeze-dried product C. tetani hannless to susceptible animals or by any other
yields a clear, colourless or pale yellow liquid. suitable in vitro test.

2441

www.webofpharma.com
TETANUS ANTITOXIN IF 2010

Tests mixed with 0.1 Unit of the Standard Preparation and injected
subcutaneously into mice causes tetanic paralysis within 4
Other tests. Complies with the tests stated under Antisera. days. Prepare a solution of the Standard Preparation in a
Potency. Cany out the biological assay of tetanus antitoxin suitable liquid such that 1 ml contains 0.5 Unit. Accurately
described below. measure or weigh a quantity ofthe test toxin and dilute it with,
or dissolve it in, a suitable liquid. Prepare mixtures such that
Biological Assay of Tetanus Antitoxin each contains 2.0 ml ofthe solution ofthe Standard Preparation
(1 Unit); one of a series of graded volumes ofthe solution of
The potency of tetanus antitoxin is determined by comparing
the toxin, and sufficient of a suitable liquid to give a final
the dose necessary to protect mice against the paralytic effects
volume of 5.0 ml. Allow the mixtures to stand at room
ofa fixed dose of tetanus toxin with the dose ofthe Standard
temperature, protected from light, for 60 minutes and then
Preparation of tetanus antitoxin necessary to give the same
inject 0.5 ml of each mixture subcutaneously into mice, six
protection. For this purpose the Standard Preparation of
mice being used for each mixture, and observe the mice for 4
tetanus antitoxin and a suitable preparation of toxin, for use
days.
as a test toxin, are required. The test dose of the toxin is
determined in relation to the Standard Preparation and the The test (Lp/lO) dose ofthe toxin is the amount present in 0.5
potency of the preparation under examination is then ml ofthatmixture that contains the smallest amount of toxin
determined in relation to the Standard Preparation using the sufficient to cause tetanic paralysis in all six mice injected
test toxin. with it within 4 days.

Standard Preparation Determination ofpotency ofthe antitoxin. Prepare a solution


bfthe StahdardPtepll.ratiCll1 in a suitableliquidsllch thatit
The Standard Preparatioil is the 2nd International Standard contains 0.5 Unit per ml. Accurately measure or weigh a
for Tetanus antitoxin, equine, established in 1969, consisting quantity of the test toxin and dilute it with, or dissolve it in, a
of freeze-dried hyperimmune horse serum (supplied in suitable liquid so that 1.0 ml contains five times the Lp/lO
ampoules containing 1400 Units), or another suitable dose as previously determined. Prepare mixtures such that
preparation the potency of which has been determined in each contains 2.0 ml of the solution of the test toxin and one
relation to the International Standard. of a series of graded volumes of the preparation under
examination, and sufficient of a suitable liquid to give a final
Suggested Method volume of5.0 ml. Prepare similar mixtures containing 2.0 ml of
NOTE - The severity ofthe tetanic paralysis to be regarded the test toxin and one of a series of graded volumes of the
as the end-point is such that the paralysis is readily solution of the Standard Preparation centred on that volume
recognised but not sL@ciently extensive to cause significant (2.0 ml) that contains 1 Unit. Allow the mixtures to stand at
suffering. For humane reasons the animals should be room temperature, protected from light, for 60 minutes and
examined at least Mice a day and should be killed as soon then inoculate 0.5 ml of each mixture subcutaneously into
as the end-point is reached. each mouse, six mice being used for each mixture, and observe
the mice for 4 days. The mixture that contains the largest
Test animals. Use healthy mice, weighing between 17 to 22 g,
volume of the preparation under examination that fails to
from the same stock.
protect the mice from paralysis contains 1 Unit. The test is not
Preparation oftest toxin. Prepare tetanus toxin from a sterile valid unless all the mice injected with mixtures containing 2.0
filtrate ofan 8 to 10 day culture of C. tetani. Test toxin may be ml or less of the solution of the Standard Preparation show
prepared by adding this filtrate to glycerin in the propOltion paralysis and all those injected with more do not. Calculate
of 1 volume ofthe filtrate to 1 or 2 volumes of glycerin. This the potency of the preparation under examination in Units
solution oftest toxin is stored below 0°. Test toxin may also be perml.
prepared in stable form by saturating the filtrate with
ammonium sulphate, collecting the resulting precipitate, Storage. As stated under Antisera.
drying it over phosphorus pentoxide at a pressure of 1.5 to 2.5 Labelling. The label states (1) the number ofUnits per ml; (2)
kPa and reducing it to a fine powder. The powder so obtained species of animal from which the preparation has been made;
is preserved in the dry condition at a low temperature either in (3) whether the preparation is for prophylactic or for therapeutic
sealed ampoules or over phosphorus pentoxirjf!~~ ~J)!~~~~!~ use; (4)Jhe_xecommendeddose;_(5)_the name and proportion
of 1.5 to 2.5 kPa. of any added preservative; (6) thl:!t the preparation, if liquid,
Determination of test dose of toxin (Lp/l0 dose). First should not be allowed to freeze; (7) that the preparation, if
determine the Limes paralyticum/l 0 (Lp/10) dose of the test dried, should be used immediately after reconstitution in the
toxin. This is the smallest quantity of the toxin which when stated quantity of the diluent supplied by the manufacturer.

2442

www.webofpharma.com
IP 2010 TETANUS VACCINE (ADSORBED)

Tetanus Vaccine (Adsorbed) sensitive assay for active tetanus toxin, such as inoculation
into mice or guinea-pigs. The toxoid complies with the test if
Tetanus Vaccine (Adsorbed) is a preparation oftetanus formol neither sample produces any sign of a toxic reaction attribut-
toxoid adsorbed on mineral carrier. The formol toxoid is able to tetanus toxin.
prepared ii-om the toxin produced by the growth of Clostridium
tetani. Antigenic purity. Not less than 1,000 Lf per mg of protein
nitrogen.
Production
FINAL BULK VACCINE
General provisions
The final bulk vaccine is prepared by adsorption of a suitable
The maximum number ofLfper single human dose oftetanus quantity ofbulk purified toxoid onto a mineral carrier such as
vaccine is 25. The production method is validated to hydrated aluminium phosphate or aluminium hydroxide; the
demonstrate that the product, if tested, would comply with resulting mixture is approximately isotonic with blood. Suitable
the following test. antimicrobial preservatives may be added. Certain antimicrobial
preservatives, particularly those of the phenolic type,
BULK PURIFIED TOXOID
adversely affect the antigenic activity and must not be used.
For the production of tetanus toxin, from which toxoid is Only final bulk vaccine that complies with the following
prepared, seed cultures are managed in a defined seed-lot requirements may be used in the preparation of the final lot.
system in which toxinogenicity is conserved and, where
necessary, restored by deliberate reselection. A highly Specific toxicity. Use five nonnal, healthy guinea pigs weigh-
toxinogenic strain of C. tetani with lmown origin and history ing between 250 g and 350 g which have been maintained for
is grown in a suitable liquid medium. At the end ofcultivation, at least one week on a uniform, unrestricted diet, have not lost
the purity of each culture is tested and contaminated cultures weight during this period and have not been previously treated
are discarded. Toxin-containing culture medium is collected with any material that will interfere with the test. Weigh the
aseptically. The toxin content (Lfper ml) is checked to monitor animals separately and record their weights. Inject subcuta-
consistency of production. Single harvests may be pooled to neously into each animal five times the dose stated on the
prepare the bulle purified toxoid. The toxin is purified to remove label. Weigh all the animals at weeldy intervals for 3 weeks.
components likely to cause adverse reactions in humans. The None of the animals shows any symptoms of tetanus tox-
purified toxin is detoxified with formaldehyde by a method aemia or dies from tetanus within 21 days or loses weight at
that avoids destruction of the immunogenic potency of the the end of the test. If more than one animal dies from non-
toxoid and reversion oftoxoid to toxin, particularly on exposure specific causes or loses weight repeat the test. If an animal
to heat. Alternatively, purification may be carried out after dies or loses weight in the second test, the vaccine fails the
detoxification. test.

Only bulk purified toxoid that complies with the following Antimicrobial preservative. Where applicable, determine the
requirements may be used in the preparation of the final bulle amount of antimicrobial preservative by a suitable chemical
vaccine. method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent ofthe intended amount.
Sterility (2.2.11). Cany out test for sterility using 10 ml of
bulle for each sterility medium. Free formaldehyde (2.3.20). Maximum 0.2 gil.
Absence of tetanus toxin. Inject subcutaneously at least 500 pH (2.4.24).6.0 to 7.0.
Lf of purified toxoid in a volume of 1 ml into each of five Sterility (2.2.11). Cany out test for sterility using 10 ml ofbulle
healthy guinea pigs, each weighing 250 to 350 g, that have not for each sterility medium.
previously been treated with any material that will interfere
with the test. None of the animals shows any symptoms of Abnormal toxicity (2.2.1). Complies with the test for
tetanus toxaemia or dies from tetanus within 21 days or loses abnormal toxicity for antisera and vaccine.
weight at the end ofthe test. Ifmore than one animal dies from
FINAL LOT
non-specific causes or loses weight repeat the test. Ifan animal
dies or loses weight in the second test the toxoid fails the test. The final bulle vaccine is distributed aseptically into sterile,
Irreversibility oftoxoid. Using the buffer for the final vaccine, tamper-proof containers. The containers are closed. so as to
prepare a dilution of the bulle purified toxoid containing the prevent cont~mination.
same toxoid concentration as the final vaccine. Divide the Only a final lot that is satisfactory with respect to each ofthe
dilution into two equal parts. Keep one ofthem at 2° to 8° and requirements given below under Identification, Tests and
the other at 37° for 6 weeks. Test both dilutions by a suitable Assay may be released for use. Provided the tests for

2443

www.webofpharma.com
TETANUS VACCINE (ADSORBED) IP 2010

antimicrobial preservative, free formaldehyde and the assay Biological assay of adsorbed tetanus vaccine
have been carried out with satisfactory results on the final
The potency of adsorbed tetanus vaccine is detennined by
bulle vaccine, they may be omitted on the final lot.
comparing the dose ofthe vaccine required to protect guinea-
Identification pigs or mice from the paralytic effects of a subcutaneous
injection of tetanus toxin with the dose of the Standard
Tetanus toxoid is identified by a suitable immunochemical preparation needed to give the same protection. For this
method (2.2.14). The following method, applicable to certain comparison, the Standard preparation of adsorbed tetanus
vaccines, is given as an example. Dissolve in the vaccine under toxoid and a suitable preparation oftetanus toxin, for use as a
examination sufficient sodium citrate to give a 10 per cent challenge toxin, are necessary.
solution. Maintain at 37° for about 16 hours and centrifuge
until a clear supernatant liquid is obtained. The clear Standard preparation
supernatant liquid reacts with a suitable tetanus antitoxin,
The Standard preparation is International standard for Tetanus
giving a precipitate.
toxoid, adsorbed or another suitable preparation, the potency
Tests of which has been determined in relation to the International
standard.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
if aluminium hydroxide or hydrated aluminium phosphate is Suggested method
used as the adsorbent. The paralysis method is not obligatory and the lethal method
Free formaldehyde (2.3.20). Maximum 0.2 gil. may be used. For this method the number of animals and the
procedure are identical with those describedfci:dhepariilysis
Antimicrobial preservative. Where applicable, determine the
method but the end-point is the death of the animal rather
amount of antimicrobial preservative by a suitable chemical
than the onset of paralysis and the LD IO dose is used instead
method. The content is not less than 85.0 per cent and not
ofthe LD 50 dose.
greater than 115.0 per cent ofthe intended amount.
Sterility (2.2.11). Complies with the test for sterility. (a) Test on guinea pigs

Abnormal toxicity (2.2.1). Complies with the test for abnormal Test animals. Use healthy guinea-pigs from the same stock
toxicity for antisera and vaccine. and weighing between 250 and 350 g. Distribute them into six
groups of sixteen each. The guinea-pigs should all be of the
Potency of tetanus component same sex or the males and females should be distributed equally
between the groups. If the challenge toxin to be used has not
Assay. Detennine by either ofthe following methods. been shown to be stable or has not been adequately
(1) Inject subcutaneously on each of two occasions separated standardised, include four further groups of five guinea pigs
by an interval of not more than 4 weeks, one-tenth of the to serve as unvaccinated controls.
stated human dose diluted to 1 ml with saline solution into Challenge toxin. Select a preparation of tetanus toxin
each of9 normal, healthy guinea pigs weighing between 250 containing not less than 50 times the 50 per cent paralytic
and 350 g. Not more than 2 weeks after the second injection, dose per ml. Ifthe challenge toxin preparation has been shown
collect the serum from each animal and carry out the biological to be stable, it is not necessary to verify the paralytic dose for
test for tetanus antitoxin, described under Tetanus antitoxin every assay.
or any other method approved by National Regulatory
. Authority. Preparation ofthe challenge toxin solution. Immediately prior
to use, prepare from the challenge toxin by dilution with
Sera of at least 6 guinea pigs out of 9 should contain not less phosphate buffered saline pH 7.4 a challenge toxin solution
than 0.5 Unit oftetanus antitoxin per ml. containing fifty times the 50 per cent paralytic dose per ml. If
(2) Carry out the biological assay ofadsorbed Tetanus Vaccine necessary, dilute portions of this challenge toxin solution 16,
(Adsorbed) described below. 50 and 160 fold with the same buffer solution.
If the lower limit of the 95.0 per cent confidence interval of Determination of potency. Prepare in saline solution three
estimated potency is less than 40 IU per single human dose dilutions ofthe vaccine under examination and three dilutions
then the limitsofthe 95;0 percent confidence interval ofthe ofasolution-ofthe-Standard-preparation such -that,.for-eaGh,
estimaterofpotency shall be within 50 to 200 per cent of the the dilutions forma series differing by-not more than 2.5 fold
estimated potency unless the lower limit of the 95.0 per cent steps and in which the dilutions ofintermediate concentration,
confidence interval of the estimated potency is greater than when injected subcutaneously in 1 ml volumes into guinea-
40 IU per single human dose. pigs, protect approximately 50 per cent ofthe animals from the

2444

www.webofpharma.com
IP 2010 TETANUS VACCINE (ADSORBED)

paralytic effects ofthe subcutaneous injection of the quantity Determination of potency. Prepare in saline solution three
of tetanus toxin prescribed for this test. Allocate the six dilutions ofthe vaccine under examination and three dilutions
dilutions one to each of the six groups of sixteen guinea pigs of a solution of the Standard preparation such that, for each,
and inject subcutaneously 1.0 ml of each dilution into each the dilutions form a series differing by not more than 2.5 fold
guinea pig in the group to which that dilution is allocated. steps and in which the dilutions ofintermediate concentration,
After 28 days inject each animal subcutaneously with 1.0 ml when injected subcutaneously in 0.5 ml volumes into mice,
of the challenge toxin solution containing fifty times the protect approximately 50 per cent. of the animals from the
50 per cent paralytic dose. Ifnecessary, allocate the challenge paralytic effects of the subcutaneous injection ofthe quantity
toxin solution and the three dilutions made from it one to each of tetanus toxin prescribed for this test. Allocate the six
of the four groups of five guinea pigs and inject dilutions one to each of the six groups of sixteen mice and
subcutaneously 1.0 ml ofeach toxin solution into each guinea- inject subcutaneously 0.5 ml ofeach dilution into each mouse
pig in the group to which that toxin solution is allocated. in the group to which the dilution is allocated. After 28 days
Examine the guinea pigs twice daily, remove and kill all animals inject each animal subcutaneously with 0.5 ml ofthe challenge
showing definite signs oftetanus paralysis. Count the number toxin solution containing fifty times the 50 per cent paralytic
of guinea pigs without paralysis 5 days after injection of the dose. If necessary, allocate the challenge toxin solution and
challenge toxin and calculate the potency ofthe vaccine under the three dilutions made from it one to each ofthe four groups
examination relative to the potency ofthe Standard preparation of six mice and inject subcutaneously 0.5 ml of each toxin
on the basis of the number of animals without paralysis in solution into each mouse in the group to which that toxin
each of the six groups of sixteen, using standard statistical solution is allocated. Count the number of mice without
methods. paralysis 4 days after injection of the challenge toxin and
calculate the potency ofthe vaccine under examination relative
The test is not valid unless (a) for both the vaccine under
to the potency ofthe Standard preparation on the basis of the
examination and the Standard preparation, the 50 per cent
numbers ofanimals without paralysis in each ofthe six groups
protective doses lie between the largest and smallest doses of
of sixteen, using standard statistical methods.
the preparations given to the guinea pigs; (b) if applicable,
the number of paralysed animals among the four groups of The test is not valid unless (a) for both the vaccine under
five injected with the challenge toxin solution and its dilutions examination and the Standard preparation, the 50 per cent
indicate that the challenge was approximately 50 times the 50 protective doses lie between the largest and smallest doses of
per cent paralytic dose; (c) the fiducial limits of assay lie the preparations given to the mice; (b) ifapplicable, the number
between.50.0 per cent and 200.0 per cent of the estimated of paralysed animals among the four groups of six injected
potency; (d) the statistical analysis shows no deviations from with the challenge toxin solution and its dilutions indicate
linearity or parallelism. The test may be repeated any number that the challenge was approximately 50 times the 50 per cent
o~times but when more than one test is performed the results paralytic dose; (c) the fiducial limits ofthe assay lie between
of all valid tests must be combined in the estimate ofpotency. 50.0 per cent and 200.0 per cent of the estimated potency;
(d) the statistical analysis shows no deviation from linearity
(b) Test on mice or parallelism. The test may be repeated any number oftimes
but when more than one test is performed the results of all
Test animals. Use healthy mice from the same stock, weighing
valid tests must be combined in the estimate of potency.
between 14 and 20 g. Distribute them into six groups ofsixteen
each. If the challenge toxin to be used has not been shown to (c) Determination ofantibodies in guinea pigs
be stable or has not been adequately standardised, include
Preparation of serum samples. For preparation of serum
four further groups of six. mice to serve as unvaccinated
samples, the following technique has been found suitable.
controls. The mice should all be of the same sex or the males
Invert the tubes containing blood samples 6 times and allow
and females should be distributed equally among the groups.
to stand at 37 0 for 2 h, then at 4 0 for 2 hours, centrifuge at room
Challenge toxin. Select a preparation of tetanus toxin temperature at 800 g for 20 min. transfer the serum to sterile
containing not less than 100 times the 50 per cent paralytic tubes and store at a temperature below -20 0 • At least 40.0 per
dose perml. cent yield of serum is obtained by this procedure.
Preparation ofthe challenge toxin solutions. Immediately prior Determination of antibody titre. The ELISA and ToBI tests
to use prepare from the challenge toxin by dilution with shown below are given as examples ofimmunochemical
phosphate buffered saline pH 7.4 a challenge toxin solution methods that have been found suitable for the determination
containing fifty times the 50 per cent paralytic dose in each 0.5 of antibody tire.
ml. Ifnecessary, dilute portions ofthis challenge toxin solution Determination of antibody titre in guinea pig serum by
16, 50 and 160 fold with the same buffer solution. enzyme-linked immunosorbent assay (ELISA). Dilutions of

2445

www.webofpharma.com
TETANUS VACClNE (ADSORBED) IP 2010

test and reference sera are made on ELISA plates coated with thoroughly with washing buffer. Block the plates by addition
tetanus toxoid. A positive guinea"pig serum control and a of 100 III of diluent block buffer to each well. Incubate in a
negative guinea-pig serum control are included on each plate humid atmosphere at 379 for I h. Wash the plates thoroughly
to monitor the assay performance. Peroxidase-conjugated with washing buffer. Place 100 III of diluent block buffer in
rabbit or goat antibody directed against guinea-pig-IgG is each well ofthe plates, except those ofrow A. Prepare suitable
added followed by a peroxidase substrate. Optical density is dilutions of negative control serum; positive control serum
measured and the relative antibody titre is calculated using (from about 0.01 IU/ml) and test sera. Allocate the negative
the usual statistical methods; control sentm to column l,positive control serum to columns
2 and 3 and test sera to columns 4-12 and add 100 III of each
Reagents and equipment
serum to the fIrst 2 wells ofthe column to which it is allocated.
ELISAplates: 96 wells, columns 1-12, rowsA-H. Using a multi channel micropipette, make twofold serial
Clostridium tetani guinea-pig antiserum (jor vaccines-human dilutions from row B down the plate to row H by transferring
use) reference preparation (positive control serum). 100 III to the following well. Discard 100 III from the last row so
that all wells contain 100 Ill. Incubate at 37° for 2 h. Wash
Peroxidase conjugate. Peroxidase-conjugated rabbit or goat thoroughly with washing buffer. Prepare a suitable dilution (a
antibody directed against guinea-pig IgG. 1 in 2000 dilution has been found suitable) of peroxidase
Tetanus toxoid. conjugate in diluent block buffer and add 100 III to each well.
Incubate at 37° in a humid atmosphere for 1 h. Wash the plates
Carbonate coating buffer pH 9.6. Dissolve 1.59 g of anhyd,;ous
thoroughly with washing bzif.[er. Add 100 III of peroxidase
sodium carbonate and 2.93 g of sodium hydrogen carbonate
substrate to each well. Allow to stand at room temperature,
in 1000 ml of wate/: Distribute into 150 m1 bottles and sterilise
protected from light, for 30 min. Read the plates at 405 nm in
by autoclaving at 121 ° for 15 min.
the same order as addition of substrate was made.
Phosphate buffered saline pH 7.4 (PBS). Dissolve with stirring
80.0 g of sodium chloride, 2.0 g of potassium dihydrogen Determination ofantibody titre in guinea-pig serum by toxin-
phosphate, 14.3 g of disodium hydrogen phosphate dihydrate or toxoid-binding inhibition(ToBI). Tetanus toxin or toxoid is
and 2.0 g ofpotassium chloride in 1000 ml ofwate/: Store at added to serial dilutions oftest and reference sera; the seruml
room temperature to prevent crystallisation. Dilute to 10 times antigen mixtures are incubated overnight. To determine
its volume with water before use. unbound toxin or toxoid, the mixtures are transferred to an
ELISA plate coated with tetanus antitoxin. Peroxidase-
Citric acid solution. Dissolve 10.51 g of citric acid in 1000 m1 conjugated equine anti-tetanus IgG is added followed by a
of water and adjust the solution to pH 4.0 with a 400 gil solution peroxidase substrate. Optical density is measured and the
of sodium hydroxide. antibody titre is calculated using the usual statistical methods.
Washing buffer. PBS containing 0.5 gil ofpolysorbate 20. A positive control serum and a negative control serum are
Diluent block buffer. PBS containing 0.5 gil ofpolysorbate 20 included on each plate to monitor assay performance.
and 25 gil ofdried skimmed milk.
Reagents and equipment
Peroxidase substrate. ShOltly before use, dissolve 10 mg of
diammonium 2, 2'-azinobis(3-ethylbenzothiazoline-6- Round-bottomed, rigid polystyrene microtitre plates.
sulphonate) (ABTS) in 20 ml of citric acid solution. Flat-bottomed ELISA plates.
Immediately before use add 5 III of strong hydrogen peroxide
Tetanus toxin or tetanus toxoid.
solution.
Clostridium tetani guinea-pig antiserum (jor vaccines-human
Method use) reference preparation.
The description below is given as an example of a suitable Equine anti-tetanus IgG.
plate lay-out but others may be used. Wells 1A-H are for
negative control serum and wells 2A-H and 3A-H are for Peroxidase-conjugated equine anti-tetanus IgG.
positive control serum for assay monitoring. Wells 4-12A-H Carbonate buffer pH 9.6. Dissolve 1.5 g of anhydrous sodium
are for test samples. carbonate, 2.39 g of sodium hydrogen carbonate and 0.2 g of
<::;Q£lt(l£l<:11 W(lll QfJhe.E1ISAplate~wi!11 100 Ill. ()f tetanus sodium azide in 1000 m1 of wateJ; adjustto pH 9.6 and autoclave
aTi2T'5Tor20J:rilii.~-~- .---- -.. --.--- -- -- ._~--~-

toxoid solution (0.5 Lflml in carbonate coating buffer).


Allow to stand overnight at 4° in a humid atmosphere. To Sodium acetate buffer pH 5.5. Dissolve 90.2 g of anhydrous
avoid interference from temperature gradient, do not stack sodium acetate in 900 ml ofwateJ; adjust to pH 5.5 using a
more than 4 plates high. On the following day, wash the plates saturated solution of citric acid monohydrate and dilute to

2446

www.webofpharma.com
IP 2010 TICK-BORNE ENCEPHALITIS VACCINE (INACTIVATED)

1000 ml with wate!: conjugated equine anti-tetanus IgG in diluent buffer. Add
100 !J.I of the dilution to each well and cover the plates with a
Phosphate buffered saline pH 7.2 (PBS). Dissolve 135.0 g of
lid. Incubate at 37° in a humid atmosphere for 1.5 h. Wash the
sodium chloride, 20.55 g of disodium hydrogen phosphate
ELISA plates thoroughly with washing solution. Add 100 !J.I
dihydrate and 4.80 g of sodium dihydrogen phosphate
ofperoxidase substrate to each well. A blue colour develops.
monohydrate in water and dilute to 15 litres with the same
Incubate the plates at room temperature. Stop the reaction at
solvent. Autoclave at 100° for 60 min.
a given time (within 10 min) by the addition of 100 !J.l of2 M
Diluent buffer. PBS containing 5 gil of bovine albumin and sulphuric acid to each well in the same order as the addition
0.5 gil of polysorbate 80. ofsubstrate. The colour changes from blue to yellow. Measure
Block buffer. PBS containing 5 gil of bovine albumin. the absorbance (2.4.7) at 450 nm immediately after addition of
the sulphuric acid or maintain the plates in the dark until
Tetramethylbenzidine solution. 6 gil solution of tetramethyl- reading.
benzidine in alcohol. The substance dissolves within 30-40
min at room temperature. (d) Any other validated serological assay in guinea-pigs/mice
Peroxidase substrate. Mix 90 ml of wate!; 10 ml of sodium approved by National RegulatoryAuthority.
acetate buffer pH 5.5, 1.67 ml of tetramethylbenzidine solution Labelling. The label states (1) the human dose (ml); (2) the
and 20 !J.I of strong hydrogen peroxide solution. minimum units per single human dose or the minimum
Washing solution. Tap water containing 0.5 gil ofpolysorbate International Units per single human dose ifpotency test done
80. by challenge method; (3) the name and the amount of the
adsorbent and preservative; (4) that the vaccine must be
Method shaken before use; (5) that the vaccine is not to be frozen.

Block the round-bottomed polystyrene microtitre plates by


placing in each well 150 !J.I of block buffer. Cover the plates
with a lid or sealer. Incubate in a humid atmosphere at 37° for Tick-borne Encephalitis Vaccine
1 h. Wash the plates thoroughly with washing solution. Place
100 !J.I ofPBS in each well. Place 100 !J.I of reference guineapig (Inactivated)
tetanus antitoxin in the first well of a row. Place 100 !J.I of Tick-borne Encephalitis Vaccine (Inactivated) is a liquid
undiluted test sera in the first well of the required number of preparation ofa suitable strain ofticle-borne encephalitis virus
rows. Using a multichannel micropipette, make it two fold serial grown in cultures of chick-embryo cells or other suitable cell
dilutions across the plate (up to column 10), by transfer of 100 cultW"es and inactivated by a suitable, validated method.
!J.I to the following well. Discard 100 !J.l from the last column so
that all wells contain 100 !J.l. Prepare 0.1 Lfiml solution oftetanus Production
toxin or toxoid using PBS a diluent. Add 40 !J.I ofthis solution
to all wells except those column 12. The wells ofrow 11 are a General provisions
positive control. Add 40 !J.I of PBS to the wells of column 12 The vaccine complies with the General Requirements of
(negative control). Shake the plates gently and cover them Vaccines for Human Use.
with lids. Coat the ELISA plates: immediately before use make
a suitable dilution of equine anti-tetanus IgG in carbonate Production of the vaccine is based on a virus seed lot system.
bufferpH 9. 6 and add 100 !J.I to all wells. Incubate the 2 series The production method shall have been shown to yield
of plates overnight in a humid atmosphere at 37°. To avoid consistently vaccines comparable with the vaccine ofproven
temperature gradient effects, do not stack more than 4 plate clinical efficacy and safety in man. The virus in the final vaccine
high. Cover the plates with lids. On the following day, wash shall not have undergone more passages from the master seed
the ELISA plates thoroughly with washing solution. Block lot than the virus in the vaccine used in clinical trials.
the plates by placing in each well 125 !J.I of block bzlffir. Incubate The production method is validated to demonstrate that the
at 37° in a humid atmosphere for 1 h. Wash _the plates product, if tested, would comply with the test for abnormal
thoroughly with washing solution. Transfer 100 !J.I ofthe pre- toxicity and immunogenicity.
incubation mixture from the polystyrene plates to the
corresponding wells ofthe ELISA plates, starting with column Substrate for virus propagation
12 and then from 1 to 11.· Cover the plates with a lid. Incubate
at 37° in a humid atmosphere for 2 h. Wash the ELISA plates The virus is propagated in chick embryo cells prepared from
thoroughly with washing solution. Make a suitable dilution eggs derived from a chicken flock free from specified pathogens
(a 1 in 4000 dilution has been found suitable) ofthe peroxidase- (2.7.7) or in other suitable cell cultures.

2447

www.webofpharma.com
TICK-BORNE ENCEPHALITIS VACCINE (INACTIVATED) IP 2010

SEED LOT Inactivation

The strain ofvirus used is identified by historical records thatTo avoid interference, viral aggregates are removed by :filtration
include information on the origin of the strain and its immediately before the inactivation process. The virus
subsequent manipulation. Virus seed lots are stored at or below suspension is inactivated by a validated method; the method
-60°. shall have been shown to be consistently capable of
Only a seed lot that complies with the following requirements inactivating tick-borne encephalitis virus without destroying
may be used for virus propagation. the antigenic and immunogenic activity; as part of the
validation studies, an inactivation curve is plotted representing
Identification residual live virus concentration measured on not fewer than
Each seed lot is identified as containing the vaccine strain of three occasions. If fonnaldehyde is used for inactivation, the
tick-borne encephalitis virus by a suitable immunochemical presence of an excess offree fonnaldehyde is verified at the
method, preferably using monoclonal antibodies. end of the inactivation process.
Virus concentration. The virus concentration of each seed Only an inactivated harvest that complies with the following
lot is determined by titration in suitable cell cultures to monitor requirements may be used in the preparation of the final bulle
consistency of production. vaccine.
Extraneous agents (2.7.3). Each seed lot complies with the Residual infective virus. Inoculate a quantity ofthe inactivated
requirements for extraneous agents in viral vaccines for human harvest equivalent to not less than ten human doses ofvaccine
use; the tests in cell cultures are carried out in human and in the final lot into primary chicken fibroblast cell cultures, or
simian cells only. other cells shown to be at least as sensitive to tick-borne
encephalitis virus with not less than 3 cmz of cell sheet per ml
PROPAGATION AND HARVEST of inoculum. Incubate at 37 ± 1° for 14 days. No cytopathic
All processing of the cell cultures ifperfonned under aseptic effect is detected at the end ofthe incubation period. Collect
conditions in an area where no other cells are being handled. the culture fluid and inoculate 0.03 ml intracerebrally into each
Serum and trypsin used in the preparation of cell suspensions of not fewer than ten mice about 4 weeks old. Observe the
and media used must be shown to be free from extraneous mice for 14 days. They show no evidence of tick-borne
agents. The cell culture media may contain a pH indicator encephalitis virus infection.
such as phenol red and approved antibiotics at the lowest Purification
effective concentration. At least 500 ml of the cell cultures
employed for vaccine production is set aside as uninfected Several inactivated single harvests may be pooled before
cell cultures (control cells). concentration and purification by suitable methods, preferably
by continuous-flow, sucrose density-gradient centrifugation.
Only a single harvest that complies with the following
requirements may be used in the preparation ofthe inactivated Only a purified, inactivated harvest that complies with the
harvest. following requirements may be used in the preparation offinal
bulle vaccine.
Identification
Sterility ( 2.2.11). Complies with the test for sterility, carried
The single harvest is shown to contain tick-borne encephalitis out using 10 ml for each medium.
virus by a suitable immunochemical method, preferably using Specific activity. Detennine the antigen content ofthe purified,
monoclonal antibodies, or by virus neutralization in cell inactivated harvest by a suitable immunochemical method
cultures. (2.2.14). Detennine the total protein content by a suitable
Sterility (2.2.11). Complies with the test for sterility carried method. The specific activity, calculated as the antigen content
out using 10 ml for each medium. per unit mass of protein, is within the limits approved for the
Mycoplasma (2.7.4). Complies with the test for mycoplasmas specific product.
carried out using I ml for each medium.
FINAL BULK VACCINE
Control cells. The control cells comply with the tests for
extraneous agents (2.7.3). If the vaccine is produced using a The final bulle vaccine is prepared from one or more purified,
for inactivated harvests.
identification. Qnly a final bulle vaccin.e that cOlnplies. with the following
Virus concentration. Determine the virus concentration by requirement may be used in the preparation ofthe final lot.
titration in suitable cell culture to monitor consistency of Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulle
production. for each sterility medium.

2448

www.webofpharma.com
IP 2010 TUBERCULIN PURIFIED PROTEIN DERIVATIVE

FINAL LOT validity criteria four to five dilutions will usually be necessary.
Prepare dilutions such that the most concentrated suspension
Only a final lot that is satisfactory with respect to each of the
is expected to protect more than 50 per cent ofthe animals and
requirements given below under Identification, Tests and
the least concentrated suspension less than 50.0 per cent.
Assay may be released for use. Provided that the tests for :free
Allocate each dilution to a different group of mice and inject
formaldehyde, bovine serum albumin (where applicable) and
subcutaneously into each mouse 0.2 ml ofthe dilution allocated
pyrogens and the assay have been carried out satisfactory
to its group. Seven days later make a second injection using
results on the final bulle vaccine, they may be omitted on the
the same dilution scale. 14 days after the second injection
final lot.
prepare a suspension of the challenge virus containing not
Identification less than 100 LD so in 0.2 m!. Inject 0.2 ml of this virus
suspension intraperitoneally into each vaccinated mouse. To
The vaccine is shown to contain tick-borne encephalitis virus verify the challenge dose, prepare a series of not fewer than
antigen by a suitable immunochemical method using specific three dilutions ofthe challenge virus suspension at not greater
antibodies or by the mouse immunogenicity test described than one-hundred fold intervals. Allocate the challenge
under Assay. suspension and the four dilutions, one to each of the five
Tests groups often mice, and inject intraperitoneally into each mouse
0.2 ml ofthe challenge suspension or the dilution allocated to
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, its group. Observe the animals for 21 days after the challenge
if aluminium hydroxide or hydrated aluminium phosphate is and record the number of mice that die in the period between
used as the adsorbent. 7 and 21 days after the challenge.
Free formaldehyde (2.3.20). Maximum 0.1 gil. Calculations. Calculate the results by the usual statistical
Bovine serum albumin. If bovine serum albumin has been methods for an assay with quantal responses (5.7).
used during production, the vaccine contains not more than Validity criteria. The test is not valid unless (l) the
50 ng per single human dose, determined by a suitable . concentration ofthe challenge virus is not less than 100 LD 50;
immunochemical method (2.2.14). (2) for both the vaccine under examination and the reference
Sterility (2.2.11). Complies with the test for sterility. preparation'the 50.0 per cent protective dose (PD so) lies
between the largest and smallest doses given to the mice; (3)
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
the statistical analysis shows a significant slope and no
into each rabbit, per kg of body mass, one dose of vaccine.
significant deviation from linearity and parallelism ofthe dose-
Assay response lines; (4) the fiducial limits (P = 0.95) are not less
than 33.0 per cent and not more than 300.0 per cent of the
The potency is determined by comparing the dose necessary
to protect a given proportion of mice against the effects of a estimated potency.
lethal dose of tick-borne encephalitis virus, administered Potency requirement. Include all valid tests to estimate the
intraperitoneally, with the quantity of a reference preparation mean potency and the fiducial limits (P = 0.95) for the mean
of tick-borne encephalitis vaccine necessary to provide the potency; compute weighed means with the inverse of the
same protection. For this comparison an approved reference squared standard error as weights. The vaccine complies with
preparation and a suitable preparation of tick-borne the test ifthe estimated potency is not less than that approved
encephalitis virus from an approved strain for use as the by the competent a)lthority, based on data from clinical efficacy
challenge preparation are necessary. trials.
The following is cited as an example ofa method that has been Labelling. The label states (1) the strain of virus used in
found suitable for a given vaccine. preparation; (2) the type of cells used for production of the
Selection and distribution oftest animals. Use healthy mice vaccine.
weighing between 11 and 17 g derived from the same stock.
Distribute the mice into not less than six groups of a suitable
size to meet the requirements for validity ofthe test; for titration Thberculin Purified Protein Derivative
of the challenge suspension, use not fewer than four groups
often mice. Use mice of the same sex or distribute males and Tuberculin PPD; Tuberculin Purified Protein Derivative for
females equally between groups. Human Use
Determination ofpotency of the vaccine. Prepare not less Tuberculin Purified Protein Derivative is a preparation made
than three suitable dilutions ofthe vaccine under examination from the heat-treated products of growth and lysis of one or
and of the reference preparation; in order to comply with more strains of Mycobacterium tuberculosis that reveal

2449

www.webofpharma.com
TUBERCULIN PURIFIED PROTEIN DERIVATIVE IP 2010

delayed hypersensitivity in animals sensitised by a micro~ Live mycobacteria


organism of the same species. A. Inject 5.0 ml intraperitoneally or subcutaneously into each
oftwo guinea-pigs weighing between 300 and 400 g. Observe
Production
the animals for not less than 42. days. Kill the animals and
It is prepared from the water-soluble fraction obtained by carry out an autopsy. No guinea-pig shows signs of infection
heating in free-flowing steam or in an autoclave and with mycobacteria.
subsequently filtering cultures of the mycobacteda grown in B. Carry out tests for live mycobacteria in the preparation
a suitable liquid medium. The active fraction in the filtrate, under examination using suitable culture media. No growth of
which is predominantly protein, is separated by precipitation, mycobacteria should occur.
washed and redissolved. The preparation is free from
Sensitising effect. Inject intradell11ally into each of three
mycobacteria. An antimicrobial preservative that does not give
guinea-pigs three times, at intervals of 5 days, a dose of the
rise to false positive reactions, such as 0.5 per cent w/v of
preparation under examination containing about 500 Units in
phenol, and a suitable stabiliser may be added. Phenol is not
a volume of0.1 m!. Two to three weeks after the third injection
added to preparations that are to be freeze-dTied. The final
inoculate the same dose intradellllally into these animals and
sterile product is distributed into sterile glass containers which
into a control group of three guinea-pigs of the same weight
are then sealed so as to prevent microbial contamination or
but that have not had any previous injections of tuberculin.
alternatively it is freeze-dTied and the containers subsequently
The reactions ofthe two groups are not significantly different
sealed.*
after 48 to 72 hours.
* To ensure availability of a preparalion of uniform pOlency, Tuberculin
Purified Prolein Derivative is produced and issued by Ihe Sialens Serum Toxicity. Inject subcutaneously into 2 healthy guinea-pigs,
Inslilule, Denmark as a powder 10 be reconslilllied as slaled on Ihe weighing not less than 250 g and which have not previously
label. been treated with any material which will interfere with the
The preparation may be issued either as a sterile liquid or as a test, 0.5 ml ofa solution containing 1,00,000 Units per m!. No
freeze-dTied product. If issued as a liquid, it is in a ready-to- hall11ful effects are produced within 7 days.
use form and 0.1 ml constitutes one intradermal dose Biological assay
containing appropriate number ofUnits. If issued as a freeze-
The potency of tuberculin purified protein derivative is
dTied product, it should yield a ready-to-use preparation when
detell11ined by comparing the dose necessary to reveal delayed
reconstituted as per manufacturer's instructions.
hypersensitivity in guinea-pigs or other animals
Description. A colourless or pale, straw-coloured liquid, or hypersensitised with mycobacteria of the same type as that
dry cream-coloured powder, or pellet. used in the preparation of the tuberculin purified protein
The preparation, reconstituted if necessary as stated on the derivative with the dose of the appropriate Standard
label, complies wi(h the follOWing requirements. Preparation necessary to give the same effect.

Identification The estimated potency is not less 80 per cent and not more
than 125 per cent ofthe stated potency. The fiducial limits of
A. When progressively increasing doses are injected error are not less than 64 per cent and not more than 156 per
intradellllally into specifically sensitised guinea-pigs, reactions cent of the stated potency.
occur at the points of injection, varying from erythema to
necrosis. When similar injections are .administered to non- Standard Preparation
sensitised guinea pigs no such reactions occur.
The Standard Preparation is the 1st International Standard for
B. The potency test described below serves as a test for Tuberculin, purified protein derivative (PPD), mammalian,
identity ifit is perfoll11ed on material from the final containers. established in 1951, consisting ofPPD derived from cultures
ofM tuberculosis (supplied in ampoules containing 5,00,000
Tests
Units) or another suitable preparation the potency of which
pH (2.4.24). 6.5 to 7.5. has been detellllined in relation to. the International Standard.
Phenol (ifpresent) (2.3.36). Not more than 0.5 per cent w/v.
Method
Sterility (2.2.11). Complies with the tests for sterility.
Sensitise 12 guinea-pigs eachweighingnotlessthan400g by
Potency. Carry out the biological assay oftuberculin purified the intramuscular injection of a total of about 0.5 ml of a
protein derivative described below.
suspension in a suitable mineral oil with or without emulsifier
Tuberculin Purified Protein Derivative in fi'eeze-dried form and containing 0.1 mg per ml of heat-inactivated, dried
complies with the following additional requirements. mycobacteria ofthe same type as that used in the preparation

2450

www.webofpharma.com
IP 2010 TYPHOID (STRAIN Ty 21a) VACCINE, LIVE (ORAL)

of tuberculin. Use phosphate-buffered saline pH 7.4 flagellar H:d antigen and does not produce hydrogen sulphide
containing 0.005 per cent w/v of polysorbate 80 to prepare on Kligler iron agar. The strain is nonvirulent for mice. Cells of
the dilutions of the Standard preparation and of the strain Ty 21 a lyse if grown in the presence of 1.0 per cent of
preparation under examination and to reconstitute freeze~dried galactose.
preparations containing no stabiliser. Not less than 1 month
SEED LOT
and not more than 6 months later carry out the following
test: The vaccine is prepared using a seed-lot system. The working
Shave the flanks to provide space for at least three reactions seed lots represent not more than one subculture from the
on each side, but not more than a total of 12 injection sites per master seed lot. The final vaccine represents not more than
animal. Use at least three doses of the Standard preparation four subcultures £i:om the Oliginal vaccine on which were made;
and at least three doses of the preparation under examination, the laboratory and clinical tests showing the strain to be
the highest dose being about 10 times as strong as the lowest. suitable.
Dilute the preparations so that the lesions produced are 8 to Only a master seed lot that complies with the following
25 mm in diameter. Allocate the doses to the available sites in requirements may be used in the preparation of working seed
a random manner, in a Latin square design. Inject each dose lots.
intradermally in the same volume (0.1 or 0.2 ml) at the sites to
Galactose metabolism
which it has been allocated. Measure the diameters of the
lesions after 24 to 48 hours and calculate the result ofthe test In a spectrophotometric assay, no activity of the enzyme
using standard statistical methods on the basis that the lesion uridine diphosphate-galactose-4-epimerase is found in the
diameters are directly proportional to the logarithm of the cytoplasm of strain Ty 21a compared to strain Ty 2.
concentration of the tuberculin.
Biosynthesis of lipopolysaccharide
Storage. Store in light-resistant containers at a temperature
between 2° and 8°. It should not be allowed to freeze. Lipopolysaccharides are extracted by the hot-phenol method
and examined by size-exclusion chromatography (2.4.16). Strain
Labelling. The label states (1) the number ofUnits per dose of
Ty 21a grown in medium free of galactose shows only the
0.1 ml or per ml or per mg; (2) the total volume in the container
rough (R) type of lipopolysaccharide.
(for liquid preparation); (3) the nature and quantity of the
reconstituting liquid (for the freeze-dried preparation); (4) the Serological characteristics
name and proportion of any added substances; (5) the species
Strain Ty 21a grown in a synthetic medium without galactose
or strain used; (6) the storage conditions; (7) the date after
does not agglutinate to specific anti-O:9 antiserum. Whatever
which the contents are not intended to be used; (8) that care
the growth conditions, strain Ty 21 a does not agglutinate to
should be taken to avoid inhaling the powder (for the freeze-
Vi antiserum. Strain Ty 21a agglutinates to H:d flagellar
dried preparation).
antiserum.

Biochemical markers
Typhoid (Strain Ty 21a) Vaccine, Live StrainTy 21a does not produce hydrogen sulphide on Kligler
(Oral) iron agar. This property serves to distinguish Ty 21 a from
other galactose-epimerase-negative S. typhi strains.
Typhoid Vaccine (Live, Oral, strain Ty 21a) is a freeze-dried
CeUgrowth
preparation of the live Salmonella typhi strain Ty 21a grown
in a suitable medium. When presented in capsules, the vaccine Strain Ty 21a cells lyse when grown in the presence of 1.0 per
complies with the tests stated under Capsules. cent of galactose.
Production PROPAGATION AND HARVEST
Choice of vaccine strain The bacteria from the working seed lot are multiplied in a
preculture, subcultured once and are then grown in a suitable
The main characteristic of the strains is the defect of the
medium containing 0.001 per cent ofgalactose at 30° for 13 to
enzyme uridine diphosphate-galactose-4 epimerase. The
15 hours. The bacteria are harvested. The harvest must be
activities of galactopermease, galactokinase and galactose-l-
free from contaminating micro-organisms.
phosphate uridyl-transferase are reduced by 50 to 90 per cent.
Whatever the growth conditions, the strain does not contain Only a single harvest that complies with the following
Vi antigen. The strain agglutinates to anti-O : 9 antiserum only requirements may be used for the preparation of the freeze-
if grown in medium containing galactose. It contains the dtied harvest.

2451

www.webofpharma.com
TYPHOID (STRAIN Ty 21a) VACCINE, LIVE (ORAL) IP 2010

pH (2.4.24).6.8 to 7.5. each dosage unit must contain not less than 4 x 109 live
bacteria.
Optical density
The optical density of the culture, measured at 546 nm, is 6.5 Identification ,
to 11.0. Before carrying out the measurement,dilute the culture Culture bacteria from the vaccine under examination on an
so that a reading in the range 0.1 to 0.5 is obtained and correct agar medium containing 1.0 per cent of galactose and
the reading to take account of the dilution. bromothymol blue. Light blue, concave colonies transparent
due to lysis of cells, should be found. No yellow colonies
Identification (galactose-fermenting) should be found.
Culture bacteria on an agar medium containing 1.0 per cent of
Tests
galactose and bromothymol blue. Light blue, concave
colonies, transparent due to lysis of cells, should be found. Microbial contamination (2.2.9). Carry out the test using
No yellow colonies (galactose-fermenting) should be found. suitable selective media. Determine the total viable count using
the plate-count method. The number of contaminating micro-
FREEZE-DRIED HARVEST organisms per dosage unit is not greater than 102 bacteria and
20 fungi. No pathogenic bacterium, particularly Escherichia
The harvest is mixed with a suitable stabilizer and freeze-dried
coli, Staphylococcus aureus, Pseudomonas aeruginosa, and
by a process that ensures the survival of at least 10.0 per cent
Salmonella other than strain Ty 21 a are found.
of the bacteria and to a water content shown to be favourable
to the stability ofthe vaccine. No antimicrobial preservative is Water (2.4.43). 1.5 to 4.0 per cent.
added to the vaccine.
Number of live bacteria
Only a freeze-dried harvest that complies with the following
Carry out the test using not less than five dosage units.
tests may be used for the preparation of the final bulle.
Homogenise the contents ofthe dosage units in a 0.9 per cent
Identification w/v solution of sodium chloride at 4 0 using a mixer in a cold
room with sufficient glass beads to emerge from the liquid.
Culture bacteria are examined on an agar medium containing Immediately after homogenization prepare a suitable dilution
1.0 per cent of galactose and bromoethymol blue. Light blue, of the suspension using cooled diluent and inoculate brain
concave colonies, transparent due to lysis of cells, should be heart infUsion agar, incubate at 36 ± 10 for 20 to 36 hours. The
found. No yellow colonies (galactose-fermenting) should be vaccine contains not less than 2 x 109 live S. typhi Ty 2la
found. bacteria per dosage unit.
Number oflive bacteria Labelling. The label states (1) the minimum number oflive
Not less than 1 x 1011 live S. lJphi strain Ty 21a per gram. bacteria per dosage unit; (2) that the vaccine is for oral use
only.
Water (2.4.43). 1.5 to 4.0 per cent, determined by the semi-
micro detennination of water or any other validated method.

FINAL BULK VACCINE Typhoid Polysaccharide Vaccine


The final bulle vaccine is prepared by aseptically mixing one or Typhoid Polysaccharide Vaccine is a preparation of purified
more freeze-dried harvests with a suitable sterile excipient. Vi capsular polysaccharide obtained from Salmonella typhi
Only a final bulle that complies with the following requirement Ty2 strain or some other suitable strain of known origin and
may be used in the preparation of the final lot. history that has the capacity to produce Vi polysaccharide,
and which have been characterized by suitable biochemical,
Number of live bacteria. Not less than 40 x 109 live S. typhi
physicochemical, serological or molecular methods.
strain Ty 21 a per gram.
Capsular polysaccharide is a partly 3-0-acetylated repeated
FINAL LOT units of 2-acetylamino-2-deoxy-D-galactopyranuronic acid
The final bulle vaccine is distributed under aseptic conditions with a-(1-74)-linkages.
c:aJ~~lllles with a shell or into suitable
containers.
Only a final lot that is satisfactory with respect to Identification,
Ginerarpiovisi()-lis'
Tests and Number of live bacteria will be released for use, The production of Vi polysaccharide is based on a defined
except that in the determination of number of live bacteria seed-lot system. The method of production shall have been

2452

www.webofpharma.com
IP 2010 TYPHOID POLYSACCHARIDE VACCINE

shown to yield consistently Viapolysaccharide typhoid Only those pools of purified Vi-polysaccharide that comply
vaccines of adequate immunogenicity and safety in man. The with the following requirements may be used in the preparation
production method is validated to demonstrate that the product, ofthe final bulle:
if tested, would comply with the tests for abnormal toxicity.
Protein (2.7.1). Not more than 10 mg per gram of
SEED LOT polysaccharide, calculated with reference to the dried
substance.
The strain of S. typhi used for the master seed lot shall be
identified by historical records that include information on its Nucleic acids (2.7.1). Not more than 20 mg per gram of
origin and by its biochemical and serological characteristics. polysaccharide, calculated with reference to the dried
Cultures from the working seed lot shall have the same substance.
characteristics as the strain that was used to prepare the master O-Acetyl groups (2.7.1). Not less than 2 mmol per gram of
seed lot, shall be demonstrated along with adequate polysaccharide, calculated with reference to the dried
documentation. substance.
Only a strain that has the following characteristics may be
Molecular size. Examine by gel filtration or size-exclusion
used in the preparation ofthe vaccine: (a) stained smears from
chromatography (2.4.16) using cross-linked agarose for
a culture are typical of S. typhi; (b) the culture utilises glucose
chromatography. Use a column 0.9 m long and 16 mm in intemal
without production of gas; (c) colonies on agar are oxidase-
diameter equilibrated with a solvent having an ionic strength
negative; (d) a suspension of the culture agglutinates
of 0.2 mol per kg and a pH of7.0 to 7.5. Apply about 5 mg of
specifically with an appropriate Vi antiserum or colonies form
polysaccharide in a volume of 1 ml to the column and elute at
haloes on an agar plate containing a suitable Vi antiserum.
about 20 ml/h. Collect fractions ofabout 2.5 ml. Determine the
PROPAGATION AND HARVEST point corresponding to K o = 0.25 and make two pools
consisting of fractions eluted before and after this point.
The worldng seed lot is cultured on a solid medium, which Determine O-acetyl groups on the two pools. Not less than 50
may contain blood-group substances, or a liquid medium; the per cent ofthe polysaccharide is found in the pool containing
inoculum obtained is transferred to a liquid medium, which is fractions eluted before K o = 0.25.
used to inoculate the fmal medium. The liquid medium used
and the final medium are semi-synthetic, fi'ee from substances Identification
that are precipitated by cetrimonium bromide and do not
contain blood-group substances or high-molecular-mass Carry out an identification test using a suitable
polysaccharides, unless it has been demonstrated that they immunochemical method (2.2.14).
are removed by the purification process. The bacterial purity Bacterial endotoxins (2.2.3). Determine by a suitable method,
ofthe culture is verified by microscopic examination ofGram- the content should not be more than 150 IU per mg of
stained smears and by inoculation into appropriate media. polysaccharide.
The culture is then inactivated at the beginning of the
stationary phase by the addition ofJormaldehyde. Bacterial FINAL BULK VACCINE
cells are eliminated by centrifugation; the polysaccharide is
precipitated from the culture medium by addition of One or more batches of purified Vi polysaccharide are
hexadecyltrimethylammonium bromide (cetrimonium dissolved in a suitable solvent, which may contain an
bromide). The precipitate is harvested and may be stored at antimicrobial preservative, so that the volume corresponding
or below -20 0 before purification. to one dose contains 25 J.tg ofpolysaccharide and the solution
is isotonic with blood (250 mosrn/kg to 350 mosrn/kg).
Purified Vi Polysaccharide
Only a final bulle vaccine that complies with the following
The polysaccharide is purified, after dissociation of the requirements may be used in the preparation of the final lot;
polysaccharide/cetrimonium bromide complex, using suitable
procedures to eliminate successively nucleic acids, proteins Sterility (2.2.11). Cany out test for sterility using 10 ml of
and lipopolysaccharides. The polysaccharide is precipitated bulle for each sterility medium.
in its salt form and dried at 5±3°; the powder obtained Antimicrobial preservative. Where applicable, deterniine the
constitutes the purified Vi polysaccharide. The loss on drying amount of antimicrobial preservative by a suitable chemical
is determined by thermogravimetry, Karl Fischer or any other method. The content is not less than 85.0 per cent and not
suitable method and is used to calculate the results of the greater than 115.0 per cent of the intended amount. Ifphenol
chemical tests shown below with reference to the dried has been used in the preparation, the content is not more than
substance. 2.5 gil.

2453

www.webofpharma.com
TYPHOID POLYSACCHARIDE VACCINE IF 2010

FINAL LOT absorbance for the five reference solutions and the
corresponding content ofacetylcholine chloride and read from
The final bulle vaccine is distributed and filled aseptically into
the curve the content of acetylcholine chloride in the test
sterile containers. The containers are closed so as to prevellt
solution for each vo1mne tested. Calculate the mean of the
contamination.
two values.
Only a final lot that is satisfactory with respect to each of the
1 mol ofacetylcholine chloride (181.7 g) is equivalent to 1 mol
requirements prescribed below under Identification, Tests and
ofO-acetyl(43.05 g).
Assay and with the requirements for Bacterial endotoxins may
be released for use. Provided the tests for free fonnaldehyde Free formaldehyde(2.3.20). Maximum 0.2 gil.
and antimicrobial preservative have been carried out on the
Antimicrobial preservative. Where applicable, detennine the
final bulle vaccine, they may be omitted on the final lot.
amount of antimicrobial preservative by a suitable chemical
The vaccine contains minimum of 25 J.Lg purified Vi- method. The content is not less than 85.0 per cent and not
Polysaccharide per dose of 0.5 ml. greater than 115.0 per cent of the intended amount.

Identification If phenol has been used in the preparation, the content is not
more than 2.5 gil.
Carry out an identification test using a suitable
immunochemical method (2.2.14). Assay
Determine Vi polysaccharide content by a suitable
Tests
immunochemical method (2.2.14) using a reference purified
Sterility (2.2.11). Complies with the tests forsterility; polysaccharide. The estimated amount of polysaccharide per
Abnormal toxicity (2.2.1). Complies with the test for abnonnal dose is 80.0 per cent to 120.0 per cent ofthe content stated on
toxicity. the label. The fiducial limits oferror (P = 0.95) ofthe estimated
amount of polysaccharide are not less than 80.0 per cent and
pH (2.4.24). 6.5 to 7.5. not more than 120.0 per cent.
O-Acetyl groups (2.7.1). 0.085 (± 25 per cent) J.Lmol per dose Labelling. The label states (1) the number of micrograms of
(25 J.Lg ofpolysaccharide). polysaccharide per human dose (25 J.Lg); (2) the total quantity
Test solution. Place 3 ml of the vaccine in each ofthree tubes of polysaccharide in the container.
(two reaction solutions and one correction solution).
Reference solutions. Dissolve 0.150 g of acetylcholine
chloride in 10 ml of water (stock solution containing 15 gil of Typhoid Paratyphoid A Vaccine
acetylcholine chloride). Immediately before use, dilute 0.5 ml Typhoid Paratyphoid A Vaccine is a sterile suspension or a
of the stock solution to 50 ml with water (working dilution freeze-dried solid prepared from one or more strains of
containing 150 J.Lg/ml of acetylcholine chloride). In ten tubes, Salmonella typhi and S. paratyphi A that are smooth and
place in duplicate (reaction and correction solutions) 0.1 ml, have the full complement of 0, H and Vi antigens in S. typhi
0.2 ml, 0.5 ml, 1.0 ml and 1.5 ml ofthe working dilution. strain, and 0 and H antigens in S. paratyphi A strain. The
Prepare a blank using 3 ml ofpurified water. bacteria are killed by heat or by a bactericide such as phenol
or fonnaldehyde or by a chemical such as acetone. The liquid
Make up the volume in each tube to 3 ml with water. Add vaccine and any diluent supplied by the manufacturer with
0.5 ml of a mixture of 1 volume of water and 2 volumes of the freeze-dried vaccine contains a suitable preservative. The
dilute hydrochloric acid to each of the correction tubes and dried vaccine contains no preservative.
to the blank. Add 1.0 m1 of alkaline hydroxylamine solution
to each tube. Allow the reaction to proceed for exactly 2 min Typhoid Paratyphoid A Vaccine for adults contains not less
and add 0.5 ml ofa mixture ofl volume of water and 2 volumes than 1000 million S. typhi and 500 million S. paratyphi A
of dilute hydrochloric acid to each of the reaction tubes. organisms per ml. The vaccine meant for children contains
Add 0.5 ml of a 20 per cent wlv solution offerric chloride in 333 million S. typhi and 167 million ofS. paratyphi A organisms
a.2M hydrochloric acid to each tube, stopper the tubes and pennl.
shCl!~~_"igQr()ll.sly torelll()"e. bll1:>1:>l~s~ ;Q~~~r:ipti()!1~ I~eliqlli(t()l:t.l!l:l.~l:l<:()l1sti!Ute<ly~<:cineis~Vv'1:>ite
Measure the absorbance of each solution at 540 TIm using the or pale yellow turbid liquid free from clumps.
blank as the compensation liquid. For each reaction solution, Identification
subtract the absorbance of the corresponding correction
solution. Draw a calibration curve from the corrected Identify by specific agglutination.

2454

www.webofpharma.com
IP 2010 TYPHUS VACCINE

Tests Typhoid Vaccine (Freeze Dried)


pH (2.2.24). 6.0 to 7 A. Freeze Dried Typhoid Vaccine is a freeze-dried preparation of
Other tests. Complies with the tests stated under Vaccines inactivated Salmonella typhi. The vaccine is reconstituted as
with the following modifications. stated on the label to give a uniform suspension containing
not less than 5 x 108 and not more than 1 x 109 bacteria per
Phenol (2.3.36) (ifpresent). Not more than 0.5 per cent w/v.
human dose. The human dose does not exceed 1.0 ml of the
Abnormal toxicity (2.2.1 ). Complies with the test for abnormal reconstituted vaccine.
toxicity injectirig subcutaneously, intramuscularly or
intraperitoneally. Production
Storage. Store at a temperature 2° to 8°. The liquid vaccine The vaccine is prepared from a seed-lot system from a suitable
must not be allowed to freeze. strain of S. typhi, such as Ty 2. The final vaccine represents
Labelling. The label states (1) the number of organisms per not more than 3 subcultures from the strain on which were
ml; (2) whether the vaccine is meant for use in adults or for use made the laboratory and clinical tests that showed it to be
in children; (3) the recommended dose; (4) the name and suitable. The bacteria are inactivated either by acetone or by
proportion ofany added preservative; (5) that in case ofliquid formaldehyde or by heat. Phenol is not used in the preparation.
preparation, the vaccine should not be allowed to freeze. The vaccine is distributed into sterile containers and freeze-
dried to a moisture content favourable to the stability of the
vaccine.
Typhoid Vaccine The production method is validated to demonstrate that the
product, iftested, would comply with test for abnormal toxicity
Typhoid Vaccine is a sterile suspension of inactivated (2.2.1), modified to the extent that 0.5 ml of the vaccine is
Salmonella typhi containing not less than 5 x 108 and not injected subcutaneously or intramuscularly or
more than 1 x 109 bacteria per human dose. The human dose intraperitoneaUy into each mouse and 1.0 ml into each guinea-
does not exceed 1.0 ml. pig.

Production Identification
The vaccine is prepared using a seed-lot system from a The vaccine reconstituted as stated on the label is identified
suitable strain of S. typhi such as, Ty 2. The final vaccine by specific agglutination.
represents not more than 3 subcultures from the strain on
Antigenic power. When injected into susceptible laboratory
which were made the laboratory and clinical tests that showed
animals, the reconstituted vaccine elicits anti-O, anti-H and,
it to be suitable. The bacteria are inactivated by acetone, by
to a lesser extent, anti-Vi agglutinins.
formaldehyde, by phenol or by heating or by a combination
of the last two methods. Sterility (2.2.11). The reconstituted vaccine complies with the
tests for sterility.
The production method is validated to demonstrate that the
product, if tested, would comply with the test for abnormal Water. Not more than 5.0 percent.
toxicity (2.2.1) modified to the extent that 0.5 rnl ofthe vaccine Labelling. The label states (1) the method used to inactivate
is injected subcutaneously or intramuscularly or the bacteria; (2) the number of bacteria per human dose; (3)
intraperitoneally into each mouse and 1.0 ml into each guinea- that the vaccine should be used within 8 hours of
pig. reconstitution.
Identification
It is identified by specific agglutination.
Typhus Vaccine
Phenol (2.3.36). If phenol has been used in the preparation,
the concentration is not more than 0.5 per cent w/v. Typhus Vaccine is a sterile suspension ofkiUed rickettsiae of
a strain or strains of epidemic typhus rickettsiae (Rickettsia
Antigenic power. When injected into susceptible laboratory prowazeki) selected for antigenic efficiency.
animals, it elicits anti-O, anti-H and, to a lesser extent, anti-Vi
agglutinins. The vaccine is prepared by injecting virulent rickettsiae into
the yolk sacs of fertile eggs which have been incubated for 7
Sterility (2.2.11). Complies with the test for sterility. days. Within 9 to 13 days after the injection, the yolk sacs are
Labelling. The label states (1) the method used to inactivate collected under aseptic conditions and subjected to suitable
the bacteria; (2) the number of bacteria per human dose. treatment to liberate the maximum number ofrickettsiae. The

2455

www.webofpharma.com
TYPHUS VACCINE IP 2010

material is suspended in saline solution or other suitable ampoules for freezing. Dilute a small amount ofthis suspension
solution isotonic with blood to which formaldehyde solution 8-, 16-, 32- and 64-fold with saline solution. Inject
has been added so that the concentration of formaldehyde is intravenously doses of 0.5 ml of each dilution into 8 white
0.2 per cent to 0.5 per cent. The suspension so formed contains mice, each weighing between 11 and 15 g. 18 hours later,
10 per cent to 15 per cent w/w ofyolk sac tissue. It is purified estimate the toxicity of the preparation as the LD so per g of
by treatment with ether. The aqueous middle layer of the yollc sac, calculated from the numbers of mice killed by each
resultant mixture is collected and distributed under aseptic dilution. Those toxic suspensions showing 160 or more LDso
conditions into sterile containers which are then sealed so as per g ofyollc sac are satisfactory for use in the neutralisation
to exclude micro-organisms. test. The remainder of the suspension may be sealed in
ampoules and preserved for 18 months in liquid nitrogen.
The vaccine may also be prepared from the lungs of small
rodents in which rickettsial pneumonias have been caused by Prepare mixtures of the toxic substance and the sera under
inhalation ofmassive doses ofvirulent rickettsiae, or from the test for the neutralisation test in mice and allow to stand for 2
peritoneal cavities of gerbils which have received hours before injection. Inject intravenously into mice, each
intraperitoneal injections of rickettsiae. weighing between 11 and 15 g, 0.5 ml containing 2 LDso ofthe
toxic substance together with serial 1 to 2 dilutions of serum.
Description. A slightly turbid liquid; a white deposit of the
Keep the mice in an incubator at a temperature between 28°
rickettsiae may separate on standing which can be readily
and 30° until the results of the test are recorded. Record the
redistributed on shaking.
deaths at the end of 24 hours and express the results in terms
Identification of the highest dilution giving complete protection.
Storage. Store at a temperature 2° to 8° ; When stored under
The vaccine specifically protects laboratory animals against
the prescribed conditions, the vaccine may be expected to
epidemic typhus.
retain its potency for at least 1 year.
Tests Labelling. The label states (1) the nature of the preparation,
Sterility (2.2.11). Complies with the tests for sterility. i.e., whether it has been prepared in eggs, rodent's lungs or
otherwise and whether it has been purified; (2) the name and
Abnormal toxicity (2.2.1). Complies with the test for abnonnal proportion of any added preservative.
toxicity injecting subcutaneously, intramuscularly or
intraperitoneally.
Potency. Carry out the biological assay of typhus vaccine Varicella Vaccine, Live
described below. The vaccine passes the test if the serum of
Varicella Vaccine (Live) is a freeze-dried preparation ofa suitable
immunised guinea-pigs, when diluted 32-fold, protects mice
attenuated strain of Helpesvirus varicellae.
against the toxin of epidemic typhus rickettsiae.
Production
Biological Assay of Typhus Vaccine
General provisions
Inject subcutaneously 0.5 ml ofthe vaccine under examination
into each of 10 or more healthy guinea-pigs, each weighing The production ofvaccine is based on a virus seed-lot system
between 400 and 500 g. After 7 days inject once again 0.5 ml of and a cell-bank system. The production method shall have
the vaccine into the animals. Fourteen days after the second been shown to yield consistently live varicella vaccines of
injection bleed the animals and pool aliquots of the sera. Test adequate immunogenicity and safety in man. The virus in the
the pools against a toxic substance prepared from injected final vaccine shall not have been passaged in cell cultures
yolle sacs of living fertile eggs as described below. beyond the 38th passage from the original isolated virus.

Incubate for 4 to 10 days after inoculation with epidemic typhus The production method is validated to demonstrate that the
rickettsiae. Harvest the yolk sac and stain a smear by the product, if tested, would comply with the test for safety and
Macchiavello technique. Use only yolle sac of living embryos efficacy.
showing 4+ or more rickettsiae. Determine the wet weight of Substrate for virus propagation
the yolk_sac, grind the yolk sac with a suitable grade of
aluminilllnoxide -anrlsuspend-il1 sterile-mi1lC'(pHadjusted-to The virus is propagated in human diploid .cells (2.7.2).
7.6) or in a mixture ofone part offresh eggyollc and 99 parts of
SEED LOT
sodium chloride injection using, for each g ofthe yolk sac, 10
ml of the vehicle. Centrifuge this suspension for 5 minutes at The strain ofvaricella virus shall be identified as being suitable
low speed, decant the supernatant liquid and distribute in by historical records which shall include information on the

2456

www.webofpharma.com
IP 2010 VARICELLAVACCINE, LIVE

origin ofthe strain and its subsequent manipulation. The virus monitor consistency of production and to determine the
shall at no time have been passaged in continuous cell lines. dilution to be used for the final bulle vaccine.
Seed lots are prepared in the same kind of cells as those used Extraneous agents (2.7.3). Use 50 ml for the test in cell cultures.
for the production of the final vaccine. To avoid the
unnecessary use of monkeys in the test for neurovirulence, Control cells. The control cells ofthe production cell culture
virus seed lots are prepared in large quantities and stored at from which the single harvest is derived comply with a test for
temperatures below -20°, iffreeze-dried, or below -60°, ifnot identity and with the requirements for extraneous agents
freeze-dried. (2.7.3).

Only a virus seed lot that complies with the following FINAL BULK VACCINE
requirements may be used for virus propagation.
Virus harvests that comply with the above tests are pooled
Identification and clarified to remove cells. A suitable stabiliser may be added
and the pooled harvests diluted as appropriate.
The master and working seed lots are identified as varicella Only a final bulle vaccine that complies with the following
virus by serum neutralisation in cell culture, using specific requirements may be used in the preparation of the final lot.
antibodies.
Sterility (2.2.11). Cany out test for sterility using 10 ml ofbulle
Virus concentration. The virus concentration of the master for each sterility medium.
and working seed lots is detelmined as prescribed underAssay
to monitor consistency of production. FINAL LOT

Extraneous agents (2.7.3). Complies with the requirements for The final bulle vaccine is distributed aseptically into sterile,
seed lots for live virus vaccines; a sample of 50 ml is taken for tamper-proofcontainers and freeze-dried to a moisture content
the test in cell cultures. shown to be favourable to the stability of the vaccine. The
containers are then closed so as to prevent contamination
Neurovirulence (2.7.5). Complies with the test for
and the introduction of moisture.
neurovirulence of live virus vaccines.
Only a final lOt that is satisfactory with respect to each of the
PROPAGATION AND HARVEST requirements given below under Identification, Tests and
Assay may be released for use. Provided that the test for
All processing ofthe cell bank and subsequent cell cultures is bovine serum albumin has been carried out with satisfactory
done under aseptic conditions in an area where no other cells results on the final bulle vaccine, it may be omitted on the final
are handled. Approved animal (but not human) serum may be lot.
used in the media. Serum and trypsin used in the preparation
of cell suspensions and media are shown to be free from Identification
extraneous agents. The cell culture medium may contain a pH When the vaccine reconstituted as stated on the label is mixed
indicator such as phenol red and approved antibiotics at the with specific Herpesvinls varicellae antibodies, it is no longer
lowest effective concentration. It is preferable to have a able to infect susceptible cell cuIhIres.
substrate free from antibiotics during production. 5.0 per cent,
but not less than 50.0 ml, of the cell cultures employed for Tests
vaccine production is set aside as uninfected cell cultures
Sterility (2.2.11) Complies with the test for sterility.
(control cells). The infected cells constituting a single harvest
are washed, released from the support surface and pooled. Abnormal toxicity (2.2.1). Complies with the test for abnormal
The cell suspension is disrupted by sonication. toxicity.
Only a virus harvest that complies with the following Bovine serum albumin. Not more than 0.5 J.Lg per human dose,
requirements may be used in the preparation of the final bulk detelmined by a suitable immunochemical method (2.2.14).
vaccine. Water (2.3.43). Not more than 3.0 per cent, detelmined by the
semi-micro determination ofwater. .
Identification
Assay
The virus harvest contains virus that is identified as varicella
Tih'ate for infective virus, using at least ten cell culhu'es for
virus by serum neutralisation in cell culture, using specific
each fourfold dilution or by a technique of equal precision.
antibodies.
Use a suitable virus reference preparation to validate each
Virus concentration. The concentration of infective virus in assay. The virus concentration is not less than the minimum
virus harvests is determined as prescribed under assay to stated on the label.

2457

www.webofpharma.com
VIPER VENOM IP 2010

Labelling. The label states (1) the strain of virus used for the intravenously into each of 10 mice weighing between 18 and
preparation ofthe vaccine; (2) the type and origin ofthe cells 20 g and observe the animals for 24 hours. Not less than 3 and
used for the preparation of the vaccine; (3) that contact with not more than 8 ofthe mice die in 2 to 24 hours. Ifthe number
disinfectants is to be avoided; (4) the minimum virus of deaths is not within this range, change the dilution of the
concentration; (5) that the vaccine is not to be administered venom suitably. .
to pregnant women; (6) the time within which the vaccine Express the result in terms ofnumber ofMouse Units per mg.
must be used after reconstitution.
NOTE - The quantity in mg ofthe venom which will/dll in
2 to 24 hours not less than 3 and not more than 8 mice
represents one Mouse Unit.
Viper Venom Storage. Store in single dose, light-resistant containers.
Daboia Venom Labelling. The label states (1) the number ofMouse Units per
Viper Venom is the dried secretion obtained from the poison container; (2) the volume of waterfor injection to be used for
glands of Viperae russelli and other species of Viperae (Fam. reconstitution.
Viperidae).
Viper Venom contains not less than 50 Mouse Units per mg.
Yellow Fever Vaccine (Live)
Production
Yellow Fever Vaccine (Live) is a freeze-dried preparation of
Immediately afterextraction;thepoisonoussecretion is dried the l7D stiairiofyellow fever virus gfoWif iIi fertilised hert
from the frozen state. The dried venom is pooled, mixed, eggs.
dissolved in ice-cold water for injection and then filtered
through a bacteria-prooffilter to give a stock solution. Further Production
dilutions of the stock solution are made with water for
General provisions
injection under aseptic conditions to give solutions with the
required number of Mouse Units per ml. These solutions are The production ofvaccine is based on a virus seed-lot system.
then distributed in single dose sterile glass containers, dried The production method shall have been shown to yield
from the frozen state and sealed at a pressure not exceeding consistently the yellow fever vaccine (live) of acceptable
2.75kPa. immunogenicity and safety for man.
Description. An almost white or very light yellow, dry powder The production method is validated to demonstrate that the
which when mixed with water yields a clear solution with product, if tested, would comply with the test for safety and
some insoluble residue. efficacy.

Identification Referencepreparation. In the test for neurotropism, a suitable


batch of vaccine known to have satisfactory properties in
A. Produces almost immediate coagulation of blood and man is used as the reference preparation.
citrated human plasma.
B. Mix the soluble fraction from at least 0.6 mg with 1 mlof Substrate for virus propagation
polyvalent antisnake venom serum and incubate the mixture Virus for the preparation ofmaster and working seed lots and
at 370 for 30 minutes. Inject 0.5 ml ofthe mixture intravenously for all vaccine batches is grown in the tissues ofchick embryos
into a group of mice weighing between 18 and 20 g. Observe from a flock free from specified pathogens (2.7.7).
the animals for 24 hours; no animal dies.
SEED LOT
Tests
The 17D strain shall be identified by historical records that
Sterility (2.2.11). Complies with tests for sterility. include information on the origin of the strain and its
Assay. Carry out the biological assay of snake venom subsequent manipulation. Virus seed lots are prepared in large
described below: . quantities and stored at a temperature below _60 0 • Master and
worklrtg seed Ioisshitu- nofcoiifain-any-humanprotein or
Biologicalassay of snake venom added serum.
Dissolve a quantity of the freeze-dried venom equivalent to Unless otherwise justified and authorised, the virus in the
50 Mouse Units in 25 ml of saline solution. Inject 0.5 ml [mal vaccine shall be between passage levels 204 and 239

2458

www.webofpharma.com
IP 2010 YELLOW FEVER VACCINE (LIVE)

from the original isolate ofstrain l7D. A working seed lot shall content is carried out. A suitable stabiliser may be added and
be only one passage from a master seed lot. A working seed the pooled harvests diluted as appropriate.
lot shall be used without intervening passage as the inoculum Only a final bulle vaccine that complies with the following
for infecting the tissues used in the production of a vaccine tests may be used in the preparation of the final lot.
lot, so that no vaccine vims is more than one passage from a
seed lot that has passed all the safety tests. Sterility (2.2.11). Carry out test for sterility using 10 ml ofbulk
for each sterility medium.
Only a vims seed lot that complies with the following
requirements may be used for virus propagation. Protein nitrogen content (2.3.30). The protein nitrogen
content, before the addition of any stabiliser, is not more than
Identification 0.25 mg per human dose.

The master and working seed lots are identified as containing FINAL LOT
yellow fever virus by semm neutralisation in cell culture, using
The final bulle vaccine is distributed aseptically into sterile,
specific antibodies.
tamper-proofcontainers and freeze-dried to a moishrre content
Extraneous agents (2.7.3). Each worldng seed lot complies shown to be favourable to the stability of the vaccine. The
with the test for extraneous agents. containers are then closed so as to prevent contamination
and the introduction of moishrre.
PROPAGATION AND HARVEST
Only a final lot that is satisfactory with respect to thermal
All processing of the fertilised eggs is done under aseptic stability and each ofthe tests given under Identification, Tests
conditions in an area where no other infectious agents or cells and Assay may be released for use. Provided that the test for
are handled at the same time. Two per cent but not less than ovalbumin has been performed with satisfactory results on
twenty and not more than fifty eggs are set aside as uninfected
the final bulk vaccine, it may be omitted on the final lot.
control eggs. After inoculation and incubation at a controlled
temperature, only living and typical chick embryos are Thermal stability. Maintain samples ofthe final lot offreeze-
harvested. The age of the embryo at the time of vims harvest dried vaccine in the dry state at 37° for 14 days. Determine the
is reckoned from the initial introduction of the egg into the vims concentration as described under Assay in parallel for
incubator and shall not be more than 12 days. After the heated vaccine and for unheated vaccine. The difference
homogenisation and clarification by centrifugation, the extract in the virus concentration between unheated and heated
of embryonic pulp is tested as described below and kept at - vaccine does not exceed 1.0 loglo' and the vims concentration
70° or colder until further processing. Vims harvests that ofthe heated vaccine is not less than the number of TCID so or
comply with the prescribed tests may be pooled. No human plaque-forming units (PFU) equivalent to 1 x 103 mouse LDso
protein is added to the vims suspension at any stage during per human dose.
production. Ifstabilisers are added, they shall have been shown
Identification
to have no antigenic or sensitising properties for man.
Only a single harvest that complies with the following tests When the vaccine reconstituted as stated on the label is mixed
may be used in the preparation of the final bulle vaccine. with specific yellow fever virus antibodies, there is a significant
reduction in its ability to infect susceptible cell culhlres.
Identification
Tests
The single harvest contains vims that is identified as yellow
fever virus by semm neutralisation in cell culture, using Sterility (2.2.11). Complies with the test for sterility.
specific antibodies. .Ovalbumin. Not more than 5 Ilgofovalbumin per hrrman dose,
Extraneous agents (2.7.3). Complies with the tests for determined by a suitable imrnunochemical method (2.2.14).
,
extraneous agents. Abnormal toxicity (2.2.1). Complies with the test for abnormal
Control eggs. Complies with the tests for extraneous agents toxicity.
(2.7.3). Bacterial endotoxins (2.2:3). Not more than 5 ill ofbacterial
Virus concentration. In order to calculate the dilution for endotoxin per human dose.
formulation ofthe final bulle, each single harvest is titrated as Water (2:3.43). Not more than 3.0 per cent, detelmined by the
described under Assay. semi-micro determination ofwater.
FINAL BULK VACCINE Assay
Single harvests that comply with the tests prescribed above Titrate for infective vims in cell cultures. Use an appropriate
are pooled and clarified again. A test for protein nitrogen vims reference preparation to validate each assay.

2459

www.webofpharma.com
YELLOW FEVER VACCINE (LIVE) IP 2010

The virus concentration is not less than the equivalent in inoculation and prepare serum from each sample. Prepare 1: 10,
TCID or PFU of 1 x 103 mouse LD 50 per human dose. The 1:100 and 1: 1000 dilutions from each serum and inoculate each
m
relationship between mouse LD 50 and TCID 50 or PFU'IS dilution into a group ofat least six cell culture vessels used for
established by each laboratory and approved by the competent the detennination of the virus concentration. The seed lot
authority. complies with the test if none of the sera contains more than
the equivalent of 500 mouse LD50 in 0.03 ml and at most one
The method shown below, or another suitable technique, may
serum contains more than the equivalent of 100 mouse LD 50 in
be used to detennine the mouse LD 50 •
O.03ml.
Mouse LD . The statistically calculated quantity of virus
suspensio~ that is expected to produce fatal specific Immunogenicity. Take blood from each monkey 30 days after
encephalitis in 50 per cent of mice of a highly susceptible the injection of the test dose and prepare serum from each
strain, 4 to 6 weeks of age, after intracerebral inoculation. sample. The seed lot complies with the test if at least 90.0 per
cent of the test monkeys are shown to be immune, as
Appropriate serial dilutions of the reconstituted vaccine are detennined by examining their sera in the test for neutralisation
made in diluent for yellow fever virus (0.75 per cent solution of yellow fever virus described below.
of bovine albumin in phosphate-buffered saline pH 7.4, or
any other diluent that has been shown to be equivalent for It has been shown that a low dilution of serum (for example,
maintaining the infectivity ofthevirus). 1: 10) may contain non-specific inhibitors that influence this
test; such serum shall be treated to remove inhibitors. Mix
Mice of a highly susceptible strain, 4 to 6 weeks of age, are dilutions of at least 1: 10, 1:40 and 1: 160 ofserum from each
injected intracerebrally under anaesthesia with 0.03 ml of the monkey with an equal volume of l7D vaccine virus at a dilution
vaccine dilution. Groups of not less than eight mice are used that will yield an optimum number ofplaques with the titration
for each dilution; the series of dilutions is chosen so as to method used. Incubate the serum-virus mixtures in a water-
cover the range 0 to 100.0 per cent mortality of the mice. bath at 37° for 1 h and then cool in iced water; add 0.2 ml of
Injection of the mice is performed immediately after the each serum-virus mixture to each of four cell-culture plates
dilutions have been made. The mice are observed for 21 days and proceed as for the detennination of virus concentration.
and all deaths are recorded. Only survivors and deaths caused Inoculate similarly ten plates with the same amount of virus
by typical yellow fever infections are counted in the plus an equal volume ofa 1: 10 dilution ofmonkey serum Imown
computations. Mice paralysed on the twenty-first day of to contain no neutralising antibodies to yellow fever virus. At
observation are counted as survivors. the end of the observation period, compare the mean number
Tests in monkeys for Yellow Fever Vaccine ofplaques in the plates receiving virus plus non-immune serum
with the mean number ofplaques in the plates receiving virus
Each master and working seed lot complies with the following plus dilutions of each monkey serum. Not more than 10 per
tests in monkeys for viraemia (viscerotropism), cent of the test monkeys have serum that fails to reduce the
immunogenicity and neurotropism. number ofplaques by 50.0 per cent at the 1: 10 dilution.
The monkeys shall be Macaca spp. susceptible to yellow Neurotropism. Neurotropism is assessed from clinical
fever virus and shall have been shown to be non-immune to evidence of encephalitis, from incidence of clinical
yellow fever at the time ofinjecting the seed virus. They shall manifestations and by evaluation of histological lesions, in
be healthy and shall not have received previously intracerebral comparison with ten monkeys injected with the reference
or intraspinal inoculation. Furthennore, they shall not have preparation. The seed lot is not acceptable if either the onset
been inoculated by other routes with neurotropic viruses or and duration of the febrile reaction or the clinical signs of
with antigens related to yellow fever virus. Not fewer than ten encephalitis and pathological findings are such as to indicate
monkeys are used for each test. a change in the propeIties of the -virus.
Use a test dose of 0.25 ml containing the equivalent of not Clinical evaluation. The monkeys are examined daily for 30
less than 5000 mouse LD 50 and not more than 50,000 mouse days by personnel familiar with clinical signs of encephalitis
LD 50' determined by a titration for infectious virus and using in primates (ifnecessary, the monkeys are removed from their
the established equivalence between virus concentration and cage and examined for signs ofmotor wealmess or spasticity).
mouse LD 50 (see under Assay). Inject the test dose into one The seed lot is not acceptable ifin the monkeys injected with
lobe of each under anaesthesia and observe l11(;J(1lenc:e of severe signs of-encephalitis,such.-as
the monkeys for not less than 30 days. paralysis or inability to stand when stimulated, or mortality is
Viraemia (Viscerotropism). Viscerotropism is indicated by the greater than for the reference vaccine. These and other signs
amount ofvirus present in serum. Take blood from each ofthe of encephalitis, such as paresis, in-coordination, lethargy,
test monkeys on the second, fourth and sixth days after tremors or spasticity are assigned numerical values for the

2460

www.webofpharma.com
IP 2010 YELLOW FEVER VACCINE (LIVE)

severity of symptoms by a grading method. Each day each anatomical fonnations of the central nervous system with
monkey in the test is given a score based on the scale: varying frequency and severity (I. S. Levenbook et al., Journal
o/Biological Standardization, 1987,15,305-313). Based on
Grade 1- rough coat, not eating,
these two indicators, the anatomical structures can be divided
Grade 2- high-pitched voice, inactive, slow moving, into target, spared and discriminator areas. Target areas are
Grade 3- shaky, tremors, unco-ordinated, limb weakness, those which show more severe specific lesions in a majority
of monkeys ilTespective of the degree of neurovirulence of
Grade 4- inability to stand, limb paralysis or death (a dead
the seed lot. Spared areas are those which show only minimal
monkey receives a daily score of4 from the day
specific lesions and in a minority of monkeys. Discriminator
of death until day 30).
areas are those where there is a significant increase in the
A clinical score for a particular monkey is the average of its frequency ofmore severe specific lesions with seed lots having
daily scores; the clinical score for the seed lot is the mean of a higher degree of neurovirulence. Discriminator and target
the individual monkey scores. The seed lot is not acceptable areas for Macaca cynomolgus and Macaca rhesus monkeys
if the mean of the clinical severity scores for the group of are shown in the table below:
monkeys inoculated with it is significantly greater (P = 0.95)
than the mean for the group of monkeys injected with the Table 1 - The discriminator and target areas for monkey.
reference preparation. In addition, special consideration is
Type ofmonkey Discriminator areas Target areas
given to any animal showing unusually severe signs when
deciding on the acceptability of the seed lot. Macaca cynomolgus globus pallidus substantia nigra
putamen anterior/
Histological evaluation. Five levels ofthe brain are examined
median thalamic
including:
nucleus lateral
Block 1 - the corpus striatum at the level of the optic thalamic nucleus
chiasma,
Macaca rhesus caudate nucleus substantia nigra
Block II - the thalamus at the level ofthe mamillary bodies, globus pallidus cervical
Block III - the mesencephalon at the level of the superior putamen anterior/ lumbm'
colliculi, median thalamic enlargement
BlockIV - the pons and cerebellum at the level of the nucleus lateral
superior olives, thalamic nucleus
cervical enlargement
Block V - the medulla oblongata and cerebellum at the
lumbar enlargement
level ofthe mid-inferior olivary nuclei.
enlargement
Cervical and lumbar enlargements of the spinal cord are each
divided equally into six blocks; 15 /lm sections are cut from Scores for discriminator and target areas are used for the final
the tissue blocks embedded in paraffin wax and stained with evaluation of the seed lot. The individual monkey score is
gallocyanin. Numerical scores are given to each hemisection calculated from the sum of individual target area scores in
of the cord and to structures in each hemisection of the brain each hemisection divided by the number ofareas examined. A
as listed below. Lesions are scored as follows: separate score is calculated similarly for the discriminator areas.
Grade 1. Minimal: 1 to 3 small focal inflammatory infiltrates. Mean scores for the test group are calculated in two ways: (1)
Degeneration or loss of a few neurons. by dividing the sum of the individual monkey discriminator
Grade 2. Moderate: 4 or more focal inflammatory infiltrates. scores by the number ofmonkeys and (2) by dividing the sum
Degeneration or loss of neurons affecting not more than one of the individual monkey target and discriminator scores by
third of cells. the number of monkeys. These two mean scores are taken
into account when deciding on the acceptability of the seed
Grade 3. Severe: moderate focal or diffuse inflammatory
lot. The seed lot is not acceptable if either ofthe mean lesion
infiltration. Degeneration or loss of up to two third of the
scores is significantly greater (P = 0.95) than for the reference
neurons.
preparation.
Grade 4. Overwhelming: vmiable but often severe inflammatory
Labelling. The label states (1) the strain of virus used in
reaction. Degeneration or loss of more than 90.0 per cent of
preparation; (2) that the vaccine has been prepared in chick
neurons. embryos; (3) the minimum virus concentration; (4) that contact
It has been found that inoculation of yellow fever vaccine with disinfectants is to be avoided; (5) the period of time
into the monkey brain causes histological lesions in different within which the vaccine is to be used after reconstitution.

2461

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

HERBS AND HERBAL PRODUCTS

General Requirements .... 2467


Monographs
Acacia 2469
Ajwain 2470
Amalaki 2471
Amla Juice Powder 2472
Amra 2473
Anantmula 2474
Arachis Oil 2475
AIjuna 2476
Aljuna Dry Extract 2477
Artemisia 2478
Ashwagandha 2479
Ashwagandha Dry Extract 2480
BelladonnaLeaf 2481
Belladonna Dry Extract 2483
Belladonna Tincture 2483
Bhibhitaki 2484
BhibhitakiAqueous Extract 2485
Bhringraj 2486
Bhuiarnla 2488
Brahmi· 2489
Brahmi Extract 2490
Castor Oil 2490
Clove Oil 2491
Coconut Oil 2492
Coleus 2492
Coleus Dry Extract 2493
Coriander Oil 2494
Daruharidra Roots 2495

2463

www.webofpharma.com
MONOGRAPHS INDIAN PHARMACOPOEIA 2010

Daruharidra Stems 2496


Eucalyptus Oil 2498
Garcinia 2499
GarciniaAqueous Extract 2500
Gokhru 2500
GuarGum ·2501
Gudmar 2502
Guduchi 2503
GuggulResin 2504
Gugulipid 2505
Gugulipid Tablets 2506
Haridra 2507
Haridra Dry Extract 2508
Haritaki 2508
Haritaki Extract· 2510
Haritaki Aqueous Extract 2510
Hydrogenated Castor Oil 2511
Ipecac Tincture 2512
Ispaghula Husk 2513
Kalmegh 2513
Kalmegh Dry Extract 2514
Kunduru 2515
Kutki 2516
Lasuna 2517
Lavang 2518
Malt Extract 2519
Mandukaparni 2520
Manjistha 2521
Maricha 2522
Mentha Oil 2523

Neem 2524
Opium 2525

2464

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

Opium Powder 2527


Papain 2528
Peppennint Oil 2529
Pippali Large 2530
Pippali Small 2531
Punarnava 2532
Sarpagandha 2533
Sarpagandha Powder 2535
Sarpagandha Tablets 2535
Saunf 2536
Senna Leaf 2537
Senna Pods 2538
Senna DIY Extract 2539
Senna Tablets 2540
Shatavari 2540
Shati 2542
Shellac 2543
Starch 2543
Sunthi 2544
Sunthi Extract 2545
ToluBalsarn 2546
Tragacanth 2547
Tulasi 2548
Tulasi DIY Extract 2549
Vasaka 2550
Vasaka Extract 2550
Yasti 2551
Yasti DIY Extract 2552

2465

www.webofpharma.com
www.webofpharma.com
IP 2010 HERBS AND HERBAL PRODUCTS

Herbs and Herbal Products which may figure in a regulated list under appropriate forest
and other laws, may still be taken up for a monograph for
Herbs and products containing herb(s) have been in trade inclusion in pharmacopoeia, if there is knowledge of efforts
and commerce and are currently used for a variety ofpurposes. to cultivate or take care ofsustainability issues and lor specific
As a country, India has a rich history ofuse ofherbs, processed permission is available under law for use ofthe herb. As already
herbs and formulations containing herbs both from traditional specified under "General Notices" in the pharmacopoeia,
wisdom as well as cultural usage. Herbs and herbal products appearance of a monograph does not mean its approval as a
are also regulated by various laws. For the purposes of drug under the law. Monograph ofa herb in the pharmacopoeia
pharmacopoeial standards various considerations have been is to provide qualitative and quantitative standards of quality
given. This monograph provides a general outline and policies for the herb for its use either as a food item or food ingredient
towards the same. or food supplement! nutraceuticals, as a drug, and/or as an
ingredient in cosmetics. Each such use would need to comply
Crude Herbs with applicable regulations. Each herb is regarded as one active
substance, irrespective of the lmowledge about the active
This term means, unless specified otherwise, mainly whole,
constituents of the herb is available of not.
fragmented or cut, plants, parts of plants, algae, fungi, and
lichen in a form which is not processed. Herbs are usually in * Ayurvedic Pharmacopoeia of India, Vol. 1-6, Ministry of Health and
dried form, but sometimes, when specified, may also be in a Family Welfare, Govt. of India.
fresh form. In specific cases exudates which have not been Herbs may be exposed to low dose gamma radiation for
processed further also are covered under the term herbs. purposes of reducing their microbial contamination. Herbs
Processing, does not include, normally expected value treated with low dose gamma radiation shall meet national
addition steps like grading, sizing, removal of weeds or parts laws related to such treatment and shall be .labeled as per law.
of plants other than those specified herb and removal of
adulterants. The term herbs, though botanically generally refer
Processed Herbs
to plants of specified height and nature, for the purposes of
pharmacopoeial reference, shall mean and include plants and Processed herbs means preparations obtained by subjecting
parts of plants not necessarily from herbs and shrubs, but herbs to treatment such as extraction, distillation, expression,
cover the entire range namely creepers, climbers, trees etc. fractionation, purification, concentration and partial or full
Each monograph ofa herb in the pharmacopoeia shali specify fermentation. Processing may also be done by way of
the botanical scientific name according the binomial system powdering herbs, preparing tincture, preparing extract,
specifying the genus, species, variety and author. In cases distilling to get essential oils, fatty oils (either expressed or
where there are controversial botanical identity, as is seen solvent extracted or a blend of both) expressedjuices, extracted
with mainly herbs known in the Indian traditional system, the exudates, gums and oleo resins, liquid extract where the
monograph shall specify the official name of the herb along solvent is evaporated to yield concentrated semi solid mass
with its botanical scientific name and guidance is taken from or dried mass. Extraction may be performed by means of
Ayurvedic Pharmacopoiea of India' to decide the same. In appropriate technology such as infusion, maceration,
cases where, the same herb is available in different grades or soxbleting, boiling under ambient or higher pressure, with or
sizes, if found appropriate and necessary, separate without specified enzymes, with or without agitation and
monographs may be introduced in the pharmacopoeia to cover combination thereof. Drying ofliquid extracts for removal of
each ofthem with appropriate standards. For example- Pippali the solvent may be done by using various appropriate
(large) and Pippali (small). technologies like air drying, sun drying, drying under vacuum
While deciding to introduce a monograph for a herb in the or with forced air circulation, drying at low temperature with
air circulation, by way of lyophilization or freeze drying.
pharmacopoeia, the criteria that would be kept in mind, but
Extracts of herbs may also be prepared by using carbon di-
not limited to are - herbs with specific name and a definitive
botanical identity up to species, availability and usage in trade oxide as a SOlvent-super critical fluid extraction.
and commerce, regulatory interest, knowledge of and Extracts may be liquid extracts and tinctures, semi solid (soft
availability of a specific chemical compound of well extracts) or solid dry extracts of known consistency obtained
characterised structure [either responsible for the biological from herbs. Standardized extract, a term commonly employed,
activity of the herb (bio-marker) or a chemical compound would for pharmacopoeial purposes, mean an extract adjusted
known to be present in the herb even if not responsible for with in an acceptable tolerance to a given content of bio-
biological activity(chemical/analytical marker)], availability of marker or chemical/ analytical marker. Standardization may be
a quantitative method for estimation of such a compound, achieved by adjusting the extracts with approved inert material
lmowledge of safety of the herb, and its sustainability. Herbs or by blending one or more batches of extracts. Wherever

2467

www.webofpharma.com
HERBS AND HERBAL PRODUCTS IP 2010

possible, extracts shall specify the defined range of the Approved preservative~ or preservatives system may be used
constituents (bio-marker or chemical! analytical marker). during preparation of extracts. The names of such
Extracts not covered in the above description would be defined preservatives used which would remain in the final extract
by the process ofproduction ofthe herb to the extract, solvent shall be listed on the label of such extract, and the proportion
used and technology applied. The difference between extracts ofpreservatives used shall not exceed normally accepted safe
and tinctures would be, in the type of solvent used for limits of their usage as per relevant laws or pharmacopoeial
extracting a herb, and tincture would normally mean an extract standards. No artificial colours may be used in extracts of
where aqueous-ethanol is used as a solvent for extraction. herbs unless and otherwise specified in the specific
Dry extracts usually have a loss on drying or water content monograph.Only approved colours shall be used.
not greater than 5 per cent w/w, unless specified otherwise in Extracts may be exposed to ethylene oxide fumigation or low
any monograph. It is normal to extrapolate safety aspects and dose gamma radiation for purposes ofreducing their microbial
history ofuse information for extracts as long as the process, contamination. In cases where they are fumigated, the final
solvents, extraction ratios are comparable to the processes extracts exposed shall meet residual levels of ethylene oxide
used in documented traditional Imowledge. Additionally in limits as applicable. Herbs treated with low dose gamma
cases of standardized extracts the inert excipients(s), if any radiation shall meet national laws related to such treatment
used for standardization or adjustment of the content of and shall be labelled as per law.
constituents should also be declared on the label of such Appearance ofa monograph ofan extract in the pharmacopoeia
extracts. Extracts shall be free from solvent used for extraction does not mean its approval as a drug under the law. Monograph
and shall comply with the respective limts as given in of an extract in the pharmacopoeia is to provide qualitative
Appendix 5.4. Harmful and carcinogenic solvents shall not be and quantitative standards ofquality for the extract for its use
used fof extraction purposes. Solvents and solvent systems either as a food item or food ingredient or food supplement!
may include use of propylene glycol, glycerin, sorbitol and nutraceuticals, as a drug and / or as an ingredient in cosmetics.
such other polyhydroxy alcohols, as long as the content of Each such use would need to comply with applicable
such polyhydroxy alcohol are within safe limit in the final regulations. Each extract is regarded as one active substance
product. irrespective of the knowledge about the active constituents
In cases where extraction and fractionation process leads to ofthe herb is available or not.
preparation of an extract, which consists of a single chemical Herbal Formulations
compound of more than at least 70 per cent purity, such
extracts shall be treated as an active pharmaceutical ingredient Herbal formulation shall mean a dosage form consisting of
or a food additive or a cosmetic ingredient and would be one or more herbs or processed herb(s) in specified quantities
required to meet relevant laws. to provide specific nutritional, cosmetic benefits, and/or other
benefits meant for use to diagnose treat, mitigate diseases of
Extracts may also be offered as purified or enriched extracts. human beings or animals and/or to alter the structure or
Such extract of!l herb is processed in such a way to provide physiology of human beings or animals. Dosage forms
higher than normal proportion ofthe active constituent (s) of commonly employed for food or cosmetic or pharmaceuticals
the herb as long as the active constituent (s) is/are Imown. may be employed to formulate one or more herb or processed
Such purified or enriched extracts may contain additiona~ herbs. Dosage forms Imown in traditional medicines may also
valuable components which may provide specific properties be adopted for preparing herbal formulations, either for external
like enhanced efficacy or stability or solubility and availability use or for internal administration. Adequate consideration for
of the active constituent (s). Purified and enriched extracts uniform distribution of herb or processed herbs as well as
may also be prepared to reduce or remove other specific stability of the same in the dosage form shall be provided
compound or group of compounds that is scientifically during formulation development.
considered undesirable in the herb extracts. Pharmacological, Herbal formulation shall be labelled to comply with relevant
toxicological, pharmaceutical considerations need to be labelling requirements under food or drug or cosmetics laws
applied while preparing such purified or enriched extracts. as applicable. Additionally, adequate information shall be
Mixed extracts may also be offered which would cover provided on label ofsuch formulations to include the name of
combination of more than one herb extract for purposes of the herb, parts used, nature and type of extract or processed
providing simplification or economical way to manufacture herb used, extraction ratios, quantity per unit dose or ,per
~nerbal-formulationS:-------~------~~--'--'---~~
-serving:name (s) ofinert-excq;ients used and any preserVatives
Herbs may also be extracted using vegetable oils (approved added shall be provided on the label. .
by Food Law) for extraction purposes and such extracts shall Appearance of a monograph of a herbal formulation in the
specify the oil used for processing. pharmacopoeia does not mean its approval as a drug under

2468

www.webofpharma.com
IP 2010 ACACIA

the law. Monograph of a herbal formulation in the C. Al 0 per cent w/v solution is either dextro-rotatory or slightly
pharmacopoeia is to provide qualitative and quantitative laevo-rotatory.
standards of quality for the formulation for its use either as a
food item or food ingredient or food supplement/ Tests
nutraceuticals, as a drug and / or as a cosmetic. Each such use
Sterculia gum and agar. To 50 mg ofthe powdered substance
would need to comply with applicable regulations.
under examination add 0.2 ml of freshly prepared ruthenium
red solution and examine microscopically; the particles do
not acquire a red colour after irrigation with water.
Agar and tragacanth. To 10 ml of a 10 per cent w/v solution
Acacia add 0.2 ml of lead acetate solution; no precipitate is produced.
Gum Acacia; Indian Gum Starch and dextrin. Boil 10 ml of a 10 per cent w/v solution
and cool, add 0.1 ml of 0.05 M iodine; no blue or brown colour
is produced.
Tannins. To 10 ml of a 10 per cent w/v solution add 0.1 mlof
ferric chloride test solution; a gelatinous precipitate is formed,
but neither the precipitate nor the liquid shows. a dark blue
colour.
Sucrose and fructose. To 1 ml of a 10 per cent w/v solution
add 4 ml of water, 0.1 g of resorcinol and 2 ml of hydrochloric
acid and heat on a water-bath; no yellow or piDk colour
develops.
Water-insoluble matter. Dissolve 5 g, in fine powder, in 100
ml of water in a 250-ml flask, add 10 ml of dilute hydrochloric
acid and boil gently for 15 minutes. Filter by suction while hot
through a sintered-glass crucible, previously tared, wash
Acacia is the air-hardened, gummy exudation from the stem thoroughly with hot water, dry at 105° and weigh; the residue
and branches of Acacia nilotica (Linn.) Del. subsp. indica does not exceed 50 mg.
(Benth.) Brenan (syn. A. arabica Willd. var. indica Benth.) Microbial contamination (2.2.9). 1.0 g is free from Escherichia
(Fam. Leguminosae), or other species ofAcacia. It is available coli.
as pieces (tears) or in the form of a powder.
Sulphated ash (2.3.1 8). Not more than 5.0 per cent.
Description
Acid-insoluble ash (2.3.19). Not more than 1.0 per cent,
Tears - Irregular and broken pieces ofvarying size, yellowish- determined on 1.0 g by Method C.
white, yellow or amber in colour, with numerous minute Loss on drying (2.4.1 9). Not more than 15.0 per cent, determined
fissures; brittle fractured surface, glassy and occasionally on 1.0 g by drying in an oven at 105°.
iridescent; odourless.
. Storage. Store protected from heat, moisture and against
Powder - A white or yellowish-white powder; odourless; on attack by insects and rodents.
treatment with water it dissolves to give a mucilaginous liquid
which is colourless or yellowish, dense, viscous, adhesive
and translucent.

Identification
A. An aqueous solution is gelatinised by the addition of lead
subacetate solution.
B. To 5 ml of a 10 per cent w/v solution add gradually, while
shaking, 10 ml of ethanol (95 per cent). The cloudy liquid, on
addition of 0.5 ml of acetic acid, gives a white precipitate.
Filter and add to the clear filtrate 50 ml of ammonium oxalate
solution; the filtrate becomes cloudy.

2469

www.webofpharma.com
AJWAlN IP 2010

Ajwain Test solution. To 1 g of the coarsely powdered substance


under examination, add 25 ml ofdichloromethane, reflux for
Bishop's weed 15 minutes, cool and filter. Reflux the residue further for two
times with 25 m1 ofdichloromethane, cool and filter. Combine
all the filtrates and concentrate under vacuum to 25 m!.
9 Reference solution. To 1 g of the qjwain RS, add 25 ml of
8 dichloromethane, reflux for 15 minutes, cool and filter. Reflux
the residue further for two times with 25 ml of
7
dichloromethane, cool and filter. Combine all the filtrates and
6 concentrate under vacuum to 25 m!.
5
Apply to the plate 10 III ofeach solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with dnisaldehyde sulphuric acid reagent. Heat at 11 0° for 10
2
minutes arid examine the plate in daylight. The
1 chromatographic profile of the test solution is similar to that
o of the 'reference solution.
~ '. .

Tests
Ajwain consists of the dried fruits of Trachyspermum ammi
Mill. (Fam.Apiaceae). Foreign organic matter{2.6.1). Not more than 2.0 per cent.

Ajwain contains not less than 1.0 per cent w/w of thymol, Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent.
calculated on the dried basis. Water-soluble extractive(2:6.3); Not less than 15.0 per cent
by method!. ' ' ,
Description. The fruits are oval-oblong, greenish brown to
yellowish brown in colour. They have an aromatic Total Ash (2.3.19). Not more than 15.0 percent.
characteristic odour and the taste is sweet aromatic.
Acid-insoluble ash (2.3 .19). Not more than 7.0 per cent.

Identification Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
A. Macroscopic - The fruit consists of two portions each
Loss on drying (2.4.19). Not more than 10 per cent, detennined
called mericarp and connected by central stalk (carpophore).
on 5 g by drying in an oven at 105°.
A single seed is seen in each mericarp. Fruit surface is glabrous
and the five primary ridges of each mericarp are prominent, Microbial contamination (2.2.9). Complies with the microbial
straight and pale straw coloured. contamination tests.
B. Microscopic - Epicarp is composed ofpolygonal cells. In Assay. Detennine by gas chromatography (2.4.13).
the mesocarpic region, reticulate and lignified parenchymas Test solution. Weigh 2.0 g ofthe coarsely powdered substance
are seen at vascular strands. Tracheids show helical under examination, add 50 ml of methanol, and reflux on a
thickening. Endocarp consists of narrow elongated cells water-bath for 15 minutes, cool and filter. Reflux the residue
having a parquetry arrangement. Polyhedral, thick walled further with methanol till the extract turns colourless, cool
endospenn contains aleurone grains and oil globules. Vittae and filter. Combine all the filtrates and concentrate to a volume
are six in number, four on the dorsal surface at the mesocarpic slightly less than 100 m!. Dilute with methanol to 100.0 m!.
region below the secondary ridges and two on the commissural
surface ofthe mericarp. Vittae long, slender composed ofthin Reference solution. A 0.01 per cent w/v solution of thymol RS
walled polygonal cells and is lined by an epithelium of small in methanol.
polygonal tubular cells; 10-15 separate, septum transverse or Chromatographic system
curved. - a capillary column 30 m x 0.25 m x 0.25 mm, packed with
_..... methylpolysiloxane,._ ..... _ ....__ .. - ._- ....
C. Detennine by thin-'layer chromatography (2.4.17), coating
the plate with silica gel GF 254.. temperature:
oven 90° to 260° @1 0° per minute (initially and finally
Mobile phase. A mixture of 93 volumes of toluene and 7 hold for 5 minutes respectively),
volumes of ethyl acetate. injector 240°,

2470

www.webofpharma.com
IP 2010 AMALAKI

detector 280°, C. Determine by thin-layer chromatography (2.4.17), coating


- flow rate. 0.8 ml per minute, the plate with silica gel GF254.
- split flow 20 ml per minute.
Mobile phase. A mixture of20 volumes of toluene, 45 volumes
Inject the reference solution and the test solution. of ethyl acetate, 20 volumes of glacial acetic acid and 5
volumes offormic acid.
Calculate the content of thymol.
Test solution. Reflux 2 g of the coarsely powdered substance
Storage. Store protected from light in well-filled containers, at
under examination with 50-75 ml of methanol for 15 minutes,
a temperature not exceeding 30°.
cool and filter. Reflux the residue further for two times with
75 mlofmethanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 50 ml.
Amalaki Reference solution. Reflux 0.4 g of the coarsely powdered
Emblic Myrobalan; Indian Gooseberry amalaki RS with 50-75 ml of methanol for 15 minutes, cool
and filter. Reflux the residue further for two times with 75 ml of
methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 10 ml.
Apply to the plate 10 /ll ofeach solutioh as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with anisaldehyde sulphuric acid reagent. Heat the place at
100° for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 3 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 30 per cent.
Water-soluble extractive (2.6.3). Not less than 40 per cent by
Method I.
Amalaki consists of 'the dried fruit pericarp of Emblica
offiCinalis Gaertn. (Phyllanthus emblica Linn.) (Fam. Total Ash (2.3.19). Not more than 5.0 per cent.
Euphorbiaceae). Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Amalaki contains not less than 1.0 per cent w/w gallic acid
Heavy metals (2.3.13). 1.0 g complies with the limit test for
calculated on the dried basis.
heavy metals, Method B (20 ppm).
Description. The dried fruit has a highly shriveled and wrinlded
Loss on drying (2.4.19). Not more than 12.0 per cent,
external surface. The taste is sour and astringent followed by
determined on 5 g by drying in an oven at 105°.
delicately sweet tinge.
Microbial contamination (2.2.9). Complies with the microbial
Identification contamination tests.
A. Macroscopic - The dried fruit shows a broad, highly Assay. Determine by liquid chromatography (2.4.14).
shriveled and wrinkled external convex surface, lateral surface
transversely wrinlded, external surface exhibits few whitish
Test solution. Weigh accurately about 0.5 g of coarsely
powdered substance under examination, add 50 ml of water,
specks, occasionally some pieces show a portion of stony
sonicate for 3 minutes and heat on a boiling water-bath for
testa.
15 minutes, cool and dilute to 100.0 ml with water and filter.
E. Microscopic - The epicarpic cells are rectangular in shape
and their walls are highly cuticularized. Anomocytic type of
Reference solution. A 0.01 per cent w/v solution of gallic
stomata is found rarely. Collateral fibrovascular bundles are
acid RS in water.
scattered throughout the inner mesocarp. Pitted and helical Chromatographic system
tracheids with tapering ends are seen. At places in the phloem, - a stainless steel column 25 cm x 4.6 mm packed with
large cavities filled with crystal mass are present. octadecylsilane bonded to porous silica (5 /lm),

2471

www.webofpharma.com
AMLA JUICE POWDER IP 2010

mobile phase: A. a solution prepared by dissolving 0.136 and examine in ultraviolet light at 254 urn. Spray the plate with
g of potassium di-hydrogen orthophosphate in 500 ml anisaldehyde-sulphuric acid reagent. Heat the plate at 110°
of water, add 0.5 ml of orthophosphoric acid and dilute for 10 minutes and examine the plate under 365 nm and in day
to 1000 ml with water, light. The chromatographic profile ofthe test solution is similar
B. acetonitrile to that of the reference solution.
a linear gradient programme using the conditions given
below, Tests
~
flow rate. 1.5 ml per minute,
Water - soluble extractive (2.6.3). Not less than 85.0 per cent
- spectrophotometer set at 270 nm,
by method I.
- injection volume. 20 Ill.
Total ash (2.3.19). Not more than 15.0 percent.
Time Mobile phase A Mobile phase B
(in min) (per cent v/v) (per cent v/v) Water (2.3.43). Not more than 5.0 per cent.
o 100 o Heavy metals (2.3.13). 1.0 g complies with the limit test for
18 55 45 heavy metals, Method B (20 ppm).
25 20 80 Microbial contamination (2.2.9). Complies with the microbial
30 . 100 o contamination tests:

Inject the reference solution. The test is not valid unless the Gallic acid. Not less than 0.1 per cent and not more than 2.0
relative standard deviation for replicate injections is not more per cent.
than2;0 per cent. Determine by liquid chromatography (2.4.14).
Inject the test solution and the reference solution. Test solution. Dissolve about 50 mg of the extract under
Calculate the content of gallic acid. examination or quantity equivalent to 10 mg of gallic acid in
Storage. Store protected from light, heat, moisture and against water, by frequent shaking and dilute to 50.0 ml with water
attack by insects and rodents. and filter.
Reference solution. A 0.01 per cent w/v solution of gallic
acid RS in water.

Amla Juice Powder Chromatographic system


- a stainless steel column 25 cm x 4.6 mm packed with
Amla Juice Powder is obtained by spray drying cold pressed octadecylsilane bonded porous silica (5 11m),
juice of fresh fruits of Phyllanthus emblica Linn. (Emblica mobile phase: A. 0.1 per centformic acid in water,
officinalis Gaertn, Fam. Euphorbiaceae) B. methanol,
Amla Juice Powder contains not less than 90.0 per cent w/w a linear gradient programme using the conditions given
and not more than 110 per cent w/w of the stated amount of below,
polyphenols, calculated on the anhydrous basis. It may contain flow rate. 0.7 ml per minute,
suitable added substances. spectrophotometer set at 272 urn,
injection volume. 20 Ill.
Description. A beige to off-white powder.
Time Mobile phase A Mobile phase B
Identification (in min) (per cent v/v) (per cent v/v)

Determine by thin-layer chromatography (2.4.17), coating the


o 95 5
plate with silica gel GF254. 25 95 5
30 0 100
Mobile phase. A mixture of 15 volumes of ethyl acetate, 1.5
volumes of methanol and 1 volume of water. 40 0 100
42 95 5
Test solution. Dissolve 0.5 g of the extract under examination
with 10 ml of water and filter. Inject the reference solution. The test is not valid unless the
Reference solution. A 0.01 per cent w/v solution of gallic ieTiitivestanaara-creviii1ion-for therep1icateiiijections isiiof
acid RS in water. more than 2.0 percent.
Apply to the plate 10 III ofeach solution as bands 10 mm by 2 Inject the reference solution and the test solution.
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air Calculate the content ofthe gallic acid in extract.

2472

www.webofpharma.com
IP 2010 AMRA

Assay. Weigh accurately about 2 g of the extract, add 50 ml Identification


boiling water and heat it on a water-bath for 30 minutes with
A. Macroscopic - The surface is rough due to longitudinal
frequent stirring. Allow the solution to settle and carefully
cracks, fissures and scattered, raised lenticels, gr~yish to dark
decant it through a piece ofcotton wool to a 500-ml volumetric
brown externally and yellowish~white to reddish internally. .
flask. Repeat the extraction for 5 times with 50 ml of boiling
water. To confirm that all tannins have been extracted, add 3- B. Microscopic - The mature bark shows a wide cork which
4 drops offerric ammonium sulphate solution to 5 ml of the has tangentially elongated cells a few outer layers are brown
extract. Blue colour will not be produced, if all tannins have and inner lighter in colour, resin canals and yellow coloured
been extracted. If blue colour develops extract again with 2 tannin sacs are found in the phloem region, stone cells are
times with 50 ml of boiling water and check with ferric thick walled and lignified, prismatic calcium oxalate crystals
ammonium sulphate solution again. Cool the extracts and are present.
make up to the mark with water. Take 50 ml into a 250-ml
C. Determine by thin-layer ohromatography (2.4.17), coating
conical flask and add 50 ml of indigo suiphonic acidsolution,
the plate with silica gel GF254.
prepared by dissolving 1 g of indigo carmin in 50 ml of
sulphuric acid, add 500 ml of water and dilute this solution to Mobile phase. A mixture of 100 volumes of ethyl acetate,
1000.0 m!. Titrate, with 0.1 M potassium permanganate using 11 volumes of formic acid, 11 volumes of acetic acid and
indigo sulphonic acid as indicator until a golden yellow colour 25 volumes of water.
is produced. Carry out a blank titration. Test solution. To 5.0 g of the coarsely powdered substance
1 ml of 0.1 M potassium permanganate is equivalent to under examination, add 50 ml of methanol and reflux for
0.004157 g ofpolyphenoIs calculated as tannic acid. 15 minutes, cool and filter. Reflux the residue further for three
times with 50 ml of methanol, cool and filter. Combine all the
Usual strength. 40 per cent w/w.
filtrates and evaporate under vacuum to 10m!.
Storage. Store protected from heat and moisture.
Reference solution. Weigh about 2.0 g of amra RS, add 50 ml
of methanol and reflux for 15 minutes, cool and filter. Reflux
the residue further three times with 50 ml of methanol, cool
Amra and filter. Combine all the filtrates and concentrate under
vacuum to 4 m!.
Mango; Mangifera indica
Apply to the plate 10 III ofeach solution as bands 10 rnm by 2
rnm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with vanillin glacial acetic acid reagent. Heat the plate at
100° for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 10.0 percent.
Water-soluble extractive (2.6.3). Not less than 10.0 per cent
by method I.
Total ash (2.3.19). Not more than 16.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Amra consists ofdried stem bark of Mangifera indica L.(Fam. Heavy metals (2.3.13). 1.0 g complies with the limit test for
Anacardiaceae). heavy metals, Method B (20 ppm).

Amra contains not less than 1.5 per cent of mangiferin Loss on drying (2.4.19). Not rllorethan 12.0 per cent,
calculated on the dried basis. determined on 5 g by drying in an oven at 105°.

Description. The dried stem bark occurs in pieces ofvariable Microbial contamination (2.2.9). Complies with the microbial
size and thickness, surface rough. Odour pleasant and taste contamination tests.
astringent. Assay. Determine by liquid chromatography (2.4.14).

2473

www.webofpharma.com
AMRA IP 2010

Test solution. Weigh 2 g of coarsely powdered substance Identification


under examination, add 10 ml ofdimethylformamide, sonicate
for 5 minutes and add 50 ml of methanol and boil on a water- A. Macroscopic - Root is 30 cm or more long and from 3 to
bath for 10 minutes, cool and dilute to 100.0 ml with methanol 6 rom thick, rigid, tortuous, cylindrical, and little branched,
and filter. Dilute 5.0 rnl ofthis solution to 50.0 ml with methanol. consisting of a ligneous center, and a brownish, corky bark,
marked with longitudinal furrows and transverse fissures. The
Reference solution. Dissolve 10 mg ofmangiferin RS in 10 ml odour is aromatic, recalling that of tonqua bean, the taste is
ofdimethylformamide and dilute to 100.0 ml with methanol. aromatic and sweetish. On one side of the root the cork is
Chromatographic system frequently separated from and raised above the cortex, and is
a stainless steel column 25 cm x 4.6 rom packed with transversely fissured.
octadecylsilane bonded to porous silica (5 /lm),
B. Microscopic - Transverse section of root shows 2-3
- mobile phase: filtered and degassed mixture of 15
layered cork cambium having compressed cells filled with
volumes of acetonitrile and 85 volumes of buffer pH
brown contents. Xylem traversed by narrow medullary rays.
2.8 prepared by dissolving 1.36 g of potassium di-
Pith absent and central region occupied by woody tissues.
hydrogen orthophosphate in 950 ml of water, adjust
Vessel shows pitted thickening. The transverse section
the pH 2.8 with orthophosphoric acid and make upto
exhibits numerous laticiferous cells in the secondary cortex.
1000rnl,
Powder in chloral hydrate solution shows unicellular and
flow rate. 1 ml per minute,
branched latex ducts; lignified elements and vessels with pitted
spectrophotometer set at 365 nm,
thickenings.
injection volume. 20 /ll.
Inject the reference solution. The test is not valid unless the C. Determine by thin-layer chromatography (2.4.17), coating
relative standard deviation for replicate injections is not more the plate with silica gel GF 254.
than 2.0 per cent. Mobile phase. A mixture of75 volumes oftoluene, 15 volumes
Inject the test solution and the reference solution. of ethyl acetate, 5 volumes of glacial acetic acid and 5
volumes of methanol.
Calculate the content of mangiferin.
Test solution. To 2 g of the coarsely powdered substance
Storage. Store protected from moisture and against attack by
under examination, add 40 rnl ofmethanol, reflux for 15 minutes,
insects and rodents.
cool and filter. Reflux the residue further for two times with 25
ml of methanol, cool and filter. Combine all the filtrates and
Anantmula concentrate under vacuum to 25 ml.
Sariva; Indian Sarsaparilla Reference solution. To 2 g of the anantmula RS, add 40 ml of
methanol, reflux for 15 minutes, cool and filter. Reflux the
9 residue further for two times with 25 ml ofmethanol cool and
filter. Combine all the filtrates and concentrate unde~ vacuum
8
t025rnl.
7
Apply to the plate 20 /ll of each soluti~n as bands 10 rom by
6
2 rom.Allow the mobile phaseto rise 8 cm. Dry the plate in air
5 and examine in ultraviolet light at 254 nm and 365 nm, spray
4 with anisaldehyde sulphuric acid reagent. Heat at 105" for 5-
10 minutes and examine the plate in day light. The
3
chromatographic profile of the test solution is similar to that
2 of the reference solution.
1
Tests
o 1
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Anantrnula consists ofthe dried roots ofHemidesmus indicus
(Linn) R.Br. (Fam.Asc1epiadaceae). Ethanol-soluble extractive (2.6.2). Not less than 8.0 per cent.

Allani:ffiulacontalnsnotfessthano.02ifper-centoflso=vanmill, Water,;;soluble extractive (2:6.3): Not less than 12:0 percent


by method I.
calculated on the dried basis.
Description. The roots are cylindrical, yellowish brown in Total ash (2.3.19). Not more than 15.0 percent.
colour. They have vanillin like odour and the taste is acrid. Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.

2474

www.webofpharma.com
IP 2010 ARACHIS OIL

.Heavy metals (2.3.13). 1.0 g complies with the limit test for Description. A clear, colourless or pale yellow oily liquid;
heavy metals, Method B (20 ppm). odour, faint and nutlike.
Loss on drying (2.4.19). Not more than 12 per cent, determined Identification
on 5.0 g by drying in an oven at 105°.
Determine by the thin-layer chromatography (2.4.17), coating
Microbial contamination (2.2.9). Complies with the microbial
the plate with Ideselguhr G.
contamination tests.
Mobile phase. Glacial acetic acid.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve 20 mg (one drop) of the substance
Test solution. Weigh 2 g ofthe coarsely powdered substance
under examination in 4 ml of chloroform.
under examination, add 30 ml of water, and reflux on a water-
bath for 15 minutes, cool and filter. Reflux the residue further Reference solution (a). Dissolve 20 mg (one drop) of arachis
with water till the extract turns colourless, cool and filter. oil in 4 ml of chloroform.
Combine all the filtrates and concentrate to a volume slightly Reference solution (b). Dissolve 20 mg (one drop) of
less than 50 ml. Dilute to 50.0 ml with methanol. cottonseed oil in 4 ml of chloroform.
Reference solution. A 0.002 per cent w/v solution of iso- Reference solution (c). Dissolve 20 mg (one drop) of sesame
vanillin RS in methanol. oil in 4 ml of chloroform.
Chromatographic system Impregnate the plate by placing it to a depth of about 5 mm in
- a stainless steel column 25 cm x 4.6 mm, packed with a tank containing a shallow layer ofa mixture of95 volumes of
octadecylsilane bonded to porous silica (5 Ilm), light petroleum (60° to 80°) and 5 volumes of liquidparqlfin,
mobile phase: A. 0.1 per cent w/v solution of formiC allowing the solvent to rise at least 14 cm, removing the plate
acid in water and allowing it to dry in air for 5 minutes; use with the flow of
B. acetonitrile, the mobile phase in the direction in which impregnation was
flow rate. 1 ml per minute, carried out.
a linear gradient programme using the conditions given
Apply to the plate 2 III of each solution. Allow the mobile
below,
phase to rise 12 cm. Dry the plate at 110° for 10 minutes, allow
spectrophotometer set at 230 urn,
to cool and expose it to iodine vapour in a saturated chamber.
injection volume. 20 Ill.
Remove the plate and allow it to stand for a few minutes until
Time Mobile phase A Mobile phase B the brown background colour has disappeared. Spray the plate
(in min.) (per cent v/v) (per cent v/v) with starch solution; blue spots appear which become brown
o 90 10 on drying and become blue again on spraying with water. The
10 70 30 spots in the chromatogram obtained with the test solution
35 10 90 correspond to those in the chromatogram obtained with
36 90 10 reference solution (a).
40 90 10
Tests
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more Weight per ml (2.4.29).0.908 g to 0.920 g.
than 2.0 per cent. Refractive index (2.4.27). 1.467 to 1.470.
Inject the test solution and the reference solution.
Alkaline impurities. Mix 10 ml of recently distilled acetone
Calculate the content ofiso-vanillin. and 0.3 ml of water in a test-tube, add 0.05 m1 ofa 0.04 per cent
Storage. Store protected from heat, moisture and against attack w/v solution of bromophenol blue in ethanol (95 per cent)
of insects and rodents. and neutralise the solution, if necessary, with 0.01 M
hydrochloric acid or 0.01 M sodium hydroxide. Add 10 m1 of
the substance under examination, shaIce and allow'to stand.
Arachis Oil Not more than 0.1 ml of 0.01 M hydrochloric acid is required
to change the colour of the upper layer to yellow. .
Groundnut Oil; Peanut Oil Semi-drying oils. Boil 1 g in a flask under a reflux condenser
Arachis Oil is the refined fixed oil obtained from the seed for 5 minutes with 5 ml of a mixture of 3 volumes of 2 M
kernels of one Of more of the cultivated varieties of Arachis ethanolic potassium hydroxide and 1 volume of ethanol
hypogaea Linn. (Fam. Leguminosae) and may contain suitable (95 per cent), add 1.5 m1 of 6 M acetic acid and 50 ml of
antioxidants as stabilisers. ethanol (70 per cent), warm until the solution is clear. Cool

2475

www.webofpharma.com
ARACHIS OIL IP 2010

slowly with a thennometer in the liquid; the temperature at AIjuna consists ofdried stem bark ofTerminalia arjuna (Roxb)
which the solution becomes turbid is not lower than 36°. Wight & Am (Fam. Combretaceae)
Peroxide value (2.3.35). Not more than 5.0. Arjuna contains not less than 0.02 per cent of arjungenin
Acid value (2.3.23). Not more than 0.5. .calculated on the dried basis.

Iodine value (2.3.28). 85 to 105. Description. A flat or minutely curved thick pieces of bark
with reddish gray colour and astringent taste.
Saponification value (2.3.37). 185 to 196.
Rancidity. Shake 1 ml of a 10 per cent v/v solution in ether Identification
with 1 ml of hydrochloric acid, add 1 ml of a 0.1 per cent w/v
solution of phloroglucinol in ether; no red or pink colour is A. Macroscopic - Stem bark pieces, flat or minutely curved,
produced. with reddish gray external surface and darker inner surface.
Internal surface has longitudinal minute ridges. Fractures
Unsaponifiable matter (2.3.39). Not more than 1.0 per cent. longitudinal.
Cottonseed oil. In the Identification test, the chromatogram
B. Microscopic - Cork consisting of6-10 layers ofelongated
obtained with the test solution does not correspond to that
cells, phloem broad, medullary rays uruseriate. Calcium oxalate
obtained with reference solution (b)
clusters abundant. Few of the parenchyma cells contain
Sesame oil. In the Identification test, the chromatogram colouring matter.
obtained with the test solution does not correspond to that
C. Detennine by thin-layer chromatography (2.4.17), coating
obtained with reference solution (c).
the plate with silica gel GF254.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method B (10 ppm). Mobile phase. A mixture of 5 volumes of toluene,S volumes
of ethyl acetate and 0.5 volume of acetic acid.
Arachis Oil intendedforuse in the manufacture ofparenteral
preparations complies with the follOWing additional Test solution. Reflux 2 g ofcoarsely powdered substance under
requirement. examination with 50 ml of chloroform for 15 minutes, cool and
filter. Reflux the residue further with 50 rnl of chloroform.
Water (2.3.43). Not more than 0.3 percent, determined on 3.0 g.
Combine the filtrate and concentrate under vacuum to dryness.
Storage. Store protected from light in a well-filled container. Dissolve the residue in 10 ml of ethanol at 50° for 10 minutes
Labelling. The label states (1) whether the contents are and filter.
suitable for use in the manufacture ofparenteral preparations; Reference solution. Reflux 1 g of arjuna RS with 50 ml of
(2) when the addition of antioxidants is authorised, the name chloroform for 15 minutes, cool and filter. Reflux the residue
and quantity of the added antioxidants. further with 50 ml of chloroform. Combine the filtrate and
concentrate under vacuum to dryness. Dissolve the residue
in 5 ml of ethanol at 50° for 10 minutes and filter.
Arjuna Apply to the plate 10 ~l ofeach solution as bands 10 mID by 2
mID. Allow the mobile phase to rise 8 cm. Dry the plate in air
Terminalia arjuna Bark and examine in ultraviolet light at 254 urn and 365 urn, spray
with 10 per cent w/v solution of sulphuric acid in methanol.
Heat the plate at 110° for 5 minutes and examine in day light.
The chromatographic profile of the test solution is similar to
that of the reference solution.

Tests

Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 20.0 per cent.
Water",solubleextractive(2.6.3).Notlessthan20per~centby­
method!.
Total ash (2.3.19). Not more than 30.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.

2476

www.webofpharma.com
IP 2010 ARJUNA DRY EXTRACT

Heavy metals (2.3.13). 1.0 g complies with the limit test for arjunolic acid on the dried basis. It may contain suitable added
heavy metals, Method B (20 ppm). . substances.
Loss on drying (2.4.19). Not more than 10.0 per cent, Description. A light brown to beige powder with or without
determined on 5 g by drying in an oven at 105°. red tinge.
Microbial contamination (2.2.9). Complies with the microbial Identification
contamination tests.
A. Determine by thin-layer chromatography (2.4.17), coating
Assay. Determine by liquid chromatography (2.4.14).
the plate with silica gel GF254.
Test solution. Reflux 5 g ofcoarsely powdered substance under
Mobile phase. A mixture of 92 volumes of chloroform and 8
examination with 50 ml of chloroform for 15 minutes, cool and
volumes of methanol.
filter. Reflux the residue further with 50 ml of chloroform, cool
and filter. Combine the filtrates and concentrate under vacuum Test solution. Dissolve 50 mg ofthe extract under examination
to dryness, then extract dried residue with 10 ml of ethanol at with 50.0 ml of methanol and filter.
50° for 10 minutes and filter.
Reference solution. A 0.1 per cent w/w solution of arjunolic
Reference solution. A 0.1 per cent w/v solution of aljungenin acid RS in the methanol.
RS in ethanol.
Apply to the plate 5 /ll of each solution as bands 10 mm by 2
Chromatographic system mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
- a stainless steel column 25 cm x 4.6 mm packed with and spray with anisaldehyde sulphuric acid reagent. Heat
octadecylsilane bonded to porous silica (l0 /lm), the plate at 110° for 10 minutes and examine the plate in
mobile phase: A. acetonitrile (70 per cent) in water, ultraviolet light at 365 nm and day light. The chromatographic
B. acetonitrile (30 per cent) in water, profile of the test solution is similar to that of the reference
flow rate. 1.2 ml per minute, solution.
spectrophotometer set at 210 urn,
B. Determine by thin-layer chromatography (2.4.17), coating
injection volume.20 III
the plate with silica gel GF254.
Time Mobile phase A Mobile phase B
(in min) (per cent v/v) (per cent v/v) Mobile phase. A mixture of 35 volumes of chlorofom, 10
volumes of methanol and 2 volumes of water.
o 30 70
10 50 50 Test solution. Dissolve 50 mg ofthe extract under examination
with 10.0 ml of methanol and filter.
30 70 30
50 30 70 Reference solution. A 0.05 per cent w/w solution of aljugenin
RS in the methanol.
Inject the reference solution. The test is not valid unless the
relative standard deviation for the replicate injections for the Apply to the plate 5 III of each solution as bands 10 mm by 2
analyte peak corresponding to atjungenin is not more than 2. 0 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
per cent. and spray with vanilline sulphuric acid reagent. Heat the
plate at 110° for 10 minutes and examine the plate in ultraviolet
Inject the reference solution and the test solution. light at 365 urn and day light. The chromatographic profile of
Calculate the contents of arjungenin. the test solution is similar to that of the reference solution.
Storage. Store protected from light, heat, moisture and against Tests
attack by insects and rodents.
Ethanol-soluble extractive (2.6.2). Not less than 80.0 per cent.
Total ash (2.3.19). Not more than 5.0 per cent.

Arjuna Dry Extract Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Arjuna Dry Extract is obtained by extracting Arjuna
(Terminalia aljuna Wight and Arn,Fam. Combretaceae) bark Loss on drying (2.4.19). Not more than 5.0 per cent, determined
with methanol or any other suitable solvent and evaporation on 1 g by drying in an oven at 105°.
of solvent. Microbial contamination (2.2.9). Complies with the microbial
Atjuna Dry Extract contains not less than 90.0 per cent w/w contamination tests.
and not more than 110.0 percent w/w of stated amount of the Assay. Determine by liquid chromatography (2.4.14).

2477

www.webofpharma.com
ARTEMISIA IP 2010

Test solution. Shake a quantity ofthe extract under examination 3-pinnatipartite, petiolate and much variable in size (2.5-
containing about 50 mg of mjunolic acid in 50.0 ml of the 10.0 cm long). Leaf lobes narrow, oblong to elliptical with
methanol, filter. acuminate tip, about 1.0 mm wide. Petioles up to 1.0 cm long,
Reference solution. A 0.1 per cent w/v solution of aljunolic base amplexicaul. Inflorescence panicled raceme. Dry capitula
acid RS in methanol. yellowish brown, pedicellate, heterogamous globose to sub-
globose or disc shaped, 2.0 mm in diameter, flower heads
Chromatographic system
arranged in lax or drooping, involucral bracts 3- seriate,
a stainless steel column 25 cm x 4.6 mm packedwith
greenish yellow, glabrous and oblong in shape, measure
octadecylsilane bonded to porous silica (5 ~m),
1.0-1.2 x 0.5 mm in size. Inner involucral bracts elliptic having
mobile phase: a mixture of 35 volumes of 5 mM
a median greenish streak on its outer surface. Ray florets
a-cyclodextrin and 65 volumes of methanol,
pistillate, 6-8 in number per capitulum and 1.0-1.2 mm long.
flow rate. 1 ml per minute,
Disc florets hermaphrodite, 20-36 florets per capitulum and
spectrophotometer set at 205 urn,
0.8-1.0 mm long. All florets possess capitate oil glands on the
injection volume. 20 ~l.
middle of the outer surface that are 54-83 ~m in diameter.
Inject the reference solution. The test is not valid unless the Stamens 0.7 mm long attached to the corolla base, anther
relative standard deviation for replicate injections is not more appendages lanceolate to triangular with acuminate tip. Anthers
than 2.0 per cent. oblong and introrse. Pollen grains tricolpate, rounded 21-33
Inject the reference solution and the test solution. ~m in diameter. In dry condition, capitula become empty
because florets/achenes come out of it. Receptacle globular
Calculate the content ofthe arjunolic acid in the extract.
to oblong. Achenes minute; possess striated surface,
Usual strength. 60 per cent w/w. yellowish brown in colour, 0.7-1.0 mm long, and oval to elliptic
Storage. Store protected from heat and moisture. in shape. Stomatal index: 8-10-12.
B. The powder ofthe herb is grayish green to greenish yellow.
Examined under a microscope using chloral hydrate solution.
Artemisia The powder shows the following diagnostic characteristics:
T-shaped and globular trichomes, epidermal cells with wavy
Artemisia annua walls, anomocytic to anisocytic stomata, minute druse crystals,
tricolpate rounded pollen grains, stone cells, annular vessels,
rod shaped palisade cells and stigma with small and club
shaped papillae.
C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Mobilephase. Amixtureofl volume of hexane and 1 volume
of diethyl ether.
Test solution. Boil 0.1 g of the coarsely powdered substance
under examination with 10 ml of hexane for 10 minutes and
filter. Evaporate the filterate to 1 ml.
Reference solution. A 0.1 per cent w/v solution of
artemisia RS in hexane.
Apply to the plate 5 ~l of each solution as bands 10 mm by 2
Artemisia consists of dried leaves or the dried leaves and mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
flowering tops of Artemisia annua L. (Fam. Asteraceae), and examine in ultraviolet light at 254 urn and 365 urn, spray
Imown as Qinghao. with anisaldehyde sulphuric acid reagent. Heat the plate at
Artemisia contains not less than 0.8 per cent of artemisinin, 100° for 15 minutes and examine in day light. The
calculated on the dried basis. chromatographic profile of the test solution is similar to that
odour and bitter in taste. of the reference solution.

Identification Tests

A. The leaves grayish green, slightly darker upper surface, Foreign organic matter (2.6.1). Not more than 2.0 per cent.
glabrous to sparsely hairy, break easily into small fragments, Total ash (2.3.19). Not more than 11.0 per cent.

2478

www.webofpharma.com
IP 2010 ASHWAGANDHA

Acid-insoluble ash (2.3.19). Not more than 3.0 per cent. Identification
Assay. Determine by high performance thin-layer A. Macroscopic - Primary roots are straight, conical or finger
chromatography (2.4.17), coating the plate with silica gel like in shape, variable in thickness with the age. Secondary
GF254. roots are thin and fibrous. Surf~ce buff to greyish-yellow
Mobile phase. A mixture of 1 volume of hexane and 1 volume with longitudinal wrinkles.
of diethyl ether. B. Microscopic - Vessels with bordered pits and horizontal
Test solution. To 0.1 g of the coarsely powdered substance perforations. Fibres aseptate with pointed ends. Wood
under examination, add 10 ml of hexane and keep for 12 hours, elements lignified. Starch grains abundant, simple, mostly
filter. Repeat the process of extraction 3 times. Combine the spherical, reniform - oval with central hilum. Microcrystals in
extracts, evaporate and dissolve in 1.0 ml of hexane. parenchyma cells.

Reference solution. A 0.1 per cent w/v solution of artemisinin C. Determine by thin-layer chromatography (2.4.17), coating
RS in hexane. the plate with silica gel GF254.

Apply to the plate 5 III of each solution as bands 10 mm by Mobile phase. A mixture of 9 volumes of chloroform and
2 rom. Allow the mobile phase to rise 8 cm. Dry the plate in air 1 volume of methanol.
and spray with a mixture of50 volumes ofglacial acetic acid, Test solution. Reflux 3 g ofcoarsely powdered substance under
1 volume of sulphuric acid and 0.5 volume of anisaldehyde. examination with 50 ml methanol for 15 minutes, cool and
Heat the plate at 100° for 15 minutes, scan the plate in filter.
absorbance mode at 540 nm. Record the chromatograms and
Reference solution. Reflux 0.6 g of coarsely powdered
measure the responses for the analyte peak.
ashwagandha RS with 10 ml methanol for 15 minutes, cool
Calculate the content of artemisinin. and filter.
,
Storage. Store protected from light and moisture and against Apply to the plate 10 III of each solution as bands 10 rom by 2
attack by insects and rodents. rom. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 urn and 365 urn, spray
with anisaldehyde sulphuric acid reagent. Heat the plate at
100° for 5-10 minutes and examine the plate in day light. The
Ashwagandba chromatographic profile of the test solution is similar to that
Indian Ginseng; Withania somnifera of the reference solution.

Tests
9
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
8

7
Ethanol-soluble extractive (2.6.2). Not less than 10.0 per cent.

6 Water-soluble extractive (2.6.3). Not less than 15 per cent by


Method I.
5

4 Total ash (2.3.19). Not more than 7.0 per cent.


3 Acid-insoluble ash (2.3.19). Not more than 1.2 per cent.
2 Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
o Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°.
Ashwagandha consists of the dried mature roots of Withania
somnifera (L.) Dunal (Fam. Solanaceae). Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Ashwagandha contains not less than 0.02 per cent w/w of
total withanolide (sum of withanolide glycosides and Assay. Determine by liquid chromatography (2.4.14).
withanolide aglycones), calculated on the dried basis. Test solution. Reflux about 5 g of the coarsely powdered
Description. Buff to greyish-yellow roots. Taste, slightly substance under examination with 50 ml of acetonitrile on a
mucilaginous, bitter and acrid. water-bath for 15 minutes, cool and filter. Reflux the residue

2479

www.webofpharma.com
ASHWAGANDHA IP 2010

further with acetonitrile till the last extract turns colourless, withanone. Sum the content of withanolide glycosides and
cool and filter. Combine all the filtrates and concentrate to 50.0 withanolide aglycones to get total withanolides.
ml. Storage. Store protected from heat, moisture and against attack
Reference solution. A solution containing 0.01 per cent by insects and rodents.
w/v each of withanolide A RS and withanoside IV RS in
acetonitrile, prepared by heating gently on a water-bath.
Chromatographic system Ashwagandha Dry Extract
- a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 Ilm), Ashwagandha Dry Extract is obtained by extracting
- mobile phase: A. a buffer solution prepared by Ashwagandha (Withania somnifera (L.) Dunal, Fam.
dissolving 1.36 g of potassium dihydrogen phosphate Solanaceae) roots with methanol or any other suitable solvent
in 900 ml water, adjust pH to 2.8 with dilute phosphoric and evaporation of solvent.
acid and diluting it to 1000 ml with water, Ashwagandha Dry Extract contains not less than 90.0 per
B. acetonitrile, cent and not more than 110.0 per cent of the stated amount of
- a linear gradient programme using the conditons given withaferin A, not less than 85.0 per cent and not more than
below, 110.0 per cent w/w ofthe total withanolides calculated as the
- flow rate. 1.5 ml per minute, sum of free withanolides and glycowithanolides calculated
- spectrophotometer set at 227nm, on the dried basis. The extract also contains not less than 1.0
- injection volume.20 Ill. per cent of alkaloids, calculated on the dried basis.
Time Mobile phase A Mobile phase B Description. A pale brown to light brown powder.
(in min) (per cent v/v) ( per cent v/v)
o 95 5 Identification
18 55 45 Determine by thin layer chromatography (2.4.17), coating the
25 20 80 plate with silica gel GF254.
27 95 5 Mobile phase. A mixture of 5 volumes of toluene, 5 volumes
37 95 5 of ethyl acetate and 1 volume offormic acid.

Inject the reference solution.The relative retention time of Test solution. Dissolve about 0.2 g of the extract under
withanolide A is 1.0 and withanoside IV is about 0.7.The test examination with 10 ml of methanol and filter.
is not valid unless the relative standard deviation for the Reference solution. A 0.05 per cent w/v solution of
replicate injections for both the analyte peaks is not more withaferin A RS in methanol.
than 2.0 per cent and resolution is not less than 2.0. The relative
retention time of withanolide glycosides and withanolide Apply to the plate 10 III of each solution as bands 10 mm by
aglycones are as follows. 2 mm. Allow the mobile phase to rise 8 em. Dry the plate in air
and examine in ultraviolet light at 254 nm. Spray the plate with
Withanolide glycoside anisaldehyde-sulphuric acid reagent. Heat the plate at 110 0
Withanoside IV 0.7 for 10 minutes and examine the plate at 365 nm and under day
Withanoside V and VI 0.89 light. The chromatographic profile ofthe test solution is similar
to that of the reference solution.
Withanolide aglycones
12, deoxywithastramonolide 0.96 Tests
Withanolide A 1.0 Water-soluble extractive (2.6.3). Not less than 70.0 per cent
Withanolide B 1.14 by method!.
Withanone 1.01 Sulphated ash (2.3.18). Not more than 10.0 per cent.
Inject the reference solution and the test solution. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Ca,lc1.!~t~!h~cont~!!t1i. Qfl1!tfhC!n~l£c.1e.glyc:g§Lrk§i!!th~ 1ia,IIlPLe
as withanoside IV by summing the peak areas of withanoside Loss on drymg(2.4: 19):Nofmorethiiii 5.Operceiit, deteiiTImed
IV,ValJ.d Vi. Calulate the content ofwithanolide aglycones in bn 1 g by drying in an ovenatl05°.
the sample as withanolide A by summing the peak areas of 12, Microbial contamination (2.2.9). Complies with the microbial
deoxywithastramonolide, withanolide A, withanolide Band contamination tests.

2480

www.webofpharma.com
IP 2010 BELLADONNA LEAF

Assay Calculate the content oftotal withanolides as the sum of free


A. Total aUmloids withanolides and glycowithanolides.

Test solution. Shake about 20 g ofthe extract under examination D.Withaferin A


with 400 ml ofa solution containing 4 volumes of ether and 1 Assay. Determine by liquid chromatography (2.4.14).
volume of ethanol. Add 20 ml of 5 per cent v/v ammonia
solution into 1000-ml conical flask and shake for one hour. Test solution. Dissolve a quantity of the extract under
Decant and filter through cotton. examination containing about 25 mg ofwithaferin A in 25.0 ml
of water, filter.
Transfer the filtrate into a separator. Wash the residue with a
mixture containing 80 volumes of ether and 20 volumes of Reference solution. A 0.1 per cent w/v solution of withaferin
ethanol. To the filtrate add 100 ml of 0.5 M sulphuric acid. A RS in methanol.
Collect the lower layer into another separator. To the ether Chromatographic system
layer; add 100 ml ofa mixture containing 80 volumes of 0.25 M a stainless steel column 25 cm x 4.6 rom packed with
sulphuric acid and 20 volumes of ethanol and extract. octadecylsilane bonded to porous silica (10 J.l.m),
Continue the extraction 3 times with 80 ml of the 0.25 M mobile phase: a mixture of65 volumes ofO.l·per cent
sulphuric acid and a mixture containing 80 volumes of ether v/v solution of orthophosphoric acid and 35 volumes
and 20 volumes of ethanol until aqueous layer is colourless. of acetonitrile,
Combine the acid solution and wash with 40 ml of chloroform - flow rate. 1ml per minute,
followed by 2 times with 20 ml of chloroform. Wash, the spectrophotometer set at 220 nm,
combined chloroform layer, with acid alcohol mixture. Discard injection volume. 20 J.l.l.
the chloroform layer. Combine the acid alcohol solutions and Inject the reference solution. The test is not valid unless the
make it alkaline with 5 per cent v/v ammonia solution and add relative standard deviation for replicate injections is not more
10 ml excess. Extract 3 times with 100 ml of chloroform. Add 2 than 2.0 per cent.
mlof 0.1 M hydrochloric acid to 0.5 ml ofthe extract, remove
the chloroform by evaporation, transfer the aqueous residue Inject the test solution and the reference solution.
to a test tube and add 0.05 ml of potassium mercury-iodide Calculate the content of the withaferin A in the extract.
solution; not more than a very faint opalescence is produced.
Usual strength. 2.5 per cent withaferin A and 7.5 to 10 per cent
If the opalescence is more, the extraction is not complete.
w/w of total withanolides and glycowithanolides.
Extract further 2 times with 100 ml of chloroform.
Combine the chloroform extract and wash with 20 ml of Storage. Store protected from heat and moisture.
distilled water. Filter the chloroform layer through cotton plug
into a tared beaker. Wash the residue with a little chloroform,
transferring the chloroform layer to the same tared beaker. Belladonna Leaf
Evaporate on a water-bath. Add 5 ml of alcohol to the residue
and dry to constant weight at 105°. Finally weigh the residue
and calculate the content of total alkaloids.
9
B. Total withanolides. Shake about 5 g ofdry extract with 25
ml of methanol and 25 ml of water in a separating funnel. It is 8

defatted by extraction 4 times with 50 m1 ofhexane. This fraction 7


is discarded. The remaining aqueous methanolic solution is 6
extracted with hexane and then with 5 times with 25 ml of
5
ether. The ether extracts are combined, washed twice with
water and dried under vacuum to give the free withanolides 4
content. Finally weigh the residue and calculate the content 3
offree withanolides.
2
C. Glycowithanolides. The above remaining aqueous
1
methanolic solution is extracted 3 times with 2 volumes of
chloroform and 1 volumes of methanol. The chloroform- o
methanol extracts are combined and evaporated on water-
bath and dried under vacuum at 80° to constant weight. Finally Belladonna Leaf consists of the dried leaf and flowering tops
weigh the residue and calculate the content of of Atropa belladonna Linn. or of A. acuminata Royle ex
glycowithanolides. Lindley (Fam. Solanaceae) or a mixture ofboth species.

2481

www.webofpharma.com
BELLADONNA LEAF IP 2010

Belladonna Herb contains not less than 0.30 per cent of total 15 minutes, filter and wash the filter with 0.05 M sulphuric
alkaloids, calculated as hyoscyamine with reference to the acid until 20 m1 of filtrate is obtained; add 1 ml of strong
material dried at 100° to 105°. ammonia solution to the filtrate, extract with two quantities,
each of 10 ml, ofperoxide-free ether, separate the ether layer,
Description. Green to greenish-brown leaves, slightly darker
by centrifugation ifnecessary, dry the combined ether extracts
on the upper surface, often crumpled and rolled and partly
over anhydrous sodium sulphate, filter, evaporate to dryness
matted together in the drug. When whole, the lamina is 5 to
on a water-bath and dissolve the residue in 0.5 ml of methanol.
25 cm long and 3 to 12 cm wide, elliptical to ovate; acuminate
at the apex, narrowing at the base; margin entire. Petiole 0.5 to Reference solution. Add 15 ml of O. 05 M sulphuric acid to 0.6
4 cm in length. The young leaves are highly pubescent, the g of the belladonna leaf RS, shake for 15 minutes, filter and
older leaves are slightly pubescent along the veins. In the wash the filter with 0.05 M sulphuric acid until 20 ml offiltrate
flowering tops, the stems are hollow and flattened, with leaves is obtained, add 1 m1 ofstrong ammonia solution to the filtrate,
in pairs ofunequal size, in the axils ofwhich are single flowers extract with two quantities, each of 10 ml, of peroxide-free
with campanulate corolla, about 2 cm long and 1.5 cm wide, ether, separate the ether layer, by centrifuging if necessary,
purple or yellow-brown in colour, with five short, reflexed lobes, dry the combined ether extracts over anhydrous sodium
five epipetalous stamens and one bilocular ovary with sulphate, filter, evaporate to dryness on a water-bath and
numerous ovules. dissolve the residue in 0.5 ml of methanol. or

Identification Dissolve 50 mg of hyoscyamine sulphate in 9 ml of methanol


(solution A) and dissolve 15 mg of hyoscine hydrobromide
A. When examined under a microscope it shows epidermal in 10 mlofmethanol (solution B), mix 8 mlofsolutionAwith
cells with sinuate anticlinal walls and cuticle which is often 1.8 ml ofsolution B.
striated and furrowed. Covering and glandular hairs infrequent,
Apply to the plate 10 III and 20 III ofeach solution as bands 10
though more frequent in the young leaves and around the
mm by 2 mm. Allow the mobile phase to rise 8 cm. Dry the plate
veins; covering hairs, multicellular, uniseriate, with thin smooth
at 105° for 15 minutes, allow to cool and examine in ultraviolet
walls; glandular hairs; short clavate glands with multicellular
light at 254 nm and 365 nm, spray with 10 ml of modified
heads and glands with a long uniseriate static and ovoid
potassium iodobismuthate solution until the bands become
unicelluar head. Stomata, anisocytic, more frequent on the
visible as orange or brown on a yellow background. The bands
lower epidermis. The midrib is characterised by an open arc of
in the chromatogram obtained with test solution have similar
vascular bundles with isolated groups ofperimedullary phloem.
Rfvalues to those in the chromatogram obtained with reference
Mesophyll dorsiventral with a single palisade layer.
solution (hyoscyamine in the lower third ofthe chromatogram;
Throughout the parenchyma and particularly just below the
hyoscine in the upper third) and are similar in colour and
palisade layer are cells containing microsphenoidal crystals
atleast equal in size. Faint secondary bands may appear,
of calcium oxalate or, very rarely, cluster crystals. The stems
particularly in the middle ofthe chromatogram obtained with
show pericyclic fibres and perimedullary bundles ofphloem,
20 III of the test solution or near the line of application in the
few trichomes; the cortical parenchymatous cells and the pith
chromatogram obtained with 10 III oftest solution. Spray the
cells contain microsphenoidal crystals of calcium oxalate.
plate with a freshly prepared 10 per cent w/v solution of
B. Powder 1 g and shake for 2 minutes with 10 ml of O. 05 M sodium nitrite until transparent and examine after 15 minutes.
sulphuric acid. Filter and add to the filtrate 1 ml of strong The colours due to hyoscyamine in the chromatogram change
ammonia solution and 5 ml of water. Extract with 15 ml of from brown to reddish-brown but not to greyish-blue
ether, taking care to prevent the formation ofan emulsion. Dry (atropine); any secondary bands are no longer visible.
the ether extract over anhydrous sodium. sulphate and filter.
Evaporate the filtrate to dryness, add 0.5 ml ofjUming nitric Tests
acid and evaporate to dryness. Add 10 ml of acetone and, Foreign organic matter (2.6.1). Not more than 3 per cent.
dropwise, a 3 per cent w/v solution ofpotassium hydroxide in
Acid-insoluble ash (2.3.19). Not more than 3 per cent.
ethanol (95 per cent); a. deep violet colour develops.
Assay. Powder 50 g and determine the loss on drying (2.4.19),
C. Determine by thin-layer chromatography (2.4.17), coating by drying 2 g, accurately weighed, in an oven at 105°. From
the plate with silica gel GF254. the remaining sample, weigh accurately about 109, moisten
MOEiilephase.Airiixfiiie 6f90 volumes ofcicelone, 7v6liimes witha·mixture···ofS·ml·ofdiluteammoniasolution,lO.mLof
of water and 3 volumes of strong ammonia solution. ethanol (95 per cent) and 30 m1 of ether and mix thoroughly.
Transfer the mixture to a percolator with the aid ofan extracting
Test solution. Add 15 ml of 0.05 Msulphuric acid to 0.6 g of solvent mixture consisting on volumes of ether and 1 volume
the material under examination, in fine powder, shake for of chloroform. Allow to macerate for 4 hours and percolate

2482

www.webofpharma.com
IP 2010 BELLADONNA TINCTURE

with the solvent mixture until complete extraction of the solutions with successive quantities ofa mixture on volumes
alkaloids is effected, (2.6.4). . of 0.1 M sulphuric acid and 1 volume of ethanol (95 per
cent) until complete extraction of the alkaloids is effected,
Concentrate the percolate to about 50 ml by distilling off the
filtering each extract through a plug of absorbent cotton
solvent mixture on a water-bath, and transfer to a separator,
previously moistened with water. Wash the mixed acid
previously rinsed with ether. Add a quantity of ether at least
solutions with 10,5 and 5 ml of chloroform, extracting each
equal to 2.1 times the volume ofthe percolate and extract with
chloroform solution with the same 20 ml of O. 05 M sulphuric
three quantities, each of 20 ml, of 0.5 M sulphuric acid.
acid and discard the chloroform. Combine the acid solutions,
Transfer each acid extract to another separating funnel.
neutralise with dilute ammonia solution, add 5 ml in excess
Combine the acid extracts, make the solution alkaline with
and shake with successive quantities, each of 25 ml, of
dilute ammonia solution and extract with chloroform until
complete extraction of the alkaloids has been effected. Wash
chloroform until complete extraction ofthe alkaloids is effected,
washing each chloroform solution with the same 10 ml of water
the combined chloroform extracts with 10 ml water, discard
and filtering into a flask through a plug of cotton wool
the watel; evaporate the chloroform layer to dryness and heat
previously moistened with chloroform. Distil most of the
the residue for 15 minutes on a water-bath. Redissolve the
chloroform from the combined extracts and transfer the
residue in successive small quantities of chloroform,
remainder ofthe chloroform to a shallow open dish. Evaporate
evaporating to dryness on a water-bath each time before
the remainder ofthe chloroform without the aid ofa current of
adding the solvent. Heat for 15 minutes on a water-bath and
air, heat the residue in an oven at 1000 for 15 minutes, dissolve
dissolve the residue in 5 ml of chloroform. Add 20.0 ml of
in a little chloroform, evaporate to dryness without the aid of
0.01 M sulphuric acid, remove the chloroform by evaporation
a current ofair and again heat in an oven at 1000 for 15 minutes.
on a water-bath and titrate the excess of acid with 0.02 M
Dissolve the residue in 2 ml of chloroform, add 5.0 ml of
sodium hydroxide using methyl red solution as indicator.
0.025 M sulphuric acid, warm to remove the chloroform, cool
1 ml of 0.01 M sulphuric acid is equivalent to 0.005788 g of and titrate the excess of acid with 0.05 M sodium hydroxide
total alkaloids calculated as hyoscyamine. Calculate the using methyl red solution as indicator.
content oftotal alkaloids with reference to the dried material.
I ml of 0.025 M sulphuric acid is equivalent to 0.01447 g of
Storage. Store protected from light and moisture. allcaloids, calculated as hyoscyamine.
Storage. Store in small, wide-mouthed, tightly-closed
containers in a cool place.
Belladonna Dry Extract
Belladonna Dry Extract is obtained from the dried leaf and
flowering top of Atropa belladonna Linn or ofA. acuminata
Royle ex Lindley (Fam. Solanaceae) by extraction with ethanol
Belladonna Tincture
or any other suitable solvent. Belladonna Tincture obtained from Belladonna leaf or roots
Belladonna DIy Extract contains not less than 0.95 per cent ofone or more ofthe cultivated varieties ofAtropa belladonna
and not more than 1.05 per cent w/w ofallcaloids, calculated as Linn. or A. acuminata Royle ex Lindley (Fam. Solanaceae) or
hyoscyamine. a mixture of both species.
Belladonna Tincture contains not less than 90 per cent w/w
Tests and not more than 110 per cent w/w oftotal alkaloids, calculated
Loss on drying (2.4.19). Not more than 5.0 per cent, determined as hyoscyamine, C 17H23N0 3 •
on 0.5 g by drying in an oven at 105°. Description. A clear green or brownish green liquid.
Assay. Weigh accurately about 3 g and wash into a separating
funnel with 12 ml of a mixture of equal 'volumes of ethanol Identification
(95 per cent) and water, shake-well and frequently for A. Determine by thin-layer chromatography (2.4.17), coating
30 minutes, add 2 ml of dilute ammonia solution and 25 mlof the plate with silica gel GF254.
chloroform. Shake well, allow to separate and filter the
Mobile phase. A mixture of 10 volumes of anhydrousformic
chloroform layer into a second separating funnel through a
acid, 10 volumes of water, 30 volumes of methyl ethyl ketone
plug ofabsorbent cotton moistened with chloroform. Continue
and 50 volumes of ethyl acetate.
the extraction with further quantities, each of 25 ml, of
chloroform until complete extraction ofthe alkaloids is effected Test solution. Evaporate 10 ml ofthe tincture under examination
(2.6.4), running each chloroform solution through the same in a water-bath at 40° under reduced pressure. Dissolve the
plug of absorbent cotton. Extract the combined chlorofonn residue in 1.0 ml ofthe methanol.

2483

www.webofpharma.com
BELLADONNA TINCTURE IP 2010

Reference solution. Dissolve 1 mg of chlorogenic acid RS free ether. Add a quantity of peroxide-free ether equal to at
and 2.5 mg of rutin RSin 10 ml ofthe methanol. least 2.1 times the volume of the solution to produce a layer
Apply to the plate 40 III ofeach solution as bands 10 mm by 2 having a density well below that ofwater. Extract the resulting
mm. Allow the mobile phase to rise 15 cm. Dry the plate in air, solution with minimum of three quantities, each of20 ml of
heat the plate at 110° for 10 minutes. Spray the warm plate with 0.25 M sulphuric acid until the alkaloids are completely
1 per cent v/v solution of diphenylboric acid aminoethyl extracted. Separate the layers by centrifugation if necessary
ester in methanol. Allow to cool and spray with 5 per cent and transfer the layers to a separating funnel. Make the
v/v solution of macrogol 400 in methanol, allow the plate to combined layers alkaline with strong ammonia solution and
dry in air for 30 minutes and examine in ultraviolet light at 365 extract with minimum of three quantities, each of 30 ml of
nm. The chromatographic profile ofthe test solution is similar dichloromethane until the alkaloids are completely extracted.
to that of the reference solution. Combine the dichloromrthane and ether extracts, add 4 g of
anhydrous sodium sulphate and allow to stand for 30 minutes
B. Atropine. Detennine by thin-layer chromatography (2.4.17), with occasional shaking. Decant the dichloromethane and
coating the plate with silica gel G. filter. Wash the sodium sulphate with three quantities, each
Mobile phase. A mixture of 3 volumes of strong ammonia of 10 ml of dichloromethane. Combine the dichloromethane
solution, 7 volumes of water and 90 volumes of acetone. and ether extracts, evaporate to dryness on a water-bath.
Heat the residue in an oven at 105° for 15 minutes. Dissolve
Test solution. To 15.0 ml of the tincture under examination,
the residue in a few ml of dichloromethane, evaporate to
add 15 ml of 0.05 M sulphuric acid and filter. Add 1 mlof
dryness on a water-bath and heat the residue in an oven at
strong ammonia solution to the filtrate, with two quantities,
105° for 15 minutes again. Dissolve the residue in a few ml of
each of 10 ml, of peroxide·free ether, separate the ether layer
dichloromethane. Add 20.0 ml of 0.01 Msulphuricacid and
by centrifugation ifnecessary, dry the combined ether extracts
remove the dichloromethane by evaporation on a water-
over anhydrous sodium sulphate, filter and evaporate to
bath. Titrate the excess ofacid with 0.02 M sodium hydroxide
dryness on a water-bath. Dissolve the residue in 0.5 ml of
using methyl red mixed solution as indicator.
methanol.
1 ml of 0.01 M sulphuric acid is equivalent to 0.005788 g of
Reference solution. Dissolve 50 mg of hyoscyamine sulphate
total alkaloids calculated as hyoscyamine. Calculate the
in 9 ml of methanol and 15 mg of hyoscine hydrobromide in
content of total alkaloids with reference to the dried material.
10 ml of methanol, separately. Mix 1.8 ml of the hyoscine
hydrobromide solution and 8 ml ofthe hyoscyamine sulphate Usual strength. 0.03 per cent w/w.
solution.
Apply to the plate 20 III and 40 III ofeach solution as bands 10
mm by 2 mm. Allow the mobile phase to rise 10 cm. Dry the
plate at 105° for 15 minutes, spray with potassium Bhibhitaki
iodobismuthate solution, dry the plate and spray with sodium Belliric Myrobalan; Terminalia bellirica
nitrite solution until the plate is transparent. Examine the plate
after 15 minutes in day light. The chromatogram obtained with
the test solution corresponds to the chromatogram obtained
9
with the reference solution.
8
Tests
7
Ethanol (2.3.45). 64 to 69 per cent v/v by method Ill. 6

Assay. Evaporate 50.0 g ofthe tincture under examination to a 5


volume ofabout 10 ml. Transfer quantitatively to a separating 4
funnel, with the minimum volume of ethanol (70 per cent
3
v/v). Add 5 ml ofstrong ammonia solution and 15 ml of water.
Extract with three quantities, each of 40 ml of a mixture of 1 2
volume of dichloromethane and 3 volumes of peroxide-fi-ee
ether, 9aIiefully toaYQig iemulsiOIl, llIlliUhe_a.ll~alQLds_are
completely extracted. Combinethe dichloromethane and ether
extracts, concentrate the solution to a volume of about 50 ml
by heating on a water-bath. Transfer the resulting solution Bhibhitald consists of the dried fruit pericarp of Terminalia
quantitatively to a separating funnel, rinsing with peroxide- bellirica (Gaertn.) Roxb. (Fam. Combretaceae).

2484

www.webofpharma.com
IP 2010 BHIBHITAKI AQUEOUS EXTRACT

Bhibhitaki contains not less than 0.3 per cent w/w of ellagic Heavy metals (2.3.13). 1.0 g complies with the limit test for
acid and 0.75 per cent w/w of gallic acid, calculated on the heavy metals, Method B (20 ppm).
dried basis. Loss on drying (2.4.19). Not more than 12.0 per cent,
Description. The dried pericarp appears as curved pieces of determined on 5.0 g by drying in an oven at 105°.
irregular shapes, the external surface is velvety, wrinkled grey Microbial contamination (2.2.9). Complies with the microbial
to brown in colour and has astringent taste contamination tests.
Identification Assay. Determine by liquid chromatography (2.4.14).

A. Macroscopic - The dried pericarp of the ripe fruit occurs Test solution. Weigh 0.5 g of coarsely powdered substance
as curved pieces of irregular shapes with convex external under examination, add 50 ml of water, sonicate for 3 minutes
surface. The external surface appears velvety, slightly wrinlded and heat on a boiling water bath for 15 minutes, cool and
grey to brown in colour. Internal surface is pale yellow. The dilute to 100.0 ml with water and filter.
cut surface is with occasional projecting threads, representing Reference solution. A solution containing 0.01 per cent w/v
the vascular bundles. each of gallic acid RS and ellagic RS in wate]:
B. Microscopic- The cells ofthe epidermis has a characteristic NOTE - Use freshly prepared solution and protectedfrom
and a slightly bulged based with a hair like prolongation. light.
S~veral vascular strands traverse the mesocarp in various
Chromatographic system
directions. Peripheral layers of mesocarp have tangentially - a stainless steel column 25 cm x 4.6 mm packed with
elongated cells, devoid of starch grains, containing rosettes octadecylsilane bonded to porous silica (5 /-lm),
of calcium oxalate crystals and few small stone cells. mobile phase: A. a gradient mixtures of acetonitrile and
C. Determine by thin-layer chromatography (2.4.17), coating a buffer solution prepared by dissolving 0.136 g of
the plate with silica gel GF254. potassium di-hydrogen orthophosphate in 500 ml of
Mobile phase. A mixture of20 volumes of toulene, 45 volumes water, add 0.5 ml of orthophosphoric acid and make
of ethyl acetate and 20 volumes of glacial acetic acid and upto 1000 ml with wate];
5 volumes offormic acid. B. acetonitrile
flow rate. 1.5 ml per minute,
Test solution. Reflux 2 g ofthe coarsely powdered substance
spectrophotometer set at 270 nm,
under examination with 50-75 ml of methanol for 15 minutes,
injection volume. 20 /-l1.
cool and filter. Reflux the residue further for two times with
Time mobile phase A mobile phase B
75 ml of methanol, cool and filteE Combine allthe filtrates and
(in min) (per cent v/v) (per cent v/v)
concentrate under vacuum to 50 m1.
Reference solution. Reflux 0.4 g of the coarsely powdered
o 95 5
18 65 35
bhibhitald RS with 50-75 ml of methanol for 15 minutes, cool
25 45 55
and filter. Reflux the residue further for two times with 75 ml of
methanol, cool and filter. Combine all the filtrates and 30 95 5
concentrate under vacuum to 10 m1. Inject the reference solution. The relative standard deviation
Apply to the plate 10 /-ll ofeach solution as bands 10 mm by 2 for the replicate injections is not more than 2.0 per cent.
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air Inject the test solution and reference solution.
and examine in ultraviolet light at 254 nm and 365 nm, spray
Calculate the content of gallic acid and ellagic acid.
with anisaldehyde sulphuric acid reagent. Heat the plate at
100° for 5-10 minutes and examine in day light. The Storage. Store protected from light, heat, moisture and against
chromatographic profile of the test solution is similar to that attack by insects and rodents.
of the reference solution.
Tests
Bhibhitaki Aqueous Extract
Foreign organic matter (2.6.1). Not more than 2 per cent.
Bhibhitaki Aqueous Extract is obtained by extracting Bhibhitaki
Ethanol-soluble extractive (2.6.2). Not less than 25 per cent. (Terminalia bellirica (Gaeltn.) Roxb., Fam. Combretaceae) fruit
Water-soluble extractive (2.6.3). Not less than 35 per cent by with water.
Method 1.
Bhibhitaki Aqueous Extract contains not less than 90 per cent
Total ash (2.3.19). Not more than 8 percent. w/w and not more than 120.0 per cent w/w ofthe stated amount
Acid-insoluble ash (2.3 .19). Not more than 2 per cent. of gallic acid and ellagic acid.

2485

www.webofpharma.com
BHIBHITAKl AQUEOUS EXTRACT IP 2010

Description. A light brown to dark brown powder. Time Mobile phase A Mobile phase B
(in min) (per cent v/v) (per cent v/v)
Identification 0-8 95 5
Determine by thin-layer chromatography (2.4.17), coating the 8-25 95~0 5 ~100
plate with silica gel G.F 254. 25-30 o 100
Mobile phase. A mixture of35 volumes of benzene, 10 volumes 30-32 0~95 5 ~5

of methanol, 4 volumes of acetone and 1 volume of water. Inject reference solution (a) and (b). The test is not valid
Test solution. Dissolve 0.5 g ofthe extract under examination unless the relative standard deviation for the replicate
in 20 ml of methanol under reflux at 80 0 on a water bath and injections is not more than 2.0 per cent.
filter. Inject the test solution, reference solution (a) and (b).
Reference solution. A 0.01 per cent w/v solution of gallic Calculate the content of gallic acid and ellagic acid in the
acid RS in methanol. extract
Apply to the plate 10 III ofeach solution as bands 10 mm by 2 Usual strength. 7 per cent w/w.
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
Storage. Store protected from heat and moisture.
and examine in ultraviolet light at 254 run. The chromatographic
profile of the test solution is similar to that of the reference
solution.
Bhringraj
Tests
Eclipta alba
Acid insoluble ash (2.3.19). Not more than 4.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
9
heavy metals, Method B (20 ppm).
Loss ofdrying (2.4.19). Not more than 5.0 per cent, determined
7
on 1.0 g by drying in an oven at 105°.
6
Microbial contamination (2.2.9). Complies with the microbial
5
contamination tests.
4
Assay. Determine by liquid chromatography (2.4.14).
3
Test solution. Dissolve about 0.2 g of the extract under
2
examination or a quantity containing 10 mg ofgallic acid and
1 mg ofellagic acid in 80 ml of methanol and dilute to 100.0 ml
with water and filter. o
Reference solution (a). A 0.01 per cent w/v solution of gallic
Bhringraj consists of the dried whole plant of Eclipta alba
acid RS in 80 per cent v/v methanol in water.
(L.) Hassle. (Fam. Asteraceae).
Reference solution (b). A 0.001 per cent wIv solution of ellagic Bhringraj contains not less than 0.1 per cent ofwedelolactone,
acid RS in 80 per cent v/v methanol in water. calculated on the dried basis.
Chromatographic system Description. A green to greenish brown colour when
- a stainless steel column 25 cm x 4.6 mm packed with completely dry.
octadecylsilane bonded to porous silica (5 Ilm),
. - mobile phase: A mixture of acetonitrile and a 0.1 per Identification
cent v/v orthophosphoric acid in water,
A. Macroscopic - Root. Well developed, a number of
- a linear gradient programme using the conditions given
secondary branches arise from main root up to about 7 mm in
flow rate. 1 ml per minute,
- spectrophotometer set at 252 nm for ellagic aCid a.nd a.t Stem. Herbaceous, branched occasionally rooting at nodes,
271 urn for gallic acid, cylindrical or flat, rough due to oppressed white hairs, node
- injection volume 20 Ill. distinct, greenish, occasionally brownish.

2486

www.webofpharma.com
IP 2010 BHRINGRAJ

Leaf. Opposite, sessile to sub sessile, usually oblong, further with 3 x 25 ml of methanol, cool and filter. Combine all
lanceolate, sub-entire, sub-acute or acute, strigose with the filtrates and concentrate under vacuum to 5 ml.
appressed hairs on both surfaces.
Apply to the plate 10 III ofeach solution as bands 10 mm by 2
Flower. Solitary or 2, together on unequal axillary peduncles, mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
involucral bracts about 8, ovate, obtuse or acute, herbaceous, and examine in ultraviolet light at 254 nm and 365 nm, spray
strigose with oppressed hairs; ray flowers ligulate, ligule small, with anisaldehyde sulphuric acid reagent. Heat the plate at
spreading, scarcely as long as bracts, not toothed; white disc 100° for 5-10 minutes and examine the plate in day light. The
flowers tubular, corolla often 4 toothed; pappus absent, except chromatographic profile of the test solution is similar to that
occasionally very minute teeth on the top of achene; stamen of the reference solution.
5, filaments epipetalous, free, anthers united into a tube with
base obtuse; pistill bicarpellary; ovary inferior; unilocular with Tests
one basal ovule.
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Fruit. Achenial cypsella, one seeded, cuneate, with a nan-ow Ethanol-soluble extractive (2.6.2). Not less than 5.0 per cent.
wing, covered with warty excrescences, brown.
Water- soluble extractive (2.6.3). Not less than 15.0 per cent
Seed. Dark brown, hairy and non endospennic. by Method 1.
B. Microscopic - Powder. Dark green; shows vessels in Total ash (2.3.19). Not more than 22 per cent.
large groups or single broken pieces with pitted walls,
Acid-insoluble ash (2.3 .19). Not more than 11 per cent.
numerous fibres entire or in pieces, trichomes entire or in
pieces, warty, a few attached with epidennal and subsidiary Heavy metals (2.3.13). 1.0 g complies with the limit test for
cells, anomocytic and anisocytic stomata. heavy metals, Method B (20 ppm).

Root. The cells of outer one or two rows of secondary cortex, Loss on drying (2.4.19). Not more than 15.0 per cent,
elongated or rounded with air cavities, while cells of inner detennined on 5 g by drying in an oven at 105°.
secondary cortex, elongated to irregular in shape. Stone cells Microbial contamination (2.2.9). Complies with the microbial
scattered in secondary cortex. Phloem rays broader towards contamination tests.
the periphery, cells rounded. Xylem rays distinct, run straight Assay. Detennine by liquid chromatography (2.4.14).
in tangential section, rarely uniseriate and biseriate, cells pitted.
Test solution. Reflux 5 g of the coarsely powdered substance
Stem. A few epidennal cells elongate to fonn characteristic under examination with 30 ml of methanol on a water- bath for
non-glandular trichomes. Secondary cortex composed oflarge, 30 minutes, cool and filter. Reflux the residue further with
rounded parenchymatous cells having wide air space. Vascular methanol till the last extract turns colourless, cool and filter.
bundle in a ring, collateral, endarch, of varying size. Vessels Combine all the filtrates and concentrate to 100 ml.
barrel-shaped, some elongated with simple perforations, pitted
Reference solution. 0.01 per cent w/v solution of
with spiral thickening. A few xylem fibres bifurcate. Xylem
wedelolactone RS in methanol.
rays uniseriate or biseriate.
Chromatographic system
Leaf. Anomocytic and anisocytic stomata and non-glandular
a stainless steel column 25 cm x 4.6 mm packed with
hairs are present on both surface, more abundant on lower
octadecylsilane bonded to porous silica (5 Ilm),
side. Vascular bundle, fine in mid rib, central one largest while
- mobile phase: 35 volumes of acetonitrile and 60 volumes
four other small flanking either side of central bundle.
of0.1 per cent v/v phosphoric acid prepared by diluting
C. Detennine by thin-layer chromatography (2.4.17), coating 1ml ofphosphoric acid to I000 ml with water,
the plate with silica gel GF254. flow rate. 1 ml per minute,
- spectrophotometer set at 249 run,
Mobile phase. A mixture of9 volumes of toluene, 6 volumes
injection volume. 20 Ill.
of acetone and 1 volume offormic acid.
Inject the reference solution. The test is not valid unless the
Test solution. Reflux Ig of the coarsely powdered substance
relative standard deviation for the replicate injections is not
under examination with 25 ml of methanol for 30 minutes, cool
more than 2.0 per cent.
and filter. Reflux the residue further with 3 x 25 ml of methanol,
cool and filter. Combine all the filtrates and concentrate under Inject the reference solution and the test solution.
vacuum to 10 ml. Calculate the content of wedelolactone.
Reference solution. Reflux 0.5 g bhringrqj RS with 25 ml of Storage. Store protected from heat, moisture and against attack
methanol for 30 minutes, cool and filter. Reflux the residue by insects and rodents.

2487

www.webofpharma.com
BHUIAMLA IP 2010

Bhuiamla Apply to the plate 10 III ofeach solution as bands 10 mm by 2


mm. Allow the mobile phase to rise 8 em. Dry the plate in air
Phyllanthus amarus and examine in ultraviolet light at 254 nm and 365 nm, spray
with methanolic sulphuric acid (10 per cent, v/v) Heat the
plate at 1200 for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 15.0 per cent
by Method 1.
Total ash (2.3.19). Not more than 8.0 per cent.
Acid-insoluble ash (~.3 .19). Not more than 5.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Bhuiamla consists of the dried aerial parts of Phyllanthus
Loss on drying (2.4.19). Not more than 12.0 per cent,
amarus Schum. & Thorn. (Fam. Euphorbiaceae).
detennined on 5 g by drying in an oven at 105°.
Bhuiamla contains not less than 0.25 per cent w/w of total
Microbial contamination (2.2.9). Complies with the microbial
phyllanthin and hypophyllanthin, calculated on the dried
contamination tests.
basis.
Assay. Determine by liquid chromatography (2.4.14).
Description. A green to greenish yellow in colour, taste,
slightly bitter. Test solution. Reflux about 2 g of the coarsely powdered
substance under examination with 50 ml of methanol on a
Identification water bath for 15 minutes, cool and filter. Reflux the residue
further with methanol till the last extract turns colorless, cool
A. Macroscopic - Stem teret, 1-4 mm in diameter. Leaves
and filter. Combine all the filtr~tes and concentrate to 10.0 ml.
oblong 5 x 3 mm, short stalked, greenish brown in colour.
Reference solution (a). 'A 0.020 per cent w/v solution of
B. Microscopic - Stem, inner cortex chlorenchymatous; xylem
phyllanthin RS in methanol.
rays 1-2-seriate. Leafstomata mostly paracytic; epidermal cell
wall markedly sinuous; rosette and prismatic crystals ofcalcium Reference solution (b). A 0.020 per cent w/v solution of
oxalate along the veins and midrib. hypophyllanthin RS in methanol.
C. Determine by thin-layer chromatography (2.4.17), coating Chromatographic system
the plate with silica gel GF254. - a stainless steel column 25 em x 4.6 mm packed with
octadecylsilane bonded to porous silica (5Ilm),
Mobile phase. A mixture of 6 volumes of toluene, 2 volumes
- mobile phase: a mixture of65 volumes of methanol and
of ethyl acetate, 1 volume offormic acid and 0.2 volume of
35 volumes of water,
methanol.
flow rate. 1.5 ml per minute,
Test solution. Reflux 2 g ofcoarsely powdered substance under - spectrophotometer set at 230 nm,
examination with 50 ml methanol on a boiling water-bath for - injection volume. 20 Ill.
30 minutes, cool and filter. Reflux the residue further with 2 x
Inject the reference solution (a) and (b). The test is not valid
50 m1 of methanol, cool and filter. Combine all the filtrates and
unless the relative standard deviation for the replicate
concentrate under vacuum to 10 ml.
injections for both the analytepeaks corresponding to
Reference solution;--Reflux: -l-gof-bhuiamla-RS-with- 50-m1 phyllanthin and·hypophyllanthin--is not-more than-2.0-per
methanol on a boiling water-bath for 30 minutes, cool and cent.
filter. Reflux the residue further with 2 x 50 ml of methanol,
Inject the test solution, reference solutions (a) and (b).
cool and filter. Combine all the filtrates and concentrate under
vacuum to 5 mL Calculate the contents of phyllanthin and hypophyllanthin.

2488

www.webofpharma.com
IP 2010 BRAHMI

Storage. Store protected from heat, moisture and against attack cool and filter. Combine all the filtrates and concentrate under
by insects and rodents. vacuum to 25 ml.
Reference solution. Reflux 0.4 g ofcoarsely powdered brahmi
RS with 5 ml methanol for 15 minutes, cool and filter.
Brahmi
Apply to the plate 10 fll of each solution as bands 10 mm by 2
Bacopa monnieri mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with methanolic sulphuric acid (20 per cent v/v). Heat the
plate at 100° for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.
D. In the Assay, the chromatogram obtained with test solution
corresponds to the chromatogram obtained with reference
solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 22 per cent by
Method l.
Total ash (2.3.19). Not more than 18 percent.
Brahmi consists of the dried whole plant, preferably leaves
and stem of Bacopa monnieri (Linn.) Pennell (Fam. Acid-insoluble ash (2.3.19). Not more than 6.0 per cent.
Scrophulariaceae). Heavy metals (2.3.13). 1.0 g complies with the limit test for
Brahmi contains not less than 2.5 per cent w/w ofbacoside A, heavy metals, Method B (20 ppm).
calculated on the dried basis. Loss on drying (2.4.19). Not more than 12.0 per cent,
Description. A brown to reddish brown colour when determined on 5 g by drying in an oven at 105°.
completely dried or green colour when partially dried with Microbial contamination (2.2.9). Complies with the microbial
slightly bitter taste. contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Identification
Test solution. Reflux about 2 g of the coarsely powdered
A. Macroscopic Herbaceous comprising ofstems, runner substance under examination with 50 ml of methanol on.a
stems and leaves. Stems glabrous, leafless towards the base; water bath for 15 minutes, cool and filter. Reflux the residue
internodes long. Leaves spatulate-obovate, sessile, and further with methanol till the last extract turns colorless, cool
glabrous. and filter. Combine all the filtrates and concentrate to 100.0 ml.
B. Microscopic - Cortex in stem composed ofparenchyma Reference solution. A 0.2 per cent w/v solution of bacoside A
cells enclosing large air spaces; xylem vessels radially arranged RS in methanol, prepared by heating gently on a water bath.
xylem rays uniseriate; pith parenchyma collapsed. In leaf,
Chromatographic system
midrib indistinct, mesophyll isobilateral ofspongy cells, a few a stainless steel column 25 cm x 4.6 rum packed with
prismatic crystals of calcium oxalate in mesophyll; stomata octadecylsilane bonded to porous silica (5 flm),
anomocytic on both the surfaces of leaf. mobile phase: A.a gradient mixtures ofacetonitrile and
C. Determine by thin-layer chromatography (2.4.17), coating a buffer solution prepared by dissolving 0.5 g
the plate with silica gel GF254. phosphoric acid in 800 ml of water, adjust pH to 2.8
with dilute phosphoric acid and dilute to 1000 ml with
Mobile phase. A mixture of 70 volumes of chloroform and
water,
30 volumes of methanol.
B. acetonitrile,
Test solution. Reflux 2 g ofcoarsely powdered substance under flow rate. 1.5 ml per minute,
examination with 25 ml methanol for 15 minutes, cool and spectrophotometer set at 205 nm,
filter. Reflux the residue further with 2 x 25 ml of methanol, injection volume. 20 fll.

2489

www.webofpharma.com
BRAHMI EXTRACT IP 2010

Time mobile phase A mobile phase B Tests


(min) ( per cent v/v) ( per cent v/v)
Loss ofdrying (2.4.19). Not more than 5.0 per cent, determined
o 70 30
on 2.0 g by drying in an oven at 105° for 3 hours.
25 60 40
Total ash (2.3.19). Not more than 5 percent.
35 40 60
36 70 30 Heavy metals (2.3.13). 1.0 g complies with the limit test for
45 70 30 heavy metals, (Method B) 20 ppm.
Microbial contamination (2.2.9). Complies with the microbial
Inject the reference solution. The relative retention times are
contamination test.
1.00 for bacoside A3, about 1.04 for bacopaside II, 1.13 for
jujubogenin isomer of bacopasaponine C and 1.19 for Assay. Determine by liquid chromatography (2.4.14).
bacopasaponine C as bacoside A. The test is not valid unless Test solution. Dissolve about 0.5 g of the extract in methanol
the relative standard deviation for the replicate injections is by gentle heating, dilute to 100 ml and filter.
not more than 2.0 per cent.
Reference solution. A 0.1 per cent w/v solution of bacoside-
Inject the test solution and reference solution. A RS in methanol.
Calculate the content of bacoside A. Chromatographic system
Storage. Store protected from heat, moisture and against attack a stainless steel column 25 cm x 4.60 mm packed with
by insects and rodents. octadecylsilane bonded to porous silica (5 /lm),
- mobile phase: A. 0.5 g ofphosphoric acid in water,
B. Acetonitrile
- a Hnear gradient programme using the conditions given
Brahmi Extract below,
- flow rate 1.5 ml per minute,
Brahmi Extract is obtained by extracting Brahmi (Bacopa
- spectrophotometer set at 205 llill,
monnieri, Fam. Plantaginaceae) from the dried leaves and stems
of with aqueous ethanol or any other suitable solvent. - injection volume.20 /ll.
Time mobile phase A mobile phase B
Brahmi Extract contains not less than 90.0 per cent w/w and
(in min) (per cent v/v) (per cent v/v)
not more than 120.0 percent w/w of bacoside-A (sum of
bacoside-AJ , bacopaside-II, bacopasaponin-C, jujubogenin
o 70 30
isomer ofbacopasaponin-C). 25 60 40
35 40 60
Description. Light green to dark green powder with
characteristic odour and bitter taste. 36 70 30
45 70 30
Identification Inject the reference solution. The test is not valid unless the
A. Determine by thin-layer chromatography (2.4.17), coating relative standard deviation for replicate injections is not more
the plate with silica gel GF 254. than 2.0 per cent.The relative retention time with refemce to
bacoside-A J for Bacopaside II is about 1.04, for jujubogenin
Mobile phase. A mixture of 7 volumes of Ethyl acetate, 2 isomer of bacopasaponin C is about 1.13 and for
volumes of methanol and 1 volume of water. bacopasaponin C is about 1.19.
Test solution. Shakewell 0.5 g ofthe extract under examination Inject the reference solution and the test solution.
with 50 ml methanol and filter.
Calculate the content of bacoside-A.
Reference solution. A 0.1 per cent w/v solution of bacoside-
Usual strengths. 10 per cent w/w; 20 per cent w/w.
A RS in methanol.
Storage. Store protected from heat and moisture.
Apply to the plate 10 /ll of each solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
andexaminein_ultravioletlight_at_25Anm.Spraythe_plate with
vanillin sulphuric acid sQJJJt!QI.l..l:Ie!3JJlle plate at 60° t070°
Castor en
for 10 minutes and examine the plate at 365 nm and in day Castor Oil is the fixed oil obtained by cold expression from the
light. The chromatographic profile ofthe test solution is similar seeds of Ricinus communis Linn. (Fam. Euphorbiaceae). It
to that of the reference solution. may contain suitable antioxidants.

2490

www.webofpharma.com
IP 2010 CLOVE OIL

Description. A pale yellowish or almost colourless, Apply to the plate 20 III of the test solution and 10 III of the
transparent, viscid liquid; odour, slight and characteristic. reference solution as bands 20 mm by 3 mm. Use an unlined
tanle, develop the chromatogram immediately after pouring
Tests the mobile phase into the tanle and allow the mobile phase to
Light absorption (2.4.7). Absorbance of a 1.0 per cent w/v rise 10 cm. Dry the plate, allow to stand for 5 minutes and
solution in ethanol (95 per cent) at the maximum at about 269 again allow the mobile phase to rise 10 cm under the same
nm, not more than 1.0. conditions. Following the second development, dry the plate
in air, examine in ultraviolet light at 254 nm and mark the
Weight per ml (2.4.29).0.945 g to 0.965 g.
quenching bands. In the chromatogram obtained with the test
Refractive index (2.4.27). 1.4758 to 1.4798. solution there is a quenching band in the middle of the plate
Optical rotation (2.4.22). +3.5°to +6.00 • corresponding to the quenching band due to eugenol in the
Peroxide value (2.3.35). Not more than 5.0. chromatogram obtained with the reference solution. A weak
quenching band may also be seen in the chromatogram
Acid value (2.3.23). Not more than 2.0.
obtained with the test solution with an Rfvalue slightly lower
Acetyl value (2.3.22). Not less than 143. than that of the band corresponding to eugenol
Hydroxyl value (2.3.27). Not less than 150. (acetyleugenol). Spray the plate with about 10 ml of
Saponification value (2.3.37).176 to 187. anisaldehyde solution, heat at 1000 to 105 0 for 10 minutes and
examine in daylight. In the chromatogram obtained with the
Iodine value (2.3.28).82 to 90.
test and reference solutions the bands corresponding to
Foreign fatty substances. A mixture of2 ml ofthe substance eugenol are strongly coloured brownish-violet and any band
under examination and 8 ml of ethanol (95 per cent) is clear. corresponding to acetyleugenol in the chromatogram obtained
B. Shake 10.0 ml with 20.0 ml of light petroleum (60 0 to 800) with the test solution is faintly violet-blue. Other coloured
and allow to separate; the volume ofthe lower layer is not less bands may be visible in the chromatogram obtained with the
than 16.0 ml. test solution, in particular a faint red band in the lower part of
Storage. Store protected from light and moisture at a the chromatogram and a reddish-violet band in the upper part
temperature not exceeding 15 0 • (caryophyllene).
Labelling. The label states (1) the name and quantity of any Tests
added antioxidant; (2) whether the contents are suitable for
use in the manufacture of parenteral preparations. Optical rotation (2.4.22). 00 to -1.50°.
Weightperml(2.4.29).1.038gto 1.060g.
Refractive index (2.4.27). 1.527 to 1.535, determined at 200 •
Clove Oil
Heavy metals (2.3.13). 0.5 g complies with the limit test for
Clove oil is the oil distilled from the dried flower buds of heavy metals, Method B (40 ppm).
Syzygium aromaticum (Linn.) Merrill and Perry Eugenia
caryophyllus (Spreng.) Bull. and Harr.(Fam. Myrtaceae). Phenol. Shake 1 ml with 20 ml ofhot water; the mixture shows
not more than a scarcely perceptible acid reaction with blue
Description. A clear, colourless or pale yellow liquid when litmus paper. Cool the mixture, pass the aqueous layer through
freshly distilled, becoming darker and thicker by ageing or a wetted filter and treat the clear filtrate with 1 drop offerric
exposure to air; odour as of clove. chloride test solution. The mixture has only a transient greyish-
Clove Oil contains not less than 85.0 per cent w/w and not green colour but not a blue or violet colour.
more than 95.0 per cent w/w of phenolic substances, chiefly Alkali-soluble matter. Place 80 ml ofa 5 per cent w/v solution
eugenol, C IOH I20 2 • of potassium hydroxide in a 150-ml flask with a long neck
which is graduated in tenths of a ml and is ofsuch a diameter
Identification that not less than 15 em in length has a capacity of 10 ml.
Determine by thin-layer chromatography (2.4.17), coating the Clean the flask with sulphuric acid and rinse well with water
plate with silica gel GF254. before use. Add 10 ml of the oil and shake thoroughly at
5 minute intervals for 30 minutes at ambient temperature. Raise
Mobile phase. Toluene
the undissolved portion of the oil into the graduated part of
Test solution. Dissolve 20 III of the substance under the neck of the flask by the gradual addition of more of the
examination in 2 ml oftoluene. potassium hydroxide solution; allow to stand for not less
Reference solution. Dissolve 20 III of eugenol RS in2 ml of than 24 hours and read off the volume of the undissolved
toluene. portion ofthe oil which measures between 1.0 and 1.5 ml.

2491

www.webofpharma.com
CLOVE OIL IP 2010

Assay. Determine by gas chromatography (2.4.14). Identification


Test solution (aj: A 0.2 per cent w/v solutiOlj ofthe oil under A. It complies with the test for melting range (2.4.21).
examination in ethanol (95 per cent).
B. It complies with the test for composition offatty acids.
Test solution (b). A 0.2 per cent w/v solution of the oil under
examination and 0.15 w/v of I-decanol (internal standard) in Tests
ethanol (95 per cent). Melting range (2.4.21). 23 ° to 26°.
Reference solution. A solution containing 0.2 per cent w/v Refractive index (2.4.27). About 1.449, at 400.
solution of eugenol RS and 0.15 per cent w/v of the internal Peroxide value (2.3.35). Not more than 5.0.
standard in ethanol (95 per cent). Acid value (2.3.23). Not more than 0.5, determined on 20.0 g
Chromatographic system Unsaponifiable matter (2.3.39). Not more than 1.0 per cent,
a glass column 1.5 m x 4 mm, packed with 3 per cent w/w determined on 5.0 g.
ofdimethyl silicone fluid on acid-washed diatomaceous Iodine value (2.3.28).82 to 90.
.support (120 mesh),
Composition offatty acids
- temperature:
column. 11 0° for 18 minutes, then increased to 170° at a Test Solution. Refined coconut oil is melted under gentle
rate of 12° per minute and maintained at this temperature heating to a homogeneous liquid.
for 2 minutes, Reference solution. Dissolve 15 mg of tricaproin RS, 80 mg of
inlet port at 220° and tristearin RS, 0.15 g of tricaprin RS, 0.2 g of tricaprylin RS,
detector at 300°, 0.45 g of trimyristin RS and 1.25 g of trilaurin RS in a mixture
- flow rate 40 ml per minute ofthe carrier gas. of2 volumes of dichloromethane and 8 volumes of heptane,
then dilute to 50 ml with the same mixture of solvents heat at
Calculate the eugenol content in the oil under examination
45°to 500. Transfer 2 ml ofthis mixture to a 10 ml centrifuge
using the ratios of the area of the peak corresponding to
tube with a screw cap and evaporate the solvent in a current
eugenol to the area ofthe peak due to the internal standard in
of nitrogen. Dissolve with 1 ml of heptane and 1 ml of dimethyl
the chromatogram obtained with test solutions (b) and the
carbonate and mix vigorously under gentle heating (500 to
reference solution.
60 0). Add, while still warm, I ml ofa 1.2 per cent w/v solution
Storage. Store protected from light in well-filled containers at . of sodium in anhydrous methanol, prepared with the
a temperature not exceeding 30°. necessary precautions, and mix vigorously for about 5 minutes.
Add 3 ml of distilled water and mix vigorously for about 30
seconds. Centrifuge for 15 minutes at 1500 rpm. Inject 1 III of
the organic phase.
Coconut Oil Storage. Store protectedfrom light in well-filled containers.
Coconut Oil is the refined fixed oil obtained from the dried,
solid part of the endosperm of Cocos nucifera L.
(Fam.Arecaceae). Coleus
Coconut Oil contains not less than 1.5 per cent w/w ofcaproic Coleus forskohlii
acid, not less than 5.0 percent and not more than 1l.0 per cent
w/w of caprylic acid, not less than 4.0 per cent and not more
9
than 9.0 per cent w/w ofcapric acid, not less than 40.0 per cent
and not more than 50.0 per cent w/w of lauric acid, not less 8

than 15.0 per cent and not more than 20.0 per cent w/w of 7
myristic acid, not less than 7.0 percent and not more than 12.0 6
per cent w/w of palmitic acid, not less than 1.5 per cent and
5
not more than 5.0 per cent w/w ofstearic acid, not less than 4.0
per cent and not more than 10.0 w/w per cent ofoleic acid, not 4
less than l.0 per cent and not more than 3.0 per cent w/w of 3
lin:(j1eicacid;~n:(jrless··than··0:2p ercent w/Wof·lin51enicacid;
2
not less than 0.2 per cent w/wof arachidic acid and not less
than 0.2 per cent w/w of eicosenoic acid. 1

Description. A white or almost white, unctuous mass.


o

2492

www.webofpharma.com
IP 2010 COLEUS DRY EXTRACT

Coleus consists of the whole or cut dried roots of Coleus Loss on drying (2.4.19). Not more than 12.0 per cent,
forskohlii Briq. (Fam. Lamiaceae). determined on 5 g by drying in an oven at 105°.
Coleus contains not less than 0.4 per cent w/w of forskolin, Microbial contamination (2.2.9). Complies with the microbial
calculated on the dried basis. contamination tests.
Description. The roots are light brown in color, generally long Assay. Determine by liquid chromatography (2.4.14).
and radially spread. They have an aromatic characteristic odor Test solution. Weigh 3 g of coarsely powdered substance
and the taste is slightly pungent. under examination, add 50 ml of acetonitrile and reflux on a
water bath for 15 minutes, cool and filter. Reflux the residue
Identification
two times with 75 ml of acetonitrile, cool and filter, Concentrate
A. Macroscopic - Roots are brown, longitudinally wrinlded, the filterate to 100.0 ml.
fracture short, cut surface yellowish white. Reference solution. A 0.1 per cent w/v solution offorskolin
B. Microscopic - The outermost layer consists ofrectangular RS in acetonitrile.
cork cells, cork cambium, rectangular parenchymatous region Chromatographic system
containing sclereids and calcium oxalate crystals. Vascular a stainless steel column 25 cm x 4.6 rom packed with
cambium is present in the form of a continuous ring. The octadecylsilane bonded to porous silica (5 /lm),
tracheids and tracheidal fibres have bordered pits. mobile phase: filtered and degassed mixture of 45
C. Determine by thin-layer chromatography (2.4.17), coating volumes of acetonitrile and 55 volumes of water,
the plate with silica gel GF254. flow rate. 1.8 ml per minute,
spectrophotometer set at 220 nm,
Mobile phase. A mixture of 75 volumes of benzene, and
injection volume. 20 /ll.
25 volumes of ethyl acetate.
Inject the reference solution. The relative standard deviation
Test solution. To· 5 g of the coarsely powdered substance
for the replicate injections is not more than 2.0 per cent.
under examination, add 50 ml of acetonitrile and reflux for
15 minutes, cool and filter. Reflux the residue further for two Inject the test solution and reference solution.
times with 50 ml of acetonitrile, cool and filter. Combine all the Calculate the content offorskolin.
filtrates and concentrate under vacuum to 100 ml.
Storage. Store protected from moisture and against attack by
Reference solution. To 1 g of coleus RS add 50 ml of insects and rodents.
acetonitrile and reflux for 15 minutes, cool and filter. Reflux
the residue further for two times with 50 ml of acetonitrile,
cool and filter. Combine all the filtrates and concentrate under
vacuum to 20 mi.
Coleus Dry Extract
Apply to the plate 20 /ll ofeach solution as bands 10 rom by 2 Coleus Dry Extract is obtained by extracting Coleus (Coleus
rom. Allow the mobile phase to rise 8 cm. Dry the plate in air Forskohlii Willd. Briq.,Fam. Lamiaceae) roots with methanol
and examine in ultraviolet light at 254 nm and 365 nm, spray or any other suitable solvent and evaporation of solvent.
with vanillin glaCial acetic acid reagent. Heat the plate at Coleus Dry Extract contains not less than 90 per cent w/w to
100° for 5-10 minutes and examine in day light. The not more than 120 per cent w/w of the stated amount of
chromatographic profile of the test solution is similar to that forskolin, calculated on the dried basis. It may contain suitable
of the reference solution. added substances.

Tests Description. A light brown to brown colour powder.

Foreign organic matter (2.6.1). Not more than 2.0 per cent. Identification
Ethanol-soluble extractive (2.6.2). Not less than 15.0 per cent. A. Detennine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Water-soluble extractive (2.6.3). Not less than 18.0 per cent
by method I. Mobile phase. A mixture of40 volumes of ethyl acetate and 60
volumes of hexane.
Total ash (2.3.19). Not more than 15.0 percent.
Test solution. Dissolve about 0.5 g of the extract under
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent. examination with 10 ml methanol, filter.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Reference solution. A 0.1 per cent w/v solution offorskolin
heavy metals, Method B (20 ppm). RS in methanol.

2493

www.webofpharma.com
COLEUS DRY EXTRACT IP 2010

Apply to the plate 10 J!l of each solution as bands 10 rom by not less than 0.5 per cent and not more than 4.0 per cent ofp-
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air cymene, not less than 3.0 per cent and not more than 6.0 per
and spray the plate with a anisaldehyde-sulphuric acid cent of camphor, not less than 0.1 per cent and not more than
reagent. Heat the plate at 110° for 10 minutes and examine in 1.5 per cent of a-terpineol, not less than 0.5 per cent and not
ultraviolet light at 365 urn and in day light. The chromatographic more than 4.0 per cent ofgeranyl acetate and not less than 0.5
profile of the test solution is similar to that of the reference per cent and not more than 3.0 per cent of geraniol.
solution.
Description. A clear, colourless or pale yellow liquid;
Tests characteristic and spicy odour .

Totalash (2.3.19). Not more than 10.0 per cent. Identification


Heavy metals (2.3.13). 1.0 g complies with the limit test for Determine by thin-layer chromatography (2.4.17), coating the
heavy metals, Method B (20 ppm). plate with silica gel GF 254.
Loss on drying (2.4.19). Not more than 6.0 per cent, determined Mobile phase. A mixture of 95 volumes of toluene and 5
on 1 g by drying in an oven at 105°. volumes of ethyl acetate.
Microbial contamination (2.2.9). Complies with the microbial Test solution. Dissolve 10 J!l of the substance under
contamination tests. examination in 1 mloftoluene.
Assay. Determine by liquid chromatography (2.4.14). Reference solution. Dissolve 10 J!l of linalol and 2 J!l of
Test solution. Dissolve a quantity of the extract under geranyl acetate in Iml of toluene.
examination containing about 50 mg offorskolin in 50.0 ml of Apply to the plate 10 J!l ofeach solution as bands of 10 rom by
methanol and filter. 2 rom. Allow the mobile phase to rise 10 cm. Dry the plate in air,
Reference solution. A 0.1 per cent w/v solution offorskolin spray with anisaldehyde sulphuric acid reagent. Heat the
RS in the methanol. plate at 100° to 105° for 10 to 15 minutes and examine the plate
in day light. The chromatographic profile of the test solution
Chromatographic system
is similar to that ofthe reference solution.
- a stainless steel column, 25 cm x 0.46 mm packed with
octadecylsilane bonded to porous silica (5J!m), Tests
- mobile phase: a mixture of50 volumes of water and 50
volumes of acetonitrile, Relative density (2.4.29.).0.860 to 0.880.
flow rate. 1 ml per minute, Refractive index (2.4.27). 1.462 to 1.470.
spectrophotometer set at 210 urn, Optical rotation (2.4.22). + 7° to + 13°.
i~ection volume. 20 J!l.
Acid value (2.3.23). Not more than 3.0.
Inject the reference solution. The test is not valid unless the
Enantiomeric purity. Not more than 14 per cent w/w of-{R) -
relative standard deviation for replicate injections is not more
linalol.
than 2.0 per cent.
Determine by gas chromatography (2.4.13).
Inject the test solution and the reference solution.
Test solution. Dissolve 0.02 g of the substance under
Calculate the content of the forskolin in the extract. examination in 10 ml ofpentane.
Usual strengths. 10 per cent w/w; 20 per cent w/w. Reference solution. Dissolve 10 J!l of linalol and 5 mg of
Storage. Store protected from heat and moisture. borneol in 10 ml ofpentane.
Chromatographic system
- a fused silica column 25 m x 0.25 mm, packed with modified
a-cyclodextrin (0.25 /lm),
Coriander Oil
- temperature:
Coriander Oil is obtained by steam distillation from the fruits column. 50° for 5 minutes, then increased to 180° at a
of Coriandrum sativum L.(Fam.Apiaceae). rate of 12° per minute and maintained at this temperature
for 65 minutes,
CorianderDiLcontainsnotJess_than..65.0_peLcenLandnoL
'iiiletport andCletectof at 1J(}0~--'~"'" .~._-~--~_....
more.tl111n 78.0 perc~llt.9Jljna1o],llQU~.~~tl111n 3.Qp~I cellt
- flowraten-mlpefmiiiliteoftlfecamer·gas:···· ....··
and not more than 7.0 per cent of ,*pinene, not less than 1.5
per cent and not more than 5.0 per cent of limonene, not less Inject I /ll of the reference solution. The test is not valid
than 1.5 per cent and not more than 8.0 per cent of 'Y -terpinene, unless the resolution between the peaks due to (R)-linalol

2494

www.webofpharma.com
IP 2010 DARUHARIDRA ROOTS

and (S) linalol is not less than 5.5 and the resolution between Description. The roots are cylindrical, yellowish brown in
the peaks due to (S)-linalol and borneol is not less than 2.9. colour. They have a slightly characteristic odour and the taste
Inject 1 fll of the reference solution and the test solution. is bitter.
Assay. Determine by gas chromatography (2.4.13). Identification
Test solution. The substance under examination dissolve in I
A. Macroscopic - Root cylindrical, more or less knotty,
ml ofhexane.
strongly branched, usually cut into pieces of varying length
Reference solution (a). Dissolve 10 fll of a-pinene, 10 fll of and up to 45 mm in diameter; externally light yellowish-brown,
limonene, 10 fll of a-terpinene, 10 fll ofp-cymene, 10 mg of longitudinally wrinlded and short scaly; fracture hard and
camphor, 20 fll oflinalol, 10 fll ofa-terpineol, 10 fll ofgeranyl tough; bark 1 mm. in thickness, easily separable into layers;
acetate and 10 fll ofgeraniol in 1 ml of hexane. wood yellow.
Reference solution (b). Dissolve 5 fll of geraniol in hexane
B. Microscopic - The powder is yellowish-brown; composed
and dilute to 10 ml with the same solvent.
chiefly of fragments of wood fibers associated with a few
Chromatographic system tracheae and medullary rays; wood fibers yellowish, scarcely
- a fused silica column 60 m x 0.25 mm, packed with giving any reaction with phloroglucinol and hydrochloric
macrogol20000 (0.25 flm), acid, and with large, simple, transverse pores; trachea chiefly
- temperature: scalariform with bordered pits, occasionally reticulate;
column. 60 0 for 10 minutes, then increased to 1900 at a medullary rays one to twelve cells wide, and in very .long
rate of 12 0 per minute and maintained at this temperature rows; starch grains simple or two- to three-compound, the
for two minutes, individual grains being irregularly spherical.
- inlet port at 220 0 and detector at 240 0 ,
C. Determine by thin-layer chromatography (2.4.17), coating
- flow rate 1ml per minute ofthe carrier gas.
the plate with silica gel GF 254.
Inject 1 fll of the reference solution: The test is not valid
Mobile phase. A mixture of 80 volumes of ethyl acetate, 10
unless the resolution between the peaks due to linalol and
volumes offormic acid, 10 volumes of glacial acetic acid
camphor is not less than 1.5.
and 20 volumes of water.
Inject 1 fll of the refernce solution and the test solution.
Test solution. To 2 g of the coarsely powdered substance
Storage. Store protected from light, at a temperature not under examination, add 40 mI ofmethanol, reflux for 15 minutes,
exceeding 300 • cool and filter. Reflux the residue further for two times with 25
ml of methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 25 ml.
Daruharidra Roots Reference solution. To 1 g of the daruharidra roots RS, add
Berberis, Berberis arisata 40 mI ofmethanol, reflux for 15 minutes, cool and filter. Reflux
the residue further for two times with 25 ml ofmethanol, cool
and filter. Combine all the filtrates and concentrate under
9 vacuum to 25 mI.
8 Apply to the plate 10 fll of each solution as bands 10 mm by 2
7 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
6
and examine in ultraviolet light at 254 urn and 365 nm and also
under day light The chromatographic profile of the test
5
solution is similar to that ofthe reference solution.
4
Tests
3

2
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent.
1
Water-soluble extractive (2.6.3). Not less than 6.0 per cent by
o methodL .
Daruharidra Roots consist of the dried roots of Berberis Total ash (2.3.19). Not more than 5.0 percent.
aristata DC (Fam. Berberidaceae). Acid-insoluble ash (2.3.19). Not more than 1.0 per cent.
Daruharidra Roots contain not less than 0.70 per cent w/w of Heavy metals (2.3.13). 1.0 g complies with the limit test for
berberine, calculated on the dried basis. heavy metals, Method B (60 ppm).

2495

www.webofpharma.com
DARUHARIDRA ROOTS IP 2010

Loss on drying (2.4.19). Not more than 10.0 per centdetennined solution of strong ammonia solution in methanol for 30
on 5.0 g by drying in an oven at 105°. minutes, cool and filter. Boil under reflux the residue with
Microbial contamination (2.2.9). Complies with the microbial further 2 quantities, each of30 mI, of 0.1 per cent v/v strong
contamination tests. . ammonia solution in methanol, cool and filter. Combine all
the filtrates and dilute to 100 m1 with 0.1 per cent v/v solution
Assay. Perform the Assay by following method I or by ofstrong ammonia solution in methanol. Dilute 5.0 ml ofthis
method I!. solution to 50 ml with a 0.1 per cent v/v strong ammonia
MethodI solution in methanol.
Determine by liquid chromatography (2.4.14). Reference solution. A 0.002 per cent w/v solution of berberine
hydrochloride RS in 0.1 per cent v/v strong ammonia solution
Test solution. Weigh 2 g ofthe coarsely powdered substance
in methanol.
under examination, add about 20 ml 2 M hydrochloric acid
and 30 ml of methanol, reflux on a water-bath for 15 minutes, Chromatographic system
cool and filter. Reflux the residue further with 2 Mhydrochloric a stainless steel column 15 cm x 4.6 mm, packed with
acid and methanol, till the extract turns colourless, cool and octadecylsilane chemically bonded to porous silica
filter. Combine all the filtrates and concentrate to a volume (5/lm),
slightly less than 50 ml. Dilute to 50.0 mI with methanol. mobile phase: a mixture of73 volumes of 1.36 per cent
w/v solution of potassium dihydrogen orthophosphate
Reference solution. A 0.01 per cent w/v solution of berberine
hydrochloride RS in methanol. in water and 27 volumes of acetonitrile,
flow rate. 1.2 ml per minute,
Chromatographic system spectrophotometer set at 235 nm,
a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 /lm), injection volume. 10 /ll.
mobile phase: A. a buffer solution pH 2.5 prepared by Inject the reference solution. The test is not valid unless the
dissolving 0.136 g of potassium dihydrogen relative standard deviation for replicate injections is not more
orthophosphate in 500 ml of water, add 0.5 ml of than 2.0 per cent.
orthophosphoric acid and make upto 1000 ml with Inject the test solution and the reference solution.
water, Calculate the content of berberine.
B. acetonitrile,
flow rate. 1 ml per minute, 1 mg ofberberine hydrochloride is equivalent to 0.9045 mg of
a linear gradient programme using the conditions given berberine.
below, Storage. Store protected from heat, moisture and against attack
spectrophotometer set at 346 nm, of insects and rodents.
injection volume. 20 /ll.
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent v/v)
Darubaridra Stems
o 80 20
25 50 50 Berberis; Berberis aristata
26 80 20
30 80 20
9
Inject the reference solution. The test is not valid unless the
relative standard deviation for the replicate injections is not 8
more than 2.0 per cent. 7

Inject the test solution and the reference solution. 6


Calculate the content of berberine. 5
1 mg of berberine hydrochloride is equivalent to 0.9045 mg of 4
berberine.
3
MethodU . ----------.-. 1=::::J "'=C7--, - ,-'----,~J"':,:'c.=:~
2
DeteriiiilieoyliqUid cbf6mat6jifapliY(2:4.T4:). ... -
Test solution. Boil under reflux 1.0 g ofthe coarsely powdered
o
substance under examination with 40 m1 of a 0.1 per cent v/v

2496

www.webofpharma.com
IP 2010 DARUHARIDRA STEMS

Daruharidra Stems consist of cut dried stems of Berberis Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent.
aristata, DC (Fam. Berberidaceae). Water-soluble extractive (2.6.3). Not less than 5 per cent by
Daruharidra Stems contain not less than 0.5 per cent w/w of Method 1.
berberine, calculated on the dried basis. Total ash (2.3.19). Not more than 5 per cent.
Description. Yellowish-brown, cut chips of varying sizes, Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
fracture fibrous, and taste bitter.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Identification heavy metals, Method B (60 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent deterrnined
A. Macroscopic - Cut tortuous pieces of varying length and
on 5 g by drying in an oven at 105°.
thickness. Bark thick, yellowish to brownish grey, wrinkled
irregularly, deeply furrowed; surface hard, rough, fracture short Microbial contamination (2.2.9). Complies with the microbial
and fibrous. contamination tests.
B. Microscopic - Cork consists of rectangular to squarish Assay. Perform the Assay by following method I or by method
yellow coloured thin walled cells, radially arranged; sieve ll.
elements irregularly shaped, few cells containing yellowish Method I
brown to orange brown masses; phloem fibers thick walled
arranged tangentially; phloem ray cells have calcium oxalate Determine by liquid chromatography (2.4.14).
crystals, scattered stone cells; secondary phloem broad, sieve Test solution. Boil under reflux 1.0 g ofthe coarsely powdered
elements arranged as tangential bands, phloem fibers short, substance under examination with 40 ml of a 0.1 per cent v/v
thick walled; secondary xylem broad, consisting of small to solution of strong ammonia solution in methanol for 30
medium sized xylem vessels, tracheids, xylem fibers. minutes, cool and filter. Boil under reflux the residue with
Powder is greenish yellow and shows the following diagnostic further 2 quantities, each 000 ml, of 0.1 per cent v/v strong
characters when observed under a microscope using chloral ammonia solution in methanol, cool and filter. Combine all
hydrate solution. Abundant thin walled parenchyma, the filtrates and dilute to 100 ml with 0.1 per cent v/v solution
fragments ofyellowish-brown cork; ovoid or spherical orange of strong ammonia solution in methanol. Dilute 5.0 m1 ofthis
brown granular masses. Examine under a microscope using solution to 50 ml with a 0.1 per cent v/v strong ammonia
50 per cent v/v solution ofglycerol. The powder shows starch solution in methanol.
granules, simple, small, spherical to ovoid. Reference solution. A 0.002 per cent w/v solution of berberine
C. Determine by thin-layer chromatography (2.4.17) coating hydrochloride RS in 0.1 per cent v/v strong ammonia solution
the plate with silica gel GF254. in methanol.

Mobile phase. A mixture of 70 volumes of n-butanol, 10 Chromatographic system


volumes of glacial acetic acid and 20 volumes of water. a stainless steel column 15 em x 4.6 mm, packed with
octadecylsilane chemically bonded to porous silica
Test solution. Shake 1 g of powdered substance under
(5Ilm),
examination with 10 ml methanol for 10 to 15 minutes and mobile phase: a mixture of73 volumes of 1.36 per cent
filter. Wash the residue with 5 ml of methanol and add washing w/v solution of potassium dihydrogen orthophosphate
to the filtrate.
in water and 27 volumes of acetonitrile,
Reference solution. Shake 1 g ofpowdered daruharidra stem flow rate. 1.2 m1 per minute,
RS with 10 ml of methanol for 10-15 minutes and filter. Wash spectrophotometer set at 235 urn,
the residue with 5 ml of methanol and add washing to the injection volume. 10 Ill.
filtrate.
Methodll
Apply to the plate, 10 III of each solution as bands of 10 mm Determine by liquid chromatography (2.4.14.).
by 2 mm. Allow the mobile phase to rise 8 em. Dry the plate in
air and observe in ultraviolet light at 254 urn and 365 urn and :Test solution. Weigh 2 g ofthe coarsely powdered substance
also under day light. The chromatographic profile of the test under examination, add about 20 ml 2 M hydrochloric acid
solution is similar to that of reference solution. and 30 ml of methanol, reflux on a water-bath for 15 minutes,
cool and filter. Reflux the residue further with 2 M hydrochloric
Tests acid and methanol, till the extract turns colourless, cool and
filter. Combine all the filtrates and concentrate to a volume
Foreign organic matter (2.6.1). Not more than 2.0 per cent. slightly less than 50 ml. Dilute to 50.0 m1 with methanol.

2497

www.webofpharma.com
DARUHARIDRA STEMS IP 2010

Reference solution. A 0.01 per cent w/v solution of berberine Eucalyptus Oil contains not less than 60 per cent w/w of
hydrochloride RS in methanol. cineole, C IOH 180.
Chromatographic system Description. A colourless or pale yellow liquid; odour, aromatic
- a stainless steel column 25 cm x 4.6 mm, packed with and camphoraceous; taste, pungent and camphoraceous,
octadecylsilane bonded to porous silica (5 Ilm), followed by a sensation of cold.
mobile phase: A. a buffer solution pH 2.5 prepared by
dissolving 0.136 g of potassium dihydrogen Identification
orthophosphate in 500 ml of water, add 0.5 ml of
orthophosphoric acid and make upto 1000 ml with Determine by thin-layer chromatography (2.4.17), coating the
water, plate with silica gel G.
B. acetonitrile, Mobile phase. A mixture of 90 volumes of toluene and
- flow rate. 1 ml per minute, 10 volumes of ethyl acetate.
- a linear gradient programme using the conditions given
Test solution. Dissolve 1 g ofthe substance under examination
below,
in 100 ml of toluene.
- spectrophotometer set at 346 Dill,
- injection volume. 20 Ill. Reference solution. A 1 per cent w/v solution of cineole RS in
Time Mobile phase A Mobile phase B toluene.
(in min.) (per cent v/v) (per cent v/v) Apply to the plate 2 III of each solution. After development,
o 80 20 dry the plate in air, spray with anisaldehyde solution, using
25 50 50 about 10 ml for a 200 mm x 200 mm plate, heat at 105° for
10 minutes and examine in daylight and in ultraviolet light at
26 80 20
365 nm. In the chromatogram obtained with the reference
30 80 20 solution a dark brown spot due to cineole is visible in daylight
Inject the reference solution. The test is not valid unless the in the middle part; when examined in ultraviolet light at
relative standard deviation for the replicate injections is not 365 nm, the spot shows a brown fluorescence. The principal
more than 2.0 per cent. spot in the chromatogram obtained with the test solution
corresponds to that ofcineole; no carmine-brown spot appears
Inject the test solution and the reference solution.
in daylight in the upper third of the chromatogram and when
Calculate the content of berberine. examined in ultraviolet light at 365 nm no spot showing a
1 mg of berberine hydrochloride is equivalent to 0.9045 mg of greenish brown fluorescence appears in the upper third
berberine. (citronellal). Other spots may be visible in the upper and lower
thirds of the chromatogram.
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
than 2.0 per cent. Tests
Inject the test solution and the reference solution. Optical rotation (2.4.22). 0° to +10°.
Calculate the content of berberine. Refractive index (2.4.27). 1.457 to 1.469.
1 mg ofberberine hydrochloride is equivalent to 0.9045 mg of Weight per ml (2.4.29).0.897 g to 0.924 g.
berberine.
Aldehydes. Place 10 ml in a glass-stoppered tube (150 mm x
Storage. Store protected from heat, moisture and against attack 25 mm) add 5 ml of toluene and 4 ml of ethanolic
of insects and rodents. hydroxylamine solution, shake vigorously and titrate
immediately with 0.5 M potassium hydroxide in ethanol
(60 per cent) until the red colour changes to yellow. Continue
Eucalyptus Oil the shaking and neutralising until the pure yellow colour of
the indicator is permanent in the lower layer after shaking
NilgiriOil vigorously for 2 minutes and allowing separation to take place;
~!1:(;alyptlIsQil is tl!(l(l§s(ll1til!IQ~l()t>:tajl1(l<it>ys!(ll!Il1<1j§tiIMi()l1 the reaction is__(;~Il1J>lelej!!_~t>_()1!t 1~_Il1il1ute_~~~l!Lt1J:~
and rectification from the fresh leaves or the fresh terminal operation using a further 10 ml of the substance under
branches of varIous speCies of ellcalyptlls iikeEucalyptus ex.amination, and as the standard for the end-point, the titrated
globulus Labill., E. fruticetorum F. von Muell., and E. smithii liquid ofthe first determination with the addition of 0.5 mlof
(R. T. Balcer)(Fam. Myrtaceae). 0.5 M potassium hydroxide in ethanol (60 per cent). Not

2498

www.webofpharma.com
IP 2010 GARCINIA

more than 2.0 ml of 0.5 M potassium hydroxide in ethanol Tests


(60 per cent) is required in the second determination.
Citric Acid. Not more than 2.0 per cent.
Phellandrene. Mix 1 ml with 2 ml of glacial acetic acid and
5 ml of light petroleum (40° to 60°), add 2 ml of a saturated Determine by liquid chromatography (2.4.14).
solution of sodium nitrite and shake gently; no crystalline Test solution, reference solutions (a), (b) and chromatographic
precipitate is produced in the upper layer within 1 hour. system as described under Assay.
Assay (2.3.24) Determine the content of cineole in the oil. Inject test solution and reference solution (b).
Storage. Store in well-filled, tightly-closed containers at a Calculate the content of citric acid.
temperature not exceeding 30°.
Foreign organic matter (2.6.1). Not more than 5.0 per cent.
Water-soluble extractive (2.6.3). Not less than 40.0 per cent
by Method I.
Garcinia
Total ash (2.3.19). Not more than 8.0 percent.
Vilayati Imli; Garcinia cambogia
Acid-insoluble ash (2.3.19). Not more than 1.5 percent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh about 5 g of the coarsely powdered
substance under examination and transfer to a 250-ml beaker.
Add 5 ml of dilute phosphoric acid and 100 ml of water,
concentrate to half the volume by heating, cool and filter.
Extract the residue further with water till the last extract turns
colorless, cool and filter. Combine all the filtrates and
concentrate under vacuum to 250.0 ml.

Garcinia is the dried deseeded fruit of Garcinia cambogia Reference solution (a). A 0.8 per cent w/v solution of
Desr. {Garciniagummi-gutia (L.) N: Robson}(Fam. Guttiferae). hydroxycitric acid calcium salt RS in dilute phosphoric acid.

Garcinia contains not less than 12.0 per cent of total Reference solution (b). A 0.1 per cent w/v solution of citric
hydroxycitric acid and hydroxycitric acid lactone, calculated acid in dilute phosphoric acid.
on the dried basis. Chromatographic system
Description. Dark brown to blackish brown fruits. Taste, acidic. - a stainless steel column 25 em x 4.6 rom packed with
octadecylsilane bonded to porous silica (5 Ilm),
Identification mobile phase: a buffer solution prepared by dissolving
1.36 g ofpotassium dihydrogen phosphate in 900 ml of
A. Macroscopic - Dark brown to blackish brown fruits,
water, adjusting the pH to 2.5 with dilute phosphoric
ovoid, longitudinally grooved.
acid and diluting to 1000.0 ml with water,
B. Microscopic - Mesocarp very wide composed of flow rate. 1 ml per minute,
parenchymatous cells ofvarious sizes and shapes. Compound - spectrophotometer set at 215 nm,
starch grains and prismatic crystals ofcalcium oxalate traverse - injection volume. 20 Ill.
throughout the parenchymatous cells of the mesocarp.
Inject the reference solution (a). The test is not valid unless
C. In the Assay, the principal peak in the chromatogram the relative retention times are about 0.8 for hydroxycitric
obtained with test solution has a retention time similar to that acid lactone, 1.0 for hydroxycitric acid and 2.0 for citric
of the peak due to hydroxycitric acid in the chromatogram acid, the resolution factor between hydroxycitric acid lactone
obtained with reference solution. and hydroxycitric acid is not less than 1.8, the tailing factor is

2499

www.webofpharma.com
GARCINIA AQUEOUS EXTRACT IP 2010

not more than 1.5 and the relative standard deviation for the Inject the referene solution and the test solution.
replicate injections is not more than 2.0 per cent. Calculate the content ofthe hydroxy citric acid in the extract.
Inject the test solution and reference solution (a). Calcium -
Calculate the sum of the contents of hydroxycitric acid and Test solution. Weigh accurately about 0.2 g ofthe extract and
hydroxycitric acid lactone. transfer into a 500 ml conical flask. Dissolve in 2 ml of 3 M
Storage. Store protected from heat, moisture and against attack hydrochloric acid and add 200 ml of water. Add 15 ml of 1 M
by insects and rodents. Sodium hydroxide and titrate with 0.05 M EDTA using hydroxy
naphthol blue as indicator until deep blue colour persist.
Each ml of 0.05 M EDTA solution is equivalent to 0.002004 g
Garcinia Aqueous Extract ofcalcium.
Garcinia Aqueous Extract is obtained by extracting Garcinia Usual strengths. 50 per cent; 60 per cent.
(Garcinia cambogia Desr., Garcinia gummi-gutta (L.) N: Storage. Store in air-tight containers, protected from light.
Robson, Fam. Guttiferoe) fruit rinds with water as calcium saIts.
Garcinia Aqueous Extract contains not less than 90.0 per cent
w/w to not more than 120.0 per cent w/w ofthe stated amount Gokhru
ofhydroxy citric acid, and calcium not less than 15.0 per cent Tribulus terrestris
w/w calculated on the dried basis. It may contain suitable
added substances.
Description. A pale brown to brown powder.

Tests
Total ash (2.3.19). Not more than 10.0 percent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 6.0 per cent determined
on I g by drying in an oven at 105°.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. hydroxy citric acid - Determine by liquid
chromatography (2.4.14).
Test Solution. Shake well 0.2 g ofthe extract under examination Gokhru consists of dried fruits of Tribulus terrestris
add 2 ml of 3 M hydrochloric acid and add 10 ml of water, L.(Fam.zygophyllaceae).
.sonicate it to dissolve, make up to volume 100.0 ml with water Gokhru contains not less than 0.5 per cent of diosgenin,
and mix. calculated on the dried basis.
Reference solution. A 0.1 per cent w/v solution of hydroxy Description. Pedicellate and globose fruits, having wedge-
citric acid RS with 2 ml of 3 M hydrochloric acid in water. shaped cocci, covered with short and stiff spines. Possesses
Chromatographic system faintly aromatic smell and acrid taste.
a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5J.1m), Identification
mobile phase: o. I per cent orthophosphoric acid in A. Macroscopic - Fruit is pedicellate, having wedge-shaped
water, filter and degas, cocci, covered with spines. Surface of schizocarp is rough.
- flow rote. 0.6 ml per minute,
___ ~ectr~~~()met~~~~tat_~~nm,- , _ B. Microscopic - Pericarp is differentiated into epicarp,
- injection volume. 20 J.11. mes6'carp and eiidocail): Epicarp issuif6uiidedoyiion'::'
. glandular trichomes;
Inject the reference solution. The test is not valid unless the
relative standard deviation for the replicate injections is not C. Determine by thin-layer chromatography (2.4.17), coating
more than 2.0 per cent. the plate with silica gel GF254.

2500

www.webofpharma.com
IP 2010 GUAR GUM

Mobile phase. A mixture of8 volumes oftoluene and 2 volumes mobile phase: 80 volumes ofacetonitrile and 20 volumes
of ethyl acetate. of methanol,
Test solution. Reflux 5 g ofcoarsely powdered substance under flow rate. 1 ml per minute,
examination with 50 ml of methanol for 15 minutes, cool and spectrophotometer set at 210 nm,
filter. Reflux the residue further with 50 ml ofmethanol, cool injection volume. 20 Ill.
and filter. Combine both the filtrates and concentrate under Inject the reference solution. The relative standard deviation
vacuum to dryness. Extract the dried residue with 10 ml of for the replicate injections is not more than 2.0 per cent.
methanol at 50° for 10 minutes, filter the solution and use
Inject the test solution.
filtrates for analysis.
Calculate the content of diosgenin.
Reference solution. Reflux 2.5 g ofgokhnl RS with 50 ml of
methanol for 15 minutes, cool and filter. Reflux the residue Storage. Store protected from heat, moisture and against attack
further with 50 ml ofmethanol, cool and filter. Combine both by insects and rodents.
the filtrates and concentrate under vacuum to dryness. Extract
the dried residue with 5 ml ofmethanol at 50° for 10 minutes,
filter the solution and use the filtrates for analysis. Guar Gum
Apply to the plate 20 III ofeach solution as bands 10 mm by 2 Guar Gum is a gum obtained from the ground endosperms of
rom. Allow the mobile phase to rise 8 cm. Dry the plate in air the seeds of Cyamopsis tetragonolobus (Linn.) Taub or other
and examine in ultraviolet light at 254 nm and 365 llill, spray species of Cyamopsis (Fam. Leguminosae). It consists mainly
with anisaldehyde sulphuric acid reagent. Heat the plate at of a high molecular weight hydrocolloidal polysaccharide,
105° for 5 minutes and examine in day light. The chromatographic composed of galactan and mannan units combined through
profile of the test solution is similar to that of the reference glycosidic linkages.
solution.
Description. An almost white to pale yellowish white powder;
Tests odour, characteristic.

Foreign organic matter (2.6.1). Not more than 2.0 per cent. Identification
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent. A. When mounted in lactophenol and examined under a
Water-soluble extractive (2.6.3). Not less than 15.0 per cent microscope, irregular, angular particles of various sizes and
by method I. shapes are seen.
Total ash (2.3 .19). Not more than 11.0 per cent. B. To 0.1 g add 1 ml of 0.2 M iodine; the mixture does not
acquire an olive-green colour.
Acid-insoluble ash (2.3.19). Not more than 1.0 per cent.
C. Dissolve 0.1 gin 20 ml ofwater by shaking and add 0.5 ml
Heavy metals (2.3.13). 1.0 g complies with the limit test for
of hydrogen peroxide solution (20 vol) and 0.5 ml ofa 1 per
heavy metals, Method B (20 ppm).
cent w/v solution of benzidine in ethanol (90 per cent), shake
Loss on drying (2.4.19). Not more than 5.0 per cent, determined and allow to stand; no blue colour is produced (distinction
on 5 g by drying in an oven at 105°. from acacia).
Microbial contamination (2.2.9). Complies with the microbial D. Mount a small quantity in ruthenium red solution and
contamination tests. examine under a microscope; the particles do not acquire a
Assay. Determine by liquid chromatography (2.4.14). pink colour (distinction from sterculia gum and agar).
Test solution. Reflux 5.0 g ofthe substance under examination E. To 2 ml of a 0.5 per cent w/v solution add 2 ml of a 20 per
with 50 m1 of sulphuric acid (10 per cent) for 4 hours. Cool cent w/v solution of lead acetate; a flocculent precipitate is
and transfer to separating funnel. Extract with 50 ml of ethyl produced (distinction from acacia, ghatti gum and sterculia).
acetate. Repeat the extraction 3 times. Pass the ethyl acetate
Tests
layer through sodium sulphate and evaporate. Dissolve the
residue with 50 rn1 of methanol. Acidity or alkalinity. A 0.5 per cent w/v solution is neutral to
Reference solution. A 0.1 per cent w/v solution of diosgenin litmus paper.
RS in methanol. Tannin. To 5 ml of a 0.5 per cent w/v solution add 0.1 ml of
Chromatographic system ferric chloride test solution; no bluish black colour is produced.
a stainless steel column 25 cm x 4.6 rom packed with Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium
octadecylsilane bonded to porous silica (5 Ilm), carbonate, add 10 ml ofbromine solution and mix thoroughly.

2501

www.webofpharma.com
·GUAR GUM IP 2010

Evaporate to dryness on a water-bath, gently ignite and Gudmar contains not less than 1.0 per cent w/w of gymnemic
dissolve the cooled residue in a mixture of 16 ml of brominated acids (calculated as gymnemagenin), calculated on dried basis.
hydrochloric acid and 45 ml of water. Remove the excess of
Description. Greenish-yellow in colour, surface pubescent
bromine with 2 ml of stannous chloride solution AsT. The
on both sides and characteristic odour with extremely bitter
resulting solution complies with the limit test for arsenic
and acrid taste.
(3 ppm).
Heavy metals (2.3.13). 1.0 g complies with the limit test for Identification
heavy metals, Method B (20 ppm).
A. Macroscopic - Leaves, simple, petiolate about 2 to 6 em
Protein. Not more than 5.0 per cent, determined by the long and 1 to 4 em broad, yellowish brown on adaxial and dark
following method. Carry out the determination of nitrogen green on abaxial side.
(2.3.30), using about 3.5 g, accurately weighed, and multiplying
the percentage of nitrogen determined by 6.25 to obtain the B. Microscopic - Upper and lower epidermis covered with
percentage of protein. cuticle having uni to tri cellular covering trichomes which are
slightly curved at the bulbous base. Below the epidermis is
Acid-insoluble matter. Not more than 3.0 per cent, determined
single layer ofpalisade cells followed by 2-3 layered spongy
by the following method. Weigh accurately about 1.5 g and
parenchyma. Starch gains are simple and present in spongy
disperse in 150 ml of water and 1.5 ml ofsulphuric acid. Warm
parenchyma. Midrib region shows 2-7 layers of
on a water-bath for 6 hours, replacing the water lost by
collenchymatous cells. Stomata are ofparacytic type, mostly
evaporation. Add about 0.5 g ofa suitable filter-aid, accurately
on lower the surface. There is a fan shaped vascular bundle in
weighed, and filter through a suitable ashless filter paper. Wash
the centre. Each vascular bundle is collateral, closed and
the residue several times with hot water, dry the filter and its
surrounded by parenchymatous sheath. Rosette crystal of
contents at 105° for 3 hours. Cool in a desiccator, weigh and
calcium oxalate present in the spongy parenchyma.
subtract the weight of the filter aid.
Microbial contamination (2.2.9). Total bacterial count: Not C. Determine by thin-layer chromatography (2.4.17), coating
more than 5000 per g. 1 g is free from Escherichia coli and 10 the plate with silica gel GF254.
g is fi'ee from salmonellae. Mobile phase. A mixture of5 volumes of chloroforms, 1 volume
Total ash (2.3.19). Not more than 2.0 per cent, determined on of methanol and 1 volume of ethyl acetate.
l.Og. Test solution. Reflux 5 g ofthe coarsely powdered substance
Loss on drying (2.4.19). Not more than 13.0 per cent, under examination with 50 ml of ethanol (50 per cent v/v) for
determined on 0.5 g by drying in an oven at 105°. 15 minutes, cool and filter. Reflux the residue further with 2 x
50 ml of ethanol (50 per cent v/v), cool and filter. Combine all
the filtrates and concentrate under vacuum to 25 ml. Take 5 ml
of resulting solution, add 5 ml ethanol and 2 ml ofpotassium
Gudmar
hydroxide and reflux for 1 hour. Cool and add 1.8 ml of 12 M
Gymnema sylvestre hydrochloric acid and heat on water bath. Cool and adjust
the pH to 7.5-8.5 with 11 per cent potassium hydroxide. Dilute
the solution with ethanol (50 per cent v/v) to 100 ml and filter.
9
Reference Solution. Reflux 1 g of gudmar RS with 50 ml of
8
ethanol (50 per cent v/v) for 15 minutes, cool and filter. Reflux
7
the residue further with 2 x 50 ml of ethanol (50 per cent v/v) ,
6 cool and filter. Combine all the filtrates and concentrate under
vacuum to 10 ml. Take 5 ml of resulting solution, add 5 ml
ethanol and 2 ml ofpotassium hydroxide and reflux for 1 hour.
4
Cool and add 1.8 ml of 12 M hydrochloric acid and heat on
water bath. Cool and adjust the pH to 7.5-8.5 with 11 per cent
2 potassium hydroxide. Dilute the solution with ethanol
(50 per cent v/v) to 50 mi and filter.
1

o Apply to the piatelO III ofeach solution as ~ands 10 mm 2


rom. Allow the mobile phase to rise 8 em. Dry the plate in air
Gudmar consists of the dried mature leaves of Gymnema and examine in ultraviolet light at 254 urn and 365 nm, spray
sylvestre R.Br. (Fam. Asclepiadaceae). with anisaldehyde sulphuric acid reagent. Heat the plate at

2502

www.webofpharma.com
IP 2010 GUDUCHI

100° for 10 minutes and examine in day light. The Inject the reference solution. The test is not valid unless the
chromatographic profile of the test solution is similar to that relative standard deviation for the replicate injections is not
of the reference solution. more than 2.0 per cent.
Inject the test solution and reference solution.
Tests
Calculate the content of gymnemagenin.
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Storage. Store protected from heat, moisture and against attack
Ethanol-soluble extractive (2.6.2). Not less than 5.0 per cent. by insects and rodents.
Water-soluble extractive (2.6.3). Not less than 20.0 per cent
by method 1.
Total ash (2.3.19). Not more than 15.0 per cent. Guduchi
Acid-insoluble ash (2.3.19). Not more than 6.0 per cent. Giloe; Amrita; Tinospora cordifolia
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
9
Loss on drying (2.4.19). Not more than 14.0 per cent, 8
determined on 5 g by drying in an oven at 105°.
7

Microbial contamination (2.2.9). Complies with the microbial 6


contamination tests.
Assay. Determine by liquid chromatography (2.4.14). 4

Test solution. Reflux 5 g of the coarsely powdered substance 3


under examination with 50 ml of ethanol (50 per cent v/v) for
15 minutes, cool and filter. Reflux the residue further with 2 x
1
50 ml of ethanol (50 per cent v/v) , cool and filter. Combine all
the filtrates and concentrate under vacuum to 25 ml. Take 5 ml o
of resulting solution, add 5 ml ethanol and 2 ml ofpotassium
hydroxide and reflux for 1 hour. Cool and add 1.8 ml of 12 M
hydrochloric acid and heat on water bath. Cool and adjust Guduchi consists of the dried, mature pieces of stem of
the pH to 7.5-8.5 with 11 per cent potassium hydroxide. Dilute Tinospora cordifolia (W~lld.) Miers (Fam. Menispermaceae).
the solution with ethanol (50 per cent v/v) to 100 ml and filter. Guduchi contains not less than 0.02 per cent w/w of
cordifolioside A, calculated on the dried basis.
Reference solution. A 0.01 per cent w/v solution of
gymnemagenin RS in methanol. Description. A greyish-black in colour, fibrous fracture and
no distinct odour with bitter taste.
Chromatographic system
- a stainless steel column 25 em x 4.6 rom packed with Identification
octadecylsilane bonded to porous silica (5 /lm),
- mobile phase (A). 80 per cent v/v acetonitrile, A. Macroscopic - Stem-pieces glabrous, cylindrical, solid,
(B). 0.1 percentw/v dihydogenpotassium lenticillate, 5-15 rom in diameter having light brown surface
phosphate, marked with warty protuberances due to circular lenticels.
- a linear gradient prograriune using the conditions given Transversely smoothened surface shows a radial structure
below, with conspicuous medullary rays traversing porous tissues.
- flow rate. 1.5 mlperminute, B. Microscopic - Transverse section of stem shows
spectrophotometer set at 210 urn, outermost layer ofcork which is differentiated in to outer zone
injection volume. 20 /ll. ofthick walled, compressed cells and inner zone ofthin walled,
Time Mobile phase A Mobile phase B tangential cells. Cork broken at some places due to lenticels.
(min) (per cent v/v) (per cent v/v) Cortex consists of 3-5 rows ofirtegularly arranged tangential,
chlorenchymatous cells with numerous intercellular spaces.
o 25 75
Inner cortex filled with plenty of starch gains. Vascular zone
20 50 50 consists of 10-12 wedge-shaped strips of xylem externally
30 25 75 surrounded by semi-circular strips of phloem. Cambium

2503

www.webofpharma.com
GUDUCHI IP 2010

composed ofone to two layers oftangentially elongated cells. Chromatographic system


Primary phloem appears crushed and obliterated; secondary - a stainless steel column 25 cm x 4.6 mm packed with
phloem groups are massive. Pith composed oflarge, thin walled octadecylsilane bonded to porous silica (5 /-lm),
cells mostly containing starch gains. - mobile phase: A. a gradient mixtures of water
B. .acetonitrile,
C. Determine by thin-layer chromatography (2.4.17), coating
- a linear gradient programme using the conditions given
the plate with silica gel GF254.
below,
Mobile phase. A mixture of 85 volumes of chloroform and - flow rate. 1 ml per minute,
15 volumes of methanol. - spectrophotometer set at 210 urn,
Test solution. Reflux 2 g of the coarsely powdered substance - injection volume. 20 /-ll.
under examination with 25 ml of methanol for 15 minutes, cool Time Mobile phase A Mobile phase B
and filter. Reflux the residue further with 2 x 25 ml of methanol, (in min) (per cent v/v) (per cent v/v)
cool and filter. Combine all the filtrates and concentrate under o 80 20
vacuum to 5 ml. 25 20 80
Reference solution. Reflux 2 g ofthe guduchi RS with 25 mlof 30 80 20
methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 x 25 ml of methanol, cool and filter. Combine all Inject the reference solution. The test is not valid unless the
the filtrates and concentrate under vacuum to 5 ml. relative standard deviation for the replicate injections is not
more than 2.0 per cent.
Apply to the plate 10 /-ll ofeach solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air Inject the test solution and reference solution.
and examine in ultraviolet light at 254 nm and 365 urn, spray Calculate the cont~nt of cordifolioside A.
with anisaldehyde sulphuric acid reagent. Heat the plate at
Storage. Store protected from heat, moisture and against attack
110° for 10 minutes and examine in day light. The
by insects and rodents.
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Guggul Resin
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Guggul; Commiphora wightii
Ethanol-soluble extractive (2.6.2). Not less than 1.5 per cent.
Water-soluble extractive (2.6.3). Not less than 9.0 per cent by
method!. 9

Total ash (2.3.19). Not more than 10.0 percent. 8

7
Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
6
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm). 5

Loss on drying (2.4.19). Not more than 10.0 per cent, 4

determined on 5 g by drying in an oven at 105°. 3


Microbial contamination (2.2.9). Complies with the microbial 2
contamination tests.
Assay. Determine by liquid chromatography (2.4.14). o
Test solution. Reflux 2 g of the coarsely powdered substance
under examination with 50 ml of methanol on a water-bath for
15 minutes, cool and filter. Reflux the residue further with Guggul Resin is the oleoresin exudation from Commiphora
methanol till the extract turns colourless, cool and filter. wightii (Arnott) Bhandari (Commiphora mukul (Am.)
. -Comoinealnhe~filtratesandcoifcentrateto~avoluinesliglit1y Bfian(Jan~73atsiiiiioaenaroii-miilCiil
H ool(~ ex ·StoCks) (Fam.
less than25 ml. Dilute with methanol to 25;0 ml. Btitsetaceae). .
Reference solution. A 0.004 per cent w/v solution of Guggul Resin contains not less than 1.0 per cent w/w and not
cordifolioside A RS in methanol. more than 1.5 per cent w/w of gugulsterones (Z and E).

2504

www.webofpharma.com
IP 2010 GUGULIPID

Description. Light to dark-brown conglomerates of tears, allowing to stand for 18 hours. Filter rapidly taking care to
rounded or irregular, slightly sticky to touch; odour, faintly avoid loss ofethanol, evaporate 25 ml ofthe filtrate to dryness,
balsamic. dry at 105° and weigh.
Sulphated ash (2.3.18). Not more than 10.0 per cent, determined
Identification
onO.5 g.
A. Prepare a 0.005 per cent w/v solution in ethanol (95 per
Assay. Determine by liquid chromatography (2.3.14).
cent) of the residue obtained in the test for Ethanol-soluble
extractive. Test solution. Weigh accUrately about 3.0 g of the substance
under examination, add 50 ml of acetonitrile, reflux on a water-
When examined in the range 230 nm to 360 nm (2.4.7), the
bath for 30 minutes, cool and filter. Reflux the residue further
resulting solution shows absorption maxima at about 245 nm
with three portions, each of 30 ml, of acetonitrile, cool and
and 327 nm.
filter. Combine the filtrates and concentrate to 100.0 ml.
B. Determine by thin-layer chromatography (2.4.17), coating
Reference solution. A 0.02 per cent w/v solution of
the plate with silica gel GF254.
gugulsterones (Z and E) RS in acetonitrile.
Mobile phase. A mixture on volumes of light petroleum (60°
Chromatographic system
to 80°) and 1 volume of ethyl acetate.
a stainless steel column 25 cm x 4.6 mm, packed with
Test solution. Dissolve 0.5 g of the residue obtained in the octadecylsilane silica gel (5 /Lm),
test for Ethanol-soluble extractive in 100 ml of ethanol (95 per mobile phase: a filtered and degassed mixture of 45
cent). volumes of acetonitrile and 55 volumes of water,
flow rate. 2 ml per minute,
Reference solution. A 0.5 per cent w/v solution ofthe residue
spectrophotometer set at 242 mn,
obtained similarly from guggul resin RS in ethanol (95 per
injection volume. 20 /Ll.
cent).
Inject the reference solution. The test is not valid unless the
Apply to the plate 20 /Ll of each solution as bands 10 mm by 2
relative retention times are about 0.69 for gugulsterone E and
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
1.0 for gugulsterone Z and the relative standard deviation for
until the odour of the solvent is no longer detectable and
replicate injections is not more than 2.0 per cent.
examine in ultraviolet light at 254 om and 365 om, spray with a
10 per cent v/v solution of sulphuric acid in methanol. Heat Inject the test solution and the reference solution. Calculate
the plate at 100° for 10 minutes and examine in day light. The the contents of gugulsterones (Z and E).
chromatographic profile of the test solution is similar to that
Storage. Store protected from light at a temperature not
of the reference solution.
exceeding 30°.
Tests
Ethyl acetate-soluble extractive. Not less than 25.0 per cent,
determined by the following method. Crush the substance
Gugulipid
under examination to a coarse powder. Shake 5.0 g of the Gugulipid is the ethyl acetate extractive ofGuggul Resin.
powder with 25 ml of light petroleum (60° to 80°) for 1 hour
Gugulipid contains not less than 4.0 per cent w/w and not
and separate the liquid by filtration. Repeat the extraction
more than 6.0 per cent w/w of gugulsterones (Z and E).
twice and dry the defatted material over phosphonLS pentoxide
at room temperature at a pressure not exceeding 2.75 kPa for Description. A brown, viscous liquid.
8 hours. Crush the dried material and extract with four quantities,
each of25 ml, of ethyl acetate by shaking each time for 1 hour Identification
followed by filtration through a sintered-glass funnel (porosity
A. When examined in the range 230 om to 360 om (2.4.7), a
No.3) and combining the filtrates. Evaporate the combined
0.005 per cent w/v solution in chloroform shows absorption
filtrates, dry over phosphorus pentoxide at room temperature
maxima at about 245 om and 327 om; absorbance at about
at a pressure not exceeding 2.75 kPa for 12 hours and weigh.
245 nm, about 0.87 and at about 327 nm, about 0.52.
Ethanol-soluble extractive (2.3.46). Not less than 35.0 percent,
determined by the following method. Crush the substance B. Determine by thin-layer chromatography (2.4.17), coating
under examination to a coarse powder. Macerate 5.0 g of the the plate with silica gel GF254.
powder with 100 ml of ethanol (95 per cent) in a closed flask Mobile phase. A mixture of 3 volumes of light petroleum
for 24 hours, shaldng frequently during the first 6 hours and (60° to 80°) and 1 volume of ethyl acetate.

2505

www.webofpharma.com
GUGULIPID IP 2010

Test solution. Dissolve 0.25 g of the substance under 15 m1, of ethyl acetate, combine the extracts, filter and
examination in 100 ml of ethanol (95 per. cent). evaporate to dryness. The residue complies with the following
tests.
Reference solution. A 0.25 per cent w/v solution of
gugulipid RS in ethanol (95 per cent). A. When examined in the range 230 om to 360 om
(2.4.7),
a 0.005 per cent w/v solution in chloroform shows absorption
Apply to the plate 5 f.ll of each solution as bands 10 mm by 2
·mm. Allow the mobile phase to rise 8 cm. Dry the plate in air maxima at about 245 nm and 327 om; absorbance at about
245 nm, about 0.87 and at about 327 om, about 0.52.
until the odour of the solvent is no longer detectable and
examine in ultraviolet light at 254 om and 365 om, spray with a B. Determine by thin-layer chromatography (2.4.17), coating
10 per cent v/v solution of sulphuric acid in methanol. Heat the plate with silica gel G.
the plate at 100° for 10 minutes and examine in day light. The
Mobile phase. A mixture of 3 volumes of light petroleum
chromatographic profile of the test solution is similar to that
(60° to 80°) and 1 volume of ethyl acetate.
of the reference solution.
Test solution. A 0.25 per cent w/v solution of the substance
Tests under examination in ethanol (95 per cent).

Assay. Detennine by liquid chromatography (2.4.14). Reference solution. A 0.25 per cent w/v solution of gugulipid
RS in ethanol (95 per cent).
Test solution. Weigh accurately about 0.75 g of the substance
under examination, add 50 ml of acetonitrile and warm on a Apply to the plate 5 f.ll of each solution. After development,
boiling water-bath for 10 minutes. Cool and add sufficient dry the plate in air until the odour of the solvent is no longer
acetonitrile to produce 100.0 m1. detectable and spray with 50 per cent w/v solution of sulphuric
acid. The principal spots in the chromatogram obtained with
Reference solution. A solution containing 0.02 per cent w/v of
the test solution correspond to those in the chromatogram
gugulsterones (Z and E) in acetonitrile.
obtained with the reference solution.
Chromatographic system
a stainless steel column 25 cm x 4.6 mm, packed with Tests
octadecylsilane silica gel (5 f.lm),
mobile phase: a filtered and degassed mixture of Disintegration (2.5.1).60 minutes.
45 volumes of acetonitrile and 55 volumes of water, Other tests. Comply with the tests stated under Tablets.
- flow rate. 2 ml per minute,
spectrophotometer set at 242nm, Assay. Determine by liquid chromatography (2.4.13).
injection volume. 20 f.l1. Test solution. Weigh and powder 20 tablets. Weigh accurately
Inject the reference solution. The test is not valid unless the a quantity of the powder containing about 10 mg of
relative retention times are about 0.69 for gugulsterone E and gugulsterones (Z and E) and extract with five quantities, each
1.0 for gugulsterone Z and the relative standard deviation for of 20 ml, of acetonitrile, with the aid of heat. Combine the
replicate injections is not more than 2.0 per cent. extracts and concentrate to 50.0 ml. Filter the solution through
a membrane filter disc with an average pore diameter not greater
Inject alternately the test solution and the reference solution. than 1.0 f.lm and use the filtrate.
Calculate the contents of gugulsterones (Z and E).
Reference solution. A solution containing 0.02 per cent w/v of
Storage. Store protected from light at a temperature not gugulsterones (Z and E) in acetonitrile.
exceeding 30°.
Chromatographic system
a stainless steel colwI1ll 25 cm x 4.6 mm, packed with
octadecylsilyl silica gel (5 f.lm),
mobile phase: a filtered and degassed mixture of
Gugulipid Tablets 45 volumes of acetonitrile and 55 volumes of water,
Gugulipid Tablets contain not less than 90.0 per cent and not - flow rate. 2 ml per minute,
more than 110.0 per cent ofthe stated amount ofgugulsterones spectrophotometer set at 242 om,
(Z and E). The tablets may be coated. injection volume. 20 f.ll.
the reference solution. The test is not valid unless the
Identification
relative retention times are about 0.69 for gugulsterone E and
Extract a quantity of the powdered tablets containing about 1.0 for gugulsterone Z and the relative standard deviation for
20 mg of gugulsterones (Z and E) with two quantities; each of replicate injections is not more than 2.0 per cent.

2506

www.webofpharma.com
IP 2010 HARlDRA

Inject the test solution and the reference solution. Calculate Reference solution. Reflux 1 g of coarsely powdered haridra
the contents of gugulsterones (Z and E) in the tablets. RS with 5 ml methanol for 15 minutes, cool and filter.
Storage. Store protected from light at a temperature not Apply to the plate 10 f..ll of each solution as bands 10 rom by 2
exceeding 30°. mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
Labelling. The label states the strength in terms of the and examine in ultraviolet light 254 run, 365 run and also under
equivalent amount of gugulsterones (Z and E). day light. The chromatographic profile of the test solution is
similar to that ofthe reference solution.

Tests
Haridra
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Haldi; Turmeric; Curcuma longa
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 12.0 per cent
9 by Method 1.
8 Total ash (2.3.19). Not more than 10.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
6
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
4
Water (2.3.43). Not more than 12.0 per cent, determined on
0.2g.
2 Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
o Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux about 1 g of the coarsely powdered
Haridra consists ofthe dried rhizomes of Curcuma longa Linn. substance under examination with 50 ml of methanol on a
(Fam. Zingiberaceae). .
water bath for 15 minutes cool and filter. Reflux the residue
Haridra contains not less than 1.5 per cent w/w of curcumin, further with 5 x 25 ml of methanol, cool and filter. Combine all
calculated on the dried basis. the filtrates and concentrate to 100.Omi.
Description. Externally yellowish to yellowish brown with Reference solution. A 0.01 per cent w/v solution of curcumin
root scars and annulations. Odour, aromatic; taste, warmly RS in methanol.
aromatic and bitter. Chromatographic system
a stainless steel column 25 cm x 4.6 mm packed with
Identification silicagel consisting of porous spherical particles with
A. Macroscopic - Rhizome oblong, conical or cylindrical to chemically bonded nitrile group,
elongate, finger-like; internally orange yellow. Texture hard - mobile phase: a mixture of35 volumes of tetrahydrofuran
and heavy; fracture short. 65 volumes of a buffer solution prepared by dissolving
109 of citric acid in 1000 ml of water, adjusting the pH
B. Microscopic - Ground tissue of parenchyma cells; cells
to 3.0 with dilute ammonia solution,
filled with gelatinized starch grains and yellow pigment.
flow rate. 1.2 ml per minute,
Fibrovascular bundles and oil cells scattered through out
spectrophotometer set at 430 run,
ground tissue.
injection volume. 20 f..lI.
C. Determine by thin-layer chromatography (2.4.17), coating
Inject the reference solution. The test is not valid unless the
the plate with silica gel GF254.
relative standard deviation for the replicate injections is not
Mobile phase. A mixture of 94 volumes of chloroform, more than 2.0 per cent.
5 volumes of ethanol and 1 volume glacial acetic acid.
Inject the reference solution and the test solution.
Test solution. Extract 1 g of the coarsely powdered substance
under examination with 5 ml methanol for 10 minutes with Calculate the content of curcumin.
slight warming. Filter and use the filtrate. Storage. Store protected from moisture.

2507

www.webofpharma.com
HAiuDRA DRY EXTRACT IP 2010

Haridra Dry Extract Test Solution. Dissolve a quantity of the extract under
examination containing about 50 mg curcumin in 100.0 ml of
Haridra Dry Extract is a partially purified natural complex of tetrahydrofuran. Dilute 5.0 ml ofthis solution to 50.0 ml with
diaryl heptanoid derivatives isolated by extracting Haridra the mobile phase.
(Curcuma longa L., Fam. Zingiberaceae), rhizomes with
Reference solution. A solution 'containing 0.01 per cent w/v
acetone or ethanol or any other suitable solvent and
each of bisdemethoxycurcumin RS, demethoxycurcumin RS,
evaporation of solvent under vacuum..
curcumin RSin tetrahydrojitran. Dilute 5ml ofthis solution to
Haridra Dry Extract contains not less than 95.0 per cent w/w 50 ml with the mobile phase.
and not more than 102.0 per cent w/w ofthe stated amount of
Chromatographic system
total curcuminoids calculated on the dried basis, as the sum
a stainless steel column, 25 em x 0.46 mm packed with
ofcurcumin, demethoxycurcumin and bisdemethoxycurcumin.
octadecylsilane bonded to porous silica (5f..lm),
It contains not less than 70.0 per cent w/w to not more than
mobile phase: a mixture of 40 volumes of 0.1 per cent
80.0 per cent w/w ofcurcumin, not less than 15.0 per cent w/w
w/v solution of citric acid and 60 volumes of
to not more than 25.0 per cent w/w ofdemethoxycurcumin and
not less than 2.5 per cent w/w to not more than 6.5 per cent
tetrahydrofuran,
flow rate. 1.0 ml per minute,
w/w ofbisdemethoxycurcumin.
spectrophotometer set at 420 nm,
Description. An orange yellow crystalline powder. injection volume. 20 f..ll.
Inject the reference solution. The test is not valid unless the
Identification
relative standard deviation for replicate injections is not more
A. Determine by thin-layer chromatography (2.4.17) coating than 2.0 per cent. The relative retention time with reference to
the plate with silica gel GF254. curcumin for bisdemethoxycurcumin is about 1.3 and for
demethoxycurcumin is about 1.14.
Mobile phase. A mixture of 10 volumes of chloroform, 1.0
volume of methanol, 0.5 volume of glacial acetic acid and 0.5 Inject the test solution and the reference solution.
volume offormic acid. Calculate the content of the bisdemethoxycurcumin,
Test solution. Dissolve about 50 mg of the extract under demethoxycurcumin and curcumin in extract.
examination with 100.0 ml of methanol and filter. Storage. Store protected from heat and moisture.
Reference solution. A solution containing 0.01 per cent w/v
each of bisdemethoxycurcumin RS, demethoxycurcumin RS,
curcumin RS in the methanol.
Apply to the plate 10 f..ll ofeach ofsolution as bands 10 mm by Haritaki
2 mm. Allow the mobile phase to rise 8 em. Dry the plate in air
Harad; Chebulic myrobalan; Terminalia chebula
and examine in ultraviolet light at 254 nm. Spray the plate with
a anisaldehyde- sulphuric acid reagent. Heat the plate at
110° for 10 minutes and examine the plate at 365 nm and day
light. The chromatographic profile ofthe test solution is similar
to that of the reference solution.

Tests

Sulphated ash (2.3.18). Not more than 1.0 per cent.


Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 2.0 per cent, determined
on 1 g by drying in an oven at 105°.
Microbialcontarnination(2:2:9): Complies withthemicrobial
contamination 'tests;- -,
Assay. Total curcuminoids - Determine by liquid Haritaki consists of pericarp of the dried fruit of Terminalia
chromatography (2.4.14). chebula Retz. (Fam. Combretaceae).

2508

www.webofpharma.com
IP 2010 HARITAKI

Haritaki contains not less than 5 per cent w/w of chebulagic Ethanol-soluble extractive (2.6.2). Not less than 35.0 per cent.
acid and not less than 12.5 per cent w/w of chebulinic acid, Water-soluble extractive (2.6.3). Not less than 50 per cent by
calculated on the dried basis. Method I.
Description. It has a shine on its external part and has Total ash (2.3.19). Not more than 6.0 percent.
longitudinal ridges. The colour varies from yellowish brown
to light black. It has a astringent taste and is also slightly Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
bitter Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm)
Identification
Loss on drying (2.4.19). Not more than 12 per cent, determined
Test C may be omitted if tests A, BandD are carried out and on 5 g by drying in an oven at 105°.
test D may be omitted if tests A, Band C are carried out. Microbial contamination (2.2.9). Complies with the microbial
A. Macroscopic - The fruit is 2 to 3 cm in length and 1 to contamination tests.
2 cm in diameter with hard stony appearance. Externally it is Assay. Detennine by liquid chromatography (2.4.14).
shining and is adorned with longitudinal ridges. Color of the
fruit rind varies from yellowish brown, uniform brown to light Test solution. Weigh 0.5 g of coarsely powdered sample, add
black. Internally the fruit is light yellow. 50 ml of water, sonicate for 3 minutes and heat on a boiling
water bath for 15 minutes, cool and dilute to 100.0 ml with
B. Microscopic - Epicarp has thick walls covered with cuticle.
water and·filter. Dilute 10.0 ml ofthe solution to 25.0 ml with
The mesocarp has many stone cells of various sizes and
water.
shapes which forms a reticulum. Large quantity of tannin is
present in the mesocarp..Simple starch granules are present in Reference solution (a). Weigh 0.5 g of haritaki RS, add 50 ml
plenty. of water, sonicate for 3 minutes and heat on a boiling water
bath for 15 minutes, cool and dilute to 100.0 ml with water and
C. Determine by thin-layer chromatography (2.4.17), coating
filter. Dilute 10.0 ml ofthe solution to 25.0ml with water.
the plate with silica gel GF254.
Reference solution (b). A 0.1 per cent w/v solution of
Mobile phase. Amixture of 35 volumes of toulene, 50 volumes
chebulagic acid RS in water.
of acetone, 15 volumes of glacial acetic acid and 5 volumes
offormic acid. Reference solution (c). A 0.1 per cent w/v solution of
chebulinic acid RS in wate]:
Test solution. To 1 g of the coarsely powdered substance
being examined, add 50-75 ml of methanol and reflux for Chromatographic system
15 minutes, cooland filter. Reflux the residue further for two - a stainless steel column 25 cm x 4.6 mm packed with
times with 75 ml of methanol, cool and filter. Combined all the octadecylsilane bonded to porous silica (5 11m).
filtrates and concentrate under vacuum to 100 ml. - mobile phase: A.a buffer solution pH 2.5 prepared by
dissolving 0.136 g of potassium di~hydrogen
Reference solution. To 0.1 g ofthe haritald RS, add 50-75 ml
orthophosphate in 500 ml of water, add 0.5 ml of
of methanol and reflux for 15 minutes, cool and filter. Refluf(
orthophosphoric acid and make upto 1000 ml with
the residue further for two times with 75 ml of methanol, cool
water,
and filter. Combine all the filtrates and concentrate under
B. acetonitrile,
vacuum to 10 ml.
flow rate. 1.5 ml per minute
Apply to the plate 10 III or'each solution as bands 10 mm by 2 - spectrophotometer set at 270 nm,
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air - injection volume. 20 Ill.
and examine in ultraviolet light at 254 nm and 365 nm, spray
Time Mobile phase A Mobile phase B
with 10 per cent w/v ferric chloride solution in water. Heat
(in min.) (per cent v/v) (per cent v/v)
the plate at 100° for 10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that a 95 5
of the reference solution. 18 65 35
D. In the Assay, the chromatogram obtained with the test 25 45 55
solution corresponds to the chromatogram obtained with 28 45 55
reference solution (a). 35 95 5

Tests Inject the reference solution (b) and (c). The relative standard
deviation for the replicate injections is not more than 2.0 per
Foreign organic matter (2.6.1). Not more than 2.0 per cent. cent:

2509

www.webofpharma.com
HARITAKI EXTRACT IP 2010

Inject the test solution and reference solution (a). Assay. Detennine by liquid chromatography (2.4.14).
Calculate the content of Chebulagic acid and Chebulinic Test solution. Dissolve about 0.5 g ofthe extract or quantity
acid. equivalent to 100mg ofpolyphenoIs in water, make up to 100
Storage. Store protected from heat, moishrre and against attack ml and filter.
by insects and rodents. Reference solution (a). 0.01 per cent w/v solution of
chebulagic acid RS in water.
Reference solution (b). 0.01 per cent w/v solution of
Haritaki Extract chebulinic acid RS in watel:
Haritaki extract is obtained by extracting Haritaki (Terminalia Chromatographic system
chebula Retz., Fam. Combretaceae) from the dried fruit pericarp - a stainless steel column 25 cm x 4.6 mm packed with
with ethanol or any other suitable solvent. octadecylsilane bonded to porous silica (5 Ilm),
mobile phase:A. a buffer solution prepared by dissolving
Haritaki extract contains not less than 90 per cent w/w and not
0.136 g of potassium di-hydrogen orthophosphate in
more than 120 percent w/w ofthe stated amount ofchebulinic
500 ml of watel; add 0.5 mI of orthophosphoric acid and
acid and chebulagic acid.
dilute to 1000 ml with water,
Description. Light yellowish to yellowish brown powder with B. acetonitrile,
odour, characteristic; taste, bitter. a linear gradient programme using the conditions given
below,
Identification flow rate. 1.5 ml per minute,
A. Detennine by thin-layer chromatography (2.4.17), coating spectrophotometer set at 270 nm,
the plate with silica gel GF 254. injection volume. 20 Ill.
Time mobile phase A mobile phase B
Mobile phase. A mixture of 6 volumes of ethyl acetate, 2
(in min) (per cent v/v) (per cent v/v)
volumes offormic acid, 2 volumes of toluene and 1 volume of
methanol. o 95 5
Test solution. Dissolve 0.5 g ofextract under examination with 18 75 25
100 ml methanol and filter. 25 65 35
Reference solution. A. 0.1 per cent w/v solution of chebulagic 28 65 35
acid RS and 0.1 per cent w/v solution of chebulinic acid RS 35 95 5
in methanol.
Inject reference solution (a) and (b). The test is not valid rrnless
Apply to the plate 10 III of each solution as bands 10 mm by 2 the relative standard deviation for the replicate injections is
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air not more than 2.0 per cent.
and examine in ultra-violet light at 254 urn. Spray the plate with
ferric chloride reagent. Heat the plate at I 10° for 10 minutes Inject the reference solution and the test solution.
and examine the plates at 365 nm and in day light. The Calculate the content of chebulinic acid and chebuliagic
chromatographic profile of the test solution is similar to that acid.
of the reference solution.
Usual strength. 15 per cent w/w.
B. In the assay, the peaks due to c hebulinic acid and chebulagic
Storage. Store protected from heat and moisture.
acid in the chromatogram obtained with test solution
corresponds to the peak obtained with reference solutions.

Tests
Haritaki Aqueous Extract
Loss on drying (2.4.19). Not more than 5.0 per cent, determined
Haritaki Aqueous Extract is obtained from the dried fruit
on 2.0 g by drying in an oven at 105° for 3 hours.
pericarp of Haritaki (Terminalia chebula Retz., Fam.
Total ash (2.3.19). Not more than 2.0 percent. Combretaceae) by extraction with water.
--_._-", .. _-.,'------"'--_._----
Heavy metals (2~3 .13). 1.0· gcomplies with the limit fest for Haritaki Aqueous Extract contains not less than 90.0 per cent
heavy metals, (Method B) 20 ppm; w!waIlcl [lot more than 120.0 percen1:w/w ()filie stated am()rrnt
Microbial contamination (2.2.9). Complies with the microbial of total polyphenols (sum of chebulagic acid, chebulic acid,
contamination tests. gallic acid, coriagin, 1,3,6 trigalloyl glucose and ellagic acid).

2510

www.webofpharma.com
IP 2010 HYDROGENATED CASTOR OIL

Description. Brown to brown powder; with characteristic odour B. acetonitrile


and bitter taste. a linear gradient programme using the conditions given
below,
Identification - flow rate. 1.5 ml per minute,
- spectrophotometer set at 270 nm,
A. Determine by thin-layer chromatography (2.4.17), coating
injection volume. 20 Ill.
the plate with silica gel GF 254.
Time mobile phase A mobile phase B
Mobile phase. A mixture of 6 volumes of ethyl acetate, 2
(in min) (per cent v/v) (per cent v/v)
volumes offormic acid, 2 volumes of toluene and 1 volume of
methanol. o 95 5
18 75 25
Test solution. Dissolve 0.5 g ofextract under examination with
100 ml methanol and filter. 25 65 35

Reference solution. A 0.1 per cent w/v solution of chebulagic 28 65 35


acid RS and 0.1 per cent w/v solution of gallic acid in 35 95 5
methanol. Inject the reference solutions (a) and (b). The test is not valid
Apply to the plate 10 III ofeach solution as bands 10 mm by 2 unless the relative standard deviation for the replicate
mm. Allow the mobile phase to rise 8 em. Dry the plate in air injections is not more than 2.0 per cent.
and examine in ultra-violet light at 254 nm: Spray the plate with Inject the reference solution and the test solution.
ferriC chloride reagent. Heat the plate at 1100 for 10 minutes
and examine the plates under 365nm and under day light. The Calculate the content of Total polyphenols by summing the
chromatographic profile of the test solution is similar to that peak areas of chebuliagic acid, el/agic acid, 1,3,6 tri gal/oyl
of the reference solution. glucose, corilagin, gallic acid and chebulic acid using
chebulagic acid. The relative retention times of various
B. In the assay the peak in the chromatogram obtained with polyphenols with reference to gallic acid and chebulagic acid
the test solution corresponds to the peak in the chromatogram are as follows.
obtained with the chebulinic acid and gallic acid.
Chebulic acid 0.80 025
Tests Gallic acid 1.00 0.32
Loss on drying (2.4.19). Not more than 5.0 per cent, determined Corlagin 2.57 0.81
on 2.0 g by drying in an oven at 105° for 3 hours. 1,3,6 Trigalloyl glucose 2.81 0.89
Total ash (2.3.19). Not more than 2.0 per cent. Chebulagic acid 3.17 1.00
Heavy metals (2.3.13). 1.0 g complies with the limit test for Ellagic acid 3.49 1.10
heavy metals, (Method B) 20 ppm. Chebulinic acid 3.64 1.15
Microbial contamination (2.2.9). Complies with the microbial Usual strength. 40 per cent w/w.
contamination test.
Storage. Store protected from heat and moisture.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve about 0.5 g of the extract containing
100 mg ofpolyphenols in 100 m1 of water,and filter.
Reference solution (a). 0.01 per cent w/v solution of Hydrogenated Castor Oil
chebulagic acid RS in water. Castor Wax; Opalwax
Reference solution (b). 0.01 per cent w/v solution of gallic
Hydrogenated Castor Oil is refined, bleached, hydrogenated
acid RS in water.
and deodorised castor oil. It consists mainly ofthe triglyceride
Chromatographic system ofhydroxystearic acid.
- a stainless steel column 25 em x 4.6 mm packed with
Description. A white to yellow powder ofuniform consistency
octadecylsilane bonded to porous silica (5 Ilm),
and texture. It may have a hard, waxy consistency.
- mobile phase:A.a buffer solution prepared by mixtures
dissolving 0.136 g of Potassium di-hydrogen Tests
orthophosphate in 500 ml of water, add 0.5 ml of
orthophosphoric acid and dilute to 1000 ml with water. Melting range (2.4.21). 85 ° to 88°, determined by Method II.

2511

www.webofpharma.com
IPECAC TINCTURE IP 2010

Free fatty acids. Weigh accurately about 20 g, melt on a water- in day light. An intense yellow, fluorescent zone of emetine is
bath, add 75 ml of hot ethanol (95 per cent), previously observed in chromatogram of test solution corresponding to
neutralised to phenolphthalein solution with 0.1 M sodium intense yellow fluorescent zone of emetine in chromatogram
hydroxide, swirl, add 1 ml of phenolphthalein solution and of reference solution.
titrate with 0.1 M sodium hydroxide, swirling vigorously until
the solution remains faintly pink after being shaken for Tests
60 seconds; not more than 11.0 mlofO.I Msodiumhydroxide
Ethanol. Not less than 24.0 per cent and not more than 28.0 per
is required.
cent.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
Determine by gas chromatography (2.4.13).
heavy metals, Method B (10 ppm).
Test solution. A 0.8 per cent v/v solution ofthe sample under
Acetyl value (2.3.22). Not less than 143.
examination and 02 per cent v/v of acteonitrile as internal
Acid value (2.3.23). Not more than 2.0. standard.
Hydroxyl value (2.3.27). 154 to 162. Reference solution. A solution in water containing 0.2 per
Iodine value (2.3.28). Not more than 5.0. cent v/v of ethanol and 2 per cent v/v of acetonitrile (internal
standard).
Peroxide value (2.3.35). Not more than 5.0.
Chromatographic system
Saponification value (2.3.37). 176 to 182. a stainless steel column 1.8 m x 3 mm, packed with
Storage. Store at a temperature not exceeding 30°. Avoid copolymer of ethylvinylbenzene and divinylbenzene
exposure to excessive heat. (100 to 150 mesh),
temperature:
column. 1200 ,
inlet port. 21 00 ,
Ipecac Tincture - a flame ionization detector at 210 0 ,
- flow rate. Adjust the carrier flow so that acetonitrile,
Ipecac Tincture obtained from roots and rhizomes ofCephaelis internal standard, elutes in 5 to 10 minutes.
ipecacuanha A. Rich (Fam. Rubiaceae).
Calculate the percentage v/v of ethanol.
Ipecac Tincture contains not less than 90 per cent w/w and
not more than 110 per cent w/w ofalkaloids ofIpecac calculated Assay. Determine by liquid chromatography (2.4.14).
as emetine. Test solution. Transfer 5 ml ofsample into a 50 ml volumetric
Description. A yellowish brown liquid. flask. Dilute to volume with O.OlM hydrochloric acid and
mix.
Identification Reference solution. A 0.010 per cent emetine hydrochloride
Determine by thin layer chromatography (2.4.17), coating the heptahydrate RS and 0.010 per cent of cephaeline in O.OlM
plate with silica gel GF 254. hydrochloric acid.

Mobilephase. A mixture of 2 volumes of ammonia, 15 volumes Chromatographic system


of methanol, 18 volumes of ethyl acetate and 65 volumes of a stainless steel column 25 cm x 4.6 mm, packed with
toluene. octadecyl silane chemically bonded to porous silica
(5!-Lm).
Test solution. Shake 2.0 ml oftincture under examination with - mobile phase: a mixture of50 volumes ofmethanol and
2 ml ofwater and 0.1 ml ofammonia. Add 10 ml ofether and 50 volumes of buffer prepared by dissolving 2.0 g of
shake. Separate the ether layer, dry it over about 2 g of I-heptane sulfonate in 500 volumes water,
anhydrous sodium sulphate & filter. - column temperature: 50 0
Reference solution. Dissolve 2.5 mg ofemetine hydrochloride - flow rate. Adjust the flow rate so that the retention time
RS in methanol and dilute to 10 ml with the same solvent. of emetine is about 14 minutes,
spectrophotometer set at 283 urn,
Applytotheplate~IO_!-LlQfeach.sQlution~as_bandsj)LLQmmby
inje~ctiatfvolurn~:tO··f.l.t
2111l]1.l\.llQW the mQl:>ilepl1l1~e t()ris~toJOCIl1' .J)ryt.l1~plat.e
in air. Spray the plate with 0.5 per cent w/v solution of iodine Injecti:he referencesoiution. fhe test IS not valid unless the
in ethanol (95 per cent v/v) solution. Heat the plate at 60 0 for relative standard deviation for replicate injections is not more
10 min and examine the plate in ultraviolet light at 365 urn and than 2.0 per cent.

2512

www.webofpharma.com
IP 2010 KALMEGH

Inject the reference solution and the test solution. layers ofmucilage are more resistant and swell to form rounded
papillae. When mounted in 0.005 M iodine, occasional simple
Calculate the content of cephaeline and emetine as total
alkaloids in the sample. and two- to four-compound starch granules, about 2 mm to
10 mm, can be seen in some ofthe cells. Occasional fragments
1 mg of emetine hydrochloride contains 0.868 mg of emetine of thick-walled, reddish brown endosperm, cells with pitted
and correction factor for cephaeline is 0.971. walls and elongated fragments ofgrey embryo may be present.
Usual strengths. 0.1 per cent w/w; 0.2 per cent w/w. Swelling power. Transfer 1 g to a 100-ml stoppered cylinder
Storage. Store protected from light and moisture. containing 90 ml of water, shake well for 30 seconds and allow
to stand 24 hours, shaking gently on three occasions during
this period. Add sufficient water to produce 100 mI, mix gently
for 30 seconds, avoiding the entrapment of air, allow to stand
Ispaghula Husk for 5 hours and measure the volume ofn:mcilage. Repeat the
determination three times. The average offour detenninations
Isapgol Husk; Plantago ovata
is not less than 40 m!.
Total ash (2.3.19). Not more than 4.5 per cent, determined on
9 1 g.
8 Acid-insoluble ash (2.3.19). Not more than 0.45 percent.
7 Loss on drying (2.4.19). Not more than 12.0 per cent,
6 determined on 0.5 g by drying in an oven at 105° for 5 hours.
5 Storage. Store protected from moisture and from attack by
insects and. rodents.

Kalmegh
o
Andrographis paniculata

Ispaghula Husk consists of the epidermis and collapsed


adjacent layers removed from the dried ripe seeds ofPlantago
ovata Forssk.(Fam. Plantaginaceae).
Description. Pale buff, brittle flakes, more or less lanceolate,
up to 2 mm long and 1 mm wide at the centre, much broken into
smaller fragments; many ofthe flakes having a small, brownish,
oval spot, about 0.8 to 1.0 mm long, in the centre; the material
swells rapidly in water, forming a stiffmucilage.

Identification
When mounted in cresol and examined under a microscope,
the particles are found to be transparent and angular, the edges
straight or curved and sometimes rolled. They are composed
ofpolygonal prismatic cells with four to six straight or slightly
curved walls; the cells vary in size in different parts of the
seed coat, from about 25 mm to 60 mm at the summit of the Kalmegh consists of the· dried aerial parts, mainly stems and
seed, that is, near and over the brown spot, to 25 mm to leaves, of Andrographis paniculata Nees. (Fam,
100 mm for the remainder ofthe epidermis except at the edges Acanthaceae).
ofthe seed, where the cells are smaller, about 45 mm to 70 mm.
Kalmegh contains not less than 1.0 per cent w/w of
When mounted in ethanol (95 per cent) and irrigated with
andrographo1ide, calculated on the dried basis.
water, t4e mucilage in the outer part of the epidermal cells
swells rapidly and goes into solution, while the two inner Description.Taste, intensely bitter.

2513

www.webofpharma.com
KALMEGH IP 2010

Identification water bath for 15 minutes, cool and filter. Reflux the residue
further with methanol till the last extract turns colorless, cool
A. Macroscopic - Mixture of crisp, dark green-coloured and filter. Combine all the filtrates and concentrate to 50.0 ml.
broken leaves and quadrangular stems; leaves brittle. Stem
fracture short, fibrous. Reference solution. A 0.1 per cent w/v solution of
andrographolide RS in methanol.
B. Microscopic - Stems quadrangular with collenchyma
strands at angles and on side; small acicular crystals ofcalcium Chromatographic system
oxalate present in pith and cortex. Trichomes 1-3 celled, - a stainless steel column 25 cm x 4.6 mm packed with
glandular hair disc-shaped and multicellular. octadecylsilane bonded to porous silica (5 Ilm),
- mobile phase: 65 volumes of methanol and 35 volumes
C. Determine by thin-layer chromatography (2.4.17), coating of water,
the plate with silica gel GF254. - flow rate. 1 ml per minute,
Mobile phase. A mixture of 7 volumes of chloroform and - spectrophotometer set at 223 nm,
1 volume of methanol. - injection volume. 20 Ill.

Test solution. Reflux 1 g ofcoarsely powdered substance under Inject the reference solution. The test is not valid unless the
examination with 50 ml methanol for 15 minutes, cool and relative standard deviation for the replicate injections is not
filter. Reflux the residue further with 2 x 50 ml of methanol, more than 2.0 per cent.
cool and filter. Combine all the filtrates and concentrate to Inject the test solution and reference solution.
lOml.
Calculate the content of andrographolide.
Reference solution. Reflux 0.5 g of kalmegh RS with 50 ml
methanol for 15 minutes, cool and filter. Reflux the residue Storage. Store protected from heat, moisture and against attack
further with 2 x 50 ml of methanol, cool and filter. Combine all by insects and rodents.
the filtrates and concentrate to 5 ml.
Apply to the plate 10 III of each solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 Dill and 365 Dill, spray Kalmegh Dry Extract
with methanolic sulphuric acid (20 per cent, v/v). Heat the
plate at 1200 for 5-10 minutes and examine in day light. The Kalmegh Dry Extract is obtained by extracting Kalmegh with
chromatographic profile of the test solution is similar to that aqueous ethanol or ethanol or any other suitable solvent.
ofthe reference solution. Kalmegh Dry Extract contains not less than 90.0 per cent w/w
and not more than 120.0 per cent w/w of the labelled amount
Tests of andrographolides (sum of andrographolide, neo-
andrographolide and andrograpanin) The content of 14-deoxy-
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
11,12-didehydroandrogapholide content shall not be more than
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent. one sixth of the amount of andrographolide.
Water-soluble extractive (2.6.3). Not less than 12.0 per cent Description. A light green to dark green powder.
by Method 1.
Total ash (2.3.19), Not more than 15 per cent.
Identification
Acid-insoluble ash (2.3.19). Not more than 3.0 per cent. A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm). Mobile phase. A mixture of 7 volumes of chloroform and
1 volume of methanol.
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°. Test solution. Dissolve 200 mg ofthe extract under examination
with 50 ml methanol and filter.
Microbial contamination (2.2.9). Complies with the microbial
Reference solution. A 0.02 per cent w/v solution of
contamination tests.
andrographo1ldeJISinliieth7inol.
Assay: DefeItrline by liqtlidchrbmatography(2:4:14): .... Apply to the plate 10 III of each solution as 10 mm
Test solution. Reflux about 2.5 g of the coarsely powdered 2 mll. Allow the mobile phase to rise 8 cm. Dry the plate in air
substance under examination with 50 ml of methanol on a and examine in ultraviolet light at 254 Dill and 365 Dill and also

2514

www.webofpharma.com
IP 2010 KUNDURU

under day light. Spray the plate with methanolic sulphuric Calculate the content of l4-deoxy-ll, l2-didehydroandro-
acid (20 per cent) and heat at 120° for 10 minutes. The grapholide and andrographolides in the extract.
chromatogram obtained with test solution shows a band
Calculate andrographolide by summing the peak areas of
corresponding to the band obtained using reference solution
andrographolide, neo-andrographolide, and andrograpanin.
indicating the presence of andrographolide.
Storage. Store protected from heat and moisture.
Tests
Acid insoluble ash (2.3.19). Not more than 3.0 percent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm). Kunduru
Loss of drying (2.4.19). Not more than 5.0 percent. Sallaki Gum; Gum of Boswellia serrata
Microbial contamination (2.2.9). Complies with the microbial
contamination test.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve about 250 mg of the extract under
examination or quantity equivalent to 20 mg of
andrographolide in methano~ by gently heating, make it up to
100.0 ml and filter.
Reference solution (a). A 0.02 per cent w/v solution of
andrographolide RS in methanol.
Reference solution (b). A 0.005 per cent w/v solution of
14-deoxy-ll,12-didehydroandrographolide RS in methanol.
Chromatographic system
- a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 /lm),
- mobile phase: A. a buffer solution prepared by Kunduru is the gum-resin from Boswellia serrata Roxb. (Fam.
dissolving 0.136 g of potassium dihydrogen Burseraceae).
orthophosphate in 500 ml of water and 0.5 ml of Kunduru contains not less than 1.0 per cent w/w of total 11-
orthophosphoric acid, dilute to 1000 ml with water, keto-~-boswellic acid and acetyl-ll-keto-~-boswellicacid,
B. acetonitrile, calculated on the dried basis.
- flow rate. 1.5 ml per minute,
Description. Translucent, brittle, whitish yellow substance,
- a linear gradient programme using the conditions given
in roundish, club-shaped, pear-shaped, or irregular tears.
below,
- spectrophotometer set at 223 nm, Identification
- injection volume. 20 /ll.
Time Mobile phase Mobile phase A. Macroscopic - Fracture dull. Slightly sticky to touch;
(in min.) (per cent v/v) (per cent v/v) odour, balsamic; taste slightly mucilaginous, bitter and
aromatic.
o 95 5
B. Determine by thin-layer chromatography (2.4.17), coating
8 55 45
the plate with silica gel GF254.
25 20 80
Mobile phase. A mixture of7 volumes of hexane and 3 volumes
30 95 5 of ethyl acetate.
Inject reference solution (b). The test is not valid unless the Test solution. Reflux I g ofcoarsely powdered substance under
relative standard deviation for the replicate injections is not examination with 50 ml methc;mol on a boiling water-bath for
more than 2.0 per cent. The relative retention time with respect 30 minutes, cool and filter. Evaporate the filtrate to dryness
to andrographolide, for neo-andrographolide is about 1.2 and and dissolve the residue in 10 ml of methanol.
for andrograpanin is about 1.6.
Reference solution. Reflux 1 g ofcoarsely powdered lamduru
Inject the test solution and reference solution (a). RS with 50 ml methanol on a boiling water-bath for 30 minutes,

2515

www.webofpharma.com
KUNDURU IP 2010

cool and filter. Evaporate the filtrate to dryness and dissolve Calculate the sum ofthe contents of ll-keto-p-boswellic acid
the residue in 10 ml of methanol. and acetyl-ll-keto-p-boswellic acid.
Apply to the plate 10 Jll ofeach solution as bands 10 rom by 2 Storage. Store protected from heat, moisture and against attack
rom. Allow the mobile phase to rise 8 cm. Dry the plate in air by insects and rodents.
and examine in ultraviolet light 254 urn and 365 urn, spray with
methanolic sulphuric acid (1 0 per cent, v/v). Heat the plate at
110° for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that Kutki
of the reference solution. Picrorhiza kurroa
Tests
9
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
8
Ethanol-soluble extractive (2.6.2). Not less than 35 per cent.
7
Total ash (2.3.19). Not more than 10.0 per cent.
6
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
5
Heavy metals (2.3.13). 1.0 g complies with the limit test for
4
heavy metals, Method B (20 ppm).
3
Water (2.3.43). Not more than 12.0 per cent, determined on
0.2g.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests. o
Assay. Determine by liquid chromatography (2.4.14).
Kutlci consists of dried roots of Picrorhiza lalrroa Royle ex
Test solution. Reflux about 2 g of the coarsely powdered
Benth.(Fam. Scrophulariaceae).
substance under examination with 50 ml of methanol on a
water bath for 15 minutes, cool and filter. Reflux the residue Kutki contains not less than 5 per cent w/w ofkutkin, calculated
further with methanol till the last extract tums colorless, cool on the dried basis.
and filter. Combine all the filtrates and concentrate to 100.0 ml.
Description. Rhizomes are sub cylindIical, straight or slightly
Reference solution (a). A 0.01 per cent w/v solution of curved, externally greyish with wrinkled surfaces, circular scars
ll-keto-p-boswellic acid RS in methanol. of roots and bud scales, with cork exposed at places.
Reference solution (b). A 0.05 per cent w/v solution of
acetyl-ll-keto-p-boswellic acid RS in methanol. Identification
Chromatographic system A. Macroscopic - 3-6 cm long and about 1 cm thick, sub-
- a stainless steel column 25 cm x 4.6 rom packed with cylindrical, straight, grayish brown, wrinkled roots. Odour is
octadecylsilane bonded to porous silica (5 l..un), pleasant and tastes bitter.
mobile phase: 90 volumes of methanol and 10 volumes
B. Microscopic - There is well-developed periderm. About
ofa mixture containing 5 ml of acetonitrile and 95 ml of
7-10 layers of cork cells are seen. Cells ofphelloderm loosely
water, adjusting the pH to 2.8 with dilute
arranged.
orthophosphoric acid,
flow rate. 1.5 ml per minute, C. Determine by thin-layer chromatography (2.4.17), coating
spectrophotometer set at 247 nm, the plate with silica gel GF254.
injection volume. 20 Jll.
Mobile phase. A mixture of 7.5 volumes of ethyl acetate,
Inject the reference solution (a) and (b). The test is not valid 2.2 volumes of methanol and 0.1 volume glacial acetic acid.
unless the relative retention times are about 0.65 for ll-keto-
Testsolution.Refluxl.. g.ofcoarselypowdered.substance~unde r
P~oo~swellicacid and 1.0 foracetyr.:n~kao,:;p~boswelIic acid examiIJatiQI), withSQml ofm?thm1Q! forI Smj!1lJt~~,c.gQLl!n~d
arid thefelativestandard deviation for theteplicateihjectioIiS
filter. Reflux the residue fmther with 50 ml of methanol, cool
is not more than 2.0 per cent.
and filter. Combine both the filtrates and concentrate under
Inject the test solution. vacuum to dryness. Extract the dried residue with 50 ml of

2516

www.webofpharma.com
IP 2010 LASUNA

methanol at 50° for 10 minutes, filter the solution and use the Inject the test solution and reference solution.
filtrate for analysis.
Calculate the content ofkutkin.
Reference solution. Reflux 5 g of leutlei RS with 50 ml of
Storage. Store protected from heat, moisture and against attack
methanol for 15 minutes, cool and filter. Reflux the residue
. by insects and rodents.
further with 50 ml of methanol, cool and filter. Combine both
the filtrates and concentrate under vacuum to dryness. Extract
the dried residue with 10 ml of methanol at 50° for 10 minutes,
filter the solution and use the filtrate for analysis. Lasuna
Apply to the plate 20 )..ll ofeach solution as bands 10 rom by 2
Garlic; Allium sativum bulb
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with anisaldehyde sulphuric acid reagent. Heat the plate at
100° for 10 minutes and examin in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 15.0 per cent.
Water-soluble extractive (2.6.3). Not less than 25.0 per cent
by method I.
Total ash (2.3 .19). Not more than 6.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 1.0 per cent.
Lasuna consists of the fresh or dried compound bulbs of
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Allium sativum Linn. (Fam. Liliaceae).
heavy metals, Method B (20 ppm).
Lasuna contains not less than 0.2 per cent w/w of alliin,
Loss on drying (2.4.19). Not more than 5 per cent, determined
calculated on the dried basis.
on 5 g by drying in an oven at 105°.
Description. Bulbs made up of cloves and is wrapped in a
Microbial contamination (2.2.9). Complies with the microbial white papery sheath with pungent taste and odour
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Identification

Test solution. Dissolve 100 mg of the substance under A. Macroscopic - Each bulb has several cloves which are
examination in 25.0 ml of methanol, filter. arranged in concentric rings and enclosed in a shining white
or pinkish papery envelope. The cloves are attached to a flat,
Reference solution. A 0.1 per cent wN solution of leut/an RS circular hard disc with numerous thin wiry roots from its
in methanol. underside and short, cylindrical out growth from the upper
Chromatographic system surface. Each clove is ovoid and further covered by papery
sheath with a tail like structure at one and opposite to its
a stainless steel column 15 cm x 4.6 rom packed with
attachment. The cloves in the outer ring are loose and white in
octadecylsilane bonded to porous silica (5 )..lm),
colour where as cloves in the inner ring are adherent and pale
mobile phase: 83 volumes of 1 per cent v/v.
pinkish in colour.' Each clove covered with a white scale leaf
orthophosphoric acid in water and 17 volumes of
and a pinkish white epidermis, easily separated from the solid
acetonitrile, portion. In the middle of the bulb a hollow, cylindrical, linear
flow rate. 1 ml per minute, remnant of the scape is seen.
spectrophotometer set at 280 nm,
B. Microscopic The protective leaf contains an epiderniis
injection volume. 20 )..l1.
enclosing a mesophyll free from chlorophyll. The outer
Inject the reference solution. The relative standard deviation epidermis consists of lignified sclereid cells of thick, pitted
for the replicate injections is not more than 2.0 per cent. walls, elongated, covered with thin cuticle. The cortical cells

2517

www.webofpharma.com
LASUNA IP2010

are thick walled, non lignified, tending to collapse on maturity, Microbial contamination (2.2.9). Complies with the microbial
isodiametric and contain purple pigments. The vascular contamination tests.
bundles consist of lignified spiral and annular vessels. The Assay. Determine by liquid chromatography (2.4.14).
storage leaves show an outer epidermis of thin, delicate cells
ofvariable shape. Stomata are present on the outer epidermis Test solution. Weigh accurately about 2 gofthe freshly peeled
only at extreme tip near the base of the foliage leaves.The substance under examination. Add 15 ml of hot water and
mesophyll consists ofswollen storage parenchyma cells filled grind in a porcelain mortor and filter. Grindthe residue further
with fme granular reserve material. with 2 x 15 ml ofhot water and filter. Combine all the filtrates
and make up to volume 50 ml with distilled water.
C. Determine by thin-layer chromatography (2.4.17), coating
Reference solution. A 0.004 per cent w/v solution of alliin RS
the plate with silica gel GF254.
in water.
Mobile phase. A mixture of 3 volumes of butyl alcohol,
Chromatographic system
1 volume of n-propyl alcohol, 1 volume ofglacial acetic acid
- a stainless steel column 25 cm x 4.6 mm packed with
and 1 volume of water.
octadecylsilane bonded to porous silica (5 Ilm),
Test solution. Reflux 1 g ofsubstance under examination with - mobile phase: 0.1 per cent v/vphosphoric acid prepared
20 ml of methanol (50 per cent) for 10 minutes, cool and filter. by diluting 1ml ofphosphoric acid to 1000 ml with water,
Reflux the residue with another 20 ml of methanol (50 per - flow rate. 0.5 ml per minute,
cent), cool and filter. Combine all the filtrates and concentrate - spectrophotometer set at 210 urn,
under vacuum to 5 ml. - injection volume. 20 Ill.

Reference solution. Reflux 1 g of lasuna RS with 20 ml of Inject the reference solution. The test is not valid unless the
methanol (50 per cent) for 10 minutes, cool and filter. Reflux relative standard deviation for the replicate injections is not
the residue with another 20 ml of methanol (50 per cent), cool more than 2.0 per cent.
and filter. Combine all the filtrates and concentrate under Inject the test and reference solution.
vacuum to 5 m!.
Calculate the content of alliin.
Apply to the plate 10 III of each solution as bands 10 mm by 2
Storage. Store protected from heat, moisture and against attack
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
by insects and rodents.
and examine in ultraviolet light at 254 urn and 365 urn, spray
with 0.2 per cent w/v solution of ninhydrin in mixture of 95
volumes of butyl alcohol and 5 volumes of 2 M acetic acid.
Heat the plate at 100° to 105° for about 10 minutes and examine Lavang
in day light. The chromatographic profile ofthe test solution
Syzigium aromaticum
is similar to that ofthe reference solution.
D. Transfer about 10 g of garlic bulbs that have been cut into
small pieces to a beaker, add 10 ml of 1 M sodium hydroxide 9
and 10 ml of water, heat the beaker in a boiling water for 8
10 minutes, cool and filter. To 2 ml ofthis filtrate add few drops 7
of freshly prepared solution of sodium nitroprusside.
6
Appearance ofa red or orange red colour indicates the presence
of sulphur containing compounds in the sample. 5

4
Tests
3
Foreign organic matter (2.6.1). Not more than 2.0 per cent. 2

Total ash (2.3.19). Not more than 5.0 percent.


Acid-insoluble ash (2.3.19). Not more than 1.0 per cent. o
Heavymefals (2.3.n): LO gcompliesWithtnelimit test for Liiviiiig consists·ofdriea-floweroudsofSyzigiulrfCi;'OmaticuiiI
heavy metals; Method B (20 ppm). (L.) Merr. & L.M. Perry. (Fam.Myrtaceae}-
Loss on drying (2.4.19). Not more than 65.0 per cent for fresh Lavang contains not less than 7.0 per cent w/w of eugenol,
bulbs, determined on 5 g by drying in an oven at 105°. calculated on the dry basis.

2518

www.webofpharma.com
IP 2010 MALT EXTRACT

Description. Aromatic odour and spicy taste. Test solution. A 0.5 per cent w/v of substance under
examination in methanol.
Identification
Reference solution. A 0.04 per cent w/v of eugenol RS in
A. Macroscopic-The flower bud is reddish brown in colour methanol.
and consist of quadrangular stalked portion, the hypanthium,
Chromatographic system
surrounded by four lobes of sepals.
a stainless steel 30m xO.53mm capillary column
B.Microscopic-Epidermis followed by thick temperature:
cuticle,characteristic zone of bilateral vascular bundles, column.70° for 1minute, then increased to 100° at a rate
numerous schylogenous oil gl!mds present beneath the of 10° per minute, then increased to 200° at a rate of20°
epidermis. Central collumella and air spaces or lacuna in per minute and maintained at this temperature for 2
parenchyma regeion. minutes,
inlet port. 220°,
C. Determine by thin-layer chromatography (2.4.17), coating
detector. 250°,
the plate with silica gel GF254.
flow rate 1.5 ml per minute ofthe carrier gas.
Mobile phase. A mixture of 93 volumes of toluene and 7 Inject reference solution and the test solution.
volumes of ethylacetate.
Calculate the eugenol content in the substance under
Test solution. Reflux 2.0 g ofthe coarsely powdered substance examination using the ratios ofthe area ofpeak corresponding
under examination with 50 ml dichloromethane for 15 minutes, to eugenol to the area of the peak obtained with test solution
cool and filter. Reflux the residue further for two times with and the reference solution.
50ml dichloromethane, coo1and filter. Combine all the filtrates
Storage. Store protected from light, moisture and against
and concentrate under vacuum to 25 ml.
attack by rodents and insects.
Reference solution. Reflux 1.0 g of the coarsely powdered
Lavang RS with 25 ml dichloromethane for 15 minutes, cool
and filter. Reflux the residue further for two times with 50 ml
dichloromethane, cool and filter. Combine all the filtrates and Malt Extract
concentrate under vacuum to 10 ml.
Malt Extract is a product obtained by extracting malted grains
Apply to the plate 10 III ofeach solution as bands 10 mm by 2 of cereals (barley, cholam or wheat) with water at a suitable
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air temperature and evaporation of the strained liquid until a
and examine in ultraviolet light at 254 mn. Spray the plate with viscous product is obtained. Itmay be mixed with 10 per cent
a vanillin sulphuric acid reagent. Heat the plate at 105° for 5 by weight ofGlycerin.
minutes and examine the plate at 365 mn and in day light. The Malt Extract contains nitrogen equivalent to not less than
chromatographic profile of the test solution is similar to that
4.0 per cent w/w ofprotein.
of the reference solution.
Description. A sweet, viscous, light brown liquid; odour,
Tests pleasant and characteristic.
Foreign organic matter (2.6.1 ).Not more than 2.0 per cent. Tests
Ethanol soluble extractive (2.6.2). Not less than 7.0 per cent.
Refractive index (2.4.27). 1.489 to 1.498, determined at 20°.
Water soluble extractive (2.6.3). NotJess than 20.0 per cent
by Method 1. Arsenic (2.3.10). Dissolve 10.0 g in 10 ml of water, add 10ml
of brominated hydrochloric acid, allow to stand for 5 minutes
Total ash (2.3.19). Not more than 7.0 per cent. and remove the excess ofbromine with a few drops ofstannous
Acid-insoluble ash (2.3.19). Not more than 1.0 per cent. chloride solution AsT. The resulting solution complies with
Heavy metals (2.3.13). LOg complies with the limit test for the limit test for arsenic (1 ppm). '
heavy metals, Method B (20ppm): Lipase. To 95 ml of water add 6.5 ml of triacetin and 0.2 mlof
Loss on drying (2.4.19). Not more than 12.0 per cent determined a 0.1 per cent w/v solution of bromocresol purple, neutralise
on 5.0 g by drying in an oven at 105°. with 0.5 M sodium hydroxide and add sufficient water to
produce 110 ml. Place 50 ml of this solution in each of two
Microbial contamination (2,2.9). Complies with the microbial large test-tubes (20 cm X 3 cm)AandB keptina water-bath at
contamination tests.
30° ± 1°. Insert in each tube a rubber stopper having two
Assay. Determine by gas chromatography (2.4.14). holes, one for the tip of a burette and the other for a short

2519

www.webofpharma.com
MANDUKAPARNI IP 2010

glass tube through which passes a thread operating a glass B. Microscopic - Animocytic stomata on both surfaces,
stirring coil. Stir the contents of the tube untilthey attain the more on lower surface; petiole epidermis has calcium oxalate
temperature of the water-bath. Prepare a solution of 5.0 g of prisms; vascular bundles seven arranged in 'U' shape,
the substance under examination in 10 ml of water. To tube A parenchyma cells contain simple and compound starch
add 1 ml of this solution; to tube B add 1 ml of this solution granules.
after previous boiling. Adjust and maintain the pH ofthe two
C. Determine by thin-layer chromatography (2.4.17), coating
tubes to between 6.2 and 6.4 by the dropwise addition of 0.05
the plate with silica gel GF254.
M sodium hydroxide, stirring frequently. After 6 hours, the
difference between the amounts of 0.05 M sodium hydroxide Mobile phase. A mixture of 60 volumes of chloroform,
added to the tubes is not more than 1.0 ml. 32 volumes of glacial acetic acid, 12 volumes of methanol
and 8 volumes of water.
Assay. Weigh accurately about 5.0 g into a 200-rnllong-necked
flask and carry out the determination of nitrogen, Method A Test solution. Reflux 2 g ofcoarsely powdered substance under
(2.3.30), using 0.05 M sulphuric acid instead of 0.1 M examination with 50 ml methanol for 15 minutes, cool and
sulphuric acid. filter. Reflux the residue further with 2 x 50 ml of methanol,
1 ml of 0.05 Msulphuricacidis equivalentto 0.001401 g ofN cool and filter. Combine all the filtrates and concentrate to
and multiply the result by 6.25 to obtain the protein content. 10 ml under reduced pressure.

Storage. Store protected from moisture. Reference solution. Reflux 1 g of mandukaparni RS with
50 rnl methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 x 50 ml of methanol, cool and filter. Combine all
the filtrates and concentrate to 5 ml.

Mandukaparni Apply to the plate 10 )ll ofeach solution as bands 10 rom by 2


rom. Allow the mobile phase to rise 8 cm. Dry the plate in air
Gotu Kola; Centella asiatica and examine in ultraviolet light at 254 nm and 365 nm, spray
with anisaldehyde sulphuric acid reagent. Heat the plate at
100° for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 15.0 per cent
by Method I.
Total ash (2.3.19). Not more than 24 per cent.
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Mandukaparni consists of the dried aerial parts of Centella
asiatica (Linn.) Urban. (Fam. Umbelliferae). Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°.
Mandukaparni contains not less than 0.5 per cent w/w of
asiaticoside, calculated on the dried basis. Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Description. A green to greenish yellow in colour, taste,
slightly bitter and sweet. Assay. Detelmine by liquid chromatography (2.4.14).

Identification· Test sdutiQIJ. RdhlJI. alwut 19_Qf the.-J<9arseJY_P-QW<;l~.l'ed


substance under examination with 50 ml of methanol on a
_ ..• __ .••.•.••• .• __• • - - w·· ·_.____ _ ~ __ • _ ...• _ _ _ . _ _ _ __ _ _ _ .... ~

A. Macroscopic - A slender trailing herb with rooted nodes water bath for 15 minutes, cool and filter. Reflux the residue
and internodes. Leaves with elongated petioles and sheathing further with methanol till the last extract turns colorless, cool
leafbases; lamina reniform with crenate margin. and filter. Combine all the filtrates and concentrate to 100.0 rnl.

2520

www.webofpharma.com
IP 2010 MANJISTHA

Reference solution. A 0.1 per cent w/v solution of C. Determine by thin-layer chromatography (2.4.17), coating
asiaticoside RS in methanol. the plate with silica gel GF254.
Chromatographic system Mobile phase. A mixture of7 volumes of toluene, 25 volumes
- a stainless steel column 25 cm x 4.6 rom packed with of ethyl acetate and 0.5 volumes glacial acetic acid.
octylsilane bonded to porous.silica (10 J..1m),
Test solution. To 4 g of coarsely powdered substance under
mobile phase: 25 volumes of acetonitrile and 75 volumes
examination, add 100 ml of methanol, reflux for 15 minutes,
of water,
cool and filter. Reflux the residue further for two times with 50
flow rate. 1.5 ml per minute,
ml methanol, cool and filter. Combine all the filtrates and
spectrophotometer set at 210 nm,
concentrate under vacuum to 100 ml.
injection volume. 20 J..1l.
Reference solution. To I g of coarsely powdered manjistha,
Inject the reference solution. The test is not valid unless the
add 100 ml of methanol, reflux for 15 minutes, cool and filter.
relative standard deviation for the replicate injections is not
Reflux the residue further for two times with 50 ml methanol,
more than 2.0 per cent.
cool and filter. Combine all the filtrates and concentrate under
Inject the reference solution and the test solution. vacuum to 25 ml.
Calculate the content of asiaticoside. Apply to the plate 20 J..11 of each solution as bands 10 mm by 2
Storage. Store protected from heat, moisture and against attack mm. Allow the mobile phase to rise 8 em. Dry the plate in air
by insects and rodents. and examine in ultraviolet light at 254 urn and 365 urn, spray
the plate with anisaldehyde sulphuric acid reagent. Heat the
plate at 100° for 5 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
Manjistha of the reference solution.
Indian Madder; Rubia cordifolia Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
9
Ethanol-soluble extractive (2.6.2). Not less than 4.0 per cent.
8
Water-soluble extractive (2.6.3). Not less than 20.0 per cent
7
by method I.
6
Total ash (2.3.19). Not more than 10.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 0.5 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 5 per cent, detennined
on 5 g by drying in an oven at 105°.
o Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Manjistha consists of dried stem of Rubia cordifolia Assay. Determine by liquid chromatography (2.4.14).
Linn.sensu Hook.£ (Fam. Rubiaceae).
Test solution. Weigh 4 g of coarsely powdered substance
Manjistha contains not less than 0.02 per centw/w ofrubiadin, under examination, add 100 ml of methanol, reflux on a water-
calculated on the dried basis. bath 15 minutes, cool and filter. Reflux the residue 2 x 50 ml
Description. A cylindrical, slightly flattened, wiry pieces of methanol, cool and filter. Combine all the filtrates and
brown to purple coloured root with mild bitter taste. concentrate under vacuum to 100.0 m!.
Reference solution. A 0.002 per cent w/v solution of rubiadin
Identification
RS in methanol.
A. Microscopic - Exfoliating cork, consisting of4-12 or more Chromatographic system
layered radially arranged, thin walled cells. a stainless steel column 25 em x 4.6 rom packed with
B. Macroscopic ~ Stem, slender, cylindrical, wiry and about octadecylsilane bonded to porous silica (5 J..1m),
0.5 cm thiclc. It is brown to purple coloured and with mobile phase: A. a buffer solution pH 2.5 prepared by
longitudinal cracks. dissolving 0.136 g of potassium di-hydrogen

2521

www.webofpharma.com
MANJISTHA IP 2010

orthophosphate in 900 ml of water, add 0.5 ml of Identification


orthophosphoric acid and dilute to 1000 ml with water,
B. acetonitrile, A. Macroscopic - The fruits are globular or oblong, 4-6 mm
- a linear gradient programme using the conditions given in diameter. The outer cover is blackish brown, with raised
below, reticulated wrinkles. One seeded, seeds are white and hollow.
- flow rate. 1.5 ml per minute, B. Microscopic - The fruit has a well differentiated pericarp,
- spectrophotometer set at 278 om, testa and perisperm. Isolated, tangentially elongated oil cells
- injection volume. 20 J..tl. are in the outer region of the mesocarp. Endocarp has beaker
Time Mobile phase A Mobile phase B shaped stone cells and numerous polyhedral masses of starch
(in min.) (per cent v/v) (per cent v/v) grains. Testa has a single layer of yellow colored cells.
o 65 35 C. Determine by thin-layer chromatography (2.4.17), coating
5 50 50 the plate with silica gel GF254.
15 20 80 Mobile phase. A mixture of60 volumes of benzene, 30 volumes
30 20 80 of ethyl acetate and 10 volumes of diethyl ether.
35 50 50 Test solution. To 2 g of the coarsely powdered substance
40 65 35 under examination, add 50 ml of methanol and reflux for
15 minutes, cool and filter. Reflux the residue further for two
Inject the reference solution. The relative standard deviation
times with 75 ml of methanol, cool and filter. Combine all the
for the replicate injections is not more than 2.0 per cent.
filtrates and concentrate under vacuum to 50 ml.
Inject the test solution and reference solution.
Reference solution. To 2 g of the maricha RS, add 50 ml of
Calculate the content of rubiadin. methanol and reflux for 15 minutes, cool and filter. Reflux the
residue further for two times with 75 ml of methanol, cool and
Storage. Store protected from heat, moisture and against attack
filter. Combine all the filtrates and concentrate under vacuum
by insects and rodents.
t050ml.
Apply to the plate 10 J.!l ofeach solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 em. Dry the plate in air
Maricha and examine in ultraviolet light at 254 nm and 365 om, spray
Black pepper; pepper; Piper nigrum the plate with vanillin sulphuric acid reagent. Heat the plate
at 100° for 5-10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that
9 of the reference solution.
8
Tests
7
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
6
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
5
Water-soluble extractive (2.6.3). Not less than 6.0 per cent by
4
method I.
3 Total ash (2.3.19). Not more than 7.0 percent.
2 Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
o heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
Maricha consists of the unripe fruits of Piper nigrum Linn. determined on 5 g by drying in an oven at 105°.
(Fam. Piperaceae). Microbial contamination (2.2.9). Complies with the microbial
Maricha contains not less than 2.5 per cent w/w of piperine, contamination tests.
ciilCl.ihited on the drieu basis. Assay. I>eterrnme by Hqui(lclrr()niatognlphy (2.4. 14): ..
Description. Fruits are globular or oblong. They have a blackish Test solution. Weigh 2 g of coarsely powdered substance
brown cover, with raised reticulated wrinkles. The odour is under examination, add 50 ml of methanol, sonicate for
aromatic and the taste is strong and pungent. 3 minutes and heat on a boiling water bath for 15 minutes, cool

2522

www.webofpharma.com
IP 2010 METHI

and dilute to 100.0 ml with methanol and filter. Dilute further if a reflux condenser and boil for 2 hours. Cool, add 30 ml of
necessary. water and warm on a water-bath for 15 minutes with occasional
Reference solution. A 0.01 per cent w/v solution of shaking. Transfer the contents of the flask to a separating
piperine RS in methanol. funnel, reject the water layer and wash the remaining oil with
water until the last washing no longer shows acid reaction.
Chromatographic system Dry the resulting oil by shaking with 2 g of anhydrous sodium·
- a stainless steel column 25 cm x 4.6 mm packed with sulphate, allow to stand for 30 minutes and filter through a
octadecylsilane bonded to porous silica (5 Ilm), dry filter paper. Weigh accurately about 1.5 g of the dry
- mobile phase.A. a buffer solution pH 2.5 prepared by acetylated oil, add 3 ml of ethanol (95 per cent) and 0.1 mlof
dissolving 0.136 g of potassium di-hydrogen phenolphthalein solution and dropwise, 0.5 M ethanolic
orthophosphate in 500 ml of water, add 0.5 ml of potassium hydroxide until the solution acquires a faint pink
orthophosphoric acid and dilute to 1000 ml with water, colour. Add a further 20.0 ml of the alkali, attach a reflux
- flow rate. 1.5 ml per minute, condenser and boil for 1 hour on a water-bath. Cool, add 1 ml
B.acetonitrile, of phenolphthalein solution and titrate the excess of alkali
- a linear gradient programme using the conditions given . with 0.5 M hydrochloric acid: Repeat the operation with the
below, same quantities of the same reagents in the same manner
- spectrophotometer set at 345 Dill, without the oil and calculate the amount oftotal menthol from
- injection volume. 20 Ill. the following expression.
Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per centv/v) . (a-b)x7.813
Total menthol (per cent) = ----''-----'-----
o 95 5 S -(a-b)xO.021
18 55 45 where, S is the amount, in g, ofthe acetylated sample taken, a
25 20 80 is the amount, in ml, of 0.5 M hydrochloric acid consumed in
Inject the reference solution. The relative standard deviation the blank test, and b is the amount, in ml, of 0.5 M
for the replicate injections is not more than 2.0 per cent. hydrochloric acid consumed in saponification of the
acetylated oil.
Inject the test solution and reference solution. The relative
retention time ofpiperine is 1. Storage. Store protected from light and moisture.
~alculate the content ofpiperine.
Storage. Store protected from moisture and against attack by
insects and rodents.
Methi
Trigonella foenum-graecum

Mentha Oil
9
Mentha
8
Mentha Oil is the volatile oil distilled with steam from various 7
species of Mentha (Fam. Labiatae) and rectified ifnecessary.
6
Mentha Oil contains not less than 50.0 per cent w/w of total
menthol, CIOHzoO.
Description. A colourless or yellowish, clear liquid; odour,
characteristic and pleasant. 3

Tests
Acidity or alkalinity. A solution of 1 ml in 3.5 ml of ethanol
o
(70 per cent) is neutral to litmus.
Weight per ml (2.4.29). 0.892 gtoO.910 g. Methi is dried, ripe seeds of Trigonella foenum-graecum L.
Specific optical rotation (2.4.22). -18.00 to-33.0°. (Fam.Fabaceae).
Assay. Place about 10.0 gin an acetylation flask, add 10 mlof Methi contains not less than 0.1 per cent w/w of trigonelline
acetic anhydride and 1 g of anhydrous sodium acetate, attach on dry basis.

2523

www.webofpharma.com
METHI IP 2010

Description. The seed is hard, flattened, brown or reddish- Transfer 1g to a 100 ml stoppered cylinder containing 90 ml of
brown and more or less rhomboidal with rounded edges. The water, shake well for 30 seconds and allow to stand 24 hours,
widest surfaces are marked by a groove that divides the seed shaking gently on three occasions during this period. Add
into 2 unequal parts. The smaller part contains the radicle; the sufficient water to produce I00 ml, mix gently for 30 seconds,
larger part contains the cotyledons. They have a strong avoiding the entrapment of air, allow to stand for 5 hours and
aromatic characteristic odour. measure the volume of mucilage. Repeat the determination
three times. The average of four determinations is not less
Identification than40ml.
A. Macroscopic - Seed is rhomboidal with rounded edges. Ash (2.3.19). Not more than 5.0 per cent.
It is 3 mm to 5 rom long, 2 rom to 3 rom wide and 1.5 rom to 2 rom Loss on drying (2.4.19). Not more than 12.0 per cent,
thick. The widest surfaces are marked by a groove that divides determined on 1 g by drying in an oven at 105°.
the seed into two unequal parts. The smaller part contains the
radicle; the larger part contains the cotyledons.
B. Reduce to a powder. The powder is yellowish-brown.
Examine under a microscope using chloral hydrate solution.
Neem
The powder shows fragments of the testa in sectional view Azadirachta indica
with thick cuticle covering lageniform epidermal cells, with an
underlying hypodermis of large cells, narrower at the upper
9
end and constricted in the middle, with bar-like thickenings of
the radial walls; yellowish-brown fragments of the epidermis 8
in surface view, composed of small, polygonal cells with 7
thickened and pitted walls, frequently associated with the
6
hypodermal cells, circular in outline with thickened and closely
beaded walls; fragments ofthe hypodennis viewed from below, 5

composed ofpolygonal cells whose bar-like thickenings extend 4


to the upper and lower walls; parenchyma of the testa with 3
elongated, rectangular cells with slightly thickened and beaded
2
walls; fragments of endosperm with irregularly thickened,
sometimes elongated cells, containing mucilage. 1

C. Detennine by thin-layer chromatography (2.4.17), coating o


the plate with silica gel GF254.
Neem consist of dried leaves of Azadirachta indica A.Juss.
Mobile phase. A mixture of 30 volumes of water and 70 (Fam. Meliaceae).
volumes of methanol.
Neem leaves contain not less than 1.0 per cent w/w of the
Test solution. To 1.0 g of the powdered substance under stated amount of rutin, calculated on the dry basis.
examination, add 5.0 ml ofthe methanol. Heat on a water-bath
at 65° for 5 minutes, cool and filter. Description. Characteristic odour and bitter taste.
Reference solution. Dissolve 3 mg of trigonelline Identification
hydrochloride in 1.0 ml of methanol.
A. Macroscopic-The leaves dark green, the petioles are
Apply to the plate 20 III ofeach solution as bands 10 mm by 2 short.The shape of mature leaflets is more assymetrical and
mm. Allow the mobile phase to rise 10 cm. Dry the plate in air their margins are dentate with the exception of the base of
and examine in ultraviolet light at 254 nm and 365 nm, spray their basiscopal half, which is very strongly reduced and
with potassium iodobismuthate solution. Heat the plate at cuneate or wedge-shaped leaves.
105° for 10 minutes and examine in day light. The
chromatographic profile of the test solution is similar to that B. Microscopic-Upper and lower epidermis, exhibiting two
of the reference solution. It also shows in its upper half, a layers ofpalisade cells below the upper epidermis. The spongy
broad light brownish-yellow zone (triglycerides). parenchyma with intercellular spaces abundant on the border
line of palisade cells. Midrib shows numerous
Tests ConeiiCliym~atouscen1n)elowi.Ipperand~loweTepidermis:A

Swelling Power. Not less than 6.0, determined on the powdered characteristic Zone· of vascUlar bundles is present.
substance under examination, determined by the following C. Determine by thin-layer chromatography (2.4.17) coating
method. the plate with silica gel GF254.

2524

www.webofpharma.com
IP 2010 OPIUM

Mobile phase. A mixture of 25 volumes of ethyl acetate, 15 Inject the reference solution .The test is not valid unless the
volumes of n-butanol ,5 volumes of formic acid and 5 relative standard deviation for the replicate injections is not
volumes of water. more than 2 per cent.
Test solution. Reflux 5.0 g ofthe coarsely powdered substance Inject the reference solution and the test solution.
under examination with 100 ml of n-hexane for 2 hours, cool
Calculate the content of rutin.
and filter. Reflux the residue further for 1hour with 100 ml of
methanol, cool and filter. Concentrate under vacuum to 25ml. Storage. Store protected from heat, moisture and against attack
by rodents and insects.
Reference solution. Reflux 2.5g ofthe coarsely powdered drug
with 100 ml n-hexane for 2 hours, cool and filter. Reflux the
residue further for Ihour with 100 ml of methanol, cool and
filter. Concentrate under vacuum to 10 ml. Opium
Apply to the plate 1Of.ll of each solution as bands 10mm by 2 Raw Opium; Papaver somniferum
mill. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254. Spray the plate with
anisaldehyde sulphuric acid reagent. Heat the plate at 105°
for 5 minutes and examine the plate at 365 urn and in day light. 9

The chromatographic profile oftest solution is similar to that 8


of the reference solution. 7
Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent. 5

Ethanol soluble extractive (2.6.2). Not less than 7.0 per cent. 4

Water soluble extractive (2.6.3). Not less than 19.0 per cent 3
by Method I.
Total ash (2.3.19). Not more than 12.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent. o
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20ppm). Opium is the air-dried latex obtained by incision from the unripe
Loss on drying (2.4.19).Not more than 12.0 per cent, determined capsules of Papaver somniferum Linn (Fam. Papaveraceae).
on 5.0 g by drying in an oven at 105°.
Opium contains not less than 10.0 per cent w/w of morphine,
Microbial contamination (2.2.9). Complies with the microbial C 17H I9N03, and not less than 2.0 per cent w/w of codeine,
contamination tests. ClsH2IN03, both calculated on the dried basis.
Assay. Determine by liquid chromatography (2.4.14). Description. Masses of various sizes which tend to be soft
Test solution. Reflux 5.0 g ofthe coarsely powdered substance and shiny and, after drying, hard and brittle; usually in
under examination with 100 ml of n-hexane for 2 hours, cool somewhat irregularly shaped masses (natural opium) or
and filter. Reflux the residue further for 1 hour with 100 ml of moulded into masses of more uniform size and shape
methanol, cool and filter. Concentrate under vacuum to 25ml. (manipulated opium); colour, blacldsh brown; odour, strong
and characteristic.
Reference solution. A 0.05 percent w/v of rutin RS in
methanol. Identification
Chromatographic system
Strip off any covering, cut the substance under examination
stainless steel column 25cm x4.6mm packed with
into thin slices, ifnecessary, dry at about 60° for 48 hours and
octadecylsilane bonded to porous silica(5f.lm),
reduce to a powder.
mobile phase:30 volumes of acetonitrile and 70 volumes
of 0.5 per cent formic acid prepared by diluting 5ml of A. When examined under a microscope, a suspension in a
formic acid to 1000ml with water. 2 per cent w/v solution of potassium hydroxide appears as
flow rate, 1ml per minute, granules of latex agglomerated in irregular masses and light
spectrophotometer set at 340nm, brown elongated filaments. Some fragments of vessels and
injectiion volume.20 f.ll. rather elongated, refringent crystals are also visible, as well as

2525

www.webofpharma.com
OPIUM IP 2010

a smaller number ofround pollen grains and fragments of Chromatographic system as described in the Assay.
elongated fibres. Hairs of various lengths with sharp points
The test is not valid unless the capacity factor for thebaine is
and a few grains of starch introduced during the handling of
at least 3.0 and the number of theoretical plates is at least
the latex may be present. Fragments of epicarp consisting of
3000. Calculate the percentage content of thebaine from the
polygonal cells with thick walls defining a stellate lumen may
expression given in the assay.
also be present.
Total ash (2.3.19). Not more than 6.0 percent, determined on
B. Determine by thin-layer chromatography (2.4.17), coating
0.5g.
the plate with silica gel G.
Loss on drying (2.4.19). Not more than 15.0 per cent,
Mobile phase. A freshly prepared mixture of 20 volUmes of
determined on 0.25 g cut into thin slices, by drying in an oven
acetone, 20 volumes of toluene, 3 volumes of ethanol (95 per
at 105° for 4 hours.
cent) and 1 volume of strong ammonia solution.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Triturate OJ g of the powdered substance with
5 ml of ethanol (70 per cent), add 3 ml of ethanol (70 per Test solution. Suspend about 1.0 g, accurately weighed
cent), transfer to a 25-ml conical flask and heat in a water-bath substance under examination, cut into thin slices, in 50 ml of
at 50° to 60° for 30 minutes, with stirring. Cool, filter, wash the ethanol (50 per cent), mix with the aid of ultrasound for
filter with ethanol (70 per cent) and dilute the filtrate to 10 ml 1 hour, allow to cool, dilute to 100.0 ml with the same solvent
with the same solvent. and allow to stand. To 10.0 ml of the supernatant liquid add
5 ml of ammonia buffer pH 9.5, dilute to 25.0 ml with water,
Reference solution. Dissolve 2 mg of papaverine
mix, transfer 20.0 ml ofthe solution to a column (about 15 cmX
hydrochloride RS, 12 mg of codeine phosphate RS, 12 mg of
30 mm) containing 15 g of kieselguhr for column
noscapine hydrochloride RS, and 25 mg of morphine
chromatography and allow to stand for 15 minutes. Elute with
hydrochloride RSin ethanol (70 per cent) and dilute to 25 ml
two quantities, each of 40 ml, of a mixture of 85 volumes of
with the same solvent.
dichloromethane and 15 volumes of 2-propanol, evaporate
Apply to the plate 20 ).tl of each solution as 20 mm bands. the eluate to dryness at 40° at a pressure of2 kPa, transfer the
After development, dry the plate at 100° to 105° for 15 minutes, residue to a 25-ml volumetric flask with the aid ofthe mobile
allow to cool and spray with potassium iodobismuthate phase and dilute to volume with the same solvent.
solution and then with a 0.4 per cent w/v solution ofsulphuric
Reference solution. Weigh accurately 0.1 g of morphine
acid. The chromatogram obtained with the reference solution
hydrochloride and 25 mg of codeine, dissolve in sufficient of
shows in the lower part an orange-red or red band (morphine),
the mobile phase to produce 25.0 ml and dilute 10.0 ml ofthis
above it a similarly coloured band (codeine) and in the upper
solution to 100.0 ml with the same solvent.
part an orange-red or red band (papaverine) and above it a
similarly coloured band (noscapine). The chromatogram Chromatographic system
obtained with the test solution shows orange-red or red bands a stainless steel column 25 cm x 4.6 mm, packed with
corresponding to those in the chromatogram obtained with octylsilane bonded to porous silica (5 ).tm), fitted with a
the reference solution. The chromatogram obtained with the guard column 4 cm x 4.6 mm packed with octylsilane
test solution may also show a dark red band (thebaine) situated bonded to porous silica (5 ).tm),
between those due to codeine and to papaverine. mobile phase: 1.0 g of sodium heptanesulphonate in
420 ml of water, adjusting to pH 3.2 with phosphoric
C. To 1 g ofthe powdered substance add 5 ml of water, shake
acid that has been diluted to contain 0.49 per cent w/v
for 5 minutes, filter and add to the filtrate 0.25 ml offerric
solution of H3P04 (about 5 ml) and adding 180 ml of
chloride solution; a red colour develops which does not
acetonitrile,
disappear on the addition of 0.5 ml of 2 M hydrochloric acid.
flow rate. 1.5 ml per minute,
Tests spectrophotometer set at 280 nID.
Inject suitable volumes of each solution. The assay is not
Thebaine. Not more than 3 per cent, calculated on the dried
valid unless the resolution between the peaks corresponding
basis.
to morphine and codeine is at least 2.5. If necessary, adjust
Determine by liquid chromatography (2.4.14). the volume of acetonitrile in the mobile phase. Iiiject the
Teslsoluiion:Use·fheTesf·soliifioii·preparediii·the·assay. reference solution six times; The assayisnotvalid unless the
relative standard deviation ofthe peak area for morphine is
Reference solution. Dissolve 25 mg of thebaine in sufficient
not more than 1.0 per cent.
mobile phase to produce 25.0 ml and dilute 10.0 ml of this
solution to 100.0 mlwith the mobile phase. Inject the test solution and the reference solution.

2526

www.webofpharma.com
IP 2010 OPIUM POWDER

Calculate the percentage content of each alkaloid from the wide, in groups of from one to six, traversing a spongy
expression parenchyma of thin-walled cells about 40 to 60 rom in either
direction and united by arm-like projections enclosing almost
WI xA z x625xl00
circular intercellular spaces; fragments of the
W z xA I x5(lOO-h)
sclerenchymatous layer, consisting of thick-walled lignified
brown, rectangular to polyhedral cells in a single layer,
where, WI weight, in g, ofthe alkaloid used to prepare the
individual cells about 5 to 10 J!m wide and 10 to 30 J!m long;
reference solution,
fragments of mucilage staining in ruthenium red solution.
Wz weight, in g, of the substance under
examination used to prepare the test solution, B. Determine by thin-layer chromatography (2.4.17), coating
AI area ofthe peak corresponding to the alkaloid the plate with silica gel OF 254.
in the chromatogram obtained with the Mobile phase. A mixture of20 volumes of acetone, 20 volumes
reference solution, of toluene, 3 .volumes of ethanol (95 per cent) and I volume
A z = area ofthe peak corresponding to the alkaloid of strong ammonia solution.
in the chromatogram obtained with the test
Test solution. Triturate 0.10 g of the powdered substance with
solution,
5 ml of ethanol (70 per cent), add 3 ml of ethanol (70 per
h = percentage loss on drying.
cent), transfer to a 25-ml conical flask and heat in a water-bath
For calculation, I mg Of morphine hydrochloride may be taken at 50° to 60° for 30 minutes, with stirring. Cool, filter, wash the
as equivalent to 0.759 mg of morphine and I mg of codeine filter with ethanol (70 per cent) and dilute the filtrate to 10 ml
phosphate may be taken as equivalent to 0.943 mg ofcodeine. with the same solvent.
Storage. Store protected from light and moisture. Reference solution. Dissolve 2 mg of papaverine
hydrochloride RS, 12 mg of codeine phosphate RS, 12 mg of
noscapine hydrochloride RS, and 25 mg of morphine
hydrochloride RS in ethanol (70 per cent) and dilute to 25 ml
Opium Powder with the same solvent.

Opium Powder is Opium dried at a temperature not exceeding Apply to the plate 20 J!l of each solution as 20 mm bands.
70°, reduced to a fine or moderately fine powder and adjusted After development, dry the plate at 100° to 105° for 15 minutes,
by the addition ofLactose, suitably coloured with Caramel, or allow to cool al1d spray with potassium iodobismuthate
other suitable diluent to contaIn about 10 per cent ofmorphine solution and then with a 0.4 per cent w/v solution of sulphuric
and 2 per cent of codeine. acid. The chromatogram obtained with the reference solution
shows in the lower part an orange-red orred band (morphine),
Opium Powder contains not less than 9.5 per cent w/w and
above it a similarly coloured band (codeine) and in the upper
not more than 10.5 per cent w/w ofmorphine, C 17H I9N03, and
part an orange-red or red band (papaverine) and above it a
not less than 1.9 per cent w/w and not more than 2.1 per cent
similarly coloured band (noscapine). The chromatogram
w/w ofcodeine, ClsHzIN03, both calculated on the dried basis.
obtained with the test solution shows orange-red or red bands
Description. A light brown powder consisting of yellowish corresponding to those in the chromatogram obtained with
brown or brownish red particles; odour, characteristic. the reference solution. The chromatogram obtained with the
test solution may also show a dark red band (thebaine) situated
Identification between those due to codeine and to papaverine.
A. When examined under a microscope, the residue obtained C. To I g ofthe powdered substance add 5 ml of water, shake
after extraction with water, appears as granules of latex for 5 minutes, filter and add to the filtrate 0.25 ml offerric
agglomerated in irregular masses and light brown elongated chloride solution; a red colour develops which does not
filaments. Some fragments of vessels and rather elongated, disappear on the addition of 0.5 ml of2 M hydrochloric acid.
refringent crystals are also visible, as well as a smaller number
ofround pollen grains and fragments ofelongated fibres. Hairs Tests
ofvarious lengths with sharp points and a few grains ofstarch
introduced during the handling of the latex may be present. Thebaine. Not more than 3.0 per cent, calculated on the dried
Fragments ofepicarp consisting ofpolygonal cells with thick basis.
walls defining a stellate lumen may also be present and if
Determine by liquid chromatography (2.4.14).
powdered cocoa husk is present, the following: brown colour
of the fragments; narrow spiral vessels about 10 to 20 rom Test solution. Use the test solution prepared in the Assay.

2527

www.webofpharma.com
OPIUM POWDER IP 2010

Reference solution. Dissolve 25 mg of thebaine in sufficient Inject the test solution and the reference solution.
mobile phase to produce 25.0 ml and dilute 10.0 ml of this Calculate the percentage content of each alkaloid from the
solution to 100.0 ml with the mobile phase. expression
Chromatographic condition as described in the Assay.
WI xA z x625x100
The test is not valid unless the capacity factor for thebaine is
at least 3.0 and the number of theoretical plates is at least W z xA I x5(100-h)
3000. Calculate the percentage content of thebaine from the
where, WI weight, in g, ofthe alkaloid used to prepare the
expression given in the Assay.
reference solution,
Total ash (2.3.19). Not more than 6.0 per cent, determined on Wz = weight, in g, of the substance under
0.5g. examination used to prepare the test solution,
Loss on drying (2.4.19). Not more than 15.0 per cent, AI = area ofthe peak corresponding to the alkaloid
determined on 0.25 g by drying in an oven at 105° for 4 hours. in the chromatogram obtained with the
Assay. Determine by liquid chromatography (2.4.14). reference solution,
Test solution. Suspend about 1.0 g, accurately weighed, of Az = area ofthe peak corresponding to the alkaloid
the substance under examination, cut into thin slices, in 50 ml in the chromatogram obtained with the test
of ethanol (50 per cent), mix with the aid of ultrasound for solution,h
1 hour, allow to cool, dilute to 100.0 ml with the same solvent h = percentage loss on drying.
and allow to stand. To 10.0 ml of the supernatant liquid add For calculation, 1 mg of morphine hydrochloride may be taken
5 ml of ammonia buffer pH 9.5, dilute to 25.0 ml with water, as equivalent to 0.759 mg of morphine and 1 mg of codeine
mix, transfer20.0ml ofthe solution to a column (about 15 cmX phosphate may be taken as equivalent to 0.943 mg ofcodeine.
30 mm) containing 15 g of kieselguhr for column
chromatography and allow to stand for 15 minutes. Elute with Storage. Store protected from light and moisture.
two quantities, each of 40 ml, of a mixture of 85 volumes of
dichloromethane and 15 volumes of 2-propanol, evaporate
the eluate to dryness at 40° at a pressure of2 kPa, transfer the Papain
residue to a 25-ml volumetric flask with the aid ofthe mobile
phase and dilute to volume with the same solvent. Carica papaya
Reference solution. Weigh accurately 0.1 g of morphine Papain is an enzyme or a mixture ofenzymes obtained from the
hydrochloride RS and 25 mg of codeine phosphate RS, juice of the unripe fruit of Carica papaya Linn. (Fam.
dissolve in sufficient of the mobile phase to produce 25.0 ml Caricaceae). It may contain a suitable diluent such as Lactose.
and dilute 10.0 ml of this solution to 100.0 ml with the same Papain contains not less than the minimum protease activity
solvent. determined under the conditions of the Assay.
Chromatographic system Description. A white to light brown, amorphous or slightly
a stainless steel column 25 cm x 4.6 rom, packed with granular powder; odour, characteristic.
octylsilane bonded to porous silica (5 /lm), fitted with a
guard column 4 cm x 4.6 rom packed with octylsilane Tests
bonded to porous silica (5 /lm),
_ mobile phase: 1.0 g of sodium heptanesulphonate in Microbial contamination (2.2.9). 1.0 g is free from Escherichia
420 m1 of water, adjusting to pH 3.2 with phosphoric coli and 10.0 g is free from salmonellae.
acid that has been diluted to contain 0.49 per cent w/v Loss on drying (2.4.19). Not more than 7.0 per cent, determined
solution of H3P0 4 (about 5 ml) and adding 180 ml of on 1 g by drying in an oven at 60° at a pressure not exceeding
acetonitrile, 0.7 kPa for 4 hours.
- flow rate. 1.5 ml per minute, Assay. Weigh accurately about 0.5 g, triturate with 10 ml of
- spectrophotometer set at 280 urn. cysteine hydrochloride solution and dilute to 100.0 ml with
Inject suitable volume of reference solution. The test is not water. To 30 ml of water in each oftwo flasks add 15.0 m1 of
valid unless the resolution between the peaks corresponding . . casein solution andmaintain at 60° by heating on a water-
~~~to~m(;rp1ilnean(rcodeIne~is aTieast-2:S. If nec-essary, ~adjust ~ bath.~ To~thetlfstllasl(liaa:~5~0 n:ironllesolution~ortlie~
thevohimeofacetoniirlle iii themooilepnase: ThefeIative sli5stiiiice u.11def exaiiiiiiiitiori;and .fOthesecond flask-add
standard deviation of the peak area for morphine is not more 5.0 ml of the same solution, previously boiled for 2 minutes
than 1.0 per cent. and cooled. Maintain the solutions at 60° for 30 minutes, cool

2528

www.webofpharma.com
IP 2010 PEPPERMINT OIL

rapidly to room temperature and add to each flask 0.75 ml of (menthyl acetate). The chromatogram obtained with test
phenolphthalein solution and 10 ml offormaldehyde solution, solution shows an intense band corresponding to menthol, a
previously neutralised to phenolphthalein solution. Titrate band corresponding to cineole, a violet-blue band
both solutions with 0.1 M sodium hydroxide to the same corresponding to menthyl acetate in the middle of the
definite pink colour; the difference between the two titrations chromatogram and at a slightly lower Rfvalue a greenish band
is not less than 4.5 ml. (menthone). The chromatogram obtained with test solution
does not show intense greyish green or faint bluish grey bands
Storage. Store protected from light and moisture.
at Rf values between those of cineole and thymol in the
Labelling. The label states the name of any added substance. chromatogram obtained with reference solution (carvone,
pulegone, isomenthone). The chromatogram obtained with
test solution shows an intense reddish violet band near the
solvent front (hydrocarbons) and a brownish yellow band
Peppermint Oil (menthofuran) at a slightly lower Rfvalue; other less intensely
coloured bands may also be seen.
Peppermint Oil is obtained by steam distillation from the aerial
parts of the flowering plant of Mentha piperita L. and M. Tests
arvensis var. piperascens (Fam. Labiatae).
Acidity. To 2 g add 0.25 ml of phenolphthalein solution; not
. Peppermint Oil contains not less than 4.5 per cent w/w and more than 0.1 ml of 0.5 M ethanolic potassium hydroxide is
not more than 10.0 per cent w/w ofesters, calculated as menthyl required to change the colour of the solution.
acetate, C 12H 22 0 2, not less than 44.0 per cent w/w of free
Optical rotation (2.4.22). -10° to -30°.
alcohols, calculated as menthol, C IOH 200, and not less than
15.0 per cent w/w and not more than 32.0 per cent w/w of Refractive index (2.4.27). 1.460 to 1.467.
ketones, calculated as menthone, CIOH1SO.
Weight per ml (2.4.29).0.900 g to 0.916 g.
Description. A colourless, pale yellow or pale greenish yellow
Dimethyl sulphide. Distil 25 ml, collect the first 1 ml of the
liquid; odour, characteristic; taste, characteristic followed by
distillate and carefully superimpose it on 5 ml ofa 6.5 per cent
sensation of cold.
w/v solution of mercuric chloride; no white film is produced
within 1 minute at the interface of the two liquids.
Identification
Fixed oils and resinified volatile oils. Allow 0.05 m1 to fall on
Determine by thin-layer chromatography (2.4.17), coating the a filter paper. The oil evaporates completely within 24 hours
plate with silica gel G F254. without leaving a translucent or greasy mark.
Mobile phase. A mixture of 95 volumes of toluene and
Assay. For esters - To 2 g in a borosilicate glass flask add
5 volumes of ethyl acetate.
2 ml of ethanol (90 per cent) and 0.25 ml ofphenolphthalein
Test solution. Dissolve 1 g of the oil under examination in solution, neutralise with 0.5M ethanolic potassium
100 ml of toluene. hydroxide, add an additional 25.0 m1 and a little pumice powder
Reference solution. Dissolve 50 mg of(-)menthol RS, 20 III of or a few pieces ofporous pot and heat under a reflux condenser
cineole RS, 10 mg of thymol RS and 10 III of menthyl acetate on a water-bath for 30 minutes. Add 1 ml ofphenolphthalein
RS in sufficient toluene to produce 10 ml. solution and immediately titrate with 0.5 M hydrochloric acid.
Repeat the operation without the substance under examination.
Apply to the plate 20 III oftest solution and 10 III ofreference The difference between the titrations represents the volume
solution as bands 20 mrn by 3 mrn. After development, dry the of alkali required to saponifY the esters.
plate in air until the odour of solvent is no longer detectable
and examine in ultraviolet light at 254 urn. In the chromatogram 1 ml of 0.5 M ethanolic potassium hydroxide is equivalent to
obtained with test solution there are no quenching bands at 0.09915 g ofesters, calculated as menthyl acetate, CI2H2202
Rf values slightly lower than that of the faint band due to Forfree alcohols - To 1 g in a dry, 150-ml acetylation flask,
thymol in the chromatogram obtained with reference solution add 3 ml ofa mixture oD volumes ofpyridine and I volume of
(carvone and pulegone). Spray the plate with anisaldehyde acetic anhydride. Determine the weight of the acetylation
solution and examine in daylight after heating at 105° for mixture to the nearest mg, keeping the flask closed while
5 minutes. The chromatogram obtained with reference solution weighing. Boil under a reflux condenser in a water-bath for
shows, in order of increasing Rf value, an intense blue to 3 hours, maintaining the water level 2 to 3 cm above the level
violet band (menthol) in the lower third, a violet-blue to brown of the liquid in the flask throughout. Remove the flask from
band (cineole), a pink band (thymol) and a violet-blue band the water-bath and add 50 ml of water through the condenser,

2529

www.webofpharma.com
PEPPERMINT OIL IP 2010

remove the condenser and wash the walls of the flask with Pippali, Large contains not less than 1.0 per cent w/w of
10 ml of water. Allow to stand for 15 minutes and titrate with piperine, calculated on the dried basis.
0.5 M sodium hydroxide using 1 ml of phenolphthalein
Description. The spikes are blacking green to green in colour,
solution as indicator. Repeat the operation without the
cylindrical, erect and blunt. It has pungent taste and the odour
substance under examination. The difference between the
is aromatic and characteristic. The spikes are 2 to 4 cm long
titrations represents the volume ofsodimn hydroxide required.
and 0.4-0.7 cm in diameter.
1 ml of 0.5 M sodium hydroxide is equivalent to 0.07815 g of
free alcohols, calculated as menthol, C IOH2o O. Ifthe quantities Identification
ofacetic anhydride in pyridine used in the two determinations
differ by more than 5 mg, adjust the volume of alkali used in A. Macroscopic - The spikes are blackish green to green in
the second titration by multiplying with alb where a is the colour, surface rough. The spikes bear bracts and numerous
weight, in g, of acetic anhydride in pyridine used in the fIrst small fruits sunle in solid spike.
determination and b is the weight, in g, of acetic anhydride in
B. Microscopic - The penduncle is circular in outline,
pyridine used in the second test.
trichomes are absent, epidermis is a single layer oftangentially
Forketones- To 2 g add 25 ml ofa 5.0 per centw/v solution elongated cells. Vascular traces are in fIve to six bundles. Pith
of hydroxylamine hydrochloride in ethanol (95 per cent), is star shaped. Endosperm cells are larger, cells are packed
heat on a water-bath for 1 hour, allow to cool, add about 1 mg with abundant starch grains and oil droplets. Crystals are
of methyl orange and titrate with 0.5 M ethanolic potassium present in some of the cells, they appear unequal in size. The
hydroxide until an orange-yellow colour is obtained. Repeat epicarp is a single layer ofthick walled cells having greenish
the heating for further periods of 1 hour until, after cooling, content. Endocarp is wavy in outline. Mostly, endocarp and
not more than 0.1 ml of 0.5 M ethanolic potassium hydroxide seed coat are fused together to form a deep zone with hyaline
is required to neutralise the solution. content in outer layer and orange red (brown) inner region.

1 ml of 0.5 M ethanolic potassium hydroxide is equivalent to C. Determine by thin-layer chromatography (2.4.17), coating
0.07710 g ofketones, calculated as menthone, CIOH1SO. the plate with silica gel GF254.
Storage. Store protected from light and moisture, in well-fIlled Mobile phase. A mixture of60 volumes of benzene, 30 volumes
containers. of ethyl acetate and ·10 volumes of diethyl ether.

Test solution. Reflux 2 g of the coarsely powdered substance


under examination with 50-75 ml of methanol for 15 minutes,
Pippali, Large cool and fIlter. Reflux the residue further for two times with
75 ml of methanol, cool and fIlter. Combine all the fIltrates and
Long Pepper; Catkins (Big); Piper retrofractum
concentrate under vacuum to 50 ml.

Reference solution. Reflux 0.4 g of the coarsely powdered


9
pippali, Big RSwith 50-75 rnl of methanol for 15 minutes, cool
and fIlter. Reflux the residue further for two times with 75 ml of
8
methanol, cool and fIlter. Combine all the filtrates and
7 concentrate under vacuum to 10 ml.
6
Apply to the plate 10 I-LI ofeach solution as bands 10 mm by 2
5 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air

11~1
4 and examine in ultraviolet light at 254 urn and 365 urn, spray
the plate with vanillin sulphuric acid reagent.. Heat the plate
3
at 100° for 5-10 minutes and examine in day light. The
2 chromatographic profIle of the test solution is similar to that
1 of the reference solution.
I
0 1

Pippali, Large consists of the fruiting of Piper Foreign organic matter (2.6.1). Not more than 2.0 per cent.
retrofractum Vahl.(Syn. P. latifolium Hunter, (Fam.
Piperaceae). Ethanol-soluble extractive (2.6.2). Not less than 8.0 per cent.

2530

www.webofpharma.com
IP 2010 PIPPALI, SMALL

Water-soluble extractive (2.6.3). Not less than 10.0 per cent Pippali, Small
by Method I.
Small Pepper; Catkins (small); Piper longum
Total ash (2.3.19). Not more than 8.0 per cent.
I.'

Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.

Heavy metals (2.3.13). 1.0 g complies'·with the limit test for


heavy metals, Method B (20 ppm). ,

Loss on drying (2.4.19). Not more than 12.0 per cent,


determined on 5 g by drying in an oven at 105°.

Microbial contamination (2.2.9). Complies with the microbial


contamination tests.

Assay. Determine by liquid chromatography (2.4.14)..

Test solution. Weigh 2 g of coarsely powdered substance


under examination, add 50 ml of methanol, sonicate for
3 minutes and heat on a boiling water bath for 15 minutes, cool Pippali, small consists of the fruiting spike of Piper longllln
and dilute to 100.0 ml with methanol and filter. Dilute further if Linn. (Fam. Piperaceae).
necessary.
Pippali, small contains not less than 0.4 per cent w/w ofpiperine,
Reference solution. A ,0.01 per cent w/v solution of calculated on the dried basis.
piperine RS in methanol. Description. The spikes are greenish black to black in colour,
Chromatographic system cylindrical, erect and blunt. It has pungent taste and the odour
- a stainless steel column 25 cm x 4.6 mm packed with is aromatic and characteristic. The spikes are 1.0 to 1.9 cm
long and 0.2 to 0.3 cm in diameter.
octadecylsilane bonded to porous silica (5 /-un),
mobile phase: A.a buffer solution prepared by dissolving Identification
0.136 g of potassium di-hydrogen orthophosphate in
900 ml of water, adjust the pH to 2.5 with A. Macroscopic - The spikes are greenish black to black in
orthophosphoric acid and dilute to 1000 ml with water, colour, surface rough. The spikes bear bracts and numerous
B. acetonitrile, small fruits sunk in solid spike.
a linear gradient programme using the conditions given B. Microscopic - The peduncle has uniseriate septate
below, trichomes, epidermis is a single layer oftangentially elongated
flow rate. 1.5 ml per minute, cells. The penduncle vascular traces are in 2-3 bundles, pith is
spectrophotometer set at 270 nm, dumbbell shaped. Cells are small, packed with abundant strach
injection volume. 20 iiI. grains and oil droplets. Crystal are present in some of the
Time Mobile phase A Mobile phase B cells, they appear unequal in size. The epicarp is a single layer
(in min) (per cent v/v) (per cent v/v) of thick walled cells having greenish content. Endocarp is
o 95 5 wavy in outline. Mostly, endocarp and seed coat are fused
together to form a deep zone with hyaline content in outer
18 55 45
layer and orange red (brown) inner region.
25 20 80
C. Determine by thin-layer c:hromatography (2.4.17), coating
30 95 5 the plate with silica gel GF254.
Inject the reference solution. The relative standard deviation Mobile phase. A mixture of60 volumes of benzene, 30 volumes
for the replicate injections is not more than 2.0 per cent. of ethyl acetate and 10 volumes of diethyl ether.
Inject the test solution and the reference solution. Test solution. Reflux 2 g of the coarsely powdered substance
under examination with 50-75 ml of methanol for 15 minutes,
Calculate the content of piperine.
cool and filter. Reflux the residue further for two times with
Storage. Store protected from heat, moisture and against attach 75 ml of methanol, cool and filter. Combine all the :filtrates !;lnd
by insects and rodents. ' concentrate under vacuum to 50 mI.

2531

www.webofpharma.com
PIPPALI, SMALL IP 2010

Reference solution. Reflux 0.4 g of the coarsely powdered Time Mobile phase A Mobile phase B
pippali, small RS with 50-75 ml of methanol for 15 minutes, (min) (per cent v/v) (per cent v/v)
cool and filter. Reflux the residue further for two times with o 95 5
75 m1 of methanol, cool and filter. Combine all the filtrates and 18 55 45
concentrate under vacuum to 10 ml.
25 20 80
Apply to the plate 10 III ofeach solution as bands 10 mm by 2 30 95 5
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray Inject the reference solution. The relative standard deviation
the plate with vanillin sulphuric acid reagent.. Heat the plate for the replicate injections is not more than 2.0 per cent.
at 100° for 5-10 minutes and examine in day light. The Inject the test solution and the reference solution. The relative
chromatographic profile of the test solution is similar to that retention time ofpiperine is 1.
of the reference solution.
Calculateihe content of piperine.
Tests Storage. Store protected from heat, moisture and against attach
Foreign organic matter (2.6.1). Not more than 2.0 per cent. by insects and rodents.

Ethanol-soluble extractive (2.6.2). Not less than 8.0 per cent.


Water-soluble extractive (2.6.3). Not less than 10.0 per cent
by Method I.
Punarnava
Total ash (2.3.19). Not more than 8.0 percent. Ragweed; Boerhaavia diffusa

Acid-insoluble ash (2.3 .19). Not more than 3.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for 9
heavy metals, Method B (20 ppm). 8

Loss on drying (2.4.19). Not more than 12.0 per cent, 7


determined on 5 g by drying in an oven at 105°. 6
Microbial contamination (2.2.9). Complies with the microbial 5
contamination tests.
4
Assay. Determine by liquid chromatography (2.4.14).
3
Test solution. Weigh 2 g of coarsely powdered substance 2
under examination, add 50 ml of methanol, sonicate for
3 minutes and heat on a boiling water bath for 15 minutes, cool
and dilute to 100.0 m1 with methanol and filter. Dilute further if o
necessary.
Reference solution. A 0.01 per cent wlv solution of Punarnava consists of the dried root of Boerhaavia diffilsa
piperine RS in methanol. Linn. (syn. B. reperis Linn) (Fam. Nyctaginaceae).

Chromatographic system Punarnava contains not less than 0.005 per cent wlw of
a stainless steel column 25 cm x 4.6 mm packed with boeravinone B, calculated on the dried basis.
octadecylsi1ane bonded to porous silica (5 Ilm), Description. A greyish-brown in colour, short and fibrous
- mobile phase: A. a buffer solution prepared by fracture and no distinct odour with bitter taste.
dissolving 0.136 g of potassium di-hydrogen
orthophosphate in 900 ml of water, adjust the pH to 2.5 Identification
with orthophosphoric acid and dilute to 1000 ml with
water, A. Macroscopic - Stout, tapering, somewhat lmotty and
B. acetonitrile, twisted roots upto 30 cm or more long and 0.5-1.5 cm thick

__ .=-~alineargradientprogramme.usingJhe.conditions.given ·;~~h ~~e;~~i~~~~:iul~~~~ffi~;~~~~:~~i:su~~c;o~~ .-

flow rate. 1.5 ml per minute,


spectrophotometer set at 270 nm, B. Microscopic - Transverse section of root shows
injection volume. 20 Ill. outermost layer of cork which consists of thin walled

2532

www.webofpharma.com
IP 2010 SARPAGANDHA

tangentially elongated cells with brownish walls followed by methanol till the extract turns colourless, cool and filter.
thin walled 1-2 layered cork cambium. Cortex many layered Combine all the filtrates and concentrate to a volume slightly
and composed of thin walled cells. Secondary cortex 2-4 less than 25 ml. Dilute to 25.0 ml with methanol.
layered and parenchymtous. Concentric bands ofxylem tissues Reference solution. A 0.002 per cent w/v solution of
alternating with parenchymatous tissues. Vessels are radial boeravinone B RS in methanol.
with reticulate thickening. Simple and compound starch grains
Chromatographic system
with centric hilum and raphide crystals of calcium oxalate in
- a stainless steel column 25 cm x 4.6 mm packed with
cortex region. Fibres are aseptate and spindle shaped with
octadecylsilane bonded to porous silica (5 /lm),
pointed ends.
- mobile phase: A.a gradient mixtures of water;
C. Determine by thin-layer chromatography (2.4.17), coating B. acetonitrile,
the plate with silica gel GF254. a linear gradient programme using the conditions given
Mobile phase. A mixture of 35 volumes of chloroform, below,
6 volumes of methanol and 1 volume of glacial acetic acid. - flow rate. 1 ml per minute,
- spectrophotometer set at 272 nm,
Test solution. Reflux 2 g of the coarsely powdered substance - injection volume. 20 /ll.
under examination with 25 ml of methanol for 15 minutes, cool
Time Mobile phase A Mobile phase B
and filter. Reflux the residue further with 2 x 25 ml of methanol,
(min) (per cent v/v) ( per cent v/v)
cool and filter. Combine all the filtrates and concentrate under
vacuum to 5 ml.
o 90 10
25 10 90
Reference solution. Reflux 2 g ofthe punarnava RS with 25 ml 35 90 10
of methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 x 25 ml of methanol, cool and filter. Combine all Inject the reference solution. The test is not valid unless the
the filtrates and concentrate under vacuum to 5 ml. relative standard deviation for the replicate injections is not
more than 2.0 per cent.
Apply to the plate 10 /ll ofeach solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air Inject the test solution and reference solution.
and examine in ultraviolet light at 254 TIm and 365 nm, spray Calculate the content of boeravinone B.
the plate with anisaldehyde sulphuric acid reagent. Heat the Storage. Store protected from heat, moisture and against attack
plate at 110° for 10 minutes and examine in day light. The by insects and rodents.
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests Sarpagandha
Foreign organic matter (2.6.1). Not more than 2.0 per cent. Rauwolfia serpentina Root
Ethanol-soluble extractive (2.6.2). Not less than 0.5 per cent.
Water-soluble extractive (2.6.3). Not less than 9.0 per cent by 9
methodl. 8
\ Total ash (2.3.19). Not more than 10.0 per cent. 7

Acid-insoluble ash (2.3.19). Not more than 3.0 per cent. 6

Heavy metals (2.3.13). 1.0 g complies with the limit test for 5
heavy metals, Method B (20 ppm). 4

Loss on drying (2.4.19). Not more than 10.0 per cent, 3


determined on 5 g by drying in an oven at 105°. 2
Microbial contamination (2.2.9). Complies with the microbial 1
contamination tests.
o 1
Assay. Determine by liquid chromatography (2'.4.14).
Test solution. Reflux 2 g of the coarsely powdered substance
under examination with 50 ml of methanol on a water-bath for Sarpagandha consists of the dried roots of Rauwolfia
15 minutes, cool and filter. Reflux the residue further with serpentina Bentham ex Kurz (Fam Apocynaceae).

2533

www.webofpharma.com
SARPAGANDHA IP 2010

Sarpagandha contains not less than 0.15 per cent of reserpine band may also appear at the line of application in the
an~ ajmalicine, calculated on the dried basis. chromatogram obtained with test solution.
Description. Taste bitter, odour indistinct.
Tests
Identification Foreign organic matter (2.6.1). Not more than 2.0 per cent
A. Macroscopic - Roots are sub cylindrical to tapering, Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent.
tortuous or curved, rarely branched. Occurs as segments Water-soluble extractive (2.6.3). Not less than 5.0 per cent.
usually from 5 to 15 cm in length and 3 to 20 mID in diameter.
Externally grayish yellow to brown, wood pale yellow. Roots Total ash (2.3.19). Not more than 8.0 perceut.
tough with longitudinal marking and slightly wrinkled surface. Acid-insoluble ash (2.3.19): Not more than 2.0 per cent.
When scraped, bark separates readily from the wood. Fracture
Heavy metals (2.3.13). 1.0 g complies with the limit test for
is short and irregular.
heavy metals, Method B (20 ppm).
B. Microscopic - In transverse section, cork cells in 2 to 8 Loss on drying (2.4.19). Not more than 12.0 per cent,
alternating bands of radically narrow and broader cells, thin,
determined on 5 g by drying in an oven at 105°.
lignified up to 75 /lm in tangential width, broader cells up to
about 90 /lm in radial length, phelloderm, tangentially elongated Microbial contamination limits (2.2.9). Complies with the
to isodimetric parenchyma cells containing starch and short microbial contamination tests.
latex cells with brown resinous matter; secondary cortex Assay. Determine by liquid chromatography (2.4.14).
consists of parenchyma cells, heavily packed with starch
grains secondary phloem contains phloem parenchyma and Test solution. Reflux about 2 g of the coarsely powdered
sieve elements, parenchyma contains starch and angular substance under examination with 50 ml of
crystals of calcium oxalate 3 to 20 /lm in length. Xylem is ethanol (95 per cent), on a water bath for 15 minutes, cool
aboiIt 4/5 ofthe diameter of the root, wood is transversed by and filter. Reflux the residue further with ethanol (95 per
medullay rays 1 to 5 cells in width. Xylem consists ofvessels, cent), till the last extract turns colorless, cool and filter. Combine
tracheids, wood parenchyma and wood fibers. Xylem vessels all the filterates and concentrate to 100.0 ml.
are elongated up to 350 /lm in length and 50 /lm in width and Reference solution (a). A solution containing a 0.002 per cent
contains simple or bordered pits; tracheids lignified, pitted; w/v reserpine RS and 0.002 percent w/v ajmalicine RS in
wood parenchyma with moderately thick, lignified and pitted ethanol (95 per cent).
walls containing starch; wood fibres highly thickened with
Reference solution (b). A 0.002 per cent w/v reserpine RS in
pointed ends, stone cells absent.
ethanol (95 per cent) for peak identification.
C. Determine by thin-layer chromatography (2.4. I7), coating
Chromatographic system
the plate with silica gel GF254.
a stainless steel column 25 cm x 4.6 mID packed with
Mobile phase. A mixture of 70 volumes of chloroform and octadecylsilane bonded to porous silica (5 /lm),
30 volumes of acetone. mobile phase: a mixture of 35 volumes of acetonitrile
and 65 volumes of a buffer solution prepared by
Test solution. To 250 mg ofthe coarsely powdered substance
dissolving 6.80 g of potassium dihydrogen phosphate
under examination, add 5 ml methanol, shake for 10 minutes,
in 1000 ml water, adjust the pH to 3.0 with dilute
and filter. Wash the residue with 5 ml of methanol and add the orthophosphoric acid, .
washing to the filtrate.
flow rate. 1 ml per minute,
Reference solution. To 250 mg of sGipagandha RS, add 5 ml spectrophotometer set at 268 urn,
methanol, shake for 10 minutes, and filter. Wash the residue injection volume. 10 Ill.
with 5 ml of methanol and add the washing to the filtrate. Inject the reference solution (a). The test is not valid unless
Apply to the plate 10 /ll of each solution as bands 10 mID by 2 the relative standard deviation for the replicate injections is
mm. Allow the mobile phase to rise 12 cm. Dry the plate in air not more than 2.0 per cent. Inject reference solution (b) for
and examine in ultraviolet light at 254 urn and 365 urn, spray peak identification.
the plate_with anisaldehYdesltlphuricaCidxeagent.Heatthe Inject the test solution·andTeference solution(a}
plllte at 100° for 5 minutes and examine the plate in day light.
CalcUlate the content of reserpine and ajrnalicine.
The chromatogram obtained with test solution shows two
pinkish-violet bands corresponding to the bands in the Storage. Store protected from heat, moisture and against attack
chromatogram obtained with reference solution. A dark brown by insects and rodents.

2534

www.webofpharma.com
IP 2010 SARPAGANDHA TABLETS

Sarpagandha Powder Reference solution (a). A solution containing a 0.002 per cent
w/v reseJpine RS and 0.002 per cent w/v ajmalicine RS in
Rauwolfia serpentina Powder ethanol (95 per cent).
Sarpagandha powder is obtained by Rauwolfia serpentina Reference solution (b). A 0.002 per cent w/v reserpine RS in
Bentham ex Kurz roots (Fam. Apocynaceae) reduced to a fine ethanol (95 per cent) for peak identification.
powder.
Chromatographic system
It contains not less than 0.15 per cent w/w and not more than - a stainless steel column 25 cm x 4.6 rom packed with
0.2 per cent w/w ofreserpine-ajmalicine alkaloid calculated on octadecylsilane bonded to porous silica (5 f..lm),
dried basis. mobile phase: a mixture of35 volumes of acetonitrile
Description. Taste bitter, odour indistinct. and 65 volumes of a buffer solution prepared by
dissolving 6.80 g of potassium dihydrogen phosphate
Identification in 1000 ml water, adjust the pH to 3.0 with dilute
Determine by thin layer chromatography (2.4.17), coating the orthophosphoric acid,
plate with silica gel OF 254. - flow rate. 1 ml per minute,
- spectrophotometer set at 268 nm,
Mobile phase. A mixture ono volumes of chloroform and 30 - injection volume. 10 f..ll.
volumes of acetone.
Inject the reference solution (a). The test is not valid unless
Test solution. To 0.25 g of the substance under examination, the relative standard deviation for the replicate injections is
add 5 ml methanol, shake for 10 minutes, and filter. Wash the not more than 2.0 per cent. Inject reference solution (b) for
residue with 5 ml of methanol and add the washing to the peak identification.
filtrate.
Inject the reference solution and the test solution.
.Reference solution. To 0.25 g ofRauwolfia serpentina powder
Calculate the content ofreserpine and ajmalicine.
RS, add 5 ml methanol, shake for 10 minutes, and filter. Wash
the residue with 5 ml of methanol and add the washing to the Usual strength. 0.175 per cent w/w.
filtrate. Storage. Store protected from heat, moisture and against attack
Apply to the plate 20 f..ll ofeach solution as bands 10 rom by 2 by insects and rodents.
rom. Allow the mobile phase to rise 12 cm. Dry the plate in air
and examine in ultraviolet light at 365 nm. Spray the plate with
anisaldehyde sulphuric acid reagent. Heat the plate at 100° Sarpagandha Tablets
for 5 minutes and examine the plate at 365 nm. The
chromatographic profile of the test solution is similar to that Rauwolfia serpentina Tablets
of the reference solution. Sarpagandha Tablets contain not less than 85.0 per cent w/w
and not more than 115.0 per cent w/w of stated amount of the
Tests alkaloids contents mainly ofreserpine together with ajmalicine.
Total ash (2.3.19). Not more than 5.0 percent. Identification
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
In the assay the principal peak in chromatogram obtained
Heavy metals (2.3.13). 1.0 gm complies with the limittest for with the test solution corresponds to the peak in the
heavy metals, method B (20 ppm). chromatogram obtained with reference solution.
Loss on drying (2.4.19). Not more than 5.0 per cent determined Tests
on 1 gm by drying in an oven at 105°.
Other tests. Comply with the tests stated under tablets.
Microbial contamination limits (2.2.9). Complies with the
microbial contamination tests. Loss on drying (2.4.19). Not more than 8.0 per cent, determined
in 1.0 gm by drying in an oven at 105°. .
Assay. Determine by liquid chromatography (2.4.14).
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux about 2.0 g of the substance under
examination with 50 ml of ethanol (95 per cent), on a water Test solution. Weigh and powder 20 tablets. Weigh accurately
bath for 15 minutes, cool and filter. Reflux the residue further a quantity of the powder containing about 1 g of rauwolfia
with ethanol (95 per cent), till the last extract turns colourless, serpentina into a 200 ml flat bottom flask. Add 50 ml of
cool and filter. Combine all the filterates and concentrate to ethanol (95 per cent), reflux on a water bath for 30 minutes,
100.Ornl. cool and filter. Reflux the residue further with ethanol

2535

www.webofpharma.com
SARPAGANDHA TABLETS IP 2010

(95 per cent) till the last extract turns colorless, cool and filter. Description. The fruits are oval-oblong, greenish brown to
Combine all the filtrates and concentrate to 100.0 ml. yellowish brown in colour. They have an aromatic
characteristic odour and the taste is sweet aromatic.
Reference solution. A 0.001 per cent w/v Reserpine RS and
0.001 % w/v Ajmalicin RS in ethanol(95 per cent). Identification
Chromatographic system A. Macroscopic - Fruit cylindrical to oval with pedicel
a stainless steel column 25 cm x 4.6 mm, packed with attached, consists of mericarps. Each mericap is 10 mm long
octadecyl silane chemically bonded to porous silica (5/l), and 4 mm broad, five sided with a wider commissural surface,
- mobile phase: a mixture of 35 volumes of acetonitrile tapered at apex and base, crowned with a conical stylepod,
and 65 volumes of a buffer (pH 3.0), prepared by glabrous, greenish or yellowish brown with five ridges.
dissolving 6.80 g of potassium dihydrogen phosphate
B. Microscopic - Transverse section of fruit shows pericarp
in 1000 ml water, pH of which is adjusted to 3.0 with
with outer epidermis of quadrangular to polygonal cells with
dilute orthophosphoric acid,
smooth cuticle. Vittae 4 dorsal and 2 commissural extending
- flow rate. Iml per minute,
with length of each mericarp, having brown cells and volatile
- spectrophotometer set at 268 nm,
oil in cavity. Mesocarp with reticulate lignified parenchyma.
- injection volume. 10 Ill.
Endocarp cells thin walled arranged parallel to one another in
Inject the reference solution. The test is not valid unless the groups of5-7. Endosperm consists of thick walled, cellulosic
relative standard deviation for replicate injections is not more parenchyma containing fixed oil cells and rosette crystals of
than 2.0 per cent. calcium oxalate.
Inject the refernce solution and the test solution. C. Determine by thin-layer chromatography (2.4.17), coating
Calculate the content of ajmalicin and reserpine. the plate with silica gel GF 254.

Usualstrengths. 0.1 mg; 0.25 mg. Mobile phase. A mixture of 93 volumes of toluene and 7
volumes of ethyl acetate.
Storage. Store protected from light at a temperature not
Test solution. To 1 g of the coarsely powdered substance
exceeding 30°.
under examination, add 25 ml of dichloromethane, reflux for
Labelling. The quantity ofactive ingredient is stated in terms 15 minutes, cool and filter. Reflux the residue further for two
ofreserpine-ajmalicine content in labelled amount. times with 25 ml ofdichloromethane, cool and filter. Combine
all the filtrates and evaporate under vacuo to 25 ml.
Reference solution. To 2 g of the saunf RS, add 40 ml of
Saunf methanol, reflux for 15 minutes, cool and filter. Reflux the
residue further with 25 ml of methanol twice, cool and filter.
Fennel; Foeniculum vulgare Combine all the filtrates and concentrate under vacuum to
25ml.
9 Apply to the plate 10 /ll of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
8
and examine in ultraviolet light at 254 nm and 365 nm, spray
7
with a anisaldehyde sulphuric acid reagent. Heat at 110° for
6 10 minutes and examine the plate in day light. The
5
chromatographic profile of the test solution is similar to that
of the reference solution.
4

3
Tests
2 Foreign organic matter (2.6.1). Not more than 2.0 per cent.
1 Ethanol-soluble extractive (2.6.2). Not less than 5.0 per cent.
o 1 Water-soluble extractive (2.6.3). Not less than 14.0 per cent
by Method 1.
Totalash (2-:-:r.T9rNOtmore tlianlKO per cent.------·-·
.~~l.l~f(;Q~~!~t§ Qfthe.. d.l'ie.4fr.t1it~Qf EQ?rz.ifltlYf!I y.y Igg!:?J'4!11-
(Fam. Apiaceae). Acld=insolubie-ashC2.iT9YNot-morethan·COper'cent.
Saunf contains not less than 0.60 per cent of anethole, Heavy metals (2.3.13). 1.0 g complies with the limit test for
calculated on the dried basis. heavy metals, Method B (20 ppm).

2536

www.webofpharma.com
IP 2010 SENNA LEAF

Loss on drying (2.4.19). Not more than 10 per cent, determined Senna leaf contains not less than 1.0 percent w/w of
on 5.0 g by drying in an oven at 105°. sennosides A and B, calculated on the dried basis.
Microbial contamination (2.2.9). Complies with the microbial Description. Pale yellowish green coloured leaflets with
contamination tests. mucilaginous and faint odour.
Assay. Determine by gas chromatography (2.4.13).
Identification
Test solution. Weigh 2.0 g ofthe coarsely powdered substance
under examination, add 50 ml of methanol, reflux on a water- A. Macroscopic -Leaflets, 2.5 to 8 cm long and 5-15 mm
bath for 15 minutes, cool and filter. Reflux the residue further wide at centre, pale yellowish green, elongated lanceolate,
with methanol till the extract turns colourless, cool and filter. slightly asymmetric at base; margins entire, flat, apex acute
Combine all the filtrates and concentrate to a volume slightly with a sharp spine; both surface smooth with sparce trichomes;
less than 100 ml. Dilute to 100.0 ml with methanol. odour, faint but distinctive; taste, mucilaginous and
disagreeable but not distinctly bitter.
Reference solution. A 0.01 per cent w/v solution of anethole
RS in methanol. B. Microscopic Transverse section shows outer single
Chromatographic system layered mucilaginous epidermal cells. Unicellular hairs
- a capillary column 30 m x 0.25 m, packed with DB 1, presents. Stomata paracytic, numerous on both surface.
temperature: Mesophyll consists of upper and lower palisade layers with
oven 90° to 260° @10° per minute (initially and finally spongy layer in between, primatic crystals ofcalcium oxalate
hold for 5 minutes respectively), present on larger veins.
injector 240°, C. In the Assay, the chromatogram obtained with test solution
detector 280°, corresponds to the chromatogram obtained with reference
flow rate. 0.8 ml per minute, solution.
split flow 20 ml per minute.
D. Determine by thin-layer chromatography (2.4.17), coating
Inject 1 fll of the reference solution. The test is not valid
the plate with silica gel GF254.
unless the relative standard deviation for the replicate
injections is not more than 10.0 per cent. Mobile phase. A mixture of 40 volumes of n- propyl alcohol,
40 volumes of ethyl acetate, 29 volumes of water and 1 volume
Inject 1 fll of the test solution and the reference solution.
of glacial acetic acid.
Calculate the content of anethole.
Test solution. Take 1 g of the dried leaves powder substance
Storage. Store protected from light in well-filled containers, at
under examination. Add 25 ml of methanol, reflux for 10 minutes,
a temperature not exceeding 30°.
cool and filter. Reflux the residue with another 20 ml of
methanol, cool and filter. Combine all the filtrates and
concentrate to 10 ml.
Senna Leaf
Reference solution. Boil 0.5 g ofdried senna leaves RS powder
Cassia leaf; Cassia angustifolia with 25 ml of methanol under reflux for 10 minutes, cool and
filter. Boil under reflux the residue with another 20 ml of
methanol, cool and filter. Combine all the filtrates and evaporate
to 5 ml.
Apply to the plate 10 fll ofeach solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
6 and examine in ultraviolet light at 254 nm and 365 urn, spray
5 the plate with 20 per cent v/v of nitric acid solution. Heat the
plate at 100° to 105° for about 10 minutes and immediately
4
examine the plate in day light. The chromatographic profile of
the test solution is similar to that of the reference solution.
2
Tests

o Foreign organic matter (2.6.1). Not more than 1.0 per cent.
Total ash (2.3.19). Not more than 14.0 percent.
Senna leaf consists of the dried compound leaves of Cassia
angustifolia or Cassia senna Vahal. (Fam.Leguminosae). Acid-insoluble ash (2.3.19). Not more than 2.5 per cent.

2537

www.webofpharma.com
SENNA LEAF IP 2010

Loss on drying (2.4.19). Not more than 12.0 per cent, Description. Pale yellowish green coloured pods with slight
determined on 5 g by drying in an oven at 105° . odour. Leaflets with mucilaginous and faint odour.
Microbial contamination (2.2.9). Complies with the microbial
Identification
contamillation tests.
Assay. Determine by liquid chromatography (2.4.14). A. Macroscopic - Flattened reniform pods, brownish yellow
at the edges, dark brown in the central area about 40 to 50 mm
Test solution. Weigh accurately 1.0 g ofthe coarsely powdered
long and about at least 20 mm wide. At one end is a stylar
substance in a round bottom flask, add about 10 ml of 1 per
point and at the other a short stalk. The pods contains 5 to 7
cent v/v acetic acid and 25 ml of methanol and reflux on a
flattened and obovate seeds, green to pale brown, with a
water bath for about 30 minutes. Cool to room temperature;
continuous network of prominent ridges on the tests andin
make up the volume up to 50.0 ml with methanol and filter.
complate wavy transverse ridges on the testa. Leaflets, 2.5 to
Reference solution. A 0.004 per cent w/v solution of 8 em long and 5-15 mm wide at centre, pale yellowish green,
sennasides RS in methanol. elongated lanceolate, slightly asymmetric at base; margins
Chromatographic system entire, flat, apex acute with a sharp spine; both surface smooth
- a stainless steel column 25 em x 4.6 mm packed with with sparce trichomes; odour, faint but distinctive; taste,
octadecylsilane bonded to porous silica (5 Ilm), . mucilaginous and disagreeable but not distinctly bitter.
mobile phase: a mixtures of 82 volumes of 1 per cent B. Microscopic - The pods present an epicarp with strongly
v/v acetic acid in water and 18 volumes of acetonitrile, cuticularised isodiametric cells, occasional anomocytic or
flow rate. 1 ml per minute, paracytic stomata, and very few conical, unicellular and warty
spectrophotometer set at 350 urn, trichomes. Hypodermis with collenchymatous cells, mesocarp
injection volume. 20 Ill. with parenchymatous tissue, a layer of prism s of calcium
Inject the reference solution. The test is not valid unless the oxalate and containing vascular bundles incompletely
relative standard deviation for the replicate injections is not surrounded by fibres with a crystals sheth of calcium oxalate
more than 2.0 per cent. prisms, endocarp consisting of thick-walled and inter lacing
fibres. The seeds present a sub epidermal layers of palisade
Inject the reference solution and the test solution.
cells with thick outer walls, endosperm composed of
Calculate the content of sennoside A and B. polyhedral cells with mucilaginous wall.
Storage. Store protected from light and moisture. Reduce to moderately fine powder examine microscopically
using chloral hydrate solution. The powder consists ofepicarp
with polygonal cells and a small number of warty trichomes
Senna Pods and occasional anomocytic stomata, fibres in two crossed
layers, accomparied by a crystal sheath of calcium oxalate
. Senna Fruit; Pods of cassia; Cassia angustifolia prism, characteristic palisade cells in the seeds and stratified
cells in the endosperm, cluster and prisms of calcium oxalate
9 C. In the Assay, the chromatogram obtained with test solution
8 corresponds to the chromatogram obtained with reference
7
solution.

6
D. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
5
Mobile phase. A mixture of40 volumes of n- propyl alcohol,
4
40 volumes of ethyl acetate, 29 volumes of water and 1volume
3 of glacial acetic acid.
Test solution. Take 1 g of the dried pods powder substance
1 under examination. Add 25 ml ofmethanol, reflux for 10 minutes,
o cool and filter. Reflux the residue with another 20 ml of
11'l~!£h'1..nc~~co~o~lJ;a~n~(d filter. Combine all the filtrates and
Se.I1Ila. PQcl~cQl1sist()f.tlle cb:teg g()J.:I1lJ()l,l!!d..:fl()As.()f(:;'qss.iCl concentrate to
angustifolia or Cassia senna Vahal. (Fam.Leguminosae). Reference solution. Boil 0.5 g ofdried senna'jJods RS powder
Senna Pods contains not less than 1.0 per cent w/w of with 25 ml of methanol under reflux for 10 minutes, cool and
sennosides A and B, claculated on the dried basis. filter. Boil under reflux the residue with another 20 ml of

2538

www.webofpharma.com
IP 2010 SENNA DRY EXTRACT

methan~l, cool and filter. Combine all the filtrates and evaporate Senna Dry Extract contains not less than 90.0 per cent w/w
t05m!. and not more than 110.0 per cent w/w of stated amount of
sennoside A and sennoside B, calculated on the dried basis.
Apply to the plate 10 III ofeach solution as bands 10 mm by 2
min. Allow the mobile phase to rise 8 cm. Dry the plate in air Description. A light brown to dark brown powder.
•' and examine in ultraviolet light at 254 nm and 365 urn, spray
.. the plate with 20 per cent v/v of nitric acidsolution. Heat the Identification
ptatt,l at r00° to 105° for about 10 minutes and immediately A. Determine by thin-layer chromatography (2.4.17), coating
examine the plate in day light. The chromatographic profile of the plate with silica gel GF254.
the ,test solution is similar to that of the reference solution.
Mobile phase. A mixture of40 volumes of n-propyl alcohol,
Tests 40 volumes of ethylacetate, 30 volumes of water and 1 volume
of glacial acetic acid.
Foreign organic matter (2.6.1). Not more than 1.0 per cent.
Test solution. Shake well 0.1 g ofthe extract under examination
Total ash (2.3.19). Not more than 14.0 per cent. with 5 ml ofa mixture ofequal volumes of methanol and water
Acid-insoluble ash (2.3.19). Not more than 2.5 per cent. for 5 minutes, heat to 60°, cool and allow to settle. Use the
supernatant liquid.
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105° . 'Reference solution. Dissolve 10 rng of senna dry extract RS
in 1 ml of a mixture of equal volumes of methanol and water.
Microbial contamination (2.2.9). Complies with the microbial
Apply to the plate, 10 III of each solution as bands of 10 mm
contamination tests.
by 2 mm. Allow the mobile phase to rise 10 cm. Dry the plate
Assay. Determine by liquid chromatography (2.4.14). in air and examine in ultraviolet light at 254 urn and 365 nm,
Test solution. Weigh accurately 1.0 g ofthe coarsely powdered spray the plate with 20 per cent v/v of nitric acid solution.
substance in a round bottom flask, add about 10 ml of 1 per Heat the plate at 110° for 10 minutes and examine the plate in
cent v/v acetic acid and 25 ml of methanol and reflux on a day light. Allow to cool and spray with a 5 per cent w/v
water bath for about 30 minutes. Cool to room temperature; solution of potassium hydroxide in ethanol (50 per cent v/v)
make up the volume up to 50 ml with methanol and filter. until the zones appear. The chromatographic profile of the
test solution is similar to that of the reference solution.
Reference solution. A 0.004 percent w/v solution of
sennosides RS in methanol. B. To about 25 mg ofthe extract add 50 ml of water and 2 ml
of hydrochloric acid. Heat in a water-bath for 15 minutes.
Chromatographic system: Cool and shake with 40 ml of ether. Separate ether layer, dry
a stainless steel column 25 cm x 4.6 mm packed with over anhydrous sodium sulphate and evaporate 5 ml to
octadecylsilane bonded to porous silica (5 Ilm), dryness. To the cooled residue add 5 ml of dilute ammonia. A
mobile phase. a mixtures of 82 volumes of 1 per cent yellow or orange colour develops. Heat on a water-bath for 2
v/v acetic acid in water and 18 volumes of acetonitrile. minutes. A reddish-violet colour develops.
flow rate. 1 ml per minute,
spectrophotometer set at 350 urn, Tests
injection volume. 20 Ill. pH (2.4.24).5.5 to 7.5, determined in a 10 per cent solution in
Inject the reference solution. The test is not valid unless the water.
relative standard deviation for the replicate injections is not Loss on drying (2.4.19). Not more than 5.0 per cent, determined
more than 2.0 per cent. on 1.0 gm by drying in an oven at 105°.
Inject the reference solution and the test solution. Microbial contamination (2.2.9). Total viable aerobic count
Calculate the content of sennoside A and B. not more than 104 micro organisms per gm, fungi not more
than 102 micro organisms per g and Escherichia coli and
Storage. Store protected from light and moisture.
salmonellae absent.
Assay. Determine by liquid chromatography (2.4.14).
Solvent mixture. A 0.3 per cent v/v solution of acetic acid
Senna Dry Extract with the pH adjusted to 5.9 with 1 M sodium hydroxide.
Senna Dry Extract is produced from Senna leaves or pods of Test solution. Dissolve an accurately weighed quantity of
Cassia angustifolia (Tinnevelly Senna) or Cassia acutifilia substance under examination containing about 10 mg of
(Cassia Senna) as calcium salts. sennosides in 50.0 ml of the solvent mixture.

2539

www.webofpharma.com
SENNA TABLETS IP 2010

Reference solution. Dissolve an accurately weighed quantity Chromatographic system


of calcium sennosides RS containing about 10 mg of a stainless steel column 15 cm x 4.6 mID, packed with
sennosides in 50.0 ml ofthe solvent mixture. octadecylsilane chemically bonded to porous silica
(5J.lm),
Chromatographic system
mobile phase: a mixture of 83 volumes of a 1 per cent
a stainless steel column 15 cm x 4.6 mm, packed with
v/v solution of glacial acetic acid and 17 volumes of
octadecylsilane chemically bonded to porous silica
acetonitrile,
(5J.lm),
flow rate. 1 ml per minute,
mobile phase: a mixture of 83 volumes of a 1 per cent
spectrophotometer set at 350 nm,
v/v solution of glacial acetic acid and 17 volumes of
- injection volume. 10 J.ll.
acetonitrile,
flow rate. 1 ml per minute, Inject the reference solution. The test is not valid unless the
spectrophotometer set at 350 nm, relative standard deviation for the replicate injections is not
injection volume. 10 J.ll. more than 2.0 per cent.

Inject the reference solution. The test is not valid unless the Inject the test solution and the reference solution.
relative standard deviation for replicate injections is not more Calculate the content of sennosides A and B, as sennoside B
than 2.0 per cent. in the tablets.
Inject the test solution and the reference solution. Storage. Store protected from light, at a temperature not
Calculate the content of sennosides A and B, as sennoside B exceeding 30°.
in the extract. Labelling. The quantity ofactive ingredient is stated in terms
Storage. Store protected from light, in air-tight containers. of total sennoside A and B, expressed as the equivalent
content of sennoside B.

Senna Tablets Shatavari


SennaTablets contain not less than 85.0 per cent w/w and not Asparagus racemosus root
more than 115.0 per cent w/w of the stated amount of
sennosides A and B, calculated as sennoside B.

Identification 9

8
In the assay, the principal peak in the chromatogram obtained
7
with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. 6

5
Tests
4
Other tests. Comply with the tests stated under tablets.
3
Loss on drying (2.4.19). Not more than 6.0 per cent, determined
2
on 1.0 g by drying in an oven at 105°.
Assay. Determine by liquid chromatography (2.4.14).
o
Solvent mixture. A 0.3 per cent v/v solution of acetic acid,
with the pH adjusted to 5.9 with 1 M sodium hydroxide.
Test solution. Weigh and powder 20 tablets. Weigh accurately Shatavari consists of the tuberous roots of Asparagus
a quantity of the powder containing about 10 mg of racemosus Willd. (Fam. Liliaceae).
sennosides, dissolve in about 40 ml ofsolvent mixture and mix Shatavari contains not less than 0.1 per cent of shatavarin IV,
with·the··ai&ofultrasound·for15-millutes:-Diluteto-50mlwitb ·ca1culated·oni:hedried·basis:- .._ - . - - - - -.....
the solventmixture and filter.
DescrIptloil:fhetuberous roofbIts are-dirtY whIte lri. color,
Reference solution. A 0.02 per cent w/v solution of calcium longitudinally wrinkled with yellow hard central core. It is
sennaside RS in the solvent mixture. starchy and slightly bitter followed by sweet taste.

2540

www.webofpharma.com
IP 2010 SHATAVARl

Identification Microbial contamination (2.2.9). Complies with the microbial


contamination tests.
A. Macroscopic - The tuberous roots are borne in a compact
Assay. Perform the Assay by following method I or by method
bunch and are fleshy, and spindle shaped. They are marketed
II. The result ofthe method II will be official in case ofdispute.
in pieces 5-15 cm in length and 2 cm in thiclmess. They are
silvery white or ash-colored externally and white internally, Method I
more or less smooth when fresh, developing longitudinal
wrinldes when dry. Determine by thin-layer chromatography (2.4.17), coating the
plate with silica gel GF 254.
B. Microscopic - The inner parenchymatous zone of cortex
Mobile phase. A mixture of 6.5 volumes of chloroform and
is composed of 18-24 layers in upper and 42-47 layers in the
3.5 volumes of methanol.
middle tuberous portion ofthe roots. The cells are thin - walled
cellulosic, with circular to oral outlines and distinct inter cellular Test solution. Reflux 4 g of the coarsely powdered substance
spaces. In some roots 3-4 layers ofcortex immediately adjacent under examination with 50 ml of methanol on a water-bath for
to the endodermis are modified into a sheath of stone cells 30 minutes, cool and filter. Reflux the residue further with
round the endodermis. The number ofvascular bundles is 30- methanol till the last extract turns colourless, cool and filter.
35 in the upper levels and 35-45 in the middle tuberous portions Combine all the filtrates and concentrate to 50 mi.
of the roots. Reference solution. A 0.008 per cent w/v solution ofshatavarin
C. Determine by thin-layer chromatography (2.4.17), coating IV RS in methanol.
the plate with silica gel GF254. Apply to the plate 5 /-ll of each solution as bands 10 mm by
Mobile phase. A mixture of 13 volumes of chloroform, 2 mm.Allowthe mobile phase to rise 8 cm.Dry the plate in air
10 volumes of methanol and 2 volumes of water. and spray with a 10 per cent v/v solution of sulphuric acid in
methanol. Heat the plate at 100° for 5 minutes, scan the plate
Test solution. Reflux 1 g ofthe coarsely powdered substance in absorbance mode at 500 urn. Record the chromatograms
under examination with 30 ml of methanol for 30 minutes, cool and measure the responses for the analyte peak.
and filter. Reflux the residue further with 2 x 30 mI of methanol,
cool and filter. Combine all the filtrates and concentrate under Calculate the content of shatavarin Iv.
vacuum to 10.0 mI. Methodll
Reference solution. Reflux 1 g of shatavari RS with 30 ml of
Determine by liquid chromatography (2.4.14).
methanol for 30 minutes, cool and filter. Reflux the residue
further with 2 x 30 ml of methanol, cool and filter. Combine all Test solution. Reflux 5 g of the coarsely powdered substance
the filtrates and concentrate under vacuum to 10.0mI. under examination with 50 ml of methanol on a water-bath for
30 minutes, cool and filter. Reflux the residue further with
Apply to the plate 5 /-l1 of each solution as bands 10 mm by 2
methanol till the last extract turns colourless, cool and filter.
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
Combine all the filtrates and concentrate to 50.0 mi.
and examine in ultraviolet light at 254 urn and 365 urn, spray
the plate with vanillin sulphuric acid reagent. Heat the plate Reference solution. A 0.1 per cent w/v solution of
at 120° for 10 minutes and examine the plate in day light. The shatavarin IV RS in methanol. Dilute suitably to prepare
chromatographic profile of the test solution is similar to that 0.0075- 0.075 percentw/v solution.
of the reference solution.
Chromatographic system
Tests a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 /-lm),
Foreign organic matter (2.6.1). Not more than 2.0 per cent. - mobile phase: a mixture of 60 volumes· of acetonitrile
Ethanol-soluble extractive (2.6.2). Not less than 15.0 per cent. and 40 volumes of water,
Water- soluble extractive (2.6.3). Not less than 20.0 per cent flow rate. 1ml per minute,
by Method I. use evaporative light scattering detector,
temperature
Total ash (2.3.19). Not more than 15.0 percent. evaporator 110°,
Acid-insoluble ash (2.3.19). Not more than 3.0 per cent. nebulizer 90°,
Heavy metals (2.3.13). 1.0 g complies with the limit test for nebulizer gas nitrogen and gas flow 1 SLM,
heavy metals, Method B (20 ppm). injection volume. 20 /-ll.
Loss on drying (2.4.19). Not more than 15.0 per cent, Inject the reference solution. The test is not valid unless the
determined on 5 g by drying in an oven at 105°. regression coefficient is not more than 0.9.

2541

www.webofpharma.com
SHATI IP 2010

Inject the test solution and reference solution. grains tissue are simple, circular or oval in shape, found in
Calculate the content of shatavarin lV. ground cells.

o Storage. Store protected from heat, moisture and against attack C. Determine by thin-layer chromatography (2.4.17), coating
by insects and rodents. the plate with silica gel GF254.
Mobile phase. A mixture of 80 volumes of n-hexane and
20 volumes ofacetone.
Test solution. Reflux 1 g of the coarsely powdered substance
Shati under examination with 25 ml of methanol for 15 minutes, cool
and filter. Reflux the residue further with 2 x 25 ml of methanol,
Hedychium spicatum
cool and filter. Combine all the filtrates and concentrate under
vacuum to 25 mi.
Reference solution. Reflux 0.5 g ofthe shati RSwith 25 mlof
9 methanol for 15 minutes, cool and filter. Reflux the residue
8 further with 2 x 25 ml of methanol, cool and filter. Combine all
'7
the filtrates and concentrate under vacuum to 12.5 mi.
6 Apply to the plate 10 J!l of each solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
5
and examine in ultraviolet light at 254 om and 365 om, spray
4 the plate with anisaldehyde sulphuric acid reagent. Heat the
3 plate at 110 0 for 10 minutes and examine the plate in day light.
The chromatographic profile of the test solution is similar to
2
that ofthe reference solution.
1

o Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Shati consists of the dried rhizomes of Hedychium spicatum Ethanol-soluble extractive (2.6.2). Not less than 0.7 per cent.
Buch.-Ham.ex Smith (Fam. Zingiberaceae). Water-soluble extractive (2.6.3). Not less than 12.0 per cent
Shati contains not less than 0.80 per cent w/w ofp-methoxy by method!.
cinnamic acid ethyl ester, calculated on the dried basis. Total ash (2.3.19). Not more than 8.0 per cent.
Description. A reddish-brown outer surface and white in side, Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
short and uneven fracture and camphoraceous odour with Heavy metals (2.3.13). 1.0 g complies with the limit test for
aromatic and pungent taste. heavy metals, Method B (20 ppm).

Identification Loss on drying (2.4.19). Not more than 14.0 per cent,
determined on 5 g by drying in an oven at 105°.
A. Macroscopic - Tuberous rhizome having reddish brown, Microbial contamination (2.2.9). Complies with the microbial
rough outer surface with round root scars or rootlets which contamination tests.
remain attached at some places. Transversely sliced pieces of
dried rhizome are spherical, flat, 1 cm in thickness and 2-3 cm Assay. Determine by liquid chromatography (2.4.14).
in diameter having white and starchy surface. Test solution. Reflux 2 g ofthe coarsely powdered substance
under examination with 50 ml of methanol on a water-bath for
B. Microscopic - Transverse section of rhizome shows 15 minutes, cool and filter. Reflux the residue further with
outermost layer of cork having 2-6 layers of isodiametic, methanol till the extract turns colourless, cool and filter.
nonlignified and suberised cells which are radially arranged. Combine all the filtrates and concentrate to a volume slightly
In old rhizome, the cork cells are exfoliated or crushed. Cortex less than 100 mi. Dilute to 100.0 ml with methanol.
is a broad zone with 20-25 layers of thin walled parenchymatous
cells. Cortex region is filled with abundant starch grains and Reference solution. A 0.01 per cent w/v solution of
~nlliiieroi.iSo1eo-resincells. Vascular bundles are closed and ·p;methoxy~cinnamic~acid~ethyl-esterRS~inmethanol~--·
··collateral arid .scattered fhrougl1ourtne-grouiid·lissues. Chromat6graphicsystem ..... -.~ .....~-~ ..... _....
Cambium has 4-5 rows oftangentially elongated cells. It forms a stainless steel column 25 cm x 4.6 mm packed with
a complete ring between the xylem and phloem groups. Starch octadecylsilane bonded to porous silica (5 J!m),

2542

www.webofpharma.com
IP 2010 STARCH

mobile phase: 40 volumes of water and 60 volumes of wash with two successive portions, each of 50 ml, of water,
acetonitrile, filter the washed light petroleum solution, and evaporate to
flow rate. 1 ml per minute, dryness; to the residue add 2 ml of a mixture of 1 volume of
spectrophotometer set at 310 nm, liquified phenol and 2 volumes of carbon tetrachloride and
- injection volume. 20 Ill. transfer to a cavity of a colour-reaction porcelain tile; fill an
Inject the reference solution. The test is not valid unless the adjacent cavity with a mixture of 1 volume of bromine and
relative standard deviation for the replicate injections is not 4 volumes of carbon tetrachloride, and cover both cavities
more than 2.0 per cent. with an inverted watch-glass; no purple or deep indigo-blue
colour is produced in the liquid containing the residue.
Inject the test solution and reference solution.
Arsenic (2.3 .10). Heat gently 5.0 g with 2 ml of nitric acid and
Calculate the content ofp-methoxy cinnamic acid ethyl ester.
0.5 ml of sulphuric acid in a long-necked flask, until the first
Storage. Store protected from heat, moisture and against attack reaction has subsided, cool, add carefully and in small portions,
by insects and rodents. 15 ml of nitric acid and 6 ml of sulphuric acid taking care to
avoid excessive foaming, and continue heating, adding further
small portions of nitric acid, if necessary, until white fumes
Shellac are evolved and the solution becomes colourless or almost
colourless. Cool, add carefully 10 ml of water, evaporate until
Lac
white fumes are evolved, and repeat the addition of water and
Shellac consists of a resinous substance prepared from a evaporation until all the nitric acid has been removed, cool,
secretion that encrusts the bodies of a scale insect, Laccifer dilute to 50 ml with water, and add 10 ml of stannated
lacca Kerr (Fam. Ciccidae). hydrochloric acid AsT. The resulting solution complies with
Description. Lemon-yellow to brownish orange thin scales or the limittest for arsenic (2 ppm). Use 0.5 ml ofarsenic standard
hard, brittle masses; odourless or with a faint odour. solution (10 ppm As) for the standard stain.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Identification
heavy metals, Method B (20 ppm).
To 50 mg add a few drops of a mixture of 1 g of ammonium Sulphated ash (2.3.18). Not more than 1.0 per cent, determined
molybdate and 3 ml of sulphuric acid; a green colour is on 0.5 g.
produced and it becomes lilac on standing for 5 minutes.
Storage. Store protected from moisture.
Tests
Acid value (2.3.23). 50 to 70, determined by the following
method. Weigh accurately about 2.0 g and dissolve with the Starch
aid ofgentle heat, in 50 ml of ethanol (95 per cent) previously Starch consists ofpolysaccharide granules obtained from the
neutralised to ethanolic thymol blue solution. Titrate with caryopsis ofmaize or com, Zea mays Linn.(Fam. Poceae), or of
0.1 M ethanolic potassium hydroxide using ethanolic thymol rice, Oryza sativa Linn., or of wheat, Triticum aestivum
blue solution as an external indicator. Calculate the acid value Linn.(Fam. Graminae) , or from the tuber ofpotato, Solanum
from the expression tuberosum Linn (Fam. Solanaceae), or from the rhizomes of
5.61 x alw tapioca, Manihot utilissima PoW.(Fam. Euphorbiaceae).

where, a number of ml of 0.1 M ethanolic potassium Description. A very fme, white or slightly yellowish powder
hydroxide and or irregular white masses which are readily reducible to powder,
w = weight, in g, ofthe sample. creaks when pressed between the fmgers; odourless and
tasteless. The presence of granules showing cracks or edge
Ethanol-insoluble matter. Not more than 2.0 per cent, irregularities is exceptional in starches other than wheat starch;
determined by the following method. Weigh accurately about wheat starch may contain granules with cracks on the edges.
5.0 g in an extraction thimble, cover with ethanol (95 per
cent) and allow to stand for 16 hours. Place in an apparatus Identification
for the continuous extraction of drugs, extract with ethanol
A. Corn or maize starch - Polyhedral granules, 2 to 23 Ilm in
(95 per cent) for 4 hours, dry the residue at 1000 for 3 hours
size, or rounded granules, 25 to 32 Ilm in diameter. The central
and weigh.
hilum consists of a distinct cavity or two- to five-rayed cleft;
Colophony. Dissolve 2.0 g by shaking with 10 ml ofethanol, no concentric striations. Viewed between crossed nicol prisms,
add slowly, with shaking 50 ml of lightpetroleum (40 0 to 60"), a distinct black cross is seen intersecting at the hilum.

2543

www.webofpharma.com
STARCH IP 2010

Potato starch - Single granules, either irregular, ovoid or Microbial Contamination (2.2.9). 1 g is free from Escherichia
pear-shaped, 30 to 100 11m in size, or rounded, 10 to 35 11m in coli and salmonellae.
size; oompound granules consisting of groups of two to four Sulphated ash (2.3.18). Not more than 0.6 per cent (for all
elements are rare. Eccentric hilum; clearly visible concentric starches except rice starch) and not more than 0.8 per cent (for
striations. Viewed between crossed nicol prisms, a distinct rice starch), determined on 2.0 g.
black cross is seen intersecting at the hilum.
Los·s on drying (2.4.19). Not more than 15.0 per cent (for all
Rice starch - Polyh<;ldral granules, 2 to 5 11m in size, either starches except potato starch) and not more than 20.0 per cent
isolated or aggregated in ovoid masses, 10 to 20 11m in size. (for potato starch), determined on 0.2 g by drying in an oven
Central hilum poorly visible; no concentric striations. Viewed at 105°.
between crossed nicol prisms, a distinct black cross is seen
intersecting at the hilum. Storage. Store protected from light and moisture.

Tapioca starch - Principally simple granules, sub-spherical, Labelling. The label states the type of starch.
muller-shaped or rounded polyhedral; smaller granules 5 to
10 11m, larger granules 20 to 35 11m in diameter; hilum, central,
punctate, linear or triradiate; striations, faint, concentric; Sunthi
compound granules, few, oftwo to three unequal components.
Saunth; Ginger; Zingiber officinale
Wheat starch - Large discoid or, more rarely, reniform
granules, 10 to 45 11m in size; profile, elliptical and fusiform, slit
along the main axis. Small rounded or polyhedral granules,
2 to 10 11m in size. Granules of intermediate size very rarely 9
occur. Hilum and striations invisible or barely visible. Viewed 8
between crossed mcol prisms, a distinct black cross is seen
7
intersecting at the hilum.
6
B. Heat to boiling for 1 minute a suspension of 1 g in 50 ml of
5
water and cool; a thin and cloudy mucilage is produced with
all starches except potato starch which gives a thick and more 4
transparent mucilage. 3

C. To 10 ml ofthe mucilage obtained in test B add 0.05 mlof 2


0.01 M iodine; a dark blue colour is produced, which 1
disappears on heating and reappears on cooling.
o
Tests
Acidity. Add 10.0 g to 100 ml of ethanol (70 per cent) Sunthi is the whole or cut scraped or unscraped, dried rhizomes
previously neutralised to phenolphthalein solution, shake of Zingiber officinale Roscoe. (Fam. Zingiberaceae).
for 1 hour, filter and titrate 50 ml of the filtrate with 0.1 M Sunthi contains not less than 0.8 per cent w/w of total
sodium hydroxide. Not more than 2.0 ml is required to change gipgerols, calculated on the dried basis.
the colour of the solution. .
Description. Odour, agreeable and aromatic; taste, agreeable
Iron (2.3.14). Dissolve the residue obtained in the test for and pungent.
sulphated ash in 4 ml of hydrochloric acid with the aid of
gentle heat, dilute with water to 50 ml and mix; 25 ml of the Identification
resulting solution complies with the limit test for iron (40 ppm).
A. Macroscopic - Rhizome laterally compressed, bearing
Fluorescence. No fluorescence should be visible under short, flattened, oblique branches; outer surface buff-coloured,
screened ultra-violet light. longitudinally striate; inner surface pale yellow, starchy and
Oxidising substances. To 5.0 gadd 10mlofwaterand 1 mlof fibrous. Fracture short with projecting fibers.
--acetic aci~ranastiruni;iiaI;omogeneoussu-sp·ensionTs B. MicroscopiC=- Fibro-vascular bundles"-tilld-6Ieore-sin cells--- -".,,-,-,,~ . _,---

·obfamea:Aaa-03 illJ. Of:.i freshly prepared safuiated solution witli-yellow·pigmenfsc:.itteredingroUiiatissue~Sfarcli·graiiis -


ofpotassium iOdide, mix and allow to stand for 5 minutes; no abundant in parenchyma cells, mostly simple, sack shaped,
distinct brown or blue colour is observed. spherical; hilum eccentric.

2544

www.webofpharma.com
IP 2010 SUNTHI EXTRACT

C. Determine by thin-layer chromatography (2.4.17), coating Inject the reference solution. The test is not valid unless the
the plate with silica gel GF254. relative standard deviation for the replicate injections is not
Mobile phase. A mixture of 30 volumes of hexane and more than 2.0 per cent.
70 volumes of diethyl ether. Inject the test solution and reference solution.
Test solution. Reflux 1 g of the coarsely powdered substance Calculate the contents oftotal gingerols by summing the peale
under examination with 25 rnl of methanol for 15 minutes, cool areas of 6-gingerol with all other peaks, which elute after 6-
and filter. Wash the residue with 10 ml of methanol. Combine gingerol and have a peak area ofat least 5 per cent ofthe peak
all the filtrates and concentrate to 10 ml. area of6-gingerol.
Reference solution. Reflux 0.5 g of coarsely powdered Storage. Store protected from heat, moisture and against attack
sunthi RS with 5 ml methanol for 15 minutes, cool and filter. by insects and rodents.
Apply to the plate 10 III of each solution as bands 10 rom by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 urn and 365 urn, spray
the plate with vanillin sulphuric acid reagent. Heat the plate Suothi Extract
at 100° for 5-10 minutes and examine the plate in day light. The Sunthi extract is obtained by extracting Sunthi (Zingiber
chromatographic profile of the test solution is similar to that officinale Roscoe, Fam. Zingiberaceae) dried rhizome ofwith
of the reference solution. ethanol or any other suitable solvent. The powdered extract
Tests may contain suitable excipients.
Sunthi extract contains not less than 90.0 per cent w/w and
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
not more than 120 percent w/w of the stated amount of total
Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent. gingerols and shogaols (sum of 6 gingerol, 8 gingerol, 10
Water-soluble extractive (2.6.3). Not less than 10.0 per cent gingerol and 6 shogaol). The content of6 shogaol shall not be
by Method I. more than the content of 6 gingerol.

Total ash (2.3.19). Not more than 8.0 percent. Description. Very light yellow to light brown powder or thick
liquid with characteristic odour and pungent taste.
Acid-:-insoluble ash (2.3 .19). Not more than 1.5 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Identification
heavy metals, Method B (20 ppm).
A. Determine by thin-layer chromatography (2.4.17), coating
Water (2.3.43). Not more than 12.0 per cent, determined on the plate with silica gel GF 254.
O.2g.
Mobile phase. A mixture of 30 volumes of hexane and 70
Microbial contamination (2.2.9). Complies with the microbial volumes of diethyl ether.
contamination tests.
Test solution. Dissolve 0.5 g ofextract under examination with
Assay. Determine by liquid chromatography (2.4.14). 50 ml of methanol and filter.
Test solution. Reflux about 3 g of the coarsely powdered Reference solution. 0.1 per cent w/v solution of 6 gingerol RS
substance under examination with 100 rnl of methanol on a in methanol.
water-bath for 15 minutes cool and filter. Reflux the residue
Apply to the plate 10 III ofeach solution as bands 10 rom by 2
further with methanol till the last extract turns colorless, cool
and filter. Combine all the filtrates and concentrate to 50.0 rnl
rom. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 urn. Spray the plate with
Reference solution. A 0.1 per cent w/v solution of vanillin sulphuric acid Heat the plate at 100° for 10 minutes
6-gingerol RS in methanol. and examine the plate at 365 nm and in day light. The
Chromatographic system chromatographic profile of the test solution is similar to that
- a stainless steel column 25 cm x 4.6 rom packed with ofthe reference solution.
octadecylsilane bonded to porous silica (5 Ilm),
mobile phase: 55 volumes of acetonitrile and 45 volumes Tests
of water,
Water (2.3.43).Notmorethan 5.0 percent, determined on2.0 g
- flow rate. 1.3 rnl per minute,
- spectrophotometer set at 278 urn, when dried at 105° for 3 hours.
- injection volume. 20 Ill. Total ash (2.3.19). Not more than 3 per cent.

2545

www.webofpharma.com
SUNTHI EXTRACT IP 2010

Heavy metals (2.3.13).1.0 g complies with the limit test for Identification
heavy metals, (Method B) 20 ppm.
A. To a solution in ethanol (90 per cent) add ferric chloride
Microbial contamination (2.2.9). Complies with the microbial test solution; a green colour is produced.
contamination test
B. To 1 g add to 5 ml ofwater, heat to boiling, filter, add 30 mg
Assay. Determine by liquid chromatography (2.4.14). of potassium permanganate and continue heating; the odour .
Test solution. Dissolve about Ig ofthe extract containing 100 of benzaldehyde is produced.
mg ofgingerols in methanol by heating, make up to 100 ml and
filter. Tests

Reference solution. A 0.1 per cent wlv solution of 6 gingerol Acidity. A solution in ethanol (90 per cent) is acidic to litmus
RS in methanol. solution.
Chromatographic system Acid value (2.3.23).97 to 160, calclliated on the dry, ethanol-
- a stainless steel column 25 cm x 4.6 mm packed with soluble matter, determined by the following method. Dissolve
octadecylsilane bonded to porous silica (5 11m), 5.0 g in 50 ml of boiling ethanol (90 per cent), add 3 mlof
- mobile phase: A mixture of 55 volumes of Acetonitrile phenolphthalein solution and titrate the hot solution with
and 45 volumes of water 1 M ethanolic potassium hydroxide. When the colour
- flow rate 1.3 ml per minute, becomes dark brown, attach to a reflux condenser, boil for a
- spectrophotometer set at 278 nm, few minutes to break up the precipitate and complete the
- injection volume.20 Ill. titration.
Inject the reference solution. The test is not valid unless the Ethanol-insoluble matter. Not more than 5 per cent, determined
relative standard deviation for the replicate injections is not by the following method. Digest 2.5 g with 50 ml of ethanol
more than 2.0 per cent. (90 per cent), filter through a sintered glass crucible, transfer
the residue to the filter crucible with the aid of more ethanol
Inject the reference solution and the test solution.
(90 per cent), wash with hot ethanol (90 per cent) until all
Calculate the content oftotal gingerols by summing the peak soluble matter is removed and dry to constant weight at 100°.
areas of 6 gingerol, 8 gingerol, 10 gingerol and 6 shogaol. The
relative retention time of 6 gingerol is 1.0, 8 gingerol is about Colophony. Add 5 g to 25 ml ofcarbon disulphide, warm gently
2.1, 10 gingerol is about 5.0 and 6 shogaol is about 2.6. The on a water-bath under a reflux condenser, filter, evaporate the
area ofthe peak of 6 gingerol in the sample chromatogram is solution to dryness, dissolve the residue in 6 ml of light
more than the area of 6 shogaol indicating the content of 6 petroleum (40° to 60°) and shake with 10 ml of a 0.5 per cent
gingerol is more than the content of 6 shogaol. wlv solution of cupric acetate; the light petroleum layer is
not coloured green.
Usual strengths. 5 per cent w/w; 20 per cent w/w.
Ester value (2.3.26).47 to 95, calculated on the dry, ethanol-
Storage. Store protected from heat and moisture. soluble basis.
Saponification value (2.3.37). 170 to 230, calculated on the
dry, ethanol-soluble basis.
Tolu Balsam Loss on drying (2.4.19). Not more than 4 per cent, determined
a
Balsam ofTolu on 2.0 g by drying in a thin layer in an oven at 60° over
phosphorus pentoxide at a pressure not exceeding 2.7 kPa.
Tolu Balsam is a solid or semi-solid, balsam obtained by incision
from the trunk of Myroxylon balsamum (Linn.) Harms (Fam. Assay. Weigh accurately about 2.0 g and boil with 25 ml of
Leguminosae). dilute ethanolic potassium hydroxide solution under a reflux
condenser for 1 hour. Remove the ethanol and digest the
Tolu Balsam contains not less than 35.0 per cent and not more residue with 50 ml ofhot water until diffused. Cool the liquid,
than 50.0 per cent of total balsamic acids, calculated as add 150 ml of water and 1.5 g of magnesium sulphate
cinnamic acid, C9H gOz, on the dry, ethanol-soluble matter. dissolved in 50 mlofwater. Mix thoroughly and set aside for
.... ~_._ ....Qes_cription.AsQft,.teml&i~:ms,b[Qwnis.hy_ellQ\YOTb.ro}'\T!:! 10 minu!es.Filt~r,\\,ashthe residue on the filter with 20 ml of
.!ll.a..~s,'Y_l!~1!fu§Lc.<:ln~.<:.t~Q;~tlQ~~qt!~I!!ly,1?-~C:<:>Il1!l1K~.~cl~~-water:a:cTCliiY the combined filtrate- an-Cl washings with--~-··­
and finally brittle. Transparent in thin films; odour, aromatic . hydroClilorlcaczdaiidextracfwiilisuccessivequanntiesoC
and vanilla-like. Warmed and pressed between pieces ofglass 50,40,30,30 and 30 ml ofether. Combine the ether extracts and
and examined with a lens, it exhibits crystals ofcinnamic acid. discard the aqueous portion. Extract with successive

2546

www.webofpharma.com
IP 2010 TRAGACANTH

quantities of 20, 20, 10, 10 and 10 ml of sodium bicarbonate solution. A slightly flocculent precipitate is fonned which,
solution, washing each aqueous extract with the same 20 ml when heated for 10 minutes on a water-bath, gives an intense
of ether. Discard the ether layers, acidify the combined yellow colour.
aqueous extracts with hydrochloric acid and extract with
D. Add 4 ml ofa 0.5 per cent w/v dispersion in water to 0.5 m1
successive quantities of 30, 20, 20 and 10 ml of chloroform, of hydrochloric acid and heat on a water-bath for 30 minutes.
filtering each chlorofOlm extract through a plug ofcotton wool
To one half of the resulting liquid add 1.5 ml of sodium
on which a layer of anhydrous sodium sulphate is placed.
hydroxide solution and 3 ml of alkaline cupric-tartrate
Evaporate the chlorofonn on a water-bath until about 10 ml
solution and heat on a water-bath; a reddish brown precipitate
remains and remove the remainder in a current ofair stopping
is fonned. To the other half of the liquid, add a few drops of
immediately when the last trace ofsolvent is removed. Dissolve
barium chloride solution; no precipitate is fonned (freedom
the residue by wanning with 10 ml of ethanol (95 per cent),
from agar).
previously neutralised to phenol red solution, cool and titrate
with 0.1 M sodium hydroxide using phenol red solution as Tests
indicator.
Acacia and other soluble gums. To 20 ml ofa 2.5 per cent w/v
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01482 g of
suspension of the powdered material prepared with freshly
total balsamic acids, calculated as cinnamic acid, C9H sOz.
boiled water add 10 ml of lead acetate solution; a flocculent
Storage. Store protected from light and moisture. Avoid precipitate is fonned. Filter and add to the filtrate 10 ml of lead
exposure to excessive heat. subacetate solution; a slight cloudiness may appear, but there
is no precipitate.
Karaya gum. Boil 1 g with 20 ml of water until a mucilage is
Tragacanth fonned, add 5 ml of hydrochloric acid and again boil for
5 minutes; no pink or red colour develops.
Tragacanth is the air-hardened gummy exudate, flowing
naturally or obtained by incision, from the trunk and branches Sterculia. A. Shake 0.2 g ofthe powdered material with 10 ml
of Astragalus gummifer Labill. and certain other species of of ethanol (60 per cent) in a 10-ml stoppered cylinder; any
Astragalus (Fam. Fabaceae). gel fOlmed occupies not more than 1.5 ml.
Description. Pale yellow, thin, flattened ribbons or brittle B. Shake 1 g ofthe powdered material with 100 m1 of water and
pieces; odourless and almost tasteless. On the addition of titrate with 0.01 M sodium hydroxide, using methyl red
about 10 times its weight of water, it forms a mucilaginous gel. solution as indicator. Not more than 5.0 ml is required to change
the colour of the solution.
It has the macroscopic and microscopic characteristics
described under Identification tests A and B. Foreign matter. Not more than 1.0 per cent, detennined by the
following method. To 2.0 g ofthe powdered material in a 250-
Identification . ml round-bottomed flask add 95 ml of methanol, swirl to
A. Macroscopic - Occurs as thin, flattened pieces, 30 mm moisten the powder and add 60 m1 of 7 M hydrochloric acid.
long, 10 mm wide and up to I mm in thic1mess, more or less Add a few glass beads and heat under a reflux condenser in a
curved, marked on the surface by fine longitudinal striae and water-bath for 3 hours, shaking occasionally. Remove the glass
concentric transverse ridges; white, translucent, horny; beads and filter the hot suspension under reduced pressure
fracture, short. May also be in the fonn ofthicker, less brittle through a sintered-glass crucible (porosity No.1). Rinse the
pieces, white to pale yellow and more opaque. flask with a small quantity of water, passing the rinsings
through the filter. Wash the residue on the filter with about
B. Microscopic - Reduce to powder. Examine under a 40 ml of methanol and dry to constant weight at 110°.
microscope; the powder shows in the gummy mass numerous
stratified cellular membranes which turn violet on the addition Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium
of iodinated zinc chloride solution. The mass includes starch carbonate, add 10 ml of bromine solution and mix thoroughly.
granules, isolated or in small clusters, rounded or occasionally Evaporate to dryness on a water-bath, gently ignite, and
defonned, diameter 4 to 10 /lm, and up to 20 /lm, with a central dissolve the cooled residue in 16 ml of brominated
hilum, visible in polarised light. hydrochloric acidAsT and 45 ml of water. Remove the excess
ofbromine with 2 ml of stannous chloride AsT. The resulting
C. Moisten 0.5 g ofthe powdered material with 1 ml of ethanol
solution complies with the limit test for arsenic (3 ppm).
(95 per cent) and add gradually, while shaking, 50 ml of water
until a homogeneous mucilage is obtained. To 5 ml of the Heavy metals (2.3.13). 0.5 g complies with the limit test for
mucilage add 5 m1 of water and 2 ml of barium hydroxide heavy metals, Method B (40 ppm).

2547

www.webofpharma.com
TULASI IP 2010

Total ash (2.3.19). Not more than 4.0 per cent, determined on and thin smooth cuticle. Trichomes bent, consisting of 2-6
·1.0g. cells; glandular trichomes short, lamiaceae type, consisting of
Microbial contamination (2.2.9). 1 g is free from Escherichia one stock cell and 2-4 cells with rounded heads. Palisade
coli and 10 g is free from salmonellae. parenchyma consists of layer of long cylindrical cells
containing chlorophyll; spongy parenchyma consists of
Storage. Store protected from moisture. polygonal cells with thin, straight or slightly wavy side walls.
Vascular bundles collateral type. Stomata diacytic.
C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Tulasi Mobile phase. A mixture of 97 volumes of toluene and
3 volumes of ethyl acetate.
Basil; Ocimum sanctum
Test solution. Reflux 2 g ofthe coarsely powdered substance
under examination with 25 ml of methanol for 15 minutes, cool
and filter. Reflux the residue further with 2 x 25 ml of methanol,
cool and filter. Combine all the filtrates and concentrate under
vacuum to 10 ml.
Reference solution. Reflux 1 g of tulasi RS with 25 ml of
methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 x 25 ml of methanol, cool and filter. Combine all
the filtrates and concentrate under vacuum to 5 ml.
Apply to the plate 10 fJ.I of each solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with anisaldehyde sulphuric acid reagent. Heat the plate
110° for 10 minutes and examine the plate in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.
Tulasi consists of leaves of Ocimum sanctum Linn (Fam.
Lamiaceae). Tests
Tulasi contains not less than 0.40 per cent w/w of eugenol, Foreign organic matter (2.6.1). Not more than 2.0 per cent.
calculated on the dried basis.
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent.
Description. Greyish-black in colour having characteristic
odour with slightly pungent and aromatic taste. Water-soluble extractive (2.6.3). Not less than 10.0 per cent
by method!.
Identification Total ash (2.3.19). Not more than 15.0 per cent.
A. Macroscopic - Leaves simple, elliptic, 2.7-7.5 cm long, Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
1-3 cm wide, with acute top, cuneate, obtuse to rounded base,
margin entire, undulate or serrate, both surfaces thinly Heavy metals (2.3.13). 1.0 g complies with the limit testfor
pubescent and dotted; petiole 0.2-3.0 cm long. Flowers are heavy metals, Method B(20 ppm).
5-7 mm in length. It has both male and female parts. Calyx: Loss on drying (2.4.19). Not more than 12.0 per cent,
There are 5 sepals and it is greenish in colour. Corolla: There determined on 5 g by drying in an oven at 105°.
are 5 petals, bilabiate in shape and covered with scattered
h' P t 1 h't' h 1 Microbial contamination (2.2.9). Complies with the microbial
aIrs. e a s w 1 IS -purp e. contamination tests.

.... _._. ~hap~::{i~ppe~:~~~::~s~e~~~~:s~~i:~~;:~::s~~~:


__ Ass~~:.~~t~~i~~~!'~~~~~~~~:~~a~~~ (2:~:13).. . .... _.. .__
... Jlll!!clral1glJJ~rtr!!!l~12!!1:{ll1t9_e..U~.witJ;1Jmll_W!!11~!!!lgJ.bi!t~.rllQQt1L
. r?.§£..s:Qltl!j()I1,R~j'lIIX_Q~~g.Q.f1b.e <::QaI§~IYP()w..Q~r<:l.g§1.!1:>~!IlJ1'<::~ .....
cuticle. On tangential view, these cells are polygonal with under examination with 50 ml of methanol on a water-bath for
straight or wavy walls. Lower epidermis consists ofa layer of 15 minutes, cool and filter. Reflux the residue further with
small, quadrangular transparent cells with thin walls and thin methanol till the extract turns colourless, cooi and filter.

2548

www.webofpharma.com
IP 2010 TULASI DRY EXTRACT

Combine all the filtrates and concentrate to a volume slightly at 365 nm and in day light. The chromatographic profile ofthe
less than 100 ml. Dilute to 100.0 ml with methanol. test solution is similar to that of the reference solution.
Reference solution. A 0.004 per cent w/v solution of
Tests
eugenol RS in methanol.
Chromatographic system Ethanol-soluble extractive (2.6.2). Not less than 15.0 per cent.
- a capillary column 30 m x 0.25 x 0.25 mm coated with100 Water-soluble extractive (2.6.3). Not less than 30.0 per cent
per cent dimethylpolysiloxane by method I.
temperature:
Total ash (2.3.19). Not more than 10.0 per cent.
oven 60° to 260° @l 0° per minute, (Initially and finally
hold for 5 minutes respectively) Heavy metals (2.3.13). 1.0 g complies with the limit test for
Injector 240°, heavy metals, Method B (20 ppm).
detector 280°,
Loss on drying (2.4.19). Not more than 6.0 per cent determined
flow rate. 0.8 ml per minute,
on 1 g by drying in an oven at 105°.
- split flow 20 ml per minute.
Microbial contamination (2.2.9). Complies with the microbial
Inject 1 III of the reference solution. The test is not valid
contamination tests.
unless the relative standard deviation for the replicate
injections is not more than 2.0 per cent. Assay. Determine by liquid chromatography (2.4.14).

Inject the refemce solution and the test solution. Test solution. Shakewell about 0.5 g of the extract under
examination in 100 ml ofthe methanol.
Calculate the content of eugenol.
Reference solution. A 0.03 per cent w/v solution of ursolic
Storage. Store protected from heat, moisture and against attack
acid RS in the methanol.
by insects and rodents.
Chromatographic system
- a stainless steel column, 25 cm x 0.46 mm packed with
octadescylsilane bonded to porous (0.5 Ilm),
Thlasi Dry Extract mobile phase: a mixture of30 volumes of mefhanol and
70 volumes of acetonitrile,
Tulasi Dry Extract is obtained from the Tulasi leaves ( Ocimum - flow rate. 0.6mlperminute,
sanctum Linn, Fam. Laminaceae) by extraction with methanol - spectrophotometer set at 210 urn,
or other any suitable solvent and evaporation of solvent.
- injection volume. 20 Ill.
Tulasi Dry Extract contains not less than 90.0 per cent w/w Inject the reference solution. The test is not valid unless the
and not more than 120.0 per cent w/w ofthe stated amount of relative standard deviation for replicate injections is not more
the ursolic acid, calculated on the dried basis. It may contain than 2.0 per cent.
suitable added substances.
Inject the reference solution and the test solution.
Description. A pale green powder.
Calculate the content of the ursolic acid in extract.
Identification Usual strengths. 2 per cent w/w; 3 per cent w/w.
A. Determine by thin layer chromatography (2.4.17) coating Storage. Store in a container purged with Nitrogen under the
the plate with silica gel GF254. refrigerated condition, protected from heat and moisture.
Mobile phase. A mixture of 95 volumes of chloroform and 5
volume of methanol.
Test solution. Shakewell 0.2 g ofthe extract under examination
with 10.0 ml methanol, filter.
Reference solution. A 0.02 per cent w/v solution of ursolic
acid RS in the methanol.
Apply to the plate 10 III of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and spray with anisaldehyde-sulphuric acid reagent. Heat
the plate at 110° for 10 minutes and examine in ultraviolet light

2549

www.webofpharma.com
VASAKA IP 2010

Vasaka Tests
Adulasa; Adhatoda vasica Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent.
Water-soluble extractive (2.6.3). Not less than 22 per cent by
Method I.
Total ash (2.3.19). Not more than 21.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux about 2 g of the coarsely powdered
Vasaka consists ofthe dried mature leaves ofAdhatoda vasica
,substance under examination with 50 ml of methanol on a
Neese (Fam. Acanthaceae).
water bath for 15 minutes, cool and filter. Reflux the residue
Vasaka contains not less than 0.6 per cent w/w of vasicine, further with methanol till the last extract turns colorless, cool
calculated on the dried basis. and filter. Combine all the filtrates and concentrate to 100.0 ml.
Description. Taste, bitter. Reference solution. Dissolve 25 mg ofvasicine hydrochloride
RS in 50 ml of methanol. Dilute 5.0 ml ofthis solution to 50.0 ml
Identification with methanol.
A. Macroscopic - Leafpieces membranous, brittle, greyish- Chromatographic system
brown, a few pieces green coloured. Floral bracts leaf-lilee. - a stainless steel column 25 em x 4.6 rom packed with
octadecylsilane bonded to porous silica (5 f.!m),
B. Microscopic - Stomata diacytic, more on the lower
- mobile phase: a mixture of 3 volumes of the solution
epidermis; glandular and non-glandular hair on both surfaces
ofleaf; elongated cystoliths present in palisade cells. prepared by dissolving 1 g of sodium hexane-
sulphonate in 1000 ml of water, 1 volume of acetonitrile
C. Determine by thin-layer chromatography (2.4.17), coating and 20 volumes of glacial acetic acid,
the plate with silica gel GF254. flow rate. 1 ml per minute,
Mobile phase. A mixture of 8 volumes of ethyl acetate, spectrophotometer set at 300 nm,
2 volumes of methanol and 0.2 volume of strong ammonia injection volume. 20 f.!l.
solution.
Inject the reference solution. The test is not valid unless the
Test solution. Reflux 1 g ofcoarsely powdered substance under relative standard deviation for the replicate injections is not
examination with 50 ml methanol for 15 minutes, cool and more than 2.0 per cent.
filter. Reflux the residue further with 2 x 50 ml of methanol,
Inject the reference solution and the test solution.
cool and filter. Combine all the filtrates and concentrate to
IOml. Calculate the content of vasicine.
Reference solution. Reflux 0.5 g of vasaka RS with 50 ml Storage. Store protected from heat, moisture and against attack
methanol for 15 minutes, cool and filter. Reflux the residue by insects and rodents.
further with 2 x 50 ml of methanol, cool and filter. Combine all
the filtrates and concentrate to 5 ml.
Apply to the plate 10 f.!l ofeach solution as bands 10 rom by 2 Vasaka Extract
.~~._.~ mm.Allo.wthemobilephasetorise.8cm.Dry the.plateinaiL.~__ ._ ~......................_ _~ __ .~_._
llIld tl:x:alllille iIlllltrayioM ligl1tllt254IllllaIl<l36.5Illll,spraYYll~alelle:x:trll<::tj.s()1J!lliJJ~41Jye:x:trll<::til1gya,sllka(4d,hgtQclg
with dragendOiff's reagent. Heat the plate at 100° for 5-10 vasica Nees, Fam. Acanthaceae) dried matured leaves with
minutes and examine in day light. The chromatographic profile ethanol or any other suitable solvent. The powdered extract
ofthe test solution is similar to that ofthe reference solution. may contain suitable excipients.

2550

www.webofpharma.com
IP 2010 YASTI .

Vasaka extract contains not less than 90.0 per cent w/w and - injection volume. 20 Ill.
not more than 120.0 percent w/w of the stated amount of
Inject the reference solution. The test is not valid unless the
vasicine.
relative standard deviation for the replicate injections is not
Description. Light green to greenish brown powder; more than 2.0 per cent.
characteristic odour and bitter taste.
Inject the refernce solution and the test solution.
Identification Calculate the content of vasicine.
A. Determine by thin-layer chromatography (2.4.17), coating Usual strengths. 1 per cent w/w; 1.5 per cent w/w.
the plate with silica gel GF254. Storage. Store protected from heat and moisture.
Mobile phase. A mixture of 8 volumes of ethyl acetate, 2
volumes of methanol and 1 volume of strong ammonia.
Test solution. Dissolve 1.0 g ofeJ:(.tract under examination with Yasti
25 ml of methanol and filter.
Liquorice root; Mulethi; Glycyrrhiza glabra
Reference solution. A 0.1 per cent w/v solution of vasicine
hydrochloride RS in methanol.
Apply to the plate 10 III of each solution as bands 10 mm by 9

2 mm. Allow the mobile phase to rise 8 cm.Spray the plate with 8
Dragendorff reagent.Dry the plate in air and examine in 7
ultraviolet light at 254nm and 365 urn.The chromatographic
6
profile of the test solution is similar to that of the reference
solution.
4
Tests
3
Loss ofdrying (2.4.19). Not more than 6.0 per cent, detennined
on 2.0 g when dried at 105° for 3 hours.
Total ash (2.3.19). Not more than 2.0 percent.
o
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, (Method B) 20 ppm. Yasti consists of the dried, unpeeled roots and stolons of
. Microbial contamination (2.2.9). Complies with the microbial Glycyrrhiza glabra Linn. (Fam. Leguminosae).
contamination test. Yasti contains not less than 3.0 per cent w/w of glycyrrhizinic
Assay. Detennine by liquid chromatography (2.4.14). acid.
Test solution. Dissolve about 1.5 g of the extract containing Description. Odour, characteristic and slightly aromatic; taste,
20 mg ofvasicine in 100 ml of methanol by gently heating, and very sweet and faintly astringent; the bark is not bitter.
filter.
Identification
Reference solution. A 0.025 per cent w/v solution ofvasicine
hydrochloride RS in methanol. A. Macroscopic - Root with few branches, up to 1 m long
and 0.5 to 3 cm in diameter. Bark, brownish-grey to brown with
Chromatographic system
longitudinal striations, bearing traces oflateral roots. Stolons,
a stainless steel column 250 cm x 4.6 mm packed with
cylindrical, 1 to 2 cm in diameter and up to several meters long,
silica bonded with cyanopropyl groups (5 Ilm),
but may be cut into lengths of 10 to 15 cm; similar in external
mobilephase: A mixture of 92 volumes ofbuffer solution
appearance to the root but with occasional small buds. Fracture
prepared by dissolving 1.36 g ofpotassium dihydrogen
of the root and stolon, granular and fibrous. Cork layer, thin;
ortho phosphate in 500 ml of water and 2 ml of
secondary phloem region, wide, light yellow with radial
orthophosphoric acid, dilute to 1000 ml with water, 5
volumes of acetonitrile and 3 volumes of striations; xylem, compact, yellow, with radiate structure. The
tetrahydrofitran, stolon has a central pith which is absent from the root.
flow rate. 1.0 ml per minute, E. Microscopic - Cork and phelloderm are narrow. Phloem
spectrophotometer set at 300 nm, consisting ofbundles ofthick-walled, yellow fibres with narrow

2551

www.webofpharma.com
YASTI IP 2010

lumina surrounded by cells each containing a calciumoxalate Assay. Determine by liquid chromatography (2.4.14).
prism, alternatingin the external layers with areas of strongly Test solution. Weigh accurately about 1.0 g of the coarsely
hyaline keratenchyma; functional sieve tissue near the powdered substance under examination in a 250-ml conical
cambium. Medullary rays parenchymatous, widening towards flask, add 100 ml of 0.1 M ammonia and mix with the aid of
the exterior, 3 to 12 cells wide. Xylem composed ofradial rows ultrasound for 30 minutes. Centrifuge a part ofthe supernatant
oftracheids and vessels alternating with bundles of lignified liquid and dilute 1.0 m1 to 5;0 rn1 with 0.1 M ammonia. Filter the
fibres with crystal sheaths similar to those of the secondary solution through a membrane filter disc with an average pore
phloem; vessels 30 J-lm to 150 J-lm in diameter with thick walls diameter not greater than 1.0 J-lm and use the filtrate.
(5 J-lm to 10 J-lm) having reticulate thickenings or numerous
Reference solution. A 0.005 per cent w/v solution of
bordered pits with slit-shaped openings associated with
glycyrrhizinic acid RS in 0.1 M ammonia.
lignified xylem parenchyma. Medullary rays, 2 to 5 cells wide.
Parenchymatous cells throughout containing simple, round, Chromatographic system
oval or fusiform starch granules 2 J-lm to 20 J-lm, mostly 5 J-lm to a stainless steel column 25 cm x 4.6 mm, packed with
12 J-lm, in diameter; parenchymatous pith present solely in the octadecylsilane bonded to porous silica (5 J-lm),
stolon. mobile phase: a mixture of 6 volumes of glacial acetic
acid, 30 volumes of acetonitrile and 64 volumes of water,
C. Determine by thin-layer chromatography (2.4.17), coating
- flow rate. 1.5 ml per minute,
the plate with silica gel GF254. - spectrophotometer set at 254 nm,
Mobile phase. A mixture of 70 volumes of butyl alcohol, 20 injection volume. 20 J-ll.
volumes of water and 10 volumes of acetic acid.
Inject the reference solution. The test is not valid unless the
Test solution. Add 10 ml of 70 per cent v/v methanol to 1 g of relative standard deviation for replicate injections is not more
dried yasti powder, heat by shaking on a water bath for than 2.0 per cent.
5 minutes, cool and filter.
Inject the test solution and the reference solution arid measure
Reference solution. Add 10 ml ono per cent v/v methanol to the responses for the analyte peak.
1 g of dried yasti RS powder, heat by shaking on a. water bath
Calculate the content of glycyrrhizinic acid.
for 5 minutes, cool and filter
Storage. Store protected from light and moisture.
Apply to the plate 10 J-ll of each solution as bands 10 mm by 2
mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultraviolet light at 254 nm and 365 nm, spray
with anisaldehyde sulphuric acid reagent. Heat the plate at
105° for 10 minutes and examine the plate in day light. The
Yasti Dry Extract
chromatographic profile of the test solution is similar to that Yasti Dry Extract is obtained by extracting Yasti with water or
of the reference solution. aqueous ethanol or any other suitable solvent.
D. Mix a small quantity, in powder, with 0.05 ml of sulphuric Yasti Dry Extract contains not less than 90.0 per cent w/w and
acid; the powder particles become orange-yellow and some not more than 120.0 per cent w/w of the stated amount of
fragments change, more slowly, to pinkish red. glycyrrhizinic acid.
Description. A yellowish-brown to dark brown powder.
Tests
Identification
Curcuma. When examined under a microscope, none of the
fragments ofthe powder in sulphuric acid (see Identification A. Determine by thin-layer chromatography (2.4.17), coating
test D) should immediately take on a carmine-red colour. the plate with silica gel G.
Water-soluble extractive (2.6.3). Not less than 20 per cent, Mobile phase. A mixture of70 volumes of butanol, 10 volumes
determined by the following method. Mix 2.5 g of the finely of acetic acid and 20 volumes of water.
powdered drug with 50 ml of water and allow to stand for Test solution. Dissolve 200 mg ofthe extract under examination
2 hours, shaking frequently. Filter, evaporate 10.0 g of the with 50 ml of methanol (70 per cent) and filter.
-----filtrateto-·dryness-on-a-water-bath,drytheresidueatl05()and -Rejerencesolflti{fii.AO.1percent-w/v·solijlionofglycytthizin·--
weigh._ --- --------.---.-.---.-.-.--...-----.-.---.- .-- -ammonical hydrateRS in methanol 00 percent);
Acid-insoluble ash (2.3.19). Not more than 2.0 percent. Apply to the plate 10 J-ll of each solution as bands 10 mm by
Sulphated ash (2.3.18). Not more than 10.0 percent. 2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air

2552

www.webofpharma.com
IP 2010 YASTI DRY EXTRACT

and examine in ultraviolet light at 254 nm and 365 nm. Spray mobile phase by mixing for 30 minutes with the aid ofultrasound
the plate with anisaldehyde sulphuric acid reagent and heat and dilute to 1OO.Oml with the mobile phase and filter.
the plate at 105° for 10 minutes and examine the plate in day
Reference solution. A 0.01 per cent w/v solution of
light. The chromatogram obtained with test solution shows a
glycyrrhizin ammonical hydrate RS in the mobile phase.
band corresponding to the band obtained with the reference
solution indicating the presence of glycyrrhizin. Chromatographic system
a stainless steel column 25 cm x 4.6 llllll, packed with
Tests octadecylsilane bonded to porous silica (5 /lm),
- mobile phase: a mixture of 1 volume of glacial acetic
Acid insoluble ash (2.3.19). Not more than 2.0 per cent. acid, 67 volumes of methanol and 33 volumes of 0.2 M
Heavy metals (2.3.13). 1.0 g complies with the limit test for ammonium acetate in water,
heavy metals, Method B (20 ppm). flow rate. Iml per minute,
spectrophotometer set at 250 nm,
Loss ofdrying (2.4.19). Not more than 5.0 per cent, determined injection volume. 20 Ill.
on 1.0 g by drying in an oven at 105°.
Inject the reference solution. The test is not valid unless the
Microbial contamination (2.2.9). Complies with the microbial relative standard deviation for the replicate injections is not
contamination tests. more than 2.0 per cent.
Assay. Determine by liquid chromatography (2.4.14). Inject the reference solution and the test solution.
Test solution. Dissolve about 500 mg ofthe extract or a quantity Calculate the content of glycyrrhizinic acid in the extract.
containing 7 mg of glycyrrhizinic acid and dissolve in the Storage. Store protected from heat and moisture.

2553

www.webofpharma.com
www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

BLOOD AND BLOOD-RELATED PRODUCTS

Anti-A Blood Grouping Serum 2557


Anti-B Blood Grouping Serum 2557
Anti-D (Rho) ImrnlUloglobulin 2558
Anti-D ImrnlUloglobulin for Intravenous Use 2559
Anti-Hmnan Globulin Serum 2560
Blood Grouping Serums Anti-D, Anti-C, Anti-E, Anti-c, Anti-e 2560
Concentrated Hmnan Red Blood Corpuscles 2562
CryoprecipitatedAntihaemophilic Factor 2562
Dried HmnanAntihaemophilic Fraction 2563
Fibrin Sealant Kit 2566
HumanAlbumin 2568
Hmnan Coagulation Factor IX 2570
Hmnan Coagulation Factor vn 2571
Hmnan Coagulation Factor VIII (rDNA) 2572
HumanNonnal ImmlUloglobulin 2574
Human Plasma Protein Fraction 2576
Hmnan Prothrombin Complex 2578
HmnanNonnallmmlUloglobulin for Intravenous Use 2579
Plasma for Fractionation 2586
Platelet Concentrate 2588
Whole Human Blood 2589

2555

www.webofpharma.com
www.webofpharma.com
IP 2010 ANTI-B BLOOD GROUPING SERUM

Anti-A Blood Grouping Serum after the date of issue from manufacturer's cold storage (5°,1
year; or 0°, 2 years), provided that the expiration date for dried
Anti-A Blood Grouping Serum is a sterile, liquid or dried serum is not later than 1 year after constitution.
preparation containing the particular blood group antibodies
Storage. Store at a temperature between 2° and go.
derived from high-titered blood plasma or serum of human
subjects, with or without stimulation by the injection ofBlood Labelling. The label states that the source material was not
Group Specific Substance A (or AB). It contains a suitable reactive for hepatitis B surface antigen, but that no known
antimicrobial preservative. It is of two types polyclonal & test method offers assurance that products derived from human
monoclonal. blood will not transmit hepatitis in case of polyclonal. Label
Production also states that it is for in vitro diagnostic use.

The monoclonal antibody technique devised by Kohler and NOTE-The labeling is in black lettering imprinted on paper
Milsten has proved useful in producing high titre and specific that is white or is coloured completely or in part to match
antibodies. Laboratory animals, usually mice are immunized the specified blue colour standard.
for the production of monoclonal antibodies. After suitable
immune response, mouse spleen cells containing antibody
secreting lymphocytes are fused to neoplastic plasma cells of Anti-B Blood Grouping Serum
infinite reproductive capacity from a mouse (that is myeloma
cells). The resulting hybridomas are screened for antibody Anti-B Blood Grouping Serum is a sterile, liquid or dried
with the required specificity and affinity. The antibody preparation containing the particular blood group antibodies
secreting clones may then be propagated in tissue culture or derived from high-titered blood plasma or serum of human
by inoculation into mice in which case the antibodies are subjects, with or without stimulation by the injection ofBlood
collected as ascites. The clonal line produces a single antibody, Group Specific Substance B (or AB). It contains a suitable
there is no need for absorption or to remove heterospecific antimicrobial preservative.
antibodies. All antibody molecules produced by a clone of
Production
hybridoma cells are identical in terms of antibody structure
and antigen specificity. Once one antibody secreting clone of The monoclonal antibody technique devised by Kohler and
cells has been established, antibody with same specificity Milsten has proved useful in producing high titre and specific
and reaction characteristics will be available indefmitely. antibodies. Laboratory animals, usuallymice are immunized
for the production of monoclonal antibodies. After suitable
Identification immune response, mouse spleen cells containing antibody
It agglutinates human red cells containing A-antigens that is secreting lymphocytes are fused to neoplastic plasma cells of
blood groups A and AB (including subgroups A lo A2, AlB, infinite reproductive capacity from a mouse (that is myeloma
and A2B but not necessarily weaker subgroups). cells). The resulting hybridomas are screened for antibody
with the required specificity and affinity. The antibody
Tests secreting clones may then be propagated in tissue culture or
by inoculation into mice in which case the antibodies are
It complies with the test for potency, in parallel with, and not
collected as ascites. The clonal line produces a single antibody,
less than equivalent to, the Reference Blood Grouping Serum
there is no need for absorption or to remove heterospecific
Anti-A, in agglutinating red blood cells from Group Al and
antibodies. All antibody molecules produced by a clone of
Group A2B donors. It complies with the tests for specificity
hybridoma cells are identical in terms of antibody structure
with Group AI, A2B, B, and 0 cells and confirms the absence
and antigen specificity. Once one antibody secreting clone of
of contaminating antibodies reactive with Mg, Wra antigens
cells has been established, antibody with same specificity
as well as other antigens having an incidence of I per cent or
and reaction characteristics will be available indefinitely.
greater in the general population (see under Blood Grouping
Serums Anti-D, Anti-C, Anti-E, Anti-c, rAnti-e). It complies Identification
with the tests for avidity with Group Al and A2B cells. All
fresh or frozen red blood cell suspensions used for these It agglutinates human red cells containing B-antigens, that is
tests are prepared under specified conditions and meet blood groups Band AB (including subgroups AlB and A2B).
specified criteria. Anti-A Blood Grouping Serum may be
artificially coloured blue. "Tests
Expiration date. The expiration date for liquid serum is not It complies with the test for potency, in parallel with, and not
later than I year, and for dried serum not later than 5 years less than equivalent to, the Reference Blood Grouping Serum

2557

www.webofpharma.com
ANTI-B BLOOD GROUPING SERUM IP 2010

Anti-B, in agglutinating red blood cells from Group B donors. with D-positive erythrocytes that are compatible in relevant
It complies with the tests for specificity with Group AI> B, and blood group systems in order to avoid fonnation ofundesirable
o cells and confirms the absence of contaminating antibodies antibodies.
reactive with Mg, WI" antigens as well as other antigens having
Erythrocyte donors
an incidence of 1.0 per cent or greater in the general population
(see under Blood Grouping Serums Anti-D, Anti-C, Anti-E, Erythrocyte donors comply with the requirements for donors
Anti-c, Anti-e). It complies with the test for avidity with Group prescribed in the monograph on Human Plasma for
B cells. All fresh or frozen red blood cell suspensions used for Fractionation.
these tests are prepared under specified conditions and meet Immunisation
specified criteria. Anti-B Blood Grouping Serum may be
artificially coloured yellow. Immunisation ofthe plasma donor is carried out under proper
medical supervision. Recommendations concerning donor
Expiration date. The expiration date for liquid serum is not immunisation, including testing of erythrocyte donors, have
later than 1 year and for dried serum not later than 5 years after been formulated by the World Health Organisation
the date of issue from manufacturer's cold storage (5°, 1 year; (Requirements for the collection, processing and quality
or 0°, 2 years), provided that the expiration date for dried serum control ofblood, blood components and plasma derivatives,
is not later than 1 year after constitution. WHO Technical Report Series, No. 840, 1994 or subsequent
Storage. Store at a temperature between 2° and 8°. revision).
Pooled plasma
Labelling. The label states that the source material was not
reactive for hepatitis B smface antigen, but that no known To limit the potential B19 virus burden in plasma pools used
test method offers assurance that products derived from human for the manufacture ofanti-D immunoglobulin, the plasma pool
blood will not transmit hepatitis in case of polyclonal. Label is tested for B 19 virus using validated nucleic acid
also states that it is for in vitro diagnostic use. amplification techniques (2.8.1).
NOTE-The labelling is in black lettering imprinted on paper B19 virus DNA. Maximum 104 ill per ml.
that is white or is coloured completely or in part to match A positive control with 104 ill ofB 19 virus DNA per ml and, to
the specified yellow colour standard. test for inhibitors, an internal control prepared by addition of
a suitable marker to a sample ofthe plasma pool are included
in the test. The test is invalid if the positive control is non-
Anti-D (Rho) Immunoglobulin reactive or if the result obtained with the internal control
indicates the presence of inhibitors.
Human Anti-D Immunoglobulin
BI9 virus DNA for NAT testing reference preparation is
Human anti-D immunoglobulin is a liquid or freeze-dried suitable for use as a positive control.
preparation containing immunoglobulins, mainly
IfHuman Normal Immunoglobulin is added to the preparation,
immunoglobulin G. The preparation is intended for
the plasma pool from which it is derived complies with the
intramuscular administration. It contains specific antibodies
above requirement for B 19 virus DNA.
against erythrocyte D-antigen and may also contain small
quantities of other blood-group antibodies. Human normal Tests
immunoglobulin may be added.
Potency. Determine the assay of human anti-D
It complies with the monograph on Human Normal immunoglobulin by Method A (2.8.2). The estimated potency
Immunoglobulin, except for the minimum number of donors is not less than 90.0 per cent of the stated potency. The
and the minimum total protein content. For products prepared confidence limits (P = 0.95) are not less than 80.0 per cent and
by a method that eliminates hmnunoglobulins with specificities not more than 120.0 per cent ofthe estimated potency.
other than anti-D, where authorised, the test for antibodies to Method B or C (2.8.3) may be used for potency determination
hepatitis B surface antigen is not required. if a satisfactory correlation with the results obtained by
Production Method A has been established for the particular product.
Storage. For the liquid preparation, store protected from light,
..... _H!1!I!!ll1!ll1ti:I::> i!I!!I!1ll1og1()1:>ulil1i~.J:lr:t'lfeI'l1lJ1Y()1:>tl1i.l1t'ltlft()fl1 .···in·a-plastic·container;·Forthefreeze-driedpreparation,store _.
the plasma of donors with a sufficient titre of previously
. ·acqulre(ranti.~D anti.bbClIes. ·Where necessary; In order to ··-protectedfromlight,inaplastic container;· .
ensure an adequate supply ofhuman anti-Dimmunoglobulin, Labelling. The label states (1) for liquid preparations, the
it is obtained from plasma derived from donors immunised volume of the preparation in the container and the protein

2558

www.webofpharma.com
IP 2010 ANTI-D IMMUNOGLOBULIN FOR INTRAVENOUS USE

content expressed in grams per litre; (2) for freeze-dried 3. Antibodies against hepatitis C virus (anti-HCV).
preparations, the quantity of protein in the container; (3) the
4. Hepatitis B surface antigen (HBsAg),
route of administration; (4) for freeze-dried preparations, the
name or composition and the volume of the reconstituting Pending complete harmonisation of the laboratory tests to be
liquid to be added; (5) where applicable, that the preparation carried out, the competent authority may require that a test for
is suitable for use in the prophylaxis of hepatitis A infection; alanine aminotransferase (ALT) also be carried out.
(6) where applicable, the anti-hepatitis A virus activity in The test methods used are of suitable sensitivity and
International Units per ml; (7) where applicable, the name and specificity and comply with the regulations in force. Ifa repeat-
amount of antimicrobial preservative in the preparation; (8) reactive result is found in any of these tests, the donation is
the number ofInternational Units per container. not accepted.

Immunisation
Immunisation ofthe plasma donor is carried out under proper
Anti-D Immu.noglobulin for medical supervision. Recommendations concerning donor
Intravenous Use immunisation, including testing of erythrocyte donors, have
been formulated by the World Health Organisation
Anti-D Immunoglobulin Human for Intravenous Use (Requirements for the collection, processing and quality
Anti-D Immunoglobulin for intravenous administration is a control ofblood, blood components and plasma derivatives,
liquid or freeze-dried preparation containing immunoglobulins, WHO Technical Report Series, No. 840, 1994 or subsequent
mainly immunoglobulin G. It contains specific antibodies revision).
against erythrocyte D-antigen and may also contain small
Pooled plasma
quantities of other blood-group antibodies. Human normal
immunoglobulin for intravenous use may be added. To limit the potential B 19 virus burden in plasma pools used
for the manufacture of anti-D immunoglobulin, the plasma
It complies with the monograph on Human Normal
pool is tested for B 19 virus using validated nucleic acid
Immunoglobulin for Intravenous Administration, except for
amplification techniques (2.8.1).
the minimum number of donors, the minimum total protein
content, the limit for osmolality and the limit for prekallikrein Identification
activator. For products prepared by a method that eliminates
immunoglobulins with specificities other than anti-D where Examine by a suitable immunoelectrophoresis technique. Using
authorised, the test for antibodies to hepatitis B surface antigen antiserum to normal human serum, compare normal human
is not required; a suitable test for Fc function is carried out. serum and sample under examination in a dilution of 109 per
litre ofprotein. The main component ofthe sample corresponds
Production to the IgG component ofnormal human serum.
Polyclonal (human) anti Rh (D) serum
Tests
Human anti-D immunoglobulin is preferably obtained from
B19 virus DNA. Maximum 104 IU per ml.
the plasma of donors with a sufficient titre of previously
acquired anti-D antibodies. Where necessary, in order to A positive control with 104 IU ofB 19 virus DNA per ml and,
ensure an adequate supply ofhuman anti-D immunoglobulin, to test for inhibitors, an internal control prepared by addition
it is obtained from plasma derived from donors immunised ofa suitable marker to a sample ofthe plasma pool are included
with D-positive erythrocytes that are compatible in relevant in the test. The test is invalid if the positive control is non-
blood group systems in order to avoid formation ofundesirable reactive or if the result obtained with the internal control
antibodies. indicates the presence of inhibitors.
Bi9 virus DNA for NAT testing RP is suitable for use as a
Erythrocyte donors
positive control.
Laboratory tests are carried out for each donation to detect
If Human normal immunoglobulin for intravenous
the following viral markers:
administration is added to the preparation, the plasma pool
1. Antibodies against human immunodeficiency virus 1 (anti- from which it is derived complies with the above test for B19
HlV-I), virus DNA.
2. Antibodies against human immunodeficiency virus 2 (anti- Assay. Determine by Method A of human anti-D
HlV-2), immunoglobulin (2.8.2). The estimated potency is not less

2559

www.webofpharma.com
ANTI- HUMAN GLOBULIN SERUM IP 2010

than 90.0 per cent ofthe stated potency. The confidence limits suspended in isotonic saline sensitized with decreasing
(P = 0.95) are not less than. 80.0 per cent and not more than amounts of nonagglutinating Anti-D (Anti-Rho) serum, and
120.0 per cent of the estimated potency. with cells sensitized in the same manner with an
immunoglobulin IgG Anti- Fya serum ofsimilar potency. Anti-
MethodB or C (2.8.3) may be used for potency detennination
Human Globulin Serum containing one or more Anti-
ifa satisfactory correlation with the results obtained by Method
complement components complies with the tests for potency
A has been established for the particular product.
in giving a 2+ agglutination reaction (Le., agglutinated cells
Storage. For the liquid preparation, store protected from light, dislodged into many small clumps of equal size) by the low-
in a colourless glass container, at the temperature stated on ionic sucrose or sucrose-trypsin procedures when tested as
the label. For the freeze-dried preparation, store protected from recommended in the labelling. Anti-Human Globulin Serum
light, in colourless glass container, at a temperature not containing Anti-3Cd activity meets the requirements for
exceeding 25 0 • stability, by potency testing of representative lots every 3
months during the dating period.
Labelling. The label states (1) for liquid preparations, the
volume of the preparation in the container and the protein Expiration date. Its expiration date is not later than 1year after
content expressed in gram per litre; (2) for freeze-dried the date of issue from manufacturer's cold storage (5°, I year;
preparations, the quantity of protein in the container; (3) the or 0°, 2 years).
amount of immunoglobulin in the container; (4) the route of
administration, (5) for freeze-dried preparations, the name or Storage. Store at a temperature between 2° and 8°.
composition and the volume of the reconstituting liquid to be
added; (6) the distribution of subclasses of immunoglobulin Labelling. The label states the animal source of the product,
G present in the preparation; (7) where applicable, the amount the specific antibody activities present; the application for
of albumin added as a stabilizer; (8) the maximum content of which the reagent is intended; a cautionary statement that it
immunoglobulin A; (9) the number ofIntemational Units per does not contain antibodies to immunoglobulins or that it
container. does not contain antibodies to complement components,
wherever and whichever is applicable; and states that it is. for
in vitro diagnostic use.

Anti-Human Globulin Serum NOTE-The lettering on the label of the general-purpose


polyspecijic reagent is black on a white background. The
Anti-Human Globulin Serum is a sterile, liquid preparation of label ofall other Anti-Human Globulin Serum containers is
serum produced by immunizing lower animals such as rabbits in white lettering on a black background.
or goats with human serum or plasma, or with selected human
plasma proteins. It is free from agglutinins and from hemolysins
to nonsensitized human red cells of all blood groups. It
contains a suitable antimicrobial preservative. Blood Grouping Serums Anti-D, Anti-
Three varieties ofAnti-Human Globulin Selum are recognized: C, Anti-E, Anti-c, Anti-e
(1) a general-purpose polyspecific reagent which, as a
minimum, contains antibodies specific for immunoglobulin IgG, Anti-Rh Blood Grouping Serums
and at least the C3d component of human complement (for Blood Grouping Serums Anti-D, Anti-C, Anti-E, Anti-c, Anti-
use in the direct antiglobulin test, it contains this Anti-C3d e (Anti-Rh Group) are sterile, liquid or dried preparations derived
and Anti-IgG activity) and which may be artificially coloured from the blood plasma or serum of human subjects who have
green; (2) a reagent containing antibodies only against developed specific Rh antibodies. They are free from
immunoglobulin IgG (not heavy chain specific) intended for agglutinins for the A or B antigens and from alloantibodies
use in the indirect antiglobulin test, and which may be other than those for which claims are made in the labelling.
artificially coloured green; and (3) reagents containing They contain a suitable antimicrobial preservative. Liquid
antibodies specific for individual or selected components of serums are not artificially coloured. Two varieties ofAnti-Rh
human complement, such as Anti-C3, and Anti-C3b-C3d-C4, Blood Grouping Serums are recognized, i.e., (1) saline
or a single class ofimmunoglobulins, such as Anti-IgG (heavy agglutinating "complete" antiserums, which specifically
chainspecific),used onlyt03dentify plasma components agglutinatehumalLIed blood. cells suspended in saline and
cQat~c::l Qll t1;l~sllrfac:eofI't:c::l1::>looc::lc:c:lli>.Anti~II1ll11a!1.<::i:19_1::>1lIil1 (2) ''1::>locking or inc:ol11pl<::t~"a.t;ltii>~I'lll11i>,wllicl1C:Ql1ta.inPI9t e:!11
Serum containing Anti-IgG complies with the test for potency, or other macromolecular substances, usually require the cells
in parallel with the Reference Anti-Human Globulin (Anti- to be suspended in serum or plasma,and generally are for
IgG) Serum (at a 1:4 dilution) when tested with red cells slide or rapid tube tests. The most commonly used of these

2560

www.webofpharma.com
IP 2010 BLOOD GROUPING SERUMS ANTI-D, ANTI-C, ANTI-E, ANTI-c, ANTI-e

blood grouping serums are listed in Table 1 each reacting with antigens having an incidence of 1.0 per cent or greater in the
the antigen(s) designated by the corresponding letter(s) with general population, except where some ofthese confirmatory
the alternative nomenclature indicated parenthetically. tests can not be done by the manufacturer, in which event
such omissions are noted.
Table - 1
Serum Antigen(s) Reacting Table-3

Anti-D (Anti-Rho) D(Rho) Serum Cells

Anti-C (Anti-rh') C(rh') Anti-D CcDe, cDe, Ccde, cdEe, Al cde, B cde, 0 cde,
and where recommended for use by indirect
Anti-E (Anti-rh") E(rh") antiglobulin technique, cde Bg(a+) cells from 3
Anti-CD (Anti-Rho') D (Rho) and C (rh') different donors

Anti-DE (Anti-Rh o") D (Rho) and E (rh") Anti-C cDe, Ccde, cdEe, C + rh i neg. cells, Al cde, B
lll cde, 0 cde
Anti-CDE (Anti-Rho ) D (Rho), C (rh'), and E (rh')
Anti-c (Anti-hr') c (hr') Anti-E cDe, Ccde, cdEe, Al cde, B cde, 0 cde
Anti-e (Anti-hr") e (hr") Anti-CD cDe, Ccde, cdEe, Al cde, B cde, 0 cde, and
where recommended for detection ofthe G
Each serum complies with the test for potency in the case of antigen, rGr
serums for saline tube test in parallel with, and not less than
equivalent to, the Reference Blood Grouping Serum for Anti- Anti-DE cDe, Ccde, cdEe, Al cde, B cde, 0 cde
D, Anti-C, or Anti-E, whichever is applicable, or, in the case of Anti-CDE cDe, Ccde, cdEe, Al cde, B cde, 0 cde, and
Anti-c and Anti-e for saline tube test which have no reference where recommended for detection ofthe G
preparations, the test for minimum agglutination reactivity at antigen, rOr
a specified dilution; and in the case of serums for slide or
rapid tube test in parallel with, and not less than equivalent to, Anti-c Ccde, Al CDe, B CDe, 0 CDe, and CDEe or
the Reference Blood Grouping Serum for Anti-D, Anti-C, Anti- CDEorCdE
E, Anti-c, or Anti-e, whichever is applicable,· in agglutinating Anti-e cdEe, Al cDE, B cDE, 0 cDE, and CcDE or
as a minimum red blood cells from the donors indicated in CDEorCdE
Table 2 (which may be from Group A, B, AB, or 0).
Each serum for slide or rapid tube test complies with the tests All fresh or frozen red blood cell suspensions used for these
for avidity with the cells as indicated under tests for potency tests are prepared under specified conditions and meet
above. specified criteria.

Table - 2 Monoclonal Rh (D) antibodies. Monoclonal antibodies are


derived from hybridoma cell lines produced by fusing mouse
Serum Phenotype of Cells antibody produced by B lymphocyte with mouse myeloma
Anti-D cDe cells or are derived from a human B cell line through Epstein-
Barr Virus (EBV) transformation. Each hybridoma cell line
Anti-C Ccde produces homogenous antibodies ofonly one immunoglobulin
Anti-E cdEe class, which are identical in their chemical structure and
immunological activity.
Anti-CD cDe,Ccde
The type of monoclonal anti-D reagents are:
Anti-DE cDe,cdEe
1. IgM anti-D monoclonal reagent,
Anti-CDE cdEe, cDe, Ccde
2. Blend ofIgM and IgG monoclonal antibodies reagent,
Anti-c CcDEe
3. Blend of monoclonal IgM and polyclonal (human)
Anti-e cdEe
IgGanti-D.
Each serum complies with the tests for specificity by the most IgM anti-D monoclonal antibodies are highly specific and
sensitive method recommended by the manufacturer, in which saline reacting equally well at room temperature and at 37°.
not less than 4 positive and 4 negative phenotypes are included They are good for slide test or immediate spin tube tests as
(see Table 3), and confirms the absence of contaminating well as routine Rh(D) typing in tube. IgM anti D are unreliable
antibodies reactive with Mg, WI:" antigens as well as other for detection ofweak D (Du) by AHG test. Blend ofIgM and

2561

www.webofpharma.com
CONCENTRATED HUMAN RED BLOOD CORPUSCLES IP 2010

IgG (monoclonal) anti-D or blend ofIgM (monoclonal) and solution equivalent to not less than 40.0 per cent of the total
polyclonal (human) IgG anti-D can be used for testing weak D volume of Whole Human Blood. A portion of the plasma,
(Du) antigen by AHG test. Mostly blended IgM and IgG sufficient to ensure optimal cell preservation, shall be left. All
(monoclonal) anti- Rh(D) or blended monoclonal IgM and surfaces that come in contact with the red cells and plasma
polyclonal (human) IgG anti Rh (D) antibodies are used now shall be sterile 'and pyrogen-free and the entire processing of
in routine. the blood shall be conducted in a sterile system or in a closed
Expiration date. The expiration date for liquid serums is not system by use of satellite bags. The final containers used for
later than 1 year and for dried serums not later than 5 years the Concentrated Human Red Blood Corpuscles shall be the
after date of issue from manufacturer's cold storage (5°, I original blood containers unless the method of processing
year; or 0°, 2 years), provided that the expiration date for dried requires a different container. Immediately after processing,
serums is not later than 1 year after constitution. the containers are stored at a temperature between 2° and 8°
and not opened until immediately before transfusion. Human
Storage. Store at a temperature between 2° and 8°.
Red Blood Corpuscles may be stored for a period not longer
Labelling. The label each to state that the source material was than that for which the Whole Human Blood from which it is
not reactive for hepatitis B surface antigen, but that no known prepared. However, if the hermetic seal is broken during
test method offers assurance that products derived from human processing, the product must be used within 24 hours. From
blood will not transmit hepatitis. Label each to state that it is the tube of the blood bag sample is taken for compatibility
for in vitro diagnostic use. and infections markers testing.
Concentrated Human Red Blood Corpuscles should be
administered only with suitable equipment meant for the
Concentrated Human Red Blood transfusion of blood and blood components.
Corpuscles Concentrated Human Red Blood Corpuscles contains not less'
than 15.5 per cent w/v of haemoglobin.
Concentrate ofHuman Red Blood Cells; Packed Red Cells
Description. A dark red fluid when prepared; after standing,
Concentrated Red Blood Cells (RBCs) are units ofwhole blood the red corpuscles may form a sediment, leaving a small
with most of the plasma removed. Additive red cell supernatant layer of yellowish plasma.
preservative systems consist of a primary collection bag
containing an anticoagulant preservative with at least two Tests
satellite bags integrally attached, one is empty and one
Sterility (2.2.11 ). Complies with the tests for sterility,
contains an additive solution (AS).
determined by Method B.
Each 100 ml citrate phosphate dextrose (CPD) contains Assay. Determine the haemoglobin content by photometric
Citric acid (monohydrate) 0.327 g haemoglobinometly (2.8.12).
Sodium citrate (dihydrate) 2.63 g Storage. Store in containers which are made ofcolourless and
transparent glass, or of a suitable plastic material, are sterile
Sodium acid phosphate (dihydrate) 0.251 g and sealed so as to exclude micro-organisms. Store at a
Dextrose (monohydate) 2.55 g temperature between 2° and 8°.
Water for injection 100 ml Labelling. The label states (1) the reference number of the
Whole Human Blood from which the preparation was made;
AS contains sodium chloride, dextrose, adenine and other (2) the ABO and Rh groups of the Whole Human Blood; (3)
substances that support red cell survival and function up to the date ofcollection ofthe Whole Human Blood from which
42 days. The volume of AS in 350 ml is 49 ml and in 450 ml is the preparation was made; (4) the storage conditions; (5) the
63 m!. AS is added to the red cells remaining in the primary bag date after which the preparation is not suitable for transfusion;
after most ofthe plasma has been removed. This allows blood (6) that the preparation should not be used if there is any
centres to use or recover a maximum amount of plasma, yet visible evidence ofhaemolysis or other deterioration.
still prepare a red cell component with a final haematocrit
between 55.0 per cent and 65.0 per cent, a level that facilitates
excellentflowratesand allows_easy administration. .__ Cryoprecipitated,Antihaemophilic
Factor
It may be prepared by centrifugation or undisturbed Cryoprecipitated Antihaemophilic Factor is a sterile, frozen
sedimentation for the separation ofplasma and anticoagulant concentrate of human antihaemophilic factor prepared from

2562

www.webofpharma.com
IP 2010 DRIED HUMAN ANTIHAEMOPHILIC FRACTION

the Factor VIII-rich cryoprotein fraction of human venous human donors who are, as far as can be ascertained after
plasma obtained from suitable whole-blood donors from a clinical examination, laboratory tests on their blood and
single unit of plasma derived from whole blood or by consideration of their medical history, free from detectable
plasmapheresis, collected and processed in a closed system. agents of infection transmissible by blood transfusion. The
It contains no preservative. It complies with the test for examinations and tests to be carried out are decided by the
potency by comparison with the Standard Antihaemophilic National Regulatory Authority. In particular, the blood must
Factor (Factor VIII) or with a working reference that has been be tested with negative results for (a) evidence of syphilitic
calibrated with it, in having an average potency of not less infection; (b) hepatitis B surface antigen and (c) HIV antibodies
than 80 Antihaemophilic Factor Units per container, made at by suitably sensitive methods. The haemoglobin value of the
intervals of not more than 1 month during the dating period. donor's blood is not less than 12.5 per cent w/v.
Expiration date. The expiration date is not later than I year The blood is withdrawn aseptically through a closed system
from the date of collection of source material. of sterile tubing into a sterile container in which a suitable
Storage. Store in hennetic containers at a temperature of - 18° anticoagulant solution has been placed before sterilisation.
or lower. During the withdrawal there is.no interruption in the flow from
the donor, and the container is gently agitated. Immediately
Labelling. Label it to indicate (1) the ABO blood group after the withdrawal is completed, the blood is cooled t04°; if
designation and the identification number of the donor from the plasma is to be stored frozen it is separated from the cellular
whom the source material was obtained; (2) with the type and components by centrifugation and frozen to -30 0 or below,
result of a serologic test for syphilis, or to indicate that it was preferably within 12 hours ofcollection; ifthe plasma is not to
non-reactive in such test; with the type and result of a test for be frozen it is separated from the cellular components by
hepatitis B surface antigen, or to indicate that it was non- centrifugation as so.on as possible and not later than 18 hours
reactive in such test; with a warning not to use it if there is after collection, and fractionation begun without delay.
evidence of breakage or thawing; with instructions to thaw it
before use to a temperature between 20° and 37°, after which Dried Human Antihaemophilic Fraction may be prepared :Ii-om
it is to be stored at room temperature and used as soon as human plasma so obtained by precipitation under controlled
possible but within 6 hours after thawing; (3) to state that it is conditions ofpH, ionic strength and temperature with organic
to be used within 4 hours after the container is entered; (4) to solvents, or by freezing and thawing. The precipitate may be
state that it is for intravenous administration; (5) that a filter is washed by extraction with suitable solvents, dissolved in a
to be used in the administration equipment. solution ofsodium citrate adjusted to a pH of6.8 to 7.2, which
may also contain sodium chloride. The solution is sterilised
by filtration through a membrane filter, distributed in sterile
containers and dried from the frozen state. The air is removed
or replaced by oxygen-free nitrogen and the containers are
Dried Human Antihaemophilic sealed so as to exclude micro-organisms. No antimicrobial
Fraction preservative is added but an antiviral agent may be added
provided that it can be demonstrated to have no deleterious
Dried Factor VIII Fraction; Freeze-dried Human Coagulation effect on the final product in the amount present and to cause
FactorVII1 no adverse reaction in man. Heparin may be used.
Dried Human Antihaemophilic Fraction is a preparation of For the following tests, where it is directed that a solution is to
antihaemophilic factor which is obtained from human plasma. be used, dissolve the contents ofa sealed container in a volume
It is rich in clotting factor VIII. of the appropriate solvent equal to the volume of water for
When the contents of a sealed container of Dried Human injection stated on the label.
Antihaemophilic Fraction are dissolved in a volume ofwater Description. A white or pale yellow powder or friable solid.
equal to the volume of waterfor injection stated on the label,
the resulting solution contains not less than 3.0 Units per ml, Identification
not less than 0.1 Unit per mg ofprotein, not more than 80.0 per
cent ofwhich is fibrinogen, and not more than 200 millimoles A. Precipitation tests with a suitable range of species specific
of sodium ions per litre. antisera give positive results for the presence of plasma
proteins of human origin and negative results with antisera
Production specific to plasma proteins of the other species.
The plasma to be used for preparing Dried Human B. A fi:eshly prepared solution in water causes a reduction in
Antihaemophilic Fraction is 'obtained from blood of healthy clotting time when treated as directed under Assay.

2563

www.webofpharma.com
DRIED HUMAN ANTIHAEMOPHILIC FRACTION IP 2010

Tests to· produce the same effect under the conditions of the
following method ofassay.
pH (2.4.24).6.8 to 7.4, determined on the reconstituted solution.
Loss on drying (2.4.19). Not morethan 2.0 per cent, determined Standard preparation
by drying 0.5 g over phosphorus pentoxide at a pressure not
The Standard preparation is the 4th International Standard for
exceeding 3 kPa for 24 hours. Blood coagulation factor VIII:C,concentrate, human,
Haemagglutinins, anti-A and anti-B. Dissolve in water. Dilute established in 1989, consisting of an intermediate purity
the solution with saline solution to produce a solution concentrate of human blood clotting factor VIII (supplied in
containing 3 Units per ml. Carry out the test for ampoules containing 6.3 Units of clotting factor VIII), or
haemagglutinins anti-A and anti-B using a suitable indirect another suitable preparation the potency of which has been
method such as that described below. determined in relation to the International Standard
Prepare in duplicate serial dilutions of the preparation under The Unit is the specific antihaemophilic factor contained in
examination in saline solution. To each dilution of one series such an amount of the Standard preparation as the Ministry
add an equal volume ofa 5.0 per cent v/v suspension ofgroup ofHealth & Family Welfare, Govt. ofIndia may from time to
AI red blood cells previously washed three times with saline time indicate as the quantity exactly equivalent to the Unit
solution. To each dilution of the other series add an equal accepted for international use.
volume of a 5.0 per cent v/v suspension of group B red blood
cells previously washed three times with saline solution. Special reagents
Incubate the suspensions at 37° for 30 minutes and then wash Normal serum reagent. Collect normal human blood in a dry,
the cells three times with saline solution. Leave the cells in sterile, glass bottle, shake continuously until coagulation is
contact with a polyvalent anti-human globulin reagent for 30 complete, incubate at 37° for 3 hours, maintain at 4° overnight,
minutes. Without centrifuging, examine each suspension for remove the serum, store at -20°, dry from the frozen state and
agglutination under a microscope. The 1 in 64 dilutions do not keep in a vacuum desiccator over phosphorus pentoxide.
show agglutination. Dissolve a quantity of the dried serum calculated to have
Hepatitis B surface antigen. Dissolve in water. Examine the been obtained from 1 ml of the serum in sufficient imidazole
solution by a suitably sensitive method such as bufferpH 7.4 to produce 10 ml and allow to stand at 4° for 16
radioimmunoassay. Hepatitis B surface antigen is not detected. to 24 hours.
Abnormal toxicity (2.2.1). When dissolved in water for Phospholipid. Wash a quantity of normal human or bovine
injection, complies with the test for abnormal toxicity, Method brain freed from meninges and blood vessels and macerate in
B, injecting into each mouse a volume containing 1.5 Units a suitable blender. Weigh 1,000 to 1,300 g ofthe macerate and
and into each guinea-pig a volume containing 15 Units. measure its volume (V). Extract with three quantities, each of
4 V ml, of acetone, filter by suction and dry the precipitate at
Pyrogens (2.2.8). When dissolved in water for injection,
37° for 18 hours. Extract the dried precipitate with two
complies with the test for pyrogens, using a volume containing
quantities, each of2 V ml, ofa mixture oftwo volumes of light
10 Units per kg of the rabbit's weight in rabbits that have not
petroleum (boiling range 30° to 40°) and 3 volumes of light
previously received blood products.
petroleum (boiling range 40° to 60°), filtering each extract
Sterility (2.2.11). Complies with the tests for sterility. through a filter paper previously washed with the light
petroleum mixture. Combine the extracts' and evaporate to
Assay
dryness at 45° at a pressure not exceeding 0.7 kPa. Dissolve
For potency - Carry out the biological assay of human the residue in 0.2 V ml of ether and allow to stand at 4° until a
antihaemophilic fraction described below. The estimated deposit forms. Centrifuge and evaporate the clear supernatant
potency is not less than 80.0 per cent and not more than 125.0 liquid under reduced pressure until the volume is about 100 ml
per cent of the stated potency. The fiducial limits of error are per kg of the original macerate. Allow to stand at 4° until a
not less than 64.0 per cent and not more than 156.0 per cent of precipitate forms (12 to 24 hours) and centrifuge. To the clear
the stated potency. supernatant liquid add 5 times its volume of acetone, centrifuge,
discard the supernatant liquid, dry the precipitate and store
Biological assay protected from light in a vacuum desiccator.
. . . I'ltosPIt()lipilil"eagellt.. ~llspen4 0,125 g ()fphospho.Pp.(cijl1
Thep()tel1cy.()fl111111al1al1ti1Jfl~p:1()Pl1ili()fracti()l1js4~!~l1J1iJ.1~(1 S.
by comparing the amount necessary to reduce the clotting ml of water, shake and stir until a uniform suspension is
time ofa test mixture containing substances that cause clotting obtained. Prepare a dilution with saline solution that will give
ofblood with the amount ofthe Standard preparation necessary minimum clotting time consistent with the largest clotting time

2564

www.webofpharma.com
IP 2010 DRIED HUMAN ANTIHAEMOPHILIC FRACTION

differences between consecutive dilutions of the Standard should have a clotting time, when tested by the assay method
preparation and the preparation under examination. The . given under clottingfactor V solution, of 70 to 100 seconds;
concentration usually lies between 50 and 250 Ilg per ml. The ifthe clotting time is less than 70 seconds, incubate the plasma
diluted suspension may be kept at -20° for 6 weeks. for a further 12 to 24 hours.
Clotting factor V solution. Prepare from fresh oxalated bovine Storage. Store in small amounts, at -20° or below.
plasma by fractionation at 4° with a saturated solution of
ammonium sulphate prepared at 4°. Use the fraction Suggested method
precipitating between 38 per cent and 50 per cent saturation Dissolve the contents of the sealed container of the substance
(which contains clotting factor V not significantly contaminated under examination in the volume of the liquid stated on the
with clotting factor VIII), dialysed to remove ammonium label and use immediately. Reconstitute the entire contents of
sulphate and diluted with saline solution to give a solution one ampoule of the Standard preparation as stated on the
containing between 10 per cent and 20 per cent ofthe amount label and use immediately.
of clotting factor V present in fresh normal human plasma.
To the reconstituted Standard preparation and the preparation
Detelmine the clotting factor V content of the solution as under examination, add sufficient imidazole blif.fer pH 7.4 to
follows. Prepare two dilutions in imidazole blif.fer pH 7.4 to produce solutions containing between 0.5 and 2 Units per ml;
contain 1 volume of the solution under examination in 10 these solutions are stable for 15 minutes at200. Using a mixture
volumes and 20 volumes respectively. Test each dilution as of 1 volume of a 3.8 per cent w/v solution of sodium citrate
follows. Mix 0.1 ml each of substrate plasma deficient in and 5 volumes of saline solution as the diluent, make from the
clotting factor V, the dilution under test, thrombokinase solutions three successive 2-fold dilutions in the range 1 in16
extract and O.025M calcium chloride. Record as the clotting to 1 in 256 so that all the clotting times are between 17 and 35
time the interval between the addition ofthe calcium chloride seconds; the dilutions must be accurately made and used
solution and the first indication of fibrin formation, which immediately.
may be observed visually or by mechanical means.
Introduce into each of six glass incubation tubes (75 rom to
Similarly determine the clotting times, in duplicate, for four 100 min) 0.1 ml each of clottingfactor V solution,phospholipid
dilutions ofpooled normal human plasma in imidazole blif.fer reagent and normal serum reagent. To the first tube add 0.1
pH 7.4 containing 1 volume in 10 volumes (equivalent to 100 ml of the highest dilution of the Standard preparation, place
per cent of clotting factor V), in 50 volumes (20 per cent), in the tube in a water-bath at 37°, add 0.1 ml ofO.05M calcium
100 volumes (10 per cent), and in 1,000 volumes (1 per cent), chloride and start a. stop-watch. During the next minute add
respectively. 0.1 ml of the second highest dilution of the standard to a
second tube, place it in the water-bath, and add 0.1 ml of
To calculate the result, plot the mean of the clotting times for
O.05M calcium chloride at exactly 1 minute by the stop-watch.
each dilution of human plasma on double cycle log/log paper
Repeat the procedure with the lowest dilution of the standard
against the equivalent percentage of clotting factor V and
and the highest to lowest dilutions of the preparation under
read the percentage of clotting factor V for the two dilutions
examination so that the calcium chloride solution is added at
of clottingfactor V solution by interpolation from the curve.
2,3,4 and 5 minutes by the stop-watch, respectively.
The mean of the two results is taken as the percentage of
clotting factor V in the solution. Place in a water-bath at 37°, twelve glass tubes each containing
0.2 ml of O.025M calcium chloride and a further tube
Substrate plasma. Separate the plasma from 9 volumes of
containing about 3 ml of substrate plasma. At 14 minutes, 40
human or bovine blood collected in 1 volume ofa 3.8 per cent
seconds by the stop-watch, transfer 0.1 ml ofthe mixture from
w/v solution of sodium citrate or from 3.5 volumes ofhuman
the first incubation tube to one ofthe tubes containing 0.2 ml
or bovine blood collected in 1 volume ofa solution containing
of O.025M calcium chloride solution and mix. At 15 minutes
2.0 per cent w/v of sodium acid citrate and 2.5 per cent w/v of
add 0.2 ml ofthe warmed substrate plasma and, using a second
dextrose. In the former case, prepare the substrate plasma on
stop-watch, record as the clotting time the interval between
the day of collection of the blood. In the latter case, the
the addition ofthe substrate plasma and the first indication of
substrate plasma may be prepared up to 2 days after collection
fibrin formation, which may be observed visually or by
ofthe blood. Store at-20°.
mechanical means. Repeat the procedure with the other
Substrate plasma deficient in clotting factor V. Preferably incubation tubes at I-minute intervals and carry out a second
use congenitally deficient plasma or, alternatively, prepare as series of determinations at 21 to 26 minutes. The period of
follows. Separate the plasma from human blood collected in incubation should, ifnecessary, be adjusted so that the clotting
one-tenth its volume ofa 1.34 per cent w/v solution of sodium times recorded in the corresponding tests in the two series of
oxalate and incubate at 37° for 24 to 36 hours. This plasma determinations do not differ by more than 5.0 per cent, showing

2565

www.webofpharma.com
DRIED HUMAN ANTlHAEMOPHILIC FRACTION IP 2010

that a stable plateau of prothrombin activator formation has the solution is not complete or if a gel forms on constitution,
been reached. the preparation should not be used; (11) that the solution
should be used as soon as possible and in any case within 3
Carry out a blank determination using in place of the
hours of constitution and any unused solution' should be
preparation under examination, an equal quantity ofa mixture
discarded; (12) that the solution sho,!ldbe administered only
of 1 volume of a 3.8 per cent w/v solution of sodium citrate
with equipment that includes a filter; (13) the storage
and 5 volumes of saline solution. The result of the assay is
conditions.
not valid unless the dotting time in the blank determination is
more than 40 seconds. Calculate the result of the assay by
standard statistical methods.
For totalprotein. Dilute 1.0 ml to 10.0 ml with saline solution. Fibrin Sealant Kit
Determine the assay described under Human Plasma using Fibrin Sealant Kit is essentially composed oftwo components,
5.0 ml ofthe dilution and beginning atthe words "add 0.2 ml of namely fibrinogen concentrate (component 1), a protein
a 7.5 per cent w/v solution....". fraction containing human fibrinogen and a preparation
Forfibrinogen. Dilute 1.0 ml ofthe solution to 10.0 ml with a containing human thrombin (component 2). A fibrin clot is
phosphate-saline buffer pH 6.5 and ionic strength 0.15. Clot rapidly formed when the two thawed or reconstituted
5.0 ml ofthe dilution with the minimum amount of thrombin, components are mixed. Other ingredients (for example, human
collect the clot and add three drops ofa 30 per cent w/v solution coagulation factor XlII, a fibrinolysis inhibitor or calcium ions)
of copper sulphate and 1 ml of nitrogen-fi-ee sulphuric acid and stabilisers (for example, Human albumin solution may be
and boil gently for 10 minutes; cool, add 1 g of anhydrous added). No antimicrobial preservative is added.
sodium sulphate and 10 mg of selenium, boil gently for 1 hour Human constituents are obtained from plasma that complies
and cool. Transfer to an ammonia distillation apparatus, add with the requirements ofthe monograph on Human Plasma for
6 ml of a saturated solution of sodium hydroxide and pass Fractionation. No antibiotic is added to the plasma used.
steam through the flask; distil for seven minutes, collecting
the distillate in a mixture of 5 ml of a saturated solution of When thawed or reconstituted as stated on the label,
boric acid, 5 ml of water; and 1 drop of a saturated solution of component 1 contains not less than 4.0 per cent of clottable
methyl red in alcohol containing 0.1 per cent of methylene protein; the thrombin activity of component 2 varies over a
blue, and titrate with 0.02 M hydrochloric acid wide range (approximately 4-1 000 IV per ml).

1 ml ofO.02Mhydrochloric acid is equivalent to 0.00175 g of Production


fibrinogen.
The method of preparation includes a step or steps that have
For sodium ions. To 10.0 ml of the solution add sufficient
been shown to remove or to inactivate known agents of
water to produce 100 ml, dilute 10.0 ml to 500 ml with water infection; if substances are used for inactivation of viruses
and detennine the content of sodium ions by Method B tior
during production, the subsequent purification procedure
flame photomeny (2.4.4), measuring at about 589 urn and using must be validated to demonstrate that the concentration of
sodium solution FP suitably diluted with water as the standard these substances is reduced to a suitable level and any
solution.
residues are such as not to compromise the safety of the
Storage. Store protected from light, in an atmosphere of preparation for patients.
nitrogen at a temperature below 8°. The containers are sterile Constituents or mixtures of constituents are passed through
and sealed so as to exclude micro-organisms.
a bacteria-retentive filter and distributed aseptically into sterile
Labelling. The label states (1) the ABO blood group containers. Containers of freeze-dried constituents are closed
designation of the source ofblood; (2) the number ofUnits in under vacuum or filled with oxygen-free nitrogen or other
the container; (3) that 1 Unit is approximately equivalent to suitable inert gas before being closed. In either case, they are
the antihaemophilic activity of 1 ml ofaverage normal plasma; closed so as to exclude micro-organisms.
(4) the concentration of protein in g per litre and of sodium
Ifthe human coagulation factor XIII content in component 1
ions in millimoles per litre of the solution constituted as
is greater than 10 Units per ml, the assay ofcoagulation factor
directed; (5) the maximum fibrinogen content; (6) where XIII is carried out.
.applicahle,Jhe.numhecoLUnits_ofheparin.inJhe_c.ontaineG.(1) . ._ ... .__. . ._
t1J.e .l1a111~. al1(:l.a11101l.n1.()fl:ll1y()t1J.era<i<:l~<:lsll1:Jstal1<:~_c0l1tajJ:l~cl Qt)~(:r!pfu>!1-"Ere~:Z;\'l.-4ri.e:<igQ!1sti1:ll.e:I!!~!lrejl)'gr2sc()pic,,,,1J.itel
in it; (8) the volume of Water for Injections necessary to or pale yellow powders or friable solids. Frozen constituents
constitute the solution; (9) the instructions for constitution are colourless or pale yellow, opaque solids. Liquid
and that reconstitution may take upto 30 minutes; (10) that if constituents are colourless or pale yellow.

2566

www.webofpharma.com
IP 2010 FIBRIN SEALANT KIT

For the freeze-dried or frozen constituents, reconstitute or plasma or another suitable diluent. Add to each dilution
thaw as stated on the label immediately before carrying out suitable amounts of the following reagents:
the Identijication and the Tests, except those for solubility
a. activator reagent, containing bovine or human thrombin,
and water.
a suitable buffel; calcium chloride and a suitable inhibitor
Component 1 (Fibrinogen Concentrate) such as Gly-Pro-Arg-Pro-Ala-NH2 which inhibits clotting of
the sample but does not prevent coagulation factor XIII
Identification activation by thrombin,
b. detection reagent, containing a suitable factor XIIIa-specijic
A. Complies with the limits of the assay of fibrinogen.
peptide substrate, such as Leu-Gly-Pro-Gly-Glu-Ser-Lys-Val-
B. Complies with the limits ofthe assay offactor XIII Ile-Gly-NH2 andglycine ethyl ester as 2nd substrate in a suitable
(where applicable). buffer solution,

Tests c. NADH reagent, containing glutamate dehydrogenase, a-


ketoglutarate and NADH in a suitable buffer solution.
pH (2.4.24). 6.5 to 8.0.
After mixing, the absorbance changes (M per min) are
Stability of solution. No gel formation appears at room
measured at a wavelength of 340 nm, after the linear phase of
temperature during 120 minutes following thawing or
the reaction is reached.
reconstitution.
I Unit of factor XIII is equal to the activity of 1 mlofhuman
Water. Determine by semi-microdetermination (2.3.43), loss
normal plasma.
on drying (2.4.19) or near infrared spectrophotometry (2.4.6),
the water content is within the limits approved by the Calculate the activity of the test preparation by the usual
competent authority. statistical methods. The confidence limits (P = 0.95) are not
less than 80.0 per cent and not more than 125.0 per cent ofthe
Sterility (2.2.11). Complies with the test for sterility. estimated activity.
Assay Component 2 (Thrombin Preparation)
Fibrinogen (Clottable Protein)
Identification
The estimated content in mg of clottable protein is not less
than 70.0 per cent and not more than 130.0 per cent of the Complies with the limits ofthe assay ofthrombin.
content stated on the label.
Tests
Mix 0.2 ml of the reconstituted preparation with 2 ml of a
suitable buffer solution pH 6.6 to 7.4 containing sufficient pH (2.4.24). 5.0 to 8.0.
human thrombin (approximately 3 IU per ml) and calcium Water. Determine by semi-microdetermination of water
(0.05mol per I). Maintain at 37° for 20 minutes, separate the (2.3.43), loss on. drying (2.4.19) or near infrared
precipitate by centrifugation (5000 g, 20 minutes), wash spectrophotometry (2.4.6), the water content is within the
thoroughly with a 0.9 per cent solution of sodium chloride limits approved by the competent authority.
and determine the protein as nitrogen by sulphuric acid
digestion (2.3.30)..Calculate the protein content by multiplying Sterility (2.2.11). Complies with the test for sterility.
the result by 6.0. If for a particular preparation this method
Assay
cannot be applied, use another validated method for
determination of fibrinogen. Thrombin
The estimated activity is not less than 80.0 per cent and not
Factor XIII
more than 125.0 per cent of the activity stated on the label.
Where the label indicates that the human coagulation factor
If necessary, dilute the reconstituted preparation under
XIII activity is greater than 10 Units per ml, the estimated
examination to approximately 2-20 IV ofthrombin per ml using
activity is not less than 80.0 per cent and not more than 120.0
as diluent a suitable buffer pH 7.3 to 7.5, such as imidazole
per cent of the activity stated on the label.
buffer solution pH 7.3 containing 1.0 per cent of human
Make at least 3 suitable dilutions of thawed or reconstituted albumin or bovine albumin. To a suitable volume of the
component 1 and of human normal plasma (reference dilution, add a suitable volume offibrinogen solution (0.1 per
preparation) using as diluent coagulation factor XIII deficient cent ofclottable protein) warmed to 37° and start measurement

2567

www.webofpharma.com
. HUMAN ALBUMIN IF 2010

of the clotting time immediately. Repeat the procedure with then heated to and maintained at 60° ± 0;5° for 10 hours so as
each of at least 3 dilutions, in the range stated above, of a to prevent the transmission ofagents ofinfection transmissible
reference preparation ofthrombin, calibrated in International by transfusion of blood or blood derivatives. Finally, the
Units. Calculate the activity of the test preparation by the containers are stored for not less than 14 days at 30° to 32° or
usual s.tatistical methods. The confidence limits (P = 0.95) are for not less than 4 weeks at 20° and examined visually. Those
not less than 80.0 per cent and not more than 125.0 per cent of showing abnormalities such as abnormal colour, turbidity,
the estimated activity. microbial contamination, or presence ofatypical particles must
be discarded.
Storage. Store protected from light.
Labelling. The label states (I) the amount of fibrinogen (mg Albumin Solution should be tested in accordance with the
of clottable protein), thrombin (International Units) per requirements decided by the National Regulatory Authority;
container, and coagulation factor XIII, ifthis is greater than 10 in particular, tests for the absence ofhepatitis B surface antigen
Units per ml; (2) where applicable, the name and volume of and HIV antibodies are carried out by suitably sensitive
solvent to be used to reconstitute the components. methods.
Human Albumin contains not less than 95.0 per cent and not
more than 105.0 per cent of the stated amount of protein. It
contains not less than 95.0 per cent and not more than 105.0
Human Albumin per cent ofthe contents ofNa and K stated on the label which
are, in any case, not more than 160 millimoles ofNa per litre
Human Normal Albumin; Human Albumin Solution
and 2 millimoles ofK per litre.
Human Albumin is a sterile non-pyrogenic solution of the
Description. A clear, slightly viscous liquid, ranging in colour
albumin component obtained from pooled human blood or
from almost colourless to greenish-yellow or amber depending
from normal placentae frozen immediately after collection. It is
on protein concentration and the method offractionation used.
obtained by fractionating source material such as blood,
plasma, serum or placentae from healthy human donors and
tested individually for the absence of hepatitis B surface Identification
antigen, HCV antibodies and HIV antibodies and complies A. It contains plasma proteins of human origin only as
with other tests and requirements prescribed by the appropriate determined by precipitation tests with specific antisera.
national control authority. Source material obtained from
donors who do not meet all the requirements stated may be B. Determine the cellulose acetate by electrophoresis (2.4.12),
used provided that it has been demonstrated to the national using barbitone buffer pH 8.6, ionic strength 0.1 and human
control authority that the process offractionation will remove albumin for electrophoresis RS; 96.0 per cent of the protein
any known agent capable of adversely affecting the health of has the mobility of human albumin.
subjects treated with the preparation. It may be prepared from
pooled source materials by precipitation with organic solvents Tests
under controlled conditions of pH, ionic strength and
Acidity or alkalinity (2.4.24). Dilute with sufficient saline
temperature or by chromatography or by any other method
solution to produce a solution containing 1.0 per cent w/v of
which does not affect the integrity of the product and has
protein; pH of the resulting solution, 6.7 to 7.3.
been shown to yield consistently a product containing not
less than 95.0 per cent w/v of the total protein as albumin Alkaline phosphatase. Not more than 0.1 Unit per g ofprotein,
which is safe for intravenous injection. Residual organic determined by the following method. Transfer a mixture of
solvent, if present, is removed by freeze-drying or other 0.5 ml of the substance under examination and 0.5 ml of
suitable treatment. The product is dissolved in sufficient water diethanolamine buffer pH 10.0 to a spectrophotometer cell
to obtain a suitable concentration, and a suitable stabilising maintained at a temperature of37° ± 0.2 0 and add 0.1 mlof
agent is added to stabilise it to heat. It is prepared as a solution nitrophenyl phosphate solution. Using a continuously
containing 15.0 to 25.0 per cent w/v of total protein or as an recording spectrophotometer record the absorbance of the
isotonic solution containing 4.0 to 5.0 per cent w/v of total solution at about 405 nm (2.4.7), over a period of at least 30
protein. No antimicrobial agent is added at any stage during seconds from the time of addition of the nitrophenyl
preparation.and.allprocessing stepsareconducted.in.. a.rnanner .phosphate solutian .__CakulaJe_Jb.e_alkaJifle_pb_Q.sp.h.atas.s<.
to.miI:liIllist;:ri:;lc.ofcolltamiI:latiOil frOm t;:itl1er micI:O:QrgllIlisIl1s .11ctiyity .at37° iIllJIlitsper g ofPI:ot<eillfrOIl1!I1t) t))(pr(;:§siQI1
or other deleterious matter. The solution is sterilised by 118.3xJP, where x is the rate of increase of absorbance per
filtration and distributed aseptically into containers which are minute and P is the content of total protein in g per litre, as
then sealed so as to exclude micro-organisms. The solution is determined in the Assay:

2568

www.webofpharma.com
LP 2010 HUMAN ALBUMIN

Haem content. Dilute with sufficient saline solution to produce Reference solution. Dilute human albuminfor electrophoresis
a solution containing 1.0 per cent w/v of protein; absorbance RS with saline solution to obtain a solution containing 2.0 per
of the resulting solution at about 403 nm, not more than 0.15 cent w/v of total protein.
(2.4.7).
Not more than 5.0 per cent of total protein is contained in
Denatured protein. Equilibrate a column (60 to 75 cm x 2.5 to bands other than the principal band in the strip obtained with
3.0 cm) of a gel of a cross-linked dextran suitable for test solution. The test is not valid if the proportion of the
fractionation ofproteins in the range ofmolecular weight from protein in the principal band is not within the limits stated in
5,000 to 3,50,000 with a lower molecular weight for complete the leaflet supplied with human albumin for electrophoresis
exclusion of globulin proteins of molecular weight between RS.
4,00,000 and 5,00,000 (Such as Sephadex G 150) at 20° to 25° Stability. The contents ofthe final containerremain unchanged,
with a saline-phosphate solution prepared by mixing 2 as determined by visual inspection, after heating at 57° for 50
volumes of saline solution and I volume of mixedphosphate hours, when compared to its control consisting of a sample
bufferpH 7.0 with azide. Apply to the column 2.5 ml ofnormal from the same lot which has not undergone this heating.
human serum, previously clarified by centrifuging, and elute
with the saline-phosphate solution ata rate of20 ml per hour. Pyrogens (2.2.8). Complies with the test for pyrogens, using
Prepare a chromatogram by recording the absorbance (2.4.7), 3 ml per kg of the rabbit's weight, inespective of the protein
of the eluate at about 280 nm in relation to its volume. The content, in rabbits that have not previously received blood
chromatogram exhibits three well-defined peaks. Determine products.
the volume, V, of the eluate from the entry of the sample into Sterility (2.2.11). Complies with the tests for sterility.
the column to the apex of the first peak. ,
Abnormal toxicity (2.2.1). Complies with the test for abnormal
Dilute the substance under examination with the saline- toxicity, using Method Band 0.5 ml of the solution for each
phosphate solution to contain about 5.0 per cent w Iv ofprotein, mouse and 5 ml for each guinea-pig irrespective ofthe protein
apply 2.5 ml to the column and elute under the above content.
conditions, collecting the eluate in 5-ml portions. Three peaks
may appear in similar positions to those in the chromatogram Assay
obtained from the normal human serum but the relative peak For protein. Dilute to about 0.75 per cent w/v of total protein
heights may be different. To the fraction eluted between with saline solution. Take 2 ml of this solution in a round-
volume 0.85 V and 1.15 V, add for each 10 ml, 0.4 rnl ofa 7.5 per bottomed centrifuge tube, add 2 ml of a 7.5 per cent w/v
cent w/v solution of sodium molybdate and 0.4 ml ofa mixture solution of sodium molybdate and 2 ml of a mixture of 30
of 1 part of nitrogen-fi-ee sulphuric acid and 30 parts of water, volumes of water and I volume of nitrogen-free sulphuric
shake, centrifuge for 5 minutes and complete the Assay acid. Shake, centrifuge for 5 minutes, decant the supernatant
described under Human Plasma beginning at the words liquid and let the inverted tube stand on a filter paper to drain
"decant the supernatant...". The weight of protein in the the fluid. Carry out Method E for determination of nitrogen
fraction of the eluate is not more than 3.5 per cent of the (2.3.30), on the residue thus obtained and multiply the result
weight of protein in the volume of the substance under by 6.25 to obtain the protein content.
examination applied to the column.
For sodium. Dilute toO.Ol per cent w/v ofprotein with water
Heat the substance under examination for 50 hours at 56.5 0 to and determine by Method A for atomic absorption
57.5 and repeat the chromatographic separation and the
0
spectrophotometry (2.4.2), or by Method B for flame
detennination of the weight of protein in the fraction eluted photometry (2.4.4), measuring at about 589 urn and using
between 0.85 V and 1.15 V. When expressed as a percentage of sodiwn solution FP suitably diluted with water as the standard
the weight of protein in the volume of the substance under solution.
examination applied to the column, it exceeds the percentage
obtained before heating by not more than 1.5 per cent w/v.
For potassium. Dilute to 0.25 per cent w/v of protein with
water and determine by Method A for atomic absorption
Protein composition. Not less than 95.0 per cent w/v of the spectrophotometry (2.4.2), or by Method B for flame
total protein as albumin, when determined by the following photometry (2.4.4), measuring at about 767 nm and using
method. Carry out Method II for cellulose acetate potassium solution FP suitably diluted with water as the
electrophoresis (2.4.12), using one strip of cellulose for each standard solution.
solution.
Human Albumin intended for administration to patients
Test solution. Dilute the substance under examination with undergoing dialysis or to premature infants complies with
saline solution to contain 2.0 per cent w/v of total protein. the following additional test.

2569

www.webofpharma.com
HUMAN ALBUMIN IP 2010

Aluminium (2.3.8). Not more than 200 Ilg of Al per litre. The potency of the preparation, reconstituted as stated on
Determine by atomic absorption spectrophotometry (2.4.2), the label, is not less than 20 IV of factor IX per ml.
with a furnace as atomic generator and measuring at 309.3 nm
and using as standard solutions a suitable range of dilutions Production
in water of aluminium standard solution (10 ppm AI) further The method ofpreparation is designed to maintain functional
diluted, as necessary, with a solution containing 0.17 per cent integrity offactor IX, to minimise activation ofany coagulation
w/v of magnesium nitrate and 0.05 per cent w/v of octoxinol factor (to minimise potential thrombogenicity) and includes a
lOin a solution of nitric acid containing 1 per cent w/v of step or steps that have been shown to remove or to inactivate
nitric acid. Prepare suitable dilutions ofthe preparation under known agents of infection; if substances are used for
examination and human albumin for aluminium validation inactivation of viruses during production, the subsequent
RS with water. Dilute the solutions, as necessary, with the purification procedure must be validated to demonstrate that
magnesium nitrate-octoxinol 10-nitric acid solution used the concentration of these substances is reduced to a suitable
for dilution of the standard solution. The test is valid only if level and that any residues are such as not to compromise the
the aluminium content determined for human albumin for safety of the preparation for patients.
aluminium validation RS is within 20 per cent of the stated
The specific activity is not less than 50 IV offactor IX per mg
value.
of total protein, before the addition of any protein stabiliser.
NOTE - Wash all equipments with a solution containing The factor IX fraction is dissolved in a suitable liquid. Heparin,
20.0 per cent w/v of nitric acid before use and use plastic antithrombin and other auxiliary substances such as a
containers only to prepare all solutions. stabiliser may be included. No antimicrobial preservative is
Storage. Store protected from light, at a temperature between added. The solution is passed through a bacteria-retentive
2° and 25°. Human Albumin stored at 2° to 8° may be expected filter, distributed aseptically into the final containers and
to continue to meet the requirements of the monograph for 5 immediately frozen. It is subsequently freeze-dried and the
years from the date on which it was heated at 60° for 10 hours. containers are closed under vacuum or under an inert gas.
Human Albumin stored at a temperature not exceeding 25° Consistency ofthe method
may be expected to continue to meet the requirements of the
The consistency of the method of production is evaluated by
monograph for 3 years from the date on which it was heated at
suitable analytical procedures that are determined during
60° for 10 hours.
process development and which normally include (1) assay
Labelling. The label states (1) the volume in the container; (2) offactor IX; (2) determination ofactivated coagulation factors;
the total amount ofprotein in the container expressed in g per (3) determination of activities offactors II, VII and X which
litre or as percentage; (3) the concentration of sodium and shall be shown to be not more than 5.0 per cent ofthe activity
potassium ions expressed in millimoles per litre; (4) the names offaetor IX.
and concentrations of any stabilising agents and any other Description. A white or pale yellow, hygroscopic powder or
additives in the final solution; (5) the type of source material friable solid.
used to manufacture the product; (6) the words "Do not use if
turbid"; (7) that the contents must not be used more than 4
Reconstitute the preparation under examination as stated
hours after the container has been penetrated and any remnant
on the label, immediately before carrying out the
portion must be discarded; (8) the storage conditions; (9) the
Identification, Tests (except those for solubility and water)
date after which the solution is not intended to be used; (10)
and Assay.
either that the preparation is suitable for administration to Identification
patients undergoing dialysis and to premature infants or that
it is not intended for such purpose. It complies with the limits ofthe Assay.

Tests
pH (2.4.24). 6.5 to 7.5.
Human Coagulation Factor IX Osmolality (2.4.23). Minimum 240 mosmol per kg.
Human Coagulation Factor IX is a plasma protein fraction Total protein. If necessary, dilute an accurately measured
containing coagulation .factor. IX,preparedby_a.methodjhat :v:olume_ofthe_pr,eparation_undecexamination_w.ith_a_Q.2..p.eI
effectively seParates factor lXJrom other .. prothrombin , c:ellLw/v .SQ1lltiQIl ofs.o.dJz1l11 fh]oTic/e, tQ()lJtaiI1a,~Qlll,ti.QI1.
complex factors (factors II, VII and X). It is obtained from which may be expected to contain about 15 mg ofprotein in 2
human plasma that complies with the monograph on Human ml. To 2.0 ml ofthat solution, in a round-bottomed centrifuge
Plasma for Fractionation. tube, add 2 rnl ofa 7.5 per cent w/v solution ofsodium molybdate

2570

www.webofpharma.com
IP 2010 HUMAN COAGULATION FACTOR VII

and 2 ml of a mixture of 1 volume of nitrogen-:fi-ee sulphuric also contain small amounts of the activated fonn, the two-
acid and 30 volumes of water. Shake, centrifuge for 5 minutes chain derivative factor VIla. It may also contain coagulation
decant the supernatant liquid and allow the inverted tube to factors II, IX and X and protein C and protein seconds. It is
drain on filter paper. Determine the nitrogen in the residue by obtained from human plasma that complies with the
the method of sulphuric acid digestion (2.3.30) and calculate monograph on Human Plasma for Fractionation.
the amount of protein by multiplying the result by 6.25. The potency of the preparation, reconstituted as stated on
For some products, especially those without a protein the label, is not less than 15 ill of factor VII per ml.
stabiliseI' such as albumin, this methodmay not be applicable.
Another validated method for protein determination must Production
therefore be pe/jormed. The method ofpreparation is designed to minimise activation
Activated coagulation factors (2.8.4). Ifnecessary, dilute the of any coagulation factor (to minimise potential
preparation under examination to contain 20 IU of factor IX thrombogenicity) and includes a step or steps that have been
per ml. For each of the dilutions the coagulation time is not shown to remove or to inactivate known agents of infection;
less than 150 seconds. if substances are used for inactivation of viruses during
production, the subsequent purification procedure must be
Heparin. If heparin has been added during preparation,
validated to demonstrate that the concentration of these
determine the amount by the assay of heparin in coagulation
substances is reduced to a suitable level and that any residues
factor concentrates (2.8.10). The preparation under
are such as not to compromise the safety of the preparation
examination contains not more than the amount of heparin
for patients.
stated on the label and in any case not more than 0.5 ill of
heparin per International Unit of factor IX. The specific activity is not less than 2 IU offactor VII per mg
of total protein, before the addition of any protein stabiliser.
Water. Detennine by semi-micro determination of water
(2.3.43), loss on drying (2.4.19) or near infrared The factor VII fraction is dissolved in a suitable liquid. Heparin,
spectrophotometry (2.4.6), the water content is withinthe limits antithrombin and other auxiliary substances such as a
approved by the competent authority.' stabi1iser may be added. No antimicrobial preservative is added.
The solution is passed through a bacteria-retentive filter,
Sterility (2.2.11). Complies with the test for sterility. distributed aseptically into the final containers and
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject immediately frozen. It is subsequently freeze-dried and the
per kg of the rabbit's mass a volume equivalent to not less containers are closed under vacuum or under an inert gas.
than 50 ill offactor IX.
Consistency ofthe method
Assay. Determine the assay ofhuman blood coagulation factor
The consistency of the method of production with respect to
IX (2.8.8).
the activities of factors II, IX and X of the preparation,
The estimated potency is not less than 80.0 per cent and not expressed in International Units relative to the activity of~actor
more than 125.0 per cent ofthe stated potency. The confidence VII, shall be demonstrated.
limits (P = 0.95) are not less than 80.0 per cent and not more
The consistency of the method of production with respect to
than 125.0 per cent ofthe estimated potency.
the activity of factor VIla of the preparation shall be
Storage. Store protected from light. demonstrated. The activity offaCtor VIla may be detennined,
Labelling. The label states (1) the number ofInternational for example, using a recombinant soluble tissue factor that
Units offactor IX per container; (2) the amount ofprotein per does not activate factor VII but possesses a cofactor function
container; (3) the name and quantity of any added substances specific for factor VIla; after incubation of a mixture of the
including, where applicable, heparin; (4) the name and volume recombinant soluble tissue factor with phospholipids reagent
of the liquid to be used for reconstitution; (5) that the and the dilution of the test sample in factor VII-deficient
transmission of infectious agents cannot be totally excluded plasma, calcium chloride is added and the clotting time
when medicinal products prepared from human blood or determined; the clotting time is inversely related to the factor
plasma are administered. VIla activity of the test sample.
Description. A hygroscopic powder or friable solid that may
be white, pale yellow, green or blue.

Human Coagulation Factor VII Reconstitute the preparation under examination as stated
on the label immediately before carrying out the
Human Coagulation Factor VII is a plasma protein fraction Identification, Tests (except those for solubility and water)
that contains the single-chain glycoprotein factor VII and may and Assay.

2571

www.webofpharma.com
HUMAN COAGULATION FACTOR VII IP 2010

Identification The estimated content is not more than 125.0 per cent of the
stated content. The confidence limits (P = 0.95) are not less
It complies with the limits ofthe Assay. than 80.0 per cent and not more than 125.0 per cent of the
estimated potency.
Tests
pH (2.4.24). 6.5 to 7.5. Factor X

Osmolality (2.4.23). Minimum 240 mosmol per kg. Determine the assay ofhuman coagulation factor X (2.8.9).

Total protein. If necessary, dilute an accurately measured The estimated content is not more than 125.0 per cent of the
volume of the reconstituted preparation with a 0.9 per cent stated content. The confidence limits (P = 0.95) are not less
solution w/v of sodium chloride to obtain a solution expected than 90.0 per cent and not more than 111.0 per cent of the
to contain about 15 mg of protein in 2 ml. To 2.0 ml of the estimated potency.
solution in a round-bottomed centrifuge tube add 2 ml ofa 7.5
Water. Determine by semi-micro determination ofwater (2.3 .43),
per cent w/v solution of sodium molybdate and 2 ml of a
loss on drying (2.4.19) or near-infrared spectrophotometry
mixture of 1 volume of nitrogen-free sulphuric acid and 30
(2.4.6), the water content is within the limits approved by the
volumes of water. Shake, centrifuge for 5 minutes, decant the
competent authority.
supernatant liquid and allow the inverted tube to drain on
filter paper. Determine the nitrogen in the residue by the Sterility (2.2.11). Complies with the test for sterility.
method of sulphuric acid digestion (2.3.30) and calculate the
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
amount ofprotein by multiplying the result by 6.25.
per kg of the rabbit's mass a volume equivalent to not less
Activated coagulation factors (2.8.4). For each ofthe dilutions, than 30 ill offactor VII.
the coagulation time is not less than 150 seconds.
A~say
Heparin. If heparin has been added during preparation,
determine the amount present by the assay of heparin in Determine the assay ofhuman coagulation factor VII (2.8.6).
coagulation factor concentrates (2.8.10). The preparation under
examination contains not more than the amount of heparin The estimated potency is not less than 80.0 per cent and not
stated on the label and in any case not more than 0.5 ill of more than 125.0 per cent ofthe stated potency. The confidence
heparin per International Unit offactor VII. limits (P = 0.95) are not less than 80.0 per cent and not more
than 125.0 per cent of the estimated potency.
Thrombin. If the preparation under examination contains
heparin, determine the amount present as described in the Storage. Store protected from light.
test for heparin and neutralise the heparin by addition of Labelling. The label states (1) the number of International
protamine sulphate (10 /lg ofprotamine sulphate neutralises Units of factor VII per container; (2) the maximum content of
1 ill ofheparin). In each of2 test-tubes, mix equal volumes of International Units of factor II, factor IX and factor X per
the reconstituted preparation and a 0.3 per cent solution of container; (3) the amount of protein per container; (4) the
fibrinogen. Keep one of the tubes at 37° for 6hours and the name and quantity of any added substances, including where
other at room temperature for 24 hours. In a third tube, mix a applicable, heparin; (5) the name and volume of the liquid to
volume of the fibrinogen solution with an equal volume of a be used for reconstitution; (6) that the transmission of
solution of human thrombin (l IU per ml) and place the tube infectious agents cannot be totally excluded when medicinal
in a water-bath at 37°. No coagulation occurs in the tubes products prepared from human blood or plasma are
containing the preparation under examination. Coagulation
administered.
occurs within 30 seconds in the tube containing thrombin.

Factorll
Determine the assay of human coagulation factor II (2.8.5). Human Coagulation Factor VIII
The estimated content is not more than 125.0 per cent of the (rDNA)
stated content. The confidence limits (P = 0.95) are not less Human Coagulation Factor VIII (rDNA) is a freeze-dried
t~aIl90.0 per cf:llt and n()t l11()rethall 111 ~ 0 per (;~nt of the preparationofglycoproteinshaving the same activity as
estimated potency. coagulation fa.ctor VUI in.humanplasma.Jtactsas_a.c_Qfactor
of the activation of factor X in the presence of factor IXa,
Factor IX
phospholipids and calcium ions. It circulates in plasma
Determine the assay of human coagulation factor IX (2.8.8). mainly as a two-chain glycosylated protein with 1 heavy

2572

www.webofpharma.com
IP 2010 HUMAN COAGULATION FACTOR VIII (rDNA)

(relative molecular mass ofabout 2,00,000) and 1 light (relative applicable, the characterisation tests may alternatively be
molecular mass 80,000) chain held together by divalent metal carried out on the finished product.
ions. Human coagulation factor VIII (rDNA) is prepared as
full-length factor VIII (octocog alfa), or as a shortened two- Specific biological activity or ratio offactor VITI activity to
chain structure (relative molecular mass 90,000 and 80,000), in factor VITI antigen
which the B-domain has been deleted from the heavy chain Determine the assay ofhuman coagulation factor VIII (2.8.7).
(moroctocog alfa). The protein content, or where a protein stabiliser is present,
Full-length human rDNA coagulation factor VIII contains 25 the factor VIII antigen content, is determined by a suitable
potential N-glycosylation sites, 19 in the B domain of the method and the specific biological activity or the ratio offactor
heavy chain, 3 in the remaining part ofthe heavy chain (relative VIII activity to factor VIII antigen is calculated.
molecular mass 90,000) and 3 in the light chain (relative
Protein composition
molecular maSS 80,000). The different products are
characterised by their molecular size and post-translational The protein composition is determined by a selection of
modification and/or other modifications. appropriate characterisation techniques which may include
peptide mapping, Western blots, HPLC, gel electrophoresis,
Production capillary electrophoresis, mass spectrometry or other
techniques to monitor integrity and purity. The protein
Human coagulation factor VIII (rDNA) is produced by
composition is comparable to that ofthe reference preparation.
recombinant DNA technology in mammalian cell culture. It is
produced under conditions designed to minimise microbial Molecular size. Using size-exclusion chromatography
contamination. (2.4.16), the molecular size distribution is comparable to that
Purified bulle factor VIII (rDNA) may contain added human of the reference preparation.
albumin and/or other stabilising agents, as well· as other
Peptide mapping (2.3.47). There is no significant difference
auxiliary substances to provide, for example, correct pH and
between the test protein and the reference preparation.
osmolality.
Carbohydrates/sialic acid. To monitor batch-to-batch
The specific activity is not less than 2,000 ill offactor VIII:C
consistency, the monosaccharide content and the degree of
per mg of total protein before the addition of any protein
sialylation or the oligosaccharide profile are monitored and
stabiliser, and varies depending on purity and the type of
correspond to those of the reference preparation.
modification ofmolecular structure offactor VIII.
Final lot
The quality of the bulk preparation is controlled using
reference preparations. It complies with the tests under Identification, Tests and
Assay.
Reference preparations
During development, reference preparations are established Excipients. 80.0 per cent to 120.0 per cent ofthe stated content,
for subsequent verification of batch consistency during determined by a suitable method, where applicable.
production, and for control ofbull( and final preparation. They
are derived from representative batches ofpurified bulk factor Description. A white or slightly yellow powder or friable mass.
VIII (rDNA) that are extensively characterised by tests
including those described below and whose procoagulant
Identification
and other relevant functional properties have been A. It complies with the limits of the Assay.
ascertained and compared, wherever possible, with the
International Standard for factor VIII concentrate. The B. The distribution ofcharacteristic peptide bands corresponds .
reference preparations are suitably characterised for their with that ofthe reference preparation (SDS-PAGE or Western
intended purpose and are stored in suitably sized aliquots blot).
under conditions ensuring their stability. Tests
Purified bulk factor VITI (rDNA)
Reconstitute the preparation as stated on the label
The purified bulk complies with a suitable combination of immediately before canying out the Tests (except those for
the following tests for characterisation of integrity of the solubility and water) and Assay.
factor VIII (rDNA). Where any substance added during
pR(2.4.24).6.5t07.5.
preparation of the purified bulk interferes with a test, the
test is carried out before addition ofthat substance. Where Osmolality (2.4.23). Minimum 240 mosmol per kg.

2573

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN IP 2010

Water. Determine by serrli-rrlicro determination ofwater (2.3.43), It is prepared from pooled material ofa minimum volume of25
loss on drying (2.4.19) or near infrared spectrophotometry litres by a method which has been shown (a) to be capable of
(2.4.6), the water content is within the limits approved by the concentrating tenfold from source material at least two different
competent authority. antibodies, one viral and one bacterial", for which an
International Standard or Reference Preparation is available;
Sterility (2.2.11). Complies with the test for sterility.
(b) not to affect the integrity of the globulins; (c) to
Bacterial endotoxins (2.2.3). Less than 3 ill in the volume consistently yield a product which is safe for intramuscular
that contains 100 ill offactor VIII activity. injection and; (d) to yield a product .that does not transmit
viral hepatitis or any other infection.
Assay
The liquid preparation is prepared as a stabilised solution in
Determine the assay ofhuman coagulation factor VIII (2.8.7). saline solution or a 2.25 per cent w/v solution of glycine or
The estimated potency is not less than 80.0 per cent and not other suitable agent and is sterilised by filtration and
more than 125.0 per cent ofthe stated potency. The confidence distributed into previously sterilised containers which are then
limits (P = 0.95) are not less than 80.0 per cent and not more sealed so as to exclude micro-organisms. An antimicrobial
than 120.0 per cent of the estimated potency. preservative may be added except when the preparation is to
be freeze-dried. Any antimicrobial preservative or stabilising
Storage. Store protected from light. agent added must be such that neither deleterious effect on
Labelling. The label states (1) the factor VIII content in the final product in the amounts present nor capability to
International Units; (2) the name and amount ofany excipient; cause untoward reactions in human beings is demonstrated.
(3) the composition and volume of the liquid to be used for An accelerated degradation test is can-ied out on the final
reconstitution. liquid or freeze-dried preparation by heating at 37° for 4 weeks.
The difference between the percentages of protein eluted in
the fractions following the main peak as determined by size-
exclusion chromatography (2.4.16), before and after exposurt?
Human Normal Immunoglobulin at 37° does not exceed 5.0 per cent.
Normal Immunoglobulin; Immune Human Serum Globulin;
Human Normal Immunoglobulin contains not less than 90.0
Human Gamma Globulin
per cent and not more than 110.0 per cent of the quantity of
Human Normal Immunoglobulin is a sterile solution or freeze- protein stated on the label and in any case, not less than 10.0
dried preparation containing immunoglobulins, mainly per cent w/v and not more than 18.0 per cent w/v of protein.
immunoglobulin G (IgG), together with smaller amounts of
other plasma proteins. Description. The liquid preparation is clear pale yellow or
brownish in colour; on storage it may show turbidity or a
Production small amount ofparticulate matter. The freeze-dried preparation
is a white to slightly yellow powder or solid, friable mass.
It is obtained from source materials such as the blood, plasma,
serum or placentae frozen immediately after collection from Identification
healthy donors who must as far as can be ascertained after
clinical examination, laboratory tests on their blood and a study A. Precipitation tests with a suitable range ofspecies-specific
of their medical history, be free from disease transmissible by antisera which give positive results for the presence ofproteins
transfusion of blood or blood products. The examinations of human origin and negative results with antisera specific to
and tests to be carried out are decided by the National plasma proteins of the other species.
Regulatory Authority; in particular, tests for hepatitis B surface
B. Examine by electrophoresis (2.4.12), using the moving
antigen, HCV antibodies and for HIV antibodies must be
boundary technique and a 1.0 per cent w/v solution in
can-ied out by suitable sensitive methods and must show
barbitone buffer solution pH 8.6 of ionic strength 0.1. At
negative results in both cases. Plasma, serum or placentae
least 90.0 per cent w/v ofthe protein has a mobility not greater
obtained from donors who do not meet all the requirements than -2.8 x 10-5 cm2V- 1S-I .
stated above may be used as source material provided that it
has_bee~demonstratedjo_the-llationaLauthority.thaLprocess'l'ests--------~----- ---------.------._-.-..-. -----------
Qfft~<::tiQnllJjQl11111<:l_.prQ<:ll1<::.tiQl1.rf)mQ~fill.l1Y
.mQ\:Y.l111gfJ.l1t
capable of adversely affecting the health of subjects treated pH (2.LCi4Y.-6.4 to 7.2, deterrnlnedTn a solution prepared by
with the preparation. No antibiotic is added to the source dilution of a quantity with saline solution so as to contain 1.0
materials used for the preparation of immunoglobulin. per cent w/v of protein.

2574

www.webofpharma.com
IP 2010 HUMAN NORMAL IMMUNOGLOBULIN

Protein composition. Determine by cellulose acetate the areas ofthe peaks containing IgG monomer, dimer, albumin
electrophoresis (2.4.12), but applying an electric field such and other proteins ofsimilar molecular size (area B) is not less
that the albumin band of normal human serum applied in a than 85.0 per cent ofthe total area of the chromatogram. Not
control strip migrates at least 30 mm and using one strip of more than 10.0 per cent of the total area ofthe chromatogram
cellulose acetate for each of the following solutions: represents proteins eluted ahead ofigG dimer (area A); ifarea
A can be subdivided into two distinct areas, the area
Test solution. Dilute the preparation under examination with
corresponding to larger proteins does not exceed 5.0 per cent
saline solution to produce a solution containing 5 per cent
of the total area ofthe chromatogram. Not more than 5.0 per
w/v ofprotein.
cent ofthe total area ofthe chromatogram represents proteins
Reference solution. Reconstitute human immunoglobulinfor eluted after IgG monomer and albumin (area C).
electrophoresis RS with saline solution to produce a solution
Stability. Heat approximately 2 ml at 57° for 4 hours in a
containing 5 per cent w/v of protein.
stoppered glass tube (75 mm x 12 mm); no gelation or
Calculate the result as the mean ofthree measurements of the flocculation occurs.
absorbance of each strip. In the electrophoretogram obtained
Pyrogens (2.2.8). Complies with the test for pyrogens, using
with test solution not more than 10.0 per cent ofthe protein is
1 ml ofthe preparation under examination per kg ofthe rabbit's
contained in bands other than the principal band. The test is
weight.
not valid unless the proportion of protein in the principal
band in the electrophoretogram obtained with reference Sterility (2.2.11). Complies with the tests for sterility.
solution is within the limits stated in the leaflet supplied with
human immunoglobulin for electrophoresis RS. Abnormal toxicity (2.2.1). Complies with the test for abnormal
toxicity, Method A, injecting 0.5 ml into each mouse and 5 ml
Molecular size. Determine by size-exclusion chromatography into each guinea-pig.
(2.4.16), applying 2 ml of the preparation under examination
diluted with mixed phosphate buffer pH 7.0 with azide to Other tests. Complies with the tests stated under Parenteral
produce a solution containing 4.0 to 5.0 per cent wIv ofprotein. Preparations (Injections).

Chromatographic system Assay. Dilute a suitable volume with water to produce a


a column 1 m x 25 mm packed with agarose trapped solution containing 1.0 per cent w/v ofprotein. Take 1.5 ml of
within a cross-linked polyacrylamide network and the dilution in a round-bottomed centrifuge tube. Add 5 ml of
having a linear fraction range suitable for fractionation wate/; mix, add 0.2 ml on.5 per cent w/v solution of sodium
of globular proteins in the range of molecular weights molybdate and 2 ml of a mixture consisting of 1 volume
1i-om 20,000 to 350,000, nitrogen free sulphuric acid and 30 volumes of water. Shake,
- mobile phase: mixedphosphate buffer pH 7.0 with centrifuge for five minutes, decant the supernatant liquid and
azide, allow the inverted tube to drain on filter paper. To the residue
flow rate. 20 ml per hour (4 ml per cm2 ofcolumn in the tube add three drops of a 30 per cent w/v solution of
cross-sectional area), copper sulphate and 1 ml of nitrogen-free sulphuric acid
spectrophotometer set at 280 nm. and boil gently for 10 minutes; cool, add 1 g of anhydrous
sodium sulphate and 10 mg of selenium, boil gently for 1 hour
Collect the eluate in fractions of about 4 ml. Examine the and cool. Transfer to an ammonia distillation apparatus, add
chromatogram in comparison with that in Fig.l. The sum of 6 ml of a saturated solution of sodium hydroxide and pass
steam through the flask; distil for seven minutes, collecting
f\ the distillate in a mixture of 5 ml of a saturated solution of
boric acid, 5 ml of water, and 1 drop saturated solution of
methyl red in alcohol containing 0.1 per cent of methylene
blue, and titrate with 0.02 M hydrochloric acid.
1 ml of 0.02 Mhydrochloric acid is equivalentto 0.00175 g of
protein.

/'~!
Freeze-dried Human Normal Immunoglobulin complies with
\ the following additional requirements.
llicOO:::i'1:-'-~~==::-iiB-::=~~~~C~:;;;a,~5~OOIll1 Solubility rate. Add the volume of the liquid stated on the
ELUATE VOLUME label and allow it to stand for 15 minutes at a temperature of
Fig.l Typical Chromatogram for Human Normal Immunoglobulin 20° to 25°; it dissolves completely.

2575

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN IP 2010

Loss on drying (2.4:1 9). Not more than 2.0 per cent, determined in particular tests for hepatitis B surface antigen and for HIV
on 0.5 g, by drying over phosphorus pentoxide at a pressure antibodies are carried outby suitable sensitive methods and
not exceeding 3 Pa for 24 hours. must give negative results in .both cases. Other disease-
causative agents that are not destroyed or removed by the
Human Normal Immunoglobulin intended for use in the
processing method must not be present. Plasma or serum
prevention of infective hepatitis (hepatitis A) complies with
obtained from donors who do not meet all the stated
the follOWing additional requirement.
requirements may be used as source material provided that it
Anti-hepatitis Aactivity. Determine the anti-hepatitis A activity has been demonstrated to the national authority that the
by comparison with the activity of the Standard preparation, process offractionation will remove any lmown agent capable
using an immunoassay of suitable sensitivity and specificity. of adversely affecting the health of recipients of the Human
The stated potency is not less than 100 Units per m!. The Plasma Protein Fraction.
estimated potency is not less than the stated potency. The
The separation of the protein may be done by precipitation
fiducial limits of error are not less than 80.0 per cent and not
with suitable organic solvents under controlled conditions,
more than 125.0 per cent.
particularly of pH, ionic strength and temperature, so that in
Standard Preparation. The Standard Preparation is the 1st the final product not less than 85.0 per cent ofthe total protein
International Reference Preparation for Hepatitis A is albumin. Residual solvent, if present, may be removed by
immunoglobulin, established in 1981, consisting offreeze-dried freeze-drying or other suitable treatment. Alternative methods
material derived from fractionated plasma (supplied in ofpreparation which shall not affectthe integrity ofthe product
ampoules containing 100 Units), or another suitable and shall have been shown to yield consistently a product
preparation the antigen binding ofwhich has been determined which is safe for intravenous injection may be adopted.
in relation to the International Reference Preparation.
The product is dissolved in water and sufficient quantities of
Storage. Store protected from light, the liquid preparation in a suitable stabiliser against the effect of heat, like sodium
sealed, colourless, glass containers, at a temperature between caprylate in a suitable concentration, and sufficient sodium
2° and 8°. Store the freeze-dried preparation under vacuum or chloride to adjust the sodium ions to between 130 and 160
under an inert gas. millimoles per litre may be added but no antibiotic or
Labelling. The label states (1) the volume and the protein antimicrobial preservative is added at any stage during
concentration expressed in g per litre or, for freeze-dried preparation. The solution is sterilised by filtration through a
preparations, the total amount of protein in the container; (2) bacteria-retentive filter and distributed aseptically into sterile
the type of source material; (3) the name and quantity of any containers which are then closed so as to prevent microbial
added preservative or stabilising agent; (4) the recommended contamination. The solution in its final container is heated at
human dose; (5) that it is meant for intramuscular injection 60° ± 0.5° and maintained at this temperature for 10 hours. The
only; (6) the storage conditions. containers are then incubated at 30° to 32° for not less than 14
days or at 20° to 25° for not less than 4 weeks and examined
visually for evidence of microbial contamination. Those
showing abnormalities such as abnormal colour, turbidity,
Human Plasma Protein Fraction presence of atypical particles or microbial contamination are
discarded.
Plasma Protein Solution; Human Albumin Fraction (Saline);
Plasma Protein Fraction; PPF Human Plasma Protein Fraction contains not less than 95.0
per cent and not more than 105.0 per cent ofthe stated amount
Human Plasma Protein Fraction is a sterile isotonic aqueous
of total protein.
solution of proteins of plasma or serum containing albumin
and globulins. It is prepared as an isotonic solution containing Description. A clear, almost colourless or pale yellow liquid;
4.0 to 5.0 per cent w/v oftotal protein. It contains no fibrinogen almost odourless. On storage a dust-like precipitate may
or antibodies. develop but it disappears on shaking.

Production Identification
A. Precipitation tests with specific antisera show that the
The plasma or serum is obtained froin healthy human donors
preparation consists only ofplasma proteins of human origin
who.must, .after cJinicaLexarninaJiQl1,la.b.otat.oryJ:esJs_QDJ:heir
bl(){)cLiil1c1 a. .~tllcly()f tl1()ir .l11ecli()all1i~t()ry ,J:>()ft-()()ft-()l11
onl)lano gives negativefesiiltsWitli-antiseraspeeificto·plasma
proteins of"other species: "
detectable agents of infection transmissible by transfusion of
blood or blood derivatives. The examinations and tests to be B. Examine by a suitable immunoelectrophoresis technique.
carried out are decided by the National Regulatory Authority; Using antiserum to normal human serum, compare nOlmal

2576

www.webofpharma.com
IP 2010 HUMAN PLASMA PROTEIN FRACTION

human serum and the preparation under examination, both Sodium. Not less than 95.0 per cent and not more than 105.0
diluted to contain 1.0 per cent w/v of protein. The main per cent of the stated amount and, in any case not more than
component ofthe preparation under examination corresponds 160 millimoles ofNa per litre, determined by Method A for
to the main component of the normal human serum. The atomic absorption spectrophotometry (2.4.2), measuring at
solution may show the presence of small quantities of other 589 nm and using sodium solution AAS suitably diluted with
plasma proteins. water to prepare the standard solutions.

Tests Potassium. Not more than 50 !!mol of K per g of protein,


determined by atomic absorption spectrophotometry (2.4.2),
pH (2.4.24).6.7 to 7.3, determined in a solution prepared by measuring at 766 nm and using potassium solution AAS,
diluting with sufficient of saline solution to produce a solution suitably diluted with water to prepare the standard solutions.
containing 1.0 per cent w/v of protein.
Alkaline phosphatase. Not more than 0.1 Unit per g ofprotein,
Polymers and aggregates. Determine by size-exclusion detennined by the following method. Transfer a mixture of0.5
chromatography (2.4.16), applying 2 ml ofthe preparation under ml of the substance under examination and 0.5 ml of
examination. diethanolamine buffer pH 10.0 to a spectrophotometer cell
Chromatographic system maintained at a temperature of 37° ± 0.2° and add 0.1 ml of
- a column 1mx25 mmpacked with a cross-linked dextran nitrophenyl phosphate solution. Record the absorbance of
suitable for fractionation ofglobular proteins in the range the solution at about 405 nm (2.4.7), over a period ofat least 30
of molecular weights from 5,000 to 3,50,000 (such as seconds from the time of addition of the nitrophenyl
Sephadex 0-150), phosphate solution. Calculate the alkaline phosphatase
mobile phase: mixedphosphate bufferpH 7.0 with azide, activity at 37° in Units per g of protein from the expression
- flow rate. 20 ml per hour (4 ml per square centimeter of 118.3x1P, where x is the rate of increase of absorbance per
column cross-sectional area), minute and P is the content of total protein in g per litre, as
spectrophotometer set at 280 nm. determined in the Assay.

Collect the eluate in fractions of about 4 ml and combine the Haem content. Dilute with sufficient saline solution to produce
fractions corresponding to each peak. For each combined a solution containing 1.0 per cent w/v of protein; absorbance
fraction, determine by Method E for determination ofnitrogen of the resulting solution at about 403 nm, not more than 0.15
(2.3.30). (2.4.7).
1 ml of 0.02Mhydrochloric acid is equivalent to 0.00028 g of Sterility (2.2.11). Complies with the tests for sterility.
nitrogen. Not more than 10.0 per cent of the total nitrogen is
Abnormal toxicity (2.2.1). Complies with the test for abnormal
present in the combined fraction associated with non-retained
toxicity, using Method Band 0.5 ml of the solution for each
proteins.
mouse and 5 ml for each guinea-pig irrespective ofthe protein
Protein composition. Carry out by cellulose acetate content.
electrophoresis (2.4.12), Method II using one strip for each
Pyrogens (2.2.8). Complies with the test for pyrogens, using
solution.
3 ml per kg of the rabbit's weight in rabbits that have not
Test solution. Dilute the preparation under examination with previously received blood products.
saline solution to obtain a solution containing 2.0 per cent
w/v of protein. Assay
Reference solution. Dilute human plasma protein fraction For protein. Dilute to about 0.75 percent w/v oftotalprotein
for electrophoresis RS with saline solution to obtain a solution with saline solution. Take 2 ml of this solution in a round-
containing 2.0 per cent w/v of protein. bottomed centrifuge tube, add 2 mlof a 7.5 per cent w/v
Calculate the result as the mean ofthree measurements ofthe solution of sodium molybdate and 2 ml of a mixture of 30
absorbance ofeach ofthe 10 strips. In the electrophoretogram volumes of water and 1 volume of nitrogen-free sulphuric
obtained with test solution not more than 15.0 per cent of the acid. Shake, centrifuge for 5 minutes, decant the supernatant
protein is contained in bands other than the principal band. liquid and let the inverted tube stand on a filter paper to drain
The test is not valid unless the proportion of protein in the the fluid. Carry out Method E for determination of nitrogen
principal band in the electrophoretogram obtained with (2.3.30), on the residue thus obtained and multiply the result
reference solution is within the limits stated in the leaflet by 6.25 to obtain the protein content.
supplied with human plasma protein fraction for Storage. Store protected from light at a temperature between
electrophoresis RS. 2° and 25°.

2577

www.webofpharma.com
HUMAN PROTHROMBIN COMPLEX IP 2010

Labelling. The label states (1) the volume in the container; (2) Identification
the total amount of protein in the container expressed as a
percentage or in grams per litre; (3) the concentration ofsodium It complies with the limits of the assay for coagulation factor
ions in millimoles per litre; (4) the names and concentrations IX activity and, where applicable, those for factors II, VII and
of stabilising agents and of any other added substances X
present in the final solution; (5) that the solution should not
be used if the solution is cloudy or shows a deposit which Tests
does not disappear on shaking; (6) that, once the container pH (2.4.24). 6.5 to 7.5.
has been penetrated, the contents must be used within 4 hours
and any unused solution discarded; (7) the storage conditions. Osmolality (2.4.23). Minimum 240 mosmol per kg.
Total protein. If necessary, dilute an accurately measured
volume of the reconstituted preparation with a 0.9 per cent
Human Prothrombin Complex w/v solution of sodium chloride to obtain a solution expected
to contain about 15 mg of protein in 2 ml. To 2.0 ml of the
Human Prothrombin Complex is a plasma protein fraction
solution in a round-bottomed centrifuge tube add 2 ml ofa 7.5
containing blood coagulation factor IX together with variable
amounts of coagulation factors II, VII and X; the presence
per cent w/v solution of sodium molybdate and 2 ml of a
mixture of 1 volume of nitrogen-free sulphuric acid and 30
and proportion of these additional factors depends on the
volumes of water. Shake, centrifuge for 5 minutes, decant the
method of fractionation. It is obtained from human plasma
supernatant liquid and allow the inverted tube to drain on
that complies with the monograph on Human Plasma for
filter paper. Determine the nitrogen in the residue by the
Fractionation.
method of sulphuric acid digestion (2.3.30) and calculate the
The potency of the preparation, reconstituted as stated on amount of protein by multiplying the result by 6.25.
the label, is not less than 20 ill offactor IX per ml.
Activated coagulation factors (2.8.4). Ifnecessary, dilute the
Production preparation under examination to contain 20 ill of factor IX
per ml. For each of the dilutions, the coagulation time is not
The method ofpreparation is designed to minimise activation less than 150 seconds.
of any coagulation factor (to minimise potential
thrombogenicity) and includes a step or steps that have been Heparin. If heparin has been added during preparation,
shown to remove or to inactivate known agents of infection; determine the amount present by the assay of heparin in
if substances are used for inactivation of viruses during coagulation factor concentrates (2.8.10). The preparation under
production, the subsequent purification procedure must be examination contains not more than the amount of heparin
validated to demonstrate that the concentration of these stated on the label and in any case not more than 0.5 ill of
substances is reduced to a suitable level and that any residues heparin per International Unit of factor IX.
are such as not to compromise the safety of the preparation Thrombin. If the preparation under examination contains
for patients. heparin, determine the amount present as described in the
The specific activity is not less than 0.6 ill offactor IX per mg test for heparin and neutralise it by addition of protamine
of total protein, before the addition of any protein stabiliser. sulphate (10 Jlg of protamine sulphate neutralises 1 ill of
The prothrombin complex fraction is dissolved in a heparin). In each of 2 test-tubes, mix equal volumes of the
suitable liquid. Heparin, antithrombin and other auxiliary reconstituted preparation and a 0.3 per cent w/v solution of
substances such as a stabiliser may be added. No antimicrobial fibrinogen. Keep one of the tubes at 37° for 6 hours and the
preservative is added. The solution is passed through a other at room temperature for 24 hours. In a third tube, mix a
bacteria-retentive filter, distributed aseptically into the final volume of the fibrinogen solution with an equal volume of a
containers and immediately frozen. It is subsequently freeze- solution of human thrombin (1 IU per ml) and place the tube
dried and the containers are closed under vacuum or under in a water-bath at 37°. No coagulation occurs in the tubes
an inert gas. containing the preparation under examination. Coagulation
. occurs within 30 seconds in the tube containing thrombin.
Description. A white or slightly coloured powder or friable
s~lid, ~e_l}'~~~'<:>~~?l'i.c-.:_ ... ... ._~ ._.~.__ . . __._.. Water. Determine by semi-micro determination of water
.... R~GQ/Js.titljt~ th~ . p,.epq[(JljQ!J..ljnr1er.~~Clmill.qtion as stated ···(23.43)~loss-on-drying·(2:4·.-19)or·near~infraredspectrometry
..... _.__.
on the label immediately before carryi~g~~~ -;iz-; (2;4;6); the watercontentiswithinthelimitsapprovedbythe
Identification, Tests (except those for solubility and water) competent authority.
and Assay. Sterility (2.2.11). Complies with the test for sterility.

2578

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE
IP 2010
Q

Pyrogens (2.2.8). Complies with the test for pyrogens. Inject Human Normal Immunoglobulin for
per kg of the rabbit's mass a volume of the reconstituted
preparation equivalent to not less than 30 IU of factor IX. Intravenous Use
Human Normal Immunoglobulin for Intravenous
Assay
Administration
Factor IX Human Normal Immunoglobulin for Intravenous
Administration is a liquid or freeze-dried preparation containing
Determine the assay ofhuman coagulation factor IX (2.8.8).
immunoglobulins, mainly immunoglobulin G (IgG). Other
The estimated potency is not less than 80.0 per cent and not proteins may be present. Human normal immunoglobulin for
more than 125.0 per cent ofthe stated potency. The confidence intravenous administration contains the IgG antibodies of
interval (P = 0.95) ofthe estimated potency is not greater than normal subjects. This monograph does not apply to products
80.0 per cent to 125.0 per cent. intentionally prepared to contain fragments or chemically
modified IgG.
Factor II
Human normal immunoglobulin for intravenous administration
Determine the assay of human coagulation factor II (2.8.5). is obtained ii-om plasma that complies with the requirements
of the monograph on Human plasma for fractionation. No
The estimated potency is not less than 80.0 per cent and not antibiotic is added to the plasma used.
more than 125.0 per cent ofthe stated potency. The confidence
interval (P = 0.95) ofthe estimated potency is not greater than Production
90.0 per cent to 111.0 per cent.
The method of preparation includes a step or steps that have
Factor VII been shown to remove or to inactivate known agents of
infection; if substances are used for inactivation of viruses, it
If the label states that the preparation contains factor VII, shall have been shown that any residues present in the final
Determine the assay of human coagulation factor VII product have no adverse effects on the patients treated with
(2.8.6). the immunoglobulin.
The estimated potency is not less than 80.0 per cent and not
The product shall have been shown, by suitable tests in
more than 125.0 per cent ofthe stated potency. The confidence
animals and evaluation during clinical trials, to be well tolerated
interval (P = 0.95) ofthe estimated potency is not greater than
when administered intravenously.
80.0 per centto 125.0 per cent.
Human normal immunoglobulin for intravenous administration
Factor X is prepared from pooled material from not fewer than 1,000
doriors by a method that has been shown to yield a product
Determine the assay of human coagulation factor X (2.8.9).
that (a) does not transmit infection; (b) at an immunoglobulin
The estimated potency is not less than 80.0 per cent and not concentration of 50 g per litre, contains antibodies for at least
more than 125.0 per cent ofthe stated potency. The confidence 2 of which (one viral and one bacterial) an International
interval (P = 0.95) ofthe estimated potency is not greater than Standard or Reference Preparation is available, the
90.0 per cent to 111.0 per cent. concentration of such antibodies being at least 3 times that in
the initial pooled material; (c) has a defined distribution of
Storage. Store protected from light. immunoglobulin G subclasses; (d) complies with the test for
Fc function of immunoglobulin.
Labelling. The label states (1) the number of International
Units of factor IX, factor II and factor X per container; Test for Fc function ofimmunoglobulin
(2) where applicable, the number of International Units of
Stabilised human blood. Collect group 0 human red blood
factor VII per container; (3) where applicable, that the
into ACD anticoagulant solution. Store the stabilised blood
preparation contains protein C and/or protein S; (4) the amount
at 4° for not more than 3 weeks.
of protein per container; (5) the name and quantity of any
added substances, including where applicable, heparin; Phosphate buffered saline pH 7.2. Dissolve 1.022 g of
(6) the name and quantity of the liquid to be used for anhydrous disodium hydrogen phosphate, 0.336 g of
reconstitution; (7) that the transmission of infectious agents anhydrous sodium dihydrogen phosphate and 8.766 g of
cannot be totally excluded when medicinal products prepared sodium chloride in 800 ml of water and dilute to 1,000 ml with
from human blood or plasma are administered. the same solvent.

2579

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE IP 2010

Magnesium and calcium stock solution. Dissolve 1.103 g of barbital buffer solution, thereby obtaining the final adjusted
calcium chloride and 5.083 g of magnesium chloride in water volume Vf = Vr x A of sensitised human red blood cells and
and dilute to 25 ml with the same solvent. adjusting A to 1.0 ± 0.1 for a tenfold dilution.
Barbital buffer stock solution. Dissolve 207.5 g of sodium Antibody binding ofantigen-coated tanned human red blood
chloride and 25.48 g of barbital sodium in 4,000 ml of water cells. Prepare the following solutions in succession and in
and adjust to pH 7.3 using 1 M hydrochloric acid. Add 12.5 duplicate, using for each solution a separate half-micro cuvette
ml of magnesium and calcium stock solution and dilute to (for example, disposable type) or test-tube:
5,000 m1 with water. Filter through a membrane filter (pore size
Test solutions. Ifnecessary, adjust the immunoglobulin under
0.22 gm). Store at 4° in glass containers.
examination to pH 7, for example by addition of 1 M sodium
Albumin barbital buffer solution. Dissolve 0.150 g of bovine hydroxide. Dilute volumes of the preparation under
albumin in 20 ml of barbital buffer stock solution and dilute examination containing 30 mg and 40 mg ofimmunoglobulin
to 100 ml with water. with albumin barbital buffer solution and adjust the volume
Tannic acid solution. Dissolve 10 mg of tannic acid in 100 ml to 900 Ill.
of phosphate-buffered saline pH 7.2. Prepare immediately Reference solutions. Prepare as for the test solutions using
before use. human immunoglobulin RS.
Guinea-pig complement. Prepare a pool of serum from the Complement control. 900 III of albumin barbital buffer
blood of not fewer than 10 guinea-pigs. Separate the serum solution.
from the clotted blood by centrifugation at about 4°. Store the
serum in small amounts below _70°. Immediately before starting Add to each cuvette/test-tube 100 III of sensitised human red
complement-initiated haemolysis, dilute to 125 to 200 CHso blood cells and mix well.
per ml with albumin barbital buffer solution and store in an Incubate at room temperature for 15 minutes, add 1,000 III of
ice-bath during the test. albumin barbital buffer solution, collect the cells by
Rubella antigen. Suitable rubella antigen for centrifugation (1,000 g for 10 min) of the cuvette/test-tube
haemagglutination-inhibition titre (HIT). Titre> 256 HA units. and remove 1,900 III ofthe supernatant. Replace the 1,900 III
with albumin barbital buffer solution and repeat the whole
Preparation oftanned human red blood cells. Separate human ofthe washing procedure, finally leaving a volume of200 Ill.
red blood cells by centrifuging an appropriate volume of Test samples may be stored in sealed cuvette/test-tubes at 4°
stabilised human blood and wash the cells at least 3 times for 24 hours.
with phosphate-buffered saline pH 7.2 and suspend at 2 per
cent v/v in phosphate-bufferedsaline pH 7.2. Dilute 0.1 mlof Complement-initiated haemolysis. To measure haemolysis,
tannic acid solution to 7.5 ml with phosphate-bufferedsaline add 600 gl of albumin barbital buffer solution wanned to 37°
pH 7.2 (final concentration 1.3 mg perlitre). Mix 1 volume of to the test sample, re-suspend the cells carefully by repeated
the freshly prepared dilution with 1 volume of human red pipetting (not fewer than 5 times) and place the cuvette in the
blood cell suspension and incubate at 37° for 10 minutes. thermostatted cuvette holder of a spectrophotometer. After 2
Collect the cells by centrifugation (400 to 800 g for 10 minutes), minutes, add 200 III of diluted guinea-pig complement (125
discard the supernatant and wash the cells once with to 200 CHso/ml), mix thoroughly by pipetting twice and start
phosphate-buffered saline pH 7.2. Resuspend the tanned immediately after the second pipetting the time-dependent
cells at 1.0 per cent v/v in phosphate-buffered saline pH 7.2. recording of absorbance at 541 nm, using albumin barbital
buffer solution as the compensation liquid. Stop the
Antigen coating of tanned human red blood cells. Take a
measurement if absorbance as a function of time has clearly
suitable volume (Vs) of tanned cells, add 0.2 ml of rubella
passed the inflexion point.
antigen per 1.0 ml of tanned cells and incubate at 37° for 30
minutes Collect the cells by centrifugation (400 to 800 g for 10 Evaluation. Determine the slope (8) of the haemolysis curve
minutes) and discard the supernatant, leaving a volume of at the approximate inflexion point by segmenting the steepest
200 gl. Add a volume of albumin barbital buffer solution section in suitable time intervals At (for example, At = 1minute)
equivalent to the discarded supernatant, resuspend and collect
and calculate S between adjacent intersection points,
the cells as described and repeat the washing procedure.
Make.upJheremaining200g1Jothree::quartersofV".thereby <::xp!_e~~~d_~~_.&tE~~_Il1!l111!eJ:T~eJ.!~~~eJ~t."-ll}ll_e. .t'.o~!! seJrveJs.ll:~
ob4tiD4Ig tl1<;l ipitialYQIl1fQ<;l.( Vi),.MJx._9QQ.fllQfqlbUl11il1.QqrbjtClI ($cxp)·. IIJ,aA~ition, .~(lt(ll1:l1il1t::th(lll~~()!~lll1c:elltthe:~t~!!()f
buffer solution with 100 gl of Vi, which is thereby reduced to measurement (As) by extrapolating the curve, which is almost
the residual volume (V,), and determine the initial absorbance linear and parallel to the time axis within the first few minutes.
. at 541 nm (A). Dilute Vr by a factor equal to A using albumin Correct (Scxp) using the expression:

2580

www.webofpharma.com
IP 2010 HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE

Tests
pH (2.4.24). 4.0 to 7.4.
Dilute the preparation under examination with a 0.9 per cent
Calculate the arithmetic mean of the values of S' for each w/v solution of sodium chloride to obtain a solution
preparation. Calculate the index ofFc function (he) from the containing 1.0 per cent of protein.
expression: Osmolality (2.4.23). Minimum 240 mosmol per kg.
Total protein. Minimum 3.0 per cent w/v and between 90.0 to
100 x (Si -
S' e )
110.0 per cent of the quantity of protein stated on the label.
I - -=--'---,=---'-
Fe - S' -S'
s e Dilute the preparation under examination with a 0.9 per cent
solution of sodium chloride to obtain a solution containing
about 15 mg ofprotein in 2 ml. To 2.0 ml ofthis solution in a
S' = arithmetic mean ofthe corrected slope for the round-bottomed centrifuge tube add 2 ml of a 7.5 per cent
preparation under examination, w/v solution of sodium molybdate and 2 ml of a mixture of 1
S's = arithmetic mean of the corrected slope for the volume of nitrogen-free sulphuric acid and 30 volumes of
reference preparation, water. Shake, centrifuge for 5 minutes, decant the supernatant
S'c = arithmetic mean of the corrected slope for the liquid and allow the inverted tube to drain on filter paper.
complement control. Determine the nitrogen in the centrifugation residue. by the
method of sulphuric acid digestion (2.3.30) and calculate the
Calculate the index ofFc function for the preparation under content of protein by multiplying the result by 6.25.
examination; the value is not less than that stated in the leaflet Protein composition. Determine by zone electrophoresis
accompanying the reference preparation. (2.4.12).
Human normal immunoglobulin for intravenous administration Use strips of suitable cellulose acetate gel as the supporting
is prepared as a stabilised solution or as a freeze-dried medium and barbital buffer solution pH 8.6 as the electrolyte
preparation. A stabiliser may be added. In both cases the solution.
preparation is passed through a bacteria-retentive filter. The
Test solution. Dilute the preparation under examination with a
preparation may subsequently be freeze-dried and the
0.9 per cent w/v solution of sodium chloride to an
containers closed under vacuum or under an inert gas. No
antimicrobial preservative is added either during fractionation immunoglobulin concentration of3.0 per cent w/v.
or at the stage of the final bulk solution. Reference solution. Reconstitute human immunoglobulin for -
electrophoresis reference preparation and dilute with a 0.9
The stability of the preparation is demonstrated by suitable
per cent w/v solution of sodium chloride to- a protein
tests carried out during development studies.
concentration of 3.0 per cent w/v.
Description. The liquid preparation is clear or slightly
To a strip apply 4 III of the test solution as a 10 rom band or
opalescent and colourless or pale yellow. The freeze-dried
apply 0.4 III per mm ifa narrower strip is used. To another strip
preparation is a hygroscopic, white or slightly yellow powder
apply in the same manner the same volume of the reference
or solid friable mass.
solution. Apply a suitable electric field such that the albumin
For the freeze-dried preparation, reconstitute as stated on band ofnormal human serum applied on a control strip migrates
the label immediately before carrying out the identification at least 30 mm. Stain the strips with amido black lOB solution
and the tests, except those for solubility and water. for 5 minutes. Decolourise with a mixture of 10 volumes of
glacial acetic acid and 90 volumes of methanol so that the
Identification background is just free of colour. Develop the transparency
Examine by a suitable immunoelectrophoresis technique. Using of the strips with a mixture of 19 volumes of glacial acetic
antiserum to normal human serum, compare normal human acid and 81 volumes of methanol. Measure the absorbance
serum and the preparation under examination, both diluted to ofthe bands at 600 nm in an instrument having a linear response
contain 1.0 per cent w/v of protein. The main component of over the range of measurement. Calculate the result as the
the preparation under examination corresponds to the IgG mean of3 measurements of each strip.
component of normal human serum. The preparation under System suitability. In the electropherogram obtained with the
examination may show the presence of small quantities of reference preparation, the proportion ofprotein in the principal
other plasma proteins; ifhuman albumin has been added as a band is within the limits stated in the leaflet accompanying
stabiliser, it may be seen as a major component. the reference preparation.

2581

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE IP 2010

Results. In the electropherogram obtained with the test immunoglobulin) is incubated with a defined amount of
solution, not more than 5.0 per cent ofprotein has a mobility guinea-pig complement (20 CH so) and the remaining
different from that of the principal band. This limit is not complement is titrated; the anticomplementary activity is
applicable if albumin has been added to the preparation as a expressed as the percentage consumption of complement
stabiliser; for such· preparations, a test for protein composition relative to the complement control considered as 100.0 per
is carried out during manufacture before addition of the cent.
stabiliser.
The haemolytic unit of complement activity (CHso) is the
Molecular size. Determine by liquid chromatography (2.4.14). amount of complement that, in the given reaction conditions,
Test solution. Dilute the preparation under examination with a will produce the lysis of 2.5 x 108 out of a total of 5 x 108
0.9 per cent w/v solution of sodium chloride to obtain a optimally sensitised red blood cells.
concentration in the range of 0.4 to1.2 per cent w/v and
Magnesium and calcium stock solution. Prepare as stated
injection of 50 to 600 Jlg ofprotein are usually suitable.
earlier.
Reference solution. Dilute human immunoglobulin RS with a
0.9 per cent w/v solution of sodium chloride to the same Barbital buffer stock solution. Prepare as stated earlier.
protein concentration as the test solution. Gelatin solution. Dissolve 12.5 g ofgelatin in about 800 ml of
Chromatographic system water and heat to boiling in a water-bath. Cool to 20° and
- a stainless steel column 60 cmx 7.5 mmor30 cmx 7.8 mm dilute to 10 litres with water. Filter through a membrane filter
packed with hydrophilic silica, (pore size: 0.22 Jlm). Store at4°. Use clear solutions only.
mobile phase. dissolve 4.873 g ofdisodium hydrogen
Citrate solution. Dissolve 8.0 g of sodium citrate, 4.2 g of
phosphate dihydrate, 1.741 g of sodium dihydrogen
sodium chloride and 20.5 g of glucose in 750 ml of water.
phosphate monohydrate, 11.688 g of sodium chloride
Adjust to pH 6.1 using a 10.0 per cent w/v solution of citric
and 50 mg ofsodium azide in 1000 ml ofwater.
acid and dilute to 1,000 ml with water.
- flow rate. 0.5 ml per minute,
- spectrophotometer set at 280 urn. Gelatin barbital buffer solution. Add 4 volumes ofgelatin
In the chromatogram obtained with the reference solution, the solution to 1 volume of barbital buffer stock solution and
principal peak corresponds to IgG monomer and there is a mix. Adjust to pH 7.3, ifnecessary, using 1 M sodium hydroxide
peak corresponding to dimer with a relative retention to the or 1 M hydrochloric acid. Maintain at 4°. Prepare fresh
principal peak of about 0.85. Identify the peaks in the solutions daily.
chromatogram obtained with the test solution by comparison Stabilised sheep blood. Collect one volume of sheep blood
with the chromatogram obtained with the reference solution; into one volume ofcitrate solution and mix. Store at 4° for not
any peak with a retention time shorter than that of dimer less than 7 days and not more than 28 days. (Stabilised sheep
corresponds to polymers and aggregates. blood and sheep red blood cells are available from a number
Results. In the chromatogram obtained with the test solution: ofcommercial sources.)
a. relative retention: for monomer and dimer, the relative Haemolysin. Antiserum against sheep red blood cells prepared
retention to the corresponding peak in the chromatogram in rabbits.
obtained with the reference solution is 1 ± 02;
Guinea-pig complement. Prepare a pool of serum from the
b. peak area: the sum ofthe peak areas ofmonomer and dimer blood of not fewer than ten guinea-pigs. Separate the serum
represent not less than 90.0 per cent of the total area of the from the clotted blood by centrifugation at about 4 0 • Store the
chromatogram and the sum of the peak area of polymers and serum in small amounts below -70°.
aggregates represents not more than 3.0 per cent of the total
area ofthe chromatogram. This requirement does not apply to Method
products where albumin has been added as a stabiliser; for
products stabilised with albumin, a test for distribution of Preparation ofstandardised 5 per cent sheep red blood cell
molecular size is carried out during manufacture before suspension. Separate sheep red blood cells by centrifuging
addition ofthe stabiliser. an appropriate volume of stabilised sheep blood and wash
the cells at least three times with gelatin barbital buffer
Anticomplementary activity. The consumption ofcomplement
sQlutiQnand_prepare..as_a_5.0_pJ~LC_enLyLv_s.usp_ensiolljnths<
isnof greafeffhan 5U:OperceriI(rCH;07mgofiIDiliilii6g1615U1ill):
s.a.mesgMi<:m,.Ml'laslll"l'lJlle. c:t)n<il'll1sjty()fJI1f:Sll~Pf:l1si()l1a~
TesHorantlcomplementary'actlViijofii:i:lmiinoglobullii follows: add 0.2 to 2.8 ml of water and centrifuge the lysed
For the measurement ofanticomplementary activity (ACA) of solution for 5 minutes at 1,000 g; the cell density is suitable if
immunoglobulin, a defined amount oftest material (10 mg of the absorbance (2.4.7) ofthe supernatant liquid at 541 urn is

2582

www.webofpharma.com
IP 2010 HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE

0.62 ± 0.01. Correct the cell density by adding gelatin barbital maximum degree ofhaemolysis is not in this range, repeat the
buffer solution according to the fonnula: titration with more or less diluted complement solution.

v _ xA . Table-l
f - 0.62 Required Prepared using
dilution of
Vr = final adjusted volume,
haemolysin
V; = initial volume, Gelatin barbital buffer solution Haemolysin
A = absorbance of the original suspension at 541 nm. Volume Dilution Volume
The adjusted suspension contains about 1 x 109 cells per m!. ml (l : ... ) ml

Haemolysin titration 7.5 0.65 undiluted 0.1


Prepare haemolysin dilutions as shown in Table 1. 10 0.90 undiluted 0.1
75 1.80 7.5 0.2
Add 1.0 ml of 5.0 per cent sheep red blood cell suspension to
each tube of the haemolysin dilution series, starting at the 100 1.80 10 0.2
1:75 dilution, and mix. Incubate at 37° for 30 minutes. 150 1.00 75 1.0
Transfer 0.2 ml of each of these incubated mixtures to new 200 1.00 100 1.0
tubes and add 1.10 ml of gelatin barbital buffer solution and 300 1.00 150 1.0
0.2 ml ofdiluted guinea-pig complement (for example, 1: 150). 400 1.00 200 1.0
Perform this in duplicate. 600 1.00 300 1.0
As the unhaemolysed cell control, prepare three tubes with 800 1.00 400 1.0
1.4 ml of gelatin barbital buffer solution and 0.1 ml of 5.0 per 1200 1.00 600 1.0
cent sheep red blood cell suspension. 1600 1.00 800 1.0
As the fully haemolysed control, prepare three tubes with 1.4 2400 1.00 1200 1.0
ml of water and 0.1 ml of 5.0 per cent sheep red cell 3200* 1.00 1600 1.0
suspension.
4800* 1.00 2400 1.0
Incubate all tubes at 37° for 60 minutes and centrifuge at 1,000
g for 5 minutes. Measure the absorbance (2.4.7) of the * discard 1.0 ml ofthe mixture
supernatants at 541 urn and calculate the percentage degree
of haemolysis in each tube using the expression: Preparation of optimised sensitised sheep red blood cells
(haemolytic system)

Prepare an appropriate volume of diluted haemolysin


containing 2 MHU/ml and an equal volume of standardised
Aa absorbance oftubes with haemolysin dilution, 5.0 per cent sheep red blood cell suspension. Add the
haemolysin dilution to the standardised cell suspension and
Ab mean absorbance of the three tubes with full
mix. Incubate at 37° for 15 minutes, store at 2° to 8° and use
haemolysis,
within 6 hours.
AI mean absorbance of the three tubes with no
haemolysis. Titration ofcomplement. Prepare an appropriate dilution of
Plot the percentage degree of haemolysis as the ordinate complement (for example, 1:250) with gelatin barbital buffer
against the corresponding reciprocal value ofthe haemolysin solution and perform the titration in duplicate as shown in
dilution as the abscissa on linear graph paper. Determine the Table 2.
optimal dilution of the haemolysin from the graph by Add 0.2 ml of sensitised sheep red blood cells to each tube,
inspection. Select a dilution such that further increase in the mix well and incubate at 37° for 60 minutes. Cool the tubes in
amount of haemolysin does not cause appreciable change in an ice-bath and centrifuge at 1,000 g for 5 minutes. Measure
the degree ofhaemolysis. This dilution is defined as one minimal the absorbance of the supernatant liquid at 541 nm and
haemolytic unit (1 MHU) in 1.0 m!. The optimal haemolytic calculate the degree ofhaemolysis (Y) using the expression:
haemolysin dilution for preparation of sensitised sheep red
blood cells contains 2 MHU per m!.
The haemolysin titration is not valid unless the maximum
degree ofhaemolysis is 50.0 per cent to 70.0 per cent. If the

2583

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE IP 2010

Ac absorbance of tubes 1 to 12, Test for anticomplementary activity


Ab mean absorbance of tubes with 100 per cent Prepare a complement· dilution having 100 CHso per ml by
haemolysis, diluting titrated guinea-pig complement with gelatin barbital
AI mean. absorbance of cell controls with 0 per buffer solution. Ifnecessary, adjust the immunoglobulin under
cent haemolysis. examination to pH 7. Prepare incubation mixtures as follows
Plot Y/(l - Y) as the abscissa against the amount of diluted for an immunoglobulin containing 50 mg per ml:
complement in ml as the ordinate on log-log graph paper. Fit Table- 3
the best line to the points and determine the ordinate for the
50.0 per cent haemolytic complement dose where Y/(l - Y) = Immunoglobulin Complement
1.0. Calculate the activity in haemolytic units (CHso/ml) from under examination control
the expression: (in duplicate)

Immunoglobulin (50 mg per ml) O.2ml


Gelatin barbital buffer 0.6ml 0.8ml
Complement O.2ml O..2ml
Cd reciprocal value ofthe complement dilution,
Co volume of diluted complement in ml resulting Carry out the test on the immunoglobulin under examination
in 50.0 per cent haemolysis, and prepare ACA negative and positive controls using human
5 = scaling factor to take account ofthe number of immunoglobulin RS, as indicated in the leaflet accompanying
red blood cells. the reference preparation. Higher or lower volumes of sample
and of gelatin barbital buffer solution are added if the
The test is not valid unless the plot is a straight line between
immunoglobulin concentration varies from 50 mg per ml; for
15.0 per cent and 85.0 per cent haemolysis and the slope is
example, 0047 ml of gelatin barbital buffer solution is added
0.15 to 0040, and preferably 0.18 to 0.30.
to 0.33 ml ofimmunoglobulin containing 30 mg per ml to give
Table - 2 0.8 ml. Close the tubes and incubate at 37° for 60 minutes.
Add 0.2 ml of each incubation mixture to 9.8 ml of gelatin
Tube Number Volume ofdiluted Volume ofgelatin
barbital buffer solution to dilute the complement. Perform
complement in ml barbital buffer
complement titrations as described above on each tube to
(for example 1:250) solution in ml
determine the remaining complement activity (Table 2).
1 0.1 1.2 Calculate the anticomplementary activity of the preparation
under examination relative to the complement control
2 0.2 1.1
considered as 100.0 per cent, from the expression:
3 0.3 1.0
4 0.4 0.9 a - b xlOO
a
5 0.5 0.8 a = mean complement activity (CHso/ml) of
6 0.6 0.7 complement control,
7 0.7 0.6 b complement activity (CHso/ml) oftested sample.
The test is not valid unless:
8 0.8 0.5
a. the anticomplementary activities found for ACA negative
9 0.9 004
control and ACA positive control are within the limits stated
10 1.0 0.3 in the leaflet accompanying the reference preparation,
11 1.1 0.2 b. the complement activity ofthe complement control (a) is in
the range 80 to 120 CH so perml.
12 1.2 0.1
Prekallikrein activator. Maximum 35 IV per ml, calculated
Three tube as cell 1.3 with reference to a dilution ofthe preparation under examination
c0 l11:r01 11t Qperc;ent containing.3.O_per centw/v ofimmunoglobulin.
haemolysis
......... .,.• .. __ _--_
__ --_ .•.... .
Testforprekllllilrreifillctivator
Three tube at 100 1.3 ml of water
per cent haemolysis Prekallikrein activator (PKA) activates prekallikrein to kallikrein
and may be assayed by its ability to cleave a chromophore

2584

www.webofpharma.com
IP 2010 HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE

from a synthetic peptide substrate so that the rate of cleavage Carry out all procedures from the beginning of the
. can be measured spectrophotometrically and the chromatography to freezing in portions during a single working
concentration of PKA calculated by comparison with a day.
reference preparation calibrated in International Units.
Method. The assay may be carried out using an automated
The International Unit is the activity ofa stated amount ofthe enzyme analyser or a suitable microtitre plate system allowing
International Standard which consists of freeze-dried kinetic measurements, with appropriate software for calculation
prekallikrein activator. The equivalence in International Units ofresults. Standards, samples and prekallikrein substrate may
of the International Standard is stated by the World Health be diluted as necessary using buffer B.
Organisation.
Incubate diluted standards or samples with prekallikrein
Reagents. Prekallikrein activator in albumin RS is calibrated substrate for 10 minutes such that the volume ofthe undiluted
in International Units by comparison with the International sample does not exceed 1/10 of the total volume of the
Standard. incubation mixture to avoid errors caused by variation in ionic
strength and pH in the incubation mixture. Incubate the
Buffer A. Dissolve 6.055 g of tris(hydroxymethyl)-
aminomethane, 1.17 g of sodium chloride, 50 mg of mixture or a part thereof with at least an equal volume of a
solution ofa suitable synthetic chromogenic substrate, known
hexadimethrine bromide and 0.100 g ofsodium azide in water.
to be specific for kallikrein (for example, N-benzoyl-L-prolyl-
Adjust to pH 8.0 with 2 M hydrochloric acid and dilute to
L-phenylalanyl~L-arginine 4-nitroanilide acetate or D-
1000 ml with water.
pro ly 1- L-p h eny Ia Iany 1- L-argi n in e- 4 -n itroan iiide-
Buffer B. Dissolve 6.055 g of tris(hydroxymethyl)- dihydrochloride), dissolved in buffer B. Record the rate of
aminomethane and 8.77 g of sodium chloride in water. Adjust change in absorbance per minute for 2 to 10 minutes at the
to pH 8.0 with 2 M hydrochloric acid and dilute to 1,000 ml wavelength specific for the substrate used. Prepare a blank
with water. for each mixture ofsample or standard using buffer B instead
Preparation of prekallikrein substrate ofprekallikrein substrate.

To avoid coagulation activation, blood or plasma used for Depending on the method used, ~ A per minutes has to be
the preparation of prekallikrein must come into contact corrected by subtracting the value obtained for the
only with plastics or silicone-treated glass sUifaces. corresponding blank without the prekallikrein substrate. The
Draw 9 volumes ofhuman blood into 1volume ofanticoagulant results may be calculated using a standard curve, a parallel-
solution (ACD, CPD or 3.8 per cent w/v solution of sodium line or a slope ratio assay or any other suitable statistical
citrate) to which 1 mg per ml of hexadimethrine bromide has method. Plot a calibration curve using the values thus obtained
been added. Centrifuge the mixture at 3,600 g for 5 minutes. for the reference preparation and the respective
Separate the plasma and centrifuge again at 6,000 g for 20 concentrations; use the curve to determine the PKA activity
minutes to sediment platelets. Separate the platelet-poor plasma ofthe preparation under examiantion.
and dialyse against 10 volumes ofbuffer A for 20 hours. Apply Anti-A and anti-B haemagglutinins. Carry out the tests for
the dialysed plasma to a chromatography column containing anti-A and anti-B haemagglutinins as stated under Dried
agarose-DEAE for ion exchange chromatography which has human haemophilic fraction. If the preparation under
been equilibrated in buffer A and is equal to twice the volume examination contains more than 3.0 per cent of
ofthe plasma. Elute from the column with bufferA at20 ml per immunoglobulin, dilute to this concentration before preparing
cm 2 per hours. Collect the eluate in fractions and record the the dilutions to be used in the test. The 1 to 64 dilutions do
absorbance (2.4.7) at 280 nm. Pool the fractions containing not show agglutination.
the first protein peak so that the volume of the pool is about
1.2 times the volume ofthe platelet-poor plasma. Anti-D antibodies. It complies with the test for anti7D
antibodies in human immunoglobulin for intravenous
\
Test the substrate pool for absence of kallikrein activity by administration. .
mixing I part with 20 parts of the pre-warmed chromogenic
Test for anti-D antibodies in human immunoglobulin for
substrate solution to be used in the assay and incubate at 37°
intravenous administration '
for 2 minutes. The substrate is suitable if the increase in
absorbance is less than 0.001 per minute. Add to the pooled Materials
solution 0.7 per cent w/v ofsodium chloride and filter using a
membrane filter (porosity 0.45 /lm). Freeze the filtrate in portions Phosphate-buffered saline (PBS). Dissolve 8.0 g ofsodium
and store at _25°; the substrate may be freeze-dried before chloride, 0.76 g of anhydrous disodium hydrogen phosphate,
storage. 0.2 g ofpotassium chloride, 0.2 g of potassium dihydrogen

2585

www.webofpharma.com
HUMAN NORMAL IMMUNOGLOBULIN FOR INTRAVENOUS USE IP 2010

phosphate and 0.2 g of sodium azide in water and dilute to of the well indicates a positive result. A stream of cells
1,000 ml with the same solvent. represents a negative result.
Papain solution. Use serological grade papain from a Record the endpoint titre as the reciprocal of the highest
commercial source, the activity of which has been validated. dilution that gives rise to a positive result.
Red blood cells. Use pooled red blood cells from not fewer The titre of the preparation under examination is not greater
than 3 donors of group ORzRz and 3 donors of group Orr than the titre of the reference preparation.
respectively. Wash the cells 4 times with PBS or until the Water. Determine by semi-micro determination ofwater (2.3.43),
supematant is clear. Centrifuge the cells at 1, 800 g for 5 minutes loss on drying (2.4.19) or near infrared spectrophotometry
to pack. Treat the packed red cells with papain solution (2.4.6), the water content is within the limits approved by the
according to the manufacturer's instructions. Store in competent authority.
Alsever's solution for not more than 1 week.
Sterility (2.2.11). Complies with the test for sterility.
Microtitre plates. Use V-bottomed rigid micro-titre plates. Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
Reference standards. Immunoglobulin (anti-D antibodies per kg of the rabbit's mass a volume equivalent to 0.5 g of
test) reference preparation and Immunoglobulin (anti-D immunoglobulin but not more than 10 ml per kg of body mass.
antibodies test negative control) reference preparation are
Antibody to hepatitis B surface antigen
suitable for use as the reference preparation and negative
control, respectively. Minimum 0.5 ill per g of immunoglobulin, determined by a
suitable immunochemical method.
Method
Storage. For the liquid preparation, store in a colourless glass
Reference preparation and negative control solutions. container, protected from light, at the temperature stated on
Reconstitute the reference preparation and the negative the label. For the freeze-dried preparation, store in an airtight
control according to instructions. Dilute the reconstituted colourless glass container, protected from light, at a
preparations with an equal volume of PBS containing 0.2 per temperature not exceeding 25°.
cent w/v of bovine albumin and then prepare a further 7 serial Labelling. The label states (1) for liquid preparations, the
two-fold dilutions using PBS containing 0.2 per cent w/v of volume of the preparation in the container and the protein
bovine albumin to give a total dilution range from 1/2 to content expressed in grams per litre; (2) for freeze-dried
1/256. Make 2 independent sets of dilutions for each preparations, the quantity of protein in the container; (3) the
preparation. Add 20 III ofeach dilution to the microtitre plate. amount of immunoglobulin in the container; (4) the route of
Test solutions. Initially dilute the test samples to give a starting administration; (5) for freeze-dried preparations, the name or
immunoglobulin G (IgG) concentration of 2.5 per cent w/v composition and the volume ofthe reconstituting liquid to be
using PBS containing 0.2 per cent w/v of bovine albumin and added; (6) the distribution of subclasses of immunoglobulin
then prepare a further 7 serial two-fold dilutions using PBS G present in the preparation; (7) where applicable, the amount
containing 0.2 per cent w/v of bovine albumin to give a total of albumin added as a stabilizer; (8) the maximum content of
dilution range from 1/2 to 1/256. Make 2 independent sets of immunoglobulin A.
dilutions for each test sample. Add 20 III of each dilution to
the microtitre plate.
Plasma for Fractionation
Prepare 3.0 per cent v/v suspensions of papain-treated D-
positive (ORzR z) and D-negative (Orr) red cells in PBS Human Plasma for Fractionation
containing 0.2 per cent w/v of bovine albumin. Add 20 III of Human Plasma for Fractionation is the liquid part of human
D-positive cells to one dilution series ofeach ofthe test sample, blood remaining after separation ofthe cellular elements from
the reference preparation and the negative control, and 20 III blood collected in a receptacle containing an anticoagulant,
of D-negative cells to the other dilution series of each of the or separated by continuous filtration or centrifugation of
test samples, the reference preparation and the negative anticoagulated blood in an apheresis procedure; it is intended
control. Mix by shaking the plate on a shaker for 10 seconds. for the manufacture of plasma-derived products.
Centrifuge the plate at 80 g for 1 minutes to pellet the cells.
Place the plate at an angle ofapproximately 70°. Read after 4 to
mInutes {or.untli.tl1eceIIs·-Iiave··strealnedlntile weIls Donors
containing the negative control and the wells where the Only a carefully selected, healthy donor who, as far as can be
D-negative cells have been added). A cell button at the bottom ascertained after medical examination, laboratory blood tests

2586

www.webofpharma.com
IP 2010 PLASMA FOR FRACTIONATION

and a study of the donor's medical history, is free from If 2 or more units are pooled prior to freezing, the operations
detectable agents ofinfection transmissible by plasma-derived are carried out using sterile connecting devices or under
products may be used. aseptic conditions and using containers that have not
previously been used.
Immunisation ofdonors
When obtained by plasmapheresis, plasma intended for the
Immunisation of donors to obtain immunoglobulins with recovery of proteins that are labile in plasma is frozen by
specific activities may be carried out when sufficient supplies cooling rapidly in a chamber at _30 0 or below as soon as
of material of suitable quality can not be obtained from possible and at the latest within 24 hours of collection.
naturally immunised donors. Recommendations for such When obtained from whole blood, plasma intended for the
immunisations are formulated by the World Health recovery of proteins that are labile in plasma is separated
Organisation (Requirements for the collection, processing from cellular elements and is frozen by cooling rapidly in a
and quality control ofblood, blood components and plasma chamber at -30 0 or below as soon as possible and at the latest
derivatives, WHO Technical Report Series, No. 840, 1994 or within 24 hours of collection.
subsequent revision).
When obtained from whole blood, plasma intended solely for
Records the recovery of proteins that are not labile in plasma is
separated from cellular elements and frozen in a chamber at -
Records ofdonors and donations made are kept in such a way
20 0 or below as soon as possible and at the latest within 72
that, while maintaining the required degree of confidentiality
hours of collection.
concerning the donor's identity, the origin of each donation
in a plasma pool and the results of the corresponding It is not intended that the determination oftotal protein and
acceptance procedures and laboratory tests can be traced. factor VIII shown below be carried out on each unit ofplasma.
They are rather given as guidelines for good manufacturing
Laboratory tests practice, the test for factor VIII being relevant for plasma
Laboratory tests are carried out for each donation to detect intendedfor use in the preparation ofconcentrates oflabile
the following viral markers: proteins.

1. Antibodies against human immunodeficiency virus 1(anti- The total protein content ofa unit ofplasma depends on the
HIV-l), serum protein content ofthe donor and the degree ofdilution
inherent in the donation procedure. When plasma is obtained
2. Antibodies against human immunodeficiency virus 2 (anti- fi-om a suitable donor and using the intended proportion of
HIV-2), anticoagulant solution, a total protein content complying
3. Antibodies against hepatitis C virus (anti-HCV), with the limit of5 per cent is obtained. Ifa volume ofblood or
plasma smaller than intended is collected into the
4. Hepatitis B surface antigen (HBsAg). anticoagulant solution, the resulting plasma is not
Pending complete harmonisation of the laboratory tests to be necessarily unsuitable for poolingfor fractionation. The aim
carried out, the competent authority may require that a test for of good manufacturing practice must be to achieve the
alanine aminotransferase (ALT) also be carried out. prescribed limit for all normal donations.
The test methods used are of suitable sensitivity and Preservation offactor VIII in the donatio,n depends on the
specificity and comply with the regulations in force. Ifa repeat- collection procedure and the subsequent handling of the
reactive result is found in any of these tests, the donation is blood andplasma. With goodpractice, O. 71U1ml can usually
not accepted. be achieved, but units ofplasma with a lower activity may
still be suitable for use in the production of coagulation
Individual plasma units factor concentrates. The aim ofgood manufacturingpractice
is to conserve labile proteins as much as possible.
The plasma is prepared by a method that removes cells and
cell debris as completely as possible. Whether prepared from Total protein. Carry out the test using a pool ofnot less than
whole blood or by plasmapheresis, the plasma is separated 10 units. Dilute the pool with a 0.9 per cent solution of sodium
from the cells by a method designed to prevent the introduction chloride to obtain a solution containing about 15 mg ofprotein
of micro-organisms. No antibacterial or antifungal agent is in 2 ml. To 2.0 ml ofthis solution in a round-bottomed centrifuge
added to the plasma. The containers comply with the tube add 2 ml of a 7.5 per cent solution of sodium molybdate
requirements for plastic containers for blood and blood and 2 ml of a mixture of 1 volume of nitrogen-free sulphuric
components (6.2). The containers are closed so as to prevent acid and 30 volumes of water. Shake, centrifuge for 5 minutes,
any possibility of contamination. decant the supernatant liquid and allow the inverted tube to

2587

www.webofpharma.com
PLASMA FOR FRACTIONATION IP 2010

drain on filterpaper. Determine the nitrogen in the residue by Production


the method of sulphuric acid digestion (2.3.30) and calculate
the protein.content by multiplying the quantity ofnitrogen by Platelet Concentrate is prepared from units. of whole blood
6.25. The total protein content is not less than 5.0 per cent. that have not been allowed to cool below 20°. Platelet rich
plasma (PRP) is separated within 4-6 hours after completion
FactorVID of the phlebotomy. The final component should contain
resuspended platelets in an amount of plasma adequate to
Carry out the test using a pool ofnot less than 10 units. Thaw maintain an acceptable pH - generally 40 to 70 ml is used.
the samples under examination, ifnecessary, at 37°. Determine
the assay of factor VIII (2.8.7), using a reference plasma Tests
calibrated against the International Standard for human
coagulation factor VIII in plasma. The activity is not less than Four PC per month should be assayed for pH and for platelet,
0.7IUperml. RBC and leucocyte counts.

Pooled plasma PRP


During the manufacture of plasma products, the first Counts 3.0 - 4.5 x 105per JlI (approximately)
homogeneous pool ofplasma (for example, after removal of
Volume 170 - 250 ml (approximately)
cryoprecipitate) is tested for HBsAg, HCV antibodies and for
Total Count 9 x 10 10 per bag (approximately)
HIV antibodies using test methods of suitable sensitivity and
specificity; the pool must give negative results in these tests. PRC
The plasma pool is also testedfor hepatitis C virus RNA using Counts 8 -10 x 105per Jll350 ml bag
a validated nucleic acid amplification technique (2.8.1). A (approximately)
positive control with 100 IU per ml of hepatitis C virus RNA
10 -12 x 105per Jll450ml bag
and, to test for inhibitors, an internal control prepared by
(approximately)
addition of a suitable marker toa sample of the plasma pool
are included in the test. The test is invalid if the positive Volume 40 - 50 ml per bag
control is non-reactive or ifthe result obtained with the internal 10
Platelet counts> 5.5 x 10 per bag in 75 per cent ofbags
control indicates the presence of inhibitors. The plasma pool (ifprepared fi:om 450 ml bags)
complies with the test ifit is found non-reactive for hepatitis C
virus RNA. > 4.2 X 10 10 per bag in 75 per cent of bags
(ifprepared from 350 ml bags)
Description. Before freezing, a clear to slightly turbid liquid
without visible signs of haemolysis; it may vary in colour Leucocyte count < 0.12 x 109 per bag.
from light yellow to green. RBC counts < 1.2 x 109 per bag.
Storage. Frozen plasma is stored in conditions designed to pH < 6.3 at the end of 5 days storage.
maintain the temperature at or below -20°; for accidental
reasons, the storage temperature may rise above -20° on one Expiration time. The expiration time is not more than 72 hours
or more occasions during storage but the plasma is from the time of collection ofthe source material.
nevertheless considered suitable for fractionation if all the Storage. Store at 20° to 22° in polyvinylchloride plastic bags.
following conditions are fulfilled (1) the total period of time Preserve at the temperature relevant to the volume of
during which the temperature exceeds -20° does not exceed 72 resuspension plasma, either between 20° and 22° or between
hours; (2) the temperature does not exceed -15° on more than I ° and 6°, the latter except during shipment, when the
one occasion; (3) the temperature at no time exceeds _5°. temperature may be between 1° and 10°.
Labelling. The label enables each individual unit to be traced Labelling. In addition to the labelling requirements ofWhole
to a specific donor. Blood applicable to this product, label it to state the volume of
original plasma present, the kind and volume ofanticoagulant
solution present in the original plasma, the blood group
designation ofthe source blood, and the hour ofexpiration on
Platelet Concentrate th~~till~(L~pjIl!tiQ!1gl:lt~,Wb£:r£:.ll:lJ?~~U~MQr§tQ@g~.l:ltlQo
ill
~~o, .11l~~1 ital~()to state t~llta c:ontinu()lls gentleagitlltioll
PlafeIetsseparated from wholehlood withiri4 to 6 hollis of shall be maintained, or where labelled for storage at I° to 6°, to
collection and suspended in 40 to 50 ml of plasma are state that such agitation is optional. Label it also with the type
designated as platelet concentrate. and result of a serologic test for syphilis, or to indicate that it

2588

www.webofpharma.com
IP 2010 WHOLE HUMAN BLOOD

was nonreactive in such test; with the type and result of a test The blood is drawn aseptically through a closed system into
for hepatitis B. surface antigen, or to indicate that it was a suitable sterile container containing a specific amount of
nonreactive in such test; with a warning that it is to be used as Anticoagulant Citrate Dextrose Solution (ACD Solution) or
soon as possible but not more than 6 hours after entering the Anticoagulant Citrate Phosphate Dextrose Solution (CPD
container; to state that a filter is to be used in the administration Solution) which is placed before the container is sterilised.
equipment; and to state that the instruction circular provided The quantity of anticoagulant solution should not exceed
is to be consulted for directions for use. 22.0 per cent v/v of the final volume of the mixture. No
antimicrobial preservative is added.
During the withdrawal ofblood the container is gently agitated
to ensure thorough mixing. When withdrawal is complete the
Whole Human Blood container is immediately sealed and cooled to 2° to 8°. It is not
Whole Blood (Human) opened until immediately before transfusion. With every
container of blood, a separate sample mixed with the
Whole Human Blood is blood drawn aseptically from selected
appropriate quantity of anticoagulant solution, is collected
human donors and mixed with a suitable anticoagulant.
for compatibility and other tests; this small container is firmly
Whole Human Blood is the final mixture of blood and attached to the main container.
anticoagulant solution contains not less than 9.7 per cent
Whole Human Blood in containers from which samples have
w/v ofhaemoglobin, calculated from the haemoglobin content
been removed for tests should not be used for transfusion.
ofthe donor's blood and the dilution due to the anticoagulant
Consequently, it is not intended that the Tests and the Assay
solution. It is obtained from healthy donors who must:
should be carried out on the contents of the container. The
(a) be in the age range of 18 to 60 years and be in good health Blood Banle or the service collecting the blood is responsible
as indicated in part by normal temperature and blood for ensuring that the conditions in which the blood is collected
pressure within normal limits; and stored are such that, if and when tested, the blood will
(b) not be pregnant, if females; comply with the requirements ofthe monograph.

(c) not have undergone major surgery within 6 months of Blood group. Determine the blood group (in the sample
donation; accompanying each donation) under the ABO system (2.8.11),
by examination of both corpuscles and serum, and under the
(d) as far as can be ascertained after clinical and laboratory
Rh system (2.8.11), by examination ofthe corpuscles.
examination and the study ofmedical history ofthe donor
be free from disease transmissible by blood transfusion; Description. Deep red fluid which, on standing separates into
a lower layer of sedimented-red cells and a yellowish, almost
(e) have blood containing not less than 12.5 per cent w/v of
clear upper layer of plasma, free from visible signs of
haemoglobin;
haemolysis, with a greyish layer between the two consisting
(f) be free from acute respiratory diseases; ofleucocytes and thrombocytes. A layer containing emulsified
(g) be free from any infectious skin disease at the site of fat may form on the surface.
phlebotomy;
Tests
(h) have no history of malarial fever within 12 months of
donation; Sterility (2.2.11). Complies with the tests for sterility,
determined by Method B.
(i) have no history of viral hepatitis or of close contact with
an individual having viral hepatitis within 12 months of Assay. Detennine the haemoglobin content by photometric
donation and have blood that has given negative results haemoglobinometry (2.8.12).
in tests for the presence of hepatitis-B antigen;
Storage. Store in colourless, transparent and sterile containers
G) have blood that has been tested with negative results
into which it was originally drawn. The containers should be
for evidence ofsyphilitic infection, HCV antibodies, HIV
provided with a hermetic, contamination-proofclosure. Store
antibodies and malarial parasites.
at a temperature between 2°to 8°.
The examinations and tests to be carried out are decided by
Labelling. The label states (1) the distinctive code number by
the National Regulatory Authority.
reference to which the details of the donor are available; (2)
The frequency of donations of whole blood shall not exceed the ABO group with the approved colour scheme for different
once every 3 months with a maximum volume of 1.5 litres in groups as specified by the National Regulatory Authority; (3)
any consecutive 12 month period. the Rh group; (4) the total volume of fluid, the proportion of

2589

www.webofpharma.com
WHOLE HUMAN BLOOD IP 2010

blood, and the nature and volume of anticoagulant solution; group 0 whether haemolysins are present or not and if they
(5) the date on which the blood was withdrawn; (6) the expiry , are, that the blood must be administered only to recipients of
date which should not exceed 21 days from the date of blood group 0; (10) that the blood has given negative results
withdrawal of blood; (7) the storage conditions; (8) that the in the tests for the presence of malarial parasites, hepatitis-B
blood must not be used for transfusion if there is any visible antigen, syphilis and HIV antibodies and any other tests
evidence ofhaemolysis or other deterioration; (9) for blood of prescribed by the National Regulatory Authority.

2590

www.webofpharma.com
INDIAN PHARMACOPOEIA 2010 MONOGRAPHS

BIOTECHNOLOGY PRODUCTS

General Monograph
Biotechnology Derived Therapeutic Products .... 2593
Monographs
Erythropoietin Concentrated Solution 2602
Filgrastim Concentrated Solution 2610
Interferon Alfa-2 Concentrated Solution 2613
Streptokinase Bulle Solution 2617

2591

www.webofpharma.com
www.webofpharma.com
IP 2010 BIOTECHNOLOGY DERIvED THERAPEUTIC PRODUCTS

Biotechnology Derived Therapeutic the current edition of the Indian Pharmacopoeia does not
contain products produced by this process and hence no
Products further discussion on this aspect is included in this chapter
Biotechnology has brought about the discovery and MonoclonalAntibodies
development of a new class ofhuman therapeutics. Advances
in genetics, cellular and molecular biology have allowed Antibodies are proteins produced by differentiated b-
scientists to identitY and develop a number of new products. lymphocytes. Kohler and Milstein first fused a normally short-
These products provide significant clinical benefits, and in lived antibody producing b-cell obtained from the spleen of
many cases, provide therapies where no effective treatment mice immunized with an antigen to a rapidly dividing cancer
existed. cell (mouse myeloma) to produce a cell line (hybridoma), which
was both immortal and a producer of antibodies that
Proteins are the worldlOrse molecules ofthe cell. Due to their recognized a single epitope on an antigen, hence, the name
biological significance, proteins can make excellent drug monoclonal antibodies (mAbs). Monoclonal antibodies can
candidates. In the living cell, genes, in the form of DNA, be produced in cell culture or in live animals. When the
provide the basic instructions in creating biologically hybridoma cells are injected into the peritoneal cavity ofmice,
functional proteins. Biotechnology is the process that they produce tumors containing an antibody-rich fluid called
produces protein molecules in sufficient quantities for ascites fluid. Production in cell culture is usually preferred
therapeutic use, which have very specific physiological roles since cell culture in fermentation chambers can be used to
that ah'eady exist in the human body albeit in limited quantities. produce antibodies on a larger scale.
Techniques of genetic engineering using bacteria, yeast and
Humanized monoclonal antibodies
eukaryotic cells in culture or cultured animal cells are employed
to incorporate in these cells the information needed to produce The standard procedure of producing monoclonal antibodies
a human protein with therapeutic potential. Once engineered, yields mouse antibodies. For therapeutic applications in
these cells can be grown in large quantities by fermentation or humans these proteins are unsuitable since the human immune
large-scale cultivation of animal or human cells. The addition system recognizes mouse antibodies as foreign, causing
ofgenetic material to cells gives the engineered proteins their systemic inflammatory effects and are removed rapidly from
name, recOl;nbinant proteins circulation. Application ofrecombinant DNA technology has
overcome this problem. The DNA that encodes the binding
Gene Cloning and Protein Expression portion of monoclonal mouse antibodies is ligated to the
human antibody producing DNA. Such a construct is then
clo~ed suitably in a vector and introduced into a mammalian
The basic principle involved in gene cloning includes the
expression system to produce these half-mouse and half-
insertion of a select fragment of DNA representing the gene
human antibodies. Depending on how big a part ofthe mouse
for a peptide/protein into an episomal circular DNA, called the
antibody is used,such proteins are eitherreferred to as Chimeric
vector, which transports the gene into a host cell. The vehicle
or humanized monoclonal antibodies. Monoclonal antibodies
containing the inserted gene then replicates along with the
hav:e been generated and approved to treat cancer,
host cell. The cellular machinery of the host cell transcribes
cardiovascular disease, inflamatory diseases, muscular
and translates the genes including the inserted gene into
degeneration, transplant rejection, viral infection and others.
proteins. The vector with an inserted foreign gene is called a
recombinant DNA molecule and the protein produced as a Therapeutic recombinant monoclonal antibodies
consequence of the inserted gene by the host cell is called
recombinant Protein. The commonly used vectors for Recombinant monoclonal antibodies for therapeutic
applications belong to the IgG class of immunoglobulins.
bacterial transformations are Plasmids, which are small circular
Immunoglobulins are made up of four polypeptide chains,
DNAs found in bacteria and other microorganisms. Other
vectors include viruses such as bacteriophages (for bacterial two light (A. and K) and two heavy chains (a, ~, 'Y, e, !J.). The
type of heavy chain determines the immunoglobulin isotype
hosts) or animal viruses for eukaryotic host cells (e.g., Epstein-
(IgA, IgD, IgG, IgE and IgM respectively). The four-
Barr virus, Simian Virus etc). Major advances have been made
polypeptide chains are cross-linked by disulfide bridges (-S-
in the past decade for efficient and stable expression offoreign
proteins in mammalian cells in culture. S- bonds). Light chains comprise 220 amino acid residues
whereas heavy chains are made of440-550 amino acids. Each
In addition, transgenic animals and plants have been produced chain has two distinct regions namely, "constant" and
which express foreign genes inserted into their genome for "variable", the former being proximal to the C-terminal region
tissue/organ specific expression. Although large-scale (111-220 or 440-550 residues) whereas the latteris towards the
production of therapeutic proteins has been accomplished, N- terminal sequence of the polypeptide (1-110). The amino

2593

www.webofpharma.com
BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS IP 2010

acids of the constant region of immunoglobulin are uniform paralleled by growing concerns about the safety ofthese novel
from one antibody to another within the same isotype (e.g., pharmaceuticals, which has resulted in the regulatory
IgG), while the variable region found in both. the light and authorities setting high standards for certification. One ofthe
heavy chains consist of different amino acids. These regions strategies used by researchers in this field involves sourcing
are referred to as hypervariable region or "Complementary new genetic elements for incorporation into expression
Determining Regions (CDRs)"representing antibody diversity, systems by systematically analyzing the rich natural diversity
each specific for the recognition of the antigen. An antibody ofmicroorganisms. There are, in addition, numerous tool~ for
molecule is generally represented as a "Y" shaped structure. modifying microorganisms and for re-engineering existing
The.arms of the Y structure are connected to the stem region biological pathways or processes to meet the needs of the
by a flexible hinge. IgG can be proteolytically cleaved by pharmaceutical industry. Although many prokaryotic hosts
partial digestion with papain, which results in three distinct are described in literature for the production of recombinant
~50 KD fragments: two identical fragments designated Fab proteins, E.coli is the most preferred host because its genetics
fragments and one Fc fragment. The Fab fragments are the are well understood. The popular cloning host has been E. coli
arms ofthe antibody containing the entire light (L) chain and BL21 and its derivative BL 21A.DE3. However there are some
the N-terminal half ofthe Heavy (H) chain. These fragments disadvantages such as the recombinant protein produced may
contain the IgG's antigen binding domains. The Fc fragment not have a proper secondary structure due to the non-formation
derives from the stem portion of the Y structure and contains of intra-molecular disulfide bonds. Many recombinant
the c-terminal halves of the two H chains. The Fc region is proteins tend to aggregate when over expressed in E.coli.
responsible for the activation of phagocytosis, complement E.coli proteins begin their sequence with an N-formyl
dependent cellular toxicity and antibody dependent cellular methionine, which may not be removed by its enzyme systems.
cytotoxicity (ADCC). Proteolytic enzymes of E.coli may partially degrade the
The IgG-Fc region is glycosylated atAsn 297, which is essential recombinant protein.
for the conformation of the antibody molecule that allows The principal element in recombinant protein expression
interaction with effector ligands. The IgG-Fc glycoform pattern technology is the recombinant plasmid, which contains the
of monoclonal chimeric and "humanized" recombinant gene that codes for the protein ofinterest. The gl:!ne expression
therapeutic antibodies produced in CHO or NSO cells show regulatory elements such as the promoter sequence in the
wide variation from clone to clone, which is dependent on the plasmid play a key role in the production of mRNA of the
process and cell culture conditions. Under certain conditions recombinant gene. The other important regulatory process
a number of abnormally glycosylated products that lack employed in gene expression is the application of gene
potency and potentially immunogenic products are produced induction. Induction is the phenomenon wherein the
(e.g., galactose-a (1,3)-galactose and N-glycolylneuraminic transcription of the gene is turned on by the addition of a
acid) which are not therapeutically acceptable. However, chemical to the growth medium. The most popular system
successive rounds of selection of cell lines overcome these used in academic research is the lac promoter, which is induced
problems. Another important variable in efficacy due to by Isopropyl thiogalactopyranoside (IPTG). However, in the
glycosylation is the ability of the IgG molecule to mediate industrial production of recombinant therapeutic proteins,
ADCC. This property is particularly important in antibody addition of chemical inducers is not desired, hence auto-
therapeutics for oncology indications. The ability to elicit inducible promoters which are up-regulated upon changes in
ADCC greatly varies with different glycoforms. Therefore, the culture medium or the growth phase of the bacteria or the
the cell lines have been engineered by the introduction of a regulation of cell cycle, are employed for improvement in
glycosyl transferase (GnTIII) and/or inactivation/deletion of production efficiency. Overproduction ofproteins most often
fucosyl transferase in the CHO cells, which has resulted in results in misfolded structures, which are insoluble and
products with 20 to 100-fold increase in ADCC. accumulate in the cytoplasm as "inclusion bodies". Cloning
and expressing a group of proteins collectively called
Processes for the production of recombinant therapeutic "Chaperones" overcome this problem. Some ofthe chaperones
proteins including mAbs whose mode ofaction is partially understood include, Gro ELI
Prokaryotes Gro ES, DnaK, Dnal and GrpE. Fusion ofthe gene of interest
with a partner gene results in a recombinant protein, which
The qualitative and quantitative demand for recombinant produces both gene products linked in tandem. Such fusions
proteins is. steadi!y'jl!fr_~l!§.i!!g. MQ1~ulllX.1:>!ologjst~_~~ also increase.expression levels as. well as provide ameansfor
constalltly challenged by. the. need to iIIlproveaIldoptiIIlize 1l.CQllVj:: l1 if)IJ,t . proces~ . oJ pll:rificatioll pyaffinity
the existing expression systems to meet the steadily increasing chromatography. A number offusion partner genes are reported
qualitative and quantitative demand for recombinant proteins. in literature but the most common partner proteins are maltose
The continuous evolution of novel expression systems is binding protein, glutathione-S-transferase and thioredoxin.

2594

www.webofpharma.com
IP 2010 BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS

For ease of recombinant protein purification, short sequence Mammalian Cell Lines
tags at N- or C-tenninal are genetically attached which result
in recombinant protein products with additional amino acid Animal Cell expression systems produce proteins, which have
sequences. These additional sequences have to be cleaved high degree ofsimilarity to human proteins with respect to the
from the final therapeutic product. Several methods for precise pattern and capacity of post translational modifications. The
excision of tag sequences have been described which include most commonly used cel1lines for the expression oftherapeutic
enzymatic and chemical processes. Another strategy employed proteins are as follows:
for enhancing protein expression is the use of "bicistronic" Chinese Hamster Ovary (CHO) Cell Line
vectors. In this strategy, a short sequence of a gene efficiently
Baby Hamster Kidney (BHK) Cell Line
expressed is linked as a non-translatable transcriptional fusion
to the gene of interest. Secretion of recombinant proteins or HEK Human Embryo Kidney (HEK) Cell Line
their translocation to periplasmic space of bacteria is a Human Retinal Embryonic Cell Line (pER C.6)
procedure employed in the expression of foreign proteins in
Mouse Myeloma Cel1 Line (NSO)
prokaryotes. This procedure requires the addition of signal
sequences from secretory pathways or leader sequences for Mammalian cel1lines produce therapeutic proteins with proper
transport to periplamic space. OmpA and PelB signal peptides folding, assembly and post-translational modifications, which
have been shown to efficiently transport recombinant proteins results in phannacological1y active products in tenns ofquality
to bacterial periplasm. A good example is the production of and reproducibility. However, there remains a possibility of
correctly folded full-length antibodies within the periplasm of transmission of viral or pyrogenic substances carried over in
E. coli. the final product. There is a trend now to use cell lines, which
are of human origin since they possess all mechanisms to
Kukaryotes produce products most similar if not identical to naturally
occurring proteins in the human body.
Yeasts
Immortalized cel1 lines transfected with foreign therapeutic
Similar to prokaryotes in growth kinetics achieving high
protein genes are used for the production of the respective
density, yeasts secrete large amounts ofrecombinant proteins.
proteins. There are basical1y two processes of immortalizing
Saccharomyces sp, Pichia pastoris, Hansenula polymorpha,
mammalian cells; (1) Viral transformation: Induction of
and Kluyveromyces lactis are some of the yeasts, which are
immortalization by inactivating the tumor suppressor genes
currently in use for the production oftherapeutic recombinant
ofmammalian cel1s. Viral genes including Epstein - Barr virus
proteins. Yeasts are the preferred eukaryotes for the
(EBV), Simian Virus 40 (SV 40) T antigen, adenovirus and
production of recombinant therapeutic proteins that cannot
human papillomavirus (HPV) can induce immortalization.
be produced from prokaryotes because ofincorrect folding or
Although the method is quite reliable, there are reports of
the requirement for glycosylation. Sacharomyces cerevisiae
aneuploidy and loss of some properties of the primary cells.
is genetically well characterized, its growth physiolQgy well
(2) Modification of telomere of somatic cells. Telomerase
understood and hence, it is the prevalent yeast species in
reverse transcriptase protein (TERT), which is inactive in most
phannaceutical production process. However, Pichia pastoris
somatic cells, when exogenously expressed, the cells are able
is also gaining ground as a preferred host for heterologous
to maintain telomere lengths sufficient to avoid replicative
protein expression because of its superior secretion efficiency
senescence thus, rendering the cells immortal. The latter
and high yields. Pichia pastoris and Hansenula polymorpha
method is preferred for therapeutic protein expression.
are methanol metabolizing yeasts (methylotrophic yeasts),
Analysis ofseveral telomere-immortalized cel1lines has verified
which contain strong promoters for the genes of methanol
that the cells maintain a stable genotype and retain critical
assimilating enzymes such as methanol oxidase and alcohol
phenotype markers.
oxidase. As the enzymes ofthese pathways account for more
than 30% of the total protein content of the organism, the Development ofa cell line stably expressing a desired protein
metabolic efficiency with respect to secreted protein is gene starts with the construction of expression vectors.
significantly higher. Although attempts are being made to Different expression vectors both viral and non-viral have
humanize N-linked glycosylation in the yeastPichiapastoris, been designed to transfer foreign genes into mammalian cells.
the employability of yeasts for the expression of human The choice of the vector is dependent on the host cell, the
therapeutic proteins might reach a limit if the product has a level of the desired product fonned and safety. Typically an
highly specific post-translational modification for its expression vector consists of (i) a constitutive or inducible
phannacological activity/properties. In such cases, mammalian promoter (ii) a transcription tenninator and (iii) a cassette with
cell expression systems are employed for the production of an origin of replication and a selection marker for vector
therapeutic proteins. production in bacteria. Strong constitutive promoters are of

2595

www.webofpharma.com
BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS IP 2010

viral genomes (e.g., human cytomegalovirus (HCMV), simian General Techniques used in down-stream processing of
virus 40 and Rous sarcoma virus). Amongst the non-viral recombina.nt proteins
promoters the promoter of human elongation factor-I-a (EF-
Ia) is similar in strength to the HCMV promoter. The desired The design ofthe purification process ofa recombinant protein
therapeutic protein gene is isolated as a cDNA, with one intron and its scale-up capabilities assumes significant importance
sequence, which is preferably located between the promoter since the regulatory authorities would approve the "product
and the coding sequence of the gene of interest and inserted by process". Thus, the approved process must be followed
into the vector at a suitable site. The vector containing the even ifnew developments in separation teclmologies provide
gene insert is then propogated in the bacterial host. The vector significant advantages.
is then recovered from the bacteria, linearized and introduced Liquid chromatography is the core of preparative protein
into the mammalian cell by transfection. There are many purification and all supplementary procedures like extraction,
reagents/methods to effectively mediate gene transfer. Calcium centrifugation, ultrafiltration, and dialysis serve to prepare
phosphate facilitated transfection, electroporation, lipofection, the protein solution for chromatography. A series of
micro iiDection, biolistic and polymer mediated transfer are chromatographic steps usually tenned as capture, intennediate
routinely used. For selection of recombinant cell, a second purification and polishing, make use of different intrinsic
gene is transferred that confers to the recipient cells a selective features of proteins, is usually required to achieve sufficient
advantage. The most common selector genes are dihydrofolate separation of the target from contaminants. Common modes
reductase (DHFR) and glutamine synthetase (OS). In both of chromatography include ion exchange chromatography,
cases, selection occurs in the absence of appropriate affinity chromatography, hydrophobic interaction
metabolite (hypoxanthine and thymidine in the case ofDHFR chromatography, gel filtration and reverse phase
and glutamine in the case of OS), thus preventing the growth chromatography. The selection of chromatographic
ofnon-transfonned cells. When the transfected foreign gene procedures and their arrangements in a suitable order are based
is integrated into a "good site" on the chromosome of the on the properties of the recombinant protein particularly its
host by recombination, the resultant cell line will stably express tolerance to organic solvents, its susceptibility to denaturation
the desired gene. Selection pressure results in several fold due to changes in pH, temperature, degradation and oxidation.
amplification of genes including the integrated genes when
high concentrations ofrespective inhibitors are used (for. e.g., In the case of secreted recombinant proteins, downstream
Methotrexate inhibition of DHFR and Methionine processing starts with the collection of the fermentation
sulphoximine inhibition ofOS). supernatant after separating the cell mass while if the
recombinant protein is intracellular, the process begins with
There are two main ways of culturing animal cells for the the extraction of soluble proteins from the cell mass. In the
production of recombInant proteins, viz., (a) Adherent cell fonner case the target protein may be highly dilute with large
culture and (b) Suspension culture. In adherent cell culture amounts of inorganic salts and media proteins such as fetal
technology the mammalian cells (e.g., CHO cells) are seeded calf serum, yeast extract and other growth promoting
into roller bottles that are filled to about 25 per cent capacity substances, which may limit the choice ofthe capture step. In
with the medium and slowly rotated allowing cells to adhere. the case of the target protein expressed as an intracellular
The rotation provides the continuous contact of the cells protein, the process of extraction from the cell mass is
with the medium and oxygen is supplied by the "head space" employed, which leads to a highly complex mixture of
in the bottle. Following attainment of confluency, the product recombinant and host biomolecules including host proteins,
is harvested from the decanted supernatant. The suspension nucleic acids, lipids, carbohydrates and other cell components.
culture method is more widely used for the mass production In addition to commonly employed technologies such as
ofrecombinant proteins, where the capacity ofthe mammalian continuous centrifugation, tangential flow filtration and fast
cells for single cell suspension growth is exploited for flow chromatography two novel techniques viz.; affinity tags
scalability to very large volumes. The transition of adherent and expanded bed adsorption techniques are gaining
cells to suspension cultivation requires a selection of media importance in the purification protocols for recombinant
fonnulations. The seeding inoculum begins with a small volume proteins. Affinity tags are short peptide sequences genetically
of cell suspension, which is gradually expanded so that fused to a target protein. A peptide ofsix consecutive histidine
sufficient cell numbers are generated for the final production residues (His-6-tag) is the most widely used tag peptide, which
~~ase. J~~~~p~nsi()~'<::l:llt.1:!!:~p~~~~~~!~"~~~~ll~g!gg is fused either at the N- or C-tenninal ofthe protein ofinterest.
composition of the cell medium during the production phase This tag
facllitateSthe punflcation ofthe proteIn by irnrrlObilized
can "iiffectiiie"quaHijo:i:' iiie~product.dueiodegradatfve metii!ioii{generiillYNiClcel)iiffiiiiiY·colmnii·(lMAC).·Forthis
enzymes released by the cells as well as the molecular technique, a metal chelating ligand, e.g., iminodiacetic acid or
composition of the product due to limitations of nutrients. nitrilotriacetic acid is immobilized on a chromatographic matrix

2596

www.webofpharma.com
IP 2010 BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS

and charged with transition metal ions leaving one or more xiI) Formation ofgamma carboxyglutamic acid
coordinating sites free for interaction with the analyte. Proteins xiii) Methylation
with the His-6-tag will bind to the ligand tightly to allow
xiv) Succinimide forms
quantitative capture of the target molecule from a complex
feed stream. The bound target protein is recovered by step- xv) Aspartate isomerization
gradient elution using imidazole or by acidification. A potential xvi) Disulfide linkage
problem is the presence of leached metal ions in the eluate, xvii) Oxidation
which must be removed in a subsequent step of purification.
Amongst these only a few are relevant from the perspective
His tag facilitated purification process has yielded therapeutic
oftherapeutic recombinant proteins. Glycosylation represents
recombinant proteins without affecting the folding, bioactivity
the most important PTM since a great diversity exists among
and biodistribution profiles.
different expression systems. For example, prokaryotic
Expanded bed adsorption (EBA) is another novel technique, expression systems result in aglycosylated proteins often
which is employed for preparative protein purification when resulting in insoluble inclusion bodies. Yeasts on the other
large volumes ofparticulate raw materials are involved in the hand, add carbohydrate side chains of high mannose content,
initial purification step. EBA combines the advantages ofbatch- CHO and murine cells can also add sugars not normally found
or fluidized bed methods with superior adsorption in human proteins some ofthem are known to be immunogenic.
characteristics of packed bed columns by a novel desi,gn of
Posttranslational modifications of recombinant therapeutic
matrix particles. With the exception ofgel filtration, all modes
proteins are significant in terms of potency, stability and
ofbiochromatography are compatible for EBA. However, ion-
toxicity including immunogenecity of the biopharmaceutical
exchange media are most extensively used.
product/so This emphasizes the need for inclusion in the
Post-Translational Modifications ofProteins pharmacopoeial monographs, meticulous description of
specifications and rigorous analytical data to characterize each
The primary structure ofproteins, which is the linear sequence biopharmaceutical product.
of amino acids, is encoded by the gene sequence of the cell.
The mere demonstration that variations in the post translational
However, proteins undergo a variety of modifications
modifications are found in recombinant glycoproteins
subsequent to their synthesis. Enzymes within the cell mediate
these modifications and such modifications are attributed to produced in different expression systems, does not
certain properties, such as solubility, transportability, automatically negate the biological efficacy or the safety of
the product. In many instances it has been clearly demonstrated
resistance to proteolysis, folding, receptor binding, signal
transduction etc., in the native milieu of the cell. However, ' through clinical trials that the products are safe and efficacious
(e.g. recombinant factor VIII produced by BHK cells and CHO
with the advent of production. of proteins for therapeutic
cells have different glycosylation profiles but hoth are safe
applications in heterologous expression systems, post-
and efficacious). Therefore, comparability ofthe same protein
translational modifications (PTMs) have assumed special
significance from the point of view of setting specifications produced by different processes has to be governed by a
pragmatic approach. However, the secretory pathway ofPichia
and quality assurance. Proteins undergo a broad range of
PTMs; the most common ofthem are listed below.
pastoris has been genetically re-engineered to perform
sequential glycosylation reactions that mimic early processing
Post-Translational Modifications of Proteins ofN-glycans in humans'
D Acylation
Glycosylation ofProteins
ii). Phosphorylation
iii) Sulphonation More than one third of the biopharmaceticals currently in
clinical use have an absolute requirement for glycosylation
iv) Proteolysis (terminal or domain deletion)
and hence they are produced in eukaryote expression systems.
v) Glycosylation (N-linked, O-linked) Oligosaccharides are attached co-translationally through
vi) Aggregation specific aspargine (N-linked) or serine or threonine residues (
vii) High order structural change (conformational change or O-linked). While the consensus sequence for N-glycosylation
denaturation) has been shown to be Asn-x-ser/Thr as an essential but not
viii) Arnidation or deamidation sufficient sequence, there is no such consensus sequence
Imown for O-glycosylation sites. In some cases ofrecombinant
ix) Carbamylation
proteins, glycosylation may not be essential for therapeutic
x) Carboxylation application, but as pharmaceutical products, glycosylated
Xl) Formylation preparations have been found to have lowered protein

2597

www.webofpharma.com
BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS IP 2010

aggregation and better thermal stability which are desired to proteolytic degradation, less immunogenic due to masking
properties. of epitopes and have reduced renal clearance rates. Early
procedures of PEgylation resulted in mixtures of PEG
Immunogenicity is yet another parameter, which is a matter of
substituted positional isomers. But later developments in the
concern with regard to recombinant therapeutic proteins,
chemistry of ligation which includes different coupling
produced in different expression systems.
techniques using different activated polyethylene glycols
'Y-Carboxylation and j3-hydroxylation ofProteins and introduction of linkers between PEG and the protein
targets, have resulted in site-specific PEgylation. Several
These are particularly found in proteins involved in the
PEGylated biopharmaceuticals have received regulatory
process of Blood Coagulation. An enzyme, carboxylase,
approvals, which include PEG-IFN, PEG-human growth
converts target glutamic acid residues in the protein to 'Y-
hormone and PEG-granulocyte macrophage colony-
carboxyglutamic acid residues, while hydroxylase enzyme
stimulating factor. In all cases PEGylation increased product
catalyses the conversion of either the target aspartic acid or
plasma half-life which resulted in reduced frequency of
the target aspargine residues to their corresponding~-hydroxy
administration.
derivatives (~-hydroxyaspartate and ~-hydroxyaspargine
respectively Proteins by nature are heterogeneous but in the context of
recombinant DNA derived protein products produced in
O-sulfonation various systems add to more complexities. In the chain of
Tyrosine O-sulfonation is a post-translational modification processes starting from cloning of the gene to the production
limited to a few proteins. Tyrosine O-sulfonation is catalyzed of therapeutic protein, there could be many products formed
by two enzymes, viz., tyrosylprotein sulfotransferases (TPST- such as truncated polypeptides, differently folded proteins,
I and TPST-2) that transfer the sulfuryl (-S03') group from scrambled disulfide linkages, varying extents and different
phosphoadenosylphosphosulphate to certain tyrosyl residues pattern of glycosylation. In addition, there could be process
in the protein. Tyrosine sulfation occurs in all mammalian cell related contaminants resulting in impurities in the fmal product.
lines but not in yeasts and prokaryotes. The functional Proteins have pleomorphic physiological effects. Thus,
importance oftyrosine sulfation is presumed to be in protein- although the recombinant therapeutic protein is optimized for
protein interactions. one biological activity, it may have a deleterious effect on
another activity, which may result in adverse effects. Therefore,
Amidation pharmacopoeial monographs include precise specifications
for each formulation of therapeutic proteins, and analytical
Modification ofthe carboxy terminal ofproteins and peptides
characterization.
with an amide group is widely found in vertebrate and
invertebrate proteins and peptides but not in yeasts and Specifications
prokaryotes. Bioactive peptides such as vasopressin,
They comprise ICH recommendations of characterization
oxytocin, gastrin, calcitonin, substance P and Neuropeptide
parameters that include appearance, identity, quantity, and
Yare amidated at their C-terminal. Although amidation is of
purity including impurity profile, stability, and biological
widespread occurrence in mammalian bioactive peptides and
potency. These parameters that are numerical limits are
polypeptides, the biological significance of amidation is less
established by a list of tests, analytical procedures, and other
understood. It is presumed that this PTM may contribute to
criteria, to which a drug substance or drug product should
stability and in increasing the hydrophobicity of the peptide
conform. Conformity of each batch of production to
to facilitate binding to its receptor. Most bioactive peptides
"specification test accept-criteria" is an important part of the
are chemically synthesized and subsequently amidated
batch quality control release (certificate of analysis, COA).
enzymatically using a-amidating enzyme.
The specifications are developed in the early part of process
Covalent modification of Therapeutic Proteins and product development, scale up, non-GMP manufacture,
and cGMP manufacture for clinical trials wherein the active
Many therapeutic recombinant proteins are characterized by drug is fully characterized for its physiochemical properties,
low protease stability, quick renal excretion and high biological activity, immunochemical properties, purity, and
immunogenicity. Covalent modification ofproteins is a process, quantity. The acceptance criteria are related to the analysis
which to some extent, ifnot all overcomes problems. Covalent method chosen and established during the development
conjugation_oLpolyethyleneglycoL(EEG)totherapeutic process ----- ------------ -----------------
pJ:Qteinl'; is one of the: most important tegbniql.!es (Mole:cule: _
Altering Structural Chemistry (MASC)), employed for the IdentitY -
development of second-generation biopharmaceuticals. An important part ofthe overall characterization program for a
PEgylated proteins have been shown to be less susceptible recombinant protein is the identity tests confirming molecular

2598

www.webofpharma.com
IP 2010 BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS

weight, isoelectric point, primary structure, higher order The spectroscopic methods are very powerful when used for
structures (secondary, tertiary and quaternary) and possible comparison analysis with the natural counterpart. The
posttranslational modifications. Protein structure and function confrrmation ofcorrect disulfide linkage is usually carried out
are closely related. Even minor deviations in the three- on appropriate enzymatic or chemical digests of the target
dimensional conformation or in the posttranslational protein followed by HP-RPC purification ofthe fragments and
modifications (e.g., glycosylation, phosphorylation, or subsequent mass analysis using ESI-MS or MALDI-TOF.
acylation pattern) may result in altered biological activity or in The tertiary structure is the three~dimensionalstructure ofthe
an adverse imunogenic/allergic response. During the drug molecule. The standard methods for tertiary structure
development phase extensive characterizations are carried out determination are NMR (in solution), X-ray diffraction using
in order to confirm the chemical and physical properties ofthe crystals at high atomic resolution « 3A), and near UV circular
recombinant product with its natural counterpart. Only a dicroism. The latter method is very powerful for comparison
minority of the identity tests will be used for batch releases analysis with the natural counterpart.
that are specified in the relevant monographs.
The quaternary structure ofa protein represents the interaction
The standard methods for determination of molecular weight between individual polypeptide chains (Subunits) resulting
(MW) are Electrospray Mass Spectra (EMS), Matrix Assisted in larger assemblies. The standard methods for determination
Laser Desorption/lonization (MALDI)-Time ofFlight (TOF), of quaternary structure are HP-SEC, Raman scattering, and
High Performance Size Exclusion Chromatography (HP-SEC), light scattering, useful for determination of large
and Analytical Ultracentrifugation (AVC). Various macromolecular assemblies.
sedimentation velocity measurements (sedimentation velocity,
difference sedimentation, and sedimentation equilibrium) are Posttranslational modifications include chemical modifications
used to gain information on shape and conformation. This is of side groups (e.g., oxidation of
a useful check of homogeneity. Met, de-amidation of Asn of Gin); phosphorylation,
glycosylation, fatty acid acylation, farnesylation, sialic acid
The isoelectric point (PI) is the pH level where the protein has
capping, N-methylation, and acetylation. Typical standard
no net charge. The standard methods for determination of pI
methods for the detection of posttranslational modifications
are Isoelectric focusing in polyacrylamide gels (IEF) and
are HP-RPC, HP-IEC, or mass spectrometry. In mass
Capillary Electrophoresis (CE)-IEF. The separation principle
spectrometry, the MW of the molecule can be determined
is based on charge heterogeneity caused by differences in
with high accuracy and precision (better than 0.01 %). This
amino acid residue charges. The pI may also be determined by
performance is normally sufficient to identify modifications
means of2D-electrophoresis, where molecules are separated
such as missing residues or additional groups. Peptide
according to pI and MW.
mapping, using specific enzymes and subsequent HP-RPC
The primary structure provides information on the amino acid purification of the fragments prior to MW detection is also
sequence ofthe protein. For most proteins a primary structure used, for example, for determination ofglycosylation patterns.
analysis comprises N-terminal sequencing by Edman Heterologous glycosylated products are often identified by
degradation, C-terminal analysis, and peptide mapping their isoelectric focusing slab gel pattern or more rarely by
followed by High Performance Reversed Phase 2D-electrophoresis. A thorough carbohydrate structural
Chromatography (HP-RPC) purification and subsequent analysis may include glycosylation site(s), carbohydrate chain
determination of the MW of the fragments by mass structure, the oligosaccharide pattern, and the content of
spectrometry. The amino acid sequence is often supported by neutral sugars, amino sugars, and sialic acids.
total amino acid analysis, comparison of the cloned gene
The extinction coefficient can be determined from a known
sequence, and the molecular weight determination.
protein quantity (protein concentration) and the absorbance
Comparative fingerprints between the natural and the
at 277 to 280 urn. In chromatographic evaluation, thetarget
recombinant protein are also used to confrrm primary structure
protein retention time using HP-IEC or HP-RPC can be used
identity.
as an identity marker. For biomolecular interaction analysis,
The secondary structure provides information on disulfide the method uses surface plasmon resonance to detect
bond arrangement, a-helix, and ~-sheet content. The standard biomolecular interactions.
methods for determination of disulfide arrangements are
peptide mapping by HP-RPC or Sodium dodecyl sulphate Biological Activity
(SDS)-Polyacrylamide Gel Electrophoresis (pAGE). Structural
information is obtained by means of fluorescence, far UV The biological activity (Potency) describes the ability of the
circular dichroism, Raman scattering, and infrared absorption drug substance to achieve a defined biological effect. Examples
using Fourier Transformed Infra Red Spectroscopy (FTIR). ofprocedures used to measure the biological activity include

2599

www.webofpharma.com
BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS IP 2010

animal based biological assays; cell culture-based biological forms are detected by analytical HP-lEC, HP-RPC, native PAGE,
assays, biochemical assays, and ligand binding assays. A lEF, MS, or CEo The content accepted depends on the nature
biological assay may be replaced by physicochemical tests of the drug product and the dose.
provided sufficient information and correlation between the
Oxidized Forms. Oxidized forms are target protein derivatives
bioassay and the said tests can be demonstrated and there
in which one or several Met, Cys, His, Trp, or Tyr residues
exists a well-established manufacturing history (ICH
have been oxidized. The oxidation ofcystinyl residues results
Harinonized Tripartite Guideline. Specifications: Test
in formation of a disulfide bond (cystinyl residue). Oxidized
Procedures and Acceptance Criteria for Biotechnological
forms are detected by analytical HP-lEC, HP-RPC, native PAGE,
Products (Q6B).
lEF, MS, or CEo The content accepted depends on the nature
Structural variants of therapeutic proteins in general and of the drug produet and the dose.
recombinant proteins in particular which include isoforms (e.g.,
Scrambled Forms. Scrambled forms· are target protein
variations in post translational modifications, varying degrees
molecules with a disulfide bond pattern different from that of
of processing of C- and N- terminal substitutions etc) often
the native molecule. Scrambled forms are typically formed
show variations in biological activity. Thus, the batch release
during in vitro folding ofproteins, but disulfide bond shuffling
of the product should be shown to possess the expected
at neutral pH or above also occurs. This requires studies on
degree ofbiological activity using relevant bioassays. In order
control of protein stability during downstream processing.
to use the same dosing protocol as a reference medicinal
The formation of scrambled forms is closely linked to the
product, the content and specific activity of the clinically
folding procedure and is protein specific, which are detected
comparable product should be compared to that of one or
by analytical HP-lEC, HP-RPC, CE, or peptide mapping. The
more reference medicinal products, or to an international
content accepted depends on the nature of the drug product
reference standard to which the reference product is calibrated.
and the dose.
(WHO Guideline for abbreviated licensing pathways for certain
therapeutic proteins) Cleaved Forms. Cleaved forms are those where a peptide bond
is cleaved, resulting in loss ofa N- or C-terminal site or where
Purity an internal peptide bond is cleaved while at the same time the
Protein purity has been historically linked to the specific resulting fragments are kept together by means of disulfide
biological activity in terms of units of biological activity per bonds. These forms are detected by N- and C- terminal amino
mass unit of the product. The purest product is that of the acid analysis.
highest specific biological activity. In contrast to drugs based
Aggregates. Aggregates are target protein derivatives in which
on small molecules, which could be controlled on the drug two or more molecules are linked together either by covalent
product level, protein-based pharmaceuticals are closely linked
inter-disulfide bonds or by hydrophobic interaction. Target
to the process itself due to the complexity of the active protein aggregates are formed as a result of hydrophobic
pharmaceutical ingredient and the lack of proper intermolecular reactions or because ofintermolecular disulfide
characterization ofthe final product. With the introduction of
bond formation under oxidizing conditions. Aggregates are
recombinant technology and modem analytical methods, a very often antigenic, resulting in formation of antibodies
much better drug substance/product characterization has against the target protein. Proteins exposed to even mildly
become possible resulting in the well-characterized protein
denaturing conditions may partially unfold, resulting in the
concept and the widespread use of comparability studies. exposure of hydrophobic residues to the aqueous-solvent,
The importance of a: stronger focus on presence of favoring aggregation. It is generally believed that the
adventitious agents and specific impurities are also aggregation process is controlled by the initial dimerization
recognized, as the presence of even minor amounts oftoxic,step in a second order reaction. Consequently, high protein
immunogenic, or adventitious compounds proved to have concentrations will increase the aggregation rate.
severe side effects. The acceptable level ofimpurities depends
Intermolecular disulfide bond formation between cystinyl
on the nature of the drug product and the dose. residues takes place at alkaline pH under oxidizing conditions.
Proteins with reactive free thiol groups ~hould be purified
Impurity ProfIle under reducing conditions (typically 1 to 10 mM reducing
agent) in the presence of EDTA. Even proteins with disulfide
Product related impurities
.bonds-may-participate--in-intermolecular-disulfide-bond
DeS:3midoForms. Des-amido forms are target protein reactions due todisulfide bond shuffling at neutraland alkaline
derivatives in which one or several of the glutaminyl or pH. The aggregation reaction based on intermolecular disulfide
asparagyl amino acid residues are converted to. the bond formation is prevented at pH < 6 and under reducing
corresponding acids (glutamyl and asparagyl). Des-amido conditions. The hydrophobic aggregation compounds,

2600

www.webofpharma.com
lP 2010 BIOTECHNOLOGY DERIVED THERAPEUTIC PRODUCTS

enzymes, detergents, and stabilizers must be accounted for for lot release. If the assays are based on competently
and their removal validated. Large amounts of hydrophobic produced antibodies raised against cell lysates, the quality
reagents may affect hydrophobic interaction chromatography. may be adequate
Analytical HP-IEC, HP-RPC, native PAGE, IEF, MS, or CE
Viruses
detects oxidized forms. The content accepted depends on the
nature of the drug product and the dose. Non-reducing ID- Virus contamination comes from the host cell, the culture
SDS, HP-SEC, MS, or CE can detect disulfide-based medium, and infections during manufacture. The host cell may
aggregates. Hydrophobic aggregates may be detected by HP- contain a genomic virus or virus vectors used to transform
SEC. The content accepted depends on the nature of the drug the cell line. The type of viral genome or vector depends on
product and the dose as specified in the product monographs. the cell line history. Chronic or latent viruses may be present
in continuous cell lines, and the retroviruses associated with
Process Related Impurities continuous cell lines are non-infectious, but oncogenic.
Pyrogens and Endotoxins Epstein-Barr virus or Sendai virus is often used for cell
transformations. Contaminants such as BVDV, IBR, reovirus,
Pyrogens are a group of chemically diverse substances, PI-3, bovine leukemia virus, and bovine polyoma virus should
including endotoxins ofbacteria and debris of dead bacterial be expected from serum-supplemented media. The cell line
cells that cause fever and shock in severe cases. The most history reveals all information on the origin and identity ofthe
important pyrogenic substances in pharmaceutical industry cell line and the host genome vectors used to establish the
are bacterial endotoxins. Endotoxins come from Gram-negative cell line
bacteria (e.g., E. coli), if it is used as the expression system.
Presence of endotoxins indicates bacterial contamination in Prions
raw materials, columns, water, and buffers. There are two Prions come from transmissible spongiform encephalopathies
methods of detection: the pyrogen test, which is based upon (TSE), The major source of contamination of a recombinant
the measurement of body temperature of rabbits before and product is the use ofanimal-derived raw materials, which could
after injection of the specimen, and the Limulus Amebocyte harbor bovine prions (BSE agent). Currently, there are no assays
lysate (LAL) test, which is based upon the clotting reaction of that are sensitive or specific' enough to test raw materials or
an enzyme complex of cells of the horseshoe crab together sources, and the only reliable prevention is to include barriers,
with bacterial endotoxins (in vitro test). such as avoidance of animal or human raw materials (e.g.,
Nucleic acids trypsin, serum, transferin, bovine/human serum albumin,
protein supplements, peptones).
Nucleic acid contamination comes from host cell DNA/RNA
or retroviral RNA. Nucleic acids are detected by monitoring Microbial agents including Mycoplasma
absorption of light at 260 om. The residual content in drug
Microbial agents and fungi come from infection of the
substance (DS) or drug product (DP) is usually measured by
bioreactor during cell culture. Other sources are contaminated
PCR or amplification teclmiques. The maximum allowable
water, buffers, raw materials, chromatographic columns, and
content of nucleic acid per dose remains under continuous
equipment. Fermentation and cell culture bioreactors are prone
evaluation by regulatory agencies. "Lot-to-lot" testing for
to microbial infections. Viable cells can be identified by spread
DNA content in biological products produced in cell lines
out of the cell suspension or sample solution on agar plates.
should be performed and lot release limits established which
reflects a level of purity that can be achieved reasonably and Mycoplasma
consistently.
Mycoplasmas have for long been recognized as a contaminant
Host cell proteins of continuous cell cultures caused by an infection of the cell
line or bioreactor. Working in closed systems under GMP will
Host cell proteins (HCP) come from the host organism and reduce the risk ofinfection. The end ofproduction test includes
constitute a major purification problem due to variability screening for mycoplasmas. Mycoplasmas are difficult to
structure and surface properties. The amount released into detect, the only reliable way of demonstrating infection is by
the culture medium depends on the expression system used, agar plating, fluorescent dying of DNA, or by PCR
the culture conditions and the process related cell lysis.
Extraneous proteins may be introduced in the downstream Quantity
process also. Several analytical methods have been used to
Protein content
monitor HCP including SDS-PAGE, 2D-electrophoresis (IEF
in combination with SDS-PAGE), Western blot (WB), and Quantitative estimation of protein is measured by several
immunoassays. However, Generic HCP assays are preferred methods (Kjeldahl analysis, Lowry assay, Biuret assay,

2601

www.webofpharma.com
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2010

Bicinchoninic acid (BCA) assay, Bradford assay, UV Host cell-derived proteins. This must be routinely monitored
absorbance andamino acidanalysis). It is desirable to confIrm using a scientifically accepted method that demonstrates:
the quantity by more than one method.
1. The assay is sensitive (a useful target for the limit of
Stability detection is 1 to 100 ppm)
2. The assay is specifIc for the HCP (defined by proprietary
Formulation development of biopharmaceutical therapeutic detection reagents such as antibodies elicited against
protein will provide a fInal dosage form that offers ex vivo process-specifIc contaminating HCP) consistently found
stability during processing, handling, and long-term storage in the product from a specifIc manufacturing / purifIcation
under specifIed conditions (e.g., temperature, humidity etc). It process.
is also expected to provide adequate in vivo bioavailability
that meets the desired pharmacokinetic/pharmacodynamic (pK/ Limits are approved by the competent authority.
PD) properties. Therapeutic Proteins do not survive terminal
Host cell and vector-derived DNA
sterilization procedures commonly employed for small
therapeutic molecule. Therefore, formulation components and The limit is as prescribed by the competent authority.
excipients should also undergo microbiological tests including Description. A clear and colourless solution.
analysis for endotoxins and pyrogens in addition to biological,
chemical and physical functions. Identification
A. It gives the appropriate response when examined using the
conditions described under Assay.
Erythropoietin Concentrated Solution B. Determine by isoelectric focusing (2.4.33).
Test solution. Dilute, the preparation in water, if necessary
and desalt the preparation, using a membrane fIltration system
APPRLlCDSR VLERYLLEAK EAENITTGCA
I suitable for desalting proteins. Make up the volume to the
I original volume with water.
EHCSLNENIT VPDTKVNFYA WKRMEVGQQA
Reference solution. Dissolve erythropoietin RS in water to
VEVWQGLALL SEAVLRGQAL LVNSSQPWEP produce a solution containing 1mg per ml and desalt as above.
LQLHVDKAVS GLRSLTTLLR ALGAQKEAIS The isoelectric focusing procedure may be carried out using a
0.5 mm thick polyacrylamide slab gel containing ampholytes
PPDAASAAPL RTITADTFRK LFRVYSNFLR covering the pH of range 3 to 10, prepared as follows.

GKLKLYTGEA CRTGD Mix 9 g of urea, 6.0 ml of 30 per cent acrylamide/bisaclyl-


amide solution, 1.05 ml of pH 3 to 5 ampholyte, 0.45 ml of
pH 3 to 10 ampholyte and 13.5 ml of water. Degas the mixture
Structure of Erythropoietin Mol. Wt. 30,600 (approx)
. Add 15 ).11 of tetramethylethylene diamine and 0.3 ml ofa 100
Erythropoietin Concentrated Solution contains a family of g per litre freshly prepared solution of ammonium persulphate.
closely related glycoproteins which are not different from the Pour into a suitable gel cassette, with approximate dimensions
naturally occurring human erythropoietin (urinary ofl5 cmx 15 em x 0.05 em. Insert a suitable sample well comb
erythropoietin) in terms ofthe amino acid sequence (165 amino and allow to polymerize.
acids) and average glycosylation pattern, at a concentration
Use the anode solution the anolyte for isoelectric focusing
of 0.5 mg per ml to 10 mg per ml. It has a potency of not less
at pH 3 to 5 and the cathode solution the catholyte for
than 100000 ill per mg of active substance.
isoelectric focusing pH 3 to 5. Allow prefocusing to take
Production place for 1 hour at a constant power of 10 W, with maximum
voltage and current settings of 2000 V and 100 rnA,
Erythropoietin is produced in rodent cells in vitro by a method respectively.
based on recombinant DNA technology. All batches are tested
Dilute the test solution to 0.5 mg per ml with water. Apply to
as described below, prior to release (unless exemption has
granied'bythecompeteriiautIi'orlty)'. _.,--_.- the-geHOfJ;lofeach solution;-Garryout·foGusing-for-a-further
30 minutes at-thesamepowersupply settings. Take the gel
Host cell-derived proteins (Hep) out ofthe focusing chamber, and transfer the proteins onto a
membrane suitable for immobilization of proteins (such as
The limit is as prescribed by WHO. Polyvinylidene Fluoride), using commercially available

2602

www.webofpharma.com
IP 2010 ERYTHROPOIETIN CONCENTRATED SOLUTION

electrotransfer equipment and following the manufacturer's All the solutions should be filtered through a 0.45 /lm membrane
instructions. After electrotransfer, incubate the membrane in a filter before use.
neutral isotonic buffer containing a suitable blocking agent Test solution. Dilute the substance under examination with
(for example, 50 g per litre ofdried mille), for 1 hour, followed by water or concentrate it to obtain a concentration on mg per
incubation in the same blocking solution with a suitable dilution ml. Desalt 0.25 ml ofthe solution by passage through a micro-
of a polyclonal anti-erythropoietin antibody. Detect the concentrator cartridge provided with a membrane with a
erythropoietin-bound antibody using a suitable enzyme- or molecular mass cut-off not more than 10000. Add 0.2 ml of
radiolabelled antibody (for example, an alkaline phosphatase- water to the sample and desalt again. Repeat the desalting
.conjugated second antibody). The precise details of blocking procedure once more. Dilute the sample with watel; determine
agents, concentrations and incubation times should be its protein concentration as described under Tests and adjust
optimized using the principles set out in Immunochemical to a concentration ofapproximately 1 mg per ml with water.
methods.
Reference solution. Dissolve elythropoietin RS in water to
The test is not valid unless the distribution of bands in the produce a solution containing 1 mg per ml. Desalt the sample
electrophoretogram obtained with the reference solution as described for the test solution.
contains at least 6 well separated bands. If necessary, the Capillary system
voltage settings and duration may be altered to optimize the material. uncoated fused silica,
separation of the isofonns. size. effective length = about 100 cm, internal diameter
=50/lm,
In the electrophoretogram obtained with the test solution, the temperature. 35°,
pH range of the bands observed corresponds to that of the spectrophotometer set at 214 llID,
electrophoretogram obtained with the reference solution. The - injection. under pressure or vacuum.
predominant bands correspond to isoforms 4, 5, 6 and 7.
Additional, fainter bands corresponding to isoforms 1, 2, 3 CZE buffer concentrate (0.1 M sodium chloride, 0.1 M tricine,
and 8 may also be present. Other bands are present in no more 0.1 M sodium acetate). Dissolve 0.584 g of sodium chloride,
than trace amounts. 1".792 g oftricine and 0.820 g of anhydrous sodium acetate in
water and dilute to 100.0 ml with the same solvent.
C. Determine by capillary electrophoresis (capillary zone 1 M putrescine solution. Dissolve 0.882 g ofputrescine in 10
electrophoresis) (2.4.32). . ml of water. Distribute in 0.5 ml aliquots.

'"
on
<!
"'-

'"
'"~-

'"E
oS
_ "'Vl
5....." 00

e.EC1 E
8 ~

g
~HI._ _ --'-----_..J>.._.,J-. .., ~ .... _ . .:r-'I IA!~;: _

I.--,-:-r--...,..--.,.---,.....----,.--,....-.,..._,--.I=:I,_-,._...,....._.,.-_...-_,...---
g'"
Reference electropherogram ofErythropoietin

2603

www.webofpharma.com
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2010

CZE buffer. (0.01 M tricine, 0.01 M sodium chloride, 0.01 M D. Immunoblotting. Determine by electrophoresis (sodium
sodium acetate, 7 M urea, 2.5 mMputrescine). Dissolve 21.0 dodecyl sulphate polyacrylamide gel electrophoresis) (SDS-
g of urea in 25 ml of water by warming in a water-bath at 30°. PAGE) (2.4.12).
Add 5.0 ml of CZE buffer concentrate and 125 ml of 1 M
Use a 1-mm thick spacer with six-well comb, a well capacity of
putrescine solution. Dilute to 50.0 ml with water. Using dilute
451 and suitable plate size e.g. 100 x 120 mm
acetic acid, adjust the pH to 5.55 at room temperature and
filter through a 0.45 /-lm membrane filter. Assemble the gel casting as recommended by the
manufacturer.
Set the auto sampler to store the samples at 4° during
analysis. Use the composition of gel given below:
Preconditioning ofthe capillQ1Y. Rinse the capillary for 60 Separating gel 12 per cent
minutes with 0.1 M sodium hydroxide filtered through a 0.45
Add the reagents into a clean glass/plastic container in
/-lm membrane filter and for 60 minutes with CZE buffer. Apply
the same sequence as given in the table below:
voltage for 12 hours (20 kV).
Between-run rinsing. Rinse the capillary for 10 minutes with Solutions For7rnl For 14ml
water, for 5 minutes with 0.1 M sodium hydroxide filtered (for 1gel) (for 2 gels)
through a 0.45 /-lm membrane filter and for 10 minutes with Water 2.16rnl 4.32rnl
CZEbuffer.
AClylamide (30 per cent) 2.8ml 5.6rnl
Migration. Apply a field strength of 143 V/cm (15.4 kV for
capillaries of 107 cm total length) for 80 min, using CZE bliffer 1.5 MTris-CI pH8.8 1.75rnl 3.4rnl
as the electrolyte in both buffer reservoirs. 20 per cent SDS 35rnl 70rnl
System suitability. In the electropherogram obtained with the Ammonium persulphate 70rnl 140ml
reference solution, a pattern of well-separated peaks (iO per cent w/v)
corresponding to the peaks in the reference electropherogram
ofelythropoietin RS is seen, and the largest peak is atleast 50 TEMED 3rnl 6ml
times greater than the baseline noise. If necessary, adjust the
sample load to give peaks of sufficient height. Identify the Mix the above contents by swirling gently (do not mix
peaks corresponding to isoforms 1 to 8. The peak vigorously) in the container and pour into the casting unit
corresponding to isoform 8 is detected; the resolution between using a 1-ml pipette to 1 to 1.5 cm from the top edge of the
the peaks corresponding to isoforms 5 and 6 is not less than 1. plate. After addition of ammonium persulphate and TEMED,
Repeat the separation at least 3 times. The baseline is stable, mix the solution and pour quickly (in less than 1minute) or it
showing little drift, and the distribution ofpeaks is qualitatively will begin to polymerize. Pour 200 to 1000 /-ll of water-saturated
and quantitatively similar to the distribution of peaks in the butanol on the top of separation gel. Keep aside for at least
reference electropherogram ofelythropoietin RS. The relative 45 minutes for polymerization at room temperature.
standard deviation of the migration time of the peak Casting the Stadang gel: 4 per cent
corresponding to isoform 2 is less than 2 per cent.
Decant the water-saturated butanol and rinse the separating
Identify the peaks corresponding to isoforms 1 to 8 in the gel with water. (If the gel has not polymerized and flows out,
electropherogram obtained with the test solution by discard and prepare fresh). Place the comb in position above
comparison with the electropherogranl obtained with the the separating gel. Prepare the stacking mix by adding the
reference solution. Calculate the percentage content of each reagents in the same sequence as given in the table below:
isoform from the corresponding peak area. The percentages
are within the following ranges: Solutions For5rnl For lOrnl
(1 gel) (2 gels)
Isoform Number Content (per cent)
Water 1.8ml 3.6rnl
1 0-15
2 0-15 Acrylamide (30 per cent) 0.6rnl 1.2rnl
3 1-20 0.5 MTris CI pH 6.8 2.5rnl 5.0rnl
4 10'-:'35

Ammonium· pel'sulphate 50/-ll 100/-ll


1<)"-35·
(10 per cent w/v) (APS)
7 5-25
8 0-15
TEMED 5/-ll 10/-ll

2604

www.webofpharma.com
IP 2010 ERYTHROPOIETIN CONCENTRATED SOLUTION

Mix the above ingredients by swirling gently and pour over 3. Equilibrate the membrane in chilled transfer buffer for
the separating gel. Avoid bubbles. Keep aside for at least 45 15 - 20 minutes.
minutes for polymerization at room temperature. 4. Equilibrate the gel in chilled transfer buffer for 15 - 20
Preparation ofsamples and loading minutes.
Samples include: 5. Soak the nylon pads in transfer buffer and place on the
pad on the cathode.
1. Test sample 1.0 Ilg
6. Take 5 sheets of a suitable filter paper, cut to the size of
2. Reference standard RS 1.0 Ilg
the gel and soak in chilled transfer buffer. Place them on
3. Prestained Marker 20 III the nylon pad placed on cathode plate.
Test solution. Take X III of the test sample and add an equal 7. Carefully place the equilibrated gel on the filter papers.
volume of 2X non-reducing sample bU.ffer where, 8. Place the equilibrated membrane onto the gel.
X (Ill) = lllg per (concentration ofthe test sample in mg per ml) 9. Roll a clean glass rod dipped in transfer buffer on top of
Reference solution. Dilute a part of Img per ml reference the membrane to get rid of any air bubbles trapped
standard RS stock 10 times to get a final concentration of between the gel and the membrane.
0.1 mg per ml with water as follows: 10. Place 5 sheets of a suitable filter paper soaked in chilled
To 10 III of 1 mg per ml reference standardRS solution add 90 transfer buffer over the membrane. Once again roll the
mlwater. glass rod to remove any air bubbles.
To 10 III of 0.1 mg per ml reference standardRS solution add 11. Close the cassette.
10 ml of 2X non-reducing sample buffel: 12. Place the cassette in the running unit.
Prestained marker. Take 20 ml of prestained marker, 13. Connect the electrodes with the Power pack and set
reconstituted as recommended by the manufacturer. following parameters for transfer:
Loading ofsamples. Current: 200 rnA
Boil the samples for two minutes, centrifuge, bring to room - Voltage: 100 V
temperature and load the entire volume on the gel. - Time: 1 hour.
Sample Samples Amount of protein Total Vol. to be 14. After the transfer is over, disassemble the cassette.
No. loaded/well (Ilg) loaded. 15. Transfer the membrane to the blocldng solution. Incubate
1 Test Sample 1 the membrane in the blocldng buffer for 1 hour with gentle
shaking at room temperature.
2 Reference .1 20
16. Discard the blocldng buffer. Add 25 ml of rabbit anti
3 Marker 20 r-Hu EPO antibody (Primary antibody) solution.
Running the gel. 17. Incubate for Ihour with gentle agitation at room
1. Fix the gel apparatus in the running unit according to the temperature.
manufacturer's instructions. 18. Wash the membrane with 1 X transfer btifJer saline with
change of buffer in-between at room temperature with
2. Chill the IX running buffer to 4° for at least 1 hour.
gentle agitation (3 times X 5 minutes).
3. Pour IX running buffer in the upper as well as lower 19. Discard the transfer btifJer saline. Add 25 ml ofgoat anti-
chambers of the running unit. rabbit ALP conjugated antibody (secondary antibody)
4. Set parameters on the Power pack as follows: solution. Incubate for 1 hour with gentle shaking at room
- Constant Voltage: 130 V temperature.
- Max Current: 200 rnA. 20. Wash the membrane with 1 X transfer buffer saline (3
times X 5 minutes)
5. Run until dye front just goes out at the gel bottom (usually
about 1.5 hrs) 21. Develop the color by adding 5 ml ofthe substrate solution
(Usuallyittakes l5-20minutes).
Transfer, blotting and development of membrane: 22. Stop the reaction by pouring off the substrate solution
1. Disassemble the running unit according to the and washing it with water before the color of the
manufacturer's instructions. background gets dark.
2. Cut Nitrocellulose Membrane (NCM) having the same 23. Scan the blot while it is wet.
dimensions as of the gel. 24. Air dry the blot and preserve it.

2605

www.webofpharma.com
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2010

The molecular mass markers are resolved on the membrane Equilibrate at initial conditions for at least 15 minutes. Carry
into discrete bands with a linear relationship between distance out a blank run using the above-mentioned gradient.
migrated and logarithm lO ofthe molecular mass. The chromatogram obtained with each solution is qualitatively
To evaluate the linear relationship between the distance similar to the reference chromatogram oferythropoietin RS
migrated and 10garithmIO ofthe molecular mass, calculate digest.
a) log molecular weights corresponding to marker bands The profile of the chromatogram obtained with the test
b) migration distance of protein band solution corresponds to that of the chromatogram obtained
c) plot (a) vs (b) and perform linear regression analysis with the reference solution.

The graph should be linear F. Determine by N-terminal sequence analysis


The first 15 amino acids are: Alanine - Proline - Proline -
The single broad band of the test solution and of reference
Arginine - Leucine - Isoleucine - (no recovered peak) - Aspartic
standard RS match in position and intensity.
acid - Serine - Arginine - Valine - Leucine - Glutamic acid -
Peptide mapping (2.3.47). Arginine - Tyrosine.
Test solution. Dilute the substance under examination in tris- Perform the Edman degradation using an automated solid-
acetate buffer solution pH 8.5 to a concentration of 1.0 mg phase sequencer, operated in accordance with the
per ml. Equilibrate the solution in tris-acetate buffer solution manufacturer's instructions.
pH 8.5 using a suitable procedure (such as dialysis against
Desalt the equivalent of50 /-lg oferythropoietin. For example,
tris-acetate buffer solution pH 8.5, or membrane filtration using dilute a volume ofthe substance under examination containing
the procedure described under Identification C, but
50 /-lg of the active substance in 1 ml of a 0.1 per cent v/v
reconstituting the desalted sample with tris-acetate buffer solution of trifluoroacetic acid. Pre-wash C 18 reverse-phase
solution pH 8.5). Transfer the dialyzed solution to a sample preparation cartridge according to the instructions
polypropylene centrifuge tube. Freshly prepare a solution of
supplied by the manufacturer and equilibrate the cartridge in
trypsin for peptide mapping at a concentration of 1 mg per ml a 0.1 per cent v/v solution of trifluoroacetic acid. Apply the
in water, and add 5 ml to 0.25 ml ofthe dialysed solution. Cap sample to the cartridge, and wash successively with a 0.1 per
the tube and place in a water-bath at 37° for 18 hours. Remove cent v/v solution of trifluoroacetic acid containing 0 per cent,
the sample from the water-bath and stop the reaction 10 per cent and 50 per cent v/v of acetonitrile according to
immediately by freezing. the manufacturer's instructions. Lyophilise the 50 per
Reference solution. Dissolve the contents of a vial of cent v/v acetonitrile eluate.
erythropoietin RS in 0.25 ml of water. Prepare as for the test
Redissolve the desalted sample in 50 /-ll of a 0.1 per cent v/v
solution, ensuring that all procedures are carried out solution of trifluoroacetic acid and couple to a sequencing
simultaneously, and under identical conditions. cartridge using the protocol provided by the manufacturer.
Chromatographic system Run 15 sequencing cycles, using the reaction conditions for
a stainless steel column 25 cm x 4.6 mm, packed with proline when running the second and third cycles.
butylsilyl silica gel (5-10 /-lm),
Identify the phenylthiohydantoin (PTH)-amino acids released
- mobile phase: A. 0.06 per cent v/v solution of
at each sequencing cycle by reverse-phase liquid
trifluoroacetic acid,
chromatography. The procedure may be carried out using the
B. to 100 m1 of water add 0.6 ml of
column and reagents recommended by, the manufacturer of
trijluoroacetic acid and dilute to 1000 ml with
the sequencing equipment for the separation ofPTH-amino-
acetonitrile,
acids.
a linear gradient programme using the conditions given
below, The separation procedure is calibrated using:
- spectrophotometer set at 214 nm, the mixture of PTH-amino acids provided by the
- injection volume. 50 /-ll. manufacturer of the sequencer, with the gradient
Time Flow rate Mobile phase A Mobile phase B conditions adjusted as indicated to achieve optimum
(in min) (ml/min) (per cent v/v) (per cent v/v) resolution of all amino acids,
0-10 0.75 100 o a sample obtained from a blanksequencing cycle obtained
m=125' 0:75 100=+39 , 0=+'61 . asrecommended'by the equipment-manufactureE --
125"': 135 1.25 39 ~T7 61~83'
Tests
135 -145 1.25 17 --70 83 --7100
145-150 1.25 100 o Protein. 80 per cent to 120 per cent of the stated amount.

2606

www.webofpharma.com
IP 2010 ERYTHROPOIETIN CONCENTRATED SOLUTION

Test solution. Dilute the substance under examination in a 0.4 Solutions For20ml For40ml
per cent w/v solution of ammonium hydrogen carbonate or (1 gel) (2 gels)
the appropriate blank solution.
Water 6.l7ml 1234ml
When examined in the range 250 nm to 400 nm (2.4.7), the Acrylamide (30 per cent) 8.0ml 16.0ml
solution shows an absorption maximum between 276 nin and
1.5 MTris Cl pH 8.8 5.0ml 1O.Oml
282 nm. After correction of any light scattering measured up
to 400 nm, calculate the content of erythropoietin taking the 20 per cent SDS 100 III 200 III
specific absorbance to be 7.43. Ammonium persulphate 200 III 400J.lI
(10 per cent w/v) (APS)
Dimers and related substances of higher molecular mass
TEMED 8 III 16 J.lI
A. Detennine by size-exclusion chromatography (2.4.16).

You might also like