You are on page 1of 313

The Ministry of Health, Labour and

Welfare Ministerial Notification No. 395

In accordance with the provision of Paragraph 1 of Article 41 of the Pharmaceuti-


cal Affairs Law (Low No. 145, 1960), we hereby revise a part of the Japanese Phar-
macopoeia (Ministerial Notification No. 111, 2001) as follows, and the revised
Japanese Pharmacopoeia shall come into effect on January 1, 2003. The drugs in-
cluded in the Japanese Pharmacopoeia (hereinafter referred to as ``the old Japanese
Pharmacopoeia'') [limited to those included in the Japanese Pharmacopoeia whose
standards are changed in accordance with this notification (hereinafter referred to as
``the new Japanese Pharmacopoeia'')] and the approval for which is given on Janu-
ary 1, 2003 as stipulated in Paragraph 1 of Article 14 (including cases where it shall
apply mutatis mutandis under Article 23 of the Law; the same hereinafter) [including
the drugs designated as those exempted from manufacturing or import approval
(Ministerial Notification on the designation of the drugs exempted from manufactur-
ing or import approval in accordance with the provision of Paragraph 1 of Article 14
of the Law) (hereinafter referred to as ``the drugs exempted from approval'')] can be
treated, up to June 30, 2004, in such a way that the standards established by the old
Japanese Pharmacopoeia (limited to the contents concerned with the relevant drugs)
may be accepted as those established by the new Japanese Pharmacopoeia. The drugs
included in the new Japanese Pharmacopoeia (excluding those in the old Japanese
Pharmacopoeia) the approval for which is given on January 1, 2003, as stipulated in
Paragraph 1 of Article 14 of the Law (including the drugs exempted from approval),
can be treated, up to June 30, 2004, as those not included in the new Japanese Phar-
macopoeia.

Chikara Sakaguchi
The Minister of Health, Labour and Welfare

December 27, 2002

(The texts referred to by the term ``as follows'' are omitted here. All of them are
made available for public exhibition at the Evaluation and Licensing Division, Phar-
maceutical and Medical Safety Bureau, Ministry of Health, Labour and Welfare, at
each Regional Bureau of Health and Welfare and at each Prefectural Office in
Japan.)

The term ``as follows'' here indicates the contents from Part I to Ultraviolet-visible Reference
Spectra in the Supplement I to the Japanese Pharmacopoeia Fourteenth Edition
(pp.1359 – 1610).
CONTENTS
Preface ...................................................... i Infrared Reference Spectra ................ 1567–1586
Supplement I to The Japanese Pharmacopoeia, Part I ................................................ 1567
Fourteenth Edition, Part I................. 1359–1540
General Notices ................................... 1359 Ultraviolet-visible Reference Spectra .... 1587–1610
General Rules for Preparations ............... 1361 Part I ................................................ 1587
General Tests, Processes and Apparatus ... 1363
23. Infrared Spectrophotometry.............. 1363 General Information
52. Residue on Ignition Test .................. 1363 1. Aristolochic Acid............................ 1611
66. Vitamin A Assay ............................ 1364 5. International Harmonization Implemented
70. Reference Standards; Reagents, Test Solutions; in the Japanese Pharmacopoeia Fourteenth
Standard Solutions for Volumetric Analysis; Edition ......................................... 1611
Standard Solutions; Matching Fluids for Color; 12. Preservatives-Effectiveness Tests ........ 1616
Optical Filters for Wavelength and Transmis- 17. Basic Requirements for Viral Safety of
sion Rate Calibration; and Measuring Instru- Biotechnological/Biological Products
ments, Appliances ............................ 1365 listed in Japanese Pharmacopoeia ...... 1618
(1) Reference Standards................. 1365 18. Qualification of Animals as Origin of
(2) Reagents, Test Solutions ........... 1365 Animal-derived Medicinal Products
(3) Standard Solutions for Volumetric provided in the General Notices 39 of
Analysis................................. 1373 Japanese Pharmacopoeia and Other
72. Conductivity Measurement................ 1373 Standards...................................... 1631
73. Determination of Bulk and Tapped 19. SDS-Polyacrylamide Gel
Densities ....................................... 1375 Electrophoresis............................... 1634
Official Monographs............................. 1377
Appendix .............................................. 1641
Supplement I to The Japanese Pharmacopoeia,
Fourteenth Edition, Part II................ 1541–1566 Index.................................................... 1643
General Rules for Crude Drugs ............... 1541 Index in Japanese.................................... 1659
Official Monographs............................. 1543
Preface
The Fourteenth Edition of the Japanese Phar- and to securing and maintaining international con-
macopoeia was promulgated on March 30, 2001 by sistency.
Ministerial Notiˆcation No. 111 of the Ministry of It was also agreed that JP articles should cover
Health, Labour and Welfare. To keep pace with drugs which are important from the viewpoint of
progress in medical and pharmaceutical sciences, in health care and medical treatment, clinical results and
November 2001, the Council, at a meeting of the frequency of use, as soon as possible after they reach
Committee on Japanese Pharmacopoeia (JP), estab- the market.
lished the basic principles for the preparation of the It was also decided to make a deˆnite rule for selec-
JP Fifteenth Edition, setting out the characteristics tion of articles, by clarifying the signiˆcance of, and
and roles of the JP, the deˆnite measures for the revi- standards for selection. The JP Fifteenth Edition was
sion, the date of the revision, and the organization of decided to be slated for completion in April 2006.
the Subcommittee on JP. Under the Subcommittee on JP, the following
At the above meeting, the following ``ˆve pillars'' twelve panels and two provisional panels were estab-
were established as the basic principles of the JP: lished at ˆrst: Panel on Planning and Revisions; Panel
Making it more substantial by including all drugs on the Selection of Articles; Panel on Nomenclature;
which are important from the viewpoint of health care Panel on Excipients; First Panel on Medicinal Chemi-
and medical treatment; Making prompt partial revi- cals; Second Panel on Medicinal Chemicals; Panel on
sion as necessary and facilitating smooth administra- Biologically Derived Drugs; Panel on Biological Tests;
tive operation; Promoting international harmoniza- Panel on Physico-chemical Tests; Panel on Material
tion; Ensuring transparency regarding the revision and Sciences; Panel on Preparations; Panel on Crude
dissemination to the public of the JP; and Positively Drugs; Provisional First Panel on Antibiotics;
introducing contemporary analytical tests and de- Provisional First Panel on Crude Drugs. Some of the
veloping reference standards. It was decided at the names of the above panels were changed as follows,
meeting that each panel set up under the Subcommit- due to the reorganization of the Pharmaceutical
tee on JP should make eŠorts, on the basis of these AŠairs and Food Sanitation Council (PAFSC) in
principles, to ensure that the JP is used more eŠective- November 2001: Panel on Nomenclature to Panel on
ly in the ˆelds of health care and medical treatment by Nomenclature for Pharmaceutical Chemicals; First
taking appropriate measures, including getting the un- Panel on Medicinal Chemicals and Second Panel on
derstanding and cooperation of other parties con- Medicinal Chemicals were integrated into Panel on
cerned. Medicinal Chemicals; Panel on Physico-chemical
The JP should comprise an o‹cial standard being Tests, Panel on Material Sciences and Panel on Prepa-
required to assure the quality of drugs in this country rations were integrated into Panel on Physico-chemi-
in response to the progress in science and technology cal Tests; Panel on Crude Drugs and Provisional First
and clinical demands at the time, it should deˆne the Panel on Crude Drugs were integrated into Panel on
standards for speciˆcations as well as the methods of Crude Drugs; Provisional First Panel on Antibiotics
tests to assure the overall quality of all drugs in princi- to Panel on Antibiotics. Under the Panel on Planning
ple, and it should have a role in clarifying the criteria and Revisions, the following two panels were newly es-
for quality of drugs which are recognized to be im- tablished: Panel on General Revisions and Panel on
portant from the viewpoint of medical treatment. Pharmacopoeial Harmonization (PDG).
At the same time, it was agreed that the JP should In the Committee on Japanese Pharmacopoeia,
be prepared with the aid of the knowledge and ex- Tadao Terao took the role of chairman from Novem-
perience of many persons involved in the pharmaceuti- ber 1997 to December 2000 and Mitsuru Uchiyama
cals, that it should have the characteristics of an o‹- from January 2001 to December 2002.
cial standard, which might be widely used by all par- With the reform of central government ministries
ties concerned, that it should provide information and and agencies in January 2001, the Ministry of Health
understanding about the quality of drugs to the pub- and Welfare became the Ministry of Health, Labour
lic, and that it should be conducive to smooth and and Welfare, and the Committee on Japanese Phar-
eŠective government control of the quality of drugs, macopoeia (CJP) came under the authority of the
i
ii Preface Supplement I, JP XIV

Minister of Health, Labour and Welfare. At the same drafts for the Supplement I, generous cooperation was
time, the Central Pharmaceutical AŠairs Council given by the Technical Committee of the Pharmaceuti-
became the Pharmaceutical AŠairs and Food Sanita- cal Manufacturer's Association of Tokyo and of
tion Council (PAFSC) and Mitsuru Uchiyama was Osaka, the Crude Drugs Association of Tokyo, the
nominated as chairman of the CJP. Japan Pharmaceutical Excipients Council, the Federa-
It was decided that the JP will be revised not only tion of Crude Drugs Associations of Japan, the Japan
every ˆve years, in line with the basic principles for the Antibiotics Research Association, the Japan Flavor
preparation of the JP Fifteenth Edition, but also as and Fragrance Manufacturer's Association, the Japan
necessary to take account of recent progress of science Medical Plants Federation, the Japan Pharmaceutical
and in the interests of international harmonization. In Manufacturer's Association, the Japanese Society of
accordance with the revision principles, the panels Hospital Pharmacists, the Japan Pharmaceutical As-
continued discussions on selection of articles, and re- sociation, and the Japan Oilseed Processors Associa-
visions for General Notices, General Rules for Prepa- tion.
rations, General Tests, and monographs on drugs. In consequence of this revision, the JP Fourteenth
Among the items discussed, the following two revi- Edition carries 881 articles in Part I owing to the addi-
sion drafts were separately examined by the Commit- tion of 31 articles and the deletion of 8 articles; and
tee on JP in November 2001, followed by PAFSC in 481 articles in Part II owing to the addition of 15 arti-
December 2001, and then submitted to the Minister of cles and the deletion of 3 articles.
Health, Labour and Welfare, and Ministerial Notiˆca-
The principles of description and the salient points
tion No.151 promulgated these revisions on March 29,
of the revision in this volume are as follows:
2002: Addition of a paragraph ``In principle, animals
used as a source of materials for preparing phar- 1. The Supplement I to JP Fourteenth Edition
maceutical drugs must be healthy.'' to General No- comprises the following items, in order: Notiˆcation
tices; and Deletion of a monograph ``Phenacetin''. of the Ministry of Health, Labour and Welfare; Con-
Draft revisions covering subjects in General No- tents; Preface; followed by General Notices; General
tices, General Rules for Preparations, General Rules Rules for Preparations; General Tests, Processes and
for Crude Drugs, General Tests, and monographs on Apparatus; Monographs on Drugs in Part I, and
drugs, for which discussions were ˆnished between General Notices; General Rules for Crude Drugs;
June 2000 and February 2001, were prepared for a General Rules for Preparations; General Tests,
supplement to the book. They were examined by the Processes and Apparatus; Monographs on Drugs in
Committee on JP in September 2002, followed by Part II, then followed by Infrared Reference Spectra
PAFSC in December 2002, and then submitted to the in Part I; Ultraviolet-visible Reference Spectra in Part
Minister of Health, Labour and Welfare. I; General Information, and as appendix, the Ministry
Numbers of discussions in the panels to prepare of Health, Labour and Welfare Ministerial Notiˆca-
supplement drafts were as follows: Panel on the Prin- tion No. 151 (March 2002); and Cumulative Index
ciples of Revisions, 6 times; Panel on the Selection of containing references to the main volume and the sup-
Articles, 2 times; Panel on Nomenclature, 11 times; plement I.
Panel on Excipients, 10 times; First Panel on Medici-
2. The articles in General Rules for Preparations,
nal Chemicals, 10 times; Second Panel on Medicinal
in General Tests, Processes and Apparatus, Mono-
Chemicals, 16 times; Panel on Biologically Derived
graphs on Drugs, Infrared Reference Spectra and
Drugs, 8 times; Panel on Biological Tests, 9 times;
Ultraviolet-visible Reference Spectra are respectively
Panel on Physico-chemical Tests, 8 times; Panel on
placed in alphabetical order.
Material Sciences, 8 times; Panel on Preparations, 5
times; Panel on Crude Drugs, 6 times; Provisional 3. The following items in each monograph are put
First Panel on Antibiotics, 27 times; Provisional First in the order shown below, except that unnecessary i-
Panel on Crude Drugs, 7 times. Numbers of discus- tems are omitted depending on the nature of the drug:
sion in the panels, which were renamed due to the re- (1) English title
organization of PAFSC, were as follows: Panel on (2) Commonly used name(s)
Nomenclature for Pharmaceutical Chemicals, 2 times; (3) Latin title (only for Crude Drugs)
Panel on Physico-chemical Tests, 1 time; Panel on (4) Title in Japanese
Crude Drags, 3 times; Panel on General Revisions, 1 (5) Structural formula or empirical formula
time. (6) Molecular formula and molecular mass
It should be noted that in the preparation of the (7) Chemical name
Supplement I, JP XIV Preface iii

(8) Origin unnecessary items are omitted depending on the na-


(9) Limits of the content of the ingredient(s) ture of the drug:
and/or the unit of potency (1) Color
(10) Labeling requirements (2) Odor
(11) Method of preparation (3) Clarity and/or color of solution
(12) Description (4) Acidity or alkalinity
(13) Identiˆcation tests (5) Acid
(14) Speciˆc physical and/or chemical values (6) Alkali
(15) Purity tests (7) Chloride
(16) Loss on drying, loss on ignition, and/or water (8) Sulfate
(17) Residue on ignition, total ash, and/or acid-in- (9) Sulˆte
soluble ash (10) Nitrate
(18) Special tests (11) Nitrite
(19) Isomer ratio (12) Carbonate
(20) Assay or the content of the ingredient(s) (13) Bromide
(21) Containers and storage (14) Iodide
(22) Expiration date (15) Soluble halide
(23) Others (16) Thiocyanide
(17) Selenium
4. In each monograph on a drug, the following
(18) Cationic salts
physical and chemical values representing the proper-
(19) Ammonium
ties and quality of the drug are given in the order indi-
(20) Heavy metals
cated below, except that unnecessary items are omitted
(21) Iron
depending on the nature of the drug:
(22) Manganese
(1) Alcohol number
(23) Chromium
(2) Absorbance
(24) Bismuth
(3) Congealing point
(25) Tin
(4) Refractive index
(26) Aluminum
(5) Osmolarity
(27) Zinc
(6) Optical rotation
(28) Cadmium
(7) Viscosity
(29) Mercury
(8) pH
(30) Copper
(9) Speciˆc gravity
(31) Lead
(10) Boiling point
(32) Silver
(11) Melting point
(33) Alkaline earth metals
(12) Acid value
(34) Arsenic
(13) Saponiˆcation value
(35) Foreign matter
(14) Ester value
(36) Related substances
(15) Hydroxyl value
(37) Other mixtures
(16) Iodine value
(38) Readily carbonizable substances
5. Identiˆcation tests comprise the following
7. Revisions in the General Notices are as follows:
items, which are generally put in the order given be-
(1) In paragraph 6, an abbreviation ``lx'' for lux
low:
was added as the main unit.
(1) Coloration reactions
(2) In paragraph 8, the deˆnition for ``a cold
(2) Precipitation reactions
place'' was revised to as ``a place having a tem-
(3) Decomposition reactions
perature between 1 and 159 C''.
(4) Derivatives
(5) Visible, ultraviolet or infrared spectra 8. Revision in the General Rules for Preparations
(6) Special reactions is as follows:
(7) Cations (1) Injections: The description with respect to the
(8) Anions injection having 50 mL or more was deleted
from section (9).
6. Purity tests comprise the following items, which
are generally put in the order given below, except that 9. The following items of the General Tests,
iv Preface Supplement I, JP XIV

Processes and Apparatus were partially revised: Daunorubicin Hydrochloride


(1) Infrared Spectrophotometry Demethylchlortetracycline Hydrochloride
(2) Residue on Ignition Test Dibekacin Sulfate
(3) Vitamin A Assay Diethanolamine Fusidate
Doxorubicin Hydrochloride
10. The following tests were added to the General
Doxycycline Hydrochloride
Tests, Processes and Apparatus:
Enviomycin Sulfate
(1) Conductivity Measurement
Epirubicin Hydrochloride
(2) Determination of Bulk and Tapped Densities
Flomoxef Triethylammonium
11. The following Reference Standard was delet- Fradiomycin Sulfate
ed: Gentamicin Sulfate
Drostanolone Propionate Gramicidin
Griseofulvin
12. The following Reference Standard was re-
Imipenem
named:
Josamycin Propionate
Kitasamycin to Leucomycine A5
Kanamycin Monosulfate
13. The following Reference Standards were ad- Latamoxef Ammonium
ded: Lenampicillin Hydrochloride
Aceglutamide Lincomycin Hydrochloride
Acetylspiramycin II Lysozyme
Aclarubicin Micronomicin Sulfate
Actinomycin D Mitomycin C
Arbekacin Sulfate Oxytetracycline Hydrochloride
Astromicin Sulfate Peplomycin Sulfate
Bacitracin L-Phenethicillin Potassium
Bekanamycin Sulfate Pimaricin
Benzylpenicillin Potassium Pirarubicin
Benzylpenicillin Sodium Pivmecillinam Hydrochloride
Bleomycin A2 Hydrochloride Polymixin B Sulfate
Carumonam Sodium Puerarin
Cefaclor Pyrrolnitrin
Cefaloridin Ranitidine Hydrochloride
Cefalotin Sodium Retinol Acetate
Cefamandole Lithium Retinol Palmitate
Cefbuperazone Ribostamycin Sulfate
Cefmenoxime Hydrochloride Rifampicin
Cefodizime Sodium Sennoside A
Cefotaxime Sennoside B
Cefotetan Siccanin
Cefotiam Hexetil Hydrochloride Spectinomycin Hydrochloride
Cefoxitin Streptomycin Sulfate
Cefpiramide Sulbenicillin Sodium
Cefpodoxime Proxetil Talampicillin Hydrochloride
Cefroxadine Tobramycin
Cefteram Pivoxil Mesitylenesulfonate Trichomycin
Cefuroxime Axetil Vancomycin Hydrochloride
Chloramphenicol
14. English and Latin titles of drugs are derived,
Chloramphenicol Palmitate
in principle, from International Nonproprietary
Chloramphenicol Succinate
Names (INN) for Pharmaceutical Substances recom-
Ciclacillin
mended by the World Health Organization. Japanese
Clindamycin Hydrochloride
titles are derived from the Japanese version of this
Clindamycin Phosphate
book. The chemical names are based on the rules of
Colistin Sulfate
the International Union of Pure and Applied Chemis-
Supplement I, JP XIV Preface v

try (IUPAC). Erythromycin Lactobionate


Etizolam
15. Molecular formulas of organic compounds be-
Gramicidin
gin with C and then H, followed by other involved ele-
Kanamycin Monosulfate
ments in the alphabetical order of the symbols of the
Ketotifen Fumarate
elements.
Kitasamycin Tartrate
16. Structural formulas of drugs represent, as far Lenampicillin Hydrochloride
as possible, steric conˆgurations. Lysozyme Hydrochloride
O‰oxacin
17. Test procedures in monographs in Part I are,
Phenethicillin Potassium
in principle, written in full even in corresponding
Pimaricin
monographs in Part II, and vice versa. The test proce-
Pirarubicin
dures in monographs for preparations are also written
Pyrrolnitrin
in full even within the same part, except in the mono-
Ranitidine Hydrochloride
graphs for preparations having a corresponding
Siccanin
monograph of their principal material substances.
Sodium Fusidate
18. In O‹cial Monographs, names of some of the Spectinomycin Hydrochloride
reagents and the test solutions were changed to the Trimebutine Maleate
latest names based on the JIS.
Part II
19. The following articles were deleted from O‹- Alpinia O‹cinarum Rhizome
cial Monographs Asparagus Tuber
Part I Burdock Fruit
Drostanolone Propionate Chrysanthemum Flower
Drostanolone Propionate Injection Clematis Root
Floctafenine Eucommia Bark
Iopanoic Acid Fritillaria Bulb
Iopanoic Acid Tablets Gastrodia Tuber
Simˆbrate Hemp Fruit
Sodium Iopodate Jujube Seed
Sodium Iopodate Capsules Loquat Leaf
Magnolia Flower
Part II
Notopterygium Rhizome
Phenovalin and Magnesium Oxide Powder
Polygonum Root
Compound Scopolia Extract and Tannic Acid Oint-
Uncaria Thorn
ment
Compound Scopolia Extract and Tannic Acid Sup- 21. The following monographs were revised by an
positories addition or a change in the Description or other items:
Part I
20. The following articles were newly added to
Acetylkitasamycin
O‹cial Monographs:
Acetylspiramycin
Part I
Aclarubicin Hydrochloride
Aceglutamide Aluminum
Actinomycin D
Bacitracin
Amoxicillin
Benzylpenicillin Benzathine
Ampicillin
Cefodizime Sodium
Anhydrous Ampicillin
Cefpodoxime Proxetil
Ampicillin Sodium
Chloramphenicol Palmitate
Arbekacin Sulfate
Chloramphenicol Sodium Succinate
Astromicin Sulfate
Clindamycin Hydrochloride
Bacampicillin Hydrochloride
Colistin Sulfate
Bekanamycin Sulfate
Daunorubicin Hydrochloride
Benzbromarone
Demethylchlortetracycline Hydrochloride
Benzylpenicillin Potassium
Enviomycin Sulfate
Betamethasone Sodium Phosphate
Epirubicin Hydrochloride
vi Preface Supplement I, JP XIV

Betamethasone Valerate Doxycycline Hydrochloride


Bisacodyl Suppositories Droperidol
Bleomycin Hydrochloride En‰urane
Bleomycin Sulfate Ergocalciferol
Calcium Chloride Injection Erythromycin
Calcium Polystyrene Sulfonate Estradiol Benzoate
d-Camphor Estriol
dl-Camphor Famotidine for Injection
Carbazochrome Sodium Sulfonate Flomoxef Sodium
Carbidopa Flurbiprofen
Carumonam Sodium Folic Acid
Cefaclor Fradiomycin Sulfate
Cefaloridin Gentamicin Sulfate
Cefalotin Sodium Glycerin
Cefamandole Sodium Concentrated Glycerin
Cefbuperazone Sodium Griseofulvin
Cefmenoxime Hydrochloride Haloxazolam
Cefotaxime Sodium Homochlorcyclizine Hydrochloride
Cefotetan Hydrochlorothiazide
Cefotiam Hexetil Hydrochloride Hydrocortisone Acetate
Cefoxitin Sodium Hydrocortisone Sodium Phosphate
Cefpiramide Sodium Idarubicin Hydrochloride
Cefroxadine Idoxuridine Ophthalmic Solution
Cefteram Pivoxil Imipenem
Ceftibuten Indometacin Suppositories
Ceftriaxone Sodium Isoniazid Injection
Cefuroxime Axetil Isoniazid Tablets
Cetraxate Hydrochloride Josamycin
Chloramphenicol Josamycin Propionate
Chlordiazepoxide Powder Kanamycin Sulfate
Chlordiazepoxide Tablets Kitasamycin
Chlormadinone Acetate Latamoxef Sodium
Chlorphenesin Carbamate Lincomycin Hydrochloride
Ciclacillin Liothyronine Sodium Tablets
Citric Acid Magnesium Sulfate Injection
Anhydrous Citric Acid D-Mannitol Injection
Clindamycin Phosphate Menatetrenone
Clomifene Citrate Mepitiostane
Cloxacillin Sodium Methotrexate
1z Codeine Phosphate Powder Meticrane
10z Codeine Phosphate Powder Micronomicin Sulfate
Codeine Phosphate Tablets Migrenin
Dibekacin Sulfate Minocycline Hydrochloride
Diclofenamide Mitomycin C
Diethylcarbamazine Citrate Tablets Morphine Hydrochloride
1z Dihydrocodeine Phosphate Powder Morphine Hydrochloride Injection
10z Dihydrocodeine Phosphate Powder Morphine Hydrochloride Tablets
Dihydroergotoxine Mesilate Norepinephrine
Diltiazem Hydrochloride Norepinephrine Injection
Dipyridamole Norgestrel and Ethinylestradiol Tablets
Dobutamine Hydrochloride Oxytetracycline Hydrochloride
Dopamine Hydrochloride Injection Peplomycin Sulfate
Doxorubicin Hydrochloride Pethidine Hydrochloride
Supplement I, JP XIV Preface vii

Phytonadione Cod Liver Oil


Pipemidic Acid Trihydrate Coptis Rhizome
Piperacillin Sodium Powdered Coptis Rhizome
Pivmecillinam Hydrochloride Gardenia Fruit
Polymixin B Sulfate Glycyrrhiza
Potassium Clavulanate Powdered Glycyrrhiza
Povidone-Iodine Imperata Rhizome
Prednisolone Acetate Powdered Japanese Gentian
Prednisolone Sodium Succinate for Injection Magnolia Bark
Primidone Powdered Magnolia Bark
Procaine Hydrochloride Injection Morphine and Atropine Injection
Procarbazine Hydrochloride Mulberry Bark
Pyrazinamide Opium Alkaloids Hydrochlorides
Reserpine Powdered Panax Rhizome
Retinol Acetate Peony Root
Retinol Palmitate Powdered Peony Root
Ribostamycin Sulfate Phellodendron Bark
Rifampicin Powdered Phellodendron Bark
Roxithromycin Compound Phellodendron Powder for Cataplasm
Scopolamine Butylbromide Phellodendron, Albumin Tannate and Bismuth
Sisomicin Sulfate Subnitrate Powder
Sodium Bicarbonate Injection Pueraria Root
Sodium Chloride Rhubarb
10z Sodium Chloride Injection Powdered Rhubarb
Streptomycin Sulfate Scopolia Extract and Ethyl Aminobenzoate Powder
Sulbenicillin Sodium Scutellaria Root
Talampicillin Hydrochloride Powdered Scutellaria Root
Tegafur Senna Leaf
Teicoplanin Powdered Senna Leaf
Theophylline Sinomenium Stem
Thiamine Hydrochloride Swertia Herb
Thiamine Nitrate Powdered Swertia Herb
Thiopental Sodium Swertia and Sodium Bicarbonate Powder
Tipepidine Hibenzate Toad Venom
Tobramycin Vitamin A Oil
Tocopherol Vitamin A Oil Capsules
Tocopherol Acetate
22. The monographs, which use in Identiˆcation
Triamcinolone
the newly introduced Infrared Reference Spectra, are
Trichomycin
as follows:
Trimetoquinol Hydrochloride
Part I
Vancomycin Hydrochloride
Acetylspiramycin
Xylitol Injection
Aclarubicin Hydrochloride
Part II Ampicillin
Acacia Anhydrous Ampicillin
Powdered Acacia Ampicillin Sodium
Aloe Benzylpenicillin Benzathine
Powdered Aloe Benzylpenicillin Potassium
Areca Bleomycin Hydrochloride
Asiasarum Root Bleomycin Sulfate
Capsicum Carumonam Sodium
Powdered Capsicum Cefaclor
Cinnamon Bark Cefaloridin
Powdered Cinnamon Bark Cefalotin Sodium
viii Preface Supplement I, JP XIV

Cefamandole Sodium Benzbromarone


Cefmenoxime Hydrochloride Benzylpenicillin Benzathine
Cefodizime Sodium Benzylpenicillin Potassium
Cefotaxime Sodium Bleomycin Hydrochloride
Cefotetan Bleomycin Sulfate
Cefpodoxime Proxetil Carumonam Sodium
Cefuroxime Axetil Cefaclor
Chloramphenicol Cefaloridin
Chloramphenicol Sodium Succinate Cefalotin Sodium
Ciclacillin Cefamandole Sodium
Citric Acid Cefbuperazone Sodium
Anhydrous Citric Acid Cefmenoxime Hydrochloride
Clindamycin Phosphate Cefodizime Sodium
Demethylchlortetracycline Hydrochloride Cefotaxime Sodium
Doxorubicin Hydrochloride Cefotetan
Doxycycline Hydrochloride Cefotiam Hexetil Hydrochloride
Erythromycin Cefoxitin Sodium
Etizolam Cefpiramide Sodium
Flomoxef Sodium Cefpodoxime Proxetil
Glycerin Cefroxadine
Concentrated Glycerin Cefteram Pivoxil
Griseofulvin Ceftriaxone Sodium
Homochlorcyclizine Hydrochloride Cefuroxime Axetil
Imipenem Chloramphenicol
Ketotifen Fumarate Chloramphenicol Palmitate
Kitasamycin Tartrate Chloramphenicol Sodium Succinate
Latamoxef Sodium Cloxacillin Sodium
Lenampicillin Hydrochloride Daunorubicin Hydrochloride
Lincomycin Hydrochloride Demethylchlortetracycline Hydrochloride
Mitomycin C Doxorubicin Hydrochloride
Norepinephrine Enviomycin Sulfate
O‰oxacin Epirubicin Hydrochloride
Phenethicillin Potassium Etizolam
Phytonadione Flomoxef Sodium
Pivmecillinam Hydrochloride Gramicidin
Pyrazinamide Griseofulvin
Pyrrolnitrin Homochlorcyclizine Hydrochloride
Ranitidine Hydrochloride Imipenem
Rifampicin Josamycin
Siccanin Josamycin Propionate
Sodium Fusidate Ketotifen Fumarate
Sulbenicillin Sodium Kitasamycin Tartrate
Talampicillin Hydrochloride Latamoxef Sodium
Theophylline Lysozyme Hydrochloride
Trimebutine Maleate Minocycline Hydrochloride
Vancomycin Hydrochloride Mitomycin C
Norepinephrine
23. The monographs, which use in Identiˆcation
O‰oxacin
the newly introduced Ultraviolet-visible Reference
Oxytetracycline Hydrochloride
Spectra, are as follows:
Peplomycin Sulfate
Part I
Phenethicillin Potassium
Acetylspiramycin
Pimaricin
Aclarubicin Hydrochloride
Pirarubicin
Actinomycin D
Supplement I, JP XIV Preface ix

Potassium Clavulanate Gentamicin Sulfate


Pyrrolnitrin Glycerin
Ranitidine Hydrochloride Concentrated Glycerin
Rifampicin Griseofulvin
Siccanin Imipenem
Theophylline Josamycin
Trimebutine Maleate Josamycin Propionate
Vancomycin Hydrochloride Kanamycin Sulfate
Latamoxef Sodium
24. The following monographs were revised in ori-
Lincomycin Hydrochloride
gin:
Micronomicin Sulfate
Part I
Mitomycin C
Acetylspiramycin
Oxytetracycline Hydrochloride
Aclarubicin Hydrochloride
Peplomycin Sulfate
Actinomycin D
Pipemidic Acid Trihydrate
Amoxicillin
Pivmecillinam Hydrochloride
Ampicillin
Polymixin B Sulfate
Anhydrous Ampicillin
Potassium Clavulanate
Ampicillin Sodium
Procarbazine Hydrochloride
Arbekacin Sulfate
Pyrazinamide
Astromicin Sulfate
Retinol Acetate
Bacampicillin Hydrochloride
Retinol Palmitate
Bekanamycin Sulfate
Ribostamycin Sulfate
Benzbromarone
Rifampicin
Benzylpenicillin Potassium
Sodium Chloride
Bleomycin Hydrochloride
Streptomycin Sulfate
Bleomycin Sulfate
Sulbenicillin Sodium
Calcium Chloride Injection
Talampicillin Hydrochloride
Carumonam Sodium
Teicoplanin
Cefaclor
Tobramycin
Cefaloridin
Trichomycin
Cefalotin Sodium
Trimetoquinol Hydrochloride
Cefamandole Sodium
Vancomycin Hydrochloride
Cefbuperazone Sodium
Cefmenoxime Hydrochloride Part II
Cefotaxime Sodium Aloe
Cefotetan Cod Liver Oil
Cefotiam Hexetil Hydrochloride Evodia Fruit
Cefoxitin Sodium Gardenia Fruit
Cefpiramide Sodium Pueraria Root
Cefroxadine Senna Leaf
Cefteram Pivoxil Powdered Senna Leaf
Cefuroxime Axetil Vitamin A Oil
Chloramphenicol Vitamin A Oil Capsules
Ciclacillin
Citric Acid
Anhydrous Citric Acid
Clindamycin Phosphate
Dibekacin Sulfate
Doxorubicin Hydrochloride
Doxycycline Hydrochloride
Erythromycin
Flomoxef Sodium
Fradiomycin Sulfate
x Preface Supplement I, JP XIV

Those who were engaged in the preparation of the Takashi Morita Hisashi Sonobe
Supplement I to JP Fourteenth Edition are as follows: Toshimi Murai Shoko Sueyoshi
Shigeru Muraki Hisakazu Sunada
Norio Aimi Yoshiaki Kato
Emi Nakajima Hideyo Suzuki
Mitsuo Aoki Mitsunori Katoh
Hiroshi Nakamura Senji Suzuki
Kiichi Aonuki Noriko Katori
Tatsuya Nakano Yukio Tabuchi
Nobuo Aoyagi Nobuo Kawahara
Hiroyuki Nakazawa Yoshikazu Takahashi
Yoshichika Arakawa Toru Kawanishi
Masaaki Naotsuka Tadahiro Takeda
Keiko Arimoto Toshiaki Kawanishi
Masao Nasu Yasushi Takeda**
Kazuhide Ashizawa Nana Kawasaki
Koji Nishijima Hiroshi Tokunaga
Shinichiro Aso Toshisuke Kawasaki
Motohiro Nishijima Toyoshige Tanabe
Kunihiro Fujita Yoshiaki Kawashima
Tatsumi Nishiyama Haruo Tanaka
Hiroshi Fujiwara Keiji Kijima
Takashi Nomoto Toshihiro Tanaka
Goro Funamoto Takao Kiyohara
Hiroyasu Ogata Kenichi Tanamoto
Yukihiro Goda Toyohiko Kobayashi
Yoshiyuki Ogawa Tsuyoshi Tanimoto
Morio Hamashima Masayoshi Kohase
Masaru Ohno Susumu Terabayashi
Ruri Hanajiri Shigeo Kojima
Yasuo Ohno Tadao Terao*
Kouji Hasegawa Hiroyasu Kokubo
Yoshiro Ohtani Hiroshi Terashima
Ryuichi Hasegawa Seizo Kondo
Masakazu Ootani Kunikazu Teshima
Takao Hayakawa Hideki Kumakura
Minoru Okada Kiyoshi Tomioka
Masahiro Hayashi Takao Kunisada
Satoshi Okada Motowo Tomita
Fusayoshi Hirayama Mitsuo Kurashige
Tsuneo Okubo Tatsuru Tomizawa
Yukio Hiyama Takeshi Kurata
Haruhiro Okuda Nobuchika Tsumagari
Kunimoto Hotta Masaaki Kurihara
Masami Otsuka Mitsuru Uchiyama*
Takanori Ichikawa Haruo Kuriyama
Tadashi Ouchi Yoshimasa Uehara
Nobukazu Igoshi Fumiyo Kusu
Kazuhiko Sagara Takashi Unno
Toshio Imanari Masako Maeda
Eiji Sakai Morimasa Yagisawa
Kenichi Inui Tamio Maitani
Hideki Sasaki Takehiko Yajima
Mumio Ishibashi Toshio Masaoka
Tsuguo Sasaki Teruhide Yamaguchi
Shigeru Itai Toshihiko Matsubara
Motoyoshi Satake Keiichi Yamamoto
Mitsuo Ito Yoshihisa Matsuda
Akihiro Sato Keiji Yamamoto
Yuji Ito Norio Matsuki
Setsuko Sekita Kenichi Yamazaki
Takashi Itoh Shigeru Matsuki
Yasuo Shimada Takeshi Yamazaki
Shozo Iwagami Hayashi Matsukura
Kesamitsu Shimizu Hikaru Yoden
Akemi Kai Katsutoshi Mise
Kyoko Shimura Chikako Yomota
Kazuaki Kakehi Hiroto Miyamoto
Fumitoshi Shincho Hitoo Yoshida
Shozo Kamiya Naoki Miyata
Osamu Shirota Kazumasa Yoshikawa
Takemine Kanai Michinao Mizugaki
Kouichi Shudo Sumie Yoshioka
Nahoko Kaniwa Kaoru Morikawa
Motoko Kanke Osamu Morita *Chairman, Committee on JP
**Acting Chairman, Committee on JP
General Notices
Change the paragraph 6 to read: Change the paragraph 8 to read:
6. The following abbreviations are used for the 8. Standard temperature, ordinary temperature,
main units. room temperature, and lukewarm are deˆned as 209C,
meter m 15 – 259 C, 1 – 309C, and 30 – 409C, respectively. A
centimeter cm cold place, unless otherwise speciˆed, shall be a place
millimeter mm having a temperature of 1 – 159C.
micrometer mm The temperatures of cold water, lukewarm water,
nanometer nm warm water, and hot water are deˆned as not exceed-
kilogram kg ing 109 C, 30 – 409C, 60 – 709C, and about 1009C,
gram g respectively.
milligram mg The term ``heated solvent'' or ``hot solvent'' means
microgram mg a solvent heated almost to the boiling point of the sol-
nanogram ng vent, and the term ``warmed solvent'' or ``warm sol-
picogram pg vent'' usually means a solvent heated to a temperature
Celsius degree 9 C between 609C and 709C. The term ``heat on or in a
square centimeter cm2 water bath'' indicates, unless otherwise speciˆed,
liter L heating with a boiling water bath or a steam bath at
milliliter mL about 1009C.
microliter mL Cold extraction and warm extraction are usually
megahertz MHz performed at temperatures of 15 – 259 C and 35 – 45
per centimeter cm-1 9C, respectively.
newton N
kilopascal kPa
mole per liter mol/L
millipascal second mPa・s
square millimeter second mm2/s
lux lx
mass per cent z
mass parts per million ppm
mass parts per billion ppb
volume per cent volz
volume parts per million vol ppm
mass per volume per cent w/vz
hydrogen ion concentration pH
Endotoxin unit EU
Note: ``ppm'' used in the Nuclear Magnetic
Resonance Spectroscopy (1H) indicates the chemical
shift, and ``w/vz'' is used in the formula or compo-
sition of preparations.

1359
General Rules
for Preparations
11. Injections
Change the paragraph (9) to read:
(9) Unless otherwise speciˆed, Injections meet the
requirements of the Sterility Test. In the case of drugs
to be dissolved before use, carry out the test with the
solution obtained by dissolving the contents in the at-
tached solvent.

1361
General Tests, Processes
and Apparatus
soluble ash, extract content, essential oil content of crude
Change to read: drugs are performed as directed in the corresponding items
under the Crude Drugs Test.
General Tests, Processes and Apparatus includes common
methods for tests and other articles related to them. Unless
otherwise speciˆed, the procedures for absorbance determi-
nation, absorbance ratio determination, acid-neutralizing 23. Infrared Spectrophotometry
capacity determination of gastrointestinal medicines, alco-
Change the Instrument and adjustment to read:
hol number determination, ammonium determination, ar-
senic determination, atomic absorption spectrophotometry, Instrument and adjustment
test for bacterial endotoxins, boiling point determination, Several models of dispersive infrared spectrophotometers
distilling range determination, chloride determination, con- or Fourier-transform infrared spectrophotometers are avail-
ductivity measurement, congealing point determination, test able.
for content uniformity, determination of bulk and tapped The instruments, adjusted according to the instruction
densities, digestion test, disintegration test, dissolution test, manual of each individual instrument, should comply with
endpoint detection in titrimetry, ‰ame coloration, ‰uoro- the following test for resolving power, transmittance repro-
metry, foreign insoluble matter test for injections, gas chro- ducibility and wave number reproducibility. When the spec-
matography, heavy metals determination, infrared spec- trum of a polystyrene ˆlm about 0.04 mm thick is recorded,
trophotometry, insoluble particulate matter test for injec- the depth of the trough from the maximum absorption at
tions, insoluble particulate matter test for ophthalmic solu- about 2850 cm-1 to the minimum at about 2870 cm-1 should
tions, iron determination, liquid chromatography, loss on be not less than 18z transmittance and that from the maxi-
drying determination, loss on ignition determination, mass mum at about 1583 cm-1 to the minimum at about 1589
variation test, melting point determination, methanol deter- cm-1 should be not less than 12z transmittance.
mination, methoxyl assay, test for microbial limit, test for The wave number (cm-1) scale is usually calibrated by the
microbial limit for crude drugs, microbiological potency de- use of several characteristic absorption wave numbers
termination for antibiotics, mineral oil determination, nitro- (cm-1) of a polystyrene ˆlm shown below. The number in
gen determination, nuclear magnetic resonance spec- parentheses indicates the permissible range.
troscopy, optical rotation determination, osmolarity deter-
3060.0 (±1.5) 2849.5 (±1.5) 1942.9 (±1.5)
mination, oxygen ‰ask combustion method, paper chro-
1601.2 (±1.0) 1583.0 (±1.0) 1154.5 (±1.0)
matography, particle size distribution test for preparations,
1028.3 (±1.0)
pH determination, powder particle size determination, test
for pyrogen, qualitative test, test for readily carbonizable When the dispersive infrared spectrophotometer is used,
substances, refractive index determination, residual solvents the permissible range of the absorption wave unmbers at
test, residue on ignition determination, speciˆc gravity and 1601.2 cm-1 and at 1028.3 cm-1 should be both within ±2.0
density determination, speciˆc surface area determination, cm-1.
test for sterility, sulfate determination, test for glass contain- As the repeatability of transmittance and wave number,
ers for injections, test for metal particles in ophthalmic oint- the diŠerence of transmittance should be within 0.5z when
ments, test for plastic containers, test for rubber closure for the spectrum of a polystyrene ˆlm is measured twice at sever-
aqueous infusions, test for total organic carbon, thermal al wave numbers from 3000 to 1000 cm-1, and the diŠerence
analysis, thin-layer chromatography, viscosity determina- of wave number should be withun 5 cm-1 at about 3000
tion, vitamin A assay, test for volatile contaminants in cm-1 and within 1 cm-1 at about 1000 cm-1.
ethanol, water determination, and X-ray powder diŠraction
are performed as directed in the corresponding articles under
the General Tests, Processes and Apparatus. The tests for Change to read:
melting point of fats, congealing point of fatty acids, speciˆc
gravity, acid value, saponiˆcation value, ester value, 52. Residue on Ignition Test
hydroxyl value, unsaponiˆable matter and iodine value of
fats and fatty oils are performed as directed in the corre-
The Residue on Ignition Test is a method to measure the
sponding items under the Fats and Fatty oils Test, and the mass of the residual substance not volatilized when the sam-
tests for foreign matter and loss on drying, total ash, acid-in- ple is ignited with sulfuric acid by the method described be-

1363
1364 General Tests, Processes and Apparatus Supplement I, JP XIV

low. Generally, this test is intended for determining the con- Generally, for synthetic vitamin A esters apply Method
tent of inorganic substances contained as impurities in an or- 1-1 or Method 1-2, but if the assay conditions required for
ganic substance. Method 1-1 are not suitable, apply Method 2.
The description, for example, ``not more than 0.10z (1
Method 1-1
g),'' in a monograph, indicates that the mass of the residue is
Weigh accurately about 0.1 g of the sample, and dissolve
not more than 1.0 mg per 1 g of the substance in the test in
in 2-propanol for vitamin A assay to make exactly 50 mL.
which about 1 g of the substance is weighed accurately and
Dilute this solution with 2-propanol for vitamin A assay to
ignited by the procedure described below, and ``after
make a solution so that each mL contains 10 to 15 vitamin A
drying'' indicates that the sample is tested after being dried
Units, and use this solution as the sample solution. Deter-
under the conditions speciˆed in the test for Loss on drying.
mine the absorption spectrum of the sample solution be-
Procedure tween 220 nm and 400 nm as directed under the Ultraviolet-
Previously ignite a crucible of platinum, quartz or por- visible Spectrophotometry to obtain the wavelength of the
celain at 600 ± 509C for 30 minutes, and weigh accurately maximum absorption and the absorbances at 300 nm, 310
after cooling in a desiccator (silica gel). nm, 320 nm, 326 nm, 330 nm, 340 nm and 350 nm. When
Take the sample of the amount directed in the mono- the maximum absorption lies between 325 nm and 328 nm,
graph, transfer into the ignited crucible, and weigh accurate- and the ratios, Ali/A326, of each absorbance, Ali, at 300 nm,
ly. When the quantity of the sample to be taken is indicated 310 nm, 320 nm, 330 nm, 340 nm and 350 nm to the absor-
in a volume, pipet exactly the amount directed in the mono- bance, A326, at 326 nm are within the range of ±0.030 of the
graph and transfer into the above crucible. When directed as values in the Table, the potency of vitamin A in Units per g
``after evaporating,'' heat properly to evaporate the solu- of the sample is calculated from the following equation.
tion. A V
Units of vitamin A in 1 g = 326 × × 1900
Moisten the sample with a small amount of sulfuric acid, W 100
usually 1 mL, then heat slowly at a temperature as low as A326: Absorbance at 326 nm
practicable until the sample is completely carbonized, and V: Total volume (mL) of the sample solution
cool. Moisten again with a small amount of sulfuric acid, W: Amount (g) of sample in V mL of the sample solution
heat gently until white fumes are no longer evolved, and ig- 1900: Conversion factor from speciˆc absorbance of
nite at 600 ± 509 C until the residue is completely incinerat- retinol ester to IU (Unit/g)
ed. Proceed with care to not burn with a ‰ame. Cool the
This method is applied to drugs or preparations contain-
crucible in a desiccator (silica gel), and reweigh accurately to
ing vitamin A esters (retinol acetate or retinol palmitate) as
calculate the amount of the residue.
the main component. However, when the wavelength of
When the amount of the residue obtained above exceeds
maximum absorption does not lie between 325 nm and 328
the limit speciˆed in the monograph, unless otherwise speci-
nm, or when the absorbance ratio Ali/A326 is not within the
ˆed, ignite repeatedly to constant mass.
range of ±0.030 of the values in the Table, apply Method 2.
Table Absorbance Ratio, Ali/A326, of retinol acetate and
Change to read: retinol palmitate

Ali/A326
66. Vitamin A Assay li (nm)
Retinol acetate Retinol palmitate
The Vitamin A Assay is a method to determine vitamin A 300 0.578 0.590
in Retinol Acetate, Retinol Palmitate, Vitamin A Oil, Cod 310 0.815 0.825
Liver Oil and other preparations. Method 1 is for the assay 320 0.948 0.950
of synthetic vitamin A esters, using the ultraviolet-visible 330 0.972 0.981
spectrophotometry (Method 1-1) or the liquid chro- 340 0.786 0.795
matography (Method 1-2). Method 2 is for the assay of vita- 350 0.523 0.527
min A of natural origin, containing many geometrical
isomers, using the ultraviolet-visible spectrophotometry to Method 1-2
determine vitamin A as vitamin A alcohol obtained by Proceed with an appropriate amount of sample as directed
saponiˆcation in an alkaline solution and extraction. under the Liquid Chromatography.
One Vitamin A Unit (equal to 1 vitamin A I.U.) is equiva- For the assay of retinol acetate and retinol palmitate use
lent to 0.300 mg of vitamin A (all-trans vitamin A alcohol). Retinol Acetate Reference Standard and Retinol Palmitate
Reference Standard, respectively, and ˆx appropriately the
Procedure
operating procedure, the operating conditions and the sys-
All procedures should be carried out quickly and care
tem suitability based on the characteristics of the substance
should be taken as far as possible to avoid exposure to light,
to be tested and the species and amount of coexisting sub-
air, oxidants, oxidizing catalysts (e.g. copper, iron), acids
stances.
and heat. If necessary, light-resistant vessels may be used.
Supplement I, JP XIV General Tests, Processes and Apparatus 1365

Method 2 70. Reference Standards;


Unless otherwise speciˆed, weigh accurately a sample con-
taining not less than 500 Units of vitamin A, and not more Reagents, Test Solutions; Standard
than 1 g of fat, transfer to a ‰ask, and add 30 mL of alde- Solutions for Volumetric Analysis;
hyde-free ethanol and 1 mL of a solution of pyrogallol in
ethanol (95) (1 in 10). Then add 3 mL of a solution of potas- Standard Solutions; Matching
sium hydroxide (9 in 10), attach a re‰ux condenser, and heat Fluids for Color; Optical Filters for
on a water bath for 30 minutes to saponify. Cool quickly to
ordinary temperature, add 30 mL of water, transfer to a Wavelength and Transmission Rate
separator A, wash the ‰ask with 10 mL of water and then 40 Calibration; and Measuring
mL of diethyl ether, transfer the washings to the separator
A, shake well, and allow to stand. Transfer the water layer Instruments, Appliances
so obtained to a separator B, wash the ‰ask with 30 mL of
diethyl ether, add the washing to the separator B, and ex- (1) Reference Standards
tract by shaking. Transfer the water layer to a ‰ask, add the
diethyl ether layer to the separator A, transfer the water lay-
er in the ‰ask to the separator B, add 30 mL of diethyl ether,
Delete the following Reference Standards:
and extract by shaking. Transfer the diethyl ether layer so Drostanolone Propionate, Kitasamycin
obtained to the separator A, add 10 mL of water, allow the
separator A to stand after gentle turning upside-down 2 or 3
times, and remove the water layer. Wash the content of the Add the following:
separator A with three 50-mL portions of water with increas-
Aceglutamide, Acetylspiramycin II, Aclarubicin, Actino-
ingly vigorous shaking as the washing proceeds. Further
mycin D, Arbekacin Sulfate, Astromicin Sulfate, Bacitracin,
wash with 50-mL portions of water until the washing no lon-
Bekanamycin Sulfate, Benzylpenicillin Potassium, Benzyl-
ger shows a pink color with phenolphthalein TS, and allow
penicillin Sodium, Bleomycin A2 Hydrochloride, Carumon-
to stand for 10 minutes. Remove remaining water as far as
am Sodium, Cefaclor, Cefaloridin, Cefalotin Sodium, Cefa-
possible, transfer the diethyl ether to an Erlenmeyer ‰ask,
mandole Lithium, Cefbuperazone, Cefmenoxime Hydro-
wash the separator with two 10-mL portions of diethyl ether,
chloride, Cefodizime Sodium, Cefotaxime, Cefotetan,
add the washings to the ‰ask, add 5 g of anhydrous sodium
Cefotiam Hexetil Hydrochloride, Cefoxitin, Cefpiramide,
sulfate to the ‰ask, mix by shaking, and transfer the diethyl
Cefpodoxime Proxetil, Cefroxadine, Cefteram Pivoxil Mesit-
ether to a round-bottomed ‰ask by decantation. Wash the
ylenesulfonate, Cefuroxime Axetil, Chloramphenicol, Chlo-
remaining sodium sulfate in the ‰ask with two or more
ramphenicol Palmitate, Chloramphenicol Succinate, Ciclacil-
10-mL portions of diethyl ether, and transfer the washings
lin, Clindamycin Hydrochloride, Clindamycin Phosphate,
to the ‰ask. Evaporate the diethyl ether in a water bath at 45
Colistin Sulfate, Daunorubicin Hydrochloride, Demethyl-
9C while swirling the ‰ask, using an aspirator, to about 1
chlortetracycline Hydrochloride, Dibekacin Sulfate, Diethan-
mL, immediately add an exactly measured amount of 2-
olamine Fusidate, Doxorubicin Hydrochloride, Doxycycline
propanol for vitamin A assay to make a solution containing
Hydrochloride, Enviomycin Sulfate, Epirubicin Hydrochlo-
6 to 10 vitamin A Units per mL, and designate the solution
ride, Flomoxef Triethylammonium, Fradiomycin Sulfate,
as the sample solution. Determine the absorbances, A310 at
Gentamicin Sulfate, Gramicidin, Griseofulvin, Imipenem,
310 nm, A325 at 325 nm, and A334 at 334 nm, of the sample
Josamycin Propionate, Kanamycin Monosulfate, Latamoxef
solution as directed under the Ultraviolet-visible Spec-
Ammonium, Lenampicillin Hydrochloride, Leucomycin A5,
trophotometry.
Lincomycin Hydrochloride, Lysozyme, Micronomicin Sul-
Units of vitamin A in 1 g of the sample fate, Mitomycin C, Oxytetracycline Hydrochloride, Pepl-
A V omycin Sulfate, L-Phenethicillin Potassium, Pimaricin,
= 325 × × f × 1830
W 100 Pirarubicin, Pivmecillinam Hydrochloride, Polymixin B Sul-
A A fate, Puerarin, Pyrrolnitrin, Ranitidine Hydrochloride,
f = 6.815 - 2.555 × 310 - 4.260 × 334
A325 A325 Retinol Acetate, Retinol Palmitate, Ribostamycin Sulfate,
Rifampicin, Sennoside A, Sennoside B, Siccanin, Spec-
A325: Absorbance at 325 nm
tinomycin Hydrochloride, Streptomycin Sulfate, Sulbenicil-
V: Total volume (mL) of the sample solution
lin Sodium, Talampicillin Hydrochloride, Tobramycin,
W: Amount (g) of sample in V mL of the sample solution
Trichomycin, Vancomycin Hydrochloride
f: Correction factor
1830: Conversion factor from speciˆc absorbance of
retinol alcohol to IU (Unit/g)
(2) Reagents, Test Solutions
Delete the following Solution:
0.1 mol/L Phosphate buŠer solution for ceftibuten, pH
8.0
1366 General Tests, Processes and Apparatus Supplement I, JP XIV

Change the following: Each mL of 0.01 mol/L sulfuric acid VS


= 0.8509 mg of C7H10N2O3
Arecoline hydrobromide for thin-layer chromatography
C8H13NO2.HBr White crystals. Freely soluble in water, Acetaminophen C8H9NO2 [Same as the namesake
soluble in methanol, and practically insoluble in diethyl monograph]
ether.
Acetate buŠer solution, pH 5.4 To 5.78 mL of acetic
Melting point: 169 – 1719C
acid (100) add water to make 1000 mL (solution A). Dissolve
Purity Related substances—Dissolve 50 mg of arecoline
8.2 g of anhydrous sodium acetate in water to make 1000
hydrobromide for thin-layer chromatography in exactly 10
mL (solution B). Mix 176 mL of the solution A and 824 mL
mL of methanol. Perform the test with 10 mL of this solution
of the solution B, and adjust, if necessary, the pH to 5.4
as directed in the Identiˆcation under Areca: any spot other
with the solution A or the solution B.
than the principal spot at the Rf value of about 0.4 does not
appear. 0.02 mol/L Acetic acid-sodium acetate TS Dissolve 2.74
g of sodium acetate trihydrate in a suitable amount of water,
N-Demethylroxithromycin C40H74N2O15 White pow-
and add 2 mL of acetic acid (100) and water to make 1000
der.
mL.
Identiˆcation—Determine the infrared absorption spec-
trum of a solution of the substance to be tested in chlo- Albi‰orin C23H28O11.x H2O Colorless powder having
roform (1 in 20) as directed in the solution method under the no odor. Freely soluble in water and in methanol, and prac-
Infrared Spectrophotometry using a 0.1-mm cell made of tically insoluble in diethyl ether.
potassium bromide: it exhibits absorption at the wave num- Purity—Dissolve 1 mg in diluted methanol (1 in 2) to
bers of about 3600 cm-1, 3520 cm-1, 3450 cm-1, 3340 cm-1, make 10 mL, and use this solution as the sample solution.
1730 cm-1 and 1627 cm-1. Perform the test with 10 mL of the sample solution as direct-
ed in the Assay under Peony Root; the total area of the
Geniposide for thin-layer chromatography C17H24O10
peaks other than albi‰orin and other than the solvent is not
White crystals or crystalline powder. Melting point: 159 –
larger than 1/10 of the total area of the peaks other than the
1639 C.
solvent peak.
Purity Related substances—Dissolve 1.0 mg of genipo-
side for thin-layer chromatography in exactly 1 mL of 0.05 mol/L Ammonium formate buŠer solution, pH 4.0
methanol, and perform the test with 20 mL of this solution as Dissolve 3.5 g of ammonium formate in about 750 mL of
directed in the Identiˆcation (2) under Gardenia Fruit: any water, adjust the pH to 4.0 with formic acid, and add water
spot other than the principal spot at the Rf value of about to make 1000 mL.
0.3 does not appear.
Amoxicillin C16H19N3O5S.3H2O [Same as the name-
Imidazole TS Dissolve 8.25 g of imidazole in 65 mL of sake monograph]
water, adjust the pH to 6.8 with 5 mol/L hydrochloric acid
a-Apooxytetracycline C22H22N2O8 Yellow-brown to
TS, and add water to make 100 mL.
green powder.
Melting point : 200 – 2059C
Add the following:
b-Apooxytetracycline C22H22N2O8 Yellow-brown to
2-Acetamidoglutarimide C7H10N2O3: 170.17
brown powder.
Identiˆcation—Determine the infrared absorption spec-
Purity Related substances—Dissolve 8 mg of b-apoox-
trum of 2-acetamidoglutarimide as directed in the potassium
ytetracycline in 5 mL of 0.01 mol/L sodium hydroxide TS,
bromide disk method under the Infrared Spectrophotomet-
add 0.01 mol/L hydrochloric acid TS to make 100 mL, and
ry: it exhibits absorption at the wave numbers of about 3350
use this solution as the sample solution. Proceed the test
cm-1, 1707 cm-1, 1639 cm-1 and 1545 cm-1.
with 20 mL of the sample solution as directed in the Purity
Purity Related substances—Dissolve 10 mg of 2-
(2) under Oxytetracycline Hydrochloride, determine each
acetamidoglutarimide in 100 mL of the mobile phase, and
peak area by the automatic integration method, and calcu-
use this solution as the sample solution. Pipet 1 mL of the
late the amounts of them by the area percentage method: the
sample solution, add the mobile phase to make exactly 100
total amount of the peaks other than β-apooxytetracycline
mL, and use this solution as the standard solution. Proceed
is not more than 10z.
with 20 mL each of the sample solution and the standard so-
lution as directed in the Purity (3) under Aceglutamide Alu- L-Arabinose C5H10O5 [K 8054: 1991, L(+)-Arabinose,
minum: the total of the peak areas other than 2- Special class]
acetamidoglutarimide from the sample solution is not more
Aristolochic acid I for crude drugs purity test
than the peak area from the standard solution.
C17H11NO7 Yellow crystalline powder. Melting point:
Content : not less than 98.0z. Assay—Weigh accurately
about 2809 C (with decomposition).
about 20 mg of 2-acetamidoglutarimide, and perform the z
Absorbance E 11cm (318 nm): 384 – 424 (1 mg, methanol,
test as directed under the Nitrogen Determination.
100 mL).
Purity Related substances—Dissolve 1.0 mg of aristo-
Supplement I, JP XIV General Tests, Processes and Apparatus 1367

lochic acid I for crude drugs purity test in 100 mL of diluted Benzalphthalide C15H10O2 Yellow crystalline powder.
methanol (3 in 4), and use this solution as the sample solu- Melting point: 99 – 1029C.
tion. Pipet 1 mL of this solution, add diluted methanol (3 in
4-Chlorobenzenediazonium TS Dissolve 0.5 g of 4-chlo-
4) to make exactly 100 mL, and use this solution as the stan-
roaniline in 1.5 mL of hydrochloric acid, and add water to
dard solution. Perform the test with exactly 10 mL each of
make 100 mL. To 10 mL of this solution add 10 mL of sodi-
the sample solution and the standard solution as directed un-
um nitrite TS and 5 mL of acetone. Prepare before use.
der the Liquid Chromatography according to the following
conditions, and determine each peak area by the automatic Cinnamic acid C9H8O2 White crystalline powder, hav-
integration method: the total area of the peaks other than ing a characteristic odor.
aristolochic acid I obtained from the sample solution is not Melting point : 132 – 1359C
more than the peak area of aristolochic acid I from the stan-
Citric acid-phosphate-acetonitrile TS Dissolve 2.1 g of
dard solution.
citric acid monohydrate, 13.4 g of dipotassium hydrogen
Operating conditions
phosphate and 3.1 g of potassium dihydrogen phosphate in
Detector, column, column temperature, mobile phase,
1000 mL of a mixture of water and acetonitrile (3:1).
and ‰ow rate: Proceed as directed in the operating condi-
tions in the Purity (3) under Asiasarum Root. N-Demethylerythromycin C36H65NO13 White to light
Time span of measurement: About 3 times as long as the yellowish white powder.
retention time of aristolochic acid I after the solvent peak.
Deuterated formic acid for nuclear magnetic resonance
System suitability
spectroscopy DCOOD Prepared for nuclear magnetic
Proceed as directed in the system suitability in the Purity
resonance spectroscopy.
(3) under Asiasarum Root.
Deuterated pyridine for nuclear magnetic resonance spec-
Barbaloin for component determination Use barbaloin
troscopy C5D5N Prepared for nuclear magnetic
for thin-layer chromatography meeting the following addi-
resonance spectroscopy.
tional speciˆcations.
z
Absorbance E 11cm (360 nm): 260 – 290 [10 mg dried in a Dibekacin sulfate [Same as the namesake monograph]
desiccator (in vacuum, phosphorus (V) oxide) for not less
1,3-Dihydroxynaphthalene C10H8O2 Purple-brown,
than 24 hours, methanol, 500 mL].
crystals or powder. Freely soluble in water and in ethanol
Purity Related substances—Dissolve 10 mg of the sub-
(95).
stance to be tested in 10 mL of methanol, and use this solu-
Melting point : about 1259C
tion as the sample solution. Pipet 1 mL of the sample solu-
tion, add methanol to make exactly 100 mL, and use this so- N,N-Dimethylacetamide CH3CON(CH3)2 Clear and
lution as the standard solution (1). Perform the test with colorless liquid.
exactly 20 mL each of the sample solution and the standard Boiling point : 163 – 1659C
solution (1) as directed under the Liquid Chromatography Speciˆc gravity : 0.938 – 0.945 (Method 3).
according to the following conditions, and measure each Water : not more than 0.2z (0.1 g, Coulometric titra-
peak area of the both solutions by the automatic integration tion).
method: the total area of the peaks other than barbaloin Purity—Perform the test with 3 mL of N, N-
from the sample solution is not larger than the peak area of dimethylacetamide as directed under the Gas Chro-
barbaloin from the standard solution (1). matography according to the following conditions, deter-
Operating conditions mine each peak area by the automatic integration method,
Proceed the operating conditions in the Component deter- and calculate the amount of N, N-dimethylacetamide by the
mination under Aloe except wavelength, detection sensitivity area percentage method: not less than 98.0z.
and time span of measurement. Operating conditions
Wavelength: 300 nm Detector: A hydrogen ‰ame-ionization detector.
Detection sensitivity: Pipet 1 mL of the standard solution Column: A fused silica column 0.25 mm in inside di-
(1), add methanol to make exactly 20 mL, and use this solu- ameter and 30 m in length, coated the inside surface 0.5 mm
tion as the standard solution (2). Adjust the detection sen- in thickness with polyethylene glycol 20 M for gas chro-
sitivity so that the peak area of barbaloin obtained from 20 matography.
mL of the standard solution (2) can be measured by the auto- Column temperature: The sample is injected at a constant
matic integration method and the peak height of barbaloin temperature of about 709C, keep this temperature for 1
obtained from 20 mL of the standard solution (1) shows minute, then raise to 2009 C in a rate of 109C per minute,
about 20z of the full scale. and keep 2009 C for 3 minutes.
Time span of measurement: About 3 times as long as the Carrier gas: Helium
retention time of barbaloin after the solvent peak. Flow rate (linear velocity): About 30 cm/sec.
Time span of measurement: About 2 times as long as the
Becanamycin sulfate [Same as the namesake mono-
retention time of N, N-dimethylacetamide.
graph]
System suitability
1368 General Tests, Processes and Apparatus Supplement I, JP XIV

Test for required detection: To exactly 1.0 g of N, N- total amount of the peaks other than 4-epioxytetracycline is
dimethylacetamide add acetone to make exactly 100 mL. not more than 10z.
Pipet 5 mL of this solution, and add acetone to make exactly
Erythromycin B C37H67NO12 White to light yellowish
50 mL. Conˆrm that the peak area of N, N-
white powder.
dimethylacetamide obtained from 3 mL of this solution is e-
Purity Related substances—Dissolve 10 mg of erythro-
quivalent to 40 to 60z of the full-scale.
mycin B in 1 mL of methanol, add a mixture of phosphate
System repeatability: When the test is repeated with 3 mL
buŠer solution, pH 7.0 and methanol (15:1) to make 5 mL,
of N, N-dimethylacetamide under the above operating condi-
and use this solution as the sample solution. Pipet 1 mL of
tions, the relative standard deviation of the peak area of
the sample solution, add a mixture of phosphate buŠer solu-
N, N-dimethylacetamide is not more than 2.0z.
tion, pH 7.0 and methanol (15:1) to make exactly 20 mL,
4-Dimethylaminoantipyrine C13H17N3O Colorless or and use this solution as the standard solution. Proceed with
white crystals, or a white crystalline powder. exactly 100 mL each of the sample solution and the standard
Purity—Proceed the test with 5 mL of a solution of 4- solution as directed in the Purity (3) Related substances un-
dimethylaminoantipyrine (1 in 2000) as directed in the Assay der Erythromycin, and determine each peak area from the
under Cefpiramide Sodium, determine each peak area in a solutions by the automatic integration method: the total of
range of about 2 times as long as the retention time of 4- areas of the peaks other than erythromycin B from the sam-
dimethylaminoantipyrine after the solvent peak by the auto- ple solution is not more than the peak area of erythromycin
matic integration method, and calculate the total amount of B from the standard solution.
the peaks other than 4-dimethylaminoantipyrine by the area
Erythromycin C C36H65NO13 White to light yellowish
percentage method: not more than 1.0z.
white powder.
9,10-Diphenylanthracene C26H18 Yellow crystalline Purity Related substances—Dissolve 10 mg of erythro-
powder. Soluble in diethyl ether, and practically insoluble in mycin C in 1 mL of methanol, add a mixture of phosphate
water. buŠer solution, pH 7.0 and methanol (15:1) to make 5 mL,
Melting point : about 2489C and use this solution as the sample solution. Pipet 1 mL of
the sample solution, add a mixture of phosphate buŠer solu-
1,4-Diphenylbenzene C18H14 White scaly crystals, hav-
tion, pH 7.0 and methanol (15:1) to make exactly 20 mL,
ing a slight aromatic odor. It is freely soluble in ethanol
and use this solution as the standard solution. Proceed with
(99.5), and slightly soluble in water.
exactly 100 mL each of the sample solution and the standard
Identiˆcation—Determine the infrared absorption spec-
solution as directed in the Purity (3) Related substances un-
trum of 1,4-diphenylbenzene as directed in the potassium
der Erythromycin, and determine each peak area from the
bromide disk method under the Infrared Spectrophotomet-
solutions by the automatic integration method: the total of
ry: it exhibits absorption at the wave numbers of about 3050
areas of the peaks other than erythromycin C from the sam-
cm-1, 3020 cm-1, 1585 cm-1, 1565 cm-1, 1476 cm-1, 1450
ple solution is not more than the peak area of erythromycin
cm-1, 995 cm-1, 834 cm-1, 740 cm-1 and 680 cm-1.
C from the standard solution.
Divinylbenzene-methacrylate co-polymer for liquid chro-
3-Ethoxy-4-hydroxybenzaldehyde C9H10O3 White to
matography Prepared for liquid chromatography.
pale yellowish white crystalline. Freely soluble in ethanol
6-Epidoxycycline hydrochloride C22H24N2O8.HCl (95), and slightly soluble in water.
Yellow to dark yellow, crystals or crystalline powder. Melting point : 76 – 789C
Purity Related substances—Dissolve 20 mg of 6-epiox- Content : not less than 98.0z. Assay—Weigh accurately
ycycline hydrochloride in 25 mL of 0.01 mol/L hydrochloric about 0.3 g of 3-ethoxy-4-hydroxybenzaldehyde, previously
acid TS, and use this solution as the sample solution. Pro- dried in a desiccator (phosphorous (V) oxide) for 4 hours,
ceed the test with 20 mL of the sample solution as directed in dissolve in 50 mL of N, N-dimethylacetamide, and titrate
the Purity (2) under Doxycycline Hydrochloride, determine with 0.1 mol/L sodium methoxide VS (indicator: thymol
each peak area by the automatic integration method, and blue TS).
calculate the amounts of them by the area percentage
Each mL of 0.1 mol/L sodium methoxide VS
method: the total area of the peaks other than 6-epidoxycy-
= 16.62 mg of C9H10O3
cline is not more than 10z.
2-Formylbenzoic acid CHOC6H4COOH White crys-
4-Epioxytetracycline C22H24N2O9 Green-brown to bro-
tals. Melting point: 97 – 999C
wn powder.
Content : not less than 99.0z. Assay—Weigh accurately
Purity Related substances—Dissolve 20 mg of 4-epiox-
about 0.3 g of 2-formylbenzoic acid, previously dried (in
ytetracycline in 25 mL of 0.01 mol/L hydrochloric acid TS,
vacuum, phosphorus (V) oxide, 3 hours), dissolve in 50 mL
and use this solution as the sample solution. Proceed the test
of freshly boiled and cooled water, and titrate with 0.1
with 20 mL of the sample solution as directed in the Purity
mol/L sodium hydroxide VS (indicator: 3 drops of phenol
(2) under Oxytetracycline Hydrochloride, determine each
red TS).
peak area by the automatic integration method, and calcu-
late the amounts of them by the area percentage method: the
Supplement I, JP XIV General Tests, Processes and Apparatus 1369

Each mL of 0.1 mol/L sodium hydroxide VS 0.05 mol/L Hydrochloric acid-methanol TS To 100 mL
= 15.01 mg of C8H6O3 of 0.5 mol/L hydrochloric acid add methanol to make 1000
mL.
Geniposide for component determination Use genipo-
side for thin-layer chromatography meeting the following 1-(2-Hydroxyethyl)-1H-tetrazol-5-thiol C3H6N4OS
additional speciˆcations. White, crystals or powder.
z
Absorbance E 11cm (240 nm): 249 – 269 [10 mg dried in a Melting point : 136 – 1419C
desiccator (reduced pressure of not exceeding 0.67 kPa, Purity Related substances—Dissolve 0.10 g of 1-(2-
phosphorus (V) oxide) for 24 hours, diluted methanol (1 in hydroxyethyl)-1H-tetrazol-5-thiol in 1 mL of water, and use
2), 500 mL]. this solution as the sample solution. Pipet 0.5 mL of the
Purity Related substances—Dissolve 5 mg of geniposide sample solution, add water to make exactly 25 mL, and use
for component determination in 50 mL of diluted methanol this solution as the standard solution. Perform the test with
(1 in 2), and use this solution as the sample solution. Pipet 1 these solutions as directed under the Thin-layer Chro-
mL of the sample solution, add diluted methanol (1 in 2) to matography. Spot 1 mL each of the sample solution and the
make exactly 100 mL, and use this solution as the standard standard solution on a plate of silica gel with ‰uorescent in-
solution (1). Perform the test with exactly 10 mL each of the dicator for thin-layer chromatography, develop with a mix-
sample solution and the standard solution (1) as directed un- ture of ethyl acetate, water, methanol and formic acid
der the Liquid Chromatography according to the following (60:10:7:6) to a distance of about 10 cm, and air-dry the
conditions, and measure each peak area of the both solu- plate. Examine under ultraviolet light (main wavelength: 254
tions by the automatic integration method: the total area of nm): the spot other than the principal spot obtained from
the peaks other than geniposide from the sample solution is the sample solution is not more intense than the spot from
not larger than the peak area of geniposide from the stan- the standard solution.
dard solution (1).
Isoniazid C6 H 7 N 3 O [Same as the namesake mono-
Operating conditions
graph]
Proceed as directed in the Component determination un-
der Gardenia Fruit except detection sensitivity and time span Josamycin C42H69NO15 [Same as the namesake mono-
of measurement. graph]
Detection sensitivity: Pipet 1 mL of the standard solution
Josamycin propionate C45H73NO16 [Same as the
(1), add diluted methanol (1 in 2) to make exactly 20 mL,
namesake monograph]
and use this solution as the standard solution (2). Adjust the
detection sensitivity so that the peak area of geniposide ob- Lactobionic acid C12H22O12 Colorless crystals or white
tained from 10 mL of the standard solution (2) can be meas- crystalline powder, having no odor.
ured by the automatic integration method and the peak Melting point : 113 – 1189C
height of geniposide obtained from 10 mL of the standard Purity—Dissolve 0.10 g of lactobionic acid in 10 mL of a
solution (1) shows about 20z of the full scale. mixture of methanol and water (3:2), and perform the test
Time span of measurement: About 3 times as long as the with 10 mL of this solution as directed in the Identiˆcation
retention time of geniposide after the solvent peak. (2) under Erythromycin Lactobionate: the spot other than
the principal spot is not found.
Guaifenesin C10H14O4 [Same as the namesake mono-
graph] Luteolin for thin-layer chromatography C15H10O6
Light yellow to yellow-brown crystalline powder. Slightly
Hirsutine C22H28N2O3 White to light yellow, crystals
soluble in methanol and in ethanol (99.5), and practically in-
or crystalline powder. Melting point: about 1009 C.
soluble in water. Melting point: about 3109C (with decom-
Optical rotation [a]20D : about + 57 9 (10 mg, methanol, 1
position).
mL).
z Purity Related substances—Dissolve 1.0 mg of luteolin
Absorbance E 11cm (245 nm): 354 – 379 (5 mg calculated
for thin-layer chromatography in 1 mL of methanol. Pro-
on the anhydrous basis, a mixture of methanol and dilute
ceed the test with 10 mL of this solution as directed in the
acetic acid (7:3), 500 mL).
Identiˆcation under Chrysanthemum Flower: any spot other
Purity Related substances-Dissolve 5 mg of hirsutine
than the principal spot of Rf about 0.7 does not appear.
in 100 mL of a mixture of methanol and dilute acetic acid
(7:3), proceed with 20 mL of this solution as directed in the D-Mannose C6H12O6 White crystal or crystalline pow-
Component determination under Uncaria Thorn, and per- der. It is very soluble in water.
form the Liquid Chromatography: the sum of the peak areas Melting point: about 1329C (with decomposition).
except the areas of hirsutine and the solvent is not more than Optical rotation [a]20D : +13.7 – +14.79(4 g, diluted am-
1/10 of the sum of the peak areas except the solvent. monia TS (1 in 200), 20 mL, 100 mm).

0.01 mol/L Hydrochloric acid-methanol TS To 20 mL Metacycline hydrochloride C22H22N2O8.HCl Yellow


of 0.5 mol/L hydrochloric acid TS add methanol to make to dark yellow, crystals or crystalline powder.
1000 mL. Purity Related substances—Dissolve 20 mg of metacy-
cline hydrochloride in 25 mL of 0.01 mol/L hydrochloric
1370 General Tests, Processes and Apparatus Supplement I, JP XIV

acid TS, and use this solution as the sample solution. Pro- to light green-yellowish white, crystals or crystalline powder.
ceed the test with 20 mL of the sample solution as directed in Identiˆcation—Determine the infrared absorption spec-
the Purity (2) under Doxycycline Hydrochloride, determine trum of o‰oxacin demethyl substance as directed in the
each peak area by the automatic integration method, and potassium bromide disk method under the Infrared Spec-
calculate the amounts of them by the area percentage trophotometry: it exhibits absorption at the wave numbers
method: the total area of peaks other than metacycline is not of about 3050 cm-1, 2840 cm-1, 1619 cm-1, 1581 cm-1,
more than 10z. 1466 cm-1, 1267 cm-1, 1090 cm-1, 1051 cm-1 and 816
cm-1.
1-Methyl-1H-tetrazole-5-thiol for liquid chromatography
C2H4N4S White, crystals or crystalline powder. Very solu- Phenol red TS, dilute To 235 mL of a solution of ammo-
ble in methanol, and freely soluble in water. nium nitrate (1 in 9400) add 105 mL of 2 mol/L sodium
Melting point : 123 – 1279C hydroxide TS and 135 mL of a solution prepared by dissolv-
Loss on drying: not more than 1.0z (1 g, in vacuum, ing 24 g of acetic acid (100) in water to make 200 mL. To this
phosphorous (V) oxide, 2 hours). solution add 25 mL of a solution prepared by dissolving 33
Content : not less than 99.0z. Assay—Weigh accurately mg of phenol red in 1.5 mL of 2 mol/L sodium hydroxide
about 0.2 g of 1-methyl-1H-tetrazole-5-thiol, previously d- TS and adding water to make 100 mL. If necessary, adjust
ried, dissolve in 80 mL of N, N-dimethylformamide, and ti- the pH to 4.7.
trate with 0.1 mol/L sodium methoxide VS (indicator: 3
50z Phenyl-50z methylpolysiloxane for gas chro-
drops of thymol blue-N, N-dimethylformamide TS). Per-
matography Prepared for gas chromatography.
form a blank determination, and make any necessary correc-
tion. 0.01 mol/L Phosphate buŠer solution, pH 6.8 Dissolve
1.36 g of potassium dihydrogen phosphate in 900 mL of
Each mL of 0.1 mol/L sodium methoxide VS
water, adjust the pH to 6.8 with 0.2 mol/L sodium
= 11.61 mg of C2H4N4S
hydroxide TS, and add water to make 1000 mL.
Morphine Hydrochloride [Same as the namesake mono-
0.03 mol/L Phosphate buŠer solution, pH 7.5 Dissolve
graph]
4.083 g of potassium dihydrogen phosphate in 800 mL of
2-Naphthalenesulfonic acid C10H8O3S.H2O White to water, adjust the pH to 7.5 with 0.2 mol/L sodium
pale yellowish white powder. Very soluble in water, in hydroxide TS, and add water to make 1000 mL.
methanol and in ethanol (95), and sparingly soluble in
1/15 mol/L Phosphate buŠer solution, pH 5.6 Dissolve
diethyl ether and in chloroform.
9.07 g of potassium dihydrogen phosphate in about 750 mL
Water : 7.0 – 11.5z (0.5 g, volumetric titration, direct
of water, adjust the pH to 5.6 with potassium hydroxide TS,
titration).
and add water to make 1000 mL.
Content : not less than 95.0z, calculated on the anhy-
drous basis. Assay—Weigh accurately about 0.5 g of 2- 0.1 mol/L Phosphate buŠer solution, pH 5.3 Dissolve
naphthalenesulfonic acid, dissolve in 30 mL of water, and ti- 0.44 g of disodium hydrogen phosphate 12-water and 13.32
trate with 0.1 mol/L sodium hydroxide VS (indicator: 3 g of potassium dihydrogen phosphate in 750 mL of water,
drops of bromothymol blue TS). Perform a blank determi- adjust the pH to 5.3 with sodium hydroxide TS or phosphor-
nation, and make any necessary correction. ic acid, and add water to make 1000 mL.

Each mL of 0.1 mol/L soduim hydroxide VS 0.1 mol/L Phosphate buŠer solution, pH 6.8 Dissolve
= 20.82 mg of C10H8O3S 6.4 g of potassium dihydrogen phosphate and 18.9 g of diso-
dium hydrogen phosphate 12-water in about 750 mL of
1-Naphthol-sulfuric acid TS Dissolve 1.5 g of 1-
water, adjust the pH to 6.8 with sodium hydroxide TS if
naphthol in 50 mL of ethanol (95), add 3 mL of water and 7
necessary, and add water to make 1000 mL.
mL of sulfuric acid, and mix well. Prepare before use.
0.1 mol/L Phosphate buŠer solution for antibiotics, pH
Ninhydrin-acetic acid TS Dissolve 1.0 g of ninhydrin in
8.0 Dissolve 16.73 g of dipotassium hydrogen phosphate
50 mL of ethanol (95), and add 10 mL of acetic acid (100).
and 0.523 g of potassium dihydrogen phosphate in about
Octadecylsilanized silica gel for thin-layer chro- 750 mL of water, adjust the pH to 8.0 with phosphoric acid,
matography Octadecylsilanized silica gel prepared for and add water to make 1000 mL.
thin-layer chromatography.
Phosphate buŠer solution, pH 3.1 Dissolve 136.1 g of
Octadecylsilanized silica gel with ‰uorescent indicator for potassium dihydrogen phosphate in 500 mL of water, and
thin-layer chromatography Octadecylsilanized silica gel add 6.3 mL of phosphoric acid and water to make 1000 mL.
for thin-layer chromatography containing ‰uorescent indi-
Phosphate buŠer solution, pH 5.9 Dissolve 6.8 g of
cator.
potassium dihydrogen phosphate in 800 mL of water, adjust
O‰oxacin demethyl substance (±)-9-Fluoro-2,3-dihydro- the pH to 5.9 with diluted potassium hydroxide TS (1 in 10),
3-methyl-7-oxo-7H-10-(1-piperazinyl)-pirido[1, 2, 3-de] [1, and add water to make 1000 mL.
4]benzoxazine-6-carboxylic acid C17H18FN3O4 White
Supplement I, JP XIV General Tests, Processes and Apparatus 1371

Phosphate buŠer solution, pH 6.2 Dissolve 9.08 g of of water, adjust the pH to 3.5 with phosphoric acid, and add
potassium dihydrogen phosphate in 1000 mL of water (solu- water to make 1000 mL.
tion A). Dissolve 9.46 g of disodium hydrogen phosphate in
0.33 mol/L Potassium dihydrogen phosphate TS Dis-
1000 mL of water (solution B). Mix 800 mL of the solution
solve 4.491 g of potassium dihydrogen phosphate in water to
A and 200 mL of the solution B, and adjust the pH to 6.2
make 100 mL.
with the solution A or the solution B if necessary.
Potassium iodide TS, saturated Saturate 20 g of potassi-
Phosphate buŠer solution for antibiotics, pH 6.5
um iodide in 10 mL of ‰eshly boiled and cooled water. Pre-
Dissolve 10.5 g of disodium hydrogen phosphate 12-water
pare before use.
and 5.8 g of potassium dihydrogen phosphate in 750 mL of
water, adjust the pH to 6.5 with sodium hydroxide TS, and 2-Propanol for vitamin A assay (CH3)2CHOH [K
add water to make 1000 mL. 8839, Special class] When the absorbances at 300 nm and
between 320 nm and 350 nm are determined as directed un-
Phosphate TS Dissolve 2.0 g of dipotassium hydrogen
der the Ultraviolet-visible Spectrophotometry, using water
phosphate and 8.0 g of potassium dihydrogen phosphate in
as the control, they are not more than 0.05 and not more
water to make 1000 mL.
than 0.01, respectively. If necessary, purify by distillation.
Phthalein purple C32H32N2O12.x H2O Yellowish white
Ranitidinediamine (C10H18N2OS)2.C4H4O4 White to
to brown power. Soluble in ethanol (95), and practically in-
pale yellow crystalline powder.
soluble in water.
Identiˆcation—Determine the infrared absorption spec-
Sensitivity test—Dissolve 10 mg of phthalein purple in 1
trum of ranitidinediamine as directed in the paste method
mL of ammonia solution (28), and add water to make 100
under the Infrared Spectrophotometry: it exhibits absorp-
mL. To 5 mL of this solution add 95 mL of water, 4 mL of
tion at the wave numbers of about 2780 cm-1, 1637 cm-1,
ammonia solution (28), 50 mL of ethanol (95) and 0.1 mL of
1015 cm-1 and 788 cm-1.
diluted barium chloride TS (1 in 5): the solution shows a
Content : not less than 95z. Assay—Weigh accurately
blue-purple color which disappears on the addition of 0.15
about 0.1 g of ranitidinediamine, dissolve in 50 mL of acetic
mL of 0.1 mol/L disodium dihydrogen ethylenediamine
acid (100), and titrate with 0.1 mol/L perchloric acid VS un-
tetraacetate TS.
til the color of the solution changes from purple to green
Phthalimide C8H5NO2 White to pale brown crystals or through blue (indicator: crystal violet TS). Perform the
powder. blank determination in the same manner, and make any
Melting point : 232 – 2379C necessary correction.
Clarity—1.0 g of phthalimide dissolves in 20 mL of sodi-
Each mL of 0.1 mol/L perchloric acid VS
um hydroxide TS as a slight turbid solution.
= 13.62 mg of (C10H18N2OS)2.C4H4O4
Content : not less than 98.0z. Assay—Weigh accurately
about 0.3 g of the substance to be tested, dissolve in 40 mL Resibufogenin for thin-layer chromatography
of N, N-dimethylformamide, and titrate with 0.1 mol/L so- C24H32O4.nH2O White crystalline powder having no odor.
dium methoxide VS (potentiometric titration). Perform a It is freely soluble in acetone and in methanol.
blank determination, and make any necessary correction. Purity Related substances—Dissolve 5.0 mg of the sub-
stance to be tested in exactly 5 mL of acetone. Perform the
Each mL of 0.1 mol/L sodium methoxide VS
test with 5 mL of this solution as directed in the Identiˆcation
= 14.71 mg of C8H5NO2
under Toad Venom: no other spots than the principal spot
Polyethylene glycol 600 for gas chromatography Pre- of around Rf 0.4 appear.
pared for gas chromatography.
L-Rhamnose monohydrate C6H 12O.H2O White crys-
Polyethylene glycol 1500 for gas chromatography Pre- talline powder having sweet taste. Freely soluble in water,
pared for gas chromatography. and sparingly soluble in ethanol (95).
Optical rotation [a]20 D : +7.8 – +8.39 (1 g, 20 mL of
Polyoxyethylene (23) lauryl ether
water, 2 drops of ammonia TS, 100 mm).
C12H25(OCH2CH2)nOH White masses. Melting point:
Melting point : 87 – 919C
about 409C
Purity Related substances—Dissolve 1.0 mg of L-rham-
Potassium chloride for conductivity measurement nose monohydrate in 1 mL of water, and add methanol to
[K 8121, Potassium chloride for conductivity measurement] make exactly 10 mL. Proceed with 20 mL of this solution as
directed in the Identiˆcation (2) under Acacia: any spot
0.1 mol/L Potassium dihydrogen phosphate TS, pH 2.0
other than the principal spot at the Rf value of about 0.5
Dissolve 13.6 g of potassium dihydrogen phosphate in water
does not appear.
to make 1000 mL. Adjust the pH to 2.0 with phosphoric
acid. Rhynchophylline for component determination
C22H28N2O4 White, crystals or crystalline powder. Spar-
0.25 mol/L Potassium dihydrogen phosphate TS, pH 3.5
ingly soluble in ethanol (99.5) and in acetone, and practically
Dissolve 34 g of potassium dihydrogen phosphate in 900 mL
insoluble in water. Melting point: 205 – 2099C
1372 General Tests, Processes and Apparatus Supplement I, JP XIV
z
Absorbance E 11cm (245 nm): 473 – 502 (5 mg dried in a this solution as the sample solution. Perform the test with
desiccator (silica gel) for 24 hours, a mixture of methanol this solution as directed under the Thin-layer Chro-
and dilute acetic acid (7:3), 500 mL). matography. Spot 5 mL of the sample solution on a plate of
Purity Related substances— silica gel with ‰uorescent indicator for thin-layer chro-
(1) Dissolve 1.0 mg of rhynchophylline for component matography. Develop the plate with a mixture of ethyl
determination in 1 mL of acetone, and perform the test with acetate, acetone, water and acetic acid (100) (10:2:1:1) to a
this solution as directed under the Thin-layer Chro- distance of about 10 cm, and air-dry the plate. Examine un-
matography. Spot 10 mL of the solution on a plate of silica der ultraviolet light (main wavelength: 254 nm): any spot
gel with ‰uorescent indicator for thin-layer chro- other than the principal spot does not appear.
matography, develop with a mixture of 1-butanol, water and
Sodium 2-naphthalenesulfonate C10H7NaO3S Pale
acetic acid (100) (7:2:1) to a distance of about 10 cm, and
brown, crystals or powder.
air-dry the plate. Examine under ultraviolet light (main
Content: not less than 98.0z.
wavelength: 254 nm): any spot other than the principal spot
of Rf about 0.5 does not appear. Sodium periodate TS Dissolve 60.0 g of sodium perio-
(2) Dissolve 5 mg of rhynchophylline for component de- date in 120 mL of 0.05 mol/L sulfuric acid TS, and add
termination in 100 mL of a mixture of methanol and dilute water to make 1000 mL. If the solution is not clear, ˆlter this
acetic acid (7:3), and use this solution as the sample solution. through a glass-ˆlter. Keep in a light-resistant vessel.
Pipet 1 mL of the sample solution, add a mixture of
Strongly acidic ion-exchange silica gel for liquid chro-
methanol and dilute acetic acid (7:3) to make exactly 100
matography Prepared for liquid chromatography.
mL, and use this solution as the standard solution. Perform
the test with exactly 20 mL each of the sample solution and Substrate solution for lysozyme hydrochloride To a suit-
the standard solution as directed under the Liquid Chro- able amount of dried cells of Micrococcus luteus add a suita-
matography according to the following conditions. Deter- ble amount of phosphate buŠer solution, pH 6.2, gently
mine each peak area obtained from these solutions by the shake to make a suspension, and add the substrate cells or
automatic integration method: the sum of the peak areas ex- the same buŠer solution so that the absorbance of the sus-
cept the areas of rhynchophylline and the solvent obtained pension at 640 nm is about 0.65. Prepare before use.
from the sample solution is not more than the peak area of
Tetracycline Hydrochloride C22H24N2O8.HCl Yellow,
rhynchophylline from the standard solution.
crystals or crystalline powder.
Operating conditions
Purity Related substances—Dissolve 20 mg of tetracy-
Detector, column, column temperature, mobile phase,
cline hydrochloride in 25 mL of 0.01 mol/L hydrochloric
and ‰ow rate: Proceed as directed in the operating condi-
acid TS, and use this solution as the sample solution. Pro-
tions in the Component determination under Uncaria
ceed the test with 20 mL of the sample solution as directed in
Thorn.
the Purity (2) under Oxytetracycline Hydrochloride, deter-
Time span of measurement: About 4 times as long as the
mine each peak area by the automatic integration method,
retention time of rhynchophylline after the solvent peak.
and calculate the amounts of them by the area percentage
System suitability
method: the total amount of the peaks other than tetracy-
Test for required detectability: Measure exactly 1 mL of
cline is not more than 10z.
the standard solution, add a mixture of methanol and dilute
acetic acid (7:3) to make exactly 20 mL. Conˆrm that the Tetra‰uoroethylene polymer for gas chromatography
peak area of rhynchophylline obtained from 20 mL of this Prepared for gas chromatography.
solution is equivalent to 3.5 to 6.5z of that from 20 mL of
Tetrahydrofuran for liquid chromatography C4H8O
the standard solution.
Clear and colorless liquid.
System performance, and system repeatability: Proceed as
Refractive index n20 D : 1.406 – 1.409
directed in the operating conditions in the Component deter-
Density (209C): 0.884 – 0.889 g/mL
mination under Uncaria Thorn.
Purity Ultraviolet absorbing substances—Determine the
Sodium dihydrogen phosphate TS, pH 2.5 Dissolve 2.7 absorption spectrum of tetrahydrofuran for liquid chro-
g of sodium dihydrogen phosphate dihydrate in 1000 mL of matography as directed under the Ultraviolet-visible Spec-
water, and adjust the pH to 2.5 with phosphoric acid. trophotometry, using water as the blank: the absorbences at
240 nm, 254 nm, 280 nm, 290 nm, and between 300 nm and
6 mol/L Sodium hydroxide TS Dissolve 252 g of sodium
400 nm are not more than 0.35, 0.20, 0.05, 0.02 and 0.01, re-
hydroxide in water to make 1000 mL. Preserve in a polyethy-
spectively.
lene bottle.
Peroxide—Perform the test according to the method de-
Sodium 1-methyl-1H-tetrazole-5-thiolate scribed in JIS K 9705: not more than 0.01z.
C2H3N4NaS.2H2O White, crystals or crystalline powder.
Tetrakishydroxypropylethylenediamine for gas chro-
Melting point : 90 – 949C
matography Prepared for gas chromatography.
Purity Related substances—Dissolve 10 mg of sodium 1-
methyl-1H-tetrazole-5-thiolate in 10 mL of water, and use
Supplement I, JP XIV General Tests, Processes and Apparatus 1373

Tetra-n-propylammonium bromide precipitate on the ˆlter paper with water until the last wash-
[CH3CH2CH2]4NBr White, crystals or crystalline powder. ing shows no turbidity with silver nitrate TS, transfer the
Purity Clarity and color of solution—Dissolve 1.0 g of precipitate together with the ˆlter paper to a tared crucible,
tetra-n-propylammonium bromide in 20 mL of water: the and then heat strongly to ashes. After cooling, add 2 drops
solution is clear and colorless. of sulfuric acid, and heat again at about 7009C for 2 hours.
Content : not less than 98.0z. Assay—Weigh accurately After cooling, weigh accurately the mass of the residue, and
about 0.4 g of tetra-n-propylammonium bromide, dissolve calculate the molarity factor as barium sulfate (BaSO4).
in 50 mL of water, add 5 mL of dilute nitric acid, and titrate
Each mL of 0.1 mol/L barium chloride VS
with 0.1 mol/L silver nitrate VS while shaking strongly
= 23.34 mg of BaSO4
(potentiometric titration).

Each mL of 0.1 mol/L silver nitrate VS


= 26.63 mg of C12H28NBr Add the following:
Thymine C5H6N2O2: 126.11
Identiˆcation—Determine the infrared absorption spec-
72. Conductivity Measurement
trum of thymine, previously dried at 1059C for 3 hours, as
directed in the potassium bromide disk method under the In- Conductivity measurement is a method for the measuring
frared Spectrophotometry: it exhibits absorption at the wave the ‰owability of electric current in an aqueous solution.
numbers of about 3030 cm-1, 1734 cm-1, 1676 cm-1, 1446 The measurement is made with a conductivity meter or a re-
cm-1 and 814 cm-1. sistivity meter, and is used, for example, in the purity tests in
Purity Related substances—Dissolve 50 mg of thymine monographs. The method is applied to evaluate the test item
in 100 mL of methanol. To 10 mL of this solution add the ``Conductivity (Electrical Conductivity)'' speciˆed in the
mobile phase to make 100 mL, and use this solution as the monographs. Further it is also used for monitoring the qual-
sample solution. Proceed with 10 mL of the sample solution ity of water in the preparation of highly puriˆed water.
as directed in the Purity (3) under Aceglutamide Aluminum: However, when applying this method for monitoring the
any peak does not appear at the retention time of quality of water, the details of measurement should be speci-
aceglutamide. ˆed by the user, based on the principles described here.
Conductivity of a solution k (S・m-1) is deˆned as the
Tin (II) chloride-hydrochloric acid TS To 20 g of tin add reciprocal of resistivity r (Q・m), which is an indicator of the
85 mL of hydrochloric acid, heat until hydrogen gas no lon- strength of ionic conductivity for a ‰uid conductor. Resistiv-
ger are evolved, and allow to cool. Mix 1 volume of this so- ity is deˆned as the product of electrical resistance per unit
lution and 10 volume of dilute hydrochloric acid. Prepare length and cross-sectional area of a conductor. When resis-
before use. tivity is r, cross-section area A (m2), and length l (m),
Triethylamine buŠer solution, pH 3.2 To 4 mL of resistance R (Q) can be expressed by the following equation.
triethylamine add 2000 mL of water, and adjust the pH to R = r(l/A )
3.2 with phosphoric acid.
Thus, conductivity k is expressed as follows,
Trimethylsilanized silica gel for liquid chromatography
Prepared for liquid chromatography. k = 1/r = (1/R )(l/A )

Water, sterile puriˆed [Same as the namesake mono- If l/A is known, the conductivity k can be obtained by
graph in Part II] measuring resistance R or conductance G (= R-1).
In the International System (SI), the unit of conductivity
is the Siemens per meter (S・m-1). In practice, conductivity
(3) Standard Solutions
of a solution is generally expressed by m S・cm-1, and resistiv-
for Volumetric Analysis ity by Q・cm.
Unless otherwise speciˆed, the reference temperature for
Add the following:
the expression of conductivity or resistivity is 209C.
Barium chloride, 0.1 mol/L
Apparatus
1000 mL of this solution contains 24.426 g of barium
A conductivity meter or a resistivity meter is composed of
chloride dihydrate (BaCl2.2H2O: 244.26).
an indicator part (operating panel, display, recording unit)
Preparation—Dissolve 24.5 g of barium chloride dihy-
and a detector part, the latter of which includes a conductivi-
drate in water to make 1000 mL, and standardize the solu-
ty cell. In the conductivity cell a pair of platinum electrodes
tion as follows:
is embedded. The cell is immersed in a solution, and the
Standardization—Measure exactly 20 mL of the prepared
resistance or the resistivity of the liquid column between the
solution, add 3 mL of hydrochloric acid, and warm the mix-
electrodes is measured. Alternating current is supplied to
ture. Add 40 mL of diluted sulfuric acid (1 in 130), previous-
this apparatus to avoid the eŠects of electrode polarization.
ly warmed, heat the mixture on a water bath for 30 minutes,
Further, a temperature compensation system is generally
and allow it to stand overnight. Filter the mixture, wash the
1374 General Tests, Processes and Apparatus Supplement I, JP XIV

contained in the apparatus. er of 0.1 cm-1, 1 cm-1, or 10 cm-1, are generally used.
Conductivity measurement is generally performed by us- For determination or conˆrmation of the cell constant, an
ing an immersion-type cell. A pair of platinum electrodes, appropriate KCl standard solution should be chosen and
the surfaces of which are coated with platinum black, is ˆxed prepared, taking account of the expected conductivity of the
in parallel. Both electrodes are generally protected by a glass sample solution to be measured. Rinse the cell several times
tube to prevent physical shocks. with distilled water. Next, after rinsing the cell 2 – 3 times
When the surface area of the electrode is A (cm2), and the with the standard solution used for the cell constant determi-
separation distance of the two electrodes is l (cm), the cell nation, immerse the cell in the standard solution contained
constant C (cm-1) is given by the following equation. in a measuring vessel. After conˆrming that the temperature
of the standard solution is maintained at 20 ± 0.19 C or at
C = a・(l/A )
the temperature speciˆed in the monograph, measure the
a is a dimensionless numerical coe‹cient, and it is character- resistance RKCl or the conductance GKCl of the standard solu-
istic of the cell design. tion, and calculate the cell constant C (cm-1) by use of the
In addition to the immersion-type cell, there are ‰ow- following equation.
through-type and insert-in-pipe-type cells. These cells are set
C = RKCl・kKCl or C = kKCl/GKCl
or inserted in an appropriate position in the ‰ow system for
monitoring the quality of water continuously or intermit- RKCl: Measured resistance (MQ)
tently, during the preparation of highly puriˆed water. GKCl: Measured conductance ( mS)
kKCl: Conductivity of the standard solution being used
Standard Solution of Potassium Chloride
( mS・cm-1)
After pulverizing an appropriate amount of potassium
chloride for conductivity measurement, dry it at 500 – 6009C The measured cell constant should be consistent with the
for 4 hours. Take an indicated amount of the dried potassi- given value within 5z. If it is not consistent, coat the elec-
um chloride, as shown in Table 1, dissolve it in distilled or trodes with platinum black again, or replace the cell with a
puriˆed water (conductivity less than 2 m S・cm-1), previous- new one.
ly boiled and cooled, and adjust to make 1000.0 g, for (2) Suitability Test for the Apparatus
preparation of the standard solutions. The conductivity and Using an appropriate KCl standard solution according to
the resistivity of the respective standard solutions at 209C the expected conductivity of the sample solution, perform
are indicated in Table 1. These standard solutions should be the suitability test for the apparatus. Rinse the conductivity
kept in tightly closed polyethylene or hard glass bottles. cell several times with distilled water, and rinse again 2 – 3
times with the selected standard solution. Fill the standard
Table 1. Conductivity and Resistivity of the Standard
solution in the measuring vessel. After conˆrming that the
Solutions of Potassium Chloride at 209C
temperature of the measuring system is maintained at 20 ±
Concentration Conductivity k Resistivity r 0.19C, measure the conductivity of the standard solution.
(g/1000.0 g) (m S・cm-1) (Q・cm) When this measuring procedure is repeated several times, the
0.7455 1330 752 average conductivity should be consistent with an indicated
0.0746 133.0 7519 value in Table 1 within 5z. Further, the relative standard
0.0149 26.6 37594 deviation should be less than 2z.
(3) Measurement
When measurement at 209C can not be done, the indicat- After conˆrmation of the suitability of the apparatus, per-
ed value of conductivity for the respective standard solution form the conductivity measurement for the sample solution.
(Table 1), can be corrected by using the equation below. Unless otherwise speciˆed, the preparation method for sam-
However, the equation is valid only within the range of 20 ± ple solution should be as speciˆed in the respective mono-
59C. graph. Rinse the conductivity cell several times with distilled
water, and rinse again 2 – 3 times with sample solution. Im-
kT = k20[1 + 0.021(T - 20)]
merse the cell in the sample solution placed in a measuring
T: Measuring temperature speciˆed in the monograph vessel. If necessary, agitate gently the sample solution. After
kT: Calculated conductivity of the KCl standard solution conˆrming that the temperature of the sample solution is
at T9C maintained at 20 ± 0.19 C or at the temperature speciˆed in
k20: Conductivity of the KCl standard solution at 209C the monograph, measure the resistance RT (MQ) or conduc-
tance GT ( mS) of the sample solution, and calculate the con-
Operating Procedure
ductivity kT by using the following equation.
(1) Cell Constant
An appropriate conductivity cell should be chosen accord- kT = CGT or kT = C / R T
ing to the expected conductivity of the sample solution. The
Items such as the sample preparation method, the necessi-
higher the expected conductivity, the larger the cell constant
ty of blank correction, the calculation method, the speciˆca-
required for the conductivity cell, so that the electrical
tion value, and the measuring temperature should be de-
resistance is within the measuring range of the apparatus
scribed in the monograph, if necessary.
being used. Conductivity cells with a cell constant of the ord-
Supplement I, JP XIV General Tests, Processes and Apparatus 1375

sel by smoothly moving across it the edge of a slide glass or


Add the following: other tool. Remove any material from the sides of the vessel,
and determine the total mass Mt. Calculate the bulk density
73. Determination of Bulk and rB by the formula:
M - M0
Tapped Densities rB = t
V
rB: Bulk density by constant volume method (g/mL)
Determination of bulk and tapped densities is a method to Mt: Total mass of powder and measuring vessel (g)
determine the bulk densities of powdered drugs under loose M0: Mass of measuring vessel (g)
and tapped packing conditions respectively. Loose packing V : Volume of measuring vessel (mL)
is deˆned as the state obtained by pouring a powder sample
into a vessel without any consolidation, and tapped packing Record the average of 3 determinations using 3 diŠerent
is deˆned as the state obtained when the vessel containing powder samples.
the powder sample is to be repeatedly dropped a speciˆed Tapped density
distance at a constant drop rate until the apparent volume of Tapped density is an apparent density obtained by
sample in the vessel becomes almost constant. The bulk den- mechanically tapping a measuring vessel containing a pow-
sity is expressed in mass per unit apparent volume of powder der sample. The determination of tapped density is achieved
(g/mL). Because the bulk density is one of the measures of by measuring the apparent volume of a powder sample hav-
packing properties, compressibility and ‰ow properties, and ing a known mass in a vessel after tapping (Method 1) or by
is dependent on the ``history'' of the powder, it is essential measuring the mass of powder in a vessel having a known
to report the bulk density to specify how the determination volume after tapping (Method 2).
was made.
Method 1 (Constant mass method)
Bulk density Unless otherwise speciˆed, pass a quantity of sample
The bulk density is an apparent density obtained by pour- su‹cient to complete the test through a 1000-mm (No.16) or
ing a powder sample into a vessel without any consolidation. a 710-mm (No.22) screen to break up agglomerates that may
The determination of bulk density is achieved by measuring have formed during storage. Weigh accurately about 100 g
the apparent volume of a powder sample having a known of test sample, and pour it into a 250-mL graduated glass
mass in a graduated cylinder (Method 1) or by measuring the cylinder (readable to 2 mL) without consolidation. If it is not
mass of powder in a vessel having a known volume (Method possible to use 100 g, proceed according to the same proce-
2). dure as that described above by using a 100-mL graduated g-
Method 1 (Constant mass method) lass cylinder (readable to 1 mL). It is essential to select ap-
Unless otherwise speciˆed, pass a quantity of sample propriate masses of the cylinder support, holder and cylin-
su‹cient to complete the test through a 1000-mm (No.16) der so as to ensure the dynamic stability of the apparatus
screen to break up agglomerates that may have formed dur- during tapping. After attaching the glass cylinder containing
ing storage. Weigh accurately about 30 g of test sample, and the powder sample to the tapping apparatus, carry out tap-
pour it into a dry 100-mL graduated glass cylinder (readable ping under the measuring conditions (tapping rate and drop
to 1 mL). Carefully level the powder without consolidation, height) speciˆed for each apparatus.
if necessary, and read the unsettled apparent volume, V0, to Unless otherwise speciˆed, repeat increments of 50 taps or
the nearest graduated unit. Calculate the bulk density rB by 1 minute until the diŠerence between succeeding measure-
the formula: ments is less than 2z, and determine the ˆnal apparent
M volume, Vf. Calculate the tapped density rT by the formula:
rB = M
V0 rT =
rB: Bulk density by constant mass method (g/mL) Vf
M : Mass of powder sample (g) rT: Tapped density by constant mass method (g/mL)
V0: Apparent volume of powder sample (mL) M : Mass of powder sample (g)
Vf: Final apparent volume of sample after tapping (mL)
Record the average of 3 determinations using 3 diŠerent
powder samples. If a 30-g sample is too large to determine, Record the average of 3 determinations using 3 diŠerent
adjust the mass of sample so as to provide an apparent powder samples.
volume of 60 – 100 mL. Method 2 (Constant volume method)
Method 2 (Constant volume method) Unless otherwise speciˆed, pass a quantity of sample
Unless otherwise speciˆed, pass a quantity of sample su‹cient to complete the test through a 1000-mm (No.16)
su‹cient to complete the test through a 1000-mm (No.16) screen to break up agglomerates that may have formed dur-
screen to break up agglomerates that may have formed dur- ing storage. Attach a supplementary cylinder to the stainless
ing storage. Allow an excess of sample powder to pour into steel measuring vessel having a known mass of M0 and a
the measuring vessel having the volume of V and mass of volume of V (Fig. 1), and then pour an excess of the sample
M0. Carefully scrape excess powder from the top of the ves- into the vessel. After setting up the vessel in an adequate tap-
1376 General Tests, Processes and Apparatus Supplement I, JP XIV

ping apparatus with a ˆxed drop height, carry out tapping at Mt: Total mass of powder and measuring vessel (g)
the rate and cumulative tap number speciˆed for each ap- M0: Mass of measuring vessel (g)
paratus. Then remove the supplementary cylinder from the V : Volume of measuring vessel (mL)
vessel and carefully scrape excess powder from the top of the
Record the average of 3 determinations and the relative
vessel by smoothly moving across it the edge of a slide glass
standard deviation using 3 diŠerent powder samples. If the
or other tool. Remove any material from the sides of the ves-
relative standard deviation is not less than 2z, repeat the
sel, and determine the total mass Mt. Calculate the tapped
test with further tapping.
density rT by the formula:
M - M0 Balances: Use balances readable to the nearest 0.1 g.
rT = t
V
rT: Tapped density by constant volume method (g/mL)

Fig. 1 Measuring vessel (left) and supplementary cylinder (right)


O‹cial Monographs
for Part I
Purity (1) Heavy metals—Put 1.0 g of Aceglutamide
Add the following:
Aluminum in a porcelain crucible, cover the crucible loosely,
and heat gently to carbonize. After cooling, add 2 mL of
Aceglutamide Aluminum nitric acid and 1 mL of sulfuric acid, heat gently until the
white fumes no more evolve, and heat to incinerate at 500 to
アセグルタミドアルミニウム
6009 C. If the incineration is not accomplished, add 2 mL of
nitric acid and 1 mL of sulfuric acid, heat in the same man-
ner as above, then ignite at 500 to 6009C to incinerate. After
cooling, add 2 mL of hydrochloric acid, proceed with this
solution according to Method 2, and perform the test. Pre-
pare the control solution as follows: proceed in the same
manner as the preparation of the test solution with the same
C35H59Al3N10O24: 1084.84 amount of the reagents, and add 2.0 mL of Standard Lead
Pentakis[(2S )-2-acetylamino-4- Solution and water to make 50 mL (not more than 20 ppm).
carbamoylbutanoato]tetrahydroxotrialuminium (2) Arsenic—Prepare the test solution with 1.0 g of
[12607-92-0] Aceglutamide Aluminum according to Method 1, and per-
form the test (not more than 2 ppm).
Aceglutamide Aluminum contains not less than (3) Related substances—Dissolve 0.10 g of Acegluta-
85.4z and not more than 87.6z of aceglutamide mide Aluminum in the mobile phase to make exactly
(C7H12N2O4: 188.18), and not less than 7.0z and not 100 mL, and use this solution as the sample solution. Pipet
more than 8.0z of aluminum (Al: 26.98), calculated 1 mL of the sample solution, add the mobile phase to make
on the dried basis. exactly 100 mL, and use this solution as the standard
solution (1). Separately, dissolve 10 mg of 2-acetamido-
Description Aceglutamide Aluminum occurs as a white
glutarimide in the mobile phase to make exactly 100 mL.
powder, having astringent bitter taste.
Pipet 3 mL of this solution, add the mobile phase to make
It is freely soluble in water, and practically insoluble in
exactly 100 mL, and use this solution as the standard solu-
ethanol (99.5).
tion (2). Perform the test with exactly 20 mL each of the
It dissolves in dilute hydrochloric acid.
sample solution, the standard solution (1) and the standard
It is hygroscopic.
solution (2) as directed under the Liquid Chromatography
Identiˆcation (1) Dissolve 0.03 g each of Aceglutamide according to the following conditions, and determine each
Aluminum and Aceglutamide Reference Standard in 5 mL peak area by the automatic integration method: the peak
of water, and use these solutions as the sample solution and area of 2-acetamidoglutarimide from the sample solution is
the standard solution. Perform the test with these solutions not more than that from the standard solution (2), the peak
as directed under the Thin-layer Chromatography. Spot areas other than aceglutamide and 2-acetamidoglutarimide
5 mL each of the sample solution and the standard solution from the sample solution are not more than 3/10 times the
on a plate of cellulose for thin-layer chromatography. peak area of aceglutamide from the standard solution (1),
Develop the plate with a mixture of 1-propanol, water and and the total of the peak areas other than aceglutamide and
acetic acid (100) (16:8:1) to a distance of about 10 cm, and 2-acetamidoglutarimide from the sample solution is not
air-dry the plate. Spray evenly a solution of bromocresol more than the peak area of aceglutamide from the standard
green in ethanol (95) (1 in 1000), then spray evenly diluted solution (1).
ammonia solution (28) (1 in 100): the spots from the sample Operating conditions—
solution and the standard solution show a light yellow and Detector, column, column temperature, mobile phase,
have the same Rf value. and ‰ow rate: Proceed as directed in the operating condi-
(2) A solution of Aceglutamide Aluminum in dilute tions in the Assay.
hydrochloric acid (1 in 20) responds to the Qualitative Tests Time span of measurement: About 3 times as long as the
for aluminum salt. retention time of aceglutamide.
System suitability—
Optical rotation [a]20D : -5.5 – -7.59(2 g calculated on the Test for required detection: To exactly 5 mL of the
dried basis, water, 50 mL, 100 mm).
standard solution (1) add the mobile phase to make exactly

1377
1378 O‹cial Monographs for Part I Supplement I, JP XIV

50 mL. Conˆrm that the peak area of aceglutamide obtained acid, and heat on a water bath for 60 minutes. After cooling,
from 20 mL of this solution is equivalent to 7 to 13z of that add water to make exactly 200 mL. Pipet 20 mL of this solu-
of aceglutamide obtained from 20 mL of the standard solu- tion, add exactly 25 mL of 0.05 mol/L disodium dihydrogen
tion. ethylenediamine tetraacetate VS and 20 mL of acetic acid-
System performance: Proceed as directed in the system ammonium acetate buŠer solution, pH 4.8, and boil for
suitability in the Assay. 5 minutes. After cooling, add 50 mL of ethanol (95), and
System repeatability: When the test is repeated 6 times titrate with 0.05 mol/L zinc acetate VS until the color of the
with 20 mL of the standard solution (1) under the above solution changes from light dark green to light red (indica-
operating conditions, the relative standard deviation of the tor: 2 mL of dithizone TS). Perform a blank determination
peak area of aceglutamide is not more than 2.0z. in the same manner.

Loss on drying Not more than 5.0z (1 g, 1309


C, 5 hours). Each mL of 0.05 mol/L disodium dihydrogen
ethylenediamine tetraacetate VS = 1.349 mg of Al
Assay (1) Aceglutamide—Weigh accurately about 50 mg
of Aceglutamide Aluminum, dissolve in a suitable amount Containers and storage Containers—Tight containers.
of the mobile phase, add exactly 10 mL of the internal stan-
dard solution and the mobile phase to make 50 mL, and use
this solution as the sample solution. Separately, weigh ac- Acetylkitasamycin
curately about 45 mg of Aceglutamide Reference Standard,
dissolve in a suitable amount of the mobile phase, add ex- アセチルキタサマイシン
actly 10 mL of the internal standard solution and the mobile
phase to make 50 mL, and use this solution as the standard Change the Assay (3) to read:
solution. Perform the test with 10 mL each of the sample
Assay
solution and the standard solution as directed under the Liq-
(3) Standard solution-Weigh accurately an amount of
uid Chromatography according to the following conditions,
Leucomycin A5 Reference Standard equivalent to about
and determine the ratios, QT and QS, of the peak area of
30 mg (potency), dissolve in 10 mL of methanol, add water
aceglutamide to that of the internal standard.
to make exactly 100 mL, and use this solution as the stan-
Q
Amount (mg) of aceglutamide (C7H12N2O4) = WS × T dard stock solution. Keep the standard stock solution at 59C
QS
or below and use within 3 days. Take exactly a suitable
WS: Amount (mg) of Aceglutamide Reference Standard amount of the standard stock solution before use, add
0.1 mol/L phosphate buŠer solution, pH 8.0 to make solu-
Internal standard solution—A solution of thymine in
tions so that each mL contains 30 mg (potency) and 7.5 mg
methanol (1 in 4000).
(potency), and use these solutions as the high concentration
Operating conditions—
standard solution and the low concentration standard solu-
Detector: An ultraviolet absorption photometer (wave-
tion, respectively.
length: 210 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Acetylspiramycin
ameter). アセチルスピラマイシン
Column temperature: A constant temperature of about
259C.
Change to read except the structural formula
Mobile phase: A mixture of diluted perchloric acid (1 in and chemical name:
1000) and methanol (99:1).
Flow rate: Adjust the ‰ow rate so that the retention time Acetylspiramycin contains not less than 900 mg
of aceglutamide is about 5 minutes. (potency) per mg, calculated on the dried basis. The
System suitability— potency of Acetylspiramycin is expressed as mass
System performance: When the procedure is run with (potency) of acetylspiramycin II (C47H78N2O16:
10 mL of the standard solution under the above operating
927.13). 1 mg (potency) of Acetylspiramycin is
conditions, aceglutamide and the internal standard are elut- equivalent to 0.7225 mg of acetylspiramycin II
ed in this order with the resolution between these peaks (C47H78N2O16).
being not less than 11.
System repeatability: When the test is repeated 6 times Description Acetylspiramycin occurs as a white to light
with 10 mL of the standard solution under the above operat- yellowish white powder.
ing conditions, the relative standard deviation of the ratios It is very soluble in acetonitrile and in methanol, freely
of the peak area of aceglutamide to that of the internal stan- soluble in ethanol (99.5), and practically insoluble in water.
dard is not more than 1.0z. Identiˆcation (1) Determine the absorption spectrum of
(2) Aluminum—Weigh accurately about 3.0 g of a solution of Acetylspiramycin in methanol (1 in 50,000) as
Aceglutamide Aluminum, add 20 mL of dilute hydrochloric
Supplement I, JP XIV O‹cial Monographs for Part I 1379

directed under the Ultraviolet-visible Spectrophotometry, (2) Arsenic—Prepare the test solution with 2.0 g of
and compare the spectrum with the Reference Spectrum: Acetylspiramycin according to Method 3, and perform the
both spectra exhibit similar intensities of absorption at the test (not more than 1 ppm).
same wavelengths.
Loss on drying Not more than 3.0z (1 g, in vacuum,
(2) Determine the infrared absorption spectrum of
phosphorus (V) oxide, 609
C, 3 hours).
Acetylspiramycin as directed in the potassium bromide disk
method under the Infrared Spectrophotometry, and com- Residue on ignition Not more than 0.50z (1.0 g).
pare the spectrum with the Reference Spectrum: both spec-
Assay Perform the test according to the Cylinder-plate
tra exhibit similar intensities of absorption at the same wave
method as directed under the Microbial Assay for Antibiot-
numbers.
ics according to the following conditions.
Content ratio of the active principle Dissolve 25 mg of (1) Test organism—Bacillus subtilis ATCC 6633
Acetylspiramycin in 25 mL of the mobile phase, and use this (2) Culture medium—Use the medium i in 1) Medium
solution as the sample solution. Perform the test with 5 mL for test organism [5] under (1) Agar media for seed and base
of the sample solution as directed under the Liquid Chro- layer.
matography according to the following conditions, and (3) Standard solutions—Weigh accurately an amount
determine the areas, AII, AIII, AIV, AV, AVI and AVII, of the of Acetylspiramycin II Reference Standard, equivalent to
peaks of acetylspiramycin II, acetylspiramycin III, acetyl- about 50 mg (potency), dissolve in 20 mL of methanol, add
spiramycin IV, acetylspiramycin V, acetylspiramycin VI and 0.1 mol/L phosphate buŠer solution for antibiotics, pH 8.0
acetylspiramycin VII, respectively, by the automatic integra- to make exactly 50 mL, and use this solution as the standard
tion method, and calculate the ratios of the amounts of AII, stock solution. Keep the standard stock solution at not ex-
AIV and the total of AIII and AV to the total amount of all ceeding 59 C, and use within 3 days. Take exactly a suitable
these peaks: the amount of AII is 30 – 45z, AIV is 30 – 45z, amount of the standard stock solution before use, add
and the total of AIII and AV is not more than 25z. The rela- 0.1 mol/L phosphate buŠer solution for antibiotics, pH 8.0
tive retention times of acetylspiramycin III, acetylspiramy- to make solutions so that each mL contains 80 mg (potency)
cin IV, acetylspiramycin V, acetylspiramycin VI and acetyl- and 20 mg (potency), and use these solutions as the high con-
spiramycin VII with respect to acetylspiramycin II are 1.3, centration standard solution and the low concentration stan-
1.7, 2.3, 0.85 and 1.4, respectively. dard solution, respectively.
Operating conditions— (4) Sample solutions—Weigh accurately an amount of
Detector: An ultraviolet absorption photometer (wave- Acetylspiramycin, equivalent to about 50 mg (potency), dis-
length: 231 nm). solve in 20 mL of methanol, and add 0.1 mol/L phosphate
Column: A stainless steel column 6 mm in inside diameter buŠer solution for antibiotics, pH 8.0 to make exactly
and 15 cm in length, packed with octadecylsilanized silica gel 50 mL. Take exactly a suitable amount of this solution, add
for liquid chromatography (3 mm in particle diameter). 0.1 mol/L phosphate buŠer solution for antibiotics, pH 8.0
Column temperature: A constant temperature of about to make solutions so that each mL contains 80 mg (potency)
359C. and 20 mg (potency), and use these solutions as the high con-
Mobile phase: A mixture of acetonitrile, 0.02 mol/L centration sample solution and the low concentration sample
potassium dihydrogen phosphate TS and a solution of solution, respectively.
dipotassium hydrogen phosphate (87 in 25,000) (26:7:7).
Containers and storage Containers—Tight containers.
Flow rate: Adjust the ‰ow rate so that the retention time
of acetylspiramycin II is about 10 minutes.
System suitability—
System performance: Dissolve 25 mg of Acetylspiramycin Aclarubicin Hydrochloride
II Reference Standard in the mobile phase to make 100 mL.
塩酸アクラルビシン
When the procedure is run with 5 mL of this solution under
the above operating conditions, the number of theoretical
Change to read except the structural formula
plates and the symmetry coe‹cient of the peak of acetyl-
and chemical name:
spiramycin II are not less than 14,500 and not more than 2.0,
respectively.
Aclarubicin Hydrochloride contains not less than
System repeatability: When the test is repeated 6 times
860 mg (potency) per mg, calculated on the anhydrous
with 5 mL of the sample solution under the above operating
basis. The potency of Aclarubicin Hydrochloride
conditions, the relative standard deviation of the peak area
is expressed as mass (potency) of aclarubicin
of acetylspiramycin II is not more than 2.0z.
(C42H53NO15: 811.87).
Purity (1) Heavy metals—Proceed with 1.0 g of Acetyl-
Description Aclarubicin Hydrochloride occurs as a yellow
spiramycin according to Method 2, and perform the test.
to pale orange-yellow powder.
Prepare the control solution with 1.0 mL of Standard Lead
It is very soluble in chloroform and in methanol, freely
Solution (not more than 10 ppm).
soluble in water, and slightly soluble in ethanol (95).
1380 O‹cial Monographs for Part I Supplement I, JP XIV

Identiˆcation (1) Determine the absorption spectrum of Flow rate: Adjust the ‰ow rate so that the retention time
a solution of Aclarubicin Hydrochloride in diluted methanol of aclarubicin is about 5 minutes.
(4 in 5) (3 in 100,000) as directed under the Ultraviolet- Time span of measurement: As long as about 4 times of
visible Spectrophotometry, and compare the spectrum with the retention time of aclarubicin after the solvent peak.
the Reference Spectrum: both spectra exhibit similar intensi- System suitability—
ties of absorption at the same wavelengths. Test for required detectability: Pipet 1 mL of the sample
(2) Determine the infrared absorption spectrum of solution, add the mobile phase to make exactly 100 mL, and
Aclarubicin Hydrochloride as directed in the potassium use this solution as the solution for system suitability test.
bromide disk method under the Infrared Spectrophotomet- Pipet 1 mL of the solution for system suitability test, and
ry, and compare the spectrum with the Reference Spectrum: add the mobile phase to make exactly 10 mL. Conˆrm that
both spectra exhibit similar intensities of absorption at the the peak area of aclarubicin obtained from 20 mL of this so-
same wave numbers. lution is equivalent to 7 to 13z of that from 20 mL of the so-
(3) A solution of Aclarubicin Hydrochloride in lution for system suitability test.
methanol (1 in 200) responds to the Qualitative Test (2) for System performance: Dissolve 5 mg of Aclarubicin
chloride. Hydrochloride in 10 mL of 0.1 mol/L hydrochloric acid TS,
and allow to stand for 60 minutes. To 1.0 mL of this solu-
Optical rotation [a]20
D : -146 – -1629(0.05 g calculated on
tion add 1.0 mL of 0.2 mol/L sodium hydroxide TS, 1.0 mL
the anhydrous basis, water, 10 mL, 100 mm).
of phosphate buŠer solution, pH 8.0 and 1.0 mL of chlo-
pH The pH of a solution obtained by dissolving 0.05 g of roform, shake vigorously, and take the chloroform layer.
Aclarubicin Hydrochloride in 10 mL of water is between 5.5 When the procedure is run with 20 mL of the chloroform
and 6.5. under the above operating conditions, aclarubicin and 1-
deoxypyrromycin are eluted in this order with the resolution
Purity (1) Clarity and color of solution-Dissolve 0.10 g
between these peaks being not less than 3.0.
of Aclarubicin Hydrochloride in 10 mL of water: the solu-
System repeatability: When the test is repeated 5 times
tion is clear and yellow to pale orange-yellow.
with 20 mL of the sample solution under the above operating
(2) Heavy metals—Proceed with 1.0 g of Aclarubicin
conditions, the relative standard deviation of the peak area
Hydrochloride according to Method 2, and perform the test.
of aclarubicin is not more than 2.0z.
Prepare the control solution with 2.0 mL of Standard Lead
Solution (not more than 20 ppm). Water Not more than 3.5z (0.1 g, volumetric titration,
(3) Related substances—Dissolve 10 mg of Aclarubicin direct titration).
Hydrochloride in 10 mL of the mobile phase, and use this
Residue on ignition Not more than 0.1z (1 g).
solution as the sample solution. Perform the test with 20 mL
of the sample solution as directed under the Liquid Chro- Assay Weigh accurately an amount of Aclarubicin
matography according to the following conditions, and Hydrochloride, equivalent to about 20 mg (potency), and
determine each peak area by the automatic integration dissolve in diluted methanol (4 in 5) to make exactly 100 mL.
method. Calculate the amount of the related substances by Pipet 15 mL of this solution, add diluted methanol (4 in 5) to
the area percentage method: the amount of aklavinone hav- make exactly 100 mL, and use this solution as the sample
ing the relative retention time of about 0.6 to aclarubicin is solution. Separately, weigh accurately an amount of
not more than 0.2z, aclacinomycin L1 having the relative Aclarubicin Reference Standard, equivalent to about 20 mg
retention time of about 0.75 to aclarubicin is not more than (potency), add 0.6 mL of diluted hydrochloric acid (1 in 250)
0.5z, 1-deoxypyrromycin having the relative retention time and diluted methanol (4 in 5) to make exactly 100 mL. Pipet
of about 1.7 to aclarubicin is not more than 1.5z and 15 mL of this solution, add diluted methanol (4 in 5) to make
aclacinomycin S1 having the relative retention time of about exactly 100 mL, and use this solution as the standard solu-
2.3 to aclarubicin is not more than 0.5z, and the total tion. Perform the test with the sample solution and the stan-
amount of the peaks other than aclarubicin and the peaks dard solution as directed under the Ultraviolet-visible Spec-
mentioned above is not more than 1.0z of the peak area of trophotometry, and determine the absorbencies, AT and AS,
aclarubicin. at 433 nm.
Operating conditions—
Amount [mg (potency)] of aclarubicin (C42H53NO15)
Detector: A visible absorption photometer (wavelength:
A
436 nm). = WS × T × 1000
AS
Column: A stainless steel column 3.9 mm in inside di-
WS: Amount [mg (potency)] of Aclarubicin Reference
ameter and 30 cm in length, packed with silica gel for liquid
Standard
chromatography (10 mm in particle diameter).
Column temperature: A constant temperature of about Containers and storage Containers—Tight containers.
259C. Storage—Light-resistant and at 59
C or below.
Mobile phase: A mixture of chloroform, methanol, acetic
acid (100), water and triethylamine (6800:2000:1000:200:1).
Supplement I, JP XIV O‹cial Monographs for Part I 1381

Change to read: the test with exactly 25 mL each of the sample solution and
the standard solution as directed under the Liquid Chro-
Actinomycin D matography according to the following conditions, and
determine the peak area of actinomycin D, AT and AS, of
アクチノマイシンD both solutions.

Amount [mg (potency)] of C62H86N12O16


A
= WS × T × 1000
AS
WS: Amount [mg (potency)] of Actinomycin D Reference
Standard

Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
C62H86N12O16: 1255.42 Column: A stainless steel column 3.9 mm in inside di-
[50-76-0] ameter and 30 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (10 mm in particle
Actinomycin D, when dried, contains not less than diameter).
950 mg (potency) per mg. The potency of Actinomycin Column temperature: A constant temperature of about
D is expressed as mass (potency) of actinomycin D 259C.
(C62H86N12O16). Mobile phase: A mixture of 0.02 mol/L acetic acid-
sodium acetate TS and acetonitrile (25:23).
Description Actinomycin D occurs as an orange-red to red
Flow rate: Adjust the ‰ow rate so that the retention time
crystalline powder.
of actinomycin D is about 23 minutes.
It is freely soluble in acetone, sparingly soluble in acetoni-
System suitability—
trile and in methanol, slightly soluble in ethanol (99.5), and
System performance: When the procedure is run with
very slightly soluble in water.
25 mL of the standard solution under the above operating
Identiˆcation (1) Determine the absorption spectrum of conditions, the number of theoretical plates and the sym-
a solution of Actinomycin D in methanol (3 in 100,000) as metrical coe‹cient of the peak of actinomycin D are not less
directed under the Ultraviolet-visible Spectrophotometry, than 2000 steps and not more than 1.5, respectively.
and compare the spectrum with the Reference Spectrum or System repeatability: When the test is repeated 5 times
the spectrum of a solution of Actinomycin D Reference with 25 mL of the standard solution under the above operat-
Standard prepared in the same manner as the sample solu- ing conditions, the relative standard deviation of the peak
tion: both spectra exhibit similar intensities of absorption at area of actinomycin D is not more than 2.0z.
the same wavelengths.
Containers and storage Containers—Tight containers.
(2) Dissolve 0.1 g each of Actinomycin D and Actinomy-
Storage—Light-resistant.
cin D Reference Standard in 10 mL of acetone, and use these
solutions as the sample solution and the standard solution.
Perform the test with these solutions as directed under the
Thin-layer Chromatography. Spot 10 mL each of the sample Amoxicillin
solution and the standard solution on a plate of silica gel
アモキシシリン
with ‰uorescent indicator for thin-layer chromatography.
Develop the plate with a mixture of 1-butanol, water and
Change the origin/limits of content to read:
methanol (4:2:1) to a distance of about 10 cm, and air-dry
the plate. Examine under ultraviolet light (main wavelength:
Amoxicillin contains not less than 950 mg (potency)
254 nm): the Rf value of the principal spot from the sample
per mg, calculated on the anhydrous basis. The poten-
solution is the same as that from the standard solution.
cy of Amoxicillin is expressed as mass (potency) of
Optical rotation [a]20
D : -292 – -3179(after drying, 10 mg,
amoxicillin (C16H19N3O5S: 365.40).
methanol, 10 mL, 100 mm).
Add the following next to Identiˆcation:
Loss on drying Not more than 5.0z (1 g, in vacuum,
609C, 3 hours). Optical rotation [a]20
D : +290 – +3159(0.1 g calculated on
the anhydrous basis, water, 100 mL, 100 mm).
Assay Weigh accurately an amount of Actinomycin D
and Actinomycin D Reference Standard, previously dried,
Add the following next to Purity (2):
equivalent to about 60 mg (potency), dissolve each in the
mobile phase to make exactly 50 mL, and use these solutions Purity
as the sample solution and the standard solution. Perform (3) Related substances—Dissolve 0.10 g of Amoxicillin
1382 O‹cial Monographs for Part I Supplement I, JP XIV

in 50 mL of a solution of sodium tetraborate decahydrate WS: Amount [mg (potency)] of Amoxicillin Reference
(1 in 200), and use this solution as the sample solution. Pipet Standard
1 mL of the sample solution, add a solution of sodium
Operating conditions—
tetraborate decahydrate (1 in 200) to make exactly 100 mL,
Detector: An ultraviolet absorption photometer (wave-
and use this solution as the standard solution. Perform the
length: 230 nm).
test with exactly 10 mL each of the sample solution and the
Column: A stainless steel column 4.6 mm in inside di-
standard solution as directed under the Liquid Chro-
ameter and 15 cm in length, packed with octadecylsilanized
matography according to the following conditions, and
silica gel for liquid chromatography (5 mm in particle
determine each peak area by the automatic integration
diameter).
method: the area of the peak other than amoxicillin obtained
Column temperature: A constant temperature of about
from the sample solution is not more than the peak area of
259C.
amoxicillin from the standard solution.
Mobile phase: Dissolve 1.361 g of sodium acetate trihy-
Operating conditions—
drate in 750 mL of water, adjust the pH to 4.5 with acetic
Detector: An ultraviolet absorption photometer (wave-
acid (31), and add water to make 1000 mL. To 950 mL of
length: 254 nm).
this solution add 50 mL of methanol.
Column: A stainless steel column 4 mm in inside diameter
Flow rate: Adjust the ‰ow rate so that the retention time
and 30 cm in length, packed with octadecylsilanized silica gel
of amoxicillin is about 8 minutes.
for liquid chromatography (10 mm in particle diameter).
System suitability—
Column temperature: A constant temperature of about
System performance: When the procedure is run with
259C.
10 mL of the standard solution under the above operating
Mobile phase: Dissolve 1.361 g of sodium acetate trihy-
conditions, the number of theoretical plates of the peak of
drate in 750 mL of water, adjust the pH to 4.5 with acetic
amoxicillin is not less than 2500.
acid (31), and add water to make 1000 mL. To 950 mL of
System repeatability: When the test is repeated 6 times
this solution add 50 mL of methanol.
with 10 mL of the standard solution under the above operat-
Flow rate: Adjust the ‰ow rate so that the retention time
ing conditions, the relative standard deviation of the peak
of amoxicillin is about 8 minutes.
area of amoxicillin is not more than 1.0z.
Time span of measurement: About 4 times as long as the
retention time of amoxicillin.
System suitability—
Test for required detection: To exactly 1 mL of the stan- Ampicillin
dard solution add a solution of sodium tetraborate decahy-
アンピシリン
drate (1 in 200) to make exactly 10 mL. Conˆrm that the
peak area of amoxicillin obtained from 10 mL of this solu-
Change to read except the structural formula
tion is equivalent to 7 to 13z of that of amoxicillin obtained
and chemical name:
from 10 mL of the standard solution.
System performance: When the procedure is run with
Ampicillin contains not less than 960 mg (potency)
10 mL of the standard solution under the above operating
per mg, calculated on the anhydrous basis. The poten-
conditions, the number of theoretical plates of the peak of
cy of Ampicillin is expressed as mass (potency) of
amoxicillin is not less than 2500.
ampicillin (C16H19N3O4S: 349.40).
System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat- Description Ampicillin occurs as a white to light yellowish
ing conditions, the relative standard deviation of the peak white, crystals or crystalline powder.
area of amoxicillin is not more than 1.0z. It is sparingly soluble in water, slightly soluble in
methanol, very slightly soluble in ethanol (95), and practical-
Change the Assay to read: ly insoluble in acetonitrile.

Assay Weigh accurately an amount of Amoxicillin and Identiˆcation Determine the infrared absorption spectrum
Amoxicillin Reference Standard, equivalent to about 30 mg of Ampicillin as directed in the potassium bromide disk
(potency), dissolve each in a solution of boric acid (1 in 200) method under the Infrared Spectrophotometry, and com-
to make exactly 100 mL, and use these solutions as the sam- pare the spectrum with the Reference Spectrum or the spec-
ple solution and the standard solution. Perform the test with trum of Ampicillin Reference Standard: both spectra exhibit
exactly 10 mL each of the sample solution and the standard similar intensities of absorption at the same wave numbers.
solution as directed under the Liquid Chromatography ac-
Optical rotation [a]20
D : +280 – +3059(0.5 g calculated on
cording to the following conditions, and calculate the peak
the anhydrous basis, water, 100 mL, 100 mm).
areas, AT and AS, of amoxicillin of each solution.
pH The pH of a solution obtained by dissolving 1.0 g of
Amount [mg (potency)] of C16H19N3O5S
Ampicillin in 400 mL of water is between 3.5 and 5.5.
A
= WS × T × 1000
AS Purity (1) Heavy metals—Proceed with 1.0 g of Ampicil-
Supplement I, JP XIV O‹cial Monographs for Part I 1383

lin according to Method 2, and perform the test. Prepare the Internal standard solution—A solution of naphthalene in
control solution with 2.0 mL of Standard Lead Solution (not cyclohexane (1 in 20,000).
more than 20 ppm). Operating conditions—
(2) Arsenic—Prepare the test solution with 1.0 g of Detector: A hydrogen ‰ame-ionization detector.
Ampicillin according to Method 3, and perform the test (not Column: A glass column 2.6 mm in inside diameter and
more than 2 ppm). 2 m in length, packed with siliceous earth for gas chro-
(3) Related substances—Dissolve 50 mg of Ampicillin in matography (180 – 250 mm in particle diameter) coated with
the mobile phase to make 50 mL, and use this solution as the 50z phenyl-50z methyl polysiloxane for gas chro-
sample solution. Pipet 1 mL of the sample solution, add the matography at the ratio of 3z.
mobile phase to make exactly 100 mL, and use this solution Column temperature: A constant temperature of about
as the standard solution. Perform the test with exactly 10 mL 1209 C.
each of the sample solution and the standard solution as Carrier gas: Helium
directed under the Liquid Chromatography according to the Flow rate: Adjust the ‰ow rate so that the retention time
following conditions, and determine each peak area by the of N, N-dimethylaniline is about 5 minutes.
automatic integration method: each peak area other than System suitability—
that of ampicillin obtained from the sample solution is not Test for required detectability: Measure exactly 1 mL of
more than the peak area of ampicillin from the standard the standard stock solution, and add water to make exactly
solution. 250 mL. Pipet 1 mL of this solution, add 5 mL of sodium
Operating conditions— hydroxide TS and exactly 1 mL of the internal standard
Detector, column, column temperature, mobile phase, solution, shake vigorously for 1 minute, and use the upper
and ‰ow rate: Proceed as directed in the operating condi- layer liquid obtained after allowing it to stand for the test.
tions in the Assay. Conˆrm that when the procedure is run with 1 mL of the
Time span of measurement: About 10 times as long as the upper layer liquid under the above operating conditions, the
retention time of ampicillin. ratio of the peak area of N, N-dimethylaniline to that of the
System suitability— internal standard is equivalent to 15 – 25z of the ratio of
System performance, and system repeatability: Proceed as the peak area of N, N-dimethylaniline to that of the internal
directed in the system suitability in the Assay. standard obtained from the standard solution.
Test for required detectability: Measure exactly 1 mL of System performance: Dissolve 50 mg of N, N-dimethylani-
the standard solution, and add the mobile phase to make line in cyclohexane to make 50 mL. To 1 mL of this solution
exactly 10 mL. Conˆrm that the peak area of ampicillin add the internal standard solution to make 50 mL, and use
obtained from 10 mL of this solution is equivalent to 7 to this solution as the solution for system performance test.
13z of that from 10 mL of the standard solution. When the procedure is run with 1 mL of the solution for sys-
(4) N, N-Dimethylaniline—Weigh accurately about 1 g tem performance test under the above operating conditions,
of Ampicillin, dissolve in 5 mL of sodium hydroxide TS, N, N-dimethylaniline and the internal standard are eluted in
add exactly 1 mL of the internal standard solution, shake this order with the resolution between these peaks being not
vigorously for 1 minute, and use the upper layer liquid less than 3.
obtained after allowing it to stand as the sample solution. System repeatability: When the test is repeated 6 times
Separately, weigh accurately about 50 mg of N, N- with 1 mL of the solution for system performance test under
dimethylaniline, dissolve in 2 mL of hydrochloric acid and the above operating conditions, the relative standard devia-
20 mL of water, add water to make exactly 50 mL, and use tion of the ratios of the peak area of N, N-dimethylaniline to
this solution as the standard stock solution. Pipet 5 mL of that of the internal standard is not more than 2.0z.
the standard stock solution, and add water to make exactly
Water 12.0 – 15.0z (0.1 g, volumetric titration, direct
250 mL. Pipet 1 mL of this solution, add 5 mL of sodium
titration).
hydroxide TS and exactly 1 mL of the internal standard
solution, shake vigorously for 1 minute, and use the upper Assay Weigh accurately an amount of Ampicillin and
layer liquid obtained after allowing it to stand as the stan- Ampicillin Reference Standard, equivalent to about 50 mg
dard solution. Perform the test with 1 mL each of the sample (potency), dissolve in a suitable volume of the mobile phase,
solution and the standard solution as directed under the Gas add exactly 5 mL each of the internal standard solution and
Chromatography according to the following conditions, the mobile phase to make 50 mL, and use these solutions as
determine the ratios, QT and QS, of the peak area of N ,N- the sample solution and the standard solution. Perform the
dimethylaniline to that of the internal standard, and calcu- test with 10 mL each of the sample solution and the standard
late the amount of N, N-dimethylaniline by the following solution as directed under the Liquid Chromatography
equation: not more than 20 ppm. according to the following conditions, and determine the
W Q ratios, QT and QS, of the peak area of ampicillin to that of
Amount (ppm) of N ,N-dimethylaniline = S × T × 400
WT QS the internal standard.
Q
WS: Amount (g) of N ,N-dimethylaniline Amount [mg (potency)] of C16H19N3O4S = WS × T × 1000
QS
WT: Amount (g) of the sample
WS: Amount [mg (potency)] of Ampicillin Reference
1384 O‹cial Monographs for Part I Supplement I, JP XIV

Standard the anhydrous basis, water, 100 mL, 100 mm).

Internal standard solution—A solution of guaifenesin in the pH The pH of a solution obtained by dissolving 1.0 g of
mobile phase (1 in 200). Anhydrous Ampicillin in 100 mL of water is between 4.0 and
Operating conditions— 5.5.
Detector: An ultraviolet absorption photometer (wave-
Purity (1) Heavy metals—Proceed with 1.0 g of Anhy-
length: 230 nm).
drous Ampicillin according to Method 2, and perform the
Column: A stainless steel column 4.6 mm in inside di-
test. Prepare the control solution with 2.0 mL of Standard
ameter and 15 cm in length, packed with octadecylsilanized
Lead Solution (not more than 20 ppm).
silica gel for liquid chromatography (5 mm in particle
(2) Arsenic—Prepare the test solution with 1.0 g of An-
diameter).
hydrous Ampicillin according to Method 3, and perform the
Column temperature: A constant temperature of about
test (not more than 2 ppm).
259C.
(3) Related substances—Dissolve 0.05 g of Anhydrous
Mobile phase: Dissolve 5.94 g of diammonium hydrogen
Ampicillin in the mobile phase to make 50 mL, and use this
phosphate in 850 mL of water, add 100 mL of acetonitrile,
solution as the sample solution. Pipet 1 mL of the sample
adjust the pH to 5.0 with phosphoric acid, and add water to
solution, add the mobile phase to make exactly 100 mL, and
make exactly 1000 mL.
use this solution as the standard solution. Perform the test
Flow rate: Adjust the ‰ow rate so that the retention time
with exactly 10 mL each of the sample solution and the stan-
of ampicillin is about 6 minutes.
dard solution as directed under the Liquid Chromatography
System suitability—
according to the following conditions, and determine each
System performance: When the procedure is run with
peak area by the automatic integration method: the area of
10 mL of the standard solution under the above operating
each peak other than ampicillin from the sample solution is
conditions, ampicillin and the internal standard are eluted in
not more than the peak area of ampicillin from the standard
this order with the resolution between these peaks being not
solution.
less than 40.
Operating conditions—
System repeatability: When the test is repeated 6 times
Detector, column, column temperature, mobile phase,
with 10 mL of the standard solution under the above operat-
and ‰ow rate: Proceed as directed in the operating condi-
ing conditions, the relative standard deviation of the ratios
tions in the Assay.
of the peak area of ampicillin to that of the internal standard
Time span of measurement: As long as about 10 times of
is not more than 1.0z.
the retention time of ampicillin.
Containers and storage Containers—Tight containers. System suitability—
Test for required detectability: To exactly 1 mL of the
standard solution add the mobile phase to make exactly
Anhydrous Ampicillin 10 mL. Conˆrm that the peak area of ampicillin obtained
from 10 mL of this solution is equivalent to 7 to 13z of that
無水アンピシリン from 10 mL of the standard solution.
System performance, and system repeatability: Proceed as
Change to read except the structural formula directed in the system suitability in the Assay.
and chemical name:
Water Not more than 2.0z (2.5 g, volumetric titration,
direct titration).
Anhydrous Ampicillin contains not less than 960 mg
(potency) per mg, calculated on the anhydrous basis. Assay Weigh accurately an amount of Anhydrous Am-
The potency of Anhydrous Ampicillin is expressed as picillin and Ampicillin Reference Standard, equivalent to
mass (potency) of ampicillin (C16H19N3O4S). about 50 mg (potency), add exactly 5 mL each of the internal
standard solution and the mobile phase to make 50 mL, and
Description Anhydrous Ampicillin occurs as white to light
use these solutions as the sample solution and the standard
yellowish white, crystals or crystalline powder. It is sparingly
solution. Perform the test with 10 mL each of the sample
soluble in water, slightly soluble in methanol, very slightly
solution and the standard solution as directed under the
soluble in ethanol (95), and practically insoluble in acetoni-
Liquid Chromatography according to the following condi-
trile.
tions, and determine the ratios, QT and QS, of the peak area
Identiˆcation Determine the infrared absorption spectrum of ampicillin to that of the internal standard.
of Anhydrous Ampicillin as directed in the potassium
Amount [mg (potency)] of ampicillin (C16H19N3O4S)
bromide disk method under the Infrared Spectrophotomet-
Q
ry, and compare the spectrum with the Reference Spectrum: = WS × T × 1000
QS
both spectra exhibit similar intensities of absorption at the
same wave numbers. WS: Amount [mg (potency)] of Ampicillin Reference
Standard
Optical rotation [a]20
D: +280 – +3059(0.5 g calculated on
Internal standard solution—A solution of guaifenesin in the
Supplement I, JP XIV O‹cial Monographs for Part I 1385

mobile phase (1 in 200). pH The pH of a solution obtained by dissolving 1.0 g of


Operating conditions— Ampicillin Sodium in 10 mL of water is between 8.0 and
Detector: An ultraviolet absorption photometer (wave- 10.0.
length: 230 nm).
Purity (1) Clarity and color of solution—Dissolve 1.0 g
Column: A stainless steel column 4.6 mm in inside di-
of Ampicillin Sodium in 10 mL of water: the solution is clear
ameter and 15 cm in length, packed with octadecylsilanized
and colorless or pale yellow.
silica gel for liquid chromatography (5 mm in particle
(2) Heavy metals—Proceed with 1.0 g of Ampicillin
diameter).
Sodium according to Method 1, and perform the test.
Column temperature: A constant temperature of about
Prepare the control solution with 2.0 mL of Standard Lead
259C.
Solution (not more than 20 ppm).
Mobile phase: Dissolve 5.94 g of diammonium hydrogen
(3) Arsenic—Prepare the test solution with 1.0 g of
phosphate in 850 mL of water, add 100 mL of acetonitrile,
Ampicillin Sodium according to Method 1, and perform the
adjust the pH to 5.0 with phosphoric acid, and add water to
test (not more than 2 ppm).
make exactly 1000 mL.
(4) Related substances—Dissolve 50 mg of Ampicillin
Flow rate: Adjust the ‰ow rate so that the retention time
Sodium in 50 mL of the mobile phase, and use this solution
of ampicillin is about 6 minutes.
as the sample solution. Pipet 1 mL of the sample solution,
System suitability—
add the mobile phase to make exactly 100 mL, and use this
System performance: When the procedure is run with
solution as the standard solution. Perform the test with
10 mL of the standard solution under the above operating
exactly 10 mL each of the sample solution and the standard
conditions, ampicillin and the internal standard are eluted in
solution as directed under the Liquid Chromatography
this order with the resolution between these peaks being not
according to the following conditions, and determine each
less than 40.
peak area by the automatic integration method: the peak
System repeatability: When the test is repeated 6 times
area other than ampicillin obtained from the sample solution
with 10 mL of the standard solution under the above operat-
is not more than the peak area of ampicillin from the stan-
ing conditions, the relative standard deviation of the ratios
dard solution.
of the peak area of ampicillin to that of the internal standard
Operating conditions—
is not more than 1z.
Detector: An ultraviolet absorption photometer (wave-
Containers and storage Containers—Tight containers. length: 230 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized
Ampicillin Sodium silica gel for liquid chromatography (5 mm in particle
diameter).
アンピシリンナトリウム Column temperature: A constant temperature of about
259C.
Change to read except the structural formula Mobile phase: Dissolve 5.94 g of diammonium hydrogen
and chemical name: phosphate in 850 mL of water, add 100 mL of acetonitrile,
adjust the pH to 5.0 with phosphoric acid, and add water to
Ampicillin Sodium contains not less than 850 mg make exactly 1000 mL.
(potency) per mg, calculated on the anhydrous basis. Flow rate: Adjust the ‰ow rate so that the retention time
The potency of Ampicillin Sodium is expressed as of ampicillin is about 6 minutes.
mass (potency) of ampicillin (C16H19N3O4S: 349.40). Time span of measurement: About 10 times as long as the
retention time of ampicillin.
Description Ampicillin Sodium occurs as white to light yel-
System suitability—
lowish white, crystals or crystalline powder.
Test for required detectability: Measure exactly 1 mL of
It is very soluble in water, and sparingly soluble in ethanol
the standard solution, and add the mobile phase to make
(99.5).
exactly 10 mL. Conˆrm that the peak area of ampicillin
Identiˆcation (1) Determine the infrared absorption obtained from 10 mL of this solution is equivalent to 7 to
spectrum of Ampicillin Sodium, previously dried in a desic- 13z of that of ampicillin obtained from 10 mL of the stan-
cator (reduced pressure not exceeding 0.67 kPa, 609C) for dard solution.
3 hours, as directed in the potassium bromide disk method System performance: Dissolve 50 mg of Ampicillin
under the Infrared Spectrophotometry, and compare the Reference Standard in a suitable amount of the mobile
spectrum with the Reference Spectrum: both spectra exhibit phase, add 5 mL of a solution of guaifenesin in the mobile
similar intensities of absorption at the same wave numbers. phase (1 in 200) and the mobile phase to make 50 mL, and
(2) Ampicillin Sodium responds to the Qualitative Test use this solution as the solution for system suitability test.
(1) for sodium salt. When the procedure is run with 10 mL of the solution for sys-
tem suitability test under the above operating conditions,
Optical rotation [a]20
D : +246 – +2729(1 g calculated on the
ampicillin and guaifenesin are eluted in this order with the
anhydrous basis, water, 100 mL, 100 mm).
1386 O‹cial Monographs for Part I Supplement I, JP XIV

resolution between these peaks being not less than 35. each of the sample solution and the standard solution on a
System repeatability: When the test is repeated 6 times plate of silica gel for thin-layer chromatography. Develop
with 10 mL of the solution for system suitability test under the plate with a mixture of ammonia solution (28),
the above operating conditions, the relative standard devia- methanol, chloroform and ethanol (95) (7:6:4:1) to a dis-
tion of the ratios of the peak area of ampicillin to that of tance of about 10 cm, and air-dry the plate. Spray evenly
guaifenesin is not more than 1.0z. 0.2z ninhydrin-water saturated 1-butanol TS on the plate,
and heat at 1009C for 10 minutes: the principal spot ob-
Water Not more than 2.0z (0.2 g, volumetric titration,
tained from the sample solution and the spot from the stan-
direct titration).
dard solution are purple-brown in color and their Rf values
Assay Perform the test according to the Cylinder-plate are the same.
method as directed under the Microbial Assay for Antibiot- (2) A solution of Arbekacin Sulfate (1 in 50) responds to
ics according to the following conditions. the Qualitative Test (1) for sulfate.
(1) Test organism—Bacillus subtilis ATCC 6633
Optical rotation [a]20
D : +69 – +799 (0.25 g after drying,
(2) Culture medium—Use the medium i in 1) Medium
water, 25 mL, 100 mm).
for test organism [5] under (1) Agar media for seed and base
layer, having pH 6.5 to 6.6 after sterilization. pH The pH of a solution obtained by dissolving 0.75 g of
(3) Standard solutions—Weigh accurately an amount of Arbekacin Sulfate in 10 mL of water is between 6.0 and 8.0.
Ampicillin Reference Standard, equivalent to about 25 mg
Purity (1) Clarity and color of solution—A solution
(potency), and dissolve in phosphate buŠer solution, pH 6.0
obtained by dissolving 1.0 g of Arbekacin Sulfate in 5 mL of
to make exactly 50 mL. Take exactly a suitable amount of
water is clear and colorless.
this solution, add phosphate buŠer solution, pH 6.0 to make
(2) Heavy metals—Proceed with 2.0 g of Arbekacin Sul-
solutions so that each mL contains 5 mg (potency) and
fate according to Method 1, and perform the test. Prepare
1.25 mg (potency), and use these solutions as the high con-
the control solution with 2.0 mL of Standard Lead Solution
centration standard solution and the low concentration stan-
(not more than 10 ppm).
dard solution, respectively.
(3) Dibekacin—Weigh accurately about 20 mg of
(4) Sample solutions—Weigh accurately an amount of
Arbekacin Sulfate, add exactly 10 mL of the internal stan-
Ampicillin Sodium, equivalent to about 25 mg (potency),
dard solution to dissolve, add water to make 20 mL, and use
and dissolve in phosphate buŠer solution, pH 6.0 to make
this solution as the sample solution. Separately, weigh
exactly 50 mL. Take exactly a suitable amount of this solu-
accurately an amount of Dibekacin Sulfate Reference Stan-
tion, add phosphate buŠer solution, pH 6.0 to make solu-
dard, equivalent to about 10 mg (potency), and dissolve in
tions so that each mL contains 5 mg (potency) and 1.25 mg
water to make exactly 50 mL. Pipet 5 mL of this solution,
(potency), and use these solutions as the high concentration
add exactly 10 mL of the internal standard solution and
sample solution and the low concentration sample solution,
water to make 20 mL, and use this solution as the standard
respectively.
solution. Perform the test with 5 mL each of the sample solu-
Containers and storage Containers—Tight containers. tion and the standard solution as directed under the Liquid
Chromatography according to the following conditions, and
determine the ratios, QT and QS, of the peak area of dibeka-
Arbekacin Sulfate cin to that of the internal standard. Calculate the amount of
dibekacin by the following equation: not more than 2.0z.
硫酸アルベカシン
WS Q 1
Amount (z) of dibekacin = × T× × 1000
WT QS 50
Change to read except the structural formula
and chemical name: WS: Amount [mg (potency)] of Dibekacin Sulfate Refer-
ence Standard
Arbekacin Sulfate contains not less than 670 mg WT: Amount (mg) of the sample
(potency) per mg, calculated on the dried basis. The
Internal standard solution—A solution of bekanamycin sul-
potency of Arbekacin Sulfate is expressed as mass
fate (1 in 2000).
(potency) of arbekacin (C22H44N6O10: 552.62).
Operating conditions—
Description Arbekacin Sulfate occurs as a white powder. Detector: Fluorometry (excitation wavelength: 340 nm,
It is very soluble in water, and practically insoluble in detection wavelength: 460 nm).
ethanol (99.5). Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized
Identiˆcation (1) Dissolve 10 mg each of Arbekacin Sul-
silica gel for liquid chromatography (5 mm in particle
fate and Arbekacin Sulfate Reference Standard in 1 mL of
diameter).
water, and use these solutions as the sample solution and the
Column temperature: A constant temperature of about
standard solution. Perform the test with these solutions as
409C.
directed under the Thin-layer Chromatography. Spot 2 mL
Reaction coil: A column about 0.3 mm in inside diameter
Supplement I, JP XIV O‹cial Monographs for Part I 1387

and about 3 m in length. with 5 mL of the standard solution under the above operat-
Reaction coil temperature: A constant temperature of ing conditions, the relative standard deviation of the peak
about 509 C. area of arbekacin is not more than 5.0z.
Mobile phase: Dissolve 8.70 g of sodium 1-pentane sul-
Loss on drying Not more than 5.0z (0.5 g, reduced pres-
fonate and 8.52 g of anhydrous sodium sulfate in 980 mL of
sure not exceeding 0.67 kPa, 609
C, 3 hours).
water, adjust the pH to 4.0 with acetic acid (100), and add
water to make 1000 mL. To 230 mL of this solution add Assay Perform the test according to the Cylinder-plate
20 mL of methanol. method as directed under the Microbial Assay for Antibiot-
Reagent: Dissolve 12.36 g of boric acid in 960 mL of ics according to the following conditions.
water, add 10 mL of a solution of o-phthalaldehyde in (1) Test organism—Bacillus subtilis ATCC 6633
ethanol (99.5) (1 in 25), adjust the pH to 10.5 with 8 mol/L (2) Culture medium—Use the medium i in 1) Medium
potassium hydroxide TS, and add water to make 1000 mL. for test organisms [5] under (1) Agar media for seed and
To this solution add 1 mL of 2-mercaptoethanol. base layer, having pH 7.8 – 8.0 after sterilization.
Reaction temperature: A constant temperature of about (3) Standard solutions—Weigh accurately an amount of
509C. Arbekacin Sulfate Reference Standard, previously dried,
Flow rate of the mobile phase: 0.5 mL per minute. equivalent to about 20 mg (potency), dissolve in diluted
Flow rate of the reagent: 1 mL per minute. phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly
System suitability— 50 mL, and use this solution as the standard stock solution.
System performance: Dissolve 20 mg each of Arbekacin Keep the standard stock solution at 5 to 159 C and use within
Sulfate, becanamycin sulfate and dibekacin sulfate in 30 days. Take exactly a suitable amount of the standard
200 mL of water. When the procedure is run with 5 mL of stock solution before use, add 0.1 mol/L phosphate buŠer
this solution under the above operating conditions, be- solution, pH 8.0 to make solutions so that each mL contains
canamycin, arbekacin and dibekacin are eluted in this order, 20 mg (potency) and 5 mg (potency), and use these solutions
and the resolution between the peaks, becanamycin and as the high concentration standard solution and the low con-
arbekacin is not less than 5 and arbekacin and dibekacin is centration standard solution, respectively.
not less than 1.5, respectively. (4) Sample solutions—Weigh accurately an amount of
System repeatability: When the test is repeated 6 times Arbekacin Sulfate, equivalent to about 20 mg (potency), and
with 5 mL of the standard solution under the above operat- dissolve in water to make exactly 50 mL. Take exactly a suit-
ing conditions, the relative standard deviation of the ratio of able amount of this solution, add 0.1 mol/L phosphate
the peak area of dibekacin to that of the internal standard is buŠer solution, pH 8.0 to make solutions so that each mL
not more than 2.0z. contains 20 mg (potency) and 5 mg (potency), and use these
(4) Related substances—Dissolve 20 mg of Arbekacin solutions as the high concentration sample solution and the
Sulfate in 20 mL of water, and use this solution as the sam- low concentration sample solution, respectively.
ple solution. Pipet 3 mL of the sample solution, add water to
Containers and storage Containers—Tight containers.
make exactly 250 mL, and use this solution as the standard
solution. Perform the test with 5 mL each of the sample solu-
tion and the standard solution as directed under the Liquid
Chromatography according to the following conditions, and Astromicin Sulfate
determine the area of each peak by the automatic integration
硫酸アストロマイシン
method: the total area of the peaks other than arbekacin and
dibekacin obtained from the sample solution is not more
Change to read except the structural formula
than the peak area of arbekacin from the standard solution.
and chemical name:
Operating conditions—
Detector, column, column temperature, reaction coil,
Astromicin Sulfate contains not less than 606 mg
reaction coil temperature, mobile phase, reagent, reaction
(potency) per mg, calculated on the anhydrous basis.
temperature, ‰ow rate of mobile phase, and ‰ow rate of
The potency of Astromicin Sulfate is expressed as
reagent: Proceed as directed in the operating conditions in
mass (potency) of astromicin (C17H35N5O6: 405.49).
the Purity (3).
Time span of measurement: About 1.5 times as long as the Description Astromicin Sulfate occurs as a white to light
retention time of arbekacin. yellowish white, powder or masses.
System suitability— It is very soluble in water, sparingly soluble in ethylene
System performance: Dissolve 10 mg each of Arbekacin glycol, and practically insoluble in methanol and in ethanol
Sulfate and dibekacin sulfate in 200 mL of water. When the (99.5).
procedure is run with 5 mL of this solution under the above It is hygroscopic.
operating conditions, arbekacin and dibekacin are eluted in
Identiˆcation (1) Dissolve 10 mg each of Astromicin Sul-
this order with the resolution between these peaks being not
fate and Astromicin Sulfate Reference Standard in 10 mL of
less than 1.5.
water. To 5 mL each of these solutions add water to make
System repeatability: When the test is repeated 6 times
1388 O‹cial Monographs for Part I Supplement I, JP XIV

100 mL, and use these solutions as the sample solution and diameter and 150 cm in length.
the standard solution. Perform the test with these solutions Temperature of reaction coil: 509C
as directed under the Liquid Chromatography according to Mobile phase: To 800 mL of a solution of anhydrous sodi-
the following conditions: the retention time of astromicin um sulfate (71 in 2000) add 25 mL of a solution of sodium 1-
obtained from the sample solution is the same with that heptanesulfonate (1 in 1000) and 1 mL of acetic acid (100),
from the standard solution. and add water to make 1000 mL.
Operating conditions— Reaction reagent: Dissolve 11.2 g of potassium hydroxide,
Detector, column, column temperature, reaction coil, 0.458 g of polyoxyethylene (23) lauryl ether, 0.300 g of o-
temperature of reaction coil, mobile phase, reaction reagent, phthalaldehyde and 1 mL of 2-mercaptoethanol in 400 mL
reaction temperature, ‰ow rate of mobile phase, and ‰ow of a solution of boric acid (31 in 1000), and add water to
rate of reaction reagent: Proceed as directed in the operating make 500 mL.
conditions in the Purity (3). Reaction temperature: 509 C
(2) To 2 mL of a solution of Astromicin Sulfate (1 in Flow rate of mobile phase: 0.7 mL per minute
100) add 2 to 3 drops of barium chloride TS: a white Flow rate of reaction reagent: 0.2 mL per minute
precipitate is formed, and it does not dissolve by addition of Time span of measurement: About 2 times as long as the
dilute nitric acid. retention time of astromicin.
System suitability—
Optical rotation [a]20
D : +90 – +1109(0.25 g calculated on
Test for required detectability: To 5 mL of the sample
the anhydrous basis, water, 25 mL, 100 mm).
solution add water to make 100 mL, and use this solution as
pH The pH of a solution obtained by dissolving 1.0 g of the solution for system suitability test. Pipet 2 mL of the
Astromicin Sulfate in 10 mL of water is between 4.5 and 6.5. solution for system suitability test, and add water to make
exactly 100 mL. Conˆrm that the peak area of astromicin
Purity (1) Clarity and color of solution—Dissolve 1.0 g
obtained from 10 mL of this solution is equivalent to 1.5 to
of Astromicin Sulfate in 10 mL of water: the solution is clear
2.5z of that from 10 mL of the solution for system suitabil-
and colorless to pale yellow.
ity test.
(2) Heavy metals—Proceed with 1.0 g of Astromicin
System performance: To 100 mL of water add 5 mL of
Sulfate according to Method 1, and perform the test.
the sample solution and 2 mL of a solution of L-valine (1 in
Prepare the control solution with 2.0 mL of Standard Lead
5000). When the procedure is run with 10 mL of this solution
Solution (not more than 20 ppm).
under the above operating conditions, L-valine and astromi-
(3) Related substances—Dissolve 0.10 g of Astromicin
cin are eluted in this order with the resolution between these
Sulfate in 100 mL of water, and use this solution as the sam-
peaks being not less than 1.5, and when the procedure is run
ple solution. Pipet 2 mL of the sample solution, add water to
with 10 mL of the solution for system suitability test under
make exactly 100 mL, and use this solution as the standard
the above operating conditions, the symmetry coe‹cient of
solution. Perform the test with 10 mL each of the sample so-
the peak of astromicin is not more than 2.0.
lution and the standard solution as directed under the Liquid
System repeatability: When the test is repeated 6 times
Chromatography according to the following conditions, and
with 10 mL of the solution for system suitability test under
determine each peak area by the automatic integration
the above operating conditions, the relative standard devia-
method: the peak areas of the related substance III, having
tion of the peak area of astromicin is not more than 2.0z.
the relative retention time of about 0.1, and the related sub-
stance I, having the relative retention time of about 1.2 with Water Not more than 8.0z (0.2 g, volumetric titration,
respect to the peak of astromicin, from the sample solution back titration). Use a mixture of methanol for water deter-
are not more than the peak area of astromicin from the mination and ethylene glycol for water determination (1:1)
standard solution, the peak area of the related substance II, instead of methanol for water determination.
having the related retention time of about 0.8, is not more
Assay Perform the test according to the Cylinder-plate
than 2.0 times the peak area of astromicin from the standard
method as directed under the Microbial Assay for Antibiot-
solution, and the total area of the peaks other than astromi-
ics according to the following conditions.
cin from the sample solution is not more than 3.5 times the
(1) Test organism—Bacillus subtilis ATCC 6633
peak area of astromicin from the standard solution.
(2) Culture medium—Use the medium i in 1) Medium
Operating conditions—
for test organism [5] under (1) Agar media for seed and base
Detector: A ‰uorophotometer (excitation wavelength:
layer.
340 nm; detection wavelength: 430 nm).
(3) Standard solutions—Weigh accurately an amount of
Column: A stainless steel column 4.6 mm in inside di-
Astromicin Sulfate Reference Standard, equivalent to about
ameter and 25 cm in length, packed with octadecylsilanized
25 mg (potency), dissolve in diluted hydrochloric acid (1 in
silica gel for liquid chromatography (5 mm in particle
1000) to make exactly 25 mL, and use this solution as the
diameter).
standard stock solution. Keep the standard stock solution at
Column temperature: A constant temperature of about
5 – 159C, and use within 30 days. Take exactly a suitable
259C.
amount of the standard stock solution before use, add
Reaction coil: A stainless steel tube 0.25 mm in inside
0.1 mol/L phosphate buŠer solution for antibiotics, pH 8.0
Supplement I, JP XIV O‹cial Monographs for Part I 1389

to make solutions so that each mL contains 4 mg (potency) water to make exactly 25 mL, and use this solution as the
and 1 mg (potency), and use these solutions as the high con- standard solution. To exactly 10 mL each of the sample
centration standard solution and the low concentration stan- solution and the standard solution add exactly 2 mL of
dard solution, respectively. sodium hydroxide TS, allow to stand for exactly 15 minutes,
(4) Sample solutions—Weigh accurately an amount of add exactly 2 mL of 1 mol/L hydrochloric acid TS, exactly
Astromicin Sulfate, equivalent to about 25 mg (potency), 10 mL of 0.3 mol/L potassium hydrogen phthalate buŠer
and dissolve in 0.1 mol/L phosphate buŠer solution for solution, pH 4.6, and exactly 10 mL of 0.005 mol/L iodine
antibiotics, pH 8.0 to make exactly 25 mL. Take exactly a VS, allow to stand for exactly 20 minutes without exposure
suitable amount of this solution, add 0.1 mol/L phosphate to light. Titrate these solutions with 0.01 mol/L sodium
buŠer solution for antibiotics, pH 8.0 to make solutions so thiosulfate VS until the color of the solution changes to
that each mL contains 4 mg (potency) and 1 mg (potency), colorless. Separately, to exactly 10 mL each of the sample
and use these solutions as the high concentration sample solution and the standard solution add exactly 10 mL of
solution and the low concentration sample solution, respec- 0.3 mol/L potassium hydrogen phthalate buŠer solution,
tively. pH 4.6 and exactly 10 mL of 0.005 mol/L iodine VS, and
perform a blank determination with the same manner. De-
Containers and storage Containers—Tight containers.
termine the consumed amounts (mL) of 0.005 mol/L iodine
VS, VT and VS, of the sample solution and the standard solu-
tion: the amount of ampicillin is not more than 1.0z.
Bacampicillin Hydrochloride
Amount (mg) of ampicillin (C16H19N3O4S)
塩酸バカンピシリン V 1
= WS × T ×
VS 20
Change the origin/limits of content to read: WS: Amount (mg) of Ampicillin Reference Standard

Bacampicillin Hydrochloride is a hydrochloride of Change the Assay to read:


ampicilline ethoxycarbonyloxyethyl ester.
Assay Weigh accurately an amount of Bacampicillin
Bacampicillin Hydrochloride contains not less than
Hydrochloride and Bacampicillin Hydrochloride Reference
626 mg (potency) per mg, calculated on the anhydrous
Standard, equivalent to about 40 mg (potency), dissolve
basis. The potency of Bacampicillin Hydrochloride
each in water to make exactly 100 mL, and use these solu-
is expressed as mass (potency) of ampicillin
tions as the sample solution and the standard solution. Per-
(C16H19N3O4S: 349.40).
form the test with exactly 20 mL each of the sample solution
and the standard solution as directed under the Liquid Chro-
Add the following next to Identiˆcation:
matography according to the following conditions, and cal-
Optical rotation [a]20
D : +140 – +1709(0.1 g calculated on culate the peak areas, AT and AS, of bacampicillin of these
the anhydrous basis, ethanol (95), 25 mL, 100 mm). solutions.

Amount [mg (potency)] of ampicillin (C16H19N3O4S)


Change the Purity to read:
A
= WS × T × 1000
Purity (1) Heavy metals—Proceed with 1.0 g of Bacam- AS
picillin Hydrochloride according to Method 2, and perform
WS: Amount [mg (potency)] of Bacampicillin Hydrochlo-
the test. Prepare the control solution with 2.0 mL of Stan-
ride Reference Standard
dard Lead Solution (not more than 20 ppm).
(2) Arsenic—Prepare the test solution with 1.0 g of Operating conditions—
Bacampicillin Hydrochloride according to Method 3, and Detector: An ultraviolet absorption photometer (wave-
perform the test (not more than 2 ppm). length: 254 nm).
(3) Free ampicillin—Weigh accurately about 0.1 g of Column: A stainless steel column 4.6 mm in inside
Bacampicillin Hydrochloride, transfer into a 100-mL diameter and 15 cm in length, packed with octadecyl-
separater, add exactly 15 mL of ice-cold water to dissolve, silanized silica gel for liquid chromatography (5 mm in parti-
add and mix with exactly 10 mL of ice-cold 0.05 mol/L cle diameter).
phosphate buŠer solution, pH 7.0, then add 25 mL of ice- Column temperature: A constant temperature of about
cold chloroform, shake, and abandon the chloroform layer. 259C.
Repeat the procedure twice with two 25-mL portions of ice- Mobile phase: To 500 mL of diluted 2 mol/L sodium
cold chloroform. Centrifuge the water layer, ˆlter the super- dihydrogen phosphate TS (1 in 100), add diluted 0.05 mol/L
natant, and use the ˆltrate as the sample solution. Separate- disodium hydrogen phosphate TS (2 in 5) to adjust the pH to
ly, weigh accurately an amount of Ampicillin Reference 6.8. To 500 mL of this solution add 500 mL of acetonitrile.
Standard, equivalent to about 20 mg, and dissolve in water Flow rate: Adjust the ‰ow rate so that the retention time
to make exactly 100 mL. Pipet 5 mL of this solution, add of bacampicillin is about 6.5 minutes.
10 mL of 0.05 mol/L phosphate buŠer solution, pH 7.0 and
1390 O‹cial Monographs for Part I Supplement I, JP XIV

System suitability— 0.05 mol/L sulfuric acid TS to make 10 mL, and determine
System performance: When the procedure is run with the absorbances of this solution, AT and AF, at 252 nm and
20 mL of this solution under the above operating conditions, 290 nm as directed under the Ultraviolet-visible Spec-
the number of theoretical plates and the symmetry constant trophotometry: AF/AT is not more than 0.20.
of the peak of bacampicillin are not less than 10,000 and not
Loss on drying Not more than 5.0z (1 g, in vacuum,
more than 2, respectively.
609C, 3 hours).
System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution under the above operat- Residue on ignition Not more than 1.0z (1 g).
ing conditions, the relative standard deviation of peak areas
Assay Perform the test according to the Cylinder-plate
of bacampicillin is not more than 2.0z.
method as directed under the Microbial Assay for Antibiot-
ics according to the following conditions.
(1) Test organism—Micrococcus luteus ATCC 10240.
Add the following:
(2) Culture medium—Use the medium iii in 3) Medium
for other organisms under (1) Agar media for seed and base
Bacitracin layer.
(3) Standard solutions—Weigh accurately an amount of
バシトラシン
Bacitracin Reference Standard, equivalent to about 400
[1405-87-4] units, dissolve in phosphate buŠer solution, pH 6.0 to make
exactly 20 mL, and use this solution as the standard stock
Bacitracin is a mixture of peptide substances having solution. Keep the standard stock solution at not exceeding
antibacterial activity including bacitracin A as the 109C and use within 2 days. Take exactly a suitable amount
main component produced by the growth of Bacillus of the standard stock solution before use, add phosphate
subtilis or Bacillus licheniformis. buŠer solution, pH 6.0 to make solutions so that each mL
It contains not less than 40 Units per mg. The contains 2 units and 0.5 units, and use these solutions as the
potency of Bacitracin is expressed as unit calculated high concentration standard solution and the low concentra-
from the amount of bacitracin A (C66H103N17O16S: tion standard solution, respectively.
1422.69). 1 unit of Bacitracin is equivalent to 23.8 mg (4) Sample solutions—Weigh accurately an amount of
of bacitracin A (C66H103N17O16S). Bacitracin, equivalent about 400 units, dissolve in phosphate
buŠer solution, pH 6.0 to make exactly 20 mL. Take exactly
Description Bacitracin occurs as a white to light brown
a suitable amount of this solution, add phosphate buŠer
powder.
solution, pH 6.0 to make solutions so that each mL contains
It is freely soluble in water, and slightly soluble in ethanol
2 units and 0.5 units, and use these solutions as the high con-
(99.5).
centration sample solution and the low concentration sample
Identiˆcation (1) To 3 mL of a solution of Bacitracin (1 solution, respectively.
in 100) add 3 mL of 4-dimethylaminobenzaldehyde TS,
Containers and storage Containers—Tight containers.
shake until red-rosy to red-purple color appears, then add
Storage—In a cold place.
several drops of a solution of sodium nitrite (1 in 100), and
shake: a green to dark green color is produced.
(2) Dissolve 60 mg each of Bacitracin and Bacitracin
Reference Standard in 10 mL of water, and use these Bekanamycin Sulfate
solutions as the sample solution and the standard solution.
硫酸ベカナマイシン
Perform the test with these solutions as directed under the
Thin-layer Chromatography. Spot 1 mL each of the sample
Change to read except the structural formula
solution and the standard solution on a plate of silica gel for
and chemical name:
thin-layer chromatograph. Develop the plate with a mixture
of 1-butanol, acetic acid (100), water, pyridine and ethanol
Bekanamycin Sulfate contains not less than 680 mg
(99.5) (30:15:10:6:5) to a distance of about 10 cm, and air-
(potency) per mg, calculated on the dried basis. The
dry the plate. Spray evenly ninhydrin TS on the plate, and
potency of Bekanamycin Sulfate is expressed as mass
heat at 1109 C for 5 minutes: the spots obtained from the
(potency) of bekanamycin (C18H37N5O10: 483.51).
sample solution and the standard solution show the same Rf
value. Description Bekanamycin Sulfate occurs as a white pow-
der.
Purity (1) Heavy metals—Proceed with 1.0 g of Bacitra-
It is freely soluble in water, and practically insoluble in
cin according to Method 2, and perform the test. Prepare the
ethanol (99.5).
control solution with 2.0 mL of Standard Lead Solution (not
more than 20 ppm). Identiˆcation (1) Dissolve 20 mg of Bekanamycin Sulfate
(2) Dissolve 0.15 g of Bacitracin in 0.05 mol/L sulfuric in 2 mL of 1/15 mol/L phosphate buŠer solution, pH 5.6,
acid TS to make 100 mL. To 2 mL of this solution add add 1 mL of ninhydrin TS, and boil: a blue-purple color
Supplement I, JP XIV O‹cial Monographs for Part I 1391

develops. (3) Standard solutions—Weigh accurately an amount of


(2) Dissolve 30 mg each of Bekanamycin Sulfate and Bekanamycin Sulfate Reference Standard, previously dried,
Bekanamycin Sulfate Reference Standard in 5 mL of water, equivalent to about 20 mg (potency), dissolve in diluted
and use these solutions as the sample solution and the phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly
standard solution. Perform the test with these solutions as 50 mL, and use this solution as the standard stock solution.
directed under the Thin-layer Chromatography. Spot 5 mL Keep the standard stock solution at 5 to 159C and use within
each of the sample solution and the standard solution on a 30 days. Take exactly a suitable amount of the standard
plate of silica gel for thin-layer chromatography. Develop stock solution before use, add 0.1 mol/L phosphate buŠer
the plate with a solution of potassium dihydrogen phosphate solution, pH 8.0 to make solutions so that each mL contains
(3 in 40) to a distance of about 10 cm, and air-dry the plate. 10 mg (potency) and 2.5 mg (potency), and use these solutions
Spray evenly 0.2z ninhydrin-water saturated 1-butanol TS, as the high concentration standard solution and the low con-
and heat at 1009 C for 10 minutes: the principal spots ob- centration standard solution, respectively.
tained from the sample solution and the standard solution (4) Sample solutions—Weigh accurately an amount of
show a purple-brown color and the same Rf value. Bekanamycin Sulfate, equivalent to about 20 mg (potency),
(3) To a solution of Bekanamycin Sulfate (1 in 5) add 1 and dissolve in water to make exactly 50 mL. Take exactly a
drop of barium chloride TS: a white turbidity is produced. suitable amount of this solution, add 0.1 mol/L phosphate
buŠer solution, pH 8.0 to make solutions so that each mL
Optical rotation [a]20
D : +102 – +1169(after drying, 0.25 g,
contains 10 mg (potency) and 2.5 mg (potency), and use these
water, 25 mL, 100 mm).
solutions as the high concentration sample solution and the
pH The pH of a solution obtained by dissolving 0.50 g of low concentration sample solution, respectively.
Bekanamycin Sulfate in 10 mL of water is between 6.0 and
Containers and storage Containers—Tight containers.
8.5.

Purity (1) Clarity and color of solution—Dissolve 0.5 g


of Bekanamycin Sulfate in 5 mL of water: the solution is Benzbromarone
clear and colorless.
(2) Heavy metals—Proceed with 1.0 g of Bekanamycin ベンズブロマロン
Sulfate according to Method 4, and perform the test. Pre-
pare the control solution with 3.0 mL of Standard Lead Change the origin/limits of content to read:
Solution (not more than 30 ppm).
(3) Arsenic—Prepare the test solution with 2.0 g of Benzbromarone, when dried, contains not less than
Bekanamycin Sulfate according to Method 1, and perform 98.5z and not more than 101.0z of C17H12Br2O3.
the test (not more than 1 ppm).
(4) Related substances—Dissolve 60 mg of Bekanamycin Change the Description to read:
Sulfate in 10 mL of water, and use this solution as the sam-
Description Benzbromarone occurs as a white to light yel-
ple solution. Pipet 3 mL of the sample solution, add water to
low, crystalline powder.
make exactly 100 mL, and use this solution as the standard
It is very soluble in N, N-dimethylformamide, freely solu-
solution. Perform the test with these solutions as directed
ble in acetone, sparingly soluble in ethanol (99.5), and prac-
under the Thin-layer Chromatography. Spot 5 mL each of
tically insoluble in water.
the sample solution and the standard solution on a plate of
It dissolves in dilute sodium hydroxide TS.
silica gel for thin-layer chromatography. Develop the plate
with a solution of potassium dihydrogen phosphate (3 in 40)
Change the Identiˆcation to read:
to a distance of about 10 cm, and air-dry the plate. Spray
evenly 0.2z ninhydrin-water saturated 1-butanol TS on the Identiˆcation (1) Determine the absorption spectrum of
plate, and heat at 1009 C for 10 minutes: the spot other than a solution of Benzbromarone in 0.01 mol/L sodium
the principal spot obtained from the sample solution is not hydroxide TS (1 in 100,000) as directed under the Ultrav-
more intense than the spot from the standard solution. iolet-visible Spectrophotometry, and compare the spectrum
with the Reference Spectrum: both spectra exhibit similar
Loss on drying Not more than 5.0z (0.5 g, reduced pres-
intensities of absorption at the same wavelengths.
sure not exceeding 0.67 kPa, 609
C, 3 hours).
(2) Determine the infrared absorption spectrum of Ben-
Residue on ignition Not more than 0.5z (1 g). zbromarone, previously dried, as directed in the potassium
bromide disk method under the Infrared Spectrophotomet-
Assay Perform the test according to the Cylinder-plate
ry, and compare the spectrum with the Reference Spectrum:
method as directed under the Microbial Assay for Antibiot-
both spectra exhibit similar intensities of absorption at the
ics according to the following conditions.
same wave numbers.
(1) Test organism—Bacillus subtilis ATCC 6633
(2) Culture medium—Use the medium i in 1) Medium
for test organism [5] under (1) Agar media for seed and base
layer having pH 7.8 to 8.0 after sterilization.
1392 O‹cial Monographs for Part I Supplement I, JP XIV

Change the Purity to read: Add the following:


Purity (1) Sulfate—Dissolve 1.0 g of Benzbromarone in
40 mL of acetone, and add 1 mL of dilute hydrochloric acid Benzylpenicillin Benzathine
and water to make 50 mL. Perform the test using this solu-
ベンジルペニシリンベンザチン
tion as the test solution. Prepare the control solution as fol-
lows: to 0.40 mL of 0.005 mol/L sulfuric acid VS add 40 mL
of acetone, 1 mL of dilute hydrochloric acid and water to
make 50 mL (not more than 0.019z).
(2) Soluble halides—Dissolve 0.5 g of Benzbromarone in
40 mL of acetone, and add 6 mL of dilute nitric acid and
water to make 50 mL. Proceed with this solution as directed (C16H18N2O4S)2.C16H20N2.4H2O: 981.18
under the Chloride Limit Test. Prepare the control solution (2S,5R,6R )-3,3-Dimethyl-7-oxo-6-phenylacetylamino-4-
as follows: to 0.25 mL of 0.01 mol/L hydrochloric acid VS thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid
add 40 mL of acetone, 6 mL of dilute nitric acid and water hemi(N,N ?-dibenzylethylenediamine) dihydrate
to make 50 mL. [41372-02-5]
(3) Heavy metals—Proceed with 2.0 g of Benzbroma-
rone according to Method 2, and perform the test. Prepare Benzylpenicillin Benzathine contains not less than
the control solution with 2.0 mL of Standard Lead Solution 1152 units per mg, calculated on the anhydrous basis.
(not more than 10 ppm). The potency of Benzylpenicillin Benzathine is ex-
(4) Iron—Prepare the test solution with 1.0 g of Ben- pressed as unit calculated from the amount of benzyl-
zbromarone according to Method 3, and perform the test penicillin sodium (C16H17N2NaO4S: 356.37). 1 unit of
according to Method A. Prepare the control solution with Benzylpenicillin Benzathine is equivalent to 0.6 mg of
2.0 mL of Standard Iron Solution (not more than 20 ppm). benzylpenicillin sodium (C16H17N2NaO4S).
(5) Related substances—Dissolve 0.10 g of Benzbroma-
Description Benzylpenicillin Benzathine occurs as a white
rone in 10 mL of acetone, and use this solution as the sample
crystalline powder.
solution. Pipet 1 mL of the sample solution, add acetone to
It is slightly soluble in methanol and in ethanol (99.5), and
make exactly 100 mL, and use this solution as the standard
practically insoluble in water.
solution. Perform the test with these solutions as directed
under the Thin-layer Chromatography. Spot 10 mL each of Identiˆcation (1) Determine the absorption spectrum of
the sample solution and the standard solution on a plate of a solution of Benzylpenicillin Benzathine in methanol (1 in
silica gel with ‰uorescent indicator for thin-layer chro- 2000) as directed under the Ultraviolet-visible Spectrophoto-
matography. Develop the plate with a mixture of cyclo- metry, and compare the spectrum with the Reference Spec-
hexane, 4-methyl-2-pentanone, ethanol (99.5) and acetic trum: both spectra exhibit similar intensities of absorption at
acid (100) (100:20:2:1) to a distance of about 15 cm, and air- the same wavelengths.
dry the plate. Examine under ultraviolet light (main wave- (2) Determine the infrared absorption spectrum of
length: 254 nm): the spots other than the principal spot from Benzylpenicillin Benzathine as directed in the potassium
the sample solution are not more intense than the spot from bromide disk method under the Infrared Spectrophotomet-
the standard solution. ry, and compare the spectrum with the Reference Spectrum:
both spectra exhibit similar intensities of absorption at the
same wave numbers.

Optical rotation [a]20


D : +217 – +2339(0.1 g calculated on
the anhydrous basis, methanol, 20 mL, 100 mm).

Purity (1) Heavy metals—Proceed with 1.0 g of Benzyl-


penicillin Benzathine according to Method 2, and perform
the test. Prepare the control solution with 2.0 mL of Stan-
dard Lead Solution (not more than 20 ppm).
(2) Arsenic—Prepare the test solution with 1.0 g of
Benzylpenicillin Benzathine according to Method 3, and
perform the test (not more than 2 ppm).
(3) Related substances—Dissolve 70 mg of Benzyl-
penicillin Benzathine in 25 mL of methanol, add a solution
prepared by dissolving 1.02 g of disodium hydrogen phos-
phate and 6.80 g of potassium dihydrogen phosphate in
water to make 1000 mL to make 50 mL, and use this solu-
tion as the sample solution. Pipet 1 mL of the sample solu-
tion, add the mobile phase A to make exactly 100 mL, and
Supplement I, JP XIV O‹cial Monographs for Part I 1393

use this solution as the standard solution. Perform the test tion).
with 20 mL each of the sample solution and the standard
Assay Perform the test according to the Cylinder-plate
solution as directed under the Liquid Chromatography
method as directed under the Microbial Assay for Antibiot-
according to the following conditions, and determine each
ics according to the following conditions.
peak area by the automatic integration method: the area of
(1) Test organism—Staphylococcus aureus ATCC 6538
the peak having the relative retention time of about 2.4 with
P
respect to benzylpenicillin obtained from the sample solu-
(2) Culture medium—Use the medium iii in 3) Medium
tion is not more than 2 times the total area of the peaks of
for other organisms under (1) Agar media for seed and base
benzylpenicillin and benzathine obtained from the standard
layer.
solution, and the total area of the peaks other than benzyl-
(3) Standard solutions—Weigh accurately an amount of
penicillin, benzathine and the peak having the relative reten-
Benzylpenicillin Sodium Reference Standard, equivalent to
tion time of about 2.4 obtained from the sample solution is
about 20,000 units, dissolve in phosphate buŠer solution,
not more than the total area of the peaks of benzylpenicillin
pH 6.0 to make exactly 10 mL, and use this solution as the
and benzathine obtained from the standard solution.
standard stock solution. Keep the standard stock solution at
Operating conditions—
not exceeding 59 C and use within 2 days. Take exactly a suit-
Detector: An ultraviolet absorption photometer (wave-
able amount of the standard stock solution before use, add
length: 220 nm).
phosphate buŠer solution, pH 6.0 to make solutions so that
Column: A stainless steel column 4.0 mm in inside di-
each mL contains 2 units and 0.5 units, and use these solu-
ameter and 25 cm in length, packed with octadecylsilanized
tions as the high concentration standard solution and the
silica gel for liquid chromatography (5 mm in particle
low concentration standard solution, respectively.
diameter).
(4) Sample solutions—Weigh accurately an amount of
Column temperature: A constant temperature of about
Benzylpenicillin Benzathine, equivalent about 20,000 units,
409C.
and dissolve in N, N-dimethylformamide to make exactly
Mobile phase A: A mixture of water, methanol and
10 mL. Take exactly a suitable amount of this solution, add
0.25 mol/L potassium dihydrogen phosphate TS, pH 3.5
phosphate buŠer solution, pH 6.0 to make solutions so that
(6:3:1).
each mL contains 2 units and 0.5 units, and use these solu-
Mobile phase B: A mixture of methanol, water and
tions as the high concentration sample solution and the low
0.25 mol/L potassium dihydrogen phosphate TS, pH 3.5
concentration sample solution, respectively.
(6:3:1).
Flowing of the mobile phase: Control the gradient by mix- Containers and storage Containers—Tight containers.
ing the mobile phases A and B as directed in the following Storage—Light-resistant.
table.

Time after injection


of sample (min)
Mobile phase
A (z )
Mobile phase
B (z ) Benzylpenicillin Potassium
0 – 10 75 25 ベンジルペニシリンカリウム
10 – 20 75 → 0 25 → 100
20 – 55 0 100 Change to read except the structural formula
and chemical name:
Flow rate: 1.0 mL/min
Time span of measurement: About 3 times as long as the
Benzylpenicillin Potassium contains not less than
retention time of benzylpenicillin after the solvent peak.
1430 units per mg, calculated on the dried basis. The
System suitability—
potency of Benzylpenicillin Potassium is expressed
Test for required detectability: To exactly 1 mL of the
as mass unit of benzylpenicillin potassium
standard solution add the mobile phase A to make exactly
(C16H17KN2O4S). One unit of Benzylpenicillin Potas-
20 mL. Conˆrm that the peak area of benzylpenicillin
sium is equivalent to 0.57 mg of benzylpenicillin potas-
obtained from 20 mL of this solution is equivalent to 3.5 to
sium.
6.5z of that from the standard solution.
System performance: When the procedure is run with Description Benzylpenicillin Potassium occurs as white,
20 mL of the sample solution under the above operating con- crystals or crystalline powder.
ditions, benzathine and benzylpenicillin are eluted in this It is very soluble in water, and slightly soluble in ethanol
order with the resolution between these peaks being not less (99.5).
than 25.
Identiˆcation (1) Determine the absorption spectrum of
System repeatability: When the test is repeated 3 times
a solution of Benzylpenicillin Potassium (1 in 1000) as
with 20 mL of the standard solution under the above operat-
directed under the Ultraviolet-visible Spectrophotometry,
ing conditions, the relative standard deviation of the peak
and compare the spectrum with the Reference Spectrum or
area of benzylpenicillin is not more than 2.0z.
the spectrum of a solution of Benzylpenicillin Potassium
Water 5.0 – 8.0z (1 g, volumetric titration, direct titra- Reference Standard prepared in the same manner as the
1394 O‹cial Monographs for Part I Supplement I, JP XIV

sample solution: both spectra exhibit similar intensities of adjusted the pH to 8.0 with phosphoric acid.
absorption at the same wavelengths. Flow rate: Adjust the ‰ow rate so that the retention time
(2) Determine the infrared absorption spectrum of Ben- of benzylpenicillin is about 7.5 minutes.
zylpenicillin Potassium as directed in the potassium bromide Time span of measurement: About 5 times as long as the
disk method under the Infrared Spectrophotometry, and retention time of benzylpenicillin.
compare the spectrum with the Reference Spectrum or the System suitability—
spectrum of Benzylpenicillin Potassium Reference Standard: Test for required detection: Pipet 10 mL of the standard
both spectra exhibit similar intensities of absorption at the solution, and add water to make exactly 100 mL. Conˆrm
same wave numbers. that the peak area of benzylpenicillin obtained from 20 mL
(3) Benzylpenicillin Potassium responds to the Qualita- of this solution is equivalent to 7 to 13z of that from 20 mL
tive Test (1) for potassium salt. of the standard solution.
System performance: Dissolve 40 mg of Benzylpenicillin
Optical rotation [a]20
D : +270 – +3009(1.0 g calculated on
Potassium in 20 mL of water. Separately, dissolve 10 mg of
the dried basis, water, 50 mL, 100 mm).
methyl parahydroxybenzoate in 20 mL of acetonitrile. To
pH The pH of a solution obtained by dissolving 1.0 g of 1 mL of this solution add water to make 20 mL. Mix 1 mL
Benzylpenicillin Potassium in 100 mL of water is between each of these solutions, and add water to make 100 mL.
5.0 and 7.5. When the procedure is run with 20 mL of this solution under
the above operating conditions, benzylpenicillin and methyl
Purity (1) Clarity and color of solution-A solution
parahydroxybenzoate are eluted in this order with the reso-
obtained by dissolving 1 g of Benzylpenicillin Potassium in
lution between these peaks being not less than 8.
10 mL of water is clear, and colorless or light yellow.
System repeatability: When the test is repeated 5 times
(2) Heavy metals—Proceed with 2.0 g of Benzylpenicil-
with 20 mL of the standard solution under the above operat-
lin Potassium according to Method 4, and perform the test.
ing conditions, the relative standard deviation of the peak
Prepare the control solution with 2.0 mL of Standard Lead
area of benzylpenicillin is not more than 2.0z.
Solution (not more than 10 ppm).
(3) Arsenic—Prepare the test solution by incinerating Loss on drying Not more than 1.0z (3 g, reduced pressure
1.0 g of Benzylpenicillin Potassium according to Method 4, not exceeding 0.67 kPa, 609C, 3 hours).
and perform the test. In the incineration, use a crucible of
Assay Perform the test according to the Cylinder-plate
porcelain, and after addition of 10 mL of a solution of
method as directed under the Microbial Assay for Antibiot-
magnesium nitrate hexahydrate in ethanol (95) (1 in 10) add
ics according to the following conditions.
1 mL of hydrogen peroxide (30), then burn the ethanol (not
(1) Test organism—Staphylococcus aureus ATCC 6538
more than 2 ppm).
P
(4) Related substances—Dissolve 40 mg of Benzyl-
(2) Culture medium—Use the medium iii in 3) Medium
penicillin Potassium in 20 mL of water, and use this solution
for other organisms under (1) Agar media for seed and base
as the sample solution. Pipet 1 mL of the sample solution,
layer.
add water to make exactly 100 mL, and use this solution as
(3) Standard solutions—Weigh accurately an amount of
the standard solution. Perform the test with 20 mL each of
Benzylpenicillin Potassium Reference Standard, equivalent
the sample solution and the standard solution as directed
to about 40,000 units, dissolve in phosphate buŠer solution,
under the Liquid Chromatography according to the follow-
pH 6.0 to make exactly 100 mL, and use this solution as the
ing conditions, and determine each peak area by the auto-
standard stock solution. Keep the standard stock solution at
matic integration method: the area of the peak other than
not exceeding 59 C and use within 2 days. Take exactly a suit-
benzylpenicillin obtained from the sample solution is not
able amount of the standard stock solution before use, add
more than the peak area of benzylpenicillin from the stan-
phosphate buŠer solution, pH 6.0 to make solutions so that
dard solution, and the total area of the peaks other than ben-
each mL contains 2 units and 0.5 units, and use these solu-
zylpenicillin from the sample solution is not more than 3
tions as the high concentration standard solution and the
times the peak area of benzylpenicillin from the standard
low concentration standard solution, respectively.
solution.
(4) Sample solutions—Weigh accurately an amount of
Operating conditions—
Benzylpenicillin Potassium, equivalent to about 40,000
Detector: An ultraviolet absorption photometer (wave-
units, and dissolve in phosphate buŠer solution, pH 6.0 to
length: 254 nm).
make exactly 100 mL. Take exactly a suitable amount of this
Column: A stainless steel column 4.6 mm in inside di-
solution, add phosphate buŠer solution, pH 6.0 to make
ameter and 25 cm in length, packed with octadecylsilanized
solutions so that each mL contains 2 units and 0.5 units, and
silica gel for liquid chromatography (7 mm in particle di-
use these solutions as the high concentration sample solution
ameter).
and the low concentration sample solution, respectively.
Column temperature: A constant temperature of about
259C. Containers and storage Containers—Tight containers.
Mobile phase: A mixture of a solution of diammonium
hydrogen phosphate (33 in 5000) and acetonitrile (19:6),
Supplement I, JP XIV O‹cial Monographs for Part I 1395

Betamethasone Sodium Phosphate Betamethasone Valerate


リン酸ベタメタゾンナトリウム 吉草酸ベタメタゾン

Change the Assay to read: Change the Assay to read:


Assay Weigh accurately about 20 mg each of Betametha- Assay Dissolve about 10 mg each of Betamethasone Valer-
sone Sodium Phosphate and Betamethasone Sodium Phos- ate and Betamethasone Valerate Reference Standard, previ-
phate Reference Standard (determine its water content be- ously dried and accurately weighed, in methanol to make
fore using in the same manner as Betamethasone Sodium exactly 100 mL. Pipet 10 mL each of these solutions, add
Phosphate), and dissolve each in methanol to make exactly 10 mL each of the internal standard solution, and use these
20 mL. Pipet 5 mL each of these solutions, and exactly 5 mL solutions as the sample solution and the standard solution,
of the internal standard solution, then add methanol to respectively. Perform the test with 10 mL each of the sample
make 50 mL, and use these solutions as the sample solution solution and the standard solution as directed under the
and the standard solution, respectively. Perform the test Liquid Chromatography according to the following condi-
with 10 mL each of the sample solution and the standard so- tions, and calculate the ratios, QT and QS, of the peak area
lution as directed under the Liquid Chromatography accord- of betamethasone valerate to that of the internal standard,
ing to the following conditions, and calculate the ratios, QT respectively.
and QS, of the peak area of betamethasone phosphate to that
of the internal standard, respectively. QT
Amount (mg) of C27H37FO6 = WS ×
QS
QT
Amount (mg) of C22H28FNa2O8P = WS × WS: Amount (mg) of Betamethasone Valerate Reference
QS
Standard
WS: Amount (mg) of Betamethasone Sodium Phosphate
Reference Standard, calculated on the anhydrous Internal standard solution—A solution of isoamyl benzoate
basis in methanol (1 in 1000).
Operating conditions—
Internal standard solution—A solution of butyl parahydrox- Detector: An ultraviolet absorption photometer (wave-
ybenzoate in methanol (1 in 5000). length: 254 nm).
Operating conditions— Column: A stainless steel column 4.0 mm in inside di-
Detector: An ultraviolet absorption photometer (wave- ameter and 20 cm in length, packed with octadecylsilanized
length: 254 nm). silica gel for liquid chromatography (7 mm in particle di-
Column: A stainless steel column 4.0 mm in inside di- ameter).
ameter and 25 cm in length, packed with octadecylsilanized Column temperature: A constant temperature of about
silica gel for liquid chromatography (7 mm in particle 259C.
diameter). Mobile phase: A mixture of methanol and water (7:3).
Column temperature: A constant temperature of about Flow rate: Adjust the ‰ow rate so that the retention time
259C. of betamethasone valerate is about 10 minutes.
Mobile phase: Dissolve 1.6 g of tetra-n-butylammonium System suitability—
bromide, 3.2 g of disodium hydrogen phosphate 12-water System performance: When the procedure is run with
and 6.9 g of potassium dihydrogen phosphate in 1000 mL of 10 mL of the standard solution under the above operating
water, and add 1500 mL of methanol. conditions, betamethasone valerate and the internal stan-
Flow rate: Adjust the ‰ow rate so that the retention time dard are eluted in this order with the resolution between
of betamethasone phosphate is about 5 minutes. these peaks being not less than 5.
System suitability— System repeatability: When the test is repeated 6 times
System performance: When the procedure is run with with 10 mL of the standard solution under the above operat-
10 mL of the standard solution under the above operating ing conditions, the relative standard deviation of the ratios
conditions, betamethasone phosphate and the internal stan- of the peak area of betamethasone valerate to that of the in-
dard are eluted in this order with the resolution between ternal standard is not more than 1.0z.
these peaks being not less than 10.
System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios
of the peak area of betamethasone phosphate to that of the
internal standard is not more than 1.0z.
1396 O‹cial Monographs for Part I Supplement I, JP XIV

Bisacodyl Suppositories Bleomycin Hydrochloride


ビサコジル坐剤 塩酸ブレオマイシン

Change the Assay to read: Change to read except the structural formula
and chemical name:
Assay Weigh accurately not less than 20 Bisacodyl Sup-
positories, make them ˆne fragments carefully, and mix
Bleomycin Hydrochloride contains not less than
uniformly. Weigh accurately a portion of the fragments,
1400 mg (potency) and not more than 2000 mg
equivalent to about 10 mg of bisacodyl (C22H19NO4), add
(potency) per mg. The potency of Bleomycin
40 mL of tetrahydrofuran, warm to 409C, dissolve by shak-
Hydrochloride is expressed as mass (potency) of
ing, cool, and add tetrahydrofuran to make exactly 50 mL.
bleomycin A2 (C55H84ClN17O21S3: 1451.00).
Pipet 5 mL of this solution, add exactly 5 mL of the internal
standard solution, and add the mobile phase to make Description Bleomycin Hydrochloride occurs as a white to
100 mL. Cool this solution in ice for 30 minutes, centrifuge, yellowish white powder.
ˆlter the supernatant liquid through a membrane ˆlter with It is freely soluble in water, and slightly soluble in ethanol
pore size of 0.5 mm, discard the ˆrst 10 mL of the ˆltrate, (95).
and use the subsequent ˆltrate as the sample solution. It is hygroscopic.
Separately, weigh accurately about 10 mg of Bisacodyl
Identiˆcation (1) To 4 mg of Bleomycin Hydrochloride
Reference Standard, previously dried at 1059C for 2 hours,
add 5 mL of copper (II) sulfate TS, and dissolve in water to
and dissolve in tetrahydrofuran to make exactly 50 mL.
make 100 mL. Determine the absorption spectrum of this
Pipet 5 mL of this solution, proceed in the same manner as
solution as directed under the Ultraviolet-visible Spec-
the sample solution, and use this solution as the standard
trophotometry, and compare the spectrum with the Refer-
solution. Perform the test with 20 mL each of the sample so-
ence Spectrum: both spectra exhibit similar intensities of
lution and the standard solution as directed under the Liquid
absorption at the same wavelengths.
Chromatography according to the following conditions, and
(2) Determine the infrared absorption spectrum of
calculate the ratios, QT and QS, of the peak area of bisacodyl
Bleomycin Hydrochloride as directed in the potassium
to that of the internal standard, respectively.
bromide disk method under the Infrared Spectrophotomet-
QT ry, and compare the spectrum with the Reference Spectrum:
Amount (mg) of bisacodyl (C22H19NO4) = WS ×
QS both spectra exhibit similar intensities of absorption at the
same wave numbers.
WS: Amount (mg) of Bisacodyl Reference Standard
(3) A solution of Bleomycin Hydrochloride (1 in 100)
Internal standard solution—A solution of ethyl parahydrox- responds to the Qualitative Test (2) for chloride.
ybenzoate in acetonitrile (3 in 100,000).
pH The pH of a solution obtained by dissolving 0.10 g of
Operating conditions—
Bleomycin Hydrochloride in 20 mL of water is between 4.5
Detector: An ultraviolet absorption photometer (wave-
and 6.0.
length: 254 nm).
Column: A stainless steel column 4 mm in inside diameter Content ratio of the active principle Dissolve 10 mg of
and 30 cm in length, packed with octadecylsilanized silica gel Bleomycin Hydrochloride in 20 mL of water, and use this
for liquid chromatography (10 mm in particle diameter). solution as the sample solution. Perform the test with 20 mL
Column temperature: A constant temperature of about of the sample solution as directed under the Liquid Chro-
259C. matography according to the following conditions, and de-
Mobile phase: A mixture of 0.01 mol/L citric acid TS, termine each peak area by the automatic integration
acetonitrile and methanol (2:1:1). method: the peak area of bleomycin A2 (the ˆrst principal
Flow rate: Adjust the ‰ow rate so that the retention time peak) is between 55z and 70z, that of bleomycin B2 (the
of bisacodyl is about 8 minutes. second principal peak) is between 25z and 32z, the total
System suitability— peak area of bleomycin A2 and bleomycin B2 is not less than
System performance: When the procedure is run with 85z, the peak area of demethylbleomycin A2 (a peak having
20 mL of the standard solution under the above operating the relative retention time of 1.5 – 2.5 to bleomycin A2) is
conditions, the internal standard and bisacodyl are eluted in not more than 5.5z, and the total area of the rest peaks is
this order with the resolution between these peaks being not not more than 9.5z.
less than 2.0. Operating conditions—
System repeatability: When the test is repeated 6 times Detector: An ultraviolet absorption photometer (wave-
with 20 mL of the standard solution under the above operat- length: 254 nm).
ing conditions, the relative standard deviation of the ratios Column: A stainless steel column 4.6 mm in inside di-
of the peak area of bisacodyl to that of the internal standard ameter and 25 cm in length, packed with octadecylsilanized
is not more than 1.0z. silica gel for liquid chromatography (7 mm in particle
Supplement I, JP XIV O‹cial Monographs for Part I 1397

diameter). ics according to the following conditions.


Column temperature: A constant temperature of about (1) Test organism—Mycobacterium smegmatis ATCC
409C. 607
Mobile phase stock solution: Dissolve 0.96 g of sodium (2) Agar medium for seed, base layer and transferring
1-pentanesulfonate and 1.86 g of disodium dihydrogen the test organism
ethylenediamine tetraacetate dihydrate in 1000 mL of water Glycerin 10.0 g
and 5 mL of acetic acid (100), and adjust the pH to 4.3 with Peptone 10.0 g
ammonia TS. Meat extract 10.0 g
Mobile phase A: A mixture of the mobile phase stock Sodium chloride 3.0 g
solution and methanol (9:1). Agar 15.0 g
Mobile phase B: A mixture of the mobile phase stock solu- Water 1000 mL
tion and methanol (3:2). Mix all the components, and sterilize. Adjust the pH after
Flowing of the mobile phase: Control the gradient by sterilization to 6.9 – 7.1 with sodium hydroxide TS.
mixing the mobile phases A and B as directed in the follow- (3) Liquid media for suspending the test organism
ing table. Glycerin 10.0 g
Peptone 10.0 g
Time after injection Mobile phase Mobile phase Meat extract 10.0 g
of sample (min) A (z ) B (z )
Sodium chloride 3.0 g
0 – 60 100 → 0 0 → 100 Water 1000 mL
60 – 75 0 100 Mix all the components, and sterilize. Adjust the pH after
sterilization to 6.9 – 7.1 with sodium hydroxide TS.
Flow rate: About 1.2 mL per minute.
(4) Preparation of seeded agar layer—Cultivate the test
Time span of measurement: 20 minutes after elution of
organism on the slant of the agar medium for transferring
the peak of demethylbreomycin A2 after the solvent peak.
the test organism at 279C for 40 to 48 hours, then inoculate
System suitability—
the test organism thus obtained in 100 mL of the liquid
System performance: When the procedure is run with
media for suspending the test organism, cultivate with shak-
20 mL of the sample solution under the above operating con-
ing at between 259 C and 279C for 5 days, and use this as the
ditions, bleomycin A2 and bleomycin B2 are eluted in this
suspension of test organism. Store the suspension of test
order with the resolution between these peaks being not less
organism at a temperature not exceeding 59 C, and use
than 5.
within 14 days. Add 0.5 mL of the suspension of test organ-
System repeatability: When the test is repeated 6 times
ism in 100 mL of the agar medium for seed previously kept
with 20 mL of the sample solution under the above operating
at 489C, mix thoroughly, and use as the seeded agar layer.
conditions, the relative standard deviation of the peak area
(5) Preparation of cylinder-agar plate—Proceed as
of bleomycin A2 is not more than 2.0z.
directed in the Preparation of cylinder-agar plate under the
Purity (1) Clarity and color of solution—A solution Microbial Assay for Antibiotics, dispensing 5.0 mL of agar
obtained by dissolving 0.08 g of Bleomycin Hydrochloride medium for base layer and 8.0 mL of the agar medium for
in 4 mL of water is clear and colorless. seed into the Petri dish.
(2) Copper—Dissolve exactly 75 mg of Bleomycin (6) Standard solutions—Weigh accurately an amount of
Hydrochloride in exactly 10 mL of diluted nitric acid (1 in Bleomycin A2 Hydrochloride Reference Standard, previous-
100), and use this solution as the sample solution. Separate- ly dried under reduced pressure not exceeding 0.67 kPa at an
ly, to exactly 15 mL of Standard Copper Solution add dilut- ordinary temperature for 3 hours, equivalent to about 15 mg
ed nitric acid (1 in 100) to make exactly 100 mL, and use this (potency), dissolve in 0.1 mol/L phosphate buŠer solution,
solution as the standard solution. Perform the test with the pH 6.8 to make exactly 100 mL, and use this solution as the
sample solution and the standard solution as directed under standard stock solution. Keep the standard stock solution at
the Atomic Absorption Spectrophotometry according to the 59 C or below, and use within 30 days. Take exactly a suita-
following conditions: the absorbance of the sample solution ble amount of the standard stock solution before use, add
is not more than that of the standard solution (not more 0.1 mol/L phosphate buŠer solution, pH 6.8 to make solu-
than 200 ppm). tions so that each mL contains 30 mg (potency) and 15 mg
Gas: Combustible gas—Acetylene (potency), and use these solutions as the high concentration
Supporting gas—Air standard solution and the low concentration standard solu-
Lamp: Copper hollow-cathode lamp tion, respectively.
Wavelength: 324.8 nm (7) Sample solutions—Weigh accurately an amount of
Bleomycin Hydrochloride, equivalent to about 15 mg
Loss on drying Not more than 5.0z (60 mg, in vacuum,
(potency), and dissolve in 0.1 mol/L phosphate buŠer solu-
phosphorus (V) oxide, 609C, 3 hours). Take the sample to be
tion, pH 6.8 to make exactly 100 mL. Take exactly a suitable
tested while avoiding moisture absorption.
amount of this solution, add 0.1 mol/L phosphate buŠer
Assay Perform the test according to the Cylinder-plate solution, pH 6.8 to make solutions so that each mL contains
method as directed under the Microbial Assay for Antibiot- 30 mg (potency) and 15 mg (potency), and use these solutions
1398 O‹cial Monographs for Part I Supplement I, JP XIV

as the high concentration sample solution and the low con- length: 254 nm).
centration sample solution, respectively. Column: A stainless steel column 4.6 mm in inside di-
ameter and 25 cm in length, packed with octadecylsilanized
Containers and storage Containers—Tight containers.
silica gel for liquid chromatography (7 mm in particle di-
ameter).
Column temperature: A constant temperature of about
Bleomycin Sulfate 409C.
Mobile phase stock solution: Dissolve 0.96 g of sodium 1-
硫酸ブレオマイシン
pentanesulfonate and 1.86 g of disodium dihydrogen
ethylenediamine tetraacetate dihydrate in 1000 mL of water
Change to read except the structural formula
and 5 mL of acetic acid (100), and adjust the pH to 4.3 with
and chemical name:
ammonia TS.
Mobile phase A: A mixture of the mobile phase stock
Bleomycin Sulfate contains not less than 1400 mg
solution and methanol (9:1).
(potency) and not more than 2000 mg (potency) per
Mobile phase B: A mixture of the mobile phase stock solu-
mg. The potency of Bleomycin Sulfate is expressed as
tion and methanol (3:2).
mass (potency) of bleomycin A2 (C55H84ClN17O21S3:
Flowing of the mobile phase: Control the gradient by
1451.00).
mixing the mobile phases A and B as directed in the follow-
Description Bleomycin Sulfate occurs as a white to yellow- ing table.
ish white powder.
It is freely soluble in water, and slightly soluble in ethanol Time after injection Mobile phase Mobile phase
of sample (min) A (z ) B (z )
(95).
It is hygroscopic. 0 – 60 100 → 0 0 → 100
60 – 75 0 100
Identiˆcation (1) To 4 mg of Bleomycin Sulfate add 5 mL
of copper (II) sulfate TS, and dissolve in water to make Flow rate: About 1.2 mL/min
100 mL. Determine the absorption spectrum of this solution Time span of measurement: Twenty minutes after elution
as directed under the Ultraviolet-visible Spectrophotometry, of the peak of demethylbleomycin A2 after the solvent peak.
and compare the spectrum with the Reference Spectrum: System suitability—
both spectra exhibit similar intensities of absorption at the System performance: When the procedure is run with
same wavelengths. 20 mL of the sample solution under the above operating con-
(2) Determine the infrared absorption spectrum of ditions, bleomycin A2 and bleomycin B2 are eluted in this
Bleomycin Sulfate as directed in the potassium bromide disk order with the resolution between these peaks being not less
method under the Infrared Spectrophotometry, and com- than 5.
pare the spectrum with the Reference Spectrum: both spec- System repeatability: When the test is repeated 6 times
tra exhibit similar intensities of absorption at the same wave with 20 mL of the sample solution under the above operating
numbers. conditions, the relative standard deviation of the peak area
(3) Bleomycin Sulfate responds to the Qualitative Tests of bleomycin A2 is not more than 2.0z.
(1) and (2) for sulfate.
Purity (1) Clarity and color of solution—A solution
pH The pH of a solution obtained by dissolving 10 mg of obtained by dissolving 0.08 g of Bleomycin Sulfate in 4 mL
Bleomycin Sulfate in 20 mL of water is between 4.5 and 6.0. of water is clear and colorless.
(2) Copper—Dissolve exactly 75 mg of Bleomycin Sul-
Content ratio of the active principle Dissolve 10 mg of
fate in 10 mL of diluted nitric acid (1 in 100), and use this
Bleomycin Sulfate in 20 mL of water, and use this solution
solution as the sample solution. Separately, to exactly 15 mL
as the sample solution. Perform the test with 20 mL of the
of Standard Copper Solution add diluted nitric acid (1 in
sample solution as directed under the Liquid Chro-
100) to make exactly 100 mL, and use this solution as the
matography according to the following conditions, and de-
standard solution. Perform the test with the sample solution
termine each peak area by the automatic integration
and the standard solution as directed under the Atomic Ab-
method: the peak area of bleomycin A2 (the ˆrst principal
sorption Spectrophotometry according to the following con-
peak) is between 55z and 70z, that of bleomycin B2 (the
ditions: the absorbance of the sample solution is not more
second principal peak) is between 25z and 32z, the total
than that of the standard solution (not more than 200 ppm).
peak area of bleomycin A2 and bleomycin B2 is not less than
Gas: Combustible gas—Acetylene
85z, the peak area of demethylbleomycin A2 (a peak having
Supporting gas—Air
the relative retention time of 1.5 – 2.5 against bleomycin A2)
Lamp: Copper hollow-cathode lamp
is not more than 5.5z, and the total area of the rest peaks is
Wavelength: 324.8 nm
not more than 9.5z.
Operating conditions— Loss on drying Not more than 3.0z (60 mg, in vacuum,
Detector: An ultraviolet absorption photometer (wave- phosphorus (V) oxide, 609
C, 3 hours). Take the sample to be
Supplement I, JP XIV O‹cial Monographs for Part I 1399

tested while avoiding moisture absorption. make exactly 100 mL. Take exactly a suitable amount of this
solution, add 0.1 mol/L phosphate buŠer solution, pH 6.8
Assay Perform the test according to the Cylinder-plate
to make solutions so that each mL contains 30 mg (potency)
method as directed under the Microbial Assay for Antibiot-
and 15 mg (potency), and use these solutions as the high con-
ics according to the following conditions.
centration sample solution and the low concentration sample
(1) Test organism—Mycrobacterium smegmatis ATCC
solution, respectively.
607
(2) Agar medium for seed, base layer and transferring Containers and storage Containers—Tight containers.
the test organism
Glycerin 10.0 g
Peptone 10.0 g Calcium Chloride Injection
Meat extract 10.0 g
Sodium chloride 3.0 g 塩化カルシウム注射液
Agar 15.0 g
Water 1000 mL Change the origin/limits of content to read:
Mix all the components and sterilize. Adjust the pH after
sterilization to 6.9 – 7.1 with sodium hydroxide TS. Calcium Chloride Injection is an aqueous solution
(3) Liquid media for suspending the test organism for injection. It contains not less than 95.0z and not
Glycerin 10.0 g more than 105.0z of the labeled amount of calcium
Peptone 10.0 g chloride (CaCl2: 110.98).
Meat extract 10.0 g The concentration of Calcium Chloride Injection is
Sodium chloride 3.0 g expressed as the quantity of calcium chloride (CaCl2).
Water 1000 mL
Mix all the components and sterilize. Adjust the pH after Change the Description to read:
sterilization to 6.9 – 7.1 with sodium hydroxide TS.
Description Calcium Chloride Injection is a clear, colorless
(4) Preparation of seeded agar layer-Cultivate the test
liquid.
organism on the slant of the agar medium for transferring
the test organism at 279C for 40 to 48 hours, then inoculate
Add the following next to Identiˆcation:
the test organism thus obtained in 100 mL of the liquid
media for suspending the test organism, cultivate with shak- pH 4.5 – 7.5
ing at between 259 C and 279C for 5 days, and use this as the
Bacterial endotoxins Less than 0.30 EU/mg.
suspension of test organism. Store the suspension of test
organism at a temperature not exceeding 59 C, and use
Change the Containers and storage to read:
within 14 days. Add 0.5 mL of the suspension of test organ-
ism in 100 mL of the agar medium for seed previously kept Containers and storage Containers—Hermetic containers.
at 489C, mix thoroughly, and use as the seeded agar layer. Plastic containers for aqueous injections may be used.
(5) Preparation of cylinder-agar plate—Proceed as
directed in 7. Preparation of cylinder-agar plate under the
Microbial Assay for Antibiotics, dispensing 5.0 mL of agar Calcium Polystyrene Sulfonate
medium for base layer and 8.0 mL of the agar medium for
seed into the Petri dish. ポリスチレンスルホン酸カルシウム
(6) Standard solutions—Weigh accurately an amount of
Bleomycin A2 Hydrochloride Reference Standard, previous- Change the Purity (4) to read:
ly dried under reduced pressure not exceeding 0.67 kPa at an Purity
ordinary temperature for 3 hours, equivalent to about 15 mg (4) Styrene—To 10.0 g of Calcium Polystyrene Sul-
(potency), dissolve in 0.1 mol/L phosphate buŠer solution, fonate add 10 mL of acetone, shake for 30 minutes, cen-
pH 6.8 to make exactly 100 mL, and use this solution as the trifuge, and use the supernatant liquid as the sample solu-
standard stock solution. Keep the standard stock solution at tion. Separately, dissolve 10 mg of styrene in acetone to
59 C or below, and use within 30 days. Take exactly a suita- make exactly 100 mL. Pipet 1 mL of this solution, dilute
ble amount of the standard stock solution before use, add with acetone to make exactly 100 mL, and use this solution
0.1 mol/L phosphate buŠer solution, pH 6.8 to make solu- as the standard solution. Perform the test with exactly 5 mL
tions so that each mL contains 30 mg (potency) and 15 mg each of the sample solution and the standard solution as
(potency), and use these solutions as the high concentration directed under the Gas Chromatography according to the
standard solution and the low concentration standard solu- following conditions. Determine the peak heights, HT and
tion, respectively. HS, of styrene in each solution: HT is not larger than HS.
(7) Sample solutions—Weigh accurately an amount of Operating conditions—
Bleomycin Sulfate, equivalent to about 15 mg (potency), dis- Detector: A hydrogen ‰ame-ionization detector.
solve in 0.1 mol/L phosphate buŠer solution, pH 6.8 to Column: A stainless steel column 3 mm in inside diameter
1400 O‹cial Monographs for Part I Supplement I, JP XIV

and 2 m in length, having polyethylene glycol 20 M coated at allow to stand for 5 minutes: the turbidity of the solution
the ratio of 15z on siliceous earth for gas chromatography does not exceed that of the following control solution.
(150 to 180 mm in particle diameter). Control solution: Prepare in the same manner as de-
Column temperature: A constant temperature of about scribed above, using 0.20 mL of 0.01 mol/L hydrochloric
909C. acid VS.
Carrier gas: Nitrogen
Flow rate: Adjust the ‰ow rate so that the retention time
of styrene is about 9 minutes. Carbazochrome Sodium Sulfonate
System suitability—
System performance: Mix 10 mg of styrene with 1000 mL カルバゾクロムスルホン酸ナトリウム
of acetone. When the procedure is run with 5 mL of this solu-
tion under the above operating conditions, the number of Change the Purity (3) to read:
theoretical plates and the symmetry coe‹cient of the peak of
Purity
styrene are not less than 800 and 0.8 to 1.2, respectively.
(3) Related substances—Dissolve 50 mg of Carbazo-
System repeatability: When the test is repeated 6 times
chrome Sodium Sulfonate in 100 mL of water, and use this
with 5 mL of the standard solution under the above operat-
solution as the sample solution. Pipet 2 mL of the sample
ing conditions, the relative standard deviation of the peak
solution, add water to make exactly 200 mL, and use this
heights of styrene is not more than 5z.
solution as the standard solution. Perform the test with ex-
actly 10 mL each of the sample solution and the standard so-
lution as directed under the Liquid Chromatography accord-
d-Camphor ing to the following conditions. Determine each peak area of
these solutions by the automatic integration method: the
d-カンフル
total area of the peaks other than the peak of car-
bazochrome sulfonate from the sample solution is not larger
Change the Purity (2) to read:
than the peak area of carbazochrome sulfonate from the
Purity standard solution.
(2) Chlorinated compounds—Mix 0.20 g of ˆnely pow- Operating conditions—
dered d-Camphor with 0.4 g of sodium peroxide in a dried Detector: An ultraviolet absorption photometer (wave-
porcelain crucible. Heat the crucible gently by the open length: 360 nm).
‰ame until the incineration is complete. Dissolve the residue Column: A stainless steel column 4.6 mm in inside di-
in 20 mL of warm water, acidify with 12 mL of dilute nitric ameter and 25 cm in length, packed with octadecylsilanized
acid, and ˆlter the solution into a Nessler tube. Wash the silica gel for liquid chromatography (7 mm in particle di-
ˆlter paper with three 5-mL portions of hot water, adding ameter).
the washings to the ˆltrate. After cooling, add water to make Column temperature: A constant temperature of about
50 mL, then add 1 mL of silver nitrate TS, mix well, and 409C.
allow to stand for 5 minutes: the turbidity of the solution Mobile phase: Dissolve 1.2 g of ammonium dihydrogen
does not exceed that of the following control solution. phosphate in 1000 mL of water, and ˆlter through a mem-
Control solution: Prepare in the same manner as de- brane ˆlter (0.4 mm in pore size) if necessary. To 925 mL of
scribed above, using 0.20 mL of 0.01 mol/L hydrochloric this solution add 75 mL of ethanol (95), shake, and adjust
acid VS. the pH to 3 with phosphoric acid.
Flow rate: Adjust the ‰ow rate so that the retention time
of carbazochrome sulfonate is about 7 minutes.
dl-Camphor Time span of measurement: About 3 times as long as the
retention time of carbazochrome sulfonate after the solvent
dl-カンフル peak.
System suitability—
Change the Purity (2) to read: Test for required detection: To exactly 2 mL of the stan-
dard solution add the mobile phase to make exactly 20 mL.
Purity
Conˆrm that the peak area of carbazochrome sulfonate
(2) Chlorinated compounds—Mix 0.20 g of ˆnely pow-
obtained from 10 mL of this solution is equivalent to 7 to
dered dl-Camphor with 0.4 g of sodium peroxide in a dried
13z of that of carbazochrome sulfonate obtained from
porcelain crucible. Heat the crucible gently by the open
10 mL of the standard solution.
‰ame until the incineration is complete. Dissolve the residue
System performance: Dissolve 10 mg each of Car-
in 20 mL of warm water, acidify with 12 mL of dilute nitric
bazochrome Sodium Sulfonate and carbazochrome in
acid, and ˆlter the solution into a Nessler tube. Wash the
100 mL of water by warming. When the procedure is run
ˆlter paper with three 5-mL portions of hot water, adding
with 10 mL of this solution under the above operating condi-
the washings to the ˆltrate. After cooling, add water to make
tions, carbazochrome sulfonate and carbazochrome are elut-
50 mL, then add 1 mL of silver nitrate TS, mix well, and
Supplement I, JP XIV O‹cial Monographs for Part I 1401
AT
ed in this order with the resolution between these peaks Amount (mg) of C10H14N2O4.H2O = WS × × 1.080
AS
being not less than 3.
System repeatability: When the test is repeated 6 times WS: Amount (mg) of Carbidopa Reference Standard, cal-
with 10 mL of the standard solution under the above operat- culated on the dried basis
ing conditions, the relative standard deviation of the peak
Operating conditions—
area of carbazochrome sulfonate is not more than 2.0z.
Detector: An ultraviolet absorption photometer (wave-
length: 280 nm).
Column: A stainless steel column 4 mm in inside diameter
Carbidopa and 25 cm in length, packed with octadecylsilanized silica gel
for liquid chromatography (7 mm in particle diameter).
カルビドパ
Column temperature: A constant temperature of about
259C.
Change the Purity (2) to read:
Mobile phase: To 950 mL of 0.05 mol/L sodium dihydro-
Purity gen phosphate TS add 50 mL of ethanol (95), and adjust the
(2) Related substances—Dissolve 50 mg of Carbidopa in pH to 2.7 with phosphoric acid.
70 mL of the mobile phase, by warming and using ultrasoni- Flow rate: Adjust the ‰ow rate so that the retention time
cation, if necessary. After cooling, add the mobile phase to of carbidopa is about 6 minutes.
make 100 mL, and use this solution as the sample solution. System suitability—
Pipet 1 mL of the sample solution, add the mobile phase to System performance: Dissolve 50 mg each of Carbidopa
make exactly 100 mL, and use this solution as the standard and methyldopa in 100 mL of the mobile phase. When the
solution. Perform the test with exactly 20 mL each of the procedure is run with 20 mL of this solution under the above
sample solution and the standard solution as directed under operating conditions, methyldopa and carbidopa are eluted
the Liquid Chromatography according to the following con- in this order with the resolution between these peaks being
ditions. Determine each peak area from both solutions by not less than 0.9.
the automatic integration method: the total area of all peaks System repeatability: When the test is repeated 6 times
other than the peak of carbidopa from the sample solution is with 20 mL of the standard solution under the above operat-
not larger than the peak area of carbidopa from the standard ing conditions, the relative standard deviation of the peak
solution. area of carbidopa is not more than 1.0z.
Operating conditions—
Detector, column, column temperature, mobile phase,
and ‰ow rate: Proceed as directed in the operating condi- Carumonam Sodium
tions in the Assay.
Time span of measurement: About 3 times as long as the カルモナムナトリウム
retention time of carbidopa.
System suitability— Change to read except the structural formula
Test for required detection: To exactly 2 mL of the stan- and chemical name:
dard solution add the mobile phase to make exactly 20 mL.
Conˆrm that the peak area of carbidopa obtained from Carumonam Sodium contains not less than 850 mg
20 mL of this solution is equivalent to 7 to 13z of that of (potency) per mg, calculated on the anhydrous basis.
carbidopa obtained from 20 mL of the standard solution. The potency of Carumonam Sodium is expressed as
System performance, and system repeatability: Proceed as mass (potency) of carumonam (C12H14N6O10S2:
directed in the system suitability in the Assay. 466.40).
Description Carumonam Sodium occurs as a white to yel-
Change the Assay to read:
lowish white, crystals or crystalline powder.
Assay Weigh accurately about 50 mg each of Carbidopa It is freely soluble in water, soluble in formamide, very
and Carbidopa Reference Standard (determined separately slightly soluble in methanol, and practically insoluble in
the loss on drying), and dissolve each in 70 mL of the mobile acetic acid (100) and in ethanol (99.5).
phase, by warming and using ultrasonication if necessary.
Identiˆcation (1) Determine the absorption spectrum of
After cooling, add the mobile phase to make exactly 100
a solution of Carumonam Sodium (3 in 100,000) as directed
mL, and use these solutions as the sample solution and the
under the Ultraviolet-visible Spectrophotometry, and com-
standard solution, respectively. Perform the test with exactly
pare the spectrum with the Reference Spectrum or the spec-
20 mL each of the sample solution and the standard solution
trum of a solution of Carumonam Sodium Reference Stan-
as directed under the Liquid Chromatography according to
dard prepared in the same manner as the sample solution:
the following conditions, and determine the peak areas, AT
both spectra exhibit similar intensities of absorption at the
and AS, of carbidopa in each solution.
same wavelengths.
1402 O‹cial Monographs for Part I Supplement I, JP XIV

(2) Determine the infrared absorption spectrum of WS: Amount (g) of Carumonam Sodium Reference Stan-
Carumonam Sodium as directed in the potassium bromide dard
disk method under the Infrared Spectrophotometry, and WT: Amount (g) of the sample
compare the spectrum with the Reference Spectrum or the AS: Peak area of carumonam from the standard solution
spectrum of Carumonam Sodium Reference Standard: both AT: Each peak area other than carumonam from the sam-
spectra exhibit similar intensities of absorption at the same ple solution
wave numbers.
Operating conditions—
(3) Determine the spectrum of a solution of Carumonam
Detector, column, column temperature, mobile phase,
Sodium in heavy water for nuclear magnetic resonance spec-
and ‰ow rate: Proceed as directed in the operating condi-
troscopy (1 in 10) as directed under the Nuclear Magnetic
tions in the Assay.
Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl-
Time span of measurement: About 3 times as long as the
propionate-d4 for nuclear magnetic resonance spectroscopy
retention time of carumonam.
as an internal reference compound: it exhibits a double sig-
System suitability—
nal A at around d 5.5 ppm, and a single signal B at around
Test for required detectability: Measure exactly 5 mL of
d 7.0 ppm. The ratio of the integrated intensity of these sig-
the standard solution, and add the mobile phase to make
nals, A:B, is about 1:1.
exactly 50 mL. Conˆrm that the peak area of carumonam
(4) Carumonam Sodium responds to the Qualitative
obtained from 10 mL of this solution is equivalent to 7 to
Test (1) for sodium salt.
13z of that from 10 mL of the standard solution.
Optical rotation [a]20
D : +18.5 – +21.09(0.1 g calculated on System performance: Dissolve 40 mg of Carumonam So-
the anhydrous basis, water, 10 mL, 100 mm). dium in 20 mL of the mobile phase. To 5 mL of this solution
add 5 mL of a solution of resorcinol in the mobile phase (9 in
pH The pH of a solution obtained by dissolving 1.0 g of
1000) and the mobile phase to make 25 mL. When the proce-
Carumonam Sodium in 10 mL of water is between 5.0 and
dure is run with 10 mL of this solution under the above oper-
6.5.
ating conditions, resorcinol and carumonam are eluted in
Purity (1) Clarity and color of solution—Dissolve 0.5 g this order with the resolution between these peaks being not
of Carumonam Sodium in 5 mL of water: the solution is less than 2.5.
clear and colorless to pale yellow. System repeatability: When the test is repeated 3 times
(2) Heavy metals—Proceed with 2.0 g of Carumonam with 10 mL of the standard solution under the above operat-
Sodium according to Method 2, and perform the test. Pre- ing conditions, the relative standard deviation of the peak
pare the control solution with 3.0 mL of Standard Lead area of carumonam is not more than 2.9z.
Solution (not more than 15 ppm). (5) Related substance 2—Weigh accurately about 0.1 g
(3) Arsenic—Prepare the test solution with 2.0 g of of Carumonam Sodium, and dissolve in the mobile phase to
Carumonam Sodium according to Method 4, and perform make exactly 50 mL. Pipet 5 mL of this solution, add the
the test (not more than 1 ppm). mobile phase to make exactly 25 mL, and use this solution as
(4) Related substance 1—Weigh accurately about 0.1 g the sample solution. Separately, weigh accurately about
of Carumonam Sodium, and dissolve in the mobile phase to 0.1 g of Carumonam Sodium Reference Standard, and dis-
make exactly 50 mL. Pipet 5 mL of this solution, add the solve in the mobile phase to make exactly 50 mL. Pipet 5 mL
mobile phase to make exactly 25 mL, and use this solution as of this solution, and add the mobile phase to make exactly
the sample solution. Separately, weigh accurately about 25 mL. Pipet 1 mL of this solution, add the mobile phase to
0.1 g of Carumonam Sodium Reference Standard, and dis- make exactly 100 mL, and use this solution as the standard
solve in the mobile phase to make exactly 50 mL. Pipet 5 mL solution. Perform the test with exactly 10 mL each of the
of this solution, and add the mobile phase to make exactly sample solution and the standard solution as directed under
25 mL. Pipet 1 mL of this solution, add the mobile phase to the Liquid Chromatography according to the following con-
make exactly 100 mL, and use this solution as the standard ditions, and determine each peak area by the automatic in-
solution. Perform the test with exactly 10 mL each of the tegration method. Calculate the amount of the related sub-
sample solution and the standard solution as directed under stances by the following equation: the amount of each relat-
the Liquid Chromatography according to the following con- ed substance is not more than 1.0z.
ditions, and determine each peak area by the automatic in-
WS A
tegration method. Calculate the amount of the related sub- Amount (z) of related substance = × T
WT AS
stances by the following equation: the amount of the related
substance having the relative retention time of 0.7 to the WS: Amount (g) of Carumonam Sodium Reference Stan-
peak of carumonam is not more than 4.0z, and each dard
amount of the related substances other than the related sub- WT: Amount (g) of the sample
stance having the relative retention time of 0.7 to the peak of AS: Peak area of carumonam from the standard solution
carumonam is not more than 1.0z. AT: Each area of the peaks appeared after the peak of
carumonam from the sample solution
WS A
Amount (z) of related substance = × T
WT AS
Supplement I, JP XIV O‹cial Monographs for Part I 1403

Operating conditions— Internal standard solution—A solution of resorcinol in the


Detector, column, and column temperature: Proceed as mobile phase (9 in 1000).
directed in the operating conditions in the Assay. Operating conditions—
Mobile phase: A mixture of a solution of ammonium sul- Detector: An ultraviolet absorption photometer (wave-
fate (1 in 10,000), methanol and acetic acid (100) (74:25:1). length: 254 nm).
Flow rate: Dissolve 0.01 g of phthalic acid in the mobile Column: A stainless steel column 4 mm in inside diameter
phase to make 100 mL. Adjust the ‰ow rate so that the and 15 cm in length, packed with octadecylsilanized silica gel
retention time of phthalic acid is about 6.5 minutes when the for liquid chromatography (5 mm in particle diameter).
procedure is run with 10 mL of this solution. Column temperature: A constant temperature of about
Time span of measurement: About 10 times as long as the 259C.
retention time of carumonam. Mobile phase: A mixture of a solution of ammonium sul-
System suitability— fate (1 in 10,000), methanol and acetic acid (100) (97:2:1).
Test for required detectability: Measure exactly 5 mL of Flow rate: Adjust the ‰ow rate so that the retention time
the standard solution, and add the mobile phase to make of carumonam is about 10 minutes.
exactly 50 mL. Conˆrm that the peak area of carumonam System suitability—
obtained from 10 mL of this solution is equivalent to 7 to System performance: When the procedure is run with
13z of that from 10 mL of the standard solution. 10 mL of the standard solution under the above operating
System performance: Dissolve 40 mg of Carumonam So- conditions, the internal standard and carumonam are eluted
dium in 20 mL of the mobile phase. To 5 mL of this solution in this order with the resolution between these peaks being
add 5 mL of a solution of resorcinol in the mobile phase (9 in not less than 2.5.
1000) and the mobile phase to make 25 mL. When the proce- System repeatability: When the test is repeated 6 times
dure is run with 10 mL of this solution under the above oper- with 10 mL of the standard solution under the above operat-
ating conditions, resorcinol and carumonam are eluted in ing conditions, the relative standard deviation of the ratios
this order with the resolution between these peaks being not of the peak area of carumonam to that of the internal stan-
less than 7. dard is not more than 1.0z.
System repeatability: When the test is repeated 3 times
Containers and storage Containers—Hermetic containers.
with 10 mL of the standard solution under the above operat-
Storage—Light-resistant.
ing conditions, the relative standard deviation of the peak
area of carumonam is not more than 2.9z.
(6) Total amount of related substances-The total of the
amounts of the related substances obtained in the Related Cefaclor
substance 1 and the Related substance 2 is not more than
セファクロル
6.0z.

Water Not more than 2.0z (0.2 g, volumetric titration, Change to read except the structural formula
direct titration; Use a mixture of formamide for water deter- and chemical name:
mination and methanol for water determination (3:1) in-
stead of methanol for water determination). Cefaclor contains not less than 950 mg (potency) per
mg, calculated on the anhydrous basis. The potency of
Assay Weigh accurately an amount of Carumonam
Cefaclor is expressed as mass (potency) of cefaclor
Sodium and Carumonam Sodium Reference Standard,
(C15H14ClN3O4S).
equivalent to about 40 mg (potency), and dissolve each in the
mobile phase to make exactly 20 mL. Measure exactly 5 mL Description Cefaclor occurs as a white to yellowish white
each of these solutions, add exactly 5 mL of the internal crystalline powder.
standard solution and the mobile phase to make 25 mL, and It is slightly soluble in water and in methanol, and practi-
use these solutions as the sample solution and the standard cally insoluble in ethanol (99.5) and in N, N-dimethylfor-
solution. Perform the test with 10 mL each of the sample mamide.
solution and the standard solution as directed under the Liq-
Identiˆcation (1) Determine the absorption spectrum of
uid Chromatography according to the following conditions,
a solution of Cefaclor (1 in 50,000) as directed under the
and determine the ratios, QT and QS, of the peak area of
Ultraviolet-visible Spectrophotometry, and compare the
carumonam to that of the internal standard.
spectrum with the Reference Spectrum: both spectra exhibit
Amount [mg (potency)] of carumonam (C12H14N6O10S2) similar intensities of absorption at the same wavelengths.
Q (2) Determine the infrared absorption spectrum of
= WS × T × 1000
QS Cefaclor as directed in the potassium bromide disk method
under the Infrared Spectrophotometry, and compare the
WS: Amount [mg (potency)] of Carumonam Sodium
spectrum with the Reference Spectrum: both spectra exhibit
Reference Standard
similar intensities of absorption at the same wave numbers.
1404 O‹cial Monographs for Part I Supplement I, JP XIV

(3) Dissolve 0.04 g of Cefaclor in 0.5 mL of heavy water


for nuclear magnetic resonance spectroscopy and 1 drop of Time after injection Mobile phase Mobile phase
of sample (min) A (z ) B (z )
deuterated hydrochloric acid for nuclear magnetic resonance
spectroscopy, and determine the spectrum of this solution as 0 – 30 95 → 75 5 → 25
directed under the Nuclear Magnetic Resonance Spec- 30 – 45 75 → 0 25 → 100
troscopy (1H), using sodium 3-trimethylsilylpropanesul- 45 – 55 0 100
fonate for nuclear magnetic resonance spectroscopy as an
Flow rate: 1.0 mL per minute.
internal reference compound: it exhibits an AB type quartet
Time span of measurement: About 2.5 times as long as the
signal A at around d 3.7 ppm, and a single signal or a sharp
retention time of cefaclor after the solvent peak.
multiple signal B at around d 7.6 ppm. The ratio of the
System suitability—
integrated intensity of each signal, A:B, is about 2:5.
Test for required detectability: Measure exactly 1 mL of
(4) Perform the test with Cefaclor as directed under the
the standard solution, and add sodium dihydrogen phos-
Flame Coloration Test (2): a green color appears.
phate TS, pH 2.5 to make exactly 20 mL. Conˆrm that the
Optical rotation [a]20
D : +105 – +1209(0.1 g calculated on peak area of cefaclor obtained from 20 mL of this solution is
the anhydrous basis, water, 25 mL, 100 mm). equivalent to 4 to 6z of that from 20 mL of the standard
solution.
Purity (1) Heavy metals—Proceed with 1.0 g of Cefaclor
System performance: When the procedure is run with
according to Method 2, and perform the test. Prepare the
20 mL of the standard solution under the above operating
control solution with 2.0 mL of Standard Lead Solution (not
conditions, the number of theoretical plates and the sym-
more than 20 ppm).
metry coe‹cient of the peak of cefaclor are not less than
(2) Arsenic—Prepare the test solution by suspending
40,000 steps and 0.8 to 1.3, respectively.
1.0 g of Cefaclor in 10 mL of N, N-dimethylformamide, and
System repeatability: When the test is repeated 3 times
perform the test (not more than 2 ppm).
with 20 mL of the standard solution under the above operat-
(3) Related substances—Dissolve 0.05 g of Cefaclor in
ing conditions, the relative standard deviations of the peak
10 mL of sodium dihydrogen phosphate TS, pH 2.5, and use
areas and the retention times of cefaclor are not more than
this solution as the sample solution. Pipet 1 mL of the
2.0z, respectively.
sample solution, add sodium dihydrogen phosphate TS, pH
2.5 to make exactly 100 mL, and use this solution as the Water Not more than 6.5z (0.2 g, volumetric titration,
standard solution. Perform the test with exactly 20 mL each back titration).
of the sample solution and the standard solution as directed
Assay Weigh accurately an amount of Cefaclor and
under the Liquid Chromatography according to the follow-
Cefaclor Reference Standard, equivalent to about 0.1 g
ing conditions, and determine each peak area by the auto-
(potency), and dissolve each in 0.1 mol/L phosphate buŠer
matic integration method: the peak areas other than cefaclor
solution, pH 4.5 to make exactly 100 mL. Pipet 10 mL each
from the sample solution are not more than 1/2 of the peak
of these solutions, add exactly 10 mL of the internal stan-
area of cefaclor from the standard solution, and the total of
dard solution, add 0.1 mol/L phosphate buŠer solution, pH
the peak areas other than cefaclor from the sample solution
4.5 to make 50 mL, and use these solutions as the sample
is not more than 2 times of the peak area of cefaclor from
solution and the standard solution. Perform the test with
the standard solution. If necessary, proceed with 20 mL of
10 mL each of the sample solution and the standard solution
sodium dihydrogen phosphate TS, pH 2.5 in the same man-
as directed under the Liquid Chromatography according to
ner as above to compensate the base line.
the following conditions, and determine the ratios, QT and
Operating conditions—
QS, of the peak area of cefaclor to that of the internal stan-
Detector: An ultraviolet absorption photometer (wave-
dard.
length: 220 nm).
Column: A stainless steel column 4.6 mm in inside di- Amount [mg (potency)] of C15H14ClN3O4S
ameter and 25 cm in length, packed with octadecylsilanized Q
= WS × T × 1000
silica gel for liquid chromatography (5 mm in particle di- QS
ameter).
WS: Amount [mg (potency)] of Cefaclor Reference
Column temperature: A constant temperature of about
Standard
259C.
Mobile phase A: Dissolve 7.8 g of sodium dihydrogen Internal standard solution—A solution of 4-aminoacetophe-
phosphate dihydrate in 1000 mL of water, and adjust the pH none in 0.1 mol/L phosphate buŠer solution, pH 4.5 (1 in
to 4.0 with phosphoric acid. 700).
Mobile phase B: To 550 mL of the mobile phase A add Operating conditions—
450 mL of acetonitrile for liquid chromatography. Detector: An ultraviolet absorption photometer (wave-
Flowing of the mobile phase: Control the gradient by length: 254 nm).
mixing the mobile phases A and B as directed in the follow- Column: A stainless steel column 4.6 mm in inside di-
ing table. ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Supplement I, JP XIV O‹cial Monographs for Part I 1405

ameter). Spectroscopy (1H), using sodium 3-trimethylsilylpropanesul-


Column temperature: A constant temperature of about fonate for nuclear magnetic resonance spectroscopy as an
259C. internal reference compound: it exhibits a single signal A at
Mobile phase: Dissolve 6.8 g of potassium dihydrogen around d 3.8 ppm, and two multiple signals, B and C,
phosphate in 1000 mL of water, and adjust the pH to 3.4 between d 6.9 ppm and d7.5 ppm and between d 7.9 ppm and
with diluted phosphoric acid (3 in 500). To 940 mL of this d 9.0 ppm, respectively. The ratio of the integrated intensity
solution add 60 mL of acetonitrile. of each signal, A:B:C, is about 2:3:5.
Flow rate: Adjust the ‰ow rate so that the retention time
Optical rotation [a]20
D : +46 – +519(0.5 g calculated on the
of cefaclor is about 7 minutes.
anhydrous basis, water, 50 mL, 100 mm).
System suitability—
System performance: When the procedure is run with pH Dissolve 1.0 g of Cefaloridin in 10 mL of water: the
10 mL of the standard solution under the above operating pH of the solution is between 4.0 and 6.0.
conditions, cefaclor and the internal standard are eluted in
Purity (1) Clarity and color of solution-Dissolve 1.0 g
this order with the resolution between these peaks being not
of Cefaloridin in 5 mL of water by warming: the solution is
less than 5.
clear and light yellow.
System repeatability: When the test is repeated 6 times
(2) Heavy metals—Proceed with 1.0 g of Cefaloridin
with 10 mL of the standard solution under the above operat-
according to Method 2, and perform the test. Prepare the
ing conditions, the relative standard deviation of the ratios
control solution with 2.0 mL of Standard Lead Solution (not
of the peak area of cefaclor to that of the internal standard is
more than 20 ppm).
not more than 1.0z.
(3) Arsenic—Prepare the test solution with 1.0 g of
Containers and storage Containers—Tight containers. Cefaloridin according to Method 3, and perform the test
Storage—Light-resistant. (not more than 2 ppm).
(4) Related substances—Dissolve 20 mg of Cefaloridin
in 20 mL of a solution of sodium dihydrogen phosphate di-
Cefaloridin hydrate (39 in 1250). To 2 mL of this solution add a solution
of sodium dihydrogen phosphate dihydrate (39 in 1250) to
セファロリジン make 20 mL, and use this solution as the sample solution.
Perform the test with 20 mL of the sample solution as direct-
Change to read except the structural formula ed under the Liquid Chromatography according to the fol-
and chemical name: lowing conditions, determine each peak area by the auto-
matic integration method, and calculate the amount of each
Cefaloridin contains not less than 940 mg (potency) peak by the area percentage method: the amount of each
per mg, calculated on the anhydrous basis. The poten- peak other than cefaloridin is not more than 0.5z, and the
cy of Cefaloridin is expressed as mass (potency) of total amount of the peaks other than cefaloridin is not more
cefaloridin (C19H17N3O4S2). than 2.0z.
Operating conditions—
Description Cefaloridin occurs as white to light yellowish
Detector: An ultraviolet absorption photometer (wave-
white, crystals or crystalline powder.
length: 254 nm).
It is soluble in water, slightly soluble in methanol, and
Column: A stainless steel column 4 mm in inside diameter
very slightly soluble in ethanol (95).
and 15 cm in length, packed with octadecylsilanized silica gel
Identiˆcation (1) Determine the absorption spectrum of for liquid chromatography (5 mm in particle diameter).
a solution of Cefaloridin (1 in 50,000) as directed under the Column temperature: A constant temperature of about
Ultraviolet-visible Spectrophotometry, and compare the 259C.
spectrum with the Reference Spectrum or the spectrum of a Mobile phase: To 25.9 g of sodium acetate trihydrate add
solution of Cefaloridin Reference Standard prepared in the 0.6 mL of acetic acid (100) and water to make 1000 mL. To
same manner as the sample solution: both spectra exhibit 900 mL of this solution add 50 mL of acetonitrile and 50 mL
similar intensities of absorption at the same wavelengths. of methanol.
(2) Determine the infrared absorption spectrum of Flow rate: Adjust the ‰ow rate so that the retention time
Cefaloridin as directed in the potassium bromide disk of cefaloridin is about 14 minutes.
method under the Infrared Spectrophotometry, and com- Time span of measurement: About 2 times as long as the
pare the spectrum with the Reference Spectrum or the spec- retention time of cefaloridin after the solvent peak.
trum of Cefaloridin Reference Standard: both spectra System suitability—
exhibit similar intensities of absorption at the same wave Test for required detectability: Measure exactly 1 mL of
numbers. the sample solution, and add a solution of sodium dihydro-
(3) Determine the spectrum of a solution of Cefaloridin gen phosphate dihydrate (39 in 1250) to make exactly
in heavy water for nuclear magnetic resonance spectroscopy 100 mL. To exactly 1 mL of this solution add a solution of
(1 in 10) as directed under the Nuclear Magnetic Resonance sodium dihydrogen phosphate dihydrate (39 in 1250) to
1406 O‹cial Monographs for Part I Supplement I, JP XIV

make exactly 10 mL. Conˆrm that the peak area of cefalo- of the peak area of cefaloridin to that of the internal stan-
ridin obtained from 20 mL of this solution is equivalent to dard is not more than 1.0z.
0.07 to 0.13z of that from 20 mL of the sample solution.
Containers and storage Containers—Tight containers.
System performance: When the procedure is run with
Storage—Light-resistant.
20 mL of the sample solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
coe‹cient of the peak of cefaloridin are not less than 5000
steps and not more than 1.3, respectively. Cefalotin Sodium
System repeatability: When the test is repeated 6 times
セファロチンナトリウム
with 20 mL of the sample solution under the above operating
conditions, the relative standard deviation of the peak area
Change to read except the structural formula
of cefaloridin is not more than 3.0z.
and chemical name:
Water Not more than 4.0z (0.2 g, volumetric titration,
direct titration). Cefalotin Sodium contains not less than 910 mg
(potency) per mg, calculated on the anhydrous basis.
Assay Weigh accurately an amount of Cefaloridin and
The potency of Cefalotin Sodium is expressed as mass
Cefaloridin Reference Standard, equivalent to about 45 mg
(potency) of cefalotin (C16H16N2O6S2: 396.44).
(potency), and dissolve each in water to make exactly 50 mL.
Measure exactly 20 mL each of these solutions, add exactly Description Cefalotin Sodium occurs as white to light yel-
5 mL of the internal standard solution and water to make lowish white, crystals or crystalline powder.
100 mL, and use these solutions as the sample solution and It is freely soluble in water, slightly soluble in methanol,
the standard solution. Perform the test with 5 mL each of the very slightly soluble in ethanol (95), and practically insoluble
sample solution and the standard solution as directed under in acetonitrile.
the Liquid Chromatography according to the following
Identiˆcation (1) Determine the absorption spectrum of
conditions, and determine the ratios, QT and QS, of the peak
a solution of Cefalotin Sodium (1 in 50,000) as directed un-
area of cefaloridin to that of the internal standard.
der the Ultraviolet-visible Spectrophotometry, and compare
Amount [mg (potency)] of C19H17N3O4S2 the spectrum with the Reference Spectrum or the spectrum
Q of a solution of Cefalotin Sodium Reference Standard pre-
= WS × T × 1000
QS pared in the same manner as the sample solution: both spec-
tra exhibit similar intensities of absorption at the same wave-
WS: Amount [mg (potency)] of Cefaloridin Reference
lengths.
Standard
(2) Determine the infrared absorption spectrum of
Internal standard solution—A solution of acetanilide in Cefalotin Sodium as directed in the potassium bromide disk
acetonitrile (9 in 5000). method under the Infrared Spectrophotometry, and com-
Operating conditions— pare the spectrum with the Reference Spectrum or the spec-
Detector: An ultraviolet absorption photometer (wave- trum of Cefalotin Sodium Reference Standard: both spectra
length: 254 nm). exhibit similar intensities of absorption at the same wave
Column: A stainless steel column 4 mm in inside diameter numbers.
and 20 cm in length, packed with octadecylsilanized silica gel (3) Determine the spectrum of a solution of Cefalotin
for liquid chromatography (5 mm in particle diameter). Sodium in heavy water for nuclear magnetic resonance spec-
Column temperature: A constant temperature of about troscopy (1 in 10) as directed under the Nuclear Magnetic
259C. Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl-
Mobile phase: Dissolve 25.9 g of sodium acetate trihy- propanesulfonate for nuclear magnetic resonance spec-
drate in 0.6 mL of acetic acid (100) and water to make troscopy as an internal reference compound: it exhibits a sin-
1000 mL. To 800 mL of this solution add 200 mL of acetoni- gle signal A at around d 2.1 ppm, a single or sharp multiple
trile. signal B at around d 3.9 ppm, and a multiple signal C at
Flow rate: Adjust the ‰ow rate so that the retention time around d 7.0 ppm. The ratio of the integrated intensity of
of cefaloridin is about 5 minutes. these signals, A:B:C, is about 3:2:2.
System suitability— (4) Cefalotin Sodium responds to the Qualitative Test
System performance: When the procedure is run with 5 mL (1) for sodium salt.
of the standard solution under the above operating condi-
Optical rotation [a]25
D : +124 – +1349(5 g, water, 100 mL,
tions, cefaloridin and the internal standard are eluted in this
100 mm).
order with the resolution between these peaks being not less
than 1.5. pH The pH of a solution obtained by dissolving 1.0 g of
System repeatability: When the test is repeated 6 times Cefalotin Sodium in 10 mL of water is between 4.5 and 7.0.
with 5 mL of the standard solution under the above operat-
Purity (1) Clarity and color of solution—Dissolve 1.0 g
ing conditions, the relative standard deviation of the ratios
of Cefalotin Sodium in 5 mL of water: the solution is clear
Supplement I, JP XIV O‹cial Monographs for Part I 1407

and light yellow. Amount [mg (potency)] of cefalotin (C16H16N2O6S2)


(2) Heavy metals—Proceed with 1.0 g of Cefalotin A
= WS × T × 1000
Sodium according to Method 2, and perform the test. Pre- AS
pare the control solution with 2.0 mL of Standard Lead
WS: Amount [mg (potency)] of Cefalotin Sodium Refer-
Solution (not more than 20 ppm).
ence Standard
(3) Arsenic—Prepare the test solution with 1.0 g of
Cefalotin Sodium according to Method 3, and perform the Operating conditions—
test (not more than 2 ppm). Detector: An ultraviolet absorption photometer (wave-
(4) Related substances—Pipet 1 mL of the standard length: 254 nm).
solution obtained in the Assay, add the mobile phase to Column: A stainless steel column 4.6 mm in inside di-
make exactly 100 mL, and use this solution as the standard ameter and 25 cm in length, packed with octadecylsilanized
solution. Perform the test with exactly 10 mL each of the silica gel for liquid chromatography (5 mm in particle di-
sample solution obtained in the Assay and the standard solu- ameter).
tion prepared here as directed under the Liquid Chro- Column temperature: A constant temperature of about
matography according to the following conditions, and de- 409C.
termine each peak area by the automatic integration Mobile phase: Dissolve 17 g of sodium acetate trihydrate
method: the peak area other than cefalotin from the sample in 790 mL of water, and add 0.6 mL of acetic acid (100). If
solution is not more than the peak area of cefalotin from the necessary adjust the pH to 5.9 ± 0.1 with 0.1 mol/L sodium
standard solution, and the total area of the peaks other than hydrochloride TS or acetic acid (100). To this solution add
cefalotin from the sample solution is not more than 3 times 150 mL of acetonitrile and 70 mL of ethanol (95).
the peak area of cefalotin from the standard solution. Flow rate: Adjust the ‰ow rate so that the retention time
Operating conditions— of cefalotin is about 12 minutes.
Detector, column, column temperature, mobile phase, System suitability—
and ‰ow rate: Proceed as directed in the operating condi- System performance: Heat the standard solution in a
tions in the Assay. water bath of 909 C for 10 minutes, and cool. Measure
Time span of measurement: About 4 times as long as the exactly 2.5 mL of this solution, and add the mobile phase to
retention time of cefalotin. make exactly 100 mL. When the procedure is run with 10 mL
System suitability— of this solution under the above operating conditions, the
Test for required detectability: Measure exactly 1 mL of resolution between the peak of cefalotin and the peak, hav-
the standard solution, and add the mobile phase to make ing the relative retention time of about 0.5 with respect to
exactly 10 mL. Conˆrm that the peak area of cefalotin cefalotin is not less than 9, and the symmetry coe‹cient of
obtained from 10 mL of this solution is equivalent to 7 to the peak of cefalotin is not more than 1.8.
13z of that from 10 mL of the standard solution. System repeatability: When the test is repeated 6 times
System performance: Heat the standard solution in a with 10 mL of the standard solution under the above operat-
water bath of 909 C for 10 minutes, and cool. Measure ing conditions, the relative standard deviation of the peak
exactly 2.5 mL of this solution, and add the mobile phase to area of cefalotin is not more than 1.0z.
make exactly 100 mL. When the procedure is run with 10 mL
Containers and storage Containers—Tight containers.
of this solution under the above operating conditions, the
resolution between the peak of cefalotin and the peak, hav-
ing the relative retention time of about 0.5 with respect to
cefalotin, is not less than 9, and the symmetry coe‹cient of Cefamandole Sodium
the peak of cefalotin is not more than 1.8.
セファマンドールナトリウム
System repeatability: When the test is repeated 3 times
with 10 mL of the standard solution under the above operat-
Change to read except the structural formula
ing conditions, the relative standard deviation of the peak
and chemical name:
area of cefalotin is not more than 2.0z.

Water Not more than 1.0z (0.5 g, volumetric titration, Cefamandole Sodium contains not less than 858 mg
back titration). (potency) per mg, calculated on the anhydrous basis.
The potency of Cefamandole Sodium is expressed as
Assay Weigh accurately an amount of Cefalotin Sodium
mass (potency) of cefamandole (C18H18N6O5S2:
and Cefalotin Sodium Reference Standard, equivalent to
462.50).
about 25 mg (potency), and dissolve each in the mobile
phase to make exactly 25 mL, and use these solutions as the Description Cefamandole Sodium occurs as a white to
sample solution and the standard solution. Perform the test light yellowish white crystalline powder.
with exactly 10 mL each of the sample solution and the stan- It is very soluble in water, freely soluble in N, N-dimethyl-
dard solution as directed under the Liquid Chromatography formamide, sparingly soluble in methanol, slightly soluble in
according to the following conditions, and determine the ethanol (99.5), and practically insoluble in acetonitrile.
peak areas, AT and AS, of cefalotin. It is hygroscopic.
1408 O‹cial Monographs for Part I Supplement I, JP XIV

Identiˆcation (1) Determine the absorption spectrum of Operating conditions—


a solution of Cefamandole Sodium (1 in 50,000) as directed Detector: An ultraviolet absorption photometer (wave-
under the Ultraviolet-visible Spectrophotometry, and com- length: 254 nm).
pare the spectrum with the Reference Spectrum: both spec- Column: A stainless steel column 4 mm in inside diameter
tra exhibit similar intensities of absorption at the same wave- and 15 cm in length, packed with octadecylsilanized silica gel
lengths. for liquid chromatography (10 mm in particle diameter).
(2) Determine the infrared absorption spectrum of Column temperature: A constant temperature of about
Cefamandole Sodium as directed in the potassium bromide 259C.
disk method under the Infrared Spectrophotometry, and Mobile phase: Dissolve 6.94 g of potassium dihydrogen
compare the spectrum with the Reference Spectrum: both phosphate, 3.22 g of disodium hydrogen phosphate 12-water
spectra exhibit similar intensities of absorption at the same and 1.60 g of tetra-n-butylammonium bromide in water to
wave numbers. make 1000 mL. To 600 mL of this solution add 200 mL of
(3) Determine the spectrum of a solution of Cefaman- methanol.
dole Sodium in heavy water for nuclear magnetic resonance Flow rate: Adjust the ‰ow rate so that the retention time
spectroscopy (1 in 10) as directed under the Nuclear Magnet- of 1-methyl-1H-tetrazole-5-thiol is about 4 minutes.
ic Resonance Spectroscopy (1H), using sodium 3-trimethyl- System suitability—
silylpropanesulfonate for nuclear magnetic resonance spec- System performance: When the procedure is run with 5 mL
troscopy as an internal reference compound: it exhibits two of the standard solution under the above operating condi-
single signals, A and B, at around d 4.0 ppm and at around tions, 1-methyl-1H-tetrazole-5-thiol and the internal stan-
d 5.3 ppm, and a multiple signal C at around d 7.5 ppm. The dard are eluted in this order with the resolution between
ratio of the integrated intensity of the two signals, B:C, is these peaks being not less than 14.
about 1:5. System repeatability: When the test is repeated 3 times
(4) Cefamandole Sodium responds to the Qualitative with 5 mL of the standard solution under the above operat-
Test (1) for sodium salt. ing conditions, the relative standard deviation of the ratios
of the peak area of 1-methyl-1H-tetrazole-5-thiol to that of
Optical rotation [a]20
D : -26.0 – -34.09(1 g calculated on
the internal standard is not more than 3.0z.
the anhydrous basis, water, 10 mL, 100 mm).
Water Not more than 2.0z (1 g, volumetric titration, back
pH The pH of a solution obtained by dissolving 1.0 g of
titration).
Cefamandole Sodium in 10 mL of water is between 4.3 and
6.3. Assay Weigh accurately an amount of Cefamandole
Sodium and Cefamandole Lithium Reference Standard,
Purity (1) Clarity and color of solution—Dissolve 0.5 g
equivalent to about 50 mg (potency), and dissolve each in the
of Cefamandole Sodium in 2 mL of water: the solution is
mobile phase to make exactly 100 mL. Measure exactly 2 mL
clear and pale yellow to light yellow.
each of these solutions, add exactly 4 mL of the internal
(2) Heavy metals—Proceed with 1.0 g of Cefamandole
standard solution and the mobile phase to make 20 mL, and
Sodium according to Method 4, and perform the test. Pre-
use these solutions as the sample solution and the standard
pare the control solution with 2.0 mL of Standard Lead
solution. Perform the test with 20 mL each of the sample
Solution (not more than 20 ppm).
solution and the standard solution as directed under the Liq-
(3) Arsenic—Prepare the test solution with 1.0 g of
uid Chromatography according to the following conditions,
Cefamandole Sodium according to Method 5, and perform
and determine the ratios, QT and QS, of the peak area of
the test (not more than 2 ppm).
cefamandole to that of the internal standard.
(4) 1-Methyl-1H-tetrazole-5-thiol—Dissolve 30 mg of
Cefamandole Sodium in exactly 2 mL of the internal stan- Amount [mg (potency)] of cefamandole (C18H18N6O5S2)
dard solution, add water to make 20 mL, and use this solu- Q
= WS × T × 1000
tion as the sample solution. Separately, dissolve 9 mg of QS
sodium 1-methyl-1H-tetrazole-5-thiolate in water to make
WS: Amount [mg (potency)] of Cefamandole Lithium
exactly 100 mL. Pipet 2 mL of this solution, add exactly
Reference Standard
2 mL of the internal standard solution and water to make
20 mL, and use this solution as the standard solution. Per- Internal standard solution—A solution of methyl para-
form the test with 5 mL each of the sample solution and the hydroxybenzoate in the mobile phase (1 in 12,500).
standard solution as directed under the Liquid Chro- Operating conditions—
matography according to the following conditions, and de- Detector: An ultraviolet absorption photometer (wave-
termine the ratios, QT and QS, of the peak area of 1-methyl- length: 268 nm).
1H-tetrazole-5-thiol to that of the internal standard: QT is Column: A stainless steel column 4 mm in inside diameter
not larger than QS. and 15 cm in length, packed with divinylbenzene-methacry-
Internal standard solution—A solution of m-cresol (7 in late co-polymer for liquid chromatography (5 mm in particle
10,000). diameter).
Column temperature: A constant temperature of about
Supplement I, JP XIV O‹cial Monographs for Part I 1409

259C. intensity of each signal, A:B:C, is about 3:3:1.


Mobile phase: Dissolve 15.0 g of disodium hydrogen (3) Cefbuperazone Sodium responds to the Qualitative
phosphate 12-water and 1.01 g of potassium dihydrogen Test (1) for sodium salt.
phosphate in water to make 1000 mL, and add 1.6 g of tetra-
Optical rotation [a]20
D : +48 – +569(0.4 g calculated on the
n-butylammonium bromide. To 810 mL of this solution add
anhydrous basis, water, 20 mL, 100 mm).
290 mL of acetonitrile.
Flow rate: Adjust the ‰ow rate so that the retention time pH Dissolve 1.0 g of Cefbuperazone Sodium in 4 mL of
of cefamandole is about 7 minutes. water: the pH of the solution is between 4.0 and 6.0.
System suitability—
Purity (1) Clarity and color of solution—Dissolve 1.0 g
System performance: When the procedure is run with
of Cefbuperazone Sodium in 4 mL of water: the solution is
20 mL of the standard solution under the above operating
clear and light yellow.
conditions, cefamandole and the internal standard are eluted
(2) Heavy metals—Proceed with 2.0 g of Cefbuperazone
in this order with the resolution between these peaks being
Sodium according to Method 4, and perform the test. Pre-
not less than 3.
pare the control solution with 2.0 mL of Standard Lead
System repeatability: When the test is repeated 3 times
Solution (not more than 10 ppm).
with 20 mL of the standard solution under the above operat-
(3) Arsenic—Prepare the test solution with 1.0 g of Cef-
ing conditions, the relative standard deviation of the ratios
buperazone Sodium according to Method 4, and perform
of the peak area of cefamandole to that of the internal stan-
the test (not more than 2 ppm).
dard is not more than 1.0z.
(4) Related substances—Dissolve 0.10 g of Cefbupera-
Containers and storage Containers—Tight containers. zone Sodium in 100 mL of the mobile phase, and use this
Storage—Light-resistant. solution as the sample solution. Pipet 1 mL of the sample
solution, add the mobile phase to make exactly 50 mL, and
use this solution as the standard solution. Perform the test
Cefbuperazone Sodium with exactly 25 mL each of the sample solution and the stan-
dard solution as directed under the Liquid Chromatography
セフブペラゾンナトリウム according to the following conditions, and determine each
peak area by the automatic integration method. Calculate
Change to read except the structural formula the percentages of each peak area of related substances from
and chemical name: the sample solution against 50 times of the peak area of
cefbuperazone from the standard solution; the amount of
Cefbuperazone Sodium contains not less than related substance I having the relative retention time of
870 mg (potency) per mg, calculated on the anhydrous about 0.2 to cefbuperazone is not more than 2.0z, the
basis. The potency of Cefbuperazone Sodium is amount of related substance II having the relative retention
expressed as mass (potency) of cefbuperazone time of about 0.6 to cefbuperazone is not more than 4.5z
(C22H29N9O9S2: 627.65). and the amount of related substance III having the relative
retention time of about 1.6 to cefbuperazone is not more
Description Cefbuperazone Sodium occurs as white to
than 1.0z, and the total amount of these related substances
light yellowish white, powder or masses.
is not more than 6.0z. For these calculations, use the values
It is very soluble in water, freely soluble in methanol and
of the peak areas of the related substances I and III obtained
in pyridine, sparingly soluble in ethanol (95), and very slight-
by the automatic integration method after multiplying by
ly soluble in acetonitrile.
each sensitivity coe‹cient, 0.72 and 0.69, respectively.
Identiˆcation (1) Determine the absorption spectrum of Operating conditions—
a solution of Cefbuperazone Sodium (1 in 50,000) as direct- Detector, column, column temperature, mobile phase,
ed under the Ultraviolet-visible Spectrophotometry, and and ‰ow rate: Proceed as directed in the operating condi-
compare the spectrum with the Reference Spectrum: both tions in the Assay.
spectra exhibit similar intensities of absorption at the same Time span of measurement: About 2 times as long as the
wavelengths. retention time of cefbuperazone.
(2) Dissolve 0.1 g of Cefbuperazone Sodium in 0.5 mL System suitability—
of deuterated pyridine for nuclear magnetic resonance spec- Test for required detectability: Measure exactly 1 mL of
troscopy and 1 drop of heavy water for nuclear magnetic the standard solution, and add the mobile phase to make
resonance spectroscopy, and determine the spectrum of this exactly 10 mL. Conˆrm that the peak area of cefbuperazone
solution as directed under the Nuclear Magnetic Resonance obtained from 25 mL of this solution is equivalent to 7 to
Spectroscopy (1H), using tetramethylsilane for nuclear mag- 13z of that from 25 mL of the standard solution.
netic resonance spectroscopy as an internal reference com- System performance: When the procedure is run with
pound: it exhibits a triplet signal A at around d1.1 ppm, and 25 mL of the standard solution under the above operating
two doublet signals, B and C, at around d 1.6 ppm and at conditions, the number of theoretical plates and the symmet-
around d 5.1 ppm, respectively. The ratio of the integrated ry coe‹cient of the peak of cefbuperazone are not less than
1410 O‹cial Monographs for Part I Supplement I, JP XIV

5000 steps and not more than 1.5, respectively. Cefmenoxime Hydrochloride
System repeatability: When the test is repeated 6 times
with 25 mL of the standard solution under the above operat- 塩酸セフメノキシム
ing conditions, the relative standard deviation of the peak
area of cefbuperazone is not more than 2.0z. Change to read except the structural formula
and chemical name:
Water Not more than 1.0z (3 g, volumetric titration,
direct titration).
Cefmenoxime Hydrochloride contains not less than
Assay Weigh accurately an amount of Cefbuperazone 890 mg (potency) per mg, calculated on the dehydrated
Sodium and Cefbuperazone Reference Standard, equivalent basis. The potency of Cefmenoxime Hydrochloride
to about 0.1 g (potency), and dissolve each in the mobile is expressed as mass (potency) of cefmenoxime
phase to make exactly 100 mL. Measure exactly 10 mL each (C16H17N9O5S3: 511.57).
of these solutions, add exactly 10 mL of the internal stan-
Description Cefmenoxime Hydrochloride occurs as white
dard solution and the mobile phase to make 50 mL, and use
to light orange-yellow crystals or crystalline powder.
these solutions as the sample solution and the standard solu-
It is freely soluble in formamide and in dimethylsulfoxide,
tion. Perform the test with 10 mL each of the sample solution
slightly soluble in methanol, very slightly soluble in water,
and the standard solution as directed under the Liquid
and practically insoluble in ethanol (95).
Chromatography according to the following conditions, and
determine the ratios, QT and QS, of the peak area of cef- Identiˆcation (1) Determine the absorption spectrum of
buperazone to that of the internal standard. a solution of Cefmenoxime Hydrochloride in 0.1 mol/L
phosphate buŠer solution, pH 6.8 (3 in 200,000) as directed
Amount [mg (potency)] of cefbuperazone (C22H29N9O9S2)
under the Ultraviolet-visible Spectrophotometry, and com-
Q
= WS × T × 1000 pare the spectrum with the Reference Spectrum or the spec-
QS
trum of a solution of Cefmenoxime Hydrochloride Refer-
WS: Amount [mg (potency)] of Cefbuperazone Reference ence Standard prepared in the same manner as the sample
Standard solution: both spectra exhibit similar intensities of absorp-
tion at the same wavelengths.
Internal standard solution—A solution of acetonitrile in the
(2) Determine the infrared absorption spectrum of
mobile phase (1 in 4000).
Cefmenoxime Hydrochloride as directed in the potassium
Operating conditions—
bromide disk method under the Infrared Spectrophotomet-
Detector: An ultraviolet absorption photometer (wave-
ry, and compare the spectrum with the Reference Spectrum
length: 254 nm).
or the spectrum of Cefmenoxime Hydrochloride Reference
Column: A stainless steel column 4.6 mm in inside di-
Standard: both spectra exhibit similar intensities of absorp-
ameter and 15 cm in length, packed with octadecylsilanized
tion at the same wave numbers.
silica gel for liquid chromatography (5 mm in particle di-
(3) Determine the spectrum of a solution of Cef-
ameter).
menoxime Hydrochloride in deuterated dimethylsulfoxide
Column temperature: A constant temperature of about
for nuclear magnetic resonance spectroscopy (1 in 10) as
259C.
directed under the Nuclear Magnetic Resonance Spec-
Mobile phase: Dissolve 2.0 g of tetra-n-propylammonium
troscopy (1H), using tetramethylsilane for nuclear magnetic
bromide in 1000 mL of a mixture of water, acetonitrile and
resonance spectroscopy as an internal reference compound:
acetic acid-sodium acetate buŠer solution, pH 5.0 (83:13:4).
it exhibits two single signals, A and B, at around d 3.9 ppm,
Flow rate: Adjust the ‰ow rate so that the retention time
and a single signal C at around d 6.8 ppm. The ratio of the
of cefbuperazone is about 16 minutes.
integrated intensity of each signal, A:B:C, is about 3:3:1.
System suitability—
(4) Dissolve 0.01 g of Cefmenoxime Hydrochloride in
System performance: When the procedure is run with
1 mL of diluted sodium carbonate TS (1 in 20), add 5 mL of
10 mL of the standard solution under the above operating
acetic acid (100) and 2 drops of silver nitrate TS: a white
conditions, the internal standard and cefbuperazone are
precipitate is formed.
eluted in this order with the resolution between these peaks
being not less than 3. Optical rotation [a]20
D : -27 – -359(1 g, 0.1 mol/L phos-
System repeatability: When the test is repeated 6 times phate buŠer solution, pH 6.8, 100 mL, 100 mm).
with 10 mL of the standard solution under the above operat-
pH The pH of a solution obtained by dissolving 0.10 g of
ing conditions, the relative standard deviation of the ratios
Cefmenoxime Hydrochloride in 150 mL of water is between
of the peak area of cefbuperazone to that of the internal
2.8 and 3.3.
standard is not more than 1.0z.
Purity (1) Clarity and color of solution—A solution ob-
Containers and storage Containers—Hermetic containers.
tained by dissolving 1.0 g of Cefmenoxime Hydrochloride in
Storage—In a cold place.
10 mL of diluted sodium carbonate TS (1 in 4) is clear and
colorless to light yellow.
Supplement I, JP XIV O‹cial Monographs for Part I 1411

(2) Heavy metals—Proceed with 1.0 g of Cefmenoxime and ‰ow rate: Proceed as directed in the operating condi-
Hydrochloride according to Method 4, and perform the test. tions in the Assay.
Prepare the control solution with 2.0 mL of Standard Lead Time span of measurement: About 2.5 times as long as the
Solution (not more than 20 ppm). retention time of cefmenoxime.
(3) Arsenic—Prepare the test solution by incinerating System suitability—
1.0 g of Cefmenoxime Hydrochloride according to Method Test for required detectability: Measure exactly 5 mL of
4 and adding 10 mL of dilute hydrochloric acid to the the standard solution (1), add the mobile phase to make
residue after cooling, and perform the test (not more than exactly 100 mL. Conˆrm that the peak area of 1-methyl-1H-
2 ppm). tetrazol-5-thiol obtained from 10 mL of this solution is
(4) Related substances—Weigh accurately about 0.1 g equivalent to 4.5 to 5.5z of that from the standard solution
of Cefmenoxime Hydrochloride, dissolve in 20 mL of (1). Then, measure exactly 2 mL of the standard solution (2),
0.1 mol/L phosphate buŠer solution, pH 6.8, and add the add the mobile phase to make exactly 100 mL. Conˆrm that
mobile phase to make exactly 50 mL. Pipet 4 mL of this the peak area of cefmenoxime obtained from 10 mL of this
solution, add the mobile phase to make exactly 50 mL, and solution is equivalent to 1.5 to 2.5z of that from the stan-
use this solution as the sample solution. Separately, weigh dard solution (2).
accurately about 10 mg of 1-methyl-1H-tetrazol-5-thiol, and System performance: Proceed as directed in the system
dissolve in the mobile phase to make exactly 100 mL. Pipet suitability in the Assay.
4 mL of this solution, add the mobile phase to make exactly System repeatability: When the test is repeated 6 times
250 mL, and use this solution as the standard solution (1). with 10 mL of the standard solution (1) under the above
Weigh accurately about 0.1 g of Cefmenoxime Hydrochlo- operating conditions, the relative standard deviation of the
ride Reference Standard, dissolve in 20 mL of 0.1 mol/L peak area of 1-methyl-1H-tetrazol-5-thiol is not more than
phosphate buŠer solution, pH 6.8, and add the mobile phase 1.0z.
to make exactly 100 mL. Pipet 1 mL of this solution, add the
Water Not more than 1.5z (1 g, volumetric titration,
mobile phase to make exactly 250 mL, and use this solution
direct titration. Use a mixture of formamide for Karl Fisher
as the standard solution (2). Perform the test immediately
method and methanol for Karl Fisher method (2:1)).
after preparation of these solutions with exactly 10 mL each
of the sample solution, the standard solution (1) and the Assay Weigh accurately an amount of Cefmenoxime
standard solution (2) as directed under the Liquid Chro- Hydrochloride and Cefmenoxime Hydrochloride Reference
matography according to the following conditions. Deter- Standard, equivalent to about 50 mg (potency), dissolve
mine each peak area obtained from the chromatograms of each in 10 mL of 0.1 mol/L phosphate buŠer solution, pH
these solutions by the automatic integration method, and 6.8, and add the mobile phase to make exactly 50 mL. Pipet
calculate the amounts of 1-methyl-1H-tetrazol-5-thiol and 4 mL each of these solutions, add exactly 20 mL of the inter-
the total related substance by the following formula: the nal standard solution and the mobile phase to make 50 mL,
amount of 1-methyl-1H-tetrazol-5-thiol is not more than and use these solutions as the sample solution and the stan-
1.0z, and the total related substance is not more than 3.0z. dard solution, respectively. Perform the test with 10 mL each
of the sample solution and the standard solution as directed
Amount (z) of 1-methyl-1H-tetrazol-5-thiol
under the Liquid Chromatography according to the follow-
W A
= Sa × Ta × 20 ing conditions, and determine the ratios, QT and QS, of the
WT ASa
peak area of cefmenoxime to that of the internal standard.
Amount (z) of total related substance
Amount [mg (potency)] of cefmenoxime (C16H17N9O5S3)
W A W S
= Sa × Ta × 20 + Sb + T × 5 Q
WT ASa WT ASb = WS × T × 1000
QS
WSa: Amount (g) of 1-methyl-1H-tetrazol-5-thiol
WS: Amount [mg (potency)] of Cefmenoxime Hydrochlo-
WSb: Amount (g) of Cefmenoxime Hydrochloride Refer-
ride Reference Standard
ence Standard
WT: Amount (g) of the sample Internal standard solution—A solution of phthalimide in
ASa: Peak area of 1-methyl-1H-tetrazol-5-thiol from the methanol (3 in 2000).
standard solution (1) Operating conditions—
ASb: Peak area of cefmenoxime from the standard solu- Detector: An ultraviolet absorption photometer (wave-
tion (2) length: 254 nm).
ATa: Peak area of 1-methyl-1H-tetrazol-5-thiol from the Column: A stainless steel column 4 mm in inside diameter
sample solution and 15 cm in length, packed with octadecylsilanized silica gel
ST: Total area of the peaks other than 1-methyl-1H- for liquid chromatography (5 mm in particle diameter).
tetrazol-5-thiol and other than cefmenoxime from the Column temperature: A constant temperature of about
sample solution 259C.
Mobile phase: A mixture of water, acetonitrile and acetic
Operating conditions—
acid (100) (50:10:1).
Detector, column, column temperature, mobile phase,
1412 O‹cial Monographs for Part I Supplement I, JP XIV

Flow rate: Adjust the ‰ow rate so that the retention time spectra exhibit similar intensities of absorption at the same
of cefmenoxime is about 8 minutes. wave numbers.
System suitability— (3) Determine the spectrum of a solution of Cefodizime
System performance: When the procedure is run with Sodium in heavy water for nuclear magnetic resonance spec-
10 mL of the standard solution under the above operating troscopy (1 in 10) as directed under the Nuclear Magnetic
conditions, cefmenoxime and the internal standard are elut- Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl-
ed in this order with the resolution between these peaks propanesulfonate for nuclear magnetic resonance spec-
being not less than 2.3. troscopy as an internal reference compound: it exhibits
System repeatability: When the test is repeated 6 times single signals, A, B and C, at around d 2.3 ppm, at around
with 10 mL of the standard solution under the above operat- d 4.0 ppm, and at around d 7.0 ppm. The ratio of the in-
ing conditions, the relative standard deviation of the ratios tegrated intensity of these signals, A:B:C, is about 3:3:1.
of the peak areas of cefmenoxime to that of the internal (4) Cefodizime Sodium responds to the Qualitative Test
standard is not more than 1.0z. (1) for sodium salt.

Containers and storage Containers—Hermetic containers. Optical rotation [a]20


D : -56 – -629(0.2 g calculated on the
anhydrous basis and corrected by the ethanol amount,
water, 20 mL, 100 mm).
Add the following:
pH Dissolve 1.0 g of Cefodizime Sodium in 10 mL of
water: the pH of the solution is between 5.5 and 7.5.
Cefodizime Sodium
Purity (1) Clarity and color of solution—Dissolve 1.0 g
セフォジジムナトリウム of Cefodizime Sodium in 10 mL of water: the solution is
clear and pale yellow to light yellow.
(2) Heavy metals—Weigh 1.0 g of Cefodizime Sodium
in a crucible, cover loosely, and carbonize by gentle heating.
After cooling, add 2 mL of sulfuric acid, heat gradually un-
til the white fumes are no longer evolved, and ignite between
5009 C and 6009 C. Proceed according to Method 2, and per-
form the test. Prepare the control solution with 2.0 mL of
C20H18N6Na2O7S4: 628.63 Standard Lead Solution (not more than 20 ppm).
Disodium (6R,7R )-7-[(Z )-2-(2-aminothiazol-4-yl)- (3) Arsenic—Prepare the test solution with 1.0 g of
2-(methoxyimino)acetylamino]-3-[(5-carboxylatomethyl- Cefodizime Sodium according to Method 3, and perform the
4-methylthiazol-2-yl)sulfanylmethyl]-8-oxo-5-thia-1- test (not more than 2 ppm).
azabicycle[4.2.0]oct-2-ene-2-carboxylate [86329-79-5] (4) Related substances—Dissolve 30 mg of Cefodizime
Sodium in 10 mL of the mobile phase, and use this solution
Cefodizime Sodium contains not less than 890 mg as the sample solution. Pipet 1 mL of the sample solution,
(potency) per mg, calculated on the anhydrous basis add the mobile phase to make exactly 100 mL, and use this
and corrected by the ethanol amount. The potency of solution as the standard solution. Perform the test with ex-
Cefodizime Sodium is expressed as mass (potency) of actly 5 mL each of the sample solution and the standard solu-
cefodizime (C20H20N6O7S4: 584.67). tion as directed under the Liquid Chromatography accord-
ing to the following conditions, and determine each peak
Description Cefodizime Sodium occurs as a white to light
area by the automatic integration method: the peak area
yellowish white crystalline powder.
other than cefodizime from the sample solution is not more
It is very soluble in water, and practically insoluble in
than the peak area of cefodizime from the standard solution,
acetonitrile and in ethanol (99.5).
and the total area of the peaks other than cefodizime from
Identiˆcation (1) Determine the absorption spectrum of the sample solution is not more than 3 times the peak area of
a solution of Cefodizime Sodium (1 in 50,000) as directed cefodizime from the standard solution.
under the Ultraviolet-visible Spectrophotometry, and com- Operating conditions—
pare the spectrum with the Reference Spectrum or the spec- Detector, column, column temperature, mobile phase,
trum of a solution of Cefodizime Sodium Reference Stan- and ‰ow rate: Proceed as directed in the operating condi-
dard prepared in the same manner as the sample solution: tions in the Assay.
both spectra exhibit similar intensities of absorption at the Time span of measurement: About 4 times as long as the
same wavelengths. retention time of cefodizime after the solvent peak.
(2) Determine the infrared absorption spectrum of System suitability—
Cefodizime Sodium as directed in the potassium bromide Test for required detectability: Measure exactly 2 mL of
disk method under the Infrared Spectrophotometry, and the standard solution, and add the mobile phase to make
compare the spectrum with the Reference Spectrum or the exactly 20 mL. Conˆrm that the peak area of cefodizime
spectrum of Cefodizime Sodium Reference Standard: both obtained from 5 mL of this solution is equivalent to 7 to 13z
Supplement I, JP XIV O‹cial Monographs for Part I 1413

of that from 5 mL of the standard solution. tions, and determine the ratios, QT and QS, of the peak area
System performance, and system repeatability: Proceed as of cefodizime to that of the internal standard.
directed in the system suitability in the Assay.
Amount [mg (potency)] of cefodizime (C20H20N6O7S4)
(5) Ethanol—Weigh accurately about 1 g of Cefodizime
Q
Sodium, and dissolve in water to make exactly 10 mL. Pipet = WS × T × 1000
QS
2 mL of this solution, add exactly 2 mL of the internal stan-
dard solution, and use this solution as the sample solution. WS: Amount [mg (potency)] of Cefodizime Sodium Ref-
Separately, weigh accurately about 2 g of ethanol for gas erence Standard
chromatography, and add water to make exactly 1000 mL.
Internal standard solution—A solution of anhydrous
Pipet 2 mL of this solution, add exactly 2 mL of the internal
caŠeine (3 in 400).
standard solution, and use this solution as the standard solu-
Operating conditions—
tion. Perform the test with 10 mL each of the sample solution
Detector: An ultraviolet absorption photometer (wave-
and the standard solution as directed under the Gas Chro-
length: 254 nm).
matography according to the following conditions, and cal-
Column: A stainless steel column 4.6 mm in inside di-
culate the ratios, QT and QS, of the peak area of ethanol to
ameter and 25 cm in length, packed with octadecylsilanized
that of the internal standard: the amount of ethanol is not
silica gel for liquid chromatography (10 mm in particle
more than 2.0z.
diameter).
WS Q Column temperature: A constant temperature of about
Amount (z) of ethanol = × T
WT QS 259C.
Mobile phase: Dissolve 0.80 g of potassium dihydrogen
WS: Amount (g) of ethanol for gas chromatography
phosphate and 0.20 g of anhydrous disodium hydrogen
WT: Amount (g) of the sample
phosphate in a suitable amount of water, and add 80 mL of
Internal standard solution—A solution of 1-propanol (1 in acetonitrile and water to make 1000 mL.
400). Flow rate: Adjust the ‰ow rate so that the retention time
Operating conditions— of cefodizime is about 5 minutes.
Detector: A hydrogen ‰ame-ionization detector. System suitability—
Column: A glass column 3.2 mm in inside diameter and System performance: When the procedure is run with
3 m in length, packed with tetra‰uoroethylene polymer for 10 mL of the standard solution under the above operating
gas chromatography (180 – 250 mm in particle diameter) conditions, cefodizime and the internal standard are eluted
coated in 15z with polyethylene glycol 20 M. in this order with the resolution between these peaks being
Column temperature: A constant temperature of about not less than 6.
1009 C. System repeatability: When the test is repeated 6 times
Carrier gas: Nitrogen with 10 mL of the standard solution under the above operat-
Flow rate: Adjust the ‰ow rate so that the retention time ing conditions, the relative standard deviation of the ratios
of ethanol is about 3 minutes. of the peak area of cefodizime to that of the internal stan-
System suitability— dard is not more than 2.0z.
System performance: When the procedure is run with
Containers and storage Containers—Tight containers.
10 mL of the standard solution under the above operating
conditions, ethanol and the internal standard are eluted in
this order with the resolution between these peaks being not
less than 2.5. Cefotaxime Sodium
System repeatability: When the test is repeated 6 times
セフォタキシムナトリウム
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios
Change to read except the structural formula
of the peak area of ethanol to that of the internal standard is
and chemical name:
not more than 2.0z.

Water Not more than 4.0z (0.5 g, volumetric titration, Cefotaxime Sodium contains not less than 916 mg
direct titration). (potency) per mg, calculated on the dried basis. The
potency of Cefotaxime Sodium is expressed as mass
Assay Weigh accurately an amount of Cefodizime Sodium
(potency) of cefotaxime (C16H17N5O7S2: 455.47).
and Cefodizime Sodium Reference Standard, equivalent to
about 50 mg (potency), add exactly 10 mL of the internal Description Cefotaxime Sodium occurs as white to light
standard solution to dissolve, add water to make 100 mL, yellowish white crystalline powder.
and use these solutions as the sample solution and the stan- It is freely soluble in water, sparingly soluble in methanol,
dard solution. Perform the test with 10 mL each of the sam- and very slightly soluble in ethanol (95).
ple solution and the standard solution as directed under the
Identiˆcation (1) Dissolve 2 mg of Cefotaxime Sodium in
Liquid Chromatography according to the following condi-
0.01 mol/L hydrochloric acid TS to make 100 mL. Deter-
1414 O‹cial Monographs for Part I Supplement I, JP XIV

mine the absorption spectrum of this solution as directed un- Proceed as directed in the operating conditions in the Assay.
der the Ultraviolet-visible Spectrophotometry, and compare Time span of measurement: About 3.5 times as long as the
the spectrum with the Reference Spectrum: both spectra retention time of cefotaxime after the solvent peak.
exhibit similar intensities of absorption at the same wave- System suitability—
lengths. System performance, and system repeatability: Proceed as
(2) Determine the infrared absorption spectrum of directed in the system suitability in the Assay.
Cefotaxime Sodium as directed in the potassium bromide Test for required detectability: Measure exactly 2 mL of
disk method under the Infrared Spectrophotometry, and the standard solution, and add the mobile phase A to make
compare the spectrum with the Reference Spectrum: both exactly 100 mL. Pipet 2 mL of this solution, and add the
spectra exhibit similar intensities of absorption at the same mobile phase A to make exactly 20 mL. Conˆrm that the
wave numbers. peak area of cefotaxime obtained from 10 mL of this solu-
(3) Determine the spectrum of a solution of Cefotaxime tion is equivalent to 0.15 to 0.25z of that obtained from
Sodium in heavy water for nuclear magnetic resonance spec- 10 mL of the standard solution.
troscopy (1 in 125) as directed under the Nuclear Magnetic
Loss on drying Not more than 3.0z (1 g, 1059
C, 3 hours).
Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl-
propanesulfonate for nuclear magnetic resonance spec- Assay Weigh accurately an amount of Cefotaxime Sodium
troscopy as an internal reference compound: it exhibits three and Cefotaxime Reference Standard, equivalent to about
single signals, A, B and C, at around d 2.1 ppm, at around 40 mg (potency), dissolve each in the mobile phase A to
d 4.0 ppm and at around d 7.0 ppm. The ratio of the integrat- make exactly 50 mL, and use these solutions as the sample
ed intensity of each signal, A:B:C, is about 3:3:1. solution and the standard solution. Perform the test with
(4) Cefotaxime Sodium responds to the Qualitative Test exactly 10 mL each of the sample solution and the standard
(1) for sodium salt. solution as directed under the Liquid Chromatography ac-
cording to the following conditions, and determine the peak
Optical rotation [a]20
D : +58 – +649(0.25 g calculated on the
areas, AT and AS, of cefotaxime of these solutions.
dried basis, water, 25 mL, 100 mm).
Amount [mg (potency)] of cefotaxime (C16H17N5O7S2)
pH The pH of a solution obtained by dissolving 1.0 g of
A
Cefotaxime Sodium in 10 mL of water is between 4.5 and = WS × T × 1000
AS
6.5.
WS: Amount [mg (potency)] of Cefotaxime Reference
Purity (1) Clarity and color of solution—A solution
Standard
obtained by dissolving 1.0 g of Cefotaxime Sodium in 10 mL
of water is clear and light yellow. Operating conditions—
(2) Sulfate—Dissolve 2.0 g of Cefotaxime Sodium in Detector: An ultraviolet absorption photometer (wave-
40 mL of water, add 2 mL of dilute hydrochloric acid and length: 235 nm).
water to make 50 mL, shake well, and ˆlter. Discard ˆrst Column: A stainless steel column 4.6 mm in inside di-
10 mL of the ˆltrate, and to the subsequent 25 mL of the ameter and 15 cm in length, packed with octadecylsilanized
ˆltrate add water to make 50 mL. Perform the test with this silica gel for liquid chromatography (5 mm in particle di-
solution as the test solution. Prepare the control solution as ameter).
follows: To 1.0 mL of 0.005 mol/L sulfuric acid VS add Column temperature: A constant temperature of about
1 mL of dilute hydrochloric acid and water to make 50 mL 309C.
(not more than 0.048z). Mobile phase A: To 0.05 mol/L disodium hydrogen
(3) Heavy metals—Proceed with 1.0 g of Cefotaxime phosphate TS add phosphoric acid to adjust the pH to 6.25.
Sodium according to Method 2, and perform the test. Pre- To 860 mL of this solution add 140 mL of methanol.
pare the control solution with 2.0 mL of Standard Lead Mobile phase B: To 0.05 mol/L disodium hydrogen
Solution (not more than 20 ppm). phosphate TS add phosphoric acid to adjust the pH to 6.25.
(4) Arsenic—Prepare the test solution with 1.0 g of To 600 mL of this solution add 400 mL of methanol.
Cefotaxime Sodium according to Method 3, and perform Flowing of the mobile phase: Control the gradient by mix-
the test (not more than 2 ppm). ing the mobile phases A and B as directed in the following
(5) Related substances—Perform the test with 10 mL of table.
the sample solution obtained in the Assay as directed under
the Liquid Chromatography according to the following con- Time after injection Mobile phase Mobile phase
of sample (min) A (z ) B (z )
ditions, and determine each peak area obtained from the
chromatogram by the automatic integration method: the 0– 7 100 0
each peak area other than cefotaxime is not more than 1.0z 7– 9 100 → 80 0 → 20
and the total of these peak areas is not more than 3.0z. 9 – 16 80 20
Operating conditions— 16 – 45 80 → 0 20 → 100
Detector, column, column temperature, mobile phase A, 45 – 50 0 100
mobile phase B, ‰owing of the mobile phase, and ‰ow rate:
Supplement I, JP XIV O‹cial Monographs for Part I 1415

Flow rate: Adjust the ‰ow rate so that the retention time resonance spectroscopy as an internal reference compound:
of cefotaxime is about 14 minutes (about 1.3 mL/min). it exhibits single signals, A, B, C and D, at around d
System suitability— 3.6 ppm, at around d 4.0 ppm, at around d 5.1 ppm and at
System performance: To 1 mL of the standard solution around d 5.2 ppm, respectively. The ratio of the integrated
add 7.0 mL of water and 2.0 mL of methanol, mix, then add intensity of each signal, A:B:C:D, is about 3:3:1:1.
25 mg of sodium carbonate decahydrate, and shake. After
Optical rotation [a]20
D : +112 – +1249(0.5 g calculated on
allowing to stand for 10 minutes, add 3 drops of acetic acid
the anhydrous basis, a solution of sodium hydrogen car-
(100) and 1 mL of the standard solution, and mix. When the
bonate (1 in 200), 50 mL, 100 mm).
procedure is run with 10 mL of this solution under the above
operating conditions, desacetyl cefotaxime with the relative Purity (1) Clarity and color of solution—Dissolve 1.0 g
retention time being about 0.3 to cefotaxime and cefotaxime of Cefotetan in 10 mL of a solution of sodium hydrogen
are eluted in this order with the resolution between these carbonate (1 in 30): the solution is clear, and colorless or
peaks being not less than 20, and the symmetry coe‹cient of light yellow.
the peak of cefotaxime is not more than 2. (2) Heavy metals—Proceed with 1.0 g of Cefotetan
System repeatability: When the test is repeated 6 times according to Method 2, and perform the test. Prepare the
with 10 mL of the standard solution under the above operat- control solution with 2.0 mL of Standard Lead Solution (not
ing conditions, the relative standard deviation of the peak more than 20 ppm).
area of cefotaxime is not more than 2.0z. (3) Related substances—Weigh accurately about 0.1 g of
Cefotetan, dissolve in a suitable amount of methanol, add
Containers and storage Containers—Tight containers.
exactly 2 mL of the internal standard solution and methanol
to make 20 mL, and use this solution as the sample solution.
Separately, weigh accurately about 3 mg of 1-methyl-1H-
Cefotetan tetrazole-5-thiol for liquid chromatography, previously
dried in a desiccator (in vacuum, silica gel) for 2 hours, and
セフォテタン
about 2 mg of Cefotetan Reference Standard, calculated on
the anhydrous basis, dissolve in methanol to make exactly 20
Change to read except the structural formula
mL. Pipet 2 mL of this solution, add exactly 2 mL of the in-
and chemical name:
ternal standard solution and methanol to make 20 mL, and
use this solution as the standard solution. Perform the test
Cefotetan contains not less than 960 mg (potency)
with 5 mL of the sample solution and the standard solution
per mg, calculated on the anhydrous basis. The
as directed under the Liquid Chromatography according to
potency of Cefotetan is expressed as mass (potency) of
the following conditions, and determine the ratios, QTa, QTb,
cefotetan (C17H17N7O8S4).
QTc, QTd, QTe and QTf, of the peak areas of 1-methyl-1H-
Description Cefotetan occurs as white to light yellowish tetrazole-5-thiol, cefotetan lactone having the relative reten-
white powder. tion time of about 0.5 with respect to cefotetan, D2-cefote-
It is sparingly soluble in methanol, and slightly soluble in tan having the relative retention time of about 1.2 with
water and in ethanol (99.5). respect to cefotetan, isothiazole substance having the rela-
tive retention time of about 1.3 with respect to cefotetan,
Identiˆcation (1) Determine the absorption spectrum of
each of other related substances and the total of other relat-
a solution of Cefotetan in phosphate buŠer solution for
ed substances, to the peak area of the internal standard, re-
antibiotics, pH 6.5 (1 in 100,000) as directed under the
spectively, obtained from the sample solution, and the ra-
Ultraviolet-visible Spectrophotometry, and compare the
tios, QSa and QSb, of the peak areas of 1-methyl-1H-
spectrum with the Reference Spectrum or the spectrum of a
tetrazole-5-thiol and cefotetan, to the peak area of the inter-
solution of Cefotetan Reference Standard prepared in the
nal standard, respectively, obtained from the standard solu-
same manner as the sample solution: both spectra exhibit
tion. Calculate the amount of 1-methyl-1H-tetrazole-5-thiol,
similar intensities of absorption at the same wavelengths.
cefotetan lactone, D2-cefotetan, isothiazole substance, each
(2) Determine the infrared absorption spectrum of
of other related substances and the total of other related sub-
Cefotetan as directed in the potassium bromide disk method
stances from the following equations: the amount of 1-
under the Infrared Spectrophotometry, and compare the
methyl-1H-tetrazole-5-thiol is not more than 0.3z, cefote-
spectrum with the Reference Spectrum or the spectrum of
tan lactone is not more than 0.3z, D2-cefotetan is not more
Cefotetan Reference Standard: both spectra exhibit similar
than 0.5z, isothiazole substance is not more than 0.5z,
intensities of absorption at the same wavelengths.
each of other related substances is not more than 0.2z and
(3) Dissolve 50 mg of Cefotetan in 0.5 mL of a solution
the total of other related substances is not more than 0.4z.
of sodium hydrogen carbonate in heavy water for nuclear
magnetic resonance spectroscopy (1 in 25). Determine the
spectrum of this solution as directed under the Nuclear
Magnetic Resonance Spectroscopy (1H), using sodium
3-trimethylsilylpropanesulfonate for nuclear magnetic
1416 O‹cial Monographs for Part I Supplement I, JP XIV

WSa Q 1 and 15 cm in length, packed with octadecylsilanized silica gel


1-Methyl-1H-tetrazole-5-thiol (z) = × Ta ×
WT QSa 100 for liquid chromatography (5 mm in particle diameter).
W Q 1 Column temperature: A constant temperature of about
Cefotetan lactone (z) = Sb × Tb ×
WT QSb 100 409C.
W Q 1 Mobile phase: A mixture of 0.1 mol/L phosphate buŠer
D2-Cefotetan (z) = Sb × Tc ×
WT QSb 100 solution, pH 7.0, water and a solution of tetrabutylammoni-
W Q 1 um hydrogensulfate in acetonitrile (1 in 150) (9:9:2).
Isothiazole substance (z) = Sb × Td ×
WT QSb 100 Flow rate: Adjust the ‰ow rate so that the retention time
W Q 1 of l-substance is about 40 minutes.
Each of other related substances (z) = Sb × Te ×
WT QSb 100 System suitability—
W Q 1 System performance: When the procedure is run with 5 mL
Total of other related substances (z) = Sb × Tf ×
WT QSb 100 of the sample solution under the above operating conditions,
l-substance and d-substance are eluted in this order with the
WSa: Amount (mg) of 1-methyl-1H-tetrazole-5-thiol
resolution between these peaks being not less than 1.5.
WSb: Amount (mg) of Cefotetan Reference Standard, cal-
System repeatability: To exactly 1 mL of the sample solu-
culated on the anhydrous basis
tion add methanol to make exactly 10 mL. When the test is
WT: Amount (g) of the sample
repeated 6 times with 5 mL of this solution under the above
Internal standard solution—A solution of anhydrous operating conditions, the relative standard deviation of the
caŠeine in methanol (3 in 10,000). peak area of l-substance is not more than 5z.
Operating conditions—
Assay Weigh accurately an amount of Cefotetan and
Detector, column, column temperature, mobile phase,
Cefotetan Reference Standard, equivalent to about 50 mg
and ‰ow rate: Proceed as directed in the operating condi-
(potency), and dissolve each in phosphate buŠer solution for
tions in the Assay.
antibiotics, pH 6.5 to make exactly 50 mL. Pipet 15 mL each
Time span of measurement: About 3.5 times as long as the
of these solutions, add exactly 10 mL of the internal stan-
retention time of cefotetan.
dard solution and phosphate buŠer solution for antibiotics,
System suitability—
pH 6.5 to make 50 mL, and use these solutions as the sample
Test for required detectability: Measure exactly 15 mL of
solution and the standard solution. Perform the test with
the standard solution, and add methanol to make exactly
5 mL each of the sample solution and the standard solution
100 mL. Conˆrm that the peak area of cefotetan obtained
as directed under the Liquid Chromatography according to
from 5 mL of this solution is equivalent to 12 to 18z of that
the following conditions, and determine the ratios, QT and
from 5 mL of the standard solution.
QS, of the peak area of cefotetan to that of the internal
System performance: Proceed as directed in the system
standard.
suitability in the Assay.
System repeatability: When the test is repeated 6 times Amount [mg (potency)] of C17H17N7O8S4
with 5 mL of the standard solution under the above operat- Q
= WS × T × 1000
ing conditions, the relative standard deviation of the ratio of QS
the peak area of cefotetan to that of the internal standard is
WS: Amount [mg (potency)] of Cefotetan Reference Stan-
not more than 2.0z.
dard
Water Not more than 2.5z (1 g, volumetric titration, Internal standard solution—A solution of anhydrous
direct titration). caŠeine (1 in 1000).
Operating conditions—
Residue on ignition Not more than 0.10z (1 g).
Detector: An ultraviolet absorption photometer (wave-
Isomer ratio Dissolve 10 mg of Cefotetan in 20 mL of length: 254 nm).
methanol, and use this solution as the sample solution. Per- Column: A stainless steel column 4.6 mm in inside di-
form the test with 5 mL of the sample solution as directed un- ameter and 15 cm in length, packed with octadecylsilanized
der the Liquid Chromatography according to the following silica gel for liquid chromatography (5 mm in particle di-
conditions, and determine each peak area by the automatic ameter).
integration method. Calculate the amount of the adjacent Column temperature: A constant temperature of about
two peaks appeared at around the retention time of 40 409C.
minutes, one having shorter retention time is l-substance and Mobile phase: Dissolve 11.53 g of phosphoric acid in
another having longer retention time is d-substance, by the 1000 mL of water. To 850 mL of this solution add 50 mL of
area percentage method: the amount of l-substance is not acetonitrile, 50 mL of acetic acid (100) and 50 mL of
less than 35z and not more than 45z. methanol.
Operating conditions— Flow rate: Adjust the ‰ow rate so that the retention time
Detector: An ultraviolet absorption photometer (wave- of cefotetan is about 17 minutes.
length: 254 nm). System suitability—
Column: A stainless steel column 4 mm in inside diameter System performance: When the procedure is run with 5 mL
Supplement I, JP XIV O‹cial Monographs for Part I 1417

of the standard solution under the above operating condi- anhydrous basis, 0.1 mol/L hydrochloric acid TS, 10 mL,
tions, the internal standard and cefotetan are eluted in this 100 mm).
order with the resolution between these peaks being not less
Purity (1) Heavy metals—Proceed with 2.0 g of Cefo-
than 8.
tiam Hexetil Hydrochloride according to Method 2, and
System repeatability: When the test is repeated 5 times
perform the test. Prepare the control solution with 2.0 mL
with 5 mL of the standard solution under the above operat-
of Standard Lead Solution (not more than 10 ppm).
ing conditions, the relative standard deviation of the ratio of
(2) Arsenic—Prepare the test solution with 2.0 g of
the peak area of cefotetan to that of the internal standard is
Cefotiam Hexetil Hydrochloride according to Method 3,
not more than 1.0z.
and perform the test, using a solution of magnesium nitrate
Containers and storage Containers—Tight containers. hexahydrate in ethanol (95) (1 in 5) (not more than 1 ppm).
Storage—Light-resistant, and at a temperature not ex- (3) Related substance 1—Weigh accurately about 50 mg
ceeding 59C. of Cefotiam Hexetil Hydrochloride, and dissolve in a mix-
ture of diluted phosphoric acid (1 in 100) and acetonitrile
(4:1) to make exactly 50 mL. Pipet 10 mL of this solution,
Cefotiam Hexetil Hydrochloride add a mixture of diluted phosphoric acid (1 in 100) and
acetonitrile (4:1) to make exactly 25 mL, and use this solu-
塩酸セフォチアムヘキセチル tion as the sample solution. Separately, weigh accurately
about 50 mg of Cefotiam Hexetil Hydrochloride Reference
Change to read except the structural formula Standard, and dissolve in a mixture of diluted phosphoric
and chemical name: acid (1 in 100) and acetonitrile (4:1) to make exactly 50 mL.
Pipet 1 mL of this solution, add a mixture of diluted phos-
Cefotiam Hexetil Hydrochloride contains not less phoric acid (1 in 100) and acetonitrile (4:1) to make exactly
than 615 mg (potency) per mg, calculated on the anhy- 50 mL, and use this solution as the standard solution.
drous basis. The potency of Cefotiam Hexetil Perform the test with exactly 10 mL each of the sample solu-
Hydrochloride is expressed as mass (potency) of tion and the standard solution as directed under the Liquid
cefotiam (C18H23N9O4S3: 525.63). Chromatography according to the following conditions, and
determine each peak area by the automatic integration
Description Cefotiam Hexetil Hydrochloride occurs as a
method. Calculate the amount of the related substances by
white to light yellow powder.
the following equation: the amount of the related substance
It is very soluble in water, in methanol and in ethanol (95),
having the relative retention time of about 1.2 to one of the
freely soluble in dimethylsulfoxide, and slightly soluble in
peaks of cefotiam hexetil, which has the larger retention
acetonitrile.
time, is not more than 2.0z, and each amount of the other
It dissolves in 0.1 mol/L hydrochloric acid TS.
related substances is not more than 0.5z. For this calcula-
It is hygroscopic.
tion, use the value of the peak area obtained by the automat-
Identiˆcation (1) Determine the absorption spectrum of ic integration method of the related substance having the rel-
a solution of Cefotiam Hexetil Hydrochloride in 0.1 mol/L ative retention time of about 1.2 to one of the peaks of
hydrochloric acid TS (3 in 125,000) as directed under the cefotiam hexetil, which has the larger retention time, after
Ultraviolet-visible Spectrophotometry, and compare the multiplying by its sensitivity coe‹cient, 0.78.
spectrum with the Reference Spectrum or the spectrum of a
WS A
solution of Cefotiam Hexetil Hydrochloride Reference Amount (z) of each related substance = × T×5
WT AS
Standard prepared in the same manner as the sample solu-
tion: both spectra exhibit similar intensities of absorption at WS: Amount (g) of Cefotiam Hexetil Hydrochloride
the same wavelengths. Reference Standard
(2) Determine the spectrum of a solution of Cefotiam WT: Amount (g) of the sample
Hexetil Hydrochloride in deuterated dimethylsulfoxide for AS: Total of two peak areas of cefotiam hexetil from the
nuclear magnetic resonance spectroscopy (1 in 20) as direct- standard solution
ed under the Nuclear Magnetic Resonance Spectroscopy AT: Each peak area of related substance from the sample
(1H), using tetramethylsilane for nuclear magnetic resonance solution
spectroscopy as an internal reference compound: it exhibits
Operating conditions—
two single signals, A and B, at around d 2.8 ppm and at
Detector: An ultraviolet absorption photometer (wave-
around d 6.6 ppm, and a multiple signal, C, at around d 6.9
length: 254 nm).
ppm. The ratio of the integrated intensity of each signal,
Column: A stainless steel column 4 mm in inside diameter
A:B:C, is about 6:1:1.
and 15 cm in length, packed with octadecylsilanized silica gel
(3) To a solution of Cefotiam Hexetil Hydrochloride
for liquid chromatography (5 mm in particle diameter).
(1 in 200) add 2 mL of dilute nitric acid and 1 mL of silver
Column temperature: A constant temperature of about
nitrate TS, and mix: a white precipitate is formed.
259C.
Optical rotation [a]20
D : +52 – +609(0.1 g calculated on the Mobile phase A: A mixture of diluted 0.2 mol/L potassi-
1418 O‹cial Monographs for Part I Supplement I, JP XIV

um dihydrogen phosphate TS (1 in 2), acetonitrile and acetic AS: Peak area of cefotiam from the standard solution
acid (100) (72:28:1). AT: Each peak area from the sample solution
Mobile phase B: A mixture of acetonitrile, diluted
Operating conditions—
0.2 mol/L potassium dihydrogen phosphate TS (1 in 2) and
Detector: An ultraviolet absorption photometer (wave-
acetic acid (100) (60:40:1).
length: 254 nm).
Flowing of the mobile phase: Adjust so that the mixing
Column: A stainless steel column 4 mm in inside diameter
rate of the mobile phase A and the mobile phase B is
and 15 cm in length, packed with octadecylsilanized silica gel
changed lineally from 1:0 to 0:1 for 30 minutes.
for liquid chromatography (5 mm in particle diameter).
Flow rate: 0.7 mL per minute.
Column temperature: A constant temperature of about
Time span of measurement: As long as about 3 times of
259C.
the retention time of one of the cefotiam hexetil peaks,
Mobile phase: A mixture of a solution of diammonium
which appears ˆrst, after the solvent peak.
hydrogen phosphate (79 in 20,000), methanol and acetic acid
System suitability—
(100) (200:10:3).
Test for required detectability: Measure exactly 1 mL of
Flow rate: Adjust the ‰ow rate so that the retention time
the standard solution, and add a mixture of diluted phos-
of cefotiam is about 15 minutes.
phoric acid (1 in 100) and acetonitrile (4:1) to make exactly
Time span of measurement: As long as about 2 times of
50 mL. Conˆrm that each area of the two peaks of cefotiam
the retention time of cefotiam after the solvent peak.
hexetil obtained from 10 mL of this solution is equivalent to
System suitability—
1.6 to 2.4z of that from 10 mL of the standard solution.
Test for required detectability: Measure exactly 1 mL of
System performance: When the procedure is run with
the standard solution, and add the mobile phase to make
10 mL of the standard solution under the above operating
exactly 50 mL. Conˆrm that the peak area of cefotiam
conditions, the resolution between the two peaks of cefotiam
obtained from 10 mL of this solution is equivalent to 1.6 to
hexetil is not less than 2.0.
2.4z of that from 10 mL of the standard solution.
System repeatability: When the test is repeated 6 times
System performance: To 1 mL of a solution of
with 10 mL of the standard solution under the above operat-
acetaminophen in the mobile phase (1 in 50,000) add 3 mL
ing conditions, the relative standard deviation of the total of
of the standard solution, and mix well. When the procedure
the two peak areas of cefotiam hexetil is not more than
is run with 10 mL of this solution under the above operating
2.0z.
conditions, acetaminophen and cefotiam are eluted in this
(4) Related substance 2—Weigh accurately about 20 mg
order with the resolution between these peaks being not less
of Cefotiam Hexetil Hydrochloride, dissolve in exactly 2 mL
than 4.
of methanol, add a mixture of a solution of diammonium
System repeatability: When the test is repeated 6 times
hydrogen phosphate (79 in 20,000) and acetic acid (100)
with 10 mL of the standard solution under the above operat-
(200:3) to make exactly 50 mL, and use this solution as the
ing conditions, the relative standard deviation of the peak
sample solution. Separately, weigh accurately about 20 mg
area of cefotiam is not more than 2.0z.
of Cefotiam Hydrochloride Reference Standard, and dis-
(5) Total amount of related substances—The total of the
solve in the mobile phase to make exactly 50 mL. Pipet 2 mL
amount of related substances obtained in the Related sub-
of this solution, add the mobile phase to make exactly
stance 1 and the Related substance 2 is not more than 6.5z.
50 mL, and use this solution as the standard solution. Per-
form the test with exactly 10 mL each of the sample solution Water Not more than 3.5z (0.1 g, volumetric titration,
and the standard solution as directed under the Liquid Chro- direct titration).
matography according to the following conditions, and
Residue on ignition Not more than 0.10z (1 g).
determine each peak area by the automatic integration
method. Calculate the amount of the related substances by Isomer ratio Proceed the test with 20 mL of the sample
the following equation: the amounts of the related sub- solution obtained in the Assay as directed under the Liquid
stances having the relative retention time of about 0.1 and Chromatography according to the conditions directed in the
0.9 to cefotiam are not more than 1.0z, respectively, and Assay, and determine the areas of the two peaks, Aa for the
each amount of the related substances other than the related faster peak and Ab for the later peak, closely appeared each
substances having the relative retention time of about 0.1 other at the retention time of around 10 minutes: Aa/(Aa +
and 0.9 to cefotiam is not more than 0.5z. For this calcula- Ab) is not less than 0.45 and not more than 0.55.
tion, use the value of the peak area of the related substance
Assay Weigh accurately an amount of Cefotiam Hexetil
having the relative retention time of about 0.9 to cefotiam
Hydrochloride and Cefotiam Hexetil Hydrochloride Refer-
after multiplying by its sensitivity coe‹cient, 0.76.
ence Standard, equivalent to about 30 mg (potency), and
WS A dissolve each in a mixture of diluted phosphoric acid (1 in
Amount (z) of each related substance = × T×4
WT AS 100) and acetonitrile (4:1) to make exactly 50 mL. Measure
exactly 5 mL each of these solutions, add exactly 5 mL of the
WS: Amount (g) of Cefotiam Hydrochloride Reference
internal standard solution and a mixture of diluted phos-
Standard
phoric acid (1 in 100) and acetonitrile (4:1) to make exactly
WT: Amount (g) of the sample
Supplement I, JP XIV O‹cial Monographs for Part I 1419

50 mL, and use these solutions as the sample solution and Cefoxitin Sodium
the standard solution. Perform the test with 20 mL each of
the sample solution and the standard solution as directed セフォキシチンナトリウム
under the Liquid Chromatography according to the follow-
ing conditions, and determine the ratios, QT and QS, of the Change to read except the structural formula
peak area of cefotiam hexetil to that of the internal stand- and chemical name:
ard. For this calculation, the total of the areas of the two
peaks appeared closely each other at the retention time of Cefoxitin Sodium contains not less than 927 mg
around 10 minutes is used as the peak area of cefotiam (potency) and not more than 970 mg (potency) per mg,
hexetil. calculated on the dehydrated, de-acetone and de-
Amount [mg (potency)] of cefotiam (C18H23N9O4S3)
methanol basis. The potency of Cefoxitin Sodium
is expressed as mass (potency) of cefoxitin
Q
= WS × T × 1000 (C16H17N3O7S2: 427.45).
QS
Description Cefoxitin Sodium occurs as white to light yel-
WS: Amount [mg (potency)] of Cefotiam Hexetil Hydro-
lowish white granules or powder.
chloride Reference Standard
It is very soluble in water, soluble in methanol, and slight-
Internal standard solution—A solution of benzoic acid in a ly soluble in ethanol (95).
mixture of diluted phosphoric acid (1 in 100) and acetonitrile
Identiˆcation (1) Determine the absorption spectrum of
(4:1) (7 in 10,000).
a solution of Cefoxitin Sodium in 0.05 mol/L phosphate
Operating conditions—
buŠer solution, pH 7.0 (1 in 40,000) as directed under the
Detector: An ultraviolet absorption photometer (wave-
Ultraviolet-visible Spectrophotometry, and compare the
length: 254 nm).
spectrum with the Reference Spectrum: both spectra exhibit
Column: A stainless steel column 4 mm in inside diameter
similar intensities of absorption at the same wavelengths.
and 15 cm in length, packed with octadecylsilanized silica gel
(2) Determine the spectrum of a solution of Cefoxitin
for liquid chromatography (5 mm in particle diameter).
Sodium in heavy water for nuclear magnetic resonance spec-
Column temperature: A constant temperature of about
troscopy (1 in 10) as directed under the Nuclear Magnetic
259C.
Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl-
Mobile phase: A mixture of diluted 0.2 mol/L potassium
propanesulfonate for nuclear magnetic resonance spec-
dihydrogen phosphate TS (1 in 2), acetonitrile and acetic
troscopy as an internal reference compound: it exhibits
acid (100) (72:28:1).
single signals, A, B and C, at around d 3.5 ppm, at around
Flow rate: Adjust the ‰ow rate so that the retention time
d 3.9 ppm and at around d 5.1 ppm, respectively, and a mul-
of the faster peak of cefotiam hexetil is about 9 minutes.
tiple signal D between d 6.9 ppm and d 7.5 ppm. The ratio of
System suitability—
the integrated intensity of each signal, A:B:C:D, is about
System performance: When the procedure is run with
3:2:1:3.
20 mL of the standard solution under the above operating
(3) A solution of Cefoxitin Sodium (1 in 10) responds to
conditions, the internal standard and cefotiam hexetil are
the Qualitative Tests for sodium salt.
eluted in this order with the resolution between the two
peaks of cefotiam hexetil being not less than 2.0. Optical rotation [a]20
D : +206 – +2149(0.25 g calculated on
System repeatability: When the test is repeated 6 times the dehydrated, de-acetone and de-methanol basis,
with 20 mL of the standard solution under the above operat- methanol, 25 mL, 100 mm).
ing conditions, the relative standard deviation of the ratios
pH The pH of a solution obtained by dissolving 1.0 g of
of the peak area of cefotiam hexetil to that of the internal
Cefoxitin Sodium in 10 mL of water is between 4.5 and 6.5.
standard is not more than 1.0z.
Purity (1) Clarity and color of solution—A solution
Containers and storage Containers—Tight containers.
obtained by dissolving 1.0 g of Cefoxitin Sodium in 10 mL
of water is clear and a pale yellow to yellow.
(2) Heavy metals—Proceed with 1.0 g of Cefoxitin
Sodium according to Method 2, and perform the test. Pre-
pare the control solution with 2.0 mL of Standard Lead
Solution (not more than 20 ppm).
(3) Arsenic—Prepare the test solution with 1.0 g of
Cefoxitin Sodium according to Method 3, and perform the
test (not more than 2 ppm).
(4) Acetone and methanol—Dissolve 5.0 g of Cefoxitin
Sodium in water to make exactly 50 mL. Pipet 3 mL of this
solution in a centrifuge tube, allow to stand in an ice-cold
water for 2 minutes, and add exactly 3 mL of diluted
1420 O‹cial Monographs for Part I Supplement I, JP XIV

hydrochloric acid (1 in 50). After mixing, centrifuge, and use Water Not more than 1.0z (1 g, volumetric titration,
the supernatant liquid as the sample solution. Separately, to direct titration).
exactly 5 mL of acetone add water to make exactly 1000 mL,
Assay Weigh accurately an amount of Cefoxitin Sodium
and use this solution as the standard stock solution (1).
and Cefoxitin Reference Standard, equivalent to about 0.2 g
Separately, to exactly 5 mL of methanol add water to make
(potency), and dissolve each in phosphate buŠer solution,
exactly 1000 mL, and use this solution as the standard stock
pH 6.0 to make exactly 100 mL. Pipet 15 mL each of these
solution (2). To exactly 50 mL of the standard stock solution
solutions, add exactly 5 mL of the internal standard solution
(1) and exactly 5 mL of the standard stock solution (2) add
and phosphate buŠer solution, pH 6.0 to make 50 mL, and
water to make exactly 500 mL, and use this solution as the
use these solutions as the sample solution and the standard
standard solution. Perform the test with exactly 2 mL each of
solution, respectively. Perform the test with 10 mL each of
the sample solution and the standard solution as directed un-
the sample solution and the standard solution as directed
der the Gas Chromatography according to the following
under the Liquid Chromatography according to the follow-
conditions, and determine the peak areas of acetone, ATa
ing conditions, and determine the ratios, QT and QS, of the
and ASa, and the peak areas of methanol, ATb and ASb, of
peak area of cefoxitin to that of the internal standard.
these solutions: the amounts of acetone and methanol are
not more than 0.7z and not more than 0.1z, respectively. Amount [mg (potency)] of cefoxitin (C16H17N3O7S2)
Q
ATa = WS × T × 1000
Amount (z) of acetone = × 0.791 QS
ASa
A WS: Amount [mg (potency)] of Cefoxitin Reference
Amount (z) of methanol = Tb × 0.0791
ASb Standard

Operating conditions— Internal standard solution—A solution of anhydrous


Detector: An hydrogen ‰ame-ionization detector. caŠeine in phosphate buŠer solution, pH 6.0 (1 in 200).
Column: A glass column 3 mm in inside diameter and 2 m Operating conditions—
in length, packed with porous styrene-divinylbenzene Detector: An ultraviolet absorption photometer (wave-
copolymer for gas chromatography (150 – 180 mm in particle length: 254 nm).
diameter, 0.0085 mm in average pore size, 300 – 400 m2/g). Column: A stainless steel column 3.9 mm in inside di-
Column temperature: A constant temperature of about ameter and 30 cm in length, packed with octadecylsilanized
1109 C. silica gel for liquid chromatography (10 mm in particle di-
Carrier gas: Nitrogen ameter).
Flow rate: Adjust the ‰ow rate so that the retention time Column temperature: A constant temperature of about
of acetone is about 7 minutes. 259C.
System suitability— Mobile phase: A mixture of water, acetonitrile and acetic
System performance: When the procedure is run with 2 mL acid (100) (80:19:1).
of the standard solution under the above operating condi- Flow rate: Adjust the ‰ow rate so that the retention time
tions, methanol and acetone are eluted in this order, and the of cefoxitin is about 7 minutes.
number of theoretical plates of these peaks are not less than System suitability—
160, respectively, and the symmetry constant of these peaks System performance: When the procedure is run with
are not more than 2.3 and not more than 1.3, respectively. 10 mL of the standard solution under the above operating
System repeatability: When the test is repeated 5 times conditions, the internal standard and cefoxitin are eluted in
with 2 mL of the standard solution under the above operat- this order with the resolution between these peaks being not
ing conditions, the relative standard deviation of the peak less than 2.5.
area of acetone is not more than 5z. System repeatability: When the test is repeated 5 times
(5) Lactone substance—Dissolve 0.25 g of Cefoxitin with 10 mL of the standard solution under the above operat-
Sodium in 10 mL of water, and use this solution as the sam- ing conditions, the relative standard deviation of the ratios
ple solution. Pipet 0.5 mL of the sample solution, add water of the peak areas of cefoxitin to that of the internal standard
to make exactly 100 mL, and use this solution as the stan- is not more than 2.0z.
dard solution. Perform the test with these solutions as
Containers and storage Containers—Tight containers.
directed under the Thin-layer Chromatography. Spot 5 mL
Storage—Light-resistant.
each of the sample solution and the standard solution on a
plate of silica gel with ‰uorescent indicator for thin-layer
chromatography, develop with a mixture of ethyl acetate,
acetone, water and acetic acid (100) (5:2:1:1) to a distance of
about 15 cm, and air-dry the plate. Examine under ultravio-
let light (main wavelength: 254 nm): the spot other than the
principal spot from the sample solution is not more intense
than the principal spot from the standard solution.
Supplement I, JP XIV O‹cial Monographs for Part I 1421

Cefpiramide Sodium Reference Standard, equivalent to about 75 mg (potency),


dissolve them in 0.03 mol/L phosphate buŠer solution, pH
セフピラミドナトリウム 7.5 to make exactly 100 mL. Pipet 2 mL of this solution, add
0.03 mol/L phosphate buŠer solution, pH 7.5 to make ex-
Change to read except the structural formula actly 100 mL, and use this solution as the standard solution.
and chemical name: Perform the test with exactly 5 mL of the sample solution
and the standard solution as directed under the Liquid Chro-
Cefpiramide Sodium contains not less than 900 mg matography according to the following conditions, and de-
(potency) per mg, calculated on the anhydrous basis. termine each peak area by the automatic integration
The potency of Cefpiramide Sodium is expressed method. Calculate the amount of 1-methyl-1H-tetrazole-5-
as mass (potency) of cefpiramide (C25H24N8O7S2: thiol, each of the other related substances and the total of
612.64). the other related substances by the following equations: the
amount of 1-methyl-1H-tetrazole-5-thiol, each of the other
Description Cefpiramide Sodium occurs as white to yel-
related substances and the total of the other related sub-
lowish white powder.
stances are not more than 1.0z, not more than 1.5z and
It is very soluble in dimethylsulfoxide, freely soluble in
not more than 4.0z, respectively.
water, sparingly soluble in methanol, and slightly soluble in
ethanol (95). Amount (z) of 1-methyl-1H-tetrazole-5-thiol (C2H4N4S)
W A
Identiˆcation (1) Determine the absorption spectrum of = Sa × Ta
WT ASa
a solution of Cefpiramide Sodium in 0.05 mol/L phosphate
buŠer solution, pH 7.0 (1 in 50,000) as directed under the Amount (z) of each of other related substances
Ultraviolet-visible Spectrophotometry, and compare the W A
= Sb × Tc
spectrum with the Reference Spectrum: both spectra exhibit WT ASb
similar intensities of absorption at the same wavelengths.
WSa: Amount (mg) of 1-methyl-1H-tetrazole-5-thiol
(2) Determine the spectrum of a solution of Cefpiramide
WSb: Amount [mg (potency)] of Cefpiramide Reference
Sodium in deuterated dimethylsulfoxide for nuclear magnet-
Standard
ic resonance spectroscopy (1 in 10) as directed under the
WT: Amount (mg) of the sample
Nuclear Magnetic Resonance Spectroscopy (1H), using tetra-
ASa: Peak area of 1-methyl-1H-tetrazole-5-thiol from the
methylsilane for nuclear magnetic resonance spectroscopy as
standard solution
an internal reference compound: it exhibits single signals, A,
ASb: Peak area of cefpiramide from the standard solution
B and C, at around d 2.3 ppm, at around d 3.9 ppm and at
ATa: Peak area of 1-methyl-1H-tetrazole-5-thiol from the
around d 8.2 ppm, respectively. The ratio of the integrated
sample solution
intensity of each signal, A:B:C, is about 3:3:1.
ATc: Area of each peak other than 1-methyl-1H-tetrazole-
(3) Cefpiramide Sodium responds to the Qualitative Test
5-thiol and cefpiramide from the sample solution
(1) for sodium salt.
Operating conditions—
Optical rotation [a]20
D : -33 – -409(0.2 g calculated on the
Detector: An ultraviolet absorption photometer (wave-
anhydrous basis, 0.05 mol/L phosphate buŠer solution, pH
length: 254 nm).
7.0, 10 mL, 100 mm).
Column: A stainless steel column 4 mm in inside diameter
pH The pH of a solution obtained by dissolving 2.0 g of and 30 cm in length, packed with octylsilanized silica gel for
Cefpiramide Sodium in 20 mL of water is between 5.5 and liquid chromatography (10 mm in particle diameter).
8.0. Column temperature: A constant temperature of about
259C.
Purity (1) Clarity and color of solution—Dissolve 1.0 g
Mobile phase: A mixture of 0.03 mol/L phosphate buŠer
of Cefpiramide Sodium in 10 mL of 0.05 mol/L phosphate
solution, pH 7.5 and methanol (3:1).
buŠer solution, pH 7.0: the solution is clear, and colorless or
Flow rate: Adjust the ‰ow rate so that the retention time
light yellow.
of cefpiramide is about 11 minutes.
(2) Heavy metals—Proceed with 1.0 g of Cefpiramide
Time span of measurement: About 2 times as long as the
Sodium according to Method 2, and perform the test.
retention time of cefpiramide.
Prepare the control solution with 2.0 mL of Standard Lead
System suitability—
Solution (not more than 20 ppm).
Test for required detectability: Measure exactly 5 mL of
(3) Related substances—Weigh accurately about 25 mg
the standard solution, and add 0.03 mol/L phosphate buŠer
of Cefpiramide Sodium, dissolve in 0.03 mol/L phosphate
solution, pH 7.5 to make exactly 50 mL. Conˆrm that the
buŠer solution, pH 7.5 to make exactly 50 mL, and use this
peak area of 1-methyl-1H-tetrazole-5-thiol obtained from
solution as the sample solution. Separately, weigh accurately
5 mL of this solution is equivalent to 8 to 12z of that from
about 25 mg of 1-methyl-1H-tetrazole-5-thiol for liquid
5 mL of the standard solution.
chromatography, previously dried in a desiccator (in vacu-
System performance: Dissolve 25 mg of Cefpiramide
um, silica gel) for 2 hours, and an amount of Cefpiramide
Reference Standard and 7 mg of cinnamic acid in the mobile
1422 O‹cial Monographs for Part I Supplement I, JP XIV

phase to make 50 mL. When the procedure is run with 5 mL Add the following:
of this solution under the above operating conditions, cin-
namic acid and cefpiramide are eluted in this order with the Cefpodoxime Proxetil
resolution between these peaks being not less than 3.
System repeatability: When the test is repeated 6 times セフポドキシムプロキセチル
with 5 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
area of 1-methyl-1H-tetrazole-5-thiol is not more than
2.0z.

Water Not more than 7.0z (0.35 g, volumetric titration,


direct titration).

Assay Weigh accurately an amount of Cefpiramide


C21H27N5O9S2: 557.60
Sodium and Cefpiramide Reference Standard, equivalent to
(1RS )-1-(Isopropoxycarbonyloxy)ethyl (6R,7R )-7-[(Z )-
about 50 mg (potency), add exactly 5 mL of the internal
2-(2-aminothiazol-4-yl)-2-(methoxyimino)acetylamino]-
standard solution to dissolve, then add the mobile phase to
3-methoxymethyl-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-
make 100 mL, and use these solutions as the sample solution
2-ene-2-carboxylate [87239-81-4]
and the standard solution. Perform the test with 5 mL each
of the sample solution and the standard solution as directed
Cefpodoxime Proxetil contains not less than 706 mg
under the Liquid Chromatography according to the follow-
(potency) per mg, calculated on the anhydrous basis.
ing conditions, and determine the ratios, QT and QS, of the
The potency of Cefpodoxime Proxetil is expressed
peak area of cefpiramide to that of the internal standard.
as mass (potency) of cefpodoxime (C15H17N5O6S2:
Amount [mg (potency)] of cefpiramide (C25H24N8O7S2) 427.46).
Q
= WS × T × 1000 Description Cefpodoxime Proxetil occurs as a white to
QS
light brownish white powder.
WS: Amount [mg (potency)] of Cefpiramide Reference It is very soluble in acetonitrile, in methanol and in chlo-
Standard roform, freely soluble in ethanol (99.5), and very slightly
soluble in water.
Internal standard solution—A solution of 4-dimethylamino-
antipyrine (1 in 100). Identiˆcation (1) Determine the absorption spectrum of
Operating conditions— a solution of Cefpodoxime Proxetil in acetonitrile (3 in
Detector: An ultraviolet absorption photometer (wave- 200,000) as directed under the Ultraviolet-visible Spec-
length: 254 nm). trophotometry, and compare the spectrum with the Refer-
Column: A stainless steel column 4 mm in inside diameter ence Spectrum or the spectrum of a solution of Cef-
and 30 cm in length, packed with octylsilanized silica gel for podoxime Proxetil Reference Standard prepared in the same
liquid chromatography (10 mm in particle diameter). manner as the sample solution: both spectra exhibit similar
Column temperature: A constant temperature of about intensities of absorption at the same wavelengths.
259C. (2) Determine the infrared absorption spectrum of Cef-
Mobile phase: A mixture of 0.01 mol/L phosphate podoxime Proxetil as directed in the potassium bromide disk
buŠer solution, pH 6.8, acetonitrile, methanol and tetra- method under the Infrared Spectrophotometry, and com-
hydrofuran (22:1:1:1). pare the spectrum with the Reference Spectrum or the spec-
Flow rate: Adjust the ‰ow rate so that the retention time trum of Cefpodoxime Proxetil Reference Standard: both
of cefpiramide is about 7 minutes. spectra exhibit similar intensities of absorption at the same
System suitability— wave numbers.
System performance: When the procedure is run with 5 mL (3) Determine the spectrum of a solution of Cef-
of the standard solution under the above operating condi- podoxime Proxetil in deuterochloroform for nuclear
tions, cefpiramide and the internal standard are eluted in magnetic resonance spectroscopy (1 in 10) as directed under
this order with the resolution between these peaks being not the Nuclear Magnetic Resonance Spectroscopy (1H), using
less than 7. tetramethylsilane for nuclear magnetic resonance spec-
System repeatability: When the test is repeated 6 times troscopy as an internal reference compound: it exhibits dou-
with 5 mL of the standard solution under the above operat- ble signals, A and B, at around d 1.3 ppm and at around
ing conditions, the relative standard deviation of the ratio of d 1.6 ppm, and single signals, C and D, at around d 3.3 ppm
the peak area of cefpiramide to that of the internal standard and at around d 4.0 ppm. The ratio of the integrated inten-
is not more than 2.0z. sity of these signals, A:B:C:D, is about 2:1:1:1.

Containers and storage Containers—Tight containers. Optical rotation [a]20


D : +24.0 – +31.49(0.1 g calculated on
Storage—Light-resistant, and at a temperature not ex- the anhydrous basis, acetonitrile, 20 mL, 100 mm).
ceeding 59C.
Supplement I, JP XIV O‹cial Monographs for Part I 1423

Purity (1) Heavy metals—Proceed with 1.0 g of Cef- equivalent to 1.4 to 2.6z of them from 20 mL of the solution
podoxime Proxetil according to Method 2, and perform the for required detectability test, respectively.
test. Prepare the control solution with 2.0 mL of Standard System performance: Dissolve 1 mg of Cefpodoxime
Lead Solution (not more than 20 ppm). Proxetil in 100 mL of the mixture of water, acetonitrile and
(2) Related substances—Dissolve 50 mg of Cefpodoxime acetic acid (100) (99:99:2). When the procedure is run with
Proxetil in 50 mL of a mixture of water, acetonitrile and 20 mL of this solution under the above operating conditions,
acetic acid (100) (99:99:2), and use this solution as the sam- the isomer A and the isomer B of cefpodoxime proxetil are
ple solution. Perform the test with 20 mL of the sample solu- eluted in this order with the resolution between these peaks
tion as directed under the Liquid Chromatography accord- being not less than 6.
ing to the following conditions. If necessary, perform the System repeatability: Dissolve 1 mg of Cefpodoxime
test in the same manner with 20 mL of the mixture of water, Proxetil in 100 mL of the mixture of water, acetonitrile and
acetonitrile and acetic acid (100) (99:99:2) to compensate for acetic acid (100) (99:99:2). When the test is repeated 5 times
the base line. Determine each peak area by the automatic with 20 mL of this solution under the above operating condi-
integration method, and calculate the amounts of them by tions, the relative standard deviations of the peak areas of
the area percentage method: the peak, having the relative the isomer A and the isomer B are not more than 2.0z,
retention time of about 0.8 with respect to the isomer B of respectively.
cefpodoxime proxetil, is not more than 2.0z, the peak other
Water Not more than 2.5z (0.5 g, volumetric titration,
than cefpodoxime proxetil is not more than 1.0z, and the
direct titration).
sum of the peaks other than cefpodoxime proxetil is not
more than 6.0z. Residue on ignition Not more than 0.20z (1 g).
Operating conditions—
Isomer ratio Perform the test with 5 mL of the sample solu-
Detector: An ultraviolet absorption photometer (wave-
tion obtained in the Assay as directed under the Liquid
length: 254 nm).
Chromatography according to the following conditions, and
Column: A stainless steel column 4.6 mm in inside di-
determine the peak areas, Aa and Ab, of the two isomers of
ameter and 15 cm in length, packed with octadecylsilanized
cefpodoxime proxetil, having the smaller and larger reten-
silica gel for liquid chromatography (5 mm in particle di-
tion times, respectively, by the automatic integration
ameter).
method: Ab/(Aa + Ab) is between 0.50 and 0.60.
Column temperature: A constant temperature of about
Operating conditions—
229C.
Detector, column, column temperature, mobile phase,
Mobile phase A: A mixture of water, methanol and a solu-
and ‰ow rate: Proceed as directed in the operating condi-
tion of formic acid (1 in 50) (11:8:1).
tions in the Assay.
Mobile phase B: A mixture of methanol and a solution of
System suitability—
formic acid (1 in 50) (19:1).
System performance, and system repeatability: Proceed as
Flowing of the mobile phase: Control the gradient by
directed in the system suitability in the Assay.
mixing the mobile phases A and B as directed in the follow-
ing table. Assay Weigh accurately an amount of Cefpodoxime Prox-
etil and Cefpodoxime Proxetil Reference Standard, equiva-
Time after injection Mobile phase Mobile phase lent to about 60 mg (potency), dissolve in 80 mL of acetoni-
of sample (min) A (z ) B (z )
trile, add exactly 4 mL of the internal standard solution, add
0 – 65 95 5 acetonitrile to make 100 mL, and use these solutions as the
65 – 145 95 → 15 5 → 85 sample solution and the standard solution. Perform the test
145 – 155 15 85 with 5 mL each of the sample solution and the standard solu-
tion as directed under the Liquid Chromatography accord-
Flow rate: Adjust the ‰ow rate so that the retention time ing to the following conditions, and determine the ratios,
of the isomer B of cefpodoxime proxetil is about 60 minutes. QT1, QS1, QT2 and QS2, of the areas of the two peaks of the
Time span of measurement: About 2.5 times as long as the isomers of cefpodoxime proxetil to the peak area of the in-
retention time of the isomer B of cefpodoxime proxetil after ternal standard.
the solvent peak.
System suitability— Amount [mg (potency)] of cefpodoxime (C15H17N5O6S2)
Test for required detectability: Measure exactly 5 mL of Q + QT2
= WS × T1 × 1000
the sample solution, add the mixture of water, acetonitrile QS1 + QS2
and acetic acid (100) (99:99:2) to make exactly 200 mL, and WS: Amount [mg (potency)] of Cefpodoxime Proxetil
use this solution as the solution for required detectability Reference Standard
test. Pipet 2 mL of the solution for required detectability
test, and add the mixture of water, acetonitrile and acetic Internal standard solution—Dissolve 0.3 g of ethyl para-
acid (100) (99:99:2) to make exactly 100 mL. Conˆrm that hydroxybenzoate in a solution of citric acid in acetonitrile (1
the peak areas of the isomer A and the isomer B of cef- in 2000) to make 100 mL.
podoxime proxetil obtained from 20 mL of this solution are
1424 O‹cial Monographs for Part I Supplement I, JP XIV

Operating conditions— nuclear magnetic resonance spectroscopy as an internal


Detector: An ultraviolet absorption photometer (wave- reference compound: it exhibits three sharp single signals,
length: 240 nm). A, B and C, at around d 2.8 ppm, at around d 4.1 ppm and at
Column: A stainless steel column 4.6 mm in inside di- around d 6.3 ppm. The ratio of the integrated intensity of
ameter and 15 cm in length, packed with octadecylsilanized each signal, A:B:C, is about 4:3:1.
silica gel for liquid chromatography (5 mm in particle di-
Optical rotation [a]20
D : +95 – +1089(0.1 g calculated on the
ameter).
dehydrated basis, diluted acetic acid (100) (3 in 25), 100 mL,
Column temperature: A constant temperature of about
100 mm).
409C.
Mobile phase: A mixture of water and methanol (11:9). Purity (1) Heavy metals—Weigh 1.0 g of Cefroxadine in
Flow rate: Adjust the ‰ow rate so that the retention time a porcelain crucible, add 10 mL of a solution of magnesium
of the internal standard is about 11 minutes. nitrate hexahydrate in ethanol (95) (1 in 10), mix, burn the
System suitability— ethanol, and carbonize by gently heating. After cooling, add
System performance: When the procedure is run with 5 mL 2 mL of nitric acid, heat carefully, and incinerate by ignition
of the standard solution under the above operating condi- at 500 – 6009 C. If a carbonized substance is still remained,
tions, the internal standard, the isomer A and the isomer B moisten it with a small amount of nitric acid, and incinerate
are eluted in this order with the resolution between these again by ignition. After cooling, add 6 mL of hydrochloric
peaks being not less than 4. acid, and evaporate on a water bath to dryness. Moisten the
System repeatability: When the test is repeated 5 times residue with 3 drops of hydrochloric acid, and add 10 mL of
with 5 mL of the standard solution under the above operat- hot water to dissolve the residue by heating on a water bath.
ing conditions, the relative standard deviation of the ratios After cooling, adjust the pH between 3 and 4 with ammonia
of the peak area of the isomer B of cefpodoxime proxetil to TS, add 2 mL of dilute acetic acid, ˆlter if necessary, trans-
that of the internal standard is not more than 1.0z. fer to a Nessler tube, wash the crucible with 10 mL of water,
and add the washing and water to the tube to make 50 mL.
Containers and storage Containers—Tight containers.
Perform the test with this solution. Prepare the control solu-
tion as follows: Put 2.0 mL of Standard Lead Solution and
10 mL of a solution of magnesium nitrate hexahydrate in
Cefroxadine ethanol (95) (1 in 10) in a porcelain crucible, and proceed as
directed for the preparation of the test solution (not more
セフロキサジン
than 20 ppm).
(2) Arsenic—Prepare the test solution with 1.0 g of
Change to read except the structural formula
Cefroxadine according to Method 4, and perform the test
and chemical name:
(not more than 2 ppm).
(3) Related substances—Dissolve 10 mg of Cefroxadine
Cefroxadine contains not less than 921 mg (potency)
in 100 mL of the mobile phase, and use this solution as the
per mg, calculated on the dehydrated basis. The
sample solution. Pipet 1 mL of the sample solution, add the
potency of Cefroxadine is expressed as mass (potency)
mobile phase to make exactly 100 mL, and use this solution
of cefroxadine (C16H19N3O5S: 365.41).
as the standard solution. Perform the test with exactly 40 mL
Description Cefroxadine occurs as pale yellowish white to each of the sample solution and the standard solution as
light yellow, crystalline particles or powder. directed under the Liquid Chromatography according to the
It is very soluble in formic acid, slightly soluble in water following conditions, and determine each peak area ob-
and in methanol, and very slightly soluble in acetonitrile tained from the chromatograms of these solutions by the au-
and in ethanol (95). tomatic integration method: the areas of the peaks appeared
It dissolves in 0.001 mol/L hydrochloric acid TS and in at the relative retention times of 0.07, 0.6 and 0.8 against the
dilute acetic acid. peak of cefroxadine from the sample solution are not more
than 2 times, 4 times and 1 time of the peak area of cefroxa-
Identiˆcation (1) Determine the absorption spectrum of
dine from the standard solution, respectively, and any peak
a solution of Cefroxadine in 0.001 mol/L hydrochloric acid
area other than cefroxadine and other than the peaks men-
TS (1 in 50,000) as directed under the Ultraviolet-visible
tioned above from the sample solution is not more than 1/2
Spectrophotometry, and compare the spectrum with the
of the peak area of cefroxadine from the standard solution,
Reference Spectrum or the spectrum of a solution of Cefrox-
and the total area of the peaks other than cefroxadine from
adine Reference Standard prepared in the same manner as
the sample solution is not more than 6 times of the peak area
the sample solution: both spectra exhibit similar intensities
of cefroxadine from the standard solution.
of absorption at the same wavelengths.
Operating conditions—
(2) Determine the spectrum of a solution of Cefroxadine
Detector: An ultraviolet absorption photometer (wave-
in deuterated formic acid for nuclear magnetic resonance
length: 254 nm).
spectroscopy (1 in 10) as directed under the Nuclear Magnet-
Column: A stainless steel column 4.6 mm in inside di-
ic Resonance Spectroscopy (1H), using tetramethylsilane for
ameter and 10 cm in length, packed with octadecylsilanized
Supplement I, JP XIV O‹cial Monographs for Part I 1425

silica gel for liquid chromatography (5 mm in particle di- Column temperature: A constant temperature of about
ameter). 259C.
Column temperature: A constant temperature of about Mobile phase: A mixture of a solution of ammonium sul-
259C. fate (1 in 50) and acetonitrile (97:3).
Mobile phase: Dissolve 1.4 g of sodium perchlorate in Flow rate: Adjust the ‰ow rate so that the retention time
1000 mL of a mixture of water and acetonitrile (489:11). of cefroxadine is about 10 minutes.
Flow rate: Adjust the ‰ow rate so that the retention time System suitability—
of cefroxadine is about 20 minutes. System performance: When the procedure is run with
Time span of measurement: About 2 times as long as the 10 mL of the standard solution under the above operating
retention time of cefroxadine. conditions, cefroxadine and the internal standard are eluted
System suitability— in this order with the resolution between these peaks being
Test for required detectability: Measure exactly 2 mL of not less than 1.5.
the standard solution, and add the mobile phase to make System repeatability: When the test is repeated 6 times
exactly 20 mL. Conˆrm that the peak area of cefroxadine with 10 mL of the standard solution under the above operat-
obtained from 40 mL of this solution is equivalent to 7 to ing conditions, the relative standard deviation of the ratios
13z of that obtained from 40 mL of the standard solution. of the peak areas of cefroxadine to that of the internal stan-
System performance: Dissolve 3 mg of Cefroxadine and dard is not more than 1.0z.
15 mg of orcin in 100 mL of the mobile phase. When the
Containers and storage Containers—Tight containers.
procedure is run with 40 mL of this solution under the above
operating conditions, orcin and cefroxadine are eluted in
this order with the resolution between these peaks being not
less than 3. Cefteram Pivoxil
System repeatability: When the test is repeated 6 times
セフテラムピボキシル
with 40 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
Change to read except the structural formula
area of cefroxadine is not more than 2.0z.
and chemical name:
Water Not less than 8.5z and not more than 12.0z
(0.1 g, volumetric titration, direct titration). Cefteram Pivoxil contains not less than 743 mg
(potency) per mg, calculated on the anhydrous basis.
Assay Weigh accurately an amount of Cefroxadine and
The potency of Cefteram Pivoxil is expressed as mass
Cefroxadine Reference Standard, equivalent to about 50 mg
(potency) of cefteram (C16H17N9O5S2: 479.49).
(potency), dissolve each in a suitable amount of a mixture of
dilute acetic acid and phosphoric acid (500:1), add exactly Description Cefteram Pivoxil occurs as a white to pale
5 mL of the internal standard solution and a mixture of yellowish white powder.
dilute acetic acid and phosphoric acid (500:1) to make It is very soluble in acetonitrile, freely soluble in
200 mL, and use these solutions as the sample solution and methanol, in ethanol (95) and in chloroform, and practically
the standard solution. Perform the test with 10 mL each of insoluble in water.
the sample solution and the standard solution as directed
Identiˆcation (1) Determine the absorption spectrum of
under the Liquid Chromatography according to the follow-
a solution of Cefteram Pivoxil in 0.05 mol/L hydrochloric
ing conditions, and determine the ratios, QT and QS, of the
acid-methanol TS (1 in 100,000) as directed under the
peak area of cefroxadine to that of the internal standard.
Ultraviolet-visible Spectrophotometry, and compare the
Amount [mg (potency)] of C16H19N3O5S spectrum with the Reference Spectrum: both spectra exhibit
Q similar intensities of absorption at the same wavelengths.
= WS × T × 1000
QS (2) Determine the spectrum of a solution of Cefteram
Pivoxil in deuterated chloroform for nuclear magnetic
WS: Amount [mg (potency)] of Cefroxadine Reference
resonance spectroscopy (1 in 10) as directed under the
Standard
Nuclear Magnetic Resonance Spectroscopy (1H), using
Internal standard solution—Dissolve 1.6 g of vanillin in tetramethylsilane for nuclear magnetic resonance spec-
5 mL of methanol, and add a mixture of dilute acetic acid troscopy as an internal reference compound: it exhibits
and phosphoric acid (500:1) to make 100 mL. single signals A, B and C, at around d 1.2 ppm, at around
Operating conditions— d 2.5 ppm and at around d 4.0 ppm, respectively. The ratio
Detector: An ultraviolet absorption photometer (wave- of the integrated intensity of these signals, A:B:C, is about
length: 254 nm). 3:1:1.
Column: A stainless steel column 4.6 mm in inside di-
Optical rotation [a]20
D : +35 – +439(0.4 g calculated on the
ameter and 10 cm in length, packed with octadecylsilanized
anhydrous basis, methanol, 20 mL, 100 mm).
silica gel for liquid chromatography (5 mm in particle di-
ameter). Purity (1) Heavy metals—Proceed with 1.0 g of Cefter-
1426 O‹cial Monographs for Part I Supplement I, JP XIV

am Pivoxil according to Method 2, and perform the test. each of the sample solution and the standard solution as
Prepare the control solution with 2.0 mL of Standard Lead directed under the Liquid Chromatography according to the
Solution (not more than 20 ppm). following conditions, and determine the ratios, QT and QS,
(2) Arsenic—Prepare the test solution with 1.0 g of of the peak area of cefteram pivoxil to that of the internal
Cefteram Pivoxil according to Method 4, and perform the standard.
test (not more than 2 ppm).
Amount [mg (potency)] of cefteram (C16H17N9O5S2)
(3) Related substances—Dissolve 50 mg of Cefteram
Q
Pivoxilin in 50 mL of the mobile phase, and use this solution = WS × T × 1000
QS
as the sample solution. Pipet 1 mL of the sample solution,
add the mobile phase to make exactly 50 mL, and use this WS: Amount [mg (potency)] of Cefteram Pivoxil Mesity-
solution as the standard solution. Perform the test with ex- lenesulfonate Reference Standard
actly 10 mL each of the sample solution and the standard so-
Internal standard solution—A solution of methyl para-
lution as directed under the Liquid Chromatography accord-
hydroxybenzoate in diluted acetonitrile (1 in 2) (1 in 1000).
ing to the following conditions, and determine each peak
Operating conditions—
area by the automatic integration method: the area of the
Detector: An ultraviolet absorption photometer (wave-
peak, having the relative retention time of about 0.9 with
length: 254 nm).
respect to cefteram pivoxil from the sample solution is not
Column: A stainless steel column 4.6 mm in inside di-
more than 1.25 times the peak area of cefteram pivoxil from
ameter and 15 cm in length, packed with octadecylsilanized
the standard solution, the area of the peak, having the rela-
silica gel for liquid chromatography (5 mm in particle di-
tive retention time of about 0.1 with respect to cefteram
ameter).
pivoxil from the sample solution is not more than 0.25 times
Column temperature: A constant temperature of about
the peak area of cefteram pivoxil from the standard solu-
259C.
tion, and the total area of the peaks other than cefteram
Mobile phase: To 100 mL of acetic acid-sodium acetate
pivoxil from the sample solution is not more than 2.75 times
buŠer solution, pH 5.0 add 375 mL of acetonitrile and water
the peak area of cefteram pivoxil from the standard solu-
to make 1000 mL.
tion. For the above calculation, use the area of the peak,
Flow rate: Adjust the ‰ow rate so that the retention time
having the relative retention time of about 0.1, after mul-
of cefteram pivoxil is about 14 minutes.
tiplying by its sensitivity coe‹cient, 0.74.
System suitability—
Operating conditions—
System performance: When the procedure is run with
Detector, column, column temperature, mobile phase,
10 mL of the standard solution under the above operating
and ‰ow rate: Proceed as directed in the operating condi-
conditions, the internal standard and cefteram pivoxil are
tions in the Assay.
eluted in this order with the resolution between these peaks
Time span of measurement: About 2 times as long as the
being not less than 3.
retention time of cefteram pivoxil.
System repeatability: When the test is repeated 6 times
System suitability—
with 10 mL of the standard solution under the above operat-
Test for required detectability: Measure exactly 1 mL of
ing conditions, the relative standard deviation of the ratios
the standard solution, and add the mobile phase to make
of the peak area of cefteram pivoxil to that of the internal
exactly 10 mL. Conˆrm that the peak area of cefteram
standard is not more than 1.0z.
pivoxil obtained from 10 mL of this solution is equivalent to
7 to 13z of that from 10 mL of the standard solution. Containers and storage Containers—Tight containers.
System performance: When the procedure is run with Storage—In a cold place.
10 mL of the standard solution under the above operating
conditions, the number of theoretical plates and the symmet-
ry coe‹cient of the peak of cefteram pivoxil are not less than Ceftibuten
5000 and not more than 1.5, respectively.
System repeatability: When the test is repeated 6 times セフチブテン
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak Change the Identiˆcation (1) to read:
area of cefteram pivoxil is not more than 3.0z.
Identiˆcation (1) Determine the absorption spectrum of
Water Not more than 3.0z (0.3 g, coulometric titration). a solution of Ceftibuten in 0.1 mol/L phosphate buŠer solu-
tion for antibiotics, pH 8.0 (1 in 50,000) as directed under
Assay Weigh accurately an amount of Cefteram Pivoxil
the Ultraviolet-visible Spectrophotometry: it exhibits a max-
and Cefteram Pivoxil Mesitylenesulfonate Reference Stan-
imum between 261 nm and 265 nm.
dard, equivalent to about 40 mg (potency), dissolve each in
20 mL of diluted acetonitrile (1 in 2), add exactly 5 mL of
Change the Absorbance to read:
the internal standard solution and diluted acetonitrile (1 in 2)
to make 50 mL, and use these solutions as the sample solu- Absorbance E 11zcm (263 nm): 320 – 345 (20 mg calculated on
tion and the standard solution. Perform the test with 10 mL the anhydrous basis, 0.1 mol/L phosphate buŠer solution
Supplement I, JP XIV O‹cial Monographs for Part I 1427

for antibiotics, pH 8.0, 1000 mL). Ceftriaxone Sodium


Change the Optical rotation to read: セフトリアキソンナトリウム
Optical rotation [a]20
D : +135 – +1559(0.3 g calculated on
the anhydrous basis, 0.1 mol/L phosphate buŠer solution
Change the Identiˆcation (1) to read:
for antibiotics, pH 8.0, 50 mL, 100 mm). Identiˆcation (1) Determine the absorption spectrum of
a solution of Ceftriaxone Sodium (1 in 100,000) as directed
Change the Assay to read: under the Ultraviolet-visible Spectrophotometry, and com-
pare the spectrum with the Reference Spectrum or the spec-
Assay Weigh accurately an amount of Ceftibuten and
trum of a solution of Ceftriaxone Sodium Reference Stan-
Ceftibuten Hydrochloride Reference Standard, equivalent
dard prepared in the same manner as the sample solution:
to about 10 mg (potency), dissolve each in 36 mL of
both spectra exhibit similar intensities of absorption at the
0.1 mol/L phosphate buŠer solution for antibiotics, pH 8.0,
same wavelengths.
add exactly 4 mL each of the internal standard solution,
shake, and use these solutions as the sample solution and the
standard solution. Perform the test with 5 mL of the sample
Add the following next to Purity (3):
solution and the standard solution as directed under the Liq- Purity
uid Chromatography according to the following conditions, (4) Related substances 1—Dissolve 20 mg of Ceftriaxone
and calculate the ratios, QT and QS, of the peak area of Sodium in 10 mL of the mobile phase, and use this solution
ceftibuten to that of the internal standard. Keep the sample as the sample solution. Pipet 1 mL of the sample solution,
solution and the standard solution at 59C or below and use add a mixture of water and acetonitrile for liquid chro-
within 2 hours. matography (11:9) to make exactly 100 mL, and use this so-
lution as the standard solution. Perform the test with exactly
Amount [mg (potency)] of C15H14N4O6S2
10 mL each of the sample solution and the standard solution
Q
= WS × T × 1000 as directed under the Liquid Chromatography according to
QS
the following conditions, and determine each peak area by
WS: Amount [mg (potency)] of Ceftibuten Hydrochloride the automatic integration method: the peak areas of the
Reference Standard impurity 1 having the relative retention time of about 0.5
and the impurity 2 having the relative retention time of
Internal standard solution—A solution of methyl p-hydrox-
about 1.3 to ceftriaxone from the sample solution are not
ybenzoate in acetonitrile (3 in 4000).
more than the peak area of ceftriaxone from the standard
Operating conditions—
solution. In this case, these peak areas for the impurity 1 and
Detector: An ultraviolet absorption photometer (wave-
the impurity 2 are used after multiplying by 0.9 and 1.2,
length: 263 nm).
respectively.
Column: A stainless steel column 4 mm in inside diameter
Operating conditions—
and 20 cm in length, packed with octadecylsilanized silica gel
Detector: An ultraviolet absorption photometer (wave-
for liquid chromatography (7 mm in particle diameter).
length: 254 nm).
Column temperature: A constant temperature of about
Column: A stainless steel column 4.6 mm in inside di-
259C.
ameter and 25 cm in length, packed with octadecylsilanized
Mobile phase: A mixture of 0.005 mol/L n-decyl
silica gel for liquid chromatography (10 mm in particle di-
trimethylammonium bromide TS and acetonitrile (4:1).
ameter).
Flow rate: Adjust the ‰ow rate so that the retention time
Column temperature: A constant temperature of about
of ceftibuten is about 10 minutes.
259C.
System suitability—
Mobile phase: Dissolve 5.796 g of anhydrous disodium
System performance: Dissolve 5 mg of Ceftibuten in
hydrogen phosphate and 3.522 g of potassium dihydrogen
1 mol/L Hydrochloric acid TS to make 50 mL, and allow to
phosphate in water to make exactly 1000 mL, and use this
stand for 4 hours at room temperature. To 10 mL of this
solution as the solution A. Separately, dissolve 20.256 g of
solution add 0.1 mol/L phosphate buŠer solution for
citric acid monohydrate and 7.840 g of sodium hydroxide in
antibiotics, pH 8.0 to make 25 mL. When the procedure is
water to make exactly 1000 mL, and use this solution as the
run with 5 mL of this solution under the above operating
solution B. Dissolve 4.00 g of tetra-n-heptylammonium
conditions, trans-isomer and ceftibuten are eluted in this
bromide in 450 mL of acetonitrile for liquid chromato-
order with the resolution between these peaks being not less
graphy, add 55 mL of the solution A, 5 mL of the solution B
than 1.5.
and 490 mL of water.
System repeatability: When the test is repeated 6 times
Flow rate: Adjust the ‰ow rate so that the retention time
with 5 mL of the standard solution under the above operat-
of ceftriaxone is about 7 minutes.
ing conditions, the relative standard deviation of the ratios
Time span of measurement: About 2 times as long as the
of the peak area of ceftibuten to that of the internal standard
retention time of ceftriaxone.
is not more than 1.0z.
1428 O‹cial Monographs for Part I Supplement I, JP XIV

System suitability— matography, and add 55 mL of the solution A, 5 mL of the


Test for required detectability: Measure exactly 5 mL of solution B, 490 mL of water and 700 mL of acetonitrile for
the sample solution, add a mixture of water and acetonitrile liquid chromatography.
for liquid chromatography (11:9) to make exactly 200 mL, Flow rate: Adjust the ‰ow rate so that the retention time
and use this solution as the solution for system suitability of ceftriaxone is about 3 minutes.
test. Pipet 1 mL of the solution for system suitability test, Time span of measurement: About 10 times as long as the
and add a mixture of water and acetonitrile for liquid chro- retention time of ceftriaxone.
matography (11:9) to make exactly 100 mL. Conˆrm that System suitability—
the peak area of ceftriaxone obtained from 10 mL of this Test for required detectability: Measure exactly 5 mL of
solution is equivalent to 0.9 to 1.1z of that from 10 mL of the sample solution, add a mixture of acetonitrile for liquid
the solution for system suitability test. chromatography and water (23:11) to make exactly 100 mL,
System performance: Dissolve 10 mg of Ceftriaxone and use this solution as the solution for system suitability
Sodium in a mixture of water and acetonitrile for liquid test. Measure exactly 1 mL of the solution for system
chromatography (11:9) to make 5 mL, add 5 mL of a solu- suitability test, and add a mixture of acetonitrile for liquid
tion of diethyl terephthalate in a mixture of water and chromatography and water (23:11) to make exactly 100 mL.
acetonitrile for liquid chromatography (11:9) (9 in 5000), Conˆrm that the peak area of ceftriaxone obtained from
and add a mixture of water and acetonitrile for liquid chro- 10 mL of this solution is equivalent to 0.9 to 1.1z of that
matography (11:9) to make 200 mL. When the procedure is from 10 mL of the solution for system suitability test.
run with 10 mL of this solution under the above operating System performance: Dissolve 10 mg of Ceftriaxone
conditions, ceftriaxone and diethyl terephthalate are eluted Sodium in a mixture of acetonitrile for liquid chro-
in this order, with the resolution between these peaks being matography and water (23:11) to make 5 mL, add 5 mL of a
not less than 6. solution of diethyl terephthalate in a mixture of water and
System repeatability: When the test is repeated 6 times acetonitrile for liquid chromatography (11:9) (9 in 5000),
with 10 mL of the solution for system suitability test under and add a mixture of acetonitrile for liquid chromatography
the above operating conditions, the relative standard devia- and water (23:11) to make 200 mL. When the procedure is
tion of the peak area of ceftriaxone is not more than 1.0z. run with 10 mL of this solution under the above operating
(5) Related substances 2—Dissolve 10 mg of Ceftriaxone conditions, ceftriaxone and diethyl terephthalate are eluted
Sodium in 10 mL of the mobile phase, and use this solution in this order with the resolution between these peaks being
as the sample solution. Pipet 1 mL of the sample solution, not less than 3.
add a mixture of acetonitrile for liquid chromatography and System repeatability: When the test is repeated 6 times
water (23:11) to make exactly 100 mL, and use this solution with 10 mL of the solution for system suitability test under
as the standard solution. Perform the test with exactly 10 mL the above operating conditions, the relative standard devia-
each of the sample solution and the standard solution as tion of the peak area of ceftriaxone is not more than 1.0z.
directed under the Liquid Chromatography according to the
following conditions, and determine each peak area by the Change the Assay to read:
automatic integration method: the each peak area of the im-
Assay Weigh accurately an amount of Ceftriaxone Sodium
purities which appear after the peak of ceftriaxone from the
and Ceftriaxone Sodium Reference Standard, equivalent to
sample solution is not more than the peak area of
about 0.1 g (potency), dissolve each in a mixture of water
ceftriaxone from the standard solution, and the total peak
and acetonitrile for liquid chromatography (11:9) to make
area of these impurities is not more than 2.5 times of the
exactly 50 mL. Pipet 5 mL of each solution, add exactly
peak area from the standard solution.
5 mL of the internal standard solution and a mixture of
Operating conditions—
water and acetonitrile for liquid chromatography (11:9) to
Detector: An ultraviolet absorption photometer (wave-
make 200 mL, and use these solutions as the sample solution
length: 254 nm).
and the standard solution, respectively. Perform the test
Column: A stainless steel column 4.6 mm in inside di-
with 10 mL each of these solutions as directed under the Liq-
ameter and 25 cm in length, packed with octadecylsilanized
uid Chromatography according to the following conditions,
silica gel for liquid chromatography (10 mm in particle di-
and calculate the ratios, QT and QS, of the peak area of
ameter).
ceftriaxone to that of the internal standard.
Column temperature: A constant temperature of about
259C. Amount [mg (potency)] of ceftriaxone (C18H18N8O7S3)
Mobile phase: Dissolve 5.796 g of anhydrous disodium Q
= WS × T × 1000
hydrogen phosphate and 3.522 g of potassium dihydrogen QS
phosphate in water to make exactly 1000 mL, and use this
WS: Amount [mg (potency)] of Ceftriaxone Sodium Ref-
solution as the solution A. Separately, dissolve 20.256 g of
erence Standard
citric acid monohydrate and 7.840 g of sodium hydroxide in
water to make exactly 1000 mL, and use this solution as the Internal standard solution—A solution of diethyl terephtha-
solution B. Dissolve 4.00 g of tetra-n-heptylammonium late in a mixture of water and acetonitrile for liquid chro-
bromide in 450 mL of acetonitrile for liquid chro- matography (11:9) (9 in 5000).
Supplement I, JP XIV O‹cial Monographs for Part I 1429

Operating conditions— tion: both spectra exhibit similar intensities of absorption at


Detector: An ultraviolet absorption photometer (wave- the same wavelengths.
length: 254 nm). (2) Determine the infrared absorption spectrum of
Column: A stainless steel column 4.6 mm in inside di- Cefuroxime Axetil as directed in the potassium bromide disk
ameter and 25 cm in length, packed with octadecylsilanized method under the Infrared Spectrophotometry, and com-
silica gel for liquid chromatography (10 mm in particle pare the spectrum with the Reference Spectrum or the spec-
diameter). trum of Cefuroxime Axetil Reference Standard: both spec-
Column temperature: A constant temperature of about tra exhibit similar intensities of absorption at the same wave
259C. numbers.
Mobile phase: Dissolve 5.796 g of anhydrous disodium (3) Determine the spectrum of a solution of Cefuroxime
hydrogen phosphate and 3.522 g of potassium dihydrogen Axetil in deuterated dimethylsulfoxide for nuclear magnetic
phosphate in water to make exactly 1000 mL, and use this resonance spectroscopy (1 in 20) as directed under the
solution as solution A. Dissolve 20.256 g of citric acid Nuclear Magnetic Resonance Spectroscopy (1H), using
monohydrate and 7.840 g of sodium hydroxide in water to tetramethylsilane for nuclear magnetic resonance spec-
make exactly 1000 mL, and use this solution as solution B. troscopy as an internal reference compound: it exhibits a
Dissolve 4.00 g of tetra-n-heptylammonium bromide in double signal or a pair of double signals A at around
450 mL of acetonitrile for liquid chromatography, and add d 1.5 ppm, a pair of single signals B at around d 2.1 ppm,
490 mL of water, 55 mL of solution A, and 5 mL of solution and a single signal C at around d 3.9 ppm. The ratio of the
B. integrated intensity of each signal, A:B:C, is about 1:1:1.
Flow rate: Adjust the ‰ow rate so that the retention time
Optical rotation [a]20
D : +41 – +479 (0.5 g, methanol,
of ceftriaxone is about 7 minutes.
50 mL, 100 mm).
System suitability—
System performance: When the procedure is run with Purity (1) Heavy metals—Proceed with 2.0 g of
10 mL of the standard solution under the above operating Cefuroxime Axetil according to Method 2, and perform the
conditions, ceftriaxone and the internal standard are eluted test. Prepare the control solution with 2.0 mL of Standard
in this order with the resolution between these peaks being Lead Solution (not more than 10 ppm).
not less than 6. (2) Arsenic—Put 1.0 g of Cefuroxime Axetil in a cruci-
System repeatability: When the test is repeated 6 times ble, add 10 mL of a solution of magnesium nitrate hexahy-
with 10 mL of the standard solution under the above operat- drate in ethanol (95) (1 in 10), burn the ethanol, then heat
ing conditions, the relative standard deviation of the ratios gradually to incinerate. If a carbonized substance remains,
of the peak area of ceftriaxone to that of the internal stan- moisten with a small amount of nitric acid, and ignite to in-
dard is not more than 1.0z. cinerate. Cool, add 10 mL of dilute hydrochloric acid to the
residue, dissolve by warming on a water bath, and perform
the test using this solution as the test solution (not more than
Cefuroxime Axetil 2 ppm).
(3) Related substance—Dissolve 25 mg of Cefuroxime
セフロキシムアキセチル Axetil in 4 mL of methanol, add a solution of ammonium
dihydrogen phosphate (23 in 1000) to make 10 mL, and use
Change to read except the structural formula this solution as the sample solution. Pipet 1 mL of the
and chemical name: sample solution, add 40 mL of methanol and a solution of
ammonium dihydrogen phosphate (23 in 1000) to make ex-
Cefuroxime Axetil contains not less than 760 mg actly 100 mL, and use this solution as the standard solution.
(potency) per mg, calculated on the anhydrous basis Perform the test with exactly 2 mL each of the sample solu-
and corrected by the amount of acetone. The potency tion and the standard solution as directed under the Liquid
of Cefuroxime Axetil is expressed as mass (potency) of Chromatography according to the following conditions, and
cefuroxime (C16H16N4O8S: 424.39). determine each peak area by the automatic integration
method: the area of the peak other than cefuroxime axetil
Description Cefuroxime Axetil occurs as white to yellow-
obtained from the sample solution is not more than 1.5 times
ish white, non-crystalline powder.
the sum area of two peaks of cefuroxime axetil obtained
It is freely soluble in dimethylsulfoxide, soluble in
from the standard solution, and the sum area of the peaks
methanol, sparingly soluble in ethanol (95), and very slightly
other than cefuroxime axetil from the sample solution is not
soluble in water.
more than 4 times the sum area of two peaks of cefuroxime
Identiˆcation (1) Determine the absorption spectrum of axetil from the standard solution.
a solution of Cefuroxime Axetil in methanol (3 in 200,000) Operating conditions—
as directed under the Ultraviolet-visible Spectrophotometry, Detector, column, column temperature, mobile phase,
and compare the spectrum with the Reference Spectrum or and ‰ow rate: Proceed as directed in the operating condi-
the spectrum of a solution of Cefuroxime Axetil Reference tions in the Assay.
Standard prepared in the same manner as the sample solu- Time span of measurement: About 3 times as long as the
1430 O‹cial Monographs for Part I Supplement I, JP XIV

retention time of the peak having the larger retention time of tions, acetone and the internal standard are eluted in this
the two peaks of cefuroxime axetil after the solvent peak. order with the resolution between these peaks being not less
System suitability— than 5.
Test for required detectability: Measure exactly 1 mL of System repeatability: When the test is repeated 6 times
the standard solution, and add 4 mL of methanol and a solu- with 1 mL of the standard solution under the above operat-
tion of ammonium dihydrogen phosphate (23 in 1000) to ing conditions, the relative standard deviation of the ratios
make exactly 10 mL. Conˆrm that the sum area of the two of the peak area of acetone to that of the internal standard is
peaks of cefuroxime axetil obtained from 2 mL of this solu- not more than 5.0z.
tion is equivalent to 7 to 13z of that obtained from 2 mL of
Water Not more than 2.0z (0.4 g, volumetric titration,
the standard solution.
direct titration).
System performance: Proceed as directed in the system
suitability in the Assay. Residue on ignition Not more than 0.20z (0.5 g).
System repeatability: When the test is repeated 6 times
Isomer ratio Perform the test with 10 mL of the sample so-
with 2 mL of the standard solution under the above operat-
lution obtained in the Assay as directed under the Liquid
ing conditions, the relative standard deviation of the sum
Chromatography according to the following conditions, and
area of the two peaks of cefuroxime axetil is not more than
determine the area, Aa, of the peak having the smaller reten-
2.0z.
tion time and the area, Ab, of the peak having the bigger
(4) Acetone—Weigh accurately about 1.0 g of
retention time of the two peaks of cefuroxime axetil:
Cefuroxime Axetil, add exactly 0.2 mL of the internal stan-
Ab/(Aa + Ab) is between 0.48 and 0.55.
dard solution and dimethylsulfoxide to make exactly 10 mL,
Operating conditions—
and use this solution as the sample solution. Separately,
Detector, column, column temperature, mobile phase,
weigh accurately about 0.5 g of acetone, and add dimethyl-
and ‰ow rate: Proceed as directed in the operating condi-
sulfoxide to make exactly 100 mL. Pipet 0.2 mL of this solu-
tions in the Assay.
tion, add exactly 0.2 mL of the internal standard solution
System suitability—
and dimethylsulfoxide to make exactly 10 mL, and use this
System performance, and system repeatability: Proceed as
solution as the standard solution. Perform the test with 1 mL
directed in the system suitability in the Assay.
each of the sample solution and the standard solution as
directed under the Gas Chromatography according to the Assay Weigh accurately an amount of Cefuroxime Axetil
following conditions, determine each peak area by the and Cefuroxime Axetil Reference Standard, equivalent to
automatic integration method, and calculate the ratios, QT about 50 mg (potency), and dissolve each in methanol to
and QS, of the peak area of acetone to that of the internal make exactly 50 mL. Pipet 10 mL each of these solutions,
standard: the amount of acetone is not more than 1.3z. add exactly 5 mL of the internal standard solution, 5 mL of
methanol and a solution of ammonium dihydrogen phos-
WS Q
Amount (z) of acetone = × T × 0.2 phate (23 in 1000) to make 50 mL, and use these solutions as
WT QS
the sample solution and the standard solution. Perform the
WS: Amount (g) of acetone test with 10 mL each of the sample solution and the standard
WT: Amount (g) of the sample solution as directed under the Liquid Chromatography
according to the following conditions, and determine the
Internal standard solution—A solution of 1-propanol in
ratios, QT and QS, of the sum area of the two peaks of
dimethylsulfoxide (1 in 200).
cefuroxime axetil to the peak area of the internal standard.
Operating conditions—
Detector: A hydrogen ‰ame-ionization detector. Amount [mg (potency)] of cefuroxime (C16H16N4O8S)
Column: A glass column 3 mm in inside diameter and 2 m Q
= WS × T × 1000
in length, packed with siliceous earth for gas chromato- QS
graphy coated with a mixture of polyethylene glycol 600 for
WS: Amount [mg (potency)] of Cefuroxime Axetil Refer-
gas chromatography and polyethylene glycol 1500 for gas
ence Standard
chromatography (1:1) in the ratio of 20z (125 – 150 mm in
particle diameter). Internal standard solution—A solution of acetanilide in
Column temperature: A constant temperature of about methanol (27 in 5000).
909C. Operating conditions—
Temperature of injection port: A constant temperature of Detector: An ultraviolet absorption photometer (wave-
about 1159 C. length: 278 nm).
Carrier gas: Nitrogen Column: A stainless steel column 4.6 mm in inside di-
Flow rate: Adjust the ‰ow rate so that the retention time ameter and 20 cm in length, packed with trimethylsilanized
of the internal standard is about 4 minutes. silica gel for liquid chromatography (5 mm in particle di-
System suitability— ameter).
System performance: When the procedure is run with 1 mL Column temperature: A constant temperature of about
of the standard solution under the above operating condi- 259C.
Supplement I, JP XIV O‹cial Monographs for Part I 1431

Mobile phase: A mixture of a solution of ammonium operating conditions, cetraxate and phenol are eluted in this
dihydrogen phosphate (23 in 1000) and methanol (5:3). order with the resolution between these peaks being not less
Flow rate: Adjust the ‰ow rate so that the retention time than 5.
of the peak having the smaller retention time of the two System repeatability: When the test is repeated 6 times
peaks of cefuroxime axetil is about 8 minutes. with 10 mL of the standard solution under the above operat-
System suitability— ing conditions, the relative standard deviation of the peak
System performance: When the procedure is run with area of cetraxate is not more than 2.0z.
10 mL of the standard solution under the above operating (4) 3-( p-Hydroxyphenyl)propionic acid—To 0.10 g of
conditions, the internal standard and cefuroxime axetil are Cetraxate Hydrochloride add exactly 2 mL of the internal
eluted in this order with the resolution between the two standard solution and methanol to make 10 mL, and use this
peaks of cefuroxime axetil being not less than 1.5. solution as the sample solution. Separately, dissolve 25 mg
System repeatability: When the test is repeated 6 times of 3-( p-hydroxyphenyl)propionic acid in methanol to make
with 10 mL of the standard solution under the above operat- exactly 100 mL. To exactly 2 mL of this solution add exactly
ing conditions, the relative standard deviation of the ratios 2 mL of the internal standard solution and methanol to
of the sum area of the two peaks of cefuroxime axetil to the make 10 mL, and use this solution as the standard solution.
peak area of the internal standard is not more than 1.0z. Perform the test with 10 mL each of the sample solution and
the standard solution as directed under the Liquid Chro-
Containers and storage Containers—Tight containers.
matography according to the following conditions, and
Storage—Light-resistant.
calculate the ratios, QT and QS, of the peak area of 3-( p-
hydroxyphenyl)propionic acid to that of the internal stan-
dard: QT is not larger than QS.
Cetraxate Hydrochloride Internal standard solution—A solution of caŠeine in
methanol (1 in 4000).
塩酸セトラキサート
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Change the Purity (3) and (4) to read:
length: 230 nm).
Purity Column: A stainless steel column 6 mm in inside diameter
(3) cis Isomer—Dissolve 0.10 g of Cetraxate Hydrochlo- and 15 cm in length, packed with octadecylsilanized silica gel
ride in 10 mL of water, and use this solution as the sample for liquid chromatography (5 mm in particle diameter).
solution. To exactly 5 mL of the sample solution add water Column temperature: A constant temperature of about
to make exactly 100 mL. To exactly 2 mL of this solution 409C.
add water to make exactly 50 mL, and use this solution as Mobile phase: Adjust the pH of a mixture of water,
the standard solution. Perform the test with exactly 10 mL methanol and 0.5 mol/L ammonium acetate TS (15:5:2) to
each of the sample solution and the standard solution as 5.5 with acetic acid (31).
directed under the Liquid Chromatography according to the Flow rate: Adjust the ‰ow rate so that the retention time
following conditions. Determine each peak area of both so- of 3-( p-hydroxyphenyl)propionic acid is about 7 minutes.
lutions by the automatic integration method: the area of the System suitability—
peak which has a retention time 1.3 to 1.6 times that of System performance: When the procedure is run with
cetraxate from the sample solution is not larger than the 10 mL of the standard solution under the above operating
peak area of cetraxate from the standard solution. conditions, 3-( p-hydroxyphenyl)propionic acid and the in-
Operating conditions— ternal standard are eluted in this order with the resolution
Detector: An ultraviolet absorption photometer (wave- between these peaks being not less than 5.
length: 220 nm). System repeatability: When the test is repeated 6 times
Column: A stainless steel column 6 mm in inside diameter with 10 mL of the standard solution under the above operat-
and 15 cm in length, packed with octadecylsilanized silica gel ing conditions, the relative standard deviation of the ratios
for liquid chromatography (5 mm in particle diameter). of the peak area of 3-( p-hydroxyphenyl)propionic acid to
Column temperature: A constant temperature of about that of the internal standard is not more than 1.0z.
259C.
Mobile phase: Adjust the pH of a mixture of water,
methanol and 0.5 mol/L ammonium acetate TS (15:10:4) to
6.0 with acetic acid (31).
Flow rate: Adjust the ‰ow rate so that the retention time
of cetraxate is about 10 minutes.
System suitability—
System performance: Dissolve 0.02 g of Cetraxate
Hydrochloride and 0.01 g of phenol in 100 mL of water. To
2 mL of this solution add water to make 20 mL. When the
procedure is run with 10 mL of this solution under the above
1432 O‹cial Monographs for Part I Supplement I, JP XIV

Chloramphenicol and the spot on the original obtained from the sample solu-
tion are not more intense than the spot from the standard
クロラムフェニコール solution (1), and the total amount of these spots is not more
than 2.0z.
Change to read except the structural formula Loss on drying Not more than 0.5z (1 g, 1059
C, 3 hours).
and chemical name:
Residue on ignition Not more than 0.10z (1 g).
Chloramphenicol contains not less than 980 mg Assay Weigh accurately an amount of Chloramphenicol
(potency) per mg, calculated on the dried basis. The and Chloramphenicol Reference Standard, equivalent to
potency of Chloramphenicol is expressed as mass about 0.1 g (potency), dissolve each in 20 mL of methanol,
(potency) of chloramphenicol (C11H12Cl2N2O5). and add water to make exactly 100 mL. Pipet 20 mL each of
Description Chloramphenicol occurs as white to yellowish these solutions, and add water to make exactly 100 mL.
white, crystals or crystalline powder. Pipet 10 mL each of these solutions, add water to make ex-
It is freely soluble in methanol and in ethanol (99.5), and actly 100 mL, and use these solutions as the sample solution
slightly soluble in water. and the standard solution. Determine the absorbances, AT
and AS, at 278 nm of the sample solution and the standard
Identiˆcation (1) Determine the absorption spectrum of
solution as directed under the Ultraviolet-visible Spec-
the sample solution obtained in the Assay as directed under
trophotometry.
the Ultraviolet-visible Spectrophotometry, and compare the
spectrum with the Reference Spectrum or the spectrum of a Amount [mg (potency)] of C11H12Cl2N2O5
solution of Chloramphenicol Reference Standard prepared A
= WS × T × 1000
in the same manner as the sample solution: both spectra AS
exhibit similar intensities of absorption at the same wave-
WS: Amount [mg (potency)] of Chloramphenicol Refer-
lengths.
ence Standard
(2) Determine the infrared absorption spectrum of
Chloramphenicol as directed in the potassium bromide disk Containers and storage Containers—Tight containers.
method under the Infrared Spectrophotometry, and com-
pare the spectrum with the Reference Spectrum or the spec-
trum of Chloramphenicol Reference Standard: both spectra Add the following:
exhibit similar intensities of absorption at the same wave
numbers. Chloramphenicol Palmitate
Optical rotation [a]20
D : +18.5 – +21.59 (1.25 g, ethanol パルミチン酸クロラムフェニコール
(99.5), 25 mL, 100 mm).

Melting point 150 – 1559


C

Purity (1) Heavy metals—Proceed with 1.0 g of Chlo-


ramphenicol according to Method 2, and perform the test.
Prepare the control solution with 2.5 mL of Standard Lead
Solution (not more than 25 ppm).
(2) Arsenic—Prepare the test solution with 2.0 g of C27H42Cl2N2O6: 561.54
Chloramphenicol according to Method 4, and perform the (2R,3R )-2-(Dichloroacetyl)amino-3-hydroxy-3-(4-
test (not more than 1 ppm). nitrophenyl)propan-1-yl palmitate [530-43-8]
(3) Related substances—Dissolve 0.10 g of Chloram-
phenicol in 10 mL of methanol, and use this solution as the Chloramphenicol Palmitate contains not less than
sample solution. Pipet 1 mL of the sample solution, add 558 mg (potency) per mg, calculated on the dried basis.
methanol to make exactly 100 mL, and use this solution as The potency of Chloramphenicol Palmitate is ex-
the standard solution (1). Pipet 10 mL of the standard solu- pressed as mass (potency) of chloramphenicol
tion (1), add methanol to make exactly 20 mL, and use this (C11H12Cl2N2O5: 323.13).
solution as the standard solution (2). Perform the test with Description Chloramphenicol Palmitate occurs as a white
these solutions as directed under the Thin-layer Chro- to grayish white, crystalline powder.
matography. Spot 20 mL each of the sample solution and the It is freely soluble in acetone, sparingly soluble in
standard solutions (1) and (2) on a plate of silica gel with methanol and in ethanol (99.5), and practically insoluble in
‰uorescent indicator for thin-layer chromatography, de- water.
velop the plate with a mixture of chloroform, methanol and
acetic acid (100) (79:14:7) to a distance of about 15 cm, and Identiˆcation (1) Determine the absorption spectrum of
air-dry the plate. Examine under ultraviolet light (main a solution of Chloramphenicol Palmitate in ethanol (99.5) (1
wavelength: 254 nm): the spots other than the principal spot in 33,000) as directed under the Ultraviolet-visible Spec-
Supplement I, JP XIV O‹cial Monographs for Part I 1433

trophotometry, and compare the spectrum with the Refer- Column temperature: A constant temperature of about
ence Spectrum or the spectrum of a solution of Chloram- 209C.
phenicol Palmitate Reference Standard prepared in the same Mobile phase: Methanol
manner as the sample solution: both spectra exhibit similar Flow rate: Adjust the ‰ow rate so that the retention time
intensities of absorption at the same wavelengths. of chloramphenicol palmitate is about 5 minutes.
(2) Dissolve 5 mg each of Chloramphenicol Palmitate Time span of measurement: About 6 times as long as the
and Chloramphenicol Palmitate Reference Standard in retention time of chloramphenicol palmitate.
1 mL of acetone, and use these solutions as the sample solu- System suitability—
tion and the standard soution. Perform the test with these Test for required detectability: Dissolve 50 mg of Chlo-
solutions as directed under the Thin-layer Chromatography. ramphenicol Palmitate in 50 mL of methanol. Pipet 1 mL of
Spot 5 mL each of the sample solution and the standard solu- this solution, add methanol to make exactly 100 mL, and use
tion on a plate of silica gel with ‰uorescent indicator for this solution as the solution for system suitability test. Pipet
thin-layer chromatography. Develop the plate with a mix- 5 mL of the solution for system suitabillity test, and add
ture of acetone and cyclohexane (1:1) to a distance of about methanol to make exactly 50 mL. Conˆrm that the peak area
10 cm, and air-dry the plate. Examine under ultraviolet light of chloramphenicol palmitate obtained from 20 mL of this
(main wavelength: 254 nm): the principal spot obtained solution is equivalent to 7 to 13z of that obtained from
from the sample solution has the same Rf value as the spot 20 mL of the solution for system suitability test.
from the standard solution. System performance: When the procedure is run with
20 mL of the solution for system suitability test under the
Optical rotation [a]25
D : +21 – +259(1 g calculated on the
above operating conditions, the number of theoretical plates
dried basis, ethanol (99.5), 20 mL, 100 mm).
of the peak of chloramphenicol palmitate is not less than
Melting point 91 – 969C 5000.
System repeatability: When the test is repeated 6 times
Purity (1) Heavy metals—Proceed with 1.0 g of Chlo-
with 20 mL of the solution for system suitability test under
ramphenicol Palmitate according to Method 4, and perform
the above operating conditions, the relative standard devia-
the test. Prepare the control solution with 2.0 mL of Stan-
tion of the peak area of chloramphenicol palmitate is not
dard Lead Solution (not more than 20 ppm).
more than 1.0z.
(2) Arsenic—Prepare the test solution with 1.0 g of
Chloramphenicol Palmitate according to Method 3, and per- Loss on drying Not more than 1.0z (1 g, reduced pressure
form the test (not more than 2 ppm). not exceeding 0.67 kPa, 609C, 3 hours).
(3) Related substances—Dissolve 50 mg of Chloram-
Assay Weigh accurately an amount of Chloramphenicol
phenicol Palmitate in 50 mL of methanol, and use this solu-
Palmitate and Chloramphenicol Palmitate Reference Stan-
tion as the sample solution. Pipet 1 mL of the sample solu-
dard, equivalent to about 37 mg (potency), dissolve each in
tion, add methanol to make exactly 100 mL, and use this
40 mL of methanol and exactly 1 mL of acetic acid (100),
solution as the standard solution. Perform the test with ex-
and add methanol to make exactly 50 mL. Pipet 10 mL each
actly 20 mL each of the sample solution and the standard so-
of these solutions, add the mobile phase to make exactly
lution as directed under the Liquid Chromatography accord-
25 mL, and use these solutions as the sample solution and
ing to the following conditions. The test should be per-
the standard solution. Perform the test with exactly 10 mL
formed within 30 minutes after the sample solution and the
each of the sample solution and the standard solution as
standard solution are prepared. Determine each peak area
directed under the Liquid Chromatography according to the
by the automatic integration method: the total area of the
following conditions, and determine the peak areas, AT and
peaks other than the peak of chloramphenicol palmitate
A S.
from the sample solution is not more than 3.5 times the peak
area of chloramphenicol palmitate from the standard solu- Amount [mg (potency)] of chloramphenicol (C11H12Cl2N2O5)
tion. For this calculation, use the peak areas for chloram- A
= WS × T × 1000
phenicol, having the relative retention time of about 0.5 with AS
respect to chloramphenicol palmitate, and for chloram-
WS: Amount [mg (potency)] of Chloramphenicol Palmi-
phenicol dipalmitate, having the relative retention time of
tate Reference Standard
about 5.0 with respect to chloramphenicol palmitate, after
multiplying by their response factors, 0.5 and 1.4, respec- Operating conditions—
tively. Detector: An ultraviolet absorption photometer (wave-
Operating conditions— length: 280 nm).
Detector: An ultraviolet absorption photometer (wave- Column: A stainless steel column 3.9 mm in inside di-
length: 270 nm). ameter and 30 cm in length, packed with octadecylsilanized
Column: A stainless steel column 6.0 mm in inside di- silica gel for liquid chromatography (10 mm in particle di-
ameter and 15 cm in length, packed with octadecylsilanized ameter).
silica gel for liquid chromatography (5 mm in particle di- Column temperature: A constant temperature of about
ameter). 409C.
1434 O‹cial Monographs for Part I Supplement I, JP XIV

Mobile phase: A mixture of methanol, water and acetic (3) Chloramphenicol Sodium Succinate responds to the
acid (100) (172:27:1). Qualitative Test (1) for sodium salt.
Flow rate: Adjust the ‰ow rate so that the retention time
Optical rotation [a]25
D : +5 – +89(1.25 g calculated on the
of chloramphenicol palmitate is about 7 minutes.
anhydrous basis, water, 25 mL, 100 mm).
System suitability—
System performance: When the procedure is run with pH The pH of a solution obtained by dissolving 1.4 g of
10 mL of the standard solution under the above operating Chloramphenicol Sodium Succinate in 5 mL of water is
conditions, the number of theoretical plates of the peak of between 6.0 and 7.0.
chloramphenicol palmitate is not less than 2400.
Purity (1) Clarity and color of solution—Dissolve 1.0 g
System repeatability: When the test is repeated 6 times
of Chloramphenicol Sodium Succinate in 10 mL of water:
with 10 mL of the standard solution under the above operat-
the solution is clear and colorless to yellowish.
ing conditions, the relative standard deviation of the peak
(2) Heavy metals—Proceed with 1.0 g of Chloram-
area of chloramphenicol palmitate is not more than 1.0z.
phenicol Sodium Succinate according to Method 2, and
Containers and storage Containers—Tight containers. perform the test. Prepare the control solution with 2.0 mL
Storage—Light-resistant. of Standard Lead Solution (not more than 20 ppm).
(3) Arsenic—Prepare the test solution with 1.0 g of
Chloramphenicol Sodium Succinate according to Method 1,
Add the following: and perform the test (not more than 2 ppm).

Water Not more than 2.0z (1.0 g, volumetric titration,


Chloramphenicol Sodium Succinate direct titration).
コハク酸クロラムフェニコールナトリウム Assay Weigh accurately an amount of Chloramphenicol
Sodium Succinate, equivalent to about 20 mg (potency), dis-
solve in water to make exactly 1000 mL, and use this solu-
tion as the sample solution. Separately, weigh accurately an
amount of Chloramphenicol Succinate Reference Standard,
equivalent to about 20 mg (potency), add about 50 mL of
water to make a suspension, and add gradually about 7 mL
of 0.01 mol/L sodium hydroxide TS while stirring to adjust
C15H15Cl2N2NaO8: 445.18 the pH to 7.0. To this solution add water to make exactly
Monosodium (2R,3R )-2-(dichloroacetyl)amino- 1000 mL, and use this solution as the standard solution.
3-hydroxy-3-(4-nitrophenyl)propan-1-yl succinate Determine the absorbances, AT and AS, at 276 nm of the
[982-57-0] sample solution and the standard solution as directed under
the Ultraviolet-visible Spectrophotometry.
Chloramphenicol Sodium Succinate contains not
less than 711 mg (potency) per mg, calculated on the Amount [mg (potency)] of chloramphenicol (C11H12Cl2N2O5)
anhydrous basis. The potency of Chloramphenicol A
= WS × T × 1000
Sodium Succinate is expressed as mass (potency) of AS
chloramphenicol (C11H12Cl2N2O5: 323.13). WS: Amount [mg (potency)] of Chloramphenicol Suc-
Description Chloramphenicol Sodium Succinate occurs as cinate Reference Standard
white to yellowish white, crystals or crystalline powder. Containers and storage Containers—Hermetic containers.
It is very soluble in water, and freely soluble in methanol
and in ethanol (99.5).
It is hygroscopic.
Chlordiazepoxide Powder
Identiˆcation (1) Determine the absorption spectrum of
a solution of Chloramphenicol Sodium Succinate (1 in クロルジアゼポキシド散
50,000) as directed under the Ultraviolet-visible Spec-
trophotometry, and compare the spectrum with the Refer- Change the Assay to read:
ence Spectrum: both spectra exhibit similar intensities of Assay Conduct this procedure without exposure to day-
absorption at the same wavelengths. light, using light-resistant vessels. Weigh accurately a quan-
(2) Determine the infrared absorption spectrum of Chlo- tity of Chlordiazepoxide Powder, equivalent to about 0.1 g
ramphenicol Sodium Succinate as directed in the potassium of Chlordiazepoxide (C16H14ClN3O), transfer to a glass-
bromide disk method under the Infrared Spectrophotomet- stoppered ‰ask, wet with exactly 10 mL of water, add exact-
ry, and compare the spectrum with the Reference Spectrum: ly 90 mL of methanol, stopper, shake vigorously for 15
both spectra exhibit similar intensities of absorption at the minutes, and centrifuge. Pipet 10 mL of the supernatant liq-
same wave numbers. uid, add exactly 5 mL of the internal standard solution, add
Supplement I, JP XIV O‹cial Monographs for Part I 1435

methanol to make exactly 100 mL, and use this solution as to make exactly 100 mL, and centrifuge. Pipet 10 mL of the
the sample solution. Separately, weigh accurately about 0.1 supernatant liquid, add exactly 5 mL of the internal stan-
g of Chlordiazepoxide Reference Standard, previously dried dard solution, add methanol to make exactly 100 mL, and
in a desiccator (in vacuum, phosphorus (V) oxide, 609C) for use this solution as the sample solution. Separately, weigh
4 hours, and dissolve in exactly 10 mL of water and 90 mL accurately about 10 mg of Chlordiazepoxide Reference Stan-
of methanol. Pipet 10 mL of this solution, add exactly 5 mL dard, previously dried in a desiccator (in vacuum, phospho-
of the internal standard solution, add methanol to make 100 rus (V) oxide, 609C) for 4 hours, dissolve in 1 mL of water
mL, and use this solution as the standard solution. Perform and a suitable amount of methanol, add exactly 5 mL of the
the test with 10 mL each of the sample solution and the stan- internal standard solution, add methanol to make 100 mL,
dard solution as directed under the Liquid Chromatography and use this solution as the standard solution. Perform the
according to the following conditions, and calculate the ra- test with 10 mL each of the sample solution and the standard
tios, Q T and QS, of the peak area of chlordiazepoxide to that solution as directed under the Liquid Chromatography ac-
of the internal standard. cording to the following conditions, and calculate the ratios,
Q T and Q S, of the peak area of chlordiazepoxide to that of
Amount (mg) of chlordiazepoxide (C16H14ClN3O)
the internal standard.
Q
= WS × T × 10
QS Amount (mg) of chlordiazepoxide (C16H14ClN3O)
Q
WS: Amount (mg) of Chlordiazepoxide Reference = WS × T × 10
QS
Standard
WS: Amount (mg) of Chlordiazepoxide Reference
Internal standard solution—A solution of isobutyl salicylate
Standard
in methanol (1 in 20).
Operating conditions— Internal standard solution—A solution of isobutyl salicylate
Detector: An ultraviolet absorption photometer (wave- in methanol (1 in 20).
length: 254 nm). Operating conditions—
Column: A stainless steel column 4 mm in inside diameter Detector: An ultraviolet absorption photometer (wave-
and 25 cm in length, packed with octadecylsilanized silica gel length: 254 nm).
for liquid chromatography (10 mm in particle diameter). Column: A stainless steel column 4 mm in inside diameter
Column temperature: A constant temperature of about and 25 cm in length, packed with octadecylsilanized silica gel
259C. for liquid chromatography (10 mm in particle diameter).
Mobile phase: A mixture of methanol and 0.02 mol/L Column temperature: A constant temperature of about
ammonium dihydrogen phosphate TS (7:3). 259C.
Flow rate: Adjust the ‰ow rate so that the retention time Mobile phase: A mixture of methanol and 0.02 mol/L
of chlordiazepoxide is about 5 minutes. ammonium dihydrogen phosphate TS (7:3).
System suitability— Flow rate: Adjust the ‰ow rate so that the retention time
System performance: When the procedure is run with of chlordiazepoxide is about 5 minutes.
10 mL of the standard solution under the above operating System suitability—
conditions, chlordiazepoxide and the internal standard are System performance: When the procedure is run with
eluted in this order with the resolution between these peaks 10 mL of the standard solution under the above operating
being not less than 9. conditions, chlordiazepoxide and the internal standard are
System repeatability: When the test is repeated 6 times eluted in this order with the resolution between these peaks
with 10 mL of the standard solution under the above operat- being not less than 9.
ing conditions, the relative standard deviation of the ratios System repeatability: When the test is repeated 6 times
of the peak area of chlordiazepoxide to that of the internal with 10 mL of the standard solution under the above operat-
standard is not more than 1.0z. ing conditions, the relative standard deviation of the ratios
of the peak area of chlordiazepoxide to that of the internal
standard is not more than 1.0z.
Chlordiazepoxide Tablets
クロルジアゼポキシド錠 Chlormadinone Acetate
Change the Assay to read: 酢酸クロルマジノン

Assay Conduct this procedure without exposure to day-


Change the Purity (3) to read:
light, using light-resistant vessels. Weigh accurately a quan-
tity of Chlordiazepoxide Tablets, equivalent to about 0.1 g Purity
of Chlordiazepoxide (C16H14ClN3O), transfer to a glass- (3) Other steroids—Dissolve 20 mg of Chlormadinone
stoppered ‰ask, add 10 mL of water, and shake well to disin- Acetate in 10 mL of acetonitrile, and use this solution as the
tegrate. Add 60 mL of methanol, shake well, add methanol sample solution. Pipet 1 mL of the sample solution, add
1436 O‹cial Monographs for Part I Supplement I, JP XIV

acetonitrile to make exactly 100 mL, and use this solution as phenesin-2-carbamate by the automatic integration method:
the standard solution. Perform the test with exactly 10 mL the ratio, Ab/( Aa + Ab), is not larger than 0.007.
each of the sample solution and the standard solution as Operating conditions—
directed under the Liquid Chromatography according to the Detector: An ultraviolet absorption photometer (wave-
following conditions, and determine each peak area by the length: 280 nm).
automatic integration method: the total area of peaks other Column: A stainless steel column 4 mm in inside diameter
than the peak of chlormadinone acetate from the sample so- and 30 cm in length, packed with silica gel for liquid chro-
lution is not larger than the peak area of chlormadinone matography (5 mm in particle diameter).
acetate from the standard solution. Column temperature: A constant temperature of about
Operating conditions— 409C.
Detector: An ultraviolet absorption photometer (wave- Mobile phase: A mixture of hexane for liquid chro-
length: 236 nm). matography, 2-propanol and acetic acid (100) (700:300:1).
Column: A stainless steel column 6 mm in inside diameter Flow rate: Adjust the ‰ow rate so that the retention time
and 15 cm in length, packed with octadecylsilanized silica gel of chlorphenesin carbamate is about 9 minutes.
for liquid chromatography (5 mm in particle diameter). System suitability—
Column temperature: A constant temperature of about Test for required detection: Pipet 1 mL of the sample
309C. solution, add a mixture of hexane for liquid chromato-
Mobile phase: A mixture of acetonitrile and water (13:7). graphy and 2-propanol (7:3) to make exactly 100 mL, and
Flow rate: Adjust the ‰ow rate so that the retention time use this solution as the solution for system suitability test.
of chlormadinone acetate is about 10 minutes. To exactly 5 mL of the solution for system suitability test
Time span of measurement: About 1.5 times as long as the add a mixture of hexane for liquid chromatography and 2-
retention time of chlormadinone acetate after the solvent propanol (7:3) to make exactly 10 mL. Conˆrm that the
peak. peak area of chlorphenesin carbamate obtained from 10 mL
System suitability— of this solution is equivalent to 40 to 60z of that of chlor-
Test for required detection: To exactly 5 mL of the stan- phenesin carbamate obtained from 10 mL of the solution for
dard solution add acetonitorile to make exactly 50 mL. Con- system suitability test.
ˆrm that the peak area of chlormadinone acetate obtained System performance: Dissolve 0.1 g of Chlorphenesin
from 10 mL of this solution is equivalent to 7 to 13z of that Carbamate in 50 mL of methanol. To 25 mL of this solution
of chlormadinone acetate obtained from 10 mL of the stan- add 25 mL of dilute sodium hydroxide TS, and warm at
dard solution. 609C for 20 minutes. To 20 mL of this solution add 5 mL of
System performance: Dissolve 8 mg of Chlormadinone 1 mol/L hydrochloric acid TS, shake well with 20 mL of
Acetate and 2 mg of butyl parahydroxybenzoate in 100 mL ethyl acetate, and allow to stand to separate the ethyl acetate
of acetonitrile. When the procedure is run with 10 mL of this layer. When the procedure is run with 10 mL of this layer
solution under the above operating conditions, butyl para- under the above operating conditions, chlorphenesin, chlor-
hydroxybenzoate and chlormadinone acetate are eluted in phenesin carbamate and chlorphenesin-2-carbamate are
this order with the resolution between these peaks being not eluted in this order, with the ratios of the retention time of
less than 8. chlorphenesin and chlorphenesin-2-carbamate with respect
System repeatability: When the test is repeated 6 times to chlorphenesin carbamate are about 0.7 and about 1.2,
with 10 mL of the standard solution under the above operat- respectively, and with the resolution between the peaks of
ing conditions, the relative standard deviation of the peak chlorphenesin and chlorphenesin carbamate being not less
area of chlormadinone acetate is not more than 1.0z. than 2.0.
System repeatability: When the test is repeated 6 times
with 10 mL of the solution for system suitability test under
Chlorphenesin Carbamate the above operating conditions, the relative standard devia-
tion of the peak area of chlorphenesin carbamate is not more
カルバミン酸クロルフェネシン than 2.0z.
(ii) Other related substances: Dissolve 0.10 g of Chlor-
Change the Purity (3) to read: phenesin Carbamate in 10 mL of ethanol (95), and use this
solution as the sample solution. Pipet 1 mL of the sample
Purity
solution, add ethanol (95) to make exactly 20 mL. Pipet
(3) Related substances—(i) Chlorphenesin-2-carba-
2 mL of this solution, add ethanol (95) to make exactly
mate: Dissolve 0.10 g of Chlorphenesin Carbamate in 20 mL
20 mL, and use this solution as the standard solution. Per-
of a mixture of hexane for liquid chromatography and 2-
form the test with these solutions as directed under the Thin-
propanol (7:3), and use this solution as the sample solution.
layer Chromatography. Spot 50 mL each of the sample solu-
Perform the test with 10 mL of the sample solution as direct-
tion and the standard solution on a plate of silica gel for
ed under the Liquid Chromatography according to the
thin-layer chromatography. Develop the plate with a mix-
following conditions. Determine the peak area, Aa, of chlor-
ture of ethyl acetate, methanol and ammonia solution (28)
phenesin carbamate and the peak area, Ab, of chlor-
(17:2:1) to a distance of about 10 cm, and air-dry the plate.
Supplement I, JP XIV O‹cial Monographs for Part I 1437

Allow the plate to stand in iodine vapor for 20 minutes: the Standard
spots other than the principal spot from the sample solution
Internal standard solution—A solution of orcin in the
are not more intense than the spot from the standard solu-
mobile phase (1 in 500).
tion.
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Ciclacillin Column: A stainless steel column 4 mm in inside diameter
and 15 cm in length, packed with octadecylsilanized silica gel
シクラシリン
for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
Change to read except the structural formula
259C.
and chemical name:
Mobile phase: Dissolve 0.771 g of ammonium acetate in
about 900 mL of water, adjust the pH to 4.0 with acetic acid
Ciclacillin contains not less than 920 mg (potency)
(100), and add water to make 1000 mL. To 850 mL of this
per mg, calculated on the dehydrated basis. The
solution add 150 mL of acetonitrile.
potency of Ciclacillin is expressed as mass (potency) of
Flow rate: Adjust the ‰ow rate so that the retention time
ciclacillin (C15H23N3O4S).
of ciclacillin is about 4 minutes.
Description Ciclacillin occurs as white to light yellowish System suitability—
white crystalline powder. System performance: When the procedure is run with
It is sparingly soluble in water, slightly soluble in 10 mL of the standard solution under the above operating
methanol, and practically insoluble in acetonitrile and in conditions, ciclacillin and the internal standard are eluted in
ethanol (99.5). this order with the resolution between these peaks being not
less than 8.
Identiˆcation Determine the infrared absorption spectrum
System repeatability: When the test is repeated 6 times
of Ciclacillin as directed in the potassium bromide disk
with 10 mL of the standard solution under the above operat-
method under the Infrared Spectrophotometry, and com-
ing conditions, the relative standard deviation of the ratios
pare the spectrum with the Reference Spectrum or the spec-
of the peak areas of ciclacillin to that of the internal stan-
trum of Ciclacillin Reference Standard: both spectra exhibit
dard is not more than 1.0z.
similar intensities of absorption at the same wave numbers.
Containers and storage Containers—Tight containers.
Optical rotation [a]20
D : +300 – +3159(2 g, water, 100 mL,
100 mm).

Purity (1) Heavy metals—Proceed with 1.0 g of Ciclacil- Citric Acid


lin according to Method 2, and perform the test. Prepare the
control solution with 2.0 mL of Standard Lead Solution (not クエン酸
more than 20 ppm).
(2) Arsenic—Prepare the test solution with 1.0 g of Change to read except the structural formula
Ciclacillin according to Method 3, and perform the test (not and chemical name:
more than 2 ppm).
Citric Acid contains not less than 99.5z and not
Water Not more than 2.0z (1 g, volumetric titration,
more than 100.5z of anhydrous citric acid (C6H8O7:
direct titration).
192.12), calculated on the anhydrous basis.
Assay Weigh accurately an amount of Ciclacillin and
Description Citric Acid occurs as colorless crystals, white
Ciclacillin Reference Standard, equivalent to about 50 mg
granules or crystalline powder.
(potency), dissolve each in a suitable amount of the mobile
It is very soluble in water, and freely soluble in ethanol
phase, add exactly 5 mL of the internal standard solution
(95).
and the mobile phase to make 50 mL, and use these solutions
It is eŒorescent in dry air.
as the sample solution and the standard solution. Perform
the test with 10 mL each of the sample solution and the stan- Identiˆcation Determine the infrared absorption spectrum
dard solution as directed under the Liquid Chromatography of Citric Acid, previously dried at 1059C for 2 hours, as
according to the following conditions, and determine the directed in the potassium bromide disk method under the
ratios, QT and QS, of the peak area of ciclacillin to that of Infrared Spectrophotometry, and compare the spectrum
the internal standard. with the Reference Spectrum: both spectra exhibit similar
intensities of absorption at the same wave numbers.
Amount [mg (potency)] of C15H23N3O4S
Q Purity (1) Clarity and color of solution—Dissolve 2.0 g
= WS × T × 1000
QS of Citric Acid in water to make 10 mL: the solution is clear
and has no more color than the following control solutions
WS: Amount [mg (potency)] of Ciclacillin Reference
1438 O‹cial Monographs for Part I Supplement I, JP XIV

(1), (2) or (3). Assay Weigh accurately about 0.55 g of Citric Acid,
Control solution (1): To 1.5 mL of Cobalt (II) Chloride dissolve in 50 mL of water, and titrate with 1 mol/L sodium
Colorimetric Stock Solution and 6.0 mL of Iron (III) hydroxide VS (indicator: 2 drops of phenolphthalein TS).
Chloride Colorimetric Stock Solution add water to make
Each mL of 1 mol/L sodium hydroxide VS
1000 mL.
= 64.04 mg of C6H8O7
Control solution (2): To 0.15 mL of Cobalt (II) Chloride
Colorimetric Stock Solution, 7.2 mL of Iron (III) Chloride Containers and storage Containers—Tight containers.
Colorimetric Stock Solution and 0.15 mL of Copper (II)
Sulfate Colorimetric Stock Solution add water to make
1000 mL. Anhydrous Citric Acid
Control solution (3): To 2.5 mL of Cobalt (II) Chloride
Colorimetric Stock Solution, 6.0 mL of Iron (III) Chloride 無水クエン酸
Colorimetric Stock Solution and 1.0 mL of Copper (II)
Sulfate Colorimetric Stock Solution add water to make Change to read except the structural formula
1000 mL. and chemical name:
(2) Sulfates—Dissolve 2.0 g of Citric Acid in water to
make 30 mL, and use this solution as the sample solution. Anhydrous Citric Acid contains not less than 99.5z
Separately, dissolve 0.181 g of potassium sulfate in diluted and not more than 100.5z of C6H8O7, calculated on
ethanol (99.5) (3 in 10) to make exactly 500 mL. Pipet 5 mL the anhydrous basis.
of this solution, and add diluted ethanol (99.5) (3 in 10) to
Description Anhydrous Citric Acid occurs as colorless
make exactly 100 mL. To 4.5 mL of this solution add 3 mL
crystals, white granules or crystalline powder.
of a solution of barium chloride dihydrate (1 in 4), shake,
It is very soluble in water, and freely soluble in ethanol
and allow to stand for 1 minute. To 2.5 mL of this solution
(95).
add 15 mL of the sample solution and 0.5 mL of acetic acid
(31), and allow to stand for 5 minutes: the solution has no Identiˆcation Determine the infrared absorption spectrum
more turbidity than the following control solution. of Anhydrous Citric Acid, previously dried at 1059 C for 24
Control solution: Dissolve 0.181 g of potassium sulfate in hours, as directed in the potassium bromide disk method
water to make exactly 500 mL. Pipet 5 mL of this solution, under the Infrared Spectrophotometry, and compare the
add water to make exactly 100 mL, and proceed in the same spectrum with the Reference Spectrum: both spectra exhibit
manner as above using this solution instead of the sample similar intensities of absorption at the same wave numbers.
solution.
Purity (1) Clarity and color of solution—Dissolve 2.0 g
(3) Oxalate—Dissolve 0.80 g of Citric Acid in 4 mL of
of Anhydrous Citric Acid in water to make 10 mL: the solu-
water, add 3 mL of hydrochloric acid and 1 g of zinc, and
tion is clear and has no more color than the following con-
boil for 1 minute. After allowing to stand for 2 minutes, take
trol solutions (1), (2) or (3).
the supernatant liquid, add 0.25 mL of a solution of phenyl-
Control solution (1): To 1.5 mL of Cobalt (II) Chloride
hydrazinium hydrochloride (1 in 100), heat to boil, and then
Colorimetric Stock Solution and 6.0 mL of Iron (III)
cool quickly. To this solution add the equal volume of
Chloride Colorimetric Stock Solution add water to make
hydrochloric acid and 0.25 mL of a solution of potassium
1000 mL.
hexacyanoferrate (III) (1 in 20), mix, and allow to stand for
Control solution (2): To 0.15 mL of Cobalt (II) Chloride
30 minutes: the solution has no more color than the follow-
Colorimetric Stock Solution, 7.2 mL of Iron (III) Chloride
ing control solution prepared at the same time.
Colorimetric Stock Solution and 0.15 mL of Copper (II)
Control solution: To 4 mL of a solution of oxalic acid
Sulfate Colorimetric Stock Solution add water to make
dihydrate (1 in 10,000) add 3 mL of hydrochloric acid and
1000 mL.
1 g of zinc, and proceed in the same manner as the test solu-
Control solution (3): To 2.5 mL of Cobalt (II) Chloride
tion.
Colorimetric Stock Solution, 6.0 mL of Iron (III) Chloride
(4) Heavy metals—Proceed with 2.0 g of Citric Acid
Colorimetric Stock Solution and 1.0 mL of Copper (II)
according to Method 2, and perform the test. Prepare the
Sulfate Colorimetric Stock Solution add water to make 1000
control solution with 2.0 mL of Standard Lead Solution (not
mL.
more than 10 ppm).
(2) Sulfates—Dissolve 2.0 g of Anhydrous Citric Acid in
(5) Readily carbonizable substances—Perform the test
water to make 30 mL, and use this solution as the sample
with 0.5 g of Citric Acid, provided that the solution is heated
solution. Separately, dissolve 0.181 g of potassium sulfate in
at 909C for 1 hour and then cool quickly: the solution has no
diluted ethanol (99.5) (3 in 10) to make exactly 500 mL.
more color than Matching Fluid K.
Pipet 5 mL of this solution, and add diluted ethanol (99.5) (3
Water Not less than 7.5z and not more than 9.0z (0.5 g, in 10) to make exactly 100 mL. To 4.5 mL of this solution
volumetric titration, direct titration). add 3 mL of a solution of barium chloride dihydrate (1 in 4),
shake, and allow to stand for 1 minute. To 2.5 mL of this so-
Residue on ignition Not more than 0.10z (1 g).
lution add 15 mL of the sample solution and 0.5 mL of acet-
Supplement I, JP XIV O‹cial Monographs for Part I 1439

ic acid (31), and allow to stand for 5 minutes: the solution Add the following:
has no more turbidity than the following control solution.
Control solution: Dissolve 0.181 g of potassium sulfate in Clindamycin Hydrochloride
water to make exactly 500 mL. Pipet 5 mL of this solution,
add water to make exactly 100 mL, and proceed in the same 塩酸クリンダマイシン
manner as above using this solution instead of the sample
solution.
(3) Oxalate—Dissolve 0.80 g of Anhydrous Citric Acid
in 4 mL of water, add 3 mL of hydrochloric acid and 1 g of
zinc, and boil for 1 minute. After allowing to stand for 2
minutes, take the supernatant liquid, add 0.25 mL of a solu-
tion of phenylhydrazinium hydrochloride (1 in 100), heat to
boil, and then cool quickly. To this solution add the equal
C18H33ClN2O5S.HCl: 461.44
volume of hydrochloric acid and 0.25 mL of a solution of
Methyl 7-chloro-6,7,8-trideoxy-6-[(2S,4R )-1-methyl-4-
potassium hexacyanoferrate (III) (1 in 20), mix, and allow to
propylpyrrolidine-2-carboxamido]-1-thio-L-threo-a-D-
stand for 30 minutes: the solution has no more color than
galacto-octopyranoside monohydrochloride [21462-39-5]
the following control solution prepared at the same time.
Control solution: To 4 mL of a solution of oxalic acid
Clindamycin Hydrochloride contains not less than
dihydrate (1 in 10,000) add 3 mL of hydrochloric acid and
759 mg (potency) per mg. The potency of Clindamycin
1 g of zinc, and proceed in the same manner as the test solu-
Hydrochloride is expressed as mass (potency) of clin-
tion.
damycin (C18H33ClN2O5S: 424.98).
(4) Heavy metals—Proceed with 2.0 g of Anhydrous
Citric Acid according to Method 2, and perform the test. Description Clindamycin Hydrochloride occurs as white to
Prepare the control solution with 2.0 mL of Standard Lead grayish white, crystals or crystalline powder.
Solution (not more than 10 ppm). It is freely soluble in water and in methanol, and slightly
(5) Readily carbonizable substances—Perform the test soluble in ethanol (95).
with 0.5 g of Anhydrous Citric Acid, provided that the solu-
Identiˆcation Dissolve 0.1 g of Clindamycin Hydrochlo-
tion is heated at 909C for 1 hour and then cool quickly: the
ride in 5 mL of water, add 2 mL of sodium hydroxide TS,
solution has no more color than Matching Fluid K.
and mix: a white turbidity is produced. To this solution add
Water Not more than 1.0z (2 g, volumetric titration, 0.3 mL of sodium pentacyanonitrosylferrate (III) TS, mix,
direct titration). allow to stand at 60 to 659C for 10 minutes, and add 2 mL of
dilute hydrochloric acid: a blue-green color develops.
Residue on ignition Not more than 0.10z (1 g).
Optical rotation [a]25
D : +135 – +1509(0.5 g calculated on
Assay Weigh accurately about 0.55 g of Anhydrous Citric
the anhydrous basis, water, 25 mL, 100 mm).
Acid, dissolve in 50 mL of water, and titrate with 1 mol/L
sodium hydroxide VS (indicator: 2 drops of phenolphthalein Water Not more than 6.0z (0.3 g, volumetric titration,
TS). direct titration).

Each mL of 1 mol/L sodium hydroxide VS Assay Perform the test according to the Cylinder-plate
= 64.04 mg of C6H8O7 method as directed under the Microbial Assay for Antibiot-
ics according to the following conditions.
Containers and storage Containers—Tight containers.
(1) Test organism—Micrococcus luteus ATCC 9341
(2) Culture medium—Use the medium i in 3) Medium
for other organisms under (1) Agar media for seed and base
layer.
(3) Standard solutions—Weigh accurately an amount of
Clindamycin Hydrochloride Reference Standard, equivalent
to about 25 mg (potency), dissolve in 0.1 mol/L phosphate
buŠer solution, pH 7.0 to make exactly 250 mL, and use this
solution as the standard stock solution. Keep the standard
stock solution at a temperature not exceeding 159C and use
within 14 days. Take exactly a suitable amount of the stan-
dard stock solution before use, add 0.1 mol/L phosphate
buŠer solution, pH 7.0 to make solutions so that each mL
contains 2 mg (potency) and 1 mg (potency), and use these
solutions as the high concentration standard solution and
the low concentration standard solution, respectively.
(4) Sample solutions—Weigh accurately an amount of
1440 O‹cial Monographs for Part I Supplement I, JP XIV

Clindamycin Hydrochloride, equivalent to about 25 mg solution as directed under the Liquid Chromatography ac-
(potency), and dissolve in 0.1 mol/L phosphate buŠer solu- cording to the following conditions, and determine each
tion, pH 7.0 to make exactly 250 mL. Take exactly a suitable peak area by the automatic integration method: the peak
amount of this solution, add 0.1 mol/L phosphate buŠer area of clindamycin, having the relative retention time of
solution, pH 7.0 to make solutions so that each mL contains about 1.8 with respect to clindamycin phosphate, obtained
2 mg (potency) and 1 mg (potency), and use these solutions as from the sample solution is not more than 1/2 of the peak
the high concentration sample solution and the low concen- area of clindamycin phosphate from the standard solution,
tration sample solution, respectively. and the total area of the peaks other than clindamycin phos-
phate from the sample solution is not more than 4 times the
Containers and storage Containers—Tight containers.
peak area of clindamycin phosphate from the standard solu-
tion.
Operating conditions—
Clindamycin Phosphate Detector, column, column temperature, mobile phase,
and ‰ow rate: Proceed as directed in the operating condi-
リン酸クリンダマイシン
tions in the Assay.
Time span of measurement: About 2 times as long as the
Change to read except the structural formula
retention time of clindamycin phosphate after the solvent
and chemical name:
peak.
System suitability—
Clindamycin Phosphate contains not less than
Test for required detectability: Measure exactly 1 mL of
758 mg (potency) per mg, calculated on the anhydrous
the standard solution, and add the mobile phase to make
basis. The potency of Clindamycin Phosphate is
exactly 10 mL. Conˆrm that the peak area of clindamycin
expressed as mass (potency) of clindamycin
phosphate obtained from 20 mL of this solution is equivalent
(C18H33ClN2O5S: 424.98).
to 7 to 13z of that from 20 mL of the standard solution.
Description Clindamycin Phosphate occurs as a white to System performance, and system repeatability: Proceed as
pale yellowish white crystalline powder. directed in the system suitability in the Assay.
It is freely soluble in water, sparingly soluble in methanol,
Water Not more than 6.0z (0.5 g, volumetric titration,
and practically insoluble in ethanol (95).
direct titration).
Identiˆcation Determine the infrared absorption spectrum
Assay Weigh accurately an amount of Clindamycin Phos-
of Clindamycin Phosphate, previously dried at 1009 C for 2
phate and Clindamycin Phosphate Reference Standard,
hours, as directed in the paste method under the Infrared
equivalent to about 20 mg (potency), add exactly 25 mL of
Spectrophotometry, and compare the spectrum with the
the internal standard solution and the mobile phase to make
Reference Spectrum or the spectrum of Clindamycin Phos-
100 mL, and use these solutions as the sample solution and
phate Reference Standard previously dried at 1009C for 2
the standard solution. Perform the test with 20 mL each of
hours: both spectra exhibit similar intensities of absorption
the sample solution and the standard solution as directed un-
at the same wavelengths.
der the Liquid Chromatography according to the following
Optical rotation [a]20
D : +115 – +1309(0.25 g calculated on conditions, and determine the ratios, QT and QS, of the peak
the anhydrous basis, water, 25 mL, 100 mm). area of clindamycin phosphate to that of the internal stan-
dard.
pH Dissolve 0.10 g of Clindamycin Phosphate in 10 mL of
water. The pH of the solution is between 3.5 and 4.5. Amount [mg (potency)] of clindamycin (C18H33ClN2O5S)
Q
Purity (1) Clarity and color of solution—Dissolve 1.0 g = WS × T × 1000
QS
of Clindamycin Phosphate in 10 mL of freshly boiled and
cooled water: the solution is clear and colorless. WS: Amount [mg (potency)] of Clindamycin Phosphate
(2) Heavy metals—Proceed with 2.0 g of Clindamycin Reference Standard
Phosphate according to Method 4, and perform the test.
Internal standard solution—A solution of methyl para-
Prepare the control solution with 1.0 mL of Standard Lead
hydroxybenzoate in the mobile phase (3 in 50,000).
Solution (not more than 5 ppm).
Operating conditions—
(3) Arsenic—Prepare the test solution with 1.0 g of Clin-
Detector: An ultraviolet absorption photometer (wave-
damycin Phosphate according to Method 4, and perform the
length: 210 nm).
test (not more than 2 ppm).
Column: A stainless steel column 4 mm in inside diameter
(4) Related substances—Dissolve 0.1 g of Clindamycin
and 25 cm in length, packed with octylsilanized silica gel for
Phosphate in 100 mL of the mobile phase, and use this solu-
liquid chromatography (5 mm in particle diameter).
tion as the sample solution. Pipet 1 mL of the sample solu-
Column temperature: A constant temperature of about
tion, add the mobile phase to make exactly 100 mL, and use
259C.
this solution as the standard solution. Perform the test with
Mobile phase: Dissolve 10.54 g of potassium dihydrogen
exactly 20 mL each of the sample solution and the standard
Supplement I, JP XIV O‹cial Monographs for Part I 1441

phosphate in 775 mL of water, adjust the pH to 2.5 with Cloxacillin Sodium


phosphoric acid, and add 225 mL of acetonitrile.
Flow rate: Adjust the ‰ow rate so that the retention time クロキサシリンナトリウム
of clindamycin phosphate is about 8 minutes.
System suitability— Change the Description to read:
System performance: When the procedure is run with
Description Cloxacillin Sodium occurs as white to light
20 mL of the standard solution under the above operating
yellowish white, crystals or crystalline powder.
conditions, clindamycin phosphate and the internal standard
It is freely soluble in water, in N, N-dimethylformamide
are eluted in this order with the resolution between these
and in methanol, and sparingly soluble in ethanol (95).
peaks being not less than 4.
System repeatability: When the test is repeated 6 times
Change the Identiˆcation (1) to read:
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios Identiˆcation (1) Determine the absorption spectrum of
of the peak area of clindamycin phosphate to that of the a solution of Cloxacillin Sodium in methanol (1 in 2500) as
internal standard is not more than 2.5z. directed under the Ultraviolet-visible Spectrophotometry,
and compare the spectrum with the Reference Spectrum or
Containers and storage Containers—Tight containers.
the spectrum of a solution of Cloxacillin Sodium Reference
Standard prepared in the same manner as the sample solu-
tion: both spectra exhibit similar intensities of absorption at
Clomifene Citrate the same wavelength.

クエン酸クロミフェン
Add the following next to Identiˆcation:
Change the Isomer ratio to read: Optical rotation [a]20
D : +163 – +1719(1 g calculated on the
anhydrous basis, water, 100 mL, 100 mm).
Isomer ratio To 0.10 g of Clomifene Citrate add 10 mL of
water and 1 mL of sodium hydroxide TS, and extract with
Change the pH to read:
three 15-mL portions of diethyl ether. Wash the combined
diethyl ether extracts with 20 mL of water, add 10 g of anhy- pH Dissolve 1.0 g of Cloxacillin Sodium in 10 mL of
drous sodium sulfate to the combined diethyl ether extracts, water: the pH of the solution is between 6.0 and 7.5.
shake for 1 minute, ˆlter, and evaporate the diethyl ether of
the ˆltrate. Dissolve the residue in 10 mL of chloroform, and Change the Purity to read:
use this solution as the sample solution. Perform the test
Purity (1) Clarity and color of solution—A solution
with 2 mL of the sample solution as directed under the Gas
obtained by dissolving 1.0 g of Cloxacillin Sodium in 10 mL
Chromatography according to the following conditions.
of water is clear and colorless to light yellow.
Determine the areas of two adjacent peaks, Aa and Ab, hav-
(2) Heavy metals—Proceed with 1.0 g of Cloxacillin
ing retention times of about 20 minutes, where Aa is the peak
Sodium according to Method 2, and perform the test. Pre-
area of shorter retention time and Ab is the peak area of
pare the control solution with 2.0 mL of Standard Lead
longer retention time: Ab W (Aa + Ab) is between 0.3 and 0.5.
Solution (not more than 20 ppm).
Operating conditions—
(3) Arsenic—Prepare the test solution with 1.0 g of
Detector: A hydrogen ‰ame-ionization detector.
Cloxacillin Sodium according to Method 5, and perform the
Column: A column 3 mm in inside diameter and 1 m in
test (not more than 2 ppm).
length, having methylsilicone polymer coated at the ratio
(4) Related substances—Dissolve 50 mg of Cloxacillin
of 1z on siliceous earth for gas chromatography (125 to
Sodium in 50 mL of the mobile phase, and use this solution
150 mm in particle diameter).
as the sample solution. Pipet 1 mL of the sample solution,
Column temperature: A constant temperature of about
add the mobile phase to make exactly 100 mL, and use this
1959 C.
solution as the standard solution. Perform the test with ex-
Carrier gas: Nitrogen
actly 10 mL each of the sample solution and the standard so-
Flow rate: Adjust the ‰ow rate so that the retention time
lution as directed under the Liquid Chromatography accord-
of the ˆrst peak of clomifene citrate is about 20 minutes.
ing to the following conditions, and determine each peak
System suitability—
area by the automatic integration method: the area of the
System performance: When the procedure is run with 2 mL
peak other than cloxacillin obtained from the sample solu-
of the sample solution under the above operating conditions,
tion is not more than the peak area of cloxacillin obtained
the resolution between the two peaks is not less than 1.3.
from the standard solution.
System repeatability: When the test is repeated 5 times
Operating conditions—
with 2 mL of the sample solution under the above operating
Detector: An ultraviolet absorption photometer (wave-
conditions, the relative standard deviation of Ab/(Aa + Ab)
length: 230 nm).
is not more than 5.0z.
Column: A stainless steel column 6 mm in inside diameter
1442 O‹cial Monographs for Part I Supplement I, JP XIV

and 15 cm in length, packed with octadecylsilanized silica gel Q T and Q S, of the peak area of codeine to that of the inter-
for liquid chromatography (5 mm in particle diameter). nal standard.
Column temperature: A constant temperature of about
Amount (mg) of codeine phosphate
259C.
(C18H21NO3.H3PO4. 1/2 H2O)
Mobile phase: Dissolve 4.953 g of diammonium hydrogen
Q
phosphate in 700 mL of water, and add 250 mL of acetoni- = WS × T × 1.0227
QS
trile. Adjust the pH to 4.0 with phosphoric acid, and add
water to make exactly 1000 mL. WS: Amount (mg) of codeine phosphate for assay, calcu-
Flow rate: Adjust the ‰ow rate so that the retention time lated on the anhydrous basis
of cloxacillin is about 24 minutes.
Internal standard solution—A solution of etilefrine
Time span of measurement: About 3 times as long as the
hydrochloride (3 in 10,000).
retention time of cloxacillin.
Operating conditions—
System suitability—
Detector: An ultraviolet absorption photometer (wave-
Test for required detectability: Measure exactly 1 mL of
length: 280 nm).
the standard solution, and add the mobile phase to make
Column: A stainless steel column 4.6 mm in inside di-
exactly 10 mL. Conˆrm that the peak area of cloxacillin
ameter and 15 cm in length, packed with octadecylsilanized
obtained from 10 mL of this solution is equivalent to 7 to
silica gel for liquid chromatography (5 mm in particle di-
13z of that obtained from the standard solution.
ameter).
System performance: Weigh accurately about 50 mg of
Column temperature: A constant temperature of about
Cloxacillin Sodium Reference Standard, dissolve in a suita-
409C.
ble amount of the mobile phase, add 5 mL of a solution of
Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in
guaifenesin in the mobile phase (1 in 200), then add the
500 mL of diluted phosphoric acid (1 in 1000), and adjust
mobile phase to make exactly 50 mL, and use this solution as
the pH to 3.0 with sodium hydroxide TS. To 240 mL of this
the solution for system suitability test. When the procedure
solution add 70 mL of tetrahydrofuran, and mix.
is run with 10 mL of the solution for system suitability test
Flow rate: Adjust the ‰ow rate so that the retention time
under the above operating conditions, guaifenesin and clox-
of codeine is about 10 minutes.
acillin are eluted in this order with the resolution between
System suitability—
these peaks being not less than 25.
System performance: When the procedure is run with
System repeatability: When the test is repeated 6 times
20 mL of the standard solution under the above operating
with 10 mL of the solution for system suitability test under
conditions, codeine and the internal standard are eluted in
the above operating conditions, the relative standard devia-
this order with the resolution between these peaks being not
tion of the ratios of the peak area of cloxacillin to that of
less than 4.
guaifenesin is not more than 1.0z.
System repeatability: When the test is repeated 5 times
with 20 mL of the standard solution under the above operat-
Change the Water to read: ing conditions, the relative standard deviation of the ratios
Water 3.0 – 4.5z (0.2 g, volumetric titration, direct titra- of the peak area of codeine to that of the internal standard is
tion). not more than 1.0z.

1z Codeine Phosphate Powder 10z Codeine Phosphate Powder


リン酸コデイン散 1 リン酸コデイン散 10

Change the Assay to read: Change the Assay to read:


Assay Weigh accurately about 5 g of 1z Codeine Phos- Assay Weigh accurately about 2.5 g of 10z Codeine Phos-
phate Powder, dissolve in water to make exactly 100 mL, phate Powder, dissolve in water to make exactly 100 mL,
then pipet 10 mL of this solution, add exactly 10 mL of the then pipet 2 mL of this solution, add exactly 10 mL of the
internal standard solution, and use this solution as the sam- internal standard solution and water to make 20 mL, and
ple solution. Separately, weigh accurately about 50 mg of use this solution as the sample solution. Separately, weigh
codeine phosphate for assay, separately determined the accurately about 50 mg of codeine phosphate for assay,
water in the same manner as Codeine Phosphate, dissolve in separately determined the water in the same manner as
water to make exactly 100 mL, then pipet 10 mL of this solu- Codeine Phosphate, dissolve in water to make exactly
tion, add exactly 10 mL of the internal standard solution, 100 mL, then pipet 10 mL of this solution, add exactly
and use this solution as the standard solution. Perform the 10 mL of the internal standard solution, and use this solu-
test with 20 mL each of the sample solution and the standard tion as the standard solution. Perform the test with 20 mL
solution as directed under the Liquid Chromatography ac- each of the sample solution and the standard solution as
cording to the following conditions, and calculate the ratios, directed under the Liquid Chromatography according to the
Supplement I, JP XIV O‹cial Monographs for Part I 1443

following conditions, and calculate the ratios, Q T and QS, of of this solution, add exactly 10 mL of the internal standard
the peak area of codeine to that of the internal standard: solution, and use this solution as the standard solution. Per-
form the test with 20 mL each of the sample solution and the
Amount (mg) of codeine phosphate
standard solution as directed under the Liquid Chro-
(C18H21NO3.H3PO4. 1/2 H2O)
matography according to the following conditions, and cal-
Q
= WS × T × 5 × 1.0227 culate the ratios, Q T and QS, of the peak area of codeine to
QS
that of the internal standard.
WS: Amount (mg) of codeine phosphate for assay, calcu-
Amount (mg) of codeine phosphate
lated on the anhydrous basis
(C18H21NO3.H3PO4. 1/2 H2O)
Internal standard solution—A solution of etilefrine Q
= WS × T × 2 × 1.0227
hydrochloride (3 in 10,000). QS
Operating conditions—
WS: Amount (mg) of codeine phosphate for assay, calcu-
Detector: An ultraviolet absorption photometer (wave-
lated on the anhydrous basis
length: 280 nm).
Column: A stainless steel column 4.6 mm in inside di- Internal standard solution—A solution of etilefrine
ameter and 15 cm in length, packed with octadecylsilanized hydrochloride (3 in 10,000).
silica gel for liquid chromatography (5 mm in particle di- Operating conditions—
ameter). Detector: An ultraviolet absorption photometer (wave-
Column temperature: A constant temperature of about length: 280 nm).
409C. Column: A stainless steel column 4.6 mm in inside di-
Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in ameter and 15 cm in length, packed with octadecylsilanized
500 mL of diluted phosphoric acid (1 in 1000), and adjust silica gel for liquid chromatography (5 mm in particle di-
the pH to 3.0 with sodium hydroxide TS. To 240 mL of this ameter).
solution add 70 mL of tetrahydrofuran, and mix. Column temperature: A constant temperature of about
Flow rate: Adjust the ‰ow rate so that the retention time 409C.
of codeine is about 10 minutes. Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in
System suitability— 500 mL of diluted phosphoric acid (1 in 1000), and adjust
System performance: When the procedure is run with the pH to 3.0 with sodium hydroxide TS. To 240 mL of this
20 mL of the standard solution under the above operating solution add 70 mL of tetrahydrofuran, and mix.
conditions, codeine and the internal standard are eluted in Flow rate: Adjust the ‰ow rate so that the retention time
this order with the resolution between these peaks being not of codeine is about 10 minutes.
less than 4. System suitability—
System repeatability: When the test is repeated 5 times System performance: When the procedure is run with
with 20 mL of the standard solution under the above operat- 20 mL of the standard solution under the above operating
ing conditions, the relative standard deviation of the ratios conditions, codeine and the internal standard are eluted in
of the peak area of codeine to that of the internal standard is this order with the resolution between these peaks being not
not more than 1.0z. less than 4.
System repeatability: When the test is repeated 5 times
with 20 mL of the standard solution under the above operat-
Codeine Phosphate Tablets ing conditions, the relative standard deviation of the ratios
of the peak area of codeine to that of the internal standard is
リン酸コデイン錠 not more than 1.0z.

Change the Assay to read:


Assay Weigh accurately and powder not less than 20
Codeine Phosphate Tablets. Weigh accurately a portion of
the powder, equivalent to about 0.1 g of codeine phosphate
(C18H21NO3.H3PO4. 1/2 H2O), add 30 mL of water, shake,
add 20 mL of diluted dilute sulfuric acid (1 in 20), treat the
mixture with ultrasonic waves for 10 minutes, and add water
to make exactly 100 mL. Filter the solution, then pipet 5 mL
of the ˆltrate, add exactly 10 mL of the internal standard
solution and water to make 20 mL, and use this solution as
the sample solution. Separately, weigh accurately about 50
mg of codeine phosphate for assay, separately determined its
water content in the same manner as Codeine Phosphate,
dissolve in water to make exactly 100 mL, then pipet 10 mL
1444 O‹cial Monographs for Part I Supplement I, JP XIV

Add the following: dard solution (2), and the Rf value of the rest principal spot
is about 0.1. No spot is observed at the position corre-
Colistin Sulfate sponding to the spots obtained from the standard solution
(3) and the standard solution (4).
硫酸コリスチン (3) A solution of Colistin Sulfate (1 in 20) responds to
the Qualitative Test (1) for sulfate.

Optical rotation [a]20


D : -63 – -739(1.25 g, after drying,
water, 25 mL, 100 mm).

pH The pH of a solution obtained by dissolving 0.10 g of


Colistin Sulfate in 10 mL of water is between 4.0 and 6.0.

Purity (1) Sulfuric acid—Weigh accurately about 0.25 g


of previously dried Colistin Sulfate, dissolve in a suitable
1 amount of water, adjust the pH to 11 with ammonia solu-
Colistin A Sulfate C53H100N16O13.2 H SO : 1414.66
2 2 4 tion (28), and add water to make 100 mL. To this solution
1 add exactly 10 mL of 0.1 mol/L barium chloride VS and
Colistin B Sulfate C52H98N16O13.2 H2SO4: 1400.63
2 50 mL of ethanol (99.5), and titrate with 0.1 mol/L disodi-
[1264-72-8] um dihydrogen ethylenediamine tetraacetate VS until the
blue-purple color of the solution disappears (indicator:
Colistin Sulfate, when dried, contains not less than 0.5 mg of phthalein purple): the amount of sulfuric acid
16,000 units per mg. The potency of Colistin Sulfate is (SO4) is 16.0 to 18.0z.
expressed as unit calculated from the amount of
Each mL of 0.1 mol/L barium chloride VS
colistin A (C53H100N16O13: 1169.46). One unit of
= 9.606 mg of SO4
Colistin Sulfate is equivalent to 0.04 mg of colistin A
(C53H100N16O13). (2) Related substances-Dissolve 50 mg of Colistin Sul-
fate in 10 mL of water, and use this solution as the sample
Description Colistin Sulfate occurs as a white to light yel-
solution. Pipet 1 mL of the sample solution, add water to
lowish white powder.
make exactly 50 mL, and use this solution as the standard
It is freely soluble in water, and practically insoluble in
solution. Perform the test with these solutions as directed
ethanol (99.5).
under the Thin-layer Chromatography. Spot 1 mL each of
It is hygroscopic.
the sample solution and the standard solution on a plate of
Identiˆcation (1) Dissolve 20 mg of Colistin Sulfate in silica gel for thin-layer chromatography. Develop the plate
2 mL of water, add 0.5 mL of sodium hydroxide TS, then with a mixture of pyridine, 1-butanol, water and acetic acid
add 5 drops of copper (II) sulfate TS while shaking: a purple (100) (6:5:4:1) to a distance of about 10 cm, and dry the
color develops. plate at 1009 C for 30 minutes. Spray evenly ninhydrin-
(2) Dissolve 50 mg of Colistin Sulfate in 10 mL of dilut- butanol TS on the plate, and heat at 1009 C for about 20
ed hydrochloric acid (1 in 2). Transfer 1 mL of this solution minutes: the spot other than the principal spot from the sam-
in a tube for hydrolysis, seal, and heat at 1359C for 5 hours. ple solution is not more intense than the spot from the stan-
After cooling, open the tube, and evaporate the content to dard solution.
dryness until the odor of hydrochloric acid is no more
Loss on drying Not more than 6.0z (1 g, in vacuum,
perceptible. Dissolve the residue in 0.5 mL of water, and use
609C, 3 hours).
this solution as the sample solution. Separately, dissolve
20 mg each of L-leucine, L-threonine, phenylalanine and L- Residue on ignition Not more than 1.0z (1 g).
serine in 10 mL of water, and use these solutions as the stan-
Assay Perform the test according to the Cylinder-plate
dard solution (1), the standard solution (2), the standard
method as directed under the Microbial Assay for Antibiot-
solution (3) and the standard solution (4). Perform the test
ics according to the following conditions.
with these solutions as directed under the Thin-layer Chro-
(1) Test organism—Escherichia coli NIHJ
matography. Spot 1 mL each of the sample solution and the
(2) Culture medium—Dissolve 10.0 g of peptone, 30.0 g
standard solutions on a plate of silica gel for thin-layer chro-
of sodium chloride, 3.0 g of meat extract and 15.0 g of agar
matography. Develop the plate with a mixture of 1-butanol,
in 1000 mL of water, adjust the pH with sodium hydroxide
acetic acid (100), water, pyridine and ethanol (99.5)
TS so that the solution will be 6.5 to 6.6 after sterilization,
(60:15:10:6:5) to a distance of about 10 cm, and dry the plate
and use as the agar media for seed layer and for base layer.
at 1059 C for 10 minutes. Spray evenly ninhydrin TS on the
(3) Standard solutions—Weigh accurately an amount
plate, and heat at 1109 C for 5 minutes: three principal spots
of Colistin Sulfate Reference Standard, previously dried,
are obtained from the sample solution, the Rf values of two
equivalent to about 1,000,000 units, dissolve in phosphate
spots of them are the same with those of the corresponding
buŠer solution, pH 6.0 to make exactly 10 mL, and use this
spots obtained from the standard solution (1) and the stan-
solution as the standard stock solution. Keep the standard
Supplement I, JP XIV O‹cial Monographs for Part I 1445

stock solution at not exceeding 109 C, and use within 7 days. (2) A solution of Daunorubicin Hydrochloride (1 in 50)
Take exactly a suitable amount of the standard stock solu- responds to the Qualitative Test (2) for chloride.
tion before use, add phosphate buŠer solution, pH 6.0 to
Optical rotation [a]20
D : +250 – +2759(15 mg calculated on
make solutions so that each mL contains 10,000 units and
the dried basis, methanol, 10 mL, 100 mm).
2500 units, and use these solutions as the high concentration
standard solution and the low concentration standard solu- pH Dissolve 0.15 g of Daunorubicin Hydrochloride in
tion, respectively. 30 mL of water: the pH of the solution is between 4.5 and
(4) Sample solutions—Weigh accurately an amount of 6.0.
Colistin Sulfate, previously dried, equivalent to about
Purity (1) Clarity and color of solution-Dissolve 20 mg
1,000,000 units, and dissolve in phosphate buŠer solution,
of Daunorubicin Hydrochloride in 10 mL of water: the solu-
pH 6.0 to make exactly 10 mL. Take exactly a suitable
tion is clear and red.
amount of this solution, add phosphate buŠer solution, pH
(2) Heavy metals—Proceed with 1.0 g of Daunorubicin
6.0 to make solutions so that each mL contains 10,000 units
Hydrochloride according to Method 2, and perform the test.
and 2500 units, and use these solutions as the high concen-
Prepare the control solution with 2.0 mL of Standard Lead
tration sample solution and the low concentration sample
Solution (not more than 20 ppm).
solution, respectively.
(3) Related substances—Dissolve 10 mg of Daunorubi-
Containers and storage Containers—Tight containers. cin Hydrochloride in 5 mL of methanol, and use this solu-
tion as the sample solution. Pipet 3 mL of the sample solu-
tion, add methanol to make exactly 100 mL, and use this
Add the following: solution as the standard solution. Perform the test with these
solutions as directed under the Thin-layer Chromatography.
Daunorubicin Hydrochloride Spot 10 mL each of the sample solution and the standard
solution on a plate of silica gel for thin-layer chromato-
塩酸ダウノルビシン graphy. Develop the plate with a mixture of chloroform,
methanol, water and acetic acid (100) (15:5:1:1) to a distance
of about 10 cm, and air-dry the plate. Examine the spots
with the naked eye: the spot other than the principal spot
obtained from the sample solution is not more intense than
the spot from the standard solution.

Loss on drying Not more than 7.5z (0.1 g, reduced pres-


sure not exceeding 0.67 kPa, 609
C, 3 hours).

Assay Weigh accurately an amount of Daunorubicin


C27H29NO10.HCl: 563.98 Hydrochloride and Daunorubicin Hydrochloride Reference
(2S,4S )-2-Acetyl-4-(3-amino-2,3,6-trideoxy-a-L-lyxo- Standard, equivalent to about 20 mg (potency), dissolve
hexopyranosyloxy)-1,2,3,4-tetrahydro-2,5,12- each in a suitable amount of the mobile phase, add exactly
trihydroxy-7-methoxynaphthacene-6,11-dione 4 mL of the internal standard solution and the mobile phase
monohydrochloride [23541-50-6] to make 20 mL, and use these solutions as the sample solu-
tion and the standard solution, respectively. Perform the test
Daunorubicin Hydrochloride contains not less than with 5 mL each of the sample solution and the standard solu-
940 mg (potency) per mg, calculated on the dried basis. tion as directed under the Liquid Chromatography accord-
The potency of Daunorubicin Hydrochloride is ex- ing to the following conditions, and determine the ratios, QT
pressed as mass (potency) of daunorubicin hydrochlo- and QS, of the peak area of daunorubicin to that of the inter-
ride (C27H29NO10.HCl). nal standard.

Description Daunorubicin Hydrochloride occurs as a red Amount [mg (potency)] of C27H29NO10.HCl


powder. Q
= WS × T × 1000
It is soluble in water and in methanol, and slightly soluble QS
in ethanol (99.5).
WS: Amount [mg (potency)] of Daunorubicin Hydrochlo-
Identiˆcation (1) Determine the absorption spectrum of ride Reference Standard
a solution of Daunorubicin Hydrochloride in methanol (1 in
Internal standard solution—A solution of 2-naphthalenesul-
100,000) as directed under the Ultraviolet-visible Spec-
fonic acid in the mobile phase (1 in 100).
trophotometry, and compare the spectrum with the Refer-
Operating conditions—
ence Spectrum or the spectrum of a solution of Daunorubi-
Detector: An ultraviolet absorption photometer (wave-
cin Hydrochloride Reference Standard prepared in the same
length: 254 nm).
manner as the sample solution: both spectra exhibit similar
Column: A stainless steel column 4.6 mm in inside di-
intensities of absorption at the same wavelengths.
ameter and 30 cm in length, packed with octadecylsilanized
1446 O‹cial Monographs for Part I Supplement I, JP XIV

silica gel for liquid chromatography (10 mm in particle di- Reference Spectrum or the spectrum of a solution of De-
ameter). methylchlortetracycline Hydrochloride Reference Standard
Column temperature: A constant temperature of about prepared in the same manner as the sample solution: both
259C. spectra exhibit similar intensities of absorption at the same
Mobile phase: Adjust the pH of a mixture of water and wavelengths.
acetonitrile (31:19) to 2.2 with phosphoric acid. (2) Determine the infrared absorption spectrum of De-
Flow rate: Adjust the ‰ow rate so that the retention time methylchlortetracycline Hydrochloride as directed in the
of daunorubicin is about 9 minutes. potassium chloride disk method under the Infrared Spec-
System suitability— trophotometry, and compare the spectrum with the Refer-
System performance: When the procedure is run with 5 mL ence Spectrum or the spectrum of Demethylchlortetracycline
of the standard solution under the above operating condi- Hydrochloride Reference Standard: both spectra exhibit
tions, the internal standard and daunorubicin are eluted in similar intensities of absorption at the same wave numbers.
this order with the resolution between these peaks being not (3) A solution of Demethylchlortetracycline Hydrochlo-
less than 2.0. ride (1 in 100) responds to the Qualitative Test (2) for chlo-
System repeatability: When the test is repeated 6 times ride.
with 5 mL of the standard solution under the above operat-
Optical rotation [a]20
D : -248 – -2639(0.25 g calculated on
ing conditions, the relative standard deviation of the ratios
the dried basis, 0.1 mol/L hydrochloric acid TS, 25 mL,
of the peak area of daunorubicin to that of the internal stan-
100 mm).
dard is not more than 2.0z.
Containers and storage Containers—Tight containers. pH Dissolve 1.0 g of Demethylchlortetracycline Hydro-
chloride in 100 mL of water: the pH of the solution is be-
tween 2.0 and 3.0.
Add the following:
Purity (1) Heavy metals—Proceed with 1.0 g of De-
methylchlortetracycline Hydrochloride according to Method
Demethylchlortetracycline 2, and perform the test. Prepare the control solution with
Hydrochloride 2.0 mL of Standard Lead Solution (not more than 20 ppm).
(2) Arsenic—Prepare the test solution with 1.0 g of De-
塩酸デメチルクロルテトラサイクリン methylchlortetracycline Hydrochloride according to Method
4, and perform the test (not more than 2 ppm).
(3) Related substances—Dissolve 25 mg of Demethyl-
chlortetracycline Hydrochloride in 50 mL of 0.01 mol/L
hydrochloric acid TS, and use this solution as the sample so-
lution. Pipet 5 mL of the sample solution, add 0.01 mol/L
hydrochloric acid TS to make exactly 100 mL, and use this
solution as the standard solution. Perform the test with
C21H21ClN2O8.HCl: 501.31
20 mL each of the sample solution and the standard solution
(4S,4a S,5a S,6S,12a S )-7-Chloro-4-dimethylamino-
as directed under the Liquid Chromatography according
1,4,4a,5,5a,6,11,12a-octahydro-3,6,10,12,12a-
to the following conditions. Determine each peak area
pentahydroxy-1,11-dioxonaphthacene-2-carboxamide
obtained from the chromatograms of these solutions by the
monohydrochloride [64-73-3]
automatic integration method: the peak area other than
demethylchlortetracycline obtained from the sample solu-
Demethylchlortetracycline Hydrochloride contains tion is not more than 6/5 times that of demethylchlortetracy-
not less than 900 mg (potency) per mg, calculated on cline from the standard solution, and the sum of the areas of
the dried basis. The potency of Demethylchlortetra- the peaks other than demethylchlortetracycline from the
cycline Hydrochloride is expressed as mass (potency) sample solution is not more than 2 times the peak area of
of demethylchlortetracycline hydrochloride demethylchlortetracycline from the standard solution.
(C21H21ClN2O8.HCl). Operating conditions—
Description Demethylchlortetracycline Hydrochloride oc- Detector, column, column temperature, mobile phase,
curs as a yellow crystalline powder. and ‰ow rate: Proceed as directed in the operating condi-
It is soluble in water, and slightly soluble in ethanol (99.5). tions in the Assay.
Time span of measurement: About 2 times as long as the
Identiˆcation (1) Dissolve 40 mg of Demethylchlor-
retention time of demethylchlortetracycline after the solvent
tetracycline Hydrochloride in 250 mL of water. To 10 mL of
peak.
this solution add 85 mL of water and 5 mL of a solution of
System suitability—
sodium hydroxide (1 in 5). Determine the absorption spec-
Test for required detectability: Measure exactly 10 mL of
trum of this solution as directed under the Ultraviolet-visible
the standard solution, add 0.01 mol/L hydrochloric acid TS
Spectrophotometry, and compare the spectrum with the
to make exactly 50 mL, and use this solution as the solution
Supplement I, JP XIV O‹cial Monographs for Part I 1447

for system suitability test. Pipet 5 mL of the solution for sys- ative retention time of 4-epidemethylchlortetracycline with
tem suitability test, and add 0.01 mol/L hydrochloric acid respect to demethylchlortetracycline is about 0.7.
TS to make exactly 50 mL. Conˆrm that the peak area of de- System repeatability: When the test is repeated 6 times
methylchlortetracycline obtained from 20 mL of this solution with 20 mL of the standard solution under the above operat-
is equivalent to 7 to 13z of that from 20 mL of the solution ing conditions, the relative standard deviation of the peak
for system suitability test. area of demethylchlortetracycline is not more than 1.0z.
System performance: Proceed as directed in the system
Containers and storage Containers—Tight containers.
suitability in the Assay.
Storage—Light-resistant.
System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
area of demethylchlortetracycline is not more than 1.0z. Dibekacin Sulfate
Loss on drying Not more than 2.0z (1 g, in vacuum, 硫酸ジベカシン
609C, 3 hours).
Change to read except the structural formula
Residue on ignition Not more than 0.2z (1 g).
and chemical name:
Assay Weigh accurately an amount of Demethylchlor-
tetracycline Hydrochloride and Demethylchlortetracycline Dibekacin Sulfate contains not less than 640 mg
Hydrochloride Reference Standard, equivalent to about (potency) per mg, calculated on the dried basis. The
25 mg (potency), dissolve each in 0.01 mol/L hydrochloric potency of Dibekacin Sulfate is expressed as mass
acid TS to make exactly 50 mL, and use these solutions as (potency) of dibekacin (C18H37N5O8: 451.52).
the sample solution and the standard solution, respectively.
Description Dibekacin Sulfate occurs as a white to yellow-
Perform the test with exactly 20 mL each of the sample solu-
ish white powder.
tion and the standard solution as directed under the Liquid
It is very soluble in water, and practically insoluble in
Chromatography according to the following conditions, and
ethanol (99.5).
determine the peak areas, AT and AS, of demethylchlor-
tetracycline. Identiˆcation (1) Dissolve 20 mg each of Dibekacin Sul-
fate and Dibekacin Sulfate Reference Standard in 1 mL of
Amount [mg (potency)] of C21H21ClN2O8.HCl
water, and use these solutions as the sample solution and the
A
= WS × T × 1000 standard solution. Perform the test with these solutions as
AS
directed under the Thin-layer Chromatography. Spot 5 mL
WS: Amount [mg (potency)] of Demethylchlortetracycline each of the sample solution and the standard solution on a
Hydrochloride Reference Standard plate of silica gel for thin-layer chromatography. Develop
the plate with a mixture of ammonia solution (28) and
Operating conditions—
methanol (1:1) to a distance of about 10 cm, and air-dry the
Detector: An ultraviolet absorption photometer (wave-
plate. Spray evenly 0.2z ninhydrin-water saturated 1-
length: 254 nm).
butanol TS, and heat at 1009 C for 10 minutes: the principal
Column: A stainless steel column 4.1 mm in inside di-
spots obtained from the sample solution and the standard
ameter and 25 cm in length, packed with styrene-divinylben-
solution show a purple-brown color and the same R f value.
zene copolymer for liquid chromatography (10 mm in parti-
(2) To 5 mL of a solution of Dibekacin Sulfate (1 in 50)
cle diameter).
add 1 drop of barium chloride TS: a white precipitate is pro-
Column temperature: A constant temperature of about
duced.
609C.
Mobile phase: Dissolve 3.5 g of dipotassium hydrogen Optical rotation [a]20
D : +96 – +1069(0.25 g calculated on
phosphate, 1.5 g of tetrabutylammonium hydrogensulfate the dried basis, water, 25 mL, 100 mm).
and 0.4 g of disodium dihydrogen ethylenediamine tetraa-
pH The pH of a solution obtained by dissolving 1.0 g of
cetate dihydrate in 300 mL of water, and adjust the pH to
Dibekacin Sulfate in 20 mL of water is between 6.0 and 8.0.
8.5 with sodium hydroxide TS. To this solution add 75.0 g
of t-butanol and water to make 1000 mL. Purity (1) Clarity and color of solution—Dissolve 1.0 g
Flow rate: Adjust the ‰ow rate so that the retention time of Dibekacin Sulfate in 10 mL of water: the solution is clear
of demethylchlortetracycline is about 8 minutes. and colorless to pale yellow.
System suitability— (2) Heavy metals—Proceed with 1.0 g of Dibekacin Sul-
System performance: Heat 10 mL of the standard solution fate according to Method 1, and perform the test. Prepare
on a water bath for 60 minutes. When the procedure is run the control solution with 2.0 mL of Standard Lead Solution
with 20 mL of this solution so obtained under the above (not more than 20 ppm).
operating conditions, 4-epidemethylchlortetracycline and
Loss on drying Not more than 5.0z (1 g, reduced pressure
demethylchlortetracycline are eluted in this order with the
not exceeding 0.67 kPa, 609C, 3 hours).
resolution between these peaks being not less than 3. The rel-
1448 O‹cial Monographs for Part I Supplement I, JP XIV

Assay Perform the test according to the Cylinder-plate Conˆrm that the peak area of diclofenamide obtained from
method as directed under the Microbial Assay for Antibiot- 10 mL of this solution is equivalent to 3.5 to 6.5z of that of
ics according to the following conditions. diclofenamide obtained from 10 mL of the standard solu-
(1) Test organism—Bacillus subtilis ATCC 6633 tion.
(2) Culture medium—Use the medium i in 1) Medium System performance: Proceed as directed in the system
for test organism [5] under (1) Agar media for seed and base suitability in the Assay.
layer having pH 6.5 to 6.6 after sterilization. System repeatability: When the test is repeated 6 times
(3) Standard solutions-Weigh accurately an amount of with 10 mL of the standard solution under the above operat-
Dibekacin Sulfate Reference Standard, previously dried, ing conditions, the relative standard deviation of the peak
equivalent to about 20 mg (potency), dissolve in diluted area of diclofenamide is not more than 1.0z.
phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly
50 mL, and use this solution as the standard stock solution. Change the Assay to read:
Keep the standard stock solution at 5 to 159 C and use within
Assay Weigh accurately about 50 mg each of Diclofena-
30 days. Take exactly a suitable amount of the standard
mide and Diclofenamide Reference Standard, previously
stock solution before use, add 0.1 mol/L phosphate buŠer
dried, and dissolve each in 30 mL of the mobile phase. To
solution, pH 8.0 to make solutions so that each mL contains
each add exactly 10 mL of the internal standard solution and
20 mg (potency) and 5 mg (potency), and use these solutions
the mobile phase to make 50 mL, and use these solutions as
as the high concentration standard solution and the low con-
the sample solution and the standard solution. Perform the
centration standard solution, respectively.
test with 10 mL each of the sample solution and the standard
(4) Sample solutions—Weigh accurately an amount of
solution as directed under the Liquid Chromatography ac-
Dibekacin Sulfate, equivalent to about 20 mg (potency), and
cording to the following conditions, and calculate the ratios,
dissolve in water to make exactly 50 mL. Take exactly a suit-
QT and QS, of the peak area of diclofenamide to that of the
able amount of this solution, add 0.1 mol/L phosphate
internal standard, respectively.
buŠer solution, pH 8.0 to make solutions so that each mL
contains 20 mg (potency) and 5 mg (potency), and use these QT
Amount (mg) of C6H6Cl2N2O4S2 = WS ×
solutions as the high concentration sample solution and the QS
low concentration sample solution, respectively.
WS: Amount (mg) of Diclofenamide Reference Standard
Containers and storage Containers—Tight containers.
Internal standard solution—A solution of butyl parahydroxy
benzoate in the mobile phase (3 in 5000).
Operating conditions—
Diclofenamide Detector: An ultraviolet absorption photometer (wave-
length: 280 nm).
ジクロフェナミド
Column: A stainless steel column 4 mm in inside diameter
and 30 cm in length, packed with octadecylsilanized silica gel
Change the Purity (4) to read:
for liquid chromatography (10 mm in particle diameter).
Purity Column temperature: A constant temperature of about
(4) Related substances—Dissolve 0.10 g of Diclofena- 259C.
mide in 50 mL of the mobile phase, and use this solution as Mobile phase: A mixture of sodium phosphate TS and
the sample solution. Pipet 2 mL of the sample solution, add acetonitrile (1:1).
the mobile phase to make exactly 100 mL, and use this solu- Flow rate: Adjust the ‰ow rate so that the retention time
tion as the standard solution. Perform the test with exactly of diclofenamide is about 7 minutes.
10 mL each of the sample solution and the standard solution System suitability—
as directed under the Liquid Chromatography according to System performance: When the procedure is run with
the following conditions, and determine each peak area by 10 mL of the standard solution under the above operating
the automatic integration method: the total area of the peaks conditions, diclofenamide and the internal standard are elut-
other than the peak of diclofenamide from the sample solu- ed in this order with the resolution between these peaks
tion is not larger than the peak area of diclofenamide from being not less than 9.
the standard solution. System repeatability: When the test is repeated 6 times
Operating conditions— with 10 mL of the standard solution under the above operat-
Detector, column, column temperature, mobile phase, ing conditions, the relative standard deviation of the ratios
and ‰ow rate: Proceed as directed in the operating condi- of the peak area of diclofenamide to that of the internal
tions in the Assay. standard is not more than 1.0z.
Time span of measurement: About 5 times as long as the
retention time of diclofenamide.
System suitability—
Test for required detection: To exactly 5 mL of the stand-
ard solution add the mobile phase to make exactly 100 mL.
Supplement I, JP XIV O‹cial Monographs for Part I 1449

Diethylcarbamazine Citrate Tablets 1z Dihydrocodeine Phosphate


クエン酸ジエチルカルバマジン錠 Powder
リン酸ジヒドロコデイン散 1
Change the Assay to read:
Assay Weigh accurately and powder not less than 20 Change the Assay to read:
Diethylcarbamazine Citrate Tablets. Weigh accurately a
Assay Weigh accurately about 5 g of 1z Dihydrocodeine
portion of the powder, equivalent to about 50 mg of diethyl-
Phosphate Powder, dissolve in water to make exactly
carbamazine citrate (C10H21N3O.C6H8O7), add 10 mL of
100 mL, then pipet 10 mL of this solution, add exactly
water, shake well, add 5 mL of sodium hydroxide TS, then
10 mL of the internal standard solution, and use this solu-
add exactly 20 mL of the internal standard solution, and
tion as the sample solution. Separately, weigh accurately
shake vigorously for 10 minutes. Centrifuge, discard the
about 50 mg of dihydrocodeine phosphate for assay,
aqueous layer, and use the chloroform layer as the sample
separately determined its loss on drying (1059C, 4 hours),
solution. Separately, weigh accurately about 50 mg of
dissolve in water to make exactly 100 mL, then pipet 10 mL
Diethylcarbamazine Citrate Reference Standard, previously
of this solution, add exactly 10 mL of the internal standard
dried at 1059C for 4 hours, dissolve in 10 mL of water, add
solution, and use this solution as the standard solution. Per-
5 mL of sodium hydroxide TS, proceed in the same manner
form the test with 20 mL each of the sample solution and the
as the preparation of the sample solution, and use the chlo-
standard solution as directed under the Liquid Chro-
roform layer as the standard solution. Perform the test with
matography according to the following conditions, and
2 mL of the sample solution and the standard solution as
calculate the ratios, Q T and QS, of the peak area of dihydro-
directed under the Gas Chromatography according to the
codeine to that of the internal standard.
following conditions, and calculate the ratios, QT and QS, of
the peak area of diethycarbamazine to that of the internal Amount (mg) of dihydrocodeine phosphate
standard, respectively. (C18H23NO3.H3PO4)
Q
Amount (mg) of diethylcarbamazine citrate = WS × T
QS
(C10H21N3O.C6H8O7)
Q WS: Amount (mg) of dihydrocodeine phosphate for as-
= WS × T
QS say, calculated on the dried basis

WS: Amount (mg) of Diethylcarbamazine Citrate Refer- Internal standard solution—A solution of ethylefurin
ence Standard hydrochloride (3 in 10,000).
Operating conditions—
Internal standard solution—A solution of n-octadecane in
Detector: An ultraviolet absorption photometer (wave-
chloroform (1 in 1250).
length: 280 nm).
Operating conditions—
Column: A stainless steel column 4.6 mm in inside di-
Detector: A hydrogen ‰ame-ionization detector.
ameter and 15 cm in length, packed with octadecylsilanized
Column: A glass tube 3 mm in inside diameter and 1 m in
silica gel for liquid chromatography (5 mm in particle di-
length, packed with silanized siliceous earth for gas chro-
ameter).
matography (180 to 250 mm in particle diameter) coated with
Column temperature: A constant temperature of about
35z methylphenyldimethyl silicone polymer for gas chro-
409C.
matography in the ratio of 3z.
Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in
Column temperature: A constant temperature of about
500 mL of diluted phosphoric acid (1 in 1000), and adjust
1459 C.
the pH to 3.0 with sodium hydroxide TS. To 240 mL of this
Carrier gas: Nitrogen
solution add 70 mL of tetrahydrofuran.
Flow rate: Adjust the ‰ow rate so that the retention time
Flow rate: Adjust the ‰ow rate so that the retention time
of diethylcarbamazine is about 4 minutes.
of dihydrocodeine is about 9 minutes.
System suitability—
System suitability—
System performance: When the procedure is run with 2 mL
System performance: When the procedure is run with
of the standard solution under the above operating condi-
20 mL of the standard solution under the above operating
tions, diethylcarbamazine and the internal standard are elut-
conditions, dihydrocodeine and the internal standard are
ed in this order with the resolution between these peaks
eluted in this order with the resolution between these peaks
being not less than 5.
being not less than 4.
System repeatability: When the test is repeated 6 times
System repeatability: When the test is repeated 5 times
with 2 mL of the standard solution under the above operat-
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios
ing conditions, the relative standard deviation of the ratios
of the peak area of diethylcarbamazine to that of the inter-
of the peak area of dihydrocodeine to that of the internal
nal standard is not more than 1.5z.
standard is not more than 1.0z.
1450 O‹cial Monographs for Part I Supplement I, JP XIV

10z Dihydrocodeine Phosphate Dihydroergotoxine Mesilate


Powder メシル酸ジヒドロエルゴトキシン
リン酸ジヒドロコデイン散 10
Change the Assay (1) to read:
Change the Assay to read: Assay (1) Dihydroergotoxine mesilate—Weigh accurate-
ly about 30 mg each of Dihydroergotoxine Mesilate and Di-
Assay Weigh accurately about 2.5 g of 10z Dihydro-
hydroergotoxine Mesilate Reference Standard, and dissolve
codeine Phosphate Powder, dissolve in water to make ex-
them separately in a suitable amount of a mixture of water
actly 100 mL, then pipet 2 mL of this solution, add exactly
and acetonitrile (3:1). To these solutions add exactly 10 mL
10 mL of the internal standard solution and water to make
of the internal standard solution and an amount of a mixture
20 mL, and use this solution as the sample solution.
of water and acetonitrile (3:1) to make 50 mL, and use these
Separately, weigh accurately about 50 mg of dihydrocodeine
solutions as the sample solution and the standard solution.
phosphate for assay, separately determined its loss on drying
Perform the test with 20 mL of the sample solution and the
(1059 C, 4 hours), dissolve in water to make exactly 100 mL,
standard solution as directed under the Liquid Chro-
then pipet 10 mL of this solution, add exactly 10 mL of the
matography according to the following conditions, and cal-
internal standard solution, and use this solution as the stan-
culate the ratios of the peak areas of dihydroergocornine, di-
dard solution. Perform the test with 20 mL each of the sam-
hydro-a-ergocryptine, dihydroergocristine and dihydro-b-
ple solution and the standard solution as directed under the
ergocryptine to the peak area of the internal standard of
Liquid Chromatography according to the following condi-
these solutions.
tions, and calculate the ratios, Q T and QS, of the peak area
Amount (mg) of dihydroergotoxine mesilate
of dihydrocodeine to that of the internal standard.
M + MTB + MTC + MTD
= WS × TA
Amount (mg) of dihydrocodeine phosphate MSA + MSB + MSC + MSD
(C18H23NO3.H3PO4)
WS: Amount (mg) of Dihydroergotoxine Mesilate Refer-
Q
= WS × T × 5 ence Standard, calculated on the anhydrous basis
QS
MTA: Ratio of the peak area of dihydroergocornine to
WS: Amount (mg) of dihydrocodeine phosphate for as- that of the internal standard of the sample solution
say, calculated on the dried basis × 659.80
MTB: Ratio of the peak area of dihydro-a-ergocryptine to
Internal standard solution—A solution of ethylefrine
that of the internal standard of the sample × 673.83
hydrochloride (3 in 10,000).
MTC: Ratio of the peak area of dihydroergocristine to that
Operating conditions—
of the internal standard of the sample solution ×
Detector: An ultraviolet absorption photometer (wave-
707.85
length: 280 nm).
MTD: Ratio of the peak area of dihydro-b-ergocryptine to
Column: A stainless steel column 4.6 mm in inside di-
that of the internal standard of the sample solution
ameter and 15 cm in length, packed with octadecylsilanized
× 673.83
silica gel for liquid chromatography (5 mm in particle di-
MSA: Ratio of the peak area of dihydroergocornine to that
ameter).
of the internal standard of the standard solution ×
Column temperature: A constant temperature of about
659.80
409C.
MSB: Ratio of the peak area of dihydro-a-ergocryptine to
Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in
that of the internal standard of the standard solution
500 mL of diluted phosphoric acid (1 in 1000), and adjust
× 673.83
the pH to 3.0 with sodium hydroxide TS. To 240 mL of this
MSC: Ratio of the peak area of dihydroergocristine to that
solution add 70 mL of tetrahydrofuran.
of the internal standard of the standard solution ×
Flow rate: Adjust the ‰ow rate so that the retention time
707.85
of dihydrocodeine is about 9 minutes.
MSD: Ratio of the peak area of dihydro-b-ergocryptine to
System suitability—
that of the internal standard of the standard solu-
System performance: When the procedure is run with
tion × 673.83
20 mL of the standard solution under the above operating
conditions, dihydrocodeine and the internal standard are Internal standard solution—Dissolve 0.04 g of chloram-
eluted in this order with the resolution between these peaks phenicol in a mixture of water and acetonitrile (3:1) to make
being not less than 4. 250 mL.
System repeatability: When the test is repeated 5 times Operating conditions—
with 20 mL of the standard solution under the above operat- Detector: An ultraviolet absorption photometer (wave-
ing conditions, the relative standard deviation of the ratios length: 280 nm).
of the peak area of dihydrocodeine to that of the internal Column: A stainless steel column 4.6 mm in inside di-
standard is not more than 1.0z. ameter and 15 cm in length, packed with octadecylsilanized
Supplement I, JP XIV O‹cial Monographs for Part I 1451

silica gel for liquid chromatography (5 mm in particle di- Flow rate: Adjust the ‰ow rate so that the retention time
ameter). of diltiazem is about 9 minutes.
Column temperature: A constant temperature of about Time span of measurement: About twice as long as the
259C. retention time of diltiazem after the solvent peak.
Mobile phase: A mixture of water, acetonitrile and System suitability—
triethylamine (30:10:1). Test for required detection: To exactly 2 mL of the stan-
Flow rate: Adjust the ‰ow rate so that the retention time dard solution add diluted ethanol (99.5) (4 in 5) to make
of chloramphenicol is about 5 minutes. exactly 10 mL. Conˆrm that the peak area of diltiazem
System suitability— obtained from 20 mL of this solution is equivalent to 15 to
System performance: When the procedure is run with 25z of that of diltiazem obtained from 20 mL of the stan-
20 mL of the standard solution under the above operating dard solution.
conditions, the internal standard, dihydroergocornine, di- System performance: Dissolve 0.03 g of Diltiazem
hydro-a-ergocryptine, dihydroergocristine and dihydro-b- Hydrochloride, 0.02 g of d-3-hydroxy-cis-2,3-dihydro-5-[2-
ergocryptine are eluted in this order with the resolution (dimethylamino)ethyl]-2-(4-methoxyphenyl)-1, 5-benzothia-
between the peaks of dihydro-a-ergocryptine and dihydroer- zepin-4-(5H )-one hydrochloride and 0.02 g of phenylbenzo-
gocristine being not less than 1.5. ate in 160 mL of ethanol (99.5), and add water to make 200
System repeatability: When the test is repeated 6 times mL. Perform the test with 20 mL of this solution as directed
with 20 mL of the standard solution under the above operat- under the Liquid Chromatography under the above operat-
ing conditions, the relative standard deviation of the ratios ing conditions: d-3-hydroxy-cis-2,3-dihydro-5-[2-(dimethyl-
of the peak area of dihydroergocornine, dihydro-a- amino)ethyl] - 2 - (4 - methoxyphenyl) - 1, 5 - benzothiazepin-
ergocryptine, dihydroergocristine and dihydro-b-ergocryp- 4(5H )-one, diltiazem and phenyl benzoate are eluted in this
tine to that of the internal standard is not more than 0.5z. order with the resolutions between the peaks of d-3-hydroxy-
cis - 2, 3 - dihydro - 5 - [2 - (dimethylamino)ethyl] - 2 - (4 -
methoxyphenyl)-1,5-benzothiazepin-4(5H )-one and diltia-
Diltiazem Hydrochloride zem and between the peaks of diltiazem and phenyl benzoate
being not less than 2.5, respectively.
塩酸ジルチアゼム System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution under the above operat-
Change the Purity (5) to read: ing conditions, the relative standard deviation of the peak
area of diltiazem is not more than 2.0z.
Purity
(5) Related substances—Dissolve 50 mg of Diltiazem
Hydrochloride in 50 mL of diluted ethanol (99.5) (4 in 5),
and use this solution as the sample solution. Measure exactly Dipyridamole
1 mL of the sample solution, add diluted ethanol (99.5) (4 in
ジピリダモール
5) to make exactly 200 mL, and use this solution as the stan-
dard solution. Perform the test with exactly 20 mL each of
Change the Purity (4) to read:
the sample solution and the standard solution as directed un-
der the Liquid Chromatography according to the following Purity
conditions. Determine each peak area of both solutions by (4) Related substances—Dissolve 50 mg of Dipyrida-
automatic integration method: the total peak area of peaks mole in 50 mL of the mobile phase, and use this solution as
other than the peak of diltiazem obtained from the sample the sample solution. Pipet 0.5 mL of the sample solution,
solution is not more than 3/5 times the peak area of diltia- add the mobile phase to make exactly 100 mL, and use this
zem obtained from the standard solution. solution as the standard solution. Perform the test with ex-
Operating conditions— actly 20 mL each of the sample solution and the standard so-
Detector: An ultraviolet absorption photometer (wave- lution as directed under the Liquid Chromatography accord-
length: 240 nm). ing to the following conditions, and determine each peak
Column: A stainless steel column 4.6 mm in inside diame- area by the automatic integration method: the total area of
ter and 15 cm in length, packed with octadecylsilanized silica the peaks other than the peak of dipyridamole from the sam-
gel for liquid chromatography (5 mm in particle diameter). ple solution is not larger than the peak area of dipyridamole
Column temperature: A constant temperature of about from the standard solution.
509C. Operating conditions—
Mobile phase: Dissolve 8 g of sodium acetate trihydrate Detector: An ultraviolet absorption photometer (wave-
and 1.5 g of d-camphorsulfonic acid in 500 mL of water, length: 280 nm).
and ˆlter using a membrane ˆlter (0.4 mm in pore size). Add Column: A stainless steel column 4 mm in inside diameter
250 mL each of acetonitrile and methanol to the ˆltrate, and and 15 cm in length, packed with octylsilanized silica gel for
adjust the solution to a pH of 6.6 by adding sodium acetate liquid chromatography (5 mm in particle diameter).
trihydrate. Column temperature: A constant temperature of about
1452 O‹cial Monographs for Part I Supplement I, JP XIV

409C. length: 280 nm).


Mobile phase: Dissolve 0.2 g of potassium dihydrogen Column: A stainless steel column 4.0 mm in inside di-
phosphate in 200 mL of water, and add 800 mL of ameter and 15 cm in length, packed with octadecylsilanized
methanol. silica gel for liquid chromatography (7 mm in particle di-
Flow rate: Adjust the ‰ow rate so that the retention time ameter).
of dipyridamole is about 4 minutes. Column temperature: A constant temperature of about
Time span of measurement: About 5 times as long as the 259C.
retention time of dipyridamole. Mobile phase: A mixture of tartrate buŠer solution, pH
System suitability— 3.0 and methanol (7:3).
Test for required detection: To exactly 5 mL of the stan- Flow rate: Adjust the ‰ow rate so that the retention time
dard solution add the mobile phase to make exactly 25 mL. of dobutamine is about 7 minutes.
Conˆrm that the peak area of dipyridamole obtained from System suitability—
20 mL of this solution is equivalent to 15 to 25z of that of System performance: When the procedure is run with 5 mL
dipyridamole obtained from 20 mL of the standard solution. of the standard solution under the above operating condi-
System performance: Dissolve 7 mg of Dipyridamole and tions, dobutamine and internal standard are eluted in this
3 mg of terphenyl in 50 mL of methanol. When the proce- order with the resolution between these peaks being not less
dure is run with 20 mL of this solution under the above oper- than 5.
ating conditions, dipyridamole and terphenyl are eluted in System repeatability: When the test is repeated 6 times
this order with the resolution between these peaks being not with 5 mL of the standard solution under the above operat-
less than 5. ing conditions, the relative standard deviation of the ratios
System repeatability: When the test is repeated 6 times of the peak area of dobutamine to that of the internal stan-
with 20 mL of the standard solution under the above operat- dard is not more than 1.0z.
ing conditions, the relative standard deviation of the peak
area of dipyridamole is not more than 1.0z.
Dopamine Hydrochloride Injection
Dobutamine Hydrochloride 塩酸ドパミン注射液

塩酸ドブタミン Change the Assay to read:


Assay To an exact volume of Dopamine Hydrochloride
Change the pH to read:
Injection, equivalent to about 30 mg of dopamine
pH Dissolve 1.0 g of Dobutamine Hydrochloride in hydrochloride (C8H11NO2.HCl), add the mobile phase to
100 mL of water: the pH of this solution is between 4.0 and make exactly 50 mL. Pipet 2.5 mL of this solution, add
5.5. exactly 2.5 mL of the internal standard solution and the
mobile phase to make 20 mL, and use this solution as the
Change the Assay to read: sample solution. Separately, weigh accurately about 30 mg
of dopamine hydrochloride for assay, previously dried at
Assay Weigh accurately about 0.1 g each of Dobutamine
1059 C for 3 hours, dissolve in the mobile phase to make
Hydrochloride and Dobutamine Hydrochloride Reference
exactly 50 mL. Pipet 2.5 mL of this solution, add exactly
Standard, each previously dried, dissolve separately in ex-
2.5 mL of the internal standard solution and the mobile
actly 10 mL of the internal standard solution, add diluted
phase to make 20 mL, and use this solution as the standard
methanol (1 in 2) to make 50 mL, and use these solutions as
solution. Perform the test with 10 mL each of the sample
the sample solution and the standard solution respectively.
solution and the standard solution as directed under the Liq-
Perform the test with 5 mL each of the sample solution and
uid Chromatography according to the following conditions,
the standard solution as directed under the Liquid Chro-
and calculate the ratios, QT and QS, of the peak area of
matography according to the following conditions, and cal-
dopamine to that of the internal standard.
culate the ratios, QT and QS, of the peak area of dobutamine
to that of the internal standard, respectively. Amount (mg) of dopamine hydrochloride (C8H11NO2.HCl)
Q
QT = WS × T
Amount (mg) of C18H23NO3.HCl = WS × QS
QS
WS: Amount (mg) of dopamine hydrochloride for assay
WS: Amount (mg) of Dobutamine Hydrochloride Refer-
ence Standard Internal standard solution—A solution of uracil in the
mobile phase (3 in 10,000).
Internal standard solution—A solution of salicylamide in
Operating conditions—
diluted methanol (1 in 2) (1 in 125).
Detector: An ultraviolet absorption photometer (wave-
Operating conditions—
length: 280 nm).
Detector: An ultraviolet absorption photometer (wave-
Column: A stainless steel column 4.6 mm in inside di-
Supplement I, JP XIV O‹cial Monographs for Part I 1453

ameter and 25 cm in length, packed with octadecylsilanized dard: both spectra exhibit similar intensities of absorption at
silica gel for liquid chromatography (5 mm in particle di- the same wave numbers.
ameter). (3) A solution of Doxorubicin Hydrochloride (1 in 200)
Column temperature: A constant temperature of about responds to the Qualitative Test (1) for chloride.
259C.
Optical rotation [a]20
D : +240 – +2909(20 mg calculated on
Mobile phase: Disodium hydrogen phosphate-citric acid
the anhydrous basis, methanol, 20 mL, 100 mm).
buŠer solution, pH 3.0
Flow rate: Adjust the ‰ow rate so that the retention time pH The pH of a solution obtained by dissolving 50 mg of
of dopamine is about 10 minutes. Doxorubicin Hydrochloride in 10 mL of water is between
System suitability— 4.0 and 5.5.
System performance: When the procedure is run with
Purity (1) Clarity and color of solution—Dissolve 50 mg
10 mL of the standard solution under the above operating
of Doxorubicin Hydrochloride in 10 mL of water: the solu-
conditions, the internal standard and dopamine are eluted in
tion is clear and red.
this order with the resolution between these peaks being not
(2) Related substances—Dissolve 25 mg of Doxorubicin
less than 10.
Hydrochloride in 100 mL of the mobile phase, and use this
System repeatability: When the test is repeated 6 times
solution as the sample solution. Pipet 2 mL of the sample
with 10 mL of the standard solution under the above operat-
solution, add the mobile phase to make exactly 100 mL, and
ing conditions, the relative standard deviation of the ratios
use this solution as the standard solution. Perform the test
of peak area of dopamine to that of the internal standard is
with exactly 20 mL each of the sample solution and the stan-
not more than 1.0z.
dard solution as directed under the Liquid Chromatography
according to the following conditions, and determine each
Change the Containers and storage to read:
peak area by the automatic integration method: the area of
Containers and storage Containers—Hermetic containers. the peak other than doxorubicin obtained from the sample
Plastic containers for aqueous injections may be used. solution is not more than 1/4 times the peak area of dox-
orubicin from the standard solution, and the total area of
the peaks other than doxorubicin is not more than the peak
Doxorubicin Hydrochloride area of doxorubicin from the standard solution.
Operating conditions—
塩酸ドキソルビシン Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Change to read except the structural formula Column: A stainless steel column 4.6 mm in inside di-
and chemical name: ameter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Doxorubicin Hydrochloride contains not less than ameter).
980 mg (potency) per mg, calculated on the anhydrous Column temperature: A constant temperature of about
basis. The potency of Doxorubicin Hydrochloride 259C.
is expressed as mass (potency) of doxorubicin Mobile phase: Dissolve 3 g of sodium lauryl sulfate in
hydrochloride (C27H29NO11. HCl). 1000 mL of diluted phosphoric acid (7 in 5000), and add
1000 mL of acetonitrile.
Description Doxorubicin Hydrochloride occurs as a red-
Flow rate: Adjust the ‰ow rate so that the retention time
orange crystalline powder.
of doxorubicin is about 8 minutes.
It is sparingly soluble in water, slightly soluble in
Time span of measurement: About 3 times as long as the
methanol, very slightly soluble in ethanol (99.5), and practi-
retention time of doxorubicin.
cally insoluble in acetonitrile.
System suitability—
Identiˆcation (1) Determine the absorption spectrum of Test for required detectability: Measure 1 mL of the stan-
a solution of Doxorubicin Hydrochloride in methanol (1 in dard solution, and add the mobile phase to make exactly
100,000) as directed under the Ultraviolet-visible Spec- 20 mL. Conˆrm that the peak area of doxorubicin obtained
trophotometry, and compare the spectrum with the Refer- from 20 mL of this solution is equivalent to 3.5 to 6.5z of
ence Spectrum or the spectrum of a solution of Doxorubicin that from 20 mL of the standard solution.
Hydrochloride Reference Standard prepared in the same System performance: Dissolve 5 mg of Doxorubicin
manner as the sample solution: both spectra exhibit similar Hydrochloride in 20 mL of water, add 1.5 mL of phosphoric
intensities of absorption at the same wavelengths. acid, and allow to stand at room temperature for 30
(2) Determine the infrared absorption spectrum of Dox- minutes. Adjust the pH of this solution to 2.5 with 2 mol/L
orubicin Hydrochloride as directed in the potassium chloride sodium hydroxide TS. When the procedure is run with 20 mL
disk method under the Infrared Spectrophotometry, and of this solution under the above operating conditions, dox-
compare the spectrum with the Reference Spectrum or the orubicinone, having the relative retention time of about 0.6
spectrum of Doxorubicin Hydrochloride Reference Stan- with respect to doxorubicin, and doxorubicin are eluted in
1454 O‹cial Monographs for Part I Supplement I, JP XIV

this order with the resolution between these peaks being not Optical rotation [a]20
D : -105 – -1209 (0.25 g calculated
less than 5. on the anhydrous basis and correcred by the amount of
System repeatability: When the test is repeated 6 times ethanol, 0.01 mol/L hydrochloric acid-methanol TS, 25 mL,
with 20 mL of the standard solution under the above operat- 100 mm). Determine within 5 minutes after the sample solu-
ing conditions, the relative standard deviation of the peak tion is prepared.
area of doxorubicin is not more than 2.0z.
Purity (1) Heavy metals—Proceed with 1.0 g of Doxycy-
Water Not more than 3.0z (0.3 g, volumetric titration, cline Hydrochloride according to Method 2, and perform
direct titration). the test. Prepare the control solution with 5.0 mL of Stan-
dard Lead Solution (not more than 50 ppm).
Assay Weigh accurately an amount of Doxorubicin
(2) Related substance—Dissolve 20 mg of Doxycycline
Hydrochloride and Doxorubicin Hydrochloride Reference
Hydrochloride in 0.01 mol/L hydrochloric acid TS to make
Standard, equivalent to about 10 mg (potency), dissolve
exactly 25 mL, and use this solution as the sample solution.
each in water to make exactly 25 mL. Pipet 5 mL each of
Separately, dissolve 20 mg of 6-epidoxycycline hydrochlo-
these solutions, add water to make exactly 100 mL, and use
ride in 0.01 mol/L hydrochloric acid TS to make exactly
these solutions as the sample solution and the standard solu-
25 mL, and use this solution as 6-epidoxycycline hydrochlo-
tion, respectively. Determine the absorbances at 495 nm, AT
ride stock solution. Separately, dissolve 20 mg of metacy-
and AS, of the sample solution and the standard solution as
cline hydrochloride in 0.01 mol/L hydrochloric acid TS to
directed under the Ultraviolet-visible Spectrophotometry.
make exactly 25 mL, and use this solution as metacycline
Amount [mg (potency)] of C27H29NO11.HCl hydrochloride stock solution. Pipet 2 mL each of 6-epidoxy-
A cycline hydrochloride stock solution and metacycline
= WS × T × 1000
AS hydrochloride stock solution, add 0.01 mol/L hydrochloric
acid TS to make exatly 100 mL, and use this solution as the
WS: Amount [mg (potency)] of Doxorubicin Hydrochlo-
standard solution. Perform the test with exactly 20 mL each
ride Reference Standard
of the sample solution and the standard solution as directed
Containers and storage Containers—Tight containers. under the Liquid Chromatography according to the follow-
ing conditions, and determine each peak area by the auto-
matic integration method: the peak areas of metacycline and
Doxycycline Hydrochloride 6-epidoxycycline obtained from the sample solution are not
more than the peak areas of them obtained from the stan-
塩酸ドキシサイクリン dard solution, respectively, and the areas of the two peaks,
appeared between the solvent peak and metacycline and
Change to read except the structural formula behind of doxycycline, obtained from the sample solution
and chemical name: are not more than 1/4 of the peak area of 6-epidoxycycline
from the standard solution.
Doxycycline Hydrochloride contains not less than Operating conditions—
880 mg (potency) per mg, calculated on the anhydrous Detector: An ultraviolet absorption photometer (wave-
basis and corrected by the amount of ethanol. The length: 254 nm).
potency of Doxycycline Hydrochloride is expressed as Column: A stainless steel column 4.6 mm in inside di-
mass (potency) of doxycycline (C22H24N2O8: 444.43). ameter and 25 cm in length, packed with styrene-divinylben-
zene copolymer for liquid chromatography (8 mm in particle
Description Doxycycline Hydrochloride occurs as yellow
diameter).
to dark yellow, crystals or crystalline powder.
Column temperature: A constant temperature of about
It is freely soluble in water and in methanol, and slightly
609C.
soluble in ethanol (99.5).
Mobile phase: Mix 125 mL of 0.2 mol/L potassium
Identiˆcation (1) Determine the infrared absorption dihydrogen phosphate TS, 117 mL of 0.2 mol/L sodium
spectrum of Doxycycline Hydrochloride as directed in the hydroxide TS, and add water to make 500 mL. To 400 mL
potassium bromide disk method under the Infrared Spec- of this solution add 50 mL of a solution of tetrabutylammo-
trophotometry, and compare the spectrum with the Refer- nium hydrogensulfate (1 in 100), 10 mL of a solution of dis-
ence Spectrum or the spectrum of Doxycycline Hydrochlo- odium dihydrogen ethylenediamine tetraacetate dihydrate (1
ride Reference Standard: both spectra exhibit similar intensi- in 25), 60 g of t-butanol and 200 mL of water, adjust the pH
ties of absorption at the same wave numbers. to 8.0 with 2 mol/L sodium hydroxide TS, and add water to
(2) Dissolve 10 mg of Doxycycline Hydrochloride in make 1000 mL.
10 mL of water, and add silver nitrate TS: a white turbidity Flow rate: Adjust the ‰ow rate so that the retention time
is produced. of doxycycline is about 19 minutes.
Time span of measurement: About 2.4 times as long as the
Absorbance E 11zcm (349 nm): 285 – 315 (10 mg, 0.01 mol/L
retention time of doxycycline after the solvent peak.
hydrochloric acid-methanol TS, 500 mL).
Supplement I, JP XIV O‹cial Monographs for Part I 1455

System suitability— System suitability—


Test for required detectability: Measure exactly 1 mL of System performance: When the procedure is run with 1 mL
the standard solution, and add 0.01 mol/L hydrochloric of the standard solution under the above operating condi-
acid TS to make exactly 20 mL. Conˆrm that the peak areas tions, ethanol and the internal standard are eluted in this
of 6-epidoxycycline and metacycline obtained from 20 mL of order with the resolution between these peaks being not less
this solution are equivalent to 3.5 to 6.5z of them obtained than 2.0.
from 20 mL of the standard solution, respectively. System repeatability: When the test is repeated 5 times
System performance: To 8 mL of the sample solution, with 1 mL of the standard solution under the above operat-
3 mL of 6-epidoxycycline hydrochloride stock solution and ing conditions, the relative standard deviation of the ratios
2 mL of metacycline hydrochloride stock solution add of the peak area of ethanol to that of the internal standard is
0.01 mol/L hydrochloric acid TS to make 50 mL. When the not more than 2.0z.
procedure is run with 20 mL of this solution under the above
Water Not less than 1.4z and not more than 2.8z (0.6 g,
operating conditions, metacycline, 6-epidoxycycline and
volumetric titration, direct titration).
doxycycline are eluted in this order with the resolutions
between the peaks, metacycline and 6-epidoxycycline, and 6- Residue on ignition Not more than 0.3z (1 g).
epidoxycycline and doxycycline, being not less than 1.3 and
Assay Weigh accurately an amount of Doxycycline
not less than 2.0, respectively, and the symmetry coe‹cient
Hydrochloride and Doxycycline Hydrochloride Reference
of the peak of doxycycline is not more than 1.3.
Standard, equivalent to about 50 mg (potency), dissolve
System repeatability: When the test is repeated 6 times
each in water to make exactly 50 mL, and use these solutions
with 20 mL of the standard solution under the above operat-
as the sample solution and the standard solution. Perform
ing conditions, the relative standard deviations of the peak
the test with exactly 10 mL each of the sample solution and
area of metacycline and 6-epidoxycycline are not more than
the standard solution as directed under the Liquid Chro-
3.0z and not more than 2.0z, respectively.
matography according to the following conditions, and
Ethanol Weigh accurately about 0.1 g of Doxycycline determine the peak areas, AT and AS, of doxycycline.
Hydrochloride, dissolve in the internal standard solution to
Amount [mg (potency)] of doxycycline (C22H24N2O8)
make exactly 10 mL, and use this solution as the sample
A
solution. Separately, weigh accurately about 0.4 g of ethanol = WS × T × 1000
AS
(99.5), and add the internal standard solution to make
exactly 100 mL. Pipet 1 mL of this solution, add the internal WS: Amount [mg (potency)] of Doxycycline Hydrochlo-
standard solution to make exactly 10 mL, and use this solu- ride Reference Standard
tion as the standard solution. Perform the test with 1 mL
Operating conditions—
each of the sample solution and the standard solution as
Detector: An ultraviolet absorption photometer (wave-
directed under the Gas Chromatography according to the
length: 280 nm).
following conditions, and determine the ratios, QT and QS,
Column: A stainless steel column 3.9 mm in inside di-
of the peak area of ethanol to that of the internal standard:
ameter and 30 cm in length, packed with octadecylsilanized
the amount of ethanol is not less than 4.3z and not more
silica gel for liquid chromatography (10 mm in particle di-
than 6.0z.
ameter).
W Q
Amount (z) of ethanol = S × T Column temperature: A constant temperature of about
WT QS
309C.
WS: Amount (mg) of ethanol (99.5) Mobile phase: Dissolve 7.0 g of sodium dihydrogen phos-
WT: Amount (mg) of the sample phate dihydrate in 450 mL of water, add 553 mL of a mix-
ture of methanol and N, N-dimethyl-n-octylamine (550:3),
Internal standard solution—A solution of 1-propanol (1 in
and adjust the pH to 8.0 with a solution of sodium
2000).
hydroxide (43 in 200).
Operating conditions—
Flow rate: Adjust the ‰ow rate so that the retention time
Detector: An hydrogen ‰ame-ionization detector.
of doxycycline is about 6 minutes.
Column: A glass column 3.2 mm in inside diameter
System suitability—
and 1.5 m in length, packed with porous ethylvinylbenzene-
System performance: When the procedure is run with
divinylbenzene copolymer for gas chromatography
10 mL of the standard solution under the above operating
(0.0075 mm in average pore size, 500 – 600 m2/g in speciˆc
conditions, the theoretical plates and the symmetrical
surface area) (150 – 180 mm in particle diameter).
coe‹cient of the peak of doxycycline are not less than 1000
Column temperature: A constant temperature of about
and not more than 2.0, respectively.
1359 C.
System repeatability: When the test is repeated 6 times
Carrier gas: Nitrogen
with 10 mL of the standard solution under the above operat-
Flow rate: Adjust the ‰ow rate so that the retention time
ing conditions, the relative standard deviation of the peak
of ethanol is about 5 minutes.
area of doxycycline is not more than 1.0z.
1456 O‹cial Monographs for Part I Supplement I, JP XIV

Containers and storage Containers—Tight containers. En‰urane


Storage—Light-resistant.
エンフルラン

Droperidol Change the Purity (4) to read:

ドロペリドール Purity
(4) Related substances—Proceed the test with 5 mL of
En‰urane as directed under the Gas chromatography ac-
Change the Description to read:
cording to the following conditions. Determine each peak
Description Droperidol occurs as a white to light yellow area other than the peak of air which appears soon after in-
powder. jection of the sample by the automatic integration method,
It is freely soluble in acetic acid (100), soluble in and calculate the amount of each peak by the area percen-
dichloromethane, slightly soluble in ethanol (99.5), and tage method: the amount of the substances other than en‰u-
practically insoluble in water. rane is not more than 0.10z.
It is gradually colored by light. Operating conditions—
Detector: A thermal conductivity detector.
Change the Identiˆcation to read: Column: A column 3 mm in inside diameter and 3 m in
Identiˆcation (1) Put 30 mg of Droperidol in a brown length, packed with siliceous earth for gas chromatography,
volumetric ‰ask, and dissolve in 10 mL of 0.1 mol/L 180 to 250 mm in particle diameter, coated with diethylene
hydrochloric acid TS and ethanol (95) to make 100 mL. glycol succinate ester for gas chromatography in the ratio of
Transfer 5 mL of the solution to a brown volumetric ‰ask, 20z.
and add 10 mL of 0.1 mol/L hydrochloric acid TS and Column temperature: A constant temperature of about
ethanol (95) to make 100 mL. Determine the absorption 809C.
spectrum of the solution as directed under the Ultraviolet- Carrier gas: Helium
visible Spectrophotometry, and compare the spectrum with Flow rate: Adjust the ‰ow rate so that the retention time
the Reference Spectrum: both spectra exhibit similar intensi- of en‰urane is about 3 minutes.
ties of absorption at the same wavelengths. Time span of measurement: About three times as long as
(2) Determine the infrared absorption spectrum of the retention time of en‰urane.
Droperidol, previously dried, as directed in the potassium System suitability—
bromide disk method under the Infrared Spectrophotomet- Test for required detection: Pipet exactly 1 mL of en‰u-
ry, and compare the spectrum with the Reference Spectrum: rane add 2-propanol to make exactly 100 mL. To exactly
both spectra exhibit similar intensities of absorption at the 2 mL of this solution add 2-propanol to make exactly
same wave numbers. If any diŠerence appears between the 10 mL, and use this solution as the solution for system
spectra, dissolve Droperidol in acetone, evaporate the ace- suitability test. Pipet 1 mL of the solution, and add 2-
tone, dry the residue in a desiccator (in vacuum, silica gel, propanol to make exactly 10 mL. Conˆrm that the peak area
709C) for 4 hours, and perform the test with the residue. of en‰urane obtained from 5 mL of this solution is equiva-
lent to 7 to 13z of that of en‰urane obtained from 5 mL of
Delete the Melting point. the solution for system suitability test.
System performance: Mix 5 mL of En‰urane and 5 mL of
2-propanol. When the procedure is run with 5 mL of this
mixture under the above operating conditions, en‰urane and
Delete the following Monographs: 2-propanol are eluted in this order with the resolution be-
tween these peaks being not less than 2.0.
System repeatability: When the test is repeated 6 times
Drostanolone Propionate with 5 mL of the solution for system suitability test under the
プロピオン酸ドロスタノロン above operating conditions, the relative standard deviation
of the peak area of en‰urane is not more than 2.0z.
Drostanolone Propionate Injection
プロピオン酸ドロスタノロン注射液
Supplement I, JP XIV O‹cial Monographs for Part I 1457

Add the following: 20) add 1 drop of barium chloride TS: a white precipitate is
produced.
Enviomycin Sulfate Optical rotation [a]20
D : -16 – -229(0.5 g calculated on the
dried basis, water, 50 mL, 100 mm).
硫酸エンビオマイシン
pH The pH of a solution obtained by dissolving 2.0 g of
Enviomycin Sulfate in 20 mL of water is between 5.5 and
7.5.

Content ratio of the active principle Dissolve 0.1 g of En-


viomycin Sulfate in water to make 100 mL, and use this solu-
tion as the sample solution. Perform the test with 3 mL of the
sample solution as directed under the Liquid Chro-
matography according to the following conditions, and de-
termine the peak areas, AT1 and AT2, of tuberactinomycin N
and tuberactinomycin O, having the relative retention time,
1.4 ± 0.4, with respect to tuberactinomycin N, by the auto-
matic integration method: AT2/(AT1 + AT2) is between 0.090
and 0.150.
Tuberactinomycin N Sulfate Operating conditions—
1 Detector: An ultraviolet absorption photometer (wave-
C25H43N13O10.1 H2SO4: 832.81
2 length: 254 nm).
Tuberactinomycin O Sulfate Column: A stainless steel column 4.6 mm in inside di-
1 ameter and 15 cm in length, packed with silica gel for liquid
C25H43N13O9.1 H2SO4: 816.81
2 chromatography (5 mm in particle diameter).
Tuberactinomycin N Sulfate Column temperature: A constant temperature of about
(3R,4R )-N-[(3S,9S,12S,15S )-9,12-Bis(hydroxymethyl)-3- 259C.
[(4R )-2-iminohexahydropyrimidin-4-yl]-2,5,8,11,14- Mobile phase: A mixture of ammonium acetate TS, 1,4-
pentaoxo-6-(Z )-ureidomethylene-1,4,7,10,13- dioxane, tetrahydrofuran, water and ammonia solution (28)
pentaazacyclohexadec-15-yl]-3,6-diamino-4- (100:75:50:23:2).
hydroxyhexanamide sesquisulfate Flow rate: Adjust the ‰ow rate so that the retention time
[33103-22-9, Tuberactinomycin N] of tuberactinomycin N is about 9 minutes.
Tuberactinomycin O Sulfate System suitability—
(S )-N-[(3S,9S,12S,15S )-9,12-Bis(hydroxymethyl)-3-[(4R )- System performance: When the procedure is run with 3 mL
2-iminohexahydropyrimidin-4-yl]-2,5,8,11,14-pentaoxo- of the sample solution under the above operating conditions,
6-(Z )-ureidomethylene-1,4,7,10,13-pentaazacyclohexadec- tuberactinomycin N and tuberactinomycin O are eluted in
15-yl]-3,6-diaminohexanamide sesquisulfate this order with the resolution between these peaks being not
[33137-73-4, Tuberactinomycin O] less than 1.5.
System repeatability: When the test is repeated 6 times
Enviomycin Sulfate contains not less than 770 mg with 3 mL of the sample solution under the above operating
conditions, the relative standard deviation of the peak area
(potency) per mg, calculated on the dried basis. The
potency of Enviomycin Sulfate is expressed as mass of tuberactinomycin N is not more than 2.0z.
(potency) of tuberactinomycin N (C25H43N13O10: Purity (1) Clarity and color of solution—Dissolve 1.0 g
685.69). of Enviomycin Sulfate in 10 mL of water: the solution is
Description Enviomycin Sulfate occurs as a white powder. clear and colorless.
It is very soluble in water, and practically insoluble in (2) Heavy metals—Proceed with 2.0 g of Enviomycin
ethanol (99.5). Sulfate according to Method 1, and perform the test. Pre-
pare the control solution with 2.0 mL of Standard Lead
Identiˆcation (1) To 5 mL of a solution of Enviomycin Solution (not more than 10 ppm).
Sulfate (1 in 200) add 1.5 mL of sodium hydroxide TS, and (3) Arsenic—Prepare the test solution with 2.0 g of
add 1 drop of a mixture of 0.01 mol/L citric acid TS and Enviomycin Sulfate according to Method 1, and perform the
copper (II) sulfate TS (97:3) : a blue-purple color develops. test (not more than 1 ppm).
(2) Determine the absorption spectrum of a solution of
Enviomycin Sulfate (1 in 100,000) as directed under the Loss on drying Not more than 4.0z (0.2 g, in vacuum,
Ultraviolet-visible Spectrophotometry, and compare the phosphorus (V) oxide, 609
C, 3 hours).
spectrum with the Reference Spectrum: both spectra exhibit Assay Perform the test according to the Cylinder-plate
similar intensities of absorption at the same wavelengths. method as directed under the Microbial Assay for Antibiot-
(3) To 2 mL of a solution of Enviomycin Sulfate (1 in ics according to the following conditions.
1458 O‹cial Monographs for Part I Supplement I, JP XIV

(1) Test organism—Bacillius subtilis ATCC 6633 ethanol (95), and practically insoluble in acetonitrile.
(2) Culture medium—Use the medium i in 1) Medium It is hygroscopic.
for test organism [5] under (1) Agar media for seed and base
Identiˆcation (1) Determine the absorption spectrum of
layer.
a solution of Epirubicin Hydrochloride in methanol (3 in
(3) Standard solutions—Weigh accurately an amount of
200,000) as directed under the Ultraviolet-visible Spec-
Enviomycin Sulfate Reference Standard, equivalent to
trophotometry, and compare the spectrum with the Refer-
about 20 mg (potency), dissolve in water to make exactly
ence Spectrum: both spectra exhibit similar intensities of
20 mL, and use this solution as the standard stock solution.
absorption at the same wavelengths.
Keep the standard stock solution at a temperature not ex-
(2) Determine the infrared absorption spectrum of
ceeding 59 C and use within 10 days. Take exactly a suitable
Epirubicin Hydrochloride and Epirubicin Hydrochloride
amount of the standard stock solution before use, add
Reference Standard as directed in the potassium bromide
0.1 mol/L phosphate buŠer solution, pH 8.0 to make solu-
disk method under the Infrared Spectrophotometry, and
tions so that each mL contains 400 mg (potency) and 100 mg
compare these spectra: both spectra exhibit similar intensi-
(potency), and use these solutions as the high concentration
ties of absorption at the same wave numbers.
standard solution and the low concentration standard solu-
tion, respectively. Optical rotation [a]20
D : +310 – +3409(10 mg calculated on
(4) Sample solutions—Weigh accurately an amount of the anhydrous basis and collected by the amount of the
Enviomycin Sulfate, equivalent to about 20 mg (potency), residual solvent, methanol, 20 mL, 100 mm).
and dissolve in water to make exactly 20 mL. Take exactly a
pH Dissolve 10 mg of Epirubicin Hydrochloride in 2 mL
suitable amount of this solution, add 0.1 mol/L phosphate
of water: the pH of the solution is between 4.0 and 5.5.
buŠer solution, pH 8.0 to make solutions so that each mL
contains 400 mg (potency) and 100 mg (potency), and use Purity (1) Clarity and color of solution-Dissolve 50 mg
these solutions as the high concentration sample solution of Epirubicin Hydrochloride in 5 mL of water: the solution
and the low concentration sample solution, respectively. is clear and dark red.
(2) Heavy metals—Proceed with 1.0 g of Epirubicin
Containers and storage Containers—Tight containers.
Hydrochloride according to Method 2, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead
Solution (not more than 20 ppm).
Add the following:
(3) Related substances—Perform the test with 10 mL of
the sample solution obtained in the Assay as directed under
Epirubicin Hydrochloride the Liquid Chromatography according to the following con-
ditions, determine each peak area by the automatic integra-
塩酸エピルビシン
tion method, and calculate the sum amount of the peaks
other than epirubicin and 2-naphthalenesulfonic acid by the
area percentage method: not more than 5.0z.
Operating conditions—
Detector, column, column temperature, mobile phase,
and ‰ow rate: Proceed as directed in the operating condi-
tions in the Assay.
Time span of measurement: About 3 times as long as the
retention time of epirubicin after the solvent peak.
System suitability—
C27H29NO11.HCl: 579.98 Test for required detectability: To 1 mL of the sample
(2S,4S )-4-(3-Amino-2,3,6-trideoxy-a-L-arabino- solution add the mobile phase to make 100 mL, and use this
hexopyranosyloxy)-1,2,3,4-tetrahydro-2,5,12-trihydroxy- solution as the solution for system suitability test. Pipet
2-hydroxyacetylnaphthacene-6,11-dione 1 mL of the solution for system suitability test, and add the
monohydrochloride [56390-09-1] mobile phase to make exactly 10 mL. Conˆrm that the peak
area of epirubicin obtained from 10 mL of this solution is
Epirubicin Hydrochloride contains not less than equivalent to 7 to 13z of that obtained from 10 mL of the
900 mg (potency) per mg, calculated on the anhydrous solution for system suitability test.
basis and corrected by the amount of the residual sol- System performance, and system repeatability: Proceed as
vent. The potency of Epirubicin Hydrochloride is ex- directed in the system suitability in the Assay.
pressed as mass (potency) of epirubicin hydrochloride (4) Residual solvents—Weigh accurately about 0.3 g of
(C27H29NO11.HCl). Epirubicin Hydrochloride, add exactly 0.6 mL of the inter-
Description Epirubicin Hydrochloride occurs as a pale yel- nal standard solution, add N, N-dimethylformamide to
lowish red to brownish red powder. make 6 mL, and use this solution as the sample solution.
It is soluble in water and in methanol, slightly soluble in Separately, pipet 1 mL of methanol, add N, N-dimethylfor-
mamide to make exactly 25 mL, and use this solution as
Supplement I, JP XIV O‹cial Monographs for Part I 1459

methanol standard stock solution. Take exactly 125 mL of Water Not more than 8.0z (0.1 g, volumetric titration,
acetone, 30 mL of ethanol (99.5), 32 mL of 1-propanol and direct titration).
17 mL of the methanol standard stock solution, add exactly
Residue on ignition Not more than 0.5z (0.1 g).
10 mL of the internal standard solution and N, N-dimethyl-
formamide to make 100 mL, and use this solution as the Assay Weigh accurately an amount of Epirubicin Hydro-
standard solution. Perform the test with 1 mL each of the chloride and Epirubicin Hydrochloride Reference Standard,
sample solution and the standard solution as directed under equivalent to about 50 mg (potency), dissolve each in the
the Gas Chromatography according to the following condi- internal standard solution to make exactly 50 mL, and use
tion, and determine the ratios of the peak areas of acetone, these solutions as the sample solution and the standard solu-
ethanol, 1-propanol and methanol to that of the internal tion, respectively. Perform the test with 10 mL each of the
standard, QTA and QSA, QTB and QSB, QTC and QSC, and QTD sample solution and the standard solution as directed under
and QSD, respectively. Calculate the amounts of acetone, the Liquid Chromatography according to the following con-
ethanol, 1-propanol and methanol by the following equa- ditions, and determine the ratios, QT and QS, of the peak
tions: the amounts of acetone, ethanol, 1-propanol and area of epirubicin to that of the internal standard.
methanol are not more than 1.5z, not more than 0.5z, not
Amount [mg (potency)] of C27H29NO11.HCl
more than 0.5z and not more than 0.1z, respectively.
Q
= WS × T × 1000
1 Q QS
Amount (z) of acetone = × TA × 593
WT QSA
WS: Amount [mg (potency)] of Epirubicin Hydrochloride
1 Q
Amount (z) of ethanol = × TB × 142 Reference Standard
WT QSB
1 Q Internal standard solution—A solution of sodium 2-
Amount (z) of 1-propanol = × TC × 154
WT QSC naphthalene sulfonate in a mixture of water, acetonitrile,
1 Q methanol and phosphoric acid (540:290:170:1) (1 in 2000).
Amount (z) of methanol = × TD × 2.23
WT QSD Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
WT: Amount (mg) of Epirubicin Hydrochloride
length: 254 nm).
Internal standard solution—A solution of 1,4-dioxane in Column: A stainless steel column 4 mm in inside diameter
N, N-dimethylformamide (1 in 100). and 25 cm in length, packed with trimethylsilanized silica gel
Operating conditions— for liquid chromatography (6 mm in particle diameter).
Detector: Hydrogen ‰ame-ionization detector. Column temperature: A constant temperature of about
Column: A fused silica column 0.53 mm in inside di- 359C.
ameter and 30 m in length, coated with polyethylene glycol Mobile phase: Dissolve 2 g of sodium lauryl sulfate in a
for gas-chromatography 1 mm in thickness. mixture of water, acetonitrile, methanol and phosphoric
Column temperature: 409C for 11 minutes after injection acid (540:290:170:1) to make 1000 mL.
of the sample, then rise to 909C at a rate of 109
C per minute. Flow rate: Adjust the ‰ow rate so that the retention time
If necessary, rise to 1309C at a rate of 509 C per minute and of epirubicin is about 9.5 minutes.
maintain the temperature for 30 minutes. System suitability—
Injection port temperature: A constant temperature of System performance: When the procedure is run with
about 1209 C. 10 mL of the standard solution under the above operating
Detector temperature: A constant temperature of about conditions, the internal standard and epirubicin are eluted in
1509 C. this order with the resolution between these peaks being not
Carrier gas: Herium less than 20.
Flow rate: Adjust the ‰ow rate so that the retention time System repeatability: When the test is repeated 5 times
of the internal standard is about 8 minutes. with 10 mL of the standard solution under the above operat-
Split ratio: 1:15 ing conditions, the relative standard deviation of the ratios
System suitability— of the peak area of epirubicin to that of the internal standard
System performance: When the procedure is run with 1 mL is not more than 1.0z.
of the standard solution under the above operating condi-
Containers and storage Containers—Tight containers.
tions, acetone, methanol, ethanol, 1-propanol and the inter-
Storage—At a temperature between 09
C and 59C.
nal standard are eluted in this order with the resolution be-
tween the peaks of acetone and the internal standard being
not less than 30.
System repeatability: When the test is repeated 6 times
with 1 mL of the standard solution under the above operat-
ing conditions, the relative standard deviations of the peak
areas of acetone, methanol, ethanol and 1-propanol are not
more than 4.0z, respectively.
1460 O‹cial Monographs for Part I Supplement I, JP XIV

Ergocalciferol ing conditions, the relative standard deviation of the ratios


of the peak area of ergocalciferol to that of the internal stan-
エルゴカルシフェロール dard is not more than 1.0z.

Change the Assay to read:


Assay Weigh accurately about 30 mg each of Ergocal-
Erythromycin
ciferol and Ergocalciferol Reference Standard, and dissolve エリスロマイシン
each in isooctane to make exactly 50 mL. Pipet 10 mL each
of these solutions, add exactly 3 mL each of the internal Change to read except the structural formula
standard solution, then add the mobile phase to make and chemical name:
50 mL, and use these solutions as the sample solution and
the standard solution, respectively. Perform the test with 10 Erythromycin contains not less than 930 mg
to 20 mL each of the sample solution and the standard solu-
(potency) per mg, calculated on the anhydrous basis.
tion as directed under the Liquid Chromatography accord- The potency of Erythromycin is expressed as mass
ing to the following conditions, and calculate the ratios, QT (potency) of erythromycin (C37H67NO13).
and QS, of the peak area of ergocalciferol to that of the
internal standard. Perform the procedure rapidly avoiding Description Erythromycin occurs as a white to light yel-
contact with air or other oxidizing agents and using light- lowish white powder.
resistant containers. It is very soluble in N, N-dimethylformamide, freely solu-
ble in methanol and in ethanol (95), and very slightly soluble
QT in water.
Amount (mg) of C28H44O = WS ×
QS
Identiˆcation (1) Determine the infrared absorption
WS: Amount (mg) of Ergocalciferol Reference Standard spectrum of Erythromycin as directed in the potassium
Internal standard solution—A solution of dimethyl phtha- bromide disk method under the Infrared Spectrophotomet-
late in isooctane (1 in 100). ry, and compare the spectrum with the Reference Spectrum
Operating conditions— or the spectrum of Erythromycin Reference Standard: both
Detector: An ultraviolet absorption photometer (wave- spectra exhibit similar intensities of absorption at the same
length: 254 nm). wave numbers.
Column: A stainless steel column 4.6 mm in inside (2) Dissolve 0.01 g each of Erythromycin and
diameter and 25 cm in length, packed with a silica gel for liq- Erythromycin Reference Standard in 1 mL of methanol, and
uid chromatography (10 mm particle diameter). use these solutions as the sample solution and the standard
Column temperature: A constant temperature of about solution. Perform the test with these solutions as directed
209C. under the Thin-layer Chromatography. Spot 5 mL each of
Mobile phase: A mixture of hexane and n-amylalcohol the sample solution and the standard solution on a plate of
(997:3). silica gel for thin-layer chromatography. Develop the plate
Flow rate: Adjust the ‰ow rate so that the retention time with a mixture of methanol and ammonia solution (28)
of ergocalciferol is about 25 minutes. (50:1) to a distance of about 10 cm, and air-dry the plate.
System suitability— Spray evenly 4-methoxybenzaldehyde-sulfuric acid TS on
System performance: Dissolve 15 mg of Ergocalciferol the plate, and heat at 1009 C for 15 minutes: the principal
Reference Standard in 25 mL of isooctane. Transfer this spot from the sample solution and the spot from the stan-
solution to a ‰ask, heat in an oil bath under a re‰ux con- dard solution are dark purple in color, and their R f values
denser for 2 hours, and cool immediately to room tempera- are the same.
ture. Transfer the solution to a quartz test tube, and irradi- Optical rotation [a]20
D : -71 – -789(1 g calculated on the
ate with a short-wave lamp (main wavelength: 254 nm) and a anhydrous basis, ethanol (95), 50 mL, 100 mm).
long-wave lamp (main wavelength: 365 nm) for 3 hours. To
10 mL of this solution add the mobile phase to make 50 mL. Purity (1) Heavy metals—Proceed with 1.0 g of
When the procedure is run with 10 mL of this solution under Erythromycin according to Method 4, and perform the test.
the above operating conditions. Use a column with the ratios Prepare the control solution with 2.0 mL of Standard Lead
of the retention time of previtamin D2, trans-vitamin D2 and Solution (not more than 20 ppm).
tachysterol2 to that of ergocalciferol being about 0.5, about (2) Arsenic—Prepare the test solution with 1.0 g of
0.6 and about 1.1, respectively, and with resolution between Erythromycin according to Method 5 using hydrochloric
previtamin D2 and trans-vitamin D2 being not less than 0.7, acid instead of diluted hydrochloric acid (1 in 2), and per-
and that between ergocalciferol and tachysterol2 being not form the test (not more than 2 ppm).
less than 1.0. (3) Related substances—Dissolve 40 mg of Erythromy-
System repeatability: When the test is repeated 6 times cin in 2 mL of methanol, add a mixture of phosphate buŠer
with 10 mL of the standard solution under the above operat- solution, pH 7.0 and methanol (15:1) to make exactly
10 mL, and use this solution as the sample solution.
Supplement I, JP XIV O‹cial Monographs for Part I 1461

Separately, dissolve 16 mg of Erythromycin Reference Stan- Assay Perform the test according to the Cylinder-plate
dard in 2 mL of methanol, add a mixture of phosphate method as directed under the Microbial Assay for Antibiot-
buŠer solution, pH 7.0 and methanol (15:1) to make exactly ics according to the following conditions.
10 mL, and use this solution as the standard stock solution. (1) Test organism—Staphylococcus aureus ATCC 6538
Dissolve 5 mg each of erythromycin B and erythromycin C P
in 2 mL of methanol, add exactly 2 mL of the standard stock (2) Culture medium—Use the medium i in 3) Medium
solution, add a mixture of phosphate buŠer solution, pH 7.0 for other organisms under (1) Agar media for seed and base
and methanol (15:1) to make exactly 25 mL, and use this layer. Adjust the pH of the medium so that it will be 7.8 to
solution as the standard solution. Perform the test with ex- 8.0 after sterilization.
actly 100 mL each of the sample solution and the standard (3) Standard solutions—Weigh accurately an amount of
solution as directed under the Liquid Chromatography Erythromycin Reference Standard, equivalent to about
according to the following conditions, and determine each 25 mg (potency), dissolve in 25 mL of methanol, add
peak area by the automatic integration method: the peak 0.1 mol/L phosphate buŠer solution, pH 8.0 to make
areas of erythromycin B and erythromycin C from the sam- exactly 100 mL, and use this solution as the standard stock
ple solution are not more than those of erythromycin B and solution. Keep the standard stock solution at 59 C or below,
erythromycin C from the standard solution, respectively, and use within 7 days. Take exactly a suitable amount of the
and each area of the peaks other than erythromycin, standard stock solution before use, add 0.1 mol/L phos-
erythromycin B and erythromycin C is not more than the phate buŠer solution, pH 8.0 to make solutions so that each
area of the peak of erythromycin from the standard solu- mL contains 20 mg (potency) and 5 mg (potency), and use
tion. these solutions as the high concentration standard solution
Operating conditions— and the low concentration standard solution, respectively.
Detector: An ultraviolet absorption photometer (wave- (4) Sample solutions—Weigh accurately an amount of
length: 215 nm). Erythromycin, equivalent to about 25 mg (potency), dissolve
Column: A stainless steel column 4.6 mm in inside di- in 25 mL of methanol, and add 0.1 mol/L phosphate buŠer
ameter and 25 cm in length, packed with styrene-divinylben- solution, pH 8.0 to make exactly 100 mL. Take exactly a
zene copolymer for liquid chromatography (8 mm in particle suitable amount of this solution, add 0.1 mol/L phosphate
diameter). buŠer solution, pH 8.0 to make solutions so that each mL
Column temperature: A constant temperature of about contains 20 mg (potency) and 5 mg (potency), and use these
709C. solutions as the high concentration sample solution and the
Mobile phase: Dissolve 3.5 g of dipotassium hydrogen low concentration sample solution, respectively.
phosphate in water to make 100 mL, and adjust the pH to
Containers and storage Containers—Well-closed contain-
9.0 with diluted phosphoric acid (1 in 10). To 50 mL of this
ers.
solution add 190 mL of t-butanol, 30 mL of acetonitrile and
water to make 1000 mL.
Flow rate: Adjust the ‰ow rate so that the retention time
of erythromycin is about 20 minutes.
Time span of measurement: About 4 times as long as the
retention time of erythromycin after the solvent peak.
System suitability—
System performance: Dissolve 2 mg of N-demethyl-
erythromycin in 10 mL of the standard solution. When the
procedure is run with 100 mL of this solution under the
above operating conditions, N-demethylerythromycin,
erythromycin C, erythromycin and erythromycin B are elut-
ed in this order, with the resolution between the peaks of N-
demethylerythromycin and erythromycin C being not less
than 0.8, and with the resolution between the peaks of N-
demethylerythromycin and erythromycin being not less than
5.5.
System repeatability: When the test is repeated 3 times
with 100 mL of the standard solution under the above oper-
ating conditions, the relative standard deviation of the peak
area of erythromycin is not more than 3.0z.

Water Not more than 10.0z (0.2 g, volumetric titration,


direct titration).

Residue on ignition Not more than 0.2z (1 g).


1462 O‹cial Monographs for Part I Supplement I, JP XIV

Add the following: pH The pH of a solution obtained by dissolving 0.5 g of


Erythromycin Lactobionate in 10 mL of water is between 5.0
Erythromycin Lactobionate and 7.5.

Water Not more than 5.0z (0.5 g, volumetric titration,


ラクトビオン酸エリスロマイシン
direct titration).

Assay Perform the test according to the Cylinder-plate


method as directed under the Microbial Assay for Antibiot-
ics according to the following conditions.
(1) Test organism—Staphylococcus aureus ATCC 6538
P
(2) Culture medium—Use the medium i in 3) Medium
for other organisms under (1) Agar media for seed and base
layer. Adjust the pH of the medium so that it will be 7.8 to
8.0 after sterilization.
C37H67NO13.C12H22O12: 1092.22
(3) Standard solutions—Weigh accurately an amount
(2R,3S,4S,5R,6R,8R,10R,11R,12S,13R )-5-(3,4,6-Trideoxy-3-
of Erythromycin Reference Standard, equivalent to about
dimethylamino-b-D -xylo-hexopyranosyloxy)-3-(2,6-dideoxy-
50 mg (potency), dissolve in 50 mL of methanol, add
3-C-methyl-3-O-methyl-a-L-ribo-hexopyranosyloxy)-
0.1 mol/L phosphate buŠer solution, pH 8.0 to make
6,11,12-trihydroxy-2,4,6,8,10,12-hexamethyl-9-
exactly 100 mL, and use this solution as the standard stock
oxopentadecan-13-olide mono(4-O-b-D-galactopyranosyl-
solution. Keep the standard stock solution at not exceeding
D-gluconate) [3847-29-8]
59 C and use within 7days. Take exactly a suitable amount of
the standard stock solution before use, add 0.1 mol/L phos-
Erythromycin Lactobionate contains not less than phate buŠer solution, pH 8.0 to make solutions so that each
590 mg (potency) per mg, calculated on the anhydrous mL contains 20 mg (potency) and 5 mg (potency), and use
basis. The potency of Erythromycin Lactobionate these solutions as the high concentration standard solution
is expressed as mass (potency) of erythromycin and the low concentration standard solution, respectively.
(C37H67NO13: 733.93). (4) Sample solutions—Weigh accurately an amount of
Description Erythromycin Lactobionate occurs as a white Erythromycin Lactobionate, equivalent to about 50 mg
powder. (potency), dissolve in 50 mL of methanol, and add
It is freely soluble in water, in methanol and in ethanol 0.1 mol/L phosphate buŠer solution, pH 8.0 to make
(99.5), and very slightly soluble in acetone. exactly 100 mL. Take exactly a suitable amount of this solu-
tion, add 0.1 mol/L phosphate buŠer solution, pH 8.0 to
Identiˆcation (1) To 3 mg of Erythromycin Lactobionate
make solutions so that each mL contains 20 mg (potency) and
add 2 mL of acetone, and add 2 mL of hydrochloric acid: an
5 mg (potency), and use these solutions as the high concentra-
orange color is produced, and it changes immediately to red
tion sample solution and the low concentration sample solu-
to deep purple.
tion, respectively.
(2) Transfer about 0.3 g of Erythromycin Lactobionate
to a separator, add 15 mL of ammonia TS and 15 mL of Containers and storage Containers—Tight containers.
chloroform, shake, and take the separated aqueous layer.
Wash the aqueous layer with three 15-mL portions of chlo-
roform, and evaporate the aqueous liquid on a water bath to Estradiol Benzoate
dryness. Dissolve the residue in 10 mL of a mixture of
methanol and water (3:2), and use this solution as the sample 安息香酸エストラジオール
solution. Separately, dissolve 0.10 g of lactobionic acid in
10 mL of a mixture of methanol and water (3:2), and use this Change the Assay to read:
solution as the standard solution. Perform the test with these Assay Weigh accurately about 10 mg each of Estradiol
solutions as directed under the Thin-layer Chromatography. Benzoate and Estradiol Benzoate Reference Standard, previ-
Spot 10 mL each of the sample solution and the standard ously dried, and dissolve each in methanol to make exactly
solution on a plate of silica gel for thin-layer chromato- 20 mL. Pipet 5 mL each of these solutions, add 5 mL of the
graph. Develop the plate with the upper layer obtained from internal standard solution, then add methanol to make
a mixture of water, 1-butanol and acetic acid (100) (3:3:1) to 20 mL, and use these solutions as the sample solution and
a distance of about 10 cm, and air-dry the plate. Spray the standard solution, respectively. Perform the test with
evenly dilute sulfuric acid, and heat at 1059
C for 20 minutes: 5 mL of the sample solution and the standard solution as
the principal spot obtained from the sample solution shows a directed under the Liquid Chromatography according to the
deep brown and the Rf value which are the same as those of following conditions, and calculate the ratios, Q T and QS, of
the principal spot from the standard solution. the peak area of estradiol benzoate to that of the internal
standard.
Supplement I, JP XIV O‹cial Monographs for Part I 1463

QT Column: A stainless steel column 4.6 mm in inside di-


Amount (mg) of C25H28O3 = WS ×
QS ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
WS: Amount (mg) of Estradiol Benzoate Reference
ameter).
Standard
Column temperature: A constant temperature of about
Internal standard solution—A solution of progesterone in 259C.
methanol (13 in 80,000). Mobile phase: A mixture of water and methanol (51:49).
Operating conditions— Flow rate: Adjust the ‰ow rate so that the retention time
Detector: An ultraviolet absorption photometer (wave- of estriol is about 10 minutes.
length: 230 nm). System suitability—
Column: A stainless steel column 4.6 mm in inside di- System performance: When the procedure is run with
ameter and 15 cm in length, packed with octadecylsilanized 10 mL of the standard solution under the above operating
silica gel for liquid chromatography (5 mm in particle di- conditions, estriol and the internal standard are eluted in this
ameter). order with the resolution between these peaks being not less
Column temperature: A constant temperature of about than 8.
359C. System repeatability: When the test is repeated 6 times
Mobile phase: A mixture of acetonitrile and water (7:3). with 10 mL of the standard solution under the above operat-
Flow rate: Adjust the ‰ow rate so that the retention time ing conditions, the relative standard deviation of the ratios
of estradiol benzoate is about 10 minutes. of the peak area of estriol to that of the internal standard is
System suitability— not more than 1.0z.
System performance: When the procedure is run with 5 mL
of the standard solution under the above operating condi-
tions, the internal standard and estradiol benzoate are eluted Add the following:
in this order with the resolution between these peaks being
not less than 9. Etizolam
System repeatability: When the test is repeated 6 times
with 5 mL of the standard solution under the above operat- エチゾラム
ing conditions, the relative standard deviation of the ratios
of the peak area of estradiol benzoate to that of the internal
standard is not more than 1.0z.

Estriol
エストリオール
C17H15ClN4S: 342.85
Change the Assay to read: 4-(2-Chlorophenyl)-2-ethyl-9-methyl-6H-
thieno[3,2-f ][1,2,4]triazolo[4,3-a][1,4]diazepine
Assay Weigh accurately about 25 mg each of Estriol and [40054-69-1]
Estriol Reference Standard, previously dried, and dissolve
each in methanol to make exactly 50 mL. Pipet 10 mL each Etizolam contains not less than 98.5z and not more
of these solutions, add exactly 5 mL of the internal standard than 101.0z of etizolam (C17H15ClN4S).
solution, add methanol to make 100 mL, and use these solu-
tions as the sample solution and the standard solution, Description Etizolam occurs as a white to pale yellowish
respectively. Perform the test with 10 mL each of the sample white crystalline powder.
solution and the standard solution as directed under the Liq- It is soluble in ethanol (99.5), sparingly soluble in aceto-
uid Chromatography according to the following conditions, nitrile and in acetic anhydride, and practically insoluble in
and calculate the ratios, QT and QS, of the peak area of water.
estriol to that of the internal standard, respectively. Identiˆcation (1) Determine the absorption spectrum of
Q a solution of Etizolam in ethanol (99.5) (1 in 100,000) as
Amount (mg) of C18H24O3 = WS × T
QS directed under the Ultraviolet-visible Spectrophotometry,
WS: Amount (mg) of Estriol Reference Standard and compare the spectrum with the Reference Spectrum:
both spectra exhibit similar intensities of absorption at the
Internal standard solution—A solution of methyl benzoate same wavelengths.
for estriol limit test in methanol (1 in 1000). (2) Determine the infrared absorption spectrum of
Operating conditions— Etizolam as directed in the potassium bromide disk method
Detector: An ultraviolet absorption photometer (wave- under the Infrared Spectrophotometry, and compare the
length: 280 nm). spectrum with the Reference Spectrum: both spectra exhibit
1464 O‹cial Monographs for Part I Supplement I, JP XIV

similar intensities of absorption at the same wave numbers. Assay Weigh accurately about 0.3 g of Etizolam, previous-
ly dried, dissolve in 70 mL of a mixture of acetic anhydride
Melting point 146 – 1499
C
and acetic acid (100) (7:3), and titrate with 0.1 mol/L per-
Purity (1) Heavy metals—Proceed with 2.0 g of Etizolam chloric acid VS (potentiometric titration). The end point is
according to Method 2, and perform the test. Prepare the the second equivalent point. Perform a blank determination,
control solution with 2.0 mL of Standard Lead Solution (not and make any necessary correction.
more than 10 ppm).
Each mL of 0.1 mol/L perchloric acid VS
(2) Related substances—Dissolve 20 mg of Etizolam in
= 17.14 mg of C17H15ClN4S
50 mL of acetonitrile, and use this solution as the sample
solution. Pipet 1 mL of the sample solution, and add Containers and storage Containers—Tight containers.
acetonitrile to make exactly 20 mL. Pipet 1 mL of this solu- Storage—Light-resistant.
tion, add acetonitrile to make exactly 50 mL, and use this
solution as the standard solution. Perform the test with
exactly 10 mL each of the sample solution and the standard Famotidine for Injection
solution as directed under the Liquid Chromatography
according to the following conditions, and determine each 注射用ファモチジン
peak area by the automatic integration method: the area of
the peak other than etizolam obtained from the sample solu- Change the Purity (2) to read:
tion is not more than the peak area of etizolam from the
Purity
standard solution.
(2) Related substances—Take a number of Famotidine
Operating conditions—
for Injection, equivalent to about 0.1 g of famotidine
Detector: An ultraviolet absorption photometer (wave-
(C8H15N7O2S3), dissolve each content in water, wash the
length: 240 nm).
inside of the container with water, combine the solutions of
Column: A stainless steel column 4.6 mm in inside di-
the contents with the washings, add water to the combined
ameter and 15 cm in length, packed with octadecylsilanized
solution to make exactly 100 mL, and use this solution as the
silica gel for liquid chromatography (5 mm in particle di-
sample solition. Pipet 1 mL of the sample solution, add
ameter).
water to make exactly 100 mL, and use this solution as the
Column temperature: A constant temperature of about
standard solution. Perform the test with exactly 5 mL each of
359C.
the sample solution and the standard solution as directed un-
Mobile phase: Dissolve 1.36 g of potassium dihydrogen
der the Liquid Chromatography according to the following
phosphate in water to make 1000 mL, and adjust the pH to
conditions, and determine each peak area of these solutions
3.5 with potassium hydroxide TS. To 550 mL of this solu-
by the automatic integration method: the total area of the
tion add 450 mL of acetonitrile.
peaks other than peak of famotidine from the sample solu-
Flow rate: Adjust the ‰ow rate so that the retention time
tion is not larger than peak area of famotidine from the stan-
of etizolam is about 6 minutes.
dard solution.
Time span of measurement: About 5 times as long as the
Operating conditions—
retention time of etizolam after the solvent peak.
Detector, column, column temperature, mobile phase,
System suitability—
and ‰ow rate: Proceed as directed in the operating condi-
Test for required detectability: Measure exactly 2 mL of
tions in the Assay.
the standard solution, and add acetonitrile to make exactly
Time span of measurement: About 2 times as long as the
20 mL. Conˆrm that the peak area of etizolam obtained
retention time of famotidine after the solvent peak.
from 10 mL of this solution is equivalent to 8 to 12z of that
System suitability—
from 10 mL of the standard solution.
Test for required detection: To exactly 2 mL of the stan-
System performance: Dissolve 0.02 g each of Etizolam
dard solution add the water to make exactly 20 mL. Conˆrm
and ethyl parahydroxybenzoate in the mobile phase to make
that the peak area of famotidine obtained from 5 mL of this
50 mL. To 1 mL of this solution add the mobile phase to
solution is equivalent to 8 to 12z of that of famotidine
make 50 mL. When the procedure is run with 10 mL of this
obtained from 5 mL of the standard solution.
solution under the above operating conditions, etizolam and
System performance: Proceed as directed in the system
ethyl parahydroxybenzoate are eluted in this order with the
suitability in the Assay.
resolution between these peaks being not less than 3.
System repeatability: When the test is repeated 6 times
System repeatability: When the test is repeated 6 times
with 5 mL of the standard solution under the above operat-
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
ing conditions, the relative standard deviation of the peak
area of famotidine is not more than 2.0z.
area of etizolam is not more than 2z.

Loss on drying Not more than 0.5z (1 g, 1059


C, 3 hours).

Residue on ignition Not more than 0.10z (1 g).


Supplement I, JP XIV O‹cial Monographs for Part I 1465

Delete the following Monograph: 50 mL, 100 mm).

pH The pH of a solution obtained by dissolving 0.5 g of


Floctafenine Flomoxef Sodium in 5 mL of water is between 4.0 and 5.5.
フロクタフェニン Purity (1) Clarity and color of solution—Dissolve 1.0 g
of Flomoxef Sodium in 10 mL of water: the solution is clear
and pale yellow.
(2) Heavy metals—Proceed with 1.0 g of Flomoxef
Flomoxef Sodium Sodium in a quartz crucible according to Method 2, and
perform the test. Prepare the control solution with 2.0 mL
フロモキセフナトリウム of Standard Lead Solution (not more than 20 ppm).
(3) Arsenic—To 1.0 g of Flomoxef Sodium 5 mL of sul-
Change to read except the structural formula furic acid and 5 mL of nitric acid, heat carefully until the
and chemical name: solution changes to colorless to light yellow with occasional
addition of 2 mL of nitric acid. After cooling, add 10 mL of
Flomoxef Sodium contains not less than 862 mg ammonium oxalate TS, heat until white fumes evolve, and
(potency) per mg, calculated on the anhydrous basis. concentrate to 2 to 3 mL. After cooling, add water to make
The potency of Flomoxef Sodium is expressed as mass 10 mL, and perform the test using this solution as the test
(potency) of ‰omoxef (C15H18F2N6O7S2: 496.47). solution: the color is not darker than that of the control
Description Flomoxef Sodium occurs as white to light yel- solution.
lowish white, powder or masses. Control solution: Proceed to prepare a solution in the
It is very soluble in water, freely soluble in methanol, and same manner as the test solution without Flomoxef Sodium,
sparingly soluble in ethanol (99.5). and transfer 10 mL of the solution so obtained to the genera-
tor bottle, add exactly 2 mL of Standard Arsenic Solution,
Identiˆcation (1) Decompose 0.01 g of Flomoxef Sodium and proceed in the same manner as the test solution (not
as directed under the Oxygen Flask Combustion Method, more than 2 ppm).
using a mixture of 0.5 mL of 0.01 mol/L sodium hydroxide (4) 1-(2-Hydroxyethyl)-1H-tetrazol-5-thiol—Weigh ac-
TS and 20 mL of water as the absorbing liquid. To 2 mL of curately about 20 mg of 1-(2-hydroxyethyl)-1H-tetrazol-5-
the test solution so obtained add 1.5 mL of a mixture of thiol, and dissolve in water to make exactly 100 mL. Pipet
alizarin complexone TS, acetic acid-potassium acetate buŠer 5 mL of this solution, add exactly 25 mL of the internal stan-
solution, pH 4.3 and cerium (III) nitrate TS (1:1:1): blue- dard solution and water to make 50 mL, and use this solu-
purple color develops. tion as the standard solution. Perform the test with 5 mL
(2) Determine the absorption spectrum of a solution of each of the sample solution obtained in the Assay and the
Flomoxef Sodium (3 in 100,000) as directed under the standard solution as directed under the Liquid Chro-
Ultraviolet-visible Spectrophotometry, and compare the matography according to the following conditions, and
spectrum with the Reference Spectrum: both spectra exhibit determine the ratios, QT and QS, of the peak area of 1-(2-
similar intensities of absorption at the same wavelengths. hydroxyethyl)-1H-tetrazol-5-thiol to that of the internal
(3) Determine the infrared absorption spectrum of standard: the amount of 1-(2-hydroxyethyl)-1H-tetrazol-5-
Flomoxef Sodium as directed in the potassium bromide disk thiol is not more than 1.0z of the amount of Flomoxef
method under the Infrared Spectrophotometry, and com- Sodium calculated on the anhydrous basis.
pare the spectrum with the Reference Spectrum: both spec-
tra exhibit similar intensities of absorption at the same wave Amount (mg) of 1-(2-hydroxyethyl)-1H-tetrazol-5-thiol
numbers. (C3H6N4OS)
(4) Determine the spectrum of a solution of Flomoxef Q 1
= WS × T ×
QS 10
Sodium in heavy water for nuclear magnetic resonance spec-
troscopy (1 in 100) as directed under the Nuclear Magnetic WS: Amount (mg) of 1-(2-hydroxyethyl)-1H-tetrazol-5-
Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl- thiol
propanesulfonate for nuclear magnetic resonance spec-
troscopy as an internal reference compound: it exhibits a sin- Operating conditions—
gle signal A at around d 3.5 ppm, a single signal or a sharp Proceed as directed in the operating conditions in the
multiple signal B at around d 3.7 ppm, and a single signal C Assay.
at around d 5.2 ppm. The ratio of the integrated intensity of System suitability—
these signals, A:B:C, is about 3:2:1. Proceed as directed in the system suitability in the Assay.
(5) Flomoxef Sodium responds to the Qualitative Test Water Not more than 1.5z (0.5 g, volumetric titration,
(1) for sodium salt. back titration).
Optical rotation [a]20
D : -8 – -139 (1 g calculated on the Assay Weigh accurately an amount of Flomoxef Sodium
anhydrous basis, a mixture of water and ethanol (99.5) (4:1), and Flomoxef Triethylammonium Reference Standard,
1466 O‹cial Monographs for Part I Supplement I, JP XIV

equivalent to about 50 mg (potency), and dissolve each in ex- (11:9) to make exactly 200 mL, and use this solution as the
actly 50 mL of the internal standard solution, add water to standard solution. Perform the test with exactly 20 mL each
make 100 mL, and use these solutions as the sample solution of the sample solution and the standard solution as directed
and standard solution. Perform the test with 5 mL each of under the Liquid Chromatography according to the follow-
the sample solution and the standard solution as directed un- ing conditions. Determine each peak area of both solutions
der the Liquid Chromatography according to the following by the automatic integration method: each area of the peaks
conditions, and determine the ratios, QT and QS, of the peak other than the peak of ‰urbiprofen from the sample solution
area of ‰omoxef to that of the internal standard. is not larger than the peak area of ‰urbiprofen from the
standard solution, and the total area of these peaks is not
Amount [mg (potency)] of ‰omoxef (C15H18F2N6O7S2)
larger than twice the peak area of ‰urbiprofen from the stan-
Q
= WS × T × 1000 dard solution.
QS
Operating conditions—
WS: Amount [mg (potency)] of Flomoxef Triethylammo- Detector: An ultraviolet absorption photometer (wave-
nium Reference Standard length: 254 nm).
Column: A stainless steel column 4.6 mm in inside di-
Internal standard solution—A solution of m-cresol (3 in
ameter and 15 cm in length, packed with octadecylsilanized
1000).
silica gel for liquid chromatography (5 mm in particle di-
Operating conditions—
ameter).
Detector: An ultraviolet absorption photometer (wave-
Column temperature: A constant temperature of about
length: 246 nm).
309C.
Column: A stainless steel column 4 mm in inside diameter
Mobile phase: A mixture of water, acetonitrile and acetic
and 20 cm in length, packed with octadecylsilanized silica gel
acid (100) (12:7:1).
for liquid chromatography (5 – 10 mm in particle diameter).
Flow rate: Adjust the ‰ow rate so that the retention time
Column temperature: A constant temperature of about
of ‰urbiprofen is about 20 minutes.
259C.
Time span of measurement: About twice as long as the
Mobile phase: Dissolve 6.94 g of potassium dihydrogen
retention time of ‰urbiprofen after the solvent peak.
phosphate, 3.22 g of disodium hydrogen phosphate 12-water
System suitability—
and 1.60 g of tetra-n-butylammonium bromide in water to
Test for required detection: To exactly 5 mL of the stan-
make 1000 mL. To 750 mL of this solution add 250 mL of
dard solution add a mixture of water and acetonitrile (11:9)
methanol.
to make exactly 25 mL. Conˆrm that the peak area of ‰ur-
Flow rate: Adjust the ‰ow rate so that the retention time
biprofen obtained from 20 mL of this solution is equivalent
of ‰omoxef is about 9 minutes.
to 16 to 24z of that of ‰urbiprofen obtained from 20 mL of
System suitability—
the standard solution.
System performance: When the procedure is run with 5 mL
System performance: Dissolve 0.04 g of ‰urbiprofen and
of the standard solution under the above operating condi-
0.02 g of butyl parahydroxybenzoate in 100 mL of a mixture
tions, ‰omoxef and the internal standard are eluted in this
of water and acetonitrile (11:9). To 5 mL of this solution add
order with the resolution between these peaks being not less
a mixture of water and acetonitrile (11:9) to make 50 mL.
than 10.
When the procedure is run with 20 mL of this solution under
System repeatability: When the test is repeated 3 times
the above operating conditions, butyl parahydroxybenzoate
with 5 mL of the standard solution under the above operat-
and ‰urbiprofen are eluted in this order with the resolution
ing conditions, the relative standard deviation of the ratios
between these peaks being not less than 12.
of the peak area of ‰omoxef to that of the internal standard
System repeatability: When the test is repeated 6 times
is not more than 1.0z.
with 20 mL of the standard solution under the above operat-
Containers and storage Containers—Tight containers. ing conditions, the relative standard deviation of the peak
Storage—At 59 C or below. area of ‰urbiprofen is not more than 2.0z.

Flurbiprofen Folic Acid


フルルビプロフェン 葉酸

Change the Purity (3) to read: Change the Water to read:


Purity Water Not more than 8.5z (10 mg, coulometric titration).
(3) Related substances—Dissolve 20 mg of Flurbiprofen
in 10 mL of a mixture of water and acetonitrile (11:9), and
use this solution as the sample solution. Pipet 1 mL of the
sample solution, and add a mixture of water and acetonitrile
Supplement I, JP XIV O‹cial Monographs for Part I 1467

Fradiomycin Sulfate tone (1 in 50) on the plate, and heat at 1109C for 15 minutes:
the spot at around R f 0.4 from the sample solution is not
硫酸フラジオマイシン more intense than the spot from the standard solution.

Loss on drying Not more than 8.0z (0.2 g, in vaccum,


Change to read except the structural formula 609C, 3 hours).
and chemical name:
Residue on ignition Not more than 0.30z (1 g).
Fradiomycin Sulfate, when dried, contains not less Assay Perform the test according to the Cylinder-plate
than 623 mg (potency) per mg. The potency of method as directed under the Microbial Assay for Antibio-
Fradiomycin Sulfate is expressed as mass (potency) of tics according to the following conditions.
fradiomycin (C23H46N6O13: 614.64). (1) Test organism—Staphylococcus aureus ATCC 6538
Description Fradiomycin Sulfate occurs as a white to light P
yellow powder. (2) Agar medium for seed and base layer
It is freely soluble in water, and practically insoluble in Glucose 1.0 g
ethanol (95). Peptone 6.0 g
It is hygroscopic. Meat extract 1.5 g
Yeast extract 3.0 g
Identiˆcation (1) Dissolve 0.05 g each of Fradiomycin
Sodium chloride 2.5 g
Sulfate and Fradiomycin Sulfate Reference Standard in
Agar 15.0 g
1 mL of water, and use these solutions as the sample solution
Water 1000 mL
and the standard solution. Perform the test with these solu-
Mix all the ingredients and sterilize. Adjust the pH after
tions as directed under the Thin-layer Chromatography.
sterilization to 7.8 – 8.0 with sodium hydroxide TS.
Spot 2 mL each of the sample solution and the standard solu-
(3) Standard solutions-Weigh accurately an amount of
tion on a plate of silica gel for thin-layer chromatography.
Fradiomycin Sulfate Reference Standard, previously dried,
Develop the plate with a mixture of methanol, ammonia
equivalent to about 50 mg (potency), dissolve in 0.1 mol/L
solution (28) and dichloromethane (3:2:1) to a distance of
phosphate buŠer solution for antibiotics, pH 8.0 to make ex-
about 10 cm, and air-dry the plate. Spray evenly a solution
actly 50 mL, and use this solution as the standard stock solu-
of ninhydrin in acetone (1 in 50) on the plate, and heat at
tion. Keep the standard stock solution at 59 C or below and
1109 C for 15 minutes: the Rf values of the principal spots
use within 14 days. Take exactly a suitable amount of the
from the sample solution and the standard solution are not
standard stock solution before use, add 0.1 mol/L phos-
diŠerent each other.
phate buŠer solution for antibiotics, pH 8.0 to make solu-
(2) A solution of Fradiomycin Sulfate (1 in 20) responds
tions so that each mL contains 80 mg (potency) and 20 mg
to the Qualitative Test (1) for sulfate.
(potency), and use these solutions as the high concentration
Optical rotation [a]20
D : +53.5 – +59.09(1 g calculated on
standard solution and the low concentration standard solu-
the dried basis, water, 10 mL, 100 mm). tion, respectively.
(4) Sample solutions—Weigh accurately an amount of
pH The pH of a solution obtained by dissolving 1.0 g of
Fradiomycin Sulfate, previously dried, equivalent to about
Fradiomycin Sulfate in 10 mL of water is between 5.0 and
50 mg (potency), dissolve in 0.1 mol/L phosphate buŠer
7.5.
solution for antibiotics, pH 8.0 to make exactly 50 mL. Take
Purity (1) Heavy metals—Proceed with 1.0 g of Fra- exactly a suitable amount of this solution, add 0.1 mol/L
diomycin Sulfate according to Method 2, and perform phosphate buŠer solution for antibiotics, pH 8.0 to make so-
the test. Prepare the control solution with 2.0 mL of Stan- lutions so that each mL contains 80 mg (potency) and 20 mg
dard Lead Solution (not more than 20 ppm). (potency), and use these solutions as the high concentration
(2) Arsenic—Prepare the test solution with 1.0 g of sample solution and the low concentration sample solution,
Fradiomycin Sulfate according to Method 1, and perform respectively.
the test (not more than 2 ppm).
Containers and storage Containers—Tight containers.
(3) Related substances—Dissolve 0.63 g of Fradiomycin
Storage—Light-resistant.
Sulfate in 5 mL of water, and use this solution as the sample
solution. Pipet 1 mL of the sample solution, add water to
make exactly 50 mL, and use this solution as the standard
solution. Perform the test with these solutions as directed
under the Thin-layer Chromatography. Spot 1 mL each of
the sample solution and the standard solution on a plate of
silica gel for thin-layer chromatography. Develop the plate
with a mixture of methanol, ammonia solution (28) and
dichloromethane (3:2:1) to a distance of about 10 cm, and
air-dry the plate. Spray evenly a solution of ninhydrin in ace-
1468 O‹cial Monographs for Part I Supplement I, JP XIV

Gentamicin Sulfate hour. To 20 mL of the lower layer so obtained add 0.5 mL


of methanol, and use this as the developing solvent. Develop
硫酸ゲンタマイシン the plate with the developing solvent to a distance of about
17 cm in a developing container with a cover, having an
Change to read except the structural formula opening of about 20 mm2, without putting a ˆlter paper in
and chemical name: the container, and air-dry the plate. Allow the plate to stand
in iodine vapor. Determine the integral absorbances, Aa, Ab
Gentamicin Sulfate contains not less than 590 mg and Ac, of the colored spots of gentamicin C1 (R f value:
(potency) per mg, calculated on the dried basis. The about 0.3), gentamicin C2 (R f value: about 0.2) and gen-
potency of Gentamicin Sulfate is expressed as mass tamicin C1a (R f value: about 0.1), respectively, by a den-
(potency) of gentamicin C1 (C21H43N5O7: 477.60). sitometer (wavelength: 450 nm) while covering the plate with
a glass plate, and calculate these amounts by the following
Description Gentamicin Sulfate occurs as a white to light
formulae: gentamicin C1 is between 25z and 55z, gentami-
yellowish white powder.
cin C2 is between 25z and 50z, and gentamicin C1a is
It is very soluble in water, and practically insoluble in
between 5z and 30z.
ethanol (99.5).
Aa
It is hygroscopic. Amount (z) of gentamicin C1 = × 100
Aa + 1.35Ab + Ac
Identiˆcation (1) Dissolve 50 mg of Gentamicin Sulfate 1.35Ab
Amount (z) of gentamicin C2 = × 100
in 1 mL of water, and add 2 drops of a solution of 1- Aa + 1.35Ab + Ac
naphthol in ethanol (95) (1 in 500). Gently superimpose this Ac
Amount (z) of gentamicin C1a = × 100
solution on 1 mL of sulfuric acid: a blue-purple color de- Aa + 1.35Ab + Ac
velops at the zone of contact.
Purity (1) Clarity and color of solution—Dissolve 1.0 g
(2) Dissolve 50 mg each of Gentamicin Sulfate and Gen-
of Gentamicin Sulfate in 10 mL of water: the solution is
tamicin Sulfate Reference Standard in 10 mL of water, and
clear and colorless to pale yellow.
use these solutions as the sample solution and the standard
(2) Heavy metals—Proceed with 2.0 g of Gentamicin
solution. Perform the test with these solutions as directed
Sulfate according to Method 4, and perform the test. Pre-
under the Thin-layer Chromatography. Spot 20 mL of the
pare the control solution with 2.0 mL of Standard Lead
sample solution and the standard solution on a plate of silica
Solution (not more than 10 ppm).
gel for thin-layer chromatography. Separately, shake a mix-
(3) Related substances—Dissolve 50 mg of Gentamicin
ture of chloroform, ammonia solution (28) and methanol
Sulfate in water to make 10 mL, and use this solution as the
(2:1:1) in a separator, and allow the mixture to stand for
sample solution. Pipet 1 mL of the sample solution, add
more than 1 hour. To 20 mL of the lower layer so obtained
water to make exactly 50 mL, and use this solution as the
add 0.5 mL of methanol, and use this as the developing sol-
standard solution. Perform the test with these solutions as
vent. Develop the plate with the developing solvent to a dis-
directed under the Thin-layer Chromatography. Spot 20 mL
tance of about 17 cm in a developing container with a cover,
of the sample solution and the standard solution on a plate
having an opening of about 20 mm2, and without putting a
of silica gel for thin-layer chromatography. Separately,
ˆlter paper in the container, and air-dry the plate. Allow the
shake a mixture of chloroform, ammonia solution (28) and
plate to stand in iodine vapors: three principal spots ob-
methanol (2:1:1) in a separator, and allow the mixture to
tained from the sample solution are the same with the corre-
stand for more than 1 hour. To 20 mL of the lower layer so
sponding spots from the standard solution in color tone and
obtained add 0.5 mL of methanol, and use this as the de-
the Rf value, respectively.
veloping solvent. Develop the plate with the developing sol-
(3) Dissolve 50 mg of Gentamicin Sulfate in 5 mL of
vent to a distance of about 17 cm in a developing container
water, and add 0.5 mL of barium chloride TS: a white
with a cover, having an opening of about 20 mm2, without
precipitate is formed.
putting a ˆlter paper in the container, and air-dry the plate.
Optical rotation [a]25
D : +107 – +1219(0.25 g calculated on
Allow the plate to stand in iodine vapor, and compare the
the dried basis, water, 25 mL, 100 mm). colored spots while covering with a glass plate: the spots
other than the spots of gentamicin C1 (R f value: about 0.3),
pH The pH of a solution obtained by dissolving 0.20 g of
gentamicin C2 (R f value: about 0.2) and gentamicin C1a (R f
Gentamicin Sulfate in 5 mL of water is between 3.5 and 5.5.
value: about 0.1) obtained from the sample solution are not
Content ratio of the active principle Dissolve 50 mg of more intense than the spot of gentamicin C2 from the stan-
Gentamicin Sulfate in water to make 10 mL, and use this dard solution.
solution as the sample solution. Perform the test with this
Loss on drying Not more than 18.0z (0.15 g, reduced
solution as directed under the Thin-layer Chromatography.
pressure not exceeding 0.67 kPa, 1109C, 3 hours). Handle
Spot 20 mL of the sample solution on a plate of silica gel for
the sample avoiding absorption of moisture.
thin-layer chromatography. Separately, shake a mixture of
chloroform, ammonia solution (28) and methanol (2:1:1) in Residue on ignition Not more than 1.0z (1 g).
a separator, and allow the mixture to stand for more than 1
Assay Perform the test according to the Cylinder-plate
Supplement I, JP XIV O‹cial Monographs for Part I 1469

method as directed under the Microbial Assay for Antibio- Change the Identiˆcation to read:
tics according to the following conditions.
Identiˆcation Determine the infrared absorption spectrum
(1) Test organism—Staphylococcus epidermidis ATCC
of Glycerin as directed in the liquid ˆlm method under the
12228
Infrared Spectrophotometry, and compare the spectrum
(2) Agar media for seed and base layer—
with the Reference Spectrum: both spectra exhibit similar in-
Glucose 1.0 g
tensities of absorption at the same wave numbers.
Peptone 6.0 g
Meat extract 1.5 g
Add the following next to Purity:
Yeast extract 3.0 g
Sodium chloride 10.0 g Water 13 – 17z (0.1 g, volumetric titration, direct titra-
Agar 15.0 g tion).
Water 1000 mL
Mix all the ingredients, and sterilize. Adjust the pH of the Add the following next to Residue on ignition:
solution so that it will be 7.8 to 8.0 after sterilization.
Assay Weigh accurately about 0.2 g of Glycerin, transfer
(3) Agar medium for transferring test organisms—Use
into a glass-stoppered ‰ask, add 50 mL of water, mix, add
the medium ii in 2) Medium for other organisms under (2)
exactly 50 mL of sodium periodate TS, shake, and allow to
Agar media for transferring test organisms.
stand in a dark place at a room temperature for about 30
(4) Standard solutions—Weigh accurately an amount of
minutes. Add 10 mL of a mixture of water and ethylene
Gentamicin Sulfate Reference Standard, equivalent to about
glycol (1:1), allow to stand for about 20 minutes, add 100
25 mg (potency), dissolve in 0.1 mol/L phosphate buŠer
mL of water, and titrate with 0.1 mol/L sodium hydroxide
solution, pH 8.0 to make exactly 25 mL, and use this solu-
VS (indicator: 2 drops of phenolphthalein TS). Perform a
tion as the standard stock solution. Keep the standard stock
blank determination, and make the necessary correction.
solution at 159C or lower, and use within 30 days. Take
exactly a suitable amount of the standard stock solution be- Each mL of 0.1 mol/L sodium hydroxide VS
fore use, add 0.1 mol/L phosphate buŠer solution, pH 8.0 = 9.209 mg of C3H8O3
to make solutions so that each mL contains 4 mg (potency)
and 1 mg (potency), and use these solutions as the high con-
centration standard solution and the low concentration stan- Concentrated Glycerin
dard solution, respectively.
(5) Sample solutions—Weigh accurately an amount of 濃グリセリン
Gentamicin Sulfate, equivalent to about 25 mg (potency),
and dissolve in 0.1 mol/L phosphate buŠer solution, pH 8.0 Change the origin/limits of content to read:
to make exactly 25 mL. Take exactly a suitable amount of
this solution, add 0.1 mol/L phosphate buŠer solution, pH Concentrated Glycerin contains not less than 98.0z
8.0 to make solutions so that each mL contains 4 mg and not more than 101.0z of glycerin (C3H8O3), cal-
(potency) and 1 mg (potency), and use these solutions as the culated of the anhydrous basis.
high concentration sample solution and the low concentra-
tion sample solution, respectively. Change the Description to read:
Containers and storage Containers—Tight containers. Description Concentrated Glycerin is a clear, colorless and
viscous liquid. It has a sweet taste.
It is miscible with water and with ethanol (99.5).
Glycerin It is hygroscopic.

グリセリン Change the Identiˆcation to read:


Identiˆcation Determine the infrared absorption spectrum
Change the origin/limits of content to read:
of Concentrated Glycerin as directed in the liquid ˆlm
method under the Infrared Spectrophotometry, and com-
Glycerin contains not less than 84.0z and not more
pare the spectrum with the Reference Spectrum: both spec-
than 87.0z of C3H8O3.
tra exhibit similar intensities of absorption at the same wave
numbers.
Change the Description to read:
Description Glycerin is a clear, colorless, viscous liquid. It Add the following next to Purity:
has a sweet taste.
Water Not more than 2.0z (6 g, volumetric titration,
It is miscible with water and with ethanol (99.5).
direct titration).
It is hygroscopic.
1470 O‹cial Monographs for Part I Supplement I, JP XIV

Add the following next to Residue on ignition: Glucose 10.0 g


Casein peptone 5.0 g
Assay Weigh accurately about 0.2 g of Concentrated Glyc-
Yeast extract 20.0 g
erin, transfer into a glass-stoppered ‰ask, add 50 mL of
Potassium dihydrogen phosphate 2.0 g
water, mix, add exactly 50 mL of sodium periodate TS,
Polysorbate 80 0.1 g
shake, and allow to stand in a dark place at a room tempera-
Agar 15.0 g
ture for about 30 minutes. Add 10 mL of a mixture of water
Water 1000 mL
and ethylene glycol (1:1), allow to stand for about 20
Mix all the ingredients, and sterilize. Adjust the pH of the
minutes, add 100 mL of water, and titrate with 0.1 mol/L
solution so that it will be 6.7 to 6.8 after sterilization.
sodium hydroxide VS (indicator: 2 drops of phenolphthalein
(3) Liquid medium for suspending test organism—Use
TS). Perform a blank determination, and make the necessa-
the culture medium (2).
ry correction.
(4) Preparation of the test organism suspension—Pun-
Each mL of 0.1 mol/L sodium hydroxide VS cture the test organism in the medium, prepared by dispens-
= 9.209 mg of C3H8O3 ing 10 mL of the agar medium for transferring test organism
in a test tube about 16 mm in inside diameter, incubate at
36.5 to 37.59 C for 20 to 24 hours. After sub-culturing at
Add the following: least three times, keep between 1 to 59 C. Transfer the organ-
ism so obtained in 10 mL of the liquid medium for suspend-
Gramicidin ing test organism, incubate at 36.5 to 37.59C for 20 to 24
hours, and use this medium as the test organism stock sus-
グラミシジン pension. Before use, add the test organism stock suspension
to the liquid medium for suspending test organism so that
[1405-97-6]
the transmittance at 580 nm is 50 to 60z. Mix one volume of
this suspension and 200 volume of the liquid medium for
Gramicidin is a mixture of peptide substances hav-
suspending test organism, and use this as the test organism
ing antibacterial activity produced by the growth of
suspension.
Bacillus brevis Dubos.
(5) Standard solution—Weigh accurately an amount of
It contains not less than 900 mg (potency) per mg,
Gramicidin Reference Standard, previously dried under
calculated on the dried basis. The potency of
reduced pressure not exceeding 0.67 kPa at 609 C for 3
Gramicidin is expressed as mass (potency) of
hours, equivalent to about 10 mg (potency), dissolve in
gramicidin.
ethanol (99.5) to make exactly 100 mL, and use this solution
Description Gramicidin occurs as a white to light yellowish as the standard stock solution. Keep the standard stock solu-
white crystalline powder. tion at not exceeding 59C and use within 7 days. Take ex-
It is freely soluble in methanol, soluble in ethanol (99.5), actly a suitable amount of the standard stock solution before
and practically insoluble in water. use, add the following diluting solution to make a solution
so that each mL contains 0.02 mg (potency), and use this
Identiˆcation (1) To 10 mg of Gramicidin add 2 mL of
solution as the standard solution.
6 mol/L hydrochloric acid TS, and heat in a water bath for
Diluting solution: To 390 mL of propylene glycol add 210
30 minutes with occasional stirring. After cooling, neutralize
mL of a mixture of ethanol (99.5) and acetone (9:1) and
with 6 mol/L sodium hydroxide TS, add 1 mL of ninhydrin
Sterile Puriˆed Water to make 1000 mL.
TS and 0.5 mL of pyridine, and heat for 2 minutes: a blue-
(6) Sample solution—Weigh accurately an amount of
purple to red-purple color develops.
Gramicidin, equivalent to about 10 mg (potency), and dis-
(2) Determine the absorption spectrum of a solution of
solve in ethanol (99.5) to make exactly 100 mL. Take exactly
Gramicidin in ethanol (95) (1 in 20,000), as directed under
a suitable amount of this solution, add the diluting solution
the Ultraviolet-visible Spectrophotometry, and compare the
obtained in (5) to make a solution so that each mL contains
spectrum with the Reference Spectrum or the spectrum of a
0.02 mg (potency), and use this solution as the sample solu-
solution of Gramicidin Reference Standard prepared in the
tion.
same manner as the sample solution: both spectra exhibit
(7) Procedure—Transfer 0.155 mL, 0.125 mL, 0.100
similar intensities of absorption at the same wavelengths.
mL, 0.080 mL and 0.065 mL each of the standard solution,
Loss on drying Not more than 3.0z (0.1 g, in vacuum, 0.100 mL of the sample solution and 0.100 mL of the dilut-
609C, 3 hours). ing solution, separately, in test tubes about 14 mm in inside
diameter and about 15 cm in length, and make three sets for
Residue on ignition Not more than 1.0z (1 g).
each. To each of the test tube add 10 mL of the test organism
Assay Perform the test according to the Turbidimetric suspension, stopper the tube, incubate in a water bath at
method as directed under the Microbial Assay for Antibiot- 36.5 to 37.59C for 180 to 270 minutes, add 0.5 mL of a solu-
ics according to the following conditions. tion of formaldehyde (1 in 3), and determine their transmit-
(1) Test organism—Enterococcus hirae ATCC 10541 tances at 580 nm.
(2) Agar medium for transferring test organism
Supplement I, JP XIV O‹cial Monographs for Part I 1471

Containers and storage Containers—Tight containers. tion and the standard solution as directed under the Gas
Chromatography according to the following conditions,
determine each peak area by the automatic integration
Griseofulvin method, and calculate the ratio, Q1, of the peak area of
dechlorogriseofulvin, having the relative retention time of
グリセオフルビン about 0.6 with respect to griseofulvin, to that of the internal
standard obtained from the sample solution, the ratio, Q2,
Change to read except the structural formula of the peak area of dehydrogriseofulvin, having the relative
and chemical name: retention time of about 1.2 with respect to griseofulvin, to
that of the internal standard obtained from the sample solu-
Griseofulvin contains not less than 960 mg (potency) tion and the ratio, QS, of the peak area of griseofulvin to
per mg, calculated on the dried basis. The potency that of the internal standard obtained from the standard
of Griseofulvin is expressed as mass (potency) of solution: Q1/QS is not more than 0.6, and Q2/QS is not more
griseofulvin (C17H17ClO6). than 0.15.
Internal standard solution—A solution of 9,10-
Description Griseofulvin occurs as white, crystals or crys-
diphenylanthracene in acetone (1 in 500).
talline powder.
Operating conditions—
It is soluble in N, N-dimethylformamide, sparingly soluble
Detector: An hydrogen ‰ame-ionization detector.
in acetone, slightly soluble in methanol and in ethanol (95),
Column: A glass column 4 mm in inside diameter and 1 m
and practically insoluble in water.
in length, packed with siliceous earth for gas chro-
Identiˆcation (1) Determine the absorption spectrum of matography coated with 25z phenyl-25z cyanopropyl-
a solution of Griseofulvin in ethanol (95) (1 in 100,000) as methylsilicone polymer for gas chromatography in the ratio
directed under the Ultraviolet-visible Spectrophotometry, of 1z (150 – 180 mm in particle diameter).
and compare the spectrum with the Reference Spectrum or Column temperature: A constant temperature of about
the spectrum of a solution of Griseofulvin Reference Stan- 2509 C.
dard prepared in the same manner as the sample solution: Temperature of injection port: A constant temperature of
both spectra exhibit similar intensities of absorption at the about 2709 C.
same wavelengths. Temperature of detector: A constant temperature of
(2) Determine the infrared absorption spectrum of about 3009 C.
Griseofulvin as directed in the potassium bromide disk Carrier gas: Nitrogen
method under the Infrared Spectrophotometry, and com- Flow rate: Adjust the ‰ow rate so that the retention time
pare the spectrum with the Reference Spectrum or the of griseofulvin is about 10 minutes.
spectrum of Griseofulvin Reference Standard: both spectra System suitability—
exhibit similar intensities of absorption at the same wave Test for required detectability: Measure exactly 1 mL of
numbers. the standard solution, and add the internal standard solution
diluted with acetone (1 in 10) to make exactly 10 mL. Con-
Optical rotation [a]20
D : +350 – +3649(0.25 g calculated on
ˆrm that the ratio of the peak area of griseofulvin to that of
the dried basis, N, N-dimethylformamide, 25 mL, 100 mm).
the internal standard obtained from 2 mL of this solution is
Melting point 218 – 2229
C equivalent to 7 to 13z of that obtained from 2 mL of the
standard solution.
Purity (1) Acid—Dissolve 0.25 g of Griseofulvin in
System performance: When the procedure is run with 2 mL
20 mL of neutralized ethanol, and add 2 drops of
of the standard solution under the above operating condi-
phenolphthalein TS and 1.0 mL of 0.02 mol/L sodium
tions, the internal standard and griseofulvin are eluted in
hydroxide VS: the color of the solution is red.
this order with the resolution between these peaks being not
(2) Heavy metals—Proceed with 1.0 g of Griseofulvin
less than 5.
according to Method 2, and perform the test. Prepare the
System repeatability: When the test is repeated 6 times
control solution with 2.5 mL of Standard Lead Solution (not
with 2 mL of the standard solution under the above operat-
more than 25 ppm).
ing conditions, the relative standard deviation of the ratios
(3) Arsenic—Prepare the test solution with 1.0 g of
of the peak area of griseofulvin to that of the internal stan-
Griseofulvin according to Method 3, and perform the test
dard is not more than 5.0z.
(not more than 2 ppm).
(5) Petroleum ether soluble substances—To 1.0 g of
(4) Related substances—To 0.10 g of Griseofulvin add
Griseofulvin add 20 mL of petroleum ether, shake, and boil
exactly 1 mL of the internal standard solution and acetone to
for 10 minutes under a re‰ux condenser. After cooling, ˆlter
make 10 mL, and use this solution as the sample solution.
through a dried ˆlter paper, wash the ˆlter paper with two
Separately, to 5.0 mg of Griseofulvin Reference Standard
15-mL portions of petroleum ether, combine the washings to
add exactly 1 mL of the internal standard solution and ace-
the ˆltrate, evaporate the petroleum ether on a water bath,
tone to make 10 mL, and use this solution as the standard
and dry the residue at 1059 C for 1 hour: the amount of the
solution. Perform the test with 2 mL each of the sample solu-
residue is not more than 0.2z.
1472 O‹cial Monographs for Part I Supplement I, JP XIV

Loss on drying Not more than 1.0z (1 g, reduced pressure solution. Pipet 1 mL of the sample solution, add acetonitrile
not exceeding 0.67 kPa, 609C, 3 hours). to make exactly 100 mL, and use this solution as the stan-
dard solution. Perform the test with exactly 10 mL each of
Residue on ignition Not more than 0.20z (1 g).
the sample solution and the standard solution as directed un-
Assay Weigh accurately an amount of Griseofulvin and der the Liquid Chromatography according to the following
Griseofulvin Reference Standard, equivalent to about 50 mg conditions. Determine each peak area of both solutions by
(potency), dissolve each in 50 mL of N, N-dimethylform- the automatic integration method: the total area of all peaks
amide, add exactly 20 mL of the internal standard solution other than the area of the haloxazolam from the sample
and water to make 250 mL, and use these solutions as the solution is not larger than the peak area of the haloxazolam
sample solution and the standard solution. Perform the test from the standard solution.
with exactly 10 mL each of the sample solution and the stan- Operating conditions—
dard solution as directed under the Liquid Chromatography Detector: An ultraviolet absorption photometer (wave-
according to the following conditions, and determine the length: 250 nm).
ratios, QT and QS, of the peak area of griseofulvin to that of Column: A stainless steel column 4.6 mm in inside di-
the internal standard. ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle
QT
Amount [mg (potency)] of C17H17ClO6 = WS × × 1000 diameter).
QS
Column temperature: A constant temperature of about
WS: Amount [mg (potency)] of Griseofulvin Reference 259C.
Standard Mobile phase: Dissolve 6.2 g of boric acid and 7.5 g of
potassium chloride in 900 mL of water, adjust the pH with
Internal standard solution—A solution of butyl parahydrox-
triethylamine to 8.5, and add water to make 1000 mL. To
ybenzoate in acetonitrile (1 in 400).
300 mL of this solution add 200 mL of acetonitrile.
Operating conditions—
Flow rate: Adjust the ‰ow rate so that the retention time
Detector: An ultraviolet absorption photometer (wave-
of haloxazolam is about 10 minutes.
length: 254 nm).
Time span of measurement: About 3 times as long as the
Column: A stainless steel column 4.6 mm in inside di-
retention time of haloxazolam after the solvent peak.
ameter and 25 cm in length, packed with octadecylsilanized
System suitability—
silica gel for liquid chromatography (10 mm in particle di-
Test for required detection: To exactly 5 mL of the stan-
ameter).
dard solution add acetonitrile to make exactly 50 mL. Con-
Column temperature: A constant temperature of about
ˆrm that the peak area of haloxazolam obtained from 10 mL
259C.
of this solution is equivalent to 8 to 12z of that of halox-
Mobile phase: A mixture of water and acetonitrile (3:2).
azolam obtained from 10 mL of the standard solution.
Flow rate: Adjust the ‰ow rate so that the retention time
System performance: Dissolve 10 mg each of Haloxazol-
of griseofulvin is about 6 minutes.
am and cloxazolam in 200 mL of acetonitrile. When the
System suitability—
procedure is run with 10 mL of this solution under the above
System performance: When the procedure is run with
operating conditions, haloxazolam and cloxazolam are elut-
10 mL of the standard solution under the above operating
ed in this order with the resolution between these peaks
conditions, griseofulvin and the internal standard are eluted
being not less than 1.5.
in this order with the resolution between these peaks being
System repeatability: When the test is repeated 6 times
not less than 4.
with 10 mL of the standard solution under the above operat-
System repeatability: When the test is repeated 6 times
ing conditions, the relative standard deviation of the peak
with 10 mL of the standard solution under the above operat-
area of haloxazolam is not more than 1.0z.
ing conditions, the relative standard deviation of the ratios
of the peak area of griseofulvin to that of the internal stan-
dard is not more than 1.0z.
Homochlorcyclizine Hydrochloride
Containers and storage Containers—Tight containers.
塩酸ホモクロルシクリジン

Haloxazolam Change the Description to read:


Description Homochlorcyclizine Hydrochloride occurs as
ハロキサゾラム
white to pale brown, crystals or powder.
It is very soluble in water, freely soluble in acetic acid
Change the Purity (5) to read:
(100), slightly soluble in ethanol (99.5), and very slightly
Purity soluble in acetonitrile and in acetic anhydride.
(5) Related substances—Dissolve 0.10 g of Haloxazolam It dissolves in 0.1 mol/L hydrochloric acid TS.
in 100 mL of acetonitrile, and use this solution as the sample It is hygroscopic.
Supplement I, JP XIV O‹cial Monographs for Part I 1473

It is colored slightly by light. retention time of homochlorcyclizine.


A solution of Homochlorcyclizine Hydrochloride (1 in 10) System suitability—
shows no optical rotation. Test for required detection: To exactly 5 mL of the stan-
Melting point: about 2279 C (with decomposition). dard solution add the mobile phase to make exactly 50 mL.
Conˆrm that the peak area of homochlorcyclizine obtained
Change the Identiˆcation to read: from 10 mL of this solution is equivalent to 7 to 13z of that
of homochlorcyclizine obtained from 10 mL of the standard
Identiˆcation (1) Determine the absorption spectrum
solution.
of a solution of Homochlorcyclizine Hydrochloride in
System performance: Dissolve 5 mg each of Homochlor-
0.1 mol/L hydrochloric acid TS (1 in 100,000) as directed
cyclizine Hydrochloride and methyl parahydroxybenzoic
under the Ultraviolet-visible Spectrophotometry, and com-
acid in 100 mL of the mobile phase. When the procedure is
pare the spectrum with the Reference Spectrum: both spec-
run with 10 mL of this solution under the above operating
tra exhibit similar intensities of absorption at the same wave-
conditions, methyl parahydroxybenzoic acid and homo-
lengths.
chlorcyclizine are eluted in this order with the resolution
(2) Determine the infrared absorption spectrum of
between these peaks being not less than 5.
Homochlorcyclizine Hydrochloride, previously dried, as
System repeatability: When the test is repeated 6 times
directed in the potassium chloride disk method under the In-
with 10mL of the standard solution under the above operat-
frared Spectrophotometry, and compare the spectrum with
ing conditions, the relative standard deviation of the peak
the Reference Spectrum: both spectra exhibit similar intensi-
area of homochlorcyclizine is not more than 1.0z.
ties of absorption at the same wave numbers.
(3) A solution of Homochlorcyclizine Hydrochloride (1
in 100) responds to the Qualitative Tests for chloride.
Hydrochlorothiazide
Change the Purity to read: ヒドロクロロチアジド
Purity (1) Heavy metals—Proceed with 1.0 g of Homo-
chlorcyclizine Hydrochloride according to Method 2, and Change the Assay to read:
perform the test. Prepare the control solution with 2.0 mL
Assay Weigh accurately about 30 mg each of Hydro-
of Standard Lead Solution (not more than 20 ppm).
chlorothiazide and Hydrochlorothiazide Reference Stan-
(2) Related substances—Dissolve 0.10 g of Homochlor-
dard, previously dried, and dissolve in 150 mL of the mobile
cyclizine Hydrochloride in 100 mL of the mobile phase, and
phase, add exactly 10 mL each of the internal standard solu-
use this solution as the sample solution. Measure exactly
tion, then add the mobile phase to make 200 mL, and use
1 mL of this solution, add the mobile phase to make exactly
these solutions as the sample solution and the standard solu-
100 mL, and use this solution as the standard solution.
tion, respectively. Perform the test with 20 mL each of the
Perform the test with exactly 10 mL each of the sample solu-
sample solution and the standard solution as directed under
tion and the standard solution as directed under the Liquid
the Liquid Chromatography according to the following con-
Chromatography according to the following conditions, and
ditions, and calculate the ratios, Q T and Q S, of the peak area
determine the peak areas by the automatic integration
of hydrochlorothiazide to that of the internal standard.
method: the areas of the peaks other than homochlorcycli-
Q
zine obtained from the sample solution are not more than Amount (mg) of C7H8ClN3O4S2 = WS × T
QS
1/2 times the peak area of homochlorcyclizine from the
standard solution, and the total area of the peaks other than WS: Amount (mg) of Hydrochlorothiazide Reference
homochlorcyclizine from the sample solution is not more Standard
than the peak area of homochlorcyclizine from the standard
Internal standard solution—A solution of 4-aminoacetophe-
solution.
none in acetonitrile (9 in 2000).
Operating conditions—
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Detector: An ultraviolet absorption photometer (wave-
length: 223 nm).
length: 254 nm).
Column: A stainless steel column 4.6 mm in inside di-
Column: A stainless steel column 4.6 mm in inside di-
ameter and 25 cm in length, packed with octadecylsilanized
ameter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
silica gel for liquid chromatography (5 mm in particle di-
ameter).
ameter).
Column temperature: A constant temperature of about
Column temperature: A constant temperature of about
409C.
259C.
Mobile phase: A mixture of water, acetonitrile and per-
Mobile phase: A mixture of 0.1 mol/L sodium dihydro-
chloric acid (134:66:1).
gen phosphate TS, pH 3.0 and acetonitrile (9:1).
Flow rate: Adjust the ‰ow rate so that the retention time
Flow rate: Adjust the ‰ow rate so that the retention time
of homochlorcyclizine is about 10 minutes.
of hydrochlorothiazide is about 10 minutes.
Time span of measurement: About 2 times as long as the
1474 O‹cial Monographs for Part I Supplement I, JP XIV

System suitability— of the peak area of hydrocortisone acetate to that of the


System performance: When the procedure is run with internal standard is not more than 1.0z.
20 mL of the standard solution under the above operating
conditions, hydrochlorothiazide and the internal standard
are eluted in this order with the resolution between these Hydrocortisone Sodium Phosphate
peaks being not less than 4.
System repeatability: When the test is repeated 6 times リン酸ヒドロコルチゾンナトリウム
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios Cange the Purity (6) to read:
of the peak area of hydrochlorothiazide to that of the inter-
Purity
nal standard is not more than 1.0z.
(6) Free hydrocortisone—Dissolve 25 mg of Hydrocorti-
sone Sodium Phosphate in the mobile phase to make exactly
20 mL, and use this solution as the sample solution.
Hydrocortisone Acetate Separately, weigh 25 mg of Hydrocortisone Reference Stan-
dard, previously dried at 1059C for 3 hours, and dissolve in
酢酸ヒドロコルチゾン
the mobile phase to make exactly 100 mL. Pipet 10 mL of
this solution, add the mobile phase to make exactly 200 mL,
Change the Assay to read:
and use this solution as the standard solution. Perform the
Assay Dissolve about 20 mg each of Hydrocortisone test with 20 mL each of the sample solution and the standard
Acetate and Hydrocortisone Acetate Reference Standard, solution as directed under the Liquid Chromatography ac-
previously dried and accurately weighed, in methanol, add cording to the following conditions. Determine the peak
exactly 10 mL each of the internal standard solution, then areas, AT and AS, of hydrocortisone from each solution: AT
add methanol to make 100 mL, and use these solutions as is not larger than AS.
the sample solution and the standard solution. Perform the Operating conditions—
test with 20 mL each of the sample solution and the standard Detector, column, column temperature, mobile phase,
solution as directed under the Liquid Chromatography ac- and ‰ow rate: Proceed as directed in the operating condi-
cording to the following conditions, and calculate the ratios, tions in the Assay.
Q T and Q S, of the peak area of hydrocortisone acetate to System suitability—
that of the internal standard, respectively. System performance: Proceed as directed in the system
suitability in the Assay.
QT
Amount (mg) of C23H32O6 = WS × System repeatability: When the test is repeated 6 times
QS
with 20 mL of the standard solution under the above operat-
WS: Amount (mg) of Hydrocortisone Acetate Reference ing conditions, the relative standard deviation of the peak
Standard area of hydrocortisone is not more than 1.0z.

Internal standard solution—A solution of benzyl para-


Change the Assay to read:
hydroxybenzoate in methanol (1 in 1000).
Operating conditions— Assay Weigh accurately about 20 mg each of Hydrocorti-
Detector: An ultraviolet absorption photometer (wave- sone Sodium Phosphate and Hydrocortisone Sodium Phos-
length: 254 nm). phate Reference Standard (determine its loss on drying
Column: A stainless steel column 3.9 mm in inside di- before using), dissolve each in 50 mL of the mobile phase,
ameter and 30 cm in length, packed with octadecylsilanized add exactly 10 mL of the internal standard solution, then
silica gel for liquid chromatography (10 mm in particle di- add the mobile phase to make 200 mL, and use these solu-
ameter). tions as the sample solution and the standard solution,
Column temperature: A constant temperature of about respectively. Perform the test with 20 mL each of the sample
259C. solution and the standard solution as directed under the Liq-
Mobile phase: A mixture of water and acetonitrile (13:7). uid Chromatography according to the following conditions,
Flow rate: Adjust the ‰ow rate so that the retention time and calculate the ratios, QT and QS, of the peak area of
of hydrocortisone acetate is about 8 minutes. hydrocortisone phosphate to that of the internal standard,
System suitability— respectively.
System performance: When the procedure is run with
QT
20 mL of the standard solution under the above operating Amount (mg) of C21H29Na2O8P = WS ×
QS
conditions, hydrocortisone acetate and the internal standard
are eluted in this order with the resolution between these WS: Amount (mg) of Hydrocortisone Sodium Phosphate
peaks being not less than 4. Reference Standard, calculated on the dried basis
System repeatability: When the test is repeated 6 times
Internal standard solution—A solution of isopropyl para-
with 20 mL of the standard solution under the above operat-
hydroxybenzoate in the mobile phase (3 in 5000).
ing conditions, the relative standard deviation of the ratios
Supplement I, JP XIV O‹cial Monographs for Part I 1475

Operating conditions— length: 254 nm).


Detector: An ultraviolet absorption photometer (wave- Column: A stainless steel column 3.9 mm in inside di-
length: 254 nm). ameter and 30 cm in length, packed with octadecylsilanized
Column: A stainless steel column 4.6 mm in inside di- silica gel for liquid chromatography (10 mm in particle di-
ameter and 25 cm in length, packed with octadecylsilanized ameter).
silica gel for liquid chromatography (7 mm in particle di- Column temperature: A constant temperature of about
ameter). 259C.
Column temperature: A constant temperature of about Mobile phase: A mixture of water and methanol (24:1).
259C. Flow rate: Adjust the ‰ow rate so that the retention time
Mobile phase: A mixture of 0.05 mol/L sodium dihydro- of 2?-deoxyuridine is about 6 minutes.
gen phosphate TS, pH 2.6 and methanol (1:1). System suitability—
Flow rate: Adjust the ‰ow rate so that the retention time System performance: When the procedure is run with 10 mL
of hydrocortisone phosphate is about 10 minutes. of the standard solution under the above operating condi-
System suitability— tions, 2?-deoxyuridine and 5-iodouracil are eluted in this
System performance: When the procedure is run with order with the resolution between these peaks being not less
20 mL of the standard solution under the above operating than 2.0.
conditions, hydrocortisone phosphate and isopropyl para- System repeatability: When the test is repeated 6 times
hydroxybenzoate are eluted in this order with the resolution with 10 mL of the standard solution under the above operat-
between these peaks being not less than 8. ing conditions, the relative standard deviation of the peak
System repeatability: When the test is repeated 6 times area of 2?-deoxyuridine is not more than 1.0z.
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios Change the Assay to read:
of the peak area of hydrocortisone phosphate to that of the
Assay Measure exactly a volume of Idoxuridine
internal standard is not more than 1.0z.
Ophthalmic Solution, equivalent to 3 mg of idoxuridine
(C9H11IN2O5) according to the labeled amount, add exactly
2 mL of the internal standard solution, then add water to
Idarubicin Hydrochloride make 10 mL, and use this solution as the sample solution.
Separately weigh accurately about 10 mg of Idoxuridine
塩酸イダルビシン
Reference Standard, previously dried at 609C for 3 hours,
dissolve in water to make exactly 10 mL. Measure exactly
Change the Water to read:
3 mL of this solution, add exactly 2 mL of the internal stan-
Water Not more than 5.0z (0.1 g, coulometric titration). dard solution, then add water to make 10 mL, and use this
solution as the standard solution. Perform the test with
10 mL each of the sample solution and the standard solution
Idoxuridine Ophthalmic Solution as directed under the Liquid Chromatography according to
the following conditions, and calculate the ratios, Q T and
イドクスウリジン点眼液 Q S, of the peak area of idoxuridine to that of the internal
standard, respectively.
Change the Purity to read:
Amount (mg) of idoxuridine (C9H11IN2O5)
Purity 5-Iodouracil and 2?-deoxyuridine—To a volume of Q 3
Idoxuridine Ophthalmic Solution, equivalent to 4.0 mg of = WS × T ×
QS 10
Idoxuridine according to the labeled amount, add water to
make exactly 5 mL, and use this solution as the sample solu- WS: Amount (mg) of Idoxuridine Reference Standard
tion. Separately, dissolve 12.0 mg of 5-iodouracil for liquid Internal standard solution—A solution of sulfathiazole in
chromatography and 4.0 mg of 2?-deoxyuridine for liquid the mobile phase (1 in 4000).
chromatography in water to make exactly 200 mL. Measure Operating conditions—
exactly 5 mL of this solution, add water to make exactly Detector: An ultraviolet absorption photometer (wave-
25 mL, and use this solution as the standard solution. length: 254 nm).
Perform the test with exactly 10 mL each of the sample solu- Column: A stainless steel column 3.9 mm in inside di-
tion and the standard solution as directed under the Liquid ameter and 30 cm in length, packed with octadecylsilanized
Chromatography according to the following conditions, and silica gel for liquid chromatography (10 mm in particle di-
determine the peak areas of 5-iodouracil and 2?-deoxyuri- ameter).
dine: the peak areas of 5-iodouracil and 2?-deoxyuridine of Column temperature: A constant temperature of about
the sample solution are not more than the peak areas of 5- 259C.
iodouracil and 2?-deoxyuridine of the standard solution. Mobile phase: A mixture of water and methanol (87:13).
Operating conditions— Flow rate: Adjust the ‰ow rate so that the retention time
Detector: An ultraviolet absorption photometer (wave- of idoxuridine is about 9 minutes.
1476 O‹cial Monographs for Part I Supplement I, JP XIV

System suitability— acid, heat, and repeat this procedure once more. Then add 2
System performance: When the procedure is run with mL of hydrogen peroxide (30), heat, and repeat this proce-
10 mL of the standard solution under the above operating dure several times until the color of the solution changes to
conditions, idoxuridine and the internal standard are eluted colorless to pale yellow. After cooling, heat again until white
in this order with the resolution between these peaks being fumes evolve. After cooling, add water to make 5 mL, and
not less than 2.0. perform the test with this solution as the test solution (not
System repeatability: When the test is repeated 6 times more than 1 ppm).
with 10 mL of the standard solution under the above operat- (3) Related substances—Dissolve 50 mg of Imipenem in
ing conditions, the relative standard deviation of the ratios 50 mL of 0.1 mol/L 3-( N-morpholino)propanesulfonic acid
of the peak area of idoxuridine to that of the internal stan- buŠer solution, pH 7.0, and use this solution as the sample
dard is not more than 1.0z. solution. Pipet 1 mL of the sample solution, add 0.1 mol/L
3-( N-morpholino)propanesulfonic acid buŠer solution, pH
7.0 to make exactly 100 mL, and use this solution as the
Imipenem standard solution. Perform the test with exactly 10 mL each
of the sample solution and the standard solution as directed
イミペネム under the Liquid Chromatography according to the follow-
ing conditions, and determine each peak area by the auto-
Change to read except the structural formula matic integration method: the peak area of thienamycin,
and chemical name: having the relative retention time of about 0.8 with respect
to imipenem, obtained from the sample solution is not more
Imipenem contains not less than 924 mg (potency) than the peak area of imipenem from the standard solution,
per mg, calculated on the anhydrous basis. The poten- the area of the peak other than imipenem and thienamycin
cy of Imipenem is expressed as mass (potency) of im- from the sample solution is not more than 1/3 times the peak
ipenem (C12H17N3O4S: 299.35). area of imipenem from the standard solution, and the total
area of the peaks other than imipenem and thienamycin
Description Imipenem occurs as white to light yellow crys-
from the sample solution is not more than the peak area of
talline powder.
imipenem from the standard solution.
It is sparingly soluble in water, and practically insoluble in
Operating conditions—
ethanol (99.5).
Detector, column, column temperature, mobile phase,
Identiˆcation (1) Determine the absorption spectrum of and ‰ow rate: Proceed as directed in the operating condi-
a solution of Imipenem in 0.1 mol/L 3-( N-mor- tions in the Assay.
pholino)propanesulfonic acid buŠer solution, pH 7.0 (1 in Time span of measurement: About 2 times as long as the
50,000) as directed under the Ultraviolet-visible Spec- retention time of imipenem.
trophotometry, and compare the spectrum with the Refer- System suitability—
ence Spectrum or the spectrum of a solution of Imipenem Test for required detectability: Measure exactly 5 mL of
Reference Standard prepared in the same manner as the the standard solution, add 0.1 mol/L 3-( N-morpholino)-
sample solution: both spectra exhibit similar intensities of propanesulfonic acid buŠer solution, pH 7.0 to make exactly
absorption at the same wavelengths. 50 mL. Conˆrm that the peak area of imipenem from 10 mL
(2) Determine the infrared absorption spectrum of Im- of this solution is equivalent to 7 to 13z of that from the
ipenem as directed in the potassium bromide disk method standard solution.
under the Infrared Spectrophotometry, and compare the System performance: Proceed as directed in the system
spectrum with the Reference Spectrum or the spectrum of suitability in the Assay.
Imipenem Reference Standard: both spectra exhibit similar System repeatability: When the test is repeated 6 times
intensities of absorption at the same wave numbers. with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
Optical rotation [a]20
D : +84 – +899(50 mg calculated on the
area of imipenem is not more than 2.0z.
anhydrous basis, 0.1 mol/L 3-( N-morpholino)propanesul-
fonic acid buŠer solution, pH 7.0, 10 mL, 100 mm). Water Not less than 5.0z and not more than 8.0z (20
mg, coulometric titration, water evaporation temperature:
pH The pH of a solution obtained by dissolving 1.0 g of
1409C).
Imipenem in 200 mL of water is between 4.5 and 7.0.
Residue on ignition Not more than 0.2z (1 g).
Purity (1) Heavy metals—Proceed with 1.0 g of Imipen-
em according to Method 2, and perform the test. Prepare the Assay Weigh accurately an amount of Imipenem and Im-
control solution with 2.0 mL of Standard Lead Solution (not ipenem Reference Standard, equivalent to about 50 mg
more than 20 ppm). (potency), dissolve each in 0.1 mol/L 3-( N-morpholino)-
(2) Arsenic—Put 2.0 g of Imipenem in a crucible, add 5 propanesulfonic acid buŠer solution, pH 7.0 to make exactly
mL of nitric acid and 1 mL of sulfuric acid, and heat careful- 50 mL, and use these solutions as the sample solution and
ly until white fumes evolve. After cooling, add 2 mL of nitric the standard solution. Perform the test with exactly 10 mL
Supplement I, JP XIV O‹cial Monographs for Part I 1477

each of the sample solution and the standard solution, the sample solution. Separately, weigh accurately about
within 30 minutes after preparation of these solutions, as 50 mg of Indometacin Reference Standard, previously dried
directed under the Liquid Chromatography according to the at 1059 C for 4 hours, and dissolve in tetrahydrofuran to
following conditions, and determine the peak areas, AT and make exactly 50 mL. Pipet 5 mL of the solution, proceed in
AS, of imipenem of these solutions. the same manner as the sample solution, and use as the stan-
dard solution. Perform the test with 20 mL each of the sam-
Amount [ mg (potency)] of C12H17N3O4S
ple solution and the standard solution as directed under the
A
= WS × T × 1000 Liquid Chromatography according to the following condi-
AS
tions, and calculate the ratios, Q T and QS, of the peak area
WS: Amount [mg (potency)] of Imipenem Reference
of indometacin to that of the internal standard, respectively.
Standard
Amount (mg) of indometacin (C19H16ClNO4)
Operating conditions—
Q
Detector: An ultraviolet absorption photometer = WS × T
QS
(wavelength: 280 nm).
Column: A stainless steel column 3.9 mm in inside di- WS: Amount (mg) of Indometacin Reference Standard
ameter and 30 cm in length, packed with octadecylsilanized
Internal standard solution—A solution of butyl parahydrox-
silica gel for liquid chromatography (10 mm in particle di-
ybenzoate in methanol (1 in 1000).
ameter).
Operating conditions—
Column temperature: A constant temperature of about
Detector: An ultraviolet absorption photometer
259C.
(wavelength: 254 nm).
Mobile phase: A mixture of 0.1 mol/L 3-( N-mor-
Column: A stainless steel column 4.0 mm in inside di-
pholino)propanesulfonic acid buŠer solution, pH 7.0 and
ameter and 25 cm in length, packed with octadecylsilanized
acetonitrile (100:1).
silica gel for liquid chromatography (7 mm in particle di-
Flow rate: Adjust the ‰ow rate so that the retention time
ameter).
of imipenem is about 6 minutes.
Column temperature: A constant temperature of about
System suitability—
259C.
System performance: Dissolve 50 mg of Imipenem and 75
Mobile phase: A mixture of methanol and diluted phos-
mg of resorcinol in 50 mL of 0.1 mol/L 3-( N-mor-
phoric acid (1 in 1000) (7:3).
pholino)propanesulfonic acid buŠer solution, pH 7.0. When
Flow rate: Adjust the ‰ow rate so that the retention time
the procedure is run with 10 mL of this solution under the
of indometacin is about 8 minutes.
above operating conditions, imipenem and resorcinol are
System suitability—
eluted in this order with the resolution between these peaks
System performance: Dissolve 50 mg of 4-chlorobenzoic
being not less than 4.
acid, 30 mg of butyl parahydroxybenzoate and 50 mg of in-
System repeatability: When the test is repeated 5 times
dometacin in 50 mL of methanol. To 5 mL of this solution
with 10 mL of the standard solution under the above operat-
add the mobile phase to make 100 mL. When the procedure
ing conditions, the relative standard deviation of the peak
is run with 20 mL of this solution under the above operating
area of imipenem is not more than 0.80z.
conditions, 4-chlorobenzoic acid, butyl parahydroxybenzo-
Containers and storage Containers—Hermetic containers. ate and indometacin are eluted in this order with the resolu-
tion between the peaks of 4-chlorobenzoic acid and butyl
parahydroxybenzoate being not less than 2.0 and between
Indometacin Suppositories the peaks of parahydroxybenzoate and indometacin being
not less than 5.
インドメタシン坐剤 System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution under the above operat-
Change the Assay to read: ing conditions, the relative standard deviation of the ratios
of the peak area of indometacin to that of the internal stan-
Assay Weigh accurately not less than 20 Indometacin Sup-
dard is not more than 1.0z.
positories, cut into small pieces carefully, and mix well.
Weigh accurately a portion of the mass, equivalent to about
50 mg of indometacin (C19H16ClNO4), add 40 mL of tetra-
hydrofuran, warm at 409 C, dissolve by shaking, cool, and
add tetrahydrofuran to make exactly 50 mL. Filter the solu-
tion, discard the ˆrst 10 mL of the ˆltrate, pipet the subse-
quent 5 mL of the ˆltrate, add exactly 3 mL of the internal
standard solution, and add the mobile phase to make
100 mL. Allow the solution to stand for 30 minutes, ˆlter
through a membrane ˆlter (0.5 mm pore size), discard the
ˆrst 10 mL of the ˆltrate, and use the subsequent ˆltrate as
1478 O‹cial Monographs for Part I Supplement I, JP XIV

Delete the following Monographs: ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Iopanoic Acid ameter).
Column temperature: A constant temperature of about
イオパノ酸 409C.
Mobile phase: Dissolve 6.80 g of potassium dihydrogen
Iopanoic Acid Tablets phosphate in water to make 1000 mL. Separately, to 5.76 g
of phosphoric acid add water to make 1000 mL. Mix these
イオパノ酸錠 solutions to make a solution having pH 2.5. To 500 mL of
this solution add 500 mL of methanol, and add 2.86 g of so-
dium tridecanesulfonate to dissolve.
Flow rate: Adjust the ‰ow rate so that the retention time
Isoniazid Injection of isoniazid is about 5 minutes.
System suitability—
イソニアジド注射液 System performance: When the procedure is run with 5 mL
of the standard solution under the above operating condi-
Change the Description to read: tions, isoniazid and the internal standard are eluted in this
Description Isoniazid Injection occurs as a clear, colorless order with the resolution between these peaks being not less
liquid. than 10.
pH: 6.5 – 7.5. System repeatability: When the test is repeated 6 times
with 5 mL of the standard solution under the above operat-
Change the Identiˆcation to read: ing conditions, the relative standard deviation of the ratios
of the peak area of isoniazid to that of the internal standard
Identiˆcation To a volume of Isoniazid Injection, equiva- is not more than 1.3z.
lent to 20 mg of Isoniazid according to the labeled amount,
and add water to make 200 mL. To 5 mL of the solution add
1 mL of 0.1 mol/L hydrochloric acid TS and water to make
50 mL. Determine the absorption spectrum of this solution
Isoniazid Tablets
as directed under the Ultraviolet-visible Spectrophotometry: イソニアジド錠
it exhibits a maximum between 264 nm and 268 nm.
Change the Assay to read:
Change the Assay to read:
Assay Weigh accurately and powder not less than 20
Assay To an exactly measured volume of Isoniazid Injec- Isoniazid Tablets. Weigh accurately a quantity of the pow-
tion, equivalent to about 50 mg of isoniazid (C6H7N3O), add der, equivalent to about 0.10 g of isoniazid (C6H7N3O), add
water to make exactly 100 mL. Pipet 5 mL of the solution, 150 mL of water, shake for 30 minutes, then add water to
add exactly 5 mL of the internal standard and the mobile make exactly 200 mL, and ˆlter. Discard the ˆrst 10 mL of
phase to make 50 mL, and use this solution as the sample so- the ˆltrate, pipet 5 mL of the subsequent ˆltrate, add the
lution. Separately, weigh accurately about 50 mg of isonia- mobile phase to make exactly 50 mL, and use this solution as
zid for assay, previously dried at 1059C for 2 hours, and dis- the sample solution. Separately, weigh accurately about 50
solve in water to make exactly 100 mL. Pipet 5 mL of this mg of isoniazid for assay, previously dried at 1059 C for 2
solution, add exactly 5 mL of the internal standard and the hours, dissolve in water to make exactly 100 mL. Pipet 5 mL
mobile phase to make 50 mL, and use this solution as the of this solution, add the mobile phase to make exactly 50
standard solution. Perform the test with 5 mL each of the mL, and use this solution as the standard solution. Perform
sample solution and the standard solution as directed under the test with 10 mL each of the sample solution and the stan-
the Liquid Chromatography according to the following con- dard solution as directed under the Liquid Chromatography
ditions, and determine the ratios, Q T and QS, of the peak according to the following conditions. Determine the peak
area of isoniazid to that of the internal standard. areas, AT and AS, of isoniazid of the sample solution and the
standard solution.
QT
Amount (mg) of isoniazid (C6H7N3O) = WS ×
QS AT
Amount (mg) of isoniazid (C6H7N3O) = WS × ×2
AS
WS: Amount (mg) of isoniazid for assay

Internal standard solution—A solution of propyl para- WS: Amount (mg) of isoniazid for assay
hydroxybenzoate (1 in 4000). Operating conditions—
Operating conditions— Detector: An ultraviolet absorption photometer
Detector: An ultraviolet absorption photometer (wavelength: 265 nm).
(wavelength: 265 nm). Column: A stainless steel column 4.6 mm in inside di-
Column: A stainless steel column 4.6 mm in inside di- ameter and 25 cm in length, packed with octadecylsilanized
Supplement I, JP XIV O‹cial Monographs for Part I 1479

silica gel for liquid chromatography (5 mm in particle di- the standard solution.
ameter). Operating conditions—
Column temperature: A constant temperature of about Detector, column, column temperature, mobile phase,
409C. and ‰ow rate: Proceed as directed in the operating condi-
Mobile phase: Dissolve 6.80 g of potassium dihydrogen tions in the Purity (2).
phosphate in water to make 1000 mL. Separately, to 5.76 g
Purity (1) Heavy metals—Proceed with 1.0 g of Josamy-
of phosphoric acid add water to make 1000 mL. Mix these
cin according to Method 2, and perform the test. Prepare the
solutions to adjust the pH to 2.5. To 400 mL of this solution
control solution with 3.0 mL of Standard Lead Solution (not
add 600 mL of methanol, and dissolve 2.86 g of sodium
more than 30 ppm).
tridecanesulfonate in this.
(2) Related substances—Dissolve 50 mg of Josamycin in
Flow rate: Adjust the ‰ow rate so that the retention time
5 mL of methanol, add diluted methanol (1 in 2) to make 50
of isoniazid is about 5 minutes.
mL, and use this solution as the sample solution. Perform
System suitability—
the test with 10 mL of the sample solution as directed under
System performance: Dissolve 5 mg of Isoniazid and 5 mg
the Liquid Chromatography according to the following con-
of isonicotinic acid in 100 mL of the mobile phase. When the
ditions. Determine each peak area by the automatic integra-
procedure is run with 10 mL of this solution under the above
tion method, and calculate the amounts of josamycin and
operating conditions, isonicotinic acid and isoniazid are
the related substances by the area percentage method: the
eluted in this order with the resolution between these peaks
amount of any peak other than josamycin is not more than
being not less than 1.5.
6z, and the total of these peaks is not more than 20z.
System repeatability: When the test is repeated 6 times
Operating conditions—
with 10 mL of the standard solution under the above operat-
Detector: An ultraviolet absorption photometer
ing conditions, the relative standard deviation of the peak
(wavelength: 231 nm).
area of isoniazid is not more than 1.0z.
Column: A stainless steel column 4.6 mm in inside di-
ameter and 5 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Josamycin ameter).
Column temperature: A constant temperature of about
ジョサマイシン
409C.
Mobile phase: Dissolve 119 g of sodium perchlorate in
Change to read except the structural formula
water to make 1000 mL, and adjust the pH to 2.5 with 1
and chemical name:
mol/L hydrochloric acid TS. To 600 mL of this solution add
400 mL of acetonitrile.
Josamycin contains not less than 900 mg (potency)
Flow rate: Adjust the ‰ow rate so that the retention time
per mg, calculated on the dried basis. The potency of
of josamycin is about 10 minutes.
Josamycin is expressed as mass (potency) of josamycin
Time span of measurement: About 4 times as long as the
(C42H69NO15).
retention time of josamycin after the solvent peak.
Description Josamycin occurs as a white to yellowish white System suitability—
powder. Test for required detectability: Pipet 3 mL of the sample
It is very soluble in methanol and in ethanol (99.5), and solution, add diluted methanol (1 in 2) to make exactly 50
very slightly soluble in water. mL, and use this solution as the solution for system suitabil-
ity test. Pipet 2 mL of the solution for system suitability test,
Identiˆcation (1) Determine the absorption spectrum of
and add diluted methanol (1 in 2) to make exactly 20 mL.
a solution of Josamycin in methanol (1 in 100,000) as direct-
Conˆrm that the peak area of josamycin obtained from 10
ed under the Ultraviolet-visible Spectrophotometry, and
mL of this solution is equivalent to 8 to 12z of that from 10
compare the spectrum with the Reference Spectrum or the
mL of the solution for system suitability test.
spectrum of a solution of Josamycin Reference Standard
System performance: Dissolve 0.05 g of Josamycin in 50
prepared in the same manner as the sample solution: both
mL of 0.1 mol/L potassium dihydrogen phosphate TS, pH
spectra exhibit similar intensities of absorption at the same
2.0, and allow to stand at 409 C for 3 hours. Adjust the pH
wavelengths.
of this solution to 6.8 to 7.2 with 2 mol/L sodium hydroxide
(2) Dissolve 5 mg each of Josamycin and Josamycin
TS, and add 50 mL of methanol. When the procedure is run
Reference Standard in 1 mL of methanol, add diluted
with 10 mL of this solution under the above operating condi-
methanol (1 in 2) to make 100 mL, and use these solutions as
tions, the resolution between the peaks of josamycin S1,
the sample solution and the standard solution, respectively.
which relative retention time to josamycin is about 0.9, and
Perform the test with 10 mL each of the sample solution and
josamycin is not less than 2.0.
the standard solution as directed under the Liquid Chro-
System repeatability: When the test is repeated 6 times
matography according to the following conditions: the
with 10 mL of the solution for system suitability test under
retention time of the main peak obtained from the sample
the above operating conditions, the relative standard devia-
solution is the same with that of the peak of josamycin from
1480 O‹cial Monographs for Part I Supplement I, JP XIV

tion of the peak area of josamycin is not more than 1.5z. Propionate Reference Standard prepared in the same man-
ner as the sample solution: both spectra exhibit similar inten-
Loss on drying Not more than 1.0z (0.5 g, in vacuum,
sities of absorption at the same wavelengths.
phosphorus (V) oxide, 609
C, 3 hours).
(2) Dissolve 5 mg each of Josamycin Propionate and
Residue on ignition Not more than 0.10z (1 g). Josamycin Propionate Reference Standard in 50 mL of
diluted acetonitrile (1 in 2), and use these solutions as the
Assay Perform the test according to the Cylinder-plate
sample solution and the standard solution, respectively. Per-
method as directed under the Microbial Assay for Antibiot-
form the test with 10 mL each of the sample solution and the
ics according to the following conditions.
standard solution as directed under the Liquid Chro-
(1) Test organism—Bacillus subtilis ATCC 6633
matography according to the following conditions: the
(2) Culture medium—Use the medium ii in 3) Medium
retention time of the peak of josamycin propionate obtained
for other organisms under (1) Agar media for seed and base
from the sample solution is the same with that of the peak of
layer. Adjust the pH of the medium so that it will be 7.9 to
josamycin propionate from the standard solution.
8.1 after sterilization.
Operating conditions—
(3) Standard solutions—Weigh accurately an amount of
Detector, column, column temperature, mobile phase,
Josamycin Reference Standard, equivalent to about 30 mg
and ‰ow rate: Proceed as directed in the operating condi-
(potency), dissolve in 5 mL of methanol, add water to make
tions in the Purity (2).
exactly 100 mL, and use this solution as the standard stock
solution. Keep the standard stock solution at 59 C or below, Purity (1) Heavy metals—Proceed with 1.0 g of Josamy-
and use within 7 days. Take exactly a suitable amount of the cin Propionate according to Method 2, and perform the test.
standard stock solution before use, add water to make solu- Prepare the control solution with 3.0 mL of Standard Lead
tions so that each mL contains 30 mg (potency) and 7.5 mg Solution (not more than 30 ppm).
(potency), and use these solutions as the high concentration (2) Related substances—Dissolve 0.05 g of Josamycin
standard solution and the low concentration standard solu- Propionate in the mobile phase to make 50 mL, and use this
tion, respectively. solution as the sample solution. Perform the test with 10 mL
(4) Sample solutions—Weigh accurately an amount of of the sample solution as directed under the Liquid Chro-
Josamycin, equivalent to about 30 mg (potency), dissolve in matography according to the following conditions. Deter-
5 mL of methanol, and add water to make exactly 100 mL. mine each peak area by the automatic integration method,
Take exactly a suitable amount of this solution, add water to and calculate the amounts of each peak other than josamy-
make solutions so that each mL contains 30 mg (potency) and cin propionate by the area percentage method: the amount
7.5 mg (potency), and use these solutions as the high concen- of any peak other than josamycin is not more than 6z, and
tration sample solution and the low concentration sample the total of these peaks is not more than 22z.
solution, respectively. Operating conditions—
Detector: An ultraviolet absorption photometer
Containers and storage Containers—Tight containers.
(wavelength: 234 nm).
Storage—Light-resistant.
Column: A stainless steel column 6 mm in inside diameter
and 15 cm in length, packed with octadecylsilanized silica gel
for liquid chromatography (5 mm in particle diameter).
Josamycin Propionate Column temperature: A constant temperature of about
409C.
プロピオン酸ジョサマイシン
Mobile phase: To 10 mL of triethylamine add water to
make 1000 mL, and adjust the pH to 4.3 with acetic acid
Change to read except the structural formula
(100). To 500 mL of this solution add 500 mL of acetoni-
and chemical name:
trile.
Flow rate: Adjust the ‰ow rate so that the retention time
Josamycin Propionate contains not less than 843 mg
of josamycin propionate is about 24 minutes.
(potency) per mg, calculated on the dried basis. The
Time span of measurement: About 3.5 times as long as the
potency of Josamycin Propionate is expressed as mass
retention time of josamycin propionate after the solvent
(potency) of josamycin (C42H69NO15).
peak.
Description Josamycin Propionate occurs as a white to System suitability—
light yellowish white crystalline powder. Test for required detectability: Measure exactly 3 mL of
It is very soluble in acetonitrile, freely soluble in methanol the sample solution, add the mobile phase to make exactly
and in ethanol (99.5), and practically insoluble in water. 50 mL, and use this solution as the solution for system
suitability test. Measure exactly 2 mL of the solution for sys-
Identiˆcation (1) Determine the absorption spectrum of
tem suitability test, and add the mobile phase to make ex-
a solution of Josamycin Propionate in methanol (1 in
actly 20 mL. Conˆrm that the peak area of josamycin
100,000) as directed under the Ultraviolet-visible Spec-
propionate obtained from 10 mL of this solution is equiva-
trophotometry, and compare the spectrum with the Refer-
lent to 8 to 12z of that from 10 mL of the solution for sys-
ence Spectrum or the spectrum of a solution of Josamycin
Supplement I, JP XIV O‹cial Monographs for Part I 1481

tem suitability test. Add the following:


System performance: Dissolve 5 mg of josamycin pro-
pionate and 2 mg of josamycin in 50 mL of the mobile Kanamycin Monosulfate
phase. When the procedure is run with 10 mL of this solution
under the above operating conditions, josamycin and 一硫酸カナマイシン
josamycin propionate are eluted in this order with the reso-
lution between these peaks being not less than 25.
System repeatability: When the test is repeated 6 times
with 10 mL of the solution for system suitability test under
the above operating conditions, the relative standard devia-
tion of the peak area of josamycin propionate is not more
than 1.5z.

Loss on drying Not more than 1.0z (1 g, in vacuum,


phosphorus (V) oxide, 609
C, 3 hours).

Residue on ignition Not more than 0.10z (1 g).

Assay Perform the test according to the Cylinder-plate


C18H36N4O11.H2SO4: 582.58
method as directed under the Microbial Assay for Antibiot-
O-3-Amino-3-deoxy-a-D-glucopyranosyl-(1→6)-O-[6-
ics according to the following conditions.
amino-6-deoxy-a-D-glucopyranosyl-(1→4)]-2-deoxy-
(1) Test organism—Bacillus subtilis ATCC 6633
D-streptamine monosulfate [25389-94-0 ]
(2) Culture medium—Use the medium ii in 3) Medium
for other organisms under (1) Agar media for seed and base
layer. Adjust the pH of the medium so that it will be 7.9 to
Kanamycin Monosulfate contains not less than 750
8.1 after sterilization.
mg (potency) per mg, calculated on the dried basis. The
(3) Standard solutions—Weigh accurately an amount of
potency of Kanamycin Monosulfate is expressed as
Josamycin Propionate Reference Standard, equivalent to
mass (potency) of kanamycin (C18H36N4O11: 484.50).
about 20 mg (potency), dissolve in 10 mL of methanol, add Description Kanamycin Monosulfate occurs as a white
1/15 mol/L phosphate buŠer solution, pH 5.6 to make ex- crystalline powder.
actly 50 mL, and use this solution as the standard stock solu- It is freely soluble in water, and practically insoluble in
tion. Keep the standard stock solution at 59 C or below, and ethanol (99.5).
use within 3 days. Take exactly a suitable amount of the
Identiˆcation (1) Dissolve 50 mg of Kanamycin
standard stock solution before use, add 1/15 mol/L phos-
Monosulfate in 3 mL of water, and add 6 mL of anthrone
phate buŠer solution, pH 5.6 to make solutions so that each
TS: a blue-purple color develops.
mL contains 80 mg (potency) and 20 mg (potency), and use
(2) Dissolve 20 mg each of Kanamycin Monosulfate and
these solutions as the high concentration standard solution
Kanamycin Monosulfate Reference Standard in 1 mL of
and the low concentration standard solution, respectively.
water, and use these solutions as the sample solution and the
(4) Sample solutions—Weigh accurately an amount of
standard solution. Perform the test with these solutions as
Josamycin Propionate, equivalent to about 20 mg (potency),
directed under the Thin-layer Chromatography. Spot 5 mL
dissolve in 10 mL of methanol, add 1/15 mol/L phosphate
each of the sample solution and the standard solution on a
buŠer solution, pH 5.6 to make exactly 50 mL. Take exactly
plate of silica gel for thin-layer chromatography. Develop
a suitable amount of this solution, add 1/15 mol/L phos-
the plate with the supernatant layer of a mixture of chlo-
phate buŠer solution, pH 5.6 to make solutions so that each
roform, ammonia solution (28) and methanol (2:1:1) to a
mL contains 80 mg (potency) and 20 mg (potency), and use
distance of about 10 cm, and air-dry the plate. Spray evenly
these solutions as the high concentration sample solution
a solution of 0.2z ninhydrin-water saturated 1-butanol TS
and the low concentration sample solution, respectively.
on the plate, and heat at 1009 C for 10 minutes: the principal
Containers and storage Containers—Tight containers. spots obtained from the sample solution and the standard
Storage—Light-resistant. solution show a purple-brown color and the same Rf value.
(3) To a solution of Kanamycin Monosulfate (1 in 5) add
1 drop of barium chloride TS: a white precipitate is formed.

Optical rotation [a]20D : +112 – +1239(0.2 g calculated on


the dried basis, water, 20 mL, 100 mm).

Sulfuric acid Weigh accurately about 0.25 g of Kanamycin


Monosulfate, dissolve in 100 mL of water, adjust the pH to
11.0 with ammonia solution (28), add exactly 10 mL of
0.1 mol/L barium chloride VS, and titrate with 0.1 mol/L
1482 O‹cial Monographs for Part I Supplement I, JP XIV

disodium dihydrogen ethylenediamine tetraacetate VS until cy), and dissolve in water to make exactly 50 mL. Take ex-
the color of the solution, blue-purple, disappears (indicator: actly a suitable amount of this solution, add 0.1 mol/L
0.5 mg of phthalein purple). At a near of the end-point add phosphate buŠer solution, pH 8.0 to make solutions so that
50 mL of ethanol (99.5). Perform a blank determination in each mL contains 20 mg (potency) and 5 mg (potency), and
the same manner. The amount of sulfuric acid (SO4) is not use these solutions as the high concentration sample solution
less than 15.0z and not more than 17.0z, calculated on the and the low concentration sample solution, respectively.
dried basis.
Containers and storage Containers—Well-closed contain-
Each mL of 0.1 mol/L barium chloride VS ers.
= 9.606 mg of SO4

Purity (1) Heavy metals—Proceed with 2.0 g of Kanamycin Sulfate


Kanamycin Monosulfate according to Method 4, and per-
form the test. Prepare the control solution with 2.0 mL of 硫酸カナマイシン
Standard Lead Solution (not more than 10 ppm).
(2) Arsenic—Prepare the test solution with 2.0 g of Change to read except the structural formula
Kanamycin Monosulfate according to Method 4, and per- and chemical name:
form the test (not more than 1 ppm).
(3) Related substances—Dissolve 0.30 g of Kanamycin Kanamycin Sulfate contains not less than 690 mg
Monosulfate in water to make exactly 10 mL, and use this (potency) per mg, calculated on the dried basis. The
solution as the sample solution. Separately, dissolve 45 mg potency of Kanamycin Sulfate is expressed as mass
of Kanamycin Monosulfate Reference Standard in water to (potency) of kanamycin (C18H36N4O11: 484.50).
make exactly 50 mL, and use this solution as the standard
Description Kanamycin Sulfate occurs as a white to yel-
solution. Perform the test with these solutions as directed
lowish white powder.
under the Thin-layer Chromatography. Spot 1 mL each of
It is very soluble in water, and practically insoluble in
the sample solution and the standard solution on a plate of
ethanol (99.5).
silica gel for thin-layer chromatography. Develop the plate
with a solution of potassium dihydrogen phosphate (3 in 40) Identiˆcation (1) Dissolve 20 mg each of Kanamycin Sul-
to a distance of about 10 cm, and air-dry the plate. Spray fate and Kanamycin Monosulfate Reference Standard in 1
evenly a solution of ninhydrin in 1-butanol (1 in 100) on the mL of water, and use these solutions as the sample solution
plate, and heat at 1109 C for 10 minutes: the spot other than and the standard solution. Perform the test with these solu-
the principal spot obtained from the sample solution is not tions as directed under the Thin-layer Chromatography.
more intense than the spot from the standard solution. Spot 5 mL each of the sample solution and the standard solu-
tion on a plate of silica gel for thin-layer chromatography.
Loss on drying Not more than 4.0z (5 g, reduced pressure
Develop the plate with a mixture of chloroform, ammonia
not exceeding 0.67 kPa, 609C, 3 hours).
solution (28) and methanol (2:1:1) to a distance of about 10
Residue on ignition Not more than 0.5z (1 g). cm, and air-dry the plate. Spray evenly 0.2z ninhydrin-
water saturated 1-butanol TS on the plate, and heat at 1009C
Assay Perform the test according to the Cylinder-plate
for 10 minutes: the principal spots obtained from the sample
method as directed under the Microbial Assay for Antibiot-
solution and the standard solution show a purple-brown
ics according to the following conditions.
color and the same Rf value.
(1) Test organism—Bacillus subtilis ATCC 6633
(2) A solution of Kanamycin Sulfate (1 in 10) responds
(2) Culture medium—Use the medium i in 1) Medium
to the Qualitative Test (1) for sulfate.
for test organism [5] under (1) Agar media for seed and base
layer. Optical rotation [a]20D : +103 – +1159(0.5 g calculated on

(3) Standard solutions—Weigh accurately an amount of the dried basis, water, 50 mL, 100 mm).
Kanamycin Monosulfate Reference Standard, previously
pH The pH of a solution obtained by dissolving 1.0 g of
dried, equivalent to about 20 mg (potency), dissolve in dilut-
Kanamycin Sulfate in 20 mL of water is between 6.0 and 7.5.
ed phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly
50 mL, and use this solution as the standard stock solution. Purity (1) Clarity and color of solution—Dissolve 1.5 g
Keep the standard stock solution between 5 and 159 C and of Kanamycin Sulfate in 5 mL of water: the solution is clear
use within 30 days. Take exactly a suitable amount of the and colorless to pale yellow.
standard stock solution before use, add 0.1 mol/L phos- (2) Heavy metals—Proceed with 1.0 g of Kanamycin
phate buŠer solution, pH 8.0 to make solutions so that each Sulfate according to Method 4, and perform the test. Pre-
mL contains 20 mg (potency) and 5 mg (potency), and use pare the control solution with 3.0 mL of Standard Lead So-
these solutions as the high concentration standard solution lution (not more than 30 ppm).
and the low concentration standard solution, respectively. (3) Arsenic—Prepare the test solution with 2.0 g of
(4) Sample solutions—Weigh accurately an amount of Kanamycin Sulfate according to Method 3, and perform the
Kanamycin Monosulfate, equivalent to about 20 mg (poten- test (not more than 1 ppm).
Supplement I, JP XIV O‹cial Monographs for Part I 1483

(4) Related substances—Dissolve 0.30 g of Kanamycin Add the following:


Sulfate in water to make exactly 10 mL, and use this solution
as the sample solution. Separately, dissolve 9.0 mg of Ketotifen Fumarate
Kanamycin Monosulfate Reference Standard in water to
make exactly 10 mL, and use this solution as the standard フマル酸ケトチフェン
solution. Perform the test with these solutions as directed
under the Thin-layer Chromatography. Spot 1 mL each of
the sample solution and the standard solution on a plate of
silica gel for thin-layer chromatography. Develop the plate
with a solution of potassium dihydrogen phosphate (3 in 40)
to a distance of about 10 cm, and air-dry the plate. Spray
evenly a solution of ninhydrin in 1-butanol (1 in 100) on the
plate, and heat at 1109 C for 10 minutes: the spot other than
the principal spot obtained from the sample solution is not C19H19NOS.C4H4O4: 425.50
more intense than the spot from the standard solution. 4-(1-Methylpiperidin-4-ylidene)-4H-
benzo[4,5]cyclohepta[1,2-b]thiophen-10(9H )-one
Loss on drying Not more than 5.0z (0.5 g, reduced pres-
monofumarate [34580-14-8 ]
sure not exceeding 0.67 kPa, 609
C, 3 hours).

Assay Perform the test according to the Cylinder-plate Ketotifen Fumarate, when dried, contains not less
method as directed under the Microbial Assay for Antibiot- than 99.0z and not more than 101.0z of ketotifen
ics according to the following conditions. fumarate (C19H19NOS.C4H4O4).
(1) Test organism—Bacillus subtilis ATCC 6633
Description Ketotifen Fumarate occurs as a white to light
(2) Culture medium—Use the medium i in 1) Medium
yellowish white crystalline powder.
for test organism [5] under (1) Agar media for seed and base
It is sparingly soluble in methanol and in acetic acid (100),
layer having pH 7.8 to 8.0 after sterilization.
and slightly soluble in water, in ethanol (99.5) and in acetic
(3) Standard solutions—Weigh accurately an amount of
anhydride.
Kanamycin Monosulfate Reference Standard, previously d-
Melting point: about 1909 C (with decomposition).
ried, equivalent to about 20 mg (potency), dissolve in diluted
phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly 50 Identiˆcation (1) Prepare the test solution with 0.03 g of
mL, and use this solution as the standard stock solution. Ketotifen Fumarate as directed under the Oxygen Flask
Keep the standard stock solution at 5 to 159 C and use within Combustion Method using 20 mL of water as the absorbing
30 days. Take exactly a suitable amount of the standard liquid: the test solution responds to the Qualitative Tests for
stock solution before use, add 0.1 mol/L phosphate buŠer sulfate.
solution, pH 8.0 to make solutions so that each mL contains (2) Determine the absorption spectrum of a solution of
20 mg (potency) and 5 mg (potency), and use these solutions Ketotifen Fumarate in methanol (1 in 50,000) as directed un-
as the high concentration standard solution and the low con- der the Ultraviolet-visible Spectrophotometry, and compare
centration standard solution, respectively. the spectrum with the Reference Spectrum: both spectra ex-
(4) Sample solutions—Weigh accurately an amount of hibit similar intensities of absorption at the same
Kanamycin Sulfate, equivalent to about 20 mg (potency), wavelengths.
and dissolve in water to make exactly 50 mL. Take exactly a (3) Determine the infrared absorption spectrum of
suitable amount of this solution, add 0.1 mol/L phosphate Ketotifen Fumarate, previously dried, as directed in the
buŠer solution, pH 8.0 to make solutions so that each mL potassium bromide disk method under the Infrared Spec-
contains 20 mg (potency) and 5 mg (potency), and use these trophotometry, and compare the spectrum with the Refer-
solutions as the high concentration sample solution and the ence Spectrum: both spectra exhibit similar intensities of ab-
low concentration sample solution, respectively. sorption at the same wave numbers.

Containers and storage Containers—Tight containers. Purity (1) Chloride—Dissolve 0.6 g of Ketotifen
Fumarate in 2.5 mL of sodium carbonate TS in a crucible,
heat on a water bath to dryness, and ignite at about 5009 C.
Dissolve the residue in 15 mL of water, ˆlter if necessary,
neutralize with diluted nitric acid (3 in 10), and add 6 mL of
dilute nitric acid and water to make 50 mL. Perform the test
using this solution as the test solution. Prepare the control
solution as follows: To 0.25 mL of 0.01 mol/L hydrochloric
acid VS add 2.5 mL of sodium carbonate TS, the used
amount of diluted nitric acid (3 in 10) for the neutralization,
6 mL of dilute nitric acid and water to make 50 mL (not
more than 0.015z).
1484 O‹cial Monographs for Part I Supplement I, JP XIV

(2) Heavy metals—Proceed with 1.0 g of Ketotifen Operating conditions—


Fumarate according to Method 2, and perform the test. Pre- Detector: An ultraviolet absorption photometer
pare the control solution with 2.0 mL of Standard Lead So- (wavelength: 232 nm).
lution (not more than 20 ppm). Column: A stainless steel column 4.0 mm in inside di-
(3) Related substances—Dissolve 0.10 g of Ketotifen ameter and 15 cm in length, packed with octylsilanized silica
Fumarate in 10 mL of a mixture of methanol and ammonia gel for liquid chromatography (5 mm in particle diameter).
TS (99:1), and use this solution as the sample solution. Pipet Column temperature: A constant temperature of about
1 mL of the sample solution, and add a mixture of methanol 409C.
and ammonia TS (99:1) to make exactly 25 mL. Pipet 1 mL Mobile phase: To a volume of a solution of ammonium
of this solution, add a mixture of methanol and ammonia TS acetate (77 in 500) add diluted phosphoric acid (1 in 150) to
(99:1) to make exactly 20 mL, and use this solution as the adjust to pH 5.5. To 370 mL of this solution add 580 mL of
standard solution. Perform the test with these solutions as methanol and 50 mL of acetonitrile.
directed under the Thin-layer Chromatography. Spot 10 mL Flow rate: Adjust the ‰ow rate so that the retention time
each of the sample solution and the standard solution on a of leucomycin A5 is about 8 minutes.
plate of silica gel for thin-layer chromatography. Develop Time span of measurement: About 3 times as long as the
the plate with a mixture of acetonitrile, water and ammonia retention time of leucomycin A5.
solution (28) (90:10:1) to a distance of about 15 cm, and air- System suitability—
dry the plate. Spray evenly DragendorŠ's TS for spraying System performance: Dissolve about 20 mg each of Leu-
and then hydrogen peroxide TS on the plate: the number of comycin A5 Reference Standard and Josamycin Reference
the spot other than the principal spot obtained from the Standard in 20 mL of diluted acetonitrile (1 in 2). When the
sample solution is not more than four, and they are not more procedure is run with 5 mL of this solution under the above
intense than the spot from the standard solution. operating conditions, leucomycin A5 and josamycin are elut-
ed in this order with the resolution between these peaks
Loss on drying Not more than 0.5z (1 g, 1059
C, 4 hours).
being not less than 5.
Residue on ignition Not more than 0.10z (1 g). System repeatability: When the test is repeated 6 times
with 5 mL of the sample solution under the above operating
Assay Weigh accurately about 0.35 g of Ketotifen
conditions, the relative standard deviation of the peak area
Fumarate, previously dried, dissolve in 80 mL of a mixture
of leucomycin A5 is not more than 1.0z.
of acetic anhydride and acetic acid (100) (7:3), and titrate
with 0.1 mol/L perchloric acid VS (potentiometric titra-
Change the Assay (3) to read:
tion). Perform a blank determination, and make any neces-
sary correction. Assay
(3) Standard solutions—Weigh accurately an amount of
Each mL of 0.1 mol/L perchloric acid VS
Leucomycin A5 Reference Standard equivalent to about
= 42.55 mg of C19H19NOS.C4H4O4
30 mg (potency), dissolve in 10 mL of methanol, add water
to make exactly 100 mL, and use this solution as the stan-
Containers and storage Containers—Tight containers.
dard stock solution. Keep the standard stock solution at 59
C
or below and use within 3 days. Take exactly a suitable
amount of the standard stock solution before use, add phos-
Kitasamycin phate buŠer solution, pH 8.0 to make solutions so that each
mL contains 30 mg (potency) and 7.5 mg (potency), and use
キタサマイシン
these solutions as the high concentration standard solution
and the low concentration standard solution, respectively.
Change the Content ratio of the active principle
to read:
Content ratio of the active principle Dissolve 0.02 g of
Kitasamycin in diluted acetonitrile (1 in 2) to make 20 mL,
and use this solution as the sample solution. Perform the test
with 5 mL of the sample solution as directed under the Liquid
Chromatography according to the following conditions, and
measure each peak area by the automatic integration
method. Calculate the amounts of leucomycin A5, leucomy-
cin A4 and leucomycin A1 by the area percentage method:
the amounts of leucomycin A5, leucomycin A4 and leucomy-
cin A1 are 40 to 70z, 5 to 25z and 3 to 12z, respectively.
Relative retention times of leucomycin A4 and leucomycin
A1 to that of leucomycin A5 are 1.2 and 1.5, respectively.
Supplement I, JP XIV O‹cial Monographs for Part I 1485

Add the following: Kitasamycin Tartrate contains not less than 1300 mg
(potency) per mg, calculated on the anhydrous basis.
Kitasamycin Tartrate The potency of Kitasamycin Tartrate is expressed as
mass (potency) of kitasamycin based on the amount of
Leucomycin Tartrate leucomycin A5 (C39H65NO14: 771.93). 1 mg (potency)
of Kitasamycin Tartrate is equivalent to 0.530 mg of
酒石酸キタサマイシン
leucomycin A5 (C39H65NO14).
Description Kitasamycin Tartrate occurs as a white to light
yellowish white powder.
It is very soluble in water, in methanol and in ethanol
(99.5).

Identiˆcation (1) Determine the absorption spectrum of


a solution of Kitasamycin Tartrate in methanol (1 in 40,000)
as directed under the Ultraviolet-visible Spectrophotometry,
and compare the spectrum with the Reference Spectrum:
both spectra exhibit similar intensities of absorption at the
same wavelengths.
(2) Determine the infrared absorption spectrum of
Kitasamycin Tartrate as directed in the potassium bromide
disk method under the Infrared Spectrophotometry, and
compare the spectrum with the Reference Spectrum: both
spectra exhibit similar intensities of absorption at the same
wave numbers.
(3) Dissolve 1 g of Kitasamycin Tartrate in 20 mL of
water, add 3 mL of sodium hydroxide TS, add 20 mL of n-
butyl acetate, shake well, and discard the n-butyl acetate lay-
er. To the aqueous layer add 20 mL of n-butyl acetate, and
shake well. The aqueous layer so obtained responds to the
Qualitative Test (1) for tartrate.

pH Dissolve 3.0 g of Kitasamycin Tartrate in 100 mL of


water: the pH of the solution is between 3.0 and 5.0.
(Leucomycin A1, A5, A7, A9, A13 tartrate)
Content ratio of the active principle Dissolve 20 mg of
(3R,4R,5S,6R,8R,9R,10E,12E,15R )-5-[O-(4-O-Acyl-
Kitasamycin Tartrate in diluted acetonitrile (1 in 2) to make
2,6-dideoxy-3-C-methyl-a-L-ribo-hexopyranosyl)-(1→4)-
20 mL, and use this solution as the sample solution. Perform
3,6-dideoxy-3-dimethylamino-b-D-glucopyranosyloxy]-6-
the test with 5 mL of the sample solution as directed under
formylmethyl-3,9-dihydroxy-4-methoxy-8-methylhexadeca-
the Liquid Chromatography according to the following con-
10,12-dien-15-olide mono-(2R,3R )-tartrate
ditions, determine the peak areas by the automatic integra-
Leucomycin A1 tartrate: acyl = 3-methylbutanoyl
tion method, and calculate the amounts of leucomycin A5,
Leucomycin A5 tartrate: acyl = butanoyl
leucomycin A4 and leucomycin A1 by the area percentage
Leucomycin A7 tartrate: acyl = propanoyl
method: the amount of leucomycin A5 is 40 – 70z, leu-
Leucomycin A9 tartrate: acyl = acetyl
comycin A4 is 5 – 25z, and leucomycin A1 is 3 – 12z. The
Leucomycin A13 tartrate: acyl = hexanoyl
relative retention times of leucomycin A 4 and leucomycin A1
with respect to leucomycin A5 are 1.2 and 1.5, respectively.
(Leucomycin A3, A4, A6, A8 tartrate)
Operating conditions—
(3R,4R,5S,6R,8R,9R,10E,12E,15R )-3-Acetoxy-5-[O-(4-O-
Detector: An ultraviolet absorption photometer
acyl-2,6-dideoxy-3-C-methyl-a-L-ribo-hexopyranosyl)-
(wavelength: 232 nm).
(1→4)-3,6-dideoxy-3-dimethylamino-b-D-
Column: A stainless steel column 4.0 mm in inside di-
glucopyranosyloxy]-6-formylmethyl-9-hydroxy-
ameter and 15 cm in length, packed with octadecylsilanized
4-methoxy-8-methylhexadeca-10,12-dien-15-olide
silica gel for liquid chromatography (5 mm in particle di-
mono-(2R,3R )-tartrate
ameter).
Leucomycin A3 tartrate: acyl = 3-methylbutanoyl
Column temperature: A constant temperature of about
Leucomycin A4 tartrate: acyl = butanoyl
409C.
Leucomycin A6 tartrate: acyl = propanoyl
Mobile phase: To a suitable amount of a solution of am-
Leucomycin A8 tartrate: acyl = acetyl
monium acetate (77 in 500) add diluted phosphoric acid (1 in
[37280-56-1 ]
150) to adjust the pH to 5.5. To 370 mL of this solution add
580 mL of methanol and 50 mL of acetonitrile.
1486 O‹cial Monographs for Part I Supplement I, JP XIV

Flow rate: Adjust the ‰ow rate so that the retention time Latamoxef Sodium
of leucomycin A5 is about 8 minutes.
Time span of measurement: About 3 times as long as the ラタモキセフナトリウム
retention time of leucomycin A5.
System suitability— Change to read except the structural formula
System performance: Dissolve about 20 mg of Leucomy- and chemical name:
cin A5 Reference Standard and about 20 mg of Josamycin
Reference Standard in 20 mL of diluted acetonitrile (1 in 2). Latamoxef Sodium contains not less than 830 mg
When the procedure is run with 5 mL of this solution under (potency) per mg, calculated on the anhydrous basis.
the above operating conditions, leucomycin A5 and josamy- The potency of Latamoxef Sodium is expressed as
cin are eluted in this order with the resolution between these mass (potency) of latamoxef (C20H20N6O9S: 520.47).
peaks being not less than 5.
Description Latamoxef Sodium occurs as white to light
System repeatability: When the test is repeated 6 times
yellowish white, powder or masses.
with 5 mL of the sample solution under the above operating
It is very soluble in water, freely soluble in methanol, and
conditions, the relative standard deviation of the peak area
slightly soluble in ethanol (95).
of leucomycin A5 is not more than 1.0z.
Identiˆcation (1) Determine the absorption spectrum of
Purity (1) Clarity and color of solution—Dissolve 1.0 g
a solution of Latamoxef Sodium (3 in 100,000) as directed
of Kitasamycin Tartrate in 10 mL of water: the solution is
under the Ultraviolet-visible Spectrophotometry, and com-
clear and colorless or light yellow.
pare the spectrum with the Reference Spectrum: both spec-
(2) Heavy metals—Proceed with 1.0 g of Kitasamycin
tra exhibit similar intensities of absorption at the same
Tartrate according to Method 2, and perform the test. Pre-
wavelengths.
pare the control solution with 3.0 mL of Standard Lead So-
(2) Determine the infrared absorption spectrum of
lution (not more than 30 ppm).
Latamoxef Sodium as directed in the potassium bromide
Water Not more than 3.0z (0.1 g, volumetric titration, disk method under the Infrared Spectrophotometry, and
direct titration). compare the spectrum with the Reference Spectrum: both
spectra exhibit similar intensities of absorption at the same
Assay Perform the test according to the Cylinder-plate
wave numbers.
method as directed under the Microbial Assay for Antibiot-
(3) Determine the spectrum of a solution of Latamoxef
ics according to the following conditions.
Sodium in heavy water for nuclear magnetic resonance spec-
(1) Test organism—Bacillus subtilis ATCC 6633
troscopy (1 in 10) as directed under the Nuclear Magnetic
(2) Culture medium—Use the medium i in 1) Medium
Resonance Spectroscopy (1H), using sodium 3-trimethylsilyl-
for test organism [5] under (1) Agar media for seed and base
propanesulfonate for nuclear magnetic resonance spec-
layer.
troscopy as an internal reference compound: it exhibits sin-
(3) Standard solutions—Weigh accurately an amount of
gle signals, A and B, at around d 3.5 ppm and at around
Leucomycin A5 Reference Standard, equivalent to about 30
d 4.0 ppm. The ratio of the integrated intensity of these sig-
mg (potency), dissolve in 10 mL of methanol, add water to
nals, A:B, is about 1:1.
make exactly 100 mL, and use this solution as the standard
(4) Latamoxef Sodium responds to the Qualitative Test
stock solution. Keep the standard stock solution at not ex-
(1) for sodium salt.
ceeding 59 C, and use within 3 days. Take exactly a suitable
amount of the standard stock solution before use, add phos- Optical rotation [a]20D : -32 – -409(0.5 g calculated on

phate buŠer solution, pH 8.0 to make solutions so that each the anhydrous basis, phosphate buŠer solutiuon, pH 7.0, 50
mL contains 30 mg (potency) and 7.5 mg (potency), and use mL, 100 mm).
these solutions as the high concentration standard solution
pH The pH of a solution obtained by dissolving 1.0 g of
and the low concentration standard solution, respectively.
Latamoxef Sodium in 10 mL of water is between 5.0 and
(4) Sample solutions—Weigh accurately an amount of
7.0.
Kitasamycin Tartrate, equivalent to about 30 mg (potency),
and dissolve in water to make exactly 100 mL. Take exactly a Purity (1) Clarity and color of solution—Dissolve 1.0 g
suitable amount of this solution, add phosphate buŠer solu- of Latamoxef Sodium in 10 mL of water: the solution is
tion, pH 8.0 to make solutions so that each mL contains 30 clear and pale yellow.
mg (potency) and 7.5 mg (potency), and use these solutions as (2) Heavy metals—Carbonize 1.0 g of Latamoxef Sodi-
the high concentration sample solution and the low concen- um by heating gently, previously powdered if it is masses.
tration sample solution, respectively. After cooling, add 10 mL of a solution of magnesium nitrate
hexahydrate in ethanol (1 in 10), and burn the ethanol. After
Containers and storage Containers—Tight containers.
cooling, add 1 mL of sulfuric acid. Proceed according to
Method 4, and perform the test. Prepare the control solution
with 2.0 mL of Standard Lead Solution (not more than 20
ppm).
Supplement I, JP XIV O‹cial Monographs for Part I 1487

(3) Arsenic—Prepare the test solution by dissolving System suitability—


1.0 g of Latamoxef Sodium in 20 mL of water, and perform System performance: When the procedure is run with 5 mL
the test (not more than 2 ppm). of the sample solution under the above operating conditions,
(4) Related substances—Dissolve an amount of the resolution between the two peaks of latamoxef is not less
Latamoxef Sodium, equivalent to about 25 mg (potency), in than 3.
water to make exactly 50 mL, and use this solution as the System repeatability: When the test is repeated 3 times
sample solution. Pipet 2 mL of the sample solution, add with 5 mL of the sample solution under the above operating
water to make exactly 100 mL, and use this solution as the conditions, the relative standard deviation of the area of the
standard solution. Perform the test with exactly 5 mL each of ˆrst eluted peak of latamoxef is not more than 2.0z.
the sample solution and the standard solution as directed un-
Assay Weigh accurately an amount of Latamoxef Sodium
der the Liquid Chromatography according to the following
and Latamoxef Ammonium Reference Standard, equivalent
conditions, and determine each peak area by the automatic
to about 25 mg (potency) each, dissolve in exactly 5 mL of
integration method: the peak area of 1-methyl-1H-tetrazole-
the internal standard solution, add water to make 50 mL,
5-thiol, having the relative retention time of about 0.5 with
and use these solutions as the sample solution and the stan-
respect to the ˆrst eluted peak of the two peaks of latamox-
dard solution. Perform the test with 5 mL each of the sample
ef, obtained from the sample solution is not more than the
solution and the standard solution as directed under the Liq-
peak area of latamoxef from the standard solution, and the
uid Chromatography according to the following conditions,
peak area of decarboxylatamoxef, having the relative reten-
and determine the ratios, Q T and QS, of the peak area of
tion time of about 1.7 with respect to the ˆrst peak of the
latamoxef to that of the internal standard.
two peaks of latamoxef, is not more than 2 times that of
latamoxef from the standard solution. For this calculation, Amount [ mg (potency)] of latamoxef (C20H20N6O9S)
use the peak area for 1-methyl-1H-tetrazole-5-thiol after Q
= WS × T × 1000
multiplying by its response factor, 0.52. QS
Operating conditions—
WS: Amount [mg (potency)] of Latamoxef Ammonium
Proceed as directed in the operating conditions in the
Reference Standard
Assay.
System suitability— Internal standard solution—A solution of m-cresol (3 in
System performance: Proceed as directed in the system 200).
suitability in the Assay. Operating conditions—
System repeatability: When the test is repeated 6 times Detector: An ultraviolet absorption photometer
with 5 mL of the standard solution under the above operat- (wavelength: 254 nm).
ing conditions, the relative standard deviation of the peak Column: A stainless steel column 4 mm in inside diameter
area of latamoxef is not more than 2.0z. and 15 cm in length, packed with octadecylsilanized silica gel
for liquid chromatography (10 mm in particle diameter).
Water Not more than 5.0z (0.5 g, volumetric titration,
Column temperature: A constant temperature of about
back titration).
259C.
Isomer ratio Dissolve 25 mg of Latamoxef Sodium in Mobile phase: Dissolve 6.94 g of potassium dihydrogen
water to make 50 mL, and use this solution as the sample so- phosphate, 3.22 g of disodium hydrogen phosphate 12-water
lution. Perform the test with 5 mL of the sample solution as and 1.60 g of tetra n-butylammonium bromide in water to
directed under the Liquid Chromatography according to the make exactly 1000 mL. To 750 mL of this solution add
following conditions, and determine the areas, Aa and Ab, of 250 mL of methanol.
the two peaks in order of elution, which appear close to each Flow rate: Adjust the ‰ow rate so that the retention time
other at the retention time of about 10 minutes: Aa/Ab is be- of latamoxef is about 7 minutes.
tween 0.8 and 1.4. System suitability—
Operating conditions— System performance: When the procedure is run with 5 mL
Detector: An ultraviolet absorption photometer of the standard solution under the above operating condi-
(wavelength: 254 nm). tions, latamoxef and the internal standard are eluted in this
Column: A stainless steel column 4 mm in inside diameter order with the resolution between these peaks being not less
and 15 cm in length, packed with octadecylsilanized silica gel than 5.
for liquid chromatography (10 mm in particle diameter). System repeatability: When the test is repeated 6 times
Column temperature: A constant temperature of about with 5 mL of the standard solution under the above operat-
259C. ing conditions, the relative standard deviation of the ratios
Mobile phase: Dissolve 7.7 g of ammonium acetate in of the peak area of latamoxef to that of the internal standard
water to make 1000 mL. To 950 mL of this solution add is not more than 1.0z.
50 mL of methanol.
Containers and storage Containers—Tight containers.
Flow rate: Adjust the ‰ow rate so that the retention time
Storage—Not exceeding 59C.
of the ˆrst eluted peak of latamoxef is about 8 minutes.
1488 O‹cial Monographs for Part I Supplement I, JP XIV

Add the following: solution. The sample solution should be used to the follow-
ing test immediately after the solution is prepared. Perform
Lenampicillin Hydrochloride the test with 10 mL each of the sample solution and the stan-
dard solution as directed under the Liquid Chromatography
塩酸レナンピシリン according to the following conditions, and determine the ra-
tios, Q T and QS, of the peak height of ampicillin to that of
the internal standard: the amount of ampicillin is not more
than 1.0z.

Amount (z) of ampicillin (C16H19N3O4S)


W Q
= S× T×2
WT QS
C21H23N3O7S.HCl: 497.95
5-Methyl-2-oxo[1,3]dioxol-4-ylmethyl (2S,5R,6R )-6- WS: Amount [mg (potency)] of Ampicillin Reference
[(2R )-2-amino-2-phenylacetylamino]-3,3-dimethyl-7- Standard
oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylate WT: Amount (mg) of the sample
monohydrochloride [80734-02-7 ]
Internal standard solution—A solution of anhydrous
caŠeine in the mobile phase (1 in 50,000).
Lenampicillin Hydrochloride is the hydrochloride of
Operating conditions—
ampicillin methyloxodioxolenylmethyl ester.
Detector: An ultraviolet absorption photometer
It contains not less than 653 mg (potency) per mg,
(wavelength: 230 nm).
calculated on the anhydrous basis and corrected by the
Column: A stainless steel column 4 mm in inside diameter
amount of the residual solvents. The potency of
and 30 cm in length, packed with octadecylsilanized silica gel
Lenampicillin Hydrochloride is expressed as mass
for liquid chromatography (10 mm in particle diameter).
(potency) of ampicillin (C16H19N3O4S: 349.40).
Column temperature: A constant temperature of about
Description Lenampicillin Hydrochloride occurs as a white 259C.
to light yellowish white powder. Mobile phase: Dissolve 1.22 g of potassium dihydrogen
It is very soluble in water, in methanol and in ethanol (95), phosphate in water to make 900 mL, and add 100 mL of
and freely soluble in N, N-dimethylformamide. acetonitrile.
Flow rate: Adjust the ‰ow rate so that the retention time
Identiˆcation (1) Determine the infrared absorption
of ampicillin is about 7 minutes.
spectrum of Lenampicillin Hydrochloride as directed in the
System suitability—
potassium chloride disk method under the Infrared Spec-
System performance: When the procedure is run with 10
trophotometry, and compare the spectrum with the Refer-
mL of the standard solution under the above operating con-
ence Spectrum or the spectrum of Lenampicillin Hydrochlo-
ditions, ampicillin and the internal standard are eluted in
ride Reference Standard: both spectra exhibit similar intensi-
this order with the resolution between these peaks being not
ties of absorption at the same wave numbers.
less than 5.
(2) To 1 mL of a solution of Lenampicillin Hydrochlo-
System repeatability: When the test is repeated 6 times
ride (1 in 100) add 0.5 mL of dilute nitric acid and 1 drop of
with 10 mL of the standard solution under the above operat-
silver nitrate TS: a white precipitate is formed.
ing conditions, the relative standard deviation of the ratios
Optical rotation [a]20
D : +174 – +1949(0.2 g calculated on of the peak height of ampicillin to that of the internal stan-
the anhydrous de-residual solventization basis, ethanol (95), dard is not more than 5z.
20 mL, 100 mm). (4) Penicilloic acid—Weigh accurately about 0.1 g of
Lenampicillin Hydrochloride, dissolve in water to make ex-
Purity (1) Heavy metals—Proceed with 2.0 g of Lenam-
actly 100 mL, and use this solution as the sample solution.
picillin Hydrochloride according to Method 2, and perform
Pipet 10 mL of the sample solution, add 10 mL of potassium
the test. Prepare the control solution with 2.0 mL of Stan-
hydrogen phthalate buŠer solution, pH 4.6 and exactly 10
dard Lead Solution (not more than 10 ppm).
mL of 0.005 mol/L iodine VS, allow to stand for exactly 15
(2) Arsenic—Prepare the test solution with 1.0 g of
minutes while protecting from exposure to light, and titrate
Lenampicillin Hydrochloride according to Method 3, and
with 0.01 mol/L sodium thiosulfate VS (indicator: 1 mL of
perform the test (not more than 2 ppm).
starch TS). Perform a blank determination, and make any
(3) Free ampicillin—Weigh accurately about 0.1 g of
necessary correction: the amount of penicilloic acid
Lenampicillin Hydrochloridein, dissolve in exactly 10 mL of
(C16H21N3O5S: 367.42) is not more than 3.0z.
the internal standard solution, and use this solution as the
sample solution. Separately, weigh accurately an amount of Each mL of 0.01 mol/L sodium thiosulfate VS
Ampicillin Reference Standard, equivalent to about 25 mg = 0.45 mg of C16H21N3O5S
(potency), and dissolve in water to make exactly 100 mL.
(5) Residual solvent—Weigh accurately about 0.25 g of
Pipet 2 mL of this solution, add exactly 10 mL of the inter-
Lenampicillin Hydrochloride, dissolve in exactly 1 mL of
nal standard solution, and use this solution as the standard
Supplement I, JP XIV O‹cial Monographs for Part I 1489

the internal standard solution, add N,N-dimethylformamide of the peak height of ethyl acetate to that of the internal
to make 5 mL, and use this solution as the sample solution. standard is not more than 5z.
Separately, weigh accurately about 80 mg of 2-propanol and
Water Not more than 1.5z (1 g, volumetric titration,
about 0.12 g of ethyl acetate, and add N,N-dimethylfor-
direct titration).
mamide to make exactly 100 mL. Pipet 1 mL and 3 mL of
this solution, add exactly 1 mL each of the internal standard Residue on ignition Not more than 0.2z (1 g).
solution, add N,N-dimethylformamide to make 5 mL, and
Assay Weigh accurately an amount of Lenampicillin
use these solutions as the standard solution (1) and the stan-
Hydrochloride and Lenampicillin Hydrochloride Reference
dard solution (2), respectively. Perform the test with 4 mL
Standard, equivalent to about 0.1 g (potency), dissolve each
each of the sample solution, the standard solution (1) and
in the internal standard solution to make exactly 10 mL, and
the standard solution (2) as directed under the Gas Chro-
use these solutions as the sample solution and the standard
matography according to the following conditions, and de-
solution. Perform the test with 5 mL each of the sample solu-
termine the ratios, Q Ta and Q Tb, of the peak height of 2-
tion and the standard solution as directed under the Liquid
propanol and ethyl acetate to that of the internal standard of
Chromatography according to the following conditions, and
the sample solution, the ratios, QSa1 and QSb1, of the peak
determine the ratios, Q T and QS, of the peak area of lenam-
height of 2-propanol and ethyl acetate to that of the internal
picillin to that of the internal standard.
standard of the standard solution (1) and the ratios, QSa2 and
QSb2, of the peak height of 2-propanol and ethyl acetate to Amount [ mg (potency)] of ampicillin (C16H19N3O4S)
that of the internal standard of the standard solution (2). Q
= WS × T × 1000
Calculate the amounts of 2-propanol and ethyl acetate by QS
the following equations: not more than 0.7z and not more
WS: Amount [mg (potency)] of Lenampicillin Hydrochlo-
than 1.7z, respectively.
ride Reference Standard
Amount (z) of 2-propanol
Internal standard solution—A solution of ethyl aminoben-
W 2Q Ta - 3QSa1 + QSa2
= Sa × zoate in the mobile phase (1 in 4000).
WT QSa2 - QSa1
Operating conditions—
Amount (z) of ethyl acetate Detector: An ultraviolet absorption photometer
W 2Q Tb - 3QSb1 + QSb2 (wavelength: 254 nm).
= Sb ×
WT QSb2 - QSb1 Column: A stainless steel column 6 mm in inside diameter
and 15 cm in length, packed with octadecylsilanized silica gel
WSa: Amount (g) of 2-propanol
for liquid chromatography (5 mm in particle diameter).
WSb: Amount (g) of ethyl acetate
Column temperature: A constant temperature of about
WT: Amount (g) of the sample
259C.
Internal standard solution—A solution of cyclohexane in Mobile phase: Dissolve 9.53 g of potassium dihydrogen
N,N-dimethylformamide (1 in 1000). phosphate in water to make exactly 700 mL, and add
Operating conditions— acetonitrile to make exactly 1000 mL.
Detector: A hydrogen ‰ame-ionization detector. Flow rate: Adjust the ‰ow rate so that the retention time
Column: A glass column 3 mm in inside diameter and 3 m of lenampicillin is about 6 minutes.
in length, packed with siliceous earth for gas chromato- System suitability—
graphy (180 – 250 mm in particle diameter) coated with System performance: When the procedure is run with 5 mL
tetrakishydroxypropylethylenediamine for gas chromato- of the standard solution under the above operating condi-
graphy at the ratio of 10 to 15z. tions, lenampicillin and the internal standard are eluted in
Column temperature: A constant temperature of about this order with the resolution between these peaks being not
809C. less than 10.
Injection port temperature: A constant temperature of System repeatability: When the test is repeated 6 times
about 1609 C. with 5 mL of the standard solution under the above operat-
Carrier gas: Nitrogen ing conditions, the relative standard deviation of the ratios
Flow rate: Adjust the ‰ow rate so that the retention time of the peak area of lenampicillin to that of the internal stan-
of the internal standard is about 1 minute. dard is not more than 1.0z.
System suitability—
Containers and storage Containers—Tight containers.
System performance: When the procedure is run with 4 mL
of the standard solution (2) under the above operating con-
ditions, the internal standard, ethyl acetate and 2-propanol
are eluted in this order, and the resolution between the peaks
of the internal standard and ethyl acetate is not less than 2.0.
System repeatability: When the test is repeated 3 times
with 4 mL of the standard solution (2) under the above oper-
ating conditions, the relative standard deviation of the ratios
1490 O‹cial Monographs for Part I Supplement I, JP XIV

Lincomycin Hydrochloride 6.5z of that obtained from 20 mL of the sample solution.


System performance, and system repeatability: Proceed as
塩酸リンコマイシン directed in the system suitability in the Assay.

Water 3.0 – 6.0z (0.5 g, volumetric titration, direct titra-


Change to read except the structural formula tion).
and chemical name:
Assay Weigh accurately an amount of Lincomycin
Lincomycin Hydrochloride contains not less than Hydrochloride and Lincomycin Hydrochloride Reference
825 mg (potency) per mg, calculated on the anhydrous Standard, equivalent to about 10 mg (potency), dissolve
basis. The potency of Lincomycin Hydrochloride each in the mobile phase to make exactly 10 mL, and use
is expressed as mass (potency) of lincomycin these solutions as the sample solution and the standard solu-
(C18H34N2O6S: 406.54). tion. Perform the test with exactly 20 mL each of the sample
solution and the standard solution as directed under the Liq-
Description Lincomycin Hydrochloride occurs as white,
uid Chromatography according to the following conditions,
crystals or crystalline powder.
and determine the peak areas, AT and AS, of lincomycin.
It is freely soluble in water and in methanol, sparingly
soluble in ethanol (95), and very slightly soluble in acetoni- Amount [ mg (potency)] of lincomycin (C18H34N2O6S)
trile. A
=WS × T × 1000
AS
Identiˆcation (1) Determine the infrared absorption
spectrum of Lincomycin Hydrochloride as directed in the WS: Amount [mg (potency)] of Lincomycin Hydrochlo-
paste method under the Infrared Spectrophotometry, and ride Reference Standard
compare the spectrum with the Reference Spectrum or the
Operating conditions—
spectrum of Lincomycin Hydrochloride Reference Stan-
Detector: An ultraviolet absorption photometer
dard: both spectra exhibit similar intensities of absorption at
(wavelength: 210 nm).
the same wave numbers.
Column: A stainless steel column 4 mm in inside diameter
(2) A solution of Lincomycin Hydrochloride (1 in 100)
and 25 cm in length, packed with octylsilanized silica gel for
responds to the Qualitative Test (2) for chloride.
liquid chromatography (5 mm in particle diameter).
Optical rotation [a]20
D : +135 – +1509 (0.5 g, water, 25 Column temperature: A constant temperature of about
mL, 100 mm). 469C.
Mobile phase: To 13.5 mL phosphoric acid add water to
pH Dissolve 0.10 g of Lincomycin Hydrochloride in 1 mL
make 1000 mL, and adjust the pH to 6.0 with ammonia TS.
of water: 3.0 – 5.5.
To 780 mL of this solution add 150 mL of acetonitrile and
Purity (1) Clarity and color of solution—Dissolve 1.0 g 150 mL of methanol.
of Lincomycin Hydrochloride in 10 mL of water: the solu- Flow rate: Adjust the ‰ow rate so that the retention time
tion is clear and colorless. of lincomycin is about 9 minutes.
(2) Heavy metals—Proceed with 2.0 g of Lincomycin System suitability—
Hydrochloride according to Method 4, and perform the test. System performance: When the procedure is run with 20
Prepare the control solution with 1.0 mL of Standard Lead mL of the standard solution under the above operating con-
Solution (not more than 5 ppm). ditions, the theoretical plates and the symmetrical coe‹cient
(3) Lincomycin B—Perform the test with 20 mL of the of the peak of lincomycin are not less than 4000 and not
sample solution obtained in the Assay as directed under the more than 1.3, respectively.
Liquid Chromatography according to the following condi- System repeatability: When the test is repeated 6 times
tions, and determine the peak areas of lincomycin and lin- with 20 mL of the standard solution under the above operat-
comycin B, having the relative retention time of about 0.5 ing conditions, the relative standard deviation of the peak
with respect to lincomycin, by the automatic integration area of lincomycin is not more than 2.0z.
method: the peak area of lincomycin B is not more than
Containers and storage Containers—Tight containers.
5.0z of the sum of the peak areas of lincomycin and lin-
comycin B.
Operating conditions—
Detector, column, column temperature, mobile phase,
Liothyronine Sodium Tablets
and ‰ow rate: Proceed as directed in the operating condi- リオチロニンナトリウム錠
tions in the Assay.
System suitability— Change the Content uniformity to read:
Test for required detectability: Measure exactly 1 mL of
the sample solution, and add the mobile phase to make ex- Content uniformity Place 1 tablet of Liothyronine Sodium
actly 20 mL. Conˆrm that the peak area of lincomycin ob- Tablets in a glass-stoppered centrifuge tube, add exactly 10
tained from 20 mL of this solution is equivalent to 3.5 to mL of 0.01 mol W L sodium hydroxide TS, warm at 509 C for
Supplement I, JP XIV O‹cial Monographs for Part I 1491

15 minutes, and shake vigorously for 20 minutes. Centrifuge Add the following:
for 5 minutes, and ˆlter the supernatant liquid, if necessary.
Pipet a deˆnite volume of this solution, and add a volume of Lysozyme Hydrochloride
0.01 mol W L sodium hydroxide VS to prepare a deˆnite
volume of a solution containing about 0.5 mg of liothyronine 塩化リゾチーム
sodium (C15H11I3NNaO4) per mL. Pipet 5 mL of this solu-
tion, add exactly 1 mL of the internal standard solution, and
use this solution as the sample solution. Perform the test
with 200 mL of the sample solution as directed under the Liq-
uid Chromatography according to the following conditions,
and calculate the ratio of the peak area of the liothyronine to
that of the internal standard. Calculate the mean value of
the ratios of each peak area of 10 samples: the deviation (z) C616H963N193O182S10.xHCl
of each ratio of the peak area from the mean value should be [12650-88-3, egg white lysozyme]
not more than 15z. When the deviation (z) is more than
15z, and 1 sample shows not more than 25z, perform Lysozyme Hydrochloride is a hydrochloride of a
another test with 20 samples. Calculate the deviation (z) of basic polypeptide obtained from albumen of hen's
each ratio of the peak area from the mean value of the 30 egg, and has an activity to hydrolyze mucopolysaccha-
samples used in the two tests: there should be not more than rides.
1 sample with the deviation more than 15z but not more It contains not less than 0.9 mg (potency) of lyso-
than 25z, and no sample should deviate by more than 25z. zyme per mg, calculated on the dried basis.
Internal standard solution—A solution of propylparahy-
droxybenzoate in a mixture of methanol and diluted phos- Description Lysozyme Hydrochloride occurs as white,
phoric acid (1 in 10) (9:1) (1 in 250,000). crystals, or crystalline or amorphous powder.
Operating conditions— It is freely soluble in water, and practically insoluble in
Detector: An ultraviolet absorption photometer ethanol (99.5).
(wavelength: 225 nm). It is hygroscopic.
Column: A stainless steel column 4.6 mm in inside di- The pH of a solution of Lysozyme Hydrochloride (3 in
ameter and 15 cm in length, packed with octadecylsylanized 200) is between 3.0 and 5.0.
silica gel for liquid chromatography (5 mm in particle di- Identiˆcation (1) To 5 mL of a solution of Lysozyme
ameter). Hydrochloride in acetate buŠer solution, pH 5.4 (1 in 500)
Column temperature: A constant temperature of about add 1 mL of ninhydrin TS, and heat for 10 minutes: a blue-
259C. purple color develops.
Mobile phase: Diluted methanol (57 in 100). (2) Determine the absorption spectrum of a solution of
Flow rate: Adjust the ‰ow rate so that the retention time Lysozyme Hydrochloride in acetate buŠer solution, pH 5.4
of liothyronine is about 9 minutes. (1 in 10,000) as directed under the Ultraviolet-visible Spec-
System suitability— trophotometry, and compare the spectrum with the Refer-
System performance: To 5 mL of a solution of liothyro- ence Spectrum: both spectra exhibit similar intensities of ab-
nine sodium in 0.01 mol/L sodium hydroxide TS (1 in sorption at the same wavelengths.
2,000,000) add 1 mL of the internal standard solution, and
use this solution as the solution for system suitability test. Purity (1) Clarity of solution—To 5 mL of a solution of
When the procedure is run with 200 mL of this solution un- Lysozyme Hydrochloride (3 in 200) add, if necessary, dilute
der the above operating conditions, the internal standard hydrochloric acid to adjust the pH to 3: the solution is clear.
and liothyronine are eluted in this order with the resolution (2) Heavy metals—Proceed with 1.0 g of Lysozyme
between these peaks being not less than 2.0. Hydrochloride according to Method 2, and perform the test.
System repeatability: When the test is repeated 6 times Prepare the control solution with 2.0 mL of Standard Lead
with 200 mL of the solution for system suitability test under Solution (not more than 20 ppm).
the above operating conditions, the relative standard devia- Loss on drying Not more than 8.0z (0.1 g, 1059
C, 2
tion of the ratios of the peak area of liothyronine to that of hours).
the internal standard is not more than 1.0z.
Residue on ignition Not more than 2.0z (0.5 g).

Nitrogen Perform the test as directed under the Nitrogen


Determination: the amount of nitrogen (N: 14.01) is between
16.8z and 18.6z, calculated on the dried basis.

Assay Weigh accurately an amount of Lysozyme


Hydrochloride, equivalent to about 25 mg (potency), dis-
solve in phosphate buŠer solution, pH 6.2 to make exactly
1492 O‹cial Monographs for Part I Supplement I, JP XIV

100 mL. Pipet 2 mL of this solution, add phosphate buŠer D-Mannitol Injection
solution, pH 6.2 to make exactly 50 mL, and use this solu-
tion as the sample solution. Separately, weigh accurately an マンニトール注射液
amount of Lysozyme Reference Standard (separately deter-
mine its loss on drying in the same manner as Lysozyme Delete the Residue on ignition.
Hydrochloride), equivalent to about 25 mg (potency), and
dissolve in phosphate buŠer solution, pH 6.2 to make ex- Delete the Pyrogen and add the following:
actly 100 mL. Pipet 1 mL and 2 mL of this solution, add
Bacterial endotoxins Less than 0.50 EU/mL.
phosphate buŠer solution, pH 6.2 to them to make exactly
50 mL, and use these solutions as the standard solution (1)
Change the Containers and storage to read:
and the solution (2), respectively. Keep the sample solution
and the standard solutions in an ice-bath. Pipet 4 mL of sub- Containers and storage Containers—Hermetic containers.
strate solution for lysozyme hydrochloride, previously Plastic containers for aqueous injections may be used.
warmed in a water bath of 359 C for about 5 minutes, add ex-
actly 100 mL of the sample solution, previously warmed in a
water bath of 359 C for about 3 minutes, and allow to stand Menatetrenone
at 359C for exactly 10 minutes, then add exactly 0.5 mL of 1
mol/L hydrochloric acid TS, and immediately shake. Deter- メナテトレノン
mine the absorbance, AT, of this solution at 640 nm, using
water as the blank. Determine the absorbances, AS1 and AS2, Change the Description to read:
of the solutions obtained with the standard solution (1) and Description Menatetrenone occurs as yellow, crystals,
the standard solution (2) in the same manner as the sample crystalline powder, waxy mass or oily material.
solution. It is very soluble in hexane, soluble in ethanol (99.5), spar-
Amount [mg (potency)] of lysozyme per mg, ingly soluble in 2-propanol, slightly soluble in methanol, and
calculated on the dried basis practically insoluble in water.

Å œ
WS A - AT It decomposes and the color becomes more intense by
= × S1 +1 light.
2 WT A S1 - A S2
Melting point: about 379 C
WS: Amount (mg) of Lysozyme Reference Standard, cal-
culated on the dried basis.
WT: Amount (mg) of the sample, calculated on the dried
basis.
Mepitiostane
Containers and storage Containers—Tight containers. メピチオスタン

Change the Assay to read:


Magnesium Sulfate Injection Assay Weigh accurately about 0.3 g of Mepitiostane, and
dissolve in cyclohexane to make exactly 10 mL. Pipet 2 mL
硫酸マグネシウム注射液 of this solution, add 10 mL of ethanol (99.5), mix with ex-
actly 2 mL each of 0.01 mol W L hydrochloric acid TS and the
Change the Description to read: internal standard solution, add ethanol (99.5) to make 20
Description Magnesium Sulfate Injection is a clear, color- mL, allow to stand at ordinary temperature for 30 minutes,
less liquid. and use this solution as the sample solution. Separately,
weigh accurately about 45 mg of Epitiostanol Reference
Add the following next to Identiˆcation: Standard, dissolve in exactly 2 mL of the internal standard
solution, add ethanol (99.5) to make 20 mL, and use this so-
pH 5.5 – 7.0 When the labeled concentration exceeds lution as the standard solution. Perform the test with 10 mL
5z, prepare a solution of 5z with water, and perform the each of the sample solution and the standard solution as
test. directed under the Liquid Chromatography according to the
following conditions, and calculate the ratios, Q T and QS, of
Change the Bacterial endotoxins to read: the peak area of epitiostanol to that of the internal standard,
Bacterial endotoxins Less than 0.09 EU/mg. respectively.

QT
Change the Containers and storage to read: Amount (mg) of C25H40O2S = WS × × 5 × 1.3202
QS
Containers and storage Containers—Hermetic containers. WS: Amount (mg) of Epitiostanol Reference Standard,
Plastic containers for aqueous injections may be used. calculated on the anhydrous basis

Internal standard solution—A solution of n-octylbenzene in


Supplement I, JP XIV O‹cial Monographs for Part I 1493

ethanol (99.5) (1 in 300). of methotrexate is about 8 minutes.


Operating conditions— System suitability—
Detector: An ultraviolet absorption photometer System performance: Dissolve 10 mg each of Meth-
(wavelength: 265 nm). otrexate and folic acid in 100 mL of the mobile phase. When
Column: A stainless steel column 4.0 mm in inside di- the procedure is run with 10 mL of this solution under the
ameter and 15 cm in length, packed with octadecylsilanized above operating conditions, folic acid and methotrexate are
silica gel for liquid chromatography (10 mm in particle di- eluted in this order with the resolution between these peaks
ameter). being not less than 8.
Column temperature: A constant temperature of about System repeatability: When the test is repeated 6 times
259C. with 10 mL of the standard solution under the above operat-
Mobile phase: A mixture of methanol and water (20:3). ing conditions, the relative standard deviation of the peak
Flow rate: Adjust the ‰ow rate so that the retention time area of methotrexate is not more than 1.0z.
of epitiostanol is about 6 minutes.
System suitability—
System performance: When the procedure is run with Meticrane
10 mL of the standard solution under the above operating
conditions, epitiostanol and the internal standard are eluted メチクラン
in this order with the resolution between these peaks being
not less than 4. Change the Purity (4) to read:
System repeatability: When the test is repeated 6 times
Purity
with 10 mL of the standard solution under the above operat-
(4) Related substances—Dissolve 0.05 g of Meticrane in
ing conditions, the relative standard deviation of the ratios
50 mL of acetonitrile, and use this solution as the sample so-
of the peak area of epitiostanol to that of the internal stan-
lution. Pipet 1 mL of the sample solution, add acetonitrile to
dard is not more than 1.0z.
make exactly 100 mL, and use this solution as the standard
solution. Perform the test with exactly 2 mL each of the sam-
ple solution and the standard solution as directed under the
Methotrexate Liquid Chromatography according to the following condi-
tions, and determine each peak area of both solutions by the
メトトレキサート
automatic integration method: the total area of the peaks
other than meticrane from the sample solution is not larger
Change the Assay to read:
than the peak area of meticrane from the standard solution.
Assay Weigh accurately about 25 mg each of Methotrexate Operating conditions 1—
and Methotrexate Reference Standard, dissolve in the mo- Detector: An ultraviolet absorption photometer
bile phase to make exactly 250 mL, and use these solutions (wavelength: 230 nm).
as the sample solution and the standard solution. Perform Column: A stainless steel column 4.6 mm in inside di-
the test with 10 mL each of these solutions as directed under ameter and 15 cm in length, packed with octadecylsilanized
the Liquid Chromatography according to the following con- silica gel for liquid chromatography (5 mm in particle di-
ditions, and measure the peak areas, AT and AS, of meth- ameter).
otrexate in each solution. Column temperature: A constant temperature of about
409C.
AT
Amount (mg) of C20H22N8O5 = WS × Mobile phase: A mixture of water and acetonitrile (17:3).
AS
Flow rate: Adjust the ‰ow rate so that the retention time
WS: Amount (mg) of Methotrexate Reference Standard, of meticrane is about 7 minutes.
calculated on the anhydrous basis Time span of measurement: About 4 times as long as the
retention time of meticrane, after the solvent peak.
Operating conditions—
System suitability 1—
Detector: An ultraviolet absorption photometer
Test for required detection: To exactly 2 mL of the stan-
(wavelength: 302 nm).
dard solution add the mobile phase to make exactly 20 mL.
Column: A stainless steel column 4.6 mm in inside di-
Conˆrm that the peak area of meticrane obtained from
ameter and 25 cm in length, packed with octadecylsilanized
10 mL of this solution is equivalent to 7 to 13z of that of
silica gel for liquid chromatography (10 mm in particle di-
meticrane obtained from 10 mL of the standard solution.
ameter).
System performance: Dissolve 0.01 g each of Meticrane
Column temperature: A constant temperature of about
and caŠeine in 100 mL of acetonitrile. To exactly 2 mL of
259C.
this solution add the mobile phase to make exactly 10 mL.
Mobile phase: A mixture of disodium hydrogen phos-
When the procedure is run with 10 mL of this solution under
phate-citric acid buŠer solution, pH 6.0 and acetonitrile
the above operating conditions 1, caŠeine and meticrane are
(89:11).
eluted in this order with the resolution between these peaks
Flow rate: Adjust the ‰ow rate so that the retention time
1494 O‹cial Monographs for Part I Supplement I, JP XIV

being not less than 10. on a plate of silica gel for thin-layer chromatography. De-
System repeatability: When the test is repeated 6 times velop the plate with a mixture of ethanol (99.5), 1-buthanol
with 10 mL of the standard solution under the above operat- and ammonia solution (28) (10:8:7) to a distance of about 10
ing conditions 1, the relative standard deviation of the peak cm, and air-dry the plate. Spray evenly a solution of nin-
area of meticrane is not more than 2.0z. hydrin in a mixture of acetone and pyridine (25:1) (1 in 500),
Operating conditions 2— and heat at 1009 C for 10 minutes: the spots obtained from
Detector, column, and column temperature: Proceed as the sample solution and the standard solution are red-purple
directed in the operating conditions 1. to red-brown and their Rf values are the same.
Mobile phase: A mixture of water and acetonitrile (1:1). (2) To 5 mL of a solution of Micronomicin Sulfate (1 in
Flow rate: Adjust the ‰ow rate so that the retention time 100) add 1 mL of barium chloride TS: a white precipitate is
of meticrane is about 2 minutes. formed, and it does not dissolve by addition of dilute nitric
Time span of measurement: About 10 times as long as the acid.
retention time of meticrane, after the solvent peak.
Optical rotation [a]20
D : +110 – +1309 (0.25 g calculated
System suitability 2—
on the anhydrous basis, water, 25 mL, 100 mm).
Test for required detection: To exactly 2 mL of the stan-
dard solution add the mobile phase to make exactly 20 mL. pH The pH of a solution obtained by dissolving 1.0 g of
Conˆrm that the peak area of meticrane obtained from Micronomicin Sulfate in 10 mL of water is between 3.5 and
10 mL of this solution is equivalent to 7 to 13z of that of 5.5.
meticrane obtained from 10 mL of the standard solution.
Purity (1) Clarity and color of solution—Dissolve 1.5 g
System performance: Dissolve 0.02 g each of Meticrane
of Micronomicin Sulfate in 10 mL of water: the solution is
and methyl parahydroxybenzoate in 100 mL of acetonitrile.
clear and colorless to pale yellow.
To exactly 2 mL of this solution add the mobile phase to
(2) Heavy metals—Proceed with 1.0 g of Micronomicin
make exactly 10 mL. When the procedure is run with 10 mL
Sulfate according to Method 2, and perform the test. Pre-
of this solution under the above operating conditions 2,
pare the control solution with 2.0 mL of Standard Lead So-
meticrane and methyl parahydroxybenzoate are eluted in
lution (not more than 20 ppm).
this order with the resolution between these peaks being not
(3) Related substances—Dissolve 0.40 g of Micronomi-
less than 4.
cin Sulfate in 10 mL of water, and use this solution as the
System repeatability: When the test is repeated 6 times
sample solution. Pipet 1 mL of the sample solution, add
with 10 mL of the standard solution under the above operat-
water to make exactly 200 mL, and use this solution as the
ing conditions 2, the relative standard deviation of the peak
standard solution. Perform the test with these solutions as
area of meticrane is not more than 2.0z.
directed under the Thin-layer Chromatography. Spot 5 mL
of the sample solution and the standard solution on a plate
of silica gel for thin-layer chromatography. Develop the
Micronomicin Sulfate plate with a mixture of ethanol (99.5), 1-buthanol and am-
monia solution (28) (10:8:7) to a distance of about 10 cm,
硫酸ミクロノマイシン
and air-dry the plate. Spray evenly a solution of ninhydrin in
a mixture of acetone and pyridine (25:1) (1 in 500), and heat
Change to read except the structural formula
at 1009 C for 10 minutes: the spot other than the principal
and chemical name:
spot obtained from the sample solution is not more intense
than the spot from the standard solution.
Micronomicin Sulfate contains not less than 590 mg
(potency) per mg, calculated on the anhydrous basis. Water Not more than 10.0z (0.2 g, volumetric titration,
The potency of Micronomicin Sulfate is expressed as back titration). Use a mixture of methanol for water deter-
mass (potency) of micronomicin (C20H41N5O7: mination and ethylene glycol for water determination (1:1)
463.57). instead of methanol for water determination.
Description Micronomicin Sulfate occurs as a white to Assay Perform the test according to the Cylinder-plate
light yellowish white powder. method as directed under the Microbial Assay for Antibiot-
It is very soluble in water, sparingly soluble in ethylene ics according to the following conditions.
glycol, and practically insoluble in methanol and in ethanol (1) Test organism—Bacillus subtilis ATCC 6633
(99.5). (2) Culture medium—Use the medium i in 1) Medium
It is hygroscopic. for test organism [5] under (1) Agar media for seed and base
layer.
Identiˆcation (1) Dissolve 50 mg each of Micronomicin
(3) Standard solutions—Weigh accurately an amount of
Sulfate and Micronomicin Sulfate Reference Standard in 10
Micronomicin Sulfate Reference Standard, equivalent to
mL of water, and use these solutions as the sample solution
about 20 mg (potency), dissolve in 0.1 mol/L phosphate
and the standard solution. Perform the test with these solu-
buŠer solution for antibiotics, pH 8.0 to make exactly 20
tions as directed under the Thin-layer Chromatography.
mL, and use this solution as the standard stock solution.
Spot 5 mL of the sample solution and the standard solution
Supplement I, JP XIV O‹cial Monographs for Part I 1495

Keep the standard stock solution at 5 – 159C, and use within Flow rate: Adjust the ‰ow rate so that the retention time
30 days. Take exactly a suitable amount of the standard of ethenzamide is about 4 minutes.
stock solution before use, add 0.1 mol/L phosphate buŠer System suitability—
solution for antibiotics, pH 8.0 to make solutions so that System performance: Dissolve 0.9 g of antipyrine and
each mL contains 2 mg (potency) and 0.5 mg (potency), and 0.09 g of caŠeine in 10 mL of chloroform. When the proce-
use these solutions as the high concentration standard solu- dure is run with 1 mL of this solution under the above operat-
tion and the low concentration standard solution, respec- ing conditions, caŠeine and antipyrine are eluted in this ord-
tively. er with the resolution between these peaks being not less
(4) Sample solutions—Weigh accurately an amount of than 1.5.
Micronomicin Sulfate, equivalent to about 20 mg (potency), System repeatability: When the test is repeated 6 times
and dissolve in 0.1 mol/L phosphate buŠer solution for an- with 1 mL of the standard solution under the above operat-
tibiotics, pH 8.0 to make exactly 20 mL. Take exactly a suit- ing conditions, the relative standard deviation of the ratios
able amount of this solution, add 0.1 mol/L phosphate of the peak area of caŠeine to that of the internal standard is
buŠer solution for antibiotics, pH 8.0 to make solutions so not more than 1.0z.
that each mL contains 2 mg (potency) and 0.5 mg (potency),
and use these solutions as the high concentration sample so-
lution and the low concentration sample solution, respec- Minocycline Hydrochloride
tively.
塩酸ミノサイクリン
Containers and storage Containers—Tight containers.
Change the Identiˆcation to read:

Migrenin Identiˆcation (1) Determine the absorption spectrum of


a solution of Minocycline Hydrochloride in a solution of
ミグレニン hydrochloric acid in methanol (19 in 20,000) (1 in 62,500) as
directed under the Ultraviolet-visible Spectrophotometry,
Change the Assay (2) to read: and compare the spectrum with the Reference Spectrum or
the spectrum of a solution of Minocycline Hydrochloride
Assay
Reference Standard prepared in the same manner as the
(2) CaŠeine—To about 1 g of Migrenin, previously
sample solution: both spectra exhibit similar intensities of
dried and accurately weighed, add exactly 5 mL of the inter-
absorption at the same wavelengths.
nal standard solution, dissolve in chloroform to make 10
(2) Determine the infrared absorption spectrum of
mL, and use this solution as the sample solution. Separately,
Minocycline Hydrochloride as directed in the potassium
weigh accurately about 90 mg of CaŠeine Reference Stan-
chloride disk method under the Infrared Spectrophotomet-
dard, previously dried at 809C for 4 hours, add exactly 5 mL
ry, and compare the spectrum with the Reference Spectrum
of the internal standard solution, dissolve in chloroform to
or the spectrum of Minocycline Hydrochloride Reference
make 10 mL, and use this solution as the standard solution.
Standard: both spectra exhibit similar intensities of absorp-
Perform the test with 1 mL each of the sample solution and
tion at the same wave numbers.
the standard solution as directed under the Gas Chromatog-
(3) A solution of Minocycline Hydrochloride (1 in 100)
raphy according to the following conditions, and calculate
responds to the Qualitative Test (2) for chloride.
the ratios, Q T and Q S, of the peak area of caŠeine to that of
the internal standard.
Delete the Absorbance.
Q
Amount (mg) of caŠeine (C8H10N4O2) = WS × T
QS Change the Purity to read:
WS: Amount (mg) of CaŠeine Reference Standard Purity (1) A solution of Minocycline Hydrochloride (1 in
100) is clear, and when the test is performed within 1 hour
Internal standard solution—A solution of ethenzamide in
after preparation of this solution, the absorbance of the so-
chloroform (1 in 50).
lution at 560 nm, determined as directed under the Ultrav-
Operating conditions—
iolet-visible Spectrophotometry, is not more than 0.06.
Detector: A hydrogen ‰ame-ionization detector.
(2) Heavy metals—Proceed with 0.5 g of Minocycline
Column: A glass column 2.6 mm in inside diameter and
Hydrochloride according to Method 2, and perform the test.
210 cm in length, packed with siliceous earth for gas chro-
Prepare the control solution with 2.5 mL of Standard Lead
matography (180 to 250 mm in particle diameter) coated with
Solution (not more than 50 ppm).
50z phenyl-methyl silicon polymer for gas chromatography
(3) Related substances—Dissolve 50 mg of Minocycline
at the ratio of 15z.
Hydrochloride in 100 mL of the mobile phase, and use this
Column temperature: A constant temperature of about
solution as the sample solution. Perform the test immedi-
2109 C.
ately after the preparation of the sample solution with 20 mL
Carrier gas: Nitrogen
of the sample solution as directed under the Liquid Chro-
1496 O‹cial Monographs for Part I Supplement I, JP XIV

matography according to the following conditions, and (2) Determine the infrared absorption spectrum of
measure each peak area by the automatic integration Mitomycin C as directed in the potassium bromide disk
method. Calculate the amount of each peak area by the area method under the Infrared Spectrophotometry, and com-
percentage method: the amount of epiminocycline is not pare the spectrum with the Reference Spectrum or the spec-
more than 1.2z, the amount of each peak other than trum of Mitomycin C Reference Standard: both spectra
minocycline and epiminocycline is not more than 1.0z, and exhibit similar intensities of absorption at the same wave
the total area of the peaks other than minocycline and numbers.
epiminocycline is not more than 2.0z.
Purity Related substances—Conduct this procedure rap-
Operating conditions—
idly after the sample and the standard solutions are pre-
Detector, column, column temperature, and mobile
pared. Dissolve 50 mg of Mitomycin C in 10 mL of
phase: Proceed as directed in the operating conditions in the
methanol, and use this solution as the sample solution. Pipet
Assay.
1 mL of the sample solution, add methanol to make exactly
Flow rate: Adjust the ‰ow rate so that the retention time
100 mL, and use this solution as the standard solution. Per-
of minocycline is about 12 minutes. The retention time of
form the test with exactly 10 mL each of the sample solution
epiminocycline is about 10 minutes under this condition.
and the standard solution as directed under the Liquid Chro-
Time span of measurement: About 2.5 times as long as the
matography according to the following conditions, and de-
retention time of minocycline after the solvent peak.
termine each peak area by the automatic integration
System suitability—
method: each area of the peak other than mitomycin C ob-
Test for required detection: To exactly 2 mL of the sample
tained from the sample solution is not more than the peak
solution add the mobile phase to make exactly 100 mL, and
area of mitomycin C from the standard solution, and the
use this solution as the solution for system suitability test.
total area of the peaks other than mitomycin C from the
Pipet 5 mL of the solution for system suitability test, and
sample solution is not more than 3 times the peak area of
add the mobile phase to make exactly 100 mL. Conˆrm that
mitomycin C from the standard solution.
the peak area of minocycline obtained from 20 mL of this so-
Operating conditions—
lution is equivalent to 3.5 to 6.5z of that of minocycline ob-
Detector: An ultraviolet absorption photometer
tained from 20 mL of the solution for system suitability test.
(wavelength: 254 nm).
System performance: Proceed as directed in the system
Column: A stainless steel column 6 mm in inside diameter
suitability in the Assay.
and 15 cm in length, packed with octadecylsilanized silica gel
System repeatability: When the test is repeated 6 times
for liquid chromatography (5 mm in particle diameter).
with 20 mL of the solution for system suitability test under
Column temperature: A constant temperature of about
the above operating conditions, the relative standard devia-
309C.
tion of the peak area of minocycline is not more than 2.0z.
Mobile phase A: To 20 mL of 0.5 mol/L ammonium
acetate TS add water to make 1000 mL. To 800 mL of this
solution add 200 mL of methanol.
Mitomycin C Mobile phase B: To 20 mL of 0.5 mol/L ammonium
acetate TS add water to make 1000 mL. To this solution add
マイトマイシンC
1000 mL of methanol.
Flowing of the mobile phase: Control the gradient by mix-
Change to read except the structural formula
ing the mobile phases A and B as directed in the following t-
and chemical name:
able.
Mitomycin C contains not less than 970 mg (potency) Time after injection Mobile phase Mobile phase
per mg, calculated on the dried basis. The potency of of sample (min) A (z) B (z)
Mitomycin C is expressed as mass (potency) of
0 – 10 100 0
mitomycin C (C15H18N4O5).
10 – 30 100 → 0 0 → 100
Description Mitomycin C occurs as blue-purple, crystals or 30 – 45 0 100
crystalline powder.
Flow rate: About 1.0 mL/min
It is freely soluble in N,N-dimethylacetamide, slightly
Time span of measurement: About 2 times as long as the
soluble in water and in methanol, and very slightly soluble in
retention time of mitomycin C after the solvent peak.
ethanol (99.5).
System suitability—
Identiˆcation (1) Determine the absorption spectrum of Test for required detection: Pipet 10 mL of the standard
a solution of Mitomycin C (1 in 100,000) as directed under solution, and add methanol to make exactly 100 mL. Con-
the Ultraviolet-visible Spectrophotometry, and compare the ˆrm that the peak area of mitomycin C obtained from 10 mL
spectrum with the Reference Spectrum or the spectrum of a of this solution is equivalent to 7 to 13z of that from 10 mL
solution of Mitomycin C Reference Standard prepared in the of the standard solution.
same manner as the sample solution: both spectra exhibit System performance: Dissolve 25 mg of Mitomycin C and
similar intensities of absorption at the same wavelengths. 40 mg of 3-ethoxy-4-hydroxybenzaldehyde in 50 mL of
Supplement I, JP XIV O‹cial Monographs for Part I 1497

methanol. When the procedure is run with 10 mL of this so- Morphine Hydrochloride
lution under the above operating conditions, mitomycin C
and 3-ethoxy-4-hydroxybenzaldehyde are eluted in this order 塩酸モルヒネ
with the resolution between these peaks being not less than
15. Change the Description to read:
System repeatability: When the test is repeated 3 times
Description Morphine Hydrochloride occurs as white,
with 10 mL of the standard solution under the above operat-
crystals or crystalline powder.
ing conditions, the relative standard deviation of the peak
It is freely soluble in formic acid, soluble in water, spar-
area of mitomycin C is not more than 3.0z.
ingly soluble in methanol, and slightly soluble in ethanol
Loss on drying Not more than 1.0z (0.1 g, reduced pres- (95).
sure not exceeding 0.67 kPa, 609
C, 3 hours). It is colored by light.
Assay Weigh accurately an amount of Mitomycin C and
Mitomycin C Reference Standard, equivalent to about
Add the following next to Optical rotation:
25 mg (potency), dissolve each in N,N-dimethylacetamide to pH The pH of a solution obtained by dissolving 0.10 g of
make exactly 50 mL, and use these solutions as the sample Morphine Hydrochloride in 10 mL of water is between 4.0
solution and the standard solution. Perform the test with ex- and 6.0.
actly 10 mL each of the sample solution and the standard so-
lution as directed under the Liquid Chromatography accord- Change the Purity to read:
ing to the following conditions, and determine the peak
Purity (1) Clarity and color of solution—Dissolve 0.10 g
areas, AT and AS, of mitomycin C.
of Morphine Hydrochloride in 10 mL of water: the solution
Amount [ mg (potency)] of C15H18N4O5 is clear and colorless.
A (2) Sulfate—Dissolve 0.20 g of Morphine Hydrochloride
= WS × T × 1000
AS in 5 mL of water, and add 2 to 3 drops of barium chloride
TS: no turbidity is produced.
WS: Amount [mg (potency)] of Mitomycin C Reference
(3) Meconic acid—Dissolve 0.20 g of Morphine
Standard
Hydrochloride in 5 mL of water, and add 5 mL of dilute
Operating conditions— hydrochloric acid and 2 drops of iron (III) chloride TS: no
Detector: An ultraviolet absorption photometer red color develops.
(wavelength: 365 nm). (4) Other alkaloids—Dissolve 0.1 g of Morphine
Column: A stainless steel column 4 mm in inside diameter Hydrochloride in 10 mL of diluted ethanol (95) (1 in 2), and
and 30 cm in length, packed with phenylated silica gel for use this solution as the sample solution. Pipet 1 mL of the
liquid chromatography (10 mm in particle diameter). sample solution, add diluted ethanol (95) (1 in 2) to make ex-
Column temperature: A constant temperature of about actly 200 mL, and use this solution as the standard solution.
259C. Perform the test with these solutions as directed under the
Mobile phase: To 40 mL of 0.5 mol/L ammonium acetate Thin-layer Chromatography. Spot 10 mL each of the sample
TS add 5 mL of diluted acetic acid (100) (1 in 20) and water solution and the standard solution on a plate of silica gel
to make 1000 mL. To 600 mL of this solution add 200 mL of with ‰uorescent indicator for thin-layer chromatography.
methanol. Develop the plate with a mixture of ethanol (99.5), toluene,
Flow rate: Adjust the ‰ow rate so that the retention time acetone and ammonia solution (28) (14:14:7:1) to a distance
of mitomycin C is about 7 minutes. of about 15 cm, and air-dry the plate. Examine under ultrav-
System suitability— iolet light (main wavelength: 254 nm): the spots other than
System performance: Dissolve about 25 mg of Mitomycin the principal spot from the sample solution are not more in-
C Reference Standard and about 0.375 g of 3-ethoxy-4- tense than the spot from the standard solution.
hydroxybenzaldehyde in 50 mL of N,N-dimethylacetamide.
When the procedure is run with 10 mL of this solution under
the above operating conditions, mitomycin C and 3-ethoxy- Morphine Hydrochloride Injection
4-hydroxybenzaldehyde are eluted in this order with the
resolution between these peaks being not less than 3. 塩酸モルヒネ注射液
System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat- Change the Assay to read:
ing conditions, the relative standard deviation of the peak
Assay Take exactly a volume of Morphine Hydrochloride
area of mitomycin C is not more than 1.0z.
Injection, equivalent to about 80 mg of morphine
Containers and storage Containers—Tight containers. hydrochloride (C17H19NO3.HCl.3H2O), and add water to
make exactly 20 mL. Pipet 5 mL of this solution, add exactly
10 mL of the internal standard solution and water to make
50 mL, and use this solution as the sample solution.
1498 O‹cial Monographs for Part I Supplement I, JP XIV

Separately, weigh accurately about 25 mg of morphine 50 mL. Filter this solution, and use the ˆltrate as the sample
hydrochloride for assay, dissolve in exactly 10 mL of the in- solution. Separately, weigh accurately about 25 mg of mor-
ternal standard solution, add water to make 50 mL, and use phine hydrochloride for assay, dissolve in exactly 10 mL of
this solution as the standard solution. Perform the test with the internal standard solution, add water to make 50 mL,
20 mL each of the sample solution and the standard solution and use this solution as the standard solution. Perform the
as directed under the Liquid Chromatography according to test with 20 mL each of the sample solution and the standard
the following conditions, and calculate the ratios, Q T and solution as directed under the Liquid Chromatography ac-
QS, of the peak area of morphine to that of the internal stan- cording to the following conditions, and calculate the ratios,
dard. Q T and QS, of the peak area of morphine to that of the inter-
nal standard.
Amount (mg) of morphine hydrochloride
(C17H19NO3.HCl.3H2O) Amount (mg) of morphine hydrochloride
Q (C17H19NO3.HCl.3H2O)
= WS × T × 4 × 1.1679
QS Q
= WS × T × 1.1679
QS
WS: Amount (mg) of morphine hydrochloride for assay,
calculated on the anhydrous basis WS: Amount (mg) of morphine hydrochloride for assay,
calculated on the anhydrous basis
Internal standard solution—A solution of etilefrine
hydrochloride (1 in 500). Internal standard solution—A solution of etilefrine
Operating conditions— hydrochloride (1 in 500).
Detector: An ultraviolet absorption photometer Operating conditions—
(wavelength: 285 nm). Detector: An ultraviolet absorption photometer
Column: A stainless steel column 4.6 mm in inside di- (wavelength: 285 nm).
ameter and 15 cm in length, packed with octadecylsilanized Column: A stainless steel column 4.6 mm in inside di-
silica gel for liquid chromatography (5 mm in particle di- ameter and 15 cm in length, packed with octadecylsilanized
ameter). silica gel for liquid chromatography (5 mm in particle di-
Column temperature: A constant temperature of about ameter).
409C. Column temperature: A constant temperature of about
Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in 409C.
500 mL of diluted phosphoric acid (1 in 1000), and adjust Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in
the pH to 3.0 with sodium hydroxide TS. To 240 mL of this 500 mL of diluted phosphoric acid (1 in 1000), and adjust
solution add 70 mL of tetrahydrofuran, and mix. the pH to 3.0 with sodium hydroxide TS. To 240 mL of this
Flow rate: Adjust the ‰ow rate so that retention time of solution add 70 mL of tetrahydrofuran, and mix.
morphine is about 10 minutes. Flow rate: Adjust the ‰ow rate so that the retention time
System suitability— of morphine is about 10 minutes.
System performance: When the procedure is run with System suitability—
20 mL of the standard solution under the above operating System performance: When the procedure is run with
conditions, morphine and the internal standard are eluted in 20 mL of the standard solution under the above operating
this order with the resolution between these peaks being not conditions, morphine and the internal standard are eluted in
less than 3. this order with the resolution between these peaks being not
System repeatability: When the test is repeated 6 times less than 3.
with 20 mL of the standard solution under the above operat- System repeatability: When the test is repeated 6 times
ing conditions, the relative standard deviation of the ratios with 20 mL of the standard solution under the above operat-
of the peak area of morphine to that of the internal standard ing conditions, the relative standard deviation of the ratios
is not more than 1.0z. of the peak area of morphine to that of the internal standard
is not more than 1.0z.

Morphine Hydrochloride Tablets


Norepinephrine
塩酸モルヒネ錠
ノルエピネフリン
Change the Assay to read:
Change the Description to read:
Assay Take not less than 20 Morphine Hydrochloride
Tablets, weigh accurately, and powder. Weigh accurately a Description Norepinephrine occurs as a white to light
quantity of the powder, equivalent to about 20 mg of mor- brown or slightly reddish brown, crystalline powder.
phine hydrochloride (C17H19NO3.HCl.3H2O), add exactly It is freely soluble in acetic acid (100), very slightly soluble
10 mL of the internal standard solution, extract the mixture in water, and practically insoluble in ethanol (95).
with ultrasonic waves for 10 minutes, and add water to make It dissolves in dilute hydrochloric acid.
Supplement I, JP XIV O‹cial Monographs for Part I 1499

It gradually changes to brown by air and by light. tradiol to the peak area of the internal standard of the sam-
ple solution and also the ratios, QSa and QSb, of the peak
Change the Identiˆcation to read: areas of norgestrel and ethinylestradiol to the peak area of
the internal standard of the standard solution.
Identiˆcation (1) Determine the absorption spectrum of
a solution of Norepinephrine in 0.1 mol/L hydrochloric acid Amount (mg) of norgestrel (C21H28O2)
TS (3 in 100,000) as directed under the Ultraviolet-visible Q 1
= WSa × Ta ×
Spectrophotometry, and compare the spectrum with the QSa 100
Reference Spectrum: both spectra exhibit similar intensities
Amount (mg) of ethinylestradiol (C20H24O2)
of absorption at the same wavelengths.
Q 1
(2) Determine the infrared absorption spectrum of = WSb× Tb ×
QSb 100
Norepinephrine, previously dried, as directed in the potassi-
um bromide disk method under the Infrared Spectrophoto- WSa: Amount (mg) of Norgestrel Reference Standard
metry, and compare the spectrum with the Reference Spec- WSb: Amount (mg) of Ethinylestradiol Reference
trum: both spectra exhibit similar intensities of absorption at Standard
the same wave numbers.
Internal standard solution—A solution of diphenyl in dilut-
ed methanol (7 in 10) (1 in 50,000).
Operating conditions—
Norepinephrine Injection Proceed as directed in the operating conditions in the
Assay.
ノルエピネフリン注射液
System suitability—
Proceed as directed in the system suitability in the Assay.
Change the Identiˆcation to read:
Identiˆcation Transfer a volume of Norepinephrine Injec- Change the Dissolution test to read:
tion, equivalent to 1 mg of Norepinephrine according to the
Dissolution test Perform the test with 1 tablet of Norges-
labeled amount, to each of two test tubes A and B, and add
trel and Ethinylestradiol Tablets at 50 revolutions per
1 mL of water to each tube. Add 10 mL of potassium hydro-
minute according to Method 2 under the Dissolution Test,
gen phthalate buŠer solution, pH 3.5, to A, and 10 mL of
using 900 mL of water as the test solution. Take 50 mL or
phosphate buŠer solution, pH 6.5, to B. To each of these so-
more of the dissolved solution 45 minutes after starting the
lutions add 1.0 mL of iodine TS, allow to stand for 5
test, and membrane ˆlter through a membrane ˆlter with
minutes, and add 2.0 mL of sodium thiosulfate TS: no color
pore size of not more than 0.8 mm. Discard the ˆrst 10 mL of
or a pale red color develops in test tube A, and a deep red-
the ˆltrate, transfer exactly 30 mL of the subsequent into a
purple color develops in test tube B.
chromatography column [prepared by packing 0.36 g of oc-
tadecylsilanized silica gel for pretreatment (55 to 105 mm in
particle diameter) in a tube about 1 cm in inside diameter].
Norgestrel and Ethinylestradiol After washing the column with 15 mL of water, elute with
Tablets 3 mL of methanol, and evaporate the eŒuent on a water
bath to dryness at about 409C with the aid of a current air.
ノルゲストレルエチニルエストラジオール錠 Dissolve the residue in exactly 2 mL of diluted methanol (7
in 10), and use this solution as the sample solution. Separate-
Change the Content uniformity to read: ly, weigh accurately about 25 mg of Norgestrel Reference
Content uniformity Add 2 mL of diluted methanol (7 in Standard and about 2.5 mg of Ethinylestradiol Reference
10) to 1 tablet of Norgestrel and Ethinylestradiol Tablets, Standard dissolve in diluted methanol (7 in 10) to make ex-
add exactly 2 mL of the internal standard solution, shake for actly 100 mL, then pipet 3 mL of this solution, add diluted
20 minutes, and centrifuge. Filter the supernatant liquid methanol (7 in 10) to make exactly 100 mL, and use this so-
through a membrane ˆlter with pore size of not more than lution as the standard solution. Perform the test with exactly
0.2 mm, and use this ˆltrate as the sample solution. Separate- 50 mL each of the sample solution and the standard solution
ly, weigh accurately quantities of Norgestrel Reference Stan- as directed under the Liquid Chromatography according to
dard and of Ethinylestradiol Reference Standard, equivalent the following conditions. Determine the peak areas, ATa and
to 100 times each of the labeled amounts, dissolve in diluted ATb, of norgestrel and ethinylestradiol from the sample solu-
methanol (7 in 10) to make exactly 200 mL. Pipet 2 mL of tion, and the peak areas, ASa and ASb, of norgestrel and
this solution, add exactly 2 mL of the internal standard solu- ethinylestradiol from the standard solution.
tion, and use this solution as the standard solution. Perform The dissolution rate of Norgestrel and Ethinylestradiol
the test with 20 mL each of the sample solution and the stan- Tablets in 45 minutes is not less than 70z.
dard solution as directed under the Liquid Chromatography
according to the following conditions. Calculate the ratios,
Q Ta and Q Tb, of the peak areas of norgestrel and ethinyles-
1500 O‹cial Monographs for Part I Supplement I, JP XIV

Dissolution rate (z) with respect to the labeled amount Operating conditions—
of norgestrel (C21H28O2) Detector: Norgestrel—An ultraviolet absorption photom-
A 1 9 eter (wavelength: 241 nm).
= WSa × Ta × ×
ASa Ca 5 Ethinylestradiol—A ‰uorophotometer (excitation wave-
Dissolution rate (z) with respect to the labeled amount length: 281 nm, ‰uorescence wavelength: 305 nm).
of ethinylestradiol (C20H24O2) Column: A stainless steel column 4.6 mm in inside di-
A 1 9 ameter and 25 cm in length, packed with octadecylsilanized
= WSb × Tb × ×
ASb Cb 5 silica gel for liquid chromatography (10 mm in particle di-
WSa: Amount (mg) of Norgestrel Reference Standard ameter).
WSb: Amount (mg) of Ethinylestradiol Reference Stand- Column temperature: A constant temperature of about
ard 259C.
Ca: Labeled amount (mg) of norgestrel (C21H28O2) in 1 Mobile phase: A mixture of acetonitrile and water (11:9).
tablet Flow rate: Adjust the ‰ow rate so that the retention time
Cb: Labeled amount (mg) of ethinylestradiol (C20H24O2) of norgestrel is about 10 minutes.
in 1 tablet System suitability—
System performance: When the procedure is run with 20
Operating conditions—
mL of the standard solution under the above operating con-
Proceed as directed in the operating conditions in the As-
ditions, ethinylestradiol, norgestrel and the internal stan-
say.
dard are eluted in this order, and the resolution between the
System suitability—
peaks of norgestrel and the internal standard is not less than
Proceed as directed in the system suitability in the Assay.
8.
System repeatability: When the test is repeated 6 times
Change the Assay to read:
with 20 mL of the standard solution under the above operat-
Assay Weigh accurately not less than 20 Norgestrel and ing conditions, the relative standard deviation of the ratios
Ethinylestradiol Tablets, and powder. Weigh accurately a of the peak area of ethinylestradiol and norgestrel to that of
portion of the powder, equivalent to about 1 mg of norges- the internal standard are not more than 1.0z, respectively.
trel (C21H28O2), add 4 mL of diluted methanol (7 in 10), add
exactly 4 mL of the internal standard solution, shake for 20
minutes, and centrifuge. Filter the supernatant liquid Add the following:
through a membrane ˆlter with pore size of not more than
0.2 mm, and use this ˆltrate as the sample solution. Separate- O‰oxacin
ly, weigh accurately about 50 mg of Norgestrel Reference
Standard and about 5 mg of Ethinylestradiol Reference オフロキサシン
Standard, and dissolve in diluted methanol (7 in 10) to make
exactly 200 mL. Pipet 4 mL of this solution, add exactly 4
mL of the internal standard solution, and use this solution as
the standard solution. Perform the test with 20 mL each of
the sample solution and the standard solution as directed un-
der the Liquid Chromatography according to the following
conditions. Calculate the ratios, Q Ta and Q Tb, of the peak C18H20FN3O4: 361.37
areas of norgestrel and ethinylestradiol to the peak area of (3RS )-9-Fluoro-2,3-dihydro-3-methyl-10-(4-
the internal standard of the sample solution and also the ra- methylpiperazin-1-yl)-7-oxo-7H-pyrido[1,2,3-de]-1,4-
tios, QSa and QSb, of the peak areas of norgestrel and benzoxazine-6-carboxylic acid [82419-36-1 ]
ethinylestradiol to the peak area of the internal standard of
the standard solution. O‰oxacin, when dried, contains not less than
99.0z and not more than 101.0z of o‰oxacin
Amount (mg) of norgestrel (C21H28O2) (C18H20FN3O4).
Q 1
= WSa × Ta × Description O‰oxacin occurs as pale yellowish white to
QSa 50
light yellowish white, crystals or crystalline powder.
Amount (mg) of ethinylestradiol (C20H24O2) It is freely soluble in acetic acid (100), slightly soluble in
Q 1 water, and very slightly soluble in acetonitrile and in ethanol
= WSb × Tb ×
QSb 50 (99.5).
WSa: Amount (mg) of Norgestrel Reference Standard A soluton of O‰oxacin in sodium hydroxide TS (1 in 20)
WSb: Amount (mg) of Ethinylestradiol Reference Stand- does not show optical rotation.
ard It is changed in color by light.
Melting point: about 2659 C (with decomposition).
Internal standard solution—A solution of diphenyl in dilut-
ed methanol (7 in 10) (1 in 50,000). Identiˆcation (1) Determine the absorption spectrum of
Supplement I, JP XIV O‹cial Monographs for Part I 1501

a solution of O‰oxacin in 0.1 mol/L hydrochloric acid TS (1 mixture of water and acetonitrile (6:1) to make 100 mL.
in 150,000) as directed under the Ultraviolet-visible Spec- When the procedure is run with 10 mL of this solution under
trophotometry, and compare the spectrum with the Refer- the above operating conditions, o‰oxacin demethyl sub-
ence Spectrum: both spectra exhibit similar intensities of ab- stance and o‰oxacin are eluted in this order with the resolu-
sorption at the same wavelengths. tion between these peaks being not less than 2.5.
(2) Determine the infrared absorption spectrum of System repeatability: When the test is repeated 6 times
O‰oxacin as directed in the potassium bromide disk method with 10 mL of the standard solution under the above operat-
under the Infrared Spectrophotometry, and compare the ing conditions, the relative standard deviation of the peak
spectrum with the Reference Spectrum: both spectra exhibit area of o‰oxacin is not more than 2.0z.
similar intensities of absorption at the same wave numbers.
Loss on drying Not less than 0.2z (1 g, 1059
C, 4 hours).
Purity (1) Heavy metals—Proceed with 2.0 g of O‰oxa-
Residue on ignition Not more than 0.10z (1 g).
cin according to Method 4, and perform the test. Prepare the
control solution with 2.0 mL of Standard Lead Solution (not Assay Weigh accurately about 0.30 g of O‰oxacin, previ-
more than 10 ppm). ously dried, dissolve in 100 mL of acetic acid (100), and ti-
(2) Related substances—Conduct this procedure without trate with 0.1 mol/L perchloric acid VS (potentiometric
exposure to light. Dissolve 10 mg of O‰oxacin in 50 mL of a titration). Perform a blank determination, and make any
mixture of water and acetonitrile (6:1), and use this solution necessary correction.
as the sample solution. Pipet 1 mL of the sample solution,
Each mL of 0.1 mol/L perchloric acid VS
and add a mixture of water and acetonitrile (6:1) to make ex-
= 36.14 mg of C18H20FN3O4
actly 20 mL. Pipet 1 mL of this solution, add a mixture of
water and acetonitrile (6:1) to make exactly 10 mL, and use
Containers and storage Containers—Tight containers.
this solution as the standard solution. Perform the test with
Storage—Light-resistant.
exactly 10 mL each of the sample solution and the standard
solution as directed under the Liquid Chromatography ac-
cording to the following conditions, and determine each
peak area by the automatic integration method: the area of Oxytetracycline Hydrochloride
the peak other than o‰oxacin obtained from the sample so-
塩酸オキシテトラサイクリン
lution is not more than 0.4 times the peak area of o‰oxacin
from the standard solution, and the total area of the peaks
Change to read except the structural formula
other than o‰oxacin from the sample solution is not more
and chemical name:
than the peak area from the standard solution.
Operating conditions—
Oxytetracycline Hydrochloride contains not less
Detector: An ultraviolet absorption photometer
than 880 mg (potency) per mg, calculated on the dried
(wavelength: 294 nm).
basis. The potency of Oxytetracycline Hydrochloride
Column: A stainless steel column 4.6 mm in inside di-
is expressed as mass (potency) of oxytetracycline
ameter and 25 cm in length, packed with octadecylsilanized
(C22H24N2O9: 460.43).
silica gel for liquid chromatography (5 mm in particle di-
ameter). Description Oxytetracycline Hydrochloride occurs as yel-
Column temperature: A constant temperature of about low, crystals or crystalline powder.
459C. It is freely soluble in water, and slightly soluble in ethanol
Mobile phase: Dissolve 7.0 g of sodium perchlorate (99.5).
monohydrate and 4.0 g of ammonium acetate in 1300 mL of
Identiˆcation (1) Determine the absorption spectrum of
water, adjust the pH to 2.2 with phosphoric acid, and add
a solution of Oxytetracycline Hydrochloride in 0.1 mol/L
240 mL of acetonitrile.
hydrochloric acid TS (1 in 50,000) as directed under the
Flow rate: Adjust the ‰ow rate so that the retention time
Ultraviolet-visible Spectrophotometry, and compare the
of o‰oxacin is about 20 minutes.
spectrum with the Reference Spectrum or the spectrum of
Time span of measurement: About 1.8 times as long as the
Oxytetracycline Hydrochloride Reference Standard pre-
retention time of o‰oxacin after the solvent peak.
pared in the same manner as the sample solution: both spec-
System suitability—
tra exhibit similar intensities of absorption at the same
Test for required detectability: Measure 1 mL of the stan-
wavelengths.
dard solution, and add a mixture of water and acetonitrile
(2) Dissolve 20 mg of Oxytetracycline Hydrochloride in
(6:1) to make exactly 20 mL. Conˆrm that the peak area of
3 mL of water, and add 1 drop of silver nitrate TS: a white
o‰oxacin obtained from 10 mL of this solution is equivalent
turbidity is produced.
to 4 to 6z of that from 10 mL of the standard solution.
System performance: To 0.5 mL of the sample solution Optical rotation [a]20 D : -188 – -2009 (0.25 g calculated

add 1 mL of a solution of o‰oxacin demethyl substance in a on the dried basis, 0.1 mol/L hydrochloric acid, 25 mL, 100
mixture of water and acetonitrile (6:1) (1 in 20,000) and a mm).
1502 O‹cial Monographs for Part I Supplement I, JP XIV

Purity (1) Heavy metals—Proceed with 0.5 g of Oxyt- acetate dihydrate (1 in 2500) and 200 mL of water, and adjust
etracycline Hydrochloride according to Method 2, and per- the pH to 7.5 with 2 mol/L sodium hydroxide TS. To this
form the test. Prepare the control solution with 2.5 mL of solution add 100 g of t-butanol and water to make 1000 mL.
Standard Lead Solution (not more than 50 ppm). Flowing of the mobile phase: Control the gradient by mix-
(2) Related substances—Dissolve 20 mg of Oxytetracy- ing the mobile phases A and B as directed in the following
cline Hydrochloride in 0.01 mol/L hydrochloric acid TS to table.
make exactly 25 mL, and use this solution as the sample so-
lution. Separately, dissolve 20 mg of 4-epioxytetracycline in Time after injection Mobile phase Mobile phase
of sample (min) A (z) B (z)
0.01 mol/L hydrochloric acid TS to make exactly 25 mL,
and use this solution as 4-epioxytetracycline stock solution. 0 – 20 70 → 10 30 → 90
Separately, dissolve 20 mg of tetracycline hydrochloride in 20 – 35 10 → 20 90 → 80
0.01 mol/L hydrochloric acid TS to make exactly 25 mL,
Flow rate: 1.0 mL/min
and use this solution as tetracycline hydrochloride stock so-
Time span of measurement: About 3.5 times as long as the
lution. Separately, dissolve 8 mg of b-apooxytetracycline in
retention time of oxytetracycline after the solvent peak.
5 mL of 0.01 mol/L sodium hydroxide TS, add 0.01 mol/L
System suitability—
hydrochloric acid TS to make exactly 100 mL, and use this
Test for required detectability: Pipet 1 mL of 4-
solution as b-apooxytetracycline stock solution. Pipet 1 mL
epioxytetracycline stock solution, and add 0.01 mol/L
of 4-epioxytetracycline stock solution, 4 mL of tetracycline
hydrochloric acid TS to make exactly 200 mL. Pipet 4 mL of
hydrochloride stock solution and 40 mL of b-apooxytetracy-
this solution, and add 0.01 mol/L hydrochloric acid TS to
cline stock solution, add 0.01 mol/L hydrochloric acid TS to
make exactly 20 mL. Conˆrm that the peak area of 4-epiox-
make exactly 200 mL, and use this solution as the standard
ytetracycline obtained from 20 mL of this solution is equiva-
solution. Perform the test with exactly 20 mL each of the
lent to 14 to 26z of that from 20 mL of the standard
sample solution and the standard solution as directed under
solution.
the Liquid Chromatography according to the following con-
System performance: Dissolve 8 mg of a-apooxytetracy-
ditions, and determine each peak area by the automatic in-
cline in 5 mL of 0.01 mol/L sodium hydroxide TS, add 0.01
tegration method: the peak areas of 4-epioxytetracycline and
mol/L hydrochloric acid TS to make 100 mL, and use this
tetracycline obtained from the sample solution are not more
solution as a-apooxytetracycline stock solution. Mix 3 mL
than each of the peak area obtained from the standard solu-
of the sample solution, 2 mL of 4-epioxytetracycline stock
tion, and the total area of the peaks, a-apooxytetracycline
solution, 6 mL of tetracycline hydrochloride stock solution,
having the relative retention time of about 2.1 with respect
6 mL of b-apooxytetracycline stock solution and 6 mL of a-
to oxytetracycline, b-apooxytetracycline and the peaks,
apooxytetracycline stock solution, and add 0.01 mol/L
which appear between a-apooxytetracycline and b-apoox-
hydrochloric acid TS to make 50 mL. When the procedure is
ytetracycline, is not more than the peak area of b-apoox-
run with 20 mL of this solution under the above operating
ytetracycline from the standard solution. The peak area of 2-
conditions, 4-epioxytetracycline, oxytetracycline, tetracy-
acetyl-2-decarboxamide oxytetracycline, which appears after
cline, a-apooxytetracycline and b-apooxytetracycline are
the principal peak, obtained from the sample solution is not
eluted in this order with the resolutions between the peaks,
more than 4 times the peak area of 4-epioxytetracycline from
4-epioxytetracycline and oxytetracycline, oxytetracycline
the standard solution.
and tetracycline, and a-apooxytetracycline and b-apoox-
Operating conditions—
ytetracycline being not less than 4, not less than 5 and not
Detector: An ultraviolet absorption photometer
less than 4, respectively, and the symmetry coe‹cient of the
(wavelength: 254 nm).
peak of oxytetracycline is not more than 1.3.
Column: A stainless steel column 4.6 mm in inside di-
System repeatability: Pipet 1 mL of 4-epioxytetracycline
ameter and 25 cm in length, packed with styrene-divinylben-
stock solution, and add 0.01 mol/L hydrochloric acid TS to
zene copolymer for liquid chromatography (8 mm in particle
make exactly 200 mL. When the test is repeated 6 times with
diameter).
20 mL of this solution under the above operating conditions,
Column temperature: A constant temperature of about
the relative standard deviation of the peak area of 4-epiox-
609C.
ytetracycline is not more than 2.0z.
Mobile phase A: Mix 60 mL of 0.33 mol/L potassium di-
hydrogen phosphate TS, 100 mL of a solution of Loss on drying Not more than 2.0z (1 g, in vacuum,
tetrabutylammonium hydrogensulfate (1 in 100), 10 mL of a 609C, 3 hours).
solution of disodium dihydrogen ethylenediamine tetra-
Residue on ignition Not more than 0.5z (1 g).
acetate dihydrate (1 in 2500) and 200 mL of water, and adjust
the pH to 7.5 with 2 mol/L sodium hydroxide TS. To this Assay Weigh accurately an amount of Oxytetracycline
solution add 30 g of t-butanol and water to make 1000 mL. Hydrochloride and Oxytetracycline Hydrochloride Refer-
Mobile phase B: Mix 60 mL of 0.33 mol/L potassium ence Standard, equivalent to about 50 mg (potency), and
dihydrogen phosphate TS, 50 mL of a solution of dissolve each in diluted hydrochloric acid (1 in 100) to make
tetrabutylammonium hydrogensulfate (1 in 100), 10 mL of a exactly 50 mL. Pipet 5 mL each of these solutions, add dilut-
solution of disodium dihydrogen ethylenediamine tetra- ed methanol (3 in 20) to make exactly 50 mL, and use these
Supplement I, JP XIV O‹cial Monographs for Part I 1503

solutions as the sample solution and the standard solution. Identiˆcation (1) To 4 mg of Peplomycin Sulfate add
Perform the test with exactly 20 mL each of the sample solu- 5 mL of copper (II) sulfate TS, and dissolve in water to make
tion and the standard solution as directed under the Liquid 100 mL. Determine the absorption spectrum of this solution
Chromatography according to the following conditions, and as directed under the Ultraviolet-visible Spectrophotometry,
determine the peak areas, AT and AS, of oxytetracycline. and compare the spectrum with the Reference Spectrum:
both spectra exhibit similar intensities of absorption at the
Amount [ mg (potency)] of oxytetracycline (C22H24N2O9)
same wavelengths.
A
= WS × T × 1000 (2) Dissolve 10 mg each of Peplomycin Sulfate and
AS
Peplomycin Sulfate Reference Standard in 6 mL of water,
WS: Amount [mg (potency)] of Oxytetracycline add 0.5 mL of a solution of copper (II) sulfate pentahydrate
Hydrochloride Reference Standard (1 in 125), and use these solutions as the sample solution and
Operating conditions— the standard solution. Perform the test with 10 mL each of
Detector: An ultraviolet absorption photometer these solutions as directed under the Liquid Chro-
(wavelength: 263 nm). matography according to the following conditions: the
Column: A stainless steel column 4.6 mm in inside di- retention time of the principal spot obtained from the sam-
ameter and 25 cm in length, packed with strongly acidic ion ple solution is the same as that from the standard soution.
exchange resin for liquid chromatography (5 mm in particle Operating conditions—
diameter). Detector, column, column temperature, mobile phase
Column temperature: A constant temperature of about stock solution, mobile phase A, mobile phase B, ‰owing of
309C. the mobile phase, and ‰ow rate: Proceed as directed in the
Mobile phase: Dissolve 3.402 g of potassium dihydrogen operating conditions in the Purity (3).
phosphate and 9.306 g of disodium dihydrogen ethylenedia- (3) A solution of Peplomycin Sulfate (1 in 200) responds
mine tetraacetate dihydrate in 700 mL of water, add 300 mL to the Qualitative Tests (1) and (2) for sulfate.
of methanol, and adjust the pH to 4.5 with dilute
Optical rotation [a]20D : -2 – -59(0.1 g calculated on the
hydrochloric acid.
dried basis, 0.1 mol/L phosphate buŠer solution, pH 5.3, 10
Flow rate: Adjust the ‰ow rate so that the retention time
mL, 100 mm).
of oxytetracycline is about 7 minutes.
System suitability— pH The pH of a solution obtained by dissolving 0.10 g of
System performance: When the procedure is run with 20 Peplomycin Sulfate in 20 mL of water is between 4.5 and
mL of the standard solution under the above operating con- 6.0.
ditions, the theoretical plates and the symmetrical coe‹cient
Purity (1) Clarity and color of solution—Dissolve 80 mg
of the peak of oxytetracycline are not less than 1000 and not
of Peplomycin Sulfate in 4 mL of water: the solution is clear
more than 2.0, respectively.
and colorless.
System repeatability: When the test is repeated 6 times
(2) Copper—Dissolve exactly 75 mg of Peplomycin Sul-
with 20 mL of the standard solution under the above operat-
fate in exactly 10 mL of diluted nitric acid (1 in 100), and use
ing conditions, the relative standard deviation of the peak
this solution as the sample solution. Separately, to 5.0 mL of
area of oxytetracycline is not more than 1.0z.
Standard Copper Stock Solution add diluted nitric acid (1 in
Containers and storage Containers—Tight containers. 100) to make exactly 100 mL. To 3.0 mL of this solution add
Storage—Light-resistant. diluted nitric acid (1 in 100) to make exactly 100 mL, and use
this solution as the standard solution. Perform the test with
the sample solution and the standard solution as directed un-
Peplomycin Sulfate der the Atomic Absorption Spectrophotometry according to
the following conditions: the absorbance of the sample solu-
硫酸ペプロマイシン tion is not more than that of the standard solution (not more
than 200 ppm).
Change to read except the structural formula Gas: Combustible gas—Acetylene
and chemical name: Supporting gas—Air
Lamp: Copper hollow cathode lamp
Peplomycin Sulfate contains not less than 865 mg Wavelength: 324.8 nm
(potency) per mg, calculated on the dried basis. The (3) Related substances—Dissolve about 10 mg of
potency of Peplomycin Sulfate is expressed as mass Peplomycin Sulfate in 6 mL of water, add 0.5 mL of a solu-
(potency) of peplomycin (C61H88N18O21S2: 1473.59). tion of copper (II) sulfate pentahydrate (1 in 125), and use
this solution as the sample solution. Perform the test with 10
Description Peplomycin Sulfate occurs as a white to light
mL of the sample solution as directed under the Liquid Chro-
yellowish white powder.
matography according to the following conditions. Deter-
It is freely soluble in water, and practically insoluble in
mine the areas of the peaks, appeared after the peak of cop-
ethanol (95).
per sulfate, by the automatic integration method, and calcu-
It is hygroscopic.
1504 O‹cial Monographs for Part I Supplement I, JP XIV

late the amounts of them by the area percentage method: the (1) Test organism—Mycobacterium smegmatis ATCC
total amount of the peaks other than peplomycin is not more 607
than 7z. (2) Agar media for seed and base layer, and for transfer-
Operating conditions— ring test organism
Detector: An ultraviolet absorption photometer Glycerin 10.0 g
(wavelength: 254 nm). Peptone 10.0 g
Column: A stainless steel column 4.6 mm in inside di- Meat extract 10.0 g
ameter and 25 cm in length, packed with octadecylsilanized Sodium chloride 3.0 g
silica gel for liquid chromatography (7 mm in particle di- Agar 15.0 g
ameter). Water 1000 mL
Column temperature: A constant temperature of about Mix all the ingredients and adjust the pH of the solution
409C. with sodium hydroxide TS so that it will be 6.9 to 7.1 after
Mobile phase stock solution: Dissolve 0.96 g of sodium 1- sterilization.
pentanesulfonate and 1.86 g of disodium dihydrogen (3) Liquid medium for suspending test organism
ethylenediamine tetraacetate dihydrate in 1000 mL of water Glycerin 10.0 g
and 5 mL of acetic acid (100), and adjust the pH to 4.3 with Peptone 10.0 g
ammonia TS. Meat extract 10.0 g
Mobile phase A: A mixture of mobile phase stock solution Sodium chloride 3.0 g
and methanol (9:1). Water 1000 mL
Mobile phase B: A mixture of mobile phase stock solution Mix all the ingredients and adjust the pH of the solution
and methanol (3:2). with sodium hydroxide TS so that it will be 6.9 to 7.1 after
Flowing of the mobile phase: Control the gradient by mix- sterilization.
ing the mobile phases A and B as directed in the following (4) Preparation of agar medium of seeded layer—Inocu-
table. late the test organism onto the slant of the agar medium for
transferring test organism, and incubate the slant at 279C
Time after injection Mobile phase Mobile phase for 40 to 48 hours. Inoculate the subcultured test organism
of sample (min) A (z) B (z)
into 100 mL of the liquid medium for suspending test organ-
0 – 60 100 → 0 0 → 100 ism, incubate at 25 to 279C for 5 days while shaking, and use
60 – 75 0 100 this suspension as the suspension of the test organism. Keep
the suspension of the test organism at a temperature of not
Flow rate: 1.2 mL per minute.
exceeding 59 C and use within 14 days. Add 0.5 mL of the
Time span of measurement: As long as 20 minutes after
suspension of the test organism in 100 mL of the Agar medi-
elution of peplomycin after the peak of copper sulfate.
um for seed layer previously kept at 489 C, mix thoroughly,
System suitability—
and use this as the agar medium of seeded layer.
Test for required detectability: Measure exactly 1 mL of
(5) Preparation of cylinder-agar plate—Proceed as
the sample solution, add water to make exactly 100 mL, and
directed in 7. Preparation of cylinder-agar plates under the
use this solution as the solution for system suitability test.
Microbial Assay for Antibiotics with the exception of the
Pipet 1 mL of the solution for system suitability test, and
amounts of the agar medium for base layer and the agar
add water to make exactly 10 mL. Conˆrm that the peak
medium of seeded layer to put in the Petri dish, which are
area of peplomycin obtained from 10 mL of this solution is
5.0 mL and 8.0 mL, respectively.
equivalent to 7 to 13z of that from 10 mL of the solution for
(6) Standard solutions—Weigh accurately an amount of
system suitability test.
Peplomycin Sulfate Reference Standard, equivalent to about
System performance: When the procedure is run with 10
20 mg (potency), dissolve in 0.1 mol/L phosphate buŠer so-
mL of the sample solution under the above operating condi-
lution, pH 6.8 to make exactly 100 mL, and use this solution
tions, the number of theoretical plates and the symmetry
as the standard stock solution. Keep the standard stock solu-
coe‹cient of the peak of peplomycin are not less than 30,000
tion at a temperature not exceeding 59 C, and use within 15
and not more than 2.0, respectively.
days. Take exactly a suitable amount of the standard stock
System repeatability: When the test is repeated 6 times
solution before use, add 0.1 mol/L phosphate buŠer solu-
with 10 mL of the sample solution under the above operating
tion, pH 6.8 to make solutions so that each mL contains 4 mg
conditions, the relative standard deviation of the peak area
(potency) and 2 mg (potency), and use these solutions as the
of peplomycin is not more than 2.0z.
high concentration standard solution and the low concentra-
Loss on drying Not more than 3.0z (60 mg, in vacuum, tion standard solution, respectively.
phosphorus (V) oxide, 609C, 3 hours). Handle the sample (7) Sample solutions—Weigh accurately an amount of
avoiding absorption of moisture. Peplomycin Sulfate, equivalent to about 20 mg (potency),
and dissolve in 0.1 mol/L phosphate buŠer solution, pH 6.8
Assay Perform the test according to the Cylinder-plate
to make exactly 100 mL. Take exactly a suitable amount of
method as directed under the Microbial Assay for Antibiot-
this solution, add 0.1 mol/L phosphate buŠer solution, pH
ics according to the following conditions.
6.8 to make solutions so that each mL contains 4 mg (poten-
Supplement I, JP XIV O‹cial Monographs for Part I 1505

cy) and 2 mg (potency), and use these solutions as the high area of pethidine is not more than 2.0z.
concentration sample solution and the low concentration
sample solution, respectively.
Add the following:
Containers and storage Containers—Tight containers

Phenethicillin Potassium
Pethidine Hydrochloride フェネチシリンカリウム

塩酸ペチジン

Change the Purity (3) to read:


Purity
(3) Related substances—Dissolve 0.05 g of Pethidine C17H19KN2O5S: 402.51
Hydrochloride in 20 mL of the mobile phase, and use this Monopotassium (2 S,5 R,6 R )-3,3-dimethyl-7-oxo-6-
solution as the sample solution. Pipet 1 mL of the sample so- [(2 RS )-2-phenoxypropanoylamino]-4-thia-1-
lution, add the mobile phase to make exactly 100 mL, and azabicyclo[3.2.0]heptane-2-carboxylate [132-93-4 ]
use this solution as the standard solution. Perform the test
with exactly 20 mL each of the sample solution and the stan- Phenethicillin Potassium contains not less than 1400
dard solution as directed under the Liquid Chromatography units per mg, calculated on the dried basis. The poten-
according to the following conditions. Determine each peak cy of Phenethicillin Potassium is expressed as unit
area obtained from both solutions by the automatic integra- based on the amount of phenethicillin potassium
tion method: the total area of the peaks other than that of (C17H19KN2O5S). 1 unit of Phenethicillin Potassium is
pethidine from the sample solution is not larger than the equivalent to 0.68 mg of phenethicillin potassium
peak area of perthidine from the standard solution. (C17H19KN2O5S).
Operating conditions—
Description Phenethicillin Potassium occurs as a white to
Detector: An ultraviolet absorption photometer
light yellowish white crystalline powder.
(wavelength: 257 nm).
It is freely soluble in water, and slightly soluble in ethanol
Column: A stainless steel column 4.6 mm in inside di-
(99.5).
ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di- Identiˆcation (1) Determine the absorption spectrum of
ameter). a solution of Phenethicillin Potassium (1 in 5000) as directed
Column temperature: A constant temperature of about under the Ultraviolet-visible Spectrophotometry, and com-
409C. pare the spectrum with the Reference Spectrum: both spec-
Mobile phase: Dissolve 2.0 g of sodium lauryl sulfate in tra exhibit similar intensities of absorption at the same
1000 mL of diluted phosphoric acid (1 in 1000), adjust the wavelengths.
pH to 3.0 with sodium hydroxide TS, and to 550 mL of this (2) Determine the infrared absorption spectrum of
solution add 450 mL of acetonitrile. Phenethicillin Potassium as directed in the potassium
Flow rate: Adjust the ‰ow rate so that the retention time bromide disk method under the Infrared Spectrophotomet-
of pethidine is about 7 minutes. ry, and compare the spectrum with the Reference Spectrum:
Time span of measurement: About 2 times as long as the both spectra exhibit similar intensities of absorption at the
retention time of pethidine after the solvent peak. same wave numbers.
System suitability— (3) Phenethicillin Potassium responds to the Qualitative
Test for required detection: To exactly 2 mL of the stan- Test (1) for potassium salt.
dard solution add the mobile phase to make exactly 20 mL.
Optical rotation [a]20D : +217 – +2449(1 g calculated on
Conˆrm that the peak area of pethidine obtained from 20 mL
the dried basis, phosphate TS, 100 mL, 100 mm).
of this solution is equivalent to 5 to 15z of that of pethidine
obtained from 20 mL of the standard solution. L-a-Phenethicillin potassium Dissolve about 50 mg of
System performance: To 2 mL each of the sample solution Phenethicillin Potassium in the mobile phase to make ex-
and a solution of isoamyl parahydroxybenzoate in the mo- actly 50 mL, and use this solution as the sample solution.
bile phase (1 in 50,000) add the mobile phase to make 10 mL. Perform the test with 10 mL of the sample solution as direct-
When the procedure is run with 20 mL of this solution ac- ed under the Liquid Chromatography according to the fol-
cording to the above operating conditions, pethidine and lowing conditions, and determine the peak areas, AD and
isoamyl parahydroxybenzoate are eluted in this order with AL, of D -a-phenethicillin and L-a-phenethicillin by the auto-
the resolution between these peaks being not less than 2.0. matic integration method: AL/(AD + AL ) is between 0.50
System repeatability: When the test is repeated 6 times and 0.70.
with 20 mL of the standard solution under the above operat- Operating conditions—
ing conditions, the relative standard deviation of the peak Detector: An ultraviolet absorption photometer
1506 O‹cial Monographs for Part I Supplement I, JP XIV

(wavelength: 254 nm). Loss on drying Not more than 1.0z (0.1 g, in vacuum,
Column: A stainless steel column 6 mm in inside diameter 609C, 3 hours).
and 15 cm in length, packed with octadecylsilanized silica gel
Assay Weigh accurately an amount of Phenethicillin
for liquid chromatography (5 mm in particle diameter).
Potassium and dried L-Phenethicillin Potassium Reference
Column temperature: A constant temperature of about
Standard, equivalent to about 40,000 units, dissolve each in
309C.
phosphate buŠer solution, pH 6.0 to make exactly 20 mL,
Mobile phase: Adjust the pH of a mixture of a solution of
and use these solutions as the sample solution and the stan-
diammonium hydrogen phosphate (1 in 150) and acetonitrile
dard solution, respectively. Pipet 2 mL each of these solu-
(41:10) to 7.0 with phosphoric acid.
tions in 100-mL glass-stoppered ‰asks, add 2.0 mL of sodi-
Flow rate: Adjust the ‰ow rate so that the retention time
um hydroxide TS to them, and allow to stand for exactly 15
of L-a-phenethicillin is about 25 minutes.
minutes. To them add 2.0 mL of diluted hydrochloric acid (1
System suitability—
in 10) and exactly 10 mL of 0.005 mol/L iodine VS, and al-
System performance: When the procedure is run with 10
low them to stand for exactly 15 minutes. Add 0.2 – 0.5 mL
mL of the sample solution under the above operating condi-
of starch TS, and titrate with 0.01 mol/L sodium thiosulfate
tions, D-a-phenethicillin and L-a-phenethicillin are eluted in
VS until the color of the solution disappears. Separately, to
this order with the resolution between these peaks being not
exactly 2 mL each of the sample solution and the standard
less than 1.5.
solution add exactly 10 mL of 0.005 mol/L iodine VS, then
System repeatability: When the test is repeated 6 times
proceed in the same manner as above without allowing to
with 10 mL of the sample solution under the above operating
stand for 15 minutes as a blank determination, and make
conditions, the relative standard deviation of the peak area
any necessary correction. Determine the volumes, VT and
of L-a-phenethicillin is not more than 2.0z.
VS, of 0.005 mol/L iodine VS consumed in the sample solu-
Purity (1) Heavy metals—Proceed with 1.0 g of tion and the standard solution.
Phenethicillin Potassium according to Method 2, and per-
VT
form the test. Prepare the control solution with 1.0 mL of Amount (unit) of C17H19KN2O5S = WS ×
VS
Standard Lead Solution (not more than 10 ppm).
(2) Arsenic—Prepare the test solution with 1.0 g of WS: Amount (unit) of L-Phenethicillin Potassium Refer-
Phenethicillin Potassium according to Method 4 and, per- ence Standard
form the test (not more than 2 ppm).
Containers and storage Containers—Well-closed contain-
(3) Related substances—Dissolve 50 mg of Phenethicil-
ers.
lin Potassium in 50 mL of the mobile, and use this solution
as the sample solution. Pipet 1 mL of the sample solution,
add the mobile phase to make exactly 100 mL, and use this
solution as the standard solution. Perform the test with
exactly 10 mL each of the sample solution and the standard
solution as directed under the Liquid Chromatography ac-
cording to the following conditions, and determine each
peak area by the automatic integration method: the total of
the peak areas other than D-a-phenethicillin and L-a-
phenethicillin obtained from the sample solution is not more
than 5 times the total of the peak areas of D-a-phenethicillin
and L-a-phenethicillin from the standard solution.
Operating conditions—
Detector, column, column temperature, mobile phase,
and ‰ow rate: Proceed as directed in the operating condi-
tions in the L-a-Phenethicillin potassium.
Time span of measurement: About 1.5 times as long as the
retention time of L-a-phenethicillin.
System suitability—
Test for required detectability: Measure exactly 2 mL of
the standard solution, and add the mobile phase to make ex-
actly 10 mL. Conˆrm that the peak area of L-a-phenethicil-
lin obtained from 10 mL of this solution is equivalent to 14 to
26z of that from 10 mL of the standard solution.
System performance, and system repeatability: Proceed as
directed in the system suitability in the L-a-Phenethicillin
potassium.
Supplement I, JP XIV O‹cial Monographs for Part I 1507

Phytonadione Add the following:

フィトナジオン Pimaricin
Change the Description to read: Natamycin
Description Phytonadione is a clear yellow to orange-yel- ピマリシン
low, viscous liquid.
It is miscible with isooctane.
It is sparingly soluble in ethanol (99.5), and practically in-
soluble in water.
It decomposes gradually and changes to a red-brown by
light.
20
Speciˆc gravity d 20 : about 0.967

Change the Identiˆcation to read:


Identiˆcation (1) Determine the absorption spectrum of C33H47NO13: 665.73
a solution of Phytonadione in isooctane (1 in 100,000) as (1 R *,3S *,5R *,7R *,8E,12 R *,14E,16E,18E,20E,22 R *,
directed under the Ultraviolet-visible Spectrophotometry, 24S *,25R *,26S *)-22-(3-Amino-3,6-dideoxy-b-D-
and compare the spectrum with the Reference Spectrum 1: mannopyranosyloxy)-1,3,26-trihydroxy-12-methyl-
both spectra exhibit similar intensities of absorption at the 10-oxo-6,11,28-trioxatricyclo[22.3.1.05,7]octacosa-
same wavelengths. Separately, determine the absorption 8,14,16,18,20-pentaene-25-carboxylic acid [7681-93-8 ]
spectrum of a solution of Phytonadione in isooctane (1 in
10,000) as directed under the Ultraviolet-visible Spec- Pimaricin contains not less than 900 mg (potency)
trophotometry, and compare the spectrum with the Refer- per mg, calculated on the anhydrous basis. The poten-
ence Spectrum 2: both spectra exhibit similar intensities of cy of Pimaricin is expressed as mass (potency) of
absorption at the same wavelengths. pimaricin (C33H47NO13).
(2) Determine the infrared absorption spectrum of Description Pimaricin occurs as white to yellowish white
Phytonadione as directed in the liquid ˆlm method under the crystalline powder.
Infrared Spectrophotometry, and compare the spectrum It is slightly soluble in methanol and in acetic acid (100),
with the Reference Spectrum: both spectra exhibit similar in- and practically insoluble in water and in ethanol (99.5).
tensities of absorption at the same wave numbers.
Identiˆcation (1) To 3 mg of Pimaricin add 1 mL of
Change the Purity to read: hydrochloric acid, and mix: a blue-purple color appears.
(2) Dissolve 5 mg of Pimaricin in a solution of acetic
Purity (1) Ratio of absorbances—Determine the absor- acid (100) in methanol (1 in 100) to make 1000 mL. Deter-
bances, A1, A2 and A3, of a solution of Phytonadione in mine the absorption spectrum of this solution as directed un-
isooctane (1 in 100,000) at 248.5 nm, 253.5 nm and 269.5 der the Ultraviolet-visible Spectrophotometry, and compare
nm, respectively: the ratio A2/A1 is between 0.69 and 0.73, the spectrum with the Reference Spectrum or the spectrum
and the ratio A2/A3 is between 0.74 and 0.78. Determine the of a solution of Pimaricin Reference Standard prepared in
absorbances, A4 and A5, of a solution of Phytonadione in the same manner as the sample solution: both spectra exhibit
isooctane (1 in 10,000) at 284.5 nm and 326 nm, respectively: similar intensities of absorption at the same wavelengths.
the ratio A4/A5 is between 0.28 and 0.34.
(2) Heavy metals—Carbonize 1.0 g of Phytonadione by Optical rotation [a]20
D : +243 – +2599(0.1 g, acetic acid

gentle heating. Cool, add 10 mL of a solution of magnesium (100), 25 mL, 100 mm).
nitrate hexahydrate in ethanol (95) (1 in 10), and ignite the Purity (1) Heavy metals—Proceed with 1.0 g of Pimari-
ethanol to burn. Cool, add 1 mL of sulfuric acid, proceed cin according to Method 4, and perform the test. Prepare the
according to Method 4, and perform the test. Prepare the control solution with 3.0 mL of Standard Lead Solution (not
control solution with 2.0 mL of Standard Lead Solution (not more than 30 ppm).
more than 20 ppm). (2) Related substances—Dissolve 20 mg of Pimaricin in
(3) Menadione—Dissolve 0.020 g of Phytonadione in methanol to make 100 mL, and use this solution as the sam-
0.5 mL of a mixture of water and ethanol (95) (1:1), add 1 ple solution. Perform the test with 10 mL of the sample solu-
drop of a solution of 3-methyl-1-phenyl-5-pyrazolone in tion as directed under the Liquid Chromatography accord-
ethanol (95) (1 in 20) and 1 drop of ammonia solution (28), ing to the following conditions, and determine the total area
and allow to stand for 2 hours: no blue-purple color of the peaks other than pimaricin by the automatic integra-
develops. tion method: not more than 4.0z.
1508 O‹cial Monographs for Part I Supplement I, JP XIV

Operating conditions— Pipemidic Acid Trihydrate


Detector: An ultraviolet absorption photometer
(wavelength: 303 nm). ピペミド酸三水和物
Column: A stainless steel column 3.9 mm in inside di-
ameter and 30 cm in length, packed with octadecylsilanized Change the origin/limits of content to read:
silica gel for liquid chromatography (10 mm in particle di-
ameter). Pipemidic Acid Trihydrate, when dried, contains
Column temperature: A constant temperature of about not less than 98.5z and not more than 101.0z of
409C. C14H17N5O3 (mol. wt.: 303.32).
Mobile phase: Dissolve 1.0 g of ammonium acetate in
1000 mL of a mixture of water, methanol and tetrahydrofu- Change the Description to read:
ran (47:44:2).
Description Pipemidic Acid Trihydrate occurs as a pale
Flow rate: Adjust the ‰ow rate so that the retention time
yellow, crystalline powder.
of pimaricin is about 10 minutes.
It is freely soluble in acetic acid (100), very slightly soluble
Time span of measurement: About 3 times as long as the
in water and in ethanol (99.5).
retention time of pimaricin.
It dissolves in sodium hydroxide TS.
System suitability—
It is gradually colored by light.
Test for required detectability: Measure exactly 1 mL of
Melting point: about 2509 C (with decomposition).
the sample solution, add methanol to make exactly 100 mL,
and use this solution as the solution for system suitability
test. Pipet 1 mL of the solution for system suitability test,
Change the Identiˆcation to read:
and add methanol to make exactly 10 mL. Conˆrm that the Identiˆcation (1) Dissolve 0.1 g of Pipemidic Acid Trihy-
peak area of pimaricin obtained from 10 mL of this solution drate in 20 mL of sodium hydroxide TS, and dilute with
is equivalent to 7 to 13z of that from 10 mL of the solution water to make 200 mL. To 1 mL of the solution add water to
for system suitability test. make 100 mL. Determine the absorption spectrum of the
System performance: When the procedure is run with solution as directed under the Ultraviolet-visible Spec-
10 mL of the solution for system suitability test under the trophotometry, and compare the spectrum with the Refer-
above operating conditions, the number of theoretical plates ence Spectrum: both spectra exhibit similar intensities of ab-
and the symmetry coe‹cient of the peak of pimaricin are not sorption at the same wavelengths.
less than 1500 and not more than 2.0, respectively. (2) Determine the infrared absorption spectrum of
System repeatability: When the test is repeated 6 times Pipemidic Acid Trihydrate as directed in the potassium
with 10 mL of the solution for system suitability test under bromide disk method under the Infrared Spectrophotomet-
the above operating conditions, the relative standard devia- ry, and compare the spectrum with the Reference Spectrum:
tion of the peak area of pimaricin is not more than 2.0z. both spectra exhibit similar intensities of absorption at the
same wave numbers.
Water Between 6.0z and 9.0z (0.2 g, volumetric titra-
tion, direct titration).
Change the Assay to read:
Assay Weigh accurately an amount of Pimaricin and
Assay Weigh accurately about 0.3 g of Pipemidic Acid Tri-
Pimaricin Reference Standard, equivalent to about 25 mg
hydrate, previously dried, dissolve in 40 mL of acetic acid
(potency), and dissolve each in methanol to make exactly
(100), and titrate with 0.1 mol/L perchloric acid VS (poten-
100 mL. Pipet 2 mL each of these solution, add a solution of
tiometric titration). Perform a blank determination, and
acetic acid (100) in methanol (1 in 100) to make exactly 100
make any necessary correction.
mL, and use these solutions as the sample solution and the
standard solution. Determine the absorbances at 295.5 nm, Each mL of 0.1 mol/L perchloric acid VS
AT1 and AS1, at 303 nm, AT2 and AS2, and at 311 nm, AT3 = 30.33 mg of C14H17N5O3
and AS3, of the sample solution and the standard solution as
directed under the Ultraviolet-visible Spectrophotometry.

Amount [ mg (potency)] of C33H47NO13 Piperacillin Sodium


A + A T3
A T2 - T 1 ピペラシリンナトリウム
2
= WS × × 1000
A S1 + A S3
A S2 -
2
Change the Description to read:
WS: Amount [mg (potency)] of Pimaricin Reference Description Piperacillin Sodium occurs as a white powder
Standard or mass.
It is very soluble in water, freely soluble in methanol and
Containers and storage Containers—Tight containers. in ethanol (95), and practically insoluble in acetonitrile.
Storage—Light resistant.
Supplement I, JP XIV O‹cial Monographs for Part I 1509

Add the following: the solution is clear and red.


(2) Heavy metals—Proceed with 1.0 g of Pirarubicin ac-
Pirarubicin cording to Method 2, and perform the test. Prepare the con-
trol solution with 2.0 mL of Standard Lead Solution (not
ピラルビシン more than 20 ppm).
(3) Related substances—Dissolve 10 mg of Pirarubicin
in 20 mL of the mobile phase, and use this solution as the
sample solution. Pipet 1 mL of the sample solution, add the
mobile phase to make exactly 200 mL, and use this solution
as the standard solution. Perform the test with exactly 20 mL
each of the sample solution and the standard solution as
directed under the Liquid Chromatography according to the
following conditions, and determine each peak area by the
automatic integration method: the peak area of doxorubi-
cin, having the relative retention time of about 0.45 with
C32H37NO12: 627.64
respect to pirarubicin, and the area of the peak, having the
(2S,4S )-4-{3-Amino-2,3,6-trideoxy-4-O-[(2 R )-
relative retention time of about 1.2 with respect to pirarubi-
tetrahydropyran-2-yl]-a-L-lyxo-hexopyranosyloxy}-
cin, obtained from the sample solution are not more than the
1,2,3,4-tetrahydro-2,5,12-trihydroxy-2-hydroxyacetyl-
peak area of pirarubicin from the standard solution, respec-
7-methoxynaphthacene-6,11-dione [72496-41-4 ]
tively, and the sum of the areas of the peaks, having the rela-
tive retention times of about 1.9 and about 2.0 with respect
Pirarubicin contains not less than 950 mg (potency) to pirarubicin, from the sample solution is not more than 5
per mg, calculated on the anhydrous basis. The poten- times the peak area of pirarubicin from the standard solu-
cy of Pirarubicin is expressed as mass (potency) of tion. For these calculations, use the peak area for doxorubi-
pirarubicin (C32H37NO12). cin after multiplying by the response factor 0.94 and the area
Description Pirarubicin occurs as a red-orange crystalline for the two peaks, having the relative retention times of
powder. about 1.9 and about 2.0, after multiplying by their response
It is soluble in chloroform, very slightly soluble in acetoni- factors, 1.09, respectively.
trile, in methanol and in ethanol (99.5), and practically in- Operating conditions—
soluble in water. Detector, column, column temperature, mobile phase,
and ‰ow rate: Proceed as directed in the operating condi-
Identiˆcation (1) Dissolve 10 mg of Pirarubicin in 80 mL
tions in the Assay.
of methanol and 6 mL of diluted hydrochloric acid (1 in
Time span of measurement: About 4 times as long as the
5000), and add water to make 100 mL. To 10 mL of this so-
retention time of pirarubicin.
lution add diluted methanol (4 in 5) to make 100 mL. Deter-
System suitability—
mine the absorption spectrum of this solution as directed un-
Test for required detectability: Measure exactly 2 mL of
der the Ultraviolet-visible Spectrophotometry, and compare
the standard solution, and add the mobile phase to make ex-
the spectrum with the Reference Spectrum or the spectrum
actly 10 mL. Conˆrm that the peak area of pirarubicin ob-
of a solution of Pirarubicin Reference Standard prepared in
tained from 20 mL of this solution is equivalent to 14 to 26z
the same manner as the sample solution: both spectra exhibit
of that from 20 mL of the standard solution.
similar intensities of absorption at the same wavelengths.
System performance, and system repeatability: Proceed as
(2) Dissolve 5 mg each of Pirarubicin and Pirarubicin
directed in the system suitability in the Assay.
Reference Standard in 5 mL of chloroform, and use these
solutions as the sample solution and the standard solution. Water Not more than 2.0z (0.1 g, volumetric titration,
Perform the test with these solutions as directed under the direct titration).
Thin-layer Chromatography. Spot 5 mL each of the sample
Assay Weigh accurately an amount of Pirarubicin and
solution and the standard solution on a plate of silica gel for
Pirarubicin Reference Standard, equivalent to about 10 mg
thin-layer chromatography. Develop the plate with a mix-
(potency), and dissolve in the mobile phase to make exactly
ture of chloroform and methanol (5:1) to a distance of about
10 mL. Pipet 5 mL of these solutions, add exactly 5 mL of
10 cm, and air-dry the plate. Examine the spots with the
the internal standard solution, and use these solutions as the
necked eye: the principal spot obtained from the sample
sample solution and the standard solution. Perform the test
solution and the spot from the standard solution show a red-
with 20 mL each of the sample solution and the standard so-
orange color and the same Rf value.
lution as directed under the Liquid Chromatography accord-
Optical rotation [a]20
D : +195 – +2159(10 mg, chloroform, ing to the following conditions, and determine the ratios, Q T
10 mL, 100 mm). and QS, of the peak area of pirarubicin to that of the internal
standard.
Purity (1) Clarity and color of solution—Dissolve 10 mg
of Pirarubicin in 10 mL of 0.01 mol/L hydrochloric acid TS:
1510 O‹cial Monographs for Part I Supplement I, JP XIV
QT
Amount [ mg (potency)] of C32H37NO12 = WS × × 1000 (2) Dissolve 0.5 g of Pivmecillinam Hydrochloride in
QS
10 mL of water, and add 1 mL of dilute nitric acid and 1
WS: Amount [mg (potency)] of Pirarubicin Reference drop of silver nitrate TS: a white precipitate is formed.
Standard
Optical rotation [a]20
D : +200 – +2209(1 g calculated on
Internal standard solution—A solution of 2-naphthol in the the anhydrous basis, water, 100 mL, 100 mm).
mobile phase (1 in 1000).
Purity (1) Heavy metals—To 1.0 g of Pivmecillinam
Operating conditions—
Hydrochloride in a crucible add 10 mL of a solution of mag-
Detector: An ultraviolet absorption photometer
nesium nitrate hexahydrate in ethanol (95) (1 in 10), ˆre the
(wavelength: 254 nm).
ethanol to burn, and heat gradually to incinerate. If a car-
Column: A stainless steel column 6 mm in inside diameter
bonized substance remains, moisten with a small amount of
and 15 cm in length, packed with octadecylsilanized silica gel
nitric acid, and ignite to incinerate. Cool, add 3 mL of
for liquid chromatography (5 mm in particle diameter).
hydrochloric acid to the residue, dissolve by warming on a
Column temperature: A constant temperature of about
water bath, and heat to dryness. To the residue add 10 mL of
259C.
water, and dissolve by warming on a water bath. After cool-
Mobile phase: A mixture of 0.05 mol/L ammonium for-
ing, adjust the pH to 3 to 4 with ammonia TS, add 2 mL of
mate buŠer solution, pH 4.0 and acetonitrile (3:2).
dilute acetic acid, ˆlter if necessary, and wash the crucible
Flow rate: Adjust the ‰ow rate so that the retention time
and the ˆlter with 10 mL of water. Put the ˆltrate and the
of pirarubicin is about 7 minutes.
washings to a Nessler tube, add water to make 50 mL, and
System suitability—
use this solution as the test solution. Prepare the control so-
System performance: When the procedure is run with 20
lution in the same manner as the test solution with 2.0 mL of
mL of the standard solution under the above operating con-
Standard Lead Solution (not more than 20 ppm).
ditions, pirarubicin and the internal standard are eluted in
(2) Arsenic—Prepare the test solution with 1.0 g of Piv-
this order with the resolution between these peaks being not
mecillinam Hydrochloride according to Method 4, and per-
less than 9.
form the test (not more than 2 ppm).
System repeatability: When the test is repeated 6 times
(3) Related substances—Dissolve 0.05 g of Pivmecillin-
with 20 mL of the standard solution under the above operat-
am Hydrochloride in 4.0 mL of a mixture of acetonitrile and
ing conditions, the relative standard deviation of the ratios
acetic acid (100) (97:3), and use this solution as the sample
of the peak area of pirarubicin to that of the internal stan-
solution. Separately, dissolve 2.0 mg of Pivmecillinam
dard is not more than 1.0z.
Hydrochloride Reference Standard in 4.0 mL of water, and
Containers and storage Containers—Hermetic containers. use this solution as the standard solution. Perform the test
with these solutions as directed under the Thin-layer Chro-
matography. Spot 2 mL of the standard solution on a plate
Pivmecillinam Hydrochloride of silica gel for thin-layer chromatography, allow to stand
for 30 minutes, then spot 2 mL of the sample solution on the
塩酸ピブメシリナム plate. Immediately, develop the plate with a mixture of ace-
tone, water and acetic acid (100) (10:1:1) to a distance of
Change to read except the structural formula about 12 cm, and air-dry the plate. Allow the plate to stand
and chemical name: for 10 minutes in iodine vapor: the spot from the sample so-
lution appeared at the position corresponding to the spot ob-
Pivmecillinam Hydrochloride contains not less than tained from the standard solution is not larger and not more
630 mg (potency) per mg, calculated on the anhydrous intense than the spot from the standard solution, and any
basis. The potency of Pivmecillinam Hydrochloride spot other than the principal spot and the above spot is not
is expressed as mass (potency) of pivmecillinam observable.
(C21H33N3O5S: 439.57).
Water Not more than 1.0z (0.25 g, coulometric titration).
Description Pivmecillinam Hydrochloride occurs as a
Assay Weigh accurately an amount of Pivmecillinam
white to yellowish white crystalline powder.
Hydrochloride and Pivmecillinam Hydrochloride Reference
It is very soluble in methanol and in acetic acid (100), free-
Standard, equivalent to about 20 mg (potency), dissolve in a
ly soluble in water and in ethanol (99.5), and soluble in
suitable amount of the mobile phase, add exactly 10 mL of
acetonitrile.
the internal standard solution and the mobile phase to make
Identiˆcation (1) Determine the infrared absorption 100 mL, and use these solutions as the sample solution and
spectrum of Pivmecillinam Hydrochloride as directed in the the standard solution, respectively. Perform the test with 10
potassium bromide disk method under the Infrared Spec- mL each of the sample solution and the standard solution as
trophotometry, and compare the spectrum with the Refer- directed under the Liquid Chromatography according to the
ence Spectrum or the spectrum of Pivmecillinam Hydrochlo- following conditions, and determine the ratios, Q T and QS,
ride Reference Standard: both spectra exhibit similar intensi- of the peak area of pivmecillinam to that of the internal
ties of absorption at the same wave numbers. standard.
Supplement I, JP XIV O‹cial Monographs for Part I 1511

Amount [ mg (potency)] of pivmecillinam (C21H33N3O5S) tahydrate (1 in 100) while shaking: a purple color develops.
Q (2) Transfer 5 mg each of Polymixin B Sulfate and Poly-
= WS × T × 1000
QS mixin B Sulfate Reference Standard separately in two glass
stoppered test tubes, add 1 mL of diluted hydrochloric acid
WS: Amount [mg (potency)] of Pivmecillinam Hydro-
(1 in 2), stopper the tube, heat at 1359 C for 5 hours, then
chloride Reference Standard
heat to dryness on a water bath, and keep the heating until
Internal standard solution—A solution of diphenyl in the no more hydrochloric acid odor is evolved. Dissolve the
mobile phase (1 in 12,500). residue in 0.5 mL of water, and use these solutions as the
Operating conditions— sample solution and the standard solution (1). Separately,
Detector: An ultraviolet absorption photometer dissolve 20 mg each of L-leucine, L-threonine, phenylalanine
(wavelength: 254 nm). and L-serine separately in 10 mL of water, and use these so-
Column: A stainless steel column 4 mm in inside diameter lutions as the standard solution (2), the standard solution
and 30 cm in length, packed with octadecylsilanized silica gel (3), the standard solution (4) and the standard solution (5),
for liquid chromatography (10 mm in particle diameter). respectively. Perform the test with these solutions as directed
Column temperature: A constant temperature of about under the Thin-layer Chromatography. Spot 3 mL each of
259C. the sample solution, the standard solution (1), the standard
Mobile phase: Dissolve 0.771 g of ammonium acetate in solution (2), the standard solution (3), the standard solution
about 900 mL of water, adjust the pH to 3.5 with acetic acid (4) and the standard solution (5) on a plate of silica gel for
(100), and add water to make 1000 mL. To 400 mL of this thin-layer chromatography, and expose the plate to a satu-
solution add 600 mL of acetonitrile. rated vapor of the developing solvent for 15 hours. Develop
Flow rate: Adjust the ‰ow rate so that the retention time the plate with a mixture of phenol and water (3:1) to a dis-
of pivmecillinam is about 6.5 minutes. tance of about 13 cm while without exposure to light, and
System suitability— dry the plate at 1109 C for 5 minutes. Spray evenly nin-
System performance: When the procedure is run with 10 hydrin-acetic acid TS on the plate, and heat at 1109C for 5
mL of the standard solution under the above operating con- minutes: Rf value of each spot obtained from the sample so-
ditions, pivmecillinam and the internal standard are eluted lution is the same with Rf value of the corresponding spots
in this order with the resolution between these peaks being from the standard solution (1). Each of the spots from the
not less than 4. sample solution appears at the position corresponding to
System repeatability: When the test is repeated 6 times each of the spots from the standard (2), (3) and (4), but not
with 10 mL of the standard solution under the above operat- appears at the position corresponding to the spot from the
ing conditions, the relative standard deviation of the ratios standard solution (5).
of the peak area of pivmecillinam to that of the internal (3) A solution of Polymixin B Sulfate (1 in 20) responds
standard is not more than 1.0z. to the Qualitative Tests for sulfate.

Containers and storage Containers—Tight containers. Optical rotation [a]20D : -78 – -909(0.5 g calculated on
the dried basis, water, 25 mL, 100 mm).

pH The pH of a solution obtained by dissolving 1.0 g of


Polymixin B Sulfate Polymixin B Sulfate in 50 mL of water is between 5.0 and
7.0.
硫酸ポリミキシン B
Phenylalanine Weigh accurately about 0.375 g of Polymix-
Change to read except the structural formula in B Sulfate, dissolve in 0.1 mol/L hydrochloric acid TS to
and chemical name: make exactly 100 mL. Determine absorbances, A1, A2, A3,
A4 and A5, of this solution at 252 nm, at 258 nm, at 264 nm,
Polymixin B Sulfate contains not less than 6500 at 280 nm and at 300 nm, respectively, as directed under the
units per mg, calculated on the dried basis. The poten- Ultraviolet-visible Spectrophotometry, and calculate the
cy of Polymixin B Sulfate is expressed as mass unit of amount of phenylalanine by the following equation: the
polymixin B (C55-56H96-98N16O13). One unit of Poly- amount of phenylalanine calculated on the dried basis is not
mixin B Sulfate is equivalent to 0.129 mg of polymixin less than 9z and not more than 12z.
B sulfate (C55-56H96-98N16O13.1-2H2SO4).
Amount (z) of phenylalanine
Description Polymixin B Sulfate occurs as a white to yel- A -0.5A1+0.5A3-1.8A4+0.8A5
= 2 × 9.4787
low-brown powder. WT
It is freely soluble in water, and practically insoluble in
WT: Amount (g) of the sample, calculated on the dried
ethanol (99.5).
basis
Identiˆcation (1) To 5 mL of a solution of Polymixin B
Purity Heavy metals—Proceed with 1.0 g of Polymixin B
Sulfate (1 in 10) add 5 mL of a solution of sodium hydroxide
Sulfate according to Method 2, and perform the test. Pre-
(1 in 10), add 5 drops of a solution of copper (II) sulfate pen-
pare the control solution with 2.0 mL of Standard Lead
1512 O‹cial Monographs for Part I Supplement I, JP XIV

Solution (not more than 20 ppm). ed under the Ultraviolet-visible Spectrophotometry, and
compare the spectrum with the Reference Spectrum: both
Loss on drying Not more than 6.0z (1 g, in vacuum,
spectra exhibit similar intensities of absorption at the same
609C, 3 hours).
wavelengths.
Residue on ignition Not more than 0.75z (1 g).
Change the Optical rotation to read:
Assay Perform the test according to the Cylinder-plate
method as directed under the Microbial Assay for Antibiot- Optical rotation [a]20
D : +53 – +639(0.5 g calculated on
ics according to the following conditions. the anhydrous basis, water, 50 mL, 100 mm).
(1) Test organism—Escherichia coli NIHJ
(2) Agar media for seed and base layer Delete the pH.
Peptone 10.0 g
Meat extract 3.0 g Add the following next to Purity (2):
Sodium chloride 30.0 g
Purity
Agar 20.0 g
(3) Related substances—Dissolve 0.10 g of Potassium
Water 1000 mL
Clavulanate in 10 mL of the mobile phase A, and use this so-
Mix all the ingredients, and sterilize. Adjust the pH of the
lution as the sample solution. Pipet 1 mL of the sample solu-
solution so that it will be 6.5 to 6.6 after sterilization.
tion, add the mobile phase to make exactly 100 mL, and use
(3) Standard solutions—Weigh accurately an amount of
this solution as the standard solution. Perform the test with
Polymixin B Sulfate Reference Standard, equivalent to
exactly 20 mL each of the sample solution and the standard
about 200,000 units, dissolve in phosphate buŠer solution,
solution as directed under the Liquid Chromatography ac-
pH 6.0 to make exactly 20 mL, and use this solution as the
cording to the following conditions, and determine each
standard stock solution. Keep the standard stock solution at
peak area by the automatic integration method: the area of
not exceeding 59 C and use within 14 days. Take exactly a
each peak other than clavulanic acid from the sample solu-
suitable amount of the standard stock solution before use,
tion is not more than the peak area of clavulanic acid from
add phosphate buŠer solution, pH 6.0 to make solutions so
the standard solution, and the total area of the peaks other
that each mL contains 4000 units and 1000 units, and use
than clavulanic acid from the sample solution is not more
these solutions as the high concentration standard solution
than 2 times of the peak area of clavulanic acid from the
and the low concentration standard solution, respectively.
standard solution.
(4) Sample solutions—Weigh accurately an amount of
Operating conditions—
Polymixin B Sulfate, equivalent to about 200,000 units, and
Detector: An ultraviolet absorption photometer
dissolve in phosphate buŠer solution, pH 6.0 to make ex-
(wavelength: 230 nm).
actly 20 mL. Take exactly a suitable amount of this solution,
Column: A stainless steel column 4.6 mm in inside di-
add phosphate buŠer solution, pH 6.0 to make solutions so
ameter and 10 cm in length, packed with octadecylsilanized
that each mL contains 4000 units and 1000 units, and use
silica gel for liquid chromatography (5 mm in particle
these solutions as the high concentration sample solution
diameter).
and the low concentration sample solution, respectively.
Column temperature: A constant temperature of about
Containers and storage Containers—Tight containers. 409C.
Storage—Light-resistant. Mobile phase A: Adjust the pH of 0.05 mol/L sodium di-
hydrogen phosphate TS to 4.0 with phosphoric acid.
Mobile phase B: A mixture of the mobile phase A and
Potassium Clavulanate methanol (1:1).
Flowing of the mobile phase: Control the gradient by mix-
クラブラン酸カリウム ing the mobile phases A and B as directed in the following
table.
Change the origin/limits of content to read:
Time after injection Mobile phase Mobile phase
of sample (min) A (z) B (z)
Potassium Clavulanate contains not less than 810 mg
(potency) per mg, calculated on the anhydrous basis. 0– 4 100 0
The potency of Potassium Clavulanate is expressed as 4 – 15 100 → 0 0 → 100
mass (potency) of clavularic acid (C8H9NO5: 199.16). 15 – 25 0 100

Flow rate: 1.0 mL per minute.


Change the Identiˆcation (1) to read:
Time span of measurement: About 6 times as long as the
Identiˆcation (1) To 1 mL of a solution of Potassium retention time of clavulanic acid.
Clavulanate (1 in 50,000) add 5 mL of imidazole TS, and System suitability—
warm in a water bath at 309C for 12 minutes. After cooling, Test for required detectability: Pipet 1 mL of the standard
determine the absorption spectrum of this solution as direct- solution, and add the mobile phase A to make exactly 10
Supplement I, JP XIV O‹cial Monographs for Part I 1513

mL. Conˆrm that the peak area of clavulanic acid obtained that of the internal standard.
from 20 mL of this solution is equivalent to 7 to 13z of that QT
Amount (mg) of C23H30O6 = WS ×
from 20 mL of the standard solution. QS
System performance: Dissolve 10 mg each of Potassium
WS: Amount (mg) of Prednisolone Acetate Reference
Clavulanate and Amoxycillin Reference Standard in 100 mL
Standard
of the mobile phase A. When the procedure is run with 20
mL of this solution under the above operating conditions, Internal standard solution—A solution of butyl parahydrox-
clavulanic acid and amoxycillin are eluted in this order with ybenzoate in methanol (3 in 1000).
the resolution between these peaks being not less than 8 and Operating conditions—
the number of theoretical plates of the peak of clavulanic Detector: An ultraviolet absorption photometer
acid is not less than 2500 steps. (wavelength: 254 nm).
System repeatability: When the test is repeated 3 times Column: A stainless steel column 4.0 mm in inside di-
with 20 mL of the standard solution under the above operat- ameter and 15 cm in length, packed with octadecylsilanized
ing conditions, the relative standard deviation of the peak silica gel for liquid chromatography (5 mm in particle di-
area of clavulanic acid is not more than 2.0z. ameter).
Column temperature: A constant temperature of about
259C.
Povidone-Iodine Mobile phase: A mixture of water and acetonitrile (3:2).
Flow rate: Adjust the ‰ow rate so that the retention time
ポビドンヨード of prednisolone acetate is about 10 minutes.
System suitability—
Change the Description to read: System performance: When the procedure is run with 10
mL of the standard solution under the above operating con-
Description Povidone-Iodine occurs as a dark red-brown
ditions, prednisolone acetate and the internal standard are
powder. It has a faint, characteristic odor.
eluted in this order with the resolution between these peaks
It is freely soluble in water and in ethanol (99.5).
being not less than 10.
The pH of a solution obtained by dissolving 1.0 g of
System repeatability: When the test is repeated 6 times
Povidone-Iodine in 100 mL of water is between 1.5 and 3.5.
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios
Change the Purity (3) to read:
of the peak height of prednisolone acetate to that of the in-
Purity ternal standard is not more than 1.0z.
(3) Arsenic—Prepare the test solution with 1.0 g of
Povidone-Iodine according to Method 4, and perform the
test (not more than 2 ppm). Prednisolone Sodium Succinate
Change the Assay (2) to read: for Injection
Assay 注射用コハク酸プレドニゾロンナトリウム
(2) Nitrogen—Weigh accurately about 20 mg of
Povidone-Iodine, and perform the test as directed under the Change the Assay to read:
Nitrogen Determination.
Assay Take a quantity of sealed containers of Predniso-
lone Sodium Succinate for Injection, equivalent to about
0.10 g of prednisolone (C21H28O5), and dissolve the contents
Prednisolone Acetate in a suitable amount of diluted methanol (1 in 2), and trans-
酢酸プレドニゾロン fer to a 100-mL volumetric ‰ask. Wash each container with
diluted methanol (1 in 2), collect the washings in the volu-
Change the Assay to read: metric ‰ask, and add diluted methanol (1 in 2) to make
volume. Pipet 4 mL of this solution, add diluted methanol (1
Assay Dissolve about 10 mg each of Prednisolone Acetate in 2) to make exactly 50 mL. Pipet 5 mL of this solution, add
and Prednisolone Acetate Reference Standard, previously exactly 5 mL of the internal standard solution, mix, and use
dried and accurately weighed, in 60 mL each of methanol, this solution as the sample solution. Separately, weigh ac-
add exactly 2 mL each of the internal standard solution, then curately about 25 mg of Prednisolone Succinate Reference
add methanol to make 100 mL, and use these solutions as Standard, previously dried in a desiccator for 6 hours (in
the sample solution and the standard solution. Perform the vacuum, phosphorus (V) oxide, 609 C), dissolve in methanol
text with 10 mL each of the sample solution and the standard to make exactly 25 mL. Pipet 5 mL of this solution, add
solution as directed under the Liquid Chromatography ac- diluted methanol (1 in 2) to make exactly 50 mL. Pipet 5 mL
cording to the following conditions, and calculate the ratios, of this solution, add exactly 5 mL of the internal standard
Q T and QS, of the peak height of prednisolone acetate to solution, mix, and use this solution as the standard solution.
1514 O‹cial Monographs for Part I Supplement I, JP XIV

Perform the test with 10 mL of the sample solution and the the sample solution. Separately, dissolve 50 mg of 2-ethyl-2-
standard solution as directed under the Liquid Chro- phenylmalonediamide in pyridine to make exactly 100 mL.
matography according to the following conditions, and cal- Pipet 2 mL of this solution, add exactly 2 mL of the internal
culate the ratios, Q T and QS, of the peak area of predniso- standard solution, proceed in the same manner as Primi-
lone succinate to that of the internal standard. done, and use this solution as the standard solution. Per-
form the test with 2 mL of the sample solution and the stan-
Amount (mg) of prednisolone sodium succinate
dard solution as directed under the Gas Chromatography ac-
(C25H31NaO8)
cording to the following conditions, and calculate the ratios,
Q
= WS × T × 5 × 1.0477 Q T and Q S, of the peak area of 2-ethyl-2-phenylmalonedi-
QS
amide to that of the internal standard: Q T is not more than
Amount (mg) of prednisolone (C21H28O5)
Q S.
Q
= WS × T × 5 × 0.7827 Internal standard solution—A solution of stearylalcohol in
QS
pyridine (1 in 2000).
WS: Amount (mg) of Prednisolone Succinate Reference Operating conditions—
Standard Detector: A hydrogen ‰ame-ionization detector.
Column: A glass column 3 mm in inside diameter and 150
Internal standard solution—A solution of propyl para-
cm in length, packed with siliceous earth for gas chro-
hydroxybenzoate in diluted methanol (1 in 2) (1 in 25,000).
matography (125 to 150 mm in particle diameter) coated with
Operating conditions—
50z phenyl-methyl silicon polymer for gas chromatography
Detector: An ultraviolet absorption photometer
at the ratio of 3z.
(wavelength: 254 nm).
Column temperature: A constant temperature of about
Column: A stainless steel column 4.6 mm in inside di-
1959 C.
ameter and 25 cm in length, packed with octadecylsilanized
Carrier gas: Nitrogen
silica gel for liquid chromatography (5 mm in particle
Flow rate: Adjust the ‰ow rate so that the retention time
diameter).
of stearylalcohol is about 10 minutes.
Column temperature: A constant temperature of about
System suitability—
259C.
System performance: When the procedure is run with 2 mL
Mobile phase: Dissolve 0.32 g of tetra n-butylammonium
of the standard solution under the above operating condi-
bromide, 3.22 g of disodium hydrogen phosphate 12-water
tion, 2-ethyl-2-phenylmalonediamide and the internal stan-
and 6.94 g of potassium dihydrogen phosphate in 1000 mL
dard are eluted in this order with the resolution between
of water. To 840 mL of this solution add 1160 mL of
these peaks being not less than 3.
methanol.
System repeatability: When the test is repeated 5 times
Flow rate: Adjust the ‰ow rate so that the retention time
with 2 mL of the standard solution under the above operat-
of prednisolone succinate is about 15 minutes.
ing conditions, the relative standard deviation of the ratios
System suitability—
of the peak area of 2-ethyl-2-phenylmalonediamide to that
System performance: When the procedure is run with 10
of the internal standard is not more than 1.5z.
mL of the standard solution under the above operating con-
ditions, prednisolone succinate and the internal standard are
eluted in this order with the resolution between these peaks
being not less than 6. Procaine Hydrochloride Injection
System repeatability: When the test is repeated 6 times
塩酸プロカイン注射液
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios
Change the Assay to read:
of the peak area of prednisolone succinate to that of the in-
ternal standard is not more than 1.0z. Assay To an exactly measured volume of Procaine
Hydrochloride Injection, equivalent to about 20 mg of
procaine hydrochloride (C13H20N2O2.HCl), add the mobile
Primidone phase to make exactly 20 mL. Pipet 5 mL of this solution,
add exactly 5 mL of the internal standard solution and the
プリミドン mobile phase to make 20 mL, and use this solution as the
sample solution. Separately, weigh accurately about 50 mg
Change the Purity (3) to read: of procaine hydrochloride for assay, previously dried in a
desiccator (silica gel) for 4 hours, dissolve in the mobile
Purity
phase to make exactly 50 mL. Pipet 5 mL of this solution,
(3) 2-Ethyl-2-phenylmalonediamide—Dissolve 0.10 g of
add exactly 5 mL of the internal standard solution and the
Primidone in 2 mL of pyridine, add exactly 2 mL of the in-
mobile phase to make 20 mL, and use this solution as the
ternal standard solution, then add 1 mL of bis-trimethyl silyl
standard solution. Perform the test with 5 mL each of the
acetamide, shake well, and heat at 1009 C for 5 minutes.
sample solution and the standard solution as directed under
Cool, add pyridine to make 10 mL, and use this solution as
Supplement I, JP XIV O‹cial Monographs for Part I 1515

the Liquid Chromatography according to the following con- Change the Purity to read:
ditions, and calculate the ratios, Q T and QS, of the peak area
Purity (1) Heavy metals—Proceed with 1.0 g of Procar-
of procaine to that of the internal standard.
bazine Hydrochloride according to Method 4, and perform
Amount (mg) of procaine hydrochloride (C13H20N2O2.HCl) the test. Prepare the control solution with 2.0 mL of Stan-
Q dard Lead Solution (not more than 20 ppm).
= WS × T
QS (2) Related substances—Dissolve 50 mg of Procarbazine
Hydrochloride in 5.0 mL of a solution of L-cysteine
WS: Amount (mg) of procaine hydrochloride for assay
hydrochloride in diluted methanol (7 in 10) (1 in 200), and
Internal standard solution—A solution of caŠeine in the mo- use this solution as the sample solution. Pipet 1 mL of the
bile phase (1 in 1000). sample solution, add a solution of L-cysteine hydrochloride
Operating conditions— in diluted methanol (7 in 10) (1 in 200) to make exactly 50
Detector: An ultraviolet absorption photometer mL, and use this solution as the standard solution. Perform
(wavelength: 254 nm). the test with these solutions as directed under the Thin-layer
Column: A stainless steel column 6 mm in inside diameter Chromatography. Immerse slowly, by inclining, a plate of
and 15 cm in length, packed with octadecylsilanized silica gel silica gel with ‰uorescent indicator for thin-layer chro-
for liquid chromatography (5 mm in particle diameter). matography in a solution of L-cysteine hydrochloride in
Column temperature: A constant temperature of about diluted methanol (7 in 10) (1 in 200), allow to stand for 1
409C. minute, lift the plate from the solution, dry it in cold wind
Mobile phase: Adjust the pH of 0.05 mol/L potassium di- for 10 minutes, then dry in warm wind for 5 minutes, and
hydrogen phosphate TS to 3.0 with phosphoric acid, and then dry at 609 C for 5 minutes. After cooling, spot 5 mL
add an amount of sodium 1-pentane sulfonate to make a so- each of the sample solution and the standard solution on the
lution so that containing 0.1z. To 800 mL of this solution plate. Develop the plate with a mixture of methanol and
add 200 mL of methanol. ethyl acetate (1:1) to a distance of about 12 cm, and air-dry
Flow rate: Adjust the ‰ow rate so that the retention time the plate. Examine under ultraviolet light (main wavelength:
of procaine is about 10 minutes. 254 nm): not more than 1 spot other than the principal spot
System suitability— and the spot of the starting point from the sample solution
System performance: When the procedure is run with 5 mL appears, and is not more intense than the spot from the stan-
of the standard solution under the above operating condi- dard solution.
tions, procaine and the internal standard are eluted in this
order with the resolution between these peaks being not less
than 8. Pyrazinamide
System repeatability: When the test is repeated 6 times
with 5 mL of the standard solution under the above operat- ピラジナミド
ing conditions, the relative standard deviation of the ratios
of the peak area of procaine to that of the internal standard Change the origin/limits of content to read:
is not more than 1.0z.
Pyrazinamide, when dried, contains not less than
99.0z and not more than 101.0z of C5H5N3O.
Procarbazine Hydrochloride
Change the Description to read:
塩酸プロカルバジン
Description Pyrazinamide occurs as white crystals or crys-
talline powder.
Change the origin/limits of content to read:
It is sparingly soluble in water and in methanol, and
slightly soluble in ethanol (99.5).
Procarbazine Hydrochloride, when dried, contains
The pH of a solution obtained by dissolving 1.0 g of
not less than 98.5z and not more than 101.0z of
Pyrazinamide in 100 mL of water is between 5.0 and 7.0.
C12H19N3O.HCl.
Change the Identiˆcation to read:
Change the Description to read:
Identiˆcation (1) Determine the absorption spectrum of
Description Procarbazine Hydrochloride occurs as white
a solution of Pyrazinamide in 0.1 mol/L hydrochloric acid
to light yellowish white crystals or crystalline powder.
TS (1 in 100,000) as directed under the Ultraviolet-visible
It is freely soluble in water, and slightly soluble in ethanol
Spectrophotometry, and compare the spectrum with the
(99.5).
Reference Spectrum: both spectra exhibit similar intensities
It dissolves in dilute hydrochloric acid.
of absorption at the same wavelengths.
Melting point: about 2239 C (with decomposition).
(2) Determine the infrared absorption spectrum of
Pyrazinamide, previously dried, as directed in the potassium
1516 O‹cial Monographs for Part I Supplement I, JP XIV

bromide disk method under the Infrared Spectrophotomet- (2) Determine the infrared absorption spectrum of Pyr-
ry, and compare the spectrum with the Reference Spectrum: rolnitrin as directed in the potassium bromide disk method
both spectra exhibit similar intensities of absorption at the under the Infrared Spectrophotometry, and compare the
same wave numbers. spectrum with the Reference Spectrum or the spectrum of
Pyrrolnitrin Reference Standard: both spectra exhibit simi-
Change the Purity (4) and (5) to read: lar intensities of absorption at the same wave numbers.

Purity Melting point 124 – 1289


C
(4) Related substances—Dissolve 0.10 g of Pyrazina-
Purity Related substances—Dissolve 0.10 g of Pyrrolnitrin
mide in 10 mL of methanol, and use this solution as the sam-
in 10 mL of methanol, and use this solution as the sample so-
ple solution. Pipet 1 mL of the sample solution, add
lution. Pipet 1 mL of the sample solution, and add methanol
methanol to make exactly 200 mL, and use this solution as
to make exactly 100 mL. Pipet 3 mL of this solution, add
the standard solution. Perform the test with these solutions
methanol to make exactly 10 mL, and use this solution as the
as directed under the Thin-layer Chromatography. Spot 20
standard solution. Perform the test with these solutions as
mL each of the sample solution and the standard solution on
directed under the Thin-layer Chromatography. Spot 10 mL
a plate of silica gel with ‰uorescent indicator for thin-layer
each of the sample solution and the standard solution on a
chromatography. Develop the plate with a mixture of 1-
plate of silica gel for thin-layer chromatography. Develop
butanol, water and acetic acid (100) (3:1:1) to a distance of
the plate with a mixture of xylene, ethyl acetate and formic
about 10 cm, and air-dry the plate. Examine under ultravio-
acid (18:2:1) to a distance of about 10 cm, and dry the plate
let light (main wavelength: 254 nm): the spot other than the
at 809 C for 30 minutes. Spray evenly diluted sulfuric acid (1
principal spot obtained from the sample solution is not more
in 3) on the plate, and heat at 1009C for 30 minutes: the spot
intense than the spot from the standard solution.
other than the principal spot obtained from the sample solu-
(5) Readily carbonizable substances—Perform the test
tion is not more intense than the spot from the standard
with 0.20 g of Pyrazinamide: the solution has no more color
solution.
than Matching Fluid A.
Loss on drying Not more than 0.5z (1 g, reduced pressure
not exceeding 0.67 kPa, 609C, 3 hours).
Add the following:
Residue on ignition Not more than 0.10z (1 g).

Pyrrolnitrin Assay Conduct this procedure using light-resistant vessels.


Weigh accurately an amount of Pyrrolnitrin and Pyrrol-
ピロールニトリン nitrin Reference Standard, equivalent to about 50 mg
(potency), dissolve in diluted acetonitrile (3 in 5) to make ex-
actly 50 mL. Pipet 10 mL each of these solutions, add ex-
actly 10 mL of the internal standard solution, add diluted
acetonitrile (3 in 5) to make 100 mL, and use these solutions
as the sample solution and the standard solution. Perform
the test with 5 mL each of the sample solution and the stan-
C10H6Cl2N2O2: 257.07 dard solution as directed under the Liquid Chromatography
3-Chloro-4-(3-chloro-2-nitrophenyl)pyrrole [1018-71-9 ] according to the following conditions, and determine the ra-
tios, Q T and QS, of the peak area of pyrrolnitrin to that of
Pyrrolnitrin contains not less than 970 mg (potency) the internal standard.
per mg, calculated on the dried basis. The potency of Amount [ mg (potency)] of C10H6Cl2N2O2
Pyrrolnitrin is expressed as mass (potency) of pyrrol-
Q
nitrin (C10H6Cl2N2O2). = WS × T × 1000
QS
Description Pyrrolnitrin occurs as yellow to yellow-brown,
WS: Amount [mg (potency)] of Pyrrolnitrin Reference
crystals or crystalline powder.
Standard
It is freely soluble in methanol and in ethanol (95), and
practically insoluble in water. Internal standard solution—A solution of benzyl benzoate
in diluted acetonitrile (3 in 5) (3 in 500).
Identiˆcation (1) Determine the absorption spectrum of
Operating conditions—
a solution of Pyrrolnitrin in ethanol (95) (1 in 100,000) as
Detector: An ultraviolet absorption photometer
directed under the Ultraviolet-visible Spectrophotometry,
(wavelength: 254 nm).
and compare the spectrum with the Reference Spectrum or
Column: A stainless steel column 4 mm in inside diameter
the spectrum of a solution of Pyrrolnitrin Reference Stan-
and 15 cm in length, packed with octylsilanized silica gel for
dard prepared in the same manner as the sample solution:
liquid chromatography (5 mm in particle diameter).
both spectra exhibit similar intensities of absorption at the
Column temperature: A constant temperature of about
same wavelengths.
259C.
Supplement I, JP XIV O‹cial Monographs for Part I 1517

Mobile phase: A mixture of water and acetonitrile (11:9). the same wave numbers.
Flow rate: Adjust the ‰ow rate so that the retention time (3) A solution of Ranitidine Hydrochloride (1 in 50)
of pyrrolnitrin is about 9 minutes. responds to the Qualitative Tests for chloride.
System suitability—
pH The pH of a solution obtained by dissolving 1.0 g of
System performance: When the procedure is run with 5 mL
Ranitidine Hydrochloride in 100 mL of water is between 4.5
of the standard solution under the above operating condi-
and 6.0.
tions, pyrrolnitrin and the internal standard are eluted in this
order with the resolution between these peaks being not less Purity (1) Clarity and color of solution—A solution of
than 3. Ranitidine Hydrochloride (1 in 10) is clear and pale yellow to
System repeatability: When the test is repeated 6 times light yellow.
with 5 mL of the standard solution under the above operat- (2) Heavy metals—Proceed with 2.0 g of Ranitidine
ing conditions, the relative standard deviation of the ratios Hydrochloride according to Method 2, and perform the test.
of the peak area of pyrrolnitrin to that of the internal stan- Prepare the control solution with 2.0 mL of Standard Lead
dard is not more than 1.0z. Solution (not more than 10 ppm).
(3) Arsenic—Prepare the test solution with 1.0 g of
Containers and storage Containers—Tight containers.
Ranitidine Hydrochloride according to Method 4, and per-
Storage—Light-resistant.
form the test (not more than 2 ppm).
(4) Related substances—Conduct this procedure without
exposure to light, using light-resistant vessels. Dissolve 0.22
Add the following:
g of Ranitidine Hydrochloride in methanol to make exactly
10 mL, and use this solution as the sample solution. Pipet
Ranitidine Hydrochloride 0.5 mL of the sample solution, add methanol to make ex-
actly 100 mL, and use this solution as the standard solution
塩酸ラニチジン
(1). Pipet 6 mL, 4 mL, 2 mL and 1 mL of the standard solu-
tion (1), add to each methanol to make exactly 10 mL, and
use these solutions as the standard solution (2), the standard
solution (3), the standard solution (4) and the standard solu-
tion (5), respectively. Separately, dissolve 12.7 mg of
C13H22N4O3S.HCl: 350.86 ranitidinediamine in methanol to make exactly 10 mL, and
N{- 2-[({5-[(Dimethylamino)methyl]furan-2- use this solution as the standard solution (6). Perform the
yl}methyl)sulfanyl]ethyl}-N ?-methyl-2-nitroethene- test with these solutions as directed under the Thin-layer
1,1-diamine monohydrochloride [66357-59-3 ] Chromatography. Spot 10 mL each of the sample solution
and the standard solutions (1), (2), (3), (4) and (5) on a plate
Ranitidine Hydrochloride, when dried, contains not of silica gel for thin-layer chromatography. Separately, spot
less than 97.5z and not more than 102.0z of raniti- 10 mL of the sample solution on the plate, then spot 10 mL of
dine hydrochloride (C13H22N4O3S.HCl). the standard solution (6) on the spotted position of the sam-
Description Ranitidine Hydrochloride occurs as a white to ple solution. Immediately develop the plate with a mixture
pale yellow, crystalline or ˆne granular powder. of ethyl acetate, 2-propanol, ammonia solution (28) and
It is very soluble in water, freely soluble in methanol, and water (25:15:5:1) to a distance of about 15 cm, and air-dry
slightly soluble in ethanol (99.5). the plate. Allow the plate to stand in iodine vapor until the
It is hygroscopic. spot from the standard solution (5) appears: the spot ob-
It is gradually colored by light. tained from the standard solution (6) is completely separated
Melting point: about 1409 C (with decomposition). from the principal spot from the sample solution. The spot
having Rf value of about 0.7 from the sample solution is not
Identiˆcation (1) Determine the absorption spectrum of more intense than the spot from the standard solution (1),
a solution of Ranitidine Hydrochloride (1 in 100,000) as the spots other than the principal spot and the spot of Rf 0.7
directed under the Ultraviolet-visible Spectrophotometry, from the sample solution are not more intense than the spot
and compare the spectrum with the Reference Spectrum or from the standard solution (2), and the total amount of these
the spectrum of a solution of Ranitidine Hydrochloride related substances, calculated by comparison with the spots
Reference Standard prepared in the same manner as the from the standard solutions (1), (2), (3), (4) and (5), is not
sample solution: both spectra exhibit similar intensities of more than 1.0z.
absorption at the same wavelengths.
(2) Determine the infrared absorption spectrum of Loss on drying Not more than 0.75z (1 g, in vacuum,
Ranitidine Hydrochloride as directed in the paste method 609C, 3 hours).
under the Infrared Spectrophotometry, and compare the Residue on ignition Not more than 0.10z (1 g).
spectrum with the Reference Spectrum or the spectrum of
previously dried Ranitidine Hydrochloride Reference Stan- Assay Weigh accurately about 20 mg of Ranitidine
dard: both spectra exhibit similar intensities of absorption at Hydrochloride and Ranitidine Hydrochloride Reference
1518 O‹cial Monographs for Part I Supplement I, JP XIV

Standard, previously dried, dissolve each in the mobile reserpine peak from the sample solution is not larger than
phase to make exactly 200 mL, and use these solutions as the the peak area of reserpine from the standard solution.
sample solution and the standard solution. Perform the test Operating conditions—
with exactly 10 mL each of the sample solution and the stan- Detector, column, and column temperature: Proceed as
dard solution as directed under the Liquid Chromatography directed in the operating conditions in the Assay.
according to the following conditions, and determine the Mobile phase: A mixture of 0.05 mol/L potassium di-
peak areas, AT and AS, of ranitidine. hydrogen phosphate, pH 3.0 and acetonitrile (13:7).
Flow rate: Adjust the ‰ow rate so that the retention time
AT
Amount (mg) of C13H22N4O3S.HCl = WS × of reserpine is about 20 minutes.
AS
Time span of measurement: About twice as long as the
WS: Amount (mg) of Ranitidine Hydrochloride Reference retention time of reserpine.
Standard System suitability—
Test for required detection: To exactly 2 mL of the stan-
Operating conditions—
dard solution add acetonitorile to make exactly 50 mL. Con-
Detector: An ultraviolet absorption photometer
ˆrm that the peak area of reserpine obtained from 10 mL of
(wavelength: 322 nm).
this solution is equivalent to 3 to 5z of that of reserpine ob-
Column: A stainless steel column 4.6 mm in inside di-
tained from 10 mL of the standard solution.
ameter and 20 cm in length, packed with octadecylsilanized
System performance: Dissolve 0.01 g of Reserpine and 4
silica gel for liquid chromatography (10 mm in particle di-
mg of butyl parahydroxybenzoate in 100 mL of acetonitrile.
ameter).
To 5 mL of this solution add acetonitrile to make 50 mL.
Column temperature: A constant temperature of about
When the procedure is run with 20 mL of this solution ac-
259C.
cording to the operating conditions in the Assay, reserpine
Mobile phase: A mixture of methanol and diluted 0.5
and butyl parahydroxybenzoate are eluted in this order with
mol/L ammonium acetate TS (1 in 5) (17:3).
the resolution between these peaks being not less than 2.0.
Flow rate: Adjust the ‰ow rate so that the retention time
System repeatability: When the test is repeated 6 times
of ranitidine is about 5 minutes.
with 10 mL of the standard solution under the above operat-
System suitability—
ing conditions, the relative standard deviation of the peak
System performance: Dissolve 20 mg of Ranitidine
area of reserpine is not more than 2.0z.
Hydrochloride and 5 mg of benzalphthalide in 200 mL of
the mobile phase. When the procedure is run with 10 mL of
Change the Assay to read:
this solution under the above operating conditions, ben-
zalphthalide and ranitidine are eluted in this order with the Assay Conduct this procedure without exposure to day-
resolution between these peaks being not less than 2.0. light, using light-resistant vessels. Weigh accurately about 10
System repeatability: When the test is repeated 6 times mg each of Reserpine and Reserpine Reference Standard,
with 10 mL of the standard solution under the above operat- previously dried, and dissolve each in acetonitrile to make
ing conditions, the relative standard deviation of the peak exactly 100 mL. Pipet 5 mL each of these solutions, add ex-
area of ranitidine is not more than 1.0z. actly 10 mL of the internal standard solution, 5 mL of
acetonitrile and water to make 50 mL, and use these solu-
Containers and storage Containers—Tight containers.
tions as the sample solution and the standard solution, re-
Storage—Light-resistant.
spectively. Perform the test with 20 mL each of the sample
solution and the standard solution as directed under the Liq-
uid Chromatography according to the following conditions,
Reserpine and calculate the ratios, Q T and QS, of the peak area of
reserpine to that of the internal standard.
レセルピン
QT
Amount (mg) of C33H40N2O9 = WS ×
Change the Purity to read: QS
Purity Related substances—Conduct this procedure WS: Amount (mg) of Reserpine Reference Standard
without exposure to daylight, using light-resistant vessels.
Internal standard solution—A solution of butyl parahydrox-
Dissolve 50 mg of Reserpine in 50 mL of acetonitrile, and
ybenzoate in acetonitrile (1 in 50,000).
use this solution as the sample solution. Pipet 3 mL of the
Operating conditions—
sample solution, add acetonitrile to make exactly 100 mL,
Detector: An ultraviolet absorption photometer
and use this solution as the standard solution. Perform the
(wavelength: 268 nm).
test with exactly 10 mL each of the sample solution and the
Column: A stainless steel column 4 mm in inside diameter
standard solution as directed under the Liquid Chro-
and 25 cm in length, packed with octadecylsilanized silica gel
matography according to the following conditions. Deter-
for liquid chromatography (5 mm in particle diameter).
mine each peak area from these solutions by the automatic
Column temperature: A constant temperature of about
integration method: the total area of all peaks other than
409C.
Supplement I, JP XIV O‹cial Monographs for Part I 1519

Mobile phase: A mixture of 0.05 mol/L potassium di- add 0.1 mL of saturated potassium iodide TS under a cur-
hydrogen phosphate, pH 3.0 and acetonitrile (11:9). rent of Nitrogen. Immediately stopper tightly, and mix with
Flow rate: Adjust the ‰ow rate so that the retention time a swirling motion for 1 minute. Add 30 mL of water, stop-
of reserpine is about 10 minutes. per tightly, and shake vigorously for 5 to 10 seconds. Titrate
System suitability— this solution with 0.01 mol/L sodium thiosulfate VS until
System performance: When the procedure is run with 20 the blue color of the solution disappears after addition of 0.5
mL of the standard solution under the above operating con- mL of starch TS near the end point where the solution is a
ditions, reserpine and the internal standard are eluted in this pale yellow color. Calculate the amount of peroxide by the
order with the resolution between these peaks being not less following formula: not more than 10 meq/kg.
than 2.0.
V
System repeatability: When the test is repeated 6 times Amount (meq/kg) of peroxide = × 10
W
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios V: Volume (mL) of 0.01 mol/L sodium thiosulfate VS
of the peak area of reserpine to that of the internal standard consumed
is not more than 2.0z. W: Amount (g) of the sample
Assay Proceed as directed in Method 1-1 under the Vita-
min A Assay.
Retinol Acetate
Containers and storage Containers—Tight containers.
酢酸レチノール Storage—Light-resistant, and almost well-ˆlled, or under
Nitrogen atmosphere, and in a cold place.
Change to read except the structural formula
and chemical name:
Retinol Palmitate
Retinol Acetate is synthetic retinol acetate or syn-
thetic retinol acetate diluted with ˆxed oil. It contains パルミチン酸レチノール
not less than 2,500,000 Vitamin A Units per gram. A
suitable antioxidant may be added. Change to read except the structural formula
Retinol Acetate contains not less than 95.0z and and chemical name:
not more than 105.0z of the labeled Units.
Retinol Palmitate is a synthetic retinol palmitate or
Description Retinol Acetate occurs as pale yellow to yel-
a synthetic retinol palmitate diluted with ˆxed oil, and
low-red crystals or an ointment-like substance, and has a
contains not less than 1,500,000 Vitamin A Units in
faint, characteristic odor, but has no rancid odor.
each gram. It may contain a suitable antioxidant.
It is freely soluble in petroleum ether, soluble in ethanol
Retinol Palmitate contains not less than 95.0z and
(95), and practically insoluble in water.
not more than 105.0z of the labeled Units.
It is decomposed by air and by light.
Description Retinol Palmitate occurs as a light yellow to
Identiˆcation Dissolve Retinol Acetate and Retinol
yellow-red, ointment-like or an oily substance. It has a faint,
Acetate Reference Standard, equivalent to 15,000 Units, in 5
characteristic odor, but has no rancid odor.
mL of petroleum ether, and use these solutions as the sample
It is very soluble in petroleum ether, slightly soluble in
solution and the standard solution. Perform the test with
ethanol (95), and practically insoluble in water.
these solutions as directed under the Thin-layer Chro-
It is decomposed by air and by light.
matography. Spot 5 mL each of the sample solution and the
standard solution on a plate of silica gel for thin-layer chro- Identiˆcation Dissolve Retinol Palmitate and Retinol
matography. Develop with a mixture of cyclohexane and Palmitate Reference Standard, equivalent to 15,000 Units,
diethyl ether (12:1) to a distance of about 10 cm, and air-dry in 5 mL of petroleum ether, and use these solutions as the
the plate. Spray evenly antimony (III) chloride TS: the prin- sample solution and the standard solution. Perform the test
cipal spot obtained from the sample solution is the same in with these solutions as directed under the Thin-layer Chro-
color tone and Rf value with the blue spot from the standard matography. Spot 5 mL each of the sample solution and the
solution. standard solution on a plate of silica gel for thin-layer chro-
matography. Develop with a mixture of cyclohexane and
Purity (1) Acid value—Take exactly 5.0 g of Retinol
diethyl ether (12:1) to a distance of about 10 cm, and air-dry
Acetate, and perform the test: not more than 2.0.
the plate. Spray evenly antimony (III) chloride TS: the prin-
(2) Peroxide—Weigh accurately about 5 g of Retinol
cipal spot obtained from the sample solution is the same in
Acetate, transfer in a 250-mL glass-stoppered conical ‰ask,
color tone and Rf value with the blue spot from the standard
add 50 mL of a mixture of acetic acid (100) and isooctane
solution.
(3:2), and gently mix to dissolve completely. Replace the air
of the inside gradually with about 600 mL of Nitrogen, then Purity (1) Acid value—Take exactly 5.0 g of Retinol
1520 O‹cial Monographs for Part I Supplement I, JP XIV

Palmitate, and perform the test: not more than 2.0. and heat at 1009 C for 10 minutes: the principal spots ob-
(2) Peroxide—Weigh accurately about 5 g of Retinol tained from the sample solution and the standard solution
Palmitate, transfer in a 250-mL glass-stoppered conical show a purple-brown color and the same Rf value.
‰ask, add 50 mL of a mixture of acetic acid (100) and isooc- (3) To 2 mL of a solution of Ribostamycin Sulfate (1 in
tane (3:2), and gently mix to dissolve completely. Replace 5) add 1 drop of barium chloride TS: a white turbidity is pro-
the air of the inside gradually with about 600 mL of Nitro- duced.
gen, then add 0.1 mL of saturated potassium iodide TS
Optical rotation [a]20
D : +42 – +499(after drying, 0.25 g,
under a current of Nitrogen. Immediately stopper tightly,
water, 25 mL, 100 mm).
and mix with a swirling motion for 1 minute. Add 30 mL of
water, stopper tightly, and shake vigorously for 5 to 10 pH The pH of a solution obtained by dissolving 1.0 g of
seconds. Titrate this solution with 0.01 mol/L sodium Ribostamycin Sulfate in 20 mL of water is between 6.0 and
thiosulfate VS until the blue color of the solution disappears 8.0.
after addition of 0.5 mL of starch TS near the end point
Purity (1) Clarity and color of solution—Dissolve 1.0 g
where the solution is a pale yellow color. Calculate the
of Ribostamycin Sulfate in 5 mL of water: the solution is
amount of peroxide by the following formula: not more
clear, and colorless or pale yellow.
than 10 meq/kg.
(2) Heavy metals—Proceed with 1.0 g of Ribostamycin
V Sulfate according to Method 1, and perform the test. Pre-
Amount (meq/kg) of peroxide = × 10
W pare the control solution with 3.0 mL of Standard Lead So-
lution (not more than 30 ppm).
V: Volume (mL) of 0.01 mol/L sodium thiosulfate VS
(3) Arsenic—Prepare the test solution with 1.0 g of
W: Amount (g) of the sample
Ribostamycin Sulfate according to Method 1, and perform
Assay Proceed as directed in Method 1-1 under the Vita- the test (not more than 2 ppm).
min A Assay. (4) Related substances—Dissolve 0.12 g of Ribostamy-
cin Sulfate in water to make exactly 20 mL, and use this so-
Containers and storage Containers—Tight containers.
lution as the sample solution. Pipet 5 mL of the sample solu-
Storage—Light-resistant, and almost well-ˆlled, or under
tion, add water to make exactly 100 mL, and use this solu-
Nitrogen atmosphere, and in a cold place.
tion as the standard solution. Perform the test with these so-
lutions as directed under the Thin-layer Chromatography.
Spot 5 mL each of the sample solution and the standard solu-
Ribostamycin Sulfate tion on a plate of silica gel for thin-layer chromatography.
Develop the plate with a solution of potassium dihydrogen
硫酸リボスタマイシン
phosphate (3 in 40) to a distance of about 10 cm, and air-dry
the plate. Spray evenly 0.2z ninhydrin-water saturated 1-
Change to read except the structural formula
butanol TS on the plate, and heat at 1009C for 10 minutes:
and chemical name:
the spot other than the principal spot obtained from the
sample solution is not more intense than the spot from the
Ribostamycin Sulfate contains not less than 680 mg
standard solution.
(potency) per mg, calculated on the dried basis. The
potency of Ribostamycin Sulfate is expressed as mass Loss on drying Not more than 5.0z (0.5 g, reduced pres-
(potency) of ribostamycin (C17H34N4O10: 454.47). sure not exceeding 0.67 kPa, 609
C, 3 hours).
Description Ribostamycin Sulfate occurs as a white to yel- Residue on ignition Not more than 1.0z (1 g).
lowish white powder.
Assay Perform the test according to the Cylinder-plate
It is freely soluble in water, and practically insoluble in
method as directed under the Microbial Assay for Antibiot-
ethanol (95).
ics according to the following conditions.
Identiˆcation (1) Dissolve 20 mg of Ribostamycin Sul- (1) Test organism—Bacillus subtilis ATCC 6633
fate in 2 mL of phosphate buŠer solution, pH 6.0, add 1 mL (2) Culture medium—Use the medium i in 1) Medium
of ninhydrin TS, and boil: a blue-purple color develops. for test organism [5] under (1) Agar media for seed and base
(2) Dissolve 0.12 g each of Ribostamycin Sulfate and layer.
Ribostamycin Sulfate Reference Standard in 20 mL of (3) Standard solutions—Weigh accurately an amount of
water, and use these solutions as the sample solution and the Ribostamycin Sulfate Reference Standard, previously dried,
standard solution. Perform the test with these solutions as equivalent to about 20 mg (potency), dissolve in diluted
directed under the Thin-layer Chromatography. Spot 5 mL phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly 50
each of the sample solution and the standard solution on a mL, and use this solution as the standard stock solution.
plate of silica gel for thin-layer chromatography. Develop Keep the standard stock solution at 5 to 159C and use within
the plate with a solution of potassium dihydrogen phosphate 20 days. Take exactly a suitable amount of the standard
(3 in 40) to a distance of about 10 cm, and air-dry the plate. stock solution before use, add 0.1 mol/L phosphate buŠer
Spray evenly 0.2z ninhydrin-water saturated 1-butanol TS, solution, pH 8.0 to make solutions so that each mL contains
Supplement I, JP XIV O‹cial Monographs for Part I 1521

20 mg (potency) and 5 mg (potency), and use these solutions this solution as the sample stock solution. Pipet 5 mL of the
as the high concentration standard solution and the low con- sample stock solution, add citric acid-phosphate-acetonitrile
centration standard solution, respectively. TS to make exactly 50 mL, and use this solution as the sam-
(4) Sample solutions—Weigh accurately an amount of ple solution. Separately, pipet 1 mL of the sample stock so-
Ribostamycin Sulfate, equivalent to about 20 mg (potency), lution, and add acetonitrile to make exactly 100 mL. Pipet 5
and dissolve in water to make exactly 50 mL. Take exactly a mL of this solution, add citric acid-phosphate-acetonitrile
suitable amount of this solution, add 0.1 mol/L phosphate TS to make exactly 50 mL, and use this solution as the stan-
buŠer solution, pH 8.0 to make solutions so that each mL dard solution. Perform the test with exactly 50 mL each of
contains 20 mg (potency) and 5 mg (potency), and use these the sample solution and the standard solution as directed un-
solutions as the high concentration sample solution and the der the Liquid Chromatography according to the following
low concentration sample solution, respectively. conditions, and determine each peak area by the automatic
integration method: the area of the peak appeared at the rel-
Containers and storage Containers—Tight containers.
ative retention time of about 0.7 with respect to rifampicin
from the sample solution is not more than 1.5 times the peak
area of rifampicin from the standard solution, the area of
Rifampicin the peak other than rifampicin and the peak mentioned
above from the sample solution is not more than the peak
リファンピシン
area of rifampicin from the standard solution, and the total
area of the peaks other than rifampicin and the peak men-
Change to read except the structural formula
tioned above from the sample solution is not more than 3.5
and chemical name:
times the peak area of rifampicin from the standard solu-
tion.
Rifampicin contains not less than 970 mg (potency)
Operating conditions—
and not more than 1020 mg (potency) per mg, calculat-
Detector, column, column temperature, mobile phase,
ed on the dried basis. The potency of Rifampicin is ex-
and ‰ow rate: Proceed as directed in the operating condi-
pressed as mass (potency) of rifampicin (C43H58N4O12:
tions in the Assay.
822.94).
Time span of measurement: About 3 times as long as the
Description Rifampicin occurs as orange-red to red- retention time of rifampicin after the peak of the solvent.
brown, crystals or crystalline powder. System suitability—
It is slightly soluble in water, in acetonitrile, in methanol Test for required detectability: Measure exactly 2 mL of
and in ethanol (95). the standard solution, and add citric acid-phosphate-
acetonitrile TS to make exactly 20 mL. Conˆrm that the
Identiˆcation (1) To 5 mL of a solution of Rifampicin in
peak area of rifampicin obtained from 50 mL of this solution
methanol (1 in 5000) add 0.05 mol/L phosphate buŠer solu-
is equivalent to 7 to 13z of that from 50 mL of the standard
tion, pH 7.0 to make 100 mL. Determine the absorption
solution.
spectrum of this solution as directed under the Ultraviolet-
System performance: Proceed as directed in the system
visible Spectrophotometry, and compare the spectrum with
suitability in the Assay.
the Reference Spectrum or the spectrum of a solution of
System repeatability: When the test is repeated 6 times
Rifampicin Reference Standard prepared in the same man-
with 50 mL of the standard solution under the above operat-
ner as the sample solution: both spectra exhibit similar inten-
ing conditions, the relative standard deviation of the peak
sities of absorption at the same wavelengths.
area of rifampicin is not more than 2.0z.
(2) Determine the infrared absorption spectrum of
Rifampicin as directed in the potassium bromide disk Loss on drying Not more than 2.0z (1 g, reduced pressure
method under the Infrared Spectrophotometry, and com- not exceeding 0.69 kPa, 609C, 3 hours).
pare the spectrum with the Reference Spectrum or the spec-
Residue on ignition Not more than 0.10z (1 g).
trum of Rifampicin Reference Standard: both spectra
exhibit similar intensities of absorption at the same wave Assay Weigh accurately an amount of Rifampicin and
numbers. Rifampicin Reference Standard, equivalent to about 40 mg
(potency), and dissolve each in acetonitrile to make exactly
Purity (1) Heavy metals—Proceed with 1.0 g of Rifampi-
200 mL. Pipet 10 mL each of these solutions, add citric acid-
cin according to Method 2, and perform the test. Prepare the
phosphate-acetonitrile TS to make exactly 100 mL, and use
control solution with 2.0 mL of Standard Lead Solution (not
these solutions as the sample solution and the standard solu-
more than 20 ppm).
tion. Perform the test with 50 mL each of the sample solution
(2) Arsenic—Prepare the test solution with 1.0 g of
and the standard solution as directed under the Liquid Chro-
Rifampicin according to Method 3, and perform the test
matography according to the following conditions, and de-
(not more than 2 ppm).
termine the peak areas, AT and AS, of rifampicin.
(3) Related substances—Perform the test immediately
after preparing of the sample and standard solutions. Dis-
solve 0.10 g of Rifampicin in 50 mL of acetonitrile, and use
1522 O‹cial Monographs for Part I Supplement I, JP XIV

Amount [ mg (potency)] of C43H58N4O12 Scopolamine Butylbromide


A
= WS × T × 1000
AS 臭化ブチルスコポラミン
WS: Amount [mg (potency)] of Rifampicin Reference
Standard
Change the Purity (3) to read:
Purity
Operating conditions—
(3) Related substances—Dissolve 0.10 g of Scopolamine
Detector: An ultraviolet absorption photometer
Butylbromide in the mobile phase to make exactly 10 mL,
(wavelength: 254 nm).
and use this solution as the sample solution. Separately, dis-
Column: A stainless steel column 4.6 mm in inside di-
solve 10 mg of scopolamine hydrobromide in the mobile
ameter and 10 cm in length, packed with octylsilanized silica
phase to make exactly 100 mL. Pipet 10 mL of this solution,
gel for liquid chromatography (5 mm in particle diameter).
add the mobile phase to make exactly 50 mL, and use this so-
Column temperature: A constant temperature of about
lution as the standard solution (1). Pipet 5 mL of the stan-
259C.
dard solution (1), add the mobile phase to make exactly 10
Mobile phase: Dissolve 4.2 g of citric acid monohydrate
mL, and use this solution as the standard solution (2). Per-
and 1.4 g of sodium perchlorate in 1000 mL of a mixture of
form the test with exactly 20 mL each of the sample solution
water, acetonitrile and phosphate buŠer solution, pH 3.1
and the standard solutions (1) and (2) as directed under the
(11:7:2).
Liquid Chromatography according to the following condi-
Flow rate: Adjust the ‰ow rate so that the retention time
tions. Determine each peak area of these solutions by the au-
of rifampicin is about 8 minutes.
tomatic integration method: the peak area of scopolamine
System suitability—
from the sample solution is not larger than that from the
System performance: To 5 mL of a solution of Rifampicin
standard solution (2), and each area of the peaks other than
in acetonitrile (1 in 5000) add 1 mL of a solution of butyl
the peak appearing in the ˆrst elution and the peak of
parahydroxybenzoate in acetonitrile (1 in 5000) and citric
scopolamine and butylscopolamine from the sample solu-
acid-phosphate-acetonitrile TS to make 50 mL. When the
tion are not larger than the peak area from the standard so-
procedure is run with 50 mL of this solution under the above
lution (1).
operating conditions, butyl parahydroxybenzoate and
Operating conditions—
rifampicin are eluted in this order with the resolution be-
Detector: An ultraviolet absorption photometer
tween these peaks being not less than 1.5.
(wavelength: 210 nm).
System repeatability: When the test is repeated 5 times
Column: A stainless steel column 4.6 mm in inside di-
with 50 mL of the standard solution under the above operat-
ameter and 15 cm in length, packed with octylsilanized silica
ing conditions, the relative standard deviation of the peak
gel for liquid chromatography (10 mm in particle diameter).
area of rifampicin is not more than 1.0z.
Column temperature: A constant temperature of about
Containers and storage Containers—Tight containers. 309C.
Mobile phase: Dissolve 2 g of sodium lauryl sulfate in 370
mL of water and 680 mL of methanol, and adjust the pH to
Roxithromycin 3.6 with diluted phosphoric acid (1 in 10).
Flow rate: Adjust the ‰ow rate so that the retention time
ロキシスロマイシン of butylscopolamine is about 7 minutes.
Time span of measurement: About twice as long as the
Change the Description to read: retention time of butylscopolamine.
Description Roxithromycin occurs as a white crystalline System suitability—
powder. System performance: Dissolve 5 mg each of Scopolamine
It is freely soluble in ethanol (95) and in acetone, soluble Butylbromide and scopolamine hydrobromide in 50 mL of
in methanol, sparingly soluble in acetonitrile, and practically the mobile phase. When the procedure is run with 20 mL of
insoluble in water. this solution under the above operating conditions, scopola-
mine and butylscopolamine are eluted in this order with the
resolution between these peaks being not less than 5.
System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution (2) under the above
operating conditions, the relative standard deviation of the
peak area of scopolamine is not more than 2.0z.
Supplement I, JP XIV O‹cial Monographs for Part I 1523

Add the following: spots other than the principal spot obtained from the sample
solution is not more than three, and they are not more in-
Siccanin tense than the spot from the standard solution.

Loss on drying Not more than 0.5z (1 g, reduced pressure


シッカニン
not exceeding 0.67 kPa, 809C, 3 hours).

Residue on ignition Not more than 0.10z (1 g).

Assay Weigh accurately an amount of Siccanin and Sicca-


nin Reference Standard, equivalent to about 50 mg (poten-
cy), dissolve each in the internal standard solution to make
C22H30O3: 342.47
exactly 50 mL, and use these solutions as the sample solution
(4aS,6aS,11bR,13aS,13bS )-1,2,3,4,4a,5,6,6a,11b,13b-
and the standard solution. Perform the test with 10 mL each
Decahydro-4,4,6a,9-tetramethyl-13H-
of the sample solution and the standard solution as directed
benzo[a]furo[2,3,4-mn]xanthen-11-ol [22733-60-4 ]
under the Liquid Chromatography according to the follow-
ing conditions, and determine the ratios, Q T and QS, of the
Siccanin contains not less than 980 mg (potency) per
peak area of siccanin to that of the internal standard.
mg, calculated on the dried basis. The potency of
Q
Siccanin is expressed as mass (potency) of siccanin Amount [ mg (potency)] of C22H30O3 = WS × T × 1000
QS
(C22H30O3).
WS: Amount [mg (potency)] of Siccanin Reference Stand-
Description Siccanin occurs as white to light yellow, crys-
ard
tals or crystalline powder.
It is freely soluble in acetone, soluble in methanol and in Internal standard solution—A solution of 1,4-diphenylben-
ethanol (99.5), and practically insoluble in water. zene in methanol (1 in 30,000).
Operating conditions—
Identiˆcation (1) Determine the absorption spectrum of
Detector: An ultraviolet absorption photometer
a solution of Siccanin in ethanol (99.5) (1 in 10,000) as
(wavelength: 280 nm).
directed under the Ultraviolet-visible Spectrophotometry,
Column: A stainless steel column 4.6 mm in inside di-
and compare the spectrum with the Reference Spectrum or
ameter and 15 cm in length, packed with octadecylsilanized
the spectrum of a solution of Siccanin Reference Standard
silica gel for liquid chromatography (5 mm in particle di-
obtained in the same manner as the sample solution: both
ameter).
spectra exhibit similar intensities of absorption at the same
Column temperature: A constant temperature of about
wavelengths.
409C.
(2) Determine the infrared absorption spectrum of Sic-
Mobile phase: A mixture of methanol and phosphate
canin as directed in the potassium bromide disk method un-
buŠer solution, pH 5.9 (19:6).
der the Infrared Spectrophotometry, and compare the spec-
Flow rate: Adjust the ‰ow rate so that the retention time
trum with the Reference Spectrum or the spectrum of Sicca-
of siccanin is about 17 minutes.
nin Reference Standard: both spectra exhibit similar intensi-
System suitability—
ties of absorption at the same wave numbers.
System performance: When the procedure is run with 10
Optical rotation [a]20
D : -165 – -1759 (0.1 g, ethanol mL of the standard solution under the above operating con-
(99.5), 10 mL, 100 mm). ditions, siccanin and the internal standard are eluted in this
order with the resolution between these peaks being not less
Melting point 138 – 1429
C
than 3.0.
Purity (1) Heavy metals—Proceed with 1.0 g of Siccanin System repeatability: When the test is repeated 6 times
according to Method 4, and perform the test. Prepare the with 10 mL of the standard solution under the above operat-
control solution with 2.0 mL of Standard Lead Solution (not ing conditions, the relative standard deviation of the ratios
more than 20 ppm). of the peak area of siccanin to that of the internal standard is
(2) Related substances—Dissolve 0.20 g of Siccanin in not more than 1.0z.
10 mL of acetone, and use this solution as the sample solu-
Containers and storage Containers—Tight containers.
tion. Pipet 1 mL of the sample solution, add acetone to
make exactly 200 mL, and use this solution as the standard
solution. Perform the test with these solutions as directed
under the Thin-layer Chromatography. Spot 5 mL each of
the sample solution and the standard solution on a plate of
silica gel for thin-layer chromatography. Develop the plate
with a mixture of cyclohexane and acetone (5:1) to a distance
of about 10 cm, and air-dry the plate. Spray evenly 4-chlo-
robenzenediazonium TS on the plate: the number of the
1524 O‹cial Monographs for Part I Supplement I, JP XIV

Delete the following Monograph: Change to read:

Simˆbrate Sodium Chloride


シンフィブラート 塩化ナトリウム

NaCl: 58.44

Sisomicin Sulfate Sodium Chloride, when dried, contains not less than
99.0z and not more than 100.5z of NaCl.
硫酸シソマイシン
Description Sodium Chloride occurs as colorless or white,
crystals or crystalline powder.
Change the Purity (3) to read:
It is freely soluble in water, and practically insoluble in
Purity ethanol (99.5).
(3) Related substances—Dissolve 50 mg of Sisomicin
Identiˆcation (1) A solution of Sodium Chloride (1 in 20)
Sulfate, calculated on the dried basis, in water to make 10
responds to the Qualitative Tests for sodium salt.
mL, and use this solution as the sample solution. Pipet 0.5
(2) A solution of Sodium Chloride (1 in 20) responds to
mL, 1 mL and 1.5 mL of the sample solution, add water to
the Qualitative Tests for chloride.
each to make exactly 50 mL, and use these solutions as the
standard solution (1), the standard solution (2) and the stan- Purity (1) Clarity and color of solution—Dissolve 1.0 g
dard solution (3), respectively. Perform the test with these of Sodium Chloride in 5 mL of water: the solution is clear
solutions as directed under the Thin-layer Chromatography. and colorless.
Spot 10 mL each of the sample solution and the standard so- (2) Acidity or alkalinity—Dissolve 20.0 g of Sodium
lutions (1), (2), and (3) on a plate of silica gel for thin-layer Chloride in 100.0 mL of freshly boiled and cooled water,
chromatography. Develop the plate with a mixture of and use this solution as the sample solution. To 20 mL of the
methanol, chloroform, ammonia water (28) and acetone sample solution add 0.1 mL of bromothymol blue TS and
(2:2:1:1) to a distance of about 10 cm, and air-dry the plate. 0.5 mL of 0.01 mol/L hydrochloric acid VS: the color of the
Spray evenly 0.2z ninhydrin-water saturated 1-butabol TS solution is yellow. Separately, to 20 mL of the sample solu-
on the plate, and heat at 1009 C for 5 minutes. The spots cor- tion add 0.1 mL of bromothymol blue TS and 0.5 mL of
responding to Rf about 0.35 and Rf about 0.30 are not more 0.01 mol/L sodium hydroxide VS: the color of the solution
intense than that of the spot from the standard solution (3), is blue.
and the spot of gallamine corresponding to Rf about 0.25 is (3) Sulfates—To 7.5 mL of the sample solution obtained
not more intense than the spot from the standard solution in (2) add 30 mL of water, and use this solution as the sam-
(1). The total amount of the related substances is not more ple solution. Separately, dissolve 0.181 g of potassium sul-
than 6z. fate in diluted ethanol (99.5) (3 in 10) to make exactly 500
mL. Pipet 5 mL of this solution, and add diluted ethanol
(99.5) (3 in 10) to make exactly 100 mL. To 4.5 mL of this
Sodium Bicarbonate Injection solution add 3 mL of a solution of barium chloride dihy-
drate (1 in 4), shake, and allow to stand for 1 minutes. To
炭酸水素ナトリウム注射液 2.5 mL of this solution add 15 mL of the sample solution
and 0.5 mL of acetic acid (31), and allow to stand for 5
Add the following next to Identiˆcation: minutes: any turbidity produced does not more than that
pH 7.0 – 8.5 produced in the following control solution.
Control solution: Dissolve 0.181 g of potassium sulfate in
water to make exactly 500 mL. Pipet 5 mL of this solution,
Delete the Purity .
add water to make exactly 100 mL, and proceed in the same
manner as directed above using this solution instead of the
Change the Containers and storage to read:
sample solution.
Containers and storage Containers—Hermetic containers. (4) Phosphates—To 2.0 mL of the sample solution ob-
Plastic containers for aqueous injections may be used. tained in (2) add 5 mL of 2 mol/L sulfuric acid TS and water
to make 100.0 mL, then add 4 mL of ammonium molyb-
date-sulfuric acid TS and 0.1 mL of tin (II) chloride-
hydrochloric acid TS, and allow to stand for 10 minutes: the
color of the solution is not darker than the following control
solution.
Control solution: To 1.0 mL of Standard Phosphoric
Acid Solution add 12.5 mL of 2 mol/L sulfuric acid TS and
Supplement I, JP XIV O‹cial Monographs for Part I 1525

water to make exactly 250 mL. To 100 mL of this solution 0.1 mol/L zinc sulfate VS and 0.2 g of eriochrome black T-
add 4 mL of ammonium molybdate-sulfuric acid TS and 0.1 sodium chloride indicator, and warm to 409 C. Add 0.01
mL of tin (II) chloride-hydrochloric acid TS, and allow to mol/L disodium dihydrogen ethylenediamine tetraacetate
stand for 10 minutes. VS dropwise until the red-purple color of the solution
(5) Bromides—To 0.50 mL of the sample solution ob- changes to blue-purple. To this solution add a solution pre-
tained in (2) add 4.0 mL of water, 2.0 mL of dilute phenol pared by dissolving 10.0 g of Sodium Chloride in 100 mL of
red TS and 1.0 mL of a solution of sodium toluenesulfon- water, and add 2.5 mL of 0.01 mol/L disodium dihydrogen
chloramide trihydrate (1 in 10,000), and mix immediately. ethylenediamine tetraacetate VS: the color of the solution is
After allowing to stand for 2 minutes, add 0.15 mL of 0.1 a blue-purple.
mol/L sodium thiosulfate VS, mix, add water to make ex- (12) Arsenic—Prepare the test solution with 1.0 g of So-
actly 10 mL, and use this solution as the sample solution. dium Chloride according to Method 1, and perform the test
Separately, to 5.0 mL of a solution of potassium bromide (3 (not more than 2 ppm).
in 1,000,000) add 2.0 mL of dilute phenol red TS and 1.0 mL
Loss on drying Not more than 0.5z (1 g, 1059
C, 2 hours).
of a solution of sodium toluenesulfonchloramide trihydrate
(1 in 10,000), and mix immediately. Proceed in the same Assay Weigh accurately about 50 mg of Sodium Chloride,
manner as for the preparation of the sample solution, and previously dried, dissolve in 50 mL of water, and titrate with
use the solution so obtained as the standard solution. Per- 0.1 mol/L silver nitrate VS (potentiometric titration).
form the test with the sample solution and the standard solu-
Each mL of 0.1 mol/L silver nitrate VS
tion as directed under the Ultraviolet-visible Spectrophoto-
= 5.844 mg of NaCl
metry using water as the control: the absorbance at 590 nm
of the sample solution is not more than that of the standard Containers and storage Containers—Tight containers.
solution.
(6) Iodides—Wet 5 g of Sodium Chloride with drop-
wisely added 0.15 mL of a freshly prepared mixture of starch 10z Sodium Chloride Injection
TS, 0.5 mol/L sulfuric acid TS and sodium nitrite TS
(1000:40:3), allow to stand for 5 minutes, and examine un- 10 塩化ナトリウム注射液
der daylight: a blue color does not appear.
(7) Ferrocyanides—Dissolve 2.0 g of Sodium Chloride Change the Containers and storage to read:
in 6 mL of water, and add 0.5 mL of a mixture of a solution
Containers and storage Containers—Hermetic containers.
of iron (II) sulfate heptahydrate (1 in 100) and a solution of
Plastic containers for aqueous injections may be used.
ammonium iron (III) sulfate 12-water in diluted sulfuric acid
(1 in 400) (1 in 100) (19:1): a blue color does not develop wi-
thin 10 minutes.
Add the following:
(8) Heavy metals—Proceed with 5.0 g of Sodium Chlo-
ride according to Method 1, and perform the test. Prepare
the control solution with 1.5 mL of Standard Lead Solution
Sodium Fusidate
(not more than 3 ppm). フシジン酸ナトリウム
(9) Iron—To 10 mL of the sample solution obtained in
(2) add 2 mL of a solution of citric acid monohydrate (1 in 5)
and 0.1 mL of mercapto acetic acid, alkalize with ammonia
TS, add water to make 20 mL, and allow to stand for 5
minutes: the solution has not more color than the following
control solution.
Control solution: Pipet 1 mL of Standard Iron Solution,
and add water to make exactly 25 mL. To 10 mL of this so-
lution add 2 mL of a solution of citric acid monohydrate (1
in 5) and 0.1 mL of mercapto acetic acid, and proceed in the
same manner as directed for the sample solution. C31H47NaO6: 538.69
(10) Barium—To 5.0 mL of the sample solution ob- Monosodium (17Z)-ent-16a-acetoxy-3b,11b-dihydroxy-
tained in (2) add 5.0 mL of water and 2.0 mL of dilute sul- 4b,8b,14a-trimethyl-18-nor-5b,10a-cholesta-17(20),24-
furic acid, and allow to stand for 2 hours: the solution has dien-21-oate [751-94-0 ]
not more turbidity than the following control solution.
Control solution: To 5.0 mL of the sample solution ob- Sodium Fusidate contains not less than 935 mg
tained in (2) add 7.0 mL of water, and allow to stand for 2 (potency) per mg, calculated on the anhydrous basis.
hours. The potency of Sodium Fusidate is expressed as mass
(11) Magnesium and alkaline-earth materials—To 200 (potency) of fusidic acid (C31H48O6: 516.71).
mL of water add 0.1 g of hydroxylammonium chloride, 10 Description Sodium Fusidate occurs as white, crystals of
mL of ammonium chloride buŠer solution, pH 10, 1 mL of
1526 O‹cial Monographs for Part I Supplement I, JP XIV

crystalline powder. Delete the following Monographs:


It is freely soluble in water, in methanol and in ethanol
(99.5). Sodium Iopodate
Identiˆcation (1) Determine the infrared absorption
イオポダートナトリウム
spectra of Sodium Fusidate as directed in the potassium
bromide disk method under the Infrared Spectrophotomet-
ry, and compare the spectrum with the Reference Spectrum:
Sodium Iopodate Capsules
both spectra exhibit similar intensities of absorption at the イオポダートナトリウムカプセル
same wave numbers.
(2) Sodium Fusidate responds to the Qualitative Test (1)
for sodium salt.

Purity Heavy metals—Proceed with 2.0 g of Sodium Fusi- Add the following:
date according to Method 4, and perform the test. Prepare
the control solution with 2.0 mL of Standard Lead Solution Spectinomycin Hydrochloride
(not more than 10 ppm).
塩酸スペクチノマイシン
Water Not more than 2.0z (1 g, volumetric titration,
direct titration).

Assay Perform the test according to the Cylinder-plate


method as directed under the Microbial Assay for Antibiot-
ics according to the following conditions.
(1) Test organism—Staphylococcus aureus ATCC 6538
P C14H24N2O7.2HCl.5H2O: 495.35
(2) Culture medium—Use the medium ii in 3) Medium (2R,4aR,5aR,6S,7S,8R,9S,9aR,10aS )-
for other organisms under (1) Agar media for seed and base Decahydro-4a,7,9-trihydroxy-2-methyl-6,8-
layer. bis(methylamino)-4H-pyrano[2,3-b][1,4]benzodioxin-4-one
(3) Standard solutions—Weigh accurately an amount of dihydrochloride pentahydrate [22189-32-8 ]
Diethanolamine Fusidate Reference Standard, equivalent to
about 20 mg (potency), dissolve in 2 mL of ethanol (95), add Spectinomycin Hydrochloride contains not less than
water to make exactly 20 mL, and use this solution as the 603 mg (potency) per mg. The potency of Spectinomy-
standard stock solution. Keep the standard stock solution at cin Hydrochloride is expressed as mass (potency) of
a temperature not exceeding 59 C and use within 7 days. spectinomycin (C14H24N2O7: 332.35).
Take exactly a suitable amount of the standard stock solu- Description Spectinomycin Hydrochloride occurs as a
tion before use, add phosphate buŠer solution, pH 6.0 to white to light yellowish white crystalline powder.
make solutions so that each mL contains 4 mg (potency) and It is freely soluble in water, and practically insoluble in
1 mg (potency), and use these solutions as the high concentra- ethanol (95).
tion standard solution and the low concentration standard
solution, respectively. Identiˆcation (1) To 5 mL of a solution of Spectinomy-
(4) Sample solutions—Weigh accurately an amount of cin Hydrochloride (1 in 100) add gently anthrone TS: a blue
Sodium Fusidate, equivalent to about 20 mg (potency), and to blue-green color is produced at the zone of contact.
dissolve in water to make exactly 20 mL. Take exactly a suit- (2) Determine the infrared absorption spectra of Spec-
able amount of this solution, add phosphate buŠer solution, tinomycin Hydrochloride and Spectinomycin Hydrochloride
pH 6.0 to make solutions so that each mL contains 4 mg Reference Standard as directed in the paste method under
(potency) and 1 mg (potency), and use these solutions as the the Infrared Spectrophotometry, and compare these spectra:
high concentration sample solution and the low concentra- both spectra exhibit similar intensities of absorption at the
tion sample solution, respectively. same wave numbers.
(3) To 3 mL of a solution of Spectinomycin Hydrochlo-
Containers and storage Containers—Tight containers. ride (1 in 150) add 1 drop of silver nitrate TS: a white turbid-
Storage—Light-resistant, and at a temperature 2 to 89
C. ity is produced.

Optical rotation [a]20


D : +15 – +219(2.1 g calculated on
the anhydrous basis, water, 25 mL, 200 mm).

pH Dissolve 0.10 g of Spectinomycin Hydrochloride in 10


mL of water: the pH of the solution is between 4.0 and 5.6.

Water Not less than 16.0z and not more than 20.0z
(0.3 g, volumetric titration, direct titration).
Supplement I, JP XIV O‹cial Monographs for Part I 1527

Residue on ignition Not more than 1.0z (1 g). directed under the Thin-layer Chromatography. Spot 10 mL
each of the sample solution and the standard solution on a
Assay Perform the test according to the Cylinder-plate
plate of silica gel for thin-layer chromatography. Develop
method as directed under the Microbial Assay for Antibiot-
the plate with a solution of potassium dihydrogen phosphate
ics according to the following conditions.
(7 in 100) to a distance of about 12 cm, and air-dry the plate.
(1) Test organism—Klebsiella pneumoniae ATCC 10031
Spray evenly a mixture of a solution of 1,3-dihydrox-
(2) Culture medium—Use the medium i in 3) Medium
ynaphthalene in ethanol (95) (1 in 500) and diluted sulfuric
for other organisms under (1) Agar media for seed and base
acid (1 in 5) (1:1) on the plate, and heat at about 1509 C for
layer. Adjust the pH of the medium so that it will be 7.8 to
about 5 minutes: the principal spots from the sample solu-
8.0 after sterilization.
tion and the standard solution show the same in color tone
(3) Standard solutions—Weigh accurately an amount of
and Rf value.
Spectinomycin Hydrochloride Reference Standard, equiva-
(3) A solution of Streptomycin Sulfate (1 in 5) responds
lent to about 20 mg (potency), dissolve in 0.1 mol/L phos-
to the Qualitative Tests for sulfate.
phate buŠer solution, pH 8.0 to make exactly 20 mL, and
use this solution as the standard stock solution. Keep the Optical rotation [a]20D : -79 – -889(0.5 g calculated on
standard stock solution at a temperature not exceeding 59C the dried basis, water, 50 mL, 100 mm).
and use within 10 days. Take exactly a suitable amount of
pH The pH of a solution obtained by dissolving 2.0 g of
the standard stock solution before use, add 0.1 mol/L phos-
Streptomycin Sulfate in 10 mL of water is between 4.5 and
phate buŠer solution, pH 8.0 to make solutions so that each
7.0.
mL contains 200 mg (potency) and 50 mg (potency), and use
these solutions as the high concentration standard solution Purity (1) Clarity and color of solution—A solution ob-
and the low concentration standard solution, respectively. tained by dissolving 1.0 g of Streptomycin Sulfate in 5 mL of
(4) Sample solutions—Weigh accurately an amount of water is clear, and colorless or pale yellow.
Spectinomycin Hydrochloride, equivalent to about 20 mg (2) Heavy metals—Proceed with 2.0 g of Streptomycin
(potency), and dissolve in 0.1 mol/L phosphate buŠer solu- Sulfate according to Method 4, and perform the test. Pre-
tion, pH 8.0 to make exactly 20 mL. Take exactly a suitable pare the control solution with 2.0 mL of Standard Lead So-
amount of this solution, add 0.1 mol/L phosphate buŠer so- lution (not more than 10 ppm).
lution, pH 8.0 to make solutions so that each mL contains (3) Arsenic—Prepare the test solution with 2.0 g of
200 mg (potency) and 50 mg (potency), and use these solu- Streptomycin Sulfate according to Method 3 and perform
tions as the high concentration sample solution and the low the test (not more than 1 ppm).
concentration sample solution, respectively. (4) Related substances—Dissolve exactly 0.20 g of Strep-
tomycin Sulfate in a mixture of methanol and sulfuric acid
Containers and storage Containers—Tight containers.
(97:3) to make 5 mL, and heat under a re‰ux condenser for 1
hour. After cooling, wash the inside of the condenser with a
suitable amount of a mixture of methanol and sulfuric acid
Streptomycin Sulfate (97:3), add a mixture of methanol and sulfuric acid (97:3) to
make exactly 20 mL, and use this solution as the sample so-
硫酸ストレプトマイシン
lution. Separately, dissolve exactly 36 mg of D(+)-mannose
in a mixture of methanol and sulfuric acid (97:3) to make 5
Change to read except the structural formula
mL, and heat under a re‰ux condenser for 1 hour. After
and chemical name:
cooling, wash the inside of the condenser with a suitable
amount of a mixture of methanol and sulfuric acid (97:3),
Streptomycin Sulfate contains not less than 740 mg
and add a mixture of methanol and sulfuric acid (97:3) to
(potency) per mg, calculated on the dried basis. The
make exactly 50 mL. Pipet 5 mL of this solution, add a mix-
potency of Streptomycin Sulfate is expressed as mass
ture of methanol and sulfuric acid (97:3) to make exactly 50
(potency) of streptomycin (C21H39N7O12: 581.57).
mL, and use this solution as the standard solution. Perform
Description Streptomycin Sulfate occurs as a white to light the test with these solutions as directed under the Thin-layer
yellowish white powder. Chromatography. Spot 10 mL each of the sample solution
It is freely soluble in water, and very slightly soluble in and the standard solution on a plate of silica gel for thin-lay-
ethanol (95). er chromatography, develop with a mixture of toluene,
methanol and acetic acid (100) (2:1:1) to a distance of 13 to
Identiˆcation (1) Dissolve 50 mg of Streptomycin Sulfate
15 cm, and air-dry the plate. Spray evenly a mixture of a so-
in 5 mL of water, add 1 mL of ninhydrin TS and 0.5 mL of
lution of 1,3-dihydroxynaphthalene in ethanol (95) (1 in 500)
pyridine, and heat for 10 minutes: a purple color is devel-
and diluted sulfuric acid (1 in 5) (1:1) on the plate, and heat
oped.
at 1109 C for 5 minutes: the spot from the sample solution
(2) Dissolve 10 mg each of Streptomycin Sulfate and
corresponding to the spot from the standard solution is not
Streptomycin Sulfate Reference Standard in 10 mL of water,
more intense than the spot from the standard solution.
and use these solutions as the sample solution and the stan-
dard solution. Perform the test with these solutions as Loss on drying Not more than 5.0z (0.5 g, reduced pres-
1528 O‹cial Monographs for Part I Supplement I, JP XIV

sure not exceeding 0.67 kPa, 609


C, 3 hours). tion at the same wave numbers.
(2) Sulbenicillin Sodium responds to the Qualitative Test
Residue on ignition Not more than 1.0z (1 g).
(1) for sodium salt.
Assay Perform the test according to the Cylinder-plate
Optical rotation [a]20
D : +167 – +1829(1 g calculated on
method as directed under the Microbial Assay for Antibiot-
the anhydrous basis, water, 20 mL, 100 mm).
ics according to the following conditions.
(1) Test organism—Bacillus subtilis ATCC 6633 pH The pH of a solution obtained by dissolving 0.20 g of
(2) Culture medium—Use the medium i in 1) Medium Sulbenicillin Sodium in 10 mL of water is between 4.5 and
for test organism [5] under (1) Agar media for seed and base 7.0.
layer, having pH 7.8 – 8.0 after sterilization.
Purity (1) Clarity and color of solution—Dissolve 2.5 g
(3) Standard solutions—Weigh accurately an amount of
of Sulbenicillin Sodium in 5 mL of water: the solution is
Streptomycin Sulfate Reference Standard, previously dried,
clear and colorless to pale yellow.
equivalent to about 20 mg (potency), dissolve in diluted
(2) Heavy metals—Proceed with 1.0 g of Sulbenicillin
phosphate buŠer solution, pH 6.0 (1 in 2) to make exactly 50
Sodium according to Method 1, and perform the test. Pre-
mL, and use this solution as the standard stock solution.
pare the control solution with 2.0 mL of Standard Lead So-
Keep the standard stock solution between 59 C and 159 C,
lution (not more than 20 ppm).
and use within 30 days. Take exactly a suitable amount of
(3) Arsenic—Prepare the test solution with 1.0 g of Sul-
the standard stock solution before use, add 0.1 mol/L phos-
benicillin Sodium according to Method 1, and perform the
phate buŠer solution, pH 8.0 to make solutions so that each
test (not more than 2 ppm).
mL contains 8 mg (potency) and 2 mg (potency), and use
(4) Related substances—Dissolve 0.10 g of Sulbenicillin
these solutions as the high concentration standard solution
Sodium in 15 mL of the mobile phase, and use this solution
and the low concentration standard solution, respectively.
as the sample solution. Perform the test with 10 mL of the
(4) Sample solutions—Weigh accurately an amount of
sample solution as directed under the Liquid Chro-
Streptomycin Sulfate, equivalent to about 20 mg (potency),
matography according to the following conditions, deter-
dissolve in water to make exactly 50 mL. Take exactly a suit-
mine each peak area by the automatic integration method,
able amount of this solution, add 0.1 mol/L phosphate
and calculate the amount of these peaks by the area percen-
buŠer solution, pH 8.0 to make solutions so that each mL
tage method: the amount of the each peak other than the
contains 8 mg (potency) and 2 mg (potency), and use these so-
two peaks of sulbenicillin is not more than 2.0z, and the
lutions as the high concentration sample solution and the
total amount of the peaks other than the two peaks of sul-
low concentration sample solution, respectively.
benicillin is not more than 5.0z.
Containers and storage Containers—Tight containers. Operating conditions—
Detector: An ultraviolet absorption photometer
(wavelength: 254 nm).
Sulbenicillin Sodium Column: A stainless steel column 3.9 mm in inside di-
ameter and 30 cm in length, packed with octadecylsilanized
スルベニシリンナトリウム silica gel for liquid chromatography (5 mm in particle di-
ameter).
Change to read except the structural formula Column temperature: A constant temperature of about
and chemical name: 259C.
Mobile phase: Dissolve 10 g of potassium dihydrogen
Sulbenicillin Sodium contains not less than 814 mg phosphate in 750 mL of water, adjust the pH to 6.0 ± 0.1
(potency) per mg, calculated on the anhydrous basis. with sodium hydroxide TS, and add water to make 1000 mL.
The potency of Sulbenicillin Sodium is expressed as To 940 mL of this solution add 60 mL of acetonitrile.
mass (potency) of sulbenicillin (C16H18N2O7S2: Flow rate: Adjust the ‰ow rate so that the retention time
414.45). of the lately eluted peak of sulbenicillin is about 18 minutes.
Time span of measurement: About 1.5 times as long as the
Description Sulbenicillin Sodium occurs as white to light
retention time of the lately eluted peak of sulbenicillin after
yellowish white powder.
the solvent peak.
It is very soluble in water, freely soluble in methanol, and
System suitability—
slightly soluble in ethanol (99.5).
Test for required detectability: Measure exactly 1 mL of
It is hygroscopic.
the sample solution, add the mobile phase to make exactly
Identiˆcation (1) Determine the infrared absorption 100 mL, and use this solution as the solution for system
spectrum of Sulbenicillin Sodium as directed in the potassi- suitability test. Pipet 1 mL of the solution for system
um bromide disk method under the Infrared Spectrophoto- suitability test, and add the mobile phase to make exactly 10
metry, and compare the spectrum with the Reference Spec- mL. Conˆrm that the total area of the two peaks of sul-
trum or the spectrum of Sulbenicillin Sodium Reference benicillin obtained from 10 mL of this solution is equivalent
Standard: both spectra exhibit similar intensities of absorp- to 7 to 13z of that from 10 mL of the solution for system
Supplement I, JP XIV O‹cial Monographs for Part I 1529

suitability test. Talampicillin Hydrochloride


System performance: When the procedure is run with 10
mL of the sample solution under the above operating condi- 塩酸タランピシリン
tions, the resolution between the two peaks of sulbenicillin is
not less than 2.0. Change to read except the structural formula
System repeatability: When the test is repeated 6 times and chemical name:
with 10 mL of the solution for system suitability test under
the above operating conditions, the relative standard devia- Talampicillin Hydrochloride is the hydrochloride of
tion of the total areas of the two peaks of sulbenicillin is not ampicillin phthalidyl ester.
more than 5.0z. It contains not less than 600 mg (potency) per mg,
Water Not more than 6.0z (0.5 g, volumetric titration,
calculated on the anhydrous basis. The potency of
direct titration).
Talampicillin Hydrochloride is expressed as mass
(potency) of ampicillin (C16H19N3O4S: 349.40).
Assay Perform the test according to the Cylinder-plate
Description Talampicillin Hydrochloride occurs as a white
method as directed under the Microbial Assay for Antibiot-
to light yellowish white powder.
ics according to the following conditions.
It is very soluble in methanol, and freely soluble in water
(1) Test organism—Bacillus subtilis ATCC 6633
and in ethanol (99.5).
(2) Culture medium—Use the medium i in 1) Medium
for test organism [5] under (1) Agar media for seed and base Identiˆcation (1) To 1 mL of a solution of Talampicillin
layer. Adjust the pH of the medium so that it will be 6.4 to Hydrochloride (1 in 30) add 1 mL of sodium hydroxide TS,
6.6 after sterilization. mix, allow to stand for 5 minutes, and add 2 mL of dilute
(3) Standard solutions—Weigh accurately an amount of sulfuric acid and 2 to 3 drops of 2,4-dinitrophenylhydrazine
Sulbenicillin Sodium Reference Standard, equivalent to TS: an orange-yellow precipitate is formed.
about 50 mg (potency), dissolve in phosphate buŠer solu- (2) Determine the infrared absorption spectrum of
tion, pH 6.0 to make exactly 50 mL, and use this solution as Talampicillin Hydrochloride as directed in the potassium
the standard stock solution. Keep the standard stock solu- bromide disk method under the Infrared Spectrophotomet-
tion in a freezer, and use within 4 days. Take exactly a suita- ry, and compare the spectrum with the Reference Spectrum
ble amount of the standard stock solution before use, add or the spectrum of Talampicillin Hydrochloride Reference
phosphate buŠer solution, pH 6.0 to make solutions so that Standard: both spectra exhibit similar intensities of absorp-
each mL contains 40 mg (potency) and 10 mg (potency), and tion at the same wave numbers.
use these solutions as the high concentration standard solu- (3) To 10 mL of a solution of Talampicillin Hydrochlo-
tion and the low concentration standard solution, respec- ride (1 in 300) add 1 mL of dilute nitric acid, and add silver
tively. nitrate TS: a white precipitate is formed.
(4) Sample solutions—Weigh accurately an amount of
Optical rotation [a]20
D : +151 – +1719(0.2 g calculated on
Sulbenicillin Sodium, equivalent to about 50 mg (potency),
the anhydrous basis, ethanol (99.5), 20 mL, 100 mm).
and dissolve in phosphate buŠer solution, pH 6.0 to make
exactly 50 mL. Take exactly a suitable amount of this solu- Purity (1) Heavy metals—Proceed with 1.0 g of Talam-
tion, add phosphate buŠer solution, pH 6.0 to make solu- picillin Hydrochloride according to Method 2, and perform
tions so that each mL contains 40 mg (potency) and 10 mg the test. Prepare the control solution with 2.0 mL of Stan-
(potency), and use these solutions as the high concentration dard Lead Solution (not more than 20 ppm).
sample solution and the low concentration sample solution, (2) Arsenic—Prepare the test solution with 1.0 g of
respectively. Talampicillin Hydrochloride according to Method 4, and
perform the test (not more than 2 ppm).
Containers and storage Containers—Hermetic containers.
(3) Related substances—Dissolve 50 mg of Talampicillin
Hydrochloride in ethanol (99.5) to make exactly 10 mL, and
use this solution as the sample solution. Pipet 1 mL, 2 mL
and 3 mL of the sample solution, add ethanol (99.5) to each
to make exactly 100 mL, and use these solutions as the stan-
dard solution (1), the standard solution (2) and the standard
solution (3), respectively. Perform the test with these solu-
tions as directed under the Thin-layer Chromatography.
Spot 10 mL each of the sample solution and the standard so-
lutions (1), (2) and (3) on a plate of silica gel for thin-layer
chromatography. Develop the plate with a mixture of tetra-
hydrofuran, ethyl acetate, water and ethanol (95) (4:4:2:1)
to a distance of about 13 cm, and air-dry the plate. Spray
evenly a solution of ninhydrin in ethanol (99.5) (1 in 500) on
1530 O‹cial Monographs for Part I Supplement I, JP XIV

the plate, and heat at 1109 C for 5 minutes: the spots other Tegafur
than the principal spot from the sample solution is not more
intense than the spot from the standard solution (3), and the テガフール
total of the amount of each spot other than the principal
spot from the sample solution, which is calculated by the Change the Description to read:
comparison with the spots obtained from the standard solu-
Description Tegafur occurs as a white, crystalline powder.
tions (1), (2) and (3), is not more than 5z.
It is soluble in methanol and in acetone, and sparingly
(4) 2-Formylbenzoic acid—Dissolve 50 mg of Talam-
soluble in water and in ethanol (95).
picillin Hydrochloride in ethanol (99.5) to make exactly 10
It dissolves in dilute sodium hydroxide TS.
mL, and use this solution as the sample solution. Separately,
A solution of Tegafur in methanol (1 in 50) shows no opti-
dissolve 10 mg of 2-formylbenzoic acid in ethanol (99.5) to
cal rotation.
make exactly 100 mL. Pipet 5 mL of this solution, add
ethanol (99.5) to make exactly 10 mL, and use this solution
Change the Purity (4) to read:
as the standard solution. Perform the test with these solu-
tions as directed under the Thin-layer chromatography. Spot Purity
10 mL each of the sample solution and the standard solution (4) Arsenic—Prepare the test solution in a platinum
on a plate of silica gel for thin-layer chromatography, de- crucible with 1.0 g of Tegafur according to Method 4, in-
velop the plate with a mixture of chloroform and acetic acid cinerating by ignition between 7509C and 8509C, and per-
(100) (4:1) to a distance of about 13 cm, and air-dry the form the test (not more than 2 ppm).
plate. Spray evenly a solution of 2,4-dinitrophenylhydrazine
in diluted sulfuric acid (6 in 25) (1 in 500): the spot of 2-for-
mylbenzoic acid obtained from the sample solution is not Teicoplanin
more intense than that obtained from the standard solution.
テイコプラニン
Water Not more than 3.0z (0.5 g, volumetric titration,
direct titration).
Change the origin/limits of content to read:
Assay Weigh accurately an amount of Talampicillin
Hydrochloride and Talampicillin Hydrochloride Reference Teicoplanin contains not less than 900 mg (potency)
Standard, equivalent to about 20 mg (potency), dissolve in per mg, calculated on the anhydrous, de-sodium chlo-
water to make exactly 20 mL each, and use these solutions as ride and de-residual solvents basis. The potency of
the sample solution and the standard solution. The standard Teicoplanin is expressed as mass (potency) of teicopla-
solution should be prepared before use. Pipet 2 mL each of nin (C72-89H68-99Cl2N8-9O28-33).
the sample solution and the standard solution in separate
100-mL glass-stoppered ‰asks, add 2.0 mL of sodium Change the Content ratio of the active principle
hydroxide TS, and allow them to stand for exactly 15 to read:
minutes. Add 2.0 mL of diluted hydrochloric acid (1 in 10) Content ratio of the active principle Dissolve about 20 mg
and exactly 10 mL of 0.005 mol/L iodine VS, allow them to of Teicoplanin in water to make 10 mL, and use this solution
stand for exactly 15 minutes, and titrate with 0.01 mol/L so- as the sample solution. Perform the test with 20 mL of the
dium thiosulfate VS until the color of the solution is disap- sample solution as directed under the Liquid Chro-
peared. If necessary, add 0.2 to 0.5 mL of starch TS. matography according to the following conditions, and cal-
Separately, pipet 2 mL each of the sample solution and the culate the sum of peak areas of teicoplanin A2 group, Sa, the
standard solution in separate 100-mL glass-stoppered ‰asks, sum of peak areas of teicoplanin A3 group, Sb, and the sum
add exactly 10 mL of 0.005 mol/L iodine VS, titrate with of peak areas of other contents, Sc from the sample solution
0.01 mol/L sodium thiosulfate VS until the color of the so- by the automatic integration method. Calculate the content
lution is disappeared, and make any necessary correction. ratio of them by the formula given below: teicoplanin A2
For this titration, add 0.2 to 0.5 mL of starch TS, if necessa- group, teicoplanin A3 group, and the other are not less than
ry. Calculate the amount (mL) of 0.005 mol/L iodine VS, VT 80.0z, not more than 15.0z and not more than 5.0z, re-
and VS, consumed by the sample solution and the standard spectively.
solution, respectively. The elution order of each content and the relative reten-
Amount [ mg (potency)] of ampicillin (C16H19N3O4S) tion time of each content to the retention time of teicoplanin
V A2-2 are shown in the following table.
= WS × T × 1000
VS

WS: Amount [mg (potency)] of Talampicillin Hydrochlo-


ride Reference Standard

Containers and storage Containers—Tight containers.


Supplement I, JP XIV O‹cial Monographs for Part I 1531

System performance: When the procedure is run with 20


Elution Relative mL of the sample solution under the above operating condi-
Name of content
order retention time
tions, the symmetry coe‹cient of the peak of teicoplanin
teicoplanin A3 group ≦0.42 A3-1 is not more than 2.2.
teicoplanin A3-1 1 0.29
teicoplanin A2 group 0.42<,≦1.25 Change the Bacterial endotoxins to read:
teicoplanin A2-1 2 0.91
Bacterial endotoxins Less than 0.75 EU/mg (potency).
teicoplanin A2-2 3 1.00
teicoplanin A2-3 4 1.04
teicoplanin A2-4 5 1.17
teicoplanin A2-5 6 1.20 Theophylline
others 1.25<
テオフィリン

Content ratio (z) of teicoplanin A2 group


Change the Description to read:
Sa
= × 100
Sa + 0.83Sb + Sc Description Theophylline occurs as white crystals or crys-
talline powder.
Content ratio (z) of teicoplanin A3 group
It is soluble in N,N-dimethylformamide, and slightly solu-
0.83Sb
= × 100 ble in water and in ethanol (99.5).
Sa + 0.83Sb + Sc
It dissolves in 0.1 mol/L hydrochloric acid TS.
Content ratio (z) of others
Sc Change the Identiˆcation to read:
= × 100
Sa + 0.83Sb + Sc
Identiˆcation (1) Determine the absorption spectrum of
Operating conditions— a solution of Theophylline in 0.1 mol/L hydrochloric acid
Detector: An ultraviolet absorption photometer (wave- TS (1 in 200,000) as directed under the Ultraviolet-visible
length: 254 nm). Spectrophotometry, and compare the spectrum with the
Column: A stainless steel column 4.6 mm in inside di- Reference Spectrum: both spectra exhibit similar intensities
ameter and 25 cm in length, packed with octadecylsilanized of absorption at the same wavelengths.
silica gel for liquid chromatography (5 mm in particle di- (2) Determine the infrared absorption spectrum of The-
ameter). ophylline, previously dried, as directed in the potassium
Column temperature: A constant temperature of about bromide disk method under the Infrared Spectrophotomet-
259C. ry, and compare the spectrum with the Reference Spectrum:
Mobile phase A: Dissolve 7.80 g of sodium dihydrogen both spectra exhibit similar intensities of absorption at the
phosphate dihydrate in 1650 mL of water, add 300 mL of same wave numbers.
acetonitrile, adjust pH to 6.0 with sodium hydroxide TS,
and add water to make 2000 mL. Delete the Purity (5).
Mobile phase B: Dissolve 7.80 g of sodium dihydrogen
phosphate dihydrate in 550 mL of water, add 1400 mL of Change the Assay to read:
acetonitrile, adjust the pH to 6.0 with sodium hydroxide TS,
Assay Weigh accurately about 0.25 g of Theophylline,
and add water to make 2000 mL.
previously dried, and dissolve in 100 mL of water, add ex-
Flowing of the mobile phase: Flow mobile phase A for 10
actly 20 mL of 0.1 mol/L silver nitrate VS, shake the mix-
minutes before injection. After injection, control the
ture, and titrate with 0.1 mol/L sodium hydroxide VS
gradient by mixing the mobile A and B as directed in the fol-
(potentiometric titration). Perform a blank determination,
lowing table.
and make any necessary correction.
Time after injection Mobile phase Mobile phase Each mL of 0.1 mol/L sodium hydroxide VS
of sample (min) A (z) B (z)
= 18.02 mg of C7H8N4O2
0 – 32 100 → 70 0 → 30
32 – 40 70 → 50 30 → 50
40 – 42 50 → 100 50 → 0

Flow rate: 1.8 mL per minute.


Time span of measurement: About 1.7 times as long as the
retention time of teicoplanin A2-2 after the solvent peak.
System suitability—
Test for required detection: Conˆrm that peak height of
teicoplanin A2-2 obtained from the sample solution is equiva-
lent to 90z of the full scale.
1532 O‹cial Monographs for Part I Supplement I, JP XIV

Thiamine Hydrochloride with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratios
塩酸チアミン of the peak area of thiamine to that of the internal standard
is not more than 1.0z.
Change the Water to read:
Water Not more than 5.0z (30 mg, coulometric titration).
Thiopental Sodium
チオペンタールナトリウム
Thiamine Nitrate
硝酸チアミン
Change the Purity (4) to read:
Purity
Change the Assay to read: (4) Related substances—Dissolve 50 mg of Thiopental
Sodium in 50 mL of the mobile phase, and use this solution
Assay Weigh accurately about 0.1 g each of Thiamine Ni-
as the sample solution. Pipet 1 mL of the sample solution,
trate, previously dried, and Thiamine Hydrochloride Refer-
add the mobile phase to make exactly 200 mL, and use this
ence Standard (previously determine its water content in the
solution as the standard solution. Perform the test with ex-
same manner as directed under Thiamine Hydrochloride),
actly 20 mL each of the sample solution and the standard so-
and dissolve them in the mobile phase to make exactly 50
lution as directed under the Liquid Chromatography accord-
mL. To 10 mL each of the solutions, accurately measured,
ing to the following conditions. Measure each peak area of
add exactly 5 mL each of the internal standard solution, add
each solution by the automatic integration method: the total
the mobile phase to make 50 mL, and use these solutions as
area of peaks other than those of thiopental in the sample
the sample solution and the standard solution. Perform the
solution is not larger than the peak area of thiopental in the
test with 10 mL each of the sample solution and the standard
standard solution.
solution as directed under the Liquid Chromatography ac-
Operating conditions—
cording to the following conditions and calculate the ratios,
Detector: An ultraviolet absorption photometer
Q T and QS, of the peak area of thiamine to that of the inter-
(wavelength: 254 nm).
nal standard.
Column: A stainless steel column 4.6 mm in inside di-
QT ameter and 15 cm in length, packed with octadecylsilanized
Amount (mg) of C12H17N5O4S = WS × × 0.9706
QS silica gel (5 mm in particle diameter).
Column temperature: A constant temperature of about
WS: Amount (mg) of Thiamine Hydrochloride Reference
409C.
Standard, calculated on the anhydrous basis
Mobile phase: Dissolve 1 g of potassium dihydrogen phos-
Internal standard solution—A solution of methyl benzoate phate in 1000 mL of water, and adjust the pH to 3.0 with
in methanol (1 in 50). phosphoric acid. To 700 mL of this solution add 300 mL of
Operating conditions— acetonitrile.
Detector: An ultraviolet spectrophotometer (wavelength: Flow rate: Adjust the ‰ow rate so that the retention time
254 nm). of thiopental is about 15 minutes.
Column: A stainless steel column 4.6 mm in inside di- Time span of measurement: About 1.5 times as long as the
ameter and 15 cm in length, packed with octadecylsilanized retention time of thiopental.
silica gel for liquid chromatography (5 mm in particle di- System suitability—
ameter). Test for required detection: To exactly 2 mL of the stan-
Column temperature: A constant temperature of about dard solution add the mobile phase to make exactly 10 mL.
309C. Conˆrm that the peak area of thiopental obtained from 20
Mobile phase: Dissolve 1.1 g of sodium l-octanesulfonate mL of this solution is equivalent to 15 to 25z of that of
in 1000 mL of diluted acetic acid (100) (1 in 100). To 600 mL thiopental obtained from 20 mL of the standard solution.
of this solution add 400 mL of a mixture of methanol and System performance: Dissolve 5 mg each of isopropyl
acetonitrile (3:2). parahydroxybenzoate and propyl parahydroxybenzoate in
Flow rate: Adjust the ‰ow rate so that the retention time 50 mL of acetonitrile, and add water to make 100 mL. When
of thiamine is about 12 minutes. the procedure is run with 20 mL of this solution under the
System suitability— above operating conditions, isopropyl parahydroxybenzoate
System performance: When the procedure is run with 10 and propyl parahydroxybenzoate are eluted in this order
mL of the standard solution under the above operating con- with the resolution between these peaks being not less than
ditions, thiamine and the internal standard are eluted in this 1.9.
order with the resolution between these peaks being not less System repeatability: When the test is repeated 6 times
than 6. with 20 mL of the standard solution under the above operat-
System repeatability: When the test is repeated 6 times ing conditions, the relative standard deviation of the peak
Supplement I, JP XIV O‹cial Monographs for Part I 1533

area of thiopental is not more than 2.0z. tion. Pipet 1 mL of the sample solution, add the mobile
phase to make exactly 100 mL, and use this solution as the
standard solution. Perform the test with exactly 20 mL each
Tipepidine Hibenzate of the sample solution and the standard solution as directed
under the Liquid Chromatography according to the follow-
ヒベンズ酸チペピジン ing conditions, and determine each peak area by the auto-
matic integration method: the total area of all peaks other
Change the Purity (4) to read: than the area of the hibenzic acid and tipepidine from the
sample solution is not larger than 1 W 2 times the peak area of
Purity
the tipepidine from the standard solution.
(4) Related substances—(i) Dissolve 10 mg of Tipep-
Operating conditions—
idine Hibenzate in 20 mL of the mobile phase, and use this
Detector: An ultraviolet absorption photometer
solution as the sample solution. Pipet 1 mL of the sample so-
(wavelength: 254 nm).
lution, add the mobile phase to make exactly 100 mL, and
Column: A stainless steel column 4.6 mm in inside di-
use this solution as the standard solution. Perform the test
ameter and 15 cm in length, packed with octadecylsilanized
with exactly 20 mL each of the sample solution and the stan-
silica gel for liquid chromatography (5 mm in particle di-
dard solution as directed under the Liquid Chromatography
ameter).
according to the following conditions. Determine each peak
Column temperature: A constant temperature of about
area of both solutions by the automatic integration method:
409C.
the total area of all peaks other than the area of the hibenzic
Mobile phase: A mixture of methanol and a solution of
acid and tipepidine from the sample solution is not larger
ammonium acetate (1 in 500) (13:7).
than the peak area of the tipepidine from the standard solu-
Flow rate: Adjust the ‰ow rate so that the retention time
tion.
of tipepidine is about 10 minutes.
Operating conditions—
Time span of measurement: As long as the retention time
Detector: An ultraviolet absorption photometer
of tipepidine after the solvent peak.
(wavelength: 254 nm).
System suitability—
Column: A stainless steel column 4.6 mm in inside di-
Test for required detection: To exactly 2 mL of the stan-
ameter and 15 cm in length, packed with octadecylsilanized
dard solution add the mobile phase to make exactly 20 mL.
silica gel for liquid chromatography (5 mm in particle di-
Conˆrm that the peak area of tipepidine obtained from 20
ameter).
mL of this solution is equivalent to 7 to 13z of that of tipepi-
Column temperature: A constant temperature of about
dine obtained from 20 mL of the standard solution.
509C.
System performance: Dissolve 12 mg of Tipepidine
Mobile phase: A mixture of a solution of ammonium
Hibenzate and 4 mg of xanthene in 50 mL of the mobile
acetate (1 in 100) and tetrahydrofuran (32:13).
phase. When the procedure is run with 10 mL of this solution
Flow rate: Adjust the ‰ow rate so that the retention time
under the above operating conditions, hibenzic acid, tipepi-
of tipepidine is about 12 minutes.
dine and xanthene are eluted in this order with the resolution
Time span of measurement: As long as the retention time
between the peaks of tipepidine and xanthene being not less
of tipepidine after the solvent peak.
than 3.
System suitability—
System repeatability: When the test is repeated 6 times
Test for required detection: To exactly 2 mL of the stan-
with 20 mL of the standard solution under the above operat-
dard solution add the mobile phase to make exactly 20 mL.
ing conditions, the relative standard deviation of the peak
Conˆrm that the peak area of tipepidine obtained from 20
area of tipepidine is not more than 3.0z.
mL of this solution is equivalent to 7 to 13z of that of tipepi-
dine obtained from 20 mL of the standard solution.
System performance: Dissolve 10 mg of Tipepidine
Hibenzate and 3 mg of propyl parahydroxybenzoate in 100 Tobramycin
mL of the mobile phase. When the procedure is run with 20
トブラマイシン
mL of this solution under the above operating conditions,
hibenzic acid, tipepidine and propyl parahydroxybenzoate
Change to read except the structural formula
are eluted in this order with the resolution between the peaks
and chemical name:
of tipepidine and propyl parahydroxybenzoate being not less
than 3.
Tobramycin contains not less than 900 mg (potency)
System repeatability: When the test is repeated 6 times
per mg, calculated on the anhydrous basis. The poten-
with 20 mL of the standard solution under the above operat-
cy of Tobramycin is expressed as mass (potency) of
ing conditions, the relative standard deviation of the peak
tobramycin (C18H37N5O9).
area of tipepidine is not more than 1.5z.
(ii) Dissolve 10 mg of Tipepidine Hibenzate in 20 mL of Description Tobramycin occurs as a white to pale yellow-
the mobile phase, and use this solution as the sample solu- ish white powder.
1534 O‹cial Monographs for Part I Supplement I, JP XIV

It is very soluble in water, freely soluble in formamide, direct titration). Use a mixture of formamide for water de-
slightly soluble in methanol, and very slightly soluble in termination and methanol for water determination (3:1) in-
ethanol (95). stead of methanol for water determination.
It is hygroscopic.
Residue on ignition Not more than 1.0z (0.5 g).
Identiˆcation (1) Determine the spectrum of a solution
Assay Perform the test according to the Cylinder-plate
of Tobramycin in heavy water for nuclear magnetic
method as directed under the Microbial Assay for Antibiot-
resonance spectroscopy (1 in 125) as directed under the
ics according to the following conditions.
Nuclear Magnetic Resonance Spectroscopy (1H), using sodi-
(1) Test organism—Bacillus subtilis ATCC 6633
um 3-trimethylsilylpropanesulfonate for nuclear magnetic
(2) Culture medium—Use the medium i in 1) Medium
resonance spectroscopy as an internal reference compound:
for test organism [5] under (1) Agar media for seed and base
it exhibits a double signal A at around d 5.1 ppm, a multiple
layer.
signal B between d 2.6 ppm and d 4.0 ppm, and a multiple
(3) Standard solutions—Weigh accurately an amount of
signal C between d 1.0 ppm and d 2.1 ppm. The ratio of the
Tobramycin Reference Standard, equivalent to about 25 mg
integrated intensity of these signals, A:B:C, is about 1:8:2.
(potency), dissolve in 0.1 mol/L phosphate buŠer solution,
(2) Dissolve 10 mg each of Tobramycin and Tobramycin
pH 8.0 to make exactly 25 mL, and use this solution as the
Reference Standard in 1 mL of water, and use these solu-
standard stock solution. Keep the standard stock solution
tions as the sample solution and the standard solution.
between 59 C and 159 C, and use within 30 days. Take exactly
Perform the test with these solutions as directed under the
a suitable amount of the standard stock solution before use,
Thin-layer Chromatography. Spot 4 mL of the sample solu-
add 0.1 mol/L phosphate buŠer solution, pH 8.0 to make
tion and the standard solution on a plate of silica gel for
solutions so that each mL contains 8 mg (potency) and 2 mg
thin-layer chromatography. Develop the plate with a mix-
(potency), and use these solutions as the high concentration
ture of ammonia TS, 1-butanol and methanol (5:5:2) to a
standard solution and the low concentration standard solu-
distance of about 10 cm, and air-dry the plate. Spray evenly
tion, respectively.
ninhydrin TS on the plate, and heat at 1009C for 5 minutes:
(4) Sample solutions—Weigh accurately an amount of
the Rf values of the principal spots obtained from the sam-
Tobramycin, equivalent to about 25 mg (potency), and dis-
ple solution and the standard solution are the same.
solve in 0.1 mol/L phosphate buŠer solution, pH 8.0 to
Optical rotation [a]20
D : +138 – +1489(1 g calculated on the make exactly 25 mL. Take exactly a suitable amount of this
anhydrous basis, water, 25 mL, 100 mm). solution, add 0.1 mol/L phosphate buŠer solution, pH 8.0
to make solutions so that each mL contains 8 mg (potency)
pH The pH of a solution obtained by dissolving 0.10 g of
and 2 mg (potency), and use these solutions as the high con-
Tobramycin in 10 mL of water is between 9.5 and 11.5.
centration sample solution and the low concentration sample
Purity (1) Clarity and color of solution—Dissolve 1.0 g solution, respectively.
of Tobramycin in 10 mL of water: the solution is clear and
Containers and storage Containers—Tight containers.
colorless to pale yellow.
(2) Heavy metals—Proceed with 1.0 g of Tobramycin
according to Method 2, and perform the test. Prepare the
control solution with 3.0 mL of Standard Lead Solution (not Tocopherol
more than 30 ppm).
トコフェロール
(3) Related substances—Dissolve 80 mg of Tobramycin
in 10 mL of diluted ammonia solution (28) (1 in 250), and
Change the Assay to read:
use this solution as the sample solution. Pipet 1 mL of the
sample solution, add diluted ammonia solution (28) (1 in Assay Dissolve about 50 mg each of Tocopherol and To-
250) to make exactly 100 mL, and use this solution as the copherol Reference Standard, accurately weighed, in etha-
standard solution. Perform the test with these solutions as nol (99.5) to make exactly 50 mL, and use these solutions as
directed under the Thin-layer Chromatography. Spot 5 mL the sample solution and the standard solution. Perform the
of the sample solution and the standard solution on a plate test with exactly 20 mL each of these solutions as directed un-
of silica gel for thin-layer chromatography. Develop the der the Liquid Chromatography according to the following
plate with a mixture of ammonia solution (28), ethanol (95) conditions, and determine the peak heights, HT and HS, of
and 2-butanone (1:1:1) to a distance of about 10 cm, air-dry tocopherol in the sample solution and the standard solution.
the plate, then further dry at 1109C for 10 minutes. Immedi-
HT
ately spray evenly a mixture of water and sodium Amount (mg) of C29H50O2 = WS ×
HS
hypochlorite TS (4:1) on the plate, air-dry the plate, then
spray potassium iodide-starch TS on the plate: the spot other WS: Amount (mg) of Tocopherol Reference Standard
than the principal spot from the sample solution is not more
Operating conditions—
intense than the spot from the standard solution.
Detector: An ultraviolet absorption photometer (wave-
Water Not more than 11.0z (0.1 g, volumetric titration, length: 292 nm).
Supplement I, JP XIV O‹cial Monographs for Part I 1535

Column: A stainless steel column 4.6 mm in inside di- acetate are eluted in this order with the resolution between
ameter and 15 cm in length, packed with octadecylsilanized these peaks being not less than 2.6.
silica gel for liquid chromatography (5 mL in particle System repeatability: When the test is repeated 5 times
diameter). with 20 mL of the standard solution under the above operat-
Column temperature: A constant temperature of about ing conditions, the relative standard deviation of the peak
359C. heights of tocopherol acetate is not more than 0.8z.
Mobile phase: A mixture of methanol and water (49:1).
Flow rate: Adjust the ‰ow rate so that the retention time
of tocopherol is about 10 minutes. Triamcinolone
System suitability—
System performance: Dissolve 0.05 g each of Tocopherol トリアムシノロン
and tocopherol acetate in 50 mL of ethanol (99.5). When the
procedure is run with 20 mL of this solution under the above Change the Assay to read:
operating conditions, tocopherol and tocopherol acetate are
Assay Dissolve about 20 mg each of Triamcinolone and
eluted in this order with the resolution between these peaks
Triamcinolone Reference Standard, previously dried and ac-
being not less than 2.6.
curately weighed, in a solution of L-ascorbic acid in metha-
System repeatability: When the test is repeated 5 times
nol (1 in 1000) to make exactly 50 mL. Pipet 5 mL each of
with 20 mL of the standard solution under the above operat-
these solutions, add exactly 5 mL each of the internal stan-
ing conditions, the relative standard deviation of the peak
dard solution, add a solution of L-ascorbic acid in methanol
heights of tocopherol is not more than 0.8z.
(1 in 1000) to make 20 mL, and use these solutions as the
sample solution and the standard solution. Perform the test
with 10 mL each of these solutions as directed under the Liq-
Tocopherol Acetate uid Chromatography according to the following conditions,
and calculate the ratios, Q T and Q S, of the peak height of tri-
酢酸トコフェロール
amcinolone to that of the internal standard, respectively.

Change the Assay to read: QT


Amount (mg) of C21H27FO6 = WS ×
QS
Assay Dissolve 50 mg each of Tocopherol Acetate and
Tocopherol Acetate Reference Standard, accurately WS: Amount (mg) of Triamcinolone Reference Standard
weighed, in ethanol (99.5) to make exactly 50 mL, and use
Internal standard solution—Dissolve 15 mg of methyl para-
these solutions as the sample solution and the standard solu-
hydroxybenzoate in a solution of L-ascorbic acid in metha-
tion. Perform the test with exactly 20 mL each of these solu-
nol (1 in 1000) to make 100 mL.
tions as directed under the Liquid Chromatography accord-
Operating conditions—
ing to the following conditions, and determine the peak
Detector: An ultraviolet absorption photometer (wave-
heights, HT and HS, of tocopherol acetate in the sample solu-
length: 254 nm).
tion and the standard solution, respectively.
Column: A stainless steel column 4.0 mm in inside di-
HT ameter and 15 cm in length, packed with octadecylsilanized
Amount (mg) of C31H52O3 = WS ×
HS silica gel (5 mm in particle diameter).
Column temperature: A constant temperature of about
WS: Amount (mg) of Tocopherol Acetate Reference
259C.
Standard
Mobile phase: A mixture of water and acetonitrile (3:1).
Operating conditions— Flow rate: Adjust the ‰ow rate so that the retention time
Detector: An ultraviolet absorption photometer (wave- of triamcinolone is about 10 minutes.
length: 284 nm). System suitability—
Column: A stainless steel column 4.6 mm in inside di- System performance: When the procedure is run with 10
ameter and 15 cm in length, packed with octadecylsilanized mL of the standard solution under the above operating con-
silica gel (5 mm in particle diameter). ditions, triamcinolone and the internal standard are eluted in
Column temperature: A constant temperature of about this order with the resolution between these peaks being not
359C. less than 2.0.
Mobile phase: A mixture of methanol and water (49:1). System repeatability: When the test is repeated 6 times
Flow rate: Adjust the ‰ow rate so that the retention time with 10 mL of the standard solution under the above operat-
of tocopherol acetate is about 12 minutes. ing conditions, the relative standard deviation of the ratios
System suitability— of the peak height of triamcinolone to that of the internal
System performance: Dissolve 0.05 g each of Tocopherol standard is not more than 1.5z.
Acetate and tocopherol in 50 mL of ethanol (99.5). When
the procedure is run with 20 mL of this solution under the
above operating conditions, tocopherol and tocopherol
1536 O‹cial Monographs for Part I Supplement I, JP XIV

Change to read: amount of trichomycin A is between 20z and 40z, and that
of trichomycin B is between 15z and 25z. The relative
Trichomycin retention time of trichomycin B with respect to trichomycin
A is about 1.2.
トリコマイシン Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 360 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
ameter).
Column temperature: A constant temperature of about
Trichomycin A
259C.
33-(3-Amino-3,6-dideoxy-b-D-mannopyranosyloxy)-17-
Mobile phase: Dissolve 3.4 g of potassium dihydrogen
[6-(4-aminophenyl)-4-hydroxy-1-methyl-6-oxohexyl]-
phosphate and 1.7 g of sodium lauryl sulfate in a mixture of
1,3,5,9,11,37-hexahydroxy-18-methyl-13,15-dioxo-16,39-
600 mL of water and 400 mL of acetonitrile for liquid chro-
dioxabicyclo[33.3.1]nonatriaconta-19,21,23,25,27,29,31-
matography.
heptaene-36-carboxylic acid [12698-99-6]
Flow rate: Adjust the ‰ow rate so that the retention time
Trichomycin B
of trichomycin A is about 8 minutes.
33-(3-Amino-3,6-dideoxy-b-D-mannopyranosyloxy)-17-
Time span of measurement: About 4 times as long as the
[6-(4-aminophenyl)-4-hydroxy-1-methyl-6-oxohexyl]-
retention time of trichomycin A.
1,3,5,7,9,37-hexahydroxy-18-methyl-13,15-dioxo-16,39-
System suitability—
dioxabicyclo[33.3.1]nonatriaconta-19,21,23,25,27,29,31-
Test for required detectability: Measure exactly 5 mL of
heptaene-36-carboxylic acid [12699-00-2]
the sample solution, add a mixture of tetrahydrofuran for
[1394-02-1, Trichomycin]
liquid chromatography and water (3:1) to make exactly 50
mL, and use this solution as the solution for system suitabil-
Trichomycin contains not less than 7000 Units per ity test. Pipet 5 mL of the solution for system suitability test,
mg, calculated on the dried basis. The potency of and add a mixture of tetrahydrofuran for liquid chro-
Trichomycin is expressed as unit based on the amount matography and water (3:1) to make exactly 30 mL. Con-
of trichomycin. 1 unit of Trichomycin is equivalent to ˆrm that the peak area of trichomycin A obtained from 5 mL
0.05 mg of trichomycin. of this solution is equivalent to 12 to 22z of that from 5 mL
Description Trichomycin occurs as a yellow to yellow- of the solution for system suitability test.
brown powder. System performance: When the procedure is run with 5 mL
It is practically insoluble in water, in ethanol (99.5) and in of the solution for system suitability test under the above
tetrahydrofuran. operating conditions, trichomycin A and trichomycin B are
It dissolves in dilute sodium hydroxide TS. eluted in this order with the resolution between these peaks
It is hygroscopic. being not less than 2.5.
System repeatability: When the test is repeated 6 times
Identiˆcation (1) To 2 mg of Trichomycin add 2 mL of
with 5 mL of the solution for system suitability test under the
sulfuric acid: a blue color appears, and the color is changed
above operating conditions, the relative standard deviation
to a blue-purple after allowing to stand.
of the peak area of trichomycin A is not more than 2.0z.
(2) Dissolve 1 mg of Trichomycin in 50 mL of a solution
of sodium hydroxide (1 in 200). Determine the absorption Loss on drying Not more than 5.0z (1 g, in vacuum,
spectrum of this solution as directed under the Ultraviolet- 609C, 3 hours).
visible Spectrophotometry: it exhibits maxima between 359
Assay Conduct this procedure without exposure to day-
nm and 365 nm, between 378 nm and 384 nm, and between
light, using light-resistant vessels. Weigh accurately an
400 nm and 406 nm.
amount of Trichomycin and Trichomycin Reference Stan-
Content ratio of the active principle Conduct this proce- dard, equivalent to about 150,000 units, dissolve them
dure without exposure to daylight, using light-resistant ves- separately in a mixture of tetrahydrofuran for liquid chro-
sels. Dissolve about 10 mg of Trichomycin in 50 mL of a matography and water (3:1) to make exactly 100 mL, and
mixture of tetrahydrofuran for liquid chromatography and use these solutions as the sample solution and the standard
water (3:1), and use this solution as the sample solution. Per- solution. Perform the test with exactly 20 mL each of the
form the test with 5 mL of the sample solution as directed un- sample solution and the standard solution as directed under
der the Liquid Chromatography according to the following the Liquid Chromatography according to the following con-
conditions, determine the peak areas by the automatic in- ditions, and determine the peak areas, AT and AS, of
tegration method, and calculate the amount of trichomycin trichomycin.
A and trichomycin B by the area percentage method: the
Supplement I, JP XIV O‹cial Monographs for Part I 1537
AT
Amount (unit) of trichomycin = WS × A solution of it in N,N-dimethylformamide (1 in 20)
AS
shows no optical rotation.
WS: Amount (unit) of Trichomycin Reference Standard
Identiˆcation (1) Determine the absorption spectrum of
Operating conditions— a solution of Trimebutine Maleate in 0.01 mol/L
Detector: An ultraviolet absorption photometer (wave- hydrochloric acid TS (1 in 50,000) as directed under the
length: 360 nm). Ultraviolet-visible Spectrophotometry, and compare the
Column: A stainless steel column 4.6 mm in inside di- spectrum with the Reference Spectrum: both spectra exhibit
ameter and 15 cm in length, packed with silica gel for liquid similar intensities of absorption at the same wavelengths.
chromatography (10 mm in particle diameter). (2) Determine the infrared absorption spectrum of
Column temperature: A constant temperature of about Trimebutine Maleate as directed in the potassium bromide
259C. disk method under the Infrared Spectrophotometry, and
Mobile phase: Dissolve 15 g of ammonium acetate in 120 compare the spectrum with the Reference Spectrum: both
mL of water, and add 1000 mL of acetonitrile for liquid spectra exhibit similar intensities of absorption at the same
chromatography and 700 mL of methanol. wave numbers.
Flow rate: Adjust the ‰ow rate so that the retention time
Melting point 131 – 1359
C
of trichomycin is about 6 minutes.
System suitability— Purity (1) Heavy metals—Proceed with 2.0 g of Trime-
System performance: Dissolve 5 mg of Trichomycin and 1 butine Maleate according to Method 2, and perform the test.
mg of berberine chloride in 100 mL of a mixture of tetra- Prepare the control solution with 2.0 mL of Standard Lead
hydrofuran for liquid chromatography and water (3:1). Solution (not more than 10 ppm).
When the procedure is run with 20 mL of this solution under (2) Arsenic—Prepare the test solution with 2.0 g of
the above operating conditions, berberine and trichomycin Trimebutine Maleate according to Method 3, and perform
are eluted in this order with the resolution between these the test (not more than 1 ppm).
peaks being not less than 4. (3) Related substances—Dissolve 0.10 g of Trimebutine
System repeatability: When the test is repeated 6 times Maleate in 100 mL of a mixture of 0.01 mol/L hydrochloric
with 20 mL of the standard solution under the above operat- acid TS and acetonitrile (13:7), and use this solution as the
ing conditions, the relative standard deviation of the peak sample solution. Pipet 1 mL of the sample solution, add a
area of trichomycin is not more than 2.0z. mixture of 0.01 mol/L hydrochloric acid TS and acetonitrile
(13:7) to make exactly 250 mL, and use this solution as the
Containers and storage Containers—Tight containers.
standard solution. Perform the test with exactly 20 mL each
Storage—Light-resistant, and in a cold place.
of the sample solution and the standard solution as directed
under the Liquid Chromatography according to the follow-
ing conditions, and determine each peak area by the auto-
Add the following:
matic integration method: the area of the peak other than
maleic acid and trimebutine from the sample solution is not
Trimebutine Maleate more than 1/2 times the peak area of trimebutine from the
standard solution, and the total area of the peaks other than
マレイン酸トリメブチン
maleic acid and trimebutine is not more than the peak area
of trimebutine from the standard solution.
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized
C22H29NO5.C4H4O4: 503.54 silica gel for liquid chromatography (5 mm in particle di-
(2RS)-2-Dimethylamino-2-phenylbutyl 3,4,5- ameter).
trimethoxybenzoate monomaleate [34140-59-5] Column temperature: A constant temperature of about
409C.
Trimebutine Maleate, when dried, contains not less Mobile phase: To 650 mL of diluted perchloric acid (17 in
than 98.5z and not more than 101.0z of trimebutine 20,000), previously adjusted the pH to 3.0 with a solution of
maleate (C22H29NO5.C4H4O4). ammonium acetate (1 in 1000), add 1 g of sodium 1-pen-
tanesulfonate to dissolve. To 650 mL of this solution add
Description Trimebutine Maleate occurs as white, crystals
350 mL of acetonitrile.
or crystalline powder.
Flow rate: Adjust the ‰ow rate so that the retention time
It is freely soluble in N,N-dimethylformamide and in acet-
of trimebutine is about 9 minutes.
ic acid (100), soluble in acetonitrile, and slightly soluble in
Time span of measurement: About 2 times as long as the
water and in ethanol (99.5).
retention time of trimebutine after the peak of maleic acid.
It dissolves in 0.01 mol/L hydrochloric acid TS.
1538 O‹cial Monographs for Part I Supplement I, JP XIV

System suitability— Ultraviolet-visible Spectrophotometry, and compare the


Test for required detectability: Measure exactly 5 mL of spectrum with the Reference Spectrum: both spectra exhibit
the standard solution, and add a mixture of 0.01 mol/L similar intensities of absorption at the same wavelengths.
hydrochloric acid TS and acetonitrile (13:7) to make exactly (2) Determine the infrared absorption spectrum of
20 mL. Conˆrm that the peak area of trimebutine obtained Trimetoquinol Hydrochloride as directed in the potassium
from 20 mL of this solution is equivalent to 20 to 30z of that chloride disk method under the Infrared Spectrophotomet-
from 20 mL of the standard solution. ry, and compare the spectrum with the Reference Spectrum:
System performance: Dissolve 40 mg of Trimebutine both spectra exhibit similar intensities of absorption at the
Maleate and 20 mg of imipramine hydrochloride in 100 mL same wave numbers.
of a mixture of 0.01 mol/L hydrochloric acid TS and (3) A solution of Trimetoquinol Hydrochloride (1 in 50)
acetonitrile (13:7). When the procedure is run with 20mL of responds to the Qualitative Tests (1) for chloride.
this solution under the above operating conditions, trimebu-
tine and imipramine are eluted in this order with the resolu- Change the Optical rotation to read:
tion between these peaks being not less than 2.5.
Optical rotation [a]20
D : -16 – -199(0.25 g, calculated on
System repeatability: When the test is repeated 6 times
the anhydrous basis, water, after warming and cooling,
with 20 mL of the standard solution under the above operat-
25 mL, 100 mm).
ing conditions, the relative standard deviation of the peak
area of trimebutine is not more than 5z.
Change the Purity to read:
Loss on drying Not more than 0.5z (1 g, 1059
C, 3 hours).
Purity (1) Clarity and color of solution—Dissolve 0.10 g
Residue on ignition Not more than 0.10z (1 g). of Trimetoquinol Hydrochloride in 10 mL of water by
warming: the solution is clear and colorless.
Assay Weigh accurately about 0.8 g of Trimebutine Male-
(2) Sulfate—Perform the test with 0.5 g of Trimeto-
ate, previously dried, dissolve in 70 mL of acetic acid (100),
quinol Hydrochloride. Prepare the control solution with
and titrate with 0.1 mol/L perchloric acid VS until the color
0.40 mL of 0.005 mol/L sulfuric acid VS (not more than
of the solution changes from purple through blue to blue-
0.038z).
green (indicator: 3 drops of crystal violet TS). Perform a
(3) Heavy metals—Proceed with 1.0 g of Trimetoquinol
blank determination, and make any necessary correction.
Hydrochloride according to Method 2, and perform the test.
Each mL of 0.1 mol/L perchloric acid VS Prepare the control solution with 2.0 mL of Standard Lead
= 50.35 mg of C22H29NO5.C4H4O4 Solution (not more than 20 ppm).
(4) Related substances—Dissolve 50 mg of Trimeto-
Containers and storage Containers—Well-closed contain-
quinol Hydrochloride in 50 mL of the mobile phase, and use
ers.
this solution as the sample solution. Pipet 1 mL of this solu-
tion, add the mobile phase to make exactly 100 mL, and use
this solution as the standard solution. Perform the test with
Trimetoquinol Hydrochloride exactly 20 mL each of the sample solution and the standard
solution, as directed under the Liquid Chromatography ac-
塩酸トリメトキノール
cording to the following conditions. Determine each peak
area of both solutions by the automatic integration method:
Change the origin/limits of content to read:
the total area of the peaks other than that of trimetoquinol
from the sample solution is not larger than the peak area of
Trimetoquinol Hydrochloride contains not less than
trimetoquinol from the standard solution.
98.5z and not more than 101.0z of C19H23NO5.HCl
Operating conditions—
(mol. wt.: 381.85), calculated on the anhydrous basis.
Detector: An ultraviolet absorption photometer (wave-
length: 283 nm).
Change the Description to read:
Column: A stainless steel column 4.6 mm in inside di-
Description Trimetoquinol Hydrochloride occurs as white, ameter and 15 cm in length, packed with octadecylsilanized
crystals or crystalline powder. silica gel for liquid chromatography (5 mm in particle di-
It is freely soluble in methanol, and sparingly soluble in ameter).
water and in ethanol (99.5). Column temperature: A constant temperature of about
Melting point: about 1519C (with decomposition, after 409C.
drying in vacuum, 1059C, 4 hours). Mobile phase: Dissolve 2 g of potassium dihydrogen phos-
phate and 2 g of sodium 1-pentane sulfonate in 1000 mL of
Change the Identiˆcation to read: water. Adjust with phosphoric acid to a pH between 2.8 and
3.2, and ˆlter through a membrane ˆlter with pore size of 0.4
Identiˆcation (1) Determine the absorption spectrum of
mm. Add 200 mL of acetonitrile to 800 mL of the ˆltrate.
a solution of Trimetoquinol Hydrochloride in 0.01 mol/L
Flow rate: Adjust the ‰ow rate so that the retention time
hydrochloric acid TS (1 in 20,000) as directed under the
of trimetoquinol is about 7 minutes.
Supplement I, JP XIV O‹cial Monographs for Part I 1539

Time span of measurement: About twice as long as the tion at the same wave numbers.
retention time of trimetoquinol after the solvent peak. (3) Dissolve 20 mg of Vancomycin Hydrochloride in 10
System suitability— mL of water, and add 1 drop of silver nitrate TS: a white tur-
Test for required detection: To exactly 2 mL of the stan- bidity is produced.
dard solution add the mobile phase to make exactly 20 mL.
Optical rotation [a]20
D : -30 – -409 (0.2 g calculated on
Conˆrm that the peak area of trimetoquinol obtained from
the anhydrous basis, water, 20 mL, 100 mm).
20 mL of this solution is equivalent to 7 to 13z of that of
trimetoquinol obtained from 20 mL of the standard solution. pH The pH of a solution obtained by dissolving 0.25 g of
System performance: Dissolve 5 mg of Trimetoquinol Vancomycin Hydrochloride in 5 mL of water is between 2.5
Hydrochloride and 1 mg of procaine hydrochloride in 50 mL and 4.5.
of the mobile phase. When the procedure is run with 20 mL
Purity (1) Heavy metals—Proceed with 1.0 g of Van-
of this solution under the above operating conditions,
comycin Hydrochloride according to Method 2, and per-
procaine and trimetoquinol are eluted in this order with the
form the test. Prepare the control solution with 2.0 mL of
resolution between these peaks being not less than 4.
Standard Lead Solution (not more than 20 ppm).
System repeatability: When the test is repeated 6 times
(2) Related substances—Dissolve 0.10 g of Vancomycin
with 20 mL of the standard solution under the above operat-
Hydrochloride in 10 mL of the mobile phase A, and use this
ing conditions, the relative standard deviation of the peak
solution as the sample solution. Pipet 1 mL of the sample so-
area of trimetoquinol is not more than 2.0z.
lution, add the mobile phase A to make exactly 25 mL, and
use this solution as the standard solution. Perform the test
Delete the Loss on drying and add the following:
with exactly 20 mL each of the sample solution and the stan-
Water 3.5 – 5.5z (0.3 g, volumetric titration, direct titra- dard solution as directed under the Liquid Chromatography
tion). according to the following conditions. If necessary, proceed
with 20 mL of the mobile phase A in the same manner to
compensate for the base line. Determine each peak area by
Vancomycin Hydrochloride the automatic integration method: the area of each peak
other than vancomycin from the sample solution is not more
塩酸バンコマイシン than the peak area of vancomycin from the standard solu-
tion, and the total area of the peaks other than vancomycin
Change to read except the structural formula is not more than 3 times of the peak area of vancomycin
and chemical name: from the standard solution.
Operating conditions—
Vancomycin Hydrochloride contains not less than Detector: An ultraviolet absorption photometer (wave-
1000 mg (potency) per mg, calculated on the anhydrous length: 280 nm).
basis. The potency of Vancomycin Hydrochloride is Column: A stainless steel column 4.6 mm in inside di-
expressed as mass (potency) of vancomycin ameter and 25 cm in length, packed with octadecylsilanized
(C66H75Cl2N9O24: 1449.25). silica gel for liquid chromatography (5 mm in particle di-
ameter).
Description Vancomycin Hydrochloride occurs as a white
Column temperature: A constant temperature of about
powder.
259C.
It is freely soluble in water, soluble in formamide, slightly
Mobile phase A: A mixture of triethylamine buŠer solu-
soluble in methanol, very slightly soluble in ethanol (95),
tion, pH 3.2, acetonitrile and tetrahydrofuran (92:7:1). Ad-
and practically insoluble in acetonitrile.
just the amount of acetonitrile so that the retention time of
It is hygroscopic.
vancomycin is 7.5 to 10.5 minutes.
Identiˆcation (1) Determine the absorption spectrum of Mobile phase B: A mixture of triethylamine buŠer solu-
a solution of Vancomycin Hydrochloride (1 in 10,000) as tion, pH 3.2, acetonitrile and tetrahydrofuran (70:29:1).
directed under the Ultraviolet-visible Spectrophotometry, Flowing of the mobile phase: Control the gradient by mix-
and compare the spectrum with the Reference Spectrum or ing the mobile phases A and B as directed in the following
the spectrum of a solution of Vancomycin Hydrochloride table.
Reference Standard prepared in the same manner as the
sample solution: both spectra exhibit similar intensities of Time after injection Mobile phase Mobile phase
absorption at the same wavelengths. of sample (min) A (z ) B (z )
(2) Determine the infrared absorption spectrum of Van- 0 – 12 100 0
comycin Hydrochloride as directed in the potassium 12 – 20 100 → 0 0 → 100
bromide disk method under the Infrared Spectrophotomet- 20 – 22 0 100
ry, and compare the spectrum with the Reference Spectrum
or the spectrum of Vancomycin Hydrochloride Reference Flow rate: 1.5 mL per minute.
Standard: both spectra exhibit similar intensities of absorp- Time span of measurement: As long as about 2.5 times of
1540 O‹cial Monographs for Part I Supplement I, JP XIV

the retention time of vancomycin after the solvent peak. 6.4 after sterilization.
System suitability— (3) Standard solutions—Weigh accurately an amount of
Test for required detectability: Conˆrm that the peak area Vancomycin Hydrochloride Reference Standard, equivalent
of vancomycin obtained from 20 mL of the standard solution to about 25 mg (potency), dissolve in water to make exactly
is equivalent to 3 to 5z of that from 20 mL of the sample so- 25 mL, and use this solution as the standard stock solution.
lution. Keep the standard stock solution at 59 C or below, and use
System performance: Dissolve 5 mg of Vancomycin within 7 days. Take exactly a suitable amount of the stan-
Hydrochloride in 10 mL of water, heat at 659 C for 48 hours, dard stock solution before use, add 0.1 mol/L phosphate
and cool to the ordinal temperature. When the procedure is buŠer solution, pH 4.5 to make solutions so that each mL
run with 20 mL of this solution under the above operating contains 100 mg (potency) and 25 mg (potency), and use these
conditions, related substance 1, vancomycin and related sub- solutions as the high concentration standard solution and
stance 2 are eluted in this order, the resolution between the the low concentration standard solution, respectively.
peaks of the related substance 1 and vancomycin is not less (4) Sample solutions—Weigh accurately an amount of
than 3, the theoretical plates of the peak of vancomycin is Vancomycin Hydrochloride, equivalent to about 25 mg
not less than 1500 steps, and the related substance 2 is eluted (potency), and dissolve in water to make exactly 25 mL.
between 15 and 18 minutes. Take exactly a suitable amount of this solution, add 0.1
System repeatability: When the test is repeated 5 times mol/L phosphate buŠer solution, pH 4.5 to make solutions
with 20 mL of the standard solution under the above operat- so that each mL contains 100 mg (potency) and 25 mg (poten-
ing conditions, the relative standard deviation of the peak cy), and use these solutions as the high concentration sample
area of vancomycin is not more than 2.0z. solution and the low concentration sample solution, respec-
tively.
Water Not more than 5.0z (0.1 g, volumetric titration,
direct titration. Use a mixture of formamide for water deter- Containers and storage Containers—Tight containers.
mination and methanol for water determination (3:1)).

Residue on ignition Not more than 1.0z (1 g).


Xylitol Injection
Assay Perform the test according to the Cylinder-plate
method as directed under the Microbial Assay for Antibiot- キシリトール注射液
ics according to the following conditions.
(1) Test organism—Bacillus subtilis ATCC 6633 Change the Containers and storage to read:
(2) Culture medium—Use the medium i in 1) Medium
Containers and storage Containers—Hermetic containers.
for test organism [5] under (1) Agar media for seed and base
Plastic containers for aqueous injections may be used.
layer. Adjust the pH of the medium so that it will be 6.2 to
General Rules
for Crude Drugs
Change the paragraph 1 to read: Picrasma Wood, Pinellia Tuber, Plantago Herb, Plan-
tago Seed, Platycodon Root, Polygala Root, Polygo-
1. Crude drugs in the monographs include medici-
num Root, Potato Starch, Powdered Acacia, Pow-
nal parts obtained from plants or animals, cell inclu-
dered Alisma Rhizome, Powdered Aloe, Powdered
sions and secretes separated from the origins, their ex-
Amomum Seed, Powdered Atractylodes Lancea Rhi-
tracts, and minerals. General Rules for Crude Drugs
zome, Powdered Atractylodes Rhizome, Powdered
and the Crude Drugs in General Tests, Processes and
Calumba, Powdered Capsicum, Powdered Cinnamon
Apparatus are applicable to the followings:
Bark, Powdered Clove, Powdered Cnidium Rhizome,
Acacia, Achyranthes Root, Agar, Agar Powder,
Powdered Coix Seed, Powdered Coptis Rhizome,
Akebia Stem, Alisma Rhizome, Aloe, Alpinia O‹cina-
Powdered Cyperus Rhizome, Powdered Digitalis,
rum Rhizome, Amomum Seed, Anemarrhena Rhi-
Powdered Dioscorea Rhizome, Powdered Fennel,
zome, Angelica Dahurica Root, Apricot Kernel, Are-
Powdered Gambir, Powdered Gardenia Fruit, Pow-
ca, Artemisia Capillaris Flower, Asiasarum Root,
dered Gentian, Powdered Geranium Herb, Powdered
Asparagus Tuber, Astragalus Root, Atractylodes Lan-
Ginger, Powdered Ginseng, Powdered Glycyrrhiza,
cea Rhizome, Atractylodes Rhizome, Bear Bile, Bearb-
Powdered Hoelen, Powdered Ipecac, Powdered
erry Leaf, Belladonna Root, Benzoin, Bitter Carda-
Japanese Angelica Root, Powdered Japanese Gentian,
mon, Bitter Orange Peel, Bupleurum Root, Burdock
Powdered Japanese Valerian, Powdered Magnolia
Fruit, Calumba, Capsicum, Cardamon, Cassia Seed,
Bark, Powdered Moutan Bark, Powdered Oyster
Catalpa Fruit, Chrysanthemum Flower, Chuling,
Shell, Powdered Panax Rhizome, Powdered Peach
Cimicifuga Rhizome, Cinnamon Bark, Citrus Unshiu
Kernel, Powdered Peony Root, Powdered Phelloden-
Peel, Clematis Root, Clove, Cnidium Rhizome, Coix
dron Bark, Powdered Picrasma Wood, Powdered
Seed, Condurango, Coptis Rhizome, Corn Starch,
Platycodon Root, Powdered Polygala Root, Powdered
Cornus Fruit, Corydalis Tuber, Cyperus Rhizome,
Polypourus Sclerotium, Powdered Rhubarb, Pow-
Digenea, Digitalis, Dioscorea Rhizome, Ephedra
dered Rose Fruit, Powdered Scutellaria Root, Pow-
Herb, Eucommia Bark, Evodia Fruit, Fennel, For-
dered Senega, Powdered Senna Leaf, Powdered Smi-
sythia Fruit, Fritillaria Bulb, Gambir, Gardenia Fruit,
lax Rhizome, Powdered Sophora Root, Powdered
Gastrodia Tuber, Gentian, Geranium Herb, Ginger,
Sweet Hydrangea Leaf, Powdered Swertia Herb, Pow-
Ginseng, Glehnia Root, Glycyrrhiza, Gypsum, Hemp
dered Tragacanth, Powdered Zanthoxylum Fruit,
Fruit, Hoelen, Honey, Houttuynia Herb, Immature
Prunella Spike, Pueraria Root, Red Ginseng, Rehman-
Orange, Imperata Rhizome, Ipecac, Japanese Angelica
nia Root, Rhubarb, Rice Starch, Rose Fruit, Rosin,
Root, Japanese Gentian, Japanese Valerian, Jujube,
SaŒower, SaŠron, Saposhnikovia Root, Saussurea
Jujube Seed, Lithospermum Root, Longgu, Loquat
Root, Schisandra Fruit, Schizonepeta Spike, Scopolia
Leaf, Magnolia Bark, Magnolia Flower, Mallotus
Rhizome, Scutellaria Root, Senega, Senna Leaf,
Bark, Mentha Herb, Moutan Bark, Mulberry Bark,
Sinomenium Stem, Smilax Rhizome, Sophora Root,
Notopterygium Rhizome, Nuphar Rhizome, Nux
Sweet Hydrangea Leaf, Swertia Herb, Toad Venom,
Vomica, Ophiopogon Tuber, Oriental Bezoar, Oyster
Tragacanth, Trichosanthes Root, Uncaria Thorn,
Shell, Panax Rhizome, Peach Kernel, Peony Root,
Wheat Starch, Zanthoxylum Fruit, Zedoary.
Perilla Herb, Pharbitis Seed, Phellodendron Bark,

1541
O‹cial Monographs
for Part II

Acacia Powdered Acacia


アラビアゴム アラビアゴム末

Change the Identiˆcation to read: Change the Identiˆcation to read:


Identiˆcation To 1 g of powdered Acacia add 25 mL of Identiˆcation To 1 g of Powdered Acacia add 25 mL of
water and 1 mL of sulfuric acid, and heat under a re‰ux con- water and 1 mL of sulfuric acid, and heat under a re‰ux con-
denser in a boiling water bath for 60 minutes. After cooling, denser in a boiling water bath for 60 minutes. After cooling,
add gently 2.0 g of anhydrous sodium carbonate. To 1 mL add gently 2.0 g of anhydrous sodium carbonate. To 1 mL
of this solution add 9 mL of methanol, mix well, centrifuge, of this solution add 9 mL of methanol, mix well, centrifuge,
and use the supernatant liquid as the sample solution. and use the supernatant liquid as the sample solution.
Separately, dissolve 0.01 g of D-galactose in 1 mL water, add Separately, dissolve 0.01 g of D-galactose in 1 mL water, add
methanol to make 10 mL, and use this solution as the stan- methanol to make 10 mL, and use this solution as the stan-
dard solution (1). Proceed with L-arabinose and with L- dard solution (1). Proceed with L-arabinose and with L-
rhamnose monohydrate in the same manner as for the rhamnose monohydrate in the same manner as for the
preparation of the standard solution (1), and use so obtained preparation of the standard solution (1), and use so obtained
solutions as the standard solution (2) and the standard solu- solutions as the standard solution (2) and the standard solu-
tion (3), respectively. Perform the test with these solutions as tion (3), respectively. Perform the test with these solutions as
directed under the Thin-layer chromatography. Spot 10 mL directed under the Thin-layer chromatography. Spot 10 mL
each of the sample solution and the standard solutions (1), each of the sample solution and the standard solutions (1),
(2) and (3) on a plate of silica gel for thin-layer chro- (2) and (3) on a plate of silica gel for thin-layer chro-
matography. Develop the plate with a mixture of acetone matography. Develop the plate with a mixture of acetone
and water (9:1) to a distance of about 10 cm, and air-dry the and water (9:1) to a distance of about 10 cm, and air-dry the
plate. Spray evenly 1-naphthol-sulfuric acid TS on the plate, plate. Spray evenly 1-naphthol-sulfuric acid TS on the plate,
and heat at 1059 C for 5 minutes: the three spots from the and heat at 1059 C for 5 minutes: the three spots from the
sample solution are the same with the spots of D-galactose, sample solution are the same with the spots of D-galactose,
L-arabinose and L-rhamnose from the standard solution in L-arabinose and L-rhamnose from the standard solution in
the color tone and the Rf value, respectively. the color tone and the Rf value, respectively.

Change the Purity to read: Change the Purity to read:


Purity (1) Insoluble residue—To 5.0 g of pulverized Aca- Purity (1) Insoluble residue—To 5.0 g of Powdered Aca-
cia add 100 mL of water and 10 mL of dilute hydrochloric cia add 100 mL of water and 10 mL of dilute hydrochloric
acid, and dissolve by gentle boiling for 15 minutes with acid, and dissolve by gentle boiling for 15 minutes with
swirling. Filter the warm mixture through a tared glass ˆlter swirling. Filter the warm mixture through a tared glass ˆlter
(G3), wash the residue thoroughly with hot water, and dry at (G3), wash the residue thoroughly with hot water, and dry at
1059 C for 5 hours: the mass of the residue does not exceed 1059 C for 5 hours: the mass of the residue does not exceed
10.0 mg. 10.0 mg.
(2) Tannin-bearing gums—To 10 mL of a solution of (2) Tannin-bearing gums—To 10 mL of a solution of
Acacia (1 in 50) add 3 drops of iron (III) chloride TS: no Powdered Acacia (1 in 50) add 3 drops of iron (III) chloride
dark green color is produced. TS: no dark green color is produced.
(3) Glucose—Dissolve 0.01 g of glucose in 1 mL of (3) Glucose—Dissolve 0.01 g of glucose in 1 mL of
water, add methanol to make 10 mL, and use this solution as water, add methanol to make 10 mL, and use this solution as
the standard solution. Proceed with the sample solution ob- the standard solution. Proceed with the sample solution ob-
tained in the Identiˆcation and the standard solution ob- tained in the Identiˆcation and the standard solution ob-
tained here as directed in the Identiˆcation: any spot at the tained here as directed in the Identiˆcation: any spot at the
Rf value corresponding to glucose from the standard solu- Rf value corresponding to glucose from the standard solu-
tion does not appear from the sample solution. tion does not appear from the sample solution.

1543
1544 O‹cial Monographs for Part II Supplement I, JP XIV

Aloe zamide in methanol (1 in 2000) and methanol to make ex-


actly 10 mL. When the procedure is run with 5 mL of this so-
アロエ lution under the above operating conditions except the
wavelength of 300 nm, barbaloin and ethenzamide are elut-
Change the origin/limits of content to read: ed in this order with the resolution between these peaks
being not less than 2.0.
Aloe is the dried juice of the leaves mainly of Aloe System repeatability: When the test is repeated 6 times
ferox Miller, or of hybrids of the species with Aloe with 5 mL of the standard solution under the above operat-
africana Miller or Aloe spicata Baker (Liliaceae). ing conditions, the relative standard deviation of the peak
It contains not less than 4.0z of barbaloin, calcu- area of barbaloin is not more than 1.5z.
lated on the basis of dried material.

Change the Loss on drying to read: Powdered Aloe


Loss on drying Not more than 12.0z.
アロエ末

Add the following next to Extract content:


Change the Identiˆcation (2) to read:
Component determination Weigh accurately about 0.1 g
Identiˆcation (2) To 0.2 g of Powdered Aloe add 10 mL
of pulverized Aloe, add 40 mL of methanol, and heat under
of methanol, shake for 5 minutes, ˆlter, and use the ˆltrate
a re‰ex condenser on a water bath for 30 minutes. After
as the sample solution. Separately, dissolve 1 mg of bar-
cooling, ˆlter, and add methanol to the ˆltrate to make ex-
baloin for thin-layer chromatography in 1 mL of methanol,
actly 50 mL. Pipet 5 mL of the solution, add methanol to
and use this solution as the standard solution. Perform the
make exactly 10 mL, and use this solution as the sample so-
test with these solutions as directed under the Thin-layer
lution. Separately, weigh accurately about 10 mg of bar-
Chromatography. Spot 10 mL each of the sample solution
baloin for component determination, previously dried in a
and the standard solution on a plate of silica gel for thin-lay-
desiccator (in vacuum, phosphorus (V) oxide) for 24 hours,
er chromatography. Develop the plate with a mixture of
add 0.04 g of oxalic acid dihydrate, and dissolve in methanol
ethyl acetate, acetone, water and acetic acid (100) (20:5:2:2)
to make exactly 100 mL. Pipet 5 mL of the solution, add
to a distance of about 10 cm, and air-dry the plate. Examine
methanol to make exactly 10 mL, and use this solution as the
under ultraviolet light (main wavelength: 365 nm): one spot
standard solution. Perform the test with exactly 5 mL each of
among several spots from the sample solution has the same
the sample solution and the standard solution as directed un-
color tone and the same Rf value with the red ‰uorescent
der the Liquid Chromatography according to the following
spot from the standard solution.
conditions, and measure the peak areas of barbaloin, AT and
AS, of both solutions.

AT Add the following:


Amount (mg) of barbaloin = WS × ×2
AS

WS: Amount (mg) of barbaloin for component determi- Alpinia O‹cinarum Rhizome
nation
Alpiniae o‹cinari Rhizoma
Operating conditions—
リョウキョウ
Detector: An ultraviolet absorption photometer (wave-
length: 360 nm).
Column: A stainless steel column about 6 mm in inside di-
Alpinia O‹cinarum Rhizome is the rhizome of Al-
ameter and about 15 cm in length, packed with octadecyl-
pinia o‹cinarum Hance (Zingiberaceae).
silanized silica gel for liquid chromatography (5 mm in parti- Description Alpinia O‹cinarum Rhizome is a slightly
cle diameter). curved and cylindrical rhizome, sometimes branched; 2 to 8
Column temperature: A constant temperature of about cm in length, 0.6 to 1.5 cm in diameter; externally red-brown
309C. to dark brown with ˆne striped lines, grayish white nodes
Mobile phase: A mixture of water, acetonitrile and acetic and several traces of rootlet; hard to break; fracture surface,
acid (100) (74:26:1). light brown in color and thickness of cortex is as same as
Flow rate: Adjust the ‰ow rate so that the retention time that of stele.
of barbaloin is about 12 minutes. Odor, characteristic; taste, extremely pungent.
System suitability— Under a microscope, transverse section reveals epidermal
System performance: To about 10 mg of barbaloin for cells often containing resin-like substances; cortex, endoder-
component determination add 0.04 g of oxalic acid dihy- mis and stele present beneath the epidermis; cortex and stele
drate, and dissolve in methanol to make exactly 100 mL. divided by endodermis; vascular bundles surrounded by
Pipet 5 mL of the solution, add 1 mL of a solution of ethen- ˆbers, scattered throughout the cortex and stele, cortex and
Supplement I, JP XIV O‹cial Monographs for Part II 1545

stele composed of parenchyma interspersed with oil cells; about 0.1 cm in diameter, with shallow longitudinal wrinkles
parenchymatous cells containing solitary crystals of calcium on the surface, and brittle. The rhizome, 2 – 4 cm in length,
oxalate and starch grains, starch grains generally simple 0.2 – 0.3 cm in diameter, often branched, with longitudinal
(sometimes 2- to 8-compound), ovate, oblong or narrowly wrinkles on the surface; internode short; each node has
ovate, 10 – 40 mm in diameter and with an eccentric navel. several scars of petiole and peduncle, and several thin and
long roots.
Identiˆcation To 0.5 g of pulverized Alpinia O‹cinarum
Odor, characteristic; taste, acrid, with some sensation of
Rhizome add 5 mL of acetone, shake for 5 minutes, and
numbness on the tongue.
ˆlter. Perform the test with the ˆltrate as directed under the
Thin-layer Chromatography. Spot 5 mL of the ˆltrate on a
Change the Purity (1) to read:
plate of silica gel for thin-layer chromatography, develop the
plate with a mixture of cyclohexane, ethyl acetate and acetic Purity (1) Terrestrial part—Any terrestrial parts are not
acid (100) (12:8:1) to a distance of about 10 cm, and air-dry found.
the plate: two yellow-brown spots appear at around Rf 0.4 –
0.5. Add the following next to Purity (2):
Loss on drying Not more than 15.0z (6 hours). Purity
(3) Aristolochic acid I—To exactly 2.0 g of pulverized
Total ash Not more than 7.5z.
Asiasarum Root add exactly 50 mL of diluted methanol (3 in
Acid-insoluble ash Not more than 1.5z. 4), shake for 15 minutes, ˆlter, and use the ˆltrate as the
sample solution. Separately, dissolve exactly 1.0 mg of
Extract content Dilute ethanol-extract: not less than
aristolochic acid I for crude drugs purity test in diluted
14.0z.
methanol (3 in 4) to make exactly 100 mL. Pipet 1 mL of this
solution, add diluted methanol (3 in 4) to make exactly 25
mL, and use this solution as the standard solution. Perform
Areca the test with exactly 20 mL each of the sample solution and
the standard solution as directed under the Liquid Chro-
ビンロウジ
matography, according to the following conditions: the sam-
ple solution shows no peak at the retention time corre-
Change the Identiˆcation to read:
sponding to aristolochic acid I from the standard solution. If
Identiˆcation Weigh 3 g of pulverized Areca in a glass- the sample solution shows such a peak, repeat the test under
stoppered centrifuge tube, and add 30 mL of diethyl ether diŠerent conditions to conˆrm that the peak in question is
and 5 mL of sodium hydroxide TS, stopper tightly, shake not aristolochic acid I.
for 5 minutes, centrifuge, and separate the diethyl ether Operating conditions—
layer. Evaporate the diethyl ether on a water bath, dissolve Detector: An ultraviolet or visible absorption photometer
the residue in 1.5 mL of methanol, ˆlter, and use the ˆltrate (wavelength: 400 nm).
as the sample solution. Separately, dissolve 5 mg of areco- Column: A stainless steel column 4.6 mm in inside di-
line hydrobromide for thin-layer chromatography in 1 mL ameter and 25 cm in length, packed with octadecylsilanized
of methanol, and use this solution as the standard solution. silica gel for liquid chromatography (5 mm in particle
Perform the test with these solutions as directed under the diameter).
Thin-layer chromatography. Spot 5 mL each of the sample Column temperature: A constant temperature of about
solution and the standard solution on a plate of silica gel for 409C.
thin-layer chromatography. Develop the plate with a mix- Mobile phase: A mixture of a solution prepared by dis-
ture of acetone, water and acetic acid (100) (10:6:1) to a dis- solving 7.8 g of sodium dihydrogen phosphate dihydrate and
tance of about 10 cm, and air-dry the plate. Spray evenly 2 mL of phosphoric acid in water to make 1000 mL and
iodine TS on the plate: one spot among the spots from the acetonitrile (11:9).
sample solution and a red-brown spot from the standard so- Flow rate: Adjust the ‰ow rate so that the retention time
lution show the same color tone and the same Rf value. of aristolochic acid I is about 15 minutes.
System suitability—
Test for required detectability: Measure exactly 1 mL of
Asiasarum Root the standard solution, and add diluted methanol (3 in 4) to
make exactly 10 mL. Conˆrm that the ratio, S/N, of the sig-
サイシン nal (S) and noise (N) of aristolochic acid I obtained from 20
mL of this solution is not less than 3. In this case, S means
Change the Description to read: the peak height on the chromatogram not including noise
Description Asiasarum Root is a nearly cylindrical rhi- obtained by drawing an average line of the detector output,
zome with numerous thin and long roots, externally light and N is 1/2 of the diŠerence between the maximum and
brown to dark brown. The root, about 15 cm in length, minimum output signals of the baseline around the peak in
the range of 20 times the width at half-height of the peak.
1546 O‹cial Monographs for Part II Supplement I, JP XIV

System repeatability: When the test is repeated 6 times Description Burdock Fruit is slightly curved, long obovate
with 20 mL of the standard solution under the above operat- achene, 5 to 7 mm in length, 2.0 to 3.2 mm in width, 0.8 to
ing conditions, the relative standard deviation of the peak 1.5 mm in thickness; externally grayish brown to brown,
area of aristolochic acid I is not more than 5.0z. with black spots; hollow about 1 mm in diameter at one
broad end; ‰at, indistinct, longitudinal ridge at the other
narrow end. 100 fruits weighing 1.0 to 1.5 g.
Add the following: Practically odorless; taste, bitter and oily.
Under a microscope, transverse section reveals an exocarp
Asparagus Tuber of single-layered epidermal tissue, mesocarp of slightly
scleriˆed parenchyma, and endocarp of a single layer of
Asparagi Tuber stone cells; seed coat composed of radially elongated, scleri-
ˆed epidermis, and parenchyma several cells thick; paren-
テンモンドウ
chymatous cells of the mesocarp contain a brown substance;
stone cells of endocarp contain solitary, discrete crystals of
Asparagus Tuber is the tuber of Asparagus cochin- calcium oxalate; cotyledons with starch grains, oil drops,
chinensis Merrill (Liliaceae), from which most of the aleurone grains, and minute crystals of calcium oxalate.
cork layer is removed, usually, after being steamed.
Identiˆcation To 0.5 g of pulverized Burdock Fruit add 20
Description Asparagus Tuber is a fusiform to cylindrical
mL of methanol, shake for 10 minutes, ˆlter, and use ˆltrate
tuber, 5 to 15 cm in length, 0.5 to 2 cm in diameter; external-
as the sample solution. Perform the test with the sample so-
ly light yellow-brown to light brown, translucent and often
lution as directed under the Thin-layer Chromatography.
with longitudinal wrinkles; ‰exible, or hard and easily
Spot 5 mL of the sample solution on a plate of silica gel for
broken in texture; fractured surface, grayish yellow, glossy
thin-layer chromatography, develop the plate with a mixture
and horny.
of acetone, ethyl acetate and water (15:10:1) to a distance of
Odor, characteristic; taste, sweet at ˆrst, followed by a
about 10 cm, and air-dry the plate. Spray evenly dilute sul-
slightly bitter aftertaste.
furic acid on the plate, and heat at 1059 C for 5 minutes: a
Under a microscope, a transverse section of Asparagus
red-purple spot appears at around Rf 0.4.
Tuber reveals stone cells and bundles of them on outer layer
of cortex; mucilaginous cells containing raphides of calcium Loss on drying Not more than 12.0z (6 hours).
oxalate in the parenchyma cells of cortex and central
Total ash Not more than 7.0z.
cylinder; no starch grains.
Acid-insoluble ash Not more than 1.0z.
Identiˆcation To 1 g of coarsely cut Asparagus Tuber add
5 mL of a mixture of 1-butanol and water (40:7), shake for Extract content Dilute ethanol-extract: not less than
30 minutes, ˆlter, and use the ˆltrate as the sample solution. 15.0z.
Perform the test with the sample solution as directed under
the Thin-layer Chromatography. Spot 10 mL of the sample
solution on a plate of silica gel for thin-layer chro- Capsicum
matography, develop the plate with a mixture of 1-butanol,
water and acetic acid (100) (10:6:3) to a distance of about 10 トウガラシ
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
on the plate, and heat at 1059 C for 2 minutes: the spot of a Delete the Extract content.
red-brown at ˆrst then changes to brown color appears at
around Rf 0.4.

Loss on drying Not more than 18.0z (6 hours).


Powdered Capsicum
Total ash Not more than 3.0z.
トウガラシ末

Add the following: Delete the Extract content.

Burdock Fruit
Arctii Fructus
ゴボウシ

Burdock Fruit is the fruit of Arctium lappa Linn áe


(Compositae).
Supplement I, JP XIV O‹cial Monographs for Part II 1547

Add the following: Cinnamon Bark


Chrysanthemum Flower ケイヒ

Chrysanthemi Flos Change the Total ash to read:


キクカ Total ash Not more than 6.0z.

Chrysanthemum Flower is the capitulum of 1)


Chrysanthemum morifolium Ramatulle or 2) Powdered Cinnamon Bark
Chrysanthemum indicum Linn áe (Compositae).
ケイヒ末
Description 1) Chrysanthemum Flower is capitulum, 15
to 40 mm in diameter; involucre consisting of 3 to 4 rows of
Change the Total ash to read:
involucral scales; the outer involucral scale linear to lanceo-
late, inner involucral scale narrow ovate to ovate; ligulate Total ash Not more than 6.0z.
‰owers are numerous, white to yellow; tubular ‰owers in
small number, light yellow-brown; tubular ‰owers oc-
casionally degenerate; outer surface of involucre green- Add the following:
brown to brown; light in texture and easy to break.
Odor, characteristic; taste, slightly bitter. Clematis Root
2) Chrysanthemum Flower is capitulum, 3 to 10 mm in
diameter; involucre consisting of 3 to 5 rows of involucral Clematidis Radix
scales; the outer involucral scale linear to lanceolatae, inner
イレイセン
involucral scale narrow ovate to ovate; ligulate ‰ower is sin-
gle, yellow to light yellow-brown; tubular ‰owers in
Clematis Root is the root and rhizome of Clematis
numerous, light yellow-brown; outer surface of involucre
chinensis Osbeck, Clematis manshurica Ruprecht, or
yellow-brown to brown; light in texture and easy to break.
Clematis hexapetala Pallas (Ranunculaceae).
Odor, characteristic; taste, slightly bitter.
Description Clematis Root consists of short rhizome and
Identiˆcation To 1 g of pulverized Chrysanthemum Flow-
numerous slender roots. The root, 10 to 20 cm in length, 1 to
er add 20 mL of methanol, shake for 10 minutes, and ˆlter.
2 mm in diameter, externally brown to blackish brown, with
Evaporate the ˆltrate to dryness, dissolve the residue in 1 mL
ˆne longitudinal wrinkles, brittle. The cortex easily separa-
of methanol, and use this as the sample solution. Separately,
ble from central cylinder; root, grayish white to light brown
dissolve 1 mg of luteolin for thin-layer chromatography in 1
in the transverse section, light grayish yellow to yellow in the
mL of methanol, and use this as the standard solution. Per-
central cylinder; under a magnifying glass, central cylinder
form the test with these solutions as directed under the Thin-
almost round, slight 2 to 4 sinuses on xylem. The rhizome, 2
layer Chromatography. Spot 10 mL each of the sample solu-
to 4 cm in length, 5 to 20 mm in diameter, externally light
tion and the standard solution on a plate of silica gel for
grayish brown to grayish brown; cortex peeled oŠ and
thin-layer chromatography, develop the plate with a mixture
ˆbrous, often with rising node; apex having the residue of
of ethyl acetate, 2-butanone, water and formic acid
ligniˆed stem.
(25:3:1:1) to a distance of about 10 cm, and air-dry the plate.
Odor, slight; practically tasteless.
Spray evenly iron (III) chloride-methanol TS on the plate:
Under a microscope, transverse section of root reveals a
one of several spots obtained from the sample solution has
uni-layered epidermis in the outermost layer; with exodermis
the same color tone and the same Rf value with the dark
lying just inside of the epidermis; cortex and stele divided by
green spot obtained from the standard solution.
endodermis; cortex composed of parenchymatous tissue;
Loss on drying Not more than 15.0z (6 hours). xylem with 2 – 4 small concavities where phloem is present;
parenchymatous cells contain both simple and 2- to 8-com-
Total ash Not more than 8.5z.
pound starch grains.
Acid-insoluble ash Not more than 1.0z.
Identiˆcation (1) To 0.5 g of pulverized Clematis Root
Extract content Dilute ethanol-extract: not less than add 10 mL of water, and boil for 2 to 3 minutes. After cool-
33.0z. ing, shake vigorously: a lasting ˆne foam appears.
(2) To 0.5 g of pulverized Clematis Root add 3 mL of
acetic anhydride, warm on a water bath for 2 minutes, and
ˆlter. To the ˆltrate add 1 mL of sulfuric acid gently: a
brown color appears at the zone of contact.

Loss on drying Not more than 13.0z (6 hours).


1548 O‹cial Monographs for Part II Supplement I, JP XIV

Total ash Not more than 8.5z. standard solution on a plate of silica gel for thin-layer chro-
matography. Develop the chromatogram with a mixture of
Acid-insoluble ash Not more than 3.0z.
1-butanol, water and acetic acid (100) (7:2:1) to a distance of
Extract content Dilute ethanol-extract: not less than about 10 cm, and air-dry the plate. Examine under ultravio-
15.0z. let light (main wavelength: 365 nm): one spot among the
spots from the sample solution and a spot from the standard
solution with yellow to yellow-green ‰uorescence show the
Cod Liver Oil same color tone and the same Rf value.

肝油
Add the following:
Change the Description to read:
Description Cod Liver Oil is a yellow to orange oily liquid. Eucommia Bark
It has a characteristic, slightly ˆshy odor and a mild taste.
It is miscible with chloroform.
Eucommiae Cortex
It is slightly soluble in ethanol (95), and practically insolu- トチュウ
ble in water.
It is decomposed by air or by light. Eucommia Bark is the bark of Eucommia ulmoides
Oliver (Eucommiaceae).
Description Eucommia Bark is a semi-tubular or plate-like
Coptis Rhizome bark, 2 to 6 mm in thickness; externally pale grayish brown
オウレン to grayish brown, and rough in texture, sometimes reddish-
brown due to the cork layer falling oŠ; internally dark vio-
Change the Identiˆcation (2) to read: let, smooth and covered with a linear pattern that runs lon-
gitudinally, silk-like threads of gutta-percha (a thermoplas-
Identiˆcation tic rubber-like substance) appearing when broken.
(2) To 0.5 g of pulverized Coptis Rhizome add 20 mL of Odor faint but distinctive; taste slightly sweet.
methanol, shake for 2 minutes, ˆlter, and use the ˆltrate as Under a microscope, transverse section reveals paren-
the sample solution. Separately, dissolve 1 mg of Berberine chymatous cells containing gutta-percha; phloem with stone-
Chloride Reference Standard in 1 mL of methanol, and use cell and ˆber layers; rays in rows of 2 – 3 cells; calcium oxa-
this solution as the standard solution. Perform the test with late crystals absent.
these solutions as directed under the Thin-layer Chro-
matography. Spot 5 mL each of the sample solution and the Identiˆcation Put 1 g of pulverized Eucommia Bark in a
standard solution on a plate of silica gel for thin-layer chro- glass-stoppered centrifuge tube, add 10 mL of water and
matography. Develop the chromatogram with a mixture of 20 mL of diethyl ether, shake for 15 minutes, and centrifuge.
1-butanol, water and acetic acid (100) (7:2:1) to a distance of Take the diethyl ether layer so obtained, evaporate the
about 10 cm, and air-dry the plate. Examine under ultravio- diethyl ether on a water bath, and add 1 mL of ethanol
let light (main wavelength: 365 nm): one spot among the (99.5) to the residue: colloidal substances appear.
spots from the sample solution and a spot from the standard Loss on drying Not more than 12.0z (6 hours).
solution with yellow to yellow-green ‰uorescence show the
same color tone and the same Rf value. Total ash Not more than 8.0z.

Acid-insoluble ash Not more than 5.0z.

Powdered Coptis Rhizome Extract content Dilute ethanol-extract: not less than 7.0z.

オウレン末
Evodia Fruit
Change the Identiˆcation (2) to read:
ゴシュユ
Identiˆcation
(2) To 0.5 g of Powdered Coptis Rhizome add 20 mL of Change the origin/limits of content to read:
methanol, shake for 2 minutes, ˆlter, and use the ˆltrate as
the sample solution. Separately, dissolve 1 mg of Berberine Evodia Fruit is the fruit of Evodia rutaecarpa Ben-
Chloride Reference Standard in 1 mL of methanol, and use tham or Evodia o‹cinalis Dode or Evodia bodinieri
this solution as the standard solution. Perform the test with Dode (Rutaceae).
these solutions as directed under the Thin-layer Chro-
matography. Spot 5 mL each of the sample solution and the
Supplement I, JP XIV O‹cial Monographs for Part II 1549

Add the following: Gardenia Fruit


Fritillaria Bulb サンシシ

Fritillariae Bulbus Change the origin/limits of content to read:


バイモ
Gardenia Fruit is the fruit of Gardenia jasminoides
Ellis (Rubiaceae).
Fritillaria Bulb is the bulb of Fritillaria verticillata
It contains not less than 3.0z of geniposide
Willdenow var. thunbergii Baker (Liliaceae).
(C17H24O10: 388.37), calculated on the basis of dried
Description Fritillaria Bulb is a depressed spherical bulb, 2 material.
to 3 cm in diameter, 1 to 2 cm in height, consisting of 2
thickened scaly leaves often separated; externally and inter- Change the Identiˆcation (1) to read:
nally white to light yellow-brown in color; inside base is in a
Identiˆcation (1) To 1.0 g of pulverized Gardenia Fruit,
slightly dark color; the bulb sprinkled with lime before
previously dried in a desiccator (silica gel) for 24 hours, add
drying is dusted with white powder; fractured surface, white
100 mL of hot water, warm the mixture between 609 C and
in color and powdery.
709C for 30 minutes with frequent shaking, and ˆlter after
Odor, slight and characteristic; taste, bitter.
cooling. To 1.0 mL of the ˆltrate add water to make 10 mL:
Under the microscope, a transverse section reveals the
the color of the resulting solution is yellow and is not lighter
outermost layer (epidermis) to be composed of a single layer
than that of the following control solution.
of cells; numerous vascular bundles scattered throughout the
Control solution: Dissolve 9.8 mg of carbazochrome sodi-
parenchyma inside of the epidermis; parenchyma ˆlled with
um sulfonate in water to make exactly 10 mL. Pipet 1 mL of
starch grains; starch grains are mainly simple (rarely 2 – 3
this solution, and add water to make exactly 50 mL.
composite), 5 – 50 mm in diameter, narrowly ovate to ovate
or triangular to obovate, stratiform ˆgure obvious; epider-
Add the following next to Identiˆcation:
mal cells and parenchymatous cells near the vessels contain
solitary crystals of calcium oxalate. Loss on drying Not more than 13.0z.

Identiˆcation Put 2 g of pulverized Fritillaria Bulb in a


Add the following next to Total ash:
glass-stoppered centrifuge tube, add 10 mL of ammonia TS
and 20 mL of a mixture of ethyl acetate and diethyl ether Component determination Weigh accurately about 0.5 g
(1:1), shake for 20 minutes, and centrifuge. Take the upper of pulverized Gardenia Fruit, transfer into a glass-stoppered
layer, add 20 g of anhydrous sodium sulfate to the layer, centrifuge tube, add 40 mL of diluted methanol (1 in 2),
shake, and ˆlter. Evaporate the ˆltrate to dryness, dissolve shake for 15 minutes, centrifuge, and take the supernatant
the residue in 1 mL of ethanol (99.5), and use this solution as liquid. To the residue add 40 mL of diluted methanol (1 in
the sample solution. Perform the test with the sample solu- 2), and perform as the same as above. Combine the extracts
tion as directed under the Thin-layer Chromatography. Spot so obtained, and add diluted methanol (1 in 2) to make ex-
10 mL of the sample solution on a plate of silica gel for thin- actly 100 mL. Pipet 5 mL of the solution, add methanol to
layer chromatography, develop the plate with a mixture of make exactly 20 mL, use this solution as the sample solu-
ethyl acetate, methanol and ammonia solution (28) (17:2:1) tion. Separately, weigh accurately about 10 mg of genipo-
to a distance of about 10 cm, and air-dry the plate. Spray side for component determination, previously dried in a
evenly DragendorŠ's TS for spraying on the plate: two spots desiccator (in vacuum, phosphorus (V) oxide) for 24 hours,
of a yellow-red color appear at around Rf 0.4 and at around and dissolve in methanol to make exactly 100 mL. Pipet 5
Rf 0.6. mL of the solution, add methanol to make exactly 10 mL,
and use this solution as the standard solution. Perform the
Loss on drying Not more than 16.0z (6 hours).
test with exactly 10 mL each of the sample solution and the
Total ash Not more than 6.5z. standard solution as directed under the Liquid Chro-
matography according to the following conditions, and
Acid-insoluble ash Not more than 1.0z.
measure the peak areas of geniposide, AT and AS, of both
Extract content Dilute ethanol-extract: not less than 8.0z. solutions.

AT
Amount (mg) of geniposide = WS × ×2
AS

WS: Amount (mg) of geniposide for component determi-


nation

Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
1550 O‹cial Monographs for Part II Supplement I, JP XIV

length: 240 nm). Total ash Not more than 4.0z.


Column: A stainless steel column 6 mm in inside diameter
Extract content Dilute ethanol-soluble extract: not less
and 15 cm in length, packed with octadecylsilanized silica gel
than 16.0z.
for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
309C.
Mobile phase: A mixture of water and acetonitrile (22:3). Glycyrrhiza
Flow rate: Adjust the ‰ow rate so that the retention time
カンゾウ
of geniposide is about 15 minutes.
System suitability—
Change the Identiˆcation to read:
System performance: Dissolve 1 mg each of geniposide
for component determination and caŠeine in methanol to Identiˆcation To 2 g of pulverized Glycyrrhiza add 10 mL
make 15 mL. When the procedure is run with 10 mL of this of a mixture of ethanol (95) and water (7:3), heat by shaking
solution under the above operating conditions, caŠeine and on a water bath for 5 minutes, cool, ˆlter, and use the ˆltrate
geniposide are eluted in this order with the resolution be- as the sample solution. Separately, dissolve 5 mg of Glycyr-
tween these peaks being not less than 3.5. rhizinic Acid Reference Standard in 1 mL of a mixture of
System repeatability: When the test is repeated 6 times ethanol (95) and water (7:3), and use this solution as the
with 10 mL of the standard solution under the above operat- standard solution. Perform the test with these solutions as
ing conditions, the relative standard deviation of the peak directed under the Thin-layer Chromatography. Spot 2 mL
area of geniposide is not more than 1.5z. each of the sample solution and the standard solution on a
plate of silica gel with ‰uorescent indicator for thin-layer
chromatography. Develop the plate with a mixture of 1-
Add the following: butanol, water and acetic acid (100) (7:2:1) to a distance of
about 10 cm, and air-dry the plate. Examine under ultravio-
Gastrodia Tuber let light (main wavelength: 254 nm): one spot among the
spots from the sample solution and a dark purple spot from
Gastrodiae Tuber the standard solution show the same color tone and the same
Rf value.
テンマ

Gastrodia Tuber is the steamed tuber of Gastrodia


elata Blume (Orchidaceae).
Powdered Glycyrrhiza
Description Gastrodia Tuber is an irregularly curved and カンゾウ末
‰attened cylindrical to ‰attened fusiform tuber, 5 to 15 cm
in length, 2 to 5 cm in diameter, 1 to 2 cm in thickness; exter- Change the Identiˆcation to read:
nally light yellow-brown to light yellowish white; with ring Identiˆcation To 2 g of Powdered Glycyrrhiza add 10 mL
nodes, and irregular longitudinal wrinkles; hard in texture; of a mixture of ethanol (95) and water (7:3), heat by shaking
fractured surface, dark brown to yellow-brown in color, on a water bath for 5 minutes, cool, ˆlter, and use the ˆltrate
with luster, horny and gluey. as the sample solution. Separately, dissolve 5 mg of Glycyr-
Odor, characteristic; practically tasteless. rhizinic Acid Reference Standard in 1 mL of a mixture of
Under a microscope, a transverse section reveals paren- ethanol (95) and water (7:3), and use this solution as the
chyma cells containing needle raphides of calcium oxalate; standard solution. Perform the with these solutions test as
starch grain absent. directed under the Thin-layer Chromatography. Spot 2 mL
Identiˆcation To 1 g of pulverized Gastrodia Tuber add 5 each of the sample solution and the standard solution on a
mL of methanol, shake for 15 minutes, and ˆlter. Evaporate plate of silica gel with ‰uorescent indicator for thin-layer
the ˆltrate to dryness, dissolve the residue in 1 mL of chromatography. Develop the plate with a mixture of 1-
methanol, and use this solution as the sample solution. Per- butanol, water and acetic acid (100) (7:2:1) to a distance of
form the test with the sample solution as directed under the about 10 cm, and air-dry the plate. Examine under ultravio-
Thin-layer Chromatography. Spot 10 mL of the sample solu- let light (main wavelength: 254 nm): one spot among the
tion on a plate of silica gel for thin-layer chromatography, spots from the sample solution and a dark purple spot from
develop the plate with a mixture of ethyl acetate, methanol the standard solution show the same color tone and the same
and water (8:2:1) to a distance of about 10 cm, and air-dry Rf value.
the plate. Spray evenly dilute sulfuric acid on the plate, and
heat at 1059 C for 1 minutes: a red-purple spot appears at
around Rf 0.4.

Loss on drying Not more than 16.0z (6 hours).


Supplement I, JP XIV O‹cial Monographs for Part II 1551

Add the following: mL of acetic anhydride, place 0.5 mL of this solution in a


test tube, and add carefully 0.5 mL of sulfuric acid to make
Hemp Fruit two layers: a red-brown color develops at the zone of con-
tact, and the upper layer acquires a blue-green to blue-purple
Cannabis Fructus color.

マシニン

Hemp Fruit is the fruit of Cannabis sativa Linn áe


Powdered Japanese Gentian
(Cannabidaceae). リュウタン末
Description Hemp Fruit is a slightly compressed void fruit,
4 to 5 mm in length, 3 to 4 mm in diameter; externally Change the Identiˆcation to read:
grayish green to grayish brown; pointed at one end, a scar of Identiˆcation To 0.5 g of Powdered Japanese Gentian add
gynophore at the other end, and crest lines on both sides; 10 mL of methanol, shake for 20 minutes, ˆlter, and use the
outer surface lustrous with white mesh-like pattern; slightly ˆltrate as the sample solution. Separately, dissolve 1 mg of
hard pericarp; seed, slightly green in color and internally has gentiopicroside for thin-layer chromatography in 1 mL of
grayish white albumen; 100 fruits weighing 1.6 to 2.7 g. methanol, and use this solution as the standard solution.
Practically odorless, aromatic on chewing; taste, mild and Perform the test with these solutions as directed under the
oily. Thin-layer Chromatography. Spot 10 mL each of the sample
Under a microscope, a transverse section reveals the ex- solution and the standard solution on a plate of silica gel
ocarp to be a single-layered epidermis; mesocarp composed with ‰uorescent indicator for thin-layer chromatography.
of parenchyma, a pigment cell layer and rows of short, small Develop the plate with a mixture of ethyl acetate, ethanol
cells; endocarp made up of a layer of radially elongated (99.5) and water (8:2:1) to a distance of about 10 cm, and
stone cells; seed coat comprises a tubular cell layer and air-dry the plate. Examine under ultraviolet light (main
spongy tissue. Inside of the seed; exosperm consists of one wavelength: 254 nm): one spot among the spots from the
layer of parenchymatous cells, endosperm of one to several sample solution and a dark purple spot from the standard
layers of parenchymatous cells; most of the embryo com- solution show the same color tone and the same Rf value.
posed of parenchyma, vascular bundles occurring in the cen-
ter of hypocotyls and cotyledons; embryo parenchyma con-
tains aleurone grains and oil drops. Add the following:
Identiˆcation To 0.3 g of pulverized Hemp Fruit add 3 mL
of methanol, shake for 10 minutes, centrifuge, and use the Jujube Seed
supernatant liquid as the sample solution. Perform the test
with the sample solution as directed under the Thin-layer Zizyphi Semen
Chromatography. Spot 5 mL of the sample solution on a
サンソウニン
plate of silica gel for thin-layer chromatography, develop the
plate with a mixture of hexane and ethyl acetate (9:2) to a
Jujube Seed is the seed of Zizyphus jujuba Miller
distance of about 10 cm, and air-dry the plate. Spray evenly
var. spinosa (Bunge) Hu ex H. F. Chou (Rham-
vanillin-sulfuric acid TS on the plate, and heat at 1059 C for
naceae).
5 minutes: a dark blue-purple spot appears at around Rf 0.6.
Description Jujube Seed is a compressed ovate to orbicu-
Purity Bract—Hemp Fruit does not contain bract.
lar, lenticular seed, 5 to 9 mm in lengh, 4 to 6 mm in width, 2
Loss on drying Not more than 9.0z (6 hours). to 3 mm in thickness, externally brown to dark red-brown,
glossy; hilum at one end, charaza at the other end; seed coat
Total ash Not more than 7.0z.
sightly ˆexible, covering, milky white endosperm and light
Acid-insoluble ash Not more than 2.0z. yellow embryo. 100 seeds weighing 3.0 to 4.5 g.
Odor, slightly oily; taste, mild and slightly oily.
Under a microscope, transverse section reveals seed coat
Imperata Rhizome composed of an upper epidermis, parenchyma and lower
epidermis; upper epidermal cells scleriˆed and elongated in
ボウコン radial direction; lower epidermis covered with cuticle; en-
dosperm composed of parenchyma, containing aggregated
Change the Identiˆcation to read: crystals of calcium oxalate, aleurone grains and starch
grains; cotyledons composed of parenchyma that contains
Identiˆcation To 1 g of pulverized Imperata Rhizome add
aleurone grains, starch grains and oil drops.
20 mL of hexane, allow the mixture to stand for 30 minutes
with occasional shaking, and ˆlter. Evaporate the hexane of Identiˆcation To 2 g of pulverized Jujube Seed add 10 mL
the ˆltrate under reduced pressure, dissolve the residue in 5 of methanol, and heat under a re‰ux condenser for 10
1552 O‹cial Monographs for Part II Supplement I, JP XIV

minutes. After cooling, ˆlter, and use the ˆltrate as the sam- air-dry the plate. Spray evenly dilute sulfuric acid on the
ple solution. Perform the test with the sample solution as plate, and heat at 1059C for 10 minutes: a red-purple prin-
directed under the Thin-layer Chromatography. Spot 10 mL cipal spot appears at around Rf 0.5.
of the sample solution on a plate of silica gel with ‰uorescent
Loss on drying Not more than 15.0z (6 hours).
indicator for thin-layer chromatography, develop the plate
with a mixture of acetone, ethyl acetate, water and acetic Total ash Not more than 10.0z.
acid (100) (10:10:3:1) to a distance of about 10 cm, and air-
Extract content Dilute ethanol-soluble extract: not less
dry the plate. Examine under ultraviolet light (main
than 16.0z.
wavelength: 254 nm): a purple spot appears at around Rf
0.3, which shows a yellow-green to grayish green color after
spraying 1-naphthol-sulfuric acid TS on the plate and heat-
ing at 1059 C for 5 minutes. Magnolia Bark
Purity Foreign substance—Jujube Seed contains not more コウボク
than 1.0z of the endocarp and other foreign substances.
Change the Extract content to read:
Loss on drying Not more than 11.0z (6 hours).
Extract content Dilute ethanol-soluble extract: not less
Total ash Not more than 5.0z.
than 11.0z.
Extract content Dilute ethanol-extract: not less than 9.0z.

Powdered Magnolia Bark


Add the following:
コウボク末
Loquat Leaf
Change the Extract content to read:
Eriobotryae Folium Extract content Dilute ethanol-soluble extract: not less
ビワヨウ than 11.0z.

Loquat Leaf is the leaf of Eriobotrya japonica Lin-


dley (Rosaceae). Add the following:

Description Loquat Leaf is an oblong to wide lanceolate


leaf, 12 to 30 cm in length, 4 to 9 cm in width; pointed at the
Magnolia Flower
apex and wedge-shaped at the base; roughly serrate leaf with Magnoliae Flos
short petiole; occasionally being cut into strips 5 to 10 mm in
shorter diameter and several cm in longer diameter; upper シンイ
surface green to green-brown in color, lower surface light
green-brown with light brown woolly hairs; vein, light yel- Magnolia Flower is the ‰ower bud of Magnolia
low-brown in color, raised out on the lower surface of the salicifolia Maximowicz, Magnolia kobus De Candolle,
leaf. Magnolia biondii Pampanini, Magnolia sprengeri
Odor, slight; practically tasteless. Pampanini or Magnolia denudata Desrousseaux
Under a microscope, a transverse section of Loquat Leaf (Magnoliaceae).
reveals thick cuticle on both surfaces; palisade tissue, mostly Description Magnolia Flower is a fusiform ‰ower bud, 15
4 to 5 layers with several large cells without chloroplast; at to 45 mm in length, 6 to 20 mm in diameter of central part;
main vein, ring of collateral bundle partly cut by intruding often having ligneous peduncles on base; usually 3 bracts,
fundamental tissue at xylem side, and group of ˆber attach- externally with sparse hairs; brown to dark brown; or with
ing to phloem; solitary and clustered crystals of calcium oxa- dense hairs, grayish white to light yellow-brown, and the in-
late in mesophyll; woolly hair, unicellular and curved, about ner surface smooth and dark brown in color; interior
25 mm in thickness, and up to 1.5 mm in length. perianth of 9 pieces or 12 pieces, same size or exterior three
Identiˆcation To 0.3 g of pulverized Loquat Leaf add 10 pieces are smaller; 50 to 100 stamens and numerous pistils.
mL of methanol, warm on a water bath for 5 minutes with Brittle in texture.
occasional shaking, cool, ˆlter, and use the ˆltrate as the Odor, characteristic; taste, acrid and slightly bitter.
sample solution. Perform the test with this solution as Identiˆcation To 1 g of pulverized Magnolia Flower add
directed under the Thin-layer Chromatography. Spot 5 mL 10 mL of methanol, shake for 15 minutes, ˆlter, and use the
of the sample solution on a plate of silica gel for thin-layer ˆltrate as the sample solution. Perform the test with the
chromatography. Develop the plate with a mixture of sample solution as directed under the Thin-layer Chro-
acetonitrile and water (3:2) to a distance of about 10 cm, and matography. Spot 20 mL of the sample solution on a plate of
Supplement I, JP XIV O‹cial Monographs for Part II 1553

silica gel for thin-layer chromatography, develop the plate Change the Assay to read:
with a mixture of ethyl acetate, acetone, water and formic
Assay (1) Morphine hydrochloride—Pipet 2 mL of Mor-
acid (5:3:1:1) to a distance of about 10 cm, and air-dry the
phine and Atropine Injection, add exactly 10 mL of the in-
plate. Spray evenly DragendorŠ's TS for spraying on the
ternal standard solution, then add water to make 50 mL,
plate: a yellow-red spot appears at around Rf 0.3.
and use this solution as the sample solution. Separately,
Loss on drying Not more than 14.0z (6 hours). weigh accurately about 25 mg of morphine hydrochloride
for assay, add exactly 10 mL of the internal standard solu-
Total ash Not more than 5.5z.
tion to dissolve, then add water to make 50 mL, and use this
Acid-insoluble ash Not more than 1.5z. solution as the standard solution. Perform the test with 20
mL of the sample solution and the standard solution as
Extract content Dilute ethanol-extract: not less than
directed under the Liquid Chromatography according to the
13.0z.
following conditions, and calculate the ratios, QT and QS, of
Essential oil content Perform the test with 50.0 g of pul- the peak area of morphine to that of the internal standard.
verized Magnolia Flower as directed in the Essential oil con-
Amount (mg) of morphine hydrochloride
tent under the Crude Drugs Test: the volume of essential oil
(C17H19NO3.HCl.3H2O)
is not less than 0.5 mL.
Q
= WS × T × 1.1679
QS

Morphine and Atropine Injection WS: Amount (mg) of morphine hydrochloride for assay,
calculated on the anhydrous basis
モルヒネアトロピン注射液
Internal standard solution—A solution of Etilefrine Hydro-
chloride (1 in 500).
Change the Description to read:
Operating conditions—
Description Morphine and Atropine Injection is a clear, Detector: An ultraviolet absorption photometer (wave-
colorless liquid. length: 285 nm).
It is gradually colored by light. Column: A stainless steel column 4.6 mm in inside di-
pH: 2.5 – 5.0 ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle
Change the Identiˆcation to read: diameter).
Column temperature: A constant temperature of about
Identiˆcation To 2 mL of Morphine and Atropine Injec-
409C.
tion add 2 mL of ammonia TS, and extract with 10 mL of
Mobile phase: Dissolve 1.0 g of sodium lauryl sulfate in
diethyl ether. Filter the extract with a ˆlter paper, evaporate
500 mL of diluted phosphoric acid (1 in 1000), and adjust
the ˆltrate on a water bath to dryness, dissolve the residue in
the pH with sodium hydroxide TS to 3.0. To 240 mL of this
1 mL of ethanol (99.5), and use this solution as the sample
solution add 70 mL of tetrahydrofuran, and mix.
solution. Separately, dissolve 0.1 g of Morphine Hydrochlo-
Flow rate: Adjust the ‰ow rate so that the retention time
ride in 10 mL of water, perform with 2 mL of this solution
of morphine is about 10 minutes.
the same procedure as used for preparation of the sample so-
System suitability—
lution, and use the solution so obtained as the standard solu-
System performance: When the procedure is run with 20
tion (1). Separately, dissolve 3 mg of Atropine Sulfate Ref-
mL of the standard solution under the above operating con-
erence Standard in 10 mL of water, perform with 2 mL of
ditions, morphine and the internal standard are eluted in this
this solution the same procedure as used for preparation of
order with the resolution between these peaks being not less
the sample solution, and use the solution so obtained as the
than 3.
standard solution (2). Perform the test with these solutions
System repeatability: When the test is repeated 6 times
as directed under the Thin-layer Chromatography. Spot 10
with 20 mL of the standard solution under the above operat-
mL each of the sample solution, the standard solution (1) and
ing conditions, the relative standard deviation of the ratios
the standard solution (2) on a plate of silica gel for thin-layer
of the peak area of morphine to that of the internal standard
chromatography. Develop the plate with a mixture of
is not more than 1.0z.
methanol and ammonia solution (28) (200:3) to a distance of
(2) Atropine sulfate—Pipet 2 mL of Morphine and
about 10 cm, and air-dry the plate. Spray evenly Dragendor-
Atropine Injection, add exactly 2 mL of the internal stan-
Š's TS on the plate: the two spots obtained from the sample
dard solution, and use this solution as the sample solution.
solution show the same color tone and the same Rf value
Separately, weigh accurately about 15 mg of Atropine Sul-
with either spot of orange color obtained from the standard
fate Reference Standard (separately determine its loss on
solution (1) or the standard solution (2) (morphine and atro-
drying in the same manner as directed under Atropine Sul-
pine).
fate), and dissolve in water to make exactly 50 mL. Pipet
2 mL of this solution, add exactly 2 mL of the internal stan-
dard solution, and use this solution as the standard solution.
1554 O‹cial Monographs for Part II Supplement I, JP XIV

Perform the test with 20 mL each of the sample solution and Add the following:
the standard solution as directed under the Liquid Chro-
matography according to the following conditions, and cal- Notopterygium Rhizome
culate the ratios, QT and QS, of the peak areas of atropine to
that of the internal standard. Notopterygii Rhizoma
Amount (mg) of atropine sulfate [(C17H23NO3)2.H2SO4.H2O] キョウカツ
Q 1
= WS × T × × 1.027
QS 25 Notopterygium Rhizome is the rhizome and root of
WS: Amount (mg) of Atropine Sulfate Reference Stand- Notopterygium incisum Ting ex H. T. Chang or
ard, calculated on the dried basis Notopterygium forbesii Boissieu (Umbelliferae ).

Internal standard solution—A solution of Etilefrine Hydro- Description Notopterygium Rhizome is slightly curved,
chloride (1 in 12,500). cylindrical to conical, 3 to 10 cm in length, 5 to 20 mm in di-
Operating conditions— ameter; rhizome occasionally branched; externally yellow-
Column, column temperature, and mobile phase: Proceed brown to dark brown. The rhizome with nearly orbicular,
as directed in the operating conditions in the Assay (1). hollowed stem scars at the apex, sometimes having short
Detector: An ultraviolet absorption photometer (wave- residue of stem; externally node rising, internode short; root
length: 225 nm). scars in warty processes on the node; externally root has
Flow rate: Adjust the ‰ow rate so that the retention time coarse longitudinal wrinkles and lateral root scars in warty
of morphine is about 7 minutes. processes; light and slightly brittle in texture, easy to break.
System suitability— The transverse section of the rhizome reveals numerous radi-
System performance: When the procedure is run with al cracks; cortex yellow-brown to brown; xylem light yellow
20 mL of the sample solution under the above operating con- to light grayish yellow; pith grayish white to light brown.
ditions, morphine, the internal standard and atropine are Under a magnifying glass, the rhizome reveals brown, ˆne
eluted in this order, and the resolution between morphine points of resin canals in the cortex and pith.
and the internal standard is not less than 3. Odor, characteristic; taste, slightly acid at ˆrst, followed
System repeatability: When the test is repeated 6 times by a slightly pungent and slightly numbing aftertaste.
with 20 mL of the standard solution under the above operat- Under a microscope, transverse section shows the outer-
ing conditions, the relative standard deviation of the ratios most layer to be composed of a cork layer several to a dozen
of the peak area of atropine to that of the internal standard or so cells thick; collenchyma just inside of the cork layer;
is not more than 1.0z. oil canals scattered in cortex, large ones more than 300 mm in
diameter; intercellular space occurring in radial direction in
cortex; oil canals scattered in pith, large ones more than
500 mm in diameter; parenchymatous cells contain simple
Mulberry Bark and 2- to 3-compound starch grains.
ソウハクヒ Identiˆcation To 0.3 g of pulverized Notopterygium Rhi-
zome add 3 mL of hexane in a glass-stoppered centrifuge
Change the Identiˆcation to read: tube, shake for 10 minutes, centrifuge, and use the super-
Identiˆcation Heat 1 g of pulverized Mulberry Bark with natant liquid as the sample solution. Perform the test with
20 mL of hexane under a re‰ux condenser on a water bath the sample solution as directed under the Thin-layer Chro-
for 15 minutes, and ˆlter. Evaporate the hexane of the matography. Spot 10 mL of the sample solution on a plate of
ˆltrate under reduced pressure, dissolve the residue in 10 mL octadecylsilanized silica gel with ‰uorescent indicator for
of acetic anhydride, place 0.5 mL of the solution in a test thin-layer chromatography, develop the plate with a mixture
tube, and add carefully 0.5 mL of sulfuric acid to make two of methanol and water (9:1) to a distance of about 10 cm,
layers: a red-brown color develops at the zone of contact. and air-dry the plate. Examine under ultraviolet light (main
wavelength: 365 nm): a bluish white ‰uorescent spot appears
at around Rf 0.5, which shows a dark purple color under
ultraviolet light (main wavelength: 254 nm).

Loss on drying Not more than 13.0z (6 hours).

Total ash Not more than 6.5z.

Acid-insoluble ash Not more than 1.5z.

Extract content Dilute ethanol-extract: not less than


20.0z.
Supplement I, JP XIV O‹cial Monographs for Part II 1555

Opium Alkaloids Hydrochlorides evenly 4-methoxybenzaldehyde-sulfuric acid TS upon the


plate, and heat at 1059C for 5 minutes: one spot among the
塩酸アヘンアルカロイド spots from the sample solution and the purple-red spot from
the standard solution show the same color tone and the same
Change the Description to read: Rf value.
Description Opium Alkaloids Hydrochlorides occur as a
white to light brown powder.
Change the Assay to read:
It is soluble in water, and slightly soluble in ethanol (95). Assay Weigh accurately about 0.5 g of pulverized Peony
It is colored by light. Root, add 50 mL of diluted methanol (1 in 2), heat under a
re‰ux condenser on a water bath for 30 minutes, cool, and
Change the Purity (1) to read: ˆlter. To the residue add 50 mL of diluted methanol (1 in 2),
and proceed in the same manner. Combine the ˆltrates, add
Purity (1) Clarity and color of solution-Dissolve 0.5 g
diluted methanol (1 in 2) to make exactly 100 mL, and use
of Opium Alkaloids Hydrochlorides in 10 mL of water: the
this solution as the sample solution. Separately, weigh ac-
solution is clear, and its absorbance at 420 nm is not more
curately about 10 mg of Paeoni‰orin Reference Standard
than 0.20.
(separately detarmine the water content), dissolve in diluted
methanol (1 in 2) to make exactly 100 mL, and use this solu-
tion as the standard solution. Perform the test with exactly
Powdered Panax Rhizome 10 mL each of the sample solution and the standard solution
チクセツニンジン末 as directed under the Liquid Chromatography according to
the following conditions. Determine the peak areas, AT and
Change the Identiˆcation to read: AS, of paeoni‰orin in each solution.

Identiˆcation Shake 0.5 g of Powdered Panax Rhizome AT


Amount (mg) of paeoni‰orin (C23H28O11) = WS ×
AS
with 10 mL of methanol for 10 minutes, ˆlter, and use the
ˆltrate as the sample solution. Separately, dissolve 2 mg of WS: Amount (mg) of Paeoni‰orin Reference Standard,
chikusetsusaponin IV for thin-layer chromatography in calculated on the anhydrous basis
1 mL of methanol, and use this solution as the standard so-
lution. Perform the test with these solutions as directed un- Operating conditions—
der the Thin-layer Chromatography. Spot 5 mL each of the Detector: An ultraviolet absorption photometer (wave-
sample solution and the standard solution on a plate of silica length: 232 nm).
gel for thin-layer chromatography. Develop the plate with a Column: A stainless steel column 4.6 mm in inside di-
mixture of ethyl acetate, water and formic acid (5:1:1) to a ameter and 15 cm in length, packed with octadecylsilanized
distance of about 10 cm, and air-dry the plate. Spray evenly silica gel for liquid chromatography (5 mm in particle di-
dilute sulfuric acid on the plate, and heat the plate at 1109C ameter).
for 5 minutes: one of several spots obtained from the sample Column temperature: A constant temperature of about
solution shows the same color tone and the same Rf value 209C.
with the purple-red spot from the standard solution. Mobile phase: A mixture of water, acetonitrile and phos-
phoric acid (850:150:1).
Flow rate: Adjust the ‰ow rate so that the retention time
of paeoni‰orin is about 10 minutes.
Peony Root System suitability—
シャクヤク System performance: Dissolve 1 mg each of Paeoni‰orin
Reference Standard and albi‰orin in diluted methanol (1 in
Change the Identiˆcation (2) to read: 2) to make 10 mL. When the procedure is run with 10 mL of
this solution under the above operating conditions, albi‰orin
Identiˆcation and paeoni‰orin are eluted in this order with the resolution
(2) To 2 g of pulverized Peony Root add 10 mL of between these peaks being not less than 2.5.
methanol, warm on a water bath for 5 minutes, cool, ˆlter, System repeatability: When the test is repeated 6 times
and use the ˆltrate as the sample solution. Separately, dis- with the standard solution under the above operating condi-
solve 1 mg of Paeoni‰orin Reference Standard in 1 mL of tions, the relative standard deviation of the peak area of
methanol, and use this solution as the standard solution. paeoni‰orin is not more than 1.5z.
Perform the test with these solutions as directed under the
Thin-layer Chromatography. Spot 10 mL each of the sample
solution and the standard solution on a plate of silica gel for
thin-layer chromatography. Develop the plate with a mix-
ture of acetone, ethyl acetate and acetic acid (100) (10:10:1)
to a distance of about 10 cm, and air-dry the plate. Spray
1556 O‹cial Monographs for Part II Supplement I, JP XIV

Powdered Peony Root directed under the Thin-layer Chromatography. Spot 5 mL


each of the sample solution and the standard solution on a
シャクヤク末 plate of silica gel for thin-layer chromatography. Develop
the plate with a mixture of 1-butanol, water and acetic acid
Change the Identiˆcation (2) to read: (100) (7:2:1) to a distance of about 10 cm, and air-dry the
plate. Examine under ultraviolet light (main wavelength: 365
Identiˆcation
nm): one spot among the spots from the sample solution and
(2) To 2 g of Powdered Peony Root add 10 mL of
a spot with yellow to yellow-green ‰uorescence from the
methanol, warm on a water bath for 5 minutes, cool, ˆlter,
standard solution show the same color tone and the same Rf
and use the ˆltrate as the sample solution. Separately, dis-
value.
solve 1 mg of Paeoni‰orin Reference Standard in 1 mL of
methanol, and use this solution as the standard solution.
Perform the test with these solutions as directed under the
Thin-layer Chromatography. Spot 10 mL each of the sample
Compound Phellodendron Powder
solution and the standard solution on a plate of silica gel for for Cataplasm
thin-layer chromatography. Develop the plate with a mix-
ture of acetone, ethyl acetate and acetic acid (100) (10:10:1) パップ用複方オウバク散
to a distance of about 10 cm, and air-dry the plate. Spray
evenly 4-methoxybenzaldehyde-sulfuric acid TS on the Change the Identiˆcation to read:
plate, and heat at 1059C for 5 minutes: one spot among the
Identiˆcation Shake thoroughly 0.2 g of Compound Phel-
spots from the sample solution and the purple spot from the
lodendron Powder for Cataplasm with 5 mL of methanol,
standard solution show the same color tone and the same Rf
ˆlter, and use the ˆltrate as the sample solution. Dissolve 1
value.
mg of Berberine Chloride Reference Standard in 1 mL of
methanol, and use the solution as the standard solution. Per-
form the test with these solutions as directed under the Thin-
Phellodendron Bark layer Chromatography. Spot 5 mL each of the sample solu-
tion and the standard solution on a plate of silica gel for
オウバク
thin-layer chromatography. Develop the plate with a mix-
ture of 1-butanol, water and acetic acid (100) (7:2:1) to a dis-
Change the Identiˆcation (2) to read:
tance of about 10 cm, air-dry the plate. Examine under
Identiˆcation ultraviolet light (main wavelength: 365 nm): one spot among
(2) Use the ˆltrate obtained in (1) as the sample solution. the spots from the sample solution and a spot with yellow to
Separately, dissolve 1 mg of Berberine Chloride Reference yellow-green ‰uorescence from the standard solution show
Standard in 1 mL of methanol, and use this solution as the the same color tone and the same Rf value (berberine).
standard solution. Perform the test with these solutions as
directed under the Thin-layer Chromatography. Spot 5 mL
each of the sample solution and the standard solution on a Phellodendron, Albumin Tannate
plate of silica gel for thin-layer chromatography. Develop
the plate with a mixture of 1-butanol, water and acetic acid and Bismuth Subnitrate Powder
(100) (7:2:1) to a distance of about 10 cm, and air-dry the
オウバクタンナルビンビスマス散
plate. Examine under ultraviolet light (main wavelength: 365
nm): one spot among the spots from the sample solution and
Change the Identiˆcation (1) to read:
a spot with yellow to yellow-green ‰uorescence from the
standard solution show the same color tone and the same Rf Identiˆcation (1) Shake thoroughly 0.1 g of Phelloden-
value. dron, Albumin Tannate and Bismuth Subnitrate Powder
with 5 mL of methanol, ˆlter, and use the ˆltrate as the sam-
ple solution. Dissolve 1 mg of Berberine Chloride Reference
Powdered Phellodendron Bark Standard in 1 mL of methanol, and use this solution as the
standard solution. Perform the test with these solutions as
オウバク末 directed under the Thin-layer Chromatography. Spot 5 mL
each of the sample solution and the standard solution on a
Change the Identiˆcation (2) to read: plate of silica gel for thin-layer chromatography. Develop
the plate with a mixture of 1-butanol, water and acetic acid
Identiˆcation
(100) (7:2:1) to a distance of about 10 cm, air-dry the plate.
(2) Use the ˆltrate obtained in (1) as the sample solution.
Examine under ultraviolet light (main wavelength: 365 nm):
Separately, dissolve 1 mg of Berberine Chloride Reference
one spot among the spots from the sample solution and a
Standard in 1 mL of methanol, and use this solution as the
spot with yellow to yellow-green ‰uorescence from the stan-
standard solution. Perform the test with these solutions as
dard solution show the same color tone and the same Rf
Supplement I, JP XIV O‹cial Monographs for Part II 1557

value (berberine). 17.0z.

Delete the following Monograph: Pueraria Root


Phenovalin and Magnesium Oxide カッコン

Powder Change the origin W


limits of content to read:
フェノバリンマグネシア散
Pueraria Root is the root of Pueraria lobata Ohwi
(Leguminosae), from which periderm has been re-
moved.
It contains not less than 2.0z of puerarin (C21H20O9:
Add the following:
416.38), calculated on the basis of dried material.
Polygonum Root Change the Identiˆcation to read:
Polygoni Multi‰ori Radix Identiˆcation To 2 g of pulverized Pueraria Root add 10
mL of methanol, shake for 3 minutes, ˆlter, and use the
カシュウ
ˆltrate as the sample solution. Separately, dissolve 1 mg of
Puerarin Reference Standard in 1 mL of methanol, and use
Polygonum Root is the root of Polygonum multi‰o-
this solution as the standard solution. Perform the test with
rum Thunberg (Polygonaceae), often being cut into
these solutions as directed under the Thin-layer Chro-
round slices.
matography. Spot 2 mL each of the sample solution and the
Description Polygonum Root is nearly fusiform, 10 to 15 standard solution on a plate of silica gel for thin-layer chro-
cm in length, 2 to 5 cm in diameter; externally red-brown to matography. Develop the plate with a mixture of ethyl
dark brown; roughly wrinkled; a cross section light red- acetate, methanol and water (12:2:1) to a distance of about
brown or light grayish brown, with numerous abnormal vas- 10 cm, and air-dry the plate. Examine under ultraviolet light
cular bundles scattering irregularly around the large vascular (main wavelength: 365 nm): one of the spots from the sam-
bundles near center; heavy and hard in texture. ple solution and a blue-white spot from the standard solu-
Odor, slight and characteristic; taste, astringent and tion show the same color tone and the same Rf value.
slightly bitter.
Under a microscope, transverse section reveals the outer- Add the following next to Total ash:
most layer to be several cells thick and composed of cork;
Assay Weigh accurately about 0.3 g of pulverized Pueraria
cork cells contain brown substances; cortex composed of
Root, add 50 mL of diluted methanol (1 in 2), and heat un-
parenchyma; abnormal vascular bundles, exhibiting a ring
der a re‰ex condenser on a water bath for 30 minutes, cool,
of cambium; xylem lies inside of the cambium, and phloem
and ˆlter. To the residue add 50 mL of diluted methanol (1
outside; ˆbers lie outside the phloem; central portion of root
in 2), and perform as the same as above. Combine the
ligniˆed; parenchymatous cells contain aggregated crystals
ˆltrates, add diluted methanol (1 in 2) to make exactly 100
of calcium oxalate, and both simple and 2- to 8-compound
mL, and use this solution as the sample solution. Separately,
starch grains; navel of starch grain obvious.
weigh accurately about 10 mg of Puerarin Reference Stan-
Identiˆcation To 1 g of pulverized Polygonum Root add dard (separately detarmine the water content), add diluted
10 mL of methanol, shake for 15 minutes, and ˆlter. methanol (1 in 2) to make exactly 100 mL, and use this solu-
Evaporate the ˆltrate to dryness, dissolve the residue in 2 mL tion as the standard solution. Perform the test with 10 mL
of methanol, and use this as the sample solution. Perform each of the sample solution and the standard solution as
the test with the sample solution as directed under the Thin- directed under the Liquid Chromatography according to the
layer Chromatography. Spot 5 mL of the sample solution on following conditions, and measure the peak areas of puera-
a plate of silica gel for thin-layer chromatography, develop rin, AT and AS, of each solution.
the plate with a mixture of ethyl acetate, water, methanol
AT
and acetic acid (100) (200:10:10:3) to a distance of about 10 Amount (mg) of puerarin (C21H20O9) = WS ×
AS
cm, and air-dry the plate. Examine under ultraviolet light
(main wavelength: 365 nm): a ‰uorescent bluish white spot WS: Amount (mg) of Puerarin Reference Standard, calcu-
appears at around Rf 0.3. lated on the anhydrous basis

Loss on drying Not more than 14.0z (6 hours). Operating conditions-


Detector: An ultraviolet absorption photometer (wave-
Total ash Not more than 5.5z.
length: 250 nm).
Extract content Dilute ethanol-extract: not less than Column: A stainless steel column 4.6 mm in inside di-
1558 O‹cial Monographs for Part II Supplement I, JP XIV

ameter and 15 cm in length, packed with octadecylsilanized dissolve in a solution of sodium hydrogen carbonate (1 in
silica gel for liquid chromatography (5 mm in particle 1000) to make exactly 50 mL. Pipet 5 mL of this solution,
diameter). add a solution of sodium hydrogen carbonate (1 in 1000) to
Column temperature: A constant temperature of about make exactly 20 mL and use this solution as the standard so-
409C. lution. Perform the test with exactly 10 mL of the sample so-
Mobile phase: A mixture of 0.05 mol W L sodium dihydro- lution and the standard solution as directed under the Liquid
gen phosphate TS and acetonitrile (9:1). Chromatography according to the following conditions, and
Flow rate: Adjust the ‰ow rate so that the retention time determine the peak areas, AT and AS, of sennoside A in each
of puerarin is about 15 minutes. solution.
System suitability-
Amount (mg) of sennoside A (C42H38O20)
System performance: When the procedure is run with
A 1
10 mL of the standard solution under the above operating = WS × T ×
AS 4
conditions, the number of theoretical plates and the symmet-
ry coe‹cient of the peak of puerarin are not less than 3000 WS: Amount (mg) of Sennoside A Reference Standard,
and not more than 2.0, respectively. calculated on the anhydrous basis
System repeatability: When the test is repeated 6 times
Operating conditions-
with 10 mL of the standard solution under the above operat-
Detector: An ultraviolet absorption photometer (wave-
ing conditions, the relative standard deviation of the peak
length: 340 nm).
area of puerarin is not more than 1.5z.
Column: A stainless steel column about 4 to 6 mm in in-
side diameter and about 15 to 25 cm in length, packed with
octadecylsilanized silica gel for liquid chromatography (5 to
Rhubarb 10 mm in particle diameter).
Column temperature: A constant temperature of about
ダイオウ
409C.
Mobile phase: A mixture of diluted acetic acid (100) (1 in
Change the Identiˆcation to read:
80) and acetonitrile (4:1).
Identiˆcation To 2 g of pulverized Rhubarb add 40 mL of Flow rate: Adjust the ‰ow rate so that the retention time
a mixture of tetrahydrofuran and water (7:3), shake for 30 of sennoside A is about 15 minutes.
minutes, and centrifuge. Transfer the supernatant liquid to a System suitability-
separator, add 13 g of sodium chloride, and shake for 30 System performance: Dissolve 1 mg each of Sennoside A
minutes. Separate the water layer with undissolved sodium Reference Standard and naringin for thin-layer chro-
chloride, and adjust the pH to 1.5 by adding 1 mol W L matography in a solution of sodium hydrogen carbonate (1
hydrochloric acid TS. Transfer this solution to another sepa- in 1000) to make 10 mL. When the procedure is run with 20
rator, add 30 mL of tetrahydrofuran, shake for 10 minutes, mL of this solution under the above operating conditions,
separate the tetrahydrofuran layer, and use this solution as sennoside A and naringin are eluted in this order with the
the sample solution. Separately, dissolve 1 mg of Sennoside resolution between these peaks being not less than 3.
A Reference Standard in 4 mL of a mixture of tetrahydrofu- System repeatability: When the test is repeated 6 times
ran and water (7:3), and use this solution as the standard so- with 10 mL of the standard solution under the above operat-
lution. Perform the test with these solutions as directed un- ing conditions, the relative standard deviation of the peak
der the Thin-layer Chromatography. Spot 40 mL each of the area of sennoside A is not more than 1.5z.
sample solution and the standard solution on a plate of silica
gel for thin-layer chromatography at 10 mm along the initial
line. Develop the plate with a mixture of ethyl acetate, 1- Powdered Rhubarb
propanol, water and acetic acid (100) (40:40:30:1) to a dis-
tance of about 15 cm, and air-dry the plate. Examine under ダイオウ末
ultraviolet light (main wavelength: 365 nm): one of the spots
from the sample solution and a red ‰uorescent spot from the Change the Identiˆcation to read:
standard solution show the same color tone and the same Rf
Identiˆcation To 2 g of Powdered Rhubarb add 40 mL of
value.
a mixture of tetrahydrofuran and water (7:3), shake for 30
minutes, and centrifuge. Transfer the supernatant liquid to a
Delete the Component determination and add
separator, add 13 g of sodium chloride, and shake for 30
the following:
minutes. Separate the water layer with undissolved sodium
Assay Weigh accurately about 0.5 g of pulverized chloride, and adjust the pH to 1.5 with 1 mol WL hydrochlor-
Rhubarb, add exactly 50 mL of a solution of sodium hydro- ic acid TS. Transfer this solution to another separator, add
gen carbonate (1 in 1000), shake for 30 minutes, ˆlter, and 30 mL of tetrahydrofuran, shake for 10 minutes, separate
use the ˆltrate as the sample solution. Separately, weigh ac- the tetrahydrofuran layer, and use this solution as the sam-
curately about 10 mg of Sennoside A Reference Standard, ple solution. Separately, dissolve 1 mg of Sennoside A
Supplement I, JP XIV O‹cial Monographs for Part II 1559

Reference Standarad in 4 mL of a mixture of tetrahydrofu- System repeatability: When the test is repeated 6 times
ran and water (7:3), and use this solution as the standard so- with 10 mL of the standard solution under the above operat-
lution. Perform the test with these solutions as directed un- ing conditions, the relative standard deviation of the peak
der the Thin-layer Chromatography. Spot 40 mL each of the area of sennoside A is not more than 1.5z.
sample solution and the standard solution on a plate of silica
gel for thin-layer chromatography at 10 mm along the initial
line. Develop the plate with a mixture of 1-propanol, ethyl Scopolia Extract and Ethyl
acetate, water and acetic acid (100) (40:40:30:1) to a distance
of about 15 cm, and air-dry the plate. Examine under ultrav- Aminobenzoate Powder
iolet light (main wavelength: 365 nm): one of the spots from
ロートエキスアネスタミン散
the sample solution and a red ‰uorescent spot from the stan-
dard solution show the same color tone and the same Rf
Change the Identiˆcation (4) to read:
value.
Identiˆcation
Delete the Component determination and add (4) Place 30 g of Scopolia Extract and Ethyl Aminoben-
the following: zoate Powder in a glass-stoppered conical ‰ask, add 100 mL
of water, shake for 30 minutes, and ˆlter immediately by
Assay Weigh accurately about 0.5 g of Powdered
suction through a glass ˆlter (G3). Transfer the residue in the
Rhubarb, add exactly 50 mL of a solution of sodium hydro-
‰ask to the same glass ˆlter with the ˆltrate, and ˆlter the
gen carbonate (1 in 1000), shake for 30 minutes, ˆlter, and
residue by suction while pressing vigorously the residue on
use the ˆltrate as the sample solution. Separately, weigh ac-
the same glass ˆlter. Place 75 mL of the ˆltrate in a 300-mL
curately about 10 mg of Sennoside A Reference Standard,
beaker, and add cautiously 10 mL of diluted sulfuric acid (1
dissolve in a solution of sodium hydrogen carbonate (1 in
in 3). Add 0.2 mL of bromocresol green TS to this solution,
1000) to make exactly 50 mL. Pipet 5 mL of this solution,
and add dilute sulfuric acid dropwise while shaking thor-
add a solution of sodium hydrogen carbonate (1 in 1000) to
oughly, until the color of the solution changes from green to
make exactly 20 mL, and use this solution as the standard
yellow-green. After cooling, place this solution in a separa-
solution. Perform the test with exactly 10 mL each of the
tor, wash with two 25-mL portions of a mixture of hexane
sample solution and the standard solution as directed under
and diethyl ether (1:1) by shaking well, and place the water
the Liquid Chromatography according to the following con-
layer in another separator. Make slightly alkaline with am-
ditions, and determine the peak areas, AT and AS, of senno-
monia TS, add immediately 30 mL of diethyl ether, and
side A in each solution.
shake well. Wash the diethyl ether layer with two 10-mL por-
Amount (mg) of sennoside A (C42H38O20) tions of a saturated solution of sodium chloride, separate the
A 1 diethyl ether layer, add 3 g of anhydrous sodium sulfate,
= WS × T ×
AS 4 shake, and ˆlter through a pledget of cotton. Evaporate the
ˆltrate to dryness, dissolve the residue in 0.2 mL of ethanol
WS: Amount (mg) of Sennoside A Reference Standard,
(95), and use this solution as the sample solution. Separate-
calculated on the anhydrous basis
ly, dissolve 2 mg of Atropine Sulfate Reference Standard
Operating conditions— and 1 mg of Scopolamine Hydrobromide Reference Stan-
Detector: An ultraviolet absorption photometer (wave- dard in 1 mL each of ethanol (95), and use these solutions as
length: 340 nm). standard solution (1) and standard solution (2). Perform the
Column: A stainless steel column about 4 to 6 mm in in- test with these solutions as directed under the Thin-layer
side diameter and about 15 to 25 cm in length, packed with Chromatography. Spot 10 mL each of the sample solution
octadecylsilanized silica gel for liquid chromatography (5 to and the standard solutions on a plate of silica gel for thin-
10 mm in particle diameter). layer chromatography. Develop the plate with a mixture of
Column temperature: A constant temperature of about acetone, water and ammonia solution (28) (90:7:3) to a dis-
409C. tance of about 10 cm, and dry the plate at 809 C for 10
Mobile phase: A mixture of diluted acetic acid (100) (1 in minutes. After cooling, spray evenly DragendorŠ's TS for
80) and acetonitrile (4:1). spraying on the plate: two principal spots from the sample
Flow rate: Adjust the ‰ow rate so that the retention time solution show the same color tone and the same Rf value
of sennoside A is about 15 minutes. with each yellow-red spot from the standard solutions,
System suitability- respectively.
System performance: Dissolve 1 mg each of Sennoside A
Reference Standard and naringin for thin-layer chro-
matography in a solution of sodium hydrogen carbonate (1
in 1000) to make 10 mL. When the procedure is run with 20
mL of this solution under the above operating conditions,
sennoside A and naringin are eluted in this order with the
resolution between these peaks being not less than 3.
1560 O‹cial Monographs for Part II Supplement I, JP XIV

Delete the following Monographs: solution and dark green spot from the standard solution
show the same color tone and the same Rf value.
Compound Scopolia Extract and
Tannic Acid Ointment Senna Leaf
複方ロートエキスタンニン軟膏
センナ

Compound Scopolia Extract and Change the origin W


limits of content to read:
Tannic Acid Suppositories
Senna Leaf is the lea‰ets of Cassia angustifolia Vahl
複方ロートエキスタンニン坐剤 or Cassia acutifolia Delile (Leguminosae).
It contains not less than 1.0z of total sennosides
[sennoside A (C42H38O20: 862.75) and sennoside B
(C42H38O20: 862.75)], calculated on the basis of dried
Scutellaria Root material.

オウゴン Change the Identiˆcation (2) to read:


Identiˆcation
Change the Identiˆcation (2) to read:
(2) To 2 g of pulverized Senna Leaf add 40 mL of a mix-
Identiˆcation ture of tetrahydrofuran and water (7:3), shake for 30
(2) To 2 g of pulverized Scutellaria Root add 10 mL of minutes, and centrifuge. Transfer the supernatant liquid to a
methanol, warm on a water bath for 3 minutes, cool, ˆlter, separator, add 13 g of sodium chloride, and shake for 30
and use the ˆltrate as the sample solution. Separately, dis- minutes. Separate the water layer with undissolved sodium
solve 1 mg of Baicalin Reference Standard in 1 mL of L
chloride, and adjust the pH to 1.5 by adding 1 mol W
methanol, and use this solution as the standard solution. hydrochloric acid TS. Transfer this solution to another sepa-
Perform the test with these solutions as directed under the rator, shake with 30 mL of tetrahydrofuran for 10 minutes,
Thin-layer Chromatography. Spot 5 mL each of the sample separate the tetrahydrofuran layer, and use this solution as
solution and the standard solution on a plate of silica gel for the sample solution. Separately, dissolve 1 mg of Sennoside
thin-layer chromatography. Develop the plate with a mix- A Reference Standarad in 1 mL of a mixture of tetra-
ture of 1-butanol, water and acetic acid (100) (4:2:1) to a dis- hydrofuran and water (7:3), and use this solution as the stan-
tance of about 10 cm, and air-dry the plate. Spray evenly a dard solution. Perform the test as directed under the Thin-
solution of iron (III) chloride hexahydrate in methanol (1 in layer Chromatography with the sample solution and the
100) on the plate: one spot among the spots from the sample standard solution. Spot 10 mL each of these solutions on a
solution and a dark green spot from the standard solution plate of silica gel for thin-layer chromatography. Develop
show the same color tone and the same Rf value. the plate with a mixture of 1-propanol, ethyl acetate, water
and acetic acid (100) (40:40:30:1) to a distance of about 15
cm, and air-dry the plate. Examine under ultraviolet light
Powdered Scutellaria Root (main wavelength: 365 nm): one spot among the spots from
the sample solution and a red ‰uorescent spot from the stan-
オウゴン末 dard solution show the same color tone and the same Rf
value.
Change the Identiˆcation (2) to read:
Delete the Component determination and add
Identiˆcation
the following:
(2) To 2 g of Powdered Scutellaria Root add 10 mL of
methanol, warm on a water bath for 3 minutes, cool, ˆlter, Assay Weigh accurately about 0.5 g of pulverized Senna
and use the ˆltrate as the sample solution. Separately, dis- Leaf in a glass-stoppered centrifuge tube, add 25 mL of
solve 1 mg of Baicalin Reference Standard in 1 mL of diluted methanol (7 in 10), shake for 30 minutes, centrifuge,
methanol, and use this solution as the standard solution. and separate the supernatant liquid. To the residue add
Perform the test with these solutions as directed under the diluted methanol (7 in 10) twice with a 10-mL portion, shake
Thin-layer Chromatography. Spot 5 mL each of the sample for 10 minutes, centrifuge, and separate the supernatant liq-
solution and the standard solution on a plate of silica gel for uid. Combine all the extracts, add diluted methanol (7 in 10)
thin-layer chromatography. Develop the plate with a mix- to make exactly 50 mL, and use this solution as the sample
ture of 1-butanol, water and acetic acid (100) (4:2:1) to a dis- solution. Separately, weigh accurately about 10 mg of Sen-
tance of about 10 cm, and air-dry the plate. Spray evenly a noside A Reference Standard, dissolve in a solution of sodi-
solution of iron (III) chloride hexahydrate in methanol (1 in um hydrogen carbonate (1 in 100) to make exactly 20 mL,
100) on the plate: one spot among the spots from the sample and use this solution as standard stock solution (1). Weigh
Supplement I, JP XIV O‹cial Monographs for Part II 1561

accurately about 10 mg of Sennoside B Reference Standard, Powdered Senna Leaf


dissolve in a solution of sodium hydrogen carbonate (1 in
100) to make exactly 20 mL, and use this solution as stan- センナ末
dard stock solution (2). Pipet 5 mL of the standard stock so-
lution (1) and 10 mL of the standard stock solution (2), add Change the origin W
limits of content to read:
methanol to make exactly 50 mL, and use this solution as the
standard solution. Perform the test with exactly 10 mL each Powdered Senna Leaf is the powder of Senna Leaf.
of the sample solution and the standard solution as directed It contains not less than 1.0z of total sennosides
under the Liquid Chromatography according to the follow- [sennoside A (C42H38O20: 862.75) and sennoside B
ing conditions. Determine the peak areas, ATa and ASa, of (C42H38O20: 862.75)], calculated on the basis of dried
sennoside A, and the peak areas, ATb and ASb, of sennoside material.
B in each solution, calculate the amounts of sennoside A and
sennoside B by the following equations, and designate the Change the Identiˆcation (2) to read:
total as the amount of total sennosides.
Identiˆcation
Amount (mg) of sennoside A (C42H38O20) (2) To 2 g of Powdered Senna Leaf add 40 mL of a mix-
A 1 ture of tetrahydrofuran and water (7:3), shake for 30
= WSa × Ta ×
ASa 4 minutes, and centrifuge. Transfer the supernatant liquid to a
Amount (mg) of sennoside B (C42H38O20) separator, add 13 g of sodium chloride, and shake for 30
A 1 minutes. Separate the water layer with undissolved sodium
= WSb × Tb ×
ASb 2 chloride, and adjust the pH to 1.5 with 1 mol W L hydrochlor-
ic acid TS. Transfer this solution to another separator, shake
WSa: Amount (mg) of Sennoside A Reference Standard,
with 30 mL of tetrahydrofuran for 10 minutes, separate the
calculated on the anhydrous basis
tetrahydrofuran layer, and use this solution as the sample
WSb: Amount (mg) of Sennoside B Reference Standard,
solution. Separately, dissolve 1 mg of Sennoside A Refer-
calculated on the anhydrous basis
ence Standard in 1 mL of a mixture of tetrahydrofuran and
Operating conditions- water (7:3), and use this solution as the standard solution.
Detector: An ultraviolet aborption photometer (wave- Spot 10 mL each of these solutions on a plate of silica gel for
length: 340 nm). thin-layer chromatography. Develop the plate with a mix-
Column: A stainless steel column 4.6 mm in inside di- ture of ethyl acetate, 1-propanol, water and acetic acid (100)
ameter and 15 cm in length, packed with octadecylsilanized (40:40:30:1) to a distance of about 15 cm, and air-dry the
silica gel for liquid chromatography (5 mm in particle di- plate. Examine under ultraviolet light (main wavelength: 365
ameter). nm): one spot among the spots from the sample solution and
Column temperature: A constant temperature of about a red ‰uorescent spot from the standard solution show the
509C. same color tone and the same Rf value.
Mobile phase: Dissolve 2.45 g of tetra-n-heptylammoni-
um bromide in 1000 mL of a mixture of diluted 1 mol W L Delete the Component determination and add
acetic acid-sodium acetate buŠer solution, pH 5.0 (1 in 10) the following:
and acetonitrile (17:8).
Assay Weigh accurately about 0.5 g of Powdered Senna
Flow rate: Adjust the ‰ow rate so that the retention time
Leaf in a glass-stoppered centrifuge tube, add 25 mL of
of sennoside A is about 26 minutes.
diluted methanol (7 in 10), shake for 30 minutes, centrifuge,
System suitability-
and separate the supernatant liquid. To the residue add
System performance: When the procedure is run with 10
diluted methanol (7 in 10) twice with a 10-mL portoin, shake
mL of the standard solution under the above operating con-
for 10 minutes, centrifuge, and separate the supernatant liq-
ditions, sennoside B and sennoside A are eluted in this order
uid. Combine all the extracts, add diluted methanol (7 in 10)
with the resolution between these peaks being not less than
to make exactly 50 mL, and use this solution as the sample
15, and the number of theoretical plates of the peak of sen-
solution. Separately, weigh accurately about 10 mg of Sen-
noside A being not less than 8000.
noside A Reference Standard, dissolve in a solution of sodi-
System repeatability: When the test is repeated 6 times
um hydrogen carbonate (1 in 100) to make exactly 20 mL,
with 10 mL of the standard solution under the above operat-
and use this solution as standard stock solution (1). Weigh
ing conditions, the relative standard deviation of the peak
accurately about 10 mg of Sennoside B Reference Standard,
area of sennoside A is not more than 1.5z.
dissolve in a solution of sodium hydrogen carbonate (1 in
100) to make exactly 20 mL, and use this solution as stan-
dard stock solution (2). Pipet 5 mL of the standard stock so-
lution (1) and 10 mL of the standard stock solution (2), add
methanol to make exactly 50 mL, and use this solution as the
standard solution. Perform the test with exactly 10 mL each
1562 O‹cial Monographs for Part II Supplement I, JP XIV

of the sample solution and the standard solution as directed Sinomenium Stem
under the Liquid Chromatography according to the follow-
ing conditions. Determine the peak areas, ATa and ASa, of ボウイ
sennoside A, and the peak areas, ATb and ASb, of sennoside
B in each solution, calculate the amounts of sennoside A and Change the Total ash to read:
sennoside B by the following equations, and designate the
Total ash Not more than 7.0z.
total as the amount of total sennoside.

Amount (mg) of sennoside A (C42H38O20)


A
= WSa × Ta ×
1 Swertia Herb
ASa 4
Amount (mg) of sennoside B (C42H38O20) センブリ
A 1
= WSb × Tb ×
ASb 2 Change the Identiˆcation to read:
WSa: Amount (mg) of Sennoside A Reference Standard, Identiˆcation To 2 g of pulverized Swertia Herb add 10
calculated on the anhydrous basis mL of ethanol (95), shake for 5 minutes, ˆlter, and use the
WSb: Amount (mg) of Sennoside B Reference Standard, ˆltrate as the sample solution. Separately, dissolve 2 mg of
calculated on the anhydrous basis Swertiamarin Refereance Standard in 1 mL of ethanol (95),
and use this solution as the standard solution. Perform the
Operating conditions—
test with these solutions as directed under the Thin-layer
Detector: An ultraviolet absorption photometer (wave-
Chromatography. Spot 10 mL each of the sample solution
length: 340 nm).
and the standard solution on a plate of silica gel with com-
Column: A stainless steel column 4.6 mm in inside di-
plex ‰uorescent indicator for thin-layer chromatography.
ameter and 15 cm in length, packed with octadecylsilanized
Develop the plate with a mixture of ethyl acetate, 1-propanol
silica gel for liquid chromatography (5 mm in particle
and water (6:4:3) to a distance of about 10 cm, and air-dry
diameter).
the plate. Examine under ultraviolet light (broad spectrum
Column temperature: A constant temperature of about
wavelength): one spot among several spots from the sample
509C.
solution and a red spot from the standard solution show the
Mobile phase: Dissolve 2.45 g of tetra-n-heptylammoni-
same color tone and the same Rf value.
um bromide in 1000 mL of a mixture of diluted 1 mol W L
acetic acid-sodium acetate buŠer solution, pH 5.0 (1 in 10)
and acetonitrile (17:8).
Flow rate: Adjust the ‰ow rate so that the retention time Powdered Swertia Herb
of sennoside A is about 26 minutes.
センブリ末
System suitability-
System performance: When the procedure is run with 10
Change the Identiˆcation to read:
mL of the standard solution under the above operating con-
ditions, sennoside B and sennoside A are eluted in this order Identiˆcation To 2 g of Powdered Swertia Herb add 10 mL
with the resolution between these peaks being not less than of ethanol (95), shake for 5 minutes, ˆlter, and use the
15, and the number of theoretical plates of the peak of sen- ˆltrate as the sample solution. Separately, dissolve 2 mg of
noside A being not less than 8000. Swertiamarin Reference Standard in 1 mL of ethanol (95),
System repeatability: When the test is repeated 6 times and use this solution as the standard solution. Perform the
with 10 mL of the standard solution under the above operat- test with these solutions as directed under the Thin-layer
ing conditions, the relative standard deviation of the peak Chromatography. Spot 10 mL each of the sample solution
area of sennoside A is not more than 1.5z. and the standard solution on a plate of silica gel with com-
plex ‰uorescent indicator for thin-layer chromatography.
Develop the plate with a mixture of ethyl acetate, 1-propanol
and water (6:4:3) to a distance of about 10 cm, and air-dry
the plate. Examine under ultraviolet light (broad spectrum
wavelength): one spot among several spots from the sample
solution and a red spot from the standard solution show the
same color tone and the same Rf value.
Supplement I, JP XIV O‹cial Monographs for Part II 1563

Swertia and Sodium Bicarbonate Add the following:

Powder Uncaria Thorn


センブリ重曹散
Uncariae Uncis Cum Ramulus
Change the Identiˆcation (1) and (2) to read: チョウトウコウ
Identiˆcation (1) To 10 g of Swertia and Sodium Bicar-
bonate Powder add 10 mL of ethanol (95), shake for 15
Uncaria Thorn is, usually, the prickle of Uncaria
minutes, ˆlter, and use the ˆltrate as the sample solution.
rhynchophylla Miquel, Uncaria sinensis Haviland or
Separately, dissolve 1 mg of Swertiamarin Reference Stan-
Uncaria macrophylla Wallich (Rubiaceae).
dard in 1 mL of ethanol (95), and use this solution as the
Uncaria Thorn contains not less than 0.03z of total
standard solution. Perform the test with these solutions as
rhynchophylline (rhynchophylline and hirstine), calcu-
directed under the Thin-layer Chromatography. Spot 30 mL
lated on the dried basis.
each of the sample solution and the standard solution on a Description Uncaria Thorn is uncinate prickle or short
plate of silica gel with ‰uorescent indicator for thin-layer stem with opposite or single prickle; the prickle, 1 to 4 cm in
chromatography. Proceed as directed in the Identiˆcation length, curved and acuminate; externally red-brown to dark
under Powdered Swertia Herb. brown or yellow-brown, some one with hairs, the transverse
(2) To 0.5 g of Swertia and Sodium Bicarbonate Powder section oblong to elliptical, light brown; stem thin and pris-
add 10 mL of water. After stirring, centrifuge the mixture matic square to cylindrical, 2 to 5 mm in diameter, external-
with 500 revolutions per minute. Smear, using a small glass ly, red-brown to dark brown or yellow-brown; the transverse
rod, the slide glass with a small amount of the precipitate, section, square to elliptical; the pith light brown, square to
add 1 drop of a mixture of water and glycerin (1:1), and put elliptical; hard in texture.
a cover glass on it so that the tissue section spreads evenly Odorless and practically tasteless.
without overlapping each other, taking precaution against Under a microscope, a transverse section of the prickle
inclusion of bubbles, and use this as the preparation for reveals vascular bundles in the cortex, unevenly distributed
microscopic examination. If the precipitate separates into and arranged in a ring. Parenchyma cells in the secondary
two layers, proceed with the upper layer in the same manner, cortex containing sand crystals of calcium oxalate.
and use as the preparation for microscopic examination.
Identiˆcation To 1 g of pulverized Uncaria Thorn add 20
Heat the preparation for microscopic examination in a short
mL of methanol, boil under a re‰ux condenser on a water
time: the preparation reveals the yellow-green to yellow-
bath for 5 minutes, and ˆlter. Evaporate the ˆltrate to dry-
brown, approximately spherical pollen grains with granular
ness, add 5 mL of dilute acetic acid to the residue, warm the
patterns under a microscope. The pollen grains are 25 – 34
mixture on a water bath for 1 minute, and ˆlter after cool-
mm in diameter.
ing. Spot 1 drop of the ˆltrate on a ˆlter paper, air-dry,
spray DragendorŠ's TS for spraying on it, and allow to
stand: a yellow-red color develops.
Toad Venom
Loss on drying Not more than 12.0z (6 hours).
センソ
Total ash Not more than 4.0z.
Change the Identiˆcation to read: Extract content Dilute ethanol-extract: not less than 8.5z.
Identiˆcation To 1 g of pulverized Toad Venom add 10 Component determination Weigh accurately about 0.2 g
mL of acetone, shake for 10 minutes, ˆlter, and use the of medium powdered Uncaria Thorn, transfer into a glass-
ˆltrate as the sample solution. Separately, dissolve 5 mg of stoppered centrifuge tube, add 30 mL of a mixture of
resibufogenin for thin-layer chromatography in 5 mL of ace- methanol and dilute acetic acid (7:3), shake for 30 minutes,
tone, and use this solution as the standard solution. Perform centrifuge, and separate the supernatant liquid. To the
the test with these solutions as directed under the Thin-layer residue add a mixture of methanol and dilute acetic acid
Chromatography. Spot 10 mL each of the sample solution (7:3) twice with a 10-mL portion, proceed in the same man-
and the standard solution on a plate of silica gel for thin-lay- ner, and combine all of the supernatant liquid. To the com-
er chromatography, develop the plate with a mixture of cy- bined liquid add a mixture of methanol and dilute acetic acid
clohexane and acetone (3:2) to a distance of about 10 cm, (7:3) to make exactly 50 mL, and use this as the sample solu-
and air-dry the plate. Spray evenly dilute sulfuric acid on the tion. Separately, weigh accurately about 5 mg of rhyn-
plate, and heat at 1059C for 5 minutes: one of several spots chophylline for component determination, previously dried
obtained from the sample solution has the same color tone in a desiccator (silica gel) for 24 hours, and dissolve in a mix-
and the same Rf value with the blue-green spot obtained ture of methanol and dilute acetic acid (7:3) to make exactly
from the standard solution. 100 mL. Pipet 1 mL of this solution, add a mixture of
methanol and dilute acetic acid (7:3) to make exactly 10 mL,
1564 O‹cial Monographs for Part II Supplement I, JP XIV

and use this solution as the standard solution (1). Separately, Change to read:
dissolve 1 mg of hirsutine in 100 mL of a mixture of
methanol and dilute acetic acid (7:3), and use this as the Vitamin A Oil
standard solution (2). Perform the test with exactly 20 mL
each of the sample solution and the standard solution (1) ビタミンA油
and (2) as directed under the Liquid Chromatography ac-
cording to the following conditions, and determine the peak Vitamin A Oil is synthetic vitamin A esters diluted
areas, ATa and ATb, of rhynchophylline and hirsutine ob- with ˆxed oils. It contains not less than 30,000 Vita-
tained from the sample solution, and the peak area, AS, of min A Units per g. It may contain suitable antiox-
rhynchophylline from the standard solution (1). idants.
Amount (mg) of total rhynchophylline
It contains not less than 90.0z and not more than
120.0z of the labeled amount of vitamin A.
A + 1.405ATb 1
= WS × Ta ×
AS 20 Description Vitamin A Oil is a yellow to yellow-brown,
clear or slightly turbid oil. It is odorless or has a faint, char-
WS: Amount (mg) of rhynchophylline for component de-
acteristic odor.
termination
It is decomposed upon exposure to air or light.
Operating conditions-
Identiˆcation Dissolve Vitamin A Oil, Retinol Acetate
Detector: An ultraviolet absorption photometer (wave-
Reference Standard and Retinol Palmitate Reference Stan-
length: 245 nm).
dard, equivalent to 15,000 Units, in 5 mL of petroleum
Column: A stainless steel column 4.6 mm in inside di-
ether, and use these solutions as the sample solution, the
ameter and 25 cm in length, packed with octadecylsilanized
standard solution (1) and the standard solution (2), respec-
silica gel for liquid chromatography (5 mm in particle
tively. Perform the test with these solutions as directed un-
diameter).
der the Thin-layer Chromatography. Spot 5 mL each of the
Column temperature: A constant temperature of about
sample solution and the standard solutions (1) and (2) on a
409C.
plate of silica gel for thin-layer chromatography. Develop
Mobile phase: Dissolve 3.85 g of ammonium acetate in
with a mixture of cyclohexane and diethyl ether (12:1) to a
about 200 mL of water, add 10 mL of acetic acid (100) and
distance of about 10 cm, and air-dry the plate. Spray evenly
water to make 1000 mL, and add 350 mL of acetonitrile.
antimony (III) chloride TS: the principal spot obtained from
Flow rate: Adjust the ‰ow rate so that the retention time
the sample solution has the same color tone and the same Rf
of rhynchophylline is about 17 minutes.
value with the blue spot obtained from the standard solution
System suitability—
(1) or the standard solution (2).
System performance: Dissolve 5 mg of rhynchophylline
for component determination in 100 mL of a mixture of Purity (1) Acid-Dissolve 1.2 g of Vitamin A Oil in 30
methanol and dilute acetic acid (7:3). To 5 mL of this solu- mL of a mixture of neutralized ethanol and diethyl ether
tion add 1 mL of ammonia solution (28), and re‰ux for 10 (1:1), boil gently for 10 minutes under a re‰ux condenser,
minutes or heat at about 509 C for 2 hours. After cooling, to cool, and add 5 drops of phenolphthalein TS and 0.60 mL of
1 mL of the solution so obtained add a mixture of methanol 0.1 mol WL sodium hydroxide VS: a red color develops.
and dilute acetic acid (7:3) to make 5 mL. When the proce- (2) Rancidity—No unpleasant odor of rancid oil is per-
dure is run with 20 mL of this solution under the above oper- ceptible by warming Vitamin A Oil.
ating conditions, the peak of isorhynchophylline is appears
Assay Proceed as directed in Method 1-1 under the Vita-
in addition to the peak of rhynchophylline, and the resolu-
min A Assay.
tion between these peaks is not less than 1.5.
System repeatability: When the test is repeated 6 times Containers and storage Containers—Tight containers.
with 20 mL of the standard solution (1) under the above Storage—Light-resistant, and almost well-ˆlled, or under
operating conditions, the relative standard deviation of the Nitrogen atmosphere.
peak area of rhynchophylline is not more than 1.5z.
Supplement I, JP XIV O‹cial Monographs for Part II 1565

Change to read: Identiˆcation Proceed the test with the content of Vitamin
A Oil Capsules as directed in the Identiˆcation under Vita-
Vitamin A Oil Capsules min A Oil.

Vitamin A Capsules Assay Weigh accurately 20 Vitamin A Oil Capsules, and


open the capsules to take out the content. Wash the capsules
ビタミンA油カプセル well with a small amount of diethyl ether, allow the capsules
to stand at ordinal temperature to vaporize the diethyl ether,
Vitamin A Oil Capsules contain not less than 90.0z and weigh accurately. Perform the test with the content as
and not more than 130.0z of the labeled Units of directed under the Vitamin A Assay, and calculate the units
Vitamin A. of vitamin A per capsule. Before applying Method 1-1, it is
Method of preparation Prepare as directed under Cap- necessary to know which the sample is, retinol acetate or
sules, using Vitamin A Oil. retinol palmitate.

Description The content of Vitamin A Oil Capsules con- Containers and storage Containers—Well-closed contain-
forms to the requirements of the Description under Vitamin ers.
A Oil. Storage—Light-resistant.
Supplement I, JP XIV Infrared Reference Spectra 1567

Add the following:


Part I
1568 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1569
1570 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1571
1572 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1573
1574 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1575
1576 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1577
1578 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1579
1580 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1581
1582 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1583
1584 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Infrared Reference Spectra 1585
1586 Infrared Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1587

Delete the following Ultraviolet-visible Reference Spectra:

Part I

Homochlorcyclizine Hydrochloride 1
Homochlorcyclizine Hydrochloride 2
1588 Ultraviolet-visible Reference Spectra Supplement I, JP XIV

Change to read:
Part I
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1589

Add the following:


Part I
1590 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1591
1592 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1593
1594 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1595
1596 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1597
1598 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1599
1600 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1601
1602 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1603
1604 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1605
1606 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1607
1608 Ultraviolet-visible Reference Spectra Supplement I, JP XIV
Supplement I, JP XIV Ultraviolet-visible Reference Spectra 1609
General Information

Change to read: Change to read:

1. Aristolochic acid 5. International Harmonization


Aristolochic acid, which occurs in plants of Aristolochia- Implemented in the Japanese
ceae, is suspected to cause renal damage. It is also reported Pharmacopoeia Fourteenth
to be oncogenic (see References).
Aristolochic acid toxicity will not be a problem if crude Edition
drugs of the origin and parts designated in the JP are used,
Items for which harmonization has been agreed among
but there may be diŠerences in crude drug nomenclature be-
the European Pharmacopoeia, the United States Phar-
tween diŠerent countries, and it is known that crude drug
macopeia and the Japanese Pharmacopoeia are implement-
preparations not meeting the speciˆcations of the JP are cir-
ed in the Japanese Pharmacopoeia Fourteenth Edition (JP
culating in some countries. Consequently, when crude drugs
14). They are shown in the tables below. The column headed
or their preparations are used, it is important that the
Harmonized items shows the harmonized items written in
materials should not include any plant containing
the Pharmacopoeial Harmonization Agreement Document,
aristolochic acid.
and the column headed JP 14 shows the items as they appear
In the Supplement I to JP14, the test for aristolochic acid
in JP 14. In the Remarks column, notes on any diŠerences
I was added to the Purity under Asiasarum Root, which con-
between JP 14 and the agreement are shown as occasion de-
sists of the rhizome and root. Because the aerial part of the
mands.
plant may contain aristolochic acid and may have been
improperly contaminated in Asiasarum Root.
It is considered that Akebia Stem, Sinomenium Stem and
Saussurea Root do not contain aristolochic acid, unless
plants of origin other than that designated in the JP are
used. However, contamination of aristolochic acid might oc-
cur, as mentioned above. In this case, the test described in
the Purity under Asiasarum Root is useful for checking the
presence of aristolochic acid.
References:
Drug & Medical Device Safety Information (No.161)
(July, 2000).
New England Journal of Medicine (June 8, 2000).
Mutation Research 515, 63–72 (2002).

1611
1612 General Information Supplement I, JP XIV

Harmonized items JP 14 Remarks

Bacterial Endotoxin Test Bacterial Endotoxins Test


Apparatus Apparatus
Preparation of Standard Endotoxin Preparation of Standard Endotoxin
Stock solution Stock Solution
Preparation of Standard Endotoxin Preparation of Standard Endotoxin
solution Solution
Preparation of sample solutions Preparation of sample solutions
Determination of Maximum Valid Di- Determination of Maximum Valid Di-
lution lution
Gel-clot technique Gel-clot techniques
(1) Preparatory testing (1) Preparatory testing
(2) Limit test (2) Limit test
(3) Assay (3) Assay
Photometric techniques Photometric techniques
(1) Turbidimetric technique (1) Turbidimetric technique
(2) Chromogenic technique (2) Chromogenic technique
(3) Preparatory testing (3) Preparatory testing
(4) Assay (4) Assay
Reagents, Test Solutions Reagents, Test Solutions
Amebocyte lysate Lysate reagent
Lysate TS Lysate TS
Water for bacterial endotoxins test Water for bacterial endotoxins test
(BET)

Note: The method for decision of the limit for bacterial endotoxins was agreed between the three pharmacopoeias, but in the
Decision of Limit for Bacterial Endotoxins under the General Information in JP 14, the maximum adult dose is calculated
based on an average body mass of an adult of 60 kg.
Supplement I, JP XIV General Information 1613

Harmonized items JP 14 Remarks

Residue on Ignition W
Sulphated Ash Residue on Ignition Test
Test
(Introduction) (Introduction) Explanation of JP's particular expres-
sions in the introduction: The descrip-
tion, for example, ``not more than 0.10
z (1 g),'' in a monograph, indicates
that the mass of the residue is not more
than 1.0 mg per g of the substance to be
tested in which about 1 g of the sub-
stance is weighed accurately and ignited
by the procedure described below, and
``after drying'' indicates that the sam-
ple is tested after being dried under the
conditions speciˆed in the test for Loss
on drying.
Procedure Procedure Explanation of JP's particular expres-
sions for taking the sample: When the
quantity of the sample to be taken is in-
dicated in a volume, pipet exactly the
amount directed in the monograph and
transfer to the above crucible. When
directed as ``after evaporating,'' heat
properly to evaporate the solution.

Harmonized items JP 14 Remarks

Sodium Chloride Sodium Chloride


Deˆnition limits of the content
Identiˆcation A Identiˆcation (1)
Identiˆcation B Identiˆcation (2)
Acidity or alkalinity Purity (2) Acidity or alkalinity
Bromides Purity (5) Bromides
Ferrocyanides Purity (7) Ferrocyanides
Iodides Purity (6) Iodides
Nitrites not speciˆed
Phosphates Purity (4) Phosphates
Sulphates Purity (3) Sulfates
Aluminium not speciˆed
Barium Purity (10) Barium
Iron Purity (9) Iron
Magnesium and alkaline-earth metals Purity (11) Magnesium and alkaline-
earth materials
Potassium not speciˆed
Loss on drying Loss on drying
Bacterial endotoxins not speciˆed
Assay Assay
1614 General Information Supplement I, JP XIV

Harmonized items JP 14 Remarks

Citric Acid Monohydrate Citric Acid


Deˆnition limits of the content
Identiˆcation Identiˆcation
Appearance of solution Purity (1) Clarity and color of solu-
tion
Readily carbonisable substances Purity (5) Readily carbonizable sub-
stances
Oxalic acid Purity (3) Oxalate
Sulphates Purity (2) Sulfates
Aluminium not speciˆed
Water Water
Sulphated ash Residue on ignition
Bacterial endotoxins not speciˆed
Assay Assay
Storage Containers and storage

Harmonized items JP 14 Remarks

Citric Acid, Anhydrous Anhydrous Citric Acid


Deˆnition limits of the content
Identiˆcation Identiˆcation
Appearance of solution Purity (1) Clarity and color of solu-
tion
Readily carbonisable substances Purity (5) Readily carbonizable sub-
stances
Oxalic acid Purity (3) Oxalate
Sulphates Purity (2) Sulfates
Aluminium not speciˆed
Water Water
Sulphated ash Residue on ignition
Bacterial endotoxins not speciˆed
Assay Assay
Storage Containers and storage
Supplement I, JP XIV General Information 1615

Harmonized items JP 14 Remarks

General Information General Information


Sodium Dodecyl Sulphate Poly- SDS-Polyacrylamide Gel Electrophore-
acrylamide Gel Electrophoresis sis
(SDS-PAGE)
Characteristics of Polyacrylamide 1. Characteristics of Polyacrylamide
Gels Gels
Denaturing Polyacrylamide Gel Elec- 2. Polyacrylamide Gel Electrophore-
trophoresis sis under Denaturing Conditions
Reducing conditions 1) Reducing conditions
Non-reducing conditions 2) Non-reducing conditions
Characteristics of Discontinuous 3. Characteristics of Discontinuous
BuŠer System Gel Electrophoresis BuŠer System Gel Electrophoresis
Preparing Vertical Discontinuous 4. Preparing Vertical Discontinuous
BuŠer SDS Polyacrylamide Gels BuŠer SDS-Polyacrylamide Gels
Assembling of the gel moulding cas- 1) Assembling of the gel moulding
sette cassette
Preparation of the gel 2) Preparation of the gel
Mounting the gel in the electrophore- 3) Mounting the gel in the electro-
sis apparatus and electrophoretic phoresis apparatus and electro-
separation phoretic separation
Detection of Protein in Gels 5. Detection of Proteins in Gels
Coomassie staining 1) Coomassie staining
Silver staining 2) Silver staining
Drying of Stained SDS Poly- 6. Drying of Stained SDS-
acrylamide Gels Polyacrylamide Gels
Molecular-Mass Determination 7. Molecular-Mass Determination
Validation of the Test 8. Suitability of the Test
Quantiˆcation of Impurities 9. Quantiˆcation of Impurities
Reagents, Test Solutions Test Solutions
Blocking solution Blocking TS
Coomassie staining solution Coomassie staining TS
Destaining solution Destaining TS
Developer solution Developer TS
Fixing solution Fixing TS
Silver nitrate reagent Silver nitrate TS for silver staining
Trichloroacetic acid reagent Trichloroacetic acid TS for ˆxing
Table 1 - Preparation of resolving Table 1. Preparation of resolving gel
gel
Table 2 - Preparation of stacking gel Table 2. Preparation of stacking gel
1616 General Information Supplement I, JP XIV

Change to read: microorganisms, one passage is deˆned as the transfer of


microorganisms from an established culture to fresh medi-
12. Preservatives-EŠectiveness um, and microorganisms subjected to not more than ˆve
passages should be used for the tests. Single-strain
Tests challenges rather than mixed cultures should be used. The
test strains can be harvested by growth on solid agar or liq-
The purpose of the Preservatives-EŠectiveness Tests is to
uid media.
assess microbiologically the preservative e‹cacy, either due
Cultures on agar plate media: Inoculate each of the ˆve
to the action of product components themselves or any
test strains on the surface of agar plates or agar slants. For
added preservative(s), for multi-dose containers. The e‹ca-
growth of bacteria, use Soybean-Casein Digest Agar Medi-
cy of the preservatives is assessed by direct inoculation and
um, and for yeasts and moulds, use Sabouraud Agar,
mixing of the test strains in the product, and titration of sur-
Glucose-Peptone Agar or Potato Dextrose Agar Medium.
vival of the test strains with time.
Incubate bacterial cultures at 309 C to 359 C for 18 to 24
Preservatives must not be used solely to comply with
hours, the culture of C. albicans at 209 C to 259 C for 40 to 48
GMP for drugs or to reduce viable aerobic counts. In addi-
hours and the culture of A. niger at 209 C to 259C for one
tion, preservatives themselves are toxic substances. There-
week or until good sporulation is obtained. Harvest these
fore, preservatives must not be added to products in
cultured cells aseptically using a platinum loop, etc. Suspend
amounts which might jeopardize the safety of human
the collected cells in sterile physiological saline or in 0.1z
beings, and consideration must be given to minimizing the
peptone water and adjust the viable cell count to about 108
amounts of preservative used. These tests are commonly
microorganisms per mL. In the case of A. niger, suspend the
used to verify that products maintain their preservative eŠec-
cultured cells in sterile physiological saline or 0.1z peptone
tiveness at the design phase of formulation or in the case of
water containing 0.05 w W vz of polysorbate 80 and adjust
periodic monitoring. Although these tests are not performed
the spore count to about 108 per mL. Use these suspensions
for lot release testing, the e‹cacy of the preservative present
as the inocula.
in the product packaged in the ˆnal containers should be
Liquid cultures: After culturing each of the four strains
veriˆed throughout the entire dating period.
except for A. niger in a suitable medium, remove the medi-
1. Products and their Categories um by centrifugation. Wash the cells in sterile physiological
The products have been divided into two categories for saline or 0.1z peptone water and resuspend them in the
these tests. Category I products are those made with aqueous same solution with the viable cell or spore count of the in-
bases or vehicles, and Category II products are those made oculum adjusted to about 108 per mL.
with nonaqueous bases or vehicles. Oil-in-water emulsions When strains other than the ˆve listed above are cultured,
are considered Category I products, and water-in-oil emul- select a culture medium suitable for growth of the strain con-
sions Category II products. Category I is further divided cerned. The cell suspension may also be prepared by a
into three subtypes depending on the dosage forms. method suitable for that strain. Use the inoculum suspen-
Category IA: Injections and other sterile parenterals sions within 24 hours after they have been prepared from the
Category IB: Nonsterile parenterals cultivations on agar plate media or in liquid media. Store the
Category IC: Oral products made with aqueous bases (in- inoculum suspensions in a refrigerator if it is not possible to
cluding syrup products to be dissolved or suspended before inoculate them into the test specimens within 2 hours. Ti-
use) trate the viable cell count of the inocula immediately before
Category II: All the dosage forms listed under Category I use, and then calculate the theoretical viable cell count per
made with nonaqueous bases or vehicles. mL (g) of the product present just after inoculation.
2. Test Microorganisms and Culture Media 3. Test Procedure
The following strains or those considered to be equivalent 3.1 Category I products
are used as the test microorganisms. Inject each of the cell suspensions aseptically into ˆve con-
Escherichia coli ATCC 8739, NBRC 3972 tainers containing the product and mix uniformly. When it is
Pseudomonas aeruginosa ATCC 9027, NBRC 13275 di‹cult to inject the cell suspension into the container asep-
Staphylococcus aureus ATCC 6538, NBRC 13276 tically or the volume of the product in each container is too
Candida albicans ATCC 10231, NBRC 1594, JCM 2085 small to be tested, transfer aseptically a su‹cient volume of
Aspergillus niger ATCC 16404, NBRC 9455 the product into each of alternative sterile containers, and
These test microorganisms are representative of those that mix the inoculum. When the product is not sterile, incubate
might be found in the environment in which the product is additional containers containing the uninoculated product
manufactured, used or stored, and they are also recognized as controls and calculate their viable cell counts (the viable
as opportunistic pathogens. In addition to these strains counts of bacteria and those of yeasts and moulds). A sterile
designated as test microorganisms, it is further recommend- syringe, spatula or glass rod may be used to mix the cell sus-
ed to use strains that might contaminate the product and pension uniformly in the product. The volume of the suspen-
grow on or in it, depending on its characteristics. For the test sion mixed in the product must not exceed 1 W 100 of the
microorganisms received from coordinated collections of volume of the product. Generally, the cell suspension is in-
Supplement I, JP XIV General Information 1617

oculated and mixed so that the concentration of viable cells limit speciˆed in ``Microbial Attributes of Nonsterile Phar-
is 105 to 106 cells per mL or per gram of the product. Incu- maceutical Products'' in General Information, caution is
bate these inoculated containers at 209C to 259 C with pro- also required in the test procedures and W
or the control of the
tection from light, and calculate the viable cell count of 1 manufacturing process of the product.
mL or 1 g of the product taken at 0, 14 and 28 days subse-
Table 1. Interpretation criteria by product category
quent to inoculation. Record any marked changes (e.g.,
changes in color or the development of a bad odor) when ob- Interpretation criteria
Product
served in the mixed samples during this time. Such changes Microorganisms
category
should be considered when assessing the preservative e‹cacy After 14 days After 28 days
of the product concerned. Express sequential changes in the
Bacteria 0.1z of inocu- Same or less
viable counts as percentages, with the count at the start of lum count or than level after
the test taken as 100. Titration of the viable cell counts is less 14 days
based, in principle, on the Pour Plate Methods in ``Microbi- Category IA
al Limit Tests''. In this case, conˆrm whether any an- Yeasts W
moulds Same or less Same or less
timicrobial substance is present in the test specimen. If a than inoculum than inoculum
count count
conˆrmed antimicrobial substance needs to be eliminated,
incorporate an eŠective inactivator of the substance in the Bacteria 1z of inoculum Same or less
buŠer solution or liquid medium to be used for dilution of count or less than level after
the test specimen, as well as in the agar plate count medium. 14 days
However, it is necessary to conˆrm that the inactivator has Category IB
Yeasts W
moulds Same or less Same or less
no eŠect on the growth of the microorganisms. When the oc- than inoculum than inoculum
currence of the preservative or the product itself aŠects titra- count count
tion of the viable cell count and there is no suitable inactiva-
tor available, calculate the viable cell counts by the Mem- Bacteria 10z of inocu- Same or less
brane Filtration Method in ``Microbial Limit Tests''. lum count or than level after
less 14 days
3.2 Category II products
Category IC
The procedures are the same as those described for Cate- Yeasts W
moulds Same or less Same or less
gory I products, but special procedures and considerations than inoculum than inoculum
are required for both uniform dispersion of the test microor- count count
ganism in the product and titration of viable cell counts in
Bacteria Same or less Same or less
the samples. than inoculum than inoculum
For semisolid ointment bases, heat the sample to 459 C to count count
509C until it becomes oily, add the cell suspension and dis- Category II
perse the inoculum uniformly with a sterile glass rod or spat- Yeasts W
moulds Same or less Same or less
ula. Surfactants may also be added to achieve uniform dis- than inoculum than inoculum
count count
persion, but it is necessary to conˆrm that the surfactant
added has no eŠect on survival or growth of the test
microorganisms and that it does not potentiate the preserva- 5. Culture Media
tive e‹cacy of the product. For titration of the viable cell Culture media and buŠer solution used for Preservatives-
count, a surfactant or emulsiˆer may be added to disperse EŠectiveness Tests are described below. Other media may be
the product uniformly in the buŠer solution or liquid medi- used if they have similar nutritive ingredients and selective
um. Sorbitan monooleate, polysorbate 80 or lecithin may be and growth-promoting properties for the microorganisms to
added to improve miscibility between the liquid medium and be tested.
semisolid ointments or oils in which test microorganisms Soybean Casein Digest Agar Medium
were inoculated. These agents serve to inactivate or neutral- Casein peptone 15.0 g
ize many of the most commonly used preservatives. Soybean peptone 5.0 g
4. Interpretation Sodium chloride 5.0 g
Interpret the preservative e‹cacy of the product accord- Agar 15.0 g
ing to Table 1. When the results described in Table 1 are ob- Water 1000 mL
tained, the product examined is considered to be eŠectively Mix all of the components and sterilize at 1219 C for 15 –
preserved. There is a strong possibility of massive microbial 20 minutes in an autoclave. pH after sterilization: 7.1 – 7.3.
contamination having occurred when microorganisms other Sabouraud Glucose Agar Medium
than the inoculated ones are found in the sterile product to Peptone (animal tissue and casein) 10.0 g
be examined, and caution is required in the test procedures Glucose 40.0 g
and W or the control of the manufacturing process of the Agar 15.0 g
product. When the contamination level in a nonsterile Water 1000 mL
product to be examined exceeds the microbial enumeration Mix all of the components and sterilize at 1219C for 15 –
1618 General Information Supplement I, JP XIV

20 minutes in an autoclave. pH after sterilization: 5.4 – 5.8. the highest level of current scientiˆc speculation. It is expect-
ed that this information will contribute to promotion of
Glucose Peptone (GP) Agar Medium
scientiˆc understanding of quality and safety assurance of
Glucose 20.0 g
not only JP listed products but also the other biotechnologi-
Yeast extract 2.0 g
cal W
biological products and to promotion of active discus-
Magnesium sulfate heptahydrate 0.5 g
sion of each O‹cial Monograph in JP.
Peptone 5.0 g
Monobasic potassium phosphate 1.0 g 1. Fundamental measures to ensure viral safety of JP listed
Agar 15.0 g biotechnological W biological products
Water 1000 mL The biotechnological W biological product JP includes the
Mix all of the components and sterilize at 1219 C for 15 – products derived from living tissue and body ‰uid (urine,
20 minutes in an autoclave. pH after sterilization: 5.6 – 5.8. blood, etc.) of mammals, etc. In near future, protein drugs
derived from cell lines of human or animal origin (e.g.,
Potato Dextrose Agar Medium
recombinant DNA drug, cell culture drug) will be included.
Potato extract 4.0 g
The fundamental measures required for comprehensive viral
Glucose 20.0 g
safety of JP listed biotechnological Wbiological products are
Agar 15.0 g
as follows: 1) acquaintance of possible virus contamination
Water 1000 mL
(source of contamination); 2) careful examination of eligibil-
Mix all of the components and sterilize at 1219 C for 15 –
ity of raw materials and their sources, e.g. human W animal,
20 minutes in an autoclave. pH after sterilization: 5.4 – 5.8.
and thorough analysis and screening of the sample chosen as
0.1z Peptone Water a substrate for drug production (e.g., pooled body ‰uid, cell
Peptone 1.0 g bank, etc.) to determine any virus contamination and deter-
Sodium chloride 8.0 g mination of type and nature of the virus, if contaminated; 3)
Water 1000 mL evaluation to determine virus titer and virus-like particles
Mix all of the components and sterilize at 1219 C for 15 – hazardous to human, if exists; 4) selection of production
20 minutes in an autoclave. pH after sterilization: 7.2 – 7.4. related material (e.g., reagent, immune antibody column)
free from infectious or pathogenic virus; 5) performance of
virus free test at an appropriate stage of manufacturing in-
Add the following: cluding the ˆnal product, if necessary; 6) adoption of eŠec-
tive viral clearance method in the manufacturing process to
17. Basic Requirements for Viral remove W inactivate virus. Combined method sometimes
achieves higher level of clearance; 7) development of a
Safety of Biotechnological W deliberate viral clearance scheme; 8) performance of the test
Biological Products listed in to evaluate viral removal and inactivation. It is considered
that the stepwise and supplemental adoption of the said
Japanese Pharmacopoeia measures will contribute to ensure viral safety and its im-
provement.
Introduction
The primary role of speciˆcation of biotechnological W bio- 2. Safety assurance measures described in the O‹cial
logical products listed in Japanese Pharmacopoeia (JP) is Monograph and this General Information
not only for securing quality control or consistency of the As mentioned in above 1, this General Information
quality but also for assuring their e‹cacy and safety. In the describes, in package, points to be concerned with and con-
meantime, the requirements to assure quality and safety of crete information on the measures taken for viral safety of
drugs have come to be quite strict recently, and a rigid atti- JP listed products. Except where any speciˆc caution is pro-
tude addressing safety assurance is expected for biotechno- vided in O‹cial Monograph of a product in question, O‹-
logical W biological products. The key points for quality and cial Monograph provides in general that ``Any raw material,
safety assurance of biotechnological W biological products are substrate for drug production and production related
selection and appropriate evaluation of source material, ap- material used for production of drug should be derived from
propriate evaluation of manufacturing process and main- healthy animals and should be shown to be free of latent vi-
tenance of manufacturing consistency, and control of rus which is infectious or pathogenic to human'', ``Cell line
speciˆc physical properties of the products. Now, how to as- and culture method well evaluated in aspects of appropriate-
sure quality and safety of such drugs within a scope of JP ness and rationality on viral safety are used for production,
has come to be questioned. This General Information and the presence of infectious or pathogenic latent virus to
describes what sorts of approaches are available to overcome human in process related materials derived from living or-
these issues. ganisms should be denied''. and ``biotechnological W biologi-
It is desired that quality and safety assurance of JP listed cal drug should be produced through a manufacturing proc-
products are achieved by state-of-the-art methods and con- ess which is capable of removing infectious or pathogenic vi-
cepts which re‰ect progress of science and accumulation of rus'', etc., to raise awareness on viral safety and on necessity
experiences. This General Information challenges to show to conduct test and process evaluation for viral safety.
Supplement I, JP XIV General Information 1619

3. Items and contents described in this General Informa- 1. Rationale, objective and general items to be concerned
tion with respect to viral clearance process evaluation
As for viral safety of protein drug derived from cell line of 2. Selection of virus
human or animal origin, there is a Notice in Japan entitled 3. Design of viral clearance studies
``Viral safety evaluation of biotechnology products derived 4. Interpretation of viral clearance studies
from cell lines of human or animal origin'' (Iyakushin No. 1) Evaluation of viral clearance factor
329 issued on February 22, 2000 by Director, Evaluation and 2) Calculation of viral clearance index
Licensing Division, Pharmaceutical and Medical Safety 3) Interpretation of results and items to be concerned at
Bureau, Ministry of Health and Welfare) to re‰ect the inter- evaluation
nationally harmonized ICH Guideline, and as for blood VI. Statistics
plasma protein fraction preparations, there is a document 1. Statistical considerations for assessing virus assays
entitled ``Guideline for ensuring viral safety of blood plasma 2. Reproducibility and conˆdence limit of viral clearance
protein fraction preparations''. This General Information studies
for ensuring viral safety of JP listed biotechnological W bio- VII. Re-evaluation of viral clearance
logical products has been written, referencing the contents VIII. Measurement for viral clearance studies
of those guidelines, to cover general points and their details 1. Measurement of virus infective titer
to be concerned for ensuring viral safety of not only JP list- 2. Testing by nucleic-acid ampliˆcation test (NAT)
ed biotechnological W biological products but also all IX. Reporting and preservation
products which would be listed in JP in future, i.e., biologi- X. Others
cal products derived from living tissue and body ‰uid, such
as urine, and protein drugs derived from cell line of human 3.1 Purpose
or animal origin (Table 1). The purpose of this document is to propose the compre-
hensive concepts of the measures to be taken for ensuring
Table 1. Items described in General Information viral safety of biotechnological W biological products derived
for Viral Safety Assurance of JP listed from living tissue or body ‰uid of mammals, etc. and of pro-
Biotechnological W Biological Product tein drugs derived from cell lines of human or animal origin.
I. Introduction That is to say, this document describes the measures and the
1. Fundamental measures to ensure viral safety of JP list- points of concern on the items, such as  consideration of
ed biotechnological W biological products the source of virus contamination;  appropriate evaluation
2. Safety assurance measures described in the O‹cial on eligibility at selecting the raw material and on qualiˆca-
Monograph and this General Information tion of its source, e.g. human or animal;  virus test, and its
3. Items and contents described in this General Informa- analysis and evaluation at a stage of cell substrate for drug
tion production;  appropriate evaluation to choose product
II. General Matters related materials derived from living organisms (e.g. rea-
1. Purpose gent, immune antibody column, etc.);  conduct of neces-
2. Background sary virus test on the product at an appropriate stage of
3. Unknown risk on the measures taken for ensuring viral manufacturing;  development of viral clearance test
safety scheme;  performance and evaluation of viral clearance
4. Applicable range test. This document is also purposed to comprehensively
5. Possible viral contamination to a JP listed biotechno- describe in details that supplemental and combining adop-
logical W
biological product (source of virus contamina- tion of the said measures will contribute to secure viral safe-
tion) ty and its improvement.
6. Basis for ensuring viral safety 3.2 Background
7. Limit of virus test One of the most important issues to be cautioned for safe-
8. Roles of viral clearance studies ty of a biological product, which is directly derived from hu-
III. Raw material W substrate for drug production man or animal, or of a protein drug, which is derived from
1. Issues relating to animal species and its region as a cell line of human or animal origin (recombinant DNA der-
source of raw material W substrate for drug production ived product, cell culture derived product, etc.), is risk of vi-
and countermeasures to be taken thereto rus contamination. Virus contamination may cause serious
2. Qualiˆcation evaluation test on human or animal as a situation at clinical use once it occurs. Virus contamination
source of raw material W substrate for drug production may be from a raw material or from a cell substrate for drug
IV. Points of concern with respect to manufacturing and production, or may be from an adventitious factor in-
virus testing troduced to the manufacturing process.
1. Virus test conducted in advance of puriˆcation process JP listed biological drugs or protein drugs derived from
2. Virus test as an acceptance test of an intermediate cell line have achieved drastic contribution to the medical so-
material, etc. ciety, and to date, there has not been any evidence of any
3. Virus test on a ˆnal product safety problem on them caused by virus. But, social require-
V. Process evaluation on viral clearance ment of health hazard prevention is strong, and it is now
1620 General Information Supplement I, JP XIV

very important to prevent accidental incidence, taking considered with the relative comparison to alternative drugs
security measures carefully supported by scientiˆc rationali- or medical treatment. The usefulness of certain drug should
ty. It is always great concern among the persons involved be reviewed ˆnally after the competitive assessment on the
that under what sort of viewpoint and to what extent we risk and beneˆt on the alternative drugs, relevant drugs and W
have to pursue for ensuring viral safety of a biotechnological or alternative medical treatment.
W biological product. Before discussing these issues, two fun- Under such background, the purpose of this article is to
damental points have to be reconˆrmed. One is that; we describe the scientiˆc and rational measures to be taken for
have to consider scientiˆc, medical, and social proˆles a ensuring viral safety of JP listed biotechnological W biological
drug has. In other words, ``Medicine is a social asset which is products. Giving scientiˆc and rational measures mean that;
utilized in medical practice paying attention to the risk and appropriate and eŠective measures, elaborated from the cur-
beneˆt from the standpoints of science and society''. It is the rent scientiˆc level, are given to the issues assumable under
destine and the mission of the medical W pharmaceutical soci- the current scientiˆc knowledge. In other words, possible
ety to realize prompt and stable supply of such a social asset, contaminant virus is assumed to have the natures of genus,
drug, among the medical work front to bring gospel to the morph, particle size, physical W chemical properties, etc.
patients. which are within the range of knowledge of existing virolo-
The other is that; issue of viral safety is independent from gy, and is those assumed to exist in human and animal, tis-
safety of the components of a drug per se (narrow sense of sue and body ‰uid, which are the source of biotechnological
safety). It is important to consider that this is the matter of W biological product, reagent, material, additives, etc. Ac-
general safety of drug (broad sense of safety). In case of a cordingly, viral clearance studies using a detection method
drug which has been used for a long time in the medical which target those viruses have to be designed.
front, such as a JP listed product, its broad sense of safety is 3.3 Unknown risk on the measures taken for ensuring viral
considered to have been established epidemiologically, and safety
its usage past records have a great meaning. However, diŠer- There are known and unknown risks.
ent from safety of drug per se (its components), taking into It is easy to determine a test method and an evaluation
account any possibility of virus contamination, we have to standard on the known risk, which exists in the drug per se
say that only the results accumulated can not always assure (pharmaceutical component) or inevitably exists due to a
viral safety of a drug used in future. Accordingly, the basis quality threshold, and quantiˆcation of such risk is possible.
for securing broad sense of viral safety of JP listed In other words, it is easy to evaluate the known risk on a
biotechnological W biological products is to pay every atten- balance sheet in relation to the beneˆt, and we can say that
tion to the measures to take for prevention, while evaluating valuation even in this respect has been established to some
the accumulated results. extent.
Adopting strict regulations and conducting tests at maxi- On the other hand, as for the unknown risk which is in-
mum level to the extent theoretically considered may be the evitable for ensuring viral safety, the subject of the risk can
ways oŠ assuring safety, but applying such way generally, not be deˆned and quantitative concept is hard to introduce,
without su‹cient scientiˆc review of the ways and evalua- and, therefore, taking a counter measure and evaluating its
tion of usage results, causes excessive requirement of regula- eŠect are not so easy. Therefore, this is the subject to be
tion and test not having scientiˆc rationality. As the results, challenged calling upon wisdom of the related parties among
eŠective and prompt supply of an important drug, already the society of drug.
having enough accumulation of experiences, to the medical Talking about the unknown risk, there are view points
work front will be hampered, and the drug, a social asset, that say ``It is risky because it is unknown.'' and ``What are
may not to be utilized eŠectively. Medicine is a sword used in the unknowns, and how do we cope with them in ensuring
medical ˆeld having double-edge named eŠectiveness and safety?''.
safety. EŠectiveness and safety factors have to be derived as The view of ``It is risky because it is unknown.'' is already
the fruits of leading edge of science, and relatively evaluated nothing but a sort of evaluation result, and directly connects
on a balance sheet of usefulness. Usefulness evaluation to a ˆnal decision if it can be used as a drug. Such evaluation
should not be unbalanced in a way that too much emphasis W decision has to be made based upon a rational, scientiˆc or
is placed on safety concern without back-up of appropriate social judgment.
scientiˆc rationality. A drug can play an important role as a For example, in the case that ``In a manufacturing process
social asset only when well balanced appropriate scientiˆc of drug, virus, virus-like particle or retrovirus was detected,
usefulness evaluation in addition to social concern of the age but its identiˆcation could not be conˆrmed, and, therefore,
are given. In other words, drug is a common asset utilized by its risk can not be denied.'', the evaluation of ``It is risky be-
society for medication as a fruit of science of the age, and cause it is unknown.'' is scientiˆcally rational and reasona-
the key point of its utilization lies on a balance of risk and ble. On the other hand, however, if we reach a decision of
beneˆt produced from scientiˆc and social evaluation. So, ``It is risky because it is unknown.'' due to the reason that
those factors have to be taken into account when target and ``In a manufacturing process of drug, virus, virus-like parti-
pursuance levels for ensuring viral safety of a JP listed cle or retrovirus was not detected, but there is a `concern'
biotechnological W biological product are reviewed. that something unknown may exist.'', it can not be said that
And, in general, the risk and beneˆt of drugs should be such evaluation is based upon a rational, scientiˆc or social
Supplement I, JP XIV General Information 1621

judgment. It goes without saying that the utmost care has to ures can not be taken on the subject of ``unknown virus,
be taken for viral safety, but the substance of `concern' has which can not be detected by currently available screening
to be at least clearly explainable. Otherwise, the `concern' method, may exist'', and conducting any virus detection test
may result in causing contradiction in the meaningful mis- at any stage will be useless ``for ensuring safety''.
sion to utilize a social asset, drug, in medical practice. The requirement of regulations or tests excessively over
From scientiˆc view point, we should not be narrow mind- scientiˆc rationality will raise human, economical and tem-
ed by saying ``it is risky'' because ``there is a `concern' that poral burden to the pharmaceutical companies, and will ad-
something unknown may exists'', but challenge to clarify the versely aŠect prompt, eŠective and economical supply of a
subject of ``What is unknown, and how to cope with it for drug to the medical front. As drug is a sort of social asset,
ensuring safety'' using wisdom. What is important at the which has to be scientiˆcally evaluated, how to assure max-
time is to deˆne ``what is unknown'' based upon current imization of its safety by means of scientiˆcally rational ap-
scientiˆc knowledge. Only through this way, is it possible for proach at minimum human, economical and time resources
us to elaborate the measures for ensuring safety. is important.
Once we chase up the substance of unknown risk for viral It is also important to reconˆrm that achievement of those
safety without premises of ``what is unknown'', issues is on the premise that appropriate measures are taken
``unknown'' will be an endless question because it theoreti- on the supply source of drugs. For example, in a case of ``In
cally remains unresolved forever. If this kind of approach is a manufacturing process of drug, virus, virus-like particle or
taken, the issue and the measure can not be scientiˆcally retrovirus was not detected, but there is a `concern' that
connected to each other, which will result in the excessive re- something unknown may exist.'', appropriateness of the
quirement of regulation and of test to be conducted. Yet, it test, which resulted in the judgment that ``virus, virus-like
is unlikely that the measure which has no relation with particle or retrovirus was not detected in a process of drug
science will be eŠective to the subject of ``What is unknown production'', should be a prerequisite premise when judged
is unknown.'' by science standard ate the time. If there is any question on
For example, ``what is unknown'' at the ``evaluation of a the premise, it is quite natural that the question of ``there is a
puriˆcation process which can completely clear up every vi- `concern' that unknown something may exist.'' will be eŠec-
rus that contaminated in a manufacturing process'' should tive.
be the subject of ``what sort of existing virus that contami- 3.4 Applicable range
nated is unknown'', not on the subject of ``what sort of This General Information is on JP listed biological
virus that exist in the world is unknown. In the former sub- products, derived from living tissue or body ‰uid, and pro-
ject, the premise of the study is based on all the knowledge tein drugs, derived from human or animal cell line, that in
on viruses including DNA W RNA-virus, virus with W without Japan. In the case of protein drugs derived from human or
envelope, particle size, physical W chemical properties, etc. animal cell line, the products developed and approved be-
The premise is that the virus contaminated should be within fore enforcement of the Notice Iyakushin No. 329 entitled
range of existing wisdom and knowledge of virus such as ``Viral safety evaluation of biotechnology products derived
species, type, nature, etc., even though the virus that con- from cell lines of human or animal origin'' should have been
taminated is unknown. Under such premise, when evalua- treated under the Notice had there been one, and it is inevita-
tion is made on a puriˆcation process to decide its capability ble that some products approved after the Notice might not
of clearing a derived virus, which is within the range of exist- have been su‹ciently treated. It is expected that such
ing wisdom and learning, speciˆc viral clearance studies biodrug will be su‹ciently examined to meet such General
designed to combine a few model viruses with diŠerent na- Information before being listed in JP. On the other hand,
tures, such as type of nucleic acid, with W without envelope, blood preparations listed in the biological products standard
particle size, physical Wchemical properties, etc., would be and covered by ``Guideline for securing safety of blood plas-
enough to simulate every sort of the virus already known, ma protein fraction preparations against virus'', are out of
and will be a ``good measure for ensuring safety''. the scope of this General Information. Further, in case of a
The issue of ``the sort of viruses that exist in the world is relatively lower molecular biogenous substance, such as ami-
unknown'' may be a future study item, but it is not an ap- no acid, saccharide and glycerin, and of gelatin, which is
propriate subject for the viral clearance test. Further, even if even classiˆed as infectious or pathogenic polymer, there are
the subject of ``unknown viruses, which have a particle size cases that viral contamination can not be considered due to
smaller than that of currently known viruses, may exists'' or its manufacturing or puriˆcation process, and that potent
``unknown viruses, which have special physical W chemical viral inactivation Wremoval procedure that can not be applied
properties that can not be matched to any of the currently to protein, can be used, and, therefore, it is considered
known viruses, may exists'' is set up as an armchair theory, reasonable to omit such substances from the subject for ap-
any experimental work can not be pursued under the current plication. However, some part of this General Information
scientiˆc level, since such virus model is not available. Fur- may be used as reference. Further, a comprehensive assur-
ther, any viral clearance test performed by using the current- ance measure for viral safety is recommendable on a
ly available methods and technologies will be meaningless biotechnological W biological product not listed in JP using
``for ensuring safety'', since particle size or natures of such this document as a reference so long as it is similar to the
speculated virus are unknown. Likewise, any counter meas- biotechnological W biological product JP.
1622 General Information Supplement I, JP XIV

3.5 Possible viral contamination to a JP listed biotechno- products. The pathogenic infectious viruses, currently
logical Wbiological product (source of virus contamination) known to contaminate to raw materials, etc. of drug and
Promoting awareness of virus contamination to a JP list- have to be cautioned, are HIV, HAV, Hepatitis B Virus
ed biotechnological W biological product (source of virus con- (HBV), HCV, Human T-Lymphotropic Virus (HTLV-I W II),
tamination) and citing countermeasure thereof are im- Human Parvovirus B19, Cytomegalovirus (CMV), etc.
portant for eradicating any possible virus contamination and Biotechnological W biological products produced from raw
raising probability of safety assurance. Many biotechnologi- material W cell substrate derived from tissue or body ‰uid of
cal Wbiological products are produced from a ``substrate'' human or animal origin always have a risk of contamination
which is derived from human or animal tissue, body ‰uid, of pathogenic or other latent virus. Therefore, safety meas-
etc. as an origin W raw material, and in puriˆcation or phar- ures should be thoroughly taken. There is also the case that a
maceutical processing of such products column materials or material, other than the biological component such as raw
additives, which are living organism origin, are occasionally material W substrate, causes virus contamination. Using en-
used. Accordingly, enough safety measures should be taken zymatic or monoclonal antibody column or using albumin
against diŠusion of the contaminant virus. Further, as men- etc. as a stabilizer, is the example of the case, in which cau-
tioned in Notice Iyakushin No. 329, any protein drug der- tion has to be taken on risk of virus contamination from the
ived from cell lines of human or animal origin should be source animal or cell. Further, there is a possibility of con-
carefully examined with respect to the risk of virus contami- tamination from environment or personnel in charge of
nation through the cell line, the cell substrate for drug production or at handling of the product. So, caution has to
production, and through the manufacturing process applied be taken on these respects as well.
thereafter. In case of protein drugs derived from cell line of human or
``Substrate for drug production'' is deˆned as a starting animal origin, there may be cases where latent or persistent
material which is at a stage where it is deemed to be in a posi- infectious viruses (e.g., herpesvirus) or endogenous
tion to ensure quality W safety of an active substance. The retroviruses exist in the cell. Further, adventitious viruses
``substrate for drug production'' is sometimes tissue, body may be introduced through the routes such as: 1) derivation
‰uid, etc. of human or animal per se and pooled material of a cell line from an infected animal; 2) use of a virus to
such as urine, and sometimes a material after some treat- drive a cell line; 3) use of a contaminated biological reagent
ment. In many cases, it is considered rational that starting (e.g., animal serum components); 4) contamination during
point of full-scale test, evaluation and control should be at cell handling. In the manufacturing process of drug, an
the stage of ``substrate for drug production''. The more adventitious virus may contaminate to the ˆnal product
strict levels of test, evaluation and control achieved at the through the routes, such as 1) contamination through a rea-
stage of ``substrate for drug production'' can more rational- gent of living being origin, such as serum component, which
ize evaluation and control of the raw material or individual is used for culturing, etc.; 2) use of a virus for introduction
level of upper stream. On the contrary, strict evaluation and of a speciˆc gene expression to code an objective protein; 3)
control of the raw material or individual level at an upper contamination through a reagent used for puriˆcation such
stream stage can rationalize tests, evaluation or quality con- as monoclonal antibody a‹nity column; 4) contamination
trol at the stage of ``substrate for drug production''. through an additive used for formulation production; 5)
The measures taken for ensuring viral safety on a contamination at handling of cell and culture media, etc. It
biotechnological W biological product currently listed in JP is reported that monitoring of cell culture parameter may be
can be assumed from the provisions of manufacturing helpful for early detection of an adventitious viral contami-
method, speciˆcation and test methods of each preparation. nation.
However, unitary principles or information with respect to 3.6 Basis for ensuring viral safety
the measures to be taken for ensuring viral safety, totally Viral safety of a biotechnological W biological product pro-
reviewing the entire process up to the ˆnal product rationally duced from a raw material W substrate, which derived from
and comprehensively, including source W raw material W sub- tissue, body ‰uid, cell line, etc. of human or animal origin,
strate, puriˆcation process, etc. have not been clariˆed. The can be achieved by supplemental and appropriate adoption
most important thing for ensuring viral safety is to take of the following plural methods.
thorough measures to eliminate the risk of virus contamina- (1) Acquaintance of possible virus contamination (source
tion at any stage of source animal, raw material and sub- of contamination).
strate. Although not the cases of a biotechnological W biologi- (2) Careful examination of eligibility of the raw material
cal product, known examples of a virus contamination from and its source, i.e., human or animal, thorough analy-
a raw material W substrate for drug production in old times sis and screening of the sample chosen as the substrate
are Hepatitis A Virus (HAV) or Hepatitis C Virus (HCV) for drug production to determine virus contamination
contamination in blood protein fraction preparations. It is and through examination of the type of virus and its
also well known that Human Immunodeˆciency Virus (HIV) nature, if contaminated.
infection caused by blood plasma protein fraction prepara- (3) Evaluation to determine hazardous properties of the vi-
tions occurred in 1980s. The aim of this General Informa- rus or virus-like particle to human, if exists.
tion is to show concrete guidelines for comprehensive viral (4) Choosing a product related material of living organism
safety assurance of the JP listed biotechnological W biological origin (e.g., reagent, immune anti-body column, etc.)
Supplement I, JP XIV General Information 1623

which is free from infectious or pathogenic virus. size, shape, with or without envelope, type of nucleic acid
(5) Conduct virus free test at an appropriate stage of (DNA type, RNA type), heat and chemical treatment toler-
manufacturing including the ˆnal product, if necessary. ance, etc., with an aim to determine removal W inactivation
(6) Adoption of an eŠective method to remove W inactivate capability of the virus that can not be detected in a raw
the virus in the manufacturing process for viral clear- material or contingently contaminated.
ance. Combined processes sometimes achieve higher As mentioned above, the role of viral clearance study is to
level of viral clearance. speculate removal W inactivation capability of a process
(7) Develop a deliberate viral clearance scheme. through a model test, and it contributes to give scientiˆc ba-
(8) Conduct test and evaluation to conˆrm removal W inacti- sis to assure that a biotechnological W biological product of
vation of the virus. human or animal origin has reached an acceptable level in
Manufacturer is responsible for explaining rationality of aspect of viral safety.
the way of approach adopted among the comprehensive At a viral clearance study, it is necessary to adopt an ap-
strategy for viral safety on each product and its manufactur- propriate approach method which is deˆnitive and rational
ing process. At the time, the approach described in this and can assure viral safety of a ˆnal product, taking into
General Information shall be applicable as far as possible. consideration the source and the properties of the raw
3.7 Limit of virus test material Wsubstrate as well as the manufacturing process.
Virus test has to be conducted to deˆne existence of virus,
4. Raw material W substrate for drug production
but it should be noted that virus test alone can not reach a
4.1 Issues relating to animal species and its region as a
conclusion of inexistence of virus nor su‹cient to secure
source of raw material W substrate for drug production and
safety of the product. Examples of a virus not being detected
countermeasures to be taken thereto
are as follows: 1) Due to statistical reason, there is an inher-
For manufacturing JP listed biotechnological W biological
ent quantitative limit, such as detection sensitivity at lower
products, which require measures for viral safety, a raw
concentration depends upon the sample size. 2) Generally,
material Wsubstrate derived mainly from human, bovine,
every virus test has a detection limit, and any negative result
swine or equine is used, and it is obvious that such human
of a virus test can not completely deny existence of a virus.
and animal has to be healthy nature. A wild animal should
3) A virus test applied is not always appropriate in terms of
be avoided, and it is recommended to use animals derived
speciˆcity or sensitivity for detection of a virus which exists
from a colony controlled by an appropriate SPF (Speciˆc
in the tissue or body ‰uid of human or animal origin.
Pathogen-Free) condition and bred under a well deigned
Virus testing method is improved as science and technolo-
hygienic control, including appropriate control for preven-
gy progress, and it is important to apply scientiˆcally the
tion of microbial contamination and contamination
most advanced technology at the time of testing so that it
monitoring system. If a meat standard for food is available,
can be possible to raise the assurance level of virus detection.
an animal meeting this standard has to be used. The type of
It should be noted, however, that the limit as mentioned
virus to be concerned about depend on animal species, but it
above can not always be completely overcome. Further, risk
may be possible to narrow down the virus for investigation
of virus contamination in a manufacturing process can not
by means of examining the hygiene control, applicability of
be completely denied, and, therefore, it is necessary to
a meat standard for food, etc. On the other hand, even with
elaborate the countermeasure taken these eŠects into ac-
the animals of the same species, a diŠerent approach may be
count.
necessary depending upon the region where the specimen for
Reliable assurance of viral free ˆnal product can not be
a raw material W substrate is taken. For example, in case of
obtained only by negative test results on the raw material W
obtaining raw material W substrate from blood or other
substrate for drug production or on the product in general, it
speciˆc region, it is necessary to be aware of the risk level, vi-
is also necessary to demonstrate inactivation W removal
rus multiplication risk, etc. which may speciˆcally exists de-
capability of the puriˆcation process.
pending upon its region. Such approach may be diŠerent
3.8 Roles of viral clearance studies
from those applied to body waste such as urine, milk, etc. as
Under the premises as mentioned in the preceding clause
a source of raw material W substrate. Further, caution has to
that there is a limit of a virus test, that there is a possibility
be taken on transmissible spongiform encephalopathy (TSE)
of existence of latent virus in a raw material W substrate for
when pituitary gland, etc. is used as a raw material. This
drug production and that there is a risk of entry of a non-en-
report does not include detailed explanation on TSE, but
dogenous virus in a manufacturing process, one of the im-
recommendations are to use raw material derived from 1)
portant measures for viral safety is how to remove or inacti-
animals originated in the countries (area) where incidence of
vate the virus, which exists in a raw material, etc. and can
TSE has not been reported; 2) animals not infected by TSE;
not be detected, or the virus, which is contingently contami-
or 3) species of animal which has not been reported on TSE.
nated in a manufacturing process. The purpose of viral
It is recommended to discuss the matters concerned with
clearance study is to experimentally evaluate the viral
TSE with the regulatory authority if there is any unclear
removal W inactivation capability of a step that mounted in a
point.
manufacturing process. So, it is necessary to conduct an ex-
Followings are the raw material W substrate used for
perimental scale spike test using an appropriate virus that is
manufacturing biotechnological W biological products in
selected by taking account the properties, such as particle
1624 General Information Supplement I, JP XIV

Japan. ˆrmed of its health by various tests. Further, it is necessary


(1) Biological products derived from human that the population, to which the animal belongs, has been
Blood plasma, placenta, urine, etc. derived from human under an appropriate breeding condition, and that no abnor-
are used as the sources of raw material of biotechnological W mal individual has been observed in the population. Further,
biological product. As for these raw materials, there are 2 it is necessary to demonstrate information or scientiˆc basis
cases: 1) Appropriateness can be conˆrmed by interview or which can deny known causes infection or disease to human,
by examination of the individual who supplies each raw or to deny such animal inherent latent virus by serologic test
material, and 2) Such su‹cient interview or examination of or by nucleic ampliˆcation test (NAT). The infectious virus
the individual can not be made due to type of raw material. that is known to be common between human and animal,
In case that su‹cient examination of individual level is not and known to cause infection in each animal are tentatively
possible, it is necessary to perform test to deny virus con- listed in Table 2. It is necessary that the table is completed
tamination at an appropriate manufacturing stage, for ex- under careful examination, and denial of all of them, by
ample, the stage to decide it as a substrate for drug produc- means of tests on individual animal, tissue, body ‰uid, etc.
tion. as a raw material, or on pooled raw material (as a direct sub-
(2) Biological products derived from animal besides hu- strate for drug production), is not always necessary. Table 2
man can be used as reference information, in addition to the
Insulin, gonadotropin, etc. are manufactured from blood other information, such as; source of animal, health condi-
plasma or from various organs of bovine, swine and equine. tion, health and breeding control, conformity to the meat
(3) Protein drug derived from cell line of human or animal standard for food, etc., to elaborate to which virus what
origin kind of test has to be performed, and for which virus it is not
It is expected that protein drugs derived from cell line of always necessary to test for, etc. It is important to clarify
human or animal origin will be listed in JP in a near future. and record the basis of choosing the virus and the test con-
In the case of these products, a cell line of human or animal ducted thereof.
is the raw material per se, and the substrate for drug produc-
Table 2. Infectious viruses known to be common
tion is a cell bank prepared from cloned cell line (master cell
between human and animal and known to
bank or working cell bank). Examination at cell bank level is
cause infection to each animal
considered enough for viral safety qualiˆcation, but it goes
without saying that the more appropriate and rational bovine swine sheep goat equine
qualiˆcation evaluation test of cell bank can be realized
when more information is available on the virus of the Cowpox virus w
source animal or on prehistory of driving the cell line, the Paravaccinia virus w w w w
base of cell bank.
4.2 Qualiˆcation evaluation test on human or animal as a Murry valley encephalitis w w
virus
source of raw material W substrate for drug production
(1) Biological products derived from human Louping-ill virus w w w w
Body ‰uid etc. obtained from healthy human must be
Wesselsbron virus w
used for biological products production. Further, in case
that interview or examination of the individual, who sup- Foot-and-mouth disease w w
plies the raw material, can be possible and is necessary, in- virus
terview under an appropriate protocol and a serologic test Japanese encephalitis w
well evaluated in aspects of speciˆcity, sensitivity and ac- virus
curacy have to be performed, so that only the raw material,
Vesicular stomatitis virus w
which is denied latent HBV, HCV and HIV, will be used. In
addition to the above, it is necessary to test for gene of HBV, Bovine papular stomatitis w
HCV and HIV by a nucleic ampliˆcation test (NAT) well virus
evaluated in aspects of speciˆcity, sensitivity and accuracy. Orf virus w
In case of the raw material (e.g., urine), which can not be
tested over the general medical examination of the individual Borna disease virus w w
who supplies the material, or of the raw material which is ir- Rabies virus w w w w w
rational to conduct individual test, the pooled raw material,
as the substrate for drug production, has to be conducted at In‰uenza virus w
least to deny existence of HBV, HCV and HIV, using a Porcine hepatitis E virus w
method well evaluated in aspects of speciˆcity, sensitivity
and accuracy, such as the antigen test or NAT. Encephalomyocarditis w w
virus
(2) Biological products derived from animal besides hu-
man Rota virus w
The animal used for manufacturing biological product has
to be under appropriate health control, and has to be con-
Supplement I, JP XIV General Information 1625

Further, it is necessary to conduct an appropriate adventiti-


Eastern equine w ous virus test (e.g., in vitro and in vivo test) and a latent en-
encephalitis virus
dogenous virus test on the cell at the limit of in vitro cell age
Western equine w (CAL) for drug production. Each WCB as a starting cell
encephalitis virus substrate for drug production should be tested for adventiti-
Venezuelan equine ous virus either by direct testing or by analysis of cells at the
w
encephalitis virus CAL, initiated from the WCB. When appropriate non-en-
dogenous virus tests have been performed on the MCB and
Morbillivirus w
cells cultured up to or beyond the CAL have been derived
Hendravirus w from the WCB and used for testing for the presence of
adventitious viruses, similar tests need not be performed on
Nipahvirus w
the initial WCB.
Transmissible gastroente- w
ritis virus 5. Points of concern with respect to manufacturing and
virus testing
Porcine respiratory w To ensure viral safety of a biological product derived from
coronavirus
tissue, body ‰uid etc. of human or animal origin, it is neces-
Porcine epidemic w sary to exclude any possibility of virus contamination from a
diarrheavirus raw material, such as tissue and body ‰uid, or a substrate,
Hemagglutinating paying attention to the source of virus contamination as
w
encephalomyelitis virus mentioned in above 3.5, and to adopt appropriate manufac-
turing conditions and technologies in addition to enhance-
Porcine respiratory and w
reproductive complex virus ment of manufacturing environment, so that virus contami-
nation in the course of process and handling and from oper-
Hog cholera virus w ators, facilities and environment can be minimized.
Parain‰uenza 3 virus w In addition to the above, eŠective virus test and viral inac-
tivation Wremoval technology, which are re‰ected by rapid
Telfan W
Teschen disease w progress of science, have to be introduced. Adoption of two
virus
or more steps with diŠerent principles is recommended for
Reoviruses w virus inactivation W removal process. Further, it is important
to minimize any possible virus derivation by using a reagent,
Endogenous retrovirus w
which quality is equivalent to that of a drug. Examples of vi-
Porcine adenovirus w rus inactivation W removal processes are  heating (It is
reported that almost viruses are inactivated by heating at 55
Porcine circovirus w
– 609C for 30 minutes with exceptions of hepatitis virus, etc.
Porcine parvovirus w and that dry heating at 609 C for 10 – 24 hours is eŠective in
case of the products of blood or urine origin.),  treatment
Swine poxvirus w
with organic solvent W surfactant (S WD treatment),  mem-
Porcine cytomegalovirus w brane ˆltration (15 – 50 nm),  acid treatment,  irradia-
tion (g-irradiation, etc.),  treatment with column chro-
Pseudorabies virus w
matograph (e.g. a‹nity chromatography, ion-exchange
Russian spring summer w w chromatography),  fractionation (e.g. organic solvent or
encephalitis virus ammonium sulfate fractionation),  extraction.
Rift Valley fever virus w w 5.1 Virus test conducted in advance of puriˆcation process
(1) Biological products derived from human
Crimean-Congo hemor- In many cases, samples for virus test before puriˆcation
rhagic fever virus w w w
(Nairovirus) process are body ‰uid or tissue of individual collected as a
raw material, or its pooled material or extraction as a sub-
Torovirus w strate. As mentioned in 4.2 (1), it is necessary to deny latent
HBV, HCV and HIV by the test evaluated enough in aspects
(3) Protein drug derived from cell line of human or animal of speciˆcity, sensitivity and accuracy. Even in a case that a
origin non-puriˆed bulk before puriˆcation process is produced
It is important to conduct thorough investigation on latent from a substrate, it is not always necessary to conduct virus
endogenous and non-endogenous virus contamination in a test again at the stage before puriˆcation, so long as the
master cell bank (MCB), which is the cell substrate for drug presence of any latent virus can be denied at the stage of sub-
production, in accordance with the Notice Iyakushin No. strate by an appropriate virus test, with cases where the non-
329 entitled ``Viral safety evaluation of biotechnology puriˆed bulk is made from the substrate by adding any rea-
products derived from cell lines of human or animal origin''. gent etc. of living organisms origin are an exception.
1626 General Information Supplement I, JP XIV

(2) Biological products derived from animal besides appropriate, a NAT test or other suitable methods may be
human used.
Similar to (1) above, samples for virus test before puriˆca- Generally, harvest material in which adventitious virus
tion process are, in many cases, body ‰uid or tissue of in- has been detected should not be used to manufacture the
dividual collected as a raw material, or its pooled material or product. If any adventitious viruses are detected at this level,
extraction as a substrate. In these cases, it is necessary to the process should be carefully checked to determine the
have a data, which can deny latent virus of probable cause of cause of the contamination, and appropriate actions taken.
human infection or disease as mentioned in the above 4.2 5.2 Virus test as an acceptance test of an intermediate
(2), or to have a result of serologic test or nucleic ampliˆca- material, etc.
tion test (NAT) evaluated enough in aspects of speciˆcity, When a biological product is manufactured from tissue,
sensitivity and accuracy. The concept, which is applied to a body ‰uid etc. of human or animal origin, there are cases
case that non-puriˆed bulk before puriˆcation process is that an intermediate material, partially processed as a raw
produced from substrate, is the same as those provided in material or substrate by outside manufacturer, is purchased
the above 4.2 (1). and used for manufacturing. In such case, if any test to meet
(3) Protein drug derived from cell line of human or animal this General Information has been conducted by such out-
origin side manufacturer, it is necessary for the manufacturer of
Generally, substrate in this case is cell bank, and the sam- the biological product, who purchased the intermediate
ple for testing before puriˆcation process is a harvested cell material, to examine what sort of virus test has to be con-
after cell culturing or unprocessed bulk which consists of ducted as acceptance tests, and to keep record on the basis of
single or pooled complex culture broth. The unprocessed rationality including the details of the test conducted.
bulk may be sometimes culture broth without cell. Denial of On the other hand, if no test to meet this General Infor-
latent virus, which is determined by virus test at a MCB or mation has been conducted by such outside manufacturer of
WCB level, does not always deny latent virus in unprocessed the raw material, all necessary virus free test has to be con-
bulk after culturing. Further, it is noted that the viral test at ducted to meet this General Information on the intermediate
the CAL is meaningful as a validation but can not guarantee material regarding it as the direct substrate for drug produc-
deˆnite assurance of latent virus denial, since the test is tion.
generally performed only once. In case of using a serum or a 5.3 Virus test on a ˆnal product
component of blood origin in a culture medium, deˆnite Virus tests to be conducted on a ˆnal product (or on a
denial of latent virus at the level of unprocessed bulk can not product to reach the ˆnal product) has to be deˆned under
be assured so long as the viral test has not been conducted on comprehensive consideration of the type of raw material or
each lot at the CAL, since lot renewal can be a variable fac- substrate, the result of virus test conducted on raw material W
tor on viral contamination. substrate, the result of evaluation on viral removal Winactiva-
A representative sample of the unprocessed bulk, removed tion process, any possibility of virus contamination in the
from the production reactor prior to further processing, manufacturing process, etc. Comprehensive viral safety as-
represents one of the most suitable levels at which the pos- surance can only be achieved by appropriate selection of the
sibility of adventitious virus contamination can be deter- raw material W substrate, an appropriate virus test conducted
mined with a high probability of detection. Appropriate test- on the raw material W substrate W intermediate material, the
ing for viruses should be performed at the unprocessed bulk virus test conducted at an appropriate stage of manufactur-
level unless virus testing is made more sensitive by initial par- ing, an appropriate viral clearance test, etc. However, there
tial processing (e.g., unprocessed bulk may be toxic in test are cases of having speciˆc backgrounds, such as 1) use of
cell cultures, whereas partially processed bulk may not be the raw material derived from unspeciˆed individual human,
toxic). In certain instances it may be more appropriate to test 2) possible existence of virus at window period, 3) speciˆc
a mixture consisting of both intact and disrupted cells and detection limit of virus test, etc. and in these cases, virus
their cell culture supernatants removed from the production contamination to the ˆnal product may occur if there is any
reactor prior to further processing. deˆciency on the manufacturing process (e.g., damage of
In case of unprocessed bulk, it is required to conduct virus membrane ˆlter) or any mix-up of the raw materials, etc. To
test on at least 3 lots obtained from pilot scale or commercial avoid such accidental virus contamination, it may be recom-
scale production. It is recommended that manufacturers mended to conduct nucleic ampliˆcation test (NAT) on the
develop programs for the ongoing assessment of adventiti- ˆnal product focusing on the most risky virus among those
ous viruses in production batches. The scope, extent and fre- that may possibly to exist in the raw material.
quency of virus testing on the unprocessed bulk should be
6. Process evaluation on viral clearance
determined by taking several points into consideration in-
6.1 Rationale, objective and general items to be concerned
cluding the nature of the cell lines used to produce the
with respect to viral clearance process evaluation
desired products, the results and extent of virus tests per-
Evaluation of a viral inactivation Wremoval process is im-
formed during the qualiˆcation of the cell lines, the cultiva-
portant for ensuring safety of a biological product derived
tion method, raw material sources and results of viral clear-
from tissue or body ‰uid of human or animal origin. Con-
ance studies. Screening in vitro tests, using one or several cell
ducting evaluation on viral clearance is to assure, even to
lines, are generally employed to test unprocessed bulk. If
Supplement I, JP XIV General Information 1627

Table 3. Example of viruses which have been used for viral clearance studies

Virus Family Genus Natural host Genome Env Size (nm) Shape Resistance

Vesicular Stomatitis Virus Rhabd Vesiculovirus Equine RNA yes 70 × 150 Bullet Low
Bovine
Parain‰uenza Virus Paramyxo Paramyxovirus Various RNA yes 100 – 200+ Pleo-Spher Low
MuLV Retro Type C Mouse RNA yes 80 – 110 Spherical Low
oncovirus
Sindbis Virus Toga Alphavirus Human RNA yes 60 – 70 Spherical Low
BVDV Flavi Pestivirus Bovine RNA yes 50 – 70 Pleo-Spher Low
Pseudorabies Virus Helpes Swine DNA yes 120 – 200 Spherical Med
Poliovirus Sabin Type 1 Picorna Enterovirus Human RNA no 25 – 30 Icosahedral Med
Encephalomyocardititis Picorna Cardiovirus Mouse RNA no 25 – 30 Icosahedral Med
Virus
Reovirus 3 Reo Orthoreovirus Various kind RNA no 60 – 80 Spherical Med
SV 40 Papova Polyomavirus Monkey DNA no 40 – 50 Icosahedral Very high
Parvovirus: canine, por- Parvo Parvovirus Canine DNA no 18 – 24 Icosahedral Very high
cine Porcine

some extent, elimination of the virus, which may exist in a to physical W chemical treatment, etc. and it is necessary to
raw material, etc. or may be derived to the process due to combine about 3 model viruses to cover these characteristics.
unexpected situation. Viral clearance studies should be made At choice of a model virus, there are also the ways to
by a carefully designed appropriate method, and has to be choose a virus closely related to or having the same charac-
rationally evaluated. teristics of the virus known to exist in the raw material. In
The objective of viral clearance studies is to assess process such case, it is in principle recommendable to choose a virus
step(s) that can be considered to be eŠective in inactivating W which demonstrates a higher resistance to inactivation W
removing viruses and to estimate quantitatively the overall removal treatment if two or more candidate viruses are
level of virus reduction obtained by the process. This should available for choice. Further, a virus which can grow at a
be achieved by the deliberate addition (``spiking'') of sig- high titer is desirable for choice, although this may not al-
niˆcant amounts of a virus at diŠerent manufacturing W ways be possible. In addition to the above, choosing a virus,
puriˆcation steps and demonstrating its removal or inactiva- which will provide eŠective and reliable assay result at each
tion during the subsequent steps. It is not necessary to evalu- step, is necessary, since sample condition to be tested at each
ate or characterize every step of a manufacturing process if step of a production process may in‰uence the detection sen-
adequate clearance is demonstrated by the use of fewer sitivity. Consideration should also be given to health hazard
steps. It should be borne in mind that other steps in the proc- which may pose to the personnel performing the clearance
ess may have an indirect eŠect on the viral inactivation W studies.
removal achieved. Manufacturers should explain and justify For the other items taken for consideration at choice of
the approach used in studies for evaluating viral clearance. virus, the Notice, Iyakushin No. 329 can be used as a refer-
The reduction of virus infectivity may be achieved by ence. Examples of the virus which have been used for viral
removal of virus particles or by inactivation of viral infectiv- clearance studies are shown in Table 3 which was derived
ity. For each production step assessed, the possible mechan- from Iyakushin No. 329. However, the Notice, Iyakushin
ism of loss of viral infectivity should be described with No. 329, is on viral safety of a product derived cell line of
regard to whether it is due to inactivation or removal. For human or animal origin, and a more appropriate model
inactivation steps, the study should be planned in such a way virus has to be chosen taking into account the origin W raw
that samples are taken at diŠerent times and an inactivation material of biological products.
curve constructed. 6.3 Design of viral clearance studies
6.2 Selection of virus The purpose of viral clearance studies is to quantitatively
To obtain broad range of information of viral inactivation evaluate removal or inactivation capability of a process, in
W removal, it is desirable that a model virus used for viral which a virus is intentionally spiked to a speciˆc step of a
clearance studies should be chosen from the viruses with manufacturing process.
broad range of characteristics in aspects of DNA W RNA, Following are the precautions to be taken at planning viral
with or without envelope, particle size, signiˆcant resistance clearance studies.
1628 General Information Supplement I, JP XIV

(1) Care should be taken in preparing the high-titer virus crude material or intermediate material should be spiked
to avoid aggregation which may enhance physical removal with infectious virus and the reduction factor calculated. It
and decrease inactivation thus distorting the correlation with should be recognized that virus inactivation is not a simple,
actual production. ˆrst order reaction and is usually more complex, with a fast
(2) Virus detection method gives great in‰uence to viral ``phase 1'' and a slow ``phase 2''. The study should, there-
clearance factor. Accordingly, it is advisable to gain detec- fore, be planned in such a way that samples are taken at
tion sensitivity of the methods available in advance, and use diŠerent times and an inactivation curve constructed. It is
a method with a detection sensitivity as high as possible. recommended that studies for inactivation include at least
Quantitative infectivity assays should have adequate sen- one time point less than the minimum exposure time and
sitivity and reproducibility in each manufacturing process, greater than zero, in addition to the minimum exposure
and should be performed with su‹cient replicates to ensure time. The reproducible clearance should be demonstrated in
adequate statistical validity of the result. Quantitative assays at least two independent studies. When there is a possibility
not associated with infectivity may be used if justiˆed. Ap- that the virus is a human pathogen, it is very important that
propriate virus controls should be included in all infectivity the eŠective inactivation process is designed and additional
assays to ensure the sensitivity of the method. Also, the data are obtained. The initial virus load should be deter-
statistics of sampling virus when at low concentrations (for mined from the virus which can be detected in the spiked
example, number of virus is 1-1000 W L) should be considered. starting material. If this is not possible, the initial virus load
(3) Viral clearance studies are performed in a miniature may be calculated from the titer of the spiking virus prepara-
size system that simulates the actual production process of tion. Where inactivation is too rapid to plot an inactivation
the biotechnological W biological product used by the curve using process conditions, appropriate controls should
manufacturer. It is inappropriate to introduce any virus into be performed to demonstrate that infectivity is indeed lost
a production facility because of GMP constraints. There- by inactivation.
fore, viral clearance studies should be conducted in a (8) If antibody against virus exists in an unprocessed
separate laboratory equipped for virological work and per- material, caution should be taken at clearance studies, since
formed by staŠ with virological expertise in conjunction it may aŠect the behavior of virus at viral removal or inacti-
with production personnel involved in designing and prepar- vation process.
ing a scaled-down version of the puriˆcation process. The (9) Virus spiked in unprocessed material should be
viral clearance studies should be performed under the basic su‹cient enough to evaluate viral removal or inactivation
concept of GLP. capability of the process. However, the virus ``spike'' to be
(4) Each factor on a viral clearance study of a process, added to the unprocessed material should be as small as pos-
which is performed in miniature size, should re‰ect that of sible in comparison with the sample volume of the unproc-
actual manufacturing as far as possible, and its rationality essed material so as not to cause characteristic change of the
should be clariˆed. In case of chromatograph process, material by addition of the virus nor to cause behavioral
length of column bed, linear velocity, ratio of bed volume change of the protein in the material.
per velocity (in other words, contact time), buŠer, type of (10) It is desirable that the virus in the sample is subject
column packing, pH, temperature, protein concentration, for quantitative determination without applying ultracen-
salt concentration and concentration of the objective trifuge, dialysis, storage, etc. as far as possible. However,
product are all correspondent to those of the actual produc- there may be a case that any handling before quantitative
tion. Further, similarity of elution proˆle should be test, such as remove procedure of inhibitor or toxic sub-
achieved. For the other process, similar concept should be stance, storage for a period to realize test at a time, etc., is
applied. If there is any factor which can not re‰ect the actual inevitable. If any manipulation, such as dilution, concentra-
production, its eŠect to the result should be examined. tion, ˆltration, dialysis, storage, etc., is applied for prepara-
(5) It is desirable that two or more inactivation W removal tion of the sample for testing, a parallel control test, which
processes of diŠerent principles are selected and examined. passes through a similar manipulation, should be conducted
(6) As for the process which is expected to inactivate W re- to assess infectivity variance at the manipulation.
move virus, each step should be evaluated in aspect of clear- (11) BuŠers and product (desired protein or other com-
ance capability, and carefully determined if it is the stage of ponent contained therein) should be evaluated independent-
inactivation, removal or their combination for designing the ly for toxicity or interference in assays used to determine the
test. Generally, in viral clearance test, a virus is spiked in virus titer, as these components may adversely aŠect the in-
each step which is the object of the test, and after passing dicator cells. If the solutions are toxic to the indicator cells,
through the process in question, the reduction level of infec- dilution, adjustment of the pH, or dialysis of the buŠer con-
tivity is evaluated. But, in some case, it is accepted that a taining spiked virus might be necessary. If the product itself
high potential virus is spiked at a step of the process, and has anti-viral activity, the clearance study may need to be
virus concentration of each succeeding step is carefully performed without the product in a ``mock'' run, although
monitored. When removal of virus is made by separation or omitting the product or substituting a similar protein that
fractionation, it is desirable to investigate how the virus is does not have anti-viral activity could aŠect the behaviour of
separated or fractionated (mass balance). the virus in some production steps.
(7) For assessment of viral inactivation, unprocessed (12) Many puriˆcation schemes use the same or similar
Supplement I, JP XIV General Information 1629

buŠers or columns, repetitively. The eŠects of this approach reference when viral clearance studies on biological products
should be taken into account when analyzing the data. The are designed.
eŠectiveness of virus elimination by a particular process may 6.4 Interpretation of viral clearance studies
vary with the stage in manufacture at which it is used. 6.4.1 Evaluation on viral clearance factor
(13) Overall reduction factors may be underestimated Viral clearance factor is a logarithm of reduction ratio of
where production conditions or buŠers are too cytotoxic or viral amount (infectious titer) between each step applied for
virucidal and should be discussed on a case-by-case basis. viral clearance of a manufacturing process. Total viral clear-
Overall reduction factors may also be overestimated due to ance factor throughout the process is sum of the viral clear-
inherent limitations or inadequate design of viral clearance ance factor of each step appropriately evaluated.
studies. Whether each and total viral clearance factor obtained are
(14) It has to be noted that clearance capability of viral acceptable or should not be evaluated in aspects of every
removal W inactivation process may vary depending upon the virus that can be realistically anticipated to derive into the
type of virus. The viral removal W inactivation process, which raw material or the manufacturing process, and its rationali-
displays viral clearance by speciˆc principle or mechanism, ty should be recorded.
may be quite eŠective to the virus, which meets such In case that existence of any viral particle is recognized in
mechanism of action, but not eŠective to the other type of a substrate for drug production, e.g., a substrate of rodent
viruses. For example, S W D treatment is generally eŠective to origin for biodrug production, it is important not only to
the virus with lipid membrane, but not eŠective to the virus demonstrate removal or inactivation of such virus, but also
not having such membrane. Further, some virus is resistant to demonstrate that the puriˆcation process has enough
to the general heating process (55 – 609C, 30 minutes). capability over the required level to assure safety of the ˆnal
When clearance is expected for such virus, introduction of a product at an appropriate level. The virus amount removed
further severe condition or process, which has diŠerent prin- or inactivated in a manufacturing process should be com-
ciple or mechanism, is necessary. Virus removal membrane pared with the virus amount assumed to exist in the substrate
ˆltration, which is diŠerent from S W D or heat treatment in etc. used for manufacturing drug, and for this purpose, it is
aspect of principle, is eŠective to a broad range of virus that necessary to obtain the virus amount in the raw materials W
can not pass through the membrane. A‹nity chro- substrate, etc. Such ˆgure can be obtained by measuring in-
matography process, which speciˆcally absorbs the objective fectious titer or by the other method such as transmission
protein, can thoroughly wash out the materials other than electron microscope (TEM). For evaluation of overall proc-
the objective protein including virus etc. and is generally ess, a virus amount, far larger than that assumed to exist in
eŠective for viral removal. Separation W fractionation of a the amount of the raw materials W substrate which is equiva-
virus from an objective protein is sometimes very di‹cult, lent to single administration of the ˆnal product, has to be
but there are not so rare that ion exchange chromatography, removed. It is quite rare that existence of virus can be as-
ethanol fractionation, etc. is eŠective for clearance of a virus sumed in a substrate for drug production, with the exception
which can not be su‹ciently inactivated or removed by the of the substrate of rodent origin, and such suspicious raw
other process. material W substrate should not be used for manufacturing
(15) EŠective clearance may be achieved by any of the drug with a special exceptional case that the drug in question
following: multiple inactivation steps, multiple complemen- is not available from the other process and is clinically in-
tary separation steps, or combinations of inactivation and dispensable, and that the information including infectious
separation steps. Separation methods may be dependent on properties of the virus particle assumed to exist has been
the extremely speciˆc physico-chemical properties of a virus clariˆed.
which in‰uence its interaction with gel matrices and precipi- 6.4.2 Calculation of viral clearance index
tation properties. However, despite these potential varia- Viral clearance factor, ``R'', for viral removal W inactiva-
bles, eŠective removal can be obtained by a combination of tion process can be calculated by the following formula.
complementary separation steps or combinations of inacti-
R = log[(V1 × T1) W
(V2 × T2)]
vation and separation steps. Well designed separation steps,
such as chromatographic procedures, ˆltration steps and In which
extractions, can be also eŠective virus removal steps pro- R: Logarithm of reduction ratio
vided that they are performed under appropriately con- V1: Sample volume of the unprocessed material
trolled conditions. T1: Virus amount (titer) of the unprocessed material
(16) An eŠective virus removal step should give V2: Sample volume of the processed material
reproducible reduction of virus load shown by at least two T2: Virus amount (titer) of the processed material
independent studies.
At the calculation of viral clearance factor, it is recom-
(17) Over time and after repeated use, the ability of
mendable to use the virus titer detected in the sample prepa-
chromatography columns and other devices used in the
ration of the unprocessed material after addition of virus,
puriˆcation scheme to clear virus may vary. Some estimate
not the viral titer added to the sample preparation wherever
of the stability of the viral clearance after several uses may
possible. If this is not possible, loaded virus amount is calcu-
provide support for repeated use of such columns.
lated from virus titer of the solution used for spike.
(18) The Notice, Iyakushin No. 329, would be used as a
1630 General Information Supplement I, JP XIV

6.4.3 Interpretation of results and items to be concerned at per mL by a factor of 8 log10 leaves zero log10 per mL or one
evaluation infectious unit per mL, taking into account the detection
At the interpretation and the evaluation of the data on limit of assay.
eŠectiveness of viral inactivation W removal process, there are (6) Variable factor of manufacturing process
various factors to be comprehensively taken into account, Minor variance of a variation factor of a manufacturing
such as  appropriateness of the virus used for the test,  process, e.g., contact time of a spiked sample to a buŠer or a
design of the viral clearance studies,  virus reduction ratio column, will sometimes give in‰uence to viral removal or in-
shown in logarithm,  time dependence of inactivation,  activation eŠect. In such case, it may be necessary to inves-
factors Witems which give in‰uence to the inactivation W tigate to what extent such variance of the factor has given in-
removal process,  sensitivity limit of virus assay method, ‰uence to the process concerned in aspect of viral inactiva-
 possible eŠect of the inactivation W removal process which tion.
is speciˆc to certain class of viruses. (7) Existence of anti-viral antiserum
Additional items to be concerned at appropriate interpre- Anti-viral antiserum that exists in the sample preparation
tation and evaluation of the viral clearance data are as fol- used for a test may aŠect sensitivity of distribution or inacti-
lows: vation of a virus, which may result in not only defusing the
(1) Behavior of virus used to the test virus titer but complicating interpretation of the test result.
At interpretation of the vial clearance results, it is necessa- So, existence of anti-viral antiserum is one of the important
ry to recognize that clearance mechanism may diŠer depend- variable factors.
ing upon the virus used for the test. Virus used for a test is (8) Introduction of a new process for removal W inactivation
generally produced in tissue culture, but behavior of the Viral clearance is an important factor for securing safety
virus prepared in the tissue culture may be diŠerent from of drug. In case that an achievement level of infective clear-
that of the native virus. Examples are possible diŠerences of ance of a process is considered insu‹cient, a process which
purity and degree of aggregation between the native and the is characterized by inactivation Wremoval mechanism to meet
cultured viruses. Further, change of surface properties of a the purpose or an inactivation W removal process which can
virus, e.g., addition of a sucrose chain which is ascribed to mutually complement to the existence process has to be in-
speciˆc nature of a separation process, may give eŠect to the troduced.
separation. These matters should be also considered at inter- (9) Limit of viral clearance studies
pretation of the results. Viral clearance studies are useful for contributing to the
(2) Design of test assurance that an acceptable level of safety in the ˆnal
Viral clearance test should have been designed taking into product is achieved but do not by themselves establish safe-
account variation factors of the manufacturing process and ty. However, a number of factors in the design and execu-
scaling down, but there still remain some variance from tion of viral clearance studies may lead to an incorrect esti-
actual production scale. It is necessary to consider such vari- mate of the ability of the process to remove virus infectivity,
ance at the interpretation of the data and limitation of the as described above.
test.
7. Statistics
(3) Acceptability of viral reduction data
The viral clearance studies should include the use of
Total viral clearance factor is expressed as a sum of
statistical analysis of the data to evaluate the results. The
logarithm of reduction ratio obtained at each step. The sum-
study results should be statistically valid to support the con-
mation of the reduction factor of multiple steps, particularly
clusions reached.
of steps with little reduction (e.g., below 1 log10), may
7.1 Statistical considerations for assessing virus assays
overestimate viral removal W inactivation capability of the
Virus titrations suŠer the problems of variation common
overall process. Therefore, virus titer of the order of 1 log10
to all biological assay systems. Assessment of the accuracy
or less has to be ignored unless justiˆed. Further, viral clear-
of the virus titrations and reduction factors derived from
ance factor achieved by repeated use of the same or similar
them and the validity of the assays should be performed to
method should be ignored for calculation unless justiˆed.
deˆne the reliability of a study. The objective of statistical
(4) Time dependence of inactivation
evaluation is to establish that the study has been carried out
Inactivation of virus infectivity frequently shows biphasic
to an acceptable level of virological competence.
curve, which consists of a rapid initial phase and subsequent
Assay
slow phase. It is possible that a virus not inactivated in a step
1. Assay methods may be either quantal or quantitative.
may be more resistant to the subsequent step. For example,
Both quantal and quantitative assays are amenable to
if an inactivated virus forms coagulation, it may be resistant
statistical evaluation.
to any chemical treatment and heating.
2. Variation can arise within an assay as a result of dilu-
(5) Evaluation of viral reduction ratio shown logarithm
tion errors, statistical eŠects and diŠerences within the assay
Viral clearance factor shown in logarithm of reduction
system which are either unknown or di‹cult to control.
ratio of virus titer can demonstrate drastic reduction of
These eŠects are likely to be greater when diŠerent assay
residual infectious virus, but there is a limit that infectious
runs are compared (between-assay variation) than when
titer can never be reduced to zero. For example, reduction in
results within a single assay run are compared (within-assay
infectivity of a preparation containing 8 log10 infectious unit
Supplement I, JP XIV General Information 1631

variation). inactivated virus still shows positive on nucleic acid. Fur-


3. The 95z conˆdence limits for results of within-assay ther, at introduction of NAT, cautions should be taken on
variation normally should be on the order of ±0.5 log10 of rationality of detection sensitivity, choice of a standard
the mean. Within-assay variation can be assessed by stan- which is used as run-control, quality assurance and main-
dard textbook methods. Between-assay variation can be tenance of a reagent used for primer, interpretation of posi-
monitored by the inclusion of a reference preparation, the tive and negative results, etc.
estimate of whose potency should be within approximately
10. Reporting and preservation
0.5 log10 of the mean estimate established in the laboratory
All the items relating to virus test and viral clearance stu-
for the assay to be acceptable. Assays with lower precision
dies should be reported and preserved.
may be acceptable with appropriate justiˆcation.
7.2 Reproducibility and conˆdence limit of viral clearance 11. Others
studies The Notice, Iyakushin No. 329, should be used as a refer-
An eŠective virus inactivation W removal step should give ence at virus test and viral clearance studies.
reproducible reduction of virus load shown by at least two
Conclusion
independent studies. The 95z conˆdence limits for the
As mentioned at the Introduction, assurance of quality W
reduction factor observed should be calculated wherever
safety etc. of JP listed drugs should be achieved by state-of-
possible in studies of viral clearance. If the 95z conˆdence
the-art methods and concepts re‰ecting the progress of
limits for the viral assays of the starting material are ±s, and
science and accumulation of experiences.
for the viral assays of the material after the step are ±a, the
The basis for ensuring viral safety of JP listed biotechno-
95z conˆdence limits for the reduction factor are
logical Wbiological products is detailed in this General Infor-
± s 2 + a 2.
mation. What is discussed here is that an almost equal level
8. Re-evaluation of viral clearance of measures are required for both development of new drugs
Whenever signiˆcant changes in the production or puriˆ- and for existing products as well, which means that similar
cation process are made, the eŠect of that change, both level of concerns should be paid on both existing and new
direct and indirect, on viral clearance should be considered products in aspect of viral safety. This document is intended
and the system re-evaluated as needed. Changes in process to introduce a basic concept that quality and safety assur-
steps may also change the extent of viral clearance. ance of JP listed product should be based upon the most ad-
vanced methods and concepts. This document has been writ-
9. Measurement for viral clearance studies
ten to cover all conceivable cases, which can be applied to all
9.1 Measurement of virus infective titer
biotechnological W biological products. Therefore, there may
Assay methods may be either quantal or quantitative.
be cases that it is not so rational to pursue virus tests and
Quantal methods include infectivity assays in animals or in
viral clearance studies in accordance with this document on
tissue-culture-infectious-dose (TCID) assays, in which the
each product, which has been used for a long time without
animal or cell culture is scored as either infected or not. In-
any safety issue. So, it will be necessary to elaborate the
fectivity titers are then measured by the proportion of
most rational ways under a case-by-case principle taking into
animals or culture infected. In quantitative methods, the in-
due consideration source, origin, type, manufacturing proc-
fectivity measured varies continuously with the virus input.
ess, characteristics, usages at clinical stage, accumulation of
Quantitative methods include plaque assays where each p-
the past usage record, etc. relating to such biotechnological W
laque counted corresponds to a single infectious unit. Both
biological products.
quantal and quantitative assays are amenable to statistical
evaluation.
9.2 Testing by nucleic-acid-ampliˆcation test (NAT)
Add the following:
NAT can detect individual or pooled raw material W cell
substrate or virus genome at a high sensitivity even in a stage
that serum test on each virus is negative. Further, it can de- 18. Qualiˆcation of Animals as
tect HBV or HCV gene, which can not be measured in cul- Origin of Animal-derived Medicinal
ture system. Window period can be drastically shortened at
the test on HBV, HCV and HIV, and the method is expected Products provided in the General
to contribute as an eŠective measure for ensuring viral safe- Notices 39 of Japanese
ty. However, depending upon a choice of primer, there may
be a case that not all the subtype of objective virus can be de- Pharmacopoeia and
tected by this method, and, therefore, it is recommendable Other Standards
to evaluate, in advance, if subtype of a broad range can be
detected. Introduction
NAT will be an eŠective evaluation method for virus The O‹cial Gazette issued on March 29, 2002 announced
removal capability at viral clearance studies. However, in that General Notices or General Rules of the Japanese Phar-
case of viral inactivation process, viral inactivation obtained macopoeia of the Standard of Drugs not required Approval,
by this method may be underrated, since there is a case that of the Biological Products Standard and of the Require-
1632 General Information Supplement I, JP XIV

ments for Antibiotic Products of Japan were amended to production, which are proved to be free from communicable
add a provision that ``When a drug product or a drug sub- disease agents after certain appropriate processing on raw
stance which is used to manufacture a drug product, is materials of animal origin.
manufactured from a raw material of animal origin, the As for raw materials of drugs of human origin, cell, tis-
animal in question should be in principle a healthy subject, if sue, blood, placenta, urine, etc. are used. Whenever it is
not otherwise provided.''. su‹cient and possible each donor, as the origin of such raw
The Notice Iyaku-hatsu No. 0329001, which was issued on materials, should be asked his (her) health condition and un-
the same date, provided that ``Healthy subject herein pro- dergoes his (her) medical examination at this stage, so that
vided is the animal which does not cause any disease or any the appropriateness as a donor can be conˆrmed from the
infection to human being at an appropriate production proc- standpoint of safety concerning communicable disease
ess and use of the drug product, and as for the oral or exter- agents such as virus.
nal drug for example, the animal, as its raw material of For example, ``Basic concept on handling and use of a
animal origin, should be conˆrmed at this stage to meet the drug product, etc. which is derived from cell W tissue''
Food Standard. It has to be noted that this standard of (Attachment 1 of the Notice Iyaku-Hatsu No. 1314 dated
healthy subject has to be revised timely taking into account December 26, 2000) and ``Guidance for quality and safety
the up-to-date information with respect to the amphixenosis assurance of a drug product, etc. which is derived from hu-
infections common between human beings and animals.''. man cell W tissue (Attachment 2 of the Notice Iyaku-Hatsu
This General Information describes safety assurance No. 1314 dated December 26, 2000)'' issued by the Director-
against infection of drugs, which are manufactured from General of the Medicinal Safety Bureau, Ministry of Health
raw materials of animal origin, to follow up the Notice as and Welfare, states that since the cell W tissue supplied by a
mentioned above. human donor comes to be applied to patients without proc-
essing through any su‹cient inactivation or removal of com-
1. Basic concept
municable disease agents, the selection and qualiˆcation
When drugs derived from raw materials of animal origin
criteria on such donor has to be established. These criteria
including human are used, it is important to take into ac-
are to be composed with the respect to the check items on the
count any possibility that communicable disease agents such
case history and the physical conditions as well as the test
as virus may cause infectious disease or any possible hazards
items on the various transmission of infectious agents
to patients. In such case, it goes without saying that the pri-
through cell W tissue, and that the appropriateness of these
mary subject that has to be considered is the absence of any
criteria has to be clariˆed. Hepatitis Type-B (HBV), Hepati-
infectious agents such as virus in the raw materials of animal
tis Type-C (HCV), Human Immune Deˆciency Viral infec-
origin including human as the source of the drug. More im-
tions (HIV), Adult T-Cell Leukemia and Parvovirus B19 In-
portant points are whether the drugs derived from such raw
fections should be denied through the interview to the donor
materials are free of such infectious agents and whether
and the tests (serologic test, nucleic-acid ampliˆcation test,
there is any possibility of transmission of infectious agents
etc.). Further, if necessary, Cytomegalovirus infection and
when the drugs are administered to patient. The eligibility of
EB Virus infection should be denied by tests. ``Infections
animals including human, as the source of raw materials of
caused by bacteria such as Treponema pallidum,
drugs, in other words ``the subject which is free from any
Chlamydia, Gonococci, Tubercule bacillus, etc.'', ``septice-
disease or transmission of infectious agents that is infectious
mia and its suspicious case'', ``vicious tumor'', ``serious
to human being at an appropriate production process and
metabolic or endocrine-related disorders'', ``collagenosis
use of the drug product'' is that ``The drug should be entire-
and haematological disorder'', ``hepatic disease'' and ``de-
ly free from any risk of infections by means of whole proce-
mentia (transmissible spongiform encephalopathies and its
dures which include evaluation of appropriateness of the
suspicious case)'' should be checked on the case history or
animals including human as the source of their raw materi-
by the interview, etc. and the experience of being transfused
als, establishment of appropriate production processes and
or Wand transplanted should be checked to conˆrm eligibility
their appropriate control, and strict adherence to the clinical
as a donor. The most appropriate check items and test
indications of the ˆnal product.''
methods then available are to be used, which need to be
2. Animals including human as the source of raw materials reconsidered at appropriate timing taking into account the
of drugs updated knowledge and the progress of the science and the
What is the most clear and appropriate preventive meas- technologies. At screening of a donor, reexaminations has to
ures against infection to human being due to administration be made at appropriate timing using the eligible check items
of drugs which are derived from animals including human is and the test methods taking into account the window period
to assure the absence of any infectious agents such as virus in (Initial period after infection, in which antibody against bac-
its raw materials or an appropriate critical raw material by teria, fungi or virus is not detected.)
each of the followings: (1) the use of raw materials of In the case of plasma derivatives produced from the do-
healthy animal origin, which are proved to be free from nated blood in Japan, the donor should be checked by
communicable disease agents to human, or (2) the use of ap- means of self-assessed report about health conditions, and a
propriate critical raw materials (e.g., cell substrate, blood serologic check and a nucleic acid ampliˆcation test (NAT)
plasma, pooled urine after some treatments) for drug on 50 pooled plasma should be performed at the stage of do-
Supplement I, JP XIV General Information 1633

nated blood. Further, the plasma material (i.e., critical raw have been followed in this case are described in detail in the
material ) for fractionation should be stored 4 months in Notice of Japanese version on the internationally accepted
minimum so that the arrangement could be taken based on ICH Guideline entitled ``Viral safety evaluation of
the information available after collection of the blood and biotechnology products derived from cell lines of human or
the blood infusion to exclude the possibility of using any animal origin'' (Iyakushin No. 329 issued on February 22,
critical raw material which might cause infection to patients. 2000 by Director, Evaluation and Licensing Division, Phar-
On the other hand, as for the materials such as urine maceutical and Medical Safety Bureau, Ministry of Health
which are taken from the unspeciˆed number of the donors and Welfare). In the meantime, it is important how to han-
and come to be critical raw materials for drug production dle the cell in case that any virus has been detected under the
after some treatments, it is unrealistic and not practical to cell level tests. This Notice describes how to cope with this
conduct the tests of virus infection, etc. on the individual situation as follows: ``It is recognised that some cell lines
donor. Consequently, appropriate tests such as virus test has used for the manufacture of product will contain en-
to be performed on such critical raw materials for drug dogenous retroviruses, other viruses or viral sequences. In
production. such circumstances, the action plan recommended for
In the case of the animals besides human, the wild ones manufacturer is described in Section V (Rationale and action
should be excluded. Only the animals, which are raised un- plan for viral clearance studies and virus tests on puriˆed
der well sanitarily controlled conditions taken to prevent bulk) of the Notice. The acceptability of cell lines containing
bacterial contamination or under the eŠective bacterial pol- viruses other than endogenous retroviruses will be consi-
lution monitoring systems, have to be used, and it is recom- dered on an individual basis by the regulatory authorities, by
mended that the animals from a colony appropriately con- taking into account a risk W beneˆt analysis based on the
trolled under speciˆc pathogen-free (SPF) environment are beneˆt of the product and its intended clinical use, the
to be used as far as possible. Further, for the animals regu- nature of the contaminating viruses, their potential for
lated under the Food Standard, only the animals that met infecting humans or for causing disease in humans, the
this standard should be used. It should be conˆrmed by ap- puriˆcation process for the product (e.g., viral clearance
propriate tests that the animals were free from pathogen, if evaluation data), and the extent of the virus tests conducted
necessary. on the puriˆed bulk.'' For example, it is well known that
The concrete measures to avoid transmittance or spread of Type A-, R- and C-endogenous particles like retrovirus are
infectivity of prion, which is considered to be the pathogen observed in the cells of the rodents used most often for drug
of transmissible spongiform encephalopathies (TSEs), as far production. It is also known that they are not infectious to
as possible are the followings:  avoidance of use of human and is not dangerous, and CHO cells are generally
animals, which are raised in the areas where high incidence used for drug production. The established cell lines (e.g.,
or high risk of TSEs (Scrapie in sheep and goat, bovine NAMALWA Cell, BALL-1 Cell, etc.) derived from cancer
spongiform encephalopathies (BSE) in cattle, chronic wast- patients are sometimes used, but through the thorough virus
ing disease (CWD) in deer, new type of Creutzfeldt-Jacob- tests, etc., their safety are conˆrmed. The established cell
Disease (CJD) in human, etc.) is reported, and humans, who lines are assumed to be safer than the primary cultured cells
have stayed long time (more than 6 months) in such areas, as which are hard to conduct the thorough virus test.
raw materials or related substances of drugs;  avoidance
4. Establishment and control of appropriate production
of use of any substances that are derived from the individual
process and adherence to the clinical indication of ˆnal
infected with scrapie, BSE, CJD, etc.;  avoidance of using
product for safety assurance
a material derived from organ, tissue and cell, etc. of high
Safety assurance against potential infections at only the
risk of TSEs; and  taking appropriate measures basing on
level of animals that are source of raw materials of drugs is
the information collected, which includes incidence of TSEs,
limited. Further, ``health of animal'' can not be deˆned
the results of epidemiological investigation and the ex-
univocally, and the various factors have to be taken into ac-
perimental research on prion, and incidence of tardive infec-
count. The ˆnal goal of this subject is to protect human
tion on donors after collecting raw materials, etc.
from any infectious disease caused by drugs. Achieving this
3. Human or animal cells which are used as critical raw goal, the establishment and control of appropriate produc-
materials for drug production tion processes of each drug and the adherence to the clinical
Cell substrates derived from humans or animals are used indications of the ˆnal product are important.
for drug production. In such case, it is desirable that the As mentioned above, the rodent cells used most often for
humans or the animals, which are the origins of the cell sub- the production of the drugs are known to have endogenous
strates, are healthy subjects. However, it is considered prac- retrovirus sometimes. The reason why such cells can be used
tical that viral safety of the drugs derived from the cell sub- for the production of the drugs is that multiple measures are
strates are evaluated on the cells, which are so called critical applied for safety in the puriˆcation stages which include ap-
raw materials for production of such drugs. In such case, the propriate inactivation or removal processes. There are cases
safety should be conˆrmed through the test and analysis on in which the production procedure involves intentional use
established cell bank thoroughly with respect to virus etc., as of a virus or a microorganism. In this case, relevant meas-
far as possible. The items and the methods of the tests that ures capable of removing or inactivating of such virus or
1634 General Information Supplement I, JP XIV

microorganism are appropriately incorporated in the puriˆ- persulfate and N,N,N?,N?-tetramethylethylenediamine


cation process, so that the risk of infection to human can be (TEMED).
fully denied and its safety can be assured when it is used as a As the acrylamide concentration of a gel increases, its
drug. Further, even in the case that it is di‹cult to clarify the eŠective pore size decreases. The eŠective pore size of a gel is
risk of contamination of the infectious agents or that the raw operationally deˆned by its sieving properties; that is, by the
material are contaminated by viruses etc., the raw material resistance it imparts to the migration of macromolecules.
in question may be used for the production of drugs so long There are limits on the acrylamide concentration that can be
as appropriate inactivation or removal processes are in- used. At high acrylamide concentrations, gels break much
troduced, their eŠectiveness can be conˆrmed and the safety more easily and are di‹cult to handle. As the pore size of a
can be assured by appropriate control of the manufacturing gel decreases, the migration rate of a protein through the gel
processes under GMP, etc. decreases. By adjusting the pore size of a gel, through
manipulating the acrylamide concentration, the resolution
5. Conclusion
of the method can be optimized for a given protein product.
The qualiˆcation of animals including human, as the
Thus, the physical characteristics of a given gel are deter-
source of raw materials of drugs, in other words ``the sub-
mined by the relative concentrations of acrylamide and bi-
ject which does not cause any infectious diseases to human
sacrylamide, used in its preparation.
being at an appropriate production process and use of the
In addition to the composition of the gel, the state of the
drug product'' is that ``the drug has to be entirely free from
protein is an important determinant of the electrophoretic
any risk of infections by means of whole procedures which
mobility. In the case of proteins, the electrophoretic mobili-
include evaluation of appropriateness of the animal includ-
ty is dependent on the pK values of the charged groups and
ing human as the source of their raw materials, establish-
the size of the molecule. It is also in‰uenced by the type,
ment of appropriate production processes and their
concentration and pH of the buŠer, the temperature and the
appropriate control, and strict adherence to the clinical indi-
ˆeld strength, as well as by the nature of the support
cation of the ˆnal product.''
material.
To cope with this subject, the advanced scientiˆc meas-
ures, which actually re‰ect the updated knowledge and 2. Polyacrylamide Gel Electrophoresis under Denaturing
progress of the science and the technology about infectious Conditions
diseases in human and infection of animal origin, have to be The method cited as an example is limited to the analysis
taken into account timely. of monomeric polypeptides with a mass range of 14,000 to
100,000 daltons. It is possible to extend this mass range by
various techniques (e.g., by using gradient gels, particular
Add the following: buŠer systems, etc.), but those techniques are not discussed
in this chapter.
19. SDS-Polyacrylamide Gel Analysis by electrophoresis on sodium dodecyl sulfate
(SDS) polyacrylamide gel (SDS-Polyacrylamide Gel Elec-
Electrophoresis trophoresis) under denaturing conditions is the most com-
mon mode of electrophoresis used in assessing the quality of
The SDS-Polyacrylamide Gel Electrophoresis is used for
proteins in biotechnological and biological products, and
the characterization of proteins in biotechnological and bio-
will be the focus of the example described here. Typically,
logical products and for control of purity and quantitative
analytical electrophoresis of proteins is carried out in poly-
determinations.
acrylamide gels under conditions that ensure dissociation of
This technique is a suitable analytical method with which
the proteins into their individual polypeptide subunits and
to identify and to assess the homogeneity of proteins in
that minimize aggregation. Most commonly, the strongly
biotechnological and biological products. The method is
anionic detergent SDS is used in combination with heat to
also routinely used for the estimation of protein subunit
dissociate the proteins before they are loaded on the gel. The
molecular masses and for determining the subunit composi-
denatured polypeptides bind to SDS, become negatively
tions of puriˆed proteins.
charged and exhibit a consistent charge-to-mass ratio
Ready-to-use gels and reagents are widely available on the
regardless of protein type. Because the amount of SDS
market and can be used instead of those described in this
bound is almost always proportional to the molecular mass
text, provided that they give equivalent results and that they
of the polypeptide and is independent of its amino acid se-
meet the validity requirements given below under Validation
quence, SDS-polypeptide complexes migrate through poly-
of the Test.
acrylamide gels with mobilities that are dependent on the
1. Characteristics of Polyacrylamide Gels size of the polypeptides.
The sieving properties of polyacrylamide gels are aŠorded The electrophoretic mobilities of the resultant SDS-
by the three-dimensional network of ˆbers and pores which polypeptide complexes all assume the same functional
is formed as the bifunctional bisacrylamide cross-links adja- relationship to their molecular masses. Migration of SDS-
cent polyacrylamide chains. Polymerization is catalyzed by a complexes occurs toward the anode in a predictable manner,
free radical-generating system composed of ammonium with low-molecular-mass complexes migrating faster than
Supplement I, JP XIV General Information 1635

larger ones. The molecular mass of a protein can therefore high-voltage gradient forms between the leading and trailing
be estimated from its relative mobility calibrated in SDS- ion fronts, causing the SDS-protein complex to form into a
Polyacrylamide Gel Electrophoresis and the occurrence of a very thin zone (called the stack) and to migrate between the
single band in such a gel is a criterion of purity. chloride and glycinate phases. Regardless of the height of
However, modiˆcations to the polypeptide backbone, the applied sample solution in the wells, all SDS-protein
such as N- or O-1inked glycosylation, have a signiˆcant im- complexes condense within broad limits and enter the resolv-
pact on the apparent molecular mass of a protein, since SDS ing gel as a well-deˆned, thin zone of high protein density.
does not bind to a carbohydrate moiety in a manner similar The large-pore stacking gel does not retard the migration of
to a polypeptide. Thus, a consistent charge-to-mass ratio is most proteins and serves mainly as an anticonvective medi-
not maintained. The apparent molecular masses of proteins um. At the interface of the stacking and resolving gels, the
that have undergone post-translational modiˆcations do not proteins experience a sharp increase in retardation due to the
truly re‰ect the masses of the polypeptides. smaller pore size of the resolving gel. Once the proteins are
1) Reducing conditions in the resolving gel, their mobility continues to be slowed
Polypeptide subunits and three-dimensional structure of down by the molecular sieving eŠect of the matrix. The
proteins are often ˆxed, at least in part, by the presence of glycinate ions overtake the proteins, which then move in a
disulˆde bonds. A goal of SDS-Polyacrylamide Gel Elec- space of uniform pH formed by the tris(hydrox-
trophoresis under reducing conditions is to disrupt this ymethyl)aminomethane and glycine. Molecular sieving
structure by reducing the disulˆde bonds. Complete denatu- causes the SDS-polypeptide complexes to separate on the
ration and dissociation of proteins by treatment with 2-mer- basis of their molecular masses.
captoethanol or dithiothreitol (DTT) will result in unfolding
4. Preparing Vertical Discontinuous BuŠer SDS-
of the polypeptide backbone and subsequent complexation
Polyacrylamide Gels
with SDS. Under these conditions, the molecular masses of
1) Assembling of the gel moulding cassette
the polypeptide subunits can be calculated by interpolation
Clean the two glass plates (size: e.g. 10 cm × 8 cm), the
in the presence of suitable molecular-mass standards.
sample comb made of polytetra‰uoroethylene, the two
2) Non-reducing conditions
spacers and the silicone rubber tubing (diameter, e.g. 0.6
For some analyses, complete dissociation of the protein of
mm × 35 cm) with mild detergent and rinse extensively with
interest into subunit peptides is not desirable. In the absence
water. Dry all the items with a paper towel or tissue. Lubri-
of treatment with reducing agents such as 2-mercap-
cate the spacers and the silicone rubber tubing with non-sili-
toethanol or DTT, disulˆde covalent bonds remain intact,
cone grease. Apply the spacers along each of the two short
preserving the oligomeric form of the protein. Oligomeric
sides of the glass plate 2 mm away from the edges and 2 mm
SDS-protein complexes migrate more slowly than their SDS-
away from the long side corresponding to the bottom of the
polypeptide subunits. In addition, non-reduced proteins
gel. Begin to lay the silicone rubber tubing on the glass plate
may not be completely saturated with SDS and, hence, may
by using one spacer as a guide. Carefully twist the silicone
not bind the detergent in the expected mass ratio. This
rubber tubing at the bottom of the spacer and follow the
makes molecular-mass determinations of these molecules by
long side of the glass plate. While holding the silicone rubber
SDS-Polyacrylamide Gel Electrophoresis less straightfor-
tubing with one ˆnger along the long side again twist the
ward than analyses of fully denatured polypeptides, since it
tubing and lay it on the second short side of the glass plate,
is necessary that both standards and unknown proteins be in
using the spacer as a guide. Place the second glass plate in
similar conˆgurations for valid comparisons. However, the
perfect alignment and hold the mould together by hand pres-
staining of a single band in such a gel is a criterion of purity.
sure. Apply two clamps on each of the two short sides of the
3. Characteristics of Discontinuous BuŠer System Gel mould. Carefully apply four clamps on the longer side of the
Electrophoresis gel mould, thus forming the bottom of the gel mould. Verify
The most widely used electrophoretic method for the anal- that the silicone rubber tubing is running along the edge of
ysis of complex mixtures of proteins involves the use of a the glass plates and has not been extruded while placing the
discontinuous buŠer system consisting of two contiguous, clamps.
but distinct gels: a resolving or separating (lower) gel and a 2) Preparation of the gel
stacking (upper) gel. The two gels are cast with diŠerent In a discontinuous buŠer SDS polyacrylamide gel, it is
porosities, pH, and ionic strengths. In addition, diŠerent recommended to pour the resolving gel, let the gel set, and
mobile ions are used in the gel and electrode buŠers. The then pour the stacking gel, since the compositions of the two
buŠer discontinuity acts to concentrate large-volume sam- gels in acrylamide-bisacrylamide, buŠer and pH are diŠer-
ples in the stacking gel, resulting in improved resolution. ent.
When power is applied, a voltage drop develops across the Preparation of the resolving gel: In a conical ‰ask, prepare
sample solution which drives the proteins into the stacking the appropriate volume of solution containing the desired
gel. Glycinate ions from the electrode buŠer follow the pro- concentration of acrylamide for the resolving gel, using the
teins into the stacking gel. A moving boundary region is rap- values given in Table 1. Mix the components in the order
idly formed with the highly mobile chloride ions in the front shown. Where appropriate, before adding the ammonium
and the relatively slow glycinate ions in the rear. A localized persulfate solution and the tetramethylethylenediamine
1636 General Information Supplement I, JP XIV

(TEMED), ˆlter the solution if necessary under vacuum Gel Electrophoresis. Prepare the test and reference solutions
through a cellulose acetate membrane (pore size: 0.45 mm); in the recommended sample buŠer and treat as speciˆed in
keep the solution under vacuum by swirling the ˆltration the individual monograph. Apply the appropriate volume of
unit until no more bubbles are formed in the solution. Add each solution to the stacking gel wells. Start the electropho-
appropriate amounts of ammonium persulfate solution and resis using suitable operating conditions for the electropho-
TEMED as indicated in Table 1, swirl and pour immediately resis equipment to be used. There are commercially available
into the gap between the two glass plates of the mould. gels of diŠerent surface area and thickness that are appropri-
Leave su‹cient space for the stacking gel (the length of the ate for various types of electrophoresis equipment. Elec-
teeth of the sample comb plus 1 cm). Using a pipette, care- trophoresis running time and current W voltage may need to
fully overlay the solution with water-saturated isobutanol. be altered depending on the type of apparatus used, in order
Leave the gel in a vertical position at room temperature to to achieve optimum separation. Check that the dye front is
allow polymerization to occur. moving into the resolving gel. When the dye is reaching the
Preparation of the stacking gel: After polymerization is bottom of the gel, stop the electrophoresis. Remove the gel
complete (about 30 minutes), pour oŠ the isobutanol and assembly from the apparatus and separate the glass plates.
wash the top of the gel several times with water to remove Remove the spacers, cut oŠ and discard the stacking gel and
the isobutanol overlay and any unpolymerized acrylamide. immediately proceed with staining.
Drain as much ‰uid as possible from the top of the gel, and
5. Detection of Proteins in Gels
then remove any remaining water with the edge of a paper
Coomassie staining is the most common protein staining
towel.
method, with a detection level of the order of 1 mg to 10 mg
In a conical ‰ask, prepare the appropriate volume of solu-
of protein per band. Silver staining is the most sensitive
tion containing the desired concentration of acrylamide,
method for staining proteins in gels and a band containing l0
using the values given in Table 2. Mix the components in the
ng to 100 ng can be detected. All of the steps in gel staining
order shown. Where appropriate, before adding the ammo-
are done at room temperature with gentle shaking in any
nium persulfate solution and the TEMED, ˆlter the solution
convenient container. Gloves must be worn when staining
if necessary under vacuum through a cellulose acetate mem-
gels, since ˆngerprints will stain.
brane (pore size: 0.45 mm); keep the solution under vacuum
1) Coomassie staining
by swirling the ˆltration unit until no more bubbles are
Immerse the gel in a large excess of Coomassie staining TS
formed in the solution. Add appropriate amounts of ammo-
and allow to stand for at least 1 hour. Remove the staining
nium persulfate solution and TEMED as indicated in Table
solution.
2, swirl and pour immediately into the gap between the two
Destain the gel with a large excess of destaining TS.
glass plates of the mould directly onto the surface of the
Change the destaining solution several times, until the
polymerized resolving gel. Immediately insert a clean sample
stained protein bands are clearly distinguishable on a clear
comb into the stacking gel solution, taking care to avoid
background. The more thoroughly the gel is destained, the
trapping air bubbles. Add more stacking gel solution to ˆll
smaller is the amount of protein that can be detected by the
completely the spaces of the sample comb. Leave the gel in a
method. Destaining can be speeded up by including 2 to 3 g
vertical position and allow to polymerize at room tempera-
of anion-exchange resin or a small sponge in the destaining
ture.
TS.
3) Mounting the gel in the electrophoresis apparatus and
NOTE: the acid-alcohol solutions used in this procedure
electrophoretic separation
do not completely ˆx proteins in the gel. This can lead to
After polymerization is complete (about 30 minutes),
losses of some low-molecular-mass proteins during the stain-
remove the sample comb carefully. Rinse the wells immedi-
ing and destaining of the gel. Permanent ˆxation is obtaina-
ately with water or with the running buŠer for SDS-
ble by allowing the gel to stand in trichloroacetic acid TS for
Polyacrylamide Gel Electrophoresis to remove any un-
ˆxing for 1 hour before it is immersed in Coomassie staining
polymerized acrylamide. If necessary, straighten the teeth of
TS.
the sample comb of the stacking gel with a blunt hypodermic
2) Silver staining
needle attached to a syringe. Remove the clamps on one
Immerse the gel in a large excess of ˆxing TS and allow to
short side, carefully pull out the silicone rubber tubing and
stand for 1 hour. Remove the ˆxing solution, add fresh ˆx-
replace the clamps. Proceed similarly on the other short side.
ing solution and incubate either for at least 1 hour or over-
Remove the silicone rubber tubing from the bottom part of
night, if convenient. Discard the ˆxing solution and wash the
the gel. Mount the gel in the electrophoresis apparatus. Add
gel in water for 1 hour. Soak the gel for 15 minutes in a 1 vol
the electrophoresis buŠers to the top and bottom reservoirs.
z glutaraldehyde solution. Wash the gel twice for 15
Remove any bubbles that become trapped at the bottom of
minutes in water. Soak the gel in fresh silver nitrate TS for
the gel between the glass plates. This is best done with a bent
silver staining for 15 minutes, in darkness. Wash the gel
hypodermic needle attached to a syringe. Never pre-run the
three times for 5 minutes in water. Immerse the gel for about
gel before loading solutions, such as samples, since this will
1 minute in developer TS until satisfactory staining has been
destroy the discontinuity of the buŠer systems. Before load-
obtained. Stop the development by incubation in blocking
ing solutions, such as samples, carefully rinse the stacking
TS for 15 minutes. Rinse the gel with water.
gel wells with the running buŠer for SDS-Polyacrylamide
Supplement I, JP XIV General Information 1637

6. Drying of Stained SDS-Polyacrylamide Gels 8. Suitability of the Test (Validation)


Depending on the staining method used, gels are pretreat- The test is not valid unless the front end of the molecular
ed in a slightly diŠerent way. For Coomassie staining, after mass marker migrates 80z of the migrating distance of the
the destaining step, allow the gel to stand in a diluted solu- dye, and over the required separation range (e.g., the range
tion of concentrated glycerin (1 in 10) for at least 2 hours covering the product and its dimer or the product and its
(overnight incubation is possible). For silver staining, add to related impurities) the separation obtained for the relevant
the ˆnal rinsing a step of 5 minutes in a diluted solution of protein bands shows a linear relationship between the
concentrated glycerin (1 in 50). logarithm of the molecular mass and the Rf as described in
Immerse two sheets of porous cellulose ˆlm in water and 7. Additional requirements with respect to the solution
incubate for 5 to 10 minutes. Place one of the sheets on a under test may be speciˆed in individual monographs.
drying frame. Carefully lift the gel and place it on the cellu-
9. Quantiˆcation of Impurities
lose ˆlm. Remove any trapped air bubbles and pour 2 to 3
Where the impurity limit is speciˆed in the individual
mL of water around the edges of the gel. Place the second
monograph, a reference solution corresponding to that level
sheet on top and remove any trapped air bubbles. Complete
of impurity should be prepared by diluting the test solution.
the assembly of the drying frame. Place in an oven or leave
For example, where the limit is 5z, a reference solution
at room temperature until dry.
would be a 1:20 dilution of the test solution. No impurity
7. Molecular-Mass Determination (any band other than the main band) in the electrophoreto-
Molecular masses of proteins are determined by compari- gram obtained with the test solution may be more intense
son of their mobilities with those of several marker proteins than the main band obtained with the reference solution.
of known molecular mass. Mixtures of proteins with pre- Under validated conditions, impurities may be quantiˆed
cisely known molecular masses blended for uniform staining by normalization to the main band, using an integrating den-
are commercially available for calibrating gels. They are ob- sitometer. In this case, the responses must be validated for
tainable in various molecular mass ranges. Concentrated linearity.
stock solutions of proteins of known molecular mass are
Test solutions:
diluted in the appropriate sample buŠer and loaded on the
Coomassie staining TS Dissolve 125 mg of coomassie
same gel as the protein sample to be studied.
brilliant blue R-250 in 100 mL of a mixture of water,
Immediately after the gel has been run, the position of the
methanol and acetic acid (100) (5:4:1), and ˆlter.
bromophenol blue tracking dye is marked to identify the
Developer TS Dissolve 2 g of citric acid monohydrate in
leading edge of the electrophoretic ion front. This can be
water to make 100 mL. To 2.5 mL of this solution add 0.27
done by cutting notches in the edges of the gel or by inserting
mL of formaldehyde solution and water to make 500 mL.
a needle soaked in India ink into the gel at the dye front.
Fixing TS To 250 mL of methanol add 0.27 mL of for-
After staining, measure the migration distances of each pro-
maldehyde solution and water to make 500 mL.
tein band (markers and unknowns) from the top of the
Silver nitrate TS for silver staining To 40 mL of sodium
resolving gel. Divide the migration distance of each protein
hydroxide TS add 3 mL of ammonia solution (28), then add
by the distance traveled by the tracking dye. The normalized
dropwise 8 mL of a solution of silver nitrate (1 in 5) while
migration distances so obtained are called the relative mobil-
stirring, and add water to make 200 mL.
ities of the proteins (relative to the dye front) and conven-
Destaining TS A mixture of water, methanol and acetic
tionally denoted as Rf. Construct a plot of the logarithm of
acid (100) (5:4:1).
the relative molecular masses (Mr) of the protein standards
Blocking TS To 10 mL of acetic acid (100) add water to
as a function of the Rf values. Note that the graphs are
make 100 mL.
slightly sigmoid. Unknown molecular masses can be esti-
Trichloroacetic acid TS for ˆxing Dissolve 10 g of
mated by linear regression analysis or interpolation from the
trichloroacetic acid in a mixture of water and methanol (5:4)
curves of log Mr against Rf as long as the values obtained for
to make 100 mL.
the unknown samples are positioned along the linear part of
the graph.
1638 General Information Supplement I, JP XIV

Table 1. Preparation of resolving gel

Component volumes (mL) per gel mould volume of


Solution components
5 mL 10 mL 15 mL 20 mL 25 mL 30 mL 40 mL 50 mL

6z Acrylamide
Water 2.6 5.3 7.9 10.6 13.2 15.9 21.2 26.5
Acrylamide solution(1) 1.0 2.0 3.0 4.0 5.0 6.0 8.0 10.0
1.5 mol W
L Tris solution (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5
100 g W
L SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
100 g W
L APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
TEMED(5) 0.004 0.008 0.012 0.016 0.02 0.024 0.032 0.04
8z Acrylamide
Water 2.3 4.6 6.9 9.3 11.5 13.9 18.5 23.2
Acrylamide solution(1) 1.3 2.7 4.0 5.3 6.7 8.0 10.7 13.3
1.5 mol W
L Tris solution (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5
100 g W
L SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
100 g W
L APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
TEMED(5) 0.003 0.006 0.009 0.012 0.015 0.018 0.024 0.03
10z Acrylamide
Water 1.9 4.0 5.9 7.9 9.9 11.9 15.9 19.8
Acrylamide solution(1) 1.7 3.3 5.0 6.7 8.3 10.0 13.3 16.7
1.5 mol W
L Tris solution (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5
100 g W
L SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
100 g W
L APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02
12z Acrylamide
Water 1.6 3.3 4.9 6.6 8.2 9.9 13.2 16.5
Acrylamide solution(1) 2.0 4.0 6.0 8.0 10.0 12.0 16.0 20.0
1.5 mol W
L Tris solution (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5
100 g W
L SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
100 g W
L APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02
14z Acrylamide
Water 1.4 2.7 3.9 5.3 6.6 8.0 10.6 13.8
Acrylamide solution(1) 2.3 4.6 7.0 9.3 11.6 13.9 18.6 23.2
1.5 mol W
L Tris solution (pH 8.8)(2) 1.2 2.5 3.6 5.0 6.3 7.5 10.0 12.5
100 g W
L SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
100 g W
L APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02
15z Acrylamide
Water 1.1 2.3 3.4 4.6 5.7 6.9 9.2 11.5
Acrylamide solution(1) 2.5 5.0 7.5 10.0 12.5 15.0 20.0 25.0
1.5 mol W
L Tris solution (pH 8.8)(2) 1.3 2.5 3.8 5.0 6.3 7.5 10.0 12.5
100 g W
L SDS(3) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
100 g W
L APS(4) 0.05 0.1 0.15 0.2 0.25 0.3 0.4 0.5
TEMED(5) 0.002 0.004 0.006 0.008 0.01 0.012 0.016 0.02

(1) Acrylamide solution: 30z acrylamide W bisacrylamide (29:1) solution


(2) 1.5 mol W
L Tris solution (pH 8.8): 1.5 mol WL tris-hydrochloride buŠer solution, pH 8.8
(3) 100 g W
L SDS: 100 g W L solution of sodium dodecyl sulfate
(4) 100 g W
L APS: 100 g W L solution of ammonium persulfate. Ammonium persulfate provides the free radicals that drive
polymerization of acrylamide and bisacrylamide. Since ammonium persulfate solution decomposes slowly, fresh solu-
tions must be prepared before use.
(5) TEMED: N,N,N?,N?-tetramethylethylenediamine
Supplement I, JP XIV General Information 1639

Table 2. Preparation of stacking gel

Component volumes (mL) per gel mould volume of


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

Water 0.68 1.4 2.1 2.7 3.4 4.1 5.5 6.8


Acrylamide solution(1) 0.17 0.33 0.5 0.67 0.83 1.0 1.3 1.7
1.0 mol W
L Tris solution (pH 6.8)(2) 0.13 0.25 0.38 0.5 0.63 0.75 1.0 1.25
W
100 g L SDS(3) 0.01 0.02 0.03 0.04 0.05 0.06 0.08 0.1
100 g W
L APS(4) 0.01 0.02 0.03 0.04 0.05 0.06 0.08 0.1
TEMED(5) 0.001 0.002 0.003 0.004 0.005 0.006 0.008 0.01

(1) Acrylamide solution: 30z acrylamide W bisacrylamide (29:1) solution


(2) 1.0 mol W
L Tris solution (pH 6.8): 1 mol WL tris-hydrochloride buŠer solution, pH 6.8
(3) 100 g W
L SDS: 100 g W L solution of sodium dodecyl sulfate
(4) 100 g W
L APS: 100 g W L solution of ammonium persulfate. Ammonium persulfate provides the free radicals that drive
polymerization of acrylamide and bisacrylamide. Since ammonium persulfate solution decomposes slowly, fresh solu-
tions must be prepared before use.
(5) TEMED: N,N,N?,N?-tetramethylethylenediamine
Supplement I, JP XIV Appendix 1641

The Ministry of Health, Labour and Welfare


The Ministerial Notiˆcation on Partial Revision
of the Japanese Pharmacopoeia
(No. 151, March 2002)

In accordance with the provision of Paragraph 1 of Add the following paragraph to the General No-
Article 41 of the Pharmaceutical AŠairs Law (Law tices of the Part I :
No. 145, 1960), we hereby revise a part of the
39. In principle, unless otherwise speciˆed,
Japanese Pharmacopoeia (Ministerial Notiˆcation
animals used as a source of materials for preparing
No. 111, March 2001) as follows and enforce the revi-
pharmaceutical preparations listed in the Japanese
sion on April 1, 2002. Although the revision shall
Pharmacopoeia must be healthy.
come into eŠect on April 1, 2002, in the case of the
drugs, which are prepared or imported before March
Delete the following Monograph:
31, 2003, the previous texts in the General Notices of
the Part I or the Part II in the Japanese Phar- Part I
macopoeia may be applied. (The paragraphs on the
Phenacetin
General Notices of the Part I are referred to the
General Notices of the Part II.)
March 29, 2002
Chikara Sakaguchi
The Minister of Health, Labour and Welfare
INDEX

Page citations refer to the pages of the Supplement I, and to pages of


the JP XIV main volume, including those where the text being revised in
the Supplement originally appeared. This Supplement I commences with
page 1359 and succeeding Supplements will continue to be paged in se-
quence.

1„1358 Main Volume of JP XIV


1359„1668 Supplement I

Diphtheria Toxoid for Adult Use, Ambenonium Chloride, 236


A 912 Amidotrizoic Acid, 237
Habu-venom Toxoid, 938 Amikacin Sulfate, 238
Absorbent Hepatitis B Vaccine, 938 Aminoacetic Acid, 931
Cotton, 851 Puriˆed Pertussis Vaccine, 1005 Aminobenzylpenicillin, 246
Cotton, Puriˆed, 851 Tetanus Toxoid, 1066 Anhydrous, 246
Cotton, Sterile, 852 A‰oqualone, 225 Sodium, 246
Cotton, Sterile, Puriˆed, 852 Agar, 859 Aminophylline, 239
Gauze, 853 Powdered, 859 Injection, 239
Gauze, Sterile, 854 Ajmaline, 226 Amitriptyline
Absorptive Ointment, 855 Tablets, 227 Hydrochloride, 240
Acacia, 855, 1543 Akebia Stem, 859 Hydrochloride Tablets, 240
Powdered, 855, 1543 Albumin Tannate, 227 Ammonia Water, 241
Acebutolol Hydrochloride, 219 Alcohol, 914 Amobarbital, 242
Aceglutamide Aluminum, 1377 Benzyl, 874 Sodium for Injection, 242
Acetaminophen, 219 Dehydrated, 914 Amomum Seed, 863
Acetazolamide, 220 for Disinfection, 916 Amorphous Insulin Zinc Injection
Acetic Acid, 856 Isopropyl, 556 (Aqueous Suspension), 542
Glacial, 856 Stearyl, 1059 Amoxapine, 243
Acetohexamide, 221 Aldioxa, 228 Amoxicillin, 244, 1381
Acetylcholine Chloride for Alimemazine Tartrate, 229 Amphotericin B, 245
Injection, 222 Alisma Rhizome, 860 Ampicillin, 246, 1382
Acetylkitasamycin, 223, 1378 Powdered, 860 Anhydrous, 246, 1384
Acetylleucomycin, 223 Allopurinol, 229 Ethoxycarbonyloxyethyl
Acetylsalicylic Acid, 252 Aloe, 860, 1544 Hydrochloride, 259
Tablets, 252 Powdered, 861, 1544 Sodium, 246, 1385
Acetylspiramycin, 224, 1378 Alprazolam, 230 Ampicillinphthalidyl
Achyranthes Root, 857 Alpinia O‹cinarum Rhizome, 1544 Hydrochloride, 782
Aclarubicin Hydrochloride, 224, 1379 Alprenolol Hydrochloride, 231 Amyl Nitrite, 246
Acrinol, 224 Alprostadil Alfadex, 231 Anemarrhena Rhizome, 864
and Zinc Oxide Oil, 857 Alum, 862 Anesthamine, 465
and Zinc Oxide Ointment, 858 Burnt, 863 Anesthetic Ether, 462
Actinomycin D, 225, 1381 Solution, 861 Angelica Dahurica Root, 864
Adhesive Plaster, 858 Aluminum Anhydrous
Adrenaline, 447 Acetylsalicylate, 253 Aminobenzylpenicillin, 246
Hydrochloride Injection, 447 Aspirin, 253 Ampicillin, 246
Hydrochloride Solution, 448 Monostearate, 861 CaŠeine, 294
Adsorbed Potassium Sulfate, 862 Citric Acid, 367, 1438
Diphtheria-Puriˆed Pertussis- Potassium Sulfate, Dried, 863 Dibasic Calcium Phosphate, 879
Tetanus Combined Vaccine, Silicate, Natural, 233 Lactose, 961
1005 Silicate, Synthetic, 235 Antiformin, Dental, 864
Diphtheria-Tetanus Combined Sucrose Sulfate Ester, 766 Antipyrine, 247
Toxoid, 913 Amantadine Hydrochloride, 235 Apricot

1643
1644 Index Supplement I, JP XIV

Kernel, 864 Beeswax Ointment, 289


Kernel Water, 865 White, 871 Bumetanide, 291
Arbekacin Sulfate, 247, 1386 Yellow, 871 Bunazosin Hydrochloride, 291
Areca, 866, 1545 Bekanamycin Sulfate, 265, 1390 Burdock Fruit, 1546
Arginine Belladonna Bupleurum Root, 876
Hydrochloride, 248 Extract, 871 Bupranolol Hydrochloride, 292
Hydrochloride Injection, 249 Root, 872 Burnt Alum, 863
L-Arginine Benoxinate Hydrochloride, 661 Busulfan, 293
Hydrochloride, 248 Benserazide Hydrochloride, 265 Butropium Bromide, 294
Hydrochloride Injection, 249 Bentonite, 873 Butyl Parahydroxybenzoate, 876
Aromatic Castor Oil, 889 Benzalkonium Butyrate, Ribo‰avin, 734
Arotinolol Hydrochloride, 249 Chloride, 266
Arsenical Paste, 866 Chloride Solution, 266 C
Arsenic Trioxide, 250 Chloride Solution 50,
Arsenous Acid, 250 Concentrated, 267 Cacao Butter, 877
Artemisia Capillaris Flower, 867 Benzbromarone, 268, 1391 CaŠeine, 294
Ascorbic Acid, 250 Benzethonium and Sodium Benzoate, 296
Injection, 251 Chloride, 269 Anhydrous, 294
Powder, 251 Chloride Solution, 269 Calciferol, 449
Asiasarum Root, 867, 1545 Benzocaine, 465 Calcium
Asparagus Tuber, 1546 Benzoic Acid, 270 Carbonate, Precipitated, 297
Aspergillus Galactosidase, 921 Benzoin, 873 Carboxymethylcellulose, 885
Aspirin, 252 Benzyl Carmellose, 885
Aluminum, 253 Alcohol, 874 Chloride, 298
Tablets, 252 Benzoate, 874 Chloride Injection, 298, 1399
Aspoxicillin, 254 Benzylpenicillin CMC, 885
Astragalus Root, 867 Benzathine, 1392 Folinate, 299
Astromicin Sulfate, 255, 1387 Potassium, 270, 1393 Fosfomycin, 494
Atractylodes Berberine Gluconate, 299
Lancea Rhizome, 869 Chloride, 271 Hydroxide, 877
Rhizome, 868 Tannate, 272 Lactate, 300
Atropine Betahistine Mesilate, 273 Leucovorin, 299
Sulfate, 255 Betamethasone, 274 Oxide, 878
Sulfate Injection, 256 Dipropionate, 275 Pantothenate, 301
Azathioprine, 256 Sodium Phosphate, 276, 1395 Para-aminosalicylate, 301
Tablets, 256 Valerate, 276, 1395 Para-aminosalicylate Granules,
Aztreonam, 258 Bethanechol Chloride, 278 302
Bifonazole, 278 Polystyrene Sulfonate, 303, 1399
B Biperiden Hydrochloride, 279 Stearate, 881
Bisacodyl, 280 Calumba, 881
Bacampicillin Hydrochloride, 259, Suppositories, 280, 1396 Powdered, 881
1389 Bismuth Camellia Oil, 882
Bacitracin, 1390 Subgallate, 281 Camostat Mesilate, 304
Baclofen, 260 Subnitrate, 282 Camphor, Synthetic, 306
Tablets, 260 Bitter d-Camphor, 305, 1400
Bamethan Sulfate, 262 Cardamon, 875 dl-Camphor, 306, 1400
Barbital 263 Orange Peel, 875 Capsicum, 882, 1546
Barium Sulfate, 264 Tincture, 875 and Salicylic Acid Spirit, 884
Bark Bleomycin Powdered, 883, 1546
Cinnamon, 896 Hydrochloride, 283, 1396 Tincture, 883
Cinnamon, Powdered, 897 Sulfate, 283, 1398 Capsules, 885
Magnolia, 970 Blood, Whole Human, 1080 Cloˆbrate, 373
Magnolia, Powdered, 971 Boric Acid, 284 Flurazepam, 488
Mallotus, 971 Botulism Antitoxin, Equine, Freeze- Indometacin, 535
Moutan, 979 dried, 876 Sodium Iodide (123I), 755
Moutan, Powdered, 980 Bromazepam, 284 Sodium Iodide (131I), 755
Mulberry, 981 Bromhexine Hydrochloride, 285 Sodium Iopodate, 757
Phellodendron, 1007 Bromocriptine Mesilate, 286 Vitamin A, 1075
Phellodendron, Powdered, 1008 Bromovalerylurea, 287 Vitamin A Oil, 1074
Bearberry Leaf, 870 Bucumolol Hydrochloride, 287 Captopril, 307
Bear Bile, 869 Bufetolol Hydrochloride, 288 Carbamazepine, 308
Beclometasone Dipropionate, 264 Bufexamac, 289 Carbazochrome Sodium Sulfonate,
Beef Tallow, 870 Cream, 289 308, 1400
Supplement I, JP XIV Index 1645

Carbetapentane Citrate, 674 Cefteram Pivoxil, 344, 1425 Hydrochloride, 1439


Carbetaoebtene Citrete, 674 Ceftibuten, 344, 1426 Phosphate, 369, 1440
Carbidopa, 309, 1401 Ceftizoxime Sodium, 345 Clinoˆbrate, 369
L-Carbocisteine, 310 Ceftriaxone Sodium, 346, 1427 Clocapramine Hydrochloride, 370
Carbolic Acid, 1010 Cefuroxime Axetil, 347, 1429 Clofedanol Hydrochloride, 371
for Disinfection, 1010 Cefuroxime Sodium, 348 Cloˆbrate, 372
Liqueˆed, 1011 Cellulose Capsules, 373
Carbon Dioxide, 312 Acetate Phthalate, 893 Clomifene
Carboxymethylcellulose, 885 Microcrystalline, 889 Citrate, 373, 1441
Calcium, 886 Powdered, 892 Citrate Tablets, 374
Sodium, 887 Cetanol, 894 Clomipramine Hydrochloride, 375
Cardamon, 885 Cetraxate Hydrochloride, 349, 1431 Clonazepam, 375
Carmellose, 885 Chloral Hydrate, 350 Clonidine Hydrochloride, 376
Calcium, 886 Chloramphenicol, 351, 1432 Cloperastine Hydrochloride, 377
Sodium, 887 Palmitate, 1432 Clotiazepam, 378
Carmofur, 313 Sodium Succinate, 1434 Clotrimazole, 379
Carnauba Wax, 888 Chlordiazepoxide, 351 Clove, 898
Carteolol Hydrochloride, 313 Powder, 351, 1434 Oil, 898
Carumonam Sodium, 314, 1401 Tablets, 352, 1435 Powdered, 898
Cassia Seed, 888 Chlorhexidine Cloxacillin Sodium, 380, 1441
Castor Oil, 888 Gluconate Solution, 353 Cloxazolam, 380
Catalpa Fruit, 889 Hydrochloride, 354 CMC, 885
Cefaclor, 314, 1403 Chlorinated Lime, 894 Calcium, 886
Cefadroxil, 314 Chlormadinone Acetate, 355, 1435 Sodium, 887
Cefalexin, 315 Chlorobutanol, 894 Cnidium Rhizome, 899
Cefaloridine, 317, 1405 Chlorphenesin Carbamate, 356, 1436 Cocaine Hydrochloride, 381
Cefalotin Sodium, 317, 1406 Chlorpheniramine Coconut Oil, 899
Cefamandole Sodium, 317, 1407 and Calcium Powder, 895 Codeine
Cefapirin Sodium, 317 Maleate, 357 Phosphate, 382
Cefatrizine Propylene Glycolate, 318 Maleate Injection, 357 Phosphate Powder, 1, 382, 1442
Cefazolin Maleate Powder, 358 Phosphate Powder, 10, 383, 1442
Sodium, 319 Maleate Tablets, 359 Phosphate Tablets, 384, 1443
Sodium Hydrate, 321 d-Chlorpheniramine Maleate, 359 Cod Liver Oil, 900, 1548
Cefbuperazone Sodium, 322, 1409 Chlorpromazine Coix Seed, 900
Cefcapene Pivoxil Hydrochloride, Hydrochloride, 360 Colchicine, 384
322 Hydrochloride Injection, 361 Colistin
Cefdinir, 324 Hydrochloride Tablets, 361 Sodium Methanesulfonate, 385
Cefditoren Pivoxil, 325 Chlorpropamide, 362 Sulfate, 1444
Cefepime Dihydrochloride, 326 Tablets, 362 Colophonium, 1029
Cefetamet Pivoxil Hydrochloride, Cholecalciferol, 363 Compound
328 Cholera Vaccine, 896 Acrinol and Zinc Oxide Oil, 857
Ceˆxime, 329 Cholesterol, 896 Diastase and Sodium Bicarbonate
Cefmenoxime Hydrochloride, 330, Chorionic Powder, 907
1410 Gonadotrophin, 934 Hycodenone Injection, 995
Cefmetazole Sodium, 321 Gonadotrophin for Injection, 936 Iodine Glycerin, 950
Cefminox Sodium, 332 Chrysanthemum Flower, 1547 Methyl Salicylate Spirit, 977
Cefodizime Sodium, 1412 Ciclacillin, 364, 1437 Oxycodone and Atropine
Cefoperazone Sodium, 332 Ciclosporin, 364 Injection, 996
Cefoselis Sulfate, 334 A, 364 Oxycodone Injection, 995
Cefotaxime Sodium, 335, 1413 Cimetidine, 365 Phellodendron Powder for
Cefotetan, 335, 1415 Cimicifuga Rhizome, 896 Cataplasm, 1009, 1556
Cefotiam Cinchocaine Hydrochloride, 404 Rhubarb and Senna Powder, 1028
Hexetil Hydrochloride, 335, 1417 Cinnamon Salicylic Acid Spirit, 1033
Hydrochloride, 335 Bark, 896, 1547 Scopolia Extract and Diastase
Cefoxitin Sodium, 336, 1419 Oil, 897 Powder, 1037
Cefozopran Hydrochloride, 337 Powdered, 1547 Scopolia Extract and Tannic Acid
Cefpiramide Sodium, 338, 1421 Citric Acid, 366, 1437 Ointment, 1039, 1560
Cefpirome Sulfate, 338 Anhydrous, 367, 1438 Scopolia Extract and Tannic Acid
Cefpodoxime Proxetil, 1422 Citrus Unshiu Peel, 898 Suppositories, 1041, 1560
Cefradine, 339 Clarithromycin, 367 Thianthol and Salicylic Acid
Cefroxadine, 340, 1424 Clemastine Fumarate, 368 Solution, 1067
Cefsulodin Sodium, 340 Clematis Root, 1547 Vitamin B Powder, 1075
Ceftazidime, 342 Clindamycin Concentrated
1646 Index Supplement I, JP XIV

Glycerin, 504 40 Injection, 398 Dioscorea Rhizome, 911


Glycerol, 504 70, 400 Diphenhydramine, 426
Condurango, 900 Sulfate Sodium Sulfur 5, 401 and Bromovalerylurea Powder,
Fluidextract, 901 Sulfate Sodium Sulfur 18, 402 911
Coptis Rhizome, 901, 1548 Dextrin, 906 Hydrochloride, 427
Powdered, 1548 Dextromethorphan Hydrobromide, Phenol and Zinc Oxide Liniment,
Corn 402 912
Oil, 903 Diagnostic Sodium Citrate Solution, Tannate, 427
Starch, 903 754 Diphenylhydantoin, 685
Cornus Fruit, 903 Diastase, 907 Powder, 686
Cortisone and Sodium Bicarbonate Powder, Sodium for Injection, 686
Acetate, 386 907 Tablets, 686
Corydalis Tuber, 904 Diazepam, 403 Diphtheria
Cotton, Absorbent, 851 Dibasic Antitoxin, Equine, Freeze-dried,
Cream, Bufexamac, 289 Calcium Phosphate, 878 912
Creosote, 904 Sodium Phosphate, 907 Toxoid, 912
Cresol, 905 Dibekacin Sulfate, 404, 1447 Diphtheria-Tetanus Combined
Solution, 905 Dibucaine Hydrochloride, 404 Toxoid, 912
Croconazole Hydrochloride, 387 Dichlorphenamide, 406 Dipyridamole, 428, 1451
Crude Glycyrrhiza Extract, 934 Tablets, 407 Disodium
Crystalline Diclofenac Sodium, 405 Edetate, 913
Insulin Zinc Injection (Aqueous Diclofenamide, 406, 1448 Ethylenediaminetetraacetate, 913
Suspension), 543 Tablets, 407 Disopyramide, 429
Penicillin G Potassium, 270 Dicloxacillin Sodium, 408 Distigmine
Crystal Violet, 617 Diethylcarbamazine Bromide, 429
Cyanamide, 387 Citrate, 408 Bromide Tablets, 430
Cyanocobalamin, 388 Citrate Tablets, 409, 1449 Disulˆram, 430
Injection, 389 Diethylstilbestrol Dobutamine Hydrochloride, 431,
Cyclopentolate Hydrochloride, 389 Diphosphate, 493 1452
Cyclophosphamide, 390 Diphosphate Tablets, 494 Dopamine
Cycloserine, 390 Difenidol Hydrochloride, 409 Hydrochloride, 432
Cyperus Rhizome, 906 Digenea, 908 Hydrochloride Injection, 433, 1452
Cyproheptadine Hydrochloride, 391 Digitalis, 908 Dover's Powder, 993
Cytarabine, 392 Powdered, 910 Doxapram Hydrochloride, 433
Digitoxin, 410 Doxorubicin Hydrochloride, 434,
D Tablets, 411 1453
Digoxin, 414 Doxycycline Hydrochloride, 434,
Dantrolene Sodium, 392 Injection, 415 1454
Daunorubicin Hydrochloride, 1445 Tablets, 416 Dried
Deferoxamine Mesilate, 393 Dihydrocodeine Aluminum Hydroxide Gel, 233
Dehydrated Phosphate, 417 Aluminum Hydroxide Gel Fine
Alcohol, 915 Phosphate Powder, 1, 417, 1449 Granules, 233
Ethanol, 915 Phosphate Powder, 10, 418, 1450 Aluminum Potassium Sulfate, 863
Dehydrocholate Sodium Injection, Dihydroergotamine Mesilate, 418, Sodium Carbonate, 1055
396 1450 Sodium Sulˆte, 1058
Dehydrocholic Acid, 394 Dihydroergotoxine Mesilate, 412 Thyroid, 1069
Injection, 396 Dihydroxyaluminum Allantoinate, Yeast, 1083
Puriˆed, 394 228 Dromostanolone
Demethylchlortetracycline Dilazep Hydrochloride, 419 Propionate, 435
Hydrochloride, 1446 Diltiazem Hydrochloride, 420, 1451 Propionate Injection, 436
Dental Dilute Droperidol, 435, 1456
Antiformin, 864 Hydrochloric Acid, 515 Drostanolone
Iodine Glycerin, 951 Iodine Tincture, 950 Propionate, 435, 1456
Paraformaldehyde Paste, 1001 Diluted Opium Powder, 987 Propionate Injection, 436, 1456
Phenol with Camphor, 1011 Dimemorfan Phosphate, 421 Dydrogesterone, 437
Sodium Hypochlorite Solution, 864 Dimenhydrinate, 422 Tablets, 438
Triozinc Paste, 1072 Dimenhydrinate Tablets, 423
Dermatol, 281 Dimercaprol, 423 E
Deslanoside, 396 Injection, 424
Injection, 397 Dimorpholamine, 424 Ecarazine Hydrochloride, 811
Dexamethasone, 397 Injection, 425 Ecothiopate Iodide, 438
Dextran 906 Dinoprost, 425 Edrophonium
40, 398 Dionin, 467 Chloride, 439
Supplement I, JP XIV Index 1647

Chloride Injection, 440 L-Cysteine Hydrochloride, 466 Tablets, 494


EDTA Sodium, 913 Parahydroxybenzoate, 916 Fosfomycin
Elcatonin, 440 Ethylenediamine, 917 Calcium, 494
En‰urane, 443, 1456 Ethylmorphine Hydrochloride, 467 Sodium, 496
Enoxacin, 444 Etilefrine Fradiomycin Sulfate, 497, 1467
Enviomycin Sulfate, 1457 Hydrochloride, 468 Freeze-dried
Ephedra Herb, 914 Hydrochloride Tablets, 468 BCG Vaccine (for Percutaneous
Ephedrine Etizolam, 1463 Use), 869
Hydrochloride, 444 Eucalyptus Oil, 917 Botulism Antitoxin, Equine, 876
Hydrochloride Injection, 445 Eucommia Bark, 1548 Diphtheria Antitoxin, Equine, 912
Hydrochloride Powder, 10z, 446 Evodia Fruit, 918, 1548 Habu Antivenom, Equine, 938
Hydrochloride Tablets, 446 Exsiccated Gypsum, 938 Inactivated Tissue Culture Rabies
Epinephrine, 447 Extract Vaccine, 1024
Hydrochloride Injection, 447 Belladonna, 871 Japanese Encephalitis Vaccine,
Hydrochloride Solution, 448 Crude Glycyrrhiza, 934 957
Injection, 447 Glycyrrhiza, 933 Live Attenuated Measles Vaccine,
Solution, 448 Nux Vomica, 984 972
Epirenamine, 447 Live Attenuated Mumps Vaccine,
Hydrochloride Injection, 447 F 981
Hydrochloride Solution, 448 Live Attenuated Rubella Vaccine,
Epirizole, 448 Famotidine, 469 1029
Epirubicin Hydrochloride, 1458 for Injection, 469, 1464 Mamushi Antivenom, Equine, 972
Ergocalciferol, 449, 1460 Powder, 469 Smallpox Vaccine, 1053
Ergometrine Tablets, 469 Smallpox Vaccine Prepared in Cell
Maleate, 450 Faropenem Sodium, 472 Culture, 1053
Maleate Injection, 451 Fenbufen, 473 Tetanus Antitoxin, Equine, 1066
Maleate Tablets, 451 Fennel, 918 Fritillaria Bulb, 1549
Ergotamine Tartrate, 452 Oil, 919 Fructose, 497
Erythromycin, 453, 1460 Powdered, 918 Injection, 498
Ethylsuccinate, 453 Fentanyl Citrate, 473 Fruit
Lactobionate, 1462 Ferrous Sulfate, 474 Catalpa, 889
Stearate, 454 Fine Granules, Aluminum Hydroxide Cornus, 903
Estazolam, 454 Gel, Dried, 233 Evodia, 918
Estradiol Flavin Adenine Dinucleotide Forsythia, 920
Benzoate, 455, 1462 Sodium, 475 Gardenia, 922
Benzoate Injection, 456 Flavoxate Hydrochloride, 476 Gardenia, Powdered, 923
Benzoate Injection (Aqueous Floctafenine, 477, 1465 Rose, 1029
Suspension), 456 Flomoxef Sodium, 478, 1465 Rose, Powdered, 1029
Estriol, 457, 1463 Flopropione, 478 Schisandra, 1035
Injection (Aqueous Suspension), Flower, Artemisia Capillaris, 867 Zanthoxylum, 1083
458 Flucytosine, 478 Zanthoxylum, Powdered, 1084
Tablets, 458 Fludiazepam, 479 Furosemide, 498
Etacrynic Acid, 459 Flunitrazepam, 480 Fursultiamine Hydrochloride, 499
Tablets, 460 Fluocinolone Acetonide, 481
Ethacridine Lactate, 224 Fluocinonide, 482 G
Ethambutol Hydrochloride, 460 Fluorescein Sodium, 483
Ethanol, 914 Fluorometholone, 484 Gabexate Mesilate, 500
Dehydrated, 915 Fluorouracil, 485 b-Galactosidase (Aspergillus), 921
for Disinfection, 916 Fluoxymesterone, 486 b-Galactosidase (Penicillium), 921
Ethenzamide, 461 Fluphenazine Enanthate, 487 Gallium (67Ga) Citrate Injection, 501
Ether, 462 Flurazepam, 487 Gambir, 920
Anesthetic, 462 Capsules, 488 Powdered, 920
Guaiacol Glyceryl, 505 Hydrochloride, 489 Gardenia Fruit, 922, 1549
Polyoxyethylene Lauryl Alcohol, Flurbiprofen, 489, 1466 Gas Gangrene Antitoxin, Equine, 923
964 Foeniculated Ammonia Spirit, 919 Gastrodia Tuber, 1550
Ethinylestradiol, 463 Folic Acid, 490, 1466 Gauze, Absorbent, 853
Tablets, 463 Injection, 491 Gel, Aluminum Hydroxide, Dried,
Ethionamide, 464 Tablets, 491 233
Ethosuximide, 465 Formalin, 919 Gelatin, 923
Ethoxybenzamide, 461 Water, 920 Puriˆed, 924
Ethyl Formoterol Fumarate, 492 Gentamicin Sulfate, 502, 1468
Aminobenzoate, 465 Forsythia Fruit, 920 Gentian, 925
Cysteine Hydrochloride, 466 Fosfestrol, 493 and Sodium Bicarbonate Powder,
1648 Index Supplement I, JP XIV

926 Homatropine Hydrobromide, 511 Indium (111In) Chloride Injection, 534


Japanese, 957 Homochlorcyclizine Hydrochloride, Indometacin, 534
Powdered, 925 512, 1472 Capsules, 535
Powdered, Japanese, 957 Honey, 938 Suppositories, 536, 1477
Geranium Herb, 926 Houttuynia Herb, 939 In‰uenza HA Vaccine, 949
Ginger, 927 Human Normal Immunoglobulin, Injection
Powdered, 927 949 Acetylcholine Chloride for, 222
Ginseng, 927 Hycoato Injection, 996 (Aqueous Suspension), Estradiol
Powdered, 929 Hydralazine Benzoate, 456
Glacial Acetic Acid, 856 Hydrochloride, 513 (Aqueous Suspension), Estriol,
Glehnia Root, 930 Hydrochloride for Injection, 513 458
Glibenclamide, 502 Hydrochloride Powder, 513 (Aqueous Suspension), Insulin Zinc,
Glucose, 503 Hydrochloride Tablets, 514 542
Injection, 503 Hydrochloric Acid, 514 (Aqueous Suspension), Insulin Zinc,
Glycerin, 504, 1469 Dilute, 515 Amorphous, 542
and Potash Solution, 930 Lemonade, 939 (Aqueous Suspension), Insulin Zinc,
Concentrated, 504, 1469 Hydrochlorothiazide, 515, 1473 Crystalline, 543
Glycerol, 504 Hydrocortisone, 516 (Aqueous Suspension), Insulin Zinc,
Concentrated, 504 Acetate, 517, 1474 Protamine, 544
Glyceryl Monostearate, 931 and Diphenhydramine Ointment, (Aqueous Suspension), Isophane
Glycine, 931 939 Insulin, 541
Glycyrrhiza, 932, 1550 Butyrate, 518 Adrenaline Hydrochloride, 447
Extract, 933 Sodium Phosphate, 519, 1474 Aminophylline, 239
Powdered, 933, 1550 Sodium Succinate, 520 Amobarbital Sodium for, 242
Gramicidin, 1470 Succinate, 521 Arginine Hydrochloride, 249
Gonadotrophin Hydrocotarnine Hydrochloride, 522 L-Arginine Hydrochloride, 249
Chorionic, 934 Hydrogenated Oil, 940 Ascorbic Acid, 251
for Injection, Chorionic, 936 Hydrophilic Atropine Sulfate, 256
for Injection, Serum, 937 Ointment, 940 Calcium Chloride, 298
Serum, 936 Petrolatum, 1005 Chlorpheniramine Maleate, 357
Gramicidin 1470 Hydrous Lanolin, 962 Chlorpromazine Hydrochloride,
Granules Hydroxocobalamin Acetate, 523 361
Calcium Para-aminosalicylate, 302 Hydroxypropylcellulose, 940 Cyanocobalamin, 389
Pas-calcium, 302 Low Substituted, 942 Dehydrocholate Sodium, 396
Griseofulvin, 505, 1471 Hydroxypropylmethylcellulose Dehydrocholic Acid, 396
Guaiacol Glyceryl Ether, 505 Phthalate, 947 Deslanoside, 397
Guaifenesin, 505 2208, 943 Dextran 40, 399
Guanabenz Acetate, 506 2906, 944 Digoxin, 415
Guanethidine Sulfate, 507 2910, 945 Dimercaprol, 424
Gypsum, 937 Hydroxyzine Dimorpholamine, 425
Exsiccated, 938 Hydrochloride, 524 Diphenylhydantoin Sodium for,
Pamoate, 524 686
H Hymecromone, 525 Dromostanolone Propionate, 436
Drostanolone Propionate, 436
Habu Antivenom, Equine, Freeze- I Edrophonium Chloride, 440
dried, 938 Ephedrine Hydrochloride, 445
Haloperidol, 507 Ibuprofen, 526 Epinephrine, 447
Halothane, 508 Ichthammol, 948 Epinephrine Hydrochloride, 447
Haloxazolam, 509, 1472 Idarubicin Hydrochloride, 527, 1475 Epirenamine Hydrochloride, 447
Hemp Fruit, 1551 Idoxuridine, 527 Ergometrine Maleate, 451
Heparin Ophthalmic Solution, 528, 1475 Estradiol Benzoate, 456
Sodium, 510 Ifenprodil Tartrate, 529 Folic Acid, 491
Sodium Injection, 511 Imipenem, 530, 1476 Fructose, 498
Herb Imipramine Gallium (67Ga) Citrate, 501
Ephedra, 914 Hydrochloride, 530 Glucose, 503
Geranium, 926 Hydrochloride Tablets, 531 Heparin Sodium, 511
Houttuynia, 939 Immature Orange, 948 Human Serum Albumin, Iodinated
Mentha, 974 Immunoglobulin, Human Normal, (131I), 546
Perilla, 1004 949 Hycoato, 996
Plantago, 1014 Imperata Rhizome, 949, 1551 Hycodenone, Compound, 995
Powdered Geranium, 926 Indenolol Hydrochloride, 532 Hydralazine Hydrochloride for,
Powdered Swertia, 1064 Indigocarmine, 533 513
Swertia, 1063 Injection, 533 Indigocarmine, 533
Supplement I, JP XIV Index 1649

Indium (111In) Chloride, 534 Thiamylal Sodium for, 796 Sodium Chloride Injection, 752
Insulin, 539 Thiopental Sodium for, 798 Sodium Chloride Solution, 752
Isoniazid, 554 Tubocurarine Chloride, 831
Levallorphan Tartrate, 570 Tubocurarine Hydrochloride, 831 J
Lidocaine, 575 Vasopressin, 837
Lidocaine Hydrochloride, 575 Vinblastine Sulfate for, 841 Japanese
Magnesium Sulfate, 586 Vitamin B12, 389 Angelica Root, 956
D -Mannite, 588 Vitamin B1 Hydrochloride, 794 Encephalitis Vaccine, 956
D -Mannitol, 588 Vitamin B2 Phosphate Ester, 736 Encephalitis Vaccine, Freeze-dried,
Meglumine Amidotrizoate, 594 Vitamin B6, 725 957
Meglumine Iotalamate, 595 Vitamin C, 251 Gentian, 957,
Meglumine Sodium Weak Opium Alkaloids and Gentian, Powdered, 1551
Amidotrizote, 596 Scopolamine, 992 Valerian, 957
Meglumine Sodium Iodamide, 597 Xylitol, 845 Josamycin, 559, 1479
Metenolone Enanthate, 608 Insulin, 536 Propionate, 559, 1480
Morphine and Atropine, 978 Insulin Human (Genetical Jujube, 958
Morphine Hydrochloride, 630 Recombination), 537 Seed, 1551
Neostigmine Methylsulfate, 638 Injection, 539
Nicotinic Acid, 645 Zinc Injection (Aqueous K
Noradrenaline Hydrochloride, 650 Suspension), 542
Norepinephrine, 650 Zinc Injection (Aqueous Kainic Acid, 560
Norepirenamine Hydrochloride, Suspension), Amorphous, 542 and Santonin Powder, 958
650 Zinc Injection (Aqueous Kallidinogenase, 560
Operidine, 679 Suspension), Crystalline, 543 Kanamycin
Opium Alkaloids and Atropine, Zinc Protamine Injection (Aqueous Monosulfate, 1481
990 Suspension), 544 Sulfate, 563, 1482
Opium Alkaloids and Iodamide, 545 Kaolin, 959
Scopolamine, 991 Iodinated (131I) Human Serum Kernel
Opium Alkaloids Hydrochlorides, Albumin Injection, 546 Apricot, 864
989 Iodine, 546 Peach, 1002
Oxycodone and Atropine, Glycerin, Compound, 950 Peach, Powdered, 1002
Compound, 996 Glycerin, Dental, 951 Ketamine Hydrochloride, 563
Oxycodone, Compound, 995 Tincture, 949 Ketoprofen, 564
Oxytocin, 665 Tincture, Dilute, 950 Ketotifen Fumarate, 1483
Paoaverine Hydrochloride, 671 Salicylic Acid and Phenol Spirit, Kitasamycin, 565, 1484
Pethidine Hydrochloride, 679 952 Tartrate, 1485
Phenolsulfonphthalein, 682 Iopamidol, 547
Phenytoin Sodium for, 686 Iopanoic Acid, 548, 1478 L
Prednisolone Sodium Succinate for, Tablets, 548, 1478
705 Iotalamic Acid, 549 Lactic Acid, 959
Procainamide Hydrochloride, 709 Iotroxic Acid, 550 Lactose, 960
Procaine Hydrochloride, 710 Ipecac, 953 Anhydrous, 961
Progesterone, 714 Powdered, 954 Lactulose, 566
Protamine Sulfate, 719 Syrup, 955 Lanatoside
Pyridoxine Hydrochloride, 725 Ipratropium Bromide, 550 C, 567
Reserpine, 730 Iproveratril Hydrochloride, 839 C Tablets, 567
Ribo‰avin Phosphate, 736 Isepamicin Sulfate, 552 Lanolin
Ribo‰avin Sodium Phosphate, L-Isoleucine, 553 Hydrous, 962
736 Isoniazid, 553 Puriˆed, 963
Sodium Bicarbonate, 750 Injection, 554, 1478 Lard, 964
Sodium Chloride, Isotonic, 752 Tablets, 554, 1478 Latamoxef Sodium, 569, 1486
Sodium Chloride, 0.9, 752 Isophane Insulin Injection (Aqueous Lauromacrogol, 964
Sodium Chloride, 10, 752 Suspension), 541 Leaf
Sodium Chromate (51Cr), 752 l-Isoprenaline Hydrochloride, 555 Bearberry, 870
Sodium Iodohippurate (131I), 756 Isopropanol, 556 Senna, Powdered, 1046
Sodium Pertechnetate (99mTc), 758 Isopropyl Alcohol, 556 Sweet Hydrangea, Powdered, 1063
Sulfobromophthalein Sodium, 775 Isopropylantipyrine, 556 Senna, 1044
Sulpyrine, 777 l-Isoproterenol Hydrochloride, 555 Sweet Hydrangea, 1063
Suxamethonium Chloride, 781 Isosorbide, 557 Lenampicillin Hydrochloride, 1488
Suxamethonium Chloride for, 781 Dinitrate, 558 L-Leucine, 569
Testosterone Enanthate, 787 Dinitrate Tablets, 558 Leucomycin, 565
Thallium (201Tl) Chloride, 791 Isotonic Levallorphan
Thiamine Hydrochloride, 794 Salt Solution, 752 Tartrate, 570
1650 Index Supplement I, JP XIV

Tartrate Injection, 570 Powdered, 1552 dl-Methylephedrine


Levodopa, 571 Mallotus Bark, 971 Hydrochloride, 613
Levomepromazine Maleate, 572 Maltose, 586 Hydrochloride Powder, 614
Levothyroxine Mamushi Antivenom, Equine, Freeze- Hydrochloride Powder, 10, 614
Sodium, 572 dried, 972 Methylergometrine
Sodium Tablets, 573 D -Mannite, 587 Maleate, 615
Lidocaine, 574 Injection, 588 Maleate Tablets, 615
Hydrochloride Injection, 575 D -Mannitol, 587 Methyl
Injection, 575 Injection, 588, 1492 Parahydroxybenzoate, 976
Light Maprotiline Hydrochloride, 588 Salicylate, 618
Anhydrous Silicic Acid, 1050 Meclofenoxate Hydrochloride, 589 Methylprednisolone, 616
Liquid Para‹n, 1001 Mecobalamin, 590 Methylrosanilinium Chloride, 617
Lime Medazepam, 591 Methyltestosterone, 618
Chlorinated, 894 Medicinal Tablets, 619
Quick, 878 Carbon, 972 Meticrane, 619, 1493
Slaked, 877 Soap, 973 Metildigoxin, 620
Lincomycin Hydrochloride, 576, 1490 Mefenamic Acid, 592 Metoclopramide, 621
Liothyronine Mefruside, 592 Metronidazole, 622
Sodium, 576 Tablets, 593 Metyrapone, 623
Sodium Tablets, 577, 1490 Meglumine, 974 Mexiletine Hydrochloride, 623
Liqueˆed Amidotrizoate Injection, 594 Miconazole, 624
Carbolic Acid, 1011 Iotalamate Injection, 595 Nitrate, 625
Phenol, 1011 Sodium Amidotrizoate Injection, Microcrystalline Cellulose, 889
Liquid Para‹n, 1000 596 Micronomicin Sulfate, 626, 1494
Lithium Carbonate, 578 Sodium Iodamide Injection, 597 Midecamycin, 626
Lithospermum Root, 964 Melphalan, 597 Acetate, 627
Live Menatetrenone, 598, 1492 Migrenin, 627, 1495
Attenuated Measles Vaccine, Freeze- Mentha Herb, 974 Minocycline Hydrochloride, 628,
dried, 972 Oil, 975 1495
Attenuated Mumps Vaccine, Freeze- Water, 975 Mitomycin C, 629, 1496
dried, 981 dl-Menthol, 975 Mixture, Magnesium Sulfate, 970
Attenuated Rubella Vaccine, Freeze- l-Menthol, 976 Monobasic Calcium Phosphate, 880
dried, 1029 Mepenzolate Bromide, 599 Monosodium
Oral Poliomyelitis Vaccine, 1015 Mepirizole, 448 Trichloroethyl Phosphate, 823
Longgu, 965 Mepitiostane, 600, 1492 Trichloroethyl Phosphate Syrup,
Loquat Leaf, 1552 Mepivacaine 824
Lorazepam, 579 Hydrochloride, 601 Morphine
Low Substituted Hydrochloride Injection, 602 and Atropine Injection, 978, 1553
Hydroxypropylcellulose, 942 Mequitazine, 602 Hydrochloride, 629, 1497
Loxoprofen Sodium, 580 Merbromin, 604 Hydrochloride Injection, 630, 1497
Lysine Hydrochloride, 581 Solution, 605 Hydrochloride Tablets, 631, 1498
Lysozyme Hydrochloride, 1491 Mercaptopurine, 603 Moutan Bark, 979
L-Lysine Hydrochloride, 581 Mercurochrome, 604 Mulberry Bark, 980, 1554
Solution, 605 Mupirocin Calcium Hydrate, 631
M Meropenem Trihydrate, 605
Mestranol, 606 N
Macrogol Metenolone
Ointment, 968 Acetate, 607 Nadolol, 633
400, 965 Enanthate, 607 Nalidixic Acid, 634
1500, 966 Enanthate Injection, 608 Naloxone Hydrochloride, 634
4000, 967 Methamphetamine Hydrochloride, Naphazoline
6000, 967 609 and Chlorpheniramine Solution,
20000, 968 L-Methionine, 609 981
Magnesium Methotrexate, 610, 1493 Hydrochloride, 635
Carbonate, 582 Methoxsalen, 611 Nitrate, 636
Oxide, 583 Methylbenactyzium Bromide, 611 Naproxen, 636
Silicate, 584 Methylcellulose, 978 Narcotine, 654
Stearate, 969 Methylchlorophenylisoxazolylpenicil- Hydrochloride, 655
Sulfate, 585 lin Sodium, 380 Natural Aluminum Silicate, 233
Sulfate Injection, 586, 1492 Methyldichlorophenylisoxazolylpenicil- Neomycin Sulfate, 497
Sulfate Mixture, 970 lin Sodium, 408 Neostigmine
Magnolia Bark, 970, 1552 Methyldopa, 612 Methylsulfate, 637
Flower, 1552 Tablets, 613 Methylsulfate Injection, 638
Supplement I, JP XIV Index 1651

Netilmicin Sulfate, 638 Turpentine, 1073 Pancuronium Bromide, 667


Nicardipine Hydrochloride, 639 Vitamin A, 1074 Panipenem, 667
Hydrochloride Injection, 640 Zinc Oxide, 1085 Pantethine, 669
Niceritrol, 641 Ointment Papaverine
Nicomol, 642 Absorptive, 855 Hydrochloride, 670
Tablets, 643 Acrinol and Zinc Oxide, 858 Hydrochloride Injection, 671
Nicotinamide, 643 Bufexamac, 290 Paracetamol, 219
Nicotinic Acid, 644 Hydrocortisone and Para‹n, 999
Injection, 645 Diphenhydramine, 939 Light Liquid, 1001
Nifedipine, 645 Hydrophilic, 940 Liquid, 1000
Nitrazepam, 646 Macrogol, 968 Paraformaldehyde, 671
Nitrogen, 982 Polyethylene Glycol, 968 Paste
Nitroglycerin Tablets, 647 Scopolia Extract and Tannic Acid Arsenical, 866
Nitrous Oxide, 648 Compound, 1039 Paraformaldehyde, Dental, 1001
Noradrenaline, 649 Simple, 1052 Triozinc, Dental, 1072
Hydrochloride, 650 Sulfur, Salicylic Acid and Pas-calcium, 301
Hydrochloride Injection, Thianthol, 1062 Granules, 302
Norepinephrine, 649, 1498 White, 1080 Peach Kernel, 1002
Injection, 650, 1499 Zinc Oxide, 1085 Peanut Oil, 1002
Norepirenamine, 649 Olive Oil, 986 Peel
Hydrochloride Injection, 650 Operidine, 678 Bitter Orange, 875
Norethisterone, 650 Injection, 679 Citrus Unshiu, 898
Nor‰oxacin, 651 Ophiopogon Tuber, 987 Penbutolol Sulfate, 672
Norgestrel, 652 Opium Penicillin
and Ethinylestradiol Tablets, 652, Alkaloids and Atropine Injection, G Potassium, 270
1499 990 Pentazocine, 672
Nortriptyline Hydrochloride, 654 Alkaloids and Scopolamine Pentobarbital Calcium, 673
Noscapine, 654 Injection, 991 Pentoxyverine Citrate, 674
Hydrochloride, 655 Alkaloids Hydrochlorides, 988, Peony Root, 1003, 1555
Notopterygium Rhizome, 1554 1555 Powdered, 1556
Nuphar Rhizome, 983 Alkaloids Hydrochlorides Peplomycin Sulfate, 675, 1503
Nux Injection, 989 Perilla Herb, 1004
Vomica, 983 Ipecac Powder, 993 Perphenazine, 675
Vomica Extract, 984 Powdered, 987 Maleate, 677
Vomica Extract Powder, 984 Tincture, 988 Maleate Tablets, 678
Vomica Tincture, 985 Orange Tablets, 676
Nystatin, 656 Immature, 948 Pethidine
Oil, 994 Hydrochloride, 678, 1505
O Peel Syrup, 994 Hydrochloride Injection, 679
Peel Tincture, 995 Petrolatum
O‰oxacin, 1500 Orciprenaline Sulfate, 656 White, 1005
Oil Oriental Bezoar, 995 Yellow, 1006
Acrinol and Zinc Oxide, 857 Oxapium Iodide, 657 Petroleum Benzin, 1006
Acrinol and Zinc Oxide, Oxaprozin, 658 Pharbitis Seed, 1007
Compound, 857 Oxazolam, 658 Phellodendron
Aromatic Castor, Oxetacaine, 659 Bark, 1007, 1556
Camellia, 889 Oxethazaine, 659 Albumin Tannate and Bismuth
Castor, 888 Oxprenolol Hydrochloride, 660 Subnitrate Powder, 1009, 1556
Cinnamon, 897 Oxybuprocaine Hydrochloride, 661 Powdered, 1556
Clove, 898 Oxycodone Hydrochloride, 662 Powder for Cataplasm, Compound,
Coconut, 899 Oxydol, 662 1556
Cod Liver, 900 Oxygen, 663 Phenacetin, 680
Corn, 903 Oxymetholone, 664 Phenazone, 247
Eucalyptus, 917 Oxytetracycline Hydrochloride, 665, Phenethicillin Potassium, 1505
Fennel, 919 1501 Phenobarbital, 681
Hydrogenated, 940 Oxytocin Injection, 665 Powder, 681
Mentha, 975 Oyster Shell, 997 Powder, 10, 681
Olive, 986 Phenol, 1010
Orange, 994 P and Zinc Oxide Liniment, 1011
Peanut, 1002 for Disinfection, 1010
Rape Seed, 1024 Panax Rhizome, 998 Liqueˆed, 1011
Sesame, 1048 Powdered, 1555 with Camphor, Dental, 1011
Soybean, 1059 Pancreatin, 999 Phenolated
1652 Index Supplement I, JP XIV

Water, 1012 Potassium Ribo‰avin, 734


Water for Disinfection, 1012 Benzylpenicillin, 270 Salicylated Alum, 1032
Phenolsulfonphthalein, 682 Bromide, 694 Scopolia Extract, 1036
Injection, 682 Canrenoate, 694 Scopolia Extract and Carbon, 1037
Phenovalin and Magnesium Oxide Carbonate, 1018 Scopolia Extract and Diastase,
Powder, 1012, 1557 Chloride, 695 Compound, 1037
L-Phenylalanine, 683 Clavulanate, 696, 1512 Scopolia Extract and Ethyl
Phenylbutazone, 684 Guaiacolsulfonate, 696 Aminobenzoate, 1037
Phenylephrine Hydrochloride, 684 Hydroxide, 1018 Scopolia Extract, Papaverine and
Phenytoin, 685 Iodide, 697 Ethyl Aminobenzoate, 1038
Powder, 686 Penicillin G, 270 Swertia and Sodium Bicarbonate,
Sodium for Injection, 686 Penicillin G, Crystalline, 270 1065
Tablets, 686 Permanganate, 698 Thiamine Hydrochloride, 794
Phytomenadione, 687 Sulfate, 1019 Vitamin B, Compound,
Phytonadione, 687, 1507 Potato Starch, 1019 Vitamin B1 Hydrochloride, 794
Picrasma Wood, 1013 Povidone, 1020 Vitamin B2, 734
Pilocarpine Hydrochloride, 687 Povidone-Iodine, 698, 1513 Vitamin C, 251
Pimaricin, 1507 Powder Zinc Oxide Starch, 1086
Pindolol, 688 Ascorbic Acid, 251 Powdered
Pinellia Tuber, 1013 Chlordiazepoxide, 351 Acacia, 855, 1543
Pipemidic Acid Trihydrate, 689, 1508 Chlorpheniramine and Calcium, Agar, 859
Piperacillin Sodium, 690, 1508 895 Alisma Rhizome, 860
Piperazine Chlorpheniramine Maleate, 358 Aloe, 861, 1544
Adipate, 691 Codeine Phosphate, 1, 382 Amomum Seed, 863
Phosphate, 692 Codeine Phosphate, 10, 383 Atractylodes Lancea Rhizome, 869
Phosphate Tablets, 692 Diastase and Sodium Bicarbonate, Atractylodes Rhizome, 868
Pirarubicin, 1509 907 Calumba, 881
Pirenoxine, 693 Diastase and Sodium Bicarbonate, Capsicum, 883, 1546
Pivmecillinam Hydrochloride, 693, Compound, 907 Cellulose, 892
1510 Dihydrocodeine Phosphate, 1, Cinnamon Bark, 897, 1547
Plantago 417 Clove, 898
Herb, 1014 Dihydrocodeine Phosphate, 10, Cnidium Rhizome, 899
Seed, 1014 418 Coix Seed, 900
Plaster, Adhesive, 858 Diphenhydramine and Coptis Rhizome, 902, 1548
Salicylic Acid, Adhesive, 1032 Bromovalerylurea, 911 Cyperus Rhizome, 906
Platycodon Diphenylhydantoin, 686 Digitalis, 910
Fluidextract, 1014 Dover's, 993 Dioscorea Rhizome, 911
Root, 1014 Ephedrine Hydrochloride, 446 Gambir, 920
Root, Powdered, 1015 Ephedrine Hydrochloride, 10, Gardenia Fruit, 923
Polyethylene 446 Gentian, 925
Glycol Ointment, 968 for Cataplasm, Phellodendron, Geranium Herb, 926
Glycol 400, 965 Compound, 1009 Ginger, 927
Glycol 1500, 966 Gentian and Sodium Bicarbonate, Ginseng, 929
Glycol 4000, 967 926 Glycyrrhiza, 933
Glycol 6000, 967 Hydralazine Hydrochloride, 513 Ipecac, 954
Glycol 20000, 968 Kainic Acid and Santonin, 958 Japanese Angelica Root, 956
Polygala dl-Methylephedrine Japanses Gentian, 957, 1551
Root, 1015 Hydrochloride, 614 Japanese Valerian, 958
Root, Powdered, 1016 dl-Methylephedrine Magnolia Bark, 971, 1552
Polygonum Root, 1557 Hydrochloride, 10, 614 Moutan Bark, 980
Polymixin B Sulfate, 694, 1511 Nux Vomica Extract, 984 Opium, 987
Polyoxyethylene Lauryl Alcohol Opium Ipecac, 993 Oyster Shell, 998
Ether, 964 Opium, Diluted, 987 Panax Rhizome, 998, 1555
Polyoxyl 40 Stearate, 1016 Phellodendron, Albumin Tannate Peach Kernel, 1002
Polyporus Sclerotium, 1016 and Bismuth Subnitrate, 1009 Peony Root, 1004, 1556
Polysorbate 80, 1017 Phenobarbital, 681 Phellodendron Bark, 1008, 1556
Polyvidone, 1020 Phenobarbital, 10, 681 Picrasma Wood, 1013
Polyvinylpyrrolidone, 1020 Phenovalin and Magnesium Oxide, Platycodon Root, 1015
K25, 1020 1012 Polygala Root, 1016
K30, 1020 Phenytoin, 686 Polyporus Sclerotium, 1016
K90, 1020 Reserpine, 731 Poria Sclerotium, 1017
Poria Sclerotium, 1017 Reserpine, 0.1, 731 Rhubarb, 1027, 1558
Potash Soap, 1018 Rhubarb and Senna, Compound, Rose Fruit, 1029
Supplement I, JP XIV Index 1653

Scutellaria Root, 1043, 1560 Lanolin, 963 Phosphate Injection, 736


Senega, 1044 Shellac, 1049 Powder, 734
Senna Leaf, 1046, 1561 Water, 1079 Sodium Phosphate, 735
Smilax Rhizome, 1053 Pyrantel Pamoate, 722 Sodium Phosphate Injection, 736
Sophora Root, 1058 Pyrazinamide, 723, 1515 Ribostamycin Sulfate, 737, 1520
Sweet Hydrangea Leaf, 1063 Pyridostigmine Bromide, 724 Rice Starch, 1028
Swertia Herb, 1064, 1562 Pyridoxine Rifampicin, 737, 1521
Tragacanth, 1072 Hydrochloride, 724 Ringer's Solution, 1028
Zanthoxylum Fruit, 1084 Hydrochloride Injection, 725 Rokitamycin, 737
Pranoprofen, 699 Pyroxylin, 1023 Root
Prazepam, 700 Pyrrolnitrin, 1516 Achyranthes, 857
Tablets, 701 Angelica Dahurica, 864
Precipitated Calcium Carbonate, 297 Q Asiasarum, 867
Prednisolone, 701 Astragalus, 867
Acetate, 703, 1513 Quick Lime, 878 Belladonna, 872
Sodium Succinate for Injection, Quinidine Sulfate, 725 Bupleurum, 876
705, 1513 Quinine Glehnia, 930
Succinate, 704 Ethyl Carbonate, 726 Japanese Angelica, 956
Tablets, 702 Hydrochloride, 727 Japanese Angelica, Powdered, 956
Primidone, 706, 1514 Sulfate, 728 Lithospermum, 964
Probenecid, 706 Peony, 1003
Tablets, 707 R Peony, Powdered, 1004
Procainamide Platycodon, 1014
Hydrochloride, 708 Ranitidine Hydrochloride, 1517 Polygala, 1015
Hydrochloride Injection, 709 Rape Seed Oil, 1024 Pueraria, 1023
Hydrochloride Tablets, 709 Red Ginseng, 1024 Rehmannia, 1025
Procaine Rehmannia Root, 1025 Saposhnikovia, 1034
Hydrochloride, 710 Reserpine, 729, 1518 Saussurea, 1034
Hydrochloride Injection, 710, 1514 Injection, 730 Scutellaria, 1042
Procarbazine Hydrochloride, 711, Powder, 731 Scutellaria, Powdered, 1043
1515 Powder, 0.1, 731 Sophora, 1058
Procaterol Hydrochloride, 712 Tablets, 731 Sophora, Powdered, 1058
Prochlorperazine Retinol Trichosanthes, 1072
Maleate, 713 Acetate, 732, 1519 Rose Fruit, 1029
Maleate Tablets, 713 Palmitate, 733, 1519 Rosin, 1029
Progesterone, 714 Rhizome Roxithromycin, 738, 1522
Injection, 714 Alisma, 860
Proglumide, 715 Alisma, Powdered, 860 S
Promethazine Hydrochloride, 716 Anemarrhena, 864
Propantheline Bromide, 716 Atractylodes, 868 Saccharated Pepsin, 1030
Propranolol Hydrochloride, 717 Atractylodes Lancea, 869 Saccharin Sodium, 1030
Propylene Atractylodes Lancea, Powdered, SaŒower, 1031
Glycol, 1022 869 SaŠron, 1031
Propyl Parahydroxybenzoate, 1021 Atractylodes, Powdered, 868 Salazosulfapyridine, 739
Propylthiouracil, 718 Cimicifuga, 896 Salbutamol Sulfate, 740
Tablets, 718 Cnidium, 899 Salicylated Alum Powder, 1032
Propyphenazone, 556 Cnidium, Powdered, 899 Salicylic Acid, 741
Prostaglandin Coptis, 901 Plaster, Adhesive, 1032
E1 a-Cyclodextrin Clathrate Coptis, Powdered, 902 Spirit, 1033
Compound, 231 Cyperus, 906 Santonin, 742
F2a, 425 Cyperus, Powdered, 906 Tablets, 742
Protamine Dioscorea, 911 Saponated Cresol Solution, 905
Sulfate, 719 Dioscorea, Powdered, 911 Saposhnikovia Root, 1034
Sulfate Injection, 719 Imperate, 949 Saussurea Root, 1034
Prothionamide, 720 Nuphar, 983 Schisandra Fruit, 1035
Protirelin, 720 Panax, 998 Schizonepeta Spike, 1035
Tartrate, 722 Panax, Powdered, 998 Sclerotium
Prunella Spike, 1023 Scopolia, 1040 Polyporus, Powdered, 1016
Pueraria Root, 1023, 1557 Rhubarb, 1026, 1558 Poria, 1017
Puriˆed Powdered, 1027, 1558 Poria, Powdered, 1017
Absorbent Cotton, 851 Ribo‰avin, 733 Scopolamine
Dehydrocholic Acid, 395 Butyrate, 734 Butylbromide, 743, 1522
Gelatin, 924 Phosphate, 735 Hydrobromide, 744
1654 Index Supplement I, JP XIV

Scopolia Aurothiomalate, 748 penicillin, 408


Extract, 1035 Benzoate, 749 Pertechnetate (99mTc) Injection, 758
Extract and Carbon Powder, 1036 Bicarbonate, 749 Phosphate, Dibasic, 907
Extract and Ethyl Aminobenzoate Bicarbonate and Bitter Tincture Picosulfate, 758
Powder, 1037, 1559 Mixture, 1054 Piperacillin, 690
Extract and Tannic Acid Bicarbonate Injection, 750, 1524 Polystyrene Sulfonate, 759
Ointment, Compound, 1560 Bisulˆte, 1054 Prasterone Sulfate, 760
Suppositories, 1039 Borate, 750 Pyrosulˆte, 1057
Suppositories, Compound, 1560 Bromide, 750 Saccharin, 1030
Extract Powder, 1036 Carbonate, 1055 Salicylate, 761
Extract, Papaverine and Ethyl Carbonate, Dried, Sulˆte, Dried, 1058
Aminobenzoate Powder, 1038 Carmellose, 887 Thiosulfate, 762
Rhizome, 1040 Cefalotin, 317 Thiosulfate Injection, 762
Scutellaria Root, 1042, 1560 Cefamandole, 317 Valproate, 762
Powdered, 1560 Cefapirin, 317 Solution
Secretin, 744 Cefazolin, 319 Adrenaline Hydrochloride, 448
Seed Cefbuperazone, 322 Alum, 861
Amomum, 863 Cefmetazole, 331 Benzalkonium Chloride, 267
Amomum, Powdered, 863 Cefoperazone, 332 Benzethonium Chloride, 269
Cassia, 888 Cefotaxime, 335 Chlorhexidine Gluconate, 353
Coix, 900 Cefoxitin, 336 Cresol, 905
Coix, Powdered, 900 Cefpiramide, 338 Cresol, Saponated, 905
Pharbitis, 1007 Cefsulodin, 340 Epinephrine, 448
Plantago, 1014 Ceftizoxime, 345 Epinephrine Hydrochloride, 448
Senega, 1043 Cefuroxime, 348 Epirenamine Hydrochloride, 448
Powdered, 1044 Chloride, 751, 1524 Glycerin and Potash, 930
Syrup, 1044 Chloride Injection, 0.9, 752 Idoxuridine Ophthalmic, 528
Senna Leaf, 1044, 1560 Chloride Injection, 10, 752, 1525 Merbromin, 605
Powdered, 1046, 1561 Chromate (51Cr) Injection, 752 Mercurochrome, 605
Serum Citrate, 753 Naphazoline and
Gonadotrophin, 936 Citrate Injection for Transfusion, Chlorpheniramine, 981
Gonadotrophin for Injection, 937 753 Ringer's, 1028
Sesame Oil, 1048 Cloxacillin, 380 Salt, Isotonic, 752
Shellac CMC, 887 Silver Nitrate Ophthalmic, 1041
Puriˆed, 1049 Cromoglicate, 754 Silver Protein, 1051
White, 1049 Dantrolene, 392 Sodium Chloride, Isotonic, 752
Shell Diclofenac, 405 Sodium Hypochlorite, Dental, 864
Oyster, 997 Dicloxacillin, 408 Sodium Iodide (131I), 756
Oyster, Powdered, 998 EDTA, 913 D-Sorbitol, 764
Siccaine, 1523 Flavin Adenine Dinucleotide, 475 Thianthol and Salicylic Acid,
Silicic Acid, Light Anhydrous, 1050 Fluorescein, 483 Compound, 1067
Silver Fosfomycin, 496 Tolnaftate, 816
Nitrate, 746 Fusidate, 1525 Zinc Sulfate Ophthalmic, 1086
Nitrate Ophthalmic Solution, 1041 Heparin, 510 50, Benzalkonium Chloride,
Protein, 1051 Hydrogen Sulˆte, 1054 Concentrated, 267
Protein Solution, 1051 Hydroxide, 1056 Sophora Root, 1058
Simˆbrate, 746, 1524 Iodide, 755 Sorbitan Sesquioleate, 1058
Simple Iodide (123I) Capsules, 755 D-Sorbitol, 763
Ointment, 1052 Iodide (131I) Capsules, 755 Solution, 764
Syrup, 1052 Iodide (131I) Solution, 756 Soybean Oil, 1059
Sinomenium Stem, 1052, 1562 Iodohippurate (131I) Injection, 756 Spectinomycin Hydrochloride, 1526
Sisomicin Sulfate, 747, 1524 Iopodate, 756, 1526 Spike
Slaked Lime, 877 Iopodate Capsules, 757, 1526 Prunella, 1023
Smilax Iotalamate Injection, 757 Schizonepeta, 1035
Rhizome, 1053 Latamoxef, 569 Spirit
Rhizome, Powdered, 1053 Lauryl Sulfate, 1056 Capsicum and Salicylic Acid, 884
Soap Levothyroxine, 572 Foeniculated Ammonia, 919
Medicinal, 973 Liothyronine, 576 Iodine, Salicylic Acid and Phenol,
Potash, 1018 Loxoprofen, 580 952
Sodium Metabisulˆte, 1057 Methyl Salicylate, Compound, 977
Acetate, 1053 Methylchlorophenylisoxazolyl- Salicylic Acid, 1033
Aminobenzylpenicillin, 246 penicillin, 380 Salicylic Acid, Compound, 1033
Ampicillin, 246 Methyldichlorophenylisoxazolyl- Spironolactone, 765
Supplement I, JP XIV Index 1655

Starch Ipecac, 955 Prednisolone, 702


Corn, 903 Orange Peel, 994 Probenecid, 707
Potato, 1019 Senega, 1044 Procainamide Hydrochloride, 709
Rice, 1028 Simple, 1052 Prochlorperazine Maleate, 713
Wheat, 1080 Trichloroethyl Phosphate, Propylthiouracil, 718
Stearic Acid, 1059 Monosodium, 824 Reserpine, 731
Stearyl Alcohol, 1059 Triclofos Sodium, 824 Santonin, 742
Stem Sulˆnpyrazone, 773
Akebia, 859 T Thiamazole, 792
Sinomenium, 1052 Tipepidine Hibenzate, 806
Sterile Tablets Tolbutamide, 814
Absorbent Cotton, 852 Acetylsalicylic Acid, 252 Trihexyphenidyl Hydrochloride,
Absorbent Gauze, 853 Ajmaline, 227 825
Puriˆed Absorbent Cotton, 852 Amitriptyline Hydrochloride, 240 Trimethadione, 827
Puriˆed Water, 1079 Aspirin, 252 Warfarin Potassium, 843
Streptomycin Sulfate, 766, 1527 Azathioprine, 258 Talampicillin Hydrochloride, 782,
Sucralfate, 766 Baclofen, 260 1529
Sucrose, 1060 Chlordiazepoxide, 352 Talc, 1065
Sulbactam Sodium, 768 Chlorpheniramine Maleate, 359 Tannalbin, 227
Sulbenicillin Sodium, 769, 1528 Chlorpromazine Hydrochloride, Tannic Acid, 782
Sulfadiazine Silver, 769 361 Tartaric Acid, 1066
Sulfafurazole, 773 Chlorpropamide, 362 Tegafur, 782, 1530
Sulfamethizole, 770 Clomifene Citrate, 374 Teicoplanin, 783, 1530
Sulfamethoxazole, 770 Codeine Phosphate, 384 Terbutaline Sulfate, 786
Sulfamonomethoxine, 771 Dichlorphenamide, 407 Testosterone
Sulfasalazine, 739 Diclofenamide, 407 Enanthate, 787
Sulˆnpyrazone, 772 Diethylcarbamazine Citrate, 409 Enanthate Injection, 787
Tablets, 773 Diethylstilbestrol Diphosphate, Propionate, 788
Sulˆsomezole, 770 494 Propionate Injection, 788
Sulˆsoxazole, 773 Digitoxin, 411 Tetanus Antitoxin, Equine, Freeze-
Sulfobromophthalein Digoxin, 416 dried, 1066
Sodium, 774 Dimenhydrinate, 423 Tetracaine Hydrochloride, 789
Sodium Injection, 775 Diphenylhydantoin, 686 Tetracycline Hydrochloride, 789
Sulfur, 775 Distigmine Bromide, 430 Thallium (201Tl) Chloride Injection,
and Camphor Lotion, 1062 Dydrogesterone, 438 791
Salicylic Acid and Thianthol Ephedrine Hydrochloride, 446 Theophylline, 791, 1531
Ointment, 1062 Ergometrine Maleate, 451 Thiamazole, 791
Sulpiride, 776 Estriol, 458 Tablets, 792
Sulpyrine, 776 Etacrynic Acid, 460 Thiamine
Injection, 777 Ethinylestradiol, 463 Hydrochloride, 793, 1532
Sultamicillin Tosilate, 778 Etilefrine Hydrochloride, 468 Hydrochloride Injection, 794
Sultiame, 779 Folic Acid, 491 Hydrochloride Powder, 794
Suppositorise Fosfestrol, 494 Nitrate, 795, 1532
Bisacodyl, 280 Hydralazine Hydrochloride, 514 Thiamylal
Indometacin, 536 Imipramine Hydrochloride, 531 Sodium, 796
Scopolia Extract and Tannic Acid, Iopanoic Acid, 548 Sodium for Injection, 796
1039 Isoniazid, 554 Thianthol, 1067
Scopolia Extract and Tannic Acid, Isosorbide Dinitrate, 558 Thiopental
Compound, 1041 Lanatoside C, 567 Sodium, 797, 1532
Suxamethonium Levothyroxine Sodium, 573 Sodium for Injection, 798
Chloride, 780 Liothyronine Sodium, 576 Thioridazine Hydrochloride, 798
Chloride for Injection, 781 Methyldopa, 613 Thiotepa, 799
Chloride Injection, 781 Methylergometrine Maleate, 615 L-Threonine, 799
Sweet Hydrangea Leaf, 1063 Methyltestosterone, 619 Thrombin, 1068
Swertia Morphine Hydrochloride, 631 Thymol, 1069
and Sodium Bicarbonate Powder, Nitroglycerin, 647 Thyroid, Dried, 1069
1065, 1563 Norgestrel and Ethinylestradiol, Tiaramide Hydrochloride, 800
Herb, 1063, 1562 652 Ticarcillin Sodium, 801
Powdered, 1562 Perphenazine, 676 Ticlopidine Hydrochloride, 802
Synthetic Perphenazine Maleate, 678 Timepidium Bromide, 803
Aluminum Silicate, 235 Phenytoin, 686 Tincture
Camphor, 306 Piperazine Phosphate, 682 Bitter, 875
Syrup Prazepam, 700 Capsicum, 883
1656 Index Supplement I, JP XIV

Iodine, 949 Trimetoquinol Hydrochloride, 827, Vincristine Sulfate, 842


Nux Vomica, 985 1538 Vitamin
Opium, 988 Tropicamide, 828 A Acetate, 732
Orange Peel, 995 L-Tryptophan, 829 A Capsules, 1075
Tinidazole, 804 Tuber A Oil, 1074, 1564
Tipepidine Corydalis, 904 A Oil Capsules, 1075, 1565
Hibenzate, 804, 1533 Ophiopogon, 987 A Palmitate, 733
Hibenzate Tablets, 806 Pinellia, 1013 B1 Hydrochloride, 793
Titanium Oxide, 1070 Tubocurarine B1 Hydrochloride Injection, 794
Toad Venom, 1071, 1563 Chloride, 830 B1 Hydrochloride Powder, 794
Tobramycin, 807, 1533 Chloride Injection, 831 B1 Nitrate, 795
Tocopherol, 807, 1534 Hydrochloride, 830 B2, 733
Acetate, 808, 1535 Hydrochloride Injection, 831 B2 Phosphate Ester, 735
Calcium Succinate, 809 Tulobuterol Hydrochloride, 831 B2 Phosphate Ester Injection, 736
Nicotinate, 810 Turpentine Oil, 1073 B2 Powder, 734
dl-a-Tocopherol, 807 B6, 724
Acetate, 808 U B6 Injection, 725
Nicotinate, 810 B12, 388
Todralazine Hydrochloride, 811 Ubidecarenone, 832 B12 Injection, 389
Toˆsopam, 812 Ulinastatin, 833 C, 250
Tolazamide, 813 Uncaria Thorn, 1563 C Injection, 251
Tolbutamide, 814 Urea, 835 C Powder, 251
Tablets, 814 Urokinase, 1073 D2, 449
Tolnaftate, 815 Ursodesoxycholic Acid, 835 D3, 363
Solution, 816 Uva Ursi Fluidextract, 1074 E, 807
Tolperisone Hydrochloride, 816 E Acetate, 808
Toxoid V E Calcium Succinate, 809
Diphtheria, 912 E Nicotinate, 810
Diphtheria-Tetanus, Combined, Vaccine K1, 687
913 Cholera, 896
Diphtheria-Tetanus, Combined, Diphtheria-Puriˆed Pertussis- W
Adsorbed, 913 Tetanus Combined, Adsorbed,
for Adult Use, Diphtheria, 1005 Warfarin
Adsorbed, 912 Hepatitis B, Adsorbed, 938 Potassium, 843
Habu-venom, Adsorbed, 938 In‰uenza HA, 949 Potassium Tablets, 843
Tetanus, Adsorbed, 1066 Japanese Encephalitis, 956 Water, 1076
Tragacanth, 1072 Japanese Encephalitis, Freeze-dried, Ammonia, 241
Powdered, 1072 957 Apricot Kernel, 865
Tranexamic Acid, 817 Live Attenuated Measles, Freeze-d- for Disinfection, Phenolated, 1012
Transfusion, Sodium Citrate Injection ried, 972 for Injection, 1078
for, 753 Live Attenuated Mumps, Freeze-d- Formalin, 920
Trapidil, 818 ried, 981 Mentha, 975
Trepibutone, 819 Live Attenuated Rubella, Freeze-d- Phenolated, 1012
Tretoquinol Hydrochloride, 827 ried, 1029 Puriˆed, 1079
Triamcinolone, 819, 1535 Live Oral Poliomyelitis, 1015 Sterile, Puriˆed, 1079
Acetonide, 820 Pertussis, Puriˆed, Adsorbed, 1005 Wax, Carnauba, 888
Triamterene, 821 Rabies, Inactivated Tissue Culture, Weak Opium Alkaloids and
Trichlormethiazide, 822 Freeze-dried, 1024 Scopolamine Injection, 992
Trichloroethyl Phosphate, Smallpox, Freeze-dried, 1053 Weil's Disease and Akiyami Combined
Monosodium, 823 Smallpox, Freeze-dried, Prepared in Vaccine, 1080
Trichomycin, 823 Cell Culture, 1053 Wheat Starch, 1080
Trichosanthes Root, 1072 Weil's Disease and Akiyami White
Triclofos Combined, 1080 Beeswax, 871
Sodium, 823 (for Percutaneous Use), BCG, Ointment, 1080
Sodium Syrup, 824 Freeze-dried, 869 Petrolatum, 1005
Trichomycin, 1536 L-Valine, 836 Shellac, 1049
Trihexyphenidyl Vancomycin Hydrochloride, 837, Soft Sugar, 1061
Hydrochloride, 824 1539 Whole Human Blood, 1080
Hydrochloride Tablets, 825 Vasopressin Injection, 837 Wine, 1081
Trimebutine Maleate, 1537 Verapamil Hydrochloride, 839 Wood
Trimetazidine Hydrochloride, 826 Vinblastine Picrasma, 1013
Trimethadione, 827 Sulfate, 840 Picrasma, Powdered, 1013
Tablets, 827 Sulfate for Injection, 841
Supplement I, JP XIV Index 1657

X Beeswax, 871 Oxide, 845


Petrolatum, 1006 Oxide Oil, 1085
Xylitol, 844 Oxide Ointment, 1085
Injection, 845, 1540 Z Oxide Starch Powder, 1086
Sulfate, 846
Y Zanthoxylum Fruit, 1083 Sulfate Ophthalmic Solution, 1086
Zedoary, 1084 Zinostatin Stimalamer, 846
Yeast, Dried, 1083 Zinc
Yellow Chloride, 1084
INDEX IN JAPANESE
アモキシシリン 244, 1381 イドクスウリジン点眼液 528, 1475
ア アモバルビタール 242 イブプロフェン 526
アラビアゴム 855, 1543 イミペネム 530, 1476
亜鉛華デンプン 1086 アラビアゴム末 855, 1543 イレイセン 1547
亜鉛華軟膏 1085 亜硫酸水素ナトリウム 1054 インジゴカルミン 533
アカメガシワ 971 アルジオキサ 228 インジゴカルミン注射液 533
アクチノマイシン D 225, 1381 アルプラゾラム 230 インスリン 536
アクリノール 224 アルプロスタジルアルファデクス インスリン亜鉛水性懸濁注射液 542
アクリノール・亜鉛華軟膏 858 231 インスリン注射液 539
アクリノール・チンク油 857 アロエ 860, 1544 インチンコウ 867
アザチオプリン 256 アロエ末 861, 1544 インドメタシン 534
アザチオプリン錠 257 アロプリノール 228 インドメタシンカプセル 535
亜酸化窒素 648 アンソッコウ 873 インドメタシン坐剤 536, 1477
アジピン酸ピペラジン 691 安息香酸 270 インフルエンザ HA ワクチン 949
アジマリン 226 安息香酸エストラジオール 455,
アジマリン錠 227 1462 ウ
亜硝酸アミル 246 安息香酸エストラジオール水性懸濁注
アスコルビン酸 250 射液 456 ウイキョウ 918
アスコルビン酸散 251 安息香酸エストラジオール注射液 ウイキョウ末 918
アスコルビン酸注射液 251 456 ウイキョウ油 918
アズトレオナム 258 安息香酸ナトリウム 749 ウリナスタチン 833
アスピリン 252 安息香酸ナトリウムカフェイン 296 ウルソデオキシコール酸 835
アスピリンアルミニウム 253 安息香酸ベンジル 874 ウロキナーゼ 1073
アスピリン錠 252 アンチピリン 247 ウワウルシ 870
アスポキシシリン 254 アンピシリン 246, 1382 ウワウルシ流エキス 1074
アセグルタミドアルミニウム 1377 アンピシリンナトリウム 246, 1385
アセタゾラミド 220 アンモニア・ウイキョウ精 919 エ
アセチルキタサマイシン 222, 1378 アンモニア水 241
アセチルスピラマイシン 224, 1378 エイジツ 1029
アセトアミノフェン 219 イ エイジツ末 1029
アセトヘキサミド 221 液状フェノール 1011
アセンヤク 920 イオウ 775 エスタゾラム 454
アセンヤク末 920 イオウ・カンフルローション 1062 エストリオール 457, 1463
亜ヒ酸パスタ 866 イオウ・サリチル酸・チアントール軟膏 エストリオール錠 458
アフロクアロン 225 1062 エストリオール水性懸濁注射液 458
アヘンアルカロイド・アトロピン注射 イオタラム酸 549 エタクリン酸 459
液 990 イオタラム酸ナトリウム注射液 757 エタクリン酸錠 460
アヘンアルカロイド・スコポラミン注 イオタラム酸メグルミン注射液 595 エタノール 814
射液 991 イオトロクス酸 550 エチオナミド 464
アヘン散 987 イオパノ酸 548, 1478 エチゾラム 1463
アヘンチンキ 988 イオパノ酸錠 548, 1478 エチニルエストラジオール 463
アヘン・トコン散 993 イオパミドール 547 エチニルエストラジオール錠 463
アヘン末 987 イオポダートナトリウム 756, 1526 エチルコハク酸エリスロマイシン
アマチャ 1063 イオポダートナトリウムカプセル 453
アマチャ末 1063 757, 1526 エチル炭酸キニーネ 727
アミドトリゾ酸 237 イクタモール 948 エチレンジアミン 917
アミドトリゾ酸ナトリウムメグルミン イソソルビド 557 エデト酸ナトリウム 913
注射液 596 イソニアジド 553 エーテル 462
アミドトリゾ酸メグルミン注射液 イソニアジド錠 554, 1478 エテンザミド 461
594 イソニアジド注射液 554, 1478 エトスクシミド 465
アミノ安息香酸エチル 465 イソフェンインスリン水性懸濁注射液 エナント酸テストステロン 787
アミノフィリン 239 541 エナント酸テストステロン注射液
アミノフィリン注射液 239 イソプロパノール 556 787
アムホテリシン B 245 イソプロピルアンチピリン 556 エナント酸フルフェナジン 487
アモキサピン 243 イドクスウリジン 527 エナント酸メテノロン 607

1659
1660 Index in Japanese Supplement I, JP XIV

エナント酸メテノロン注射液 608 塩酸エフェドリン散 10 446 塩酸ドブタミン 431, 1452


エノキサシン 444 塩酸エフェドリン錠 446 塩酸トリヘキシフェニジル 824
エピネフリン 447 塩酸エフェドリン注射液 445 塩酸トリヘキシフェニジル錠 825
エピネフリン液 447 塩酸 L-アルギニン 248 塩酸トリメタジジン 826
エピネフリン注射液 447 塩酸 L-アルギニン注射液 248 塩酸トリメトキノール 827, 1538
エピリゾール 448 塩酸 L-エチルシステイン 466 塩酸トルペリゾン 816
エリスロマイシン 453, 1460 塩酸 L-リジン 581 塩酸ナファゾリン 635
L-イソロイシン 553 塩酸オキシコドン 662 塩酸ナロキソン 634
エルカトニン 440 塩酸オキシテトラサイクリン 665, 塩酸ニカルジピン 639
L-カルボシステイン 310 1501 塩酸ニカルジピン注射液 640
エルゴカルシフェロール 449, 1460 塩酸オキシブプロカイン 661 塩酸ノスカピン 655
L-トリプトファン 829 塩酸オクスプレノロール 660 塩酸ノルトリプチリン 654
L-トレオニン 799 塩酸カルテオロール 313 塩酸バカンピシリン 259, 1389
L-バリン 836 塩酸キニーネ 727 塩酸パパベリン 670
L-フェニルアラニン 683 塩酸クリンダマイシン 1439 塩酸パパベリン注射液 671
L-メチオニン 609 塩酸クロカプラミン 370 塩酸バンコマイシン 837, 1539
L-ロイシン 569 塩酸クロコナゾール 387 塩酸ヒドララジン 513
塩化亜鉛 1084 塩酸クロニジン 376 塩酸ヒドララジン散 513
塩化アンベノニウム 236 塩酸クロフェダノール 371 塩酸ヒドララジン錠 514
塩化インジウム (111In) 注射液 534 塩酸クロペラスチン 377 塩酸ヒドロキシジン 524
塩化エドロホニウム 439 塩酸クロミプラミン 375 塩酸ヒドロコタルニン 522
塩化エドロホニウム注射液 440 塩酸クロルプロマジン 360 塩酸ピブメシリナム 693, 1510
塩化カリウム 695 塩酸クロルプロマジン錠 361 塩酸ビペリデン 279
塩化カルシウム 298 塩酸クロルプロマジン注射液 361 塩酸ピリドキシン 724
塩化カルシウム注射液 298, 1399 塩酸クロルヘキシジン 354 塩酸ピリドキシン注射液 725
塩化スキサメトニウム 781 塩酸ケタミン 563 塩酸ピロカルピン 687
塩化スキサメトニウム注射液 781 塩酸コカイン 381 塩酸フェニレフリン 684
塩化タリウム (201Tl) 注射液 791 塩酸シクロペントラート 389 塩酸ブクモロール 287
塩化ツボクラリン 830 塩酸ジフェニドール 409 塩酸ブナゾシン 291
塩化ツボクラリン注射液 831 塩酸ジフェンヒドラミン 427 塩酸ブフェトロール 288
塩化ナトリウム 751, 1524 塩酸ジブカイン 404 塩酸ブプラノロール 292
10 塩化ナトリウム注射液 752, 塩酸シプロヘプタジン 391 塩酸フラボキサート 476
1525 塩酸ジラゼプ 419 塩酸フルスルチアミン 499
塩化ベタネコール 278 塩酸ジルチアゼム 420, 1451 塩酸フルラゼパム 489
塩化ベルベリン 271 塩酸スペクチノマイシン 1526 塩酸ブレオマイシン 283, 1396
塩化ベンザルコニウム 266 塩酸セトラキサート 349, 1431 塩酸プロカイン 710
塩化ベンザルコニウム液 267 塩酸セフェタメト ピボキシル 328 塩酸プロカインアミド 708
塩化ベンゼトニウム 269 塩酸セフェピム 326 塩酸プロカインアミド錠 709
塩化ベンゼトニウム液 269 塩酸セフォゾプラン 337 塩酸プロカインアミド注射液 709
塩化メチルロザニリン 617 塩酸セフォチアム 335 塩酸プロカイン注射液 710, 1514
塩化リゾチーム 1491 塩酸セフォチアムヘキセチル 335, 塩酸プロカテロール 712
エンゴサク 904 1417 塩酸プロカルバジン 711, 1515
塩酸 514 塩酸セフカペン ピボキシル 322 塩酸プロプラノロール 717
塩酸アクラルビシン 224, 1379 塩酸セフメノキシム 330, 1410 塩酸ブロムヘキシン 285
塩酸アセブトロール 219 塩酸ダウノルビシン 1445 塩酸プロメタジン 716
塩酸アヘンアルカロイド 988, 1555 塩酸タランピシリン 782, 1529 塩酸ペチジン 678, 1505
塩酸アヘンアルカロイド注射液 989 塩酸チアミン 793, 1532 塩酸ペチジン注射液 679
塩酸アマンタジン 235 塩酸チアミン散 794 塩酸ベラパミル 839
塩酸アミトリプチリン 240 塩酸チアミン注射液 794 塩酸ベンセラジド 265
塩酸アミトリプチリン錠 240 塩酸チアラミド 800 塩酸ホモクロルシクリジン 512,
塩酸アルプレノロール 231 塩酸チオリダジン 798 1472
塩酸アロチノロール 249 塩酸チクロピジン 802 塩酸マプロチリン 588
l-塩酸イソプレナリン 555 塩酸ツロブテロール 831 塩酸ミノサイクリン 628, 1495
塩酸イダルビシン 527, 1475 塩酸テトラカイン 789 塩酸メキシレチン 623
塩酸イミプラミン 530 塩酸テトラサイクリン 789 塩酸メクロフェノキサート 589
塩酸イミプラミン錠 531 塩酸デメチルクロルテトラサイクリン 塩酸メタンフェタミン 609
塩酸インデノロール 532 1446 dl-塩酸メチルエフェドリン 613
塩酸エタンブトール 460 塩酸ドキサプラム 433 dl-塩酸メチルエフェドリン散 10
塩酸エチルモルヒネ 467 塩酸ドキシサイクリン 434, 1454 614
塩酸エチレフリン 468 塩酸ドキソルビシン 434, 1453 塩酸メピバカイン 601
塩酸エチレフリン錠 468 塩酸トドララジン 811 塩酸メピバカイン注射液 602
塩酸エピルビシン 1458 塩酸ドパミン 432 塩酸モルヒネ 629, 1497
塩酸エフェドリン 444 塩酸ドパミン注射液 433, 1452 塩酸モルヒネ錠 631, 1498
Supplement I, JP XIV Index in Japanese 1661

塩酸モルヒネ注射液 630, 1497 308, 1400 キョウニン 864


塩酸ラニチジン 1517 カルバマゼピン 308 キョウニン水 865
塩酸リモナーデ 939 カルバミン酸クロルフェネシン 356, 希ヨードチンキ 950
塩酸リンコマイシン 576, 1490 1436 金チオリンゴ酸ナトリウム 748
塩酸レナンピシリン 1488 カルビドパ 309, 1401
エンフルラン 443, 1456 カルメロース 885 ク
カルメロースカルシウム 886
オ カルメロースナトリウム 887 グアイフェネシン 505
カルモナムナトリウム 314, 1401 グアヤコールスルホン酸カリウム
オウギ 867 カルモフール 313 696
オウゴン 1042, 1560 カロコン 1072 クエン酸 366, 1437
オウゴン末 1043, 1560 カンゾウ 932, 1550 クエン酸ガリウム (67Ga) 注射液 501
黄色ワセリン 1006 カンゾウ末 1550 クエン酸クロミフェン 373, 1441
オウバク 1007, 1556 乾燥亜硫酸ナトリウム 1058 クエン酸クロミフェン錠 374
オウバク・タンナルビン・ビスマス散 カンゾウエキス 933 クエン酸ジエチルカルバマジン 408
1009, 1556 乾燥甲状腺 1069 クエン酸ジエチルカルバマジン錠
オウバク末 1008, 1556 乾燥酵母 1083 409, 1449
オウレン 901, 1548 乾燥細胞培養痘そうワクチン 1053 クエン酸ナトリウム 753
オウレン末 902, 1548 乾燥ジフテリアウマ抗毒素 912 クエン酸フェンタニル 473
オキサゾラム 658 乾燥弱毒生おたふくかぜワクチン クエン酸ペントキシベリン 674
オキサプロジン 658 981 クジン 1058
オキシトシン注射液 665 乾燥弱毒生風しんワクチン 1029 クジン末 1058
オキシドール 662 乾燥弱毒生麻しんワクチン 972 苦味重曹水 1054
オキシメトロン 664 乾燥水酸化アルミニウムゲル 233 苦味チンキ 875
オキセサゼイン 659 乾燥水酸化アルミニウムゲル細粒 クラブラン酸カリウム 696, 1512
オフロキサシン 1500 233 グラミシジン 1470
オリブ油 986 カンゾウ粗エキス 934 クラリスロマイシン 367
オレンジ油 994 乾燥組織培養不活化狂犬病ワクチン グリシン 931
オンジ 1015 1024 グリセオフルビン 505, 1471
オンジ末 1016 乾燥炭酸ナトリウム 1055 グリセリン 504, 1469
乾燥痘そうワクチン 1053 グリセリンカリ液 930
カ 乾燥日本脳炎ワクチン 957 クリノフィブラート 369
乾燥破傷風ウマ抗毒素 1066 グリベンクラミド 502
カイニン酸 560 乾燥はぶウマ抗毒素 938 グルコン酸カルシウム 299
カイニン酸・サントニン散 958 乾燥 BCG ワクチン 869 グルコン酸クロルヘキシジン液 353
カオリン 959 乾燥ボツリヌスウマ抗毒素 876 クレオソート 904
カカオ脂 877 カンゾウ末 933 クレゾール 905
加香ヒマシ油 889 乾燥まむしウマ抗毒素 972 クレゾール水 905
カゴソウ 1023 乾燥硫酸アルミニウムカリウム 863 クレゾール石ケン液 905
カシュウ 1557 カンテン 859 クロキサシリンナトリウム 380,
ガジュツ 1084 カンテン末 859 1441
加水ラノリン 962 含糖ペプシン 1030 クロキサゾラム 380
ガスえそウマ抗毒素 923 d-カンフル 305, 1400 クロチアゼパム 378
ガーゼ 853 dl-カンフル 306, 1400 クロトリマゾール 379
カッコン 1023, 1557 肝油 900, 1548 クロナゼパム 375
過テクネチウム酸ナトリウム (99mTc) カンレノ酸カリウム 694 クロフィブラート 372
注射液 758 クロフィブラートカプセル 373
果糖 497 キ クロム酸ナトリウム (51Cr) 注射液
果糖注射液 498 752
カノコソウ 957 希塩酸 515 クロモグリク酸ナトリウム 754
カノコソウ末 958 キキョウ 1014 クロラムフェニコール 351, 1432
カフェイン 294 キキョウ末 1015 クロルジアゼポキシド 351
カプセル 885 キキョウ流エキス 1014 クロルジアゼポキシド散 351, 1434
カプトプリル 307 キクカ 1547 クロルジアゼポキシド錠 352, 1435
過マンガン酸カリウム 698 キササゲ 889 クロルフェニラミン・カルシウム散
b-ガラクトシダーゼ(アスペルギルス) キジツ 948 895
921 キシリトール 844 クロルプロパミド 362
b-ガラクトシダーゼ(ペニシリウム) キシリトール注射液 845, 1540 クロルプロパミド錠 362
921 吉草酸ベタメタゾン 277, 1395 クロロブタノール 894
カリジノゲナーゼ 560 キタサマイシン 565, 1484
カリ石ケン 1018 牛脂 870 ケ
カルナウバロウ 888 吸水軟膏 855
カルバゾクロムスルホン酸ナトリウム キョウカツ 1554 ケイガイ 1035
1662 Index in Japanese Supplement I, JP XIV

経口生ポリオワクチン 1015 酢酸グアナベンズ 506 ジクロフェナミド 406, 1448


ケイ酸マグネシウム 584 酢酸クロルマジノン 355, 1435 ジクロフェナミド錠 407
軽質無水ケイ酸 1050 酢酸コルチゾン 386 シクロホスファミド 390
軽質流動パラフィン 1001 酢酸トコフェロール 808, 1535 ジゴキシン 414
ケイヒ 896, 1547 酢酸ナトリウム 1053 ジゴキシン錠 416
ケイヒ末 897, 1547 酢酸ヒドロキソコバラミン 523 ジゴキシン注射液 415
ケイヒ油 897 酢酸ヒドロコルチゾン 517, 1474 シコン 964
結晶性インスリン亜鉛水性懸濁注射液 酢酸フタル酸セルロース 893 次硝酸ビスマス 282
543 酢酸プレドニゾロン 703, 1513 ジスルフィラム 430
結晶セルロース 889 酢酸ミデカマイシン 627 ジソピラミド 429
血清性性腺刺激ホルモン 936 酢酸メテノロン 607 シタラビン 392
ケツメイシ 888 酢酸レチノール 732, 1519 シッカニン 1523
ケトプロフェン 564 サッカリンナトリウム 1030 ジドロゲステロン 437
ケンゴシ 1007 サフラン 1031 ジドロゲステロン錠 438
ゲンチアナ 925 サラシ粉 894 ジノスタチン スチマラマー 846
ゲンチアナ・重曹散 926 サラシミツロウ 871 ジノプロスト 425
ゲンチアナ末 925 サラゾスルファピリジン 739 ジピリダモール 428, 1451
ゲンノショウコ 926 サリチル・ミョウバン散 1032 ジフェンヒドラミン 426
ゲンノショウコ末 926 サリチル酸 741 ジフェンヒドラミン・フェノール・亜鉛
サリチル酸精 1033 華リニメント 912
コ サリチル酸ナトリウム 761 ジフェンヒドラミン・ワレリル尿素散
サリチル酸絆創膏 1032 911
コウカ 1031 サリチル酸メチル 618 ジフテリアトキソイド 912
硬化油 940 酸化亜鉛 845 ジフテリア破傷風混合トキソイド
コウジン 1024 酸化カルシウム 878 913
合成ケイ酸アルミニウム 235 酸化チタン 1070 ジプロピオン酸ベタメタゾン 275
コウブシ 906 酸化マグネシウム 583 シメチジン 365
コウブシ末 906 サンキライ 1053 ジメルカプロール 423
コウボク 970, 1552 サンキライ末 1053 ジメルカプロール注射液 424
コウボク末 970, 1552 三酸化ヒ素 250 ジメンヒドリナート 422
ゴオウ 995 サンシシ 922, 1549 ジメンヒドリナート錠 423
ゴシツ 857 サンシシ末 923 次没食子酸ビスマス 281
ゴシュユ 918, 1548 サンシュユ 903 ジモルホラミン 424
コハク酸クロラムフェニコールナトリ サンショウ 1083 ジモルホラミン注射液 425
ウム 1434 サンショウ末 1084 弱アヘンアルカロイド・スコポラミン
コハク酸トコフェロールカルシウム 酸素 663 注射液 992
809 サントニン 742 シャクヤク 1003, 1555
コハク酸ヒドロコルチゾン 521 サントニン錠 742 シャクヤク末 1004, 1556
コハク酸ヒドロコルチゾンナトリウム サンヤク 911 シャゼンシ 1014
520 サンヤク末 911 シャゼンソウ 1014
コハク酸プレドニゾロン 704 臭化イプラトロピウム 550
ゴボウシ 1546 シ 臭化カリウム 694
ゴマ油 1048 臭化ジスチグミン 429
ゴミシ 1035 ジアスターゼ 907 臭化ジスチグミン錠 430
コムギデンプン 1080 ジアスターゼ・重曹散 907 臭化水素酸スコポラミン 744
コメデンプン 1028 ジアゼパム 403 臭化水素酸デキストロメトルファン
コリスチンメタンスルホン酸ナトリウ シアナミド 387 402
ム 385 シアノコバラミン 388 臭化水素酸ホマトロピン 511
コルヒチン 384 シアノコバラミン注射液 389 臭化チメピジウム 803
コレカルシフェロール 363 ジオウ 1025 臭化ナトリウム 751
コレステロール 896 歯科用アンチホルミン 864 臭化パンクロニウム 667
コレラワクチン 896 歯科用トリオジンクパスタ 1072 臭化ピリドスチグミン 724
コロンボ 881 歯科用パラホルムパスタ 1001 臭化ブチルスコポラミン 743, 1522
コロンボ末 881 歯科用フェノール・カンフル 1011 臭化ブトロピウム 294
コンズランゴ 900 歯科用ヨード・グリセリン 951 臭化プロパンテリン 716
コンズランゴ流エキス 901 ジギタリス 908 臭化メチルベナクチジウム 611
ジギタリス末 910 臭化メペンゾラート 599
サ ジギトキシン 410 ジュウヤク 939
ジギトキシン錠 411 シュクシャ 863
サイクロセリン 390 シクラシリン 364, 1437 シュクシャ末 863
サイコ 876 ジクロキサシリンナトリウム 408 酒石酸 1066
サイシン 867, 1545 シクロスポリン 364 酒石酸アリメマジン 229
酢酸 856 ジクロフェナクナトリウム 405 酒石酸イフェンプロジル 529
Supplement I, JP XIV Index in Japanese 1663

酒石酸エルゴタミン 452 精製水 1079 センブリ・重曹散 1065, 1563


酒石酸キタサマイシン 1485 精製ゼラチン 924 センブリ末 1064, 1562
酒石酸プロチレリン 721 精製セラック 1049
酒石酸レバロルファン 570 精製脱脂綿 851 ソ
酒石酸レバロルファン注射液 570 精製デヒドロコール酸 395
ショウキョウ 927 精製白糖 1060 ソウジュツ 869
ショウキョウ末 927 精製ラノリン 963 ソウジュツ末 869
硝酸イソソルビド 558 生理食塩液 752 ソウハクヒ 981, 1554
硝酸イソソルビド錠 558 石油ベンジン 1006 ソヨウ 1004
硝酸銀 746 セクレチン 745
硝酸銀点眼液 1041 セスキオレイン酸ソルビタン 1058 タ
硝酸チアミン 795, 1532 セタノール 894
硝酸ナファゾリン 636 セッコウ 937 ダイオウ 1026, 1558
硝酸ミコナゾール 625 セネガ 1043 ダイオウ末 1026, 1558
常水 1076 セネガシロップ 1044 ダイズ油 1059
ショウズク 885 セネガ末 1044 タイソウ 958
消毒用エタノール 916 セファクロル 314, 1403 胎盤性性腺刺激ホルモン 934
消毒用フェノール 1010 セファゾリンナトリウム 319 タクシャ 860
消毒用フェノール水 1012 セファゾリンナトリウム水和物 319 タクシャ末 860
ショウマ 896 セファトリジンプロピレングリコール 脱脂綿 851
ジョサマイシン 559, 1478 318 タルク 1065
シンイ 1552 セファドロキシル 314 炭酸カリウム 1018
親水軟膏 940 セファピリンナトリウム 317 炭酸水素ナトリウム 749
親水ワセリン 1005 セファマンドールナトリウム 317, 炭酸水素ナトリウム注射液 750,
診断用クエン酸ナトリウム液 754 1407 1524
シンフィブラート 746, 1524 セファレキシン 315 炭酸ナトリウム 1055
セファロチンナトリウム 317, 1406 炭酸マグネシウム 582
ス セファロリジン 317, 1405 炭酸リチウム 578
セフィキシム 329 単シロップ 1052
水酸化カリウム 1018 セフォキシチンナトリウム 336, ダントロレンナトリウム 392
水酸化カルシウム 877 1419 単軟膏 1052
水酸化ナトリウム 1056 セフォジジムナトリウム 1412 タンニン酸 782
スクラルファート 766 セフォタキシムナトリウム 335, タンニン酸アルブミン 227
ステアリルアルコール 1059 1413 タンニン酸ジフェンヒドラミン 427
ステアリン酸 1059 セフォテタン 335, 1415 タンニン酸ベルベリン 272
ステアリン酸エリスロマイシン 454 セフォペラゾンナトリウム 332
ステアリン酸カルシウム 881 セフジトレン ピボキシル 325 チ
ステアリン酸ポリオキシル 40 1016 セフジニル 324
ステアリン酸マグネシウム 969 セフスロジンナトリウム 340 チアマゾール 791
スピロノラクトン 765 セフタジジム 342 チアマゾール錠 792
スルチアム 779 セフチゾキシムナトリウム 345 チアミラールナトリウム 796
スルバクタムナトリウム 768 セフチブテン 344, 1426 チアントール 1067
スルピリド 776 セフテラムピボキシル 344, 1425 チオテパ 799
スルピリン 776 セフトリアキソンナトリウム 346, チオペンタールナトリウム 797,
スルピリン注射液 777 1427 1532
スルファジアジン銀 769 セフピラミドナトリウム 338, 1421 チオ硫酸ナトリウム 762
スルファメチゾール 770 セフブペラゾンナトリウム 322, チオ硫酸ナトリウム注射液 762
スルファメトキサゾール 770 1409 チカルシリンナトリウム 801
スルファモノメトキシン 771 セフポドキシムプロキセチル 1422 チクセツニンジン 998
スルフイソキサゾール 773 セフミノクスナトリウム 332 チクセツニンジン末 998, 1555
スルフィンピラゾン 772 セフメタゾールナトリウム 331 窒素 982
スルフィンピラゾン錠 773 セフラジン 339 チニダゾール 804
スルベニシリンナトリウム 769, セフロキサジン 340, 1424 チモ 864
1528 セフロキシムアキセチル 347, 1429 チモール 1069
スルホブロモフタレインナトリウム セフロキシムナトリウム 348 注射用アモバルビタールナトリウム
774 ゼラチン 923 242
スルホブロモフタレインナトリウム注 センキュウ 899 注射用塩化アセチルコリン 222
射液 775 センキュウ末 899 注射用塩化スキサメトニウム 781
センコツ 983 注射用塩酸ヒドララジン 513
セ センソ 1071, 1563 注射用血清性性腺刺激ホルモン 937
センナ 1044, 1560 注射用コハク酸プレドニゾロンナトリ
成人用沈降ジフテリアトキソイド センナ末 1046, 1561 ウム 705, 1513
912 センブリ 1063, 1562 注射用水 1078
1664 Index in Japanese Supplement I, JP XIV

注射用胎盤性性腺刺激ホルモン 936 トウガラシチンキ 883 二酸化炭素 312


注射用チアミラールナトリウム 796 トウキ 956 ニセリトロール 641
注射用チオペンタールナトリウム トウキ末 956 ニトラゼパム 646
798 トウニン 1002 ニトログリセリン錠 647
注射用ファモチジン 470, 1464 トウニン末 1002 ニフェジピン 645
注射用フェニトインナトリウム 686 トウヒ 875 日本脳炎ワクチン 956
注射用硫酸ビンブラスチン 841 トウヒシロップ 994 乳酸 959
チョウジ 898 トウヒチンキ 995 乳酸カルシウム 300
チョウジ末 898 トウモロコシデンプン 903 乳糖 960
チョウジ油 898 トウモロコシ油 903 尿素 835
チョウトウコウ 1563 トコフェロール 807, 1534 ニンジン 927
チョレイ 1061 トコン 953 ニンジン末 929
チョレイ末 1061 トコンシロップ 955
チンク油 1085 トコン末 954 ノ
沈降ジフテリア破傷風混合トキソイド トシル酸スルタミシリン 778
913 トチュウ 1548 濃塩化ベンザルコニウム液 50 267
沈降精製百日せきジフテリア破傷風混 トフィソパム 812 濃グリセリン 504, 1469
合ワクチン 1005 トブラマイシン 807, 1533 ノスカピン 654
沈降精製百日せきワクチン 1005 トラガント 1072 ノルエチステロン 650
沈降炭酸カルシウム 297 トラガント末 1072 ノルエピネフリン 649, 1498
沈降破傷風トキソイド 1066 トラザミド 813 ノルエピネフリン注射液 650, 1499
沈降はぶトキソイド 938 トラネキサム酸 817 ノルゲストレル 652
沈降 B 型肝炎ワクチン 938 トラピジル 818 ノルゲストレル・エチニルエストラジ
チンピ 898 トリアムシノロン 819, 1535 オール錠 652, 1499
トリアムシノロンアセトニド 820 ノルフロキサシン 651
ツ トリアムテレン 821
トリクロホスナトリウム 823 ハ
ツバキ油 882 トリクロホスナトリウムシロップ
824 バイモ 1549
テ トリクロルメチアジド 822 白色セラック 1049
トリコマイシン 823, 1536 白色軟膏 1080
テイコプラニン 783, 1530 トリメタジオン 827 白色ワセリン 1005
D-ソルビトール 763 トリメタジオン錠 827 白糖 1061
D-ソルビトール液 764 トルナフタート 815 バクモンドウ 987
D-マンニトール 587 トルナフタート液 816 バクロフェン 260
D-マンニトール注射液 588 トルブタミド 814 バクロフェン錠 261
低置換度ヒドロキシプロピルセルロー トルブタミド錠 814 バシトラシン 1390
ス 942 トレピブトン 819 バソプレシン注射液 837
テオフィリン 791, 1531 トロピカミド 828 ハチミツ 938
テガフール 782, 1530 ドロペリドール 435, 1456 ハッカ 974
デキサメタゾン 397 トロンビン 1068 ハッカ水 975
デキストラン硫酸ナトリウム イオウ 豚脂 964 ハッカ油 975
5 401 パップ用複方オウバク散 1009, 1556
デキストラン硫酸ナトリウム イオウ ナ パニペネム 667
18 402 ハマボウフウ 930
デキストラン 40 398 ナイスタチン 656 パモ酸ヒドロキシジン 524
デキストラン 40 注射液 399 ナタネ油 1024 パモ酸ピランテル 722
デキストラン 70 400 ナドロール 633 パラアミノサリチル酸カルシウム
デキストリン 906 ナファゾリン・クロルフェニラミン液 301
デスラノシド 396 981 パラアミノサリチル酸カルシウム顆粒
デスラノシド注射液 397 ナプロキセン 636 302
デヒドロコール酸 394 ナリジクス酸 634 パラオキシ安息香酸エチル 916
デヒドロコール酸注射液 396 パラオキシ安息香酸ブチル 876
テレビン油 1073 ニ パラオキシ安息香酸プロピル 1021
天然ケイ酸アルミニウム 233 パラオキシ安息香酸メチル 976
テンマ 1550 ニガキ 1013 パラフィン 999
テンモンドウ 1546 ニガキ末 1013 パラホルムアルデヒド 671
ニコチン酸 644 バルビタール 263
ト ニコチン酸アミド 643 バルプロ酸ナトリウム 762
ニコチン酸注射液 645 パルミチン酸クロラムフェニコール
トウガラシ 882, 1546 ニコチン酸トコフェロール 810 1432
トウガラシ末 883, 1546 ニコモール 642 パルミチン酸レチノール 733, 1519
トウガラシ・サリチル酸精 884 ニコモール錠 643 バレイショデンプン 1019
Supplement I, JP XIV Index in Japanese 1665

ハロキサゾラム 509, 1472 フェニトイン 685 フルラゼパム 487


ハロタン 508 フェニトイン散 686 フルラゼパムカプセル 488
ハロペリドール 507 フェニトイン錠 686 フルルビプロフェン 489, 1466
パンクレアチン 999 フェニルブタゾン 684 プレドニゾロン 701
ハンゲ 1013 フェネチシリンカリウム 1505 プレドニゾロン錠 702
絆創膏 858 フェノバリン・マグネシア散 1012 フロクタフェニン 477, 1465
パンテチン 669 フェノバルビタール 681 プログルミド 715
パントテン酸カルシウム 301 フェノバルビタール散 10 681 プロゲステロン 714
フェノール 1010 プロゲステロン注射液 714
ヒ フェノール・亜鉛華リニメント 1011 フロセミド 498
フェノール水 1012 プロタミンインスリン亜鉛水性懸濁注
ピコスルファートナトリウム 758 フェノールスルホンフタレイン 682 射液 544
ビサコジル 280 フェノールスルホンフタレイン注射液 プロチオナミド 720
ビサコジル坐剤 280, 1396 682 プロチレリン 720
ビタミン A 油 1074, 1564 フェンブフェン 473 プロテイン銀 1051
ビタミン A 油カプセル 1075, 1565 複方アクリノール・チンク油 857 プロテイン銀液 1051
ヒトインスリン(遺伝子組換え) 537 複方オキシコドン・アトロピン注射液 プロピオン酸ジョサマイシン 559,
人全血液 1080 996 1480
人免疫グロブリン 949 複方オキシコドン注射液 995 プロピオン酸テストステロン 788
ヒドロキシプロピルセルロース 940 複方サリチル酸精 1033 プロピオン酸テストステロン注射液
ヒドロキシプロピルメチルセルロース 複方サリチル酸メチル精 977 788
フタレート 947 複方ジアスターゼ・重曹散 907 プロピオン酸ドロスタノロン 435,
ヒドロキシプロピルメチルセルロース 複方ダイオウ・センナ散 1028 1456
2208 943 複方チアントール・サリチル酸液 プロピオン酸ドロスタノロン注射液
ヒドロキシプロピルメチルセルロース 1067 436, 1456
2906 944 複方ビタミン B 散 1075 プロピオン酸ベクロメタゾン 264
ヒドロキシプロピルメチルセルロース 複方ヨード・グリセリン 950 プロピルチオウラシル 718
2910 945 複方ロートエキス・ジアスターゼ散 プロピルチオウラシル錠 718
ヒドロクロロチアジド 515, 1473 1037 プロピレングリコール 1022
ヒドロコルチゾン 516 複方ロートエキス・タンニン坐剤 フロプロピオン 478
ヒドロコルチゾン・ジフェンヒドラミ 1041, 1560 プロベネシド 706
ン軟膏 939 複方ロートエキス・タンニン軟膏 プロベネシド錠 707
ピペミド酸三水和物 689, 1508 1039, 1560 ブロマゼパム 284
ピペラシリンナトリウム 690, 1508 ブクリョウ 1017 ブロムワレリル尿素 287
ヒベンズ酸チペピジン 804, 1533 ブクリョウ末 1017 フロモキセフナトリウム 478, 1465
ヒベンズ酸チペピジン錠 806 フシジン酸ナトリウム 1525 粉末セルロース 892
ビホナゾール 278 ブスルファン 293
ヒマシ油 888 ブドウ酒 1081 ヘ
ピマリシン 1507 ブドウ糖 503
ヒメクロモン 525 ブドウ糖注射液 503 ベタメタゾン 274
ビャクシ 864 ブフェキサマク 289 ヘパリンナトリウム 510
ビャクジュツ 868 ブフェキサマク軟膏 290 ヘパリンナトリウム注射液 511
ビャクジュツ末 868 ブフェキサマク乳剤性軟膏 289 ベラドンナエキス 871
氷酢酸 856 フマル酸クレマスチン 368 ベラドンナコン 872
ピラジナミド 723, 1515 フマル酸ケトチフェン 1483 ペルフェナジン 675
ピラルビシン 1509 フマル酸ホルモテロール 492 ペルフェナジン錠 676
ピレノキシン 693 ブメタニド 291 ベンジルアルコール 874
ピロ亜硫酸ナトリウム 1057 プラステロン硫酸ナトリウム 760 ベンジルペニシリンカリウム 270,
ピロキシリン 1023 プラゼパム 700 1393
ピロールニトリン 1516 プラゼパム錠 701 ベンジルペニシリンベンザチン
ビワヨウ 1552 プラノプロフェン 699 1392
ピンドロール 688 フラビンアデニンジヌクレオチドナト ベンズブロマロン 268, 1391
ビンロウジ 866, 1545 リウム 475 ペンタゾシン 672
プリミドン 706, 1514 ベントナイト 873
フ フルオキシメステロン 486 ペントバルビタールカルシウム 673
フルオシノニド 482
ファノバリン・マグネシア散 1557 フルオシノロンアセトニド 481 ホ
ファモチジン 469 フルオレセインナトリウム 483
ファモチジン散 469 フルオロウラシル 485 ボウイ 1052, 1562
ファモチジン錠 471 フルオロメトロン 484 ボウコン 949, 1551
ファロペネムナトリウム 472 フルジアゼパム 479 ホウ砂 750
フィトナジオン 687, 1507 フルシトシン 478 ホウ酸 284
フェナセチン 680 フルニトラゼパム 480 抱水クロラール 350
1666 Index in Japanese Supplement I, JP XIV

ボウフウ 1034 マレイン酸レボメプロマジン 572 メルカプトプリン 603


ホスフェストロール 493 D -マンニトール注射液 1492 メルファラン 597
ホスフェストロール錠 494 メロペネム 三水和物 605
ホスホマイシンカルシウム 494 ミ dl-メントール 975
ホスホマイシンナトリウム 496 l-メントール 976
ボタンピ 979 ミグレニン 627, 1495
ボタンピ末 980 ミコナゾール 624 モ
ポビドン 1020 ミツロウ 871
ポビドンヨード 699, 1513 ミデカマイシン 626 モクツウ 859
ホミカ 983 ミョウバン水 861 モッコウ 1034
ホミカエキス 984 モノステアリン酸アルミニウム 861
ホミカエキス散 984 ム モノステアリン酸グリセリン 931
ホミカチンキ 985 モルヒネ・アトロピン注射液 978,
ポリスチレンスルホン酸カルシウム 無晶性インスリン亜鉛水性懸濁注射液 1553
303, 1399 542
ポリスチレンスルホン酸ナトリウム 無水アンピシリン 246, 1384 ヤ
759 無水エタノール 915
ポリソルベート 80 1017 無水カフェイン 295 焼セッコウ 938
ホリナートカルシウム 299 無水クエン酸 367, 1438 ヤクチ 875
ホルマリン 919 無水乳糖 961 薬用石ケン 973
ホルマリン水 920 無水リン酸水素カルシウム 879 薬用炭 972
ボレイ 997 ムピロシンカルシウム 水和物 631 ヤシ油 899
ボレイ末 998
メ ユ

メキタジン 602 ユウタン 869
マイトマイシン C 629, 1496 メグルミン 974 ユーカリ油 917
マオウ 914 メコバラミン 590 輸血用クエン酸ナトリウム注射液
マーキュロクロム 604 メシル酸ガベキサート 500 753
マーキュロクロム液 605 メシル酸カモスタット 304 ユビデカレノン 832
マクリ 908 メシル酸ジヒドロエルゴタミン 418
マクロゴール軟膏 968 メシル酸ジヒドロエルゴトキシン ヨ
マクロゴール 400 965 413, 1450
マクロゴール 1500 966 メシル酸デフェロキサミン 393 ヨウ化エコチオパート 438
マクロゴール 4000 967 メシル酸ブロモクリプチン 286 ヨウ化オキサピウム 657
マクロゴール 6000 967 メシル酸ベタヒスチン 272 ヨウ化カリウム 697
マクロゴール 20000 968 メストラノール 606 ヨウ化人血清アルブミン (131I) 注射液
マシニン 1551 メダゼパム 591 546
麻酔用エーテル 462 メチクラン 619, 1493 ヨウ化ナトリウム 755
マルトース 586 メチラポン 623 ヨウ化ナトリウム (123I) カプセル
マレイン酸エルゴメトリン 450 メチルジゴキシン 620 755
マレイン酸エルゴメトリン錠 451 メチルセルロース 978 ヨウ化ナトリウム (131I) 液 756
マレイン酸エルゴメトリン注射液 メチルテストステロン 618 ヨウ化ナトリウム (131I) カプセル
451 メチルテストステロン錠 619 755
マレイン酸クロルフェニラミン 357 メチルドパ 612 ヨウ化ヒプル酸ナトリウム (131I) 注射
d-マレイン酸クロルフェニラミン メチルドパ錠 613 液 756
359 メチルプレドニゾロン 616 葉酸 490, 1466
マレイン酸クロルフェニラミン散 メチル硫酸ネオスチグミン 637 葉酸錠 491
358 メチル硫酸ネオスチグミン注射液 葉酸注射液 491
マレイン酸クロルフェニラミン錠 638 ヨウ素 546
359 滅菌ガーゼ 854 ヨクイニン 900
マレイン酸クロルフェニラミン注射液 滅菌精製水 1079 ヨクイニン末 900
357 滅菌精製脱脂綿 852 ヨーダミド 545
マレイン酸トリメブチン 1537 滅菌脱脂綿 852 ヨーダミドナトリウムメグルミン注射
マレイン酸プロクロルペラジン 713 メトキサレン 611 液 597
マレイン酸プロクロルペラジン錠 メトクロプラミド 621 ヨード・サリチル酸・フェノール精
713 メトトレキサート 610, 1493 952
マレイン酸ペルフェナジン 677 メトロニダゾール 622 ヨードチンキ 949
マレイン酸ペルフェナジン錠 678 メナテトレノン 598, 1492 ヨードホルム 546
マレイン酸メチルエルゴメトリン メピチオスタン 600, 1492
615 メフェナム酸 592 ラ
マレイン酸メチルエルゴメトリン錠 メフルシド 592
615 メフルシド錠 593 ラウリル硫酸ナトリウム 1056
Supplement I, JP XIV Index in Japanese 1667

ラウロマクロゴール 964 硫酸ストレプトマイシン 766, 1527 リン酸ピペラジン 692


酪酸ヒドロコルチゾン 518 硫酸セフォセリス 334 リン酸ピペラジン錠 692
酪酸リボフラビン 734 硫酸セフピロム 338 リン酸ベタメタゾンナトリウム 276,
ラクツロース 566 硫酸鉄 474 1395
ラクトビオン酸エリスロマイシン 硫酸テルブタリン 786 リン酸リボフラビンナトリウム 735
1462 硫酸ネチルマイシン 630 リン酸リボフラビンナトリウム注射液
ラタモキセフナトリウム 569, 1486 硫酸バメタン 262 736
ラッカセイ油 1002 硫酸バリウム 264
ラナトシド C 567 硫酸ビンクリスチン 842 レ
ラナトシド C 錠 567 硫酸ビンブラスチン 840
硫酸フラジオマイシン 497, 1467 レセルピン 729, 1518
リ 硫酸ブレオマイシン 283, 1398 レセルピン散 0.1 731
硫酸プロタミン 719 レセルピン錠 731
リオチロニンナトリウム 576 硫酸プロタミン注射液 719 レセルピン注射液 730
リオチロニンナトリウム錠 577, 硫酸ベカナマイシン 265, 1390 レボチロキシンナトリウム 572
1490 硫酸ペプロマイシン 675, 1503 レボチロキシンナトリウム錠 573
リドカイン 574 硫酸ペンブトロール 672 レボドパ 571
リドカイン注射液 575 硫酸ポリミキシン B 694, 1511 レンギョウ 920
リファンピシン 737, 1521 硫酸マグネシウム 585
リボフラビン 733 硫酸マグネシウム水 970 ロ
リボフラビン散 734 硫酸マグネシウム注射液 586, 1492
リュウコツ 965 硫酸ミクロノマイシン 626, 1494 ロキシスロマイシン 738, 1522
硫酸亜鉛 846 硫酸リボスタマイシン 737, 1520 ロキソプロフェンナトリウム 580
硫酸亜鉛点眼液 1086 リュウタン 957 ロキタマイシン 737
硫酸アストロマイシン 255, 1387 リュウタン末 957, 1551 ロジン 1029
硫酸アトロピン 255 流動パラフィン 1000 ロートエキス 1035
硫酸アトロピン注射液 256 リョウキョウ 1544 ロートエキス・アネスタミン散 1037,
硫酸アミカシン 238 リンゲル液 1028 1559
硫酸アルベカシン 247, 1386 リン酸クリンダマイシン 369, 1440 ロートエキス・カーボン散 1037
硫酸アルミニウムカリウム 862 リン酸コデイン 382 ロートエキス・タンニン坐剤 1039
硫酸イセパマイシン 552 リン酸コデイン散 1 382, 1442 ロートエキス・パパベリン・アネスタミ
硫酸エンビオマイシン 1457 リン酸コデイン散 10 383, 1442 ン散 1038
硫酸オルシプレナリン 656 リン酸コデイン錠 384, 1443 ロートエキス散 1036
硫酸カナマイシン 563, 1482 リン酸ジヒドロコデイン 417 ロートコン 1040
一硫酸カナマイシン 1481 リン酸ジヒドロコデイン散 1 417, ロラゼパム 579
硫酸カリウム 1019 1449
硫酸キニジン 725 リン酸ジヒドロコデイン散 10 ワ
硫酸キニーネ 728 418, 1450
硫酸グアネチジン 507 リン酸ジメモルファン 421 ワイル病秋やみ混合ワクチン 1080
硫酸ゲンタマイシン 502, 1468 リン酸水素カルシウム 878 ワルファリンカリウム 843
硫酸コリスチン 1444 リン酸水素ナトリウム 907 ワルファリンカリウム錠 843
硫酸サルブタモール 740 リン酸二水素カルシウム 880
硫酸シソマイシン 747, 1524 リン酸ヒドロコルチゾンナトリウム
硫酸ジベカシン 404, 1447 519, 1474

You might also like