You are on page 1of 254

The Ministry of Health, Labour and

Welfare Ministerial Notification No. 316

Pursuant to Paragraph 1, Article 41 of the Pharmaceutical AŠairs Law (Law No.


145, 1960), we hereby revise a part of the Japanese Pharmacopoeia (Ministerial
Notiˆcation No. 285, 2006) as follows*, and the revised Japanese Pharmacopoeia
shall come into eŠect on October 1, 2007. However, in the case of drugs which are
listed in the Japanese Pharmacopoeia (hereinafter referred to as ``previous Phar-
macopoeia'') [limited to those listed in the Japanese Pharmacopoeia whose standards
are changed in accordance with this notiˆcation (hereinafter referred to as ``new
Pharmacopoeia'')] and drugs which have been approved as of October 1, 2007 as
prescribed under Paragraph 1, Article 14 of the same law [including drugs the
Minister of Health, Labour and Welfare speciˆes (the Ministry of Health and Welfare
Ministerial Notiˆcation No. 104, 1994) as those exempted from marketing approval
pursuant to Paragraph 1, Article 14 of the Pharmaceutical AŠairs Law (hereinafter
referred to as ``drugs exempted from approval'')], the Name and Standards estab-
lished in the previous Pharmacopoeia (limited to part of the Name and Standards for
the drugs concerned) may be accepted to conform to the Name and Standards estab-
lished in the new Pharmacopoeia before and on March 31, 2009. In the case of drugs
which are listed in the new Pharmacopoeia (excluding those listed in the previous
Pharmacopoeia) and drugs which have been approved as of October 1, 2007 as
prescribed under Paragraph 1, Article 14 of the same law (including those exempted
from approval), they may be accepted as those being not listed in the new Phar-
macopoeia before and on March 31, 2009. Further, Standards listed in the section
9.01 Reference Standards of the General Tests, Processes and Apparatus of the previ-
ous Pharmacopoeia at the end of application of this notiˆcation may be treated under
the previous regulation irrespective of the prescription of the section 9.01(1) Refer-
ence Standards of the General Tests, Processes and Apparatus of the new Phar-
macopoeia.

Yoichi Masuzoe
The Minister of Health, Labour and Welfare

September 28, 2007

(The text 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, Pharmaceu-
tical and Food 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 of Supplement I to the Japanese Pharmacopoeia
Fifteenth Edition from General Notice to Ultraviolet-visible Reference Spectra (pp. 1789 – 1997).
CONTENTS
Preface ...................................................... i 9.22 Standard Solutions ........................ 1817
Supplement I to The Japanese Pharmacopoeia, 9.41 Reagents, Test Solutions................. 1817
Fifteenth Edition ............................. 1789–1997 9.42 Solid Supports/Column Packings for
General Notices ................................... 1789 Chromatography........................... 1824
General Rules for Crude Drugs ............... 1791
O‹cial Monographs ................................ 1825
General Rules for Preparations ............... 1793
Crude Drugs ....................................... 1937
General Tests, Processes and Apparatus ... 1795
1.09 Qualitative Tests ........................... 1795
Infrared Reference Spectra ................ 1971–1986
2.01 Liquid Chromatography ................. 1795
2.02 Gas Chromatography..................... 1799
Ultraviolet-visible Reference Spectra .... 1987–1997
2.48 Water Determination (Karl Fischer
Method) ...................................... 1801
General Information
2.49 Optical Rotation Determination ....... 1801
8. International Harmonization Implemented
4.01 Bacterial Endotoxins Test ............... 1802
in the Japanese Pharmacopoeia Fifteenth
4.05 Microbiological Examination of Non-
Edition ......................................... 1999
sterile Products............................. 1802
12. Microbial Attributes of Non-sterile
6.01 Test for Metal Particles in Opthalmic
Pharmaceutical Products.................. 2004
Ointments.................................... 1813
21. Quality Control of Water for
6.08 Insoluble Particulate Matter Test for
Pharmaceutical Use......................... 2006
Ophthalmic Solutions..................... 1813
31. Purity Tests on Crude Drugs Using
6.10 Dissolution Test............................ 1813
Genetic Information........................ 2007
6.11 Foreign Insoluble Matter Test for
Ophthalmic Solutions..................... 1814
Index.................................................... 2011
9.01 Reference Standards ...................... 1814
Index in Latin Name................................ 2027
9.21 Standard Solutions for Volumetric
Index in Japanese.................................... 2029
Analysis ...................................... 1817
PREFACE
The 15th Edition of the Japanese Pharmacopoeia drugs, which are important from the viewpoint of
(JP) was promulgated by Ministerial Notification No. health care and medical treatment, clinical results and
285 of the Ministry of Health, Labour and Welfare frequency of use, as soon as possible after they reach
(MHLW) on March 31, 2006. the market.
In July 2006, the Committee on JP established the The target date for the publication of JP 16th Edi-
basic principles for the preparation of the JP 16th Edi- tion (the Japanese edition) was set as April 2011.
tion, setting out the roles and characteristics of the JP, JP Expert Committees are organized with the fol-
the definite measures for the revision, and the date of lowing panels: Panel on the Principles of Revisions;
the revision. Sub-committee on the Principles of Revisions; Panel
At the above Committee, the five basic principles of on Medicinal Chemicals; Panel on Antibiotics; Panel
JP, which we refer to as the ``five pillars'' were estab- on Biologicals; Panel on Crude Drugs; Panel on Phar-
lished as follows: 1) Including all drugs which are im- maceutical Excipients; Panel on Physico-Chemical
portant from the viewpoint of health care and medical Methods; Panel on Preparations; Panel on Physical
treatment; 2) Making qualitative improvement by in- Methods; Panel on Biological Tests; Panel on Nomen-
troducing the latest science and technology; 3) clature; Panel on International Harmonization; Panel
Promoting internationalization; 4) Making prompt on Pharmaceutical Water; and Panel on Reference
partial revision as necessary and facilitating smooth Standards. Furthermore, three working groups under
administrative operation; and 5) Ensuring transparen- the Panel on Medicinal Chemicals are established to
cy regarding the revision, and disseminating the JP to expedite discussion of revision drafts of Monographs.
the public. It was agreed that the Committee on JP In the Committee on JP, Takao Hayakawa took the
should make efforts, on the basis of these principles, role of chairman from March 2006 to September 2007.
to ensure that the JP is used more effectively in the In addition to the regular revision every five years in
fields of health care and medical treatment by taking line with the basic principles for the preparation of the
appropriate measurements, including getting the un- JP it was agreed that partial revision should be done as
derstanding and cooperation of other parties con- necessary to take account of recent progress of science
cerned. and in the interests of international harmonization.
It was agreed that the JP should provide an official In accordance with the above principles, the panels
standard, being required to assure the quality of medi- initiated deliberations on selection of articles, and on
cines in Japan in response to the progress of science revisions for General Notices, General Rules for
and technology and medical demands at the time. It Crude Drugs, General Rules for Preparations, Gener-
should define the standards for specifications, as well al Tests, Monographs and so on.
as the methods of testing to assure overall quality of Draft revisions covering subjects in General No-
all drugs in principle, and it should have a role in tices, General Rules for Crude Drugs, General Rules
clarifying the criteria for quality assurance of drugs for Preparations, General Tests and Monographs, for
that are recognized to be essential for public health which discussions were finished between September
and medical treatment. 2005 and March 2007, were prepared for a supplement
The JP has been prepared with the aid of the to the JP 15. They were examined by the Committee
knowledge and experience of many professionals in on JP in April 2007, followed by the Pharmaceutical
the pharmaceutical field. Therefore, the JP should Affairs and Food Sanitation Council (PAFSC) in June
have the characteristics of an official standard, which 2007, and then submitted to the Minister of MHLW.
might be widely used by all parties concerned. It Numbers of discussions in the panels to prepare the
should provide information and understanding about supplement drafts were as follows: Panel on Principles
the quality of drugs to the public, and it should be of Revisions (7); Sub-committee on the Principles of
conducive to smooth and effective regulatory control Revisions (6); Panel on Medicinal Chemicals (33, in-
of the quality of drugs, as well as promoting and cluding the working groups); Panel on Antibiotics (9);
maintaining international consistency and harmoniza- Panel on Biologicals (8); Panel on Crude Drugs (17);
tion of technical requirements. Panel on Pharmaceutical Excipients (7); Panel on
It was also agreed that JP articles should cover Physico-Chemical Methods (12); Panel on Prepara-
i
ii Preface Supplement I, JP XV

tions (10); Panel on Physical Methods (8); Panel on (15) Purity tests
Biological Tests (7); Panel on Nomenclature (9); Panel (16) Loss on drying or ignition, or water
on International Harmonization (2); and Panel on (17) Residue on ignition, total ash or acid-insoluble
Pharmaceutical Water (7). ash
It should be noted that in the preparation of the (18) Tests being required for pharmaceutical prepa-
drafts for the supplement, generous cooperation was rations and other special tests
given by the Technical Committee of the Pharmaceuti- (19) Isomer ratio
cal Manufacturer's Association of Osaka and of (20) Assay or the content of the ingredient(s)
Tokyo, the Tokyo Crude Drugs Association, the (21) Containers and storage
Japan Pharmaceutical Excipients Council, the Japan (22) Expiration date
Kampo Medicine Manufacturers' Association, the (23) Others
Japan Flavor and Fragrance Materials Association,
4. In each monograph, the following physical and
the Japan Medical Plants Federation, the Japan Phar-
chemical values representing the properties and quali-
maceutical Manufacturers Association, and the Japan
ty of the drug are given in the order indicated below,
Oilseeds Processors Association.
except that unnecessary items are omitted depending
In consequence of this revision, the JP 15th Edition
on the nature of the drug:
carries 1567 articles, owing to the addition of 90 arti-
(1) Alcohol number
cles and the deletion of 6 articles.
(2) Absorbance
The principles of description and the salient points (3) Congealing point
of the revision in this Supplement are as follows: (4) Refractive index
1. The Supplement I to JP 15th Edition comprises (5) Osmolarity
the following items, in order: Notification of MHLW; (6) Optical rotation
Contents; Preface; General Notices; General Rules for (7) Viscosity
Crude Drugs; General Rules for Preparations; Gener- (8) pH
al Tests, Processes and Apparatus; Official Mono- (9) Specific gravity
graphs; Infrared Reference Spectra; and Ultraviolet- (10) Boiling point
visible Reference Spectra; then followed by General (11) Melting point
Information; and as an appendix a Cumulative Index (12) Acid value
containing references to the main volume and the Sup- (13) Saponification value
plement I. (14) Ester value
(15) Hydroxyl value
2. The articles in General Rules for Preparations,
(16) Iodine value
Official Monographs, Infrared Reference Spectra and
Ultraviolet-visible Reference Spectra are respectively 5. Identification tests comprise the following
placed in alphabetical order. items, which are generally put in the order given below:
(1) Coloration reactions
3. The following items in each monograph are put
(2) Precipitation reactions
in the order shown below, except that unnecessary i-
(3) Decomposition reactions
tems are omitted depending on the nature of the drug:
(4) Derivatives
(1) English title
(5) Infrared and/or ultraviolet-visible absorption
(2) Commonly used name(s)
spectrometry
(3) Latin title (only for crude drugs)
(6) Special reactions
(4) Title in Japanese
(7) Cations
(5) Structural formula or empirical formula
(8) Anions
(6) Molecular formula and molecular mass
(7) Chemical name 6. Purity tests comprise the following items, which
(8) Origin are generally put in the order given below, except that
(9) Limits of the content of the ingredient(s) and/or unnecessary items are omitted depending on the na-
the unit of potency ture of the drug:
(10) Labeling requirements (1) Color
(11) Method of preparation (2) Odor
(12) Description/Description of crude drugs (3) Clarity and/or color of solution
(13) Identification tests (4) Acidity or alkalinity
(14) Specific physical and/or chemical values (5) Acidity
Supplement I, JP XV Preface iii

(6) Alkalinity (1) 1.09 Qualitative Tests


(7) Chloride (2) 2.01 Liquid Chromatography
(8) Sulfate (3) 2.02 Gas Chromatography
(9) Sulfite (4) 2.48 Water Determination (Karl Fisher Method)
(10) Nitrate (5) 2.49 Optical Rotation Determination
(11) Nitrite (6) 4.01 Bacterial Endotoxins Test
(12) Carbonate (7) 4.05 Microbial Limit Test
(13) Bromide (8) 6.01 Test for Metal Particles in Ophthalmic
(14) Iodide Ointments
(15) Soluble halide (9) 6.08 Insoluble Particulate Matter Test for
(16) Thiocyanide Ophthalmic Solutions
(17) Selenium (10) 6.10 Dissolution Test
(18) Cationic salts
11. The following Reference Standards were new-
(19) Ammonium
ly added:
(20) Heavy metals
Amlexanox
(21) Iron
Amlodipine Besilate
(22) Manganese
Clobetasol Propionate
(23) Chromium
Enalapril Maleate
(24) Bismuth
Manidipine Hydrochloride
(25) Tin
Mizoribine
(26) Aluminum
Nabumetone
(27) Zinc
Nizatidine
(28) Cadmium
Ozagrel Sodium
(29) Mercury
Vincristine Sulfate
(30) Copper
Zidovudine
(31) Lead
(32) Silver 12. The following Reference Standards were delet-
(33) Alkaline earth metals ed.
(34) Arsenic Fosfestrol
(35) Foreign matter Hypromellose Phthalate
(36) Related substances Sulfinpyrazone
(37) Residual solvent Tubocurarine Chloride Hydrochloride
(38) Other impurities
13. English and Latin titles of drugs were based, in
(39) Readily carbonizable substances
principle, on the International Nonproprietary Names
7. The abbreviations used for the principal units, for Pharmaceutical Substances, and the chemical
``mol'', ``mmol'', ``mmol/L'' and ``Pa・s'' were ad- names were based on the Rules of the International
ded to and ``pH'' was deleted from the paragraph 9 of Union of Pure and Applied Chemistry (IUPAC).
the General Notices.
14. Molecular formulas of organic compounds be-
8. The following items of the General Rules for gin with C and then H, followed by other involved ele-
Preparations were revised: ments in the alphabetical order of the symbols of the
(1) Extracts elements.
(2) Ophthalmic Ointments
15. Structural formula of drug represents, as far
(3) Tinctures
as possible, the steric configuration.
(4) Ophthalmic Solutions
(5) Fluidextracts 16. The test procedures in monographs were writ-
ten in full, except within the same monograph and in
9. The following item was added to the General
the monographs for preparations having a cor-
Tests, Processes and Apparatus:
responding monograph of their principal material sub-
6.11 Foreign Insoluble Matter Test for Ophthalmic
stances.
Solutions
17. The following monographs were added:
10. The following items of the General Tests,
Acemetacin
Processes and Apparatus were revised:
Alminoprofen
iv Preface Supplement I, JP XV

Alminoprofen Tablets Josamycin Tablets


Alprostadil Injection Labetalol Hydrochloride
Amikacin Sulfate Injection Labetalol Hydrochloride Tablets
Amlexanox Manidipine Hydrochloride
Amlexanox Tablets Manidipine Hydrochloride Tablets
Amlodipine Besilate Minocycline Hydrochloride for Injection
Amosulalol Hydrochloride Mitomycin C for Injection
Amosulalol Hydrochloride Tablets Mizoribine
Ampicillin Sodium for Injection Mizoribine Tablets
Azelastine Hydrochloride Nabumetone
Aztreonam for Injection Nabumetone Tablets
Benzylpenicillin Potassium for Injection Nafamostat Mesilate
Biotin Nizatidine
Bisoprolol Fumarate Nizatidine Capsules
Bisoprolol Fumarate Tablets Omeprazole
Bucillamine Tablets Ozagrel Sodium
Buformin Hydrochloride Ozagrel Sodium for Injection
Buformin Hydrochloride Enteric-coated Tablets Peplomycin Sulfate for Injection
Buformin Hydrochloride Tablets Piperacillin Hydrate
Buprenorphine Hydrochloride Rokitamycin Tablets
Cefadroxil Capsules L-Serine
Cefadroxil for Syrup Sodium Starch Glycolate
Cefazolin Sodium for Injection Tobramycin Injection
Cefmetazole Sodium for Injection L-Tyrosine
Ceftazidime for Injection Ubenimex
Cetirizine Hydrochloride Zidovudine
Cetirizine Hydrochloride Tablets Aralia Rhizome
Chlorphenesin Carbamate Tablets Powdered Corydalis Tuber
Cibenzoline Succinate Crataegus Fruit
Cibenzoline Succinate Tablets Hangekobokuto Extract
Cilazapril Hydrate Keishibukuryogan Extract
Cilazapril Tablets Leonurus Herb
Clindamycin Phosphate Injection Lilium Bulb
Clobetasol Propionate Peucedanum Root
Clorazepate Dipotassium Powdered Turmeric
Clorazepate Dipotassium Capsules
18. The following monographs were revised:
L-Cysteine
Acetylcholine Chloride for Injection
L-Cysteine Hydrochloride Hydrate
Ajimaline Tablets
Domperidone
Aminophylline Injection
Doxorubicin Hydrochloride for Injection
Amitriptyline Hydrochloride Tablets
Emorfazone
L-Arginine Hydrochloride Injection
Enalapril Maleate
Ascorbic Acid Injection
Enalapril Maleate Tablets
Baclofen Tablets
Erythromycin Enteric-Coated Tablets
Betahistine Mesilate
Etizolam Fine Granules
Bisacodyl Suppositories
Etizolam Tablets
Calcium Chloride Injection
Felbinac
Calcium Folinate
L-Glutamine
Camostat Mesilate
Griseofulvin Tablets
Cefalotin Sodium
Ibudilast
Cefatrizine Propylene Glycolate
Isoxsuprine Hydrochloride
Chlordiazepoxide Tablets
Isoxsuprine Hydrochloride Tablets
Chlorphenesin Carbamate
Itraconazole
Chlorpromazine Hydrochloride Injection
Supplement I, JP XV Preface v

Chlorpromazine Hydrochloride Tablets Roxithromycin


Chlorpropamide Tablets Salicylic Acid
Cilostazol Tablets Sodium Bicarbonate Injection
Creosote 10z Sodium Chloride Injection
Cyanocobalamin Sodium Citrate Injection for Transfusion
Cyanocobalamin Injection Sodium Thiosulfate Injection
Deferoxamine Mesilate Sulbactam Sodium
Dehydrocholic Acid Injection Sulpyrine Injection
Deslanoside Injection Sultamicillin Tosilate Hydrate
Dextran 40 Suxamethonium Chloride for Injection
Anhydrous Dibasic Calcium Phosphate Suxamethonium Chloride Injection
Dibasic Calcium Phosphate Hydrate Talc
Dopamine Hydrochloride Injection Teceleukin for Injection (Genetical Recombination)
Edrophonium Chloride Injection Thiamine Chloride Hydrochloride Injection
Ephedrine Hydrochloride Injection Thiopental Sodium for Injection
Ephedrine Hydrochloride Tablets Tipepidine Hibenzate Tablets
Famotidine for Injection Trimetazidine Hydrochloride
Faropenem Sodium Hydrate Vincristine Sulfate
Faropenem Sodium Tablets Water for Injection
Faropenem Sodium for Syrup Xylitol Injection
Folic Acid Injection Alpinia Offcinarum Rhizome
Folic Acid Tablets Anemarrhena Rhizome
Fructose Injection Angelica Dahurica Root
Gabexate Mesilate Apricot Kernel
Glucose Injection Asiasarum Root
Hydralazine Hydrochloride Tablets Asparagus Tuber
Hypromellose Phthalate Atractylodes Rhizome
Idoxuridine Ophthalmic Solution Powdered Atractylodes Rhizome
Imipramine Hydrochloride Tablets Belladonna Extract
Indometacin Capsules Calumba
Isotonic Sodium Chloride Solution Powdered Calumba
Anhydrous Lactose Cimicifuga Rhizome
Levallorphan Tartrate Injection Clematis Root
Magnesium Sulfate Injection Cnidium Rhizome
D-Mannitol Injection Powdered Cnidium Rhizome
Medazepam Condurango Fluidextract
Mefruside Tablets Coptis Rhizome
Methyldopa Tablets Powdered Coptis Rhizome
Morphine Hydrochloride Tablets Corydalis Tuber
Neostigmine Methylsulfate Injection Cyperus Rhizome
Nicardipine Hydrochloride Injection Powdered Cyperus Rhizome
Nicorandil Dioscorea Rhizome
Nicotinic Acid Injection Powdered Dioscorea Rhizome
Noradrenaline Injection Fritillaria Bulb
Papaverine Hydrochloride Injection Gastrodia Tuber
Pethidine Hydrochloride Injection Gentian
Prednisolone Sodium Succinate for Injection Powdered Gentian
Protamine Sulfate Glehnia Root
Protamine Sulfate Injection Glycyrrhiza Extract
Pyridoxine Hydrochloride Injection Crude Glycyrrhiza Extract
Reserpine Injection Hochuekkito Extract
Riboflavin Sodium Phosphate Injection Imperata Rhizome
Ringer's Solution Ipecac
vi Preface Supplement I, JP XV

Powdered Ipecac Tubocurarine Chloride Hydrochloride Injection


Japanese Gentian
Those who were engaged in the preparation of Sup-
Powdered Japanese Gentian
plement I to JP 15 are as follows:
Japanese Valerian
Powdered Japanese Valerian
Norio Aimi Yuuichi Kikuchi
Kakkonto Extract
Fumiaki Akahori Mitsukazu Kitada
Kamishoyosan Extract
Mitsuo Aoki Fumiyuki Kiuchi
Lindera Root
Kiichi Aonuki Junko Kizu
Lithospermum Root
Nobuo Aoyagi** Takashi Kobayashi
Lycium Bark
Keiko Arimoto Masayoshi Kohase
Magnolia Bark
Hiroshi Asama Shigeo Kojima
Powdered Magnolia Bark
Kazuhide Ashizawa Hiroyasu Kokubo
Mulberry Bark
Shinichiro Aso Katsuko Komatsu
Notopterygium Rhizome
Yukio Aso Akio Komura
Nuphar Rhizome
Takashi Bamba Tetsuya Konda
Nux Vomica Extract
Makoto Emura Toshifumi Konda
Panax Japonicus Rhizome
Hiroyuki Fuchino Kenji Kondo
Powdered Panax Japonicus Rhizome
Shigeyuki Fujikura Seizo Kondo
Peach Kernel
Akihiko Fujise Takao Kunisada
Powdered Peach Kernel
Goro Funamoto Masaaki Kurihara
Perilla Herb
Yukihiro Goda Fumiyo Kusu
Platycodon Fluidextract
Ruri Hanajiri Kumiko Kusuyama
Polygala Root
Kouji Hasegawa Masako Maeda
Powdered Polygala Root
Mitsuru Hashida Midori Makita
Polygonum Root
Rika Hatano Yoshihisa Matsuda
Polyporus Sclerotium
Takao Hayakawa* Norio Matsuki
Powdered Polyporus Sclerotium
Masahiro Hayashi Eiichi Mikami
Processed Aconite Root
Yoshinori Hayashi Tsuyoshi Miyagawa
Powdered Processed Aconite Root
Kenji Higuchi Satoshi Minobe
Processed Ginger
Fusayoshi Hirayama Tsuyoshi Miura
Rehmannia Root
Yukio Hiyama Hiroto Miyamoto
Ryokeijutsukanto Extract
Kunimoto Hotta Naoki Miyata
Saposhnikovia Root
Masashi Hyuga Michinao Mizugaki
Saussurea Root
Nobukazu Igoshi Taiichi Mizuta
Scopolia Extract
Kenichi Inui Kaoru Morikawa
Scopolia Rhizome
Akiko Ishii Osamu Morita
Scutellaria Root
Yuji Ito Takashi Morita
Powdered Scutellaria Root
Takashi Itoh Toshimi Murai
Senega
Masao Izaki Masashi Muroi
Powdered Senega
Kenichi Izutsu Hiroaki Nagashige
Smilax Rhizome
Akemi Kai Shinsaku Nakagawa
Powdered Smilax Rhizome
Kazuaki Kakehi Emi Nakajima
Sophora Root
Takemine Kanai Hiroshi Nakamura
Powdered Sophora Root
Motoko Kanke Tatsuya Nakano
Turmeric
Hirohito Katayama Tatsumi Nakashima
Uva Ursi Fluidextract
Noriko Katori Mitsuo Nanaura
Zedoary
Nobuo Kawahara Masaaki Naotsuka
19. The following monographs were deleted: Toru Kawanishi Masao Nasu
Fosfestrol Yoshihiko Kawarasaki Shingo Niimi
Fosfestrol Tablets Nana Kawasaki Hiroshi Nishimura
Sulfinpyrazone Toshisuke Kawasaki Shukichi Ochiai
Sulfinpyrazone Tablets Yoshiaki Kawashima Atsuyuki Ohtsuki
Tubocurarine Chloride Hydrochloride Hydrate Keiji Kijima Ryozo Oishi
Supplement I, JP XV Preface vii

Minoru Okada Michiko Sekiguchi Haruo Tanaka Yoshimasa Uehara


Satoshi Okada Setsuko Sekita Toshihiro Tanaka Kazuichi Umemoto
Kimiya Okazaki Yasuo Shimada Kenichi Tanamoto** Haruo Watanabe
Tsuneo Okubo Kesamitsu Shimizu Tsuyoshi Tanimoto Takehiko Yajima
Haruhiro Okuda Kyoko Shimura Susumu Terabayashi Teruhide Yamaguchi
Masami Otsuka Osamu Shirota Reiko Teraoka Keiichi Yamamoto
Tadashi Ouchi Hisashi Sonobe Keijiro Terashita Keiji Yamamoto
Kazuhiro Owada Shoko Sueyoshi Masafumi Teshigawara Tosuke Yamamoto
Kenji Saiki Shinji Sugaya Hiroshi Tokunaga Takeshi Yamazaki
Yoshikazu Sakagami Hisakazu Sunada Kiyoshi Tomioka Masato Yasuhara
Eiji Sakai Hideyo Suzuki Motowo Tomita Hikaru Yoden
Tomoaki Sakamoto Yoshikazu Takahashi Hideya Tsuge Chikako Yomota
Hideki Sasaki Toshio Takachi Yosuke Tsuji Hitoo Yoshida
Hiroshi Sasaki Kikuo Takatera Eriko Uchida Sumie Yoshioka
Tsuguo Sasaki Tadahiro Takeda
*: Chairman, the Committee on JP
Motoyoshi Satake Yasushi Takeda
**: Acting Chairman, the Committee on JP
Kyoko Sato Toyoshige Tanabe
GENERAL NOTICES
Change the paragraph 9 to read: per centimeter cm-1
newton N
9. The following abbreviations are used for the
kilopascal kPa
principal units.
pascal Pa
meter m
mole per liter mol/L
centimeter cm
millimole per liter mmol/L
millimeter mm
pascal second Pa・s
micrometer mm
millipascal second mPa・s
nanometer nm
square millimeter per second mm2/s
kilogram kg
lux lx
gram g
mass per cent z
milligram mg
mass parts per million ppm
microgram mg
mass parts per billion ppb
nanogram ng
volume per cent volz
picogram pg
volume parts per million vol ppm
mole mol
mass per volume per cent w/vz
millimole mmol
endotoxin unit EU
Celsius degree 9 C
square centimeter cm2
Note: ``ppm'' used in the Nuclear Magnetic
liter L
Resonance Spectroscopy indicates the chemical shift,
milliliter mL
and ``w/vz'' is used in the formula or composition of
microliter mL
preparations.
megahertz MHz

1789
GENERAL RULES FOR
CRUDE DRUGS
Change the paragraph 1 to read: bitis Seed, Phellodendron Bark, Picrasma Wood, Pinellia
Tuber, Plantago Herb, Plantago Seed, Platycodon Root,
1. Crude drugs in the monographs include medicinal
Polygala Root, Polygonatum Rhizome, Polygonum Root,
parts obtained from plants or animals, cell inclusions and
Polyporus Sclerotium, Poria Sclerotium, Powdered Acacia,
secretes separated from the origins, their extracts, and
Powdered Agar, Powdered Alisma Rhizome, Powdered
minerals. General Rules for Crude Drugs and Crude Drugs
Aloe, Powdered Amomum Seed, Powdered Atractylodes
Test are applicable to the following:
Lancea Rhizome, Powdered Atractylodes Rhizome, Pow-
Acacia, Achyranthes Root, Agar, Akebia Stem, Alisma
dered Calumba, Powdered Capsicum, Powdered Cinnamon
Rhizome, Aloe, Alpinia Officinarum Rhizome, Amomum
Bark, Powdered Clove, Powdered Cnidium Rhizome,
Seed, Anemarrhena Rhizome, Angelica Dahurica Root,
Powdered Coix Seed, Powdered Coptis Rhizome, Powdered
Apricot Kernel, Aralia Rhizome, Areca, Artemisia Capil-
Corydalis Tuber, Powdered Cyperus Rhizome, Powdered
laris Flower, Asiasarum Root, Asparagus Tuber, Astragalus
Dioscorea Rhizome, Powdered Fennel, Powdered Gambir,
Root, Atractylodes Lancea Rhizome, Atractylodes Rhi-
Powdered Gardenia Fruit, Powdered Gentian, Powdered
zome, Bear Bile, Bearberry Leaf, Belladonna Root, Benin-
Geranium Herb, Powdered Ginger, Powdered Ginseng,
casa Seed, Benzoin, Bitter Cardamon, Bitter Orange Peel,
Powdered Glycyrrhiza, Powdered Ipecac, Powdered
Bupleurum Root, Burdock Fruit, Calumba, Capsicum,
Japanese Angelica Root, Powdered Japanese Gentian,
Cardamon, Cassia Seed, Catalpa Fruit, Chrysanthemum
Powdered Japanese Valerian, Powdered Magnolia Bark,
Flower, Cimicifuga Rhizome, Cinnamon Bark, Citrus
Powdered Moutan Bark, Powdered Oyster Shell, Powdered
Unshiu Peel, Clematis Root, Clove, Cnidium Monnieri
Panax Japonicus Rhizome, Powdered Peach Kernel,
Fruit, Cnidium Rhizome, Coix Seed, Condurango, Coptis
Powdered Peony Root, Powdered Phellodendron Bark,
Rhizome, Cornus Fruit, Corydalis Tuber, Crataegus Fruit,
Powdered Picrasma Wood, Powdered Platycodon Root,
Cyperus Rhizome, Digenea, Dioscorea Rhizome, Dolichos
Powdered Polygala Root, Powdered Polypourus Scleroti-
Seed, Eleutherococcus Senticosus Rhizome, Ephedra Herb,
um, Powderd Poria Sclerotium, Powdered Processed
Epimedium Herb, Eucommia Bark, Evodia Fruit, Fennel,
Aconite Root, Powdered Rhubarb, Powdered Rose Fruit,
Forsythia Fruit, Fritillaria Bulb, Gambir, Gardenia Fruit,
Powdered Scutellaria Root, Powdered Senega, Powdered
Gastrodia Tuber, Gentian, Geranium Herb, Ginger,
Senna Leaf, Powdered Smilax Rhizome, Powdered Sophora
Ginseng, Glehnia Root, Glycyrrhiza, Gypsum, Hemp Fruit,
Root, Powdered Sweet Hydrangea Leaf, Powdered Swertia
Honey, Houttuynia Herb, Immature Orange, Imperata
Herb, Powdered Tragacanth, Powdered Turmeric, Pow-
Rhizome, Ipecac, Japanese Angelica Root, Japanese Genti-
dered Zanthoxylum Fruit, Processed Aconite Root,
an, Japanese Valerian, Jujube, Jujube Seed, Leonurus
Processed Ginger, Prunella Spike, Pueraria Root, Red Gin-
Herb, Lilium Bulb, Lindera Root, Lithospermum Root,
seng, Rehmannia Root, Rhubarb, Rice Starch, Rose Fruit,
Longgu, Lonicera Leaf and Stem, Loquat Leaf, Lycium
Rosin, Safflower, Saffron, Saposhnikovia Root, Sappan
Bark, Lycium Fruit, Magnolia Bark, Magnolia Flower,
Wood, Saussurea Root, Schisandra Fruit, Schizonepeta
Mallotus Bark, Mentha Herb, Moutan Bark, Mulberry
Spike, Scopolia Rhizome, Scutellaria Root, Senega, Senna
Bark, Nelumbo Seed, Notopterygium Rhizome, Nuphar
Leaf, Sinomenium Stem, Smilax Rhizome, Sophora Root,
Rhizome, Nux Vomica, Ophiopogon Tuber, Oriental
Sweet Hydrangea Leaf, Swertia Herb, Turmeric, Toad
Bezoar, Oyster Shell, Panax Japonicus Rhizome, Peach
Venom, Tragacanth, Tribulus Fruit, Trichosanthes Root,
Kernel, Peony Root, Perilla Herb, Peucedanum Root, Phar-
Uncaria Hook, Zanthoxylum Fruit, Zedoary.

1791
GENERAL RULES
FOR PREPARATIONS
a transparency which does not interfere with the test for
7. Extracts foreign matter.
(9) Unless otherwise specified, Ophthalmic Solutions
meet the Insoluble Particulate Matter Test for Ophthalmic
Change (1) to read:
Solutions <6.08>.
(1) Extracts are prepared by evaporating the extractives
of crude drugs. Generally, there are two kinds of Extracts
which are: 27. Tinctures
(i) viscous extracts (ii) dry extracts

Change (2) to read:


8. Fluidextracts (2) Unless otherwise specified, Tinctures are usually pre-
pared from coarse powder or fine cuttings of crude drug
substance(s) either by maceration or by percolation as
Change (4) to read:
described below.
(4) Unless otherwise specified, Fluidextracts meet the Maceration: Place crude drugs in a suitable container, and
requirements of the Heavy Metals Limit Test <1.07> when add the total volume or about three-fourths of the total
the test solution and the control solution are prepared as volume of a solvent to be used. Stopper, and allow the
follows. container to stand at ordinary temperature with occasional
Test solution: Ignite 1.0 g of Fluidextracts to ash, warm stirring for about 5 days or until the soluble constituents
with 3 mL of dilute hydrochloric acid, filter, and wash the have satisfactorily dissolved. Filter the liquid through cloth.
residue with two 5 mL portions of water. Add 1 drop of In the case where about three-fourths of the total volume of
phenolphthalein TS to the combined filtrate and washings, the solvent is added, wash the residue with a suitable quanti-
add ammonia TS dropwise until the solution becomes a pale ty of the solvent, press, and combine the filtrate and wash-
red, filter, if necessary, and add 2 mL of dilute acetic acid ings to make up the volume. In the case where the total
and water to make 50 mL. volume of the solvent is added, sufficient solvent may be ad-
Control solution: Proceed with 3 mL of dilute ded, if necessary, to make up for the decreasing amount. Al-
hydrochloric acid as directed in the preparation of test solu- low the mixture to stand for about 2 days, and obtain a clear
tion, and add 3.0 mL of Standard Lead Solution and water liquid by decantation or filtration.
to make 50 mL. Percolation: Pour the solvent in small portions on crude
drugs placed in a container, and mix well to moisten the
crude drugs. Stopper the container, and allow it to stand for
17. Ophthalmic Ointments about 2 hours at room temperature. Pack the contents as
tightly as possible in a suitable percolator, open the lower
Change (5) to read: opening, and slowly pour sufficient solvent to cover the
crude drugs. When the percolate begins to drip, close the
(5) Unless otherwise specified, Ophthalmic Ointments opening, and allow the mixture to stand for 2 to 3 days at
meet the requirements of the Test for Metal Particles in room temperature. Open the opening, and allow the perco-
Ophthalmic Ointments <6.01>. late to drip at a rate of 1 to 3 mL per minute. Add an
appropriate quantity of the solvent, and continue to perco-
late until the desired volume has passed. Mix thoroughly,
18. Ophthalmic Solutions allow standing for 2 days, and obtain a clear liquid by
decantation or filtration. The time of standing and the flow
Change (8), (9) to read: rate may be varied depending on the kind and amount of
crude drugs to be percolated.
(8) Ophthalmic Solutions prepared as aqueous solution Tinctures prepared by either of the above methods for
and aqueous vehicles attached to Ophthalmic Solutions, which the content of the drug substance is specified are
unless otherwise specified, meet the requirement of the prepared by assaying the drug substance using a portion of
Foreign Insoluble Matter Test for Ophthalmic Solutions the sample and adjusting, if necessary, with the percolate or
<6.11>. The containers of Ophthalmic Solutions should have with the solvent to the specified content.

1793
GENERAL TESTS, PROCESSES
AND APPARATUS
Change the introduction to read: examination, purity test, loss on drying, total ash, acid-
insoluble ash, extract content, and essential oil content of
General Tests, Processes and Apparatus includes common
crude drugs are performed as directed in the corresponding
methods for tests, useful test methods for quality recogni-
items under Crude Drugs Test.
tion of drugs and other articles related to them. Unless
The number of each test method is a category number
otherwise specified, the procedures for acid-neutralizing
given individually. The number in blackets (< >) appeared
capacity determination of gastrointestinal medicines,
in monograph indicates the number corresponding to the
alcohol number determination, ammonium determination,
general test method.
arsenic determination, atomic absorption spectrophotomet-
ry, test for bacterial endotoxins, boiling point determina-
tion, distilling range determination, chloride determination,
conductivity measurement, congealing point determination, 1.09 Qualitative Tests
determination of bulk and tapped densities, digestion test,
Add the following next to Mercurous salt:
disintegration test, dissolution test, endpoint detection in
titrimetry, test of extractable volume for injection, flame Mesilate
coloration, fluorometry, foreign insoluble matter test for (1) To mesilates add twice its mass of sodium hydroxide,
injections, foreign insoluble matter test for ophthalmic heat gently to melt, and continue heating for 20 to 30
solutions, gas chromatography, heavy metals determination, seconds. After cooling, add a little amount of water, then
test for glass containers for injections, infrared spec- add dilute hydrochloric acid, and warm: the gas evolved
trophotometry, insoluble particulate matter test for injec- changes moistened potassium iodate-starch paper to blue.
tions, insoluble particulate matter test for ophthalmic (2) To mesilates add threefold its mass of sodium nitrate
solutions, iron determination, liquid chromatography, loss and anhydrous sodium carbonate, mix, and heat gradually.
on drying determination, loss on ignition determination, After cooling, dissolve the residue in diluted hydrochloric
melting point determination, test for metal particles in acid (1 in 5), and filter if necessary. The filtrate yields a
ophthalmic ointments, methanol determination, microbial white precipitate upon addition of barium chloride TS.
assay for antibiotics, test for microbial limit, test for
microbial limit for crude drugs, mineral oil determination,
nitrogen determination, nuclear magnetic resonance spec- 2.01 Liquid Chromatography
troscopy, optical rotation determination, osmolarity deter-
mination, oxygen flask combustion method, particle size
Change to read:
distribution test for preparations, pH determination, test for
plastic containers, powder particle density determination, Liquid Chromatography is a method to develop a mixture
powder particle size determination, test for pyrogen, injected into a column prepared with a suitable stationary
qualitative test, test for readily carbonizable substances, phase by passing a liquid as a mobile phase through the
refractive index determination, residual solvents test, residue column, in order to separate the mixture into its components
on ignition determination, test for rubber closure for aque- by making use of the difference of retention capacity against
ous infusions, specific gravity and density determination, the stationary phase, and to determine the components. This
specific surface area determination, test for sterility, sulfate method can be applied to a liquid or soluble sample, and is
determination, thermal analysis, thin-layer chromatogra- used for identification, purity test, and quantitative determi-
phy, test for total organic carbon, ultravioletvisible spec- nation.
trophotometry, uniformity of dosage units (test for content A mixture injected into the column is distributed between
uniformity, mass variation test), viscosity determination, the mobile phase and the stationary phase with a characteris-
vitamin A assay, water determination, and X-ray powder tic ratio (k) for each component.
diffraction are performed as directed in the corresponding
amount of compound in the stationary phase
articles under the General Tests, Processes and Apparatus. k=
amount of compound in the mobile phase
The tests for melting point of fats, congealing point of fatty
acids, specific gravity, acid value, saponification value, ester The ratio k represents the mass distribution ratio k? in liq-
value, hydroxyl value, unsaponifiable matter and iodine uid chromatography.
value of fats and fatty oils are performed as directed in the Since the relation given below exists among the ratio (k),
corresponding items under Fats and Fatty Oils Test, and the time for which the mobile phase is passed through the
sampling, preparation of sample for analysis, microscopic column (t0: time measured from the time of injection of a

1795
1796 General Tests, Processes and Apparatus Supplement I, JP XV

compound with k=0 to the time of elution at the peak maxi- the peak shape of the sample is unchanged after mixing the
mum), and the retention time (tR: time measured from the sample with an authentic specimen.
time of injection of a compound to be determined to the In general, the purity of the sample is determined by com-
time of elution at the peak maximum), the retention time for paring the sample solution with a standard solution which is
a compound on a column has a characteristic value under prepared by diluting the sample solution to a concentration
fixed chromatographic conditions. corresponding to the specified limit amount of the impurity,
or by the peak area percentage method. Unless otherwise
tR=(1+k) t0
specified, if a sample is separated into isomers in the chro-
Apparatus matogram, the isomer ratio is calculated by using the peak
Basically, the apparatus required for the liquid chromato- area percentage method.
graphic procedure consists of a pumping system for the The peak area percentage method is a method to calculate
mobile phase, a sample injection port, a column, a detector the proportion of the components from the ratio of the peak
and a recorder. A mobile phase component regulator, a ther- area of each component to the sum of the peak areas of
mostat for the column, a pumping system for reaction every peak recorded in the chromatogram. In order to
reagents and a chemical reaction chamber are also used, if obtain accurate results in evaluating the proportion of the
necessary. The pumping system serves to deliver the mobile components, it is necessary to correct the area of each
phase and the reagents into the column and connecting tube component based on the relative response factor to the prin-
at a constant flow rate. The sample injection port is used to cipal component.
deliver a quantity of the sample to the apparatus with high
Assay
reproducibility. The column is a tube with a smooth interior,
(1) Internal standard method—In the internal standard
made of inert metal, etc., in which a packing material for
method, choose a stable compound as an internal standard
liquid chromatography is uniformly packed. A column with
which shows a retention time close to that of the compound
a stationary phase chemically bound on the inside wall
to be assayed, and whose peak is well separated from all
instead of the column packed with the packing material may
other peaks in the chromatogram. Prepare several kinds of
be used. The detector is used to detect a property of the
standard solutions containing a fixed amount of the internal
samples which is different from that of the mobile phase,
standard and several graded amounts of the authentic speci-
and may be an ultraviolet or visible spectrophotometer,
men specified in the individual monograph. Based on the
fluorometric detector, differential refractometer, elec-
chromatogram obtained by injection of a fixed volume of
trochemical detector, chemiluminescence detector, electric
individual standard solutions, calculate the ratio of peak
conductivity detector, mass spectrophotometer, etc. The
area or peak height of the authentic specimen to that of the
output signal is usually proportional to the concentration of
internal standard, and prepare a calibration curve by plot-
samples at amounts of less than a few mg. The recorder is
ting these ratios on the ordinate against the amount of the
used to record the output signals of the detector. As
authentic specimen or the ratio of the amount of the authen-
required, a data processor may be used as the recorder to
tic specimen to that of the internal standard on the abscissa.
record or output the chromatogram, retention times or
The calibration curve is usually obtained as a straight line
amounts of the components. The mobile phase component
passing through the origin. Then, prepare a sample solution
regulator is used to vary the ratio of the mobile phase
containing the internal standard in the same amount as in
components in a stepwise or gradient fashion.
the standard solutions used for the preparation of the
Procedure calibration curve according to the method specified in the
Fix the detector, column and mobile phase to the appara- individual monograph, perform the liquid chromatography
tus, and adjust the flow rate and the column temperature to under the same operating conditions as for the preparation
the values described in the operating conditions specified in of the calibration curve, calculate the ratio of the peak area
the individual monograph. Inject a volume of the sample so- or peak height of the objective compound to that of the in-
lution or the standard solution specified in the individual ternal standard, and read the amount of the compound from
monograph with the sample injector into the column the calibration curve.
through the sample injection port. The separated compo- In an individual monograph, generally one of the stan-
nents are detected by the detector, and recorded by the dard solutions with a concentration within the linear range
recorder as a chromatogram. If the components to be of the calibration curve and a sample solution with a concen-
analyzed have no readily detectable physical properties such tration close to that of the standard solution are prepared,
as absorbance or fluorescence, the detection is achieved by and the chromatography is performed with these solutions
changing the components to suitable derivatives. Usually, under fixed conditions to determine the amount of the
the derivatization is performed as a pre- or post-column objective compound.
labeling. (2) Absolute calibration curve method—Prepare stan-
dard solutions with several graded amounts of the authentic
Identification and purity test
specimen, and inject accurately a fixed volume of these stan-
Identification of a component of a sample is performed by
dard solutions. With the chromatogram obtained, prepare a
confirming agreement of the retention time of the sample
calibration curve by plotting the peak areas or peak heights
with that of an authentic specimen, or by confirming that
Supplement I, JP XV General Tests, Processes and Apparatus 1797

on the ordinate against the amount of the authentic speci- tability'' is usually required, and in order to confirm, in
men on the abscissa. The calibration curve is generally some degree, the linearity of response near its specification
obtained as a straight line passing through the origin. Then, limit, the range of expected response to the injection of a
prepare a sample solution according to the method specified certain volume of target impurity solution at the concentra-
in the individual monograph, perform the liquid chro- tion of its specification limit should be prescribed. For limit
matography under the same conditions as for the prepara- test, ``Test for required detectability'' is not required, if the
tion of the calibration curve, measure the peak area or peak test is performed by comparing the response from sample so-
height of the objective compound, and read the amount of lution with that from standard solution at the concentration
the compound from the calibration curve. of its specification limit. ``Test for required detectability'' is
In an individual monograph, generally one of the stan- also not required, if it is confirmed that the impurity can be
dard solutions with a concentration within the linear range detected at its specification limit by the evaluation of ``Sys-
of the calibration curve and a sample solution with a concen- tem repeatability'' or some other procedure.
tration close to that of the standard solution are prepared, (2) System performance
and the chromatography is performed with these solutions When it is confirmed that the specificity for determining
under a fixed condition to obtain the amount of the compo- the test ingredient is ensured, it is considered verified that
nent. In this method, all procedures, such as the injection the system used has adequate performance to achieve its in-
procedure, must be carried out under a strictly constant tended use.
condition. In assay, ``System performance'' should be defined by the
resolution between the test ingredient and a target substance
Method for peak measuring
to be separated (a closely eluting compound is preferable),
Generally, the following methods are used.
and when appropriate, by their order of elution. In purity
(1) Peak height measuring method
tests, both the resolution and the order of elution between
(i) Peak height method: Measure the distance between
the test ingredient and a target substance to be separated (a
the maximum of the peak and the intersecting point of a
closely eluting compound is preferable) should be
perpendicular line from the maximum of the peak to the
prescribed. In addition, if necessary, the symmetry factor of
horizontal axis of recording paper with a tangent linking the
the test ingredient should be prescribed together with them.
baselines on both sides of the peak.
However, if there is no suitable target substance to be sepa-
(ii) Automatic peak height method: Measure the signals
rated, it is acceptable to define ``System performance'' using
from the detector as the peak height using a data processing
the number of theoretical plates and the symmetry factor of
system.
the test ingredient.
(2) Peak area measuring method
(3) System repeatability
(i) Width at half-height method: Multiply the peak
When it is confirmed that the degree of variation (preci-
width at the half-height by the peak height.
sion) of the response of the test ingredient is at a level that
(ii) Automatic integration method: Measure the signals
meets the requirement of ``System repeatability'', it is consi-
from the detector as the peak area using a data processing
dered verified that the system used has adequate perfor-
system.
mance to achieve its intended use.
System suitability The allowable limit of ``System repeatability'' is normally
System suitability is an integral part of analytical methods defined as the relative standard deviation (RSD) of the
using chromatography, and is used to ensure that the perfor- response of the test ingredient in replicate injections of stan-
mance of the chromatographic systems used is adequate for dard solution. It is acceptable to confirm the repeatability of
the analysis of the drug to be tested, as the suitability of the the system not only by replicate injections of standard solu-
method for the evaluation of the quality of the drug was tion before sample injections, but also by divided injections
verified. System suitability test should be carried out at every of standard solution before and after sample injections, or
series of drug analysis. The test procedures and acceptance by interspersed injections of standard solution among sam-
criteria of system suitability must be prescribed in the test ple injections.
method of the drug. Sample analysis is not acceptable unless In principle, total number of replicate injections should be
the requirements of system suitability have been met. 6. However, in the case that a long time is necessary for one
In system suitability test of the chromatographic systems, analysis, such as the analysis using the gradient method, or
the evaluation of ``System performance'' and ``System the analysis of samples containing late eluting components,
repeatability'' is usually required. For purity tests, the evalu- it may be acceptable to decrease the number of replicate in-
ation of ``Test for required detectability'' may also be re- jections by adopting new allowable limit of ``System repeat-
quired. ability'' which can guarantee a level of ``System repeatabili-
(1) Test for required detectability ty'' equivalent to that at 6 replicate injections.
For purity tests, when it is confirmed that the target im- The allowable limit of ``System repeatability'' should be
purity is distinctly detected at the concentration of its set at an appropriate level based on the validation data when
specification limit, it is considered verified that the system the suitability of the method for the evaluation of the quality
used has adequate performance to achieve its intended use. of the drug was verified.
For quantitative purity tests, ``Test for required detec-
1798 General Tests, Processes and Apparatus Supplement I, JP XV

Point to consider on changing the operating conditions Relative standard deviation: Generally, it is defined as
Among the operating conditions specified in the individ- RSD (z) in the following equation.
ual monograph, inside diameter and length of the column, n
particle size of the packing material, column temperature,
100
S(x -X̃)
i= 1
i
2

composition ratio of the mobile phase, composition of RSD (z)= ×


X̃ n-1
buffer solutions in the mobile phase, pH of the mobile
phase, concentration of ion-pair forming agents in the mo- xi: Observed value,
bile phase, ionic strength of the mobile phase, flow rate of X̃: Mean of observed values,
the mobile phase, number and timing of mobile phase com- n: Number of replicate measurements.
position changes in gradient program, flow rate of mobile
Complete separation of peak: It means that the resolution
phase in gradient program, composition and flow rate of
between two peaks is not less than 1.5. It is also called as
derivatizing reagents, and reaction time and chamber tem-
``baseline separation''.
perature in chemical reaction may be modified within the
ranges in which the liquid chromatographic system used con- Peak-valley ratio: It indicates the degree of separation be-
forms to the requirements of system suitability. tween 2 peaks on a chromatogram when baseline separation
cannot be attained, and is defined as p/v by the following
Terminology
formula.
S/N ratio: It is defined by the following formula.
Hp
2H p/ v =
S/N= Hv
h
Hp: peak height from the baseline of the minor peak,
H: Peak height of the target ingredient peak from the
Hv: height from the baseline of the lowest point (peak
baseline (the median value of background noise),
valley) of the curve between major and minor peaks.
h: Width of background noise of the chromatogram of
sample solution or solvent blank around the peak of
the target ingredient.

The baseline and background noise are measured over a


range 20 times of peak width at the center point of peak
height of the target ingredient. When a solvent blank is used,
measure over almost the same range as mentioned above
around the point where the target ingredient elutes.

Separation factor: It shows the relation between the reten-


tion times of peaks in the chromatogram, and is defined as a
in the following equation.

tR2-t0
a=
tR1-t0

tR1, tR2: Retention times of two compounds used for the


resolution measurement (tR1ºtR2),
t0: Time of passage of the mobile phase through the
Symmetry factor: It shows the degree of symmetry of a column (time measured from the time of injection of a
peak in the chromatogram, and is defined as S in the follow- compound with k=0 to the time of elution at the peak
ing equation. maximum).
W0.05 h The separation factor (a) indicates thermodynamic differ-
S=
2f ence in partition of two compounds. It is basically the ratio
W0.05 h: Width of the peak at one-twentieth of the peak of their partition equilibrium coefficients or of their mass-
height, distribution ratios, and is obtained from the chromatogram
f: Distance between the perpendicular from the peak max- as the ratio of the retention times of the two compounds.
imum and the leading edge of the peak at one-twentieth Resolution: It shows the relation between the retention
of the peak height, time and the peak width of peaks in the chromatogram, and
where W0.05 h and f have the same unit. is defined as RS in the following equation.
Supplement I, JP XV General Tests, Processes and Apparatus 1799

port and flow regulator for a combustion gas, a burning


tR2-tR1
RS=1.18× supporting gas and an accessory gas and sample injection
W0.5 h1+W0.5 h2
port for headspace are also used, if necessary. The carrier
tR1, tR2: Retention times of two compounds used for the gas-introducing port and flow regulator serves to deliver the
measurement of resolution (tR1ºtR2), carrier gas into the column at a constant flow rate, and
W0.5 h1, W0.5 h2: Peak widths at half peak height, usually consist of a pressure regulation valve, a flow rate
regulation valve and a pressure gauge. The sample injection
where tR1, tR2, W0.5 h1 and W0.5 h2 have the same unit.
port is used to deliver a quantity of the sample to the flow
Number of theoretical plates: It indicates the extent of line of carrier gas with high reproducibility. There are
band broadening of a compound in the column, and is sample injection ports for packed column and for capillary
generally defined as N in the following equation. column. There are both divided injection mode and non-
divided injection mode to sample injection port for capillary
tR2
N=5.54× column. The columns are usually classified as packed
W0.5 h2
column or capillary column. The packed column is a tube
tR: Retention time of compound, made of inert metal, glass or synthetic resin, in which a
W0.5 h: Width of the peak at half peak height, packing material for gas chromatography is uniformly pack-
ed. The packed column with not more than 1 mm in inside
where tR and W0.5 h have the same unit.
diameter is also called a packed capillary column (micro
Note Avoid the use of authentic specimens, internal stan- packed column). A capillary column is a tube made of inert
dards, reagents or solvents containing substances that may metal, glass, quartz or synthetic resin, whose inside wall is
interfere with the determination. bound chemically with stationary phase for gas chro-
matography. The column oven has the setting capacity for a
column with required length and the temperature regulation
2.02 Gas Chromatography system for keeping the constant column temperature. The
detector is used to detect a component separated on the
column, and may be an alkaline thermal ionization detector,
Change to read:
a flame photometry detector, mass spectrophotometer,
Gas Chromatography is a method to develop a mixture hydrogen flame-ionization detector, an electron capture
injected into a column prepared with a suitable stationary detector, a thermal conductivity detector, etc. The recorder
phase by passing a gas (carrier gas) as a mobile phase is used to record the output signals of the detector.
through the column, in order to separate the mixture into its
Procedure
components by making use of the difference of retention
Unless otherwise specified, proceed by the following
capacity against the stationary phase, and to determine the
method. Fix the detector, column and carrier gas to the
components. This method can be applied to a gaseous or
apparatus, and adjust the flow rate and the column tempera-
vaporizable sample, and is used for identification, purity
ture to the values described in the operating conditions speci-
test, and quantitative determination.
fied in the individual monograph. Inject a volume of the
A mixture injected into the column is distributed between
sample solution or the standard solution specified in the
the mobile phase and the stationary phase with a characteris-
individual monograph with the sample injector into the
tic ratio (k) for each component.
column system through the sample injection port. The sepa-
amount of compound in the stationary phase rated components are detected by the detector, and recorded
k=
amount of compound in the mobile phase by the recorder as a chromatogram.
Since the relation given below exists among the ratio (k), Identification and purity test
the time for which the mobile phase is passed through the Identification of a component of a sample is performed by
column (t0: time measured from the time of injection of a confirming agreement of the retention time of the sample
compound with k=0 to the time of elution at the peak maxi- with that of an authentic specimen, or by confirming that
mum), and the retention time (tR: time measured from the the peak shape of the sample is unchanged after mixing the
time of injection of a compound to be determined to the sample with an authentic specimen.
time of elution at the peak maximum), the retention time for In general, the purity of the sample is determined by com-
a compound on a column has a characteristic value under paring the sample solution with a standard solution which is
fixed chromatographic conditions. prepared by diluting the sample solution to a concentration
corresponding to the specified limit amount of the impurity,
tR=(1+k)t0
or by the peak area percentage method. Unless otherwise
Apparatus specified, if a sample is separated into isomers in the chro-
Basically, the apparatus required for the gas chromato- matogram, the isomer ratio is calculated by using the peak
graphic procedure consists of a carrier gas-introducing port area percentage method.
and flow regulator, a sample injection port, a column, a The peak area percentage method is a method to calculate
column oven, a detector and a recorder. Gas introducing the proportion of the components from the ratio of the peak
1800 General Tests, Processes and Apparatus Supplement I, JP XV

area of each component to the sum of the peak areas of graphy under the same conditions as for the preparation of
every peak recorded in the chromatogram. In order to the calibration curve, measure the peak area or peak height
obtain accurate results in evaluating the proportion of the of the objective compound, and read the amount of the
components, it is necessary to correct the area of each compound from the calibration curve.
component based on its relative response factor to the prin- In an individual monograph, generally one of the stan-
cipal component. dard solutions with a concentration within the linear range
of the calibration curve and a sample solution with a concen-
Assay
tration close to that of the standard solution are prepared,
In general, perform the assay by using the internal stan-
and the chromatography is performed with these solutions
dard method. The absolute calibration curve method is used
under a fixed condition to obtain the amount of the compo-
when a suitable internal standard is not available. Perform
nent. In this method, all procedures, such as the injection
the assay by using the standard addition method when the
procedure, must be carried out under a strictly constant
effect of the component other than the compound to be
condition.
assayed on the quantitative determination is not negligible
(3) Standard addition method—Pipet a fixed volume of
against a result of the determination.
more than 4 sample solutions, add exactly the standard
(1) Internal standard method—In the internal standard
solution so that stepwise increasing amounts of the object
method, choose a stable compound as an internal standard
compound are contained in the solutions except 1 sample
which shows a retention time close to that of the compound
solution, diluted exactly each solution with and without
to be assayed, and whose peak is well separated from all
standard solution to a definite volume, and use each solution
other peaks in the chromatogram. Prepare several kinds of
as the sample solution. Based on the chromatogram ob-
standard solutions containing a fixed amount of the internal
tained by exact injection of a fixed volume of individual
standard and several graded amounts of the authentic speci-
sample solutions, measure the peak area or peak height of
men specified in the individual monograph. Based on the
individual sample solutions. Calculate the concentration of
chromatogram obtained by injection of a fixed volume of
standard objective compound added into each sample solu-
individual standard solutions, calculate the ratio of peak
tion, plot the amounts (concentration) of added standard
area or peak height of the authentic specimen to that of the
object compound on the abscissa and the peak area or peak
internal standard, and prepare a calibration curve by plot-
height on the ordinate on the graph, extend the calibration
ting these ratios on the ordinate against the amount of the
curve obtained by linking the plots, and determine the
authentic specimen or the ratio of the amount of the authen-
amount of object compound to be assayed from the distance
tic specimen to that of the internal standard on the abscissa.
between the origin and the intersecting point of the calibra-
The calibration curve is usually obtained as a straight line
tion curve with the abscissa. This method is available only in
passing through the origin. Then, prepare a sample solution
the case that the calibration curve is a straight line, and pass-
containing the internal standard in the same amount as in
es through the origin when the absolute calibration curve
the standard solutions used for the preparation of the
method is employed. In this method, all procedures must be
calibration curve according to the method specified in the
carried out under a strictly constant condition.
individual monograph, perform the gas chromatography
under the same operating conditions as for the preparation Method for peak measuring
of the calibration curve, calculate the ratio of the peak area Generally, the following methods are used.
or peak height of the objective compound to that of the (1) Peak height measuring method
internal standard, and read the amount of the compound (i) Peak height method: Measure the distance between
from the calibration curve. the maximum of the peak and the intersecting point of a per-
In an individual monograph, generally one of the stan- pendicular line from the maximum of the peak to the
dard solutions with a concentration within the linear range horizontal axis of recording paper with a tangent linking the
of the calibration curve and a sample solution with a concen- baselines on either side of the peak.
tration close to that of the standard solution are prepared, (ii) Automatic peak height method: Measure the signals
and the chromatography is performed with these solutions from the detector as the peak height using a data processing
under fixed conditions to determine the amount of the system.
objective compound. (2) Peak area measuring method
(2) Absolute calibration curve method—Prepare stan- (i) Width at half-height method: Multiply the peak
dard solutions with several graded amounts of the authentic width at the half-height by the peak height.
specimen, and inject accurately a fixed volume of these (ii) Automatic integration method: Measure the signals
standard solutions. With the chromatogram obtained, pre- from the detector as the peak area using a data processing
pare a calibration curve by plotting the peak areas or peak system.
heights on the ordinate against the amount of the authentic
System suitability
specimen on the abscissa. The calibration curve is generally
Refer to ``System suitability'' described under 2.01 Liquid
obtained as a straight line passing through the origin. Then,
Chromatography.
prepare a sample solution according to the method specified
in the individual monograph, perform the gas chromato-
Supplement I, JP XV General Tests, Processes and Apparatus 1801

Point to consider in changing the operating conditions tion, and allow to stand for more than 24 hours before use.
Among the operating conditions specified in the individ- These Karl Fischer TSs, prepared by any one of the above
ual monograph, inside diameter and length of column, parti- methods, must be standardized before every use, because of
cle size of packing material, concentration or thickness of its activity change with the lapse of time. Further preserve
stationary phase, column temperature, temperature rising- the TS in a cold place, protecting it from light and moisture.
rate, kind and flow rate of carrier gas, and split ratio may be Standardization—According to the procedure described
modified within the ranges in which the gas chromatograph- below, take a suitable quantity of methanol for Karl Fischer
ic system used conforms to the requirements of system method in a dried titration flask, and titrate the solvent with
suitability. Headspace sample injection device and its oper- a Karl Fischer TS to make the inside of the flask anhydrous.
ating conditions may be also modified, provided that they Then, weigh about 30 mg of water accurately and put it in
give equivalent or more accuracy and precision. the titration flask quickly, and titrate the water dissolved in
the solvent with a Karl Fischer TS to the end point, under
Terminology
vigorous stirring. Calculate the water equivalence factor,
The definition of terms described under 2.01 Liquid Chro-
f (mg/mL), corresponding to the amount of water (H2O) in
matography shall apply in 2.02 Gas Chromatography.
mg per 1 mL of the Karl Fischer TS by using the following
Note Avoid the use of authentic specimens, internal stan- equation:
dards, reagents or solvents containing substances that may
Amount of water taken (H2O) (mg)
interfere with the determination. f (mg/mL)=
Volume of Karl Fischer TS consumed
for titration of water (H2O) (mL)

2.48 Water Determination


(Karl Fischer Method) Change to read:

Change to read the following part under 1. 2.49 Optical Rotation


Volumetric titration:
Determination
Preparation of test solutions and standard solutions
(1) Karl Fischer TS for water determination Optical Rotation Determination is a method for the
Prepare according to the following method (i), (ii) or (iii). measurement of the angular rotation of the sample using a
Additives may be added for the purpose of improving the polarimeter.
stability or other performances if it is confirmed that they Generally, the vibrations of light take place on planes
give almost the same results as those obtained from the spe- perpendicular to the direction of the beam. In case of the or-
cified method. dinary light, the directions of the planes are unrestricted,
(i) Preparation 1 while in case of the plane polarized light, commonly called
Dissolve 63 g of iodine in 100 mL of pyridine for Karl as polarized light, the vibrations take place on only one
Fischer method, cool the solution in ice bath, and pass dried plane that includes the advancing direction of the beam.
sulfur dioxide gas through this solution until the mass in- And it is called that these beams have plane of polarization.
crease of the solution reaches 32 g. Then make up to 500 mL Some drugs in the liquid state or in solution have a property
by adding chloroform for Karl Fischer method or methanol of rotating the plane of the polarized light either to the right
for Karl Fischer method, and allow to stand for more than or to the left. This property is referred to as optical activity
24 hours before use. or optical rotation, and is inherently related to the chemical
(ii) Preparation 2 constitution of the substance.
Dissolve 102 g of imidazole for Karl Fischer method in 350 The optical rotation is a degree of rotation of polarized
mL of diethylene glycol monoethyl ether for water determi- plane, caused by the optically active substance or its solu-
nation, cool the solution in ice bath, and pass dried sulfur di- tion, and it is measured by the polarimeter. This value is
oxide gas through this solution until the mass increase of the proportional to the length of the polarimeter tube, and is
solution reaches 64 g, keeping the temperature between 259 C also related to the solution concentration, the temperature
and 309 C. Then dissolve 50 g of iodine in this solution, and and the measurement wavelength. The character of the rota-
allow to stand for more than 24 hours before use. tion is indicated by the direction of the rotation, when facing
(iii) Preparation 3 to the advancing direction of the polarized light. Thus in
Pass dried sulfur dioxide gas through 220 mL of propy- case of rotation to the right, it is called dextrorotatory and
lene carbonate for water determination until the mass in- expressed by placing plus sign (+), while in case of rotation
crease of the solvent reaches 32 g. To this solution, add 180 to the left, it is called levorotatory and expressed by placing
mL of propylene carbonate, or diethylene glycol monoethyl minus sign (-) before the figure of the angular rotation. For
ether for water determination, in which 81 g of 2-methyl- example, +209means 209of rotation to the right, while
aminopyridine for Karl Fischer method is dissolved and -209means 209of rotation to the left.
cooled in ice bath. Then dissolve 36 g of iodine in this solu- The angular rotation atx means degree of rotation, when it
1802 General Tests, Processes and Apparatus Supplement I, JP XV

is measured at t9C by using specific monochromatic light x from the contents of a sufficient number of containers. If
(expressed by wavelength of light source or the specific beam test specimens are diluted with fluid medium, the test should
name). Usually, the measurement is performed at 209 C or be performed quickly. In performing the test, precautions
259C, with a polarimeter tube of 100 mm in length, and with must be taken to prevent biohazard.
the D line of sodium lamp.
I. Microbiological Examination of Non-sterile Products:
The specific rotation is expressed by the following
Microbial Enumeration Tests
equation:
These tests are harmonized with the European Phar-
100 a macopoeia and the U.S. Pharmacopeia.
[a]tx=
lc
1 Introduction
t: The temperature of measurement. The tests described hereafter will allow quantitative
x: The wavelength or the name of the specific monochro- enumeration of mesophilic bacteria and fungi which may
matic light (in the case of the Sodium D line, it is described grow under aerobic conditions.
as D). The tests are designed primarily to determine whether a
a: The angle, in degrees, of rotation of the plane of the substance or preparation complies with an established
polarized light. specification for microbiological quality. When used for
l: The thickness of the layer of sample solution, i.e., the such purposes follow the instructions given below, including
length of the polarimeter tube (mm). the number of samples to be taken and interpret the results
c: Drug concentration in g/mL. When an intact liquid as stated below.
drug is used for the direct measurement without dilution by The methods are not applicable to products containing
an appropriate solvent, c equals to its density (g/mL). viable micro-organisms as active ingredients.
However, unless otherwise specified, the specific gravity is Alternative microbiological procedures, including auto-
conventionally used in stead of the density. mated methods, may be used, provided that their equiva-
lence to the Pharmacopoeial method has been demonstrat-
The description in the monograph, for example, ``[a]20 D:
ed.
-33.0 – -36.09 (after drying, 1 g, water, 20 mL, 100
mm),'' means the measured specific rotation [a]20D should be 2 General Procedures
in the range of -33.09and -36.09, when 1 g of accurately Carry out the determination under conditions designed to
weighed sample dried under the conditions, specified in the avoid extrinsic microbial contamination of the product to be
test item of Loss on drying, is taken, and dissolved in water examined. The precautions taken to avoid contamination
to make exactly 20 mL, then put in the polarimeter tube of must be such that they do not affect any micro-organisms
100 mm length, of which temperature is kept at 209 C. which are to be revealed in the test.
If the product to be examined has antimicrobial activity,
this is insofar as possible removed or neutralized. If inactiva-
4.01 Bacterial Endotoxins Test tors are used for this purpose their efficacy and their absence
of toxicity for micro-organisms must be demonstrated.
If surface-active substances are used for sample prepara-
Change to read the Preparation of Standard
tion, their absence of toxicity for micro-organisms and their
Endotoxin Stock Solution as follows:
compatibility with inactivators used must be demonstrated.
Preparation of Standard Endotoxin Stock Solution
3 Enumeration Methods
Prepare Standard Endotoxin Stock Solution by dissolving
Use the membrane filtration method, or the plate-count
Endotoxin Reference Standard in water for bacterial
methods, as prescribed. The most probable number (MPN)
endotoxins test (BET). Endotoxin is expressed in Endotoxin
method is generally the least accurate method for microbial
Units (EU). One EU is equal to one International Unit (IU)
counts, however, for certain product groups with very low
of endotoxin.
bioburden, it may be the most appropriate method.
The choice of a method is based on factors such as the
nature of the product and the required limit of micro-organ-
4.05 Microbial Limit Test isms. The method chosen must allow testing of a sufficient
sample size to judge compliance with the specification. The
Change to read as follows: suitability of the chosen method must be established.

4 Growth Promotion Test and Suitability of the Counting


4.05 Microbiological Examination Method
of Non-sterile Products 4-1 General considerations
The ability of the test to detect micro-organisms in the
This chapter includes microbial enumeration tests and presence of product to be tested must be established.
tests for specified micro-organisms. For the test, use a mix- Suitability must be confirmed if a change in testing perfor-
ture of several portions selected at random from the bulk or mance, or the product, which may affect the outcome of the
Supplement I, JP XV General Tests, Processes and Apparatus 1803

Table 4.05-I-1 Preparation and use of test micro-organisms

Suitability of counting method in


Growth promotion
the presence of the product
Preparation of
Micro-organism
test strain
Total aerobic Total yeasts and Total aerobic Total yeasts and
microbial count moulds count microbial count moulds count

Staphylococcus Casein soya bean Casein soya bean Casein soya bean
aureus digest agar or digest agar and digest agar/
casein soya bean casein soya bean MPN casein soya
such as ATCC digest broth digest broth bean digest broth
6538, NCIMB 30 – 359C ≦100 CFU ≦100 CFU
9518, CIP 4.83 or 18 – 24 h 30 – 359C 30 – 359C
NBRC 13276 ≦3 days ≦3 days

Pseudomonas Casein soya bean Casein soya bean Casein soya bean
aeruginosa digest agar or digest agar and digest agar/MPN
casein soya bean casein soya bean casein soya bean
such as ATCC digest broth digest broth digest broth
9027, NCIMB 30 – 359C ≦100 CFU ≦100 CFU
8626, CIP 82.118 18 – 24 h 30 – 359C 30 – 359C
or NBRC 13275 ≦3 days ≦3 days

Bacillus subtilis Casein soya bean Casein soya bean Casein soya bean
digest agar or digest agar and digest agar/MPN
such as ATCC casein soya bean casein soya bean casein soya bean
6633, NCIMB digest broth digest broth digest broth
8054, CIP 52.62 or 30 – 359C ≦100 CFU ≦100 CFU
NBRC 3134 18 – 24 h 30 – 359C 30 – 359C
≦3 days ≦3 days

Candida albicans Sabouraud-dextrose Casein soya bean Sabouraud-dex- Casein soya bean Sabouraud-
agar or Sabouraud- digest agar trose agar digest agar dextrose agar
such as ATCC dextrose broth ≦100 CFU ≦100 CFU ≦100 CFU ≦100 CFU
10231, NCPF 20 – 259C 30 – 359C 20 – 259C 30 – 359C 20 – 259C
3179, IP 48.72 or 2 – 3 days ≦5 days ≦5 days ≦5 days ≦5 days
NBRC 1594 MPN: not applicable

Aspergillus niger Sabouraud-dextrose Casein soya bean Sabouraud-dex- Casein soya bean Sabouraud-
agar or potato-dex- digest agar trose agar digest agar dextrose agar
such as ATCC trose agar ≦100 CFU ≦100 CFU ≦100 CFU ≦100 CFU
16404, IMI 20 – 259C 30 – 359C 20 – 259C 30 – 359C 20 – 259C
149007, IP 5 – 7 days, or ≦5 days ≦5 days ≦5 days ≦5 days
1431.83 or NBRC until good sporula- MPN: not applicable
9455 tion is achieved

test is introduced. prepared and then an appropriate volume of the spore


4-2 Preparation of test strains suspension is used for test inoculation. The stable spore sus-
Use standardised stable suspensions of test strains or pension may be maintained at 2 – 89C for a validated period
prepare as stated below. of time.
Seed lot culture maintenance techniques (seed-lot systems) 4-3 Negative control
are used so that the viable micro-organisms used for inocula- To verify testing conditions a negative control is per-
tion are not more than 5 passages removed from the original formed using the chosen diluent in place of the test prepara-
master seed-lot. Grow each of the bacterial and fungal test tion. There must be no growth of micro-organisms.
strains separately as described in Table 4.05-I-1. 4-4 Growth promotion of the media
Use buffered sodium chloride-peptone solution pH 7.0 or Test each batch of ready-prepared medium and each batch
phosphate buffer solution pH 7.2 to make test suspensions; of medium, prepared either from dehydrated medium or
to suspend A. niger spores, 0.05 per cent of polysorbate 80 from the ingredients described.
may be added to the buffer. Use the suspensions within 2 h Inoculate portions/plates of casein soya bean digest broth
or within 24 h if stored at 2 – 89 C. As an alternative to and casein soya bean digest agar with a small number (not
preparing and then diluting a fresh suspension of vegetative more than 100 CFU) of the micro-organisms indicated in
cells of A. niger or B. subtilis, a stable spore suspension is Table 4.05-I-1, using a separate portion/plate of medium for
1804 General Tests, Processes and Apparatus Supplement I, JP XV

each. Inoculate plates of Sabouraud-dextrose agar with a the adhesive surface with sterile porous material, for exam-
small number (not more than 100 CFU) of the micro-organ- ple sterile gauze, to prevent the patches from sticking
isms indicated in Table 4.05-I-1, using a separate plate of together, and transfer the patches to a suitable volume of the
medium for each. Incubate in the conditions described in chosen diluent containing inactivators such as polysorbate
Table 4.05-I-1. 80 and/or lecithin. Shake the preparation vigorously for at
For solid media, growth obtained must not differ by a least 30 min.
factor greater than 2 from the calculated value for a stan- 4-5-2 Inoculation and dilution
dardized inoculum. For a freshly prepared inoculum, Add to the sample prepared as described above (4-5-1) and
growth of the micro-organisms comparable to that previous- to a control (with no test material included) a sufficient
ly obtained with a previously tested and approved batch of volume of the microbial suspension to obtain an inoculum
medium occurs. of not more than 100 CFU. The volume of the suspension of
Liquid media are suitable if clearly visible growth of the the inoculum should not exceed 1 per cent of the volume of
micro-organisms comparable to that previously obtained diluted product.
with a previously tested and approved batch of medium oc- To demonstrate acceptable microbial recovery from the
curs. product, the lowest possible dilution factor of the prepared
4-5 Suitability of the counting method in the presence of sample must be used for the test. Where this is not possible
product due to antimicrobial activity or poor solubility, further
4-5-1 Preparation of the sample appropriate protocols must be developed.
The method for sample preparation depends on the physi- If inhibition of growth by the sample cannot otherwise be
cal characteristics of the product to be tested. If none of the avoided, the aliquot of the microbial suspension may be ad-
procedures described below can be demonstrated to be satis- ded after neutralization, dilution or filtration.
factory, an alternative procedure must be developed. 4-5-3 Neutralization/removal of antimicrobial activity
Water-soluble products—Dissolve or dilute (usually a 1 in 10 The number of micro-organisms recovered from the
dilution is prepared) the product to be examined in buffered prepared sample diluted as described in 4-5-2 and incubated
sodium chloride-peptone solution pH 7.0, phosphate buffer following the procedure described in 4-5-4, is compared to
solution pH 7.2 or casein soya bean digest broth. If necessa- the number of micro-organisms recovered from the control
ry adjust to pH 6 – 8. Further dilutions, where necessary, are preparation.
prepared with the same diluent. If growth is inhibited (reduction by a factor greater than
Non-fatty products insoluble in water—Suspend the product 2), then modify the procedure for the particular enumera-
to be examined (usually a 1 in 10 dilution is prepared) in tion test to ensure the validity of the results. Modification of
buffered sodium chloride-peptone solution pH 7.0, phos- the procedure may include, for example, (1) an increase in
phate buffer solution pH 7.2 or casein soya bean digest the volume of the diluent or culture medium, (2) incorpora-
broth. A surface-active agent such as 1 g/L of polysorbate tion of a specific or general neutralizing agents into the dil-
80 may be added to assist the suspension of poorly wettable uent, (3) membrane filtration or (4) a combination of the
substances. If necessary adjust to pH 6 – 8. Further dilu- above measures.
tions, where necessary, are prepared with the same diluent. Neutralizing agents—Neutralizing agents may be used to
Fatty products—Dissolve in isopropyl myristate, sterilised neutralize the activity of antimicrobial agents (Table
by filtration or mix the product to be examined with the 4.05-I-2). They may be added to the chosen diluent or the
minimum necessary quantity of sterile polysorbate 80 or medium preferably before sterilization. If used, their effica-
another non-inhibitory sterile surface-active reagent, heated cy and their absence of toxicity for micro-organisms must be
if necessary to not more than 409 C, or in exceptional cases demonstrated by carrying out a blank with neutralizer and
to not more than 459 C. Mix carefully and if necessary main- without product.
tain the temperature in a water-bath. Add sufficient of the If no suitable neutralizing method can be found, it can be
pre-warmed chosen diluent to make a 1 in 10 dilution of the assumed that the failure to isolate the inoculated organism is
original product. Mix carefully whilst maintaining the attributable to the microbicidal activity of the product. This
temperature for the shortest time necessary for the forma- information serves to indicate that the article is not likely to
tion of an emulsion. Further serial tenfold dilutions may be be contaminated with the given species of the micro-organ-
prepared using the chosen diluent containing a suitable con- ism. However, it is possible that the product only inhibits
centration of sterile polysorbate 80 or another non-inhibito- some of the micro-organisms specified herein, but does not
ry sterile surface-active reagent. inhibit others not included amongst the test strains or for
Fluids or solids in aerosol form—Aseptically transfer the which the latter are not representative. Then, perform the
product into a membrane filter apparatus or a sterile con- test with the highest dilution factor compatible with microbi-
tainer for further sampling. Use either the total contents or a al growth and the specific acceptance criterion.
defined number of metered doses from each of the contain- 4-5-4 Recovery of micro-organism in the presence of
ers tested. product
Transdermal patches—Remove the protective cover sheets For each of the micro-organisms listed in Table 4.05-I-1,
(``release liner'') of the transdermal patches and place them, separate tests are performed. Only micro-organisms of the
adhesive side upwards, on sterile glass or plastic trays. Cover added test strain are counted.
Supplement I, JP XV General Tests, Processes and Apparatus 1805

Table 4.05-I-2 Common neutralizing agents/method for interfering substances

Interfering substance Potential neutralizing agents/method

Glutaraldehyde, Mercurials Sodium hydrogen sulfite (Sodium bisulfite)

Phenolics, Alcohol, Aldehydes, Sorbate Dilution

Aldehydes Glycine

Quaternary Ammonium Compounds (QACs), Lecithin


Parahydroxybenzoates (Parabens),
Bis-biguanides

QAC, Parabens, Iodine Polysorbate

Mercurials Thioglycollate

Mercurials, Halogens, Aldehydes Thiosulfate

EDTA (edetate) Mg or Ca ions

4-5-4-1 Membrane filtration Petri dishes are used, the volume of the agar is increased
Use membrane filters having a nominal pore size not accordingly. Dry the plates, for example in a laminar-air-
greater than 0.45 mm. The type of filter material is chosen in flow cabinet or in an incubator. For each of the micro-or-
such a way that the bacteria-retaining efficiency is not af- ganisms listed in Table 4.05-I-1, at least 2 Petri dishes are
fected by the components of the sample to be investigated. used. Spread a measured volume of not less than 0.1 mL of
For each of the micro-organisms listed in Table 4.05-I-1, one the sample prepared as described under 4-5-1 to 4-5-3 over
membrane filter is used. the surface of the medium. Incubate and count as prescribed
Transfer a suitable amount of the sample prepared as under 4-5-4-2-1.
described under 4-5-1 to 4-5-3 (preferably representing 1 g of 4-5-4-3 Most-probable-number (MPN) method
the product, or less if large numbers of CFU are expected) to The precision and accuracy of the MPN method is less
the membrane filter, filter immediately and rinse the mem- than that of the membrane filtration method or the plate-
brane filter with an appropriate volume of diluent. count method. Unreliable results are obtained particularly
For the determination of total aerobic microbial count for the enumeration of moulds. For these reasons the MPN
(TAMC), transfer the membrane filter to the surface of method is reserved for the enumeration of TAMC in situa-
casein soya bean digest agar. For the determination of total tions where no other method is available. If the use of the
combined yeasts/moulds count (TYMC) transfer the mem- method is justified, proceed as follows.
brane to the surface of Sabouraud-dextrose agar. Incubate Prepare a series of at least 3 serial tenfold dilutions of the
the plates as indicated in Table 4.05-I-1. Perform the count- product as described under 4-5-1 to 4-5-3. From each level of
ing. dilution, 3 aliquots of 1 g or 1 mL are used to inoculate 3
4-5-4-2 Plate-count methods tubes with 9 – 10 mL of casein soya bean digest broth. If
Perform plate-count methods at least in duplicate for each necessary a surface-active agent such as polysorbate 80, or
medium and use the mean count of the result. an inactivator of antimicrobial agents may be added to the
4-5-4-2-1 Pour-plate method medium. Thus, if 3 levels of dilution are prepared 9 tubes are
For Petri dishes 9 cm in diameter, add to the dish 1 mL of inoculated.
the sample prepared as described under 4-5-1 to 4-5-3 and 15 Incubate all tubes at 30 – 359 C for not more than 3 days.
– 20 mL of casein soya bean digest agar or Sabouraud-dex- If reading of the results is difficult or uncertain owing to the
trose agar, both media being at not more than 459C. If larg- nature of the product to be examined, subculture in the same
er Petri dishes are used, the amount of agar medium is in- broth, or casein soya bean digest agar, for 1 – 2 days at the
creased accordingly. For each of the micro-organisms listed same temperature and use these results. Determine the most
in Table 4.05-I-1, at least 2 Petri dishes are used. probable number of micro-organisms per gram or millilitre
Incubate the plates as indicated in Table 4.05-I-1. Take of the product to be examined from Table 4.05-I-3.
the arithmetic mean of the counts per medium and calculate 4-6 Results and interpretation
the number of CFU in the original inoculum. When verifying the suitability of the membrane filtration
4-5-4-2-2 Surface-spread method method or the plate-count method, a mean count of any of
For Petri dishes 9 cm in diameter, add 15 – 20 mL of the test organisms not differing by a factor greater than 2
casein soya bean digest agar or Sabouraud-dextrose agar at from the value of the control defined in 4-5-2 in the absence
about 459 C to each Petri dish and allow to solidify. If larger of the product must be obtained. When verifying the
1806 General Tests, Processes and Apparatus Supplement I, JP XV

suitability of the MPN method the calculated value from the 5-2-2 Plate-count methods
inoculum must be within 95 per cent confidence limits of the 5-2-2-1 Pour-plate method
results obtained with the control. Prepare the sample using a method that has been shown to
If the above criteria cannot be met for one or more of the be suitable as described in section 4. Prepare for each medi-
organisms tested with any of the described methods, the um at least 2 Petri dishes for each level of dilution. Incubate
method and test conditions that come closest to the criteria the plates of casein soya bean digest agar at 30 – 359 C for
are used to test the product. 3 – 5 days and the plates of Sabouraud-dextrose agar at 20 –
259C for 5 – 7 days. Select the plates corresponding to a
5 Testing of Products
given dilution and showing the highest number of colonies
5-1 Amount used for the test
less than 250 for TAMC and 50 for TYMC. Take the
Unless otherwise prescribed, use 10 g or 10 mL of the
arithmetic mean per culture medium of the counts and calcu-
product to be examined taken with the precautions referred
late the number of CFU per gram or per millilitre of
to above. For fluids or solids in aerosol form, sample 10
product.
containers. For transdermal patches, sample 10 patches.
5-2-2-2 Surface-spread method
The amount to be tested may be reduced for active sub-
Prepare the sample using a method that has been shown to
stances that will be formulated in the following conditions:
be suitable as described in section 4. Prepare at least 2 Petri
the amount per dosage unit (e.g. tablet, capsule, injection) is
dishes for each medium and each level of dilution. For incu-
less than or equal to 1 mg or the amount per gram or mil-
bation and calculation of the number of CFU proceed as
lilitre (for preparations not presented in dose units) is less
described for the pour-plate method.
than 1 mg. In these cases, the amount of sample to be tested
5-2-2-3 Most-probable-number method
is not less than the amount present in 10 dosage units or 10 g
Prepare and dilute the sample using a method that has
or 10 mL of the product.
been shown to be suitable as described in section 4. Incubate
For materials used as active substances where sample
all tubes for 3 – 5 days at 30 – 359C. Subculture if necessary,
quantity is limited or batch size is extremely small (i.e. less
using the procedure shown to be suitable. Record for each
than 1000 mL or 1000 g), the amount tested shall be 1 per
level of dilution the number of tubes showing microbial
cent of the batch unless a lesser amount is prescribed or
growth. Determine the most probable number of micro-or-
justified and authorised.
ganisms per gram or millilitre of the product to be examined
For products where the total number of entities in a batch
from Table 4.05-I-3.
is less than 200 (e.g. samples used in clinical trials), the sam-
5-3 Interpretation of the results
ple size may be reduced to 2 units, or 1 unit if the size is less
The total aerobic microbial count (TAMC) is considered
than 100.
to be equal to the number of CFU found using casein soya
Select the sample(s) at random from the bulk material or
bean digest agar; if colonies of fungi are detected on this
from the available containers of the preparation. To obtain
medium, they are counted as part of TAMC. The total com-
the required quantity, mix the contents of a sufficient
bined yeasts/mould count (TYMC) is considered to be equal
number of containers to provide the sample.
to the number of CFU found using Sabouraud-dextrose
5-2 Examination of the product
agar; if colonies of bacteria are detected on this medium,
5-2-1 Membrane filtration
they are counted as part of TYMC. When the TYMC is
Use a filtration apparatus designed to allow the transfer of
expected to exceed the acceptance criterion due to the bac-
the filter to the medium. Prepare the sample using a method
terial growth, Sabouraud-dextrose agar containing antibiot-
that has been shown suitable as described in section 4 and
ics may be used. If the count is carried out by the MPN
transfer the appropriate amount to each of 2 membrane
method the calculated value is the TAMC.
filters and filter immediately. Wash each filter following the
When an acceptance criterion for microbiological quality
procedure shown to be suitable.
is prescribed it is interpreted as follows:
For the determination of TAMC, transfer one of the
-101 CFU: maximum acceptable count=20,
membrane filters to the surface of casein soya bean digest
-102 CFU: maximum acceptable count=200,
agar. For the determination of TYMC, transfer the other
-103 CFU: maximum acceptable count=2000, and so
membrane to the surface of Sabouraud-dextrose agar. Incu-
forth.
bate the plate of casein soya bean digest agar at 30 – 359C
The recommended solutions and media are described in
for 3 – 5 days and the plate of Sabouraud-dextrose agar at
Tests for specified micro-organisms.
20 – 259 C for 5 – 7 days. Calculate the number of CFU per
gram or per millilitre of product.
When examining transdermal patches, filter 10 per cent of II. Microbiological Examination of Non-sterile Products:
the volume of the preparation described under 4-5-1 Tests for Specified Micro-organisms
separately through each of 2 sterile filter membranes. Trans-
These tests are harmonized with the European Phar-
fer one membrane to casein soya bean digest agar for TAMC
macopoeia and the U.S. Pharmacopeia.
and the other membrane to Sabouraud-dextrose agar for
TYMC. 1 Introduction
The tests described hereafter will allow determination of
Supplement I, JP XV General Tests, Processes and Apparatus 1807

Table 4.05-I-3 Most-probable-number values of micro-organisms

Observed combinations of numbers of tubes


showing growth in each set
MPN per g or 95 per cent
Number of g or mL of product per tube per mL of product confidence limits
0.1 0.01 0.001
0 0 0 Less than 3 0 – 9.4
0 0 1 3 0.1 – 9.5
0 1 0 3 0.1 – 10
0 1 1 6.1 1.2 – 17
0 2 0 6.2 1.2 – 17
0 3 0 9.4 3.5 – 35
1 0 0 3.6 0.2 – 17
1 0 1 7.2 1.2 – 17
1 0 2 11 4 – 35
1 1 0 7.4 1.3 – 20
1 1 1 11 4 – 35
1 2 0 11 4 – 35
1 2 1 15 5 – 38
1 3 0 16 5 – 38
2 0 0 9.2 1.5 – 35
2 0 1 14 4 – 35
2 0 2 20 5 – 38
2 1 0 15 4 – 38
2 1 1 20 5 – 38
2 1 2 27 9 – 94
2 2 0 21 5 – 40
2 2 1 28 9 – 94
2 2 2 35 9 – 94
2 3 0 29 9 – 94
2 3 1 36 9 – 94
3 0 0 23 5 – 94
3 0 1 38 9 – 104
3 0 2 64 16 – 181
3 1 0 43 9 – 181
3 1 1 75 17 – 199
3 1 2 120 30 – 360
3 1 3 160 30 – 380
3 2 0 93 18 – 360
3 2 1 150 30 – 380
3 2 2 210 30 – 400
3 2 3 290 90 – 990
3 3 0 240 40 – 990
3 3 1 460 90 – 1980
3 3 2 1100 200 – 4000
3 3 3 More than 1100

the absence of, or limited occurrence of specified micro- substance or preparation complies with an established
organisms which may be detected under the conditions specification for microbiological quality. When used for
described. such purposes follow the instructions given below, including
The tests are designed primarily to determine whether a the number of samples to be taken and interpret the results
1808 General Tests, Processes and Apparatus Supplement I, JP XV

as stated below. and then diluting down a fresh suspension of vegetative cells
Alternative microbiological procedures, including auto- of Cl. sporogenes, a stable spore suspension is used for test
mated methods may be used, provided that their equivalence inoculation. The stable spore suspension may be maintained
to the Pharmacopoeial method has been demonstrated. at 2 – 89 C for a validated period.
3-2 Negative control
2 General Procedures
To verify testing conditions a negative control is per-
The preparation of samples is carried out as described in
formed using the chosen diluent in place of the test prepara-
Microbial enumeration tests.
tion. There must be no growth of micro-organisms.
If the product to be examined has antimicrobial activity,
3-3 Growth promotion and inhibitory properties of the
this is insofar as possible removed or neutralized as
media
described in Microbial enumeration tests.
Test each batch of ready-prepared medium and each batch
If surface-active substances are used for sample prepara-
of medium prepared either from dehydrated medium or
tion, their absence of toxicity for micro-organisms and their
from ingredients.
compatibility with inactivators used must be demonstrated
Verify suitable properties of relevant media as described
as described in Microbial enumeration tests.
in Table 4.05-II-1.
3 Growth Promoting and Inhibitory Properties of the Test for growth promoting properties, liquid media: in-
Media and Suitability of the Test oculate a portion of the appropriate medium with a small
The ability of the test to detect micro-organisms in the number (not more than 100 CFU) of the appropriate micro-
presence of the product to be tested must be established. organism. Incubate at the specified temperature for not
Suitability must be confirmed if a change in testing perfor- more than the shortest period of time specified in the test.
mance, or the product, which may affect the outcome of the Clearly visible growth of the micro-organism comparable to
test is introduced. that previously obtained with a previously tested and ap-
3-1 Preparation of test strains proved batch of medium occurs.
Use standardised stable suspensions of test strains or pre- Test for growth promoting properties, solid media: per-
pare as stated below. Seed lot culture maintenance tech- form surface-spread method, inoculating each plate with a
niques (seed-lot systems) are used so that the viable micro- small number (not more than 100 CFU) of the appropriate
organisms used for inoculation are not more than 5 passages micro-organism. Incubate at the specified temperature for
removed from the original master seed-lot. not more than the shortest period of time specified in the
3-1-1 Aerobic micro-organisms test. Growth of the micro-organism comparable to that
Grow each of the bacterial test strains separately in con- previously obtained with a previously tested and approved
tainers containing casein soya bean digest broth or on casein batch of medium occurs.
soya bean digest agar at 30 – 359C for 18 – 24 hours. Grow Test for inhibitory properties, liquid or solid media: in-
the test strain for Candida albicans separately on oculate the appropriate medium with at least 100 CFU of the
Sabouraud-dextrose agar or in Sabouraud-dextrose broth at appropriate micro-organism. Incubate at the specified tem-
20 – 259 C for 2–3 days. perature for not less than the longest period of time specified
Staphylococcus aureus such as ATCC 6538, NCIMB in the test. No growth of the test micro-organism occurs.
9518, CIP 4.83 or NBRC 13276, Test for indicative properties: perform surface-spread
Pseudomonas aeruginosa such as ATCC 9027, NCIMB method, inoculating each plate with a small number (not
8626, CIP 82.118 or NBRC 13275, more than 100 CFU) of the appropriate micro-organism.
Escherichia coli such as ATCC 8739, NCIMB 8545, CIP Incubate at the specified temperature for a period of time
53.126 or NBRC 3972, within the range specified in the test. Colonies are compara-
Salmonella enterica subsp. enterica serovar Typhimurium ble in appearance and indication reactions to those previous-
such as ATCC 14028 ly obtained with a previously tested and approved batch of
or, as an alternative, medium.
Salmonella enterica subsp. enterica serovar Abony such as 3-4 Suitability of the test method
NBRC 100797, NCTC 6017 or CIP 80.39, For each product to be tested perform sample preparation
Candida albicans such as ATCC 10231, NCPF 3179, IP as described in the relevant paragraph in section 4. Add each
48.72 or NBRC 1594. test strain at the time of mixing, in the prescribed growth
Use buffered sodium chloride-peptone solution pH 7.0 or medium. Inoculate the test strains individually. Use a num-
phosphate buffer solution pH 7.2 to make test suspensions. ber of micro-organisms equivalent to not more than 100
Use the suspensions within 2 hours or within 24 hours if CFU in the inoculated test preparation.
stored at 2 – 89 C. Perform the test as described in the relevant paragraph in
3-1-2 Clostridia section 4 using the shortest incubation period prescribed.
Use Clostridium sporogenes such as ATCC 11437 (NBRC The specified micro-organisms must be detected with the
14293, NCIMB 12343, CIP 100651) or ATCC 19404 (NCTC indication reactions as described in section 4.
532 or CIP 79.3). Grow the clostridial test strain under Any antimicrobial activity of the product necessitates a
anaerobic conditions in reinforced medium for Clostridia at modification of the test procedure (see 4-5-3 of Microbial
30 – 359 C for 24 – 48 hours. As an alternative to preparing Enumeration Tests).
Supplement I, JP XV General Tests, Processes and Apparatus 1809

Table 4.05-II-1 Growth promoting, inhibitory and indicative properties of media

Medium Property Test strains

Test for bile-tolerant gram-negative bacteria

Enterobacteria enrichment Growth promoting E. coli


broth-Mossel P. aeruginosa
Inhibitory S. aureus

Violet red bile glucose agar Growth promoting+ E. coli


Indicative P. aeruginosa

Test for Escherichia coli

MacConkey broth Growth promoting E. coli


Inhibitory S. aureus

MacConkey agar Growth promoting+ E. coli


Indicative

Test for Salmonella

Rappaport Vassiliadis Salmonella Growth promoting Salmonella enterica subsp. enterica serovar
enrichment broth Typhimurium or
Salmonella enterica subsp. enterica serovar
Abony

Inhibitory S. aureus

Xylose, lysine, deoxycholate agar Growth promoting+ Salmonella enterica subsp. enterica serovar
Indicative Typhimurium or
Salmonella enterica subsp. enterica serovar
Abony

Indicative E. coli
Test for Pseudomonas aeruginosa

Cetrimide agar Growth promoting P. aeruginosa


Inhibitory E. coli

Test for Staphylococcus aureus

Mannitol salt agar Growth promoting+ S. aureus


Indicative

Inhibitory E. coli
Test for Clostridia

Reinforced medium for Clostridia Growth promoting Cl. sporogenes

Columbia agar Growth promoting Cl. sporogenes


Test for Candida albicans

Sabouraud dextrose broth Growth promoting C. albicans


Sabouraud dextrose agar Growth promoting+ C. albicans
Indicative

If for a given product the antimicrobial activity with 4 Testing of Products


respect to a micro-organism for which testing is prescribed 4-1 Bile-tolerant gram-negative bacteria
cannot be neutralised, then it is to be assumed that the in- 4-1-1 Sample preparation and pre-incubation
hibited micro-organism will not be present in the product. Prepare a sample using a 1 in 10 dilution of not less than 1
1810 General Tests, Processes and Apparatus Supplement I, JP XV

Table 4.05-II-2 Interpretation of results

Results for each quantity of product


Probable number of bacteria
per gram or mL of product
0.1 g or 0.1 mL 0.01 g or 0.01 mL 0.001 g or 0.001 mL

+ + + more than 103

+ + - less than 103 and more than 102

+ - - less than 102 and more than 10

- - - less than 10

g of the product to be examined as described in Microbial 4-2-3 Interpretation


enumeration tests, but using casein soya bean digest broth as Growth of colonies indicates the possible presence of
the chosen diluent, mix and incubate at 20 – 259 C for a time E. coli. This is confirmed by identification tests.
sufficient to resuscitate the bacteria but not sufficient to en- The product complies with the test if no colonies are
courage multiplication of the organisms (usually 2 hours but present or if the identification tests are negative.
not more than 5 hours). 4-3 Salmonella
4-1-2 Test for absence 4-3-1 Sample preparation and pre-incubation
Unless otherwise prescribed use the volume corresponding Prepare the product to be examined as described in
to 1 g of the product, as prepared in 4-1-1 to inoculate Microbial enumeration tests and use the quantity cor-
enterobacteria enrichment broth-Mossel. Incubate at 30 – responding to not less than 10 g or 10 mL to inoculate a suit-
359C for 24 – 48 hours. Subculture on plates of violet red able amount (determined as described under 3-4) of casein
bile glucose agar. Incubate at 30 – 359C for 18 – 24 hours. soya bean digest broth, mix and incubate at 30 – 359C for
The product complies with the test if there is no growth of 18 – 24 hours.
colonies. 4-3-2 Selection and subculture
4-1-3 Quantitative test Transfer 0.1 mL of casein soya bean digest broth to 10 mL
4-1-3-1 Selection and subculture of Rappaport Vassiliadis Salmonella enrichment broth and
Inoculate suitable quantities of enterobacteria enrichment incubate at 30 – 359 C for 18 – 24 hours. Subculture on plates
broth-Mossel with the preparation as described under 4-1-1 of xylose, lysine, deoxycholate agar. Incubate at 30 – 359 C
and/or dilutions of it containing respectively 0.1 g, 0.01 g for 18 – 48 hours.
and 0.001 g (or 0.1 mL, 0.01 mL and 0.001 mL) of the 4-3-3 Interpretation
product to be examined. Incubate at 30 – 359C for 24 – 48 The possible presence of Salmonella is indicated by the
hours. Subculture each of the cultures on a plate of violet growth of well-developed, red colonies, with or without
red bile glucose agar. Incubate at 30 – 359C for 18 – 24 black centres. This is confirmed by identification tests.
hours. The product complies with the test if colonies of the types
4-1-3-2 Interpretation described are not present or if the confirmatory identifica-
Growth of colonies constitutes a positive result. Note the tion tests are negative.
smallest quantity of the product that gives a positive result 4-4 Pseudomonas aeruginosa
and the largest quantity that gives a negative result. Deter- 4-4-1 Sample preparation and pre-incubation
mine from Table 4.05-II-2 the probable number of bacteria. Prepare a sample using a 1 in 10 dilution of not less than
4-2 Escherichia coli 1 g of the product to be examined as described in Microbial
4-2-1 Sample preparation and pre-incubation enumeration tests and use 10 mL or the quantity correspond-
Prepare a sample using a 1 in 10 dilution of not less than ing to 1 g or 1 mL to inoculate a suitable amount (deter-
1 g of the product to be examined as described in Microbial mined as described under 3-4) of casein soya bean digest
enumeration tests and use 10 mL or the quantity correspond- broth and mix. When testing transdermal patches, filter the
ing to 1 g or 1 mL to inoculate a suitable amount (deter- volume of sample corresponding to 1 patch of the prepara-
mined as described under 3-4) of casein soya bean digest tion described in Microbial enumeration tests (4-5-1)
broth, mix and incubate at 30 – 359C for 18 – 24 hours. through a sterile filter membrane and place in 100 mL of
4-2-2 Selection and subculture casein soya bean digest broth. Incubate at 30 – 359C for 18 –
Shake the container, transfer 1 mL of casein soya bean 24 hours.
digest broth to 100 mL of MacConkey broth and incubate at 4-4-2 Selection and subculture
42 – 449 C for 24 – 48 hours. Subculture on a plate of Mac- Subculture on a plate of cetrimide agar and incubate at
Conkey agar at 30 – 359C for 18 – 72 hours. 30 – 359 C for 18 – 72 hours.
Supplement I, JP XV General Tests, Processes and Apparatus 1811

4-4-3 Interpretation mL of Sabouraud-dextrose broth and mix. Incubate at 30 –


Growth of colonies indicates the possible presence of 359C for 3-5 days.
P. aeruginosa. This is confirmed by identification tests. 4-7-2 Selection and subculture
The product complies with the test if colonies are not Subculture on a plate of Sabouraud-dextrose agar and
present or if the confirmatory identification tests are nega- incubate at 30 – 359 C for 24 – 48 hours.
tive. 4-7-3 Interpretation
4-5 Staphylococcus aureus Growth of white colonies may indicate the presence of C.
4-5-1 Sample preparation and pre-incubation albicans. This is confirmed by identification tests.
Prepare a sample using a 1 in 10 dilution of not less than 1 The product complies with the test if such colonies are not
g of the product to be examined as described in Microbial present or if the confirmatory identification tests are nega-
enumeration tests and use 10 mL or the quantity correspond- tive.
ing to 1 g or 1 mL to inoculate a suitable amount (deter- The following section is given for information.
mined as described under 3-4) of casein soya bean digest
5 Recommended Solutions and Culture Media
broth and homogenise. When testing transdermal patches,
The following solutions and culture media have been
filter the volume of sample corresponding to 1 patch of the
found satisfactory for the purposes for which they are
preparation described in Microbial enumeration tests (4-5-1)
prescribed in the test for microbial contamination in the
through a sterile filter membrane and place in 100 mL of
Pharmacopoeia. Other media may be used if they have simi-
casein soya bean digest broth. Incubate at 30 – 359C for 18 –
lar growth promoting and inhibitory properties.
24 hours.
4-5-2 Selection and subculture Stock buffer solution. Transfer 34 g of potassium dihydro-
Subculture on a plate of mannitol salt agar and incubate gen phosphate to a 1000 mL volumetric flask, dissolve in 500
at 30 – 359 C for 18 – 72 hours. mL of purified water, adjust to pH 7.2±0.2 with sodium
4-5-3 Interpretation hydroxide, add purified water to volume and mix. Dispense
The possible presence of S. aureus is indicated by the in containers and sterilize. Store at a temperature of 2 – 89
C.
growth of yellow/white colonies surrounded by a yellow
Phosphate buffer solution pH 7.2
zone. This is confirmed by identification tests.
Prepare a mixture of purified water and stock buffer solu-
The product complies with the test if colonies of the types
tion (800:1 V/V) and sterilize.
described are not present or if the confirmatory identifica-
tion tests are negative. Buffered sodium chloride-peptone solution pH 7.0
4-6 Clostridia Potassium dihydrogen phosphate 3.6 g
4-6-1 Sample preparation and heat treatment Disodium hydrogen phosphate dihydrate 7.2 g
Prepare the product to be examined as described in equivalent to
Microbial enumeration tests. 0.067 mol phosphate
Take 2 equal portions corresponding to not less than 1 g Sodium chloride 4.3 g
or 1 mL of the product to be examined. Heat 1 portion at Peptone (meat or casein) 1.0 g
809C for 10 min and cool rapidly. Do not heat the other por- Purified water 1000 mL
tion. Sterilize in an autoclave using a validated cycle.
4-6-2 Selection and subculture
Transfer the quantity corresponding to 1 g or 1 mL of the Casein soya bean digest broth
product to be examined from each of the mixed portions to 2 Pancreatic digest of casein 17.0 g
containers (38 mm×200 mm) or other containers containing Papaic digest of soya bean 3.0 g
100 mL of reinforced medium for Clostridia. Incubate Sodium chloride 5.0 g
under anaerobic conditions at 30 – 359C for 48 hours. After Dipotassium hydrogen phosphate 2.5 g
incubation, make subcultures from each tube on Columbia Glucose monohydrate 2.5 g
agar and incubate under anaerobic conditions at 30 – 359C Purified water 1000 mL
for 48 hours. Adjust the pH so that after sterilization it is 7.3±0.2 at
4-6-3 Interpretation 259C. Sterilize in an autoclave using a validated cycle.
The occurrence of anaerobic growth of rods (with or
without endospores) giving a negative catalase reaction indi- Casein soya bean digest agar
cates the presence of Clostridia. Pancreatic digest of casein 15.0 g
If no anaerobic growth of micro-organisms is detected on Papaic digest of soya bean 5.0 g
Columbia agar or the catalase test is positive, the product Sodium chloride 5.0 g
complies with the test. Agar 15.0 g
4-7 Candida albicans Purified water 1000 mL
4-7-1 Sample preparation and pre-incubation Adjust the pH so that after sterilization it is 7.3±0.2 at
Prepare the product to be examined as described in 259C. Sterilize in an autoclave using a validated cycle.
Microbial enumeration tests and use 10 mL or the quantity
corresponding to not less than 1 g or 1 mL to inoculate 100
1812 General Tests, Processes and Apparatus Supplement I, JP XV

Sabouraud-dextrose agar MacConkey agar


Glucose 40.0 g Pancreatic digest of gelatin 17.0 g
Mixture of peptic digest of animal tissue and pan- Peptones (meat and casein) 3.0 g
creatic digest of casein (1:1) 10.0 g Lactose monohydrate 10.0 g
Agar 15.0 g Sodium chloride 5.0 g
Purified water 1000 mL Bile salts 1.5 g
Adjust the pH so that after sterilization it is 5.6±0.2 at Agar 13.5 g
259C. Sterilize in an autoclave using a validated cycle. Neutral red 30 mg
Crystal violet 1 mg
Potato dextrose agar Purified water 1000 mL
Infusion from potatoes 200 g Adjust the pH so that after sterilization it is 7.1±0.2 at
Glucose 20.0 g 259C. Boil for 1 min with constant shaking then sterilize in
Agar 15.0 g an autoclave using a validated cycle.
Purified water 1000 mL
Adjust the pH so that after sterilization it is 5.6±0.2 at Rappaport Vassiliadis Salmonella enrichment broth
259C. Sterilize in an autoclave using a validated cycle. Soya peptone 4.5 g
Magnesium chloride hexahydrate 29.0 g
Sabouraud-dextrose broth Sodium chloride 8.0 g
Glucose 20.0 g Dipotassium hydrogen phosphate 0.4 g
Mixture of peptic digest of animal tissue and pan- Potassium dihydrogen phosphate 0.6 g
creatic digest of casein (1:1) 10.0 g Malachite green 36 mg
Purified water 1000 mL Purified water 1000 mL
Adjust the pH so that after sterilization it is 5.6±0.2 at Dissolve, warming slightly. Sterilize in an autoclave using
259C. Sterilize in an autoclave using a validated cycle. a validated cycle, at a temperature not exceeding 1159 C. The
pH is to be 5.2±0.2 at 259 C after heating and autoclaving.
Enterobacteria enrichment broth-Mossel
Pancreatic digest of gelatin 10.0 g Xylose, lysine, deoxycholate agar
Glucose monohydrate 5.0 g Xylose 3.5 g
Dehydrated ox bile 20.0 g L-Lysine 5.0 g
Potassium dihydrogen phosphate 2.0 g Lactose monohydrate 7.5 g
Disodium hydrogen phosphate dihydrate 8.0 g Sucrose 7.5 g
Brilliant green 15 mg Sodium chloride 5.0 g
Purified water 1000 mL Yeast extract 3.0 g
Adjust the pH so that after heating it is 7.2±0.2 at 259C. Phenol red 80 mg
Heat at 1009 C for 30 min and cool immediately. Agar 13.5 g
Sodium deoxycholate 2.5 g
Violet red bile glucose agar Sodium thiosulfate 6.8 g
Yeast extract 3.0 g Ammonium iron (III) citrate 0.8 g
Pancreatic digest of gelatin 7.0 g Purified water 1000 mL
Bile salts 1.5 g Adjust the pH so that after heating it is 7.4±0.2 at 259C.
Sodium chloride 5.0 g Heat to boiling, cool to 509
C and pour into Petri dishes. Do
Glucose monohydrate 10.0 g not heat in an autoclave.
Agar 15.0 g
Neutral red 30 mg Cetrimide agar
Crystal violet 2 mg Pancreatic digest of gelatin 20.0 g
Purified water 1000 mL Magnesium chloride 1.4 g
Adjust the pH so that after heating it is 7.4±0.2 at 259C. Dipotassium sulfate 10.0 g
Heat to boiling; do not heat in an autoclave. Cetrimide 0.3 g
Agar 13.6 g
MacConkey broth Purified water 1000 mL
Pancreatic digest of gelatin 20.0 g Glycerol 10.0 mL
Lactose monohydrate 10.0 g Heat to boiling for 1 min with shaking. Adjust the pH so
Dehydrated ox bile 5.0 g that after sterilization it is 7.2±0.2 at 259C. Sterilize in an
Bromocresol purple 10 mg autoclave using a validated cycle.
Purified water 1000 mL
Adjust the pH so that after sterilization it is 7.3±0.2 at Mannitol salt agar
259C. Sterilize in an autoclave using a validated cycle. Pancreatic digest of casein 5.0 g
Peptic digest of animal tissue 5.0 g
Supplement I, JP XV General Tests, Processes and Apparatus 1813

Beef extract 1.0 g


D-Mannitol 10.0 g Add the following next to Procedure:
Sodium chloride 75.0 g
Evaluation
Agar 15.0 g
The preparation complies with the test if the total number
Phenol red 25 mg
of metal particles of a size equal to or greater than 50 mm
Purified water 1000 mL
found in 10 units tested, is not more than 50, and also the
Heat to boiling for 1 min with shaking. Adjust the pH so
number of dishes containing more than 8 particles is not
that after sterilization it is 7.4±0.2 at 259C. Sterilize in an
more than 1. If this requirement is snot met, repeat the test
autoclave using a validated cycle.
with a further 20 units in the same manner, and if the total
number of the particles found in the total of 30 units is not
Reinforced medium for Clostridia
more than 150, and also the number of dishes containing
Beef extract 10.0 g
more than 8 particles is not more than 3, the preparation
Peptone 10.0 g
complies with the test.
Yeast extract 3.0 g
Soluble starch 1.0 g
Glucose monohydrate 5.0 g
Cysteine hydrochloride 0.5 g 6.08 Insoluble Particulate Matter
Sodium chloride 5.0 g Test for Ophthalmic Solutions
Sodium acetate 3.0 g
Agar 0.5 g Add the following next to Procedure:
Purified water 1000 mL
Evaluation
Hydrate the agar, dissolve by heating to boiling with con-
The preparation complies with the test if the calculated
tinuous stirring. If necessary, adjust the pH so that after
number per mL of insoluble particles of a size equal to or
sterilization it is about 6.8±0.2 at 259 C. Sterilize in an
greater than 300 mm is not more than 1.
autoclave using a validated cycle.

Columbia agar
Pancreatic digest of casein 10.0 g
6.10 Dissolution Test
Meat peptic digest 5.0 g
Change the Apparatus for Paddle Method
Heart pancreatic digest 3.0 g
(Apparatus 2) to read:
Yeast extract 5.0 g
Corn starch 1.0 g Apparatus for Paddle Method (Apparatus 2)—Use the
Sodium chloride 5.0 g assembly from Apparatus 1, except that a paddle formed
Agar, according to gelling power 10.0 g to 15.0 g from a blade and a shaft is used as the stirring element. The
Purified water 1000 mL shaft is positioned so that its axis is not more than 2 mm
Hydrate the agar, dissolve by heating to boiling with con- from the vertical axis of the vessel, at any point, and rotates
tinuous stirring. If necessary, adjust the pH so that after smoothly without significant wobble that could affect the
sterilization it is 7.3±0.2 at 259C. Sterilize in an autoclave results. The vertical center line of the blade passes through
using a validated cycle. Allow to cool to 45 – 509 C; add, the axis of the shaft so that the bottom of the blade is flush
where necessary, gentamicin sulfate corresponding to 20 mg with the bottom of the shaft. The paddle conforms to the
of gentamicin base and pour into Petri dishes. specifications shown in Fig. 6.10-2. The distance of 25±2
mm between the bottom of the blade and the inside bottom
of the vessel is maintained during the test. The metallic or
6.01 Test for Metal Particles in suitably inert, rigid blade and shaft comprise a single entity.
A suitable two-part detachable design may be used provided
Ophthalmic Ointments the assembly remains firmly engaged during the test. The
paddle blade and shaft may be coated with a suitable coating
Change the Preparation of test sample to read:
so as to make them inert. The dosage unit is allowed to sink
Preparation of test sample to the bottom of the vessel before rotation of the blade is
The test should be carried out in a clean place. Take 10 started. A small, loose piece of nonreactive material, such as
ophthalmic ointments to be tested, and extrude 5 g each of not more than a few turns of wire helix or such one shown in
their contents into separate flat-bottomed petri dishes 60 Fig. 6.10-2a, may be attached to the dosage unit that would
mm in diameter. Cover the dishes, and heat between 859 C otherwise float. Other validated sinker devices may also be
and 1109 C for 2 hours to dissolve bases completely. Allow used. ◆If the use of sinker is specified, unless otherwise
the samples to cool to room temperature without agitation specified, use the sinker device shown in Fig. 6.10-2a.◆
to solidify the contents. When the amount of the content is 5
g or less, extrude the contents as completely as practicable,
and proceed in the same manner as described above.
1814 General Tests, Processes and Apparatus Supplement I, JP XV

9.01 Reference Standards


Change to read:
Reference Standards are the reference substances pre-
pared to a specified quality necessary with regard to their in-
tended use as prescribed in monographs of the Phar-
macopoeia.
The Japanese Pharmacopoeia Reference Standards are as
follows:

* A: Assay
AF: Anti-factor IIa activity
B: Bacterial Endotoxins Test <4.01>
C: Content ratio of active principle
D: Dissolution
DG: Digestion Test <4.03>
HB: Heparin-binding capacity
I: Identification
IS: Isomer ratio
M: Melting Point Determination <2.60>
P: Purity
Fig. 6. 10–2 Apparatus 2, Paddle stirring element T: Thermal Analysis <2.52>
U: Uniformity of dosage units
V: Vitamin A Assay <2.55>

(1) The reference standards which are prepared by those who


have been registered to prepare them by the Minister of
Health, Labour and Welfare, according to the Ministerial
ordinance established by the Minister separately.

Reference Standard Intended Use*

Aceglutamide I, P, A
Acetaminophen I, A
Adrenaline Bitartrate P
Alprostadil I, P, A
Fig. 6. 10–2a Alternative sinker
p-Aminobenzoyl Glutamic Acid P
Amitriptyline Hydrochloride I, U, D, A
Amlexanox I, U, D, A
Amlodipine Besilate I, A
Add the following: Anhydrous Lactose I
Ascorbic Acid A
6.11 Foreign Insoluble Matter Aspirin A
Atropine Sulfate I, A
Test for Ophthalmic Solutions Azathioprine I, A
Baclofen I, U, D, A
Foreign Insoluble Matter Test for Ophthalmic Solutions is Baicalin I, A
a test method to examine foreign insoluble matters in Beclometasone Dipropionate I, A
ophthalmic solutions. Berberine Chloride I, A
When inspect with the unaided eyes at a position of Betamethasone I, P, U, D, A
luminous intensity of 3000 – 5000 lx under an incandescent Betamethasone Sodium Phosphate I, A
lamp after cleaning the exterior of containers, Ophthalmic Betamethasone Valerate I, A
Solutions must be clear and free from readily detectable for- Bisacodyl I, U, A
eign insoluble matters. Caffeine A
Calcium Folinate I, A
Calcium Oxalate Monohydrate T
Camostat Mesilate I, A
Supplement I, JP XV General Tests, Processes and Apparatus 1815

d-Camphor A Hydrochlorothiazide I, A
dl-Camphor A Hydrocortisone I, P, A
Carbidopa I, P, A Hydrocortisone Acetate I, A
Cellacefate I Hydrocortisone Sodium Phosphate I, A
Chlordiazepoxide I, P, U, A Hydrocortisone Succinate I, A
Chlormadinone Acetate I, A Idoxuridine I, A
Chlorpheniramine Maleate I, U, A Imipramine Hydrochloride I, U, D, A
Cholecalciferol I, A Indomethacin I, P, U, D, A
Ciclosporin I, P, A Insulin P, A
Cilostazol I, U, D, A Interleukin-2 A
Cisplatin I, A Isoflurane I, P, A
Clobetasol Propionate I, A Kallidinogenase A
Clofibrate I, A Lactose I
Clomifene Citrate I, A Lactulose P, A
Cortisone Acetate I, A Lanatoside C I, P, U, D, A
Cyanocobalamin I, P, A Limaprost P, A
Deferoxamine Mesilate I, A Low-molecular Mass Heparin AF, A
Deslanoside I, P, A Loxoprofen A
Dexamethasone I, A Lysozyme A
Diclofenamide I, P, D, A Maltose A
Diethylcarbamazine Citrate A Manidipine Hydrochloride I, U, D, A
Digitoxin I, U, D, A Mecobalamin I, A
Digoxin I, U, D, A Melting Point Standard-Acetanilide M
Dihydroergotoxine Mesilate A Melting Point Standard-Acetopheneti-
Dobutamine Hydrochloride I, A dine M
Edrophonium Chloride I, A Melting Point Standard-Caffeine M
Elcatonin A Melting Point Standard-Sulfanilamide M
Enalapril Maleate I, U, D, A Melting Point Standard-Sulfapyridine M
Endotoxin B Melting Point Standard-Vanillin M
Epitiostanol P, A Menatetrenone I, P, A
Ergocalciferol I, A Mestranol I, A
Ergometrine Maleate P, U, A Methotrexate I, A
Estradiol Benzoate I, P, A Methoxsalen I, A
Estriol I, U, D, A Methyldopa I, U, A
Ethenzamide A Methylergometrine Maleate I, U, D, A
Ethinylestradiol I, U, D, A Methylprednisolone Succinate I, P, A
Ethyl Aminobenzoate A Methyltestosterone I, U, A
Ethyl Icosapentate I, P, A Metildigoxin I, A
Etoposide I, A Mexiletine Hydrochloride I, P, A
Fluocinolone Acetonide I, A Mizoribine I, U, D, A
Fluocinonide I, A Nabumetone I, D, A
Fluorometholone I, A Neostigmine Methylsulfate I, A
Fluoxymesterone I, A Nicotinamide I, A
Folic Acid I, U, A Nicotinic Acid I, A
Furosemide I, U, D, A Nilvadipine I, U, D, A
Fursultiamine Hydrochloride I, A Nizatidine I, U, D, A
Gabexate Mesilate I, P, A Noradrenaline Bitartrate P, A
Ginsenoside Rb1 I, A Norgestrel I, U, D, A
Ginsenoside Rg1 I, A Oxytocin P, A
Gitoxin P Ozagrel Sodium I, A
Glycyrrhizinic Acid I, A Paeoniflorin I, A
Gonadorelin Acetate I, A Pentobarbital P, A
Guaifenesin I, A Perphenazine I, U, D, A
Heparin Sodium HB, A Phytonadione A
High-molecular Mass Urokinase A Potassium Sucrose Octasulfate P, A
Human Chorionic Gonadotrophin A Povidone I
Human Insulin I, A Pravastatin 1,1,3,3-tetramethylbutylam-
Human Menopausal Gonadotrophin P, A monium I, A
1816 General Tests, Processes and Apparatus Supplement I, JP XV

Prednisolone I, U, D, A Amoxicillin I, A
Prednisolone Acetate I, A Amphotericin B I, P, A
Prednisolone Succinate I, A Ampicillin I, P, A
Primidone A Arbekacin Sulfate I, A
Probenecid I, D, A Aspoxicillin I, P, A
Prochlorperazine Maleate I, A Astromicin Sulfate I, A
Progesterone I, A Azithromycin I, A
Protamine Sulfate P Aztreonam I, P, A
Puerarin I, A Bacampicillin Hydrochloride I, A
Pyridoxine Hydrochloride I, A Bacitracin I, A
Ranitidine Hydrochloride I, A Bekanamycin Sulfate I, A
Reserpine I, U, D, A Benzylpenicillin Potassium I, A
Retinol Acetate I, V Bleomycin A2 Hydrochloride A
Retinol Palmitate I, V Carumonam Sodium I, P, A
Riboflavin I, A Cefaclor I, P, U, D, A
Ritodrine Hydrochloride I, U, D, A Cefadroxil I, U, D, A
Roxatidine Acetate Hydrochloride I, U, D, A Cefalexin A
Saccharated Pepsin A Cefalotin Sodium I, P, A
Scopolamine Hydrobromide I, A Cefapirin Sodium I, A
Sennoside A I, A Cefatrizine Propylene Glycolate I, A
Sennoside B A Cefazolin P, A
Serum Gonadotrophin A Cefbuperazone A
Spironolactone I, A Cefcapene Pivoxil Hydrochloride I, U, A
Sulfadiazine Silver I, A Cefdinir I, P, D, A
Swertiamarin I, A Cefditoren Pivoxil I, U, D, A
Testosterone Propionate I, A Cefepime Dihydrochloride I, A
Thiamine Chloride Hydrochloride I, P, A Cefixime I, P, A
Thiamylal A Cefmenoxime Hydrochloride I, P, A
Thrombin A Cefmetazole A
Tocopherol I, P, A Cefminox Sodium I, A
Tocopherol Acetate I, A Cefodizime Sodium I, P, A
Tocopherol Nicotinate I, A Cefoperazone A
Tocopherol Succinate A Cefotaxime P, A
Tolazamide I, A Cefotetan I, P, A
Tolbutamide D Cefotiam Hexetil Hydrochloride I, P, IS, A
Tolnaftate I, A Cefotiam Hydrochloride I, P, A
Tranexamic Acid I, P, D, A Cefozopran Hydrochloride I, A
Triamcinolone I, A Cefpiramide P, A
Triamcinolone Acetonide I, A Cefpirome Sulfate I, A
Trichlormethiazide I, U, D, A Cefpodoxime Proxetil I, IS, A
Trihexyphenidyl Hydrochloride I, U, D, A Cefroxadine I, A
Tyrosine A, DG Cefsulodin Sodium I, P, A
Ubidecarenone I, A Ceftazidime I, A
Ulinastatin A Cefteram Pivoxil Mesitylene Sulfonate A
Vasopressin A Ceftibuten Hydrochloride A
Vinblastine Sulfate I, U, A Ceftizoxime P, A
Vincristine Sulfate I, A Ceftriaxone Sodium I, A
Warfarin Potassium I, U, A Cefuroxime Axetil I, P, IS, A
Zidovudine I, A Cefuroxime Sodium I, A
Chloramphenicol I, A
(2) The reference standards which are prepared by National Chloramphenicol Palmitate I, A
Institute of Infectious Diseases. Chloramphenicol Succinate A
Ciclacillin I, A
Reference Standard Intended Use* Clarithromycin I, P, U, D, A
Clindamycin Hydrochloride I, U, D, A
Aclarubicin A Clindamycin Phosphate I, P, A
Actinomycin D I, A Cloxacillin Sodium I, P, A
Amikacin Sulfate I, A Colistin Sodium Methanesulfonate I, A
Supplement I, JP XV General Tests, Processes and Apparatus 1817

Colistin Sulfate A Streptomycin Sulfate I, A


Cycloserine I, A Sulbactam P, A
Daunorubicin Hydrochloride I, A Sulbenicillin Sodium I, A
Demethylchlortetracycline Hydrochlo- Sultamicillin Tosilate I, A
ride I, P, A Talampicillin Hydrochloride I, A
Dibekacin Sulfate I, P, A Teicoplanin I, A
Dicloxacillin Sodium I, A Tetracycline Hydrochloride I, P, A
Diethanolammonium Fusidate A Tobramycin I, A
Doxorubicin Hydrochloride I, A Trichomycin A
Doxycycline Hydrochloride I, A Vancomycin Hydrochloride I, A
Enviomycin Sulfate A Zinostatin Stimalamer I, A
Epirubicin Hydrochloride I, P, A
Erythromycin I, P, A
Faropenem Sodium I, P, U, A 9.21 Standard Solutions for
Flomoxef Triethylammonium P, A
Fosfomycin Phenethylammonium A
Volumetric Analysis
Fradiomycin Sulfate I, A
Add the following:
Gentamicin Sulfate I, A
Gramicidin I, A Zinc sulfate, 0.02 mol/L
Griseofulvin I, P, U, A 1000 mL of this solution contains 5.7516 g of zinc sulfate
Idarubicin Hydrochloride I, U, A heptahydrate (ZnSO4.7H2O: 287.58).
Imipenem I, U, A Preparation—Before use, dilute 0.1 mol/L zinc sulfate VS
Isepamicin Sulfate I, P, A with water to make exactly 5 times the initial volume.
Josamycin I, C, U, A
Josamycin Propionate I, A
Kanamycin Monosulfate I, P, A 9.22 Standard Solutions
Latamoxef Ammonium P, A
Lenampicillin Hydrochloride I, A Add the following:
Leucomycin A5 C, A
Standard Aluminum Solution for Atomic Absorption
Lincomycin Hydrochloride I, P, A
Spectrophotometry To exactly 10 mL of Standard Alumi-
Lithium Clavulanate A
num Stock Solution add water to make exactly 100 mL. Pre-
Meropenem I, A
pare before use. Each mL of this solution contains 0.100 mg
Micronomicin Sulfate I, A
of aluminum (Al).
Midecamycin I, A
Midecamycin Acetate I, A Standard Iron Stock Solution Dissolve exactly 4.840 g
Minocycline Hydrochloride I, P, A of iron (III) chloride hexahydrate in diluted hydrochloric
Mitomycin C I, U, A acid (9 in 25) to make exactly 100 mL.
Mupirocin Lithium P, A
Standard Iron Solution for Atomic Absorption Spec-
Netilmicin Sulfate I, A
trophotometry To exactly 5 mL of Standard Iron Stock
Nystatin I, P, A
Solution add water to make exactly 200 mL. Prepare before
Oxytetracycline Hydrochloride I, A
use. Each mL of this solution contains 0.250 mg of iron (Fe).
Panipenem A
Peplomycin Sulfate I, A Standard Magnesium Stock Solution Dissolve exactly
Phenethicillin Potassium A 8.365 g of magnesium chloride hexahydrate in 2 mol/L
Pimaricin I, A hydrochloric acid TS to make exactly 1000 mL.
Piperacillin I, A
Standard Magnesium Solution for Atomic Absorption
Pirarubicin I, P, A
Spectrophotometry To exactly 1 mL of Standard Magnesi-
Pivmecillinam Hydrochloride I, P, A
um Stock Solution add water to make exactly 100 mL. Pre-
Polymixin B Sulfate I, A
pare before use. Each mL of this solution contains 0.0100
Pyrrolnitrin I, A
mg of magnesium (Mg).
Ribostamycin Sulfate I, A
Rifampicin I, P, A
Rokitamycin I, U, D, A
Roxithromycin I, P, A
9.41 Reagents, Test Solutions
Siccanin I, A
Change the following to read:
Sisomicin Sulfate I, A
Spectinomycin Hydrochloride I, A Albiflorin C23H28O11.xH2O White powder having no
Spiramycin Acetate II C, A odor. Freely soluble in water, in methanol and in ethanol
1818 General Tests, Processes and Apparatus Supplement I, JP XV

(99.5). dehydrocorydaline nitrate for component determination in 1


Identification Determine the absorption spectrum of a mL of a mixture of water and methanol (1:1), and use this
solution of albiflorin in diluted methanol (1 in 2) (1 in solution as the sample solution. Pipet 0.5 mL of the sample
100,000) as directed under Ultraviolet-visible Spectrophoto- solution, add a mixture of water and methanol (1:1) to make
metry <2.24>: it exhibits a maximum between 230 nm and exactly 50 mL, and use this solution as the standard solu-
234 nm. tion. Perform the test with these solutions as directed under
Purity (1) Related substances 1—Dissolve 1 mg of al- Thin-layer Chromatography <2.03>. Spot 5 mL of the sample
biflorin in 1 mL of mehthanol, and perform the test with 10 solution and standard solution on a plate of silica gel for
mL of this solution as directed in the Identification (2) under thin-layer chromatography. Develop immediately with a
Peony Root: any spot other than the principal spot which mixture of methanol, a solution of ammonium acetate (3 in
appears at around Rf 0.2 does not appear. 10) and acetic acid (100) (20:1:1) to a distance of about 10
(2) Related substances 2—Dissolve 1 mg of albiflorin in cm, and air-dry the plate. Spray Dragendorff's TS on the
10 mL of diluted methanol (1 in 2), and use this solution as plate, air-dry, and spray sodium nitrite TS: the spots other
the sample solution. Perform the test with 10 mL of the sam- than the principal spot from the sample solution are not
ple solution as directed in the Assay under Peony Root, and more intense than the spot from the standard solution.
measure the peak areas about 2 times as long as the retention (2) Related substances 2—Dissolve 5.0 mg of de-
time of peoniflorin: the total area of the peaks other than al- hydrocorydaline nitrate for component determination in 10
biflorin from the sample solution is not larger than 1/10 mL of the mobile phase, and use this solution as the sample
times the total area of the peaks other than the solvent peak. solution. Pipet 1 mL of the sample solution, add the mobile
phase to make exactly 100 mL, and use this solution as the
Amygdalin for thin-layer chromatography C20H27NO11
standard solution. Perform the test with exactly 5 mL each of
A white, odorless powder. Soluble in water, sparingly solu-
the sample solution and standard solution as directed under
ble in methanol, and practically insoluble in ethanol (99.5).
Liquid Chromatography <2.01> according to the following
Identification Determine the absorption spectrum of a
conditions, and measure each peak area from these solutions
solution of amygdalin for thin-layer chromatography in
by the automatic integration method: the total area of peaks
methanol (1 in 1000) as directed under Ultraviolet-visible
other than dehydrocorydaline from the sample solution is
Spectrophotometry <2.24>: it exhibits maxima between 250
not larger than the peak area of dehydrocorydaline from the
nm and 254 nm, between 255 nm and 259 nm, between 261
standard solution.
nm and 265 nm, and between 267 nm and 271 nm.
Operating conditions
Purity Related substances—Dissolve 5 mg of amygdalin
Column, column temperature, mobile phase, and flow
for thin-layer chromatography in 2 mL of methanol, and use
rate: Proceed as directed in the operating conditions in the
this solution as the sample solution. Pipet 1 mL of the sam-
Component determination under Corydalis Tuber.
ple solution, add methanol to make exactly 100 mL, and use
Detector: Ultraviolet absorption photometer (wavelength:
this solution as the standard solution. Perform the test with
230 nm)
10 mL each of the sample solution and standard solution as
Time span of measurement: About 3 times as long as the
directed in the Identification under Peach Kernel: any spot
retention time of dehydrocorydaline, beginning after the
other than the principal spot at the Rf value of about 0.3 ob-
peak of nitric acid.
tained from the sample solution is not more intense than the
System suitability
spot from the standard solution.
System performance and system repeatability: Proceed as
Capsaicin for component determination See (E)-capsai- directed in the system suitability in the Component determi-
cin for component determination. nation under Corydalis Tuber.
Test for required detectability: To exactly 1 mL of the
Capsaicin for thin-layer chromatography See (E)-capsai-
standard solution add the mobile phase to make exactly 20
cin for thin-layer chromatography.
mL. Confirm that the peak area of dehydrocorydaline
Chlorogenic acid for thin-layer chromatography See obtained from 5 mL of this solution is equivalent to 3.5 to
(E)-chlorogenic acid for thin-layer chromatography. 6.5z of that from 5 mL of the standard solution.

Cinnamaldehyde for thin-layer chromatography See [6]-Gingerol for thin-layer chromatography C17H26O4
(E)-cinnamaldehyde for thin-layer chromatography. A yellow-white to yellow, liquid or solid. Freely soluble in
methanol, in ethanol (99.5) and in diethyl ether, and practi-
Dehydrocorydaline nitrate for component determination
cally insoluble in water.
C22H24N2O7 Yellow, crystals or crystalline powder. It is
Identification Determine the absorption spectrum of a
sparingly soluble in methanol, and slightly soluble in water
solution of [6]-gingerol for thin-layer chromatography in
and in ethanol (99.5). Melting point: about 2409 C (with
ethanol (99.5) (7 in 200,000) as directed under Ultraviolet-
decomposition).
visible Spectrophotometry <2.24>: it exhibits a maximum be-
Absorbance <2.24> E11zcm (333 nm): 577 – 642 (3 mg,
tween 279 nm and 283 nm.
water, 500 mL). Use the sample dried in a desiccator (silica
Purity Related substances—Dissolve 1.0 mg of [6]-gin-
gel) for not less than 1 hour for the test.
gerol for thin-layer chromatography in exactly 2 mL of
Purity (1) Related substances 1—Dissolve 5.0 mg of
Supplement I, JP XV General Tests, Processes and Apparatus 1819

methanol. Perform the test with 10 mL of this solution as (C18H24N2O5S.HCl), calculated on the anhydrous basis.]
directed in the Identification under Ginger: any spot other
Amygdalin for component determination Amygdalin
than the principal spot at the Rf value of about 0.3 does not
for thin-layer chromatography. However, it meets the fol-
appear.
lowing requirements:
Magnolol for component determination Use magnolol Absorbance <2.24> E11zcm (263 nm): 55 – 58 [20 mg,
for thin-layer chromatography meeting the following addi- methanol, 20 mL; separately determine the water <2.48> (5
tional specifications. mg, coulometric titration) and calculate on the anhydrous
Absorbance <2.24> E11zcm (290 nm): 270 – 293 (10 mg, basis].
methanol, 500 mL). Use the sample dried in a desiccator (sil- Purity Related substances—Dissolve 5 mg of amygdalin
ica gel) for not less than 1 hour for the sample. for component determination in 10 mL of the mobile phase,
Purity Related substances—Dissolve 5.0 mg of mag- and use this as the sample solution. Pipet 1 mL of the sample
nolol for component determination in 10 mL of the mobile solution, add the mobile phase to make exactly 100 mL, and
phase, and use this solution as the sample solution. Pipet 1 use this as the standard solution. Perform the test with ex-
mL of the sample solution, add the mobile phase to make actly 10 mL each of the sample solution and standard solu-
exactly 100 mL, and use this solution as the standard solu- tion as directed under Liquid Chromatography <2.01> ac-
tion. Perform the test with exactly 10 mL each of the sample cording to the following conditions, and determine each
solution and standard solution as directed under Liquid peak area by the automatic integration method: the total
Chromatography <2.01> according to the following condi- area of the peaks other than amygdalin from the sample so-
tions, and determine the area of each peak from these solu- lution is not larger than the peak area of amygdalin from the
tions by the automatic integration method: the total area of standard solution.
peaks other than the peak of magnolol from the sample solu- Operating conditions
tion is not larger than the peak area of magnolol from the Detector, column, column temperature, mobile phase,
standard solution. and flow rate: Proceed as directed in the operating condi-
Operating conditions tions in the Assay (3) under Keishibukuryogan Extract.
Detector, column, column temperature, mobile phase, Time span of measurement: About 3 times as long as the
and flow rate: Proceed as directed in the operating condi- retention time of amygdalin.
tions in the component determination under Magnolia Bark. System suitability
Time span of measurement: About 3 times as long as the Test for required detectability: Pipet 1 mL of the standard
retention time of magnolol. solution, and add the mobile phase to make exactly 20 mL.
System suitability Confirm that the peak area of amygdalin obtained with 10
System performance, and system repeatability: Proceed as mL of this solution is equivalent to 3.5 to 6.5z of that with
directed in the system suitability in the component determi- 10 mL of the standard solution.
nation under Magnolia Bark. System performance and system repeatability: Proceed as
Test for required detectability: To exactly 1 mL of the directed in the system suitability in the Assay (3) under
standard solution add the mobile phase to make exactly 20 Keishibukuryogan Extract.
mL. Confirm that the peak area of magnolol obtained with
Bisoprolol fumarate for assay (C18H31NO4)2.C4H4O4
10 mL of this solution is equivalent to 3.5 to 6.5z of that
[Same as the monograph Bisoprolol Fumarate. However,
with 10 mL of the standard solution.
when dried, it contains not less than 99.0z of bisoprolol
fumarate [(C18H31NO4)2.C4H4O4]. Also, when performing
Add the following:
the Purity (2) under Bisoprolol Fumarate, the total area of
Alminoprofen for assay C13H17NO2 [Same as the the peaks other than bisoprolol is not greater than 1/5 times
monograph Alminoprofen. When dried, it contains not less the peak area of bisoprolol from the standard solution].
than 99.5z of alminoprofen (C13H17NO2).] Purify as follows if needed.
Purification method—Dissolve, with heating, 2 g of Bi-
6-Amidino-2-naphthol methanesulfonate
soprolol Fumarate in 200 mL of ethyl acetate, add 0.5 g of
C11H10N2O.CH4O3S A white to pale yellow crystalline
activated carbon, shake well, and filter using a glass filter
powder. Melting point: about 2339
C (with decomposition).
(G4). Place the filtrate in ice water for 2 hours while oc-
Purity A solution obtained by dissolving 0.5 g of 6-
casional shaking. Collect the crystals that precipitate out us-
amidino-2-naphthol methanesulfonate in 10 mL of
ing a glass filter (G3). Dry the crystals obtained in vacuum at
methanol is clear.
809C for 5 hours using phosphorus (V) oxide as a dessicant.
Aminopyrine C13H17N3O White to pale yellow crystals
Bromothymol blue-sodium hydroxide-ethanol TS Dis-
or crystalline powder.
solve 50 mg of bromothymol blue in 4 mL of diluted 0.2
Melting point <2.60>: 107 – 1099C
mol/L sodium hydroxide TS (1 in 10) and 20 mL of ethanol
Amosulalol hydrochloride for assay C18H24N2O5S.HCl (99.5), and add water to make 100 mL.
[Same as the monograph Amosulalol Hydrochloride. It con-
Bucillamine for assay C7H13NO3S2 [Same as the mono-
tains not less than 99.0z of amosulalol hydrochloride
graph Bucillamine. However, when dried, it contains not
1820 General Tests, Processes and Apparatus Supplement I, JP XV

less than 99.0z of bucillamine (C7H13NO3S2). Furthermore, (E)-Capsaicin for thin-layer chromatography
it conforms to the following test.] C18H27NO3 White crystals, having a strong irritative odor.
Purity Related substances—Dissolve 60 mg of bucilla- Very soluble in methanol, freely soluble in ethanol (95) and
mine for assay in 20 mL of a mixture of water and methanol in diethyl ether, and practically insoluble in water.
(1:1) and use this solution as the sample solution. Pipet 1 mL Melting point <2.60>: 64.5 – 66.59C
of this solution, add the mixture of water and methanol (1:1) Purity Related substances—Dissolve 20 mg of capsaicin
to make exactly 100 mL, and use this solution as the stan- for thin-layer chromatography in 2 mL of methanol, and use
dard solution. When the test is performed according to the this solution as the sample solution. Pipet 1 mL of the sam-
Purity (3) under Bucillamine, the total area of the peaks ple solution, add methanol to make exactly 100 mL, and use
other than the bucillamine peak from the sample solution is this solution as the standard solution. Perform the test with
not larger than the peak area of bucillamine from the stan- 10 mL each of the sample solution and standard solution as
dard solution. directed in the Identification under Capsicum: any spot
other than the principal spot at the Rf value of about 0.5
Buformin hydrochloride for assay C6H15N5.HCl
from the sample solution is not more intense than the spot
[Same as the monograph Buformin Hydrochloride. When
from the standard solution.
dried, it contains not less than 99.5z of buformin hydro-
chloride (C6H15N5.HCl).] Cesium chloride CsCl White crystals or crystalline
powder. Very soluble in water, and freely soluble in ethanol
Butyl benzoate C6H5COOCH2CH2CH2CH3 A clear
(99.5).
and colorless liquid.
Loss on drying <2.41>: Not more than 1.0z (1 g, 1109C, 2
Refractive index <2.45> n20D : 1.495 – 1.500
hours).
Specific gravity <2.56> d 20
20: 1.006 – 1.013
Content: not less than 99.0z. Assay—Weigh accurately
n-Butylboronic acid C4H11BO2 White flakes. about 0.5 g, previously dried, and dissolve in water to make
Melting point <2.60>: 90 – 929C exactly 200 mL. Pipet 20 mL of this solution, add 30 mL of
water, and titrate <2.50> with 0.1 mol/L silver nitrate VS (in-
(E)-Capsaicin for component determination Use (E)-
dicator: fluorescein sodium TS).
capsaicin for thin-layer chromatography meeting the follow-
ing additional specifications. Each mL of 0.1 mol/L silver nitrate VS
Absorbance <2.24> E11zcm (281 nm): 97 – 105 (10 mg, =16.84 mg of CsCl
methanol, 200 mL). Use the sample dried in a desiccator (in
Cesium chloride TS To 25.34 g of cesium chloride add
vacuum, phosphorus (V) oxide, 409 C) for 5 hours for the
water to make 1000 mL.
test.
Purity Related substances—Dissolve 10 mg of capsaicin Cetirizine hydrochloride for assay C21H25ClN2O3.2HCl
for component determination in 50 mL of methanol, and [Same as the monograph Cetirizine Hydrochloride. When
use this solution as the sample solution. Pipet 1 mL of the dried, it contains not less than 99.5z of cetirizine
sample solution, add methanol to make exactly 100 mL, and hydrochloride (C21H25ClN2O3.2HCl).]
use this solution as the standard solution. Perform the test
3?-Chloro-3?-deoxythymidine for liquid chromatography
with exactly 20 mL each of the sample solution and standard
C10H13N2O4Cl Occurs as a white powder.
solution as directed under Liquid Chromatography <2.01>
Purity—Dissolve 10 mg of 3?-chloro-3?-deoxythymidine
according to the following conditions, and measure each
for liquid chromatography in the mobile phase to make 100
peak area from these solutions by the automatic integration
mL. Perform the test with 10 mL of this solution as directed
method: the total area of the peaks other than capsaicin
in the Purity (3) under Zidovudine: a peak is not observed at
from the sample solution is not larger than the peak area of
the retention time for zidovudine.
capsaicin from the standard solution.
Operating conditions (E)-Chlorogenic acid for thin-layer chromatography
Detector, column, column temperature, mobile phase, C16H18O9.xH2O A white powder. Freely soluble in
and flow rate: Proceed the operating conditions in the Com- methanol and in ethanol (99.5), and sparingly soluble in
ponent determination under Capsicum. water. Melting point: about 2059 C (with decomposition).
Time span of measurement: About 3 times as long as the Purity Related substances—Dissolve 1.0 mg of chloro-
retention time of capsaicin beginning after the solvent peak. genic acid for thin-layer chromatography in 2 mL of
System suitability methanol, and use this solution as the sample solution. Per-
System performance, and system repeatability: Proceed form the test with this solution as directed under Thin-layer
the system suitability in the Component determination under Chromatography <2.03>. Spot 10 mL of the sample solution
Capsicum. on a plate of silica gel for thin-layer chromatography, de-
Test for required detectability: Pipet 1 mL of the standard velop the plate with a mixture of ethyl acetate, water and
solution, and add methanol to make exactly 20 mL. Con- formic acid (6:1:1) to a distance of about 10 cm, and air-dry
firm that the peak area of capsaicin from 20 mL of this solu- the plate. Examine under ultraviolet light (main wavelength:
tion is equivalent to 3.5 to 6.5z of that of capsaicin from 20 365 nm): no spot other than the principal spot at around Rf
mL of the standard solution. 0.5 appears.
Supplement I, JP XV General Tests, Processes and Apparatus 1821

Chlorphenesin carbamate for assay C10H12ClNO4 matography in 100 mL of methanol and perform the test as
[Same as the monograph Chlorphenesin Carbamate. When directed in the Purity (2) under Zidovudine: spots other than
dried, it contains not less than 99.0z of chlorphenesin car- the principal spot with an Rf value of about 0.23 are not
bamate (C10H12ClNO4).] observed.

Cibenzoline succinate for assay C18H18N2.C4H6O4 Dilute sodium pentacyanonitrosylferrate (III)-potassium


[Same as the monograph Cibenzoline Succinate. When hexacyanoferrate (III) TS To 5 mL of a solution of sodium
dried, it contains not less than 99.0z of cibenzoline suc- pentacyanonitrosylferrate (III) dihydrate (3 in 50), 5 mL of a
cinate (C18H18N2.C4H6O4) and meets the following require- solution of potassium hexacyanoferrate (III) (13 in 200) and
ment.] 2.5 mL of a solution of sodium hydroxide (1 in 10) add water
Purity: Related substances—Dissolve 0.10 g in 2 mL of to make 25 mL, and mix. Use after the color of the solution
methanol, and use this solution as the sample solution. Pipet changes from dark red to light yellow. Prepare at the time of
1 mL of the sample solution, and add methanol to make ex- use.
actly 100 mL. To exactly 1 mL of this solution add methanol
1,2-Dinitrobenzene C6H4(NO2)2 Occurs as yellowish
to make exactly 10 mL, and use this solution as the standard
white to brownish yellow crystals or a crystalline powder.
solution. Perform the test with these solutions as directed
Identification: Determine the infrared absorption spec-
under Thin-layer Chromatography <2.03>. Spot 10 mL each
trum of 1,2-dinitrobenzene as directed in the paste method
of the sample solution and standard solution on a plate of
under Infrared Spectrophotometry <2.25>: it exhibits ab-
silica gel with fluorescent indicator for thin-layer chro-
sorption at the wave numbers of about 3100 cm-1, 1585
matography. Develop the plate with a mixture of ethyl
cm-1, 1526 cm-1, 1352 cm-1, and 793 cm-1.
acetate, methanol and ammonia solution (28) (20:3:2) to a
Melting point <2.60>: 116 – 1199C
distance of about 10 cm, air-dry the plate, and dry at 809C
for 30 minutes. After cooling, examine under ultraviolet Enalapril maleate C20H28N2O5.C4H4O4 [Same as the
light (main wavelength: 254 nm): the spot other than the namesake monograph]
principal spot obtained with the sample solution is not more
2-Ethylhexyl parahydroxybenzoate C15H22O3 Pale yel-
intense than the spot with the standard solution. On stand-
low, clear viscous liquid. Miscible with methanol (99.5).
ing the plate for 30 minutes in the tank saturated with iodine
Practically insoluble in water.
vapor, the spot other than the principal spot obtained with
Content: not less than 98.0z. Assay—Proceed as
the sample solution is not more intense than the spot with
directed in the Assay under Ethyl Parahydroxybenzoate.
the standard solution.
Each mL of 1 mol/L sodium hydroxide VS
Cilazapril C22H31N3O5.H2O [Same as the monograph
=250.3 mg of C15H22O3
Cilazapril Hydrate]
Etizolam for assay C17H15ClN4S [Same as the mono-
Cilazapril for assay C22H31N3O5.H2O [Same as the
graph Etizolam. When dried, it contains not less than 99.0z
monograph Cilazapril Hydrate. It contains not less than
of etizolam (C17H15ClN4S).]
99.0z of cilazapril (C22H31N3O5), calculated on the anhy-
drous basis.] [6]-Gingerol for component determination [6]-Gingerol
for thin-layer chromatography. However, it meets the fol-
(E)-Cinnamaldehyde for thin-layer chromatography
lowing requirements:
C9H8O A colorless or light yellow liquid, having a charac-
Absorbance <2.24> E11zcm (281 nm): 101 – 112 [7 mg,
teristic aromatic odor. Very soluble in methanol and in
ethanol (99.5), 200 mL].
ethanol (99.5), and practically insoluble in water.
Purity Related substances—Dissolve 5 mg of [6]-gin-
Absorbance <2.24> E11zcm (285 nm): 1679 – 1943 (5 mg,
gerol for component determination in 5 mL of methanol,
methanol, 2000 mL)
and use this as the sample solution. Pipet 1 mL of the sample
Purity Related substances—Dissolve 10 mg in 2 mL of
solution, add methanol to make exactly 50 mL, and use this
methanol. Perform the test with 1 mL of this solution as
as the standard solution. Perform the test with exactly 10 mL
directed in the Identification (3) under Kakkonto Extract: no
each of the sample solution and standard solution as direct-
spot other than the principal spot (Rf value is about 0.4)
ed under Liquid Chromatography <2.01> according to the
appears.
following conditions, and determine each peak area by the
Clorazepate dipotassium for assay automatic integration method: the total area of the peaks
C16H10ClKN2O3.KOH [Same as the monograph Cloraze- other than [6]-gingerol from the sample solution is not larger
pate Dipotassium. When dried it contains not less than than the peak area of [6]-gingerol from the standard solu-
99.0z of clorazepate dipotassium (C16H10ClKN2O3.KOH).] tion.
Operating conditions
1-[(2R,5S)-2,5-Dihydro-5-(hydroxymethyl)-2-furyl] thy-
Detector, column, column temperature, mobile phase,
mine for thin-layer chromatography C10H12N2O4 Occurs
and flow rate: Proceed as directed in the operating condi-
as a white powder.
tions in the Assay (3) under Hangekobokuto Extract.
Purity—Dissolve 0.1 g of 1-[(2R,5S)-2,5-dihydro-5-
Time span of measurement: About 6 times as long as the
(hydroxymethyl)-2-furyl]thymine for thin-layer chro-
1822 General Tests, Processes and Apparatus Supplement I, JP XV

retention time of [6]-gingerol. Purity Related substances—Dissolve 1.0 mg of mag-


System suitability nolol for thin-layer chromatography in 1 mL of methanol,
Test for required detectability: Pipet 1 mL of the standard and use this solution as the sample solution. Perform the test
solution, and add methanol to make exactly 20 mL. Con- with the sample solution as directed under Thin-layer Chro-
firm that the peak area of [6]-gingerol obtained with 10 mL matography <2.03>. Spot 10 mL of the sample solution on a
of this solution is equivalent to 3.5 to 6.5z of that with 10 plate of silica gel with fluorescent indicator for thin-layer
mL of the standard solution. chromatography, develop the plate with a mixture of
System performance and system repeatability: Proceed as hexane, acetone and acetic acid (100) (20:15:1) to a distance
directed in the system suitability in the Assay (3) under Han- of about 10 cm, and air-dry the plate. Examine under ultrav-
gekobokuto Extract. iolet light (main wavelength: 254 nm): any spot other than
the principal spot of around Rf 0.5 does not appear.
4-Hydroxyisophthalic acid HOC6H3(COOH)2 White
crystals or powder. Miconazole nitrate C18H14Cl4N2O.HNO3 [Same as the
Content: not less than 98.0z. Assay—Weigh accurately namesake monograph]
about 0.14 g, dissolve in 50 mL of ethanol (95), and titrate
Minocycline hydrochloride C23H27N3O7.HCl [Same as
<2.50> with 0.1 mol/L sodium hydroxide VS (potentiometric
the namesake monograph]
titration). Perform a blank determination in the same man-
ner, and make any necessary correction. Nile blue C20H20ClN3O Blue-green powder.

Each mL of 0.1 mol/L sodium hydroxide VS 3-Nitroaniline C6H6N2O2 Yellow crystals or crystalline
=9.106 mg of C8H6O5 powder.
Melting point <2.60>: 112 – 1169C
Hyperoside for thin-layer chromatography C21H20O12
Yellow crystals or crystalline powder. Slightly soluble in Nodakenin for thin-layer chromatography C20H24O9
methanol, very slightly soluble in ethanol (99.5), and practi- White powder. Slightly soluble in water and in methanol,
cally insoluble in water. Melting point: about 2209 C (with and very slightly soluble in ethanol (99.5). Melting point:
decomposition). about 2209 C (with decomposition).
Identification: Determine the absorption spectrum of a Identification: Determine the absorption spectrum of a
solution of hyperoside for thin-layer chromatography in solution of nodakenin for thin-layer chromatography in
methanol (1 in 100,000) as directed under Ultraviolet-visible methanol (1 in 100,000) as directed under Ultraviolet-visible
Spectrophotometry <2.24>: it exhibits a maximum between Spectrophotometry <2.24>: it exhibits a maximum between
255 nm and 259 nm. 333 nm and 337 nm.
Purity Related substances—Dissolve 1 mg of hyperoside Optical rotation <2.49>: [a]20 D : +50 – +689 (5 mg,
for thin-layer chromatography in 20 mL of methanol. Per- methanol, 10 mL, 100 mm).
form the test with 10 mL of this solution as directed in the Purity Related substances—Dissolve 1 mg of nodakenin
Identification under Crataegus Fruit: any spot other than for thin-layer chromatography in 3 mL of methanol, and use
the principal spot of around Rf 0.5 does not appear. this solution as the sample solution. Pipet 1 mL of the sam-
ple solution, add methanol to make exactly 100 mL, and use
Isoxsuprine hydrochloride for assay C18H23NO3.HCl
this solution as the standard solution. Proceed with 5 mL
[Same as the monograph Isoxsuprine Hydrochloride]
each of these solutions as directed in the Identification (2)
Labetalol hydrochloride C19H24N2O3.HCl [Same as under Peucedanum Root: the spot other than the principal
the namesake monograph] spot of around Rf 0.3 from the sample solution is not more
intense than the spot from the standard solution.
Labetalol hydrochloride for assay C19H24N2O3.HCl
[Same as the monograph Labetalol Hydrochloride. Oleic acid C18H34O2 Occurs as a colorless or pale yel-
However, when dried, it contains not less than 99.0z of low transparent liquid and has a slightly distinct odor. It is
labetalol hydrochloride (C19H24N2O3.HCl).] miscible with ethanol (95) and with diethyl ether, and practi-
cally insoluble in water.
Lanthanum chloride TS To 58.65 g of lanthanum (III)
Specific gravity <2.56> d 20
20: about 0.9
oxide add 100 mL of hydrochloric acid, and boil. After cool-
Content: not less than 99.0z. Assay—To 40 mL of oleic
ing, add water to make 1000 mL.
acid to be examined add 1 mL of a solution of boron trifluo-
Magnolol for thin-layer chromatography C18H18O2 ride in methanol (3 in 20), mix, and heat on a water bath for
Odorless, white crystals or crystalline powder. Freely soluble 3 minutes. After cooling, add 10 mL of petroleum ether and
in methanol and in ethanol (99.5), and practically insoluble 10 mL of water, shake, collect the ether layer after allowing
in water. Melting point: about 1029 C. to stand, and use as the sample solution. Perform the test
Identification: Determine the absorption spectrum of a with 0.2 mL of the sample solution as directed under Gas
solution of magnolol for thin-layer chromatography in Chromatography <2.02> according to the following condi-
methanol (1 in 50,000) as directed under Ultraviolet-visible tions, determine each peak area by the automatic integration
Spectrophotometry <2.24>: it exhibits a maximum between method, and calculate the amount of methyl oleate by the
287 nm and 291 nm. area percentage method.
Supplement I, JP XV General Tests, Processes and Apparatus 1823

Operating conditions ameter).


Detector: A hydrogen flame-ionization detector Column temperature: A constant temperature of about
Column: A glass column 3 mm in inside diameter and 2 m 409C.
in length, packed with siliceous earth for gas chro- Mobile phase: A mixture of diluted phosphoric acid (1 in
matography (149 – 177 mm) coated with methyl polyacrylate 1000) and acetonitrile (4:1).
in a rate of 5 – 10z. Flow rate: Adjust the flow rate so that the retention time
Column temperature: A constant temperature of about of rosmarinic acid is about 14 minutes.
2209 C. Time span of measurement: About 4 times as long as the
Carrier gas: Helium retention time of rosmarinic acid.
Flow rate: Adjust the flow rate so that the retention time System suitability
of methyl oleate is about 10 minutes. Test for required detectability: Pipet 1 mL of the standard
Time span of measurement: About 2 times as long as the solution, and add methanol to make exactly 20 mL. Con-
retention time of methyl oleate, beginning after the solvent firm that the peak area of rosmarinic acid obtained with 10
peak. mL of this solution is equivalent to 3.5 to 6.5z of that with
10 mL of the standard solution.
(±)-Praeruptorin A for thin-layer chromatography
System performance: When the procedure is run with 10
C21H22O7 White crystals or crystalline powder. Soluble in
mL of the standard solution under the above operating con-
methanol, sparingly slightly soluble in ethanol (99.5), and
ditions, the number of theoretical plates and the symmetry
practically insoluble in water.
factor of the peak of rosmarinic acid are not less than 5000
Identification: Determine the absorption spectrum of a
and not more than 1.5, respectively.
solution of (±)-praeruptorin A for thin-layer chro-
System repeatability: When the test is repeated 6 times
matography in methanol (1 in 100,000) as directed under
with 10 mL of the standard solution under the above operat-
Ultraviolet-visible Spectrophotometry <2.24>: it exhibits a
ing conditions, the relative standard deviation of the peak
maximum between 320 nm and 324 nm.
area of rosmarinic acid is not more than 1.5z.
Melting point <2.60>: 152 – 1569C
Purity Related substances—Dissolve 2 mg of Rosmarinic acid for thin-layer chromatography
(±)-praeruptorin A for thin-layer chromatography in 2 mL C18H16O8 White to pale yellow crystals or crystalline pow-
of methanol, and use this solution as the sample solution. der. Freely soluble in ethanol (99.5), and slightly soluble in
Pipet 1 mL of the sample solution, add methanol to make water. Melting point: about 2059 C (with decomposition).
exactly 100 mL, and use this solution as the standard solu- Identification: Determine the absorption spectrum of a
tion. Proceed with 5 mL each of these solutions as directed in solution of rosmarinic acid for thin-layer chromatography in
the Identification (1) under Peucedanum Root: the spot ethanol (99.5) (1 in 100,000) as directed under Ultraviolet-
other than the principal spot of around Rf 0.3 from the sam- visible Spectrophotometry <2.24>: it exhibits maxima be-
ple solution is not more intense than the spot from the stan- tween 217 nm and 221 nm, between 290 nm and 294 nm, and
dard solution. between 330 nm and 334 nm.
Purity Related substances—Dissolve 10 mg of rosmarin-
Rosmarinic acid for component determination Rosmar-
ic acid for thin-layer chromatography in 2 mL of ethanol
inic acid for thin-layer chromatography. However, it meets
(99.5), and use this solution as the sample solution. Pipet 1
the following requirements:
mL of the sample solution, add ethanol (99.5) to make
Absorbance <2.24> E11zcm (332 nm): 526 – 559 [5 mg,
exactly 50 mL, and use this solution as the standard solu-
ethanol (99.5), 500 mL].
tion. Proceed with 10 mL each of the sample solution and
Purity Related substances—Dissolve 5 mg of rosmarinic
standard solution as directed in the Identification (2) under
acid for component determination in 20 mL of the mobile
Hangekobokuto Extract: the spot other than the principal
phase, and use this as the sample solution. Pipet 1 mL of the
spot of around Rf 0.5 from the sample solution is not more
sample solution, add methanol to make exactly 50 mL, and
intense than the spot from the standard solution.
use this as the standard solution. Perform the test with ex-
actly 10 mL each of the sample solution and standard solu- Sodium di-2-ethylhexyl sulfosuccinate
tion as directed under Liquid Chromatography <2.01> ac- C8H17COOCH2(C8H17COO)CHSO3Na White or translu-
cording to the following conditions, and determine each cent white mucilaginous soft masses. Sparingly soluble in
peak area by the automatic integration method: the total water.
area of the peaks other than rosmarinic acid from the sample Purity Clarity and color of solution: A solution pre-
solution is not larger than the peak area of rosmarinic acid pared by dissolving 1.0 g in 100 mL of water is clear and
from the standard solution. colorless.
Operating conditions Loss on drying <2.41>: not more than 5.0z (1 g, 1059C, 2
Detector: An ultraviolet absorption photometer (wave- hours).
length: 240 nm).
Sodium dihydrogen phosphate TS, pH 2.2 Dissolve 1.56
Column: A stainless steel column 4.6 mm in inside di-
g of sodium dihydrogen phosphate dihydrate in 800 mL of
ameter and 15 cm in length, packed with octadecylsilanized
water, adjust the pH to 2.2 with phosphoric acid, and add
silica gel for liquid chromatography (5 mm in particle di-
1824 General Tests, Processes and Apparatus Supplement I, JP XV

water to make 1000 mL. Melting point <2.60>: 49 – 529C


Purity: Other phenols—Shake vigorously 1.0 g of the sub-
1 mol/L Sulfuric acid TS Add 60 mL of sulfuric acid in
stance to be examined with 20 mL of warm water for 1
1000 mL of water slowly with stirring, then allow to cool.
minute, and filter. To 5 mL of the filtrate add 1 drop of a so-
5 mol/L Sulfuric acid TS Add 300 mL of sulfuric acid in lution of iron (III) chloride hexahydrate (27 in 100): the solu-
1000 mL of water slowly with stirring, then allow to cool. tion reveals a green but not a blue to purple color.

Thymine for liquid chromatography C5H6N2O2 Oc- Thymol-sulfuric acid-methanol TS for spraying Dissolve
curs as a white powder. 1.5 g of thymol for spraying test solution in 100 mL of
Purity—Dissolve 10 mg of the substance to be examined methanol, and add 5.7 mL of sulfuric acid.
in 100 mL of methanol, add the mobile phase to make exact-
Triphenylmethanol for thin-layer chromatography
ly 250 mL, and use this solution as the sample solution.
C19H15OH Occurs as a white powder.
Pipet 5 mL of this solution, add the mobile phase to make
Purity—Dissolve 0.1 g of triphenylmethanol for thin-lay-
exactly 100 mL, and use this solution as the standard solu-
er chromatography in 100 mL of methanol and perform the
tion. Pipet 10 mL each of these solutions and perform the
test as directed in the Purity (2) under Zidovudine: spots
test as directed in the Purity (3) under Zidovudine. Deter-
other than the principal spot with an Rf value of about 0.73
mine the area of each peak in the sample and standard solu-
are not observed.
tions by the automatic integration method: the total area of
peaks other than thymine from the sample solution is not
larger than that from the standard solution. However, the
time span of measurement is about 10 times the retention 9.42 Solid Supports/Column
time of thymine, beginning after the solvent peak. Packings for Chromatography
Thymol for spraying test solution C10H14O White crys-
tals or crystalline powder, having an aromatic odor. Very Add the following:
soluble in methanol and in ethanol (99.5), and practically in- Porous styrene-divinylbenzene copolymer for liquid chro-
soluble in water. matography A porous styrene-divinylbenzen copolymer
Identification: Determine the infrared absorption spec- prepared for liquid chromatography.
trum as directed in the potassium bromide disk method un-
der Infrared Spectrophotometry <2.25>: it exhibits absorp- Strongly basic ion exchange resin for column chro-
tion at the wave numbers of about 2960 cm-1, 1420 cm-1, matography Prepared for column chromatography.
1290 cm-1, 1090 cm-1 and 810 cm-1.
Official Monographs
Add the following: the standard solution. Perform the test with these solutions
as directed under Thin Layer Chromatography <2.03>. Spot
Acemetacin 5 mL each of the sample solution and standard solution on a
plate of silica gel with fluorescent indicator for thin layer
アセメタシン chromatography. Develop the plate with a mixture of
hexane, 4-methyl-2-pentanone and acetic acid (100) (3:2:1)
to a distance of about 10 cm, and air-dry the plate. Examine
under ultraviolet light (main wavelength: 254 nm): not more
than 2 spots other than the principal spot appear from the
sample solution, and these spots are not more intense than
the spot obtained from the standard solution.

Loss on drying <2.41> Not more than 0.5z (1 g, 1059


C, 2
C21H18ClNO6: 415.82
hours).
2-{2-[1-(4-Chlorobenzoyl)-5-methoxy-2-methyl-1H-
indol-3-yl]acetyloxy}acetic acid [53164-05-9] Residue on ignition <2.44> Not more than 0.1z (1 g).

Assay Weigh accurately about 0.35 g of Acemetacin,


Acemetacin, when dried, contains not less than
previously dried, dissolve in 20 mL of acetone, add 10 mL of
99.0z and not more than 101.0z of C21H18ClNO6.
water, and then titrate <2.50> with 0.1 mol/L sodium
Description Acemetacin occurs as a light yellow crystalline hydroxide VS (potentiometric titration). Perform a blank
powder. determination in the same method, and make any necessary
It is soluble in acetone, sparingly soluble in methanol, correction.
slightly soluble in ethanol (99.5), and practically insoluble in
Each mL of 0.1 mol/L sodium hydroxide VS
water.
=41.58 mg of C21H18ClNO6
Identification (1) To 1 mg of Acemetacin add 1 mL of
Containers and storage Containers—Tight containers.
concentrated chromotropic acid TS, and heat in a water bath
for 5 minutes: a red-purple color develops.
(2) Determine the absorption spectrum of a solution of
Acemetacin in methanol (1 in 50,000) as directed under Acetylcholine Chloride for
Ultraviolet-visible Spectrophotometry <2.24>, and compare Injection
the spectrum with the Reference Spectrum: both spectra
exhibit similar intensities of absorption at the same 注射用アセチルコリン塩化物
wavelengths.
(3) Determine the infrared absorption spectrum of Add the following next to Residue on ignition:
Acemetacin as directed in the potassium bromide disk
Uniformity of dosage units <6.02> It meets the requirement
method under Infrared Spectrometry <2.25>, and compare
of the Mass variation test.
the spectrum with the Reference Spectrum: both spectra
exhibit similar intensities of absorption at the same wave Foreign insoluble matter <6.06> Perform the test according
numbers. to Method 2: it meets the requirement.
(4) Perform the test with Acemetacin as directed under
Insoluble particulate matter <6.07> It meets the require-
Flame Coloration Test <1.04> (2): a green color appears.
ment.
Melting point <2.60> 151 – 1549
C
Sterility <4.06> Perform the test according to the Mem-
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of brane filtration method: it meets the requirement.
Acemetacin 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).
(2) Related substances—Dissolve 0.40 g of Acemetacin
in 10 mL of acetone, and use this solution as the sample
solution. Pipet 1 mL of the sample solution, and add
acetone to make exactly 50 mL. Pipet 1 mL of this solution,
add acetone to make exactly 10 mL, and use this solution as

1825
1826 Official Monographs Supplement I, JP XV

<2.24>, and compare the spectrum with the Reference Spec-


Ajimaline Tablets trum: both spectra exhibit similar intensities of absorption at
the same wavelengths.
アジマリン錠 (2) Determine the infrared absorption spectrum of
Alminoprofen as directed in the potassium bromide disk
Add the following next to Identification: method under Infrared Spectrophotometry <2.25>, and com-
pare the spectrum with the Reference Spectrum: both spec-
Uniformity of dosage units <6.02> Perform the test accord-
tra exhibit similar intensities of absorption at the same wave
ing to the following method: it meets the requirement of the
numbers.
Content uniformity test.
To 1 tablet of Ajimaline Tablets add 150 mL of 2nd fluid Melting point <2.60> 106 – 1089
C
for dissolution test, shake to disintegrate the tablet, then add
Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of
2nd fluid for dissolution test to make exactly 200 mL, and
Alminoprofen according to Method 2, and perform the test.
filter this solution through a membrane filter with a pore size
Prepare the control solution with 2.0 mL of Standard Lead
not exceeding 0.8 mm. Discard the first 10 mL of the filtrate,
Solution (not more than 10 ppm).
pipet V mL of the subsequent filtrate equivalent to about 0.5
(2) Arsenic <1.11>—Prepare the test solution with 1.0 g
mg of ajimaline (C20H26N2O2), add 2nd fluid for dissolution
of Alminoprofen according to Method 3, and perform the
test to make exactly 10 mL, and use this solution as the sam-
test (not more than 2 ppm).
ple solution. Separately, weigh accurately about 25 mg of
(3) Related substances—Conduct this procedure using
ajimaline for assay, previously dried in vacuum at 809C for
light-resistant vessels. Dissolve 50 mg of Alminoprofen in
3 hours, dissolve in 2nd fluid for dissolution test to make ex-
100 mL of the mobile phase, and use this solution as the
actly 500 mL, and use this solution as the standard solution.
sample solution. Pipet 2 mL of the sample solution, add the
Determine the absorbances at 288 nm, AT and AS, of the
mobile phase to make exactly 200 mL, and use this solution
sample solution and standard solution as directed under
as the standard solution. Perform the test with exactly 5 mL
Ultraviolet-visible Spectrophotometry <2.24>.
each of the sample solution and standard solution as direct-
Amount (mg) of ajimaline (C20H26N2O2) ed under Liquid Chromatography <2.01> according to the
=WS×(AT/AS)×(1/V)×4 following conditions. Determine each peak area of these so-
lutions by the automatic integration method: the area of the
WS: Amount (mg) of ajimaline for assay
peak other than alminoprofen obtained from the sample so-
lution is not larger than 1/5 times the peak area of
alminoprofen from the standard solution. Furthermore, the
Add the following:
total area of the peaks other than alminoprofen from the
sample solution is not larger than the peak area of
Alminoprofen alminoprofen from the standard solution.
アルミノプロフェン Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Column: A stainless steel column 6.0 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
ameter).
C13H17NO2: 219.28 Column temperature: A constant temperature of about
(2RS)-2-{[4-(2-Methylprop-2-en-1- 259C.
yl)amino]phenyl}propanoic acid [39718-89-3] Mobile phase: A mixture of methanol and diluted acetic
acid (100) (1 in 1000) (4:1).
Alminoprofen, when dried, contains not less than Flow rate: Adjust the flow rate so that the retention time
99.0z and not more than 101.0z of C13H17NO2. of alminoprofen is about 5 minutes.
Description Alminoprofen occurs as white to pale yellow Time span of measurement: About 5 times as long as the
crystals or crystalline powder. retention time of alminoprofen, beginning after the solvent
It is freely soluble in ethanol (99.5) and in acetic acid peak.
(100), and very slightly soluble in water. System suitability—
It gradually turns brown on exposure to light. Test for required detectability: Pipet 1 mL of the standard
A solution of Alminoprofen in ethanol (99.5) (1 in 10) solution, and add the mobile phase to make exactly 10 mL.
shows no optical rotation. Confirm that the peak area of alminoprofen obtained from
5 mL of this solution is equivalent to 7 to 13z of that from 5
Identification (1) Determine the absorption spectrum of mL of the standard solution.
a solution of Alminoprofen in ethanol (99.5) (3 in 500,000) System performance: Dissolve 10 mg each of
as directed under Ultraviolet-visible Spectrophotometry Alminoprofen and butyl parahydroxybenzoate in 100 mL of
Supplement I, JP XV Official Monographs 1827

methanol. Pipet 10 mL of this solution, and add methanol the test with exactly 5 mL each of the sample solution and
to make exactly 50 mL. When the procedure is run with 5 mL standard solution as directed under Liquid Chromatography
of this solution under the above operating conditions, <2.01> according to the conditions described in the Purity (3)
alminoprofen and butyl parahydroxybenzoate are eluted in under Alminoprofen. Determine each peak area of each so-
this order with the resolution between these peaks being not lution by the automatic integration method: the area of the
less than 2.0. peak other than alminoprofen obtained from the sample so-
System repeatability: When the test is repeated 6 times lution is not larger than 1/2 times the peak area of
with 5 mL of the standard solution under the above operat- alminoprofen from the standard solution. Furthermore, the
ing conditions, the relative standard deviation of the peak total area of the peaks other than alminoprofen from the
area of alminoprofen is not more than 2.0z. sample solution is not larger than 2 times the peak area of
alminoprofen from the standard solution.
Loss on drying <2.41> Not more than 0.5z (1 g, in vacu-
um, phosphorus (V) oxide, 1 hour). Uniformity of dosage units <6.02> Perform the test accord-
ing to the following method: it meets the requirement of the
Residue on ignition <2.44> Not more than 0.1z (1 g).
Content uniformity test.
Assay Weigh accurately about 0.3 g of Alminoprofen, To 1 tablet of Alminoprofen Tablets add 5 mL of water,
previously dried, dissolve in 50 mL of acetic acid (100), and shake until the tablet is disintegrated, add 50 mL of ethanol
titrate <2.50> with 0.1 mol/L perchloric acid VS (potentio- (99.5), shake for 20 minutes, then add ethanol (99.5) to
metric titration). Perform a blank determination in the same make exactly 100 mL, and centrifuge. Pipet 3 mL of the
manner, and make any necessary correction. supernatant liquid, add ethanol (99.5) to make exactly 50
mL. Pipet V mL of this solution, add ethanol (99.5) to make
Each mL of 0.1 mol/L perchloric acid VS
exactly V? mL so that each mL contains about 6 mg of
=21.93 mg of C13H17NO2
alminoprofen (C13H17NO2), and use this solution as the sam-
Containers and storage Containers—Well-closed contain- ple solution. Then, proceed as directed in the Assay.
ers.
Amount (mg) of alminoprofen (C13H17NO2)
Storage—Light-resistant.
=WS×(AT/AS)×(V?/V)×(1/3)

WS: Amount (mg) of alminoprofen for assay


Add the following:
Dissolution <6.10> When the test is performed at 50 revolu-
tions per minute according to the Paddle method, using 900
Alminoprofen Tablets mL of 2nd fluid for dissolution test as the dissolution medi-
um, the dissolution rate in 45 minutes of Alminoprofen
アルミノプロフェン錠
Tablets is not less than 80z.
Start the test with 1 tablet of Alminoprofen Tablets,
Alminoprofen Tablets contain not less than 93.0z
withdraw not less than 20 mL of the medium at specified
and not more than 107.0z of the labeled amount of
minute after starting the test, and filter through a membrane
alminoprofen (C13H17NO2: 219.28).
filter with a pore size not exceeding 0.45 mm. Discard the
Method of preparation Prepare as directed under Tablets, first 10 mL of the filtrate, pipet V mL of the subsequent
with Alminoprofen. filtrate, add 0.05 mol/L sodium hydroxide TS to make
exactly V? mL so that each mL contains about 8.9 mg of
Identification Take an amount of powdered Alminoprofen
alminoprofen (C13H17NO2) according to the labeled amount,
Tablets, equivalent to 30 mg of Alminoprofen according to
and use this solution as the sample solution. Separately,
the labeled amount, add ethanol (99.5) to make 100 mL,
weigh accurately about 30 mg of alminoprofen for assay,
shake thoroughly, and centrifuge. To 2 mL of the super-
previously dried in vacuum for 1 hour using phosphorus (V)
natant liquid add ethanol (99.5) to make 100 mL, and deter-
oxide as the dessicant, and dissolve in 0.05 mol/L sodium
mine the absorption spectrum of this solution as directed un-
hydroxide TS to make exactly 100 mL. Pipet 3 mL of this
der Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
solution, add 0.05 mol/L sodium hydroxide TS to make
maxima between 253 nm and 257 nm, and between 298 nm
exactly 100 mL, and use this solution as the standard solu-
and 302 nm.
tion. Determine the absorbances, AT and AS, at 245 nm of
Purity Related substances—Conduct this procedure using the sample solution and standard solution as directed under
light-resistant vessels. Powder 10 tablets of Alminoprofen Ultraviolet-visible Spectrophotometry <2.24>.
Tablets, weigh a portion of the powder equivalent to 50 mg
Dissolution rate (z) with respect to the labeled amount of
of Alminoprofen according to the labeled amount, add 50
alminoprofen (C13H17NO2)
mL of the mobile phase, shake for 15 minutes, add the mo-
=WS×(AT/AS)×(V?/V)×(1/C)×27
bile phase to make exactly 100 mL, centrifuge, and use the
supernatant liquid as the sample solution. Pipet 2 mL of the WS: Amount (mg) of alminoprofen for assay
sample solution, add the mobile phase to make exactly 200 C: Labeled amount (mg) of alminoprofen (C13H17NO2) in
mL, and use this solution as the standard solution. Perform 1 tablet
1828 Official Monographs Supplement I, JP XV

Assay Weigh accurately the mass of not less than 20 tablets entire volume of the sample solution and 100 mL of the
of Alminoprofen Tablets, and powder. Weigh accurately an standard solution on a plate of silica gel for thin-layer chro-
amount equivalent to about 60 mg of alminoprofen matography. Then, develop the plate with a mixture of ethyl
(C13H17NO2), add ethanol (99.5) and shake well, add ethanol acetate, ethanol (99.5) and acetic acid (100) (100:5:1) to a
(99.5) to make exactly 200 mL, and centrifuge. Pipet 2 mL distance of about 10 cm, and air-dry the plate. Spray evenly
of the supernatant liquid, add ethanol (99.5) to make exactly a solution of phosphomolybdic acid n-hydrate in ethanol
100 mL, and use this solution as the sample solution. (99.5) (1 in 10) on the plate, and heat at 1009C for 5 minutes:
Separately, weigh accurately about 30 mg of alminoprofen the color of the spot obtained from the standard solution
for assay, previously dried in vacuum for 1 hour using phos- and the spot corresponding to that location obtained from
phorus (V) oxide as the dessicant, dissolve in ethanol (99.5) the sample solution is dark blue.
to make exactly 100 mL. Pipet 2 mL of this solution, add
pH Being specified separately.
ethanol (99.5) to make exactly 100 mL, and use this solution
as the standard solution. Determine the absorbances, AT and Purity (1) Heavy metals <1.07>—Proceed with 4.0 mL of
AS, at the wavelength of maximum absorption at about 255 Alprostadil Injection according to Method 2, and perform
nm of the sample solution and standard solution as directed the test. Prepare the control solution with 2.0 mL of Stan-
under Ultraviolet-visible Spectrophotometry <2.24>. dard Lead Solution (not more than 5 ppm).
(2) Prostaglandin A1—Use the sample solution obtained
Amount (mg) of alminoprofen (C13H17NO2)
in the Assay as the sample solution. Separately, weigh ac-
=WS×(AT/AS)×2
curately about 10 mg of prostaglandin A1, previously dried
WS: Amount (mg) of alminoprofen for assay for 4 hours in a desiccator (in vacuum, phosphorus (V)
oxide), and dissolve in ethanol (99.5) to make exactly 100
Containers and storage Containers—Well-closed contain-
mL. Pipet 2.5 mL of this solution, and add the mobile phase
ers.
to make exactly 50 mL. Pipet 1 mL of this solution, add ex-
actly 1 mL of the internal standard solution, and use this so-
lution as the standard solution. Perform the test with 40 mL
Add the following:
each of the sample solution and standard solution as direct-
ed under Liquid Chromatography <2.01> according to the
Alprostadil Injection following conditions, determine the ratios, QT and QS, of the
peak area of prostaglandin A1 to that of the internal stan-
アルプロスタジル注射液
dard, and calculate the amount of prostaglandin A1 convert-
ed to alprostadil using the following equation: not more
Alprostadil Injection is an emulsion-type injection.
than 3.0 mg per a volume, equivalent to 5 mg of alprostadil
It contains not less than 80.0z and not more than
(C20H34O5).
125.0z of the labeled amount of alprostadil
(C20H34O5: 354.48). Amount (mg) of prostaglandin A1 (C20H32O4), converted to
alprostadil
Method of preparation Prepare as directed under Injec-
=WS×(QT/QS)×(1/2)×1.054
tions, with Alprostadil.
WS: Amount (mg) of prostaglandin A1
Description Alprostadil Injection occurs as a white emul-
sion and is slightly viscous. It has a distinctive odor. Internal standard solution—Dissolve 50 mg of 1-naphthol in
20 mL of ethanol (99.5). To 3 mL of this solution add the
Identification To a quantity of Alprostadil Injection, cor-
mobile phase to make 100 mL.
responding to 10 mg of Alprostadil according to the labeled
Operating conditions—
amount, add 2 mL of acetonitrile, shake well, and cen-
Proceed as directed in the operating conditions in the
trifuge. To 3.5 mL of the supernatant liquid add 7 mL of
Assay.
diluted phosphoric acid (1 in 1000), and then run this solu-
System suitability—
tion on a column (prepared by filling a 10 mm inside di-
Test for required detectability: To exactly 1 mL of the
ameter, 9 mm long chromatography tube with 0.4 g of 70
standard solution add the mobile phase to make exactly 5
mm octadecylsilanized silica gel for pretreatment) prewashed
mL. Confirm that the peak area of prostaglandin A1 ob-
with 10 mL of methanol and then 10 mL of water. Wash the
tained with 40 mL of this solution is equivalent to 14 to 26z
column with 10 mL of water and then 20 mL of petroleum
of that with 40 mL of the standard solution.
ether, followed by elution with 2.5 mL of a mixture of
System performance, and system repeatability: Proceed as
methanol and water (4:1). Remove the solvent from the ef-
directed in the system suitability in the Assay.
fluent under reduced pressure, dissolve the residue in 100 mL
(3) Peroxide—Pipet 4 mL of Alprostadil Injection,
of ethyl acetate, and use this solution as the sample solution.
place in a glass-stoppered flask, add 15 mL of a mixture of
Separately, dissolve 1 mg of Alprostadil Reference Standard
acetic acid (100) and isooctane (3:2), previously having
in 10 mL of ethyl acetate, and use this solution as the
undergone a 30 minute nitrogen substitution, and dissolve
standard solution. Perform the test with these solutions as
with gentle shaking. To this solution add 0.5 mL of saturat-
directed under Thin-layer Chromatography <2.03>. Spot the
Supplement I, JP XV Official Monographs 1829

ed potassium iodide TS, replace the inside of the vessel with 1 mL of the internal standard solution, shake, and use this
nitrogen, and shake for exactly 5 minutes. Then, add 0.5 mL solution as the sample solution. Separately, weigh accurately
of starch TS, shake vigorously, add 15 mL of water, and about 5 mg of Alprostadil Reference Standard, previously
shake vigorously. Under a stream of nitrogen, titrate <2.50> dried in a desiccator (in vacuum, phosphorus (V) oxide) for
with 0.01 mol/L sodium thiosulfate VS until the color of the 4 hours, dissolve in ethanol (99.5) to make exactly 50 mL,
solution disappears. Separately, perform a blank determina- and use this solution as standard stock solution. Pipet 2.5
tion using 4 mL of water, and make any necessary correc- mL of the standard stock solution, add the mobile phase to
tion. Calculate the amount of peroxides using the following make exactly 50 mL, pipet 1 mL, add exactly 1 mL of the
equation: not more than 0.5 meq/L. internal standard solution, and use this solution as the stan-
dard solution. Perform the test with 40 mL each of the sam-
Amount (meq/L) of peroxides=V×2.5
ple solution and standard solution as directed under Liquid
V: Amount (mL) of 0.01 mol/L sodium thiosulfate VS Chromatography <2.01> according to the following condi-
consumed tions using an apparatus equipped with an automatic
pretreatment device (using a postcolumn reaction), and cal-
(4) Free fatty acids—Pipet 3 mL of Alprostadil Injec-
culate the ratios, QT and QS, of the peak area of alprostadil
tion, add exactly 15 mL of a mixture of 2-propanol, heptane
to that of the internal standard.
and 0.5 mol/L sulfuric acid TS (40:10:1), and shake for 1
minute. After leaving for 10 minutes, add exactly 9 mL of Amount (mg) of alprostadil (C20H34O5)
heptane and exactly 9 mL of water, shake the test tube by in- =WS×(QT/QS)×(1/2)
verting 10 times, leave for 15 minutes, and pipet 9 mL of the
WS: Amount (mg) of Alprostadil Reference Standard
supernatant liquid. To this solution, add 3 mL of a solution
prepared by combining 1 volume of Nile blue solution (1 in Internal standard solution—Dissolve 50 mg of 1-naphthol in
5000) washed 5 times with heptane and 9 volumes of ethanol 20 mL of ethanol (99.5). To 3 mL of this solution add the
(99.5), and use this solution as the sample solution. Titrate mobile phase to make 100 mL.
<2.50> this solution with 0.02 mol/L sodium hydroxide VS Operating conditions—
under a stream of nitrogen. Separately, dissolve 5.65 g of Equipment: Liquid chromatograph consisting of 2 pumps
oleic acid in heptane to make exactly 200 mL, and use this for pumping the mobile phase and the reaction reagent, an
solution as the standard solution. Pipet 25 mL of the stan- automatic pretreatment device, column, reaction coil, detec-
dard solution, add 2 drops of phenolphthalein TS, titrate tor, and recording apparatus. Use a reaction coil that is
<2.50> with 0.1 mol/L potassium hydroxide-ethanol VS until maintained at a constant temperature.
a light red color develops, and determine the correction fac- Detector: An ultraviolet absorption photometer (wave-
tor f. Pipet 30 mL of the standard solution and add heptane length: 278 nm).
to make exactly 200 mL. Pipet 3 mL of this solution, add ex- Column: A stainless steel column 4.6 mm in inside di-
actly 15 mL of a mixture of 2-propanol, heptane and 0.5 ameter and 15 cm in length, packed with octadecylsilanized
mol/L sulfuric acid TS (40:10:1), and shake for 1 minute. silica gel for liquid chromatography (5 mm in particle di-
After leaving for 10 minutes, add exactly 6 mL of heptane ameter).
and exactly 12 mL of water, shake the test tube by inverting Column temperature: A constant temperature of about
10 times, and then titrate <2.50> in the same manner as for 609C.
the sample solution. Determine the volume (mL), VT and VS, Reaction coil: Polytetrafluoroethylene tube 0.5 mm in
of 0.02 mol/L sodium hydroxide VS consumed by the sam- inside diameter and 10 m in length.
ple and standard solutions: the amount of free fatty acid is Mobile phase: Dissolve 9.07 g of potassium dihydrogen
not more than 12.0 meq/L. phosphate in water to make 1000 mL and adjust the pH to
6.3 by adding a solution prepared by dissolving 9.46 g of dis-
Amount (meq/L) of free fatty acids=(VT/VS)×f×15
odium hydrogen phosphate in water to make 1000 mL. To 1
Bacterial endotoxins <4.01> Less than 10 EU/mL. volume of this solution add 9 volumes of water. To 3
volumes of this solution add 1 volume of acetonitrile for liq-
Extractable volume <6.05> It meets the requirement.
uid chromatography.
Foreign insoluble matter <6.06> Perform the test according Reaction reagent: Potassium hydroxide TS.
to Method 1: no easily detectable foreign matter is observed. Reaction temperature: A constant temperature of about
609C.
Sterility <4.06> Perform the test according to the Mem-
Mobile phase flow rate: Adjust the flow rate so that the
brane filter method: it meets the requirement. However, use
retention time of alprostadil is about 7 minutes.
the sample solution consisting of equal volume of Al-
Reaction reagent flow rate: 0.5 mL per minute.
prostadil Injection and a solution prepared by adding water
Automatic pretreatment device: Composed of a pretreat-
to 0.1 g of polysorbate 80 to make 100 mL.
ment column, pump for pumping pretreatment column wash
Particle diameter Being specified separately. solution, and routing valve for 2 high pressure flow paths.
Pretreatment column: A stainless steel column 4 mm in in-
Assay Measure exactly a volume of Alprostadil Injection
side diameter and 2.5 cm in length, packed with octadecyl-
corresponding to 5 mg of alprostadil (C20H34O5), add exactly
1830 Official Monographs Supplement I, JP XV

silanized silica gel for liquid chromatography (5 mm in parti- area of alprostadil to that of the internal standard is not
cle diameter). more than 2.0z.
Pretreatment column wash solution: Ethanol (99.5).
Containers and storage Containers—Hermetic containers.
Flow rate of wash solution: A constant flow rate of about
Storage—Light-resistant, not exceeding 59C, avoiding
2.0 mL per minute.
freezing.
Flow path operating conditions: Change the flow path
operating conditions at the times shown in the table below
using the valves shown in the figure.
Add the following:

Time of switchover (minutes) Amikacin Sulfate Injection


Valve 0 9.0 9.1 *1) *2) アミカシン硫酸塩注射液
RVA 0 0 1 0 0
Amikacin Sulfate Injection is an aqueous injection.
RVB 0 1 1 1 0 It contains not less than 90.0z and not more than
*1) After the internal standard has completely eluted 115.0 z of the labeled amount of amikacin
*2) 0.1 minutes after *1) (C22H43N5O13: 585.60).
Method of preparation Prepare as directed under Injec-
tions, with Amikacin Sulfate.

Description Amikacin Sulfate Injection occurs as a color-


less or pale yellow clear liquid.

Identification To a volume of Amikacin Sulfate Injection,


equivalent to 0.1 g (potency) of Amikacin Sulfate according
to the labeled amount, add water to make 4 mL, and use this
solution as the sample solution. Separately, dissolve 25 mg
(potency) of Amikacin Sulfate Reference Standard in 1 mL
A: RVA valve of water, and use this solution as the standard solution.
B: RVB valve Then, proceed as directed in the Identifiction (2) under
C: Sample injector Amikacin Sulfate.
D: Mobile phase Osmotic pressure ratio Being specified separately.
E: Column for pressure correction
F: Column pH <2.54> 6.0 – 7.5
G: Pretreatment column Bacterial endotoxins <4.01> Less than 0.50 EU/mg (poten-
H: Wash solution cy).
I: Drain
J: Pump Extractable volume <6.05> It meets the requirement.

Foreign insoluble matter <6.06> Perform the test according


Figure Components of automatic pretreatment system to Method 1: it meets the requirement.

System suitability— Insoluble particulate matter <6.07> It meets the require-


System performance: Dissolve 10 mg of prostaglandin A1, ment.
previously dried in a desiccator (in vacuum, phosphorus (V) Sterility <4.06> Perform the test according to the Mem-
oxide) for 4 hours, in ethanol (99.5) to make 100 mL. To 2.5 brane filtration method: it meets the requirement.
mL of this solution add 2.5 mL of the standard stock solu-
tion, and add the mobile phase to make 50 mL. To 1 mL of Assay Take exactly a volume of Amikacin Sulfate Injec-
this solution add 1 mL of the internal standard solution, tion, equivalent to about 0.1 g (potency) of Amikacin Sul-
shake, and perform the test under the above conditions with fate, and add water to make exactly 100 mL. Separately,
40 mL of the solution. Alprostadil, prostaglandin A1 and the weigh accurately an amount of Amikacin Sulfate Reference
internal standard are eluted in this order, and the resolution Standard, equivalent to about 50 mg (potency), and add
between the peaks of alprostadil and prostaglandin A1 is not water to make exactly 50 mL. Take exactly 200 mL each of
less than 10, and that between prostaglandin A1 and the these solutions into stoppered test tubes, then proceed as
internal standard is not less than 2.0. directed in the Assay under Amikacin Sulfate.
System repeatability: When the test is repeated 6 times Amount [mg (potency)] of amikacin (C22H43N5O13)
with 40 mL of the standard solution under the above condi- =WS×(HT/HS)×2
tions, the relative standard deviation of the ratio of the peak
Supplement I, JP XV Official Monographs 1831

WS: Amount [mg (potency)] of Amikacin Sulfate Refer- Add the following:
ence Standard

Container and storage Containers—Hermetic containers. Amlexanox


アンレキサノクス

Aminophylline Injection
アミノフィリン注射液

Add the following next to Identification: C16H14N2O4: 298.29


2-Amino-7-(1-methylethyl)-5-oxo-
Bacterial endotoxins <4.01> Less than 0.6 EU/mg.
5H-[1]benzopyrano[2,3-b]pyridine-3-carboxylic acid
[68302-57-8]
Add the following next to Extractable volume:
Foreign insoluble matter <6.06> Perform the test according Amlexanox, when dried, contains not less than 98.0
to Method 1: it meets the requirement. z and not more than 102.0z of C16H14N2O4.
Insoluble particulate matter <6.07> It meets the require- Description Amlexanox occurs as white to yellowish white
ment. crystals or crystalline powder.
It is very slightly soluble in ethanol (99.5), and practically
Sterility <4.06> Perform the test according to the Mem-
insoluble in water.
brane filtration method: it meets the requirement.
It dissolves in diluted sodium hydroxide TS (1 in 3).

Identification (1) Determine the absorption spectrum of


Amitriptyline Hydrochloride a solution of Amlexanox in ethanol (99.5) (1 in 250,000) as
directed under Ultraviolet-visible Spectrophotometry <2.24>,
Tablets and compare the spectrum with the Reference Spectrum or
アミトリプチリン塩酸塩錠 the spectrum of a solution of Amlexanox Reference Stan-
dard prepared in the same manner as the sample solution:
Add the following next to Identification: both spectra exhibit similar intensities of absorption at the
same wavelengths.
Uniformity of dosage units <6.02> Perform the test accord- (2) Determine the infrared absorption spectrum of Am-
ing to the following method: it meets the requirement of the lexanox as directed in the potassium bromide disk method
Content uniformity test. under Infrared Spectrophotometry <2.25>, and compare the
To 1 tablet of Amitriptyline Hydrochloride Tablets add 50 spectrum with the Reference Spectrum or the spectrum of
mL of diluted methanol (1 in 2), shake to disintegrate the Amlexanox Reference Standard: both spectra exhibit similar
tablet, then add diluted methanol (1 in 2) to make exactly intensities of absorption at the same wave numbers.
100 mL, and filter. Discard the first 20 mL of the filtrate,
pipet V mL of the subsequent filtrate, add methanol to make Purity (1) Chloride <1.03>—Dissolve 1.0 g of Amlexanox
exactly V? mL so that each mL contains about 10 mg of in 20 mL of water and 10 mL of sodium hydroxide TS, add
amitriptyline hydrochloride (C20H23N.HCl), and use this so- 15 mL of dilute nitric acid and water to make 50 mL, cen-
lution as the sample solution. Then, proceed as directed in trifuge, and then filter the supernatant liquid. To 25 mL of
the Assay. this filtrate add water to make 50 mL. Perform the test using
this solution as the test solution. The control solution con-
Amount (mg) of amitriptyline hydrochloride (C20H23N.HCl) sists of 5 mL of sodium hydroxide TS, 7.5 mL of dilute
=WS×(AT/AS)×(V?/V)×(1/20) nitric acid, 0.30 mL of 0.01 mol/L hydrochloric acid VS,
WS: Amount (mg) of Amitriptyline Hydrochloride and water added to make 50 mL (not more than 0.021z).
Reference Standard (2) Heavy metals <1.07>—Proceed with 1.0 g of Amlexa-
nox 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).
(3) Related substances—(i) Dissolve 30 mg of Amlexa-
nox in 50 mL of the mobile phase, and use this solution as
the sample solution. Pipet 1 mL of the sample solution, and
add the mobile phase to make exactly 50 mL. Pipet 1 mL of
this solution, add the mobile phase to make exactly 20 mL,
and use this solution as the standard solution. Perform the
test with exactly 10 mL each of the sample solution and stan-
dard solution as directed under Liquid Chromatography
1832 Official Monographs Supplement I, JP XV

<2.01> according to the following conditions. Determine mL of this solution is equivalent to 7 to 13z of that from 10
each peak area of these solutions by the automatic integra- mL of the standard solution.
tion method: the area of the peak other than amlexanox ob- System performance: Pipet 1 mL of the sample solution,
tained from the sample solution is not larger than 2 times the and add the mobile phase to make 100 mL. To 5 mL of this
peak area of amlexanox from the standard solution. solution add 15 mL of the solution of benzophenone in the
Operating conditions— mobile phase (3 in 1,000,000). When perform the test with
The detector, column, column temperature, mobile phase, 10 mL of this solution according to the above conditions,
and flow rate: Proceed as directed in the operating amlexanox and benzophenone are eluted in this order with
conditions in the Assay. the resolution between these peaks being not less than 10.
Time span of measurement: Until completion of the System repeatability: When the test is repeated 6 times
elution of amlexanox beginning after the solvent peak. with 10 mL of the standard solution under the above operat-
System suitability— ing conditions, the relative standard deviation of the peak
Test for required detectability: Pipet 10 mL of the stan- area of amlexanox is not more than 2.0z.
dard solution, and add the mobile phase to make exactly 100 (iii) The total amount of related substances, when calcu-
mL. Confirm that the peak area of amlexanox obtained lated according to the following formula, is not more than
from 10 mL of this solution is equivalent to 7 to 13z of that 0.5z.
from 10 mL of the standard solution.
Total amount (z) of related substances
System performance: Proceed as directed in the system
={(AT1/AS1)+(AT2/AS2)}×(1/10)
suitability in the Assay.
System repeatability: When the test is repeated 6 times AT1: Total area of the peaks other than amlexanox from the
with 10 mL of the standard solution under the above operat- sample solution obtained in (i)
ing conditions, the relative standard deviation of the peak AT2: Total area of the peaks other than amlexanox from the
area of amlexanox is not more than 2.0z. sample solution obtained in (ii)
(ii) Dissolve 30 mg of Amlexanox in 50 mL of the mobile AS1: Peak area of amlexanox from the standard solution
phase, and use this solution as the sample solution. Pipet 1 obtained in (i)
mL of the sample solution, and add the mobile phase to AS2: Peak area of amlexanox from the standard solution
make exactly 50 mL. Pipet 1 mL of this solution, add the obtained in (ii)
mobile phase to make exactly 20 mL, and use this solution as
Loss on drying <2.41> Not more than 0.3z (1 g, 1059
C, 2
the standard solution. Perform the test with exactly 10 mL
hours).
each of the sample solution and standard solution as direct-
ed under Liquid Chromatography <2.01> according to the Residue on ignition <2.44> Not more than 0.1z (1 g).
following conditions, and determine each peak area of these
Assay Weigh accurately about 30 mg each of Amlexanox
solutions by the automatic integration method: the area of
and Amlexanox Reference Standard, both dried, and dis-
the peak other than amlexanox obtained from the sample
solve them separately in the mobile phase to make exactly 50
solution is not larger than 2 times the peak area of amlexa-
mL. Pipet 5 mL each of these solutions, and add exactly 15
nox from the standard solution.
mL of the internal standard solution, and use these solutions
Operating conditions—
as the sample solution and the standard solution, respective-
Detector, column, and column temperature: Proceed as
ly. Perform the test with 10 mL each of the sample solution
directed in the operating conditions in the Assay.
and standard solution as directed under Liquid Chro-
Mobile phase: Dissolve 7.2 g of disodium hydrogen phos-
matography <2.01> according to the following conditions,
phate dodecahydrate in water to make 1000 mL. Adjust the
and calculate the ratios, QT and QS, of the peak area of am-
pH of this solution to 8.0 by adding a solution prepared by
lexanox to that of the internal standard, respectively.
dissolving 3.1 g of sodium dihydrogen phosphate dihydrate
in 1000 mL of water. To 400 mL of this solution add 600 mL Amount (mg) of C16H14N2O4=WS×(QT/QS)
of acetonitrile.
WS: Amount (mg) of Amlexanox Reference Standard
Flow rate: To 15 mL of a solution of benzophenone in the
mobile phase (3 in 1,000,000) add the mobile phase to make Internal standard solution—A solution of 3-nitroaniline in
20 mL. Adjust the flow rate so that the retention time of the mobile phase (1 in 4000).
benzophenone is about 6.5 minutes when perform the test Operating conditions—
with 10 mL of this solution under the conditions described Detector: An ultraviolet absorption photometer (wave-
above. length: 254 nm).
Time span of measurement: About 3 times as long as the Column: A stainless steel column 4.0 mm in inside di-
retention time of benzophenone, beginning after the peak of ameter and 15 cm in length, packed with octadecylsilanized
amlexanox. silica gel for liquid chromatography (5 mm in particle di-
System suitability— ameter).
Test for required detectability: Pipet 5 mL of the standard Column temperature: A constant temperature of about
solution, and add the mobile phase to make exactly 50 mL. 259C.
Confirm that the peak area of amlexanox obtained from 10 Mobile phase: Dissolve 17.9 g of disodium hydrogen
Supplement I, JP XV Official Monographs 1833

phosphate dodecahydrate in water to make 1000 mL. Adjust about 30 mg of Amlexanox Reference Standard, previously
the pH of this solution to 8.0 by adding a solution prepared dried at 1059 C for 2 hours, and dissolve in the mobile phase
by dissolving 7.8 g of sodium dihydrogen phosphate dihy- to make exactly 50 mL. Pipet 25 mL of this solution, add ex-
drate in 1000 mL of water. To 760 mL of this solution add actly 10 mL of the internal standard solution, add the mo-
240 mL of acetonitrile. bile phase to make 100 mL, and use this solution as the stan-
Flow rate: Adjust the flow rate so that the retention time dard solution. Then, proceed as directed in the Assay under
of amlexanox is about 10 minutes. Amlexanox.
System suitability—
Amount (mg) of amlexanox (C16H14N2O4)
System performance: When the procedure is run with 10
=WS×(QT/QS)×(V/200)
mL of the standard solution according to the above condi-
tions, amlexanox and the internal standard are eluted in this WS: Amount (mg) of Amlexanox Reference Standard
order with the resolution between these peaks being not less
Internal standard solution—A solution of 3-nitroaniline in
than 2.0.
the mobile phase (1 in 500).
System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above condi- Dissolution <6.10> When the test is performed at 50 revolu-
tions, the relative standard deviation of the ratio of the peak tions per minute according to the Paddle method, using 900
area of amlexanox to that of the internal standard is not mL of 2nd fluid for dissolution test as the dissolution medi-
more than 1.0z. um, the dissolution rate in 45 minutes of Amlexanox Tablets
is not less than 80z.
Containers and storage Containers—Well-closed contain-
Start the test with 1 tablet of Amlexanox Tablets,
ers.
withdraw not less than 20 mL of the medium at the specified
minute after starting the test, and filter through a membrane
filter with a pore size not exceeding 0.45 mm. Discard the
Add the following:
first 10 mL of the filtrate, pipet V mL of the subsequent
filtrate, add the dissolution medium to make exactly V? mL
Amlexanox Tablets so that each mL contains about 5.6 mg of amlexanox
(C16H14N2O4) according to the labeled amount, and use this
アンレキサノクス錠
solution as the sample solution. Separately, weigh accurately
about 28 mg of Amlexanox Reference Standard, previously
Amlexanox Tablets contain not less than 93.0z and
dried at 1059C for 2 hours, and dissolve in 2 mL of dilute so-
not more than 107.0z of the labeled amount of am-
dium hydroxide TS, add the dissolution medium to make
lexanox (C16H14N2O4: 298.29).
exactly 50 mL. Pipet 1 mL of this solution, add the dissolu-
Method of preparation Prepare as directed under Tablets, tion medium to make exactly 100 mL, and use this solution
with Amlexanox. as the standard solution. Determine the absorbances, AT and
AS, at 350 nm of the sample solution and standard solution
Identification (1) Take an amount of powdered Amlexa-
as directed under Ultraviolet-visible Spectrophotometry
nox Tablets, equivalent to 10 mg of Amlexanox according to
<2.24>.
the labeled amount, add 100 mL of ethanol (99.5), shake
vigorously, and filter. Pipet 1 mL of the filtrate, add 25 mL Dissolution rate (z) with respect to the labeled amount of
of ethanol (99.5), and use this solution as the sample solu- amlexanox (C16H14N2O4)
tion. Determine the absorption spectrum of the sample solu- =WS×(AT/AS)×(V?/V)×(1/C)×18
tion as directed under Ultraviolet-visible Spectrophotometry
WS: Amount (mg) of Amlexanox Reference Standard
<2.24>: it exhibits absorption maxima between 240 nm and
C: Labeled amount (mg) of amlexanox (C16H14N2O4) in 1
244 nm, between 285 nm and 289 nm, and between 341 nm
tablet
and 352 nm.
(2) Observe the sample solution obtained in (1) under Assay Weigh accurately not less than 20 Amlexanox
ultraviolet light (main wavelength: 365 nm): the solution Tablets, and powder. Weigh accurately a portion of the
shows a bluish-white fluorescence. powder, equivalent to about 15 mg of amlexanox (C16H14N2
O4), add exactly 10 mL of the internal standard solution,
Uniformity of dosage units <6.02> Perform the test accord-
add 80 mL of the mobile phase, shake vigorously for 5
ing to the following method: it meets the requirement of the
minutes, and then add the mobile phase to make 100 mL.
Content uniformity test.
Centrifuge this solution, and use the supernatant liquid as
Take 1 tablet of Amlexanox Tablets, add exactly 0.6 mL
the sample solution. Separately, weigh accurately about 30
of the internal standard solution per 1 mg of amlexanox
mg of Amlexanox Reference Standard, previously dried at
(C16H14N2O4), add the mobile phase to make exactly V mL
1059 C for 2 hours, and dissolve in the mobile phase to make
so there is about 167 mg of amlexanox (C16H14N2O4) per 1
exactly 50 mL. Pipet 25 mL of this solution, add exactly 10
mL, disintegrate the tablet, and then shake vigorously for 5
mL of the internal standard solution, add the mobile phase
minutes. Centrifuge this solution, and use the supernatant
to make 100 mL, and use this solution as the standard solu-
liquid as the sample solution. Separately, weigh accurately
1834 Official Monographs Supplement I, JP XV

tion. Then, proceed as directed in the Assay under Amlexa- um nitrate and 0.1 g of anhydrous sodium carbonate, mix,
nox. and gradually ignite. After cooling, dissolve the residue in 2
mL of dilute hydrochloric acid and 10 mL of water, filter if
Amount (mg) of amlexanox (C16H14N2O4)
necessary, and add barium chloride TS: a white precipitate is
=WS×(QT/QS)×(1/2)
formed.
WS: Amount (mg) of Amlexanox Reference Standard
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
Internal standard solution—A solution of 3-nitroaniline in Amlodipine Besilate according to Method 4, and perform
the mobile phase (1 in 500). the test. Prepare the control solution with 2.5 mL of Stan-
dard Lead Solution (not more than 25 ppm).
Containers and storage Containers—Tight containers.
(2) Related substances—Dissolve 0.10 g of Amlodipine
Besilate in 50 mL of a mixture of water and acetonitrile
(1:1), and use this solution as the sample solution. Pipet 1
Add the following:
mL of the sample solution, and add the mixture of water and
acetonitrile (1:1) to make exactly 100 mL. Pipet 3 mL of this
Amlodipine Besilate solution, add the mixture of water and acetonitrile (1:1) to
make exactly 10 mL, and use this solution as the standard
アムロジピンベシル酸塩
solution. Perform the test with exactly 10 mL each of the
sample solution and standard solution as directed under
Liquid Chromatography <2.01> according to the following
conditions, and determine each peak area by the automatic
integration method: the area of the peak having the relative
retention time of 0.90 with respect to amlodipine, obtained
from the sample solution is not larger than the peak area of
amlodipine from the standard solution, and the area of the
C20H25ClN2O5.C6H6O3S: 567.05 peak other than amlodipine, other than benzenesulfonic acid
3-Ethyl 5-methyl (4RS)-2-[(2-aminoethoxy)methyl]- having the relative retention time of about 0.15 with respect
4-(2-chlorophenyl)-6-methyl-1,4-dihydropyridine-3,5- to amlodipine, and other than the peak mentioned above is
dicarboxylate monobenzenesulfonate [111470-99-6] not larger than 1/3 times the peak area of amlodipine from
the standard solution. Furthermore, total peak area for
Amlodipine Besilate contains not less than 98.0z peaks other than amlodipine and benzenesulfonic acid of the
and not more than 102.0z of C20H25ClN2O5.C6H6O3S, sample solution is not larger than 2.7 times the peak area of
calculated on the anhydrous basis. amlodipine from the standard solution.
Description Amlodipine Besilate occurs as a white to yel- Operating conditions—
lowish white crystalline powder. Detector: An ultraviolet absorption photometer (wave-
It is freely soluble in methanol, soluble in ethanol (99.5), length: 237 nm).
and slightly soluble in water. Column: A stainless steel column 4.6 mm in inside di-
A solution of Amlodipine Besilate in methanol (1 in 100) ameter and 15 cm in length, packed with octadecylsilanized
shows no optical rotation. silica gel for liquid chromatography (3 mm in particle di-
Melting point: about 1989 C (with decomposition). ameter).
Column temperature: A constant temperature of about
Identification (1) Determine the absorption spectrum of 359C.
a solution of Amlodipine Besilate in 0.01 mol/L hydrochlor- Mobile phase A: A mixture of water and trifluoroacetic
ic acid-methanol TS (1 in 40,000) as directed under Ultrav- acid (5000:1).
iolet-visible Spectrophotometry <2.24>, and compare the Mobile phase B: A mixture of acetonitrile and trifluoroa-
spectrum with the Reference Spectrum or the spectrum of a cetic acid (5000:1).
solution of Amlodipine Besilate Reference Standard pre- Flowing of the mobile phase: Control the gradient by mix-
pared in the same manner as the sample solution: both spec- ing the mobile phases A and B as directed in the following
tra exhibit similar intensities of absorption at the same table.
wavelengths.
(2) Determine the infrared absorption spectrum of Time after injection Mobile phase Mobile phase
Amlodipine Besilate as directed in the potassium bromide of sample (min) A (volz) B (volz)
disk method under Infrared Spectrophotometry <2.25>, and 0 – 30 80ª 20 20ª 80
compare the spectrum with the Reference Spectrum or the 30 – 45 20 80
spectrum of Amlodipine Besilate Reference Standard: both
spectra exhibit similar intensities of absorption at the same Flow rate: 1.0 mL per minute.
wave numbers. Time span of measurement: About 3 times as long as the
(3) To 30 mg of Amlodipine Besilate add 0.1 g of sodi- retention time of amlodipine, beginning after the solvent
peak.
Supplement I, JP XV Official Monographs 1835

System suitability— this order with the resolution between these peaks being not
Test for required detectability: Pipet 1 mL of the standard less than 3.
solution, and add a mixture of water and acetonitrile (1:1) to System repeatability: When the test is repeated 6 times
make exactly 10 mL. Confirm that the peak area of amlodi- with 20 mL of the standard solution under the above operat-
pine obtained with 10 mL of this solution is equivalent to 7 to ing conditions, the relative standard deviation of the ratio of
13z of that with 10 mL of the standard solution. the peak area of amlodipine to that of the internal standard
System performance: When the procedure is run with 10 is not more than 1.0z.
mL of the standard solution under the above operating con-
Containers and storage Containers—Well-closed contain-
ditions, the number of theoretical plates and the symmetry
ers.
factor of the peak of amlodipine are not less than 70,000 and
Storage—Light-resistant.
not more than 1.5, respectively.
System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat-
Add the following:
ing conditions, the relative standard deviation of the peak
area of amlodipine is not more than 2.0z.
(3) Residual solvent—Being specified separately. Amosulalol Hydrochloride
Water <2.48> Not more than 0.5z (1 g, volumetric titra- アモスラロール塩酸塩
tion, direct titration).

Residue on ignition <2.44> Not more than 0.2z (1 g).

Assay Weigh accurately about 35 mg each of Amlodipine


Besilate and Amlodipine Besilate Reference Standard
(separately determine the water <2.48> using the same man-
ner as Amlodipine Besilate), dissolve them separately in the C18H24N2O5S.HCl: 416.92
mobile phase to make exactly 250 mL. Pipet 5 mL each of 5-((1RS)-1-Hydroxy-2-{[2-(2-
these solutions, add exactly 5 mL each of the internal stan- methoxyphenoxy)ethyl]amino}ethyl)-
dard solution, add the mobile phase to make 25 mL, and use 2-methylbenzenesulfonamide monohydrochloride
these solutions as the sample solution and standard solution. [70958-86-0]
Perform the test with 20 mL of the sample solution and
standard solution as directed under Liquid Chromatography Amosulalol Hydrochloride contains not less than
<2.01> according to the following conditions, and calculate 98.5z and not more than 101.0z of C18H24N2O5S.
the ratios, QT and QS, of the peak area of amlodipine to that HCl, calculated on the anhydrous basis.
of the internal standard.
Description Amosulalol Hydrochloride occurs as white
Amount (mg) ofC20H25ClN2O5.C6H6O3S crystals or a white crystalline powder. It has a bitter taste.
= W S × ( Q T/ Q S ) It is very soluble in formic acid, freely soluble in
methanol, and sparingly soluble in water and in ethanol
WS: Amount (mg) of Amlodipine Besilate Reference Stan-
(99.5).
dard, calculated on the anhydrous basis
It is hygroscopic.
Internal standard solution—A solution of isobutyl para- A solution of Amosulalol Hydrochloride in methanol (1 in
hydroxybenzoate in the mobile phase (3 in 20,000). 100) shows no optical rotation.
Operating conditions—
Identification (1) Determine the absorption spectrum of
Detector: An ultraviolet absorption photometer (wave-
a solution of Amosulalol Hydrochloride (1 in 20,000) as
length: 237 nm).
directed under Ultraviolet-visible Spectrophotometry <2.24>,
Column: A stainless steel column 4.6 mm in inside di-
and compare the spectrum with the Reference Spectrum:
ameter and 15 cm in length, packed with octadecylsilanized
both spectra exhibit similar intensities of absorption at the
silica gel for liquid chromatography (5 mm in particle di-
same wavelengths.
ameter).
(2) Determine the infrared absorption spectrum of
Column temperature: A constant temperature of about
Amosulalol Hydrochloride as directed in the potassium
259C.
chloride disk method under Infrared Spectrophotometry
Mobile phase: A mixture of methanol and a solution of
<2.25>, and compare the spectrum with the Reference Spec-
potassium dihydrogen phosphate (41 in 10,000) (13:7).
trum: both spectra exhibit similar intensities of absorption at
Flow rate: Adjust the flow rate so that the retention time
the same wave numbers.
of amlodipine is about 8 minutes.
(3) A solution of Amosulalol Hydrochloride (1 in 100)
System suitability—
responds to the Qualitative Tests <1.09> for chloride.
System performance: When the procedure is run with 20
mL of the standard solution under the above operating con-
ditions, amlodipine and the internal standard are eluted in
1836 Official Monographs Supplement I, JP XV

Melting point <2.60> 158 – 1629


C System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat-
Purity (1) Heavy metals < 1.07 > —Place 1.0 g of
ing conditions, the relative standard deviation of the peak
Amosulalol Hydrochloride in a porcelain crucible, add 1.5
area of amosulalol is not more than 1.0z.
mL of sulfuric acid, cover loosely, and heat gently to car-
bonize. After cooling, add 2 mL of nitric acid, heat carefully Water <2.48> Not more than 4.0z (1 g, volumetric titra-
until white fumes no longer are evolved, and then heat inten- tion, direct titration).
sely to 500 – 6009C to incinerate. After cooling, add 2 mL of
Residue on ignition <2.44> Not more than 0.1z (1 g).
hydrochloric acid, proceed according to Method 2, and
perform the test. The control solution, processed in the same Assay Weigh accurately about 0.6 g of Amosulalol
manner as the test solution using the same amounts of Hydrochloride, dissolve in 3 mL of formic acid, add 80 mL
reagents, is prepared by combining 2.0 mL of Standard of a mixture of acetic acid (100) and acetic anhydride (3:2),
Lead Solution and water to make 50 mL (not more than 20 and titrate <2.50> within 5 minutes with 0.1 mol/L perchloric
ppm). acid VS (potentiometric titration). Perform a blank determi-
(2) Related substances—Dissolve 0.10 g of Amosulalol nation using the same procedure, and make any necessary
Hydrochloride in 20 mL of the mobile phase, and use this correction.
solution as the sample solution. Pipet 1 mL of the sample
Each mL of 0.1 mol/L perchloric acid VS
solution, add the mobile phase to make exactly 200 mL, and
=41.69 mg of C18H24N2O5S.HCl
use this solution as the standard solution. Perform the test
with exactly 10 mL each of the sample solution and standard Containers and storage Containers—Tight containers.
solution as directed under Liquid Chromatography <2.01>
according to the following conditions, and determine each
peak area of both solutions by the automatic integration Add the following:
method: the total area of the peaks other than amosulalol
obtained from the sample solution is not larger than 2/5 Amosulalol Hydrochloride Tablets
times the peak area of amosulalol from the standard solu-
tion. アモスラロール塩酸塩錠
Operating conditions—
Detector: An ultraviolet absorption photometer (wave- Amosulalol Hydrochloride Tablets contain not less
length: 272 nm). than 95.0z and not more than 105.0z of the labeled
Column: A stainless steel column 4.6 mm in inside di- amount of amosulalol hydrochloride (C18H24N2O5S.
ameter and 15 cm in length, packed with octadecylsilanized HCl: 416.92).
silica gel for liquid chromatography (5 mm in particle di-
Method of preparation Prepare as directed under Tablets,
ameter).
with Amosulalol Hydrochloride.
Column temperature: A constant temperature of about
309C. Identification To a quantity of powdered Amosulalol
Mobile phase: Dissolve 1.36 g of potassium dihydrogen Hydrochloride Tablets, equivalent to 50 mg of Amosulalol
phosphate in water to make 1000 mL, and adjust to pH 5.7 Hydrochloride according to the labeled amount, add 25 mL
by adding a solution prepared by dissolving 3.58 g of disodi- of 0.1 mol/L hydrochloric acid TS, shake well, and then
um hydrogen phosphate dodecahydrate in water to make centrifuge. To 2.5 mL of the supernatant liquid add water to
1000 mL. To 670 mL of this solution add 330 mL of acetoni- make 100 mL. Determine the absorption spectrum of this
trile. solution as directed under Ultraviolet-visible Spectrophoto-
Flow rate: Adjust the flow rate so that the retention time metry <2.24>: it exhibits a maximum between 270 nm and
of amosulalol is about 7 minutes. 274 nm, and a shoulder between 275 nm and 281 nm.
Time span of measurement: About 2 times as long as the
Uniformity of dosage units <6.02> Perform the test accord-
retention time of amosulalol, beginning after the solvent
ing to the following method: it meets the requirement of the
peak.
Content uniformity test.
System suitability—
Take 1 tablet of Amosulalol Hydrochloride Tablets, disin-
Test for required detectability: Pipet 1 mL of the standard
tegrate by adding 2 mL of 0.1 mol/L hydrochloric acid TS,
solution, and add the mobile phase to make exactly 10 mL.
add 15 mL of methanol, and shake well. Add methanol to
Confirm that the peak area of amosulalol obtained with 10
make exactly V mL so that each mL contains about 0.4 mg
mL of this solution is equivalent to 7 to 13z of that with 10
of amosulalol hydrochloride (C18H24N2O5S.HCl), and cen-
mL of the standard solution.
trifuge. Pipet 5 mL of the supernatant liquid, add exactly 2
System performance: When the procedure is run with 10
mL of the internal standard solution and the mobile phase to
mL of the standard solution under the above operating
make 20 mL, and use this solution as the sample solution.
conditions, the number of theoretical plates and the symmet-
Separately, weigh accurately about 20 mg of amosulalol
ry factor of the peak of amosulalol are not less than 4000
hydrochloride for assay (separately determine the water
and not more than 1.7, respectively.
<2.48> in the same manner as Amosulalol Hydrochloride),
Supplement I, JP XV Official Monographs 1837

and dissolve in methanol to make exactly 50 mL. Pipet 5 mL by adding a soluion prepared by dissolving 3.58 g of disodi-
of this solution, add exactly 2 mL of the internal standard um hydrogen phosphate dodecahydrate in water to make
solution, add the mobile phase to make 20 mL, and use this 1000 mL. To 670 mL of this solution add 330 mL of acetoni-
solution as the standard solution. Then, proceed as directed trile.
in the Assay. Flow rate: Adjust the flow rate so that the retention time
of amosulalol is about 5 minutes.
Amount (mg) of amosulalol hydrochloride
System suitability—
(C18H24N2O5S.HCl)
System performance: When the procedure is run with 50
=WS×(QT/QS)×(V/50)
mL of the standard solution under the above operating con-
WS: Amount (mg) of amosulalol hydrochloride for assay, ditions, the number of theoretical plates and the symmetry
calculated on the anhydrous basis factor of the peak of amosulalol are not less than 4000 and
not more than 1.7, respectively.
Internal standard solution—A solution of ethyl parahydrox-
System repeatability: When the test is repeated 6 times
ybenzoate in methanol (1 in 6250).
with 50 mL of the standard solution under the above operat-
Dissolution <6.10> When the test is performed at 50 revolu- ing conditions, the relative standard deviation of the peak
tions per minute according to the Paddle method, using 900 area of amosulalol is not more than 1.0z.
mL of water as the dissolution medium, the dissolution rate
Assay Take 10 Amosulalol Hydrochloride Tablets, add 20
in 30 minutes of Amosulalol Hydrochloride Tablets is not
mL of 0.1 mol/L hydrochloric acid TS, and shake well to
less than 75z.
disintegrate. Add 120 mL of methanol, again shake well,
Start the test with 1 tablet of Amosulalol Hydrochloride
add methanol to make exactly 200 mL, and then centrifuge.
Tablets, withdraw not less than 20 mL of the medium at the
Pipet a volume of supernatant liquid corresponding to about
specified minute after starting the test, and filter through a
5 mg of amosulalol hydrochloride (C18H24N2O5S.HCl), add
membrane filter with a pore size not exceeding 0.5 mm. Dis-
exactly 5 mL of the internal standard solution, add the
card the first 10 mL of the filtrate, pipet V mL of the subse-
mobile phase to make 50 mL, and use this solution as the
quent filtrate, add water to make exactly V? mL so that each
sample solution. Separately, weigh accurately about 25 mg
mL contains about 5.5 mg of amosulalol hydrochloride
of amosulalol hydrochloride for assay (separately determine
(C18H24N2O5S.HCl) according to the labeled amount, and
the water <2.48> in the same manner as Amosulalol
use this solution as the sample solution. Separately, weigh
Hydrochloride), and dissolve in methanol to make exactly
accurately about 22 mg of amosulalol hydrochloride for
25 mL. Pipet 5 mL of this solution, add exactly 5 mL of the
assay (separately determine the water <2.48> in the same
internal standard solution, add the mobile phase to make 50
manner as Amosulalol Hydrochloride), and dissolve in
mL, and use this solution as the standard solution. Perform
water to make exactly 200 mL. Pipet 5 mL of this solution,
the test with 10 mL each of the sample solution and standard
add water to make exactly 100 mL, and use this solution as
solution as directed under Liquid Chromatography <2.01>
the standard solution. Perform the test with exactly 50 mL
according to the following conditions, and calculate the
each of the sample solution and standard solution as direct-
ratios, QT and QS, of the peak area of amosulalol to that of
ed under Liquid Chromatography <2.01> according to the
the internal standard.
following conditions, and determine the amosulalol peak
areas, AT and AS, of both solutions. Amount (mg) of amosulalol hydrochloride
(C18H24N2O5S.HCl)
Dissolution rate (z) with respect to the labeled amount of
=WS×(QT/QS)×(1/5)
amosulalol hydrochloride (C18H24N2O5S.HCl)
=WS×(AT/AS)×(V?/V)×(1/C)×(45/2) WS: Amount (mg) of amosulalol hydrochloride for assay,
calculated on the anhydrous basis
WS: Amount (mg) of amosulalol hydrochloride for assay,
calculated on the anhydrous basis Internal standard solution—A solution of ethyl parahydrox-
C: Labeled amount (mg) of amosulalol hydrochloride ybenzoate in methanol (1 in 6250).
(C18H24N2O5S.HCl) in 1 tablet Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Operating conditions—
length: 272 nm).
Detector: An ultraviolet absorption photometer (wave-
Column: A stainless steel column 4.6 mm in inside di-
length: 272 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).
259C.
Column temperature: A constant temperature of about
Mobile phase: A mixture of diluted acetic acid (100) (1 in
309C.
25), acetonitrile and a solution of ammonium acetate (1 in
Mobile phase: Dissolve 1.36 g of potassium dihydrogen
250) (5:3:2).
phosphate in water to make 1000 mL, and adjust to pH 5.7
Flow rate: Adjust the flow rate so that the retention time
1838 Official Monographs Supplement I, JP XV

of amosulalol is about 4 minutes. Insoluble particulate matter <6.07> It meets the require-
System suitability— ment.
System performance: When the procedure is run with 10
Sterility <4.06> Perform the test according to the Mem-
mL of the standard solution under the above operating con-
brane filtration method: it meets the requirement.
ditions, amosulalol and the internal standard are eluted in
this order with the resolution between these peaks being not Assay Weigh accurately the mass of the contents of not
less than 7. less than 10 containers of Ampicillin Sodium for Injection.
System repeatability: When the test is repeated 6 times Weigh accurately an amount of a portion of the contents,
with 10 mL of the standard solution under the above operat- equivalent to about 50 mg (potency) of Ampicillin Sodium,
ing conditions, the relative standard deviation of the ratio of add exactly 5 mL of the internal standard solution and dis-
the peak area of amosulalol to that of the internal standard solve. Then add the mobile phase to make 50 mL, and use
is not more than 1.0z. this solution as the sample solution. Separately, weigh ac-
curately an amount of Ampicillin Reference Standard,
Containers and storage Containers—Tight containers.
equivalent to about 50 mg (potency), add exactly 5 mL of
the internal standard solution and dissolve. Then add the
mobile phase to make 50 mL, and use this solution as the
Add the following:
standard solution. Perform the test with 10 mL each of the
sample solution and standard solution as directed under Liq-
Ampicillin Sodium for Injection uid Chromatography <2.01> according to the following con-
ditions, and calculate the ratios, QT and QS, of the peak area
注射用アンピシリンナトリウム
of ampicillin to that of the internal standard.
Ampicillin Sodium for Injection is a preparation for Amount [mg (potency)] of ampicillin (C16H19N3O4S)
injection which is dissolved before use. = W S ×( Q T / Q S )
It contains not less than 90.0z and not more than
WS: Amount [mg (potency)] of Ampicillin Reference
110.0z of the labeled amount of ampicillin
Standard
(C16H19N3O4S: 349.40).
Internal standard solution—A solution of guaifenesin in the
Method of preparation Prepare as directed under Injec-
mobile phase (1 in 200).
tions, with Ampicillin Sodium.
Operating conditions—
Description Ampicillin Sodium for Injection occurs as Detector: An ultraviolet absorption photometer (wave-
white to light yellowish white crystals or crystalline powder. length: 230 nm).
Column: A stainless steel column 4.6 mm in inside di-
Identification Proceed as directed in the Identification (1)
ameter and 15 cm in length, packed with octadecylsilanized
under Ampicillin Sodium.
silica gel for liquid chromatography (5 mm in particle di-
Osmotic pressure ratio Being specified separately. ameter).
Column temperature: A constant temperature of about
pH <2.54> The pH of a solution prepared by dissolving an
259C.
amount of Ampicillin Sodium for Injection, equivalent to
Mobile phase: Dissolve 5.94 mg of diammonium hydro-
1.0 g (potency) of Ampicillin Sodium according to the
gen phosphate in 850 mL of water, add 100 mL of
labeled amount, in 10 mL of water is 8.0 to 10.0.
acetonitril, add phosphoric acid to adjust the pH to 5.0, then
Purity Clarity and color of solution—Dissolve an amount add water to make exactly 1000 mL.
of Ampicillin Sodium for Injection, equivalent to 0.25 g Flow rate: Adjust the flow rate so that the retention time
(potency) of Ampicillin Sodium according to the labeled of ampicillin is about 6 minutes.
amount, in 0.75 mL of water: the solution is clear. Perform System suitability—
the test with the solution as directed under Ultraviolet-visible System performance: When the procedure is run with 10
Spectrophotometry <2.24>: the absorbance at 400 nm is not mL of the standard solution under the above operating con-
more than 0.40. ditions, ampicillin and the internal standard are eluted in
this order with the resolution between these peaks being not
Water <2.48> Not more than 3.0z (0.2 g, volumetric titra-
less than 26.
tion, direct titration).
System repeatability: When the test is repeated 6 times
Bacterial endotoxins <4.01> Less than 0.075 EU/mg (po- with 10 mL of the standard solution under the above operat-
tency). ing conditions, the relative standard deviation of the ratio of
the peak area of ampicillin to that of the internal standard is
Uniformity of dosage units <6.02> It meets the requirement
not more than1.0z.
of the Mass variation test.
Containers and storage Containers—Hermetic containers.
Foreign insoluble matter <6.06> Perform the test according
to Method 2: it meets the requirement.
Supplement I, JP XV Official Monographs 1839

Azelastine Hydrochloride, when dried, contains not


L-Arginine Hydrochloride Injection less than 99.0z and not more than 101.0z of
C22H24ClN3O.HCl.
L-アルギニン塩酸塩注射液
Description Azelastine Hydrochloride occurs as a white
crystalline powder.
Delete the Pyrogen and add the following next
It is freely soluble in formic acid, and slightly soluble in
to pH:
water and in ethanol (99.5).
Bacterial endotoxins <4.01> Less than 0.50 EU/mL. Melting point: about 2259 C (with decomposition).
A solution of Azelastine Hydrochloride (1 in 200) shows
Add the following next to Extractable volume: no optical rotation.
Foreign insoluble matter <6.06> Perform the test according Identification (1) Determine the absorption spectrum of
to Method 1: it meets the requirement. a solution of Azelastine Hydrochloride (3 in 100,000) as
directed under Ultraviolet-visible Spectrophotometry <2.24>,
Insoluble particulate matter <6.07> It meets the require-
and compare the spectrum with the Reference Spectrum:
ment.
both spectra exhibit similar intensities of absorption at the
Sterility <4.06> Perform the test according to the Mem- same wavelengths.
brane filtration method: it meets the requirement. (2) Determine the infrared absorption spectrum of
Azelastine Hydrochloride as directed in the potassium chlo-
ride disk method under Infrared Spectrophotometry <2.25>:
Ascorbic Acid Injection both spectra exhibit similar intensities of absorption at the
same wave numbers.
アスコルビン酸注射液 (3) To 10 mL of a saturated solution of Azelastine
Hydrochloride add 1 mL of dilute nitric acid, and filter to
Add the following next to pH: separate formed crystals: the filtrate responds to the
Bacterial endotoxins <4.01> Less than 0.15 EU/mg. Qualitative Tests <1.09> (2) for chloride.

Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of


Add the following next to Extractable volume: Azelastine Hydrochloride according to Method 2, and per-
Foreign insoluble matter <6.06> Perform the test according form the test. Prepare the control solution with 2.0 mL of
to Method 1: it meets the requirement. Standard Lead Solution (not more than 20 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 1.0 g
Insoluble particulate matter <6.07> It meets the require- of Azelastine Hydrochloride according to Method 3, and
ment. perform the test (not more than 2 ppm).
Sterility <4.06> Perform the test according to the Mem- (3) Related substances—Dissolve 50 mg of Azelastine
brane filtration method: it meets the requirement. Hydrochloride in 100 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 100 mL, and
use this solution as the standard solution. Perform the test
Add the following:
with exactly 20 mL each of the sample solution and standard
solution as directed under Liquid Chromatography <2.01>
Azelastine Hydrochloride according to the following conditions. Determine each peak
アゼラスチン塩酸塩 area of these solutions by the automatic integration method:
each peak area other than azelastine obtained from the sam-
ple solution is not larger than 1/10 times the peak area of
azelastine from the standard solution, and the total area of
the peaks other than the peak of azelastine from the sample
solution is not larger than 1/2 times the peak area of
azelastine from the standard solution.
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 240 nm).
C22H24ClN3O.HCl: 418.36
Column: A stainless steel column 4.6 mm in inside di-
4-[(4-Chlorophenyl)methyl]-2-[(4RS)-
ameter and 15 cm in length, packed with octadecylsilanized
(1-methylazepan-4-yl)]phthalazin-1(2H)-one
silica gel for liquid chromatography (5 mm in particle di-
monohydrochloride [79307-93-0]
ameter).
Column temperature: A constant temperature of about
359C.
1840 Official Monographs Supplement I, JP XV

Mobile phase: A mixture of water, acetonitrile and per- cording to the labeled amount, in 1 mL of hydroxylammoni-
chloric acid (660:340:1). um chloride-ethanol TS, allow to stand for 3 minutes, add 1
Flow rate: Adjust the flow rate so that the retention time mL of acidic ammonium iron (III) sulfate TS, and mix: a
of azelastine is about 10 minutes. red-brown color develops.
Time span of measurement: About 2 times as long as the (2) Dissolve an amount of Aztreonam for Injection,
retention time of azelastine, beginning after the solvent equivalent to 3 mg (potency) of Aztreonam according to the
peak. labeled amount, in 100 mL of water, and determine the
System suitability— absorption spectrum of the solution as directed under
Test for required detectability: Pipet 5 mL of the standard Ultraviolet-visible Spectrophotometry <2.24>: it exhibits a
solution, and add the mobile phase to make exactly 50 mL. maximum between 289 nm and 293 nm.
Confirm that the peak area of azelastine obtained from 20
pH <2.54> The pH of a solution prepared by dissolving an
mL of this solution is equivalent to 7 to 13z of that from 20
amount of Aztreonam for Injection, equivalent to 1.0 g
mL of the standard solution.
(potency) of Aztreonam according to the labeled amount, in
System performance: When the procedure is run with 20
10 mL of water is 4.5 to 7.0.
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry Purity Clarity and color of solution—Dissolve an amount
factor of the peak of azelastine is not less than 5000 and not of Aztreonam for Injection, equivalent to 1.0 g (potency) of
more than 1.5, respectively. Aztreonam according to the labeled amount, in 10 mL of
System repeatability: When the test is repeated 6 times water: the solution is clear, and its absorbance <2.24> at 450
with 20 mL of the standard solution under the above operat- nm is not more than 0.06.
ing conditions, the relative standard deviation of the peak
Water <2.48> Not more than 2.0z (0.5 g, volumetric titra-
area of azelastine is not more than 1.0z.
tion, direct titration).
Loss on drying <2.41> Not more than 1.0z (1 g, 1059
C,
Bacterial endotoxins <4.01> Less than 0.10 EU/mg (poten-
2 hours).
cy).
Residue on ignition <2.44> Not more than 0.1z (1 g).
Uniformity of dosage units <6.02> It meets the requirement
Assay Weigh accurately about 0.6 g of previously dried of the Mass variation test.
Azelastine Hydrochloride, dissolve in 5 mL of formic acid,
Foreign insoluble matter <6.06> Perform the test according
add 70 mL of acetic anhydride, and titrate <2.50> with
to Method 2: it meets the requirement.
0.1 mol/L perchloric acid VS (potentiometric titration). Per-
form a blank determination in the same manner, and make Insoluble particulate matter <6.07> It meets the require-
any necessary correction. ment.

Each mL of 0.1 mol/L perchloric acid VS Sterility <4.06> Perform the test according to the Mem-
=41.84 mg of C22H24ClN3O.HCl brane filtration method: it meets the requirement.

Containers and storage Containers—Tight containers. Assay Take an amount of Aztreonam for Injection,
Storage—Light-resistant. equivalent to about 5 g (potency) of Aztreonam, dissolve the
contents with a suitable amount of water, and transfer to a
100-mL volumetric flask. Wash each container with water,
Add the following: combine the washings and the solution, and add water to
make exactly 100 mL. Pipet 10 mL of this solution, and add
Aztreonam for Injection water to make exactly 50 mL. Pipet 2 mL of this solution,
add exactly 10 mL of the internal standard solution and
注射用アズトレオナム water to make 100 mL, and use this solution as the sample
solution. Separately, weigh accurately an amount of Aztreo-
Aztreonam for Injection is a preparation for injec- nam Reference Standard, equivalent to about 20 mg (poten-
tion which is dissolved before use. cy), dissolve in a suitable amount of water, add exactly 10
It contains not less than 93.0z and not more than mL of the internal standard solution and water to make 100
107.0z of the labeled amount of aztreonam mL, and use this solution as the standard solution. Then,
(C13H17N5O8S2: 435.43). proceed as directed in the Assay under Aztreonam.
Method of preparation Prepare as directed under Amount [mg (potency)] of aztreonam (C13H17N5O8S2)
Injections, with Aztreonam. =WS×(QT/QS)×250
Description Aztreonam for Injection is white to yellowish WS: Amount [mg (potency)] of Aztreonam Reference
white masses or powder. Standard
Identification (1) Dissolve an amount of Aztreonam for Internal standard solution—A solution of 4-aminobenzoic
Injection, equivalent to 6 mg (potency) of Aztreonam ac- acid (1 in 6250).
Supplement I, JP XV Official Monographs 1841

Containers and storage Containers—Hermetic containers. Method of preparation Prepare as directed under Injec-
Storage—Light-resistant. tions, with Benzylpenicillin Potassium.

Description Benzylpenicillin Potassium for Injection oc-


curs as white crystals or crystalline powder.
Baclofen Tablets
Identification Proceed as directed in the Identification (2)
バクロフェン錠 under Benzylpenicillin Potassium.

Osmotic pressure ratio Being specified separately.


Add the following next to Identification:
pH <2.54> The pH of a solution prepared by dissolving an
Uniformity of dosage units <6.02> Perform the test accord-
amount of Benzylpenicillin Potassium for Injection, equiva-
ing to the following method: it meets the requirement of the
lent to 1.0×105 Units of Benzylpenicillin Potassium accord-
Content uniformity test.
ing to the labeled amount, in 10 mL of water is 5.0 to 7.5.
To 1 tablet of Baclofen Tablets add 5 mL of 0.1 mol/L
hydrochloric acid TS, disperse the tablet into small particles Purity Clarity and color of solution—A solution prepared
with the aid of ultrasonic waves, then shake for 10 minutes, by dissolving an amount of Benzylpenicillin Potassium for
and add 0.1 mol/L hydrochloric acid TS to make exactly V Injection, equivalent to 1.0×106 Units of Benzylpenicillin
mL so that each mL contains about 0.5 mg of baclofen Potassium according to the labeled amount, in 10 mL of
(C10H12ClNO2). Centrifuge, pipet 5 mL of the supernatant water is clear. Perform the test with this solution as directed
liquid, add 2 drops of phenolphthalein TS, neutralize with under Ultraviolet-visible Spectrophotometry <2.24>: the
dilute sodium hydroxide TS, then add water to make exactly absorbance at 400 nm is not more than 0.10.
50 mL, and use this solution as the sample solution.
Loss on drying <2.41> Not more than 1.2z (3 g, in vacu-
Separately, weigh accurately about 25 mg of Baclofen Refer-
um, below 0.67 kPa, 609
C, 3 hours).
ence Standard (separately determine the water <2.48> in the
same manner as Baclofen), and dissolve in 0.1 mol/L Bacterial endotoxins <4.01> Less than 1.25×10-4 EU/
hydrochloric acid TS to make exactly 50 mL. Pipet 5 mL of Unit.
this solution, add 2 drops of phenolphthalein TS, neutralize
Uniformity of dosage units <6.02> It meets the requirement
with dilute sodium hydroxide TS, then add water to make
of the Mass variation test.
exactly 50 mL, and use this solution as the standard solu-
tion. To exactly 2 mL each of the sample solution and stan- Foreign insoluble matter <6.06> Perform the test according
dard solution add 4 mL of ninhydrin-tin (II) chloride TS, to Method 2: it meets the requirement.
mix, heat on a water bath for 20 minutes, then immediately
Insoluble particulate matter <6.07> It meets the require-
shake vigorously for 2 minutes. After cooling, add a mixture
ment.
of water and 1-propanol (1:1) to make them exactly 25 mL,
and determine the absorbances, AT and AS, of them at 570 Sterility <4.06> Perform the test according to the Mem-
nm as directed under Ultraviolet-visible Spectrophotometry brane filtration method: it meets the requirement.
<2.24>, using a solution obtained with 2 mL of water by the
Assay Weigh accurately the mass of the contents of not
same procedure as above as the blank.
less than 10 containers of Benzylpenicillin Potassium for In-
Amount (mg) of baclofen (C10H12ClNO2) jection. Weigh accurately an amount of a portion of the con-
=WS×(AT/AS)×(V/50) tents, equivalent to about 6×104 Units of Benzylpenicillin
Potassium, dissolve in water to make exactly 20 mL, and use
WS: Amount (mg) of Baclofen Reference Standard, cal-
this solution as the sample solution. Separately, weigh ac-
culated on the anhydrous basis
curately an amount of Benzylpenicillin Potassium Reference
Standard, equivalent to about 6×104 Units, dissolve in
water to make exactly 20 mL, and use this solution as the
Add the following:
standard solution. Perform the test with exactly 5 mL each of
the sample solution and standard solution as directed under
Benzylpenicillin Potassium for Liquid Chromatography <2.01> according to the following
Injection conditions. Determine the peak area of benzylpenicillin, AT
and AS, from each solution.
注射用ベンジルペニシリンカリウム
Amount (unit) of Benzylpenicillin Potassium
Benzylpenicillin Potassium for Injection is a prepa- (C16H17KN2O4S)
ration for injection which is dissolved before use. =WS×(AT/AS)
It contains not less than 93.0z and not more than WS: Amount (unit) of Benzylpenicillin Potassium Refer-
107.0z of the labeled amount of benzylpenicillin ence Standard
potassium (C16H17KN2O4S: 372.48).
1842 Official Monographs Supplement I, JP XV

Operating conditions— Identification Determine the infrared absorption spectrum


Detector: An ultraviolet absorption photometer (wave- of Biotin as directed in the potassium bromide disc method
length: 254 nm). under Infrared Spectrophotometry <2.25>, and compare the
Column: A stainless steel column 4.6 mm in inside di- spectrum with the Reference Spectrum: both spectra exhibit
ameter and 25 cm in length, packed with octadecylsilanized similar intensities of absorption at the same wave numbers.
silica gel for liquid chromatography (7 mm in particle di-
Optical rotation <2.49> [a]20
D : +89 – +939(after drying,
ameter).
0.4 g, dilute sodium hydroxide TS, 20 mL, 100 mm).
Column temperature: A constant temperature of about
259C. Purity (1) Clarity and color of solution—Dissolve 1.0 g
Mobile phase: To a mixture of diammonium hydrogen of Biotin in 10 mL of 0.5 mol/L sodium hydroxide TS: the
phosphate solution (33 in 5000) and acetonitril (19:6), add solution is clear and colorless.
phosphoric acid to adjust the pH of this solution to 8.0. (2) Heavy metals <1.07>—Proceed with 2.0 g of Biotin
Flow rate: Adjust the flow rate so that the retention time according to Method 2, and perform the test. Prepare the
of benzylpenicillin is about 7.5 minutes. control solution with 2.0 mL of Standard Lead Solution (not
System suitability— more than 10 ppm).
System performance: When the procedure is run with 5 mL (3) Arsenic <1.11>—Place 0.7 g of Biotin in a Kjeldahl
of the standard solution under the above operating condi- flask, add 5 mL of nitric acid and 2 mL of sulfuric acid,
tions, the number of theoretical plates and the symmetry place a small funnel on the mouth of the flask, and carefully
factor of the peak of benzylpenicillin are not less than 6000 heat until white fumes are evolved. After cooling, add 2 mL
and not more than 2.0, respectively. of nitric acid twice, heat, add 2 mL of hydrogen peroxide
System repeatability: When the test is repeated 6 times (30) several times, and heat until the color of the solution
with 5 mL of the standard solution under the above operat- becomes colorless or pale yellow. After cooling, add 2 mL of
ing conditions, the relative standard deviation of the peak saturated ammonium oxalate solution, and heat to concen-
area of benzylpenicillin is not more than 1.0z. trate until white fumes are evolved again. After cooling, add
water to make 5 mL, and perform the test using this solution
Containers and storage Containers—Hermetic containers.
as the test solution (not more than 2.8 ppm).
(4) Related substances—Dissolve 0.10 g of Biotin in 10
mL of diluted ammonia solution (28) (7 in 100), and use this
Betahistine Mesilate solution as the sample solution. Pipet 1 mL of this solution,
and add diluted ammonia solution (28) (7 in 100) to make
ベタヒスチンメシル酸塩
exactly 100 mL. Pipet 10 mL of this solution, add diluted
ammonia solution (28) (7 in 100) to make exactly 50 mL, and
Change the Identification (3) to read:
use this solution as the standard solution. Perform the test
Identification (3) A 30 mg portion of Betahistine Mesi- with these solutions as directed under Thin-layer Chro-
late responds to the Qualitative Tests <1.09> (2) for mesilate. matography <2.03>. Spot 5 mL each of the sample solution
and standard solution on a plate of silica gel for thin-layer
chromatography. Develop the plate with a mixture of 1-
Add the following: butanol, water, and acetic acid (100) (5:2:1) to a distance of
about 10 cm, air-dry the plate, and then dry for 30 minutes
Biotin at 1059 C. Spray the plate evenly with a mixture of a solution
of 4-dimethylaminocinnamaldehyde in ethanol (99.5) (1 in
ビオチン 500) and a solution of sulfuric acid in ethanol (99.5) (1 in 50)
(1:1): the spots other than the principal spot obtained from
the sample solution are not more intense than the spot from
the standard solution.

Loss on drying <2.41> Not more than 0.5z (0.5 g, 1059C,


C10H16N2O3S: 244.31 4 hours).
5-[(3aS,4S,6aR)-2-Oxohexahydro-1H-
Residue on ignition <2.44> Not more than 0.1z (1 g).
thieno[3,4-d]imidazol-4-yl]pentanoic acid [58-85-5]
Assay Weigh accurately about 0.25 g of Biotin, previously
Biotin, when dried, contains not less than 98.5z dried, dissolve by adding exactly 20 mL of 0.1 mol/L sodi-
and not more than 101.0z of C10H16N2O3S. um hydroxide VS, and titrate <2.50> the excess sodium
hydroxide with 0.1 mol/L hydrochloric acid VS (indicator: 2
Description Biotin occurs as white crystals or a white crys-
drops of phenolphthalein TS). Perform a blank determina-
talline powder.
tion in the same manner.
It is very slightly soluble in water and in ethanol (99.5).
It dissolves in dilute sodium hydroxide TS. Each mL of 0.1 mol/L sodium hydroxide VS
Melting point: about 2319 C (with decomposition). =24.43 mg of C10H16N2O3S
Supplement I, JP XV Official Monographs 1843

Containers and storage Containers—Tight containers. <2.24>, and compare the spectrum with the Reference Spec-
trum: both spectra exhibit similar intensities of absorption at
the same wavelengths.
Bisacodyl Suppositories (2) Determine the infrared absorption spectrum of
Bisoprolol Fumarate as directed in the potassium bromide
ビサコジル坐剤 disc method under Infrared Spectrophotometry <2.25>, and
compare the spectrum with the Reference Spectrum: both
Add the following next to Identification: spectra exhibit similar intensities of absorption at the same
wave numbers.
Uniformity of dosage units <6.02> Perform the test accord-
ing to the following method: it meets the requirement of the Melting point <2.60> 101 – 1059
C
Content uniformity test.
Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of
To 1 suppository of Bisacodyl Suppositories add tetra-
Bisoprolol Fumarate according to Method 2, and perform
hydrofuran to make a solution containing about 0.2 mg of
the test. Prepare the control solution with 2.0 mL of Stan-
bisacodyl (C22H19NO4) in each mL, warm to 409 C, and
dard Lead Solution (not more than 10 ppm).
shake to dissolve. After cooling, add tetrahydrofuran to
(2) Related substances—Dissolve 50 mg of Bisoprolol
make exactly V mL so that each mL contains about 10 mg of
Fumarate in 100 mL of a mixture of water and acetonitrile
bisacodyl (C22H19NO4). Pipet 5 mL of this solution, and pro-
(4:1), and use this solution as the sample solution. Pipet 1
ceed as directed in the Assay.
mL of this solution, add the mixture of water and acetoni-
Amount (mg) of bisacodyl (C22H19NO4) trile (4:1) to make exactly 100 mL, and use this solution as
=WS×(QT/QS)×(V/50) the standard solution. Perform the test with exactly 20 mL
each of the sample solution and standard solution as direct-
WS: Amount (mg) of Bisacodyl Reference Standard
ed under Liquid Chromatography <2.01> according to the
Internal standard solution—A solution of ethyl parahydrox- following conditions. Determine each peak area of both
ybenzoate in acetonitrile (3 in 100,000). solutions by the automatic integration method: the area of
the peaks other than bisoprolol obtained from the sample
solution is not larger than 1/2 times the peak area of bi-
Add the following: soprolol from the standard solution. Furthermore, the total
of the areas of all peaks other than bisoprolol from the sam-
Bisoprolol Fumarate ple solution is not larger than the peak area of bisoprolol
from the standard solution.
ビソプロロールフマル酸塩 Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 225 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octylsilanized silica
gel for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
409C.
(C18H31NO4)2.C4H4O4: 766.96 Mobile phase: Dissolve 4.08 g of potassium dihydrogen
(2RS)-1-(4-{[2-(1- phosphate in 1000 mL of water, and adjust to pH 2.5 with
Methylethoxy)ethoxy]methyl}phenoxy)- phosphoric acid. To 800 mL of this solution add 200 mL of
3-[(1-methylethyl)amino]propan-2-ol hemifumarate acetonitrile.
[104344-23-2] Flow rate: Adjust the flow rate so that the retention time
of bisoprolol is about 8 minutes.
Bisoprolol Fumarate, when dried, contains not less Time span of measurement: About 2 times as long as the
than 98.5z and not more than 101.0z of retention time of bisoprolol, beginning after the fumaric
(C18H31NO4)2.C4H4O4. acid peak.
Description Bisoprolol Fumarate occurs as white crystals System suitability—
or a white crystalline powder. Test for required detectability: Pipet 2 mL of the standard
It is very soluble in water and in methanol, and freely solution, and add a mixture of water and acetonitrile (4:1) to
soluble in ethanol (99.5) and in acetic acid (100). make exactly 20 mL. Confirm that the peak area of bi-
A solution of Bisoprolol Fumarate (1 in 10) shows no opti- soprolol obtained from 20 mL of this solution is equivalent
cal rotation. to 7 to 13z of that from 20 mL of the standard solution.
System performance: When the procedure is run with 20
Identification (1) Determine the absorption spectrum of mL of the standard solution under the above operating con-
a solution of Bisoprolol Fumarate in methanol (1 in 10,000) ditions, the number of theoretical plates and the symmetry
as directed under Ultraviolet-visible Spectrophotometry factor of the peak of bisoprolol are not less than 5000 and
1844 Official Monographs Supplement I, JP XV

not more than 1.5, respectively. oxide as a dessicant, dissolve in water to make exactly 200
System repeatability: When the test is repeated 6 times mL, and use as the standard solution. Determine the absor-
with 20 mL of the standard solution under the above operat- bances, AT and AS, of the sample solution and standard so-
ing conditions, the relative standard deviation of the peak lution at 271.5 nm as directed under Ultraviolet-visible Spec-
area of bisoprolol is not more than 1.5z. trophotometry <2.24>.

Loss on drying <2.41> Not more than 0.5z (1 g, in vacu- Amount (mg) of bisoprolol fumarate [(C18H31NO4)2.C4H4O4]
um, phosphorus (V) oxide, 809
C, 5 hours). =WS×(AT/AS)×(V?/V)×(1/20)

Residue on ignition <2.44> Not more than 0.1z (1 g). WS: Amount (mg) of bisoprolol fumarate for assay
Assay Weigh accurately about 0.6 g of Bisoprolol Dissolution <6.10>—When the test is performed at 50 revolu-
Fumarate, previously dried, dissolve in 70 mL of acetic acid tions per minute according to the Paddle method, using 900
(100), and titrate <2.50> with 0.1 mol/L perchloric acid VS mL of 2nd fluid for dissolution test as the dissolution medi-
(indicator: 2 drops of crystal violet TS). The endpoint of um, the dissolution rate in 30 minutes of Bisoprolol
titration is when the purple color of the solution turns blue Fumarate Tablets is not less than 85z.
and then blue-green. Perform a blank determination in the Start the test with 1 tablet of Bisoprolol Fumarate Tablets,
same manner, and make any necessary correction. withdraw not less than 20 mL of the medium at the specified
minute after starting the test, and filter through a membrane
Each mL of 0.1 mol/L perchloric acid VS
filter with a pore size not exceeding 0.45 mm. Discard the
=38.35 mg of (C18H31NO4)2.C4H4O4
first 10 mL of the filtrate, pipet V mL of the subsequent
Containers and storage Containers—Tight containers. filtrate, add the dissolution medium to make exactly V? mL
so that each mL contains about 2.8 mg of bisoprolol
fumarate [(C18H31NO4)2.C4H4O4] according to the labeled
Add the following: amount, and use this solution as the sample solution.
Separately, weigh accurately about 14 mg of bisoprolol
Bisoprolol Fumarate Tablets fumarate for assay, previously dried in vacuum at 809C for 5
hours using phosphorus (V) oxide as a dessicant, and dis-
ビソプロロールフマル酸塩錠 solve in the dissolution medium to make exactly 100 mL.
Pipet 2 mL of this solution, add the dissolution medium to
Bisoprolol Fumarate Tablets contain not less than make exactly 100 mL, and use this solution as the standard
95.0z and not more than 105.0z of the labeled am- solution. Perform the test with exactly 50 mL each of the
ount of bisoprolol fumarate [(C18H31NO4)2.C4H4O4: sample solution and standard solution as directed under Liq-
766.96]. uid Chromatography <2.01> according to the following con-
ditions, and determine the bisoprolol peak areas, AT and AS,
Method of preparation Prepare as directed under Tablets,
of both solutions.
with Bisoprolol Fumarate.
Dissolution rate (z) with respect to the labeled amount of
Identification To a quantity of powdered Bisoprolol
bisoprolol fumarate [(C18H31NO4)2.C4H4O4]
Fumarate Tablets, equivalent to 10 mg of Bisoprolol
=WS×(AT/AS)×(V?/V)×(1/C)×18
Fumarate according to the labeled amount, add 60 mL of
methanol, shake vigorously for 10 minutes, add methanol to WS: Amount (mg) of bisoprolol fumarate for assay
make 100 mL, and filter through a membrane filter with a C: Labeled amount (mg) of bisoprolol fumarate
pore size not exceeding 0.45 mm. Determine the absorption [(C18H31NO4)2.C4H4O4] in 1 tablet
spectrum of the filtrate as directed under Ultraviolet-visible
Operating conditions—
Spectrophotometry <2.24>: it exhibits a maximum between
Detector, column, column temperature, and flow rate:
271 nm and 275 nm.
Proceed as directed in the operating conditions in the Assay.
Uniformity of dosage units <6.02>—Perform the test accord- Mobile phase: Dissolve 4.08 g of potassium dihydrogen
ing to the following method: it meets the requirement of the phosphate in 1000 mL of water, and adjust to pH 2.5 with
Content uniformity test. phosphoric acid. To 750 mL of this solution add 250 mL of
Take 1 tablet of Bisoprolol Fumarate Tablets, disintegrate acetonitrile.
by adding 8 mL of water, and add water to make exactly 10 System suitability—
mL, and then filter through a membrane filter with a pore System performance: When the procedure is run with 50
size not exceeding 0.45 mm. Discard the first 3 mL of the mL of the standard solution under the above operating con-
filtrate, pipet V mL of the subsequent filtrate, add water to ditions, the number of theoretical plates and the symmetry
make exactly V? mL so that each mL contains about 0.1 mg factor of the peak of bisoprolol are not less than 3000 and
of bisoprolol fumarate [(C18H31NO4)2.C4H4O4], and use as not more than 2.0, respectively.
the sample solution. Separately, weigh accurately about 20 System repeatability: When the test is repeated 6 times
mg of bisoprolol fumarate for assay, previously dried under with 50 mL of the standard solution under the above operat-
reduced pressure at 809 C for 5 hours, using phosphorus (V) ing conditions, the relative standard deviation of the peak
Supplement I, JP XV Official Monographs 1845

area of bisoprolol is not more than 2.0z. Add the following:


Assay Weigh accurately not less than 20 Bisoprolol
Fumarate Tablets and powder. Weigh accurately a portion Bucillamine Tablets
of the powder, equivalent to about 20 mg of bisoprolol
ブシラミン錠
fumarate [(C18H31NO4)2.C4H4O4], add exactly 70 mL of a
mixture of water and acetonitrile (3:1) and exactly 10 mL of
Bucillamine Tablets contain not less than 95.0z and
the internal standard solution, shake vigorously for 10
not more than 105.0z of the labeled amount of bucil-
minutes, and add the mixture of water and acetonitrile (3:1)
lamine (C7H13NO3S2: 223.31).
to make 100 mL. Filter this solution through a membrane
filter with a pore size not exceeding 0.45 mm, discard the first Method of preparation Prepare as directed under Tablets,
3 mL of the filtrate, and use the subsequent filtrate as the with Bucillamine.
sample solution. Separately, weigh accurately about 20 mg
Identification (1) To a quantity of powdered Bucillamine
of bisoprolol fumarate for assay, previously dried in vacuum
Tablets, equivalent to 0.1 g of Bucillamine according to the
at 809 C for 5 hours using phosphorus (V) oxide as the des-
labeled amount, add 0.1 g of sodium hydrogen carbonate
sicant, add exactly 10 mL of the internal standard solution,
and 10 mL of water, shake well, filter, and add 1 or 2 drops
dissolve in the mixture of water and acetonitrile (3:1) to
of ninhydrin TS to the filtrate: it exhibits a red-brown color.
make 100 mL, and use this solution as the standard solution.
(2) To a quantity of powdered Bucillamine Tablets,
Perform the test with 20 mL each of the sample solution and
equivalent to 0.1 g of Bucillamine according to the labeled
standard solution as directed under Liquid Chromatography
amount, add 25 mL of water, shake well, and filter. To 5
<2.01> according to the following conditions, and calculate
mL of the filtrate, add 2 mL of dilute sodium hydroxide TS
the ratios, QT and QS, of the peak area of bisoprolol to that
and 1 or 2 drops of sodium pentacyanonitrosylferrate (III)
of the internal standard.
TS: it exhibits a red-purple color.
Amount (mg) of bisoprolol fumarate [(C18H31NO4)2.C4H4O4]
Uniformity of dosage units <6.02>—Perform the test accord-
= W S ×( Q T / Q S )
ing to the following method: it meets the requirement of the
WS: Amount (mg) of bisoprolol fumarate for assay Content uniformity test.
Store the sample solution and standard solution in a cold
Internal standard solution—A solution of isopropyl para-
place until performing the measurements. Take 1 tablet of
hydroxybenzoate in the mixture of water and acetonitrile
Bucillamine Tablets, add exactly 1 mL of the internal stan-
(3:1) (1 in 250).
dard solution per 0.1 g of bucillamine (C7H13NO3S2), then
Operating conditions—
add 3 mL of water and 6 mL of methanol per 0.1 g of bucil-
Detector: An ultraviolet absorption photometer (wave-
lamine (C7H13NO3S2), and stir well until the tablet complete-
length: 225 nm).
ly disintegrated. To 1 mL of this solution add the mobile
Column: A stainless steel column 4.6 mm in inside di-
phase to make 25 mL, filter through a membrane filter with
ameter and 15 cm in length, packed with octylsilanized silica
a pore size not exceeding 0.45 mm, and use the filtrate as the
gel for liquid chromatography (5 mm in particle diameter).
sample solution. Then, proceed as directed in the Assay.
Column temperature: A constant temperature of about
409C. Amount (mg) of bucillamine (C7H13NO3S2)
Mobile phase: Dissolve 4.08 g of potassium dihydrogen =WS×(QT/QS)×C×(1/200)
phosphate in 1000 mL of water, and adjust to pH 2.5 with
WS: Amount (mg) of bucillamine for assay
phosphoric acid. To 800 mL of this solution add 200 mL of
C: Labeled amount (mg) of bucillamine (C7H13NO3S2) in
acetonitrile.
1 tablet
Flow rate: Adjust the flow rate so that the retention time
of bisoprolol is about 8 minutes. Internal standard solution—A solution of 4-fluorobenzoic
System suitability— acid in methanol (1 in 100).
System performance: When the procedure is run with 20
Dissolution <6.10>—When the test is performed at 50 revolu-
mL of the standard solution under the above operating con-
tions per minute according to the Paddle method, using 900
ditions, fumaric acid, bisoprolol and the internal standard
mL of water as the dissolution medium, the dissolution rate
are eluted in this order with the resolution between the peaks
in 30 minutes of Bucillamine Tablets is not less than 80z.
of bisoprolol and the internal standard being not less than
Store the sample solution and standard solution in a cold
12.
place until performing the measurements. Start the test with
System repeatability: When the test is repeated 6 times
1 tablet of Bucillamine Tablets, withdraw not less than 20
with 20 mL of the standard solution under the above operat-
mL of the medium at the specified minute after starting the
ing conditions, the relative standard deviation of the ratio of
test, and filter through a membrane filter with a pore size
the peak area of bisoprolol to that of the internal standard is
not exceeding 0.45 mm. Discard the first 10 mL of the
not more than 1.0z.
filtrate, and use the subsequent filtrate as the sample solu-
Containers and storage Containers—Tight containers. tion. Separately, weigh accurately an amount of bucillamine
1846 Official Monographs Supplement I, JP XV

for assay equivalent to the labeled amount of the tablet,


Amount (mg) of bucillamine (C7H13NO3S2)
previously dried in vacuum at 609 C for 6 hours using phos-
=WS×(QT/QS)×C×(1/200)
phorus (V) oxide as a dessicant, and dissolve in methanol to
make exactly 10 mL. Pipet 1 mL of this solution, add water WS: Amount (mg) of bucillamine for assay
to make exactly 100 mL, and use this solution as the stan- C: Labeled amount (mg) of bucillamine (C7H13NO3S2) in
dard solution. Perform the test with exactly 20 mL each of 1 tablet
the sample solution and standard solution as directed under
Internal standard solution—A solution of 4-fluorobenzoic
Liquid Chromatography <2.01> according to the following
acid in methanol (1 in 100).
conditions, and determine the bucillamine peak areas, AT
Operating conditions—
and AS, of both solutions.
Detector: An ultraviolet absorption photometer (wave-
Dissolution rate (z) with respect to the labeled amount of length: 254 nm).
bucillamine (C7H13NO3S2) Column: A stainless steel column 4.6 mm in inside di-
=WS×(AT/AS)×(1/C)×90 ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
WS: Amount (mg) of bucillamine for assay
ameter).
C: Labeled amount (mg) of bucillamine (C7H13NO3S2) in
Column temperature: A constant temperature of about
1 tablet
409C.
Operating conditions— Mobile phase: A mixture of diluted phosphoric acid (1 in
Detector, column, and column temperature: Proceed as 1000) and methanol (3:2).
directed in the operating conditions in the Assay. Flow rate: Adjust the flow rate so that the retention time
Mobile phase: A mixture of diluted phosphoric acid (1 in of bucillamine is about 5 minutes.
1000) and methanol (11:9). System suitability—
Flow rate: Adjust the flow rate so that the retention time System performance: When the procedure is run with 10
of bucillamine is about 4 minutes. mL of the standard solution under the above operating con-
System suitability— ditions, bucillamine and the internal standard are eluted in
System performance: When the procedure is run with 20 this order with the resolution between these peaks being not
mL of the standard solution under the above operating con- less than 3.
ditions, the number of theoretical plates and the symmetry System repeatability: When the test is repeated 6 times
factor of the peak of bucillamine are not less than 3000 and with 10 mL of the standard solution under the above operat-
not more than 1.5, respectively. ing conditions, the relative standard deviation of the ratio of
System repeatability: When the test is repeated 6 times the peak area of bucillamine to that of the internal standard
with 20 mL of the standard solution under the above operat- is not more than 1.0z.
ing conditions, the relative standard deviation of the peak
Containers and storage Containers—Tight containers.
area of bucillamine is not more than 2.0z.

Assay Store the sample solution and standard solution in a


cold place until performing the measurements. Take 10 Add the following:
tablets of Bucillamine Tablets, add exactly 1 mL of the inter-
nal standard solution per 0.1 g of bucillamine (C7H13NO3S2), Buformin Hydrochloride
add 3 mL of water and 6 mL of methanol, and stir well until
the tablets completely disintegrated. To 1 mL of this solu- ブホルミン塩酸塩
tion add the mobile phase to make 25 mL, filter through a
membrane filter with a pore size not exceeding 0.45 mm, and
use this solution as the sample solution. Separately, weigh
accurately about 0.2 g of bucillamine for assay, previously
C6H15N5.HCl: 193.68
dried in vacuum for 6 hours at 609C using phosphorus (V)
1-Butylbiguanide hydrochloride [1190-53-0]
oxide as a dessicant, add exactly 2 mL of the internal stan-
dard solution, and add 6 mL of water and 12 mL of
Buformin Hydrochloride, when dried, contains not
methanol. To 1 mL of this solution add 25 mL of the mobile
less than 98.5z and not more than 101.0z of
phase, filter through a membrane filter with a pore size not
C6H15N5.HCl.
exceeding 0.45 mm, and use this solution as the standard so-
lution. Perform the test with 10 mL each of the sample solu- Description Buformin Hydrochloride occurs as a white
tion and standard solution as directed under Liquid Chro- crystalline powder.
matography <2.01> according to the following conditions, It is freely soluble in water and in ethanol (99.5).
and calculate the ratios, QT and QS, of the peak area of
Identification (1) To 5 mL of a solution of Buformin
bucillamine to that of the internal standard.
Hydrochloride (1 in 2000) add 1 mL of dilute sodium pen-
tacyanonitrosylferrate (III)-potassium hexacyanoferrate
Supplement I, JP XV Official Monographs 1847

(III) TS: a red-brown color develops. solution, and add the mobile phase to make exactly 10 mL.
(2) Determine the absorption spectrum of a solution of Confirm that the peak area of buformin obtained from 10
Buformin Hydrochloride (1 in 125,000) as directed under mL of this solution is equivalent to 7 to 13z of that from 10
Ultraviolet-visible Spectrophotometry <2.24>, and compare mL of the standard solution.
the spectrum with the Reference Spectrum: both spectra System performance: When the procedure is run with 10
exhibit similar intensities of absorption at the same mL of the standard solution under the above operating con-
wavelengths. ditions, the number of theoretical plates and the symmetry
(3) Determin the infrared absorption spectrum of Bufor- factor of the peak of buformin are not less than 5000 and
min Hydrochloride as directed in the potassium chloride not more than 2.0, respectively.
disk method under Infrared Spectrophotometry <2.25>, and System repeatability: When the test is repeated 6 times
compare the spectrum with the Reference Spectrum: both with 10 mL of the standard solution under the above operat-
spectra exhibit similar intensities of absorption at the same ing conditions, the relative standard deviation of the peak
wave numbers. area of buformin is not more than 1.0z.
(4) A solution of Buformin Hydrochloride (1 in 20)
Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 3
responds to the Qualitative Tests <1.09> for chlorides.
hours).
Melting point <2.60> 175 – 1809
C
Residue on ignition <2.44> Not more than 0.1z (1 g).
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
Assay Weigh accurately about 0.15 g of Buformin
Buformin Hydrochloride according to Method 1, and per-
Hydrochloride, previously dried, dissolve in 50 mL of a mix-
form the test. Prepare the control solution with 2.0 mL of
ture of acetic anhydride and acetic acid (100) (7:3), and im-
Standard Lead Solution (not more than 20 ppm).
mediately titrate <2.50> with 0.1 mol/L perchloric acid VS
(2) Arsenic <1.11>—Prepare the test solution with 1.0 g
(potentiometric titration). Perform a blank determination in
of Buformin Hydrochloride according to Method 1, and
the same manner, and make any necessary correction.
perform the test (not more than 2 ppm).
(3) Related substances—Dissolve 0.10 g of Buformin Each mL of 0.1 mol/L perchloric acid VS
Hydrochloride in 200 mL of the mobile phase, and use this =9.684 mg of C6H15N5.HCl
solution as the sample solution. Pipet 1 mL of the sample
Containers and storage Containers—Tight containers.
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 standard
Add the following:
solution as directed under Liquid Chromatography <2.01>
according to the following conditions, and determine each
peak area of both solutions by the automatic integration Buformin Hydrochloride
method: the area of the peak other than buformin obtained Enteric-coated Tablets
from the sample solution is not larger than 1/5 times the
peak area of buformin from the standard solution. Further- ブホルミン塩酸塩腸溶錠
more, the total of the areas of all peaks other than the bufor-
min peak from the sample solution is not larger than 1/2 Buformin Hydrochloride Enteric-coated Tablets
times the peak area of buformin from the standard solution. contain not less than 93.0z and not more than 107.0
Operating conditions— z of the labeled amount of buformin hydrochloride
Detector: An ultraviolet absorption photometer (wave- (C6H15N5.HCl: 193.68).
length: 230 nm). Method of preparation Prepare as directed under Tablets,
Column: A stainless steel column 4.6 mm in inside di- with Buformin Hydrochloride.
ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di- Identification To a quantity of powdered Buformin
ameter). Hydrochloride Enteric-coated Tablets, equivalent to 0.1 g of
Column temperature: A constant temperature of about Buformin Hydrochloride according to the labeled amount,
359C. add 10 mL of water, shake well, and then filter. To 4 mL of
Mobile phase: A mixture of a solution of sodium the filtrate add 1 mL of a mixture of hydrogen peroxide TS,
perchlorate monohydrate in diluted phosphoric acid (1 in sodium pentacyanonitrosylferrate (III) TS and a solution of
1000) (7 in 250) and acetonitrile (7:1). sodium hydroxide (1 in 10) (2:1:1): the solution exhibits a
Flow rate: Adjust the flow rate so that the retention time red to red-purple color.
of buformin is about 6 minutes. Uniformity of dosage units <6.02> Perform the test accord-
Time span of measurement: About 2 times as long as the ing to the following method: it meets the requirement of the
retention time of buformin, beginning after the solvent Content uniformity test.
peak. To 1 tablet of Buformin Hydrochloride Enteric-coated
System suitability— Tablets add 5 mL of a mixture of ethanol (99.5) and acetone
Test for required detectability: Pipet 1 mL of the standard
1848 Official Monographs Supplement I, JP XV

(1:1), disperse the pellicle to smaller using ultrasonic waves, Column: A stainless steel column 4.6 mm in inside di-
add exactly 10 mL of the internal standard solution per 50 ameter and 15 cm in length, packed with octadecylsilanized
mg of buformin hydrochloride (C6H15N5.HCl), and then add silica gel for liquid chromatography (5 mm in particle di-
diluted acetonitrile (1 in 2) to make 13V/20 mL. Disintegrate ameter).
the tablet using ultrasonic waves, then shake for 20 minutes, Column temperature: A constant temperature of about
and add diluted acetonitrile (1 in 2) to make a solution, 359C.
volume V mL, containing about 0.5 mg of buformin Mobile phase: A mixture of a solution of sodium
hydrochloride (C6H15N5.HCl) per mL. Centrifuge this perchlorate in diluted phosphoric acid (1 in 1000) (7 in 500)
solution, pipet 1 mL of the supernatant liquid, and add the and acetonitrile (7:1).
mobile phase to make 50 mL. If necessary, filter this solu- Flow rate: Adjust the flow rate so that the retention time
tion through a membrane filter with a pore size not exceed- of buformin is about 6 minutes.
ing 0.5 mm, and use the filtrate as the sample solution. Then, System suitability—
proceed as directed in the Assay. System performance: When the procedure is run with 20
mL of the standard solution under the above operating con-
Amount (mg) of buformin hydrochloride (C6H15N5.HCl)
ditions, the number of theoretical plates and the symmetry
=WS×(QT/QS)×(V/50)
factor of the peak of buformin are not less than 3000 and
WS: Amount (mg) of buformin hydrochloride for assay not more than 2.0, respectively.
System repeatability: When the test is repeated 6 times
Internal standard solution—A solution of p-acetanisidide in
with 20 mL of the standard solution under the above operat-
diluted acetonitrile (1 in 2) (1 in 150).
ing conditions, the relative standard deviation of the peak
Dissolution <6.10> When the tests are performed at 50 area of buformin is not more than 2.0z.
revolutions per minute according to the Paddle method,
Assay Add 20 mL of a mixture of ethanol (99.5) and
using 900 mL each of 1st fluid for dissolution test and 2nd
acetone (1:1) to an amount of Buformin Hydrochloride
fluid for dissolution test as the dissolution medium, the dis-
Enteric-coated Tablets equivalent to 0.5 g of buformin
solution rate in 120 minutes of Buformin Hydrochloride En-
hydrochloride (C6H15N5.HCl), disperse the pellicles to
teric-coated Tablets using the 1st fluid is not more than 5z,
smaller using ultrasonic waves, and then add 100 mL of
and that in 90 minutes of Buformin Hydrochloride Enteric-
diluted acetonitrile (1 in 2). Disintegrate the tablets with the
coated Tablets using the 2nd fluid is not less than 80z.
aid of ultrasonic waves, shake for 20 minutes, and then add
Start the test with 1 tablet of Buformin Hydrochloride En-
diluted acetonitrile (1 in 2) to make exactly 200 mL. Cen-
teric-coated Tablets, withdraw not less than 20 mL of the
trifuge this solution, pipet 10 mL of the supernatant liquid,
medium at the specified minute after starting the test, and
add exactly 5 mL of the internal standard solution, and then
filter through a membrane filter with a pore size not exceed-
add diluted acetonitrile (1 in 2) to make 50 mL. Pipet 1 mL
ing 0.5 mm. Discard the first 10 mL of the filtrate, pipet V
of this solution, and add the mobile phase to make 50 mL. If
mL of the subsequent filtrate, add the relevant dissolution
necessary, filter this solution through a membrane filter with
medium to make exactly V? mL so that each mL contains
a pore size not exceeding 0.5 mm, and use the filtrate as the
about 56 mg of buformin hydrochloride (C6H15N5.HCl) ac-
sample solution. Separately, weigh accurately about 25 mL
cording to the labeled amount, and use this solution as the
of buformin hydrochloride for assay, previously dried at
sample solution. Separately, weigh accurately about 28 mg
1059 C for 3 hours, dissolve in an adequate amount of dilut-
of buformin hydrochloride for assay, previously dried at
ed acetonitrile (1 in 2), add exactly 5 mL of the internal stan-
1059 C for 3 hours, and dissolve in the relevant dissolution
dard solution, and then add diluted acetonitrile (1 in 2) to
medium to make exactly 100 mL. Pipet 4 mL of this solu-
make 50 mL. To 1 mL of this solution add the mobile phase
tion, add the relevant dissolution medium to make exactly 20
to make 50 mL, and use this solution as the standard solu-
mL, and use this solution as the standard solution. Perform
tion. Perform the test with 10 mL each of the sample solution
the test with exactly 20 mL each of the sample solution and
and standard solution as directed under Liquid Chro-
standard solution as directed under Liquid Chromatography
matography <2.01> according to the following conditions,
<2.01> according to the following conditions, and determine
and calculate the ratios, QT and QS, of the peak area of
the buformin peak areas, AT and AS, of both solutions.
buformin to that of the internal standard.
Dissolution rate (z) with respect to the labeled amount of
Amount (mg) of buformin hydrochloride (C6H15N5.HCl)
buformin hydrochloride (C6H15N5.HCl)
=WS×(QT/QS)×20
=WS×(AT/AS)×(V?/V)×(1/C)×180
WS: Amount (mg) of buformin hydrochloride for assay
WS: Amount (mg) of buformin hydrochloride for assay
C: Labeled amount (mg) of buformin hydrochloride Internal standard solution—A solution of p-acetanisidide in
(C6H15N5.HCl) in 1 tablet diluted acetonitrile (1 in 2) (1 in 150).
Operating conditions—
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Detector: An ultraviolet absorption photometer (wave-
length: 233 nm).
length: 230 nm).
Column: A stainless steel column 4.6 mm in inside di-
Supplement I, JP XV Official Monographs 1849

ameter and 15 cm in length, packed with octadecylsilanized 100 mL, and use this solution as the standard solution. De-
silica gel for liquid chromatography (5 mm in particle di- termine the absorbances, AT and AS, of the sample solution
ameter). and standard solution at 233 nm as directed under Ultrav-
Column temperature: A constant temperature of about iolet-visible Spectrophotometry <2.24>.
359C.
Amount (mg) of buformin hydrochloride (C6H15N5.HCl)
Mobile phase: A mixture of a solution of sodium
=WS×(AT/AS)×(2/V)
perchlorate (7 in 250) and acetonitrile (7:1).
Flow rate: Adjust the flow rate so that the retention time WS: Amount (mg) of buformin hydrochloride for assay
of buformin is about 7 minutes.
Dissolution <6.10> When the test is performed at 50 revolu-
System suitability—
tions per minute according to the Paddle method, using 900
System performance: When the procedure is run with 10
mL of water as the dissolution medium, the dissolution rate
mL of the standard solution under the above operating con-
in 15 minutes of Buformin Hydrochloride Tablets is not less
ditions, buformin and the internal standard are eluted in this
than 80z.
order with the resolution between these peaks being not less
Start the test with 1 tablet of Buformin Hydrochloride
than 5.
Tablets, withdraw not less than 20 mL of the medium at the
System repeatability: When the test is repeated 6 times
specified minute after starting the test, and filter through a
with 10 mL of the standard solution under the above operat-
membrane filter with a pore size not exceeding 0.5 mm. Dis-
ing conditions, the relative standard deviation of the ratio of
card the first 10 mL of the filtrate, pipet V mL of the subse-
the peak area of buformin to that of the internal standard is
quent filtrate, and add water to make exactly V? mL so that
not more than 1.0z.
each mL contains about 5.6 mg of buformin hydrochloride
Containers and storage Containers—Well-closed contain- (C6H15N5.HCl) according to the labeled amount, and use
ers. this solution as the sample solution. Separately, weigh ac-
curately about 28 mg of buformin hydrochloride for assay,
previously dried at 1059 C for 3 hours, and dissolve in water
Add the following: to make exactly 100 mL. Pipet 2 mL of this solution, add
water to make exactly 100 mL, and use this solution as the
Buformin Hydrochloride Tablets standard solution. Perform the test with the sample solution
and standard solution as directed under Ultraviolet-visible
ブホルミン塩酸塩錠 Spectrophotometry <2.24>, and determine the absorbances,
AT and AS, at 233 nm.
Buformin Hydrochloride Tablets contain not less
Dissolution rate (z) with respect to the labeled amount of
than 95.0z and not more than 105.0z of the labeled
buformin hydrochloride (C6H15N5.HCl)
amount of buformin hydrochloride (C6H15N5.HCl:
=WS×(AT/AS)×(V?/V)×(1/C)×18
193.68).
WS: Amount (mg) of buformin hydrochloride for assay
Method of preparation Prepare as directed under Tablets,
C: Labeled amount (mg) of buformin hydrochloride
with Buformin Hydrochloride.
(C6H15N5.HCl) in 1 tablet
Identification To a quantity of powdered Buformin
Assay Weigh accurately not less than 20 Buformin
Hydrochloride Tablets, equivalent to 1 g of Buformin
Hydrochloride Tablets, and powder. Weigh accurately a
Hydrochloride according to the labeled amount, add 100
portion of the powder, equivalent to about 60 mg of bufor-
mL of water, shake well, and then filter. To 4 mL of the
min hydrochloride (C6H15N5.HCl), add water to make
filtrate add 1 mL of dilute sodium pentacyanonitrosylferrate
exactly 200 mL, and treat with ultrasonic waves for 5
(III)-potassium hexacyanoferrate (III) TS: the solution ex-
minutes. Take 40 mL of this solution, centrifuge, pipet 2 mL
hibits a red-brown color.
of the supernatant liquid, add water to make exactly 100
Uniformity of dosage units <6.02> Perform the test accord- mL, and use this solution as the sample solution. Separately,
ing to the following method: it meets the requirement of the weigh accurately about 60 mg of buformin hydrochloride
Content uniformity test. for assay, previously dried at 1059 C for 3 hours, and dis-
Take 1 tablet of Buformin Hydrochloride Tablets, add solve in water to make exactly 200 mL. Pipet 2 mL of this
water to make exactly 200 mL, and then treat with ultrasonic solution, add water to make exactly 100 mL, and use this so-
waves for 5 minutes. Take 40 mL of this solution and cen- lution as the standard solution. Perform the test with the
trifuge. Pipet V mL of the supernatant liquid equivalent to sample solution and standard solution as directed under
about 0.5 mg of buformin hydrochloride (C6H15N5.HCl), Ultraviolet-visible Spectrophotometry <2.24>, and determine
add water to make exactly 100 mL, and use this solution as the absorbances, AT and AS, at 233 nm.
the sample solution. Separately, weigh accurately about 50
Amount (mg) of buformin hydrochloride (C6H15N5.HCl)
mg of buformin hydrochloride for assay, previously dried at
= W S × ( A T/ A S )
1059 C for 3 hours, and dissolve in water to make exactly 200
mL. Pipet 2 mL of this solution, add water to make exactly WS: Amount (mg) of buformin hydrochloride for assay
1850 Official Monographs Supplement I, JP XV

Containers and storage Containers—Well-closed contain- phine Hydrochloride in 20 mL of the mobile phase, and use
ers. this solution as the sample solution. Pipet 1 mL of the sam-
ple solution, add the mobile phase to make exactly 100 mL,
and use this solution as the standard solution. Perform the
Add the following: test with exactly 20 mL each of the sample solution and stan-
dard solution as directed under Liquid Chromatography
Buprenorphine Hydrochloride <2.01> according to the following conditions. Determine
each peak area of both solutions by the automatic integra-
ブプレノルフィン塩酸塩 tion method: the area of each peak other than buprenor-
phine obtained from the sample solution is not larger than
1/4 times the peak area of buprenorphine from the standard
solution. Furthermore, the total area of the peaks other than
buprenorphine from the sample solution is not larger than
13/20 times the peak area of buprenorphine from the stan-
dard solution.
Operating conditions—
C29H41NO4.HCl: 504.10
Detector: An ultraviolet absorption photometer (wave-
(2S)-2-[(5R,6R,7R,14S)-17-(Cyclopropylmethyl)-4,5-
length: 288 nm).
epoxy-3-hydroxy-6-methoxy-6,14-ethanomorphinan-7-yl]-
Column: A stainless steel column 4.6 mm in inside di-
3,3-dimethylbutan-2-ol monohydrochloride [53152-21-9]
ameter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Buprenorphine Hydrochloride, when dried, con-
ameter).
tains not less than 98.5z and not more than 101.0z
Column temperature: A constant temperature of about
of C29H41NO4.HCl.
409C.
Description Buprenorphine Hydrochloride occurs as white Mobile phase: A mixture of methanol, ammonium acetate
to yellowish white, crystals or a crystalline powder. solution (1 in 100), and acetic acid (100) (6000:1000:1).
It is freely soluble in methanol and in acetic acid (100), Flow rate: Adjust the flow rate so that the retention time
and sparingly soluble in water and in ethanol (99.5). of buprenorphine is about 17 minutes.
Melting point: about 2689 C (with decomposition). Time span of measurement: About 2.5 times as long as the
retention time of buprenorphine, beginning after the solvent
Identification (1) Determine the absorption spectrum of
peak.
a solution of Buprenorphine Hydrochloride (1 in 5000) as
directed under Ultraviolet-visible Spectrophotometry <2.24>,
System suitability—
Test for required detectability: Pipet 5 mL of the standard
and compare the spectrum with the Reference Spectrum:
solution, and add the mobile phase to make exactly 50 mL.
both spectra exhibit similar intensities of absorption at the
Confirm that the peak area of buprenorphine obtained from
same wavelengths.
20 mL of this solution is equivalent to 7 to 13z of that from
(2) Determine the infrared absorption spectrum of
20 mL of the standard solution.
Buprenorphine Hydrochloride as directed in the potassium
System performance: When the procedure is run with 20
chloride disk method under Infrared Spectrophotometry
<2.25>, and compare the spectrum with the Reference Spec- mL of the standard solution under the above operating
conditions, the number of theoretical plates and the symmet-
trum: both spectra exhibit similar intensities of absorption at
ry factor of the peak of buprenorphine are not less than 6500
the same wave numbers.
and not more than 1.2, respectively.
(3) A solution of Buprenorphine Hydrochloride (1 in
System repeatability: When the test is repeated 6 times
100) responds to the Qualitative Tests <1.09> for chloride.
with 20 mL of the standard solution under the above operat-
Optical rotation <2.49> [a]20
D : -92 – -989(after drying, ing conditions, the relative standard deviation of the peak
0.4 g, methanol, 20 mL, 100 mm). area of buprenorphine is not more than 2.0z.
pH <2.54> The pH of a solution prepared by dissolving 1.0 Loss on drying <2.41> Not more than 1.0z (1 g, 1159
C, 3
g of Buprenorphine Hydrochloride in 200 mL of water is hours).
between 4.0 and 6.0.
Residue on ignition <2.44> Not more than 0.1z (1 g).
Purity (1) Clarity and color of solution—A solution ob-
Assay Weigh accurately about 0.5 g of Buprenorphine
tained by dissolving 0.1 g of Buprenorphine Hydrochloride
Hydrochloride, previously dried, dissolve in 5 mL of acetic
in 10 mL of water is clear and colorless.
acid (100), add 50 mL of acetic anhydride, and titrate <2.50>
(2) Heavy metals <1.07>—Proceed with 1.0 g of
with 0.1 mol/L perchloric acid VS (potentiometric titra-
Buprenorphine Hydrochloride according to Method 4, and
tion). Perform a blank determination in the same manner,
perform the test. Prepare the control solution with 1.0 mL
and make any necessary correction.
of Standard Lead Solution (not more than 10 ppm).
(3) Related substances—Dissolve 0.10 g of Buprenor-
Supplement I, JP XV Official Monographs 1851

Each mL of 0.1 mol/L perchloric acid VS Change the Purity to read:


=50.41 mg of C29H41NO4.HCl
Purity (1) Clarity and color of solution—To 1.25 g of
Containers and storage Containers—Well-closed contain- Calcium Folinate add 50 mL of freshly boiled and cooled
ers. water, and warm to 409C, if necessary, to dissolve: the solu-
tion is clear, and the absorbance at 420 nm of it, determined
as directed under Ultraviolet-visible Spectrophotometry
Calcium Chloride Injection <2.24>, is not more than 0.25.
(2) Heavy metals <1.07>—Proceed with 0.40 g of Calci-
塩化カルシウム注射液 um Folinate according to Method 2, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead
Add the following next to Extractable volume: Solution (not more than 50 ppm).
(3) Related substances—Dissolve 10 mg of Calcium
Foreign insoluble matter <6.06> Perform the test according
Folinate in 25 mL of water, and use this solution as the
to Method 1: it meets the requirement.
sample solution. Pipet 2 mL of the sample solution, add
Insoluble particulate matter <6.07> It meets the require- water to make exactly 200 mL, and use this solution as the
ment. standard solution. Perform the test with exactly 20 mL each
of the sample solution and standard solution as directed
Sterility <4.06> Perform the test according to the Mem-
under Liquid Chromatography <2.01> according to the
brane filtration method: it meets the requirement.
following conditions, and determine each peak area by the
automatic integration method: the area of the peak other
than folinate obtained from the sample solution is not larger
Calcium Folinate than the peak area of folinate from the standard solution,
ホリナートカルシウム and the total area of the peaks other than the peak of
folinate from the sample solution is not larger than 5 times
the peak area of folinate from the standard solution.
Change the Description and the Identification to
read: Operating conditions—
Detector, column, column temperature, mobile phase,
Description Calcium Folinate occurs as a white to light yel- and flow rate: Proceed as directed in the operating condi-
low, crystalline powder. tions in the Assay.
It is sparingly soluble in water, and practically insoluble in Time span of measurement: About 2.5 times as long as the
methanol and in ethanol (99.5). retention time of folinate, beginning after the solvent peak.
Identification (1) Determine the absorption spectrum of System suitability—
a solution of Calcium Folinate (1 in 100,000) as directed Test for required detectability: Pipet 5 mL of the standard
under Ultraviolet-visible Spectrophotometry <2.24>, and solution, and add water to make exactly 50 mL. Confirm
compare the spectrum with the Reference Spectrum or the that the peak area of folinate obtained from 20 mL of this so-
spectrum of a solution of Calcium Folinate Reference Stan- lution is equivalent to 7 to 13z of that from 20 mL of the
dard prepared in the same manner as the sample solution: standard solution.
both spectra exhibit similar intensities of absorption at the System performance: Proceed as directed in the system
same wavelengths. suitability in the Assay.
(2) Determine the infrared absorption spectrum of System repeatability: When the test is repeated 6 times
Calcium Folinate as directed in the potassium bromide disk with 20 mL of the standard solution under the above operat-
method under Infrared Spectrophotometry <2.25>, and com- ing conditions, the relative standard deviation of the peak
pare the spectrum with the Reference Spectrum: both spec- area of folinate is not more than 2.0z.
tra exhibit similar intensities of absorption at the same wave
numbers. Change the Water to read:
(3) A solution of Calcium Folinate (1 in 100) responds to Water <2.48> Not less than 7.0z and not more than 17.0z
the Qualitative Tests <1.09> (2) and (3) for calcium salt. (0.2 g, volumetric titration, direct titration).

Add the following next to Identification: Change the Assay to read:


Optical rotation <2.49> [a]20
D:
+14 – +199(0.1 g calculated Assay Weigh accurately about 10 mg each of Calcium
on the anhydrous basis, water, 10 mL, 100 mm). Folinate and Calcium Folinate Reference Standard
pH <2.54> To 1.25 g of Calcium Folinate add 50 mL of (separately determine the water <2.48> in the same manner as
freshly boiled and cooled water, and warm to 409 C, if neces- Calcium Folinate), dissolve in water to make them exactly 25
sary, to dissolve: the pH of this solution is between 6.8 and mL. Pipet 5 mL each of these solutions, add the mobile
8.0. phase to make exactly 25 mL, and use these solutions as the
sample solution and the standard solution, respectively. Per-
1852 Official Monographs Supplement I, JP XV

form the test with exactly 20 mL each of the sample solution Add the following:
and standard solution as directed under Liquid Chro-
matography <2.01> according to the following conditions, Cefadroxil Capsules
and determine the peak areas, AT and AS, of folinate of both
solutions. セファドロキシルカプセル

Amount (mg) of C20H21CaN7O7


Cefadroxil Capsules contain not less than 95.0z
= W S × ( A T/ A S )
and not more than 105.0z of the labeled amount of
WS: Amount (mg) of Calcium Folinate Reference Stan- cefadroxil (C16H17N3O5S: 363.39).
dard, calculated on the anhydrous basis
Method of preparation Prepare as directed under Cap-
Operating conditions— sules, with Cefadroxil.
Detector: An ultraviolet absorption photometer (wave-
Identification Dissolve the contents of Cefadroxil Cap-
length: 254 nm).
sules, equivalent to 10 mg (potency) of Cefadroxil according
Column: A stainless steel column 4.6 mm in inside di-
to the labeled amount, in 500 mL of water, and filter. Deter-
ameter and 15 cm in length, packed with octadecylsilanized
mine the absorption spectrum of the filtrate as directed un-
silica gel for liquid chromatography (5 mm in particle di-
der Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
ameter).
maxima between 228 nm and 232 nm, and between 261 nm
Column temperature: A constant temperature of about
and 265 nm.
459C.
Mobile phase: A mixture of disodium hydrogen phos- Water <2.48> Not more than 7.0z (0.15 g, volumetric
phate dodecahydrate solution (287 in 100,000), methanol titration, direct titration).
and tetrabutylammonium hydroxide TS (385:110:4), adjust-
Uniformity of dosage units <6.02> Perform the test accord-
ed to pH7.5 with phosphoric acid.
ing to the following method: it meets the requirement of the
Flow rate: Adjust the flow rate so that the retention time
Content uniformity test.
of folinate is about 10 minutes.
Place 1 capsule of Cefadroxil Capsules in 300 mL of
System suitability—
water, disperse with the aid of ultrasonic waves, shake for 30
System performance: Dissolve 10 mg each of Calcium
minutes, and add water to make exactly 500 mL. Pipet 5 mL
Folinate and folic acid in 100 mL of the mobile phase. When
of this solution, and add water to make exactly V mL so that
the procedure is run with 20 mL of this solution under the
each mL contains about 0.1 mg (potency) of Cefadroxil.
above operating conditions, folinate and folic acid are eluted
Filter the solution, discard the first 10 mL of the filtrate, and
in this order with the resolution between these peaks being
use the subsequent filtrate as the sample solution. Separate-
not less than 10.
ly, weigh accurately about 20 mg (potency) of Cefadroxil
System repeatability: When the test is repeated 6 times
Reference Standard, dissolve in water to make exactly 200
with 20 mL of the standard solution under the above operat-
mL, and use this solution as the standard solution. Then,
ing conditions, the relative standard deviation of the peak
proceed as directed in the Assay under Cefadroxil.
area of folinate is not more than 1.0z.
Amount [mg (potency)] of cefadroxil (C16H17N3O5S)
=WS×(AT/AS)×(V/2)
Camostat Mesilate WS: Amount [mg (potency)] of Cefadroxil Reference
Standard
カモスタットメシル酸塩
Dissolution <6.10> When the test is performed at 50 revolu-
Change the Identification (3) to read: tions per minute according to the Paddle method, using 900
mL of 0.05 mol/L acetic acid-sodium acetate buffer solu-
Identification (3) A 0.1 g portion of Camostat Mesilate
tion, pH 4.0 as the dissolution medium, the dissolution rate
responds to the Qualitative Tests <1.09> (1) for mesilate.
in 90 minutes of Cefadroxil Capsules is not less than 80z.
Start the test with 1 capsule of Cefadroxil Capsules,
withdraw not less than 20 mL of the medium at the specified
minute after starting the test, and filter through a membrane
filter with a pore size not exceeding 0.45 mm. Discard the
first 10 mL of the filtrate, pipet V mL of the subsequent
filtrate, add water to make exactly V? mL so that each mL
contains about 22 mg (potency) of Cefadroxil according to
the labeled amount, and use this solution as the sample solu-
tion. Separately, weigh accurately about 22 mg (potency) of
Cefadroxil Reference Standard, and add water to make ex-
actly 100 mL. Pipet 5 mL of this solution, add water to
Supplement I, JP XV Official Monographs 1853

make exactly 50 mL, and use this solution as the standard Uniformity of dosage units <6.02> The syrup in single-unit
solution. Determine the absorbances, AT and AS, at 263 nm container meets the requirement of the Mass variation test.
of the sample solution and standard solution as directed un-
Dissolution <6.10> When the test is performed at 50 revolu-
der Ultraviolet-visible Spectrophotometry <2.24>, using
tions per minute according to the Paddle method (put the
water as the blank.
sample in the dissolution medium so that it disperses), using
Dissolution rate (z) with respect to the labeled amount of 900 mL of water as the dissolution medium, the dissolution
cefadroxil (C16H17N3O5S) rate in 15 minutes of Cefadroxil for Syrup is not less than 85
=WS×(AT/AS)×(V?/V)×(1/C)×90 z.
Start the test with accurately weighed amount of
WS: Amount [mg (potency)] of Cefadroxil Reference
Cefadroxil for Syrup, equivalent to about 0.1 g (potency) of
Standard
Cefadroxil according to the labeled amount, withdraw not
C: Labeled amount [mg (potency)] of cefadroxil in 1 cap-
less than 20 mL of the medium at the specified minute after
sule
starting the test, and filter through a membrane filter with a
Assay Take out the contents of 20 Cefadroxil Capsules, pore size not exceeding 0.45 mm. Discard the first 10 mL of
and combine. Weigh accurately the mass of the combined the filtrate, pipet 4 mL of the subsequent filtrate, add water
contents, and powder. Weigh accurately a portion of the to make exactly 20 mL, and use this solution as the sample
powder, equivalent to about 50 mg (potency) of Cefadroxil, solution. Separately, weigh accurately about 22 mg (poten-
add 300 mL of water, shake for 30 minutes, then add water cy) of Cefadroxil Reference Standard, and dissolve in water
to make exactly 500 mL, and filter. Discard the first 10 mL to make exactly 100 mL. Pipet 5 mL of this solution, add
of the filtrate, and use the subsequent filtrate as the sample water to make exactly 50 mL, and use this solution as the
solution. Separately, weigh accurately an amount of standard solution. Determine the absorbances, AT and AS,
Cefadroxil Reference Standard, equivalent to about 20 mg at 263 nm of the sample solution and standard solution as
(potency), dissolve in water to make exactly 200 mL, and use directed under Ultraviolet-visible Spectrophotometry <2.24>.
this solution as the standard solution. Then, proceed as
Dissolution rate (z) with respect to the labeled amount of
directed in the Assay under Cefadroxil.
cefadroxil (C16H17N3O5S)
Amount [mg (potency)] of cefadroxil (C16H17N3O5S) =(WS/WT)×(AT/AS)×(1/C)×450
=WS×(AT/AS)×(5/2)
WS: Amount [mg (potency)] of Cefadroxil Reference
WS: Amount [mg (potency)] of Cefadroxil Reference Standard
Standard WT: Amount (g) of sample
C: Labeled amount [mg (potency)] of cefadroxil in 1 g
Containers and storage Containers—Tight containers.
Assay Weigh accurately an amount of powdered
Cefadroxil for Syrup, equivalent to about 50 mg (potency)
Add the following: of Cefadroxil, dissolve in water to make exactly 500 mL, and
use this solution as the sample solution. Separately, weigh
Cefadroxil for Syrup accurately an amount of Cefadroxil Reference Standard,
equivalent to about 20 mg (potency), dissolve in water to
シロップ用セファドロキシル make exactly 200 mL, and use this solution as the standard
solution. Then, proceed as directed in the Assay under
Cefadroxil for Syrup is a preparation for syrup Cefadroxil.
which is suspended before use.
Amount [mg (potency)] of cefadroxil (C16H17N3O5S)
It contains not less than 95.0z and not more than
=WS×(AT/AS)×(5/2)
110.0z of the labeled amount of cefadroxil
(C16H17N3O5S: 363.39). WS: Amount [mg (potency)] of Cefadroxil Reference
Standard
Method of preparation Prepare as directed under Syrups,
with Cefadroxil. Containers and storage Containers—Tight containers.
Identification Dissolve an amount of Cefadroxil for
Syrup, equivalent to 10 mg (potency) of Cefadroxil accord-
ing to the labeled amount, in 500 mL of water, and deter-
mine the absorption spectrum of the solution as directed un-
der Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
maxima between 228 nm and 232 nm, and between 261 nm
and 265 nm.

Water <2.48> Not more than 3.0z (0.5 g, volumetric titra-


tion, direct titration).
1854 Official Monographs Supplement I, JP XV

Purity (1) Clarity and color of solution—Conduct this


Cefalotin Sodium procedure within 10 minutes after the preparation of the
solutions. A solution prepared by dissolving an amount of
セファロチンナトリウム Cefazolin Sodium for Injection, equivalent to 1.0 g (poten-
cy) of Cefazolin Sodium according to the labeled amount, in
Change the origin/limits of content to read: 10 mL of water is clear, and the absorbance of this solution
at 400 nm, determined as directed under Ultraviolet-visible
Cefalotin Sodium contains not less than 920 mg Spectrophotometry <2.24>, is not more than 0.35.
(potency) and not more than 980 mg (potency) per mg, (2) Related substances—Dissolve an amount of Cefazo-
calculated on the anhydrous basis. The potency of lin Sodium for Injection, equivalent to 0.10 g (potency) of
Cefalotin Sodium is expressed as mass (potency) of Cefazolin Sodium according to the labeled amount, in 20
cefalotin (C16H16N2O6S2: 396.44). mL of 0.1 mol/L phosphate buffer solution, pH 7.0, and
use this solution as the sample solution. Prepare the sample
solution before use. Perform the test with 5 mL of the sample
Cefatrizine Propylene Glycolate solution as directed under Liquid Chromatography <2.01>
according to the following conditions, and determine each
セファトリジンプロピレングリコール peak area by the automatic integration method. Calculate
the amount of each peak by the area percentage method:
Change the origin/limits of content to read: each area of the peaks other than cefazolin is not more than
1.5z. Furthermore the total area of the peaks other than
Cefatrizine Propylene Glycolate contains not less cefazolin is not more than 2.5z. For these calculations, use
than 816 mg (potency) and not more than 876 mg the area of the peak, having the relative retention time of
(potency) per mg, calculated on the anhydrous basis. about 0.2 with respect to cefazolin, after multiplying by the
The potency of Cefatrizine Propylene Glycolate is relative response factor, 1.43.
expressed as mass (potency) of cefatrizine Operating conditions—
(C18H18N6O5S2: 462.50). Detector, column, column temperature, mobile phase,
and flow rate: Proceed as directed in the operating condi-
tions in the Assay under Cefazolin Sodium.
Add the following: Time span of measurement: About 3 times as long as the
retention time of cefazolin, beginning after the solvent peak.
Cefazolin Sodium for Injection System suitability—
Test for required detectability: Pipet 8 mL of the sample
注射用セファゾリンナトリウム solution, add 0.1 mol/L phosphate buffer solution, pH 7.0
to make exactly 50 mL, and use this solution as the solution
Cefazolin Sodium for Injection is a preparation for for system suitability test. Pipet 1 mL of the solution for sys-
injection which is dissolved before use. tem suitability test, add 0.1 mol/L phosphate buffer solu-
It contains not less than 90.0z and not more tion, pH 7.0 to make exactly 20 mL. Confirm that the peak
than 110.0z of the labeled amount of cefazolin area of cefazolin obtained from 5 mL of this solution is
(C14H14N8O4S3: 454.51). equivalent to 3 to 7z of that from 5 mL of the solution for
Method of preparation Prepare as directed under Injec- system suitability test.
tions, with Cefazolin Sodium. System performance: Proceed as directed in the system
suitability in the Assay under Cefazolin Sodium.
Description Cefazolin Sodium for Injection occurs as System repeatability: When the test is repeated 6 times
white to light yellowish white crystals or crystalline powder with 5 mL of the solution for system suitability test under the
or masses. above operating conditions, the relative standard deviation
Identification (1) Determine the absorption spectrum of of the peak area of cefazolin is not more than 1.0z.
a solution of Cefazolin Sodium for Injection (1 in 50,000) as Water <2.48> Not more than 3.0z (0.5 g, volumetric titra-
directed under Ultraviolet-visible Spectrophotometry <2.24>: tion, direct titration). Use a mixture of formamide for Karl
it exhibits a maximum between 270 nm and 274 nm. Fischer method and methanol for Karl Fischer method (2:1)
(2) Cefazolin Sodium for Injection responds to the instead of methanol for Karl Fischer method.
Qualitative Tests <1.09> (1) for chloride.
Bacterial endotoxins <4.01> Less than 0.05 EU/mg (poten-
Osmotic pressure ratio Being specified separately. cy).
pH <2.54> The pH of a solution prepared by dissolving an Uniformity of dosage units <6.02> It meets the requirement
amount of Cefazolin Sodium for Injection, equivalent to 1.0 of the Mass variation test.
g (potency) of Cefazolin Sodium according to the labeled
amount, in 10 mL of water is 4.5 to 6.5. Foreign insoluble matter <6.06> Perform the test according
to Method 2: it meets the requirement.
Supplement I, JP XV Official Monographs 1855

Insoluble particulate matter <6.07> It meets the require- pH <2.54> Take an amount of Cefmetazole Sodium for In-
ment. jection equivalent to 1.0 g (potency) of Cefmetazole Sodium
according to the labeled amount, and dissolve in 10 mL of
Sterility <4.06> Perform the test according to the Mem-
water: the pH of the solution is 4.2 to 6.2.
brane filtration method: it meets the requirement.
Purity (1) Clarity and color of solution—Dissolve an
Assay Weigh accurately the mass of the contents of not
amount of Cefmetazole Sodium for Injection, equivalent to
less than 10 containers of Cefazolin Sodium for Injection.
1.0 g (potency) of Cefmetazole Sodium according to the
Weigh accurately an amount of the contents, equivalent to
labeled amount, in 10 mL of water: the solution is clear and
about 50 mg (potency) of Cefazolin Sodium, dissolve in the
the color is not darker than the following control solution.
internal standard solution to make exactly 50 mL, and use
Control solution: Pipet 5 mL of Iron (III) Chloride
this solution as the sample solution. Separately, weigh ac-
Colorimetric Stock Solution and 0.5 mL of Cobalt (II) Chlo-
curately an amount of Cefazolin Reference Standard,
ride Colorimetric Stock Solution, and add water to make
equivalent to about 50 mg (potency), dissolve in the internal
exactly 50 mL. Pipet 15 mL of this solution, and add water
standard solution to make exactly 50 mL, and use this solu-
to make exactly 20 mL.
tion as the standard solution. Then, proceed as directed in
(2) Related substances—Proceed as directed in the Puri-
the Assay under Cefazolin Sodium.
ty (4) under Cefmetazole Sodium.
Amount [mg (potency)] of cefazolin (C14H14N8O4S3)
Bacterial endotoxins <4.01> Less than 0.06 EU/mg (poten-
= W S × ( Q T/ Q S )
cy).
WS: Amount [mg (potency)] of Cefazolin Reference Stan-
Uniformity of dosage units <6.02> It meets the requirement
dard
of the Mass variation test.
Internal standard solution—A solution of p-acetanisidide in
Foreign particulate matter <6.06> Perform the test accord-
0.1 mol/L phosphate buffer solution, pH 7.0 (11 in 20,000).
ing to Method 2: it meets the requirement.
Containers and storage Containers—hermetic containers.
Insoluble particulate matter <6.07> It meets the require-
Plastic containers for aqueous injections may be used.
ment.

Sterility <4.06> Perform the test according to the Mem-


Add the following: brane filtration method: it meets the requirement.

Assay Take 10 containers of Cefmetazole Sodium for In-


Cefmetazole Sodium for Injection jection, dissolve the contents of each in the mobile phase,
rinse each of the containers with the mobile phase, combine
注射用セフメタゾールナトリウム
the rinse with the respective previous solution, and add the
mobile phase to make exactly 500 mL. Take exactly a
Cefmetazole Sodium for Injection is a preparation
volume of this solution equivalent to about 0.2 g (potency)
for injection which is dissolved before use.
of Cefmetazole Sodium, and add the mobile phase to make
It contains not less than 90.0z and not more than
exactly 100 mL. Pipet 1 mL of this solution, add exactly 10
110.0z of the labeled amount of cefmetazole
mL of the internal standard solution, and use this solution as
(C15H17N7O5S3: 471.53).
the sample solution. Separately, weigh accurately an amount
Method of preparation Prepare as directed under Injec- of Cefmetazole Reference Standard, equivalent to about 50
tions, with Cefmetazole Sodium. mg (potency), and dissolve in the mobile phase to make ex-
actly 25 mL. Pipet 1 mL of this solution, add exactly 10 mL
Description Cefmetazole Sodium for Injection is a white to
of the internal standard solution, and use this solution as the
light yellow powder or masses.
standard solution. Then, proceed as directed in the Assay
It is hygroscopic.
under Cefmetazole Sodium.
Identification (1) Determine the absorption spectrum of
Amount [mg (potency)] of cefmetazole (C15H17N7O5S3)
a solution of Cefmetazole Sodium for Injection (1 in 40,000)
=WS×(QT/QS)×4
as directed under Ultraviolet-visible Spectrophotometry
<2.24>, and compare the spectrum with the Reference Spec- WS: Amount [mg (potency)] of Cefmetazole Reference
trum: both spectra exhibit similar intensities of absorption at Standard
the same wavelengths.
Internal standard solution—A solution of methyl para-
(2) Determine the infrared absorption spectrum of Cef-
hydroxybenzoate in the mobile phase (1 in 10,000).
metazole Sodium for Injection as directed in the potassium
bromide disk method under Infrared Spectrophotometry Containers and storage Containers—Hermetic containers.
<2.25>, and compare the spectrum with the Reference Spec- Plastic containers for aqueous injections may be used.
trum: both spectra exhibit similar intensities of absorption at
the same wave numbers.
1856 Official Monographs Supplement I, JP XV

Add the following: buffer solution, pH 7.0, to make 50 mL, and use this solu-
tion as the sample solution. Separately, weigh accurately an
Ceftazidime for Injection amount of Ceftazidime Reference Standard, equivalent to
about 25 mg (potency), and dissolve in 0.05 mol/L phos-
注射用セフタジジム phate buffer solution, pH 7.0, to make exactly 25 mL. Pipet
10 mL of this solution, add exactly 5 mL of the internal stan-
Ceftazidime for Injection is a preparation for injec- dard solution, then add 0.05 mol/L phosphate buffer solu-
tion which is dissolved before use. tion, pH 7.0, to make 50 mL, and use this solution as the
It contains not less than 93.0z and not more than standard solution. Then, proceed as directed in the Assay
107.0z of the labeled amount of ceftazidime under Ceftazidime Hydrate.
(C22H22N6O7S2: 546.58).
Amount [mg (potency)] of ceftazidime (C22H22N6O7S2)
Method of preparation Prepare as directed under Injec- =WS×(QT/QS)×10
tions, with Ceftazidime Hydrate.
WS: Amount [mg(potency)] of Ceftazidime Reference
Description Ceftazidime for Injection is a white to pale Standard
yellowish white powder.
Internal standard solution—A solution of dimedon in 0.05
Identification Determine the absorption spectrum of a so- mol/L phosphate buffer solution, pH 7.0 (11 in 10,000).
lution of Ceftazidime for Injection (1 in 100,000) in phos-
Containers and storage Containers—Hermetic containers.
phate buffer solution, pH 6.0, as directed under Ultraviolet-
Storage—Light-resistant.
visible Spectrophotometry <2.24>: it exhibits a maximum be-
tween 255 nm and 259 nm.

pH <2.54> Dissolve an amount of Ceftazidime for Injec- Add the following:


tion, equivalent to 1.0 g (potency) of Ceftazidime Hydrate
according to the labeled amount, in 10 mL of water: the pH Cetirizine Hydrochloride
of this solution is 5.8 to 7.8.
セチリジン塩酸塩
Purity Clarity and color of solution—Dissolve 5 g of diso-
dium hydrogen phosphate and 1 g of potassium dihydrogen
phosphate in water to make 100 mL. In 10 mL of this solu-
tion dissolve an amount of Ceftazidime for Injection,
equivalent to 1.0 g (potency) of Ceftazidime Hydrate ac-
cording to the labeled amount: the solution is clear and
colorless. Also, determine the absorption spectra of this so- C21H25ClN2O3.2HCl: 461.81
lution as directed under Ultraviolet-visible Spectrophoto- 2-(2-{4-[(RS)-(4-Chlorophenyl)phenylmethyl]piperazin-
metry <2.24>: the absorbance at 420 nm is not more than 0.3. 1-yl}ethoxy)acetic acid dihydrochloride [83881-52-1]
Loss on drying <2.41> Not more than 14.0z (0.1 g, in
vacuum not exceeding 0.67 kPa, 609
C, 3 hours). Cetirizine Hydrochloride, when dried, contains not
less than 99.0z and not more than 101.0z of
Bacterial endotoxins <4.01> Less than 0.067 EU/mg (po- C21H25ClN2O3.2HCl.
tency).
Description Cetirizine Hydrochloride occurs as a white
Uniformity of dosage units <6.02> It meets the requirement crystalline powder.
of the Mass variation test. It is very soluble in water, and slightly soluble in ethanol
Foreign insoluble matter <6.06> Perform the test according (99.5).
to Method 2: it meets the requirement. It dissolves in 0.1 mol/L hydrochloric acid TS.
A solution of Cetirizine Hydrochloride (1 in 10) shows no
Insoluble particulate matter <6.07> It meets the require- optical rotation.
ment.
Identification (1) Determine the absorption spectrum of
Sterility <4.06> Perform the test according to the Mem- a solution of Cetirizine Hydrochloride in 0.1 mol/L
brane filter method: it meets the requirement. hydrochloric acid TS (1 in 50,000) as directed under Ultrav-
Assay Weigh accurately the mass of the contents of not iolet-visible Spectrophotometry <2.24>, and compare the
less than 10 containers of Ceftazidime for Injection. Weigh spectrum with the Reference Spectrum: both spectra exhibit
accurately an amount of Ceftazidime Hydrate, equivalent to similar intensities of absorption at the same wavelengths.
about 0.25 g (potency), and dissolve in 0.05 mol/L phos- (2) Determine the infrared absorption spectrum of
phate buffer solution, pH 7.0, to make exactly 250 mL. Cetirizine Hydrochloride as directed in the potassium chlo-
Pipet 10 mL of this solution, add exactly 5 mL of the inter- ride disk method under Infrared Spectrophotometry <2.25>,
nal standard solution, add more 0.05 mol/L phosphate and compare the spectrum with the Reference Spectrum:
Supplement I, JP XV Official Monographs 1857

both spectra exhibit similar intensities of absorption at the C, 3 hours).


um, 609
same wave numbers.
Residue on ignition <2.44> Not more than 0.2z (1 g).
(3) A solution of Cetirizine Hydrochloride (1 in 100)
responds to the Qualitative Tests <1.09> for chloride. Assay Weigh accurately about 0.1 g of Cetirizine
Hydrochloride, previously dried, dissolve in 70 mL of a
Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of
mixture of acetone and water (7:3), and titrate <2.50> to the
Cetirizine Hydrochloride according to Method 2, and per-
second equivalence point with 0.1 mol/L sodium hydroxide
form the test. Prepare the control solution with 2.0 mL of
VS (potentiometric titration). Perform a blank determina-
Standard Lead Solution (not more than 10 ppm).
tion in the same manner, and make any necessary correc-
(2) Related substances—Dissolve 0.10 g of Cetirizine
tion.
Hydrochloride in 50 mL of the mobile phase, and use this
solution as the sample solution. Pipet 2 mL of the sample so- Each mL of 0.1 mol/L sodium hydroxide VS
lution, add the mobile phase to make exactly 50 mL. Pipet 5 =15.39 mg of C21H25ClN2O3.2HCl
mL of this solution, add the mobile phase to make exactly
Containers and storage Containers—Well-closed contain-
100 mL, and use this solution as the standard solution. Per-
ers.
form the test with exactly 10 mL each of the sample solution
and standard solution as directed under Liquid Chro-
matography <2.01> according to the following conditions,
Add the following:
and determine each peak area of each solution by the auto-
matic integration method: the area of each peak other than
cetirizine obtained from the sample solution is not larger Cetirizine Hydrochloride Tablets
than the peak area of cetirizine from the standard solution.
セチリジン塩酸塩錠
Furthermore, the total area of the peaks other than cetirizine
from the sample solution is not larger than 2.5 times the
Cetirizine Hydrochloride Tablets contain not less
peak area of cetirizine from the standard solution.
than 95.0z and not more than 105.0z of the labeled
Operating conditions—
amount of cetirizine hydrochloride (C21H25ClN2O3.
Detector: An ultraviolet absorption photometer (wave-
2HCl: 461.81).
length: 230 nm).
Column: A stainless steel column 4.0 mm in inside di- Method of preparation Prepare as directed under Tablets,
ameter and 25 cm in length, packed with silica gel for liquid with Cetirizine Hydrochloride.
chromatography (5 mm in particle diameter).
Identification To a quantity of powdered Cetirizine
Column temperature: A constant temperature of about
Hydrochloride Tablets, equivalent to 10 mg of Cetirizine
259C.
Hydrochloride according to the labeled amount, add about
Mobile phase: A mixture of acetonitrile and diluted 0.5
70 mL of 0.1 mol/L hydrochloric acid TS, shake, add 0.1
mol/L sulfuric acid TS (2 in 25) (47:3).
mol/L hydrochloric acid TS to make 100 mL, and filter. To
Flow rate: Adjust the flow rate so that the retention time
4 mL of the filtrate add 0.1 mol/L hydrochloric acid TS to
of cetirizine is about 9 minutes.
make 25 mL, and determine the absorption spectrum of this
Time span of measurement: About 3 times as long as the
solution as directed under Ultraviolet-visible Spectrophoto-
retention time of cetirizine, beginning after the solvent peak.
metry <2.24>: it exhibits a maximum between 230 nm and
System suitability—
234 nm.
Test for required detectability: Pipet 5 mL of the standard
solution, and add the mobile phase to make exactly 10 mL. Uniformity of dosage units <6.02> Perform the test accord-
Confirm that the peak area of cetirizine obtained from 10 mL ing to the following method: it meets the requirement of the
of this solution is equivalent to 35 to 65z of that from 10 mL Content uniformity test.
of the standard solution. Take 1 tablet of Cetirizine Hydrochloride Tablets, add
System performance: Dissolve 20 mg of Cetirizine 4V/5 mL of sodium 1-heptanesulfonate solution (1 in 5000)
Hydrochloride in the mobile phase to make 100 mL. To 5 adjusted to pH 3.0 with 0.5 mol/L sulfuric acid TS, treat
mL of this solution, add 3 mL of a solution of aminopyrine with ultrasonic waves for 20 minutes, adjust the volume to
in the mobile phase (1 in 2500), and add the mobile phase to exactly V mL, by adding sodium 1-heptanesulfonate solu-
make 20 mL. When the procedure is run with 10 mL of this tion (1 in 5000) adjusted to pH 3.0 with 0.5 mol/L sulfuric
solution under the above operating conditions, cetirizine and acid TS, so that each mL contains about 0.2 mg of cetirizine
aminopyrine are eluted in this order with the resolution be- hydrochloride (C21H25ClN2O3.2HCl), and filter through a
tween these peaks being not less than 7. membrane filter with a pore size not exceeding 0.45 mm. Dis-
System repeatability: When the test is repeated 6 times card the first 3 mL of the filtrate, pipet 5 mL of the subse-
with 10 mL of the standard solution under the above operat- quent filtrate, add exactly 2 mL of the internal standard so-
ing conditions, the relative standard deviation of the peak lution, add acetonitrile to make 10 mL, and use this solution
area of cetirizine is not more than 2.0z. as the sample solution. Then, proceed as directed in the As-
say.
Loss on drying <2.41> Not more than 0.5z (1 g, in vacu-
1858 Official Monographs Supplement I, JP XV

ditions, cetirizine and the internal standard are eluted in this


Amount (mg) of cetirizine hydrochloride
order with the resolution between these peaks being not less
(C21H25ClN2O3.2HCl)
than 7.
=WS×(QT/QS)×(V/100)
System repeatability: When the test is repeated 6 times
WS: Amount (mg) of cetirizine hydrochloride for assay with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratio of
Internal standard solution—A solution of propyl para-
the peak area of cetirizine to that of the internal standard is
hydroxybenzoate in the mobile phase (1 in 1000).
not more than 1.0z.
Assay Weigh accurately not less than 20 Cetirizine
Containers and storage Containers—Well-closed contain-
Hydrochloride Tablets, and powder. Weigh accurately a
ers.
portion of the powder, equivalent to about 10 mg of cetiri-
zine hydrochloride (C21H25ClN2O3.2HCl), add 40 mL of so-
dium 1-heptanesulfonate solution (1 in 5000) adjusted to pH
3.0 with 0.5 mol/L sulfuric acid TS, treat with ultrasonic Chlordiazepoxide Tablets
waves for 20 minutes, add sodium 1-heptanesulfonate solu-
クロルジアゼポキシド錠
tion (1 in 5000), adjusted to pH 3.0 with 0.5 mol/L sulfuric
acid TS, to make exactly 50 mL, and filter through a mem-
Add the following next to Purity:
brane with a pore size not exceeding 0.45 mm. Discard the
first 3 mL of the filtrate, pipet 5 mL of the subsequent Uniformity of dosage units <6.02> Perform the test accord-
filtrate, add exactly 2 mL of the internal standard solution, ing to the following method: it meets the requirement of the
add acetonitrile to make exactly 10 mL, and use this solution Content uniformity test.
as the sample solution. Separately, weigh accurately about Conduct this procedures using light-resistant vessels. To 1
20 mg of cetirizine hydrochloride for assay, previously dried tablet of Chlordiazepoxide Tablets add 1 mL of water, shake
in vacuum at 609C for 3 hours, and add sodium 1-hep- to disintegrate the tablet, then add 20 mL of methanol,
tanesulfonate solution (1 in 5000), adjusted to pH 3.0 with shake, add methanol to make exactly 25 mL, and filter
0.5 mol/L sulfuric acid TS, to make exactly 100 mL. Pipet 5 through a membrane filter with a pore size not exceeding 0.5
mL of this solution, add exactly 2 mL of the internal stan- mm. Discard the first 5 mL of the filtrate, take exactly V mL
dard solution, add acetonitrile to make 10 mL, and use this of the subsequent filtrate equivalent to about 2 mg of chlor-
solution as the standard solution. Perform the test with 20 diazepoxide (C16H14ClN3O), add exactly 1 mL of the internal
mL each of the sample solution and standard solution as standard solution, then add methanol to make 20 mL, and
directed under Liquid Chromatography <2.01> according to use this solution as the sample solution. Then, proceed as
the following conditions, and calculate the ratios, QT and directed in the Assay.
QS, of the peak area of cetirizine to that of the internal stan-
Amount (mg) of chlordiazepoxide (C16H14ClN3O)
dard.
=WS×(QT/QS)×(5/V)
Amount (mg) of cetirizine hydrochloride
WS: Amount (mg) of Chlordiazepoxide Reference Stan-
(C21H25ClN2O3.2HCl)
dard
=WS×(QT/QS)×(1/2)
Internal standard solution—A solution of isobutyl salicylate
WS: Amount (mg) of cetirizine hydrochloride for assay
in methanol (1 in 20).
Internal standard solution—A solution of propyl para-
hydroxybenzoate in the mobile phase (1 in 1000).
Operating conditions— Chlorphenesin Carbamate
Detector: An ultraviolet absorption photometer (wave-
length: 230 nm). クロルフェネシンカルバミン酸エステル
Column: A stainless steel column 4.0 mm in inside di-
ameter and 25 cm in length, packed with octylsilanized silica Change the Description to read:
gel for liquid chromatography (5 mm in particle diameter). Description Chlorphenesin Carbamate occurs as white
Column temperature: A constant temperature of about crystals or a crystalline powder.
259C. It is freely soluble in methanol, in ethanol (95) and in pyri-
Mobile phase: A mixture of a solution of sodium 1-hep- dine, and slightly soluble in water.
tansulfonate (1 in 2900) and acetonitrile (29:21), adjusted to A solution of Chlorphenesin Carbamate in ethanol (95)
pH 3.0 with 0.5 mol/L sulfuric acid TS. (1 in 20) shows no optical rotation.
Flow rate: Adjust the flow rate so that the retention time
of cetirizine is about 5 minutes. Change the Identification (2) to read:
System suitability—
System performance: When the procedure is run with 20 Identification
mL of the standard solution under the above operating con- (2) Determine the infrared absorption spectrum of
Supplement I, JP XV Official Monographs 1859

Chlorphenesin Carbamate, as directed in the potassium with 10 mL of the solution for system suitability test under
bromide disk method under Infrared Spectrophotometry the above operating conditions, the relative standard devia-
<2.25>, and compare the spectrum with the Reference Spec- tion of the peak areas of chlorphenesin carbamate is not
trum: both spectra exhibit similar intensities of absorption at more than 2.0z.
the same wave numbers.
Add the following next to Purity (3) :
Change the Purity (3) to read:
(4) Related substances—Dissolve 0.10 g of Chlorphene-
Purity sin Carbamate in 10 mL of ethanol (95), and use this solu-
(3) Chlorphenesin-2-carbamate—Dissolve 0.10 g of tion as the sample solution. Pipet 1 mL of the sample solu-
Chlorphenesin Carbamate in 20 mL of a mixture of hexane tion, add ethanol (95) to make exactly 20 mL. Pipet 2 mL of
for liquid chromatography and 2-propanol (7:3), and use this solution, add ethanol (95) to make exactly 20 mL, and
this solution as the sample solution. Perform the test with 10 use this solution as the standard solution. Perform the test
mL of the sample solution as directed under Liquid Chro- with these solutions as directed under Thin-layer Chro-
matography <2.01> according to the following conditions. matography <2.03>. Spot 50 mL each of the sample solution
Determine the peak area, Aa, of chlorphenesin carbamate and standard solution on a plate of silica gel for thin-layer
and the peak area, Ab, of chlorphenesin-2-carbamate by the chromatography. Develop the plate with a mixture of ethyl
automatic integration method: the ratio, Ab/(Aa+Ab), is acetate, methanol and ammonia solution (28) (17:2:1) to a
not more than 0.007. distance of about 10 cm, and air-dry the plate. Allow the
Operating conditions— plate to stand in iodine vapor for 20 minutes: the spot other
Detector: An ultraviolet absorption photometer (wave- than the principal spot from the sample solution is not more
length: 280 nm). than one, and it is not more intense than the spot from the
Column: A stainless steel column 4 mm in inside diameter standard solution.
and 30 cm in length, packed with silica gel for liquid chro-
matography (5 mm in particle diameter).
Column temperature: A constant temperature of about Add the following:
409C.
Mobile phase: A mixture of hexane for liquid chro- Chlorphenesin Carbamate Tablets
matography, 2-propanol and acetic acid (100) (700:300:1).
Flow rate: Adjust the flow rate so that the retention time クロルフェネシンカルバミン酸エステル錠
of chlorphenesin carbamate is about 9 minutes.
System suitability— Chlorphenesin Carbamate Tablets contain not less
Test for required detection: Pipet 1 mL of the sample than 93.0z and not more than 107.0z of the labeled
solution, add a mixture of hexane for liquid chro- amount of chlorphenesin carbamate (C10H12ClNO4:
matography and 2-propanol (7:3) to make exactly 100 mL, 245.66).
and use this solution as the solution for system suitability
Method of preparation Prepare as directed under Tablets,
test. To exactly 5 mL of the solution for system suitability
with Chlorphenesin Carbamate.
test add the mixture of hexane for liquid chromatography
and 2-propanol (7:3) to make exactly 10 mL. Confirm that Identification To a quantity of powdered Chlorphenesin
the peak area of chlorphenesin carbamate obtained from 10 Carbamate Tablets, equivalent to 0.15 g of Chlorphenesin
mL of this solution is equivalent to 40 to 60z of that of Carbamate according to the labeled amount, add 60 mL of
chlorphenesin carbamate obtained from 10 mL of the solu- ethanol (95), treat with ultrasonic waves, and add ethanol
tion for system suitability test. (95) to make 100 mL. Centrifuge 20 mL of this solution, add
System performance: Dissolve 0.1 g of Chlorphenesin ethanol (95) to 1 mL of the supernatant liquid to make 100
Carbamate in 50 mL of methanol. To 25 mL of this solution mL, and determine the absorption spectrum of this solution
add 25 mL of dilute sodium hydroxide TS, and warm at as directed under Ultraviolet-visible Spectrophotometry
609C for 20 minutes. To 20 mL of this solution add 5 mL of <2.24>: it exhibits maxima between 226 nm and 230 nm, be-
1 mol/L hydrochloric acid TS, shake well with 20 mL of tween 279 nm and 283 nm, and between 286 nm and 290 nm.
ethyl acetate, and allow to stand to separate the upper layer.
Uniformity of dosage units <6.02> Perform the test accord-
When the procedure is run with 10 mL of this layer under the
ing to the following method: it meets the requirement of the
above operating conditions, chlorphenesin, chlorphenesin
Content uniformity test.
carbamate and chlorphenesin-2-carbamate are eluted in this
To 1 tablet of Chlorphenesin Carbamate Tablets add 10
order, with the relative retention times of chlorphenesin and
mL of water to disintegrate the tablet, add 70 mL of a mix-
chlorphenesin-2-carbamate with respect to chlorphenesin
ture of water and methanol (1:1), treat with ultrasonic waves
carbamate being about 0.7 and about 1.2, respectively, and
for 15 minutes with occasional stirring, then add the mixture
with the resolution between the peaks of chlorphenesin and
of water and methanol (1:1) to make exactly 100 mL. Cen-
chlorphenesin carbamate being not less than 2.0.
trifuge this solution, pipet V mL of the supernatant liquid
System repeatability: When the test is repeated 6 times
equivalent to about 2.5 mg of chlorphenesin carbamate
1860 Official Monographs Supplement I, JP XV

(C10H12ClNO4), add the mixture of water and methanol (1:1) ly dried in a desiccator (in vacuum, silica gel) for 4 hours,
to make exactly 25 mL, and use this solution as the sample and dissolve in ethyl acetate to make exactly 50 mL. Pipet 5
solution. Separately, weigh accurately about 50 mg of chlor- mL of this solution, add exactly 2 mL of the internal stan-
phenesin carbamate for assay, previously dried in a desicca- dard solution, then add ethyl acetate to make 20 mL, and
tor (in vacuum, silica gel) for 4 hours, and dissolve in the use this solution as the standard solution. Perform the test
mixture of water and methanol (1:1) to make exactly 50 mL. with 10 mL each of the sample solution and standard solu-
Pipet 2 mL of this solution, add the mixture of water and tion as directed under Liquid Chromatography <2.01> ac-
methanol (1:1) to make exactly 20 mL, and use this solution cording to the following conditions, and calculate the ratios,
as the standard solution. Determine the absorbances at 280 QT and QS, of the peak area of chlorphenesin carbamate to
nm, AT and AS, of the sample solution and standard solution that of the internal standard.
as directed under Ultraviolet-visible Spectrophotometry
Amount (mg) of chlorphenesin carbamate (C10H12ClNO4)
<2.24>.
=WS×(QT/QS)×(5/2)
Amount (mg) of chlorphenesin carbamate
WS: Amount (mg) of chlorphenesin carbamate for assay
(C10H12ClNO4)
=WS×(AT/AS)×(1/V)×5 Internal standard solution—A solution of ethenzamide in
ethyl acetate (1 in 400).
WS: Amount (mg) of chlorphenesin carbamate for assay
Operating conditions—
Dissolution <6.10> When the test is performed at 50 revolu- Detector: An ultraviolet absorption photometer (wave-
tions per minute according to the Paddle method, using 900 length: 280 nm).
mL of water as the dissolution medium, the dissolution rate Column: A stainless steel column 4 mm in inside diameter
in 15 minutes of Chlorphenesin Carbamate Tablets is not and 30 cm in length, packed with silica gel for liquid chro-
less than 85z. matography (5 mm in particle diameter).
Start the test with 1 tablet of Chlorphenesin Carbamate Column temperature: A constant temperature of about
Tablets, withdraw not less than 20 mL of the medium at the 409C.
specified minute after starting the test, and filter through a Mobile phase: A mixture of hexane for liquid chro-
membrane filter with a pore size not exceeding 0.45 mm. Dis- matography, 2-propanol and acetic acid (100) (700:300:1).
card the first 10 mL of the filtrate, pipet V mL of the subse- Flow rate: Adjust the flow rate so that the retention time
quent filtrate, add water to make exactly V? mL so that each of chlorphenesin carbamate is about 9 minutes.
mL contains about 0.14 mg of chlorphenesin carbamate System suitability—
(C10H12ClNO4) according to the labeled amount, and use this System performance: Proceed as directed in the Purity (3)
solution as the sample solution. Separately, weigh accurately under Chlorphenesin Carbamate.
about 28 mg of chlorphenesin carbamate for assay, previ- System repeatability: When the test is repeated 6 times
ously dried in a desiccator (in vacuum, silica gel) for 4 hours, with 10 mL of the standard solution under the above operat-
dissolve in 1 mL of methanol, and add water to make exactly ing conditions, the relative standard deviation of the ratio of
50 mL. Pipet 5 mL of this solution, add water to make ex- the peak area of chlorphenesin carbamate to that of the in-
actly 20 mL, and use this solution as the standard solution. ternal standard is not more than 1.5z.
Determine the absorbances, AT and AS, at 278 nm of the
Containers and storage Containers—Well-closed contain-
sample solution and standard solution as directed under
ers.
Ultraviolet-visible Spectrophotometry <2.24>.

Dissolution rate (z) with respect to the labeled amount of


chlorphenesin carbamate (C10H12ClNO4) Chlorpromazine Hydrochloride
=WS×(AT/AS)×(V?/V)×(1/C)×450
Injection
WS: Amount (mg) of chlorphenesin carbamate for assay
C: Labeled amount (mg) of chlorphenesin carbamate クロルプロマジン塩酸塩注射液
(C10H12ClNO4) in 1 tablet
Add the following next to Extractable volume:
Assay Weigh accurately the mass of not less than 20 Chlor-
phenesin Carbamate Tablets, and powder them in an agate Foreign insoluble matter <6.06> Perform the test according
mortar. Weigh accurately a portion of the powder, equiva- to Method 1: it meets the requirement.
lent to about 0.25 g of chlorphenesin carbamate Insoluble particulate matter <6.07> It meets the require-
(C10H12ClNO4), add 30 mL of ethyl acetate, disperse using ment.
ultrasonic waves, then add ethyl acetate to make exactly 50
mL. Centrifuge 20 mL of this solution, pipet 2 mL of the su- Sterility <4.06> Perform the test according to the Mem-
pernatant liquid, add exactly 2 mL of the internal standard brane filtration method: it meets the requirement.
solution, add ethyl acetate to make 20 mL, and use this solu-
tion as the sample solution. Separately, weigh accurately
about 0.1 g of chlorphenesin carbamate for assay, previous-
Supplement I, JP XV Official Monographs 1861

Amount (mg) of chlorpropamide (C10H13ClN2O3S)


Chlorpromazine Hydrochloride =WS×(AT/AS)×(V/20)
Tablets WS: Amount (mg) of chlorpropamide for assay
クロルプロマジン塩酸塩錠

Add the following next to Identification: Add the following:

Uniformity of dosage units <6.02> Perform the test accord- Cibenzoline Succinate
ing to the following method: it meets the requirement of the
Content uniformity test. シベンゾリンコハク酸塩
Conduct this procedures using light-resistant vessels. To 1
tablet of Chlorpromazine Hydrochloride Tablets add an
amount of a mixture of diluted phosphoric acid (1 in 500)
and ethanol (99.5) (1:1) so that each mL contains about 0.83
mg of chlorpromazine hydrochloride (C17H19ClN2S.HCl),
treat with the ultrasonic waves for 5 minutes, then shake
vigorously for 20 minutes, and add the mixture of diluted C18H18N2.C4H6O4: 380.44
phosphoric acid (1 in 500) and ethanol (99.5) (1:1) to make 2-[(1RS)-2,2-Diphenylcyclopropan-1-yl]-4,5-dihydro-1H-
exactly V mL so that each mL contains about 0.5 mg of imidazole monosuccinate [100678-32-8]
chlorpromazine hydrochloride (C17H19ClN2S.HCl). Filter
through a membrane filter with a pore size not exceeding Cibenzoline Succinate, when dried, contains not less
0.45 mm. Discard the first 3 mL of the filtrate, pipet 2.5 mL than 98.5z and not more than 101.0z of C18H18N2.
of the subsequent filtrate, add exactly 5 mL of the internal C 4 H 6 O4 .
standard solution, then add the mixture of diluted phos-
Description Cibenzoline Succinate occurs as a white crys-
phoric acid (1 in 500) and ethanol (99.5) (1:1) to make 25
talline powder.
mL, and use this solution as the sample solution. Then, pro-
It is freely soluble in methanol and in acetic acid (100),
ceed as directed in the Assay.
and sparingly soluble in water and in ethanol (99.5).
Amount (mg) of chlorpromazine hydrochloride A solution of Cibenzoline Succinate in methanol (1 in 10)
(C17H19ClN2S.HCl) shows no optical rotation.
=WS×(QT/QS)×(V/50)
Identification (1) Determine the absorption spectrum of
WS: Amount (mg) of chlorpromazine hydrochloride for a solution of Cibenzoline Succinate (1 in 50,000) as directed
assay under Ultraviolet-visible Spectrophotometry <2.24>, and
compare the spectrum with the Reference Spectrum: both
Internal standard solution—A solution of ethyl parahydrox-
spectra exhibit similar intensities of absorption at the same
ybenzoate in the mixture of diluted phosphoric acid (1 in
wavelengths.
500) and ethanol (99.5) (1:1) (1 in 4500).
(2) Determine the infrared absorption spectrum of
Cibenzoline Succinate as directed in the paste method under
Infrared Spectrophotometry <2.25>, and compare the spec-
Chlorpropamide Tablets trum with the Reference Spectrum: both spectra exhibit
similar intensities of absorption at the same wave numbers.
クロルプロパミド錠
(3) Shake 0.4 g of Cibenzoline Succinate with 2.5 mL of
sodium hydroxide TS and 5 mL of ethyl acetate, allow to
Add the following next to Identification:
stand, and to 1 mL of the water layer so obtained add 0.5
Uniformity of dosage units <6.02> Perform the test accord- mL of 1 mol/L hydrochloric acid TS and 0.5 mL of iron
ing to the following method: it meets the requirement of the (III) chloride TS: a blown precipitate is formed.
Content uniformity test.
Melting point <2.60> 163 – 1679
C
To 1 tablet of Chlorpropamide Tablets add 75 mL of the
mobile phase, treat with the ultrasonic waves for 20 minutes pH <2.54> Dissolve 0.20 g of Cibenzoline Succinate in 10
with occasional strong shaking, then add the mobile phase to mL of water: the pH of this solution is between 4.0 and 6.0.
make exactly V mL so that each mL contains about 2.5 mg
Purity (1) Clarity and color of solution—A solution ob-
of Chlorpropamide. Centrifuge the solution, pipet 2 mL of
tained by dissolving 0.20 g of Cibenzoline Succinate in 10
the supernatant liquid, add the mobile phase to make exactly
mL of water is clear and colorless.
100 mL, and use this solution as the sample solution. Then,
(2) Heavy metals <1.07>—Proceed with 1.0 g of Ciben-
proceed as directed in the Assay.
zoline Succinate according to Method 2, and perform the
test. Prepare the control solution with 2.0 mL of Standard
1862 Official Monographs Supplement I, JP XV

Lead Solution (not more than 20 ppm). Identification To a quantity of powdered Cibenzoline Suc-
(3) Arsenic <1.11>—Prepare the test solution with 1.0 g cinate Tablets, equivalent to 50 mg of Cibenzoline Succinate
of Cibenzoline Succinate according to Method 3, using a according to the labeled amount, add 100 mL of water,
solution of magnesium nitrate hexahydrate in ethanol (95) shake for 10 minutes, and centrifuge. To 2 mL of the super-
(1 in 25), and perform the test (not more than 2 ppm). natant liquid add water to make 50 mL, and determine the
(4) Related substances—Dissolve 0.10 g of Cibenzoline absorption spectrum of this solution as directed under
Succinate in 10 mL of methanol, and use this solution as the Ultraviolet-visible Spectrophotometry <2.24>: it exhibits a
sample solution. Pipet 1 mL of the sample solution, and add maximum between 221 nm and 225 nm.
methanol to make exactly 100 mL. Pipet 5 mL and 2 mL of
Uniformity of dosage units <6.02> Perform the test accord-
this solution, add methanol to make them exactly 10 mL,
ing to the following method: it meets the requirement of the
and use these solutions as the standard solution (1) and the
Content uniformity test.
standard solution (2). Perform the test with these solutions
To 1 tablet of Cibenzoline Succinate Tablets add a suita-
as directed under Thin-layer Chromatography <2.03>. Spot
ble amount of water so that each mL contains about 10 mg
10 mL each of the sample solution and standard solutions (1)
of cibenzoline succinate (C18H18N2.C4H6O4), and allow
and (2) on a plate of silica gel with fluorescent indicator for
standing for 10 minutes while occasional shaking. To this so-
thin-layer chromatography. Develop the plate with a mix-
lution add methanol so that each mL contains about 2 mg of
ture of ethyl acetate, methanol and ammonia solution (28)
cibenzoline succinate (C18H18N2.C4H6O4), add exactly 1 mL
(20:3:2) to a distance of about 10 cm, air-dry the plate, and
of the internal standard solution per 10 mg of cibenzoline
dry more at 809 C for 30 minutes. After cooling, examine un-
succinate (C18H18N2.C4H6O4), then add methanol so that
der ultraviolet light (main wavelength: 254 nm): the spot
each mL contains about 1 mg of cibenzoline succinate
other than the principal spot obtained with the sample solu-
(C18H18N2.C4H6O4). Centrifuge the solution, and use the su-
tion is not more intense than the spot with the standard solu-
pernatant liquid as the sample solution. Then, proceed as
tion (1). Allow the plate to stand for 30 minutes in iodine
directed in the Assay.
vapor: the spot other than the principal spot obtained with
the sample solution is not more intense than the spot with Amount (mg) of cibenzoline succinate (C18H18N2.C4H6O4)
the standard solution (1), and the spot, which is more intense =WS×(QT/QS)×(C/100)
than the spot with the standard solution (2), is not more than
WS: Amount (mg) of cibenzoline succinate for assay
two.
C: Labeled amount (mg) of cibenzoline succinate
Loss on drying <2.41> Not more than 0.3z (1 g, 1059C, 2 (C18H18N2.C4H6O4) in 1 tablet
hours).
Internal standard solution—Dissolve 0.1 g of 2-ethylhexyl
Residue on ignition <2.44> Not more than 0.1z (1 g). parahydroxybenzoate in methanol to make 100 mL.

Assay Weigh accurately about 0.4 g of Cibenzoline Suc- Dissolution <6.10> When the test is performed at 50 revolu-
cinate, previously dried, dissolve in 50 mL of acetic acid tions per minute according to the Paddle method, using 900
(100), and titrate <2.50> with 0.1 mol/L perchloric acid VS mL of water as the dissolution medium, the dissolution rate
until the color of the solution changes from violet to blue- in 15 minutes of Cibenzoline Succinate Tablets is not less
green through blue (indicator: 2 drops of crystal violet TS). than 80z.
Perform a blank determination in the same manner, and Start the test with 1 tablet of Cibenzoline Succinate
make any necessary correction. Tablets, withdraw not less than 20 mL of the medium at the
specified minute after starting the test, and filter through a
Each mL of 0.1 mol/L perchloric acid VS
membrane filter with a pore size not exceeding 0.45 mm. Dis-
=38.04 mg of C18H18N2.C4H6O4
card the first 10 mL of the filtrate, pipet V mL of the subse-
Containers and storage Containers—Tight containers. quent filtrate, add water to make exactly V? mL so that each
mL contains about 11 mg of cibenzoline succinate (C18H18N2.
C4H6O4) according to the labeled amount, and use this solu-
Add the following: tion as the sample solution. Separately, weigh accurately
about 28 mg of cibenzoline succinate for assay, previously
Cibenzoline Succinate Tablets dried at 1059 C for 2 hours, and dissolve in water to make
exactly 100 mL. Pipet 2 mL of this solution, add water to
シベンゾリンコハク酸塩錠 make exactly 50 mL, and use this solution as the standard
solution. Determine the absorbances, AT and AS, of the
Cibenzoline Succinate Tablets contain not less than sample solution and standard solution at 222 nm as directed
95.0z and not more than 105.0z of the labeled under Ultraviolet-visible Spectrophotometry <2.24>.
amount of cibenzoline succinate (C18H18N2.C4H6O4:
Dissolution rate (z) with respect to the labeled amount of
380.44).
cibenzoline succinate (C18H18N2.C4H6O4)
Method of preparation Prepare as directed under Tablets, =WS×(AT/AS)×(V?/V)×(1/C)×36
with Cibenzoline Succinate.
Supplement I, JP XV Official Monographs 1863

WS: Amount (mg) of cibenzoline succinate for assay Add the following:
C: Labeled amount (mg) of cibenzoline succinate
(C18H18N2.C4H6O4) in 1 tablet Cilazapril Hydrate
Assay Weigh accurately not less than 20 Cibenzoline Suc-
シラザプリル水和物
cinate Tablets, and powder. Weigh accurately a portion of
the powder, equivalent to about 0.1 g of cibenzoline suc-
cinate (C18H18N2.C4H6O4), add 10 mL of water, shake, and
add 40 mL of methanol and exactly 10 mL of the internal
standard solution. Shake for 20 minutes, add methanol to
make 100 mL, centrifuge, and use the supernatant liquid as
C22H31N3O5.H2O: 435.51
the sample solution. Separately, weigh accurately about
(1S,9S)-9-[(1S)-(1-Ethoxycarbonyl-
0.1 g of cibenzoline succinate for assay, previously dried at
3-phenylpropyl)amino]-10-oxooctahydro-
1059 C for 2 hours, add 10 mL of water and 40 mL of
6H-pyridazino[1,2-a][1,2]diazepine-1-carboxylic acid
methanol to dissolve, then add exactly 10 mL of the internal
monohydrate [92077-78-6]
standard solution and methanol to make 100 mL, and use
this solution as the standard solution. Perform the test with
Cilazapril Hydrate contains not less than 98.5z and
5 mL each of the sample solution and standard solution as
not more than 101.0z of cilazapril (C22H31N3O5:
directed under Liquid Chromatography <2.01> according to
417.50), calculated on the anhydrous basis.
the following conditions, and calculate the ratios, QT and
QS, of the peak area of cibenzoline to that of the internal Description Cilazapril Hydrate occurs as white to yellow-
standard. ish white crystals or crystalline powder.
It is very soluble in methanol, freely soluble in ethanol
Amount (mg) of cibenzoline succinate (C18H18N2.C4H6O4)
(99.5) and in acetic acid (100), and slightly soluble in water.
= W S ×( Q T / Q S )
It gradually turns yellow on exposure to light.
WS: Amount (mg) of cibenzoline succinate for assay Melting point: about 1019 C (with decomposition).

Internal standard solution—Dissolve 0.1 g of 2-ethylhexyl Identification (1) To 4 mL of a solution of Cilazapril


parahydroxybenzoate in methanol to make 100 mL. Hydrate (1 in 1000) add 2 mL of Dragendorff's TS: an
Operating conditions— orange precipitate is produced.
Detector: An ultraviolet absorption photometer (wave- (2) Determine the infrared absorption spectrum of
length: 254 nm). Cilazapril Hydrate as directed in the potassium bromide disk
Column: A stainless steel column 4.6 mm in inside di- method under Infrared Spectrophotometry <2.25>, and com-
ameter and 5 cm in length, packed with octadecylsilanized pare the spectrum with the Reference Spectrum: both spec-
silica gel for liquid chromatography (3 mm in particle di- tra exhibit similar intensities of absorption at the same wave
ameter). numbers.
Column temperature: A constant temperature of about
Optical rotation <2.49> [a]20
D : -53 – -589(0.2 g calculated
259C.
on the anhydrous basis, methanol, 20 mL, 100 mm).
Mobile phase: Dissolve 2.67 g of sodium di-2-ethylhexyl
sulfosuccinate in 2000 mL of a mixture of water, acetonitrile Purity (1) Chloride <1.03>—Perform the test using 1.0 g
and diluted phosphoric acid (1 in 10) (1000:1000:1). of Cilazapril Hydrate. Prepare the control solution with 0.25
Flow rate: Adjust the flow rate so that the retention time mL of 0.01 mol/L hydrochloric acid VS (not more than
of cibenzoline is about 3 minutes. 0.009z).
System suitability— (2) Sulfate <1.14>—Dissolve 1.0 g of Cilazapril Hydrate
System performance: When the procedure is run with 5 mL in 40 mL of water and 1.5 mL of dilute hydrochloric acid,
of the standard solution under the above operating condi- and add water to make 50 mL. Perform the test using this
tions, cibenzoline and the internal standard are eluted in this solution as the test solution. Prepare the control solution
order with the resolution between these peaks being not less with 0.40 mL of 0.005 mol/L sulfuric acid VS (not more
than 6. than 0.019z).
System repeatability: When the test is repeated 6 times (3) Heavy metals <1.07>—Proceed with 1.0 g of
with 5 mL of the standard solution under the above operat- Cilazapril Hydrate according to Method 4, and perform the
ing conditions, the relative standard deviation of the ratio of test. However, use 10 mL of a solution of magnesium nitrate
the peak area of cibenzoline to that of the internal standard hexahydrate in ethanol (95) (1 in 8). Prepare the control so-
is not more than 1.0z. lution with 2.0 mL of Standard Lead Solution (not more
than 20 ppm).
Containers and storage Containers—Tight containers.
(4) Related substances—Dissolve 0.10 g of Cilazapril
Hydrate in 20 mL of methanol, and use this solution as the
sample solution. Pipet 1 mL of the sample solution, add
methanol to make exactly 100 mL, and use this solution as
1864 Official Monographs Supplement I, JP XV

the standard solution (1). Pipet 3 mL of the standard solu- mg of cilazapril in 5 mL of the mixture of acetonitrile and
tion (1), add methanol to make exactly 10 mL, and use this ethyl acetate (3:1), and use this solution as the standard solu-
solution as the standard solution (2). Separately, pipet 2 mL tion. Perform the test with these solutions as directed under
of the standard solution (1), add methanol to make exactly Thin-layer Chromatography <2.03>. Spot 20 mL each of the
10 mL, and use this solution as the standard solution (3). sample solution and standard solution on a plate of silica gel
Perform the test with these solutions as directed under Thin- with fluorescent indicator for thin-layer chromatography.
layer Chromatography <2.03>. Spot 20 mL each of the sam- Develop the plate with a mixture of ethyl acetate, methanol,
ple solution and three standard solutions on a plate of silica acetic acid (100), hexane and water (62:15:10:10:3) to a dis-
gel with fluorescent indicator for thin-layer chro- tance of about 15 cm, and air-dry the plate. Place the plate
matography. Develop the plate with a mixture of ethyl in iodine vapor for 2 hours, and immediately examine under
acetate, methanol, acetic acid (100), hexane, and water ultraviolet light (main wavelength: 254 nm): the spots ob-
(62:15:10:10:3) to a distance of about 15 cm, and air-dry the tained from the sample and standard solutions are dark
plate. Leave the plate in iodine vapor for 2 hours, and exa- brown and they show the same Rf value.
mine the plate under ultraviolet light (main wavelength: 254
Uniformity of dosage units <6.02> Perform the test accord-
nm): of the spots other than the principal spot with an Rf
ing to the following method: it meets the requirement of the
value close to 0.40 obtained from the sample solution, the
Content uniformity test.
spot in the vicinity of Rf value 0.17 is not more intense than
To 1 tablet of Cilazapril Tablets add 5 mL of a mixture of
the spot from the standard solution (1), and the spot in the
water and acetonitrile (7:3), shake well until disintegration,
vicinity of Rf value 0.44 is not more intense than the spot
add the mixture of water and acetonitrile (7:3) to make
from the standard solution (2). The number of all other spot
exactly V mL so that each mL contains about 25 mg of
does not exceed 3, and of these spots, no more than one is
cilazapril (C22H31N3O5), and centrifuge. Pipet 4 mL of the
more intense than the spot from the standard solution (3)
supernatant liquid, add exactly 1 mL of the internal stan-
and none are more intense than the spot from the standard
dard solution, add the mixture of water and acetonitrile (7:3)
solution (2).
to make 10 mL, and use this solution as the sample solution.
Water <2.48> 3.5 – 5.0z (0.3 g, volumetric titration, direct Separately, weigh accurately about 26 mg of cilazapril for
titration). assay (separately determine the water <2.48> in the same
manner as Cilazapril Hydrate), and dissolve in the mixture
Residue on ignition <2.44> Not more than 0.1z (0.5 g).
of water and acetonitrile (7:3) to make exactly 50 mL. Pipet
Assay Weigh accurately about 0.2 g of Cilazapril Hydrate, 2 mL of this solution, add exactly 10 mL of the internal stan-
dissolve in 50 mL of acetic acid (100), and titrate <2.50> with dard solution, add the mixture of water and acetonitrile (7:3)
0.02 mol/L perchloric acid VS (potentiometric titration). to make 100 mL, and use this solution as the standard solu-
Perform a blank determination in the same manner and tion. Perform the test with 100 mL each of the sample solu-
make any necessary correction. tion and standard solution as directed under Liquid Chro-
matography <2.01> according to the following conditions,
Each mL of 0.02 mol/L perchloric acid VS
and calculate the ratios, QT and QS, of the peak area of
=8.350 mg of C22H31N3O5
cilazapril to that of the internal standard.
Containers and storage Containers—Tight containers.
Amount (mg) of cilazapril (C22H31N3O5)
Storage—Light-resistant.
=WS×(QT/QS)×(V/1000)

WS: Amount (mg) of cilazapril for assay, calculated on


Add the following:
the anhydrous basis

Cilazapril Tablets Internal standard solution—A solution of dimethyl phtha-


late in a mixture of water and acetonitrile (7:3) (1 in 12,500).
シラザプリル錠 Operating conditions—
Proceed as directed in the operating conditions in the
Cilazapril Tablets contain not less than 93.0z and Assay.
not more than 107.0z of the labeled amount of System suitability—
cilazapril (C22H31N3O5: 417.50). System performance: When the procedure is run with 100
mL of the standard solution under the above conditions,
Method of preparation Prepare as directed under Tablets,
cilazapril and the internal standard are eluted in this order
with Cilazapril Hydrate.
with the resolution between these peaks being not less than
Identification To a quantity of powdered Cilazapril 6.
Tablets, equivalent to 2 mg of cilazapril (C22H31N3O5) ac- System repeatability: When the test is repeated 6 times
cording to the labeled amount, add 2 mL of a mixture of with 100 mL of the standard solution under the above condi-
acetonitrile and ethyl acetate (3:1), shake, treat with ultra- tions, the relative standard deviation of the ratio of the peak
sonic waves for 30 seconds, centrifuge, and use the super- area of cilazapril to that of the internal standard is not more
natant liquid as the sample solution. Separately, dissolve 5 than 2.0z.
Supplement I, JP XV Official Monographs 1865

Dissolution <6.10> When the test is performed at 50 revolu- tions, the relative standard deviation of the peak area of
tions per minute according to the Paddle method, using 900 cilazapril is not more than 2.0z.
mL of water as the dissolution medium, the dissolution rate
Assay Weigh acurately 20 Cilazapril Tablets, and powder.
in 15 minutes of Cilazapril Tablets is not less than 85z.
Weigh accurately a portion of the powder, equivalent to
Start the test with 1 tablet of Cilazapril Tablets, withdraw
about 1 mg of cilazapril (C22H31N3O5), add 30 mL of a mix-
not less than 20 mL of the medium at the specified minute
ture of water and acetonitrile (7:3), and treat with ultrasonic
after starting the test, and filter through a membrane filter
waves for 5 minutes. Next, add exactly 5 mL of the internal
with a pore size not exceeding 0.45 mm. Discard the first 10
standard solution, add the mixture of water and acetonitrile
mL of the filtrate, pipet V mL of the subsequent filtrate, and
(7:3) to make 50 mL, and centrifuge. Filter the supernatant
add water to make exactly V? mL so that each mL contains
liquid through a membrane filter with a pore size not exceed-
about 0.28 mg of cilazapril (C22H31N3O5) according to the la-
ing 0.5 mm, and use the filtrate as the sample solution.
beled amount. Pipet 10 mL of the solution, add exactly 5
Separately, weigh accurately about 26 mg of cilazapril for
mL of acetonitrile, and use this solution as the sample solu-
assay (separately determine the water <2.48> in the same
tion. Separately, weigh accurately about 29 mg of cilazapril
manner as Cilazapril Hydrate), and dissolve in the mixture
for assay (separately determine the water <2.48> in the same
of water and acetonitrile (7:3) to make exactly 50 mL. Pipet
manner as Cilazapril Hydrate), and dissolve in water to
2 mL of this solution, add exactly 5 mL of the internal stan-
make exactly 100 mL. Pipet 5 mL of the solution, add water
dard solution, add the mixture of water and acetonitrile (7:3)
to make exactly 100 mL. Then, pipet 2 mL of this solution,
to make 50 mL, and use this solution as the standard solu-
add water to make exactly 100 mL. Pipet 10 mL of this solu-
tion. Perform the test with 50 mL each of the sample solution
tion, add exactly 5 mL of acetonitrile, and use this solution
and standard solution as directed under Liquid Chro-
as the standard solution. Perform the test with exactly 100
matography <2.01> according to the following conditions,
mL each of the sample solution and standard solution as
and calculate the ratios, QT and QS, of the peak area of
directed under Liquid Chromatography <2.01> according to
cilazapril to that of the internal standard.
the following conditions, and determine the peak areas of
cilazapril, AT and AS, of both solutions. Amount (mg) of cilazapril (C22H31N3O5)
=WS×(QT/QS)×(1/25)
Dissolution rate (z) with respect to the labeled amount of
cilazapril (C22H31N3O5) WS: Amount (mg) of cilazapril for assay, calculated on the
=WS×(AT/AS)×(V?/V)×(1/C)×(9/10) anhydrous basis

WS: Amount (mg) of cilazapril for assay, calculated on Internal standard solution—A solution of dimethyl phtha-
the anhydrous basis late in a mixture of water and acetonitrile (7:3) (1 in 12,500).
C: Labeled amount (mg) of cilazapril (C22H31N3O5) in 1 Operating conditions—
tablet Detector: An ultraviolet absorption photometer (wave-
length: 210 nm).
Operating conditions—
Column: A stainless steel column 6 mm in inside diameter
Detector: An ultraviolet absorption photometer (wave-
and 15 cm in length, packed with octadecylsilanized silica gel
length: 210 nm).
for liquid chromatography (5 mm in particle diameter).
Column: A stainless steel column 4.6 mm in inside di-
Column temperature: A constant temperature of about
ameter and 15 cm in length, packed with octadecylsilanized
239C.
silica gel for liquid chromatography (5 mm in particle di-
Mobile phase: To a solution consisting of 180 mL of tetra-
ameter).
hydrofuran for liquid chromatography, 120 mL of acetoni-
Column temperature: A constant temperature of about
trile for liquid chromatography and 3 mL of triethylamine
259C.
add water to make 1000 mL, and adjust the pH to 2.5 with
Mobile phase: To a solution consisting of 180 mL of tetra-
phosphoric acid.
hydrofuran for liquid chromatography, 120 mL of acetoni-
Flow rate: Adjust the flow rate so that the retention time
trile for liquid chromatography and 3 mL of triethylamine
of cilazapril is about 10 minutes.
add water to make 1000 mL, and adjust the pH to 2.5 with
System suitability—
phosphoric acid.
System performance: When the procedure is run with 50
Flow rate: Adjust the flow rate so that the retention time
mL of the standard solution under the above conditions,
of cilazapril is about 10 minutes.
cilazapril and the internal standard are eluted in this order
System suitability—
with the resolution between these peaks being not less than
System performance: When the procedure is run with 100
6.
mL of the standard solution under the above conditions, the
System repeatability: When the test is repeated 6 times
number of theoretical plates and the symmetry factor of the
with 50 mL of the standard solution under the above condi-
peak of cilazapril are not less than 3000 and not more than
tions, the relative standard deviation of the ratio of the peak
2.0, respectively.
area of cilazapril to that of the internal standard is not more
System repeatability: When the test is repeated 6 times
than 1.0z.
with 100 mL of the standard solution under the above condi-
1866 Official Monographs Supplement I, JP XV

Containers and storage Containers—Tight containers. Method of preparation Prepare as directed under
Injections, with Clindamycin Phosphate.

Description Clindamycin Phosphate Injection is a clear,


Cilostazol Tablets colorless or light yellow liquid.
シロスタゾール錠 Identification To a volume of Clindamycin Phosphate In-
jection, equivalent to 0.15 g (potency) of Clindamycin Phos-
Change the Assay to read: phate according to the labeled amount, add 4 mL of water, 2
mL of 8 mol/L sodium hydroxide TS and 0.1 mL of sodium
Assay Weigh accurately not less than 20 Cilostazol
pentacyanonitrosylferrate (III) TS, mix, heat in a water bath
Tablets, and powder. Weigh accurately a portion of the pow-
for 10 minutes, and add 2 mL of hydrochloric acid: a blue-
der, equivalent to about 50 mg of cilostazol (C20H27N5O2),
green color develops.
add exactly 5 mL of the internal standard solution and
methanol to make 50 mL, and shake well for 10 minutes. To Osmotic pressure ratio Being specified separately.
1 mL of this solution add methanol to make 10 mL, filter
pH <2.54> 6.0 – 7.0
through a membrane filter with a pore size not exceeding 0.5
mm, and use the filtrate as the sample solution. Separately, Bacterial endotoxins <4.01> Less than 0.1 EU/mg (poten-
weigh accurately about 50 mg of Cilostazol Reference Stan- cy).
dard, previously dried at 1059 C for 2 hours, dissolve in a
Extractable volume <6.05> It meets the requirement.
suitable amount of methanol, and add exactly 5 mL of the
internal standard solution, and add methanol to make 50 Foreign insoluble matter <6.06> Perform the test according
mL. To 1 mL of this solution add methanol to make 10 mL, to Method 1: it meets the requirement.
and use this solution as the standard solution. Perform the
Insoluble particulate matter <6.07> It meets the require-
test with 10 mL each of the sample solution and standard so-
ment.
lution as directed under Liquid Chromatography <2.01> ac-
cording to the following conditions, and calculate the ratios, Sterility <4.06> Perform the test according to the Mem-
QT and QS, of the peak area of cilostazol to that of the inter- brane filtration method: it meets the requirement.
nal standard.
Assay Measure exactly a volume of Clindamycin Phos-
Amount (mg) of cilostazol (C20H27N5O2)=WS×(QT/QS) phate Injection, equivalent to about 0.3 g (potency) of Clin-
damycin Phosphate, and add the mobile phase to make ex-
WS: Amount (mg) of Cilostazol Reference Standard
actly 100 mL. Pipet 7 mL of this solution, add exactly 25 mL
Internal standard solution—A solution of benzophenone in of the internal standard solution and the mobile phase to
methanol (1 in 250). make 100 mL, and use this solution as the sample solution.
Operating conditions— Separately, weigh accurately an amount of Clindamycin
Proceed as directed in the operating conditions in the As- Phosphate Reference Standard, equivalent to about 20 mg
say under Cilostazol. (potency), dissolve in exactly 25 mL of the internal standard
System suitability— solution, add the mobile phase to make 100 mL, and use this
System performance: Proceed as directed in the system solution as the standard solution. Then, proceed as directed
suitability in the Assay under Cilostazol. in the Assay under Clindamycin Phosphate.
System repeatability: When the test is repeated 6 times
Amount [mg (potency)] of clindamycin phosphate
with 10 mL of the standard solution under the above operat-
(C18H34ClN2O8PS)
ing conditions, the relative standard deviation of the ratio of
=WS×(QT/QS)×(100/7)
the peak area of cilostazol to that of the internal standard is
not more than 1.5z. WS: Amount [mg (potency)] of Clindamycin Phosphate
Reference Standard

Internal standard solution—A solution of methyl para-


Add the following:
hydroxybenzoate in the mobile phase (3 in 50,000).

Clindamycin Phosphate Injection Containers and storage Containers—Hermetic containers.

クリンダマイシンリン酸エステル注射液

Clindamycin Phosphate Injection is an aqueous in-


jection.
It contains not less than 90.0z and not more than
110.0z of the labeled amount of clindamycin phos-
phate (C18H34ClN2O8PS: 504.96).
Supplement I, JP XV Official Monographs 1867

Add the following: Operating conditions—


Detector, column, column temperature, mobile phase,
Clobetasol Propionate and flow rate: Proceed as directed in the operating condi-
tions in the Assay.
クロベタゾールプロピオン酸エステル Time span of measurement: About 2.5 times as long as the
retention time of clobetasol propionate, beginning after the
solvent peak.
System suitability—
Test for required detectability: Pipet 2 mL of the standard
solution, and add the mobile phase to make exactly 50 mL.
Confirm that the peak area of clobetasol propionate ob-
tained from 10 mL of this solution is equivalent to 2.8 to 5.2
C25H32ClFO5: 466.97 z of that from 10 mL of the standard solution.
21-Chloro-9-fluoro-11b,17-dihydroxy- System performance: Dissolve 20 mg of Clobetasol
16b-methylpregna-1,4-diene-3,20-dione 17-propanoate Propionate in 20 mL of methanol. To 5 mL of this solution
[25122-46-7] add 10 mL of a solution of beclometasone dipropionate in
methanol (1 in 1000), and then add the mobile phase to make
Clobetasol Propionate, when dried, contains not 50 mL. When the procedure is run with 10 mL of this solu-
less than 97.0z and not more than 102.0z of tion under the above conditions, clobetasol propionate and
C25H32ClFO5. beclometasone dipropionate are eluted in this order with the
resolution between these peaks being not less than 8.
Description Clobetasol Propionate occurs as a white to
System repeatability: When the test is repeated 6 times
pale yellowish white crystalline powder.
with 10 mL of the standard solution under the above condi-
It is soluble in methanol and in ethanol (99.5), and practi-
tions, the relative standard deviation of the peak area of
cally insoluble in water.
clobetasol propionate is not more than 2.0z.
It gradually turns yellow by light.
Melting point: about 1969 C (with decomposition). Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 3
hours).
Identification Determine the infrared absorption spectra
of Clobetasol Propionate as directed in the paste method Residue on ignition <2.44> Not more than 0.1z (1 g, plati-
under Infrared Spectrophotometry <2.25>, and compare the num crucible).
spectrum with the Reference Spectrum or the spectrum of
Assay Weigh accurately about 10 mg each of Clobetasol
Clobetasol Propionate Reference Standard: both spectra
Propionate and Clobetasol Propionate Reference Standard,
exhibit similar intensities of absorbance at the same wave
both previously dried, dissolve each in the mobile phase, add
numbers.
exactly 100 mL of the internal standard solution, add the
Optical rotation <2.49> [a]20
D : +109 – +1159(after drying, mobile phase to make 250 mL, and use these solutions as the
0.1 g, methanol, 10 mL, 100 mm). sample solution and standard solution. Perform the test with
10 mL each of the sample solution and standard solution as
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
directed under Liquid Chromatography <2.01> according to
Clobetasol Propionate according to Method 2, and perform
the following conditions, and calculate the ratios, QT and
the test. Prepare the control solution with 2.0 mL of Stan-
QS, of the peak area of clobetasol propionate to that of the
dard Lead Solution (not more than 20 ppm).
internal standard.
(2) Related substances—Dissolve 10 mg of Clobetasol
Propionate in 100 mL of the mobile phase, and use this solu- Amount (mg) of C25H32ClFO5
tion as the sample solution. Pipet 5 mL of the sample solu- =WS×(QT/QS)
tion, add the mobile phase to make exactly 200 mL, and use
WS: Amount (mg) of Clobetasol Propionate Reference
this solution as the standard solution. Perform the test with
Standard
exactly 10 mL each of the sample solution and standard solu-
tion as directed under Liquid Chromatography <2.01> ac- Internal standard solution—A solution of beclometasone
cording to the following conditions, and determine each dipropionate in the mobile phase (1 in 5000).
peak area of these solutions by the automatic integration Operating conditions—
method: the area of the peak other than clobetasol Detector: An ultraviolet absorption photometer (wave-
propionate obtained from the sample solution is not larger length: 240 nm).
than 2/5 times the peak area of clobetasol propionate from Column: A stainless steel column 4.6 mm in inside di-
the standard solution. Furthermore, the total area of the ameter and 15 cm in length, packed with octadecylsilanized
peaks other than clobetasol propionate from the sample so- silica gel for liquid chromatography (5 mm in particle di-
lution is not larger than the peak area of clobetasol ameter).
propionate from the standard solution. Column temperature: A constant temperature of about
259C.
1868 Official Monographs Supplement I, JP XV

Mobile phase: Dissolve 7.80 g of sodium dihydrogen (2) Determine the absorption spectrum of a solution of
phosphate dihydrate in 900 mL of water, adjust the pH to Clorazepate Dipotassium (1 in 200,000) as directed under
2.5 with phosphoric acid, and then add water to make 1000 Ultraviolet-visible Spectrophotometry <2.24>, and compare
mL. To 425 mL of this solution add 475 mL of acetonitrile the spectrum with the Reference Spectrum: both spectra
and 100 mL of methanol. exhibit similar intensities of absorption at the same wave-
Flow rate: Adjust the flow rate so that the retention time lengths.
of clobetasol propionate is about 10 minutes. (3) Determine the infrared absorption spectrum of
System suitability— Clorazepate Dipotassium as directed in the potassium
System performance: When the procedure is run with 10 bromide disk method under Infrared Spectrophotometry
mL of the standard solution under the above conditions, <2.25>, and compare the spectrum with the Reference Spec-
clobetasol propionate and the internal standard are eluted in trum: both spectra exhibit similar intensities of absorption at
this order with the resolution between these peaks being not the same wave numbers.
less than 8. (4) Clorazepate Dipotassium responds to the Qualitative
System repeatability: When the test is repeated 6 times Tests <1.09> (1) for potassium salt.
with 10 mL of the standard solution under the above condi-
Purity (1) Chloride <1.03>—Dissolve 1.0 g of Cloraze-
tions, the relative standard deviation of the ratio of the peak
pate Dipotassium in 20 mL of water, add 20 mL of acetone,
area of clobetasol propionate to that of the internal standard
6 mL of dilute nitric acid and water to make 50 mL. Perform
is not more than 1.0z.
the test with this solution as the test solution. Prepare the
Containers and storage Containers—Tight containers. control solution as follows: To 0.40 mL of 0.01 mol/L
Storage—Light-resistant. hydrochloric acid VS add 20 mL of acetone, 6 mL of dilute
nitric acid and water to make 50 mL (not more than 0.014
z).
Add the following: (2) Heavy metals <1.07>—Proceed with 1.0 g of Cloraze-
pate Dipotassium according to Method 2, and perform the
Clorazepate Dipotassium test. Prepare the control solution with 2.0 mL of Standard
Lead Solution (not more than 20 ppm).
クロラゼプ酸二カリウム (3) Arsenic <1.11>—Prepare the test solution with 1.0 g
of Clorazepate Dipotassium according to Method 3, and
perform the test (not more than 2 ppm).
(4) Related substances—Dissolve 15 mg of Clorazepate
Dipotassium in 25 mL of a mixture of water, potassium car-
bonate solution (97 in 1000) and acetonitrile (3:1:1), and use
this solution as the sample solution. Pipet 1 mL of the sam-
ple solution, add the mixture of water, potassium carbonate
C16H10ClKN2O3.KOH: 408.92 solution (97 in 1000) and acetonitrile (3:1:1) to make exactly
Monopotassium 7-chloro-2-oxo-5-phenyl-2,3-dihydro- 200 mL, and use this solution as the standard solution. Pre-
1H-1,4-benzodiazepine-3-carboxylate pare these solutions quickly and perform the test within 3
mono (potassium hydroxide) [57109-90-7] minutes. Perform the test with exactly 5 mL each of the sam-
ple solution and standard solution as directed under Liquid
Clorazepate Dipotassium, when dried, contains not Chromatography <2.01> according to the following condi-
less than 98.5z and not more than 101.0z of tions, and determine each peak area by the automatic in-
C16H10ClKN2O3.KOH. tegration method: the peak area of nordiazepam, having the
Description Clorazepate Dipotassium occurs as white to relative retention time of about 3.0 with respect to clorazepic
light yellow, crystals or crystalline powder. acid, obtained from the sample solution is not larger than
It is freely soluble in water, and very slightly soluble in the peak area of clorazepic acid from the standard solution,
ethanol (99.5). the area of the peak other than clorazepic acid and nordia-
It dissolves in acetic acid (100). zepam is not larger than 1/5 times the peak area of clorazep-
The pH of a solution obtained by dissolving 1 g of ic acid from the standard solution, and the total area of the
Clorazepate Dipotassium in 100 mL of water is between 11.5 peaks other than clorazepic acid from the sample solution is
and 12.5. not larger than 2 times the peak area of clorazepic acid from
It gradually turns yellow on exposure to light. the standard solution. For this comparison, use the peak
area of nordiazepam, having the relative retention time of
Identification (1) Carefully and gradually ignite to red- about 3.0 with respect to clorazepic acid, after multiplying
ness 30 mg of Clorazepate Dipotassium with 50 mg of sodi- by the relative response factor, 0.64.
um. After cooling, add 3 drops of ethanol (99.5) and 5 mL Operating conditions—
of water, mix well, and filter: the filtrate responds to the Detector: An ultraviolet absorption photometer (wave-
Qualitative Tests <1.09> for chloride. length: 232 nm).
Supplement I, JP XV Official Monographs 1869

Column: A stainless steel column 4.6 mm in inside di- Method of preparation Prepare as directed under Cap-
ameter and 15 cm in length, packed with octadecylsilanized sules, with Clorazepate Dipotassium.
silica gel for liquid chromatography (5 mm in particle di-
Identification To 10 mL of the sample solution obtained in
ameter).
the Assay add water to make 20 mL. Determine the absorp-
Column temperature: A constant temperature of about
tion spectrum of this solution as directed under Ultraviolet-
259C.
visible Spectrophotometry <2.24>: it exhibits a maximum be-
Mobile phase: Dissolve 13.8 g of sodium dihydrogen
tween 228 nm and 232 nm.
phosphate dihydrate in 500 mL of water, and adjust to pH
8.0 with sodium hydroxide TS. To 100 mL of this solution Purity Related substances—Take out the contents of
add 400 mL of acetonitrile and 300 mL of water. Clorazepate Dipotassium Capsules, and powder. To a por-
Flow rate: Adjust the flow rate so that the retention time tion of the powder, equivalent to 15 mg of Clorazepate
of clorazepic acid is about 1.3 minutes. Dipotassium according to the labeled amount, add a mixture
Time span of measurement: About 10 times as long as the of water, potassium carbonate solution (97 in 1000) and
retention time of clorazepic acid, beginning after the solvent acetonitrile (3:1:1) to make 25 mL, and mix for 10 minutes.
peak. Filter the solution through a membrane filter with a pore size
System suitability— not exceeding 0.45 mm, discard the first 5 mL of the filtrate,
Test for required detectability: To exactly 5 mL of the and use the subsequent filtrate as the sample solution. Pipet
standard solution add the mixture of water, potassium car- 1 mL of the sample solution, add a mixture of water, potas-
bonate solution (97 in 1000) and acetonitrile (3:1:1) to make sium carbonate solution (97 in 1000) and acetonitrile (3:1:1)
exactly 25 mL. Confirm that the peak area of clorazepic acid to make exactly 200 mL, and use this solution as the stan-
obtained from 5 mL of this solution is equivalent to 15 to 25 dard solution. Then, proceed as directed in the Purity (4) un-
z of that from 5 mL of the standard solution. der Clorazepate Dipotassium: the peak area of nordiazep-
System performance: When the procedure is run with 5 mL am, having the relative retention time of about 3.0 with
of the standard solution under the above operating condi- respect to clorazepic acid, obtained from the sample solution
tions, the number of theoretical plates and the symmetry is not larger than 3 times the peak area of clorazepic acid
factor of the peak of clorazepic acid are not less than 3000 from the standard solution, and the total area of the peaks
and not more than 1.5, respectively. other than clorazepic acid and nordiazepam from the sample
System repeatability: When the test is repeated 6 times solution is not larger than the peak area of clorazepic acid
with 5 mL of the standard solution under the above operat- from the standard solution. For this comparison, use the
ing conditions, the relative standard deviation of the peak peak area of nordiazepam after multiplying by the relative
area of clorazepic acid is not more than 1.5z. response factor, 0.64.

Loss on drying <2.41> Not more than 0.5z (1 g, in vacu- Uniformity of dosage units <6.02> Perform the test accord-
um, phosphorus (V) oxide, 609
C, 5 hours). ing to the following method: it meets the requirement of the
Content uniformity test.
Assay Weigh accurately about 0.15 g of Clorazepate
To 1 capsule of Clorazepate Dipotassium Capsules add 70
Dipotassium, previously dried, dissolve in 100 mL of acetic
mL of water, shake for 15 minutes, and add water to make
acid (100), and titrate <2.50> with 0.1 mol/L perchloric acid
exactly 100 mL. Centrifuge the solution, pipet V mL of the
VS until the color of solution changes from violet to blue-
supernatant liquid, add water to make exactly V? mL so that
green through blue (indicator: 3 drops of crystal violet TS).
each mL contains about 12 mg of clorazepate dipotassium
Perform a blank determination in the same manner, and
(C16H10ClKN2O3.KOH), and use this solution as the sample
make any necessary correction.
solution. Then, proceed as directed in the Assay.
Each mL of 0.1 mol/L perchloric acid VS
Amount (mg) of clorazepate dipotassium
=13.63 mg of C16H10ClKN2O3.KOH
(C16H10ClKN2O3.KOH)
Containers and storage Containers—Tight containers. =WS×(AT/AS)×(V?/V)×(2/25)
Storage—Light-resistant.
WS: Amount (mg) of clorazepate dipotassium for assay
Dissolution <6.10> When the test is performed at 50 revolu-
Add the following: tions per minute according to the Paddle method using the
sinker, using 900 mL of water as the dissolution medium,
Clorazepate Dipotassium Capsules the dissolution rate in 30 minutes of Clorazepate Dipotassi-
um Capsules is not less than 80z.
クロラゼプ酸二カリウムカプセル Start the test with 1 capsule of Clorazepate Dipotassium
Capsules, withdraw not less than 20 mL of the medium at
Clorazepate Dipotassium Capsules contain not less the specified minute after starting the test, and filter through
than 93.0z and not more than 107.0z of the labeled a membrane filter with a pore size not exceeding 0.45 mm.
amount of clorazepate dipotassium (C16H10ClKN2O3. Discard the first 10 mL of the filtrate, pipet V mL of the
KOH: 408.92). subsequent filtrate, add water to make exactly V? mL so that
1870 Official Monographs Supplement I, JP XV

each mL contains about 8.3 mg of clorazepate dipotassium


(C16H10ClKN2O3.KOH) according to the labeled amount, Cyanocobalamin
and use this solution as the sample solution. Separately,
weigh accurately about 21 mg of clorazepate dipotassium シアノコバラミン
for assay, previously dried in vacuum over phosphorus (V)
oxide at 609C for 5 hours, and dissolve in water to make ex- Change the Identification (2) and (3) to read:
actly 100 mL. Pipet 4 mL of this solution, add water to
Identification
make exactly 100 mL, and use this solution as the standard
(2) Mix 1 mg of Cyanocobalamin with 50 mg of potassi-
solution. Determine the absorbances, AT and AS, at 252 nm
um hydrogen sulfate, and fuse by igniting. Cool, break up
of the sample solution and standard solution as directed un-
the mass with a glass rod, add 3 mL of water, and dissolve
der Ultraviolet-visible Spectrophotometry <2.24>.
by boiling. Add 1 drop of phenolphthalein TS, then add
Dissolution rate (z) with respect to the labeled amount of dropwise sodium hydroxide TS until a light red color just
clorazepate dipotassium (C16H10ClKN2O3.KOH) develops. Add 0.5 g of sodium acetate trihydrate, 0.5 mL of
=WS×(AT/AS)×(V?/V)×(1/C)×36 dilute acetic acid and 0.5 mL of a solution of disodium 1-
nitroso-2-naphthol-3,6-disulfonate (1 in 500): a red to oran-
WS: Amount (mg) of clorazepate dipotassium for assay
ge-red color is immediately produced. Then add 0.5 mL of
C: Labeled amount (mg) of clorazepate dipotassium
hydrochloric acid, and boil for 1 minute: the red color does
(C16H10ClKN2O3.KOH) in 1 capsule
not disappear.
Assay Carefully take out the contents of not less than 20 (3) Transfer 5 mg of Cyanocobalamin to a 50-mL distil-
Clorazepate Dipotassium Capsules, weigh accurately the ling flask, dissolve in 5 mL of water, and add 2.5 mL of
mass of the contents, and powder. Weigh accurately a por- hypophosphorous acid. Connect the flask with a short con-
tion of the powder, equivalent to about 15 mg of clorazepate denser, and dips its tip into a test tube containing 1 mL of a
dipotassium (C16H10ClKN2O3.KOH), add 70 mL of water, solution of sodium hydroxide (1 in 50). Heat gently for 10
shake for 15 minutes, and add water to make exactly 100 minutes, then distil 1 mL into a test tube. To the test tube
mL. Centrifuge the solution, pipet 4 mL of the supernatant add 4 drops of a saturated solution of ammonium iron (II)
liquid, add water to make exactly 50 mL, and use this solu- sulfate hexahydrate, shake gently, then add about 30 mg of
tion as the sample solution. Separately, weigh accurately sodium fluoride, and heat the contents to boil. Immediately
about 15 mg of clorazepate dipotassium for assay, previous- add dropwise diluted sulfuric acid (1 in 7) until a clear solu-
ly dried in vacuum over phosphorus (V) oxide at 609 C for 5 tion results, then add 3 to 5 drops more of diluted sulfuric
hours, and dissolve in water to make exactly 100 mL. Pipet acid (1 in 7): a blue to blue-green color develops.
4 mL of this solution, add water to make exactly 50 mL, and
use this solution as the standard solution. Perform the test Change the Purity (2) to read:
with the sample solution and standard solution as directed
Purity
under Ultraviolet-visible Spectrophotometry <2.24>, and
(2) Related substances—Conduct this procedure using
determine the absorbances, AT and AS, at 252 nm.
light-resistant vessels. Dissolve 10 mg of Cyanocobalamin in
Amount (mg) of clorazepate dipotassium 10 mL of the mobile phase, and use this solution as the sam-
(C16H10ClKN2O3.KOH) ple solution. Pipet 3 mL of the sample solution, add the mo-
=WS×(AT/AS) bile phase to make exactly 100 mL, and use this solution as
the standard solution. Perform the test with exactly 20 mL
WS: Amount (mg) of clorazepate dipotassium for assay
each of the sample solution and standard solution as direct-
Containers and storage Containers—Tight containers. ed under Liquid Chromatography <2.01> according to the
following conditions, and determine each peak area by the
automatic integration method: the total area of the peak
Creosote other than cyanocobalamin obtained from the sample solu-
tion is not larger than the peak area of cyanocobalamin from
the standard solution.
Change the title of the monograph as follows:
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Wood Creosote length: 361 nm).
木クレオソート Column: A stainless steel column 4.6 mm in inside di-
ameter and 25 cm in length, packed with octylsilanized silica
gel for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
309C.
Mobile phase: Dissolve 10 g of anhydrous disodium
hydrogen phosphate in 1000 mL of water, and adjust to pH
3.5 with phosphoric acid. To 147 mL of this solution add 53
Supplement I, JP XV Official Monographs 1871

mL of methanol. Add the following:


Flow rate: Adjust the flow rate so that the retention time
of cyanocobalamin is about 7 minutes. L-Cysteine
Time span of measurement: About 4 times as long as the
retention time of cyanocobalamin, beginning after the sol- L-システイン
vent peak.
System suitability—
Test for required detectability: Pipet 1 mL of the sample
solution, add the mobile phase to make exactly 100 mL, and C3H7NO2S: 121.16
use this solution as the solution for system suitability test. (2R)-2-Amino-3-sulfanylpropanoic acid [52-90-4]
Pipet 1 mL of the solution for system suitability test, and
add the mobile phase to make exactly 10 mL. Confirm that L-Cysteine contains not less than 98.5z and not
the peak area of cyanocobalamin obtained with 20 mL of this more than 101.0z of C3H7NO2S, calculated on the
solution is equivalent to 7 to 13z of that with 20 mL of the dried basis.
solution for system suitability test.
Description L-Cysteine occurs as white crystals or a white
System performance: Perform this procedure quickly
crystalline powder. It has a characteristic odor and a pun-
after the solution is prepared. To 25 mg of cyanocobalamin
gent taste.
add 10 mL of water, and warm, if necessary, to dissolve.
It is freely soluble in water, and practically insoluble in
After cooling, add 0.5 mL of sodium toluenesulfon-
ethanol (99.5).
chloramide TS, 0.5 mL of 0.05 mol/L hydrochloric acid TS
It dissolves in 1 mol/L hydrochloric acid TS.
and water to make 25 mL, mix, and allow the solution to
stand for 5 minutes. To 1 mL of the solution add the mobile Identification Determine the infrared absorption spectrum
phase to make 10 mL. When the procedure is run with 20 mL of L-Cysteine as directed in the potassium bromide disk
of the solution under the above operating conditions, two method under Infrared Spectrophotometry <2.25>, and com-
principal peaks appear with the resolution between these pare the spectrum with the Reference Spectrum: both spec-
peaks being not less than 2.5. tra exhibit similar intensities of absorption at the same wave
System repeatability: When the test is repeated 6 times numbers.
with 20 mL of the solution for system suitability test under
Optical rotation <2.49> [a]20D : +8.0 – +10.09(2 g calculat-
the above operating conditions, the relative standard devia-
ed on the dried basis, 1 mol/L hydrochloric acid TS, 25 mL,
tion of the peak area of cyanocobalamin is not more than
100 mm).
3.0z.
pH <2.54> The pH of a solution prepared by dissolving
1.25 g of L-Cysteine in 50 mL of water is 4.7 to 5.7.
Cyanocobalamin Injection Purity (1) Clarity and color of solution—Dissolve 1.0 g
シアノコバラミン注射液 of L-Cysteine in 20 mL of water: the solution is clear and
colorless.
Change the Description to read: (2) Chloride <1.03>—Dissolve 0.30 g of L-Cysteine in 10
mL of diluted nitric acid (1 in 4), add 10 mL of hydrogen
Description Cyanocobalamin Injection is a clear, light red peroxide (30), heat for 20 minutes in a boiling water bath,
to red liquid. cool, and then add water to make 50 mL. Perform the test
using this solution as the test solution. Prepare the control
Add the following next to Identification: solution with 0.35 mL of 0.01 mol/L hydrochloric acid VS
Bacterial endotoxins <4.01> Less than 0.30 EU/mg. (not more than 0.041z).
(3) Sulfate <1.14>—Dissolve 0.6 g of L-Cysteine in 30
Add the following next to Extractable volume: mL of water and 3 mL of dilute hydrochloric acid, and add
water to make 50 mL. Perform the test using this solution as
Foreign insoluble matter <6.06> Perform the test according the test solution. Prepare the control solution as follows: To
to Method 1: it meets the requirement. 0.35 mL of 0.005 mol/L sulfuric acid VS add 3 mL of dilute
Insoluble particulate matter <6.07> It meets the require- hydrochloric acid and water to make 50 mL. Prepare the test
ment. solution and the control solution with 4 mL of barium chlo-
ride TS, respectively (not more than 0.028z).
Sterility <4.06> Perform the test according to the Mem- (4) Ammonium <1.02>—Perform the test with 0.25 g of
brane filtration method: it meets the requirement. L-Cysteine, using the distillation under reduced pressure.
Prepare the control solution with 5.0 mL of Standard Am-
monium Solution (not more than 0.02z).
(5) Heavy metals <1.07>—Proceed with 1.0 g of
L-Cysteine according to Method 4, and perform the test.
1872 Official Monographs Supplement I, JP XV

Prepare the control solution with 1.0 mL of Standard Lead Add the following:
Solution (not more than 10 ppm).
(6) Iron <1.10>—Prepare the test solution with 1.0 g of L-Cysteine Hydrochloride Hydrate
L-Cysteine according to Method 1, and perform the test us-
ing Method A. Prepare the control solution with 1.0 mL of L-システイン塩酸塩水和物
Standard Iron Solution (not more than 10 ppm).
(7) Related substances—Dissolve 0.10 g of L-Cysteine in
N-ethylmaleimide solution (1 in 50) to make 10 mL, leave
for 30 minutes, and use this solution as the sample solution. C3H7NO2S.HCl.H2O: 175.63
Pipet 1 mL of the sample solution, add water to make exact- (2R)-2-Amino-3-sulfanylpropanoic acid monohydrochloride
ly 10 mL, pipet 1 mL of this solution, add water to make monohydrate [7048-04-6]
exactly 50 mL, and use this solution as the standard solution
(1). Separately, dissolve 0.10 g of L-cystine in 0.5 mol/L L-Cysteine Hydrochloride Hydrate contains not less
hydrochloric acid TS to make 20 mL. Pipet 1 mL of this than 98.5z and not more than 101.0z of L-cysteine
solution, add water to make 100 mL, and use this solution as hydrochloride (C3H7NO2S.HCl: 157.62), calculated
the standard solution (2). Perform the test with these solu- on the dried basis.
tions as directed under Thin-layer Chromatography <2.03>.
Description L-Cysteine Hydrochloride Hydrate occurs as
Spot 10 mL each of the sample solution and standard solu-
white crystals or crystalline powder. It has a characteristic
tions (1) and (2) on a plate of silica gel for thin-layer chro-
odor and a strong acid taste.
matography. Develop the plate with a mixture of 1-butanol,
It is very soluble in water, and soluble in ethanol (99.5).
water, and acetic acid (100) (3:1:1) to a distance of about 10
It dissolves in 6 mol/L hydrochloric acid TS.
cm, and dry the plate for 30 minutes at 809 C. Spray the plate
evenly with a solution of ninhydrin in a mixture of methanol Identification (1) Determine the infrared absorption
and acetic acid (100) (97:3) (1 in 100), and then heat at 809C spectrum of L-Cysteine Hydrochloride Hydrate as directed
for 10 minutes: the spot obtained from the sample solution in the potassium chloride disk method under Infrared Spec-
corresponding to the spot obtained from the standard solu- trophotometry <2.25>, and compare the spectrum with the
tion (2) is not more intense than the spot from the standard Reference Spectrum: both spectra exhibit similar intensities
solution (2). Also, the spots other than the principal spot of absorption at the same wave numbers.
and the spots mentioned above from the sample solution are (2) To 10 mL of a solution of L-Cysteine Hydrochloride
not more intense than the spot from the standard solution Hydrate (1 in 50) add 1 mL of hydrogen peroxide (30), heat
(1). on a water bath for 20 minutes, and cool: the solution
responds to the Qualitative Tests <1.09> (2) for chloride.
Loss on drying <2.41> Not more than 0.5z (1 g, in vacu-
um, phosphorus (V) oxide, 3 hours). Optical rotation <2.49> [a]20D : +6.0 – +7.59(2 g, calculat-
ed on the dried basis, 6 mol/L hydrochloric acid TS, 25 mL,
Residue on ignition <2.44> Not more than 0.1z (1 g).
100 mm).
Assay Weigh accurately about 0.2 g of L-Cysteine, place it
pH <2.54> The pH of a solution prepared by dissolving
in a stoppered flask, and dissolve in 20 mL of water. Dis-
1.0 g of L-Cysteine Hydrochloride Hydrate in 100 mL of
solve 4 g of potassium iodide in this solution, immediately
water is between 1.3 and 2.3.
place in ice cold water, add 5 mL of dilute hydrochloric acid
and exactly 25 mL of 0.05 mol/L iodine VS, leave in a dark Purity (1) Clarity and color of solution—A solution
place for 20 minutes, and then titrate <2.50> an excess obtained by dissolving 1.0 g of L-Cysteine Hydrochloride
amount of iodine with 0.1 mol/L sodium thiosulfate VS (in- Hydrate in 10 mL of water is clear and colorless.
dicator: starch TS). Perform a blank determination using (2) Sulfate < 1.14 > —Dissolve 0.8 g of L-Cysteine
the same method. Hydrochloride Hydrate in 30 mL of water and 3 mL of di-
lute hydrochloric acid, and add water to make 50 mL. Per-
Each mL of 0.05 mol/L iodine VS=12.12 mg of C3H7NO2S
form the test using this solution as the test solution. Prepare
Containers and storage Containers—Tight containers. the control solution as follows: To 0.35 mL of 0.005 mol/L
sulfuric acid VS add 3 mL of dilute hydrochloric acid and
water to make 50 mL. To both of the test solution and the
control solution add 4 mL of barium chloride TS (not more
than 0.021z).
(3) Ammonium <1.02>—Perform the test with 0.25 g of
L-Cysteine Hydrochloride Hydrate using the distillation
under reduced pressure. Prepare the control solution with
5.0 mL of Standard Ammonium Solution (not more than
0.02z).
(4) Heavy metals <1.07>—Proceed with 1.0 g of
Supplement I, JP XV Official Monographs 1873

L-Cysteine Hydrochloride Hydrate according to Method 4,


and perform the test. Prepare the control solution with 1.0 Dehydrocholic Acid Injection
mL of Standard Lead Solution (not more than 10 ppm).
(5) Iron <1.10>—Prepare the test solution with 1.0 g of デヒドロコール酸注射液
L-Cysteine Hydrochloride Hydrate according to Method 1,
and perform the test according to Method A. Prepare the Add the following next to Extractable volume:
control solution with 1.0 mL of Standard Iron Solution (not
Foreign insoluble matter <6.06> Perform the test according
more than 10 ppm).
to Method 1: it meets the requirement.
(6) Related substances—Dissolve 0.10 g of L-Cysteine
Hydrochloride Hydrate in N-ethylmaleimide solution (1 in Insoluble particulate matter <6.07> It meets the require-
50) to make 10 mL, allow to stand for 30 minutes, and use ment.
this solution as the sample solution. Pipet 1 mL of the sam-
Sterility <4.06> Perform the test according to the Mem-
ple solution, add water to make exactly 10 mL. Pipet 1 mL
brane filtration method: it meets the requirement.
of this 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 Thin-Layer Chro-
matography <2.03>. Spot 5 mL each of the sample solution
Deslanoside Injection
and standard solution on a plate of silica gel for thin-layer デスラノシド注射液
chromatography. Then develop with a mixture of 1-butanol,
water and acetic acid (100) (3:1:1) to a distance of about 10 Add the following next to Identification:
cm, and dry the plate at 809C for 30 minutes. Spray evenly a
solution of ninhydrin in a mixture of methanol and acetic Bacterial endotoxins <4.01> Less than 500 EU/mg.
acid (100) (97:3) (1 in 100) on the plate, and then heat at
809C for 10 minutes: the spot other than the principal spot Add the following next to Extractable volume:
obtained with the sample solution is not more intense than Foreign insoluble matter <6.06> Perform the test according
the spot with the standard solution. to Method 1: it meets the requirement.
Loss on drying <2.41> 8.5 – 12.0z (1 g, in vacuum, phos- Insoluble particulate matter <6.07> It meets the require-
phorus (V) oxide, 20 hours). ment.
Residue on ignition <2.44> Not more than 0.1z (1 g). Sterility <4.06> Perform the test according to the Mem-
Assay Weigh accurately about 0.25 g of L-Cysteine brane filtration method: it meets the requirement.
Hydrochloride Hydrate, place in a glass-stoppered flask,
and dissolve in 20 mL of water. Dissolve 4 g of potassium
iodide in this solution, soak immediately in ice cold water, Dextran 40
add 5 mL of dilute hydrochloric acid and exactly 25 mL of
0.05 mol/L iodine VS, allow to stand for 20 minutes in a デキストラン 40
dark place, titrate <2.50> the excess of iodine with 0.1 mol/L
sodium thiosulfate VS (indicator: starch TS). Perform a Delete the Pyrogen and add the following next
blank determination in the same manner, and make any to Residue on ignition:
necessary correction. Bacterial endotoxins <4.01> Less than 2.5 EU/g.
Each mL of 0.05 mol/L iodine VS
=15.76 mg of C3H7NO2S.HCl

Containers and storage Containers—Tight containers.

Deferoxamine Mesilate
デフェロキサミンメシル酸塩

Change the Identification (2) to read:


Identification
(2) A 50 mg portion of Deferoxamine Mesilate responds
to the Qualitative Tests <1.09> (1) for mesilate.
1874 Official Monographs Supplement I, JP XV

Change to read: um Phosphate with 5 mL of freshly boiled and cooled water,


and add immediately 2 mL of hydrochloric acid: no efferves-
Anhydrous Dibasic Calcium cence occurs.
(5) Heavy metals <1.07>—Dissolve 0.65 g of Anhy-
Phosphate drous Dibasic Calcium Phosphate in a mixture of 5 mL of
無水リン酸水素カルシウム water and 5 mL of dilute hydrochloric acid by warming,
cool, and add ammonia TS until precipitates begin to form
in the solution. Dissolve the precipitates by adding a small
CaHPO4: 136.06
amount of dilute hydrochloric acid dropwise, add 10 mL of
[7757-93-9]
hydrochloric acid-ammonium acetate buffer solution, pH
This monograph is harmonized with the European Phar- 3.5, and water to make 50 mL, and perform the test using
macopoeia and the U.S. Pharmacopeia. The parts of the text this solution as the test solution. Prepare the control solu-
that are not harmonized are marked with symbols ( ). tion as follows: to 10 mL of hydrochloric acid-ammonium
acetate buffer solution, pH 3.5, add 2.0 mL of Standard
Anhydrous Dibasic Calcium Phosphate contains Lead Solution and water to make 50 mL (not more than 31
not less than 98.0z and not more than 103.0z of ppm).
CaHPO4. (6) Barium—Heat 0.5 g of Anhydrous Dibasic Calcium
Phosphate with 10 mL of water, add 1 mL of hydrochloric
Description Anhydrous Dibasic Calcium Phosphate oc-
acid dropwise with stirring, and filter after cooling, if neces-
curs as white, crystalline powder or granules.
sary. Add 2 mL of potassium sulfate TS to this solution, and
It is practically insoluble in water and in ethanol (99.5).
allow to stand for 10 minutes: no turbidity forms.
It dissolves in dilute hydrochloric acid and in dilute nitric 
(7) Arsenic <1.11>—Dissolve 1.0 g of Anhydrous Di-
acid.
basic Calcium Phosphate in 5 mL of dilute hydrochloric
Identification (1) Dissolve 0.1 g of Anhydrous Dibasic acid, and perform the test with this solution as the test solu-
Calcium Phosphate in 10 mL of 2 mol/L hydrochloric acid tion (not more than 2 ppm).
TS by warming, add 2.5 mL of ammonia TS dropwise with
Loss on ignition <2.43> Not less than 6.6z and not more
shaking, and add 5 mL of ammonium oxalate TS: a white
than 8.5z (1 g, 800 – 8259
C, constant mass).
precipitate is produced.
(2) Dissolve 0.1 g of Anhydrous Dibasic Calcium Phos- Assay Weigh accurately about 0.4 g of Anhydrous Dibasic
phate in 5 mL of dilute nitric acid, and add 2 mL of hexaam- Calcium Phosphate, dissolve in 12 mL of dilute hydrochlor-
monium heptamolybdate TS after warming at 709 C for 1 to ic acid, and add water to make exactly 200 mL. Pipet 20 mL
2 minutes: a yellow precipitate is produced. of this solution, add exactly 25 mL of 0.02 mol/L disodium
dihydrogen ethylenediamine tetraacetate VS, 50 mL of
Purity (1) Acid-insoluble substances—Dissolve 5.0 g of
water and 5 mL of ammonia-ammonium chloride buffer
Anhydrous Dibasic Calcium Phosphate in 40 mL of water
solution, pH 10.7, and titrate <2.50> the excess of disodium
and 10 mL of hydrochloric acid, and boil gently for 5
dihydrogen ethylenediamine tetraacetate with 0.02 mol/L
minutes. After cooling, collect the insoluble substance using
zinc sulfate VS (indicator: 25 mg of eriochrome black T-so-
a filter paper for assay. Wash with water until no more
dium chloride indicator). Perform a blank determination in
turbidity of the washings is produced when silver nitrate TS
the same manner.
is added. Ignite to incinerate the residue and the filter paper
at 600±509 C: the mass is not more than 10 mg (not more Each mL of 0.02 mol/L disodium dihydrogen ethylenedia-
than 0.2z). mine tetraacetate VS
(2) Chloride <1.03>—Dissolve 0.20 g of Anhydrous Di- =2.721 mg of CaHPO4
basic Calcium Phosphate in 20 mL of water and 13 mL of Containers and storage Containers—Well-closed con-
dilute nitric acid, add water to make 100 mL, and filter, if
tainers.
necessary. Perform the test using a 50-mL portion of this so-
lution as the test solution. Prepare the control solution with
0.70 mL of 0.01 mol/L hydrochloric acid VS (not more than
0.248z).
(3) Sulfate <1.14>—Dissolve 0.5 g of Anhydrous Dibasic
Calcium Phosphate in 5 mL of water and 5 mL of dilute
hydrochloric acid, add water to make 100 mL, and filter, if
necessary. Take 20 mL of this solution, add 1 mL of dilute
hydrochloric acid, and add water to make 50 mL. Perform
the test using this solution as the test solution. Prepare the
control solution with 1.0 mL of 0.005 mol/L sulfuric acid
VS (not more than 0.48z).
(4) Carbonate—Mix 1.0 g of Anhydrous Dibasic Calci-
Supplement I, JP XV Official Monographs 1875

Change to read: and add immediately 2 mL of hydrochloric acid: no efferves-


cence occurs.
Dibasic Calcium Phosphate (5) Heavy metals <1.07>—Dissolve 0.65 g of Dibasic
Calcium Phosphate Hydrate in a mixture of 5 mL of water
Hydrate and 5 mL of dilute hydrochloric acid by warming, cool, and
リン酸水素カルシウム水和物 add ammonia TS until precipitates begin to form in the solu-
tion. Dissolve the precipitates by adding a small amount of
dilute hydrochloric acid dropwise, add 10 mL of
CaHPO4.2H2O: 172.09 hydrochloric acid-ammonium acetate buffer solution, pH
[7789-77-7] 3.5, and water to make 50 mL, and perform the test using
This monograph is harmonized with the European Phar- this solution as the test solution. Prepare the control solu-
macopoeia and the U.S. Pharmacopeia. The parts of the text tion as follows: to 10 mL of hydrochloric acid-ammonium
that are not harmonized are marked with symbols ( ). acetate buffer solution, pH 3.5, add 2.0 mL of Standard
Lead Solution and water to make 50 mL (not more than 31
Dibasic Calcium Phosphate Hydrate contains not ppm).
less than 98.0z and not more than 105.0z of (6) Barium—Heat 0.5 g of Dibasic Calcium Phosphate
CaHPO4.2H2O. Hydrate with 10 mL of water, add 1 mL of hydrochloric
acid dropwise with stirring, and filter after cooling, if neces-

Description Dibasic Calcium Phosphate Hydrate occurs sary. Add 2 mL of potassium sulfate TS to this solution, and
as a white, crystalline powder. allow to stand for 10 minutes: no turbidity forms.
It is practically insoluble in water and in ethanol (99.5). (7) Arsenic <1.11>—Dissolve 1.0 g of Dibasic Calcium
It dissolves in dilute hydrochloric acid and in dilute nitric Phosphate Hydrate in 5 mL of dilute hydrochloric acid, and
acid. perform the test with this solution as the test solution (not
Identification (1) Dissolve 0.1 g of Dibasic Calcium more than 2 ppm).
Phosphate Hydrate in 10 mL of 2 mol/L hydrochloric acid Loss on ignition <2.43> Not less than 24.5z and not more
TS by warming, add 2.5 mL of ammonia TS dropwise with than 26.5z (1 g, 800 – 8259C, constant mass).
shaking, and add 5 mL of ammonium oxalate TS: a white
precipitate is produced. Assay Weigh accurately about 0.4 g of Dibasic Calcium
(2) Dissolve 0.1 g of Dibasic Calcium Phosphate Hy- Phosphate Hydrate, dissolve in 12 mL of dilute hydrochloric
drate in 5 mL of dilute nitric acid, and add 2 mL of hexaam- acid, and add water to make exactly 200 mL. Pipet 20 mL of
monium heptamolybdate TS after warming at 709 C for 1 to this solution, add exactly 25 mL of 0.02 mol/L disodium di-
2 minutes: a yellow precipitate is produced. hydrogen ethylenediamine tetraacetate VS, 50 mL of water
and 5 mL of ammonia-ammonium chloride buffer solution,
Purity (1) Acid-insoluble substance—Dissolve 5.0 g of pH 10.7, and titrate <2.50> the excess of disodium dihydro-
Dibasic Calcium Phosphate Hydrate in 40 mL of water and gen ethylenediamine tetraacetate with 0.02 mol/L zinc sul-
10 mL of hydrochloric acid, and boil gently for 5 minutes. fate VS (indicator: 25 mg of eriochrome black T-sodium
After cooling, collect the insoluble substance using a filter chloride indicator). Perform a blank determination in the
paper for assay. Wash with water until no more turbidity of same manner.
the washing is produced when silver nitrate TS is added. Ig-
nite to incinerate the residue and filter paper at 600±509C: Each mL of 0.02 mol/L disodium dihydrogen ethylenedia-
the mass is not more than 10 mg (not more than 0.2z). mine tetraacetate VS
(2) Chloride <1.03>—Dissolve 0.20 g of Dibasic Calcium =3.442 mg of CaHPO4.2H2O
Phosphate Hydrate in 20 mL of water and 13 mL of dilute Containers and storage Containers—Well-closed contain-
nitric acid, add water to make 100 mL, and filter, if necessa- ers.
ry. Perform the test using a 50-mL portion of this solution as
the test solution. Prepare the control solution with 0.70 mL
of 0.01 mol/L hydrochloric acid VS (not more than
0.248z).
(3) Sulfate <1.14>—Dissolve 0.5 g of Dibasic Calcium
Phosphate Hydrate in 5 mL of water and 5 mL of dilute
hydrochloric acid, add water to make 100 mL, and filter, if
necessary. Take 20 mL of this solution, add 1 mL of dilute
hydrochloric acid, and add water to make 50 mL. Perform
the test using this solution as the test solution. Prepare the
control solution with 1.0 mL of 0.005 mol/L sulfuric acid
VS (not more than 0.48z).
(4) Carbonate—Mix 1.0 g of Dibasic Calcium Phos-
phate Hydrate with 5 mL of freshly boiled and cooled water,
1876 Official Monographs Supplement I, JP XV

Add the following: total area of the peaks other than domperidone from the
sample solution is not larger than the peak area of domperi-
Domperidone done from the standard solution.
Operating conditions—
ドンペリドン Detector: An ultraviolet absorption photometer (wave-
length: 287 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 25 cm in length, packed with octylsilanized silica
gel for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
359C.
Mobile phase: Dissolve 2.72 g of dipotassium hydrogen
phosphate in water to make 1000 mL, and adjust the pH to
3.5 of this solution with a solution prepared by dissolving
C22H24ClN5O2: 425.91 2.31 g of phosphoric acid in water to make 1000 mL. To 500
5-Chloro-1-{1-[3-(2-oxo-2,3-dihydro-1H-benzoimidazol- mL of this solution add 500 mL of methanol.
1-yl)propyl]piperidin-4-yl}-1,3-dihydro-2H-benzoimidazol- Flow rate: Adjust the flow rate so that the retention time
2-one [57808-66-9] of domperidone is about 9 minutes.
Time span of measurement: About 4 times as long as the
Domperidone, when dried, contains not less than retention time of domperidone, beginning after the solvent
99.0z and not more than 101.0z of C22H24ClN5O2. peak.
System suitability—
Description Domperidone occurs as a white to pale yellow,
Test for required detectability: Pipet 2 mL of the standard
crystalline powder or powder.
solution, and add methanol to make exactly 5 mL. Confirm
It is freely soluble in acetic acid (100), slightly soluble in
that the peak area of domperidone obtained from 10 mL of
methanol and in ethanol (99.5), very slightly soluble in 2-
this solution is equivalent to 30 to 50z of that from 10 mL of
propanol, and practically insoluble in water.
the standard solution.
Melting point: about 2439 C (with decomposition).
System performance: Dissolve 10 mg of Domperidone
Identification (1) Determine the absorption spectrum of and 20 mg of ethyl parahydroxybenzoate in 100 mL of
a solution of Domperidone in a mixture of 2-propanol and methanol. When the procedure is run with 10 mL of this
0.1 mol/L hydrochloric acid TS (9:1) (1 in 50,000) as direct- solution under the above operating conditions, domperidone
ed under Ultraviolet-visible Spectrophotometry <2.24>, and and ethyl parahydroxybenzoate are eluted in this order with
compare the spectrum with the Reference Spectrum: both the resolution between these peaks being not less than 1.5.
spectra exhibit similar intensities of absorption at the same System repeatability: When the test is repeated 6 times
wavelengths. with 10 mL of the standard solution under the above operat-
(2) Determine the infrared absorption spectrum of ing conditions, the relative standard deviation of the peak
Domperidone as directed in the potassium bromide disk area of domperidone is not more than 3.0z.
method under Infrared Spectrophotometry <2.25>, and com-
Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C,
pare the spectrum with the Reference Spectrum: both spec-
4 hours).
tra exhibit similar intensities of absorption at the same wave
numbers. Residue on ignition <2.44> Not more than 0.1z (1 g).
Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of Assay Weigh accurately about 0.5 g of Domperidone,
Domperidone according to Method 2, and perform the test. previously dried, dissolve in 50 mL of acetic acid (100), and
Prepare the control solution with 2.0 mL of Standard Lead titrate <2.50> with 0.1 mol/L perchloric acid VS (potentio-
Solution (not more than 10 ppm). metric titration). Perform a blank determination in the same
(2) Related substances—Dissolve 30 mg of Domperi- manner, and make any necessary correction.
done in 100 mL of methanol, and use this solution as the
Each mL of 0.1 mol/L perchloric acid VS
sample solution. Pipet 1 mL of the sample solution, add
=42.59 mg of C22H24ClN5O2
methanol to make exactly 200 mL, and use this solution as
the standard solution. Perform the test with exactly 10 mL Containers and storage Containers—Well-closed contain-
each of the sample solution and standard solution as direct- ers.
ed under Liquid Chromatography <2.01> according to the Storage—Light-resistant.
following conditions, and determine each peak area of each
solution by the automatic integration method: the area of
each peak other than domperidone obtained from the sam-
ple solution is not larger than 1/2 times the peak area of
domperidone from the standard solution. Furthermore, the
Supplement I, JP XV Official Monographs 1877

Bacterial endotoxins <4.01> Less than 2.50 EU/mg (poten-


Dopamine Hydrochloride Injection cy).

Uniformity of dosage units <6.02> It meets the requirement


ドパミン塩酸塩注射液
of the Mass variation test.
Add the following next to Extractable volume: Foreign insoluble matter <6.06> Perform the test according
to Method 2: it meets the requirement.
Foreign insoluble matter <6.06> Perform the test according
to Method 1: it meets the requirement. Insoluble particulate matter <6.07> It meets the require-
ment.
Insoluble particulate matter <6.07> It meets the require-
ment. Sterility <4.06> Perform the test according to the Mem-
brane filtration method: it meets the requirement.
Sterility <4.06> Perform the test according to the Mem-
brane filtration method: it meets the requirement. Assay Weigh accurately the mass of the contents of not
less than 10 containers of Doxorubicin Hydrochloride for
Injection. Weigh accurately an amount of the contents, e-
Add the following: quivalent to about 10 mg (potency) of Doxorubicin
Hydrochloride, add exactly 5 mL of the internal standard
Doxorubicin Hydrochloride for solution and the mobile phase to make 100 mL, and use the
solution as the sample solution. Separately, weigh accurately
Injection an amount of Doxorubicin Hydrochloride Reference Stan-
注射用ドキソルビシン塩酸塩 dard, equivalent to 10 mg (potency), add exactly 5 mL of the
internal standard solution and the mobile phase to make 100
Doxorubicin Hydrochloride for Injection is a prepa- mL, and use this solution as the standard solution. Perform
ration for injection, which is dissolved before use. the test with 10 mL each of the sample solution and standard
It contains not less than 90.0z and not more than solution as directed under Liquid Chromatography <2.01>,
110.0z of the labeled amount of doxorubicin and calculate the ratios, QT and QS, of the peak area of dox-
hydrochloride (C27H29NO11.HCl: 579.98). orubicin to that of the internal standard.

Method of preparation Prepare as directed under Injec- Amount [mg (potency)] of doxorubicin hydrochloride
tions, with Doxorubicin Hydrochloride. (C27H29NO11.HCl)
=WS×(QT/QS)
Description Doxorubicin Hydrochloride for Injection oc-
curs as red-orange, powder or masses. WS: Amount [mg (potency)] of Doxorubicin Hydrochlo-
ride Reference Standard
Identification Dissolve an amount of Doxorubicin
Hydrochloride for Injection, equivalent to 10 mg (potency) Internal standard solution—A solution of butyl parahydrox-
of Doxorubicin Hydrochloride according to the labeled ybenzoate in the mobile phase (1 in 1000).
amount, in methanol to make 100 mL. To 5 mL of this solu- Operating conditions—
tion add methanol to make 50 mL, and determine the ab- Detector: An ultraviolet absorption photometer (wave-
sorption spectrum of the solution as directed under Ultrav- length: 254 nm).
iolet-visible Spectrophotometry <2.24>: it exhibits maxima Column: A stainless steel column 4.6 mm in inside di-
between 231 nm and 235 nm, between 250 nm and 254 nm, ameter and 25 cm in length, packed with octadecylsilanized
between 477 nm and 481 nm, and between 493 nm and 497 silica gel for liquid chromatography (5 mm in particle di-
nm, and exhibits a shoulder between 528 nm and 538 nm. ameter).
Column temperature: A constant temperature of about
pH <2.54> The pH of a solution, prepared by dissolving an 259C.
amount of Doxorubicin Hydrochloride for Injection equiva- Mobile phase: Dissolve 3 g of sodium lauryl sulfate in
lent to 10 mg (potency) of Doxorubicin Hydrochloride ac- 1000 mL of diluted phosphoric acid (7 in 5000). To this solu-
cording to the labeled amount in 2 mL of water, is 5.0 to 6.0. tion add 1000 mL of acetonitrile.
Purity Clarity and color of solution—Dissolve an amount Flow rate: Adjust the flow rate so that the retention time
of Doxorubicin Hydrochloride for Injection, equivalent to of doxorubicin is about 8 minutes.
50 mg (potency) of Doxorubicin Hydrochloride according to System suitability—
the labeled amount, in 10 mL of water: the solution is clear System performance: When the procedure is run with 10
and red. mL of the standard solution under the above operating con-
ditions, doxorubicin and the internal standard are eluted in
Water <2.48> Not more than 4.0z (0.25 g, volumetric this order with the resolution between these peaks being not
titration, direct titration). less than 5, and the symmetry factor of the peak of dox-
orubicin is between 0.8 and 1.2.
System repeatability: When the test is repeated 6 times
1878 Official Monographs Supplement I, JP XV

with 10 mL of the standard solution under the above operat- (3) Determine the infrared absorption spectrum of
ing conditions, the relative standard deviation of the ratio of Emorfazone as directed in the potassium bromide disk
the peak area of doxorubicin to that of the internal standard method under Infrared Spectrophotometry <2.25>, and com-
is not more than 1.0z. pare the spectrum with the Reference Spectrum: both spec-
tra exhibit similar intensities of absorption at the same wave
Containers and storage Containers—Hermetic containers.
numbers.

Melting point <2.60> 89 – 929C (after drying).


Edrophonium Chloride Injection Purity (1) Chloride <1.03>—Perform the test with 1.0 g
of Emorfazone. Prepare the control solution with 0.50 mL
エドロホニウム塩化物注射液
of 0.01 mol/L hydrochloric acid VS (not more than 0.018
z).
Add the following next to pH:
(2) Heavy metals <1.07>—Proceed with 2.0 g of Emorfa-
Bacterial endotoxins <4.01> Less than 15 EU/mg. zone according to Method 2, and perform the test. Prepare
the control solution with 2.0 mL of Standard Lead Solution
Add the following next to Extractable volume: (not more than 10 ppm).
(3) Arsenic <1.11>—Prepare the test solution with 2.0 g
Foreign insoluble matter <6.06> Perform the test according
of Emorfazone according to Method 3, and perform the test
to Method 1: it meets the requirement.
(not more than 1 ppm).
Insoluble particulate matter <6.07> It meets the require- (4) Related substances—Conduct this procedure using
ment. light-resistant vessels. Dissolve 0.5 g of Emorfazone in 50
mL of the mobile phase, and use this solution as the sample
Sterility <4.06> Perform the test according to the Mem-
solution. Pipet 1 mL of the sample solution, add the mobile
brane filtration method: it meets the requirement.
phase to make exactly 100 mL, and use this solution as the
standard solution. Perform the test with exactly 20 mL each
of the sample solution and standard solution as directed un-
Add the following:
der Liquid Chromatography <2.01> according to the follow-
ing conditions, and determine each peak area by the auto-
Emorfazone matic integration method: each peak area other than emor-
エモルファゾン fazone obtained from the sample solution is not larger than
1/10 times the peak area of emorfazone from the standard
solution, and the total area of the peaks other than the peak
of emorfazone from the sample solution is not larger than
1/2 times the peak area of emorfazone from the standard so-
lution.
C11H17N3O3: 239.27 Operating conditions—
4-Ethoxy-2-methyl-5-(morpholin-4-yl)pyridazin- Detector: An ultraviolet absorption photometer (wave-
3(2H)-one [38957-41-4] length: 254 nm).
Column: A stainless steel column 4.6 mm in inside di-
Emorfazone, when dried, contains not less than 98.5 ameter and 15 cm in length, packed with octadecylsilanized
z and not more than 101.0z of C11H17N3O3. silica gel for liquid chromatography (5 mm in particle di-
ameter).
Description Emorfazone occurs as colorless crystals or a
Column temperature: A constant temperature of about
white to light yellow crystalline powder.
259C.
It is very soluble in ethanol (99.5), and freely soluble in
Mobile phase: A mixture of water and methanol (11:10).
water and in acetic anhydride.
Flow rate: Adjust the flow rate so that the retention time
It dissolves in 1 mol/L hydrochloric acid TS.
of emorfazone is about 5 minutes.
It gradually turns yellow and decomposes on exposure to
Time span of measurement: About 2.5 times as long as the
light.
retention time of emorfazone, beginning after the solvent
Identification (1) Dissolve 20 mg of Emorfazone in 2 mL peak.
of 1 mol/L hydrochloric acid TS, and add 5 drops of System suitability—
Reinecke's TS: light red floating matters are formed. Test for required detectability: Pipet 1 mL of the standard
(2) Determine the absorption spectrum of a solution of solution, add the mobile phase to make exactly 20 mL. Con-
Emorfazone (1 in 100,000) as directed under Ultraviolet-visi- firm that the peak area of emorfazone obtained with 20 mL
ble Spectrophotometry <2.24>, and compare the spectrum of this solution is equivalent to 3.5 to 6.5z of that with 20
with the Reference Spectrum: both spectra exhibit similar mL of the standard solution.
intensities of absorption at the same wavelengths. System performance: Dissolve 16 mg of Emorfazone and
30 mg of 2,4-dinitrophenylhydrazine in 100 mL of
Supplement I, JP XV Official Monographs 1879

methanol. When the procedure is run with 20 mL of this so- hydrochloric acid TS, shake, add 5 mL of diethyl ether, and
lution under the above operating conditions, emorfazone shake for 5 minutes. Take 3 mL of the upper layer, distil off
and 2,4-dinitrophenylhydrazine are eluted in this order with the diethyl ether on a water bath, add 5 mL of water to the
the resolution between these peaks being not less than 2.5. residue with shaking, and add 1 drop of potassium perman-
System repeatability: When the test is repeated 6 times ganate TS: the red color of the test solution immediately dis-
with 20 mL of the standard solution under the above operat- appears.
ing conditions, the relative standard deviation of the peak
Optical rotation <2.49> [a]20 D: -41.0 – -43.59 (after
area of emorfazone is not more than 1.0z.
drying, 0.25 g, methanol, 25 mL, 100 mm).
Loss on drying <2.41> Not more than 0.5z (1 g, in vacu-
Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of
um, 609C, 4 hours).
Enalapril Maleate according to Method 2, and perform the
Residue on ignition <2.44> Not more than 0.1z (1 g). test. Prepare the control solution with 2.0 mL of Standard
Lead Solution (not more than 10 ppm).
Assay Weigh accurately about 0.2 g of Emorfazone, previ-
(2) Related substances—Dissolve 30 mg of Enalapril
ously dried, dissolve in 60 mL of acetic anhydride, and
Maleate in 100 mL of a mixture of sodium dihydrogen phos-
titrate <2.50> with 0.1 mol/L perchloric acid VS (potentio-
phate TS, pH 2.5 and acetonitrile (19:1), and use this solu-
metric titration). Perform a blank determination in the same
tion as the sample solution. Pipet 1 mL of the sample solu-
manner, and make any necessary correction.
tion, add the mixture of sodium dihydrogen phosphate TS,
Each mL of 0.1 mol/L perchloric acid VS pH 2.5 and acetonitrile (19:1) to make exactly 100 mL, and
=23.93 mg of C11H17N3O3 use this solution as the standard solution. Perform the test
with exactly 50 mL each of the sample solution and standard
Containers and storage Containers—Tight containers.
solution as directed under Liquid Chromatography <2.01>
Storage—Light-resistant.
acccording to the following conditions, and determine each
peak area of these solutions by the automatic integration
method: the area of the peak other than maleic acid and
Add the following:
enalapril obtained from the sample solution is not larger
than the peak area of enalapril from the standard solution.
Enalapril Maleate Furthermore, the total area of the peaks other than maleic
acid and enalapril from the sample solution is not larger
エナラプリルマレイン酸塩
than twice the peak area of enalapril from the standard solu-
tion.
Operating conditions—
Detector, column, column temperature, mobile phases,
mobile phase flow, and flow rate: Proceed as directed in the
operating conditions in the Assay.
Time span of measurement: About 2 times as long as the
C20H28N2O5.C4H4O4: 492.52
retention time of enalapril, beginning after the peak of
(2S)-1-{(2S)-2-[(1S)-1-Ethoxycarbonyl-
maleic acid.
3-phenylpropylamino]propanoyl}pyrrolidine-2-carboxylic
System suitability—
acid monomaleate [76095-16-4]
Test for required detectability: Pipet 1 mL of the standard
solution, and add a mixture of sodium dihydrogen phos-
Enalapril Maleate, when dried, contains not less phate TS, pH 2.5 and acetonitrile (19:1) to make exactly 10
than 98.0z and not more than 102.0z of mL. Confirm that the peak area of enalapril obtained from
C20H28N2O5.C4H4O4. 50 mL of this solution is equivalent to 7 to 13z of that from
Description Enalapril Maleate occurs as white crystals or a 50 mL of the standard solution.
white crystalline powder. System performance: When the procedure is run with 50
It is freely soluble in methanol, sparingly soluble in water mL of the standard solution under the above conditions, the
and in ethanol (99.5), and slightly soluble in acetonitrile. number of theoretical plates and the symmetry factor of the
Melting point: about 1459 C (with decomposition). peak of enalapril are not less than 3000 and not more than
2.0, respectively.
Identification (1) Determine the infrared absorption
System repeatability: When the test is repeated 6 times
spectra of Enalapril Maleate as directed in the potassium
with 50 mL of the standard solution under the above condi-
bromide disc method under Infrared Spectrophotometry
tions, the relative standard deviation of the peak area of
<2.25>, and compare the spectrum with the Reference Spec-
enalapril is not more than 2.0z.
trum or the spectrum of Enalapril Maleate Reference Stan-
dard: both spectra exhibit similar intensities of absorption at Loss on drying <2.41> Not more than 1.0z (1 g, in vacu-
the same wave numbers. um, 609C, 2 hours).
(2) To 20 mg of Enalapril Maleate add 5 mL of 1 mol/L
Residue on ignition <2.44> Not more than 0.2z (1 g).
1880 Official Monographs Supplement I, JP XV

Add the following:


Assay Weigh accurately about 30 mg each of Enalapril
Maleate and Enalapril Maleate Reference Standard, both
previously dried, and dissolve in a mixture of sodium di- Enalapril Maleate Tablets
hydrogen phosphate TS, pH 2.5 and acetonitrile (19:1) to
エナラプリルマレイン酸塩錠
make exactly 100 mL, and use these solutions as the sample
solution and standard solution. Perform the test with exactly
Enalapril Maleate Tablets contain not less than 93.0
50 mL each of the sample solution and standard solution as
z and not more than 107.0z of the labeled amount
directed under Liquid Chromatography <2.01> according to
of enalapril maleate (C20H28N2O5.C4H4O4: 492.52).
the following conditions, and determine the peak areas of
enalapril, AT and AS, of both solutions. Method of preparation Prepare as directed under Tablets,
with Enalapril Maleate.
Amount (mg) of C20H28N2O5.C4H4O4
=WS×(AT/AS) Identification To a quantity of powdered Enalapril Male-
ate Tablets equivalent to 50 mg of Enalapril Maleate accord-
WS: Amount (mg) of Enalapril Maleate Reference Stan-
ing to the labeled amount, add 20 mL of methanol, shake,
dard
centrifuge, and then use the supernatant liquid as the sample
Operating conditions— solution. Separately, dissolve 25 mg of enalapril maleate in
Detector: An ultraviolet absorption photometer (wave- 10 mL of methanol, and use this solution as the standard
length: 215 nm). solution. Perform the test with these solutions as directed
Column: A stainless steel column 4.1 mm in inside di- under Thin-Layer Chromatography <2.03>. Spot 20 mL each
ameter and 15 cm in length, packed with porous styrene- of the sample solution and standard solution on a plate of
divinylbenzene copolymer for liquid chromatography (5 mm silica gel with fluorescent indicator for thin-layer chro-
in particle diameter). matography. Develop the plate with a mixture of water, ace-
Column temperature: A constant temperature of about tone, 1-butanol, acetic acid (100) and toluene (1:1:1:1:1) to a
709C. distance of about 10 cm, and air-dry the plate. Examine
Mobile phase A: Dissolve 3.1 g of sodium dihydrogen under ultraviolet light (main wavelength: 254 nm): the Rf
phosphate dihydrate in 900 mL of water, adjust the pH to values of the 2 spots obtained from the sample solution and
6.8 with a solution of sodium hydroxide (1 in 4), and add the 2 spots obtained from the standard solution are equiva-
water to make 1000 mL. To 950 mL of this solution, add 50 lent.
mL of acetonitrile for liquid chromatography.
Purity Enalaprilat and enalapril diketopiperazine—Use
Mobile phase B: Dissolve 3.1 g of sodium dihydrogen
the sample solution obtained in the Assay as the sample solu-
phosphate dihydrate in 900 mL of water, adjust the pH to
tion. Pipet 1 mL of the sample solution, add sodium di-
6.8 with a solution of sodium hydroxide (1 in 4), and add
hydrogen phosphate TS, pH 2.2 to make exactly 100 mL,
water to make 1000 mL. To 340 mL of this solution, add 660
and use this solution as the standard solution. Perform the
mL of acetonitrile for liquid chromatography.
test with exactly 50 mL each of the sample solution and stan-
Mobile phase flow: Control the concentration gradient by
dard solution as directed under Liquid Chromatography
changing the ratio of the mobile phases A and B as follows.
<2.01> according to the following conditions, and determine
Time after injection Mobile phase Mobile phase each peak area of both solutions by the automatic integra-
of sample (min) A (volz) B (volz) tion method: the peak area of enalaprilat, having the relative
retention time of about 0.5 with respect to enalapril obtained
0 95 5
from the sample solution, is not larger than 2 times the peak
0 – 20 95 → 40 5 → 60
area of enalapril from the standard solution. Also, the peak
20 – 25 40 60
area of enalapril diketopiperazine, having the relative reten-
Flow rate: 1.4 mL per minute. tion time of about 1.5 is not larger than the peak area of
System suitability— enalapril from the standard solution.
System performance: When the procedure is run with 50 Operating conditions—
mL of the standard solution under the above conditions, the Proceed as directed in the operating conditions in the As-
number of theoretical plates and the symmetry factor of the say.
peak of enalapril are not less than 3000 and not more than System suitability—
2.0, respectively. Test for required detectability: Pipet 1 mL of the standard
System repeatability: When the test is repeated 6 times solution, and add sodium dihydrogen phosphate TS, pH 2.2
with 50 mL of the standard solution under the above condi- to make exactly 10 mL. Confirm that the peak area of
tions, the relative standard deviation of the peak area of enalapril obtained from 50 mL of this solution is equivalent
enalapril is not more than 1.0z. to 7 to 13z of that from 50 mL of the standard solution.
System performance: Proceed as directed in the system
Containers and storage Containers—Well-closed contain-
suitability in the Assay.
ers.
System repeatability: When the test is repeated 6 times
Supplement I, JP XV Official Monographs 1881

with 50 mL of the standard solution under the above condi- Mobile phase: Dissolve 1.88 g of sodium dihydrogen
tions, the relative standard deviation of the peak area of phosphate dihydrate in 900 mL of water, adjust the pH to
enalapril is not more than 2.0z. 2.2 with phosphoric acid, and add water to make 1000 mL.
To 750 mL of this solution add 250 mL of acetonitrile.
Uniformity of dosage units <6.02> Perform the test accord-
System suitability—
ing to the following method: it meets the requirement of the
System performance: When the procedure is run with 50
Content uniformity test.
mL of the standard solution under the above conditions, the
Take 1 tablet of Enalapril Maleate Tablets, add V/2 mL
number of theoretical plates and the symmetry factor of the
of sodium dihydrogen phosphate TS, pH 2.2, treat with
peak of enalapril are not less than 300 and not more than
ultrasonic waves for 15 minutes, shake for 30 minutes, and
2.0, respectively.
add sodium dihydrogen phosphate TS, pH 2.2 to make
System repeatability: When the test is repeated 6 times
exactly V mL so that 1 mL of the solution contains about 0.1
with 50 mL of the standard solution under the above condi-
mg of enalapril maleate (C20H28N2O5.C4H4O4). Treat this so-
tions, the relative standard deviation of the peak area of
lution with ultrasonic waves for 15 minutes, filter through a
enalapril is not more than 2.0z.
membrane filter with a pore size not exceeding 0.45 mm, and
use the filtrate as the sample solution. Then, proceed as Assay Weigh accurately not less than 20 Enalapril Maleate
directed in the Assay. Tablets, and powder. Weigh accurately a portion of the
powder equivalent to about 10 mg of enalapril maleate
Amount (mg) of enalapril maleate (C20H28N2O5.C4H4O4)
(C20H28N2O5.C4H4O4), add 50 mL of sodium dihydrogen
=WS×(AT/AS)×(V/200)
phosphate TS, pH 2.2, treat with ultrasonic waves for 15
WS: Amount (mg) of Enalapril Maleate Reference Stan- minutes, shake for 30 minutes, and then add sodium di-
dard hydrogen phosphate TS, pH 2.2 to make exactly 100 mL.
Treat this solution with ultrasonic waves for 15 minutes,
Dissolution <6.10> When the test is performed at 50 revolu-
filter through a membrane filter with a pore size not exceed-
tions per minute according to the Paddle method, using 900
ing 0.45 mm, and use this filtrate as the sample solution.
mL of water as the dissolution medium, the dissolution rates
Separately, weigh accurately about 20 mg of Enalapril Male-
in 15 minutes of a 2.5- and 5-mg tablet of Enalapril Maleate
ate Reference Standard, previously dried in vacuum at 609C
Tablets and in 30 minutes of a 10-mg tablet of Enalapril
for 2 hours, dissolve in sodium dihydrogen phosphate TS,
Maleate Tablets are not less than 85z, respectively.
pH 2.2 to make exactly 200 mL, and use this solution as the
Start the test with 1 tablet of Enalapril Maleate Tablets,
standard solution. Perform the test with exactly 50 mL each
withdraw not less than 20 mL of the medium at the specified
of the sample solution and standard solution as directed un-
minute after starting the test, and filter through a membrane
der Liquid Chromatography <2.01> according to the follow-
filter with a pore size not exceeding 0.45 mm. Discard the
ing conditions, and determine the enalapril peak areas, AT
first 10 mL of the filtrate, pipet V mL of the subsequent
and AS, of both solutions.
filtrate, add water to make exactly V? mL so that each mL
contains about 2.8 mg of enalapril maleate (C20H28N2O5. Amount (mg) of enalapril maleate (C20H28N2O5.C4H4O4)
C4H4O4) according to the labeled amount, and use this solu- =WS×(AT/AS)×(1/2)
tion as the sample solution. Separately, weigh accurately
WS: Amount (mg) of Enalapril Maleate Reference Stan-
about 14 mg of Enalapril Maleate Reference Standard,
dard
previously dried in vacuum at 609C for 2 hours, and dissolve
in water to make exactly 500 mL. Pipet 5 mL of this solu- Operating conditions—
tion, add water to make exactly 50 mL, and use this solution Detector: An ultraviolet absorption photometer (wave-
as the standard solution. Perform the test with exactly 50 mL length: 215 nm).
each of the sample solution and standard solution as direct- Column: A stainless steel column 4.6 mm in inside di-
ed under Liquid Chromatography <2.01> according to the ameter and 25 cm in length, packed with octylsilanized silica
following conditions, and determine the enalapril peak gel for liquid chromatography (5 mm in particle diameter).
areas, AT and AS, of both solutions. Column temperature: A constant temperature of about
509C.
Dissolution rate (z) with respect to the labeled amount of
Mobile phase: A mixture of sodium dihydrogen phos-
enalapril maleate (C20H28N2O5.C4H4O4)
phate TS, pH 2.2 and acetonitrile (3:1).
=WS×(AT/AS)×(V?/V)×(1/C)×18
Flow rate: Adjust the flow rate so that the retention time
WS: Amount (mg) of Enalapril Maleate Reference Stan- of enalapril is about 5 minutes.
dard System suitability—
C: Labeled amount (mg) of enalapril maleate System performance: Heat to fusion about 20 mg of
(C20H28N2O5.C4H4O4) in 1 tablet enalapril maleate. After cooling, add 50 mL of acetonitrile,
and treat with ultrasonic waves to dissolve. To 1 mL of this
Operating conditions—
solution, add the standard solution to make 50 mL, and use
Detector, column, column temperature, and flow rate:
this solution as the solution for system suitability test. When
Proceed as directed in the operating conditions in the Assay.
the procedure is run with 50 mL of the solution for system
1882 Official Monographs Supplement I, JP XV

suitability test under the above conditions, enalapril and Add the following:
enalapril diketopiperazine, which has a relative retention
time of 1.5 to enalapril, are eluted in this order with the reso- Erythromycin Enteric-Coated
lution between these peaks being not less than 2.0.
System repeatability: When the test is repeated 6 times
Tablets
with 50 mL of the solution for system suitability test under エリスロマイシン腸溶錠
the above conditions, the relative standard deviation of the
peak area of enalapril is not more than 1.0z.
Erythromycin Enteric-Coated Tablets contain not
Containers and storage Containers—Well-closed contain- less than 90.0z and not more than 110.0z of the la-
ers. beled amount of erythromycin (C37H67NO13: 733.93).
Method of preparation Prepare as directed under Tablets,
with Erythromycin.
Ephedrine Hydrochloride Injection
Identification To a quantity of powdered Erythromycin
エフェドリン塩酸塩注射液 Enteric-Coated Tablets, equivalent to 10 mg (potency) of
Erythromycin according to the labeled amount, add 1 mL of
Add the following next to Extractable volume: methanol, shake well, filter, and use the filtrate as the sam-
ple solution. Separately, dissolve 10 mg of Erythromycin
Foreign insoluble matter <6.06> Perform the test according
Reference Standard in 1 mL of methanol, and use this solu-
to Method 1: it meets the requirement.
tion as the standard solution. Then, proceed as directed in
Insoluble particulate matter <6.07> It meets the require- the Identification (2) under Erythromycin.
ment.
Loss on drying <2.41> Not more than 10.0z (0.2 g, in
Sterility <4.06> Perform the test according to the Mem- vacuum not exceeding 0.67 kPa, 609
C, 3 hours).
brane filtration method: it meets the requirement.
Uniformity of dosage units <6.02> It meets the requirement
of the Mass variation test.

Ephedrine Hydrochloride Tablets Disintegration <6.09> It meets the requirement.

Assay Perform the test according to the Cylinder-plate


エフェドリン塩酸塩錠
method as directed under Microbial Assay for Antibiotics
<4.02> according to the following conditions.
Add the following next to Identification:
(i) Test organism, culture medium, and standard solu-
Uniformity of dosage units <6.02> Perform the test accord- tions—
ing to the following method: it meets the requirement of the Proceed as directed in the Assay under Erythromycin.
Content uniformity test. (ii) Sample solutions—Weigh accurately the mass of not
To 1 tablet of Ephedrine Hydrochloride Tablets add V less than 20 Erythromycin Enteric-Coated Tablets, and pul-
mL of water so that each mL contains 0.25 mg of ephedrine verize into a powder. Weigh accurately a portion of the pow-
hydrochloride (C10H15NO.HCl), then add exactly V/4 mL of der, equivalent to about 25 mg (potency) of Erythromycin,
the internal standard solution, disperse the tablet into small add 25 mL of methanol, shake vigorously, add 0.1 mol/L
particles using ultrasonic waves, then stir for a further 10 phosphate buffer solution, pH 8.0, to make exactly 100 mL,
minutes in the same way. Shake this solution for 10 minutes, and filter. Take exactly an appropriate volume of the
centrifuge, and use the supernatant liquid as the sample so- filtrate, add 0.1 mol/L phosphate buffer solution, pH 8.0,
lution. Separately, weigh accurately about 25 mg of ephe- to prepare solutions containing 20 mg (potency) and 5 mg
drine hydrochloride for assay, previously dried at 1059 C for (potency) per mL, and use these solutions as the high and
3 hours, dissolve in water to make exactly 100 mL. Pipet 20 low concentration sample solutions, respectively.
mL of this solution, add exactly 5 mL of the internal stan-
Containers and storage Containers—Well-closed contain-
dard solution, and use this solution as the standard solution.
ers.
Then, proceed as directed in the Assay.

Amount (mg) of ephedrine hydrochloride (C10H15NO.HCl)


=WS×(QT/QS)×(V/100)

WS: Amount (mg) of ephedrine hydrochloride for assay


Internal standard solution—A solution of etilefrine
hydrochloride (1 in 2000).
Supplement I, JP XV Official Monographs 1883

Add the following: Dissolution rate (z) with respect to the labeled amount of
etizolam (C17H15ClN4S)
Etizolam Fine Granules =(WS/WT)×(AT/AS)×(1/C)×(18/5)

WS: Amount (mg) of etizolam for assay


エチゾラム細粒
WT: Amount (g) of sample
C: Labeled amount (mg) of etizolam (C17H15ClN4S) in 1 g
Etizolam Fine Granules contain not less than 93.0z
and not more than 107.0z of the labeled amount of Operating conditions—
etizolam (C17H15ClN4S: 342.85). Detector: An ultraviolet absorption photometer (wave-
length: 243 nm).
Method of preparation Prepare fine particles as directed
Column: A stainless steel column 4.6 mm in inside di-
under Powders, with Etizolam.
ameter and 15 cm in length, packed with octadecylsilanized
Identification (1) To a quantity of powdered Etizolam silica gel for liquid chromatography (5 mm in particle di-
Fine Granules, equivalent to 5 mg of Etizolam according to ameter).
the labeled amount, add 10 mL of methanol, shake, and Column temperature: A constant temperature of about
filter through a membrane filter with a pore size not exceed- 309C.
ing 0.45 mm. Evaporate the filtrate to dryness on a water Mobile phase: A mixture of water and acetonitrile (1:1).
bath, cool, and then dissolve the residue in 2 mL of sulfuric Flow rate: Adjust the flow rate so that the retention time
acid. The solution gives off a light yellow-green fluorescent of etizolam is about 7 minutes.
when exposed to ultraviolet light (main wavelength: 365 System suitability—
nm). System performance: When the procedure is run with 50
(2) To a quantity of powdered Etizolam Fine Granules, mL of the standard solution under the above operating con-
equivalent to 1 mg of Etizolam according to the labeled ditions, the number of theoretical plates and the symmetry
amount, add 80 mL of 0.1 mol/L hydrochloric acid TS, factor of the peak of etizolam are not less than 3000 and not
shake, and then filter. Determine the absorption spectrum of more than 2.0, respectively.
the filtrate as directed under Ultraviolet-visible Spec- System repeatability: When the test is repeated 6 times
trophotometry <2.24>: it exhibits absorption maxima be- with 50 mL of the standard solution under the above operat-
tween 249 nm and 253 nm, and between 292 nm and 296 nm, ing conditions, the relative standard deviation of the peak
when perform the measurement within 10 minutes. area of etizolam is not more than 2.0z.
Uniformity of dosage units <6.02> The granules in single-u- Particle size <6.03> It meets the requirement.
nit container meet the requirement of the Mass variation
Assay Weigh accurately an amount of Etizolam Fine Gran-
test.
ules, equivalent to about 4 mg of etizolam (C17H15ClN4S),
Dissolution <6.10> When the test is performed at 50 revolu- add 30 mL of water, and stir. Add 60 mL of methanol, stir
tions per minute according to the Paddle method, using 900 for 20 minutes, add methanol to make exactly 100 mL, and
mL of water as the dissolution medium, the dissolution rate centrifuge. Pipet 5 mL of the supernatant liquid, add exactly
in 30 minutes of Etizolam Fine Granules is not less than 10 mL of the internal standard solution, add diluted
75z. methanol (7 in 10) to make 25 mL, and use this solution as
Start the test with an accurately weighed amount of the sample solution. Separately, weigh accurately about 0.1
Etizolam Fine Granules, equivalent to about 1 mg of etizol- g of etizolam for assay, previously dried at 1059C for 3
am (C17H15ClN4S) according to the labeled amount, hours, and dissolve in diluted methanol (7 in 10) to make
withdraw not less than 20 mL of the medium at the specified exactly 100 mL. Pipet 2 mL of this solution, and add diluted
minute after starting the test, and filter through a membrane methanol (7 in 10) to make exactly 100 mL. Pipet 10 mL of
filter with a pore size not exceeding 0.45 mm. Discard the this solution, add exactly 10 mL of the internal standard
first 10 mL of filtrate, pipet the subsequent 2 mL, add exact- solution, add diluted methanol (7 in 10) to make 25 mL, and
ly 2 mL of acetonitrile, and use this solution as the sample use this solution as the standard solution. Perform the test
solution. Separately, weigh accurately about 28 mg of etizol- with 10 mL each of the sample solution and standard solu-
am for assay, previously dried at 1059 C for 3 hours, and dis- tion as directed under Liquid Chromatography <2.01> ac-
solve in methanol to make exactly 50 mL. Pipet 5 mL of this cording to the following conditions, and calculate the ratios,
solution, and add water to make exactly 100 mL. Pipet 4 mL QT and QS, of the peak area of etizolam to that of the inter-
of this solution, and add water to make exactly 100 mL. nal standard.
Pipet 2 mL of this solution, add exactly 2 mL of acetonitrile,
Amount (mg) of etizolam (C17H15ClN4S)
and use this solution as the standard solution. Perform the
=WS×(QT/QS)×(1/25)
test with exactly 50 mL each of the sample solution and stan-
dard solution as directed under Liquid Chromatography WS: Amount (mg) of etizolam for assay
<2.01> according to the following conditions, and determine
Internal standard solution—A solution of ethyl parahydrox-
the enalapril peak areas, AT and AS, of both solutions.
ybenzoate in diluted methanol (7 in 10) (1 in 50,000).
1884 Official Monographs Supplement I, JP XV

Operating conditions— when perform the measurement within 10 minutes.


Detector: An ultraviolet absorption photometer (wave-
Uniformity of dosage units <6.02> Perform the test accord-
length: 240 nm).
ing to the following method: it meets the requirement of the
Column: A stainless steel column 4.6 mm in inside di-
Content uniformity test.
ameter and 15 cm in length, packed with octadecylsilanized
Take 1 tablet of Etizolam Tablets, add 2.5 mL of water,
silica gel for liquid chromatography (5 mm in particle di-
and stir until it disintegrates. Add 20 mL of methanol, stir
ameter).
for 20 minutes, add methanol to make exactly 25 mL, and
Column temperature: A constant temperature of about
centrifuge. Pipet V mL of the supernatant liquid, add exact-
359C.
ly 10 mL of the internal standard solution, add diluted
Mobile phase: Dissolve 1.36 g of potassium dihydrogen
methanol (9 in 10) to make 25 mL so that each mL contains
phosphate in water to make 1000 mL, and adjust the pH to
about 8 mg of etizolam (C17H15ClN4S), and use this solution
3.5 with diluted phosphoric acid (1 in 10). To 550 mL of this
as the sample solution. Then, proceed as directed in the As-
solution add 450 mL of acetonitrile.
say.
Flow rate: Adjust the flow rate so that the retention time
of etizolam is about 6 minutes. Amount (mg) of etizolam (C17H15ClN4S)
System suitability— =WS×(QT/QS)×(1/V)×(1/20)
System performance: When the procedure is run with 10
WS: Amount (mg) of etizolam for assay
mL of the standard solution under the above operating con-
ditions, the internal standard and etizolam are eluted in this Internal standard solution—A solution of ethyl parahydrox-
order with the resolution between these peaks being not less ybenzoate in diluted methanol (9 in 10) (1 in 50,000).
than 3.
Dissolution <6.10> When the test is performed at 50 revolu-
System repeatability: When the test is repeated 6 times
tions per minute according to the Paddle method, using 900
with 10 mL of the standard solution under the above operat-
mL of water as the dissolution medium, the dissolution rate
ing conditions, the relative standard deviation of the ratio of
in 30 minutes of Etizolam Tablets is not less than 70z.
the peak area of etizolam to that of the internal standard is
Start the test with 1 tablet of Etizolam Tablets, withdraw
not more than 1.0z.
not less than 20 mL of the medium at the specified minute
Containers and storage Containers—Tight containers. after starting the test, and filter through a membrane filter
Storage—Light-resistant. with a pore size not exceeding 0.45 mm. Discard the first 10
mL of the filtrate, pipet V mL of the subsequent filtrate, add
water to make exactly V? mL so that each mL contains about
Add the following: 0.56 mg of etizolam (C17H15ClN4S) according to the labeled
amount. Pipet 2 mL of the solution, add exactly 2 mL of
Etizolam Tablets acetonitrile, and use this solution as the sample solution.
Separately, weigh accurately about 28 mg of etizolam for
エチゾラム錠 assay, previously dried at 1059 C for 3 hours, dissolve in 50
mL of methanol, and add water to make exactly 100 mL.
Etizolam Tablets contain not less than 93.0z and Pipet 5 mL of this solution, add water to make exactly 100
not more than 107.0z of the labeled amount of etizol- mL. Pipet 4 mL of this solution, add water to make exactly
am (C17H15ClN4S: 342.85). 100 mL. Pipet 2 mL of this solution, add exactly 2 mL of
acetonitrile, and use this solution as the standard solution.
Method of preparation Prepare as directed under Tablets,
Perform the test with exactly 50 mL each of the sample solu-
with Etizolam.
tion and standard solution as directed under Liquid Chro-
Identification (1) To a quantity of powdered Etizolam matography <2.01> according to the following conditions,
Tablets, equivalent to 5 mg of Etizolam according to the and determine the peak areas of etizolam, AT and AS, of
labeled amount, add 10 mL of methanol, shake, and filter. both solutions.
Evaporate the filtrate to dryness on a water bath, cool, and
Dissolution rate (z) with respect to the labeled amount
then dissolve the residue in 2 mL of sulfuric acid. The solu-
of etizolam (C17H15ClN4S)
tion gives off a light yellow-green fluorescence when exposed
=WS×(AT/AS)×(V?/V)×(1/C)×(9/5)
to ultraviolet light (main wavelength: 365 nm).
(2) To a quantity of powdered Etizolam Tablets, equiva- WS: Amount (mg) of etizolam for assay
lent to 1 mg of Etizolam according to the labeled amount, C: Labeled amount (mg) of etizolam (C17H15ClN4S) in 1
add 80 mL of 0.1 mol/L hydrochloric acid TS, shake, and tablet
then filter through a membrane filter with a pore size not
Operating conditions—
exceeding 0.45 mm. Determine the absorption spectrum of
Detector: An ultraviolet absorption photometer (wave-
this filtrate as directed under Ultraviolet-visible Spec-
length: 243 nm).
trophotometry <2.24>: it exhibits absorption maxima be-
Column: A stainless steel column 4.6 mm in inside di-
tween 249 nm and 253 nm, and between 292 nm and 296 nm
ameter and 15 cm in length, packed with octadecylsilanized
Supplement I, JP XV Official Monographs 1885

silica gel for liquid chromatography (5 mm in particle di- of etizolam is about 6 minutes.
ameter). System suitability—
Column temperature: A constant temperature of about System performance: When the procedure is run with 10
309C. mL of the standard solution under the above operating con-
Mobile phase: A mixture of water and acetonitrile (1:1). ditions, the internal standard and etizolam are eluted in this
Flow rate: Adjust the flow rate so that the retention time order with the resolution between these peaks being not less
of etizolam is about 7 minutes. than 3.
System suitability— System repeatability: When the test is repeated 6 times
System performance: When the procedure is run with 50 with 10 mL of the standard solution under the above operat-
mL of the standard solution under the above operating con- ing conditions, the relative standard deviation of the ratio of
ditions, the number of theoretical plates and the symmetry the peak area of etizolam to that of the internal standard is
factor of the peak of etizolam are not less than 3000 and not not more than 1.0z.
more than 2.0, respectively.
Containers and storage Containers—Tight containers.
System repeatability: When the test is repeated 6 times
Storage—Light-resistant.
with 50 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
area of etizolam is not more than 2.0z.
Famotidine for Injection
Assay To 20 Etizolam Tablets add 50 mL of water, and stir
until they disintegrate. Add 400 mL of methanol, stir for 20 注射用ファモチジン
minutes, add methanol to make exactly 500 mL, and cen-
trifuge. Pipet an amount of the supernatant liquid, equiva- Add the following next to Bacterial endotoxins:
lent to about 0.2 mg of etizolam (C17H15ClN4S), add exactly
Uniformity of dosage units <6.02> It meets the requirement
10 mL of the internal standard solution, add diluted
of the Mass variation test.
methanol (9 in 10) to make 25 mL, and use this solution as
the sample solution. Separately, weigh accurately about 100 Foreign insoluble matter <6.06> Perform the test according
mg of etizolam for assay, previously dried at 1059 C for 3 to Method 2: it meets the requirement.
hours, and dissolve in diluted methanol (9 in 10) to make
Insoluble particulate matter <6.07> It meets the require-
exactly 100 mL. Pipet 2 mL of this solution, and add diluted
ment.
methanol (9 in 10) to make exactly 100 mL. Pipet 10 mL of
this solution, add exactly 10 mL of the internal standard Sterility <4.06> Perform the test according to the Mem-
solution, add diluted methanol (9 in 10) to make 25 mL, and brane filtration method: it meets the requirement.
use this solution as the standard solution. Perform the test
with 10 mL each of the sample solution and standard solu-
tion as directed under Liquid Chromatography <2.01> ac- Faropenem Sodium Hydrate
cording to the following conditions, and calculate the ratios,
QT and QS, of the peak area of etizolam to that of the inter- ファロペネムナトリウム水和物
nal standard.
Change the Purity to read:
Amount (mg) of etizolam (C17H15ClN4S)
=WS×(QT/QS)×(1/500) Purity
(1) Heavy metals <1.07>—Proceed with 2.0 g of
WS: Amount (mg) of etizolam for assay Faropenem Sodium Hydrate according to Method 4, and
Internal standard solution—A solution of ethyl parahydrox- perform the test. Prepare the control solution with 2.0 mL
ybenzoate in diluted methanol (9 in 10) (1 in 50,000). of Standard Lead Solution (not more than 10 ppm).
Operating conditions— (2) Related substances—Dissolve a quantity of Faropen-
Detector: An ultraviolet absorption photometer (wave- em Sodium Hydrate equivalent to 0.10 g (potency) in 200
length: 240 nm). mL of water, and use this solution as the sample solution.
Column: A stainless steel column 4.6 mm in inside di- Pipet 2 mL of the sample solution, add water to make exact-
ameter and 15 cm in length, packed with octadecylsilanized ly 200 mL, and use this solution as the standard solution.
silica gel for liquid chromatography (5 mm particle di- Perform the test with exactly 20 mL each of the sample solu-
ameter). tion and standard solution as directed under Liquid Chro-
Column temperature: A constant temperature of about matography <2.01> according to the following conditions,
359C. and determine each peak area by the automatic integration
Mobile phase: Dissolve 1.36 g of potassium dihydrogen method: the peak area of the epimer, having the relative
phosphate in water to make 1000 mL, and adjust the pH to retention time of about 1.1 with respect to faropenem, ob-
3.5 with diluted phosphoric acid (1 in 10). To 550 mL of this tained from the sample solution is not larger than 3/10 times
solution add 450 mL of acetonitrile. the peak area of faropenem from the standard solution, and
Flow rate: Adjust the flow rate so that the retention time the total area of the peaks other than the peak of faropenem
1886 Official Monographs Supplement I, JP XV

from the sample solution is not larger than 1/2 times the Operating conditions—
peak area of faropenem from the standard solution. Detector: An ultraviolet absorption photometer (wave-
Operating conditions— length: 240 nm).
Column, column temperature, mobile phase, and flow Column: A stainless steel column 4 mm in inside diameter
rate: Proceed as directed in the operating conditions in the and 25 cm in length, packed with octadecylsilanized silica gel
Assay. for liquid chromatography (5 mm in particle diameter).
Detector: An ultraviolet absorption photometer (wave- Column temperature: A constant temperature of about
length: 240 nm). 409C.
Time span of measurement: About 6 times as long as the Mobile phase A: Dissolve 6.12 g of potassium dihydrogen
retention time of faropenem, beginning after the solvent phosphate, 1.79 g of disodium hydrogen phosphate dodeca-
peak. hydrate and 1.61 g of tetra n-butylammonium bromide in
System suitability— water to make 1000 mL.
Test for required detectability: To exactly 2 mL of the Mobile phase B: A mixture of the mobile phase A and
standard solution add water to make exactly 20 mL. Con- acetonitrile (1:1).
firm that the peak area of faropenem obtained with 20 mL of Flowing of mobile phase: Control the gradient by mixing
this solution is equivalent to 7 to 13z of that with 20 mL of the mobile phases A and B as directed in the following table.
the standard solution.
System performance: When the procedure is run with 20 Time after injection Mobile phase Mobile phase
of sample (min) A (volz) B (volz)
mL of the standard solution obtained in the Assay under the
above operating conditions, m-hydroxyacetophenone and 0 – 54 84ª 30 16ª 70
faropenem are eluted in this order with the resolution be-
Flow rate: About 1.5 mL per minute
tween these peaks being not less than 1.5.
Time span of measurement: About 2.5 times as long as the
System repeatability: When the test is repeated 6 times
retention time of faropenem, beginning after the solvent
with 20 mL of the standard solution under the above operat-
peak.
ing conditions, the relative standard deviation of the peak
System suitability—
area of faropenem is not more than 2.0z.
Test for required detectability: To exactly 2 mL of the
standard solution add water to make exactly 20 mL. Con-
firm that the peak area of faropenem obtained with 20 mL of
Faropenem Sodium for Syrup this solution is equivalent to 7 to 13z of that with 20 mL of
the standard solution.
シロップ用ファロペネムナトリウム
System performance: When the procedure is run with 20
mL of the standard solution obtained in the Assay under the
Add the following next to Identification:
above operating conditions, m-hydroxyacetophenone and
Purity Related substances—Powder Faropenem Sodium faropenem are eluted in this order with the resolution be-
for Syrup, if necessary. To a part of the powder, equivalent tween these peaks being not less than 11.
to about 25 mg (potency) of Faropenem Sodium Hydrate System repeatability: When the test is repeated 6 times
according to the labeled amount, add about 10 mL of water, with 20 mL of the standard solution under the above operat-
shake well, then add water to make exactly 50 mL, and ing conditions, the relative standard deviation of the peak
filter. Discard the first 10 mL of the filtrate, and use the sub- area of faropenem is not more than 3.0z.
sequent filtrate as the sample solution. Pipet 2 mL of the
sample solution, add water to make exactly 200 mL, and use
this solution as the standard solution. Perform the test with Faropenem Sodium Tablets
exactly 20 mL each of the sample solution and standard solu-
tion as directed under Liquid Chromatography <2.01> ac- ファロペネムナトリウム錠
cording to the following conditions, and determine each
peak area of both solutions by the automatic integration Add the following next to Identification:
method: the area of the peak of cleaved derivative, having
Purity Related substances—Powder not less than 5
the relative retention time of about 0.71 with respect to
Faropenem Sodium Tablets. To a part of the powder,
faropenem, obtained from the sample solution is not larger
equivalent to about 25 mg (potency) of Faropenem Sodium
than 1.5 times the peak area of faropenem from the standard
Hydrate according to the labeled amount, add about 10 mL
solution, and the total area of the peaks other than the peak
of water, shake well, then add water to make exactly 50 mL,
of faropenem from the sample solution is not larger than 2
and filter. Discard the first 10 mL of the filtrate, and use the
times the peak area of faropenem from the standard solu-
subsequent filtrate as the sample solution. Pipet 2 mL of the
tion. For these calculations use the area of the peak of
sample solution, add water to make exactly 200 mL, and use
cleaved derivative, having the relative retention time of 0.71,
this solution as the standard solution. Perform the test with
after multiplying by the relative response factor 0.37.
exactly 20 mL each of the sample solution and standard solu-
tion as directed under Liquid Chromatography <2.01>
Supplement I, JP XV Official Monographs 1887

according to the following conditions, and determine each Add the following:
peak area of both solutions by the automatic integration
method: the area of the peak of cleaved derivative, having Felbinac
the relative retention time of about 0.71 with respect to
faropenem, obtained from the sample solution is not larger フェルビナク
than 1.5 times the peak area of faropenem from the standard
solution, and the total area of the peaks other than the peak
of faropenem from the sample solution is not larger than 2.5
times the peak area of faropenem from the standard solu-
tion. For these calculation, use the area of the peak of
C14H12O2: 212.24
cleaved derivative, having the relative retention time of 0.71,
Biphenyl-4-ylacetic acid [5728-52-9]
after multiplying by the relative response factor 0.37.
Operating conditions—
Felbinac, when dried, contains not less than 98.5z
Detector: An ultraviolet absorption photometer (wave-
and not more than 101.0z of C14H12O2.
length: 240 nm).
Column: A stainless steel column 4 mm in inside diameter Description Felbinac occurs as white to pale yellowish
and 25 cm in length, packed with octadecylsilanized silica gel white crystals or crystalline powder.
for liquid chromatography (5 mm in particle diameter). It is soluble in methanol and in acetone, sparingly soluble
Column temperature: A constant temperature of about in ethanol (95), and practically insoluble in water.
409C.
Identification (1) Determine the absorption spectrum of
Mobile phase A: Dissolve 6.12 g of potassium dihydrogen
a solution of Felbinac in methanol (95) (1 in 200,000) as
phosphate, 1.79 g of disodium hydrogen phosphate dodeca-
directed under Ultraviolet-visible Spectrophotometry <2.24>,
hydrate and 1.61 g of tetra n-butylammonium bromide in
and compare the spectrum with the Reference Spectrum:
water to make 1000 mL.
both spectra exhibit similar intensities of absorption at the
Mobile phase B: A mixture of the mobile phase A and
same wavelengths.
acetonitrile (1:1).
(2) Determine the infrared absorption spectrum of Fel-
Flowing of mobile phase: Control the gradient by mixing
binac as directed in the potassium bromide disc method un-
the mobile phases A and B as directed in the following table.
der Infrared Spectrophotometry <2.25>, and compare the
Time after injection Mobile phase Mobile phase spectrum with the Reference Spectrum: both spectra exhibit
of sample (min) A (volz) B (volz) similar intensities of absorption at the same wave numbers.

0 – 54 84ª 30 16ª 70 Melting point <2.60> 163 – 1669


C

Flow rate: About 1.5 mL per minute Purity (1) Chloride <1.03>—Dissolve 1.0 g of Felbinac in
Time span of measurement: About 2.5 times as long as the 40 mL of acetone, add 6 mL of dilute nitric acid and water
retention time of faropenem, beginning after the solvent to make 50 mL. Perform the test using this solution as the
peak. test solution. Prepare the control solution by combining 0.3
System suitability— mL of 0.01 mol/L hydrochloric acid VS, 40 mL of acetone
Test for required detectability: To exactly 2 mL of the and 6 mL of dilute nitric acid, and add water to make 50 mL
standard solution add water to make exactly 20 mL. Con- (not more than 0.011z).
firm that the peak area of faropenem obtained with 20 mL of (2) Heavy metals <1.07>—Proceed with 1.0 g of Felbinac
this solution is equivalent to 7 to 13z of that with 20 mL of according to Method 2, and perform the test. Prepare the
the standard solution. control solution with 1.0 mL of Standard Lead Solution (not
System performance: When the procedure is run with 20 more than 10 ppm).
mL of the standard solution obtained in the Assay under the (3) Related substances—Dissolve 0.10 g of Felbinac in
above operating conditions, m-hydroxyacetophenone and 10 mL of acetone, and use this solution as the sample solu-
faropenem are eluted in this order with the resolution be- tion. Pipet 2 mL of this solution, and add acetone to make
tween these peaks being not less than 11. exactly 100 mL. Pipet 5 mL of this solution, add acetone to
System repeatability: When the test is repeated 6 times make exactly 50 mL, and use this solution as the standard
with 20 mL of the standard solution under the above operat- solution. Perform the test with these solutions as directed
ing conditions, the relative standard deviation of the peak under Thin-layer Chromatography <2.03>. Spot 10 mL each
area of faropenem is not more than 3.0z. of the sample solution and standard solution on a plate of
silica gel with fluorescent indicator for thin-layer chro-
matography. Develop the plate with a mixture of heptane,
Add the following next to Uniformity of dosage acetone, and acetic acid (100) (50:25:1) to a distance of
units: about 12 cm, and air-dry the plate. Examine the plate under
ultraviolet light (main wavelength: 254 nm): spots other than
Disintegration <6.09> It meets the requirement.
the principal spot from the sample solution are not more in-
1888 Official Monographs Supplement I, JP XV

tense than the spot from the standard solution. hydrochloric acid and water to make exactly 100 mL. Pipet
60 mL of this solution, add 0.5 g of zinc powder, shake fre-
Loss on drying <2.41> Not more than 0.3z (1 g, 1059C, 3
quently, allow to stand for 20 minutes, and filter the solu-
hours).
tion through a dried filter paper. Discard the first 10 mL of
Residue on ignition <2.44> Not more than 0.1z (1 g). the filtrate, pipet 10 mL of the subsequent filtrate, add water
to make exactly 100 mL, and use this solution as the stan-
Assay Weigh accurately about 0.5 g of Felbinac, previous-
dard solution. Pipet 4 mL each of the sample solution and
ly dried, dissolve in 50 mL of methanol, add 15 mL of water,
standard solution, add 1 mL of water, 1 mL of dilute
and titrate <2.50> with 0.1 mol/L sodium hydroxide VS
hydrochloric acid and 1 mL of sodium nitrite solution (1 in
(potentiometric titration). Perform a blank determination in
1000) to them, mix, and allow to stand for 2 minutes. To
the same manner, and make any necessary correction.
these solutions add 1 mL of a solution of ammonium
Each mL of 0.1 mol/L sodium hydroxide VS amidosulfate (1 in 200), shake, and allow them to stand for 2
=21.22 mg of C14H12O2 minutes. To these solutions add 1 mL of a solution of N,N-
diethyl-N'-1-naphthylethylenediamine oxalate (1 in 1000),
Containers and storage Containers—Tight containers.
shake, allow to stand for 10 minutes, and add water to make
exactly 20 mL. Separately, pipet 30 mL of the sample stock
solution, add 20 mL of dilute hydrochloric acid, and add
Folic Acid Injection water to make exactly 100 mL. Pipet V mL of this solution,
and add water to make exactly V? mL so that each mL con-
葉酸注射液
tains about 15 mg of folic acid (C19H19N7O6). With exactly 4
mL of this solution perform the same procedure described
Add the following next to Extractable volume:
above for obtaining the sample solution, and use the solu-
Foreign insoluble matter <6.06> Perform the test according tion so obtained as the blank solution. Determine the absor-
to Method 1: it meets the requirement. bances at 550 nm, AT, AS and AC, of the solutions obtained
from the sample solution and standard solution, and the
Insoluble particulate matter <6.07> It meets the require-
blank solution as directed under Ultraviolet-visible Spec-
ment.
trophotometry <2.24>, using a control solution obtained with
Sterility <4.06> Perform the test according to the Mem- 4 mL of water in the same manner as described above.
brane filtration method: it meets the requirement.
Amount (mg) of folic acid (C19H19N7O6)
=WS×{(AT-AC)/AS}×(V?/V)×(1/10)

Folic Acid Tablets WS: Amount (mg) of Folic Acid Reference Standard, cal-
culated on the anhydrous basis
葉酸錠

Add the following next to Identification: Delete the following two Monographs:
Uniformity of dosage units <6.02> Perform the test accord-
ing to the following method: it meets the requirement of the Fosfestrol
Content uniformity test. ホスフェストロール
To 1 tablet of Folic Acid Tablets add 50 mL of dilute sodi-
um hydroxide TS, shake frequently, and filter. Wash the Fosfestrol Tablets
residue with dilute sodium hydroxide TS, combine the
ホスフェストロール錠
filtrate and the washings, then add dilute sodium hydroxide
TS to make exactly 100 mL, and use this solution as the sam-
ple stock solution. Pipet 30 mL of this solution, add 20 mL
of dilute hydrochloric acid and water to make exactly 100 Fructose Injection
mL. Pipet 60 mL of this solution, add 0.5 g of zinc powder,
果糖注射液
shake frequently, allow to stand for 20 minutes, and filter
the solution through a dried filter paper. Discard the first 10
Delete the Pyrogen and add the following next
mL of the filtrate, pipet V mL of the subsequent filtrate, add
to Residue on ignition:
water to make exactly V? mL so that each mL contains about
15 mg of folic acid (C19H19N7O6), and use this solution as the Bacterial endotoxins <4.01> Less than 0.5 EU/mL.
sample solution. Separately, weigh accurately about 50 mg
of Folic Acid Reference Standard (separately determine the
water <2.48> in the same manner as Folic Acid), and dissolve Add the following next to Extractable volume:
in dilute sodium hydroxide TS to make exactly 100 mL.
Foreign insoluble matter <6.06> Perform the test according
Pipet 30 mL of this solutions, add 20 mL of dilute
to Method 1: it meets the requirement.
Supplement I, JP XV Official Monographs 1889

Insoluble particulate matter <6.07> It meets the require- mL of water, warm to 409 C to dissolve, and after cooling,
ment. add water to make exactly 50 mL. Determine the optical ro-
tation of this solution in a 100-mm cell, within 60 minutes.
Sterility <4.06> Perform the test according to the Mem-
brane filtration method: it meets the requirement. pH <2.54> The pH of a solution prepared by dissolving
1.0 g of L-Glutamine in 50 mL of water is between 4.5 and
6.0.
Gabexate Mesilate Purity (1) Clarity and color of solution—A solution ob-
tained by dissolving 0.5 g of L-Glutamine in 20 mL of water
ガベキサートメシル酸塩
is clear and colorless.
(2) Chloride <1.03>—Perform the test with 0.5 g of
Change the Identification (4) to read:
L-Glutamine. Prepare the control solution with 0.30 mL of
Identification (4) A 0.1 g portion of Gabexate Mesilate 0.01 mol/L hydrochloric acid VS (not more than 0.021z).
responds to the Qualitative Tests <1.09> (1) for mesilate. (3) Sulfate <1.14>—Perform the test with 0.6 g of
L-Glutamine. Prepare the control solution with 0.35 mL of
0.005 mol/L sulfuric acid VS (not more than 0.028z).
Glucose Injection (4) Ammonium <1.02>—Perform the test with 0.10 g of
L-Glutamine, using the distillation under reduced pressure.
ブドウ糖注射液 Prepare the control solution with 10.0 mL of Standard Am-
monium Solution. The temperature of the water bath is
Add the following next to Extractable volume: 459C (not more than 0.1z).
Foreign insoluble matter <6.06> Perform the test according (5) Heavy metals <1.07>—Proceed with 1.0 g of L-Gluta-
to Method 1: it meets the requirement. mine according to Method 1, and perform the test. Prepare
the control solution with 1.0 mL of Standard Lead Solution
Insoluble particulate matter <6.07> It meets the require- (not more than 10 ppm).
ment. (6) Iron <1.10>—Prepare the test solution with 1.0 g of
L-Glutamine according to Method 1, and perform the test
Sterility <4.06> Perform the test according to the Mem-
brane filtration method: it meets the requirement. according to Method A. Prepare the control solution with
1.0 mL of Standard Iron Solution (not more than 10 ppm).
(7) Related substances—Dissolve 0.10 g of L-Glutamine
in 10 mL of water, and use this solution as the sample solu-
Add the following:
tion. Pipet 1 mL of this solution, add water to make exactly
10 mL. Pipet 1 mL of this solution, add water to make ex-
L-Glutamine
actly 50 mL, and use this solution as the standard solution.
L-グルタミン Perform the test with these solutions as directed under Thin-
layer Chromatography <2.03>. Spot 5 mL each of the sample
solution and standard solution on a plate of silica gel for
thin-layer chromatography. Then develop with a mixture of
1-butanol, water and acetic acid (100) (3:1:1) to a distance of
about 10 cm, and dry the plate at 809C for 30 minutes. Spray
C5H10N2O3: 146.14
evenly a solution of ninhydrin in a mixture of methanol and
(2S)-2,5-Diamino-5-oxopentanoic acid [56-85-9]
acetic acid (100) (97:3) (1 in 100) on the plate, and heat at
809C for 10 minutes: the spot other than the principal spot
L-Glutamine, when dried, contains not less than
obtained with the sample solution is not more intense than
99.0z and not more than 101.0z of C5H10N2O3.
the spot with the standard solution.
Description L-Glutamine occurs as white crystals or a crys-
Loss on drying <2.41> Not more than 0.3z (1 g, 1059
C, 3
talline powder. It has a slight characteristic taste.
hours).
It is freely soluble in formic acid, soluble in water, and
practically insoluble in ethanol (99.5). Residue on Ignition <2.44> Not more than 0.1z (1 g).
Identification Determine the infrared absorption spectrum Assay Weigh accurately about 0.15 g of L-Glutamine,
of L-Glutamine as directed in the potassium bromide disk previously dried, dissolve in 3 mL of formic acid, add 50 mL
method under Infrared Spectrophotometry <2.25>, and com- of acetic acid (100), and titrate <2.50> with 0.1 mol/L per-
pare the spectrum with the Reference Spectrum: both spec- chloric acid VS (potentiometric titration). Perform a blank
tra exhibit similar intensities of absorption at the same wave determination in the same manner, and make any necessary
numbers. correction.
Optical rotation <2.49> [a]20D : +6.3 – +7.39 Weigh ac- Each mL of 0.1 mol/L perchloric acid VS
curately about 2 g of L-Glutamine, previously dried, add 45 =14.61 mg of C5H10N2O3
1890 Official Monographs Supplement I, JP XV

Containers and storage Containers—Tight containers. filter through a membrane filter with a pore size not exceed-
ing 0.5 mm, discard the first 5 mL of the filtrate, and use the
subsequent filtrate as the sample solution. Separately, weigh
Add the following: accurately an amount of Griseofulvin Reference Standard,
equivalent to about 40 mg (potency), and dissolve in N,N-
Griseofulvin Tablets dimethylformamide to make exactly 20 mL. Pipet 5 mL of
this solution, add exactly 20 mL of the internal standard so-
グリセオフルビン錠 lution, add water to make 100 mL, and use this solution as
the standard solution. Perform the test with 10 mL each of
Griseofulvin Tablets contain not less than 95.0z the sample solution and standard solution as directed under
and not more than 105.0z of the labeled amount of Liquid Chromatography <2.01> according to the following
griseofulvin (C17H17ClO6: 352.77). conditions, and calculate the ratios, QT and QS, of the peak
area of griseofulvin to that of the internal standard.
Method of preparation Prepare as directed under Tablets,
with Griseofulvin. Amount [mg (potency)] of griseofulvin (C17H17ClO6)
=WS×(QT/QS)×(25/2)
Identification To a quantity of powdered Griseofulvin
Tablets, equivalent to 15 mg (potency) of Griseofulvin ac- WS: Amount [mg (potency)] of Griseofulvin Reference
cording to the labeled amount, add 100 mL of ethanol (95), Standard
shake vigorously, and filter. To 1 mL of the filtrate add
Internal standard solution—A solution of butyl parahydrox-
ethanol (95) to make 10 mL, and determine the absorption
ybenzoate in acetonitrile (1 in 2000).
spectrum of this solution as directed under Ultraviolet-visi-
Operating conditions—
ble Spectrophotometry <2.24>: it exhibits maxima between
Proceed as directed in the operating conditions in the
234 nm and 238 nm, between 290 nm and 294 nm, and be-
Assay under Griseofulvin.
tween 323 nm and 328 nm.
System suitability—
Uniformity of dosage units <6.02>—Perform the test accord- System performance: When the procedure is run with 10
ing to the following method: it meets the requirement of the mL of the standard solution under the above operating con-
Content uniformity test. ditions, griseofulvin and the internal standard are eluted in
Take 1 tablet of Griseofulvin Tablets, add V/5 mL of this order with the resolution between these peaks being not
water, treat with ultrasonic waves to disintegrate the tablet, less than 4.
add N,N-dimethylformamide to make 5V/8 mL, shake System repeatability: When the test is repeated 6 times
vigorously for 20 minutes, add N,N-dimethylformamide to with 10 mL of the standard solution under the above operat-
make exactly V mL so that each mL contains 1.25 mg ing conditions, the relative standard deviation of the ratio of
(potency) of Griseofulvin, and centrifuge. Pipet 8 mL of the the peak area of griseofulvin to that of the internal standard
supernatant liquid, add exactly 20 mL of the internal stan- is not more than 1.0z.
dard solution, add water to make 100 mL, filter through a
Containers and storage Containers—Tight containers.
membrane filter with a pore size not exceeding 0.5 mm, dis-
card the first 5 mL of the filtrate, and use the subsequent
filtrate as the sample solution. Then, proceed as directed un-
der the Assay. Hydralazine Hydrochloride Tablets
Amount [mg (potency)] of griseofulvin (C17H17ClO6) ヒドララジン塩酸塩錠
=WS×(QT/QS)×(V/32)
Add the following next to Identification:
WS: Amount [mg (potency)] of Griseofulvin Reference
Standard Uniformity of dosage units <6.02> Perform the test accord-
ing to the following method: it meets the requirement of the
Internal standard solution—A solution of butyl parahydrox-
Content uniformity test.
ybenzoate in acetonitrile (1 in 2000).
To 1 tablet of Hydralazine Hydrochloride Tablets add 25
Disintegration <6.09> It meets the requirement. mL of 0.1 mol/L hydrochloric acid TS, disperse the tablet
into a small particles using ultrasonic waves, then shake
Assay Weigh accurately not less than 20 Griseofulvin
well, add 0.1 mol/L hydrochloric acid TS to make exactly 50
Tablets, and pulverize into a powder. Weigh accurately a
mL, and centrifuge. Pipet V mL of the supernatant liquid,
portion of the powder, equivalent to about 0.5 g (potency)
add 0.1 mol/L hydrochloric acid TS to make exactly V? mL
of Griseofulvin, add 50 mL of water, and treat with ultra-
so that each mL contains about 10 mg of hydralazine
sonic waves. Add 100 mL of N,N-dimethylformamide,
hydrochloride (C8H8N4.HCl), and use this solution as the
shake vigorously for 20 minutes, and add N,N-dimethylfor-
sample solution. Separately, weigh accurately about 25 mg
mamide to make exactly 250 mL. Centrifuge this solution,
of hydralazine hydrochloride for assay, previously dried at
pipet 5 mL of the supernatant liquid, add exactly 20 mL of
1059 C for 3 hours, dissolve in 0.1 mol/L hydrochloric acid
the internal standard solution, add water to make 100 mL,
Supplement I, JP XV Official Monographs 1891

TS to make exactly 50 mL. Pipet 2 mL of this solution, add ously dried at 1059C for 1 hour, add 90 g of a mixture of
0.1 mol/L hydrochloric acid TS to make exactly 100 mL, methanol and dichloromethane in equal mass ratio, and stir
and use this solution as the standard solution. Determine the to dissolve. Determine the viscosity at 20±0.19
C as directed
absorbances at 260 nm, AT1 and AS1, and at 350 nm, AT2 and in Method 1 under Viscosity Determination: the viscosity is
AS2, of the sample solution and standard solution as directed not less than 80z and not more than 120z of the labeled
under Ultraviolet-visible Spectrophotometry <2.24>. unit.

Amount (mg) of hydralazine hydrochloride (C8H8N4.HCl) Purity (1) Chloride <1.03>—Dissolve 1.0 g of Hypromel-
=WS×{(AT1-AT2)/(AS1—AS2)}×(V?/V)×(1/50) lose Phthalate in 40 mL of 0.2 mol/L sodium hydroxide VS,
add 1 drop of phenolphthalein TS, and add dilute nitric acid
WS: Amount (mg) of hydralazine hydrochloride for assay
dropwise with vigorous stirring until the red color is dis-
charged. Further add 20 mL of dilute nitric acid with stir-
ring. Heat on a water bath with stirring until the gelatinous
Change to read:
precipitate formed turns to granular particles. After cooling,
centrifuge, and take off the supernatant liquid. Wash the
Hypromellose Phthalate precipitate with three 20-mL portions of water by centrifug-
ing each time, combine the supernatant liquid and the wash-
ヒプロメロースフタル酸エステル
ings, add water to make 200 mL, and filter. Perform the test
with 50 mL of the filtrate. Control solution: To 0.50 mL of
[9050-31-1] 0.01 mol/L hydrochloric acid VS add 10 mL of 0.2 mol/L
This monograph is harmonized with the European Phar- sodium hydroxide VS and 7 mL of dilute nitric acid, and add
macopoeia and the U.S. Pharmacopeia. The parts of the text water to make 50 mL (not more than 0.07z).
that are not harmonized are marked with symbols ( ).

(2) Heavy metals <1.07>—Proceed with 2.0 g of
Hypromellose Phthalate according to Method 2, and per-
Hypromellose Phthalate is a monophthalic acid form the test. Prepare the control solution with 2.0 mL of
ester of hypromellose. Standard Lead Solution (not more than 10 ppm).
It contains methoxy group (-OCH3: 31.03), (3) Phthalic acid—Weigh accurately about 0.2 g of
hydroxypropoxy group (-OCH2CHOHCH3: 75.09), Hypromellose Phthalate, add about 50 mL of acetonitrile to
and carboxybenzoyl group ( - COC6H4COOH: dissolve partially with the aid of ultrasonic waves, add 10
149.12). mL of water, and dissolve further with the ultrasonic waves.
It contains not less than 21.0z and not more than After cooling, add acetonitrile to make exactly 100 mL, and
35.0z of carboxybenzoyl group, calculated on the use this solution as the sample solution. Separately, weigh
anhydrous basis. accurately about 12.5 mg of phthalic acid, dissolve in about
 Its substitution type and its viscosity in millipascal 125 mL of acetonitrile by mixing, add 25 mL of water, then
second (mPa・s) are shown on the label. add acetonitrile to make exactly 250 mL, and use this solu-
tion as the standard solution. Perform the test with exactly
Substitution Carboxybenzoyl group (z) 10 mL each of the sample solution and standard solution as
Type Min. Max. directed under Liquid Chromatography <2.01> according to
200731 27.0 35.0 the following conditions, and determine the peak areas of
220824 21.0 27.0 phthalic acid, AT and AS, of both solutions: amount of
 phthalic acid (C8H6O4: 166.13) is not more than 1.0z.

Description Hypromellose Phthalate occurs as white Amount (z) of phthalic acid=(WS/WT)×(AT/AS)×40


powder or granules. WS: Amount (mg) of phthalic acid
It is practically insoluble in water, in acetonitrile and in WT: Amount (mg) of sample, calculated on the anhydrous
ethanol (99.5). basis
It becomes a viscous liquid when a mixture of methanol
and dichloromethane (1:1) or a mixture of ethanol (99.5) Operating conditions—
and acetone (1:1) is added. Detector: An ultraviolet absorption photometer (wave-
It dissolves in sodium hydroxide TS. length: 235 nm).
Column: A stainless steel column about 4.6 mm in inside

Identification Determine the infrared absorption spec- diameter and 25 cm in length, packed with octadecyl-
trum of Hypromellose Phthalate as directed in the potassi- silanized silica gel for liquid chromatography (3 to 10 mm in
um bromide disk method under Infrared Spectrophotometry particle diameter).
<2.25>, and compare the spectrum with the Reference Spec- Column temperature: A constant temperature of about
trum: both spectra exhibit similar intensities of absorption at 209C.
the same wave numbers. Mobile phase: A mixture of 0.1z trifluoroacetic acid and
Viscosity <2.53> To 10 g of Hypromellose Phthalate, previ- acetonitrile (9:1).
Flow rate: About 2.0 mL per minute.
1892 Official Monographs Supplement I, JP XV

System suitability— compare the spectrum with the Reference Spectrum: both
System performance: When the procedure is run with 10
spectra exhibit similar intensities of absorption at the same
mL of the standard solution under the above operating con- wavelengths.
ditions, the number of theoretical plates and the symmetry (2) Determine the infrared absorption spectrum of
factor of the peak of phthalic acid are not less than 2500 and Ibudilast as directed in the potassium bromide disk method
not more than 1.5, respectively. under Infrared Spectrophotometry <2.25>, and compare the
System repeatability: When repeat the test 6 times with 10 spectrum with the Reference Spectrum: both spectra exhibit
mL of the standard solution under the above operating con- similar intensities of absorption at the same wave numbers.
ditions, the relative standard deviation of the peak area of
Melting point <2.60> 54 – 589C
phthalic acid is not more than 1.0z.
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
Water <2.48> Not more than 5.0z (1 g, direct titration,
Ibudilast according to Method 2, and perform the test. Pre-
using a mixture of ethanol (99.5) and dichloromethane (3:2)
pare the control solution with 2.0 mL of Standard Lead
instead of methanol for Karl Fischer method).
Solution (not more than 20 ppm).
Residue on ignition <2.44> Not more than 0.2z (1 g). (2) Related substances—Dissolve 50 mg of Ibudilast in
50 mL of the mobile phase, and use this solution as the
Assay Weigh accurately about 1 g of Hypromellose Phtha-
sample solution. Pipet 1 mL of the sample solution, and add
late, dissolve in 50 mL of a mixture of ethanol (95), acetone
the mobile phase to make exactly 50 mL. Pipet 1 mL of this
and water (2:2:1), and titrate <2.50> with 0.1 mol/L sodium
solution, add the mobile phase to make exactly 20 mL, and
hydroxide VS (indicator: 2 drops of phenolphthalein TS).
use this solution as the standard solution. Perform the test
Perform a blank determination in the same manner, and
with exactly 10 mL each of the sample solution and standard
make any necessary correction.
solution as directed under Liquid Chromatography <2.01>
Amount (z) of carboxybenzoyl group (C8H5O3) according to the following conditions, and determine each
={(0.01×149.1×V)/W}-{(2×149.1×P)/166.1} peak area by the automatic integration method: the peak
area other than ibudilast obtained from the sample solution
P: Amount (z) of phthalic acid obtained in the Purity (3)
is not larger than the peak area of ibudilast from the stan-
V: Amount (mL) of 0.1 mol/L sodium hydroxide VS con-
dard solution, and the total area of the peaks other than
sumed
ibudilast from the sample solution is not larger than 3 times
W: Amount (g) of sample, calculated on the anhydrous
the peak area of ibudilast from the standard solution.
basis
Operating conditions—
Containers and storage Containers—Tight containers. Detector: An ultraviolet absorption photometer (wave-
length: 292 nm).
Column: A stainless steel column 2.6 mm in inside di-
Add the following: ameter and 15 cm in length, packed with silica gel for liquid
chromatography (5 mm in particle diameter).
Ibudilast Column temperature: A constant temperature of about
259C.
イブジラスト Mobile phase: A mixture of hexane and ethyl acetate
(50:1)
Flow rate: Adjust the flow rate so that the retention time
of ibudilast is about 9 minutes.
Time span of measurement: About 4 times as long as the
retention time of ibudilast, beginning after the solvent peak.
System suitability—
C14H18N2O: 230.31 Test for required detectability: To exactly 5 mL of the
1-[2-(1-Methylethyl)pyrazolo[1,5-a]pyridin-3-yl]- standard solution add the mobile phase to make exactly 10
2-methylpropan-1-one [50847-11-5] mL. Confirm that the peak area of ibudilast obtained with
10 mL of this solution is equivalent to 40 to 60z of that with
Ibudilast, when dried, contains not less than 98.5z 10 mL of the standard solution.
and not more than 101.0z of C14H18N2O. System performance: To 5 mL of the sample solution add
Description Ibudilast occurs as a white crystalline powder. the mobile phase to make 50 mL. To 2 mL of this solution
It is very soluble in methanol, freely soluble in ethanol add the mobile phase to make 20 mL. When the procedure is
(99.5) and in acetic anhydride, and very slightly soluble in run with 10 mL of this solution under the above operating
water. conditions, the number of theoretical plates and the symmet-
ry factor of the peak of ibudilast are not less than 3500 and
Identification (1) Determine the absorption spectrum of not more than 2.0, respectively.
a solution of Ibudilast in methanol (1 in 250,000) as directed System repeatability: When the test is repeated 6 times
under Ultraviolet-visible Spectrophotometry <2.24>, and with 10 mL of the standard solution under the above operat-
Supplement I, JP XV Official Monographs 1893

ing conditions, the relative standard deviation of the peak WS: Amount (mg) of Imipramine Hydrochloride Refer-
area of ibudilast is not more than 3.0z. ence Standard

Loss on drying <2.41> Not more than 0.3z (1 g, in vacu-


um, 4 hours).
Indometacin Capsules
Residue on ignition <2.44> Not more than 0.1z (1 g).
インドメタシンカプセル
Assay Weigh accurately about 0.2 g of Ibudilast, previous-
ly dried, dissolve in 50 mL of acetic anhydride, and titrate
Add the following next to Purity:
<2.50> with 0.1 mol/L perchloric acid VS (potentiometric
titration). Perform a blank determination in the same Uniformity of dosage units <6.02> Perform the test accord-
manner, and make any necessary correction. ing to the following method: it meets the requirement of the
Content uniformity test.
Each mL of 0.1 mol/L perchloric acid VS
Take out the content of 1 capsule of Indometacin Cap-
=23.03 mg of C14H18N2O
sules, and dissolve in methanol to make exactly V mL so that
Containers and storage Containers—Tight containers. each mL contains about 1 mg of indometacin (C19H16ClNO4).
Filter the solution, discard the first 10 mL of the filtrate,
pipet 5 mL of the subsequent filtrate, add exactly 3 mL of
Idoxuridine Ophthalmic Solution the internal standard solution, then add the mobile phase to
make 100 mL, and use this solution as the sample solution.
イドクスウリジン点眼液 Separately, weigh accurately about 25 mg of Indometacin
Reference Standard, previously dried at 1059C for 4 hours,
Add the following next to Purity: dissolve in methanol to make exactly 25 mL. Pipet 5 mL of
this solution, add exactly 3 mL of the internal standard solu-
Foreign insoluble matter <6.11> It meets the requirement.
tion, then add the mobile phase to make 100 mL, and use
Insoluble particulate matter <6.08> It meets the require- this solution as the standard solution. Then, proceed as
ment. directed in the Assay.

Sterility <4.06> Perform the test according to the Mem- Amount (mg) of indometacin (C19H16ClNO4)
brane filtration method: it meets the requirement. =WS×(QT/QS)×(V/25)

WS: Amount (mg) of Indometacin Reference Standard

Imipramine Hydrochloride Tablets Internal standard solution—A solution of butyl parahydrox-


ybenzoate in methanol (1 in 1000).
イミプラミン塩酸塩錠

Add the following next to Identification: Isotonic Sodium Chloride Solution


Uniformity of dosage units <6.02> Perform the test accord-
生理食塩液
ing to the following method: it meets the requirement of the
Content uniformity test.
Add the following next to Extractable volume:
To 1 tablet of Imipramine Hydrochloride Tablets add ex-
actly 40 mL of 0.01 mol/L hydrochloric acid TS, disperse Foreign insoluble matter <6.06> Perform the test according
the tablet into a small particles using ultrasonic waves, then to Method 1: it meets the requirement.
shake well. Centrifuge the solution, pipet V mL of the super-
Insoluble particulate matter <6.07> It meets the require-
natant liquid, add water to make exactly V? mL so that each
ment.
mL contains about 20 mg of imipramine hydrochloride
(C19H24N2.HCl), and use this solution as the sample solu- Sterility <4.06> Perform the test according to the Mem-
tion. Separately, weigh accurately about 25 mg of Imipra- brane filtration method: it meets the requirement.
mine Hydrochloride Reference Standard, previously dried at
1059 C for 2 hours, dissolve in 0.01 mol/L hydrochloric acid
TS to make exactly 100 mL. Pipet 2 mL of this solution, add
water to make exactly 25 mL, and use this solution as the
standard solution. Determine the absorbances at 251 nm,
AT1 and AS1, and at 330 nm, AT2 and AS2, of the sample solu-
tion and standard solution as directed under Ultraviolet-visi-
ble Spectrophotometry <2.24>.

Amount (mg) of imipramine hydrochloride (C19H24N2.HCl)


=WS×{(AT1-AT2)/(AS1-AS2)}×(V?/V)×(4/125)
1894 Official Monographs Supplement I, JP XV

Add the following: solution as directed under Liquid Chromatography <2.01>


according to the following conditions. Determine each peak
Isoxsuprine Hydrochloride area by the automatic integration method: each peak area
other than isoxsuprine obtained from the sample solution is
イソクスプリン塩酸塩 not larger than the peak area of isoxsuprine from the stan-
dard solution, and the total area of the peaks other than the
peak of isoxsuprine from the sample solution is not larger
than 2 times the peak area of isoxsuprine from the standard
solution.
Operating conditions—
C18H23NO3.HCl: 337.84 Detector: An ultraviolet absorption photometer (wave-
(1RS,2SR)-1-(4-Hydroxyphenyl)-2-{[(2SR)-1- length: 269 nm).
phenoxypropan-2-yl]amino}propan-1-ol Column: A stainless steel column 4.6 mm in inside di-
monohydrochloride [579-56-6] ameter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Isoxsuprine Hydrochloride, when dried, contains ameter).
not less than 99.0z and not more than 101.0z of Column temperature: A constant temperature of about
C18H23NO3.HCl. 409C.
Mobile phase: Dissolve 4.3 g of diammonium hydrogen
Description Isoxsuprine Hydrochloride occurs as a white,
phosphate and 3.2 g of sodium 1-pentane sulfonate in water
powder or crystalline powder.
to make 1000 mL, and adjust to pH 2.5 with phosphoric
It is soluble in formic acid and in methanol, and slightly
acid. To 770 mL of this solution add 230 mL of acetonitrile.
soluble in water and in ethanol (99.5).
Flow rate: Adjust the flow rate so that the retention time
Melting point: about 2049 C (with decomposition).
of isoxsuprine is about 18 minutes.
A solution of Isoxsuprine Hydrochloride in methanol (1 in
Time span of measurement: About 3 times as long as the
50) shows no optical rotation.
retention time of isoxsuprine, beginning after the solvent
Identification (1) Determine the absorption spectrum of peak.
a solution of Isoxsuprine Hydrochloride (1 in 20,000) as System suitability—
directed under Ultraviolet-visible Spectrophotometry <2.24>, Test for required detectability: Pipet 1 mL of the standard
and compare the spectrum with the Reference Spectrum: solution, and add the mobile phase to make exactly 10 mL.
both spectra exhibit similar intensities of absorption at the Confirm that the peak area of isoxsuprine obtained with 10
same wavelengths. mL of this solution is equivalent to 7 to 13z of that with 10
(2) Determine the infrared absorption spectrum of Isox- mL of the standard solution.
suprine Hydrochloride as directed in the potassium chloride System performance: To 1 mL of the sample solution add
disk method under Infrared Spectrophotometry <2.25>, and 2.5 mL of a solution of methyl parahydroxybenzoate (1 in
compare the spectrum with the Reference Spectrum: both 25,000) and the mobile phase to make 50 mL. When the
spectra exhibit similar intensities of absorption at the same procedure is run with 10 mL of this solution under the above
wave numbers. operating conditions, methyl parahydroxybenzoate and
(3) Dissolve 0.5 g of Isoxsuprine Hydrochloride in 50 isoxsuprine are eluted in this order with the resolution be-
mL of water by warming, and cool: the solution responds to tween these peaks being not less than 4.
the Qualitative Tests <1.09> (2) for chloride. System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat-
pH <2.54> Dissolve 0.5 g of Isoxsuprine Hydrochloride in
ing conditions, the relative standard deviation of the peak
50 mL of water by warming, and cool: the pH of the solu-
area of isoxsuprine is not more than 2.5z.
tion is between 4.5 and 6.0.
Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 1
Purity (1) Clarity and color of solution—Dissolve 0.1 g
hour).
of Isoxsuprine Hydrochloride in 10 mL of water, warm if
necessary, and cool: the solution is clear and colorless. Residue on ignition <2.44> Not more than 0.2z (1 g).
(2) Heavy metals <1.07>—Proceed with 1.0 g of Isox-
Assay Weigh accurately about 0.3 g of Isoxsuprine
suprine Hydrochloride according to Method 2, and perform
Hydrochloride, previously dried, dissolve in 5 mL of formic
the test. Prepare the control solution with 2.0 mL of Stan-
acid, add 50 mL of a mixture of acetic anhydride and acetic
dard Lead Solution (not more than 20 ppm).
acid (100) (7:3), and titrate <2.50> with 0.1 mol/L perchloric
(3) Related substances—Dissolve 20 mg of Isoxsuprine
acid VS (potentiometric titration). Perform a blank determi-
Hydrochloride in 20 mL of the mobile phase, and use this
nation in the same manner, and make any necessary correc-
solution as the sample solution. Pipet 1 mL of the sample
tion.
solution, add the mobile phase to make exactly 100 mL, and
use this solution as the standard solution. Perform the test Each mL of 0.1 mol/L perchloric acid VS
with exactly 10 mL each of the sample solution and standard =33.78 mg of C18H23NO3.HCl
Supplement I, JP XV Official Monographs 1895

Containers and storage Containers—Well-closed contain- say, previously dried at 1059C for 1 hour, and dissolve in
ers. water to make exactly 100 mL. Pipet 4 mL of this solution,
add water to make exactly 100 mL, and use this solution as
the standard solution. Perform the test with exactly 10 mL
Add the following: each of the sample solution and standard solution as direct-
ed under Liquid Chromatography <2.01> according to the
Isoxsuprine Hydrochloride Tablets following conditions, and determine the isoxsuprine peak
areas, AT and AS, of both solutions.
イソクスプリン塩酸塩錠
Dissolution rate (z) with respect to the labeled amount of
isoxsuprine hydrochloride (C18H23NO3.HCl)
Isoxsuprine Hydrochloride Tablets contain not less
=WS×(AT/AS)×(V?/V)×(1/C)×36
than 95.0z and not more than 105.0z of the labeled
amount of isoxsuprine hydrochloride (C18H23NO3. WS: Amount (mg) of isoxsuprine hydrochloride for assay
HCl: 337.84). C: Labeled amount (mg) of isoxsuprine hydrochloride
(C18H23NO3.HCl) in 1 tablet
Method of preparation Prepare as directed under Tablets,
with Isoxsuprine Hydrochloride. Operating conditions—
Proceed as directed in the operating conditions in the
Identification To a quantity of powdered Isoxsuprine
Assay.
Hydrochloride Tablets, equivalent to 10 mg of Isoxsuprine
System suitability—
Hydrochloride according to the labeled amount, add 150
System performance: When the procedure is run with 10
mL of water, shake, and then add water to make 200 mL.
mL of the standard solution under the above operating con-
Centrifuge this solution, filter the supernatant liquid
ditions, the number of theoretical plates and the symmetry
through a membrane filter with a pore size not exceeding
factor of the peak of isoxsuprine are not less than 2000 and
0.45 mm, discard the first 10 mL of filtrate, and determine
not more than 2.0, respectively.
the absorption spectrum of the subsequent filtrate as direct-
System repeatability: When the test is repeated 6 times
ed under Ultraviolet-visible Spectrophotometry <2.24>: it ex-
with 10 mL of the standard solution under the above operat-
hibits maxima between 267 nm and 271 nm, and between
ing conditions, the relative standard deviation of the peak
272 nm and 276 nm.
area of isoxsuprine is not more than 2.0z.
Uniformity of dosage units <6.02> Perform the test accord-
Assay Weigh accurately not less than 20 Isoxsuprine
ing to the following method: it meets the requirement of the
Hydrochloride Tablets, and powder. Weigh accurately a
Content uniformity test.
portion of the powder equivalent to about 40 mg of isox-
Add methanol to 1 tablet of Isoxsuprine Hydrochloride
suprine hydrochloride (C18H23NO3.HCl), add 60 mL of
Tablets, and shake to disintegrate. Add methanol to make
methanol, shake for 20 minutes, and then add methanol to
exactly V mL so that each mL contains about 0.4 mg of isox-
make exactly 100 mL. Centrifuge a portion of this solution,
suprine hydrochloride (C18H23NO3.HCl). Centrifuge this so-
filter the supernatant liquid through a membrane filter with
lution, and use the supernatant liquid as the sample solution.
a pore size not exceeding 0.45 mm, discard the first 10 mL of
Then, proceed as directed in the Assay.
filtrate, and use the subsequent filtrate as the sample solu-
Amount (mg) of isoxsuprine hydrochloride tion. Separately, weigh accurately about 40 mg of isox-
(C18H23NO3.HCl) suprine hydrochloride for assay, previously dried at 1059C
=WS×(AT/AS)×V×(1/100) for 1 hour, and dissolve in methanol to make exactly 100
mL. Filter through a membrane filter with a pore size not ex-
WS: Amount (mg) of isoxsuprine hydrochloride for assay
ceeding 0.45 mm, discard the first 10 mL of the filtrate, and
Dissolution <6.10> When the test is performed at 50 revolu- use the subsequent filtrate as the standard solution. Perform
tions per minute according to the Paddle method, using 900 the test with exactly 10 mL each of the sample solution and
mL of water as the dissolution medium, the dissolution rate standard solution as directed under Liquid Chromatography
in 15 minutes of Isoxsuprine Hydrochloride Tablets is not <2.01> according to the following conditions, and determine
less than 80z. the peak areas, AT and AS, of isoxsuprine in each solution.
Start the test with 1 tablet of Isoxsuprine Hydrochloride Amount (mg) of isoxsuprine hydrochloride
Tablets, withdraw not less than 20 mL of the medium at the (C18H23NO3.HCl)
specified minute after starting the test, and filter through a =WS×(AT/AS)
membrane filter with a pore size not exceeding 0.45 mm. Dis-
card the first 10 mL of the filtrate, pipet V mL of the subse- WS: Amount (mg) of isoxsuprine hydrochloride for assay
quent filtrate, add water to make exactly V? mL so that each Operating conditions—
mL contains about 11 mg of isoxsuprine hydrochloride Detector: An ultraviolet absorption photometer (wave-
(C18H23NO3.HCl) according to the labeled amount, and use length: 269 nm).
this solution as the sample solution. Separately, weigh ac- Column: A stainless steel column 4.6 mm in inside di-
curately about 28 mg of isoxsuprine hydrochloride for as- ameter and 15 cm in length, packed with octadecylsilanized
1896 Official Monographs Supplement I, JP XV

silica gel for liquid chromatography (5 mm in particle di- Description Itraconazole occurs as a white powder.
ameter). It is soluble in N,N-dimethylformamide, very slightly
Column temperature: A constant temperature of about soluble in ethanol (99.5), and practically insoluble in water
409C. and in 2-propanol.
Mobile phase: Dissolve 4.3 g of diammonium hydrogen A solution of Itraconazole in N,N-dimethylformamide (1
phosphate and 3.2 g of sodium 1-pentane sulfonate in water in 100) shows no optical rotation.
to make 1000 mL, and adjust to pH 2.5 with phosphoric
Identification (1) Determine the absorption spectrum of
acid. To 600 mL of this solution add 400 mL of methanol.
a solution of Itraconazole in 2-propanol (1 in 100,000) as
Flow rate: Adjust the flow rate so that the retention time
directed under Ultraviolet-visible Spectrophotometry <2.24>,
of isoxsuprine is about 9 minutes.
and compare the spectrum with the Reference Spectrum:
System suitability—
both spectra exhibit similar intensities of absorption at the
System performance: To exactly 1 mL of the standard
same wavelengths.
solution add the mobile phase to make exactly 50 mL. When
(2) Determine the infrared absorption spectrum of
the procedure is run with 10 mL of this solution under the
Itraconazole, previously dried, as directed in the potassium
above operating conditions, the number of theoretical plates
bromide disk method under Infrared Spectrophotometry
and the symmetry factor of the peak of isoxsuprine are not
<2.25>, and compare the spectrum with the Reference Spec-
less than 2000 and not more than 2.0, respectively.
trum: both spectra exhibit similar intensities of absorption at
System repeatability: When the test is repeated 6 times
the same wave numbers.
with 10 mL of the standard solution under the above operat-
(3) Perform the test with Itraconazole as directed under
ing conditions, the relative standard deviation of the peak
Flame Coloration Test <1.04> (2): a green color appears.
area of isoxsuprine is not more than 1.0z.
Melting point <2.60> 166 – 1709
C
Containers and storage Containers—Well-closed contain-
ers. Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
Itraconazole according to Method 2, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead
Add the following: Solution (not more than 20 ppm).
(2) Related substances—Dissolve 0.10 g of Itraconazole
Itraconazole in 10 mL of a mixture of methanol and tetrahydrofuran
(1:1), and use this solution as the sample solution. Pipet 1
イトラコナゾール mL of the sample solution, add the mixture of methanol and
tetrahydrofuran (1:1) to make exactly 100 mL. Pipet 5 mL
of this solution, add the mixture of methanol and tetra-
hydrofuran (1:1) to make exactly 10 mL, and use this solu-
tion as the standard solution. Perform the test with exactly
10 mL each of the sample solution and standard solution as
directed under Liquid Chromatography <2.01> according to
the following conditions. Determine each peak area of each
solution by the automatic integration method: the area of
each peak other than itraconazole obtained from the sample
solution is not larger than the peak area of itraconazole from
the standard solution. Furthermore, the total area of the
peaks other than itraconazole from the sample solution is
not larger than 2.5 times the peak area of itraconazole from
the standard solution.
C35H38Cl2N8O4: 705.63 Operating conditions—
4-(4-{4-[4-({(2RS,4SR)-2-(2,4-Dichlorophenyl)- Detector: An ultraviolet absorption photometer (wave-
2-[(1H-1,2,4-triazol-1-yl)methyl]-1,3-dioxolan- length: 225 nm).
4-yl}methoxy)phenyl]piperazin-1-yl}phenyl)-2-[(1RS)- Column: A stainless steel column 4.6 mm in inside di-
1-methylpropyl]-2,4-dihydro-3H-1,2,4-triazol-3-one ameter and 10 cm in length, packed with octadecylsilanized
4-(4-{4-[4-({(2SR,4RS)-2-(2,4-Dichlorophenyl)- silica gel for liquid chromatography (3 mm in particle di-
2-[(1H-1,2,4-triazol-1-yl)methyl]-1,3-dioxolan- ameter).
4-yl}methoxy)phenyl]piperazin-1-yl}phenyl)-2-[(1RS)- Column temperature: A constant temperature of about
1-methylpropyl]-2,4-dihydro-3H-1,2,4-triazol-3-one 309C.
[84625-61-6] Mobile phase A: A solution of Tetrabutylammonium
hydrogensulfate (17 in 625).
Itraconazole contains not less than 98.5z and not Mobile phase B: Acetonitrile.
more than 101.0z of C35H38Cl2N8O4, calculated on Flowing of the mobile phase: Control the gradient by mix-
the dried basis.
Supplement I, JP XV Official Monographs 1897

ing the mobile phases A and B as directed in the following Identification To a quantity of powdered Josamycin
table. Tablets, equivalent to 10 mg (potency) of Josamycin accord-
ing to the labeled amount, add 100 mL of methanol, shake
Time after injection Mobile phase Mobile phase vigorously, and centrifuge. To 5 mL of the supernatant liq-
of sample (min) A (volz) B (volz)
uid, add methanol to make 50 mL, and determine the ab-
0 – 20 80ª 50 20ª 50 sorption spectrum of this solution as directed under Ultrav-
20 – 25 50 50 iolet-visible Spectrophotometry <2.24>: it exhibits a maxi-
mum between 229 nm and 233 nm.
Flow rate: 1.5 mL per minute.
Time span of measurement: About 2 times as long as the Loss on drying <2.41> Not more than 5.0z (0.5 g, in vacu-
retention time of itraconazole, beginning after the solvent um, 609 C, 3 hours).
peak. Uniformity of dosage units <6.02>—Perform the test accord-
System suitability— ing to the following method: it meets the requirement of the
Test for required detectability: To exactly 1 mL of the Content uniformity test.
standard solution add the mixture of methanol and tetra- Take 1 tablet of Josamycin Tablets, add 5 mL of water,
hydrofuran (1:1) to make exactly 10 mL. Confirm that the and shake vigorously to disintegrate the tablet. Add
peak area of itraconazole obtained from 10 mL of this solu- methanol and then use ultrasonic waves to disperse the parti-
tion is equivalent to 7 to 13z of that from 10 mL of the stan- cles, add methanol to make exactly V mL so that each mL
dard solution. contains about 2 mg (potency) of Josamycin, and cen-
System performance: Dissolve 1 mg of Itraconazole and 1 trifuge. Pipet 3 mL of the supernatant liquid, and add
mg of miconazole nitrate in 20 mL of the mixture of methanol to make exactly 100 mL. Pipet 10 mL of this solu-
methanol and tetrahydrofuran (1:1). When the procedure is tion, add methanol to make exactly 50 mL, and use this solu-
run with 10 mL of this solution under the above operating tion as the sample solution. Separately, accurately weigh
conditions, miconazole and itraconazole are eluted in this about 50 mg (potency) of Josamycin Reference Standard,
order with the resolution between these peaks being not less dissolve in 5 mL of water and methanol to make exactly 25
than 2.0. mL. Pipet 3 mL of this solution, and add methanol to make
System repeatability: When the test is repeated 6 times exactly 100 mL. Pipet 10 mL of this solution, add methanol
with 10 mL of the standard solution under the above operat- to make exactly 50 mL, and use this solution as the standard
ing conditions, the relative standard deviation of the peak solution. Determine the absorbances, AT and AS, of the
area of itraconazole is not more than 2.0z. sample solution and the standard solution at 231 nm as
Loss on drying <2.41> Not more than 0.5z (1 g, 1059C, 4 directed under Ultraviolet-visible Spectrophotometry <2.24>.

hours). However, X in the formula for calculation of acceptance
value is the result of the assay.
Residue on ignition <2.44> Not more than 0.1z (1 g).
Amount [mg (potency)] of josamycin (C42H69NO15)
Assay Weigh accurately about 0.3 g of Itraconazole, dis- =WS×(AT/AS)×(V/25)
solve in 70 mL of a mixture of 2-butanone and acetic acid
(100) (7:1), and titrate <2.50> with 0.1 mol/L perchloric acid WS: Amount [mg (potency)] of Josamycin Reference
VS (potentiometric titration). Perform a blank determina- Standard
tion in the same manner, and make any necessary correc- Disintegration <6.09> It meets the requirement.
tion.
Assay Perform the test according to the Cylinder-plate
Each mL of 0.1 mol/L perchloric acid VS method as directed under Microbial Assay for Antibiotics
=35.28 mg of C35H38Cl2N8O4 <4.02> according to the following conditions.
Containers and storage Containers—Tight containers. (i) Test organism, culture medium, and standard
solutions—Proceed as directed in the Assay under Josamy-
cin.
Add the following: (ii) Sample solutions—Weigh accurately the mass of not
less than 20 Josamycin Tablets and pulverize into a powder.
Josamycin Tablets Weigh accurately a portion of the powder, equivalent to
about 0.3 g (potency) of Josamycin, add 50 mL of
ジョサマイシン錠 methanol, shake vigorously, and add water to make exactly
1000 mL. Take exactly an appropriate amount of this solu-
Josamycin Tablets contain not less than 90.0z and tion, add water to prepare solutions containing 30 mg (poten-
not more than 110.0z of the labeled amount of cy) and 7.5 mg (potency) per mL, and use these solutions as
josamycin (C42H69NO15: 827.99). the high and low concentration sample solutions, respective-
ly.
Method of preparation Prepare as directed under Tablets,
with Josamycin. Containers and storage Containers—Tight containers.
1898 Official Monographs Supplement I, JP XV

Add the following: Standard Lead Solution (not more than 20 ppm).
(2) Related substances—Dissolve 0.8 g of Labetalol
Labetalol Hydrochloride Hydrochloride in 10 mL of methanol, and use this solution
as the sample solution. Pipet 1 mL of the sample solution,
ラベタロール塩酸塩 add methanol to make exactly 200 mL, and use this solution
as the standard solution. Perform the test with these solu-
tions as directed under Thin-layer Chromatography <2.03>.
Spot 5 mL each of the sample solution and standard solution
on a plate of silica gel for thin-layer chromatography. De-
velop the plate with a mixture of ethyl acetate, 2-propanol,
water, and ammonia solution (28) (25:15:8:2) to a distance
of about 10 cm, and air-dry the plate. Allow the plate to
stand in iodine vapor for 30 minutes: the spots other than
the principal spot from the sample solution do not exceed 2
in number and are not more intense than the spot obtained
from the standard solution.

Loss on drying <2.41> Not more than 1.0z (1 g, 1059


C, 3
hours).

Residue on ignition <2.44> Not more than 0.1z (1 g).


C19H24N2O3.HCl: 364.87
2-Hydroxy-5-{(1RS)-1-hydroxy-2-[(1RS)-1-methyl- Isomer ratio Dissolve 5 mg of Labetalol Hydrochloride in
3-phenylpropylamino]ethyl}benzamide monohydrochloride 0.7 mL of a solution of n-butylboronic acid in anhydrous
2-Hydroxy-5-{(1RS)-1-hydroxy-2-[(1SR)-1-methyl- pyridine (3 in 250), allow to stand for 20 minutes, and use
3-phenylpropylamino]ethyl}benzamide monohydrochloride this solution as the sample solution. Perform the test with 2
[32780-64-6] mL of the sample solution as directed under Gas Chro-
matography <2.02> according to the following conditions.
Labetalol Hydrochloride, when dried, contains not Determine the areas of two adjacent peaks, Aa and Ab,
less than 98.5z and not more than 101.0z of where Aa is the peak area of the shorter retention time and
C19H24N2O3.HCl. Ab is the peak area of the longer retention time, using the au-
tomatic integration method: the ratio Ab/(Aa+Ab) is be-
Description Labetalol Hydrochloride occurs as a white tween 0.45 and 0.55.
crystalline powder. Operating conditions—
It is freely soluble in methanol, and sparingly soluble in Detector: A hydrogen flame-ionization detector.
water and in ethanol (99.5). Column: A fused silica column 0.53 mm in inside di-
It dissolves in 0.05 mol/L sulfuric acid TS. ameter and 25 m in length, coated inside with methyl silicone
Melting point: about 1819 C (with decomposition). polymer for gas chromatography in 5 mm thickness.
Identification (1) Determine the absorption spectrum of Column temperature: A constant temperature of about
a solution of Labetalol Hydrochloride in 0.05 mol/L sulfur- 2909 C.
ic acid TS (1 in 20,000) as directed under Ultraviolet-visible Injection port temperature: A constant temperature of
Spectrophotometry <2.24>, and compare the spectrum with about 3509 C.
the Reference Spectrum: both spectra exhibit similar intensi- Detector temperature: A constant temperature of about
ties of absorption at the same wavelengths. 3509 C.
(2) Determine the infrared absorption spectrum of Carrier gas: Helium
Labetalol Hydrochloride as directed in the potassium chlo- Flow rate: Adjust the flow rate so that the retention time
ride disc method under Infrared Spectrophotometry <2.25>, of the peak showing earlier elution of the two peaks of
and compare the spectrum with the Reference Spectrum: labetalol is about 9 minutes.
both spectra exhibit similar intensities of absorption at the System suitability—
same wave numbers. System performance: Proceed with 2 mL of the sample so-
(3) A solution of Labetalol Hydrochloride (1 in 50) lution under the above conditions: the resolution between
responds to the Qualitative Tests <1.09> for chloride. the two labetalol peaks is not less than 1.5.
System repeatability: Repeat the test 6 times under the
pH <2.54> The pH of a solution prepared by dissolving 0.5 g above conditions with 2 mL of the sample solution: the rela-
of Labetalol Hydrochloride in 50 mL of water is between 4.0 tive standard deviation of the ratio of the peak area of
and 5.0. labetalol with the shorter retention time to that of the longer
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of retention time is not more than 2.0z.
Labetalol Hydrochloride according to Method 2, and per- Assay Weigh accurately about 0.3 g of Labetalol
form the test. Prepare the control solution with 2.0 mL of Hydrochloride, previously dried, dissolve in 100 mL of a
Supplement I, JP XV Official Monographs 1899

mixture of acetic anhydride and acetic acid (100) (7:3), and of labetalol hydrochloride (C19H24N2O3.HCl), and use this
titrate <2.50> with 0.1 mol/L perchloric acid VS (potentio- solution as the sample solution. Separately, weigh accurately
metric titration). Perform a blank determination in the same about 20 mg of labetalol hydrochloride for assay, previously
manner and make any necessary correction. dried at 1059 C for 3 hours, and dissolve in 0.05 mol/L sul-
furic acid TS to make exactly 50 mL. Pipet 5 mL of this so-
Each mL of 0.1 mol/L perchloric acid VS
lution, add 0.05 mol/L sulfuric acid TS to make exactly 50
=36.49 mg of C19H24N2O3.HCl
mL, and use this solution as the standard solution. Deter-
Containers and storage Containers—Tight containers. mine the absorbances, AT and AS, of the sample solution
and standard solution at 302 nm as directed under Ultrav-
iolet-visible Spectrophotometry <2.24>.
Add the following:
Amount (mg) of labetalol hydrochloride (C19H24N2O3.HCl)
=WS×(AT/AS)×(V/40)
Labetalol Hydrochloride Tablets
WS: Amount (mg) of labetalol hydrochloride for assay
ラベタロール塩酸塩錠
Dissolution <6.10> When the test is performed at 50 revolu-
tions per minute according to the Paddle method, using 900
mL of water as the dissolution medium, the dissolution rate
Labetalol Hydrochloride Tablets contain not less
in 30 minutes of Labetalol Hydrochloride Tablets is not less
than 93.0z and not more than 107.0z of the labeled
than 75z.
amount of labetalol hydrochloride (C19H24N2O3.HCl:
Start the test with 1 tablet of Labetalol Hydrochloride
364.87).
Tablets, withdraw not less than 20 mL of the medium at spe-
Method of preparation Prepare as directed under Tablets, cified minute after starting the test, and filter through a
with Labetalol Hydrochloride. membrane filter with a pore size not exceeding 0.8 mm. Dis-
card the first 10 mL of the filtrate, pipet V mL of the subse-
Identification (1) To a quantity of powdered Labetalol
quent filtrate, and add water to make exactly V? mL so that
Hydrochloride Tablets equivalent to 5 mg of Labetalol
each mL contains about 50 mg of labetalol hydrochloride
Hydrochloride according to the labeled amount, add 100
(C19H24N2O3.HCl) according to the labeled amount, and use
mL of 0.05 mol/L sulfuric acid TS, shake, and filter. Deter-
this solution as the sample solution. Separately, weigh ac-
mine the absorption spectrum of the filtrate as directed un-
curately about 50 mg of labetalol hydrochloride for assay,
der Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
previously dried at 1059 C for 3 hours, and dissolve in water
a maximum between 300 nm and 304 nm.
to make exactly 100 mL. Pipet 10 mL of this solution, add
(2) To a quantity of powdered Labetalol Hydrochloride
water to make exactly 100 mL, and use this solution as the
Tablets equivalent to 0.25 g of Labetalol Hydrochloride ac-
standard solution. Perform the test with the sample solution
cording to the labeled amount, add 25 mL of methanol,
and standard solution as directed under Ultraviolet-visible
shake vigorously for 30 minutes, filter, and use the filtrate as
Spectrophotometry <2.24>, and determine the absorbances,
the sample solution. Separately, dissolve 10 mg of labetalol
AT and AS, at 302 nm.
hydrochloride in 1 mL of methanol, and use this solution as
the standard solution. Perform the test using these solutions Dissolution rate (z) with respect to the labeled amount of
as directed under Thin-layer Chromatography <2.03>. Spot 5 labetalol hydrochloride (C19H24N2O3.HCl)
mL each of the sample solution and standard solution on a =WS×(AT/AS)×(V?/V)×(1/C)×90
plate of silica gel with fluorescent indicator for thin-layer
WS: Amount (mg) of labetalol hydrochloride for assay
chromatography. Develop the plate with a mixture of ethyl
C: Labeled amount (mg) of labetalol hydrochloride
acetate, 2-propanol, water, and ammonia solution (28)
(C19H24N2O3.HCl) in 1 tablet
(25:15:8:2) to a distance of about 10 cm, and air-dry the
plate. Examine under ultraviolet light (main wavelength: 254 Assay Weigh accurately not less than 20 Labetalol
nm): the principal spot obtained from the sample solution Hydrochloride Tablets, and powder. Weigh accurately a
and the spot obtained from the standard solution show the portion of the powder, equivalent to about 1 g of labetalol
same Rf value. hydrochloride (C19H24N2O3.HCl), add 100 mL of 0.5 mol/L
sulfuric acid TS and 600 mL of water, shake vigorously for
Uniformity of dosage units <6.02> Perform the test accord-
30 minutes, add water to make exactly 1000 mL, and filter.
ing to the following method: it meets the requirement of the
Discard the first 5 mL of the filtrate, pipet 5 mL of the sub-
Content uniformity test.
sequent filtrate, and add 0.05 mol/L sulfuric acid TS to
To 1 tablet of Labetalol Hydrochloride Tablets add 5 mL
make exactly 25 mL. Pipet 5 mL of this solution, add 0.05
of 0.5 mol/L sulfuric acid TS and 30 mL of water, shake
mol/L sulfuric acid TS to make exactly 25 mL, and use this
vigorously for 30 minutes, add water to make exactly 50 mL,
solution as the sample solution. Separately, weigh accurately
and filter. Discard the first 5 mL of the filtrate, pipet 4 mL
about 40 mg of labetalol hydrochloride for assay, previously
of the subsequent filtrate, add 0.05 mol/L sulfuric acid TS
dried at 1059 C for 3 hours, and dissolve in 0.05 mol/L sul-
to make exactly V mL so that each mL contains about 40 mg
furic acid TS to make exactly 100 mL. Pipet 5 mL of this so-
1900 Official Monographs Supplement I, JP XV

lution, add 0.05 mol/L sulfuric acid TS to make exactly 50 to Method 1: it meets the requirement.
mL, and use this solution as the standard solution. Perform
Insoluble particulate matter <6.07> It meets the require-
the test with the sample solution and standard solution as
ment.
directed under Ultraviolet-visible Spectrophotometry <2.24>,
and determine the absorbances, AT and AS, at 302 nm. Sterility <4.06> Perform the test according to the Mem-
brane filtration method: it meets the requirement.
Amount (mg) of labetalol hydrochloride (C19H24N2O3.HCl)
=WS×(AT/AS)×25

WS: Amount (mg) of labetalol hydrochloride for assay Add the following:
Containers and storage Containers—Tight containers.
Manidipine Hydrochloride
マニジピン塩酸塩
Anhydrous Lactose
無水乳糖

Change the origin/limits of content to read:

Anhydrous Lactose is b-lactose or a mixture of b-


lactose and a-lactose.
 The relative quantities of a-lactose and b-lactose
C35H38N4O6.2HCl: 683.62
in Anhydrous Lactose is labeled as the isomer ratio.
3-{2-[4-(Diphenymethyl)piperazin-1-yl]ethyl}
5-methyl (4RS)-2,6-dimethyl-4-(3-nitrophenyl)-
Change the Identification to read:
1,4-dihydropyridine-3,5-dicarboxylate dihydrochloride

Identification Determine the infrared absorption spec- [126229-12-7]
trum of Anhydrous Lactose, previously dried, as directed in
the potassium bromide disk method under Infrared Spec- Manidipine Hydrochloride, when dried, contains
trophotometry <2.25>, and compare the spectrum with the not less than 98.5z and not more than 101.0z of
Reference Spectrum or the spectrum of Anhydrous Lactose C35H38N4O6.2HCl.
Reference Standard: both spectra exhibit similar intensities
Description Manidipine Hydrochloride occurs as white to
of absorption at the same wave numbers.
pale yellow crystals or crystalline powder.
It is freely soluble in dimethylsulfoxide, sparingly soluble
in methanol, slightly soluble in ethanol (99.5), and practical-
Levallorphan Tartrate Injection ly insoluble in water.
レバロルファン酒石酸塩注射液 A solution of Manidipine Hydrochloride in dimethylsul-
foxide (1 in 100) shows no optical rotation.
Add the following next to Identification: Manidipine Hydrochloride turns slightly brown-yellowish
white on exposure to light.
Bacterial endotoxins <4.01> Less than 150 EU/mg. Melting point: about 2079 C (with decomposition).

Add the following next to Extractable volume: Identification (1) Determine the absorption spectrum of
a solution of Manidipine Hydrochloride in methanol (1 in
Foreign insoluble matter <6.06> Perform the test according 100,000) as directed under Ultraviolet-visible Spectrophoto-
to Method 1: it meets the requirement. metry <2.24>, and compare the spectrum with the Reference
Insoluble particulate matter <6.07> It meets the require- Spectrum or the spectrum of a solution of Manidipine
ment. Hydrochloride Reference Standard prepared in the same
manner as the sample solution: both spectra exhibit similar
Sterility <4.06> Perform the test according to the Mem- intensities of absorption at the same wavelengths.
brane filtration method: it meets the requirement. (2) Determine the infrared absorption spectrum of
Manidipine Hydrochloride as directed in the potassium chlo-
ride disc method under Infrared Spectrophotometry <2.25>,
Magnesium Sulfate Injection and compare the spectrum with the Reference Spectrum or
the spectrum of Manidipine Hydrochloride Reference Stan-
硫酸マグネシウム注射液
dard: both spectra exhibit similar intensities of absorption at
the same wave numbers.
Add the following next to Extractable volume: (3) Add 10 mL of water to 0.1 g of Manidipine
Foreign insoluble matter <6.06> Perform the test according Hydrochloride, shake vigorously, and filter. Add 1 drop of
Supplement I, JP XV Official Monographs 1901

ammonia TS to 3 mL of the filtrate, allow to stand 5 Hydrochloride, previously dried, and dissolve in a mixture
minutes, and filter. The filtrate responds to the Qualitative of water and acetonitrile (1:1) to make exactly 50 mL. Pipet
Tests <1.09> (2) for chlorides. 5 mL of this solution, add exactly 5 mL of the internal stan-
dard solution, add the mixture of water and acetonitrile (1:1)
Purity (1) Heavy metals <1.07>— Proceed with 1.0 g of
to make 100 mL, and use this solution as the sample solu-
Manidipine Hydrochloride according to Method 2, and per-
tion. Separately, weigh accurately about 25 mg of Manidi-
form the test. Prepare the control solution with 1.0 mL of
pine Hydrochloride Reference Standard, previously dried,
Standard Lead Solution (not more than 10 ppm).
and dissolve in the mixture of water and acetonitrile (1:1) to
(2) Arsenic <1.11>—Prepare the test solution with 2.0 g
make exactly 50 mL. Pipet 20 mL of this solution, add ex-
of Manidipine Hydrochloride according to Method 4, and
actly 5 mL of the internal standard solution, add the mixture
perform the test (not more than 1 ppm).
of water and acetonitrile (1:1) to make 100 mL, and use this
(3) Related substances—Dissolve 20 mg of Manidipine
solution as the standard solution. Perform the test with 20
Hydrochloride in 200 mL of a mixture of water and acetoni-
mL each of the sample solution and standard solution as
trile (1:1), and use this solution as the sample solution. Pipet
directed under Liquid Chromatography <2.01> according to
1 mL of the sample solution, add the mixture of water and
the following conditions, and calculate the ratios, QT and
acetonitrile (1:1) to make exactly 100 mL, and use this solu-
QS, of the peak area of manidipine to that of the internal
tion as the standard solution. Perform the test with exactly
standard.
20 mL each of the sample solution and standard solution as
directed under Liquid Chromatography <2.01> according to Amount (mg) of C35H38N4O6.2HCl
the following conditions. Determine each peak area from = W S × ( Q T / Q S ) ×4
both solutions by the automatic integration method: the area
WS: Amount (mg) of Manidipine Hydrochloride Refer-
of the peaks other than manidipine obtained from the sam-
ence Standard
ple solution is not larger than 1/5 times the manidipine peak
area from the standard solution. Furthermore, the total of Internal standard solution—A solution of butyl benzoate in
the areas of all peaks other than the manidipine peak from acetonitrile (7 in 5000).
the sample solution is not larger than 7/10 times the peak Operating conditions—
area of manidipine from the standard solution. Detector: An ultraviolet absorption photometer (wave-
Operating conditions— length: 228 nm).
Detector, column, column temperature, mobile phase, Column: A stainless steel column 4.0 mm in inside di-
and flow rate: Proceed as directed in the operating condi- ameter and 15 cm in length, packed with octadecylsilanized
tions in the Assay. silica gel for liquid chromatography (5 mm in particle di-
Time span of measurement: About 3.5 times as long as the ameter).
retention time of manidipine, beginning after the solvent Column temperature: A constant temperature of about
peak. 259C.
System suitability— Mobile phase: Dissolve 13.6 g of potassium dihydrogen
Test for required detectability: Pipet 10 mL of the stan- phosphate in water to make 1000 mL, and adjust to pH 4.6
dard solution, add a mixture of water and acetonitrile (1:1) with diluted potassium hydroxide TS (1 in 10). To 490 mL of
to make exactly 100 mL. Confirm that the peak area of this solution add 510 mL of acetonitrile.
manidipine obtained from 20 mL of this solution is equiva- Flow rate: Adjust the flow rate so that the retention time
lent to 8 to 12z of that from 20 mL of the standard solution. of manidipine is about 10 minutes.
System performance: Dissolve 50 mg of Manidipine System suitability—
Hydrochloride in a mixture of water and acetonitrile (1:1) to System performance: When the procedure is run with 20
make 50 mL. To 10 mL of this solution add 5 mL of a solu- mL of the standard solution under the above operating con-
tion of butyl benzoate in acetonitrile (7 in 5000) and the mix- ditions, manidipine and the internal standard are eluted in
ture of water and acetonitrile (1:1) to make 100 mL. When this order with the resolution between these peaks being not
the procedure is run with 20 mL of this solution under the less than 5.
above operating conditions, manidipine and butyl benzoate System repeatability: When the test is repeated 6 times
are eluted in this order with the resolution between these with 20 mL of the standard solution under the above operat-
peaks being not less than 5. ing conditions, the relative standard deviation of the ratio of
System repeatability: When the test is repeated 6 times the peak area of manidipine to that of the internal standard
with 20 mL of the standard solution under the above operat- is not more than 1.0z.
ing conditions, the relative standard deviation of the peak
Containers and storage Containers—Tight containers.
area of manidipine is not more than 2.0z.
Storage—Light-resistant.
Loss on drying <2.41> Not more than 1.5z (1 g, 1059C, 4
hours).

Residue on ignition <2.44> Not more than 0.2z (1 g).

Assay Weigh accurately about 0.1 g of Manidipine


1902 Official Monographs Supplement I, JP XV

Add the following: in 45 minutes of Manidipine Hydrochloride Tablets is not


less than 75z.
Manidipine Hydrochloride Tablets Conduct this procedure using light-resistant vessels. Start
the test with 1 tablet of Manidipine Hydrochloride Tablets,
マニジピン塩酸塩錠 withdraw not less than 20 mL of the medium at the specified
minute after starting the test, and filter through a membrane
Manidipine Hydrochloride Tablets contain not less filter with a pore size not exceeding 0.45 mm. Discard the
than 92.0z and not more than 108.0z of the labeled first 10 mL of the filtrate, pipet V mL of the subsequent
amount of manidipine hydrochloride (C35H38N4O6. filtrate, and add the dissolution medium to make exactly V?
2HCl: 683.62). mL so that each mL contains about 5.6 mg of manidipine
hydrochloride (C35H38N4O6.2HCl) according to the labeled
Method of preparation Prepare as directed under Tablets,
amount. Pipet 2 mL of this solution, add exactly 2 mL of
with Manidipine Hydrochloride.
methanol, and use this solution as the sample solution.
Identification To a quantity of powdered Manidipine Separately, weigh accurately about 25 mg of Manidipine
Hydrochloride Tablets, equivalent to 10 mg of Manidipine Hydrochloride Reference Standard, previously dried, dis-
Hydrochloride according to the labeled amount, add 5 mL solve in a mixture of water and acetonitrile (1:1) to make ex-
of methanol, shake vigorously, centrifuge, and use the su- actly 50 mL. Pipet 1 mL of this solution, and add the disso-
pernatant liquid as the sample solution. Separately, dissolve lution medium to make exactly 100 mL. Pipet 2 mL of this
10 mg of Manidipine Hydrochloride Reference Standard in solution, add exactly 2 mL of methanol, and use this solu-
5 mL of methanol, and use this solution as the standard so- tion as the standard solution. Perform the test with exactly
lution. Perform the test with these solutions as directed un- 20 mL each of the sample solution and standard solution as
der Thin-layer Chromatography <2.03>. Spot 5 mL each of directed under Liquid Chromatography <2.01> according to
the sample solution and standard solution on a plate of silica the following conditions, and determine the manidipine
gel with fluorescent indicator for thin-layer chro- peak areas, AT and AS, of both solutions.
matography. Develop the plate with a mixture of ethyl
Dissolution rate (z) with respect to the labeled amount of
acetate and diethylamine (200:1) to a distance of about 10
manidipine hydrochloride (C35H38N4O6.2HCl)
cm, and air-dry the plate. Examine under ultraviolet light
=WS×(AT/AS)×(V?/V )×(1/C)×18
(main wavelength: 254 nm): the principal spot obtained
from the sample solution and the spot obtained from the WS: Amount (mg) of Manidipine Hydrochloride Refer-
standard solution show the same Rf value. ence Standard
C: Labeled amount (mg) of manidipine hydrochloride
Uniformity of dosage units <6.02> Perform the test accord-
(C35H38N4O6.2HCl) in 1 tablet
ing to the following method: it meets the requirement of the
Content uniformity test. Operating conditions—
Conduct this procedure using light-resistant vessels. To 1 Detector: An ultraviolet absorption photometer (wave-
tablet of Manidipine Hydrochloride Tablets, add exactly 1 length: 228 nm).
mL of the internal standard solution per 1 mg of manidipine Column: A stainless steel column 4.0 mm in inside di-
hydrochloride (C35H38N4O6.2HCl), disintegrate by adding a ameter and 15 cm in length, packed with octadecylsilanized
mixture of water and acetonitrile (1:1) to make V mL so that silica gel for liquid chromatography (5 mm in particle di-
each mL contains about 0.1 mg of manidipine hydrochloride ameter).
(C35H38N4O6.2HCl), shake vigorously for 10 minutes, and Column temperature: A constant temperature of about
filter through a membrane filter with a pore size not exceed- 259C.
ing 0.45 mm. Discard the first 1 mL of the filtrate, and use Mobile phase: A mixture of acetonitrile and a solution of
the subsequent filtrate as the sample solution. Then, proceed potassium dihydrogen phosphate (681 in 100,000) (3:2).
as directed in the Assay. Flow rate: Adjust the flow rate so that the retention time
of manidipine is about 6 minutes.
Amount (mg) of manidipine hydrochloride
System suitability—
(C35H38N4O6.2HCl)
System performance: When the procedure is run with 20
=WS×(QT/QS)×(V/250)
mL of the standard solution under the above operating con-
WS: Amount (mg) of Manidipine Hydrochloride Refer- ditions, the number of theoretical plates and the symmetry
ence Standard factor of the peak of manidipine are not less than 1500 and
not more than 1.5, respectively.
Internal standard solution—A solution of butyl benzoate in
System repeatability: When the test is repeated 6 times
acetonitrile (7 in 10,000).
with 20 mL of the standard solution under the above operat-
Dissolution <6.10> When the test is performed at 50 revolu- ing conditions, the relative standard deviation of the peak
tions per minute according to the Paddle method, using 900 area of manidipine is not more than 2.0z.
mL of 0.05 mol/L acetic acid-sodium acetate buffer solu-
Assay Conduct this procedure using light-resistant vessels.
tion, pH 4.0, as the dissolution medium, the dissolution rate
Weigh accurately not less than 20 Manidipine Hydrochloride
Supplement I, JP XV Official Monographs 1903

Tablets, and powder. Weigh accurately a portion of the water.


powder, equivalent to about 10 mg of manidipine It gradually turns yellow on exposure to light.
hydrochloride (C35H38N4O6.2HCl), add exactly 10 mL of the
internal standard solution, add a mixture of water and Change the Identification to read:
acetonitrile (1:1) to make 100 mL, shake vigorously for 10
Identification (1) Dissolve 10 mg of Medazepam in 3 mL
minutes, and filter through a membrane filter with a pore
of citric acid-acetic acid TS: a deep orange color develops.
size not exceeding 0.45 mm. Discard the first 1 mL of the
Heat in a water bath for 3 minutes: the color changes to dark
filtrate, and use the subsequent filtrate as the sample solu-
red.
tion. Separately, weigh accurately about 25 mg of Manidi-
(2) Determine the absorption spectrum of a solution of
pine Hydrochloride Reference Standard, previously dried,
Medazepam in methanol (1 in 100,000) as directed under
and dissolve in the mixture of water and acetonitrile (1:1) to
Ultraviolet-visible Spectrophotometry <2.24>, and compare
make 50 mL. Pipet 20 mL of this solution, add exactly 10
the spectrum with the Reference Spectrum: both spectra ex-
mL of the internal standard solution, add the mixture of
hibit similar intensities of absorption at the same wave-
water and acetonitrile (1:1) to make 100 mL, and use this so-
lengths.
lution as the standard solution. Then, proceed as directed in
(3) Determine the infrared absorption spectrum of
the Assay under Manidipine Hydrochloride.
Medazepam as directed in the potassium bromide disk
Amount (mg) of manidipine hydrochloride method under Infrared Spectrophotometry <2.25>, and com-
(C35H38N4O6.2HCl) pare the spectrum with the Reference Spectrum: both spec-
=WS×(QT/QS)×(2/5) tra exhibit similar intensities of absorption at the same wave
numbers.
WS: Amount (mg) of Manidipine Hydrochloride Refer-
(4) Perform the test with Medazepam as directed under
ence Standard
Flame Coloration Test <1.04> (2): a green color appears.
Internal standard solution—A solution of butyl benzoate in
acetonitrile (7 in 10,000).

Containers and storage Containers—Tight containers. Mefruside Tablets


Storage—Light-resistant.
メフルシド錠

Add the following next to Identification:


D-Mannitol Injection
Uniformity of dosage units <6.02> Perform the test accord-
D-マンニトール注射液 ing to the following method: it meets the requirement of the
Content uniformity test.
Add the following next to Extractable volume: To 1 tablet of Mefruside Tablets add 40 mL of methanol,
disintegrate the tablet using ultrasonic waves with occasional
Foreign insoluble matter <6.06> Perform the test according
stirring, then further treat with ultrasonic waves for 10
to Method 1: it meets the requirement.
minutes, and add methanol to make exactly V mL of a solu-
Insoluble particulate matter <6.07> It meets the require- tion containing about 0.5 mg of mefruside (C13H19ClN2O5S2)
ment. per mL. Centrifuge the solution, pipet 5 mL of the super-
natant liquid, add methanol to make exactly 20 mL, and use
Sterility <4.06> Perform the test according to the Mem-
this solution as the sample solution. Then, proceed as direct-
brane filtration method: it meets the requirement.
ed in the Assay.

Amount (mg) of mefruside (C13H19ClN2O5S2)


Medazepam =WS×(AT/AS)×(V/125)

WS: Amount (mg) of mefruside for assay


メダゼパム

Change the origin/limits of content to read:


Methyldopa Tablets
Medazepam, when dried, contains not less than 98.5
メチルドパ錠
z and not more than 101.0z of C16H15ClN2.
Add the following next to Identification:
Change the Description to read:
Uniformity of dosage units <6.02> Perform the test accord-
Description Medazepam occurs as white to light yellow
ing to the following method: it meets the requirement of the
crystals or crystalline powder.
Content uniformity test.
It is freely soluble in methanol, in ethanol (99.5), in acetic
To 1 tablet of Methyldopa Tablets add 50 mL of 0.05
acid (100) and in diethyl ether, and practically insoluble in
1904 Official Monographs Supplement I, JP XV

mol/L sulfuric acid TS, shake for 15 minutes, then add 0.05 equivalent to 0.1 g (potency) of Minocycline Hydrochloride
mol/L sulfuric acid TS to make exactly 100 mL, and filter. according to the labeled amount, dissolve in the mobile
Discard the first 20 mL of the filtrate, pipet V mL of the phase to make 100 mL. Pipet 25 mL of this solution, add the
subsequent filtrate equivalent to about 5 mg of methyldopa mobile phase to make exactly 50 mL, and use this solution as
(C10H13NO4), add exactly 5 mL of iron (II) tartrate TS, then the sample solution. Perform the test with 20 mL of the sam-
add ammonia-ammonium acetate buffer solution, pH 8.5, ple solution as directed under Liquid Chromatography
to make exactly 100 mL, and use this solution as the sample <2.01> according to the following conditions, and determine
solution. Separately, weigh accurately about 0.11 g of each peak area by the automatic integration method. Calcu-
Methyldopa Reference Standard (separately determine the late the amounts of each peak by the area percentage
loss on drying <2.41> at 1259C for 2 hours), and dissolve in method: the amount of epiminomycine, having the relative
0.05 mol/L sulfuric acid TS to make exactly 100 mL. Pipet 5 retention time of about 0.83 with respect to minocycline, is
mL of this solution, add exactly 5 mL of iron (II) tartrate not more than 6.0z.
TS, then add ammonia-ammonium acetate buffer solution, Operating conditions—
pH 8.5, to make exactly 100 mL, and use this solution as the Detector, column, column temperature, mobile phase and
standard solution. Determine the absorbances at 520 nm, AT flow rate: Proceed as directed in the operating conditions in
and AS, of the sample solution and standard solution as the Assay.
directed under Ultraviolet-visible Spectrophotometry <2.24>. Time span of measurement: About 2.5 times as long as the
retention time of minocycline, beginning after the solvent
Amount (mg) of methyldopa (C10H13NO4)
peak.
=WS×(AT/AS)×(5/V)
System suitability—
WS: Amount (mg) of Methyldopa Reference Standard, Test for required detectability: Pipet 2 mL of the standard
calculated on the dried basis solution obtained in the Assay, add the mobile phase to
make exactly 100 mL, and use this solution as the solution
for system suitability test. Pipet 5 mL of the solution for sys-
Add the following: tem suitability test, add the mobile phase to make exactly
100 mL. Confirm that the peak area of minocycline ob-
Minocycline Hydrochloride for tained from 20 mL of this solution is equivalent to 3.5 to 6.5
z of that from 20 mL of the solution for system suitability
Injection test.
注射用ミノサイクリン塩酸塩 System performance: Proceed as directed in the system
suitability in the Assay.
Minocycline Hydrochloride for Injection is a prepa- System repeatability: When the test is repeated 6 times
with 20 mL of the solution for system suitability test under
ration for injection which is dissolved before use.
It contains not less than 90.0z and not more than the above operating conditions, the relative standard devia-
110.0z of the labeled amount of minocycline tion of the peak area of minocycline is not more than 2.0z.
(C23H27N3O7: 457.48). Water <2.48> Weigh accurately the mass of the content of
Method of preparation Prepare as directed under Injec- one container of Minocycline Hydrochloride for Injection,
tions, with Minocycline Hydrochloride. dissolve in exactly 2 mL of methanol for water determina-
tion, and preform the test with exactly 1 mL of this solution
Description Minocycline Hydrochloride for Injection oc- as directed in the Volumetric tiration (back tiration): not
curs as a yellow to yellow-brown powder or flakes. more than 3.0z.
Identification Dissolve 4 mg of Minocycline Hydrochlo- Bacterial endotoxins <4.01> Less than 1.25 EU/mg (poten-
ride for Injection in 250 mL of a solution of hydrochloric cy).
acid in methanol (19 in 20,000). Determine the absorption
spectrum of this solution as directed under Ultraviolet-visi- Uniformity of dosage units <6.02> It meets the requirement
ble Spectrophotometry <2.24>: it exhibits maxima between of the Mass variation test.
221 nm and 225 nm, between 261 nm and 265 nm, and be- Foreign insoluble matter <6.06> Perform the test according
tween 354 nm and 358 nm. to Method 2: it meets the requirement.
pH <2.54> The pH of a solution, prepared by dissolving an Insoluble particulate matter <6.07> It meets the require-
amount of Minocycline Hydrochloride for Injection, ment.
equivalent to 0.1 g (potency) of Minocycline Hydrochloride
according to the labeled amount, in 10 mL of water is 2.0 to Sterility <4.06> Perform the test according to the Mem-
3.5. brane filtration method: it meets the requirement.

Purity Related substances—Conduct this procedure rapid- Assay Weigh accurately an amount of Minocycline
ly after the preparation of the sample solution. Take an Hydrochloride for Injection, equivalent to about 0.1 g
amount of Minocycline Hydrochloride for Injection, (potency) of Minocycline Hydrochloride, dissolve in the mo-
Supplement I, JP XV Official Monographs 1905

bile phase to make exactly 100 mL. Pipet 25 mL of this solu- Identification Dissolve an amount of Mitomycin C for In-
tion, add the mobile phase to make exactly 50 mL, and use jection, equivalent to 2 mg (potency) of Mitomycin C ac-
this solution as the sample solution. Separately, weigh ac- cording to the labeled amount, in 200 mL of water, and de-
curately an amount of Minocycline Hydrochloride Refer- termine the absorption spectrum of this solution as directed
ence Standard, equivalent to about 25 mg (potency), dis- under Ultraviolet-visible Spectrophotometry <2.24>: it ex-
solve in the mobile phase to make exactly 50 mL, and use hibits maxima between 216 nm and 220 nm, and between
this solution as the standard solution. Perform the test with 362 nm and 366 nm.
exactly 20 mL each of the sample solution and standard solu-
pH <2.54> The pH of a solution, prepared by dissolving
tion as directed under Liquid Chromatography <2.01> ac-
0.25 g of Mitomycin C for Injection in 20 mL of water, is 5.5
cording to the following conditions, and determine the peak
to 8.5.
area of minocycline, AT and AS, of each solution.
Loss on drying <2.41> Not more than 1.0z (0.4 g, in vacu-
Amount [mg (potency)] of minocycline (C23H27N3O7)
um not exceeding 0.67 kPa, phosphorus (V) oxide, 609 C, 3
=WS×(AT/AS)×4
hours).
WS: Amount [mg (potency)] of Minocycline Hydrochlo-
Bacterial endotoxins <4.01> Less than 10 EU/mg (poten-
ride Reference Standard
cy).
Operating conditions—
Uniformity of dosage units <6.02> Perform the test accord-
Detector, column, column temperature, and flow rate:
ing to the following method: it meets the requirement of the
Proceed as directed in the operating conditions in the Assay
Content uniformity test.
under Minocycline Hydrochloride.
To 1 container of Mitomycin C for Injection add exactly
Mobile phase: Adjust the pH to 6.5 of a mixture of am-
V mL of N,N-dimethylacetamide so that each mL contains
monium oxalate monohydrate solution (7 in 250), N,N-
about 0.5 mg (potency) of Mitomycin C, shake, centrifuge,
dimethylformamide and 0.1 mol/L disodium dihydrogen
and use the supernatant liquid as the sample solution.
ethylenediamine tetraacetate TS (11:5:4) with tetrabutylam-
Separately, weigh accurately about 25 mg (potency) of
monium hydroxide TS.
Mitomycin C Reference Standard, add N,N-dimethylaceta-
System suitability—
mide to make exactly 50 mL, and use this solution as the
System performance: Dissolve 50 mg of minocycline
standard solution. Then, proceed as directed in the Assay
hydrochloride in water to make 25 mL. Heat 5 mL of this
under Mitomycin C.
solution on a water bath for 60 minutes, then add water to
make 25 mL. When the procedure is run with 20 mL of this Amount [mg (potency)] of mitomycin C (C15H18N4O5)
solution under the above operating conditions, epiminocy- =WS×(AT/AS)×(V/50)
cline and minocycline are eluted in this order with the resolu-
WS: Amount [mg (potency)] of Mitomycin C Reference
tion between these peaks being not less than 2.5.
Standard
System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution under the above operat- Foreign insoluble matter <6.06> Perform the test according
ing conditions, the relative standard deviation of the peak to Method 2: it meets the requirement.
area of minocycline is not more than 2.0z.
Insoluble particulate matter <6.07> It meets the require-
Containers and storage Containers—Hermetic containers. ment.

Sterility <4.06> Perform the test according to the Mem-


brane filtration method: it meets the requirement.
Add the following:
Assay Weigh accurately the mass of the contents of not
Mitomycin C for Injection less than 10 containers of Mitomycin C for Injection. Weigh
accurately an amount of the contents, equivalent to about 10
注射用マイトマイシン C mg (potency) of Mitomycin C, add exactly 20 mL of N,N-
dimethylacetamide, shake, centrifuge, and use the super-
Mitomycin C for Injection is a preparation for injec- natant liquid as the sample solution. Separately, weigh ac-
tion, which is dissolved before use. curately an amount of Mitomycin C Reference Standard,
It contains not less than 90.0z and not more than equivalent to about 25 mg (potency), dissolve in N,N-
110.0z of the labeled amount of mitomycin C dimethylacetamide to make exactly 50 mL, and use this solu-
(C15H18N4O5: 334.33). tion as the standard solution. Then, proceed as directed in
the Assay under Mitomycin C.
Method of preparation Prepare as directed under Injec-
tions, with Mitomycin C. Amount [mg (potency)] of mitomycin C (C15H18N4O5)
=WS×(AT/AS)×(2/5)
Description Mitomycin C for Injection occurs as a blue-
purple powder. WS: Amount [mg (potency)] of Mitomycin C Reference
Standard
1906 Official Monographs Supplement I, JP XV

Containers and storage Containers—Hermetic containers. the sample solution are not larger than the mizoribine peak
area from the standard solution.
Add the following: Operating conditions—
Column, column temperature, mobile phase, and flow
Mizoribine rate: Proceed as directed in the operating conditions in the
Assay.
ミゾリビン Detector: An ultraviolet absorption photometer (wave-
length: 220 nm).
Time span of measurement: About 3 times as long as the
retention time of mizoribine, beginning after the solvent
peak.
System suitability—
Test for required detectability: Pipet 1 mL of the standard
solution, and add the mobile phase to make exactly 5 mL.
C9H13N3O6: 259.22 Confirm that the peak area of mizoribine obtained from 5
5-Hydroxy-1-b-D-ribofuranosyl-1H-imidazole-4- mL of this solution is equivalent to 14 to 26z of that from 5
carboxamide [50924-49-7] mL of the standard solution.
System performance: When the procedure is run with 5 mL
Mizoribine contains not less than 98.0z and not of the standard solution under the above operating condi-
more than 102.0z of C9H13N3O6, calculated on the tions, the number of theoretical plates and the symmetry
anhydrous basis. factor of the peak of mizoribine are not less than 10,000 and
not more than 1.4, respectively.
Description Mizoribine occurs as a white to yellowish System repeatability: When the test is repeated 6 times
white crystalline powder. with 5 mL of the standard solution under the above operat-
It is freely soluble in water, and practically insoluble in ing conditions, the relative standard deviation of the peak
methanol and in ethanol (99.5). area of mizoribine is not more than 2.0z.
Identification (1) Determine the absorption spectrum of Water <2.48> Not more than 0.5z (0.5 g, volumetric titra-
a solution of Mizoribine (1 in 100,000) as directed under tion, direct titration).
Ultraviolet-visible Spectrophotometry <2.24>, and compare
the spectrum with the Reference Spectrum or the spectrum Residue on ignition <2.44> Not more than 0.1z (1 g).
of a solution of Mizoribine Reference Standard prepared in Assay Weigh accurately about 0.1 g of Mizoribine, and
the same manner as the sample solution: both spectra exhibit dissolve in the mobile phase to make exactly 50 mL. Pipet 5
similar intensities of absorption at the same wavelengths. mL of this solution, add the mobile phase to make exactly 50
(2) Determine the infrared absorption spectrum of mL, and use this solution as the sample solution. Separately,
Mizoribine as directed in the potassium bromide disk weigh accurately about 10 mg of Mizoribine Reference Stan-
method under Infrared Spectrophotometry <2.25>, and com- dard (separately determine the water <2.48> using the same
pare the spectrum with the Reference Spectrum or the spec- manner as Mizoribine), dissolve in the mobile phase to make
trum of Mizoribine Reference Standard: both spectra exhibit exactly 50 mL, and use this solution as the standard solu-
similar intensities of absorption at the same wave numbers. tion. Perform the test with exactly 5 mL each of the sample
Optical rotation <2.49> [a]20
D : -25 – -279(0.5 g calculated
solution and standard solution as directed under Liquid
on the anhydrous basis, water, 25 mL, 100 mm). Chromatography <2.01> according to the following condi-
tions, and determine the peak areas of mizoribine, AT and
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of AS, of both solutions.
Mizoribine according to Method 1, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead Amount (mg) of C9H13N3O6
Solution (not more than 20 ppm). =WS×(AT/AS)×10
(2) Related substances—Dissolve 0.10 g of Mizoribine in WS: Amount (mg) of Mizoribine Reference Standard, cal-
the mobile phase to make 50 mL, and use this solution as the culated on the anhydrous basis
sample solution. Pipet 5 mL of the sample solution, and add
the mobile phase to make exactly 50 mL. Pipet 1 mL of this Operating conditions—
solution, add the mobile phase to make exactly 100 mL, and Detector: An ultraviolet absorption photometer (wave-
use this solution as the standard solution. Perform the test length: 279 nm).
with exactly 5 mL each of the sample solution and standard Column: A stainless steel column 4.6 mm in inside di-
solution as directed under Liquid Chromatography <2.01> ameter and 25 cm in length, packed with octadecylsilanized
according to the following conditions. Determine each peak silica gel for liquid chromatography (5 mm in particle di-
area of both solutions by the automatic integration method: ameter).
the areas of the peaks other than mizoribine obtained from Column temperature: A constant temperature of about
259C.
Supplement I, JP XV Official Monographs 1907

Mobile phase: Diluted phosphoric acid (1 in 1500). solution by the automatic integration method: the area of
Flow rate: Adjust the flow rate so that the retention time the peak, having the relative retention time of about 0.3 with
of mizoribine is about 9 minutes. respect to mizoribine, obtained from the sample solution is
System suitability— not larger than the peak area of mizoribine from the stan-
System performance: When the procedure is run with 5 mL dard solution, and the area of each peak other than mizori-
of the standard solution under the above operating condi- bine from the sample solution is not larger than 2/5 times
tions, the number of theoretical plates and the symmetry the peak area of mizoribine from the standard solution.
factor of the peak of mizoribine are not less than 10,000 and Operating conditions—
not more than 1.4, respectively. Column, column temperature, mobile phase, and flow
System repeatability: When the test is repeated 6 times rate: Proceed as directed in the operating conditions in the
with 5 mL of the standard solution under the above operat- Assay under Mizoribine.
ing conditions, the relative standard deviation of the peak Detector: An ultraviolet absorption photometer (wave-
area of mizoribine is not more than 1.0z. length: 220 nm).
Time span of measurement: About 3 times as long as the
Containers and storage Containers—Tight containers.
retention time of mizoribine, beginning after the solvent
peak.
System suitability—
Add the following:
Test for required detectability: To exactly 1 mL of the
standard solution add the mobile phase to make exactly 5
Mizoribine Tablets mL. Confirm that the peak area of mizoribine obtained
from 5 mL of this solution is equivalent to 14 to 26z of that
ミゾリビン錠
from 5 mL of the standard solution.
System performance: When the procedure is run with 5 mL
Mizoribine Tablets contain not less than 93.0z and
of the standard solution under the above operating condi-
not more than 107.0z of the labeled amount of
tions, the number of theoretical plates and the symmetry
mizoribine (C9H13N3O6: 259.22).
factor of the peak of mizoribine are not less than 10,000 and
Method of preparation Prepare as directed under Tablets, not more than 1.4, respectively.
with Mizoribine. System repeatability: When the test is repeated 6 times
with 5 mL of the standard solution under the above operat-
Identification To a quantity of powdered Mizoribine
ing conditions, the relative standard deviation of the peak
Tablets, equivalent to 0.1 g of Mizoribine according to the
area of mizoribine is not more than 2.0z.
labeled amount, add 5 mL of water, shake, filter, and use
the filtrate as the sample solution. Separately, dissolve 20 Uniformity of dosage units <6.02> Perform the test accord-
mg of Mizoribine Reference Standard in 1 mL of water, and ing to the following method: it meets the requirement of the
use this solution as the standard solution. Perform the test Content uniformity test.
with the sample solution and standard solution as directed To 1 tablet of Mizoribine Tablets add 50 mL of water,
under Thin-Layer Chromatography <2.03>. Spot 1 mL each shake until the tablet is disintegrated, and add water to make
of the sample solution and standard solution on a plate of exactly 100 mL. Filter the solution, discard not less than 10
silica gel for thin-layer chromatography. Then develop the mL of the first filtrate, pipet V mL of the subsequent
plate with a mixture of methanol, ammonia solution (28) filtrate, add water to make exactly V? mL so that each mL
and 1-propanol (2:1:1) to a distance of about 10 cm, and air- contains about 5 mg of mizoribine (C9H13N3O6), and use this
dry the plate. Allow the plate to stand in iodine vapor: the solution as the sample solution. Separately, weigh accurately
principal spot from the sample solution and the spot from about 25 mg of Mizoribine Reference Standard (separately
the standard solution show a red-brown color and the same determine the water <2.48> in the same manner as Mizori-
Rf value. bine), and dissolve in water to make exactly 100 mL. Pipet 2
mL of the solution, add water to make exactly 100 mL, and
Purity Related substances—To a quantity of powdered
use this solution as the standard solution. Determine the ab-
Mizoribine Tablets, equivalent to 0.10 g of Mizoribine ac-
sorbances, AT and AS, at 279 nm of the sample solution and
cording to the labeled amount, add 30 mL of the mobile
standad solution as directed under Ultraviolet-visible Spec-
phase, shake, then add the mobile phase to make 50 mL.
trophotometry <2.24>.
Filter the solution through a membrane filter with a pore size
not exceeding 0.5 mm and use the filtrate as the sample solu- Amount of mizoribine (C9H13N3O6)
tion. Pipet 2 mL of the sample solution, add the mobile =WS×(AT/AS)×(V?/V)×(1/50)
phase to make exactly 20 mL. Pipet 1 mL of the solution,
WS: Amount (mg) of Mizoribine Reference Standard, cal-
add the mobile phase to make exactly 20 mL, and use this so-
culated on the anhydrous basis
lution as the standard solution. Perform the test with exactly
5 mL each of the sample solution and standard solution as Dissolution <6.10> When the test is performed at 50 revolu-
directed under Liquid Chromatography <2.01> according to tions per minute according to the Paddle method, using 900
the following conditions. Determine each peak area of each mL of water as the dissolution medium, the dissolution rate
1908 Official Monographs Supplement I, JP XV

in 45 minutes of Mizoribine Tablets is not less than 80z. Content uniformity test.
Start the test with 1 tablet of Mizoribine Tablets, To 1 tablet of Morphine Hydrochloride Tablets add exact-
withdraw not less than 20 mL of the medium at the specified ly 1 mL of the internal standard solution per 2 mg of mor-
minute after starting the test, and filter through a membrane phine hydrochloride hydrate (C17H19NO3.HCl.3H2O), dis-
filter with a pore size not exceeding 0.5 mm. Discard not less perse the tablet into a small particles using ultrasonic waves,
than 10 mL of the first filtrate, pipet V mL of the subse- then treat with ultrasonic waves for 15 minutes with oc-
quent filtrate, add water to make exactly V? mL so that each casional stirring, and add water to make V mL so that each
mL contains about 14 mg of mizoribine (C9H13N3O6) accord- mL contains about 0.4 mg of morphine hydrochloride hy-
ing to the labeled amount, and use this solution as the sam- drate (C17H19NO3.HCl.3H2O). Filter the solution, and use
ple solution. Separately, weigh accurately about 28 mg of the filtrate as the sample solution. Then, proceed as directed
Mizoribine Reference Standard (separately determine the in the Assay.
water <2.48> in the same manner as Mizoribine), and dissolve
Amount (mg) of morphine hydrochloride hydrate
in water to make exactly 100 mL. Pipet 1 mL of this solu-
(C17H19NO3.HCl.3H2O)
tion, add water to make exactly 20 mL, and use this solution
=WS×(QT/QS)×(V/50)×1.1679
as the standard solution. Determine the absorbances, AT and
AS, at 279 nm of the sample solution and standard solution WS: Amount (mg) of morphine hydrochloride for assay,
as directed under Ultraviolet-visible Spectrophotometry calculated on the anhydrous basis
<2.24>.
Internal standard solution—A solution of etilefrine
Dissolution rate (z) with respect to the labeled amount of hydrochloride (1 in 500).
mizoribine (C9H13N3O6)
=WS×(AT/AS)×(V?/V)×(1/C)×45
Add the following:
WS: Amount (mg) of Mizoribine Reference Standard, cal-
culated on the anhydrous basis
C: Labeled amount (mg) of mizoribine (C9H13N3O6) in 1 Nabumetone
tablet
ナブメトン
Assay Weigh accurately not less than 20 Mizoribine
Tablets, and powder. Weigh accurately a portion of the
powder, equivalent to about 25 mg of mizoribine
(C9H13N3O6), add 50 mL of water and shake, then add water
to make exactly 100 mL. Filter the solution, discard not less
C15H16O2: 228.29
than 10 mL of the first filtrate, pipet 2 mL of the subsequent
4-(6-Methoxynaphthalen-2-yl)butan-2-one [42924-53-8]
filtrate, add water to make exactly 100 mL, and use this so-
lution as the sample solution. Separately, weigh accurately
Nabumetone contains not less than 98.0z and not
about 25 mg of Mizoribine Reference Standard (separately
more than 101.0z of C15H16O2, calculated on the an-
determine the water <2.48> in the same manner as Mizori-
hydrous basis.
bine), and dissolve in water to make exactly 100 mL. Pipet 2
mL of the solution, add water to make exactly 100 mL, and Description Nabumetone occurs as white to yellowish
use this solution as the standard solution. Determine the ab- white crystals or a crystalline powder.
sorbances, AT and AS, at 279 nm of the sample solution and It is soluble in acetonitrile, sparingly soluble in methanol
standard solution as directed under Ultraviolet-visible Spec- and in ethanol (99.5), and practically insoluble in water.
trophotometry <2.24>.
Identification (1) Determine the absorption spectrum of
Amount (mg) of mizoribine (C9H13N3O6)=WS×(AT/AS) a solution of Nabumetone in methanol (1 in 30,000) as
directed under Ultraviolet-visible Spectrophotometry <2.24>,
WS: Amount (mg) of Mizoribine Reference Standard, cal-
and compare the spectrum with the Reference Spectrum or
culated on the anhydrous basis
the spectrum of a solution of Nabumetone Reference Stan-
Containers and storage Containers—Tight containers. dard prepared in the same manner as the sample solution:
both spectra exhibit similar intensities of absorption at the
same wavelengths.
Morphine Hydrochloride Tablets (2) Determine the infrared absorption spectrum of
Nabumetone as directed in the potassium bromide disk
モルヒネ塩酸塩錠 method under Infrared Spectrophotometry <2.25>, and com-
pare the spectrum with the Reference Spectrum or the spec-
Add the following next to Identification: trum of Nabumetone Reference Standard: both spectra ex-
hibit similar intensities of absorption at the same wave num-
Uniformity of dosage units <6.02> Perform the test accord-
bers.
ing to the following method: it meets the requirement of the
Supplement I, JP XV Official Monographs 1909

Melting point <2.60> 79 – 849C with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
area of nabumetone is not more than 5.0z.
Nabumetone according to Method 2, and perform the test.
Prepare the control solution with 1.0 mL of Standard Lead Water <2.48> Not more than 0.2z (1 g, volumetric titra-
Solution (not more than 10 ppm). tion, direct titration).
(2) Related substances—Dissolve 20 mg of Nabumetone
Residue on ignition <2.44> Not more than 0.1z (1 g).
in 20 mL of acetonitrile, and use this solution as the sample
solution. Pipet 5 mL of the sample solution, add acetonitrile Assay Weigh accurately about 20 mg each of Nabumetone
to make exactly 50 mL. Pipet 1 mL of this solution, add and Nabumetone Reference Standard (separately determine
acetonitrile to make exactly 20 mL, and use this solution as the water <2.48> in the same manner as Nabumetone), dis-
the standard solution. Perform the test with exactly 10 mL solve them in acetonitrile to make exactly 20 mL, and use
each of the sample solution and standard solution as direct- these solutions as the sample solution and the standard solu-
ed under Liquid Chromatography <2.01> according to the tion, respectively. Perform the test with exactly 10 mL each
following conditions. Determine each peak area of both so- of the sample solution and standard solution as directed un-
lutions by the automatic integration method: the peak area der Liquid Chromatography <2.01> according to the follow-
of the related substance G obtained from the sample solu- ing conditions, and determine the peak area of nabumetone,
tion is not larger than 3/5 times the peak area of nabume- AT and AS, from each solution.
tone from the standard solution, and each peak area other
Amount (mg) of C15H16O2=WS×(AT/AS)
than nabumetone and the related substance G from the sam-
ple solution is not larger than 1/5 times the peak area of WS: Amount (mg) of Nabumetone Reference Standard,
nabumetone from the standard solution. Furthermore, the calculated on the anhydrous basis
total area of the peaks other than nabumetone from the sam-
Operating conditions—
ple solution is not larger than 1.6 times the peak area of
Detector: An ultraviolet absorption photometer (wave-
nabumetone from the standard solution. For these calcula-
length: 254 nm).
tions, use each peak area of the related substances A, B, C,
Column: A stainless steel column 4.6 mm in inside di-
D, E, F and G, which are having the relative retention time
ameter and 15 cm in length, packed with octadecylsilanized
of about 0.73, 0.85, 0.93, 1.2, 1.9, 2.6 and 2.7 with respect
silica gel for liquid chromatography (4 mm in particle di-
to nabumetone, after multiplying by their relative response
ameter).
factors, 0.12, 0.94, 0.25, 0.42, 1.02, 0.91 and 0.1, respective-
Column temperature: A constant temperature of about
ly.
409C.
Operating conditions—
Mobile phase: To 600 mL of a mixture of water and acetic
Detector, column, and column temperature: Proceed as
acid (100) (999:1) add 400 mL of a mixture of acetonitrile
directed in the operating conditions in the Assay.
and tetrahydrofuran (7:3).
Mobile phase A: A mixture of water and acetic acid (100)
Flow rate: Adjust the flow rate so that the retention time
(999:1).
of nabumetone is about 10 minutes.
Mobile phase B: A mixture of acetonitrile and tetra-
System suitability—
hydrofuran (7:3).
System performance: When the procedure is run with 10
Flowing of mobile phase: Control the gradient by mixing
mL of the standard solution under the above operating con-
the mobile phases A and B as directed in the following table.
ditions, the number of theoretical plates and the symmetry
Time after injection Mobile phase Mobile phase factor of the peak of nabumetone are not less than 6000 and
of sample (min) A (volz) B (volz) not more than 1.5, respectively.
System repeatability: When the test is repeated 6 times
0 – 12 60 40
with 10 mL of the standard solution under the above operat-
12 – 28 60ª 20 40ª 80
ing conditions, the relative standard deviation of the peak
Flow rate: 1.3 mL per minute. area of nabumetone is not more than 1.0z.
Time span of measurement: About 3 times as long as the
Containers and storage Containers—Tight containers.
retention time of nabumetone, beginning after the solvent
peak.
System suitability—
Test for required detectability: Pipet 2 mL of the standard
solution, and add acetonitrile to make exactly 10 mL. Con-
firm that the peak area of nabumetone obtained from 10 mL
of this solution is equivalent to 14 to 26z of that from 10 mL
of the standard solution.
System performance: Proceed as directed in the system
suitability in the Assay.
System repeatability: When the test is repeated 6 times
1910 Official Monographs Supplement I, JP XV

Add the following: C: Labeled amount (mg) of nabumetone (C15H16O2) in 1


tablet
Nabumetone Tablets Assay Weigh accurately not less than 20 tablets of
Nabumetone Tablets, and powder. Weigh accurately a por-
ナブメトン錠
tion of the powder, equivalent to about 0.2 g of nabumetone
(C15H16O2), add 10 mL of water and shake, add 40 mL of
Nabumetone Tablets contain not less than 95.0z
methanol, shake for 30 minutes, and then add methanol to
and not more than 105.0z of the labeled amount of
make exactly 100 mL. Centrifuge this solution, pipet 5 mL
nabumetone (C15H16O2: 228.29).
of the supernatant liquid, add exactly 5 mL of the internal
Method of preparation Prepare as directed under Tablets, standard solution, then add methanol to make 50 mL, and
with Nabumetone. use this solution as the sample solution. Separately, weigh
accurately about 40 mg of Nabumetone Reference Standard
Identification To a quantity of powdered Nabumetone
(separately determine the water <2.48> in the same manner as
Tablets, equivalent to 80 mg of Nabumetone according to
Nabumetone), dissolve by adding 50 mL of methanol and
the labeled amount, add 50 mL of methanol, shake for 10
exactly 20 mL of the internal standard solution, then add
minutes and centrifuge the solution. To 1 mL of the super-
methanol to make 200 mL, and use this solution as the stan-
natant liquid, add methanol to make 50 mL, and determine
dard solution. Perform the test with 10 mL each of the sam-
the absorption spectrum of this solution as directed under
ple solution and standard solution as directed under Liquid
Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
Chromatography <2.01> according to the following condi-
maxima between 259 nm and 263 nm, between 268 nm and
tions, and calculate the ratios, QT and QS, of the peak area
272 nm, between 316 nm and 320 nm, and between 330 nm
of nabumetone to that of the internal standard.
and 334 nm.
Amount (mg) of nabumetone (C15H16O2)=WS×(QT/QS)×5
Uniformity of dosage units <6.02> It meets the requirement
of the Mass variation test. WS: Amount (mg) of Nabumetone Reference Standard,
calculated on the anhydrous basis
Dissolution <6.10> When the test is performed at 75 revolu-
tions per minute according to the Paddle method, using 900 Internal standard solution—Dissolve 0.12 g of 2-ethylhexyl
mL of a solution of polysorbate 80 (dissolving 3 g of poly- parahydroxybenzoate in methanol to make 100 mL.
sorbate 80 in water to make 100 mL) as the dissolution medi- Operating conditions—
um, the dissolution rate in 60 minutes of Nabumetone Detector: An ultraviolet absorption photometer (wave-
Tablets is not less than 70z. length: 254 nm).
Start the test with 1 tablet of Nabumetone Tablets, Column: A stainless steel column 4 mm in inside diameter
withdraw not less than 20 mL of the medium at the specified and 15 cm in length, packed with octadecylsilanized silica gel
minute after starting the test, and filter through a membrane for liquid chromatography (5 mm in particle diameter).
filter with a pore size not exceeding 0.5 mm. Discard the first Column temperature: A constant temperature of about
10 mL of the filtrate, pipet V mL of the subsequent filtrate, 259C.
add a solution, prepared by adding to 20 mL of ethanol Mobile phase: A mixture of acetonitrile, water and acetic
(99.5) the dissolution medium to make 50 mL, to make ex- acid (100) (550:450:1).
actly V? mL so that each mL contains about 89 mg of Flow rate: Adjust the flow rate so that the retention time
nabumetone (C15H16O2) according to the labeled amount, of nabumetone is about 6 minutes.
and use this solution as the sample solution. Separately, System suitability—
weigh accurately about 22 mg of Nabumetone Reference System performance: When the procedure is run with 10
Standard (separately determine the water <2.48> in the same mL of the standard solution under the above operating con-
manner as Nabumetone), and dissolve in ethanol (99.5) to ditions, nabumetone and the internal standard are eluted in
make exactly 100 mL. Pipet 10 mL of this solution, add the this order with the resolution between these peaks being not
dissolution medium to make exactly 25 mL, and use this so- less than 13.
lution as the standard solution. Determine the absorbances, System repeatability: When the test is repeated 6 times
AT and AS, at 331 nm of the sample solution and standard with 10 mL of the standard solution under the above operat-
solution as directed under Ultraviolet-visible Spectrophoto- ing conditions, the relative standard deviation of the ratio of
metry <2.24>, using a solution prepared by adding to 20 mL the peak area of nabumetone to that of the internal standard
of ethanol (99.5) the dissolution medium to make 50 mL as is not more than 1.0z.
the blank.
Containers and storage Containers—Well-closed contain-
Dissolution rate (z) with respect to the labeled amount of ers.
nabumetone (C15H16O2)
=WS×(AT/AS)×(V?/V)×(1/C)×360

WS: Amount (mg) of Nabumetone Reference Standard,


calculated on the anhydrous basis
Supplement I, JP XV Official Monographs 1911

Add the following: and standard solution as directed under Liquid Chro-
matography <2.01> according to the following conditions.
Nafamostat Mesilate Determine each peak area of each solution by the automatic
integration method: the area of each peak other than
ナファモスタットメシル酸塩 nafamostat obtained from the sample solution is not larger
than 1/5 times the peak area of nafamostat from the stan-
dard solution. Furthermore, the total area of the peaks other
than nafamostat from the sample solution is not larger than
the peak area of nafamostat from the standard solution.
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
C19H17N5O2.2CH4O3S: 539.58 length: 260 nm).
6-Amidinonaphthalen-2-yl 4-guanidinobenzoate Column: A stainless steel column 4.6 mm in inside di-
bis(methanesulfonate) [82956-11-4] ameter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Nafamostat Mesilate, when dried, contains not less ameter).
than 99.0z and not more than 101.0z of Column temperature: A constant temperature of about
C19H17N5O2.2CH4O3S. 409C.
Mobile phase: Dissolve 6.07 g of sodium 1-heptane sul-
Description Nafamostat Mesilate occurs as a white crystal-
fonate in 1000 mL of diluted acetic acid (100) (3 in 500). To
line powder.
700 mL of this solution add 300 mL of acetonitrile.
It is freely soluble in formic acid, soluble in water, and
Flow rate: Adjust the flow rate so that the retention time
practically insoluble in ethanol (99.5).
of nafamostat is about 7 minutes.
It dissolves in 0.01 mol/L hydrochloric acid TS.
Time span of measurement: About 4 times as long as the
Melting point: about 2629 C (with decomposition).
retention time of nafamostat, beginning after the solvent
Identification (1) Determine the absorption spectrum of peak.
a solution of Nafamostat Mesilate in 0.01 mol/L System suitability—
hydrochloric acid TS (1 in 200,000) as directed under Ultrav- Test for required detectability: Pipet 5 mL of the standard
iolet-visible Spectrophotometry <2.24>, and compare the solution, and add the mobile phase to make exactly 50 mL.
spectrum with the Reference Spectrum: both spectra exhibit Pipet 15 mL of this solution, and add the mobile phase to
similar intensities of absorption at the same wavelengths. make exactly 100 mL. Confirm that the peak area of
(2) Determine the infrared absorption spectrum of nafamostat obtained from 10 mL of this solution is equiva-
Nafamostat Mesilate as directed in the potassium bromide lent to 1.1 to 1.9z of that from 10 mL of the standard solu-
disk method under Infrared Spectrophotometry <2.25>, and tion.
compare the spectrum with the Reference Spectrum: both System performance: Dissolve 0.1 g of nafamostat mesi-
spectra exhibit similar intensities of absorption at the same late in the mobile phase to make 100 mL. To 10 mL of this
wave numbers. solution add the mobile phase to make 100 mL. To 5 mL of
(3) A 0.1-g portion of Nafamostat Mesilate responds to this solution add 5 mL of a solution of 6-amidino-2-
the Qualitative Tests <1.09> (1) for mesilate. naphthol methanesulfonate in the mobile phase (1 in
20,000). When the procedure is run with 10 mL of this solu-
pH <2.54> The pH of a solution prepared by dissolving
tion under the above operating conditions, 6-amidino-2-
1.0 g of Nafamostat Mesilate in 50 mL of water is between
naphthol and nafamostat are eluted in this order with the
4.7 and 5.7.
resolution between these peaks being not less than 6.
Purity (1) Clarity and color of solution—A solution pre- System repeatability: When the test is repeated 6 times
pared by dissolving 1.0 g of Nafamostat Mesilate in 50 mL with 10 mL of the standard solution under the above operat-
of water is clear and colorless. ing conditions, the relative standard deviation of the peak
(2) Heavy metals <1.07>—Proceed with 2.0 g of area of nafamostat is not more than 2.0z.
Nafamostat Mesilate according to Method 4, and perform
Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 3
the test. Prepare the control solution with 2.0 mL of Stan-
hours).
dard Lead Solution (not more than 10 ppm).
(3) Related substances—Conduct this procedure using Residue on ignition <2.44> Not more than 0.1z (1 g).
light-resistant vessels. Dissolve 0.10 g of Nafamostat Mesi-
Assay Weigh accurately about 0.25 g of Nafamostat Mesi-
late in 100 mL of the mobile phase, and use this solution as
late, previously dried, dissolve in 4 mL of formic acid, add
the sample solution. Pipet 10 mL of the sample solution,
50 mL of acetic anhydride, and titrate <2.50> with 0.1 mol/L
add the mobile phase to make exactly 100 mL. Then pipet 5
perchloric acid VS (potentiometric titration). Perform a
mL of this solution, add the mobile phase to make exactly
blank determination in the same manner, and make any
100 mL, and use this solution as the standard solution. Per-
necessary correction.
form the test with exactly 10 mL each of the sample solution
1912 Official Monographs Supplement I, JP XV

Each mL of 0.1 mol/L perchloric acid VS


=26.98 mg of C19H17N5O2.2CH4O3S Nicotinic Acid Injection
Containers and storage Containers—Tight containers. ニコチン酸注射液

Add the following next to Identification:


Neostigmine Methylsulfate Bacterial endotoxins <4.01> Less than 3.0 EU/mg.
Injection
Add the following next to Extractable volume:
ネオスチグミンメチル硫酸塩注射液
Foreign insoluble matter <6.06> Perform the test according
to Method 1: it meets the requirement.
Add the following next to Extractable volume:
Insoluble particulate matter <6.07> It meets the require-
Foreign insoluble matter <6.06> Perform the test according
ment.
to Method 1: it meets the requirement.
Sterility <4.06> Perform the test according to the Mem-
Insoluble particulate matter <6.07> It meets the require-
brane filtration method: it meets the requirement.
ment.

Sterility <4.06> Perform the test according to the Mem-


brane filtration method: it meets the requirement. Add the following:

Nizatidine
Nicardipine Hydrochloride
ニザチジン
Injection
ニカルジピン塩酸塩注射液

Add the following next to Extractable volume:


Foreign insoluble matter <6.06> Perform the test according C12H21N5O2S2: 331.46
to Method 1: it meets the requirement. (1EZ)-N-{2-[({2-[(Dimethylamino)methyl]thiazol-
Insoluble particulate matter <6.07> It meets the require- 4-yl}methyl)sulfanyl]ethyl}-N?-methyl-2-nitroethene-
ment. 1,1-diamine [76963-41-2]

Sterility <4.06> Perform the test according to the Mem- Nizatidine, when dried, contains not less than 98.0
brane filtration method: it meets the requirement. z and not more than 101.0z of C12H21N5O2S2.
Description Nizatidine occurs as a white to pale yellowish
white crystalline powder, and has a characteristic odor.
Nicorandil It is soluble in methanol, sparingly soluble in water, and
ニコランジル slightly soluble in ethanol (99.5).

Identification (1) Determine the absorption spectrum of


Change the Identification to read: a solution of Nizatidine in methanol (1 in 100,000) as direct-
Identification (1) Determine the absorption spectrum of ed under Ultraviolet-visible Spectrophotometry <2.24>, and
a solution of Nicorandil (1 in 50,000) as directed under compare the spectrum with the Reference Spectrum or the
Ultraviolet-visible Spectrophotometry <2.24>, and compare spectrum of a solution of Nizatidine Reference Standard
the spectrum with the Reference Spectrum: both spectra prepared in the same manner as the sample solution: both
exhibit similar intensities of absorption at the same spectra exhibit similar intensities of absorption at the same
wavelengths. wavelengths.
(2) Determine the infrared absorption spectrum of (2) Determine the infrared absorption spectrum of
Nicorandil as directed in the potassium bromide disk Nizatidine, previously dried, as directed in the potassium
method under Infrared Spectrophotometry <2.25>, and com- bromide disk method under Infrared Spectrophotometry
pare the spectrum with the Reference Spectrum: both spec- <2.25>, and compare the spectrum with the Reference Spec-
tra exhibit similar intensities of absorption at the same wave trum or the spectrum of dried Nizatidine Reference Stan-
numbers. dard: both spectra exhibit similar intensities of absorption at
the same wave numbers.

Melting point <2.60> 130 – 1359


C (after drying).
Supplement I, JP XV Official Monographs 1913

Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of with 50 mL of the standard solution under the above operat-
Nizatidine according to Method 4, and perform the test us- ing conditions, the relative standard deviation of the peak
ing 3 mL of sulfuric acid. Prepare the control solution with area of nizatidine is not more than 2.0z.
2.0 mL of Standard Lead Solution (not more than 10 ppm).
Loss on drying <2.41> Not more than 0.5z (2 g, 1009
C, 1
(2) Related substances—Dissolve 50 mg of Nizatidine in
hour).
10 mL of a mixture of the mobile phase A and mobile phase
B (19:6), and use this solution as the sample solution. Pipet 3 Residue on ignition <2.44> Not more than 0.1z (1 g).
mL of the sample solution, add the mixture of the mobile
Assay Weigh accurately about 15 mg each of Nizatidine
phase A and mobile phase B (19:6) to make exactly 200 mL,
and Nizatidine Reference Standard, both previously dried,
and use this solution as the standard solution. Perform the
dissolve each in the mobile phase to make exactly 50 mL,
test with exactly 50 mL each of the sample solution and stan-
and use these solutions as the sample solution and the stan-
dard solution as directed under Liquid Chromatography
dard solution, respectively. Perform the test with exactly 10
<2.01> according to the following conditions. Determine
mL each of the sample solution and standard solution as
each peak area from both solutions by the automatic in-
directed under Liquid Chromatography <2.01> according to
tegration method: the area of the peaks other than nizatidine
the following conditions. Determine the peak area of nizati-
peak obtained from the sample solution is not larger than
dine, AT and AS, from each solution.
1/5 times the nizatidine peak area from the standard solu-
tion. Furthermore, the total of the areas of peaks other than Amount (mg) of C12H21N5O2S2=WS×(AT/AS)
the nizatidine peak from the sample solution is not larger
WS: Amount (mg) of Nizatidine Reference Standard
than the peak area of nizatidine from the standard solution.
Operating conditions— Operating conditions—
Detector: An ultraviolet absorption photometer (wave- Detector: An ultraviolet absorption photometer (wave-
length: 254 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 15 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
259C. 409C.
Mobile phase A: Dissolve 5.9 g of ammonium acetate in Mobile phase: Dissolve 5.9 g of ammonium acetate in 760
760 mL of water, add 1 mL of diethylamine, and adjust to mL of water, add 1 mL of diethylamine, and adjust to pH
pH 7.5 with acetic acid (100). 7.5 with acetic acid (100). To this solution add 240 mL of
Mobile phase B: Methanol. methanol.
Flowing of mobile phase: Control the gradient by mixing Flow rate: Adjust the flow rate so that the retention time
the mobile phases A and B as directed in the following table. of nizatidine is about 10 minutes.
System suitability—
Time after injection Mobile phase Mobile phase System performance: When the procedure is run with 10
of sample (min) A (volz) B (volz)
mL of the standard solution under the above operating con-
0–3 76 24 ditions, the number of theoretical plates and the symmetry
3 – 20 76ª 50 24ª 50 factor of the peak of nizatidine are not less than 5000 and
20 – 45 50 50
not more than 1.5, respectively.
Flow rate: 1.0 mL per minute. System repeatability: When the test is repeated 6 times
Time span of measurement: About 3 times as long as the with 10 mL of the standard solution under the above operat-
retention time of nizatidine, beginning after the solvent ing conditions, the relative standard deviation of the peak
peak. area of nizatidine is not more than 1.0z.
System suitability—
Containers and storage Containers—Tight containers.
Test for required detectability: Pipet 5 mL of the standard
solution, and add a mixture of the mobile phase A and mo-
bile phase B (19:6) to make exactly 25 mL. Confirm that the
Add the following:
peak area of nizatidine obtained from 50 mL of this solution
is equivalent to 15 to 25z of that from 50 mL of the standard
solution. Nizatidine Capsules
System performance: When the procedure is run with 50
ニザチジンカプセル
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
Nizatidine Capsules contain not less than 95.0z and
factor of the peak of nizatidine are not less than 20,000 and
not more than 105.0z of the labeled amount of nizati-
not more than 2.0, respectively.
dine (C12H21N5O2S2: 331.46).
System repeatability: When the test is repeated 6 times
1914 Official Monographs Supplement I, JP XV

Method of preparation Prepare as directed under Cap- Assay Take out the contents of not less than 10 Nizatidine
sules, with Nizatidine. Capsules, weigh accurately the mass of the contents, and
powder. Weigh accurately a portion of the powder, equiva-
Identification Take out the contents of Nizatidine Cap-
lent to about 0.15 g of nizatidine (C12H21N5O2S2), add exact-
sules, and powder. To a portion of the powder, equivalent to
ly 50 mL of the mobile phase, shake vigorously for 10
50 mg of Nizatidine according to the labeled amount, add 50
minutes, and centrifuge. Pipet 5 mL of the supernatant liq-
mL of methanol, shake well, and filter. Pipet 1 mL of the
uid, add exactly 5 mL of the internal standard solution, add
filtrate, and add methanol to make 100 mL. Determine the
the mobile phase to make 50 mL, and use this solution as the
absorption spectrum of the solution as directed under
sample solution. Separately, weigh accurately about 15 mg
Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
of Nizatidine Reference Standard, previously dried at 1009C
maxima between 239 nm and 244 nm, and between 323 nm
for 1 hour, dissolve in 30 mL of the mobile phase, add exact-
and 327 nm.
ly 5 mL of the internal standard solution, add the mobile
Uniformity of dosage units <6.02> Perform the test accord- phase to make 50 mL, and use this solution as the standard
ing to the following method: it meets the requirement of the solution. Perform the test with 10 mL each of the sample so-
Content uniformity test. lution and standard solution as directed under Liquid Chro-
Take out the contents from 1 capsule of Nizatidine Cap- matography <2.01> according to the following conditions,
sules, add exactly 50 mL of the mobile phase per 75 mg of and calculate the ratios, QT and QS, of the peak area of
Nizatidine, and shake vigorously for 10 minutes. Centrifuge nizatidine to that of the internal standard.
this solution, pipet V mL of the supernatant liquid, add ex-
Amount (mg) of nizatidine (C12H21N5O2S2)
actly 5 mL of the internal standard solution, and add the
=WS×(QT/QS)×10
mobile phase to make V? mL so that each mL contains about
0.3 mg of nizatidine (C12H21N5O2S2). Use this solution as the WS: Amount (mg) of Nizatidine Reference Standard
sample solution. Then, proceed as directed in the Assay.
Internal standard solution—A solution of phenol in the mo-
Amount (mg) of nizatidine (C12H21N5O2S2) bile phase (1 in 100).
=WS×(QT/QS)×(V?/V ) Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
WS: Amount (mg) of Nizatidine Reference Standard
length: 254 nm).
Internal standard solution—A solution of phenol in the mo- Column: A stainless steel column 4.6 mm in inside di-
bile phase (1 in 100). ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Dissolution <6.10> When the test is performed at 50 revolu-
ameter).
tions per minute according to the Paddle method, using a
Column temperature: A constant temperature of about
sinker, using 900 mL of water as the dissolution medium,
409C.
the dissolution rate in 15 minutes of Nizatidine Capsules is
Mobile phase: Dissolve 5.9 g of ammonium acetate in 760
not less than 80z.
mL of water, add 1 mL of diethylamine, and adjust to pH
Start the test with 1 capsule of Nizatidine Capsules,
7.5 with acetic acid (100). To this solution add 240 mL of
withdraw not less than 10 mL of the medium at the specified
methanol.
minute after starting the test, and filter through a membrane
Flow rate: Adjust the flow rate so that the retention time
filter with a pore size not exceeding 0.45 mm. Discard the
of nizatidine is about 10 minutes.
first 2 mL of the filtrate, pipet V mL of the subsequent
System suitability—
filtrate, and add water to make exactly V? mL so that each
System performance: When the procedure is run with 10
mL contains about 10 mg of nizatidine (C12H21N5O2S2) ac-
mL of the standard solution under the above operating con-
cording to the labeled amount. Use this solution as the sam-
ditions, the internal standard and nizatidine are eluted in this
ple solution. Separately, weigh accurately about 25 mg of
order with the resolution between these peaks being not less
Nizatidine Reference Standard, previously dried at 1009C
than 3.
for 1 hour, and dissolve in water to make exactly 100 mL.
System repeatability: When the test is repeated 6 times
Pipet 2 mL of this solution, add water to make exactly 50
with 10 mL of the standard solution under the above operat-
mL, and use this solution as the standard solution. Perform
ing conditions, the relative standard deviation of the ratio of
the test with the sample solution and standard solution as
the peak area of nizatidine to that of the internal standard is
directed under Ultraviolet-visible Spectrophotometry <2.24>,
not more than 1.0z.
and determine the absorbances, AT and AS, at 314 nm.
Containers and storage Containers—Tight containers.
Dissolution rate (z) with respect to the labeled amount of
nizatidine (C12H21N5O2S2)
=WS×(AT/AS)×(V?/V)×(1/C)×36

WS: Amount (mg) of Nizatidine Reference Standard


C: Labeled amount (mg) of nizatidine (C12H21N5O2S2) in 1
capsule
Supplement I, JP XV Official Monographs 1915

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


Noradrenaline Injection of Omeprazole in 25 mL of N,N-dimethylformamide: the
solution is clear and colorless or light yellow. Perform the
ノルアドレナリン注射液 test with this solution as directed under Ultraviolet-visible
Spectrophotometry <2.24>: the absorbance at 420 nm is not
Add the following next to Purity: more than 0.3.
(2) Heavy metals <1.07>—Proceed with 2.0 g of
Bacterial endotoxins <4.01> Less than 300 EU/mg.
Omeprazole according to Method 2, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead
Add the following next to Extractable volume:
Solution (not more than 10 ppm).
Foreign insoluble matter <6.06> Perform the test according (3) Related substances—Conduct the procedure soon af-
to Method 1: it meets the requirement. ter preparation of the sample solution. Dissolve 50 mg of
Omeprazole in 50 mL of the mobile phase, and use this solu-
Insoluble particulate matter <6.07> It meets the require-
tion as the sample solution. Perform the test with 10 mL of
ment.
the sample solution as directed under Liquid Chro-
Sterility <4.06> Perform the test according to the Mem- matography <2.01> according to the following conditions.
brane filtration method: it meets the requirement. Determine each of the peak areas of the sample solution by
the automatic integration method, and calculate the
amounts of them by the area percentage method: each of the
Add the following: amount other than omeprazole is not more than 0.1z, and
the total amount of the peaks other than omeprazole is not
Omeprazole more than 0.5z.
Operating conditions—
オメプラゾール Detector: An ultraviolet absorption photometer (wave-
length: 280 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octylsilanized silica
gel for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
C17H19N3O3S: 345.42 259C.
(RS)-5-Methoxy-2-{[(4-methoxy-3,5-dimethylpyridin- Mobile phase: Dissolve 2.83 g of disodium hydrogen
2-yl)methyl]sulfinyl}-1H-benzoimidazole [73590-58-6] phosphate dodecahydrate and 0.21 g of sodium dihydrogen
phosphate dihydrate in water to make 1000 mL. If necessa-
Omeprazole, when dried, contains not less than 99.0 ry, adjust the pH to 7.6 with diluted phosphoric acid (1 in
z and not more than 101.0z of C17H19N3O3S. 100). Add 11 volumes of acetonitrile to 29 volumes of this
solution.
Description Omeprazole occurs as a white to yellowish
Flow rate: Adjust the flow rate so that the retention time
white crystalline powder.
of omeprazole is about 8 minutes.
It is freely soluble in N,N-dimethylformamide, sparingly
Time span of measurement: About 10 times as long as the
soluble in ethanol (99.5), and practically insoluble in water.
retention time of omeprazole, beginning after the solvent
A solution of Omeprazole in N,N-dimethylformamide (1
peak.
in 25) shows no optical rotation.
System suitability—
It gradually turns yellowish white on exposure to light.
Test for required detectability: Pipet 5 mL of the sample
Melting point: about 1509 C (with decomposition).
solution, and add the mobile phase to make exactly 100 mL.
Identification (1) Add phosphate buffer solution, pH Pipet 5 mL of this solution, add the mobile phase to make
7.4, to 1 mL of a solution of Omeprazole in ethanol (99.5) (1 exactly 50 mL, and use this solution as the solution for sys-
in 1000) to make 50 mL. Determine the absorption spectrum tem suitability test. Pipet 5 mL of the solution, and add the
of this solution as directed under Ultraviolet-visible Spec- mobile phase to make exactly 25 mL. Confirm that the peak
trophotometry <2.24>, and compare the spectrum with the area of omeprazole obtained from 10 mL of this solution is
Reference Spectrum: both spectra exhibit similar intensities equivalent to 15 to 25z of that from 10 mL of the solution
of absorption at the same wavelengths. for system suitability test.
(2) Determine the infrared absorption spectrum of System performance: Dissolve 10 mg of Omeprazole and
Omeprazole as directed in the potassium bromide disk 25 mg of 1,2-dinitrobenzene in 5 mL of sodium borate solu-
method under Infrared Spectrophotometry <2.25>, and com- tion (19 in 5000) and 95 mL of ethanol (99.5). When the
pare the spectrum with the Reference Spectrum: both spec- procedure is run with 10 mL of this solution under the above
tra exhibit similar intensities of absorption at the same wave conditions, omeprazole and 1,2-dinitrobenzene are eluted in
numbers. this order with the resolution between these peaks being not
less than 10.
1916 Official Monographs Supplement I, JP XV

System repeatability: When the test is repeated 6 times the Qualitative Tests <1.09> for sodium salt.
with 10 mL of the solution for system suitability test under
pH <2.54> The pH of a solution prepared by dissolving 0.5 g
the above operating conditions, the relative standard devia-
of Ozagrel Sodium in 10 mL of water is between 9.5 and 10.5.
tion of the peak area of omeprazole is not more than 2.0z.
Purity (1) Clarity and color of solution—Dissolve 0.5 g
Loss on drying <2.41> Not more than 0.2z (1 g, in vacu-
of Ozagrel Sodium in 10 mL of water: the solution is clear
um, phosphorus (V) oxide, 509
C, 2 hours).
and colorless.
Residue on ignition <2.44> Not more than 0.1z (1 g). (2) Chloride <1.03>—Dissolve 2.0 g of Ozagrel Sodium
in 30 mL of water, add 1 mL of acetic acid (100) and water
Assay Weigh accurately about 0.4 g of Omeprazole, previ-
to make 50 mL, shake, and allow to stand for 30 minutes.
ously dried, dissolve in 70 mL of N,N-dimethylformamide,
Filter the solution, discard the first 5 mL of the filtrate, and
and titrate <2.50> with 0.1 mol/L tetramethylammonium
to 25 mL of the subsequent filtrate add 6 mL of dilute nitric
hydroxide VS (potentiometric titration). Separately, per-
acid and water to make 50 mL. Perform the test with this so-
form a blank determination using the same method on a so-
lution as the test solution. Prepare the control solution as
lution consisting of 70 mL of N,N-dimethylformamide and
follows: To 0.35 mL of 0.01 mol/L hydrochloric acid VS
12 mL of water, and make any necessary correction.
add 0.5 mL of acetic acid (100), 6 mL of dilute nitric acid
Each mL of 0.1 mol/L tetramethylammonium hydroxide VS and water to make 50 mL (not more than 0.012z).
=34.54 mg of C17H19N3O3S (3) Heavy metals <1.07>—Proceed with 2.0 g of Ozagrel
Sodium according to Method 2, and perform the test. Pre-
Containers and storage Containers—Tight containers.
pare the control solution with 2.0 mL of Standard Lead So-
Storage—Light-resistant, in a cold place.
lution (not more than 10 ppm).
(4) Related substances—Dissolve 50 mg of Ozagrel Sodi-
um in 100 mL of the mobile phase, and use this solution as
Add the following:
the sample solution. Perform the test with 5 mL of the sam-
ple solution as directed under Liquid Chromatography
Ozagrel Sodium <2.01> according to the following conditions. Determine
each peak area by the automatic integration method, and
オザグレルナトリウム
calculate the amount of them by the area percentage
method: each of the amount other than ozagrel is not more
than 0.2z, and the total amount other than ozagrel is not
more than 0.5z.
C13H11N2NaO2: 250.23 Operating conditions—
Monosodium (2E)-3-[4-(1H-imidazol- Column, column temperature, mobile phase, and flow rate:
1-ylmethyl)phenyl]prop-2-enoate [189224-26-8] Proceed as directed in the operating conditions in the Assay.
Detector: An ultraviolet absorption photometer (wave-
Ozagrel Sodium, when dried, contains not less than length: 220 nm).
98.0z and not more than 102.0z of C13H11N2NaO2. Time span of measurement: About 2 times as long as the
retention time of ozagrel, beginning after the solvent peak.
Description Ozagrel Sodium occurs as white crystals or
System suitability—
crystalline powder.
Test for required detectability: Pipet 1 mL of the sample
It is freely soluble in water, soluble in methanol, and prac-
solution, and add the mobile phase to make exactly 200 mL,
tically insoluble in ethanol (99.5).
and use this solution as the solution for system suitability
Identification (1) Determine the absorption spectrum of test. Pipet 2 mL of the solution for system suitability test,
a solution of Ozagrel Sodium (1 in 200,000) as directed un- and add the mobile phase to make exactly 10 mL. Confirm
der Ultraviolet-visible Spectrophotometry <2.24>, and com- that the peak area of ozagrel obtained from 5 mL of this so-
pare the spectrum with the Reference Spectrum or the spec- lution is equivalent to 15 to 25z of that from 5 mL of the so-
trum of a solution of Ozagrel Sodium Reference Standard lution for system suitability test.
prepared in the same manner as the sample solution: both System performance: When the procedure is run with 5 mL
spectra exhibit similar intensities of absorption at the same of the solution for system suitability test under the above
wavelengths. operating conditions, the number of theoretical plates and
(2) Determine the infrared absorption spectrum of the symmetry factor of the peak of ozagrel are not less than
Ozagrel Sodium as directed in the potassium bromide disk 6000 and not more than 2.0, respectively.
method under Infrared Spectrophotometry <2.25>, and com- System repeatability: When the test is repeated 6 times
pare the spectrum with the Reference Spectrum or the spec- with 5 mL of the solution for system suitability test under the
trum of Ozagrel Sodium Reference Standard: both spectra above operating conditions, the relative standard deviation
exhibit similar intensities of absorption at the same wave of the peak area of ozagrel is not more than 2.0z.
numbers.
Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 4
(3) A solution of Ozagrel Sodium (1 in 20) responds to
hours).
Supplement I, JP XV Official Monographs 1917

Assay Weigh accurately about 25 mg each of Ozagrel Sodi- Method of preparation Prepare as directed under Injec-
um and Ozagrel Sodium Reference Standard, both previous- tions, with Ozagrel Sodium.
ly dried, and dissolve each in methanol to make exactly 25
Description Ozagrel Sodium for Injection occurs as white
mL. Pipet 5 mL each of these solutions, add exactly 5 mL of
masses or powder.
the internal standard solution, and use these solutions as the
sample solution and the standard solution, respectively. Per- Identification Dissolve an amount of Ozagrel Sodium for
form the test with 1 mL each of the sample solution and stan- Injection, equivalent to 40 mg of Ozagrel Sodium according
dard solution as directed under Liquid Chromatography to the labeled amount, in water to make 40 mL. To 1 mL of
<2.01> according to the following conditions, and calculate this solution add water to make 200 mL, and determine the
the ratios, QT and QS, of the peak area of ozagrel to that of absorption spectrum of this solution as directed under
the internal standard. Ultraviolet-visible Spectrophotometry <2.24>: it exhibits a
maximum between 269 nm and 273 nm.
Amount (mg) of C13H11N2NaO2
= W S × ( Q T/ Q S ) pH Being specified separately.

WS: Amount (mg) of Ozagrel Sodium Reference Standard Purity Related substances—Dissolve an amount of
Ozagrel Sodium for Injection, equivalent to 0.20 g of
Internal standard solution—A solution of benzoic acid in
Ozagrel Sodium according to the labeled amount, in the
methanol (1 in 100).
mobile phase to make 100 mL. To 5 mL of this solution add
Operating conditions—
the mobile phase to make 20 mL, and use this solution as the
Detector: An ultraviolet absorption photometer (wave-
sample solution. Then, proceed as directed in the Purity (4)
length: 272 nm).
under Ozagrel Sodium.
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized Bacterial endotoxins <4.01> Less than 3.7 EU/mg.
silica gel for liquid chromatography (5 mm in particle di-
Uniformity of dosage units <6.02> It meets the require-
ameter).
ments of the Mass variation test.
Column temperature: A constant temperature of about
259C. Foreign insoluble matter <6.06> Perform the test according
Mobile phase: A mixture of a solution of ammonium to Method 2: it meets the requirement.
acetate (3 in 1000) and methanol (4:1).
Insoluble particulate matter <6.07> It meets the require-
Flow rate: Adjust the flow rate so that the retention time
ment.
of ozagrel is about 10 minutes.
System suitability— Sterility <4.06> Perform the test according to the Mem-
System performance: When the procedure is run with 1 mL brane filtration method: it meets the requirement.
of the standard solution under the above operating condi-
Assay Dissolve an amount of Ozagrel Sodium for
tions, the internal standard and ozagrel are eluted in this
Injection, equivalent to about 0.4 g of ozagrel sodium
order with the resolution between these peaks being not less
(C13H11N2NaO2), in water to make exactly 200 mL. Pipet 5
than 2.0, and the symmetry factor of the peak of ozagrel is
mL of this solution, add exactly 10 mL of the internal stan-
not more than 2.0.
dard solution and 5 mL of water, mix, and use this solution
System repeatability: When the test is repeated 6 times
as the sample solution. Separately, weigh accurately about
with 1 mL of the standard solution under the above operat-
25 mg of Ozagrel Sodium Reference Standard, and dissolve
ing conditions, the relative standard deviation of the ratio of
in methanol to make exactly 25 mL. Pipet 5 mL of this solu-
the peak area of ozagrel to that of the internal standard is
tion, add exactly 5 mL of the internal standard solution, and
not more than 1.0z.
use this solution as the standard solution. Then, proceed as
Containers and storage Containers—Tight containers. directed in the Assay under Ozagrel Sodium.
Storage—Light-resistant.
Amount (mg) of ozagrel sodium (C13H11N2NaO2)
=WS×(QT/QS)×16

Add the following: WS: Amount (mg) of Ozagrel Sodium Reference Standard

Internal standard solution—A solution of benzoic acid in


Ozagrel Sodium for Injection methanol (1 in 100).
注射用オザグレルナトリウム Containers and storage Containers—Hermetic containers.

Ozagrel Sodium for Injection is a preparation for in-


jection which is dissolved before use.
It contains not less than 95.0z and not more than
105.0z of the labeled amount of ozagrel sodium
(C13H11N2NaO2: 250.23).
1918 Official Monographs Supplement I, JP XV

Purity Clarity and color of solution—A solution prepared


Papaverine Hydrochloride Injection by dissolving an amount of Peplomycin Sulfate for Injec-
tion, equivalent to 10 mg (potency) of Peplomycin Sulfate
パパベリン塩酸塩注射液
according to the labeled amount, in 10 mL of water is clear
and colorless.
Add the following next to Identification:
Loss on drying <2.41> Not more than 4.0z (60 mg, in
Bacterial endotoxins <4.01> Less than 6.0 EU/mg.
vacuum, phosphorus (V) oxide, 609C, 3 hours). Perform the
sampling preventing from moisture absorption.
Add the following next to Extractable volume:
Bacterial endotoxins <4.01> Less than 1.5 EU/mg (poten-
Foreign insoluble matter <6.06> Perform the test according
cy).
to Method 1: it meets the requirement.
Uniformity of dosage units <6.02> It meets the requirement
Insoluble particulate matter <6.07> It meets the require-
of the Mass variation test.
ment.
Foreign insoluble matter <6.06> Perform the test according
Sterility <4.06> Perform the test according to the Mem-
to Method 2: it meets the requirement.
brane filtration method: it meets the requirement.
Insoluble particulate matter <6.07> It meets the require-
ment.
Add the following:
Sterility <4.06> Perform the test according to the Mem-
brane filtration method: it meets the requirement.
Peplomycin Sulfate for Injection
Assay Perform the test according to the Cylinder-plate
注射用ペプロマイシン硫酸塩 method as directed under Microbial Assay for Antibiotics
<4.02> according to the following conditions.
Peplomycin Sulfate for Injection is a preparation (i) Test organism, culture medium, liquid medium for
for injection which is dissolved before use. suspending test organisms, preparation of seeded agar layer,
It contains not less than 90.0z and not more than preparation of cylinder-agar plate and the standard solu-
115.0z of the labeled amount of peplomycin tions —Proceed as directed in the Assay under Peplomycin
(C61H88N18O21S2: 1473.59). Sulfate.
Method of preparation Prepare as directed under Injec- (ii) Sample solutions—Weigh accurately the mass of the
tions, with Peplomycin Sulfate. contents of not less than 10 containers of Peplomycin Sul-
fate for Injection. Weigh accurately an amount of the con-
Description Peplomycin Sulfate for Injection occurs as tents, equivalent to about 10 mg (potency) of Peplomycin
white light masses or powder. Sulfate, dissolve in 0.1 mol/L phosphate buffer solution,
Identification Take an amount of Peplomycin Sulfate for pH 6.8, to make exactly 100 mL. Measure exactly a suitable
Injection, equivalent to 10 mg (potency) of Peplomycin Sul- quantity of this solution, add 0.1 mol/L phosphate buffer
fate according to the labeled amount, and dissolve in 15 mL solution, pH 6.8 so that each mL contains 4 mg (potency)
of Copper (II) sulfate TS and water to make 2 mL. Apply and 2 mg (potency), and use these solutions as the high con-
this solution to the column (prepared by filling a 15 mm in- centration sample solution and the low concentration sample
side diameter and 15 cm long chromatography tube with 15 solution, respectively.
mL of strongly basic ion exchange resin (Cl type) for column Containers and storage Containers—Hermetic containers.
chromatography (75 – 150 mm in particle diameter) and run
off. Then wash the column using water at 2.5 mL per
minute, collect about 30 mL of the effluent. Add water to Pethidine Hydrochloride Injection
the effluent to make 250 mL, and determine the absorption
spectrum of this solution as directed under Ultraviolet-visi- ペチジン塩酸塩注射液
ble Spectrophotometry <2.24>: it exhibits maxima between
242 nm and 246 nm, and between 291 nm and 295 nm. Fur- Add the following next to Identification:
ther determine the absorbances A1 and A2, at 243 nm and
Bacterial endotoxins <4.01> Less than 6.0 EU/mg.
293 nm, respectively: the ratio A1/A2 is 1.20 to 1.30.

Osmotic pressure ratio Being specified separately. Add the following next to Extractable volume:
pH <2.54> The pH of a solution prepared by dissolving an Foreign insoluble matter <6.06> Perform the test according
amount of Peplomycin Sulfate for Injection, equivalent to to Method 1: it meets the requirement.
50 mg (potency) of Peplomycin Sulfate according to the
Insoluble particulate matter <6.07> It meets the require-
labeled amount, in 10 mL of water is 4.5 to 6.0.
ment.
Supplement I, JP XV Official Monographs 1919

Sterility <4.06> Perform the test according to the Mem- solution. Dissolve 20 mg of Piperacillin Hydrate in 20 mL of
brane filtration method: it meets the requirement. the mobile phase, and use this solution as the sample solu-
tion. Pipet 1 mL of the sample solution, add the mobile
phase to make exactly 200 mL, and use this solution as the
Add the following: standard solution (1). Pipet 2 mL of the standard solution
(1), add the mobile phase to make exactly 10 mL, and use
Piperacillin Hydrate this solution as the standard solution (2). Perform the test
with exactly 20 mL each of the sample solution and the stan-
ピペラシリン水和物 dard solutions (1) and (2) as directed under Liquid Chro-
matography <2.01> according to the following conditions,
and determine each peak area by the automatic integration
method: the total area of the peaks, having the relative
retention time of about 0.38 and about 0.50 with respect to
piperacillin, obtained from the sample solution is not larger
than twice the peak area of piperacillin from the standard
solution (2), the total area of the peaks, having the relative
C23H27N5O7S.H2O: 535.57
retention time of about 0.82 and about 0.86 with respect to
(2S,5R,6R)-6-{(2R)-2-[(4-Ethyl-2,3-dioxopiperazine-
piperacillin, obtained from the sample solution is not larger
1-carbonyl)amino]-2-phenylacetylamino}-3,3-dimethyl-
than the peak area of piperacillin from the standard solution
7-oxo-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid
(2), and the area of the peak other than piperacillin and
monohydrate [66258-76-2]
other than the peaks having the relative retention time of
about 0.38, about 0.50, about 0.82 and about 0.86 with
Piperacillin Hydrate contains not less than 970 mg
respect to piperacillin, obtained from the sample solution, is
(potency) and not more than 1020 mg (potency) per
not larger than the peak area of piperacillin from the stan-
mg, calculated on the anhydrous basis. The potency of
dard solution (2). Furthermore, the total area of the peaks
Piperacillin Hydrate is expressed as mass (potency) of
other than piperacillin obtained from the sample solution is
piperacillin (C23H27N5O7S: 517.55).
not larger than the peak area of piperacillin from the stan-
Description Piperacillin Hydrate occurs as a white crystal- dard solution (1).
line powder. Operating conditions—
It is freely soluble in methanol, soluble in ethanol (99.5) Detector, column, column temperature, mobile phase,
and in dimethylsulfoxide, and very slightly soluble in water. and flow rate: Proceed as directed in the operating condi-
tions in the Assay.
Identification (1) Determine the infrared absorption
Time span of measurement: About 3 times as long as the
spectrum of Piperacillin Hydrate as directed in the potassi-
retention time of piperacillin, beginning after the solvent
um bromide disk method under Infrared Spectrophotometry
peak.
<2.25>, and compare the spectrum with the Reference Spec-
System suitability—
trum or the spectrum of Piperacillin Reference Standard:
Test for required detectability: Confirm that the peak area
both spectra exhibit similar intensities of absorption at the
of piperacillin obtained from 20 mL of the standard solution
same wave numbers.
(2) is equivalent to 15 to 25z of that from 20 mL of the stan-
(2) Determine the spectrum of a solution of Piperacillin
dard solution (1).
Hydrate in deuterated dimethylsulfoxide for nuclear mag-
System performance: When the procedure is run with 20
netic resonance spectroscopy (1 in 3) as directed under
mL of the standard solution (1) under the above operating
Nuclear Magnetic Resonance Spectroscopy <2.21> (1H), us-
conditions, the number of theoretical plates and the symmet-
ing tetramethylsilane for nuclear magnetic resonance spec-
ry factor of the peak of piperacillin are not less than 3000
troscopy as an internal reference compound: it exhibits a tri-
and not more than 1.5, respectively.
ple signal A at about d 1.1 ppm, a single signal B at about d
System repeatability: When the test is repeated 6 times
4.2 ppm, and a multiple signal C at about d 7.4 ppm, and the
with 20 mL of the standard solution (2) under the above
ratio of the integrated intensity of each signal, A:B:C, is
operating conditions, the relative standard deviation of the
about 3:1:5.
peak area of piperacillin is not more than 3.0z.
Optical rotation <2.49> D :+162 – +1729(0.2 g, metha-
[a]20 (3) Related substances 2—Dissolve 20 mg of Piperacillin
nol, 20 mL, 100 mm). Hydrate in 20 mL of the mobile phase, and use this solution
as the sample solution. Pipet 1 mL of the sample solution,
Purity (1) Heavy metal <1.07>—Proceed with 2.0 g of
add the mobile phase to make exactly 200 mL, and use this
Piperacillin Hydrate according to Method 2, and perform
solution as the standard solution (1). Pipet 2 mL of the stan-
the test. Prepare the control solution with 2.0 mL of Stan-
dard solution (1), add the mobile phase to make exactly 10
dard Lead Solution (not more than 10 ppm).
mL, and use this solution as the standard solution (2). Per-
(2) Related substances 1—Conduct this procedure rapid-
form the test with exactly 20 mL each of the sample solution,
ly after the preparation of the sample solution and standard
and the standard solutions (1) and (2) as directed under Liq-
1920 Official Monographs Supplement I, JP XV

uid Chromatography <2.01> according to the following con- acetate by the automatic integration method: the peak area
ditions, and determine each peak area by the automatic in- of ethyl acetate obtained from the sample gas is not larger
tegration method: the area of the peak, having the relative than that from the standard gas.
retention time of about 6.6 with respect to piperacillin, ob- Operating conditions—
tained from the sample solution is not larger than three times Detector: A hydrogen flame-ionization detector.
the peak area of piperacillin from the standard solution (2), Column: A glass column 3 mm in inside diameter and 1 m
and the area of the peaks other than the peak of piperacillin in length, packed with porous stylene-divinyl benzene
and the peak having the relative retention time of about 6.6 copolymer for gas chromatography (average pore diameter
with respect to piperacillin from the sample solution are not of 0.0085 mm, 300 – 400 m2/g) with the particle size of 125 to
larger than 1.4 times the peak area of piperacillin from the 150 mm.
standard solution (2). Furthermore, the total area of the Column temperature: A constant temperature of about
peaks other than the peak of piperacillin from the sample so- 1459 C.
lution is not larger than the area of the peak of piperacillin Carrier gas: Nitrogen
from the standard solution (1). For these calculations, use Flow rate: Adjust the flow rate so that the retention time
the area of the peak, having the relative retention time of of ethyl acetate is about 4 minutes.
about 6.6 with respect to piperacillin, after multiplying by System suitability—
the relative response factor, 2.0. System performance: Take 1 mL of saturated sodium
Operating conditions— hydrogen carbonate solution in an about 3 mL-vial, add 2
Detector, column and column temperature: Proceed as mL each of ethyl acetate solution (1 in 400) and acetone solu-
directed in the operating conditions in the Assay. tion (1 in 400), and stop the vial tightly. When the procedure
Mobile phase: Take 60.1 g of acetic acid (100) and 101.0 g is run under the above operating conditions, acetone and
of triethylamine, add water to make 1000 mL. To 25 mL of ethyl acetate are eluted in this order with the resolution be-
this solution add 300 mL of acetonitrile and 25 mL of dilute tween these peaks being not less than 2.0.
acetic acid, and add water to make 1000 mL. System repeatability: Take 1 mL of saturated sodium
Flow rate: Adjust the flow rate so that the retention time hydrogen carbonate solution in an about 3 mL-vial, add 2
of piperacillin is about 1.2 minutes. mL of ethyl acetate solution (1 in 400), stop the vial tightly,
Time span of measurement: About 8 times as long as the and perform the test under the above operating conditions.
retention time of piperacillin, beginning after the piperacillin When the procedure is repeated 6 times, the relative stan-
peak. dard deviation of the peak area of ethyl acetate is not more
System suitability— than 10z.
Test for required detectability: Confirm that the peak area
Water <2.48> Not less than 3.2z and not more than 3.8z
of piperacillin obtained from 20 mL of the standard solution
(0.5 g, volumetric titration, direct titration).
(2) is equivalent to 15 to 25z of that from 20 mL of the stan-
dard solution (1). Residue on ignition <2.44> Not more than 0.1z (1 g).
System performance: When the procedure is run with 20
Bacterial endotoxins <4.01> Less than 0.07 EU/mg (poten-
mL of the standard solution (1) under the above operating
cy).
conditions, the number of theoretical plates and the symmet-
ry factor of the peak of piperacillin are not less than 1500 Assay Weigh accurately an amount of Piperacillin Hydrate
and not more than 2.0, respectively. and Piperacillin Reference Standard, equivalent to about 50
System repeatability: When the test is repeated 6 times mg (potency), dissolve each in the mobile phase to make 50
with 20 mL of the standard solution (2) under the above mL. Pipet 5 mL each of these solutions, add exactly 5 mL of
operating conditions, the relative standard deviation of the the internal standard solution, and use these solutions as the
peak area of piperacillin is not more than 4.0z. sample solution and standard solution, respectively. Per-
(4) Residual solvents <2.46>—Transfer exactly 10 mg of form the test with 5 mL each of the sample solution and stan-
Piperacillin Hydrate to an about 3 mL-vial, add exactly 1 dard solution as directed under Liquid Chromatography
mL of saturated sodium hydrogen carbonate solution to dis- <2.01> according to the following conditions, and calculate
solve and stop the vial tightly. After heating this at 909C for the ratios, QT and QS, of the peak height of piperacillin to
10 minutes, use the gas inside the container as the sample that of the internal standard.
gas. Separately, measure exactly 1 mL of ethyl acetate, dis-
Amount [ mg (potency)] of piperacillin (C23H27N5O7S)
solve in water to make exactly 200 mL. Pipet 10 mL of this
=WS×(QT/QS)×1000
solution, add water to make exactly 20 mL. Pipet 2 mL of
this solution in an about 3-mL vial containing exactly 1 mL WS: Amount [mg (potency)] of Piperacillin Reference
of saturated sodium hydrogen carbonate solution, and stop Standard
the vial tightly. Run the procedure similarly to the sample,
Internal standard solution—A solution of acetanilide in the
and use the gas as the standard gas. Perform the test with ex-
mobile phase (1 in 5000).
actly 0.5 mL each of the sample gas and standard gas as
Operating conditions—
directed under Gas Chromatography <2.02> according to the
Detector: An ultraviolet absorption photometer (wave-
following conditions, and determine the peak area of ethyl
Supplement I, JP XV Official Monographs 1921

length: 254 nm). It binds with not less than 100 Units of heparin per
Column: A stainless steel column 4 mm in inside diameter mg, calculated on the dried basis.
and 15 cm in length, packed with octadecylsilanized silica gel
for liquid chromatography (5 mm in particle diameter). Change the Description to read:
Column temperature: A constant temperature of about
Description Protamine Sulfate occurs as a white powder.
259C.
It is sparingly soluble in water.
Mobile phase: Take 60.1 g of acetic acid (100) and 101.0 g
of triethylamine, add water to make 1000 mL. To 25 mL of
Add the following next to Identification:
this solution add 210 mL of acetonitrile and 25 mL of dilute
acetic acid, and add water to make 1000 mL. pH <2.54> Dissolve 1.0 g of Protamine Sulfate in 100 mL
Flow rate: Adjust the flow rate so that the retention time of water: the pH of this solution is between 6.5 and 7.5.
of piperacillin is about 5 minutes.
System suitability— Change the Purity (2) to read
System performance: When the procedure is run with 5 mL
Purity (2) Absorbance—Dissolve 0.10 g of Protamine
of the standard solution under the above operating condi-
Sulfate in 10 mL of water, and determine the absorption
tions, the internal standard and piperacillin are eluted in this
spectrum as directed under Ultraviolet-visible Spectrophoto-
order with the resolution between these peaks being not less
metry <2.24>: the absorbance between 260 nm and 280 nm is
than 3.
not more than 0.1.
System repeatability: When the test is repeated 6 times
with 5 mL of the standard solution under the above operat-
Delete the Potency as antiheparin, and add the
ing conditions, the relative standard deviation of the ratio of
following next to Purity:
the peak height of piperacillin to that of the internal stan-
dard is not more than 1.0z. Loss on drying <2.41> Not more than 5.0z (1 g, 1059
C, 3
hours).
Containers and storage Containers—Tight containers.
Nitrogen content Weigh accurately about 10 mg of Prota-
mine Sulfate, and perform the test as directed under Nitro-
Prednisolone Sodium Succinate for gen Determination <1.08>: the amount of nitrogen (N:14.01)
is 22.5 – 25.5z, calculated on the dried basis.
Injection
Heparin-binding capacity
注射用プレドニゾロンコハク酸エステルナトリウム (i) Sample solution (a)—Weigh accurately about 15 mg
of Protamine Sulfate, and dissolve in water to make exactly
Add the following next to Loss on drying: 100 mL. Repeat this procedure 3 times, and use the solutions
Bacterial endotoxins <4.01> Less than 2.4 EU/mg of pred- so obtained as the sample solutions (a1), (a2) and (a3).
nisolone (C21H28O5). (ii) Sample solution (b)—Pipet 10 mL each of the sam-
ple solutions (a1), (a2) and (a3), add exactly 5 mL of water to
Uniformity of dosage units <6.02> It meets the requirement them, and use these solutions as the sample solutions (b1),
of the Mass variation test. (b2) and (b3).
Foreign insoluble matter <6.06> Perform the test according (iii) Sample solution (c)—Pipet 10 mL each of the sam-
to Method 2: it meets the requirement. ple solutions (a1), (a2) and (a3), add exactly 20 mL of water
to them, and use these solutions as the sample solutions (c1),
Insoluble particulate matter <6.07> It meets the require- (c2) and (c3).
ment. (iv) Standard solution—Dissolve Heparin Sodium
Sterility <4.06> Perform the test according to the Mem- Reference Standard in water to make a solution containing
brane filtration method: it meets the requirement. exactly about 20 Units per mL.
(v) Procedure—Transfer exactly 2 mL of the sample so-
lution to a cell for spectrophotometer, add the standard so-
lution dropwise while mixing, and determine the transmit-
Protamine Sulfate tance at 500 nm as directed under Ultraviolet-visible Spec-
プロタミン硫酸塩 trophotometry <2.24>. Continue the addition until a sharp
change in the transmittance is observed, and note the
Change the origin/limits of content to read: volume, V mL, of the standard solution added. Repeat this
procedure 2 times for each sample solution.
Protamine Sulfate is the sulfate of protamine pre- (vi) Calculation—Calculate the amount of heparin
pared from the mature spermary of fish belonging to bound with 1 mg of the sample by the following formula
the family Salmonidae. from the volume of titrant on each sample solution, and cal-
It has a property to bind with heparin. culate the average of 18 results obtained. The assay is not
valid unless each relative standard deviation of 6 results ob-
1922 Official Monographs Supplement I, JP XV

tained from the sample solution (a), sample solution (b) and Sterility <4.06> Perform the test according to the Mem-
sample solution (c) is not more than 5z, respectively, and brane filtration method: it meets the requirement.
also unless each relative standard deviation of 6 results ob-
tained from 3 sets, (a1, b1, c1), (a2, b2, c2) and (a3, b3, c3) is
not more than 5z, respectively. Reserpine Injection
Amount (heparin Unit) of heparin bound to 1 mg of Prota-
レセルピン注射液
mine Sulfate
=S×V×(50/WT)×d
Add the following next to Extractable volume:
S: Amount (heparin Unit) of heparin sodium in 1 mL of
Foreign insoluble matter <6.06> Perform the test according
the standard solution
to Method 1: it meets the requirement.
WT: Amount (mg) of the sample, calculated on the dried
basis Insoluble particulate matter <6.07> It meets the require-
d: Dilution factor for each sample solution from the sam- ment.
ple solution (a)
Sterility <4.06> Perform the test according to the Mem-
Sulfate content Weigh accurately about 0.15 g of Prota- brane filtration method: it meets the requirement.
mine Sulfate, dissolve in 75 mL of water, add 5 mL of 3
mol/L hydrochloric acid TS, and heat to boil. Add gradual-
ly 10 mL of barium chloride TS while boiling, and allow to Riboflavin Sodium Phosphate In-
stand for 1 hour while heating. Filter the precipitate formed,
wash the precipitate with warm water several times, and
jection
transfer the precipitate into a tared crucible. Dry the リボフラビンリン酸エステルナトリウム注射液
precipitate, and incinerate by ignition to constant mass: the
amount of sulfate (SO4) is 16 – 22z, calculated on the dried Add the following next to Identification:
basis, where 1 g of the residue is equivalent to 0.4117 g of
SO4. Bacterial endotoxins <4.01> Less than 10 EU/mg.

Add the following next to Extractable volume:


Protamine Sulfate Injection Foreign insoluble matter <6.06> Perform the test according
to Method 1: it meets the requirement.
プロタミン硫酸塩注射液
Insoluble particulate matter <6.07> It meets the require-
Delete the Potency as antiheparin, and add the ment.
following next to Extractable volume: Sterility <4.06> Perform the test according to the Membrane
Heparin-binding capacity Proceed the test as directed in filtration method: it meets the requirement.
the Heparin-binding capacity under Protamine Sulfate,
changing the sample solution (a) as below: it binds with not
less than 100 Units of heparin per mg of the labeled amount. Ringer's Solution
(i) Sample solution (a)—Pipet as amount of Protamine
Sulfate Injection, equivalent to 15.0 mg of Protamine Sul- リンゲル液
fate according to the labeled amount, add water to make ex-
actly 100 mL. Repeat this procedure more 2 times, and use Add the following next to Extractable volume:
the solutions so obtained as the sample solutions (a1), (a2) Foreign insoluble matter <6.06> Perform the test according
and (a3). to Method 1: it meets the requirement.

Insoluble particulate matter <6.07> It meets the require-


Pyridoxine Hydrochloride Injection ment.

Sterility <4.06> Perform the test according to the Mem-


ピリドキシン塩酸塩注射液 brane filtration method: it meets the requirement.

Add the following next to Extractable volume:


Foreign insoluble matter <6.06> Perform the test according
to Method 1: it meets the requirement.

Insoluble particulate matter <6.07> It meets the require-


ment.
Supplement I, JP XV Official Monographs 1923

Add the following: Rokitamycin Reference Standard, dissolve in 10 mL of


methanol, and add water to make exactly 100 mL. Pipet 5
Rokitamycin Tablets mL of the solution, add water to make exactly 50 mL, and
use this solution as the standard solution. Determine the ab-
ロキタマイシン錠 sorbances, AT and AS, at 232 nm of the sample solution and
standard solution using water as the blank, as directed under
Rokitamycin Tablets contain not less than 90.0z Ultraviolet-visible Spectrophotometry <2.24>.
and not more than 110.0z of the labeled amount of
rokitamycin (C42H69NO15: 827.99). Dissolution rate (z) with respect to the labeled amount of
rokitamycin (C42H69NO15)
Method of preparation Prepare as directed under Tablets, =WS×(AT/AS)×(V?/V)×(1/C)×90
with Rokitamycin.
WS: Amount [mg (potency)] of Rokitamycin Reference
Identification Take an amount of powdered Rokitamycin Standard
Tablets, equivalent to 10 mg (potency) of Rokitamycin ac- C: Labeled amount [mg (potency)] of rokitamycin
cording to the labeled amount, add 20 mL of methanol, and (C42H69NO15) in 1 tablet
centrifuge if necessary. To 1 mL of this solution add
methanol to make 25 mL, and determine the absorption Assay Perform the test according to the Cylinder-plate
spectrum as directed under Ultraviolet-visible Spectrophoto- method as directed under Microbial Assay for Antibiotics
metry <2.24>: it exhibits a maximum between 230 nm and <4.02> under the following conditions.
233 nm. (i) Test organism, culture medium and standard solu-
tions—Proceed as directed in the Assay under Rokitamycin.
Uniformity of dosage units <6.02> Perform the test accord- (ii) Sample solutions—Weigh accurately not less than 20
ing to the following method: it meets the requirement of the tablets of Rokitamycin Tablets, and powder. Weigh ac-
Content uniformity test. curately an amount of contents, equivalent to about 40 mg
Add 50 mL of water to 1 tablet of Rokitamycin Tablets, (potency) of Rokitamycin, add 50 mL of methanol, shake
and disintegrate. Then add 10 mL of methanol, shake well, vigorously, then add 0.1 mol/L phosphate buffer solution,
and add water to make exactly 100 mL. Centrifuge this solu- pH 4.5 to make exactly 100 mL, and centrifuge if necessary.
tion if necessary, filter through a membrane filter with a Measure exactly a suitable quantity of this solution, add
pore size not exceeding 0.5 mm. Discard 5 mL of the first polysorbate 80 solution, prepared by adding 0.1 mol/L
filtrate, pipet V mL of the subsequent filtrate, add water to phosphate buffer solution, pH 8.0 to 0.1 g of polysorbate 80
make exactly V? mL so that each mL contains about 20 mg to make 1000 mL, so that each mL contains 2 mg (potency)
(potency) of Rokitamycin, and use this solution as the sam- and 0.5 mg (potency), and use these solutions as the high con-
ple solution. Separately, weigh accurately about 20 mg centration sample solution and the low concentration sample
(potency) of Rokitamycin Reference Standard, dissolve in 10 solution, respectively.
mL of methanol, and add water to make exactly 100 mL.
Pipet 5 mL of this solution, add water to make exactly 50 Containers and storage Containers—Tight containers.
mL, and use this solution as the standard solution. Deter-
mine the absorbances, AT and AS, at 232 nm of the sample
solution and standard solution using water as the blank, as Roxithromycin
directed under Ultraviolet-visible Spectrophotometry <2.24>.
ロキシスロマイシン
Amount [mg (potency)] of rokitamycin (C42H69NO15)
=WS×(AT/AS)×(V?/V)×(1/10) Change the origin/limits of content to read:
WS: Amount [mg (potency)] of Rokitamycin Reference
Standard
Roxithromycin is a derivative of erythromycin.
It contains not less than 970 mg (potency) and not
Dissolution <6.10> When the test is performed at 50 revolu- more than 1020 mg (potency) per mg, calculated on the
tions per minute according to the Paddle method, using 900 anhydrous basis. The potency of Roxithromycin is
mL of water as the dissolution medium, the dissolution rate expressed as mass (potency) of roxithromycin
in 30 minutes of Rokitamycin Tablets is not less than 80z. (C41H76N2O15).
Start the test with 1 tablet of Rokitamycin Tablets,
withdraw not less than 20 mL of the medium at the specified
minute after starting the test, and filter through a membrane
filter with a pore size not exceeding 0.5 mm. Discard 10 mL
of the first filtrate, pipet V mL of the subsequent filtrate,
add water to make exactly V? mL so that each mL contains
about 22 mg (potency) of Rokitamycin according to the la-
beled amount, and use this solution as the sample solution.
Separately, weigh accurately about 22 mg (potency) of
1924 Official Monographs Supplement I, JP XV

lution as the sample solution. Separately, dissolve exactly 10


Salicylic Acid mg of phenol, exactly 25 mg of 4-hydroxyisophthalic acid
and exactly 50 mg of parahydroxybenzoic acid in the mobile
サリチル酸 phase to make exactly 100 mL. Pipet 1 mL of this solution,
add the mobile phase to make exactly 100 mL, and use this
Change the origin/limits of content to read: solution as the standard solution. Perform the test with ex-
actly 10 mL each of the sample solution and standard solu-
Salicylic Acid, when dried, contains not less than tion as directed under Liquid Chromatography <2.01> ac-
99.5z and not more than 101.0z of C7H6O3. cording to the following conditions, and determine each
peak area by the automatic integration method: the peak
Change the Description to read: areas of parahydroxybenzoic acid, 4-hydroxyisophthalic
acid and phenol obtained from the sample solution are not
Description Salicylic Acid occurs as white crystals or crys-
larger than the area of each respective peak from the stan-
talline powder. It has a slightly acid, followed by an acrid
dard solution, the area of the peak other than salicylic acid
taste.
and other than the substances mentioned above is not larger
It is freely soluble in ethanol (95) and in acetone, and
than the peak area of 4-hydroxisophthalic acid from the
slightly soluble in water.
standard solution, and the total area of peaks other than
salicylic acid is not larger than 2 times the peak area of para-
Change the Identification to read:
hydroxybenzoic acid from the standard solution.
Identification (1) A solution of Salicylic Acid (1 in 500) Operating conditions—
responds to the Qualitative Tests <1.09> (1) and (3) for salicy- Detector: An ultraviolet absorption photometer (wave-
late. length: 270 nm).
(2) Determine the absorption spectrum of a solution of Column: A stainless steel column 4.6 mm in inside di-
Salicylic Acid in ethanol (95) (3 in 200,000) as directed under ameter and 15 cm in length, packed with octadecylsilanized
Ultraviolet-visible Spectrophotometry <2.24>, and compare silica gel for liquid chromatography (5 mm in particle di-
the spectrum with the Reference Spectrum: both spectra ex- ameter).
hibit similar intensities of absorption at the same Column temperature: A constant temperature of about
wavelengths. 359C.
(3) Determine the infrared absorption spectrum of Sali- Mobile phase: A mixture of water, methanol and acetic
cylic Acid as directed in the potassium bromide disk method acid (100) (60:40:1).
under Infrared Spectrophotometry <2.25>, and compare the Flow rate: Adjust the flow rate so that the retention time
spectrum with the Reference Spectrum: both spectra exhibit of salicylic acid is about 17 minutes.
similar intensities of absorption at the same wave numbers. Time span of measurement: About 2 times as long as the
retention time of salicylic acid, beginning after the solvent
Change the Purity to read: peak.
System suitability—
Purity (1) Chloride <1.03>—Dissolve 5.0 g of Salicylic
Test for required detectability: Pipet 2 mL of the standard
Acid in 90 mL of water by heating, cool, dilute with water to
solution, and add the mobile phase to make exactly 10 mL.
100 mL, and filter. Discard the first 20 mL of the filtrate,
Confirm that the peak areas of parahydroxybenzoic acid, 4-
take subsequent 30 mL of the filtrate, add 6 mL of dilute
hydroxyisophthalic acid and phenol obtained from 10 mL of
nitric acid and water to make 50 mL, and perform the test
this solution are equivalent to 14 to 26z of the area of each
using this solution as the test solution. Prepare the control
respective peak from 10 mL of the standard solution.
solution with 0.35 mL of 0.01 mol/L hydrochloric acid VS
System performance: Dissolve 10 mg of phenol, 25 mg of
(not more than 0.008z).
4-hydroxyisophthalic acid and 50 mg of parahydroxyben-
(2) Sulfate <1.14>—To 20 mL of the filtrate obtained in
zoic acid in 100 mL of the mobile phase. To 1 mL of this so-
(1) add 1 mL of dilute hydrochloric acid and water to make
lution add the mobile phase to make 10 mL. When the
50 mL, and perform the test using this solution as the test so-
procedure is run with 10 mL of this solution under the above
lution. Prepare the control solution with 0.40 mL of 0.005
operating conditions, parahydroxybenzoic acid, 4-hydrox-
mol/L sulfuric acid VS (not more than 0.019z).
yisophthalic acid and phenol are eluted in this order with the
(3) Heavy metals <1.07>—Dissolve 2.0 g of Salicylic
resolution between the peaks of 4-hydroxyisophthalic acid
Acid in 25 mL of acetone, add 4 mL of sodium hydroxide
and phenol being not less than 4.
TS, 2 mL of dilute acetic acid and water to make 50 mL, and
System repeatability: When the test is repeated 6 times
perform the test using this solution as the test solution. Pre-
with 10 mL of the standard solution under the above operat-
pare the control solution as follows: to 2.0 mL of Standard
ing conditions, the relative standard deviation of the peak
Lead Solution add 25 mL of acetone, 2 mL of dilute acetic
areas of parahydroxybenzoic acid, 4-hydroxyisophthalic
acid and water to make 50 mL (not more than 10 ppm).
acid and phenol is not more than 2.0z, respectively.
(4) Related substances—Dissolve 0.50 g of Salicylic Acid
in the mobile phase to make exactly 100 mL, and use this so-
Supplement I, JP XV Official Monographs 1925

Change the Residue on ignition to read: Pipet 1 mL of the sample solution, add water to make exact-
ly 10 mL. Pipet 1 mL of this solution, add water to make
Residue on ignition <2.44> Not more than 0.1z (1 g).
exactly 50 mL, and use this solution as the standard solu-
tion. Perform the test with these solutions as directed under
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
Add the following:
sample solution and standard solution on a plate of silica gel
for thin-layer chromatography. Then develop with a mixture
L-Serine of 1-butanol, water and acetic acid (100) (3:1:1) to a distance
of about 10 cm, and dry the plate at 809 C for 30 minutes.
L-セリン
Spray evenly a solution of ninhydrin in a mixture of
methanol and acetic acid (100) (97:3) (1 in 100) on the plate,
and heat at 809 C for 10 minutes: the spot other than the
principal spot obtained from the sample solution is not more
C3H7NO3: 105.09
intense than the spot from the standard solution.
(2S)-2-Amino-3-hydroxypropanoic acid [56-45-1]
Loss on drying <2.41> Not more than 0.3z (1 g, 1059
C, 3
L-Serine,when dried, contains not less than 98.5 and hours).
not more than 101.0z of C3H7NO3. Residue on Ignition <2.44> Not more than 0.1z (1 g).
Description L-Serine occurs as white crystals or a crystal-
Assay Weigh accurately about 0.11 g of L-Serine, previ-
line powder. It has a slight sweet taste.
ously dried, dissolve in 3 mL of formic acid, add 50 mL of a-
It is freely soluble in water and in formic acid, and practi-
cetic acid (100), and titrate <2.50> with 0.1 mol/L perchloric
cally insoluble in ethanol (99.5).
acid VS (potentiometric titration). Perform a blank determi-
It dissolves in 2 mol/L hydrochloric acid TS.
nation in the same manner, and make any necessary correc-
Identification Determine the infrared absorption spectrum tion.
of L-Serine as directed in the potassium bromide disk
Each mL of 0.1 mol/L perchloric acid VS
method under Infrared Spectrophotometry <2.25>, and com- =10.51 mg of C3H7NO3
pare the spectrum with the Reference Spectrum: both spec-
tra exhibit similar intensities of absorption at the same wave Containers and storage Containers—Tight containers.
numbers.

Optical rotation <2.49> [a]20


D :+14.0 – +16.09(After dry-
ing, 2.5 g, 2 mol/L hydrochloric acid TS, 25 mL, 100 mm).
Sodium Bicarbonate Injection
pH <2.54> The pH of a solution prepared by dissolving 炭酸水素ナトリウム注射液
1.0 g of L-Serine in 10 mL of water is between 5.2 and 6.2.
Add the following next to Extractable volume:
Purity (1) Clarity and color of solution—Dissolve 1.0 g
of L-Serine in 10 mL of water: the solution is clear and color- Foreign insoluble matter <6.06> Perform the test according
less. to Method 1: it meets the requirement.
(2) Chloride <1.03>—Perform the test with 0.5 g of L- Insoluble particulate matter <6.07> It meets the require-
Serine. Prepare the control solution with 0.30 mL of 0.01 ment.
mol/L hydrochloric acid VS (not more than 0.021z).
(3) Sulfate <1.14>—Perform the test with 0.6 g of L-Ser- Sterility <4.06> Perform the test according to the Mem-
ine. Prepare the control solution with 0.35 mL of 0.005 brane filtration method: it meets the requirement.
mol/L sulfuric acid VS (not more than 0.028z).
(4) Ammonium <1.02>—Perform the test with 0.25 g of
L-Serine. Prepare the control solution with 5.0 mL of Stan- 10z Sodium Chloride Injection
dard Ammonium Solution (not more than 0.02z).
(5) Heavy metals <1.07>—Proceed with 2.0 g of L-Serine 10z 塩化ナトリウム注射液
according to Method 1, and perform the test. Prepare the
control solution with 2.0 mL of Standard Lead Solution (not Add the following next to Extractable volume:
more than 10 ppm). Foreign insoluble matter <6.06> Perform the test according
(6) Iron <1.10>—Prepare the test solution with 1.0 g of to Method 1: it meets the requirement.
L-Serine according to Method 1, and perform the test ac-
cording to Method A. Prepare the control solution with 1.0 Insoluble particulate matter <6.07> It meets the require-
mL of Standard Iron Solution (not more than 10 ppm). ment.
(7) Related substances—Dissolve 0.10 g of L-Serine in 10 Sterility <4.06> Perform the test according to the Mem-
mL of water, and use this solution as the sample solution. brane filtration method: it meets the requirement.
1926 Official Monographs Supplement I, JP XV

of potassium hexahydroxoantimonate (V) TS.


Sodium Citrate Injection for pH <2.54> To 1 g of Sodium Starch Glycolate add 30 mL
Transfusion of water and stir: the pH of the resulting suspension of Type
A is 5.5 – 7.5, and that of Type B is 3.0 – 5.0.
輸血用クエン酸ナトリウム注射液
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
Add the following next to Extractable volume: Sodium Starch Glycolate according to Method 2, and per-
form the test. Prepare the control solution with 2.0 mL of
Foreign insoluble matter <6.06> Perform the test according Standard Lead Solution (not more than 20 ppm).
to Method 1: it meets the requirement. (2) Iron
Insoluble particulate matter <6.07> It meets the require- (i) Sample solution Take 2.5 g of Sodium Starch
ment. Glycolate in a silica or platinum crucible, add 2 mL of 5
mol/L sulfuric acid TS. Heat on a water bath, then ignite
Sterility <4.06> Perform the test according to the Mem- cautiously with a gas burner or preferably in an electric fur-
brane filtration method: it meets the requirement. nace at 600±259C, and incinerate the residue completely.
Allow to cool, add a few drops of 1 mol/L sulfuric acid TS,
and heat and ignite as above. Allow to cool, add a few drops
Add the following: of ammonium carbonate TS, evaporate to dryness on a
water bath, and heat and ignite as above. After cooling, dis-
Sodium Starch Glycolate solve the residue by adding 50 mL of water.
(ii) Standard solution Weigh accurately 863.4 mg of
デンプングリコール酸ナトリウム ammonium iron (III) sulfate dodecahydrate, dissolve in
water, add 25 mL of 1 mol/L sulfuric acid TS, and add
Sodium Starch Glycolate [9063-38-1] water to make exactly 500 mL. Pipet 10 mL of this solution,
This monograph is harmonized with the European Phar- and add water to make exactly 100 mL. Pipet 5 mL of this
macopoeia and the U.S. Pharmacopeia. The parts of the text solution, and add water to make exactly 100 mL. Each mL
that are not harmonized are marked with symbols ( ). of this solution contains 1.0 mg of iron (Fe).
(iii) Procedure Pipet 10 mL each of the sample solu-
Sodium Starch Glycolate is the sodium salt of a car- tion and standard solution, and to each solution add 2 mL
boxymethyl ether of starch or of a cross-linked car- of citric acid solution (1 in 5) and 0.1 mL of thioglycolic
boxymethyl ether of starch. acid. Then add ammonia solution (28) dropwise to render
There are two neutralization types of Sodium Starch the solution alkaline, using litmus paper as an indicator.
Glycolate, Type A and Type B, and their insoluble Add water to make 20 mL, and use these solutions as the test
matter in a mixture of ethanol (99.5) and water (8:2), solution and the control solution, respectively. Allow these
when dried, contains not less than 2.8z and not more solutions to stand for 5 minutes, and compare the color of
than 4.2z, and not less than 2.0z and not more than the solutions using white background: the color of the test
3.4z of sodium (Na: 22.99), respectively. solution is not deeper than that of the control solution (not
 The label states the type of neutralization. more than 20 ppm).

(3) Sodium glycolate—Conduct this procedure without
Description Sodium Starch Glycolate occurs as a white exposure to light, using light-resistant vessels.
powder, and has a characteristic salty taste. (i) Sample solution Weigh accurately 0.200 g of Sodi-
It practically insoluble in ethanol (99.5). um Starch Glycolate in a beaker, add 4 mL of 6 mol/L acetic
It swells with water, and becomes viscous, pasty liquid. acid TS and 5 mL of water, and stir to dissolve. Add 50 mL
It is hygroscopic. of acetone and 1 g of sodium chloride, stir, and filter
Identification (1) Acidify 5 mL of a solution of Sodium through a filter paper previously soaked with acetone. Rinse
Starch Glycolate (1 in 500) with dilute hydrochloric acid, the beaker and the filter paper with acetone, combine the
then add one drop of iodine TS, and stir: a blue to violet filtrate and washings, and add acetone to make exactly 100
color is produced. mL. Allow to stand for 24 hours, and use the supernatant
(2) Determine the infrared absorption spectrum of liquid as the sample solution.
Sodium Starch Glycolate as directed in the potassium (ii) Standard solution To exactly 0.310 g of glycolic
bromide disk method under Infrared Spectrophotometry acid, previously dried in a desiccator (silica gel) for 18 hours,
<2.25>, and compare the spectrum with the Reference Spec- add water to dissolve to make exactly 500 mL. Pipet 5 mL of
trum: both spectra exhibit similar intensities of absorption at this solution, add 4 mL of 6 mol/L acetic acid TS, and allow
the same wave numbers. to stand for 30 minutes. Add 50 mL of acetone and 1 g of so-
(3) The sample solution obtained in the Purity (2) dium chloride, proceed as (i) above, and use the supernatant
responds to the Qualitative Tests <1.09> (2) for sodium salt. liquid as the standard solution.
Perform the test using 2 mL of the sample solution and 4 mL (iii) Procedure Pipet 2.0 mL each of the sample solu-
tion and standard solution into 25-mL stoppered test tubes,
Supplement I, JP XV Official Monographs 1927

and heat on a water bath for 20 minutes to remove acetone.


After cooling, add 20.0 mL of 2,7-dihydroxynaphthalene TS Sulbactam Sodium
to the residue, stopper the test tube, and heat on a water
bath for 20 minutes. Cool under running water, and transfer スルバクタムナトリウム
whole quantity of the content to a 25-mL volumetric flask.
Maintain the flask under running water, and add sulfuric Change the origin/limits of content to read:
acid to make 25 mL. Within 10 minutes, determine the ab-
sorbance of these solutions at 540 nm using water as the Sulbactam Sodium contains not less than 875 mg
blank as directed under Ultraviolet-visible Spectrophotomet- (potency) and not more than 941 mg (potency) per mg,
ry <2.24>; the absorbance of the sample solution is not larger calculated on the anhydrous basis. The potency of Sul-
than that of the standard solution (not more than 2.0z). bactam Sodium is expressed as mass (potency) of sul-
(4) Sodium chloride—Weigh accurately about 0.5 g of bactam (C8H11NO5S: 233.24).
Sodium Starch Glycolate in a beaker, disperse in 100 mL of
water, and add 1 ml of nitric acid. Titrate <2.50> with 0.1
mol/L silver nitrate VS (potentiometric titration): the Delete the following two Monographs:
amount of sodium chloride (NaCl: 58.44) is not more than
7.0z. Sulfinpyrazone
Each mL of 0.1 mol/L silver nitrate VS=5.844 mg of NaCl スルフィンピラゾン

Loss on drying <2.41> Not more than 10.0z (1 g, 1309


C, Sulfinpyrazone Tablets
90 minutes). スルフィンピラゾン錠
Assay To about 1 g of Sodium Starch Glycolate add 20 mL
of a mixture of ethanol (99.5) and water (8:2), stir for 10
minutes, and filter. Repeat this procedure until no more tur-
Sulpyrine Injection
bidity is produced by adding silver nitrate TS, and dry the スルピリン注射液
residue on the filter paper at 1059C to constant mass. Weigh
accurately 0.7 g of the mass, add 80 mL of acetic acid (100), Add the following next to Extractable volume:
and heat the mixture under a reflux condenser on a water
bath for 2 hours. After cooling, titrate <2.50> with 0.1 Foreign insoluble matter <6.06> Perform the test according
mol/L perchloric acid VS (potentiometric titration). to Method 1: it meets the requirement.

Content (z) of sodium (Na)=(V×2.299×100)/W Insoluble particulate matter <6.07> It meets the require-
ment.
V: Consumed amount (mL) of 0.1 mol/L perchloric acid
VS Sterility <4.06> Perform the test according to the Mem-
W: Mass (mg) of the dried residue brane filtration method: it meets the requirement.


Containers and storage Containers—Tight containers.
Sultamicillin Tosilate Hydrate
Sodium Thiosulfate Injection スルタミシリントシル酸塩水和物

チオ硫酸ナトリウム注射液 Change the origin/limits of content to read:

Delete the Pyrogen and add the following next Sultamicillin Tosilate Hydrate contains not less than
to Identification: 698 mg (potency) and not more than 800 mg (potency)
Bacterial endotoxins <4.01> Less than 0.01 EU/mg. per mg, calculated on the anhydrous basis and correct-
ed by the amount of residual solvent. The potency of
Add the following next to Extractable volume: Sultamicillin Tosilate Hydrate is expressed as mass
(potency) of sultamicillin (C25H30N4O9S2: 594.66).
Foreign insoluble matter <6.06> Perform the test according
to Method 1: it meets the requirement.

Insoluble particulate matter <6.07> It meets the require-


ment.

Sterility <4.06> Perform the test according to the Mem-


brane filtration method: it meets the requirement.
1928 Official Monographs Supplement I, JP XV

It is unctuous, and adheres readily to the skin.


Suxamethonium Chloride for It is practically insoluble in water and in ethanol (99.5).

Injection Identification Determine the infrared absorption spectrum


of Talc as directed in the potassium bromide disk method
注射用スキサメトニウム塩化物 under Infrared Spectrophotometry <2.25>: it exhibits ab-
sorption at the wave numbers of about 3680 cm-1,
Add the following next to Purity: 1018 cm-1 and 669 cm-1.
Bacterial endotoxins <4.01> Less than 1.5 EU/mg. Purity (1) Acidity or alkalinity—To 2.5 g of Talc add 50
Uniformity of dosage units <6.02> It meets the requirement mL of freshly boiled and cooled water, and heat under a
of the Mass variation test. reflux condenser. Filter the liquid by suction, add 0.1 mL of
bromothymol blue-sodium hydroxide-ethanol TS to 10 mL
Foreign insoluble matter <6.06> Perform the test according of the filtrate, and add 0.01 mol/L hydrochloric acid VS
to Method 2: it meets the requirement. until the color of the solution changes: the necessary volume
Insoluble particulate matter <6.07> It meets the require- of the VS is not more than 0.4 mL. Separately, to 10 mL of
ment. the filtrate add 0.1 mL of phenolphthalein TS, and add 0.01
mol/L sodium hydroxide VS until the color of the solution
Sterility <4.06> Perform the test according to the Mem- changes to light red: the necessary volume of the VS is not
brane filtration method: it meets the requirement. more than 0.3 mL.
(2) Acid-soluble substances—Weigh accurately about
1 g of Talc, heat with 20 mL of dilute hydrochloric acid at
Suxamethonium Chloride Injection 509C for 15 minutes with stirring. Cool, add water to make
exactly 50 mL, and filter. Centrifuge, if necessary, until the
スキサメトニウム塩化物注射液 filtrate becomes clear. To 25 mL of the filtrate add 1 mL of
dilute sulfuric acid, evaporate to dryness, and ignite to con-
Add the following next to Purity: stant mass at 800±259C: the amount of the residue is not
Bacterial endotoxins <4.01> Less than 2.0 EU/mg. more than 2.0z.
(3) Water-soluble substances—To 10.0 g of Talc add 50
Add the following next to Extractable volume: mL of water, weigh the mass, and boil for 30 minutes, sup-
plying water lost by evaporation. Cool, add water to restore
Foreign insoluble matter <6.06> Perform the test according the original mass, and filter. Centrifuge, if necessary, until
to Method 1: it meets the requirement. the filtrate becomes clear. Evaporate 20 mL of the filtrate to
Insoluble particulate matter <6.07> It meets the require- dryness, and dry the residue at 1059C for 1 hour: the mass of
ment. the residue is not more than 4.0 mg.
(4) Iron—Weigh accurately about 10 g of Talc, add 50
Sterility <4.06> Perform the test according to the Mem- mL of 0.5 mol/L hydrochloric acid VS gently while stirring,
brane filtration method: it meets the requirement. and heat under a reflux condenser on a water bath for 30
minutes. After cooling, transfer the content to a flask, and
allow to precipitate the insoluble matter. Filter the super-
Talc natant liquid through a filter paper for quantitative analysis
(No. 5B), leaving the precipitate in the flask as much as pos-
タルク sible, wash the remaining precipitate in the flask with three
10-mL portions of hot water, and also wash the filter paper
Change to read: with 15 mL of hot water, and combine the washings and the
filtrate. After cooling, add water to make exactly 100 mL,
Talc is a fractured and screened native hydrous mag- and use this solution as the sample stock solution. Pipet 2.5
nesium silicate. Pure talc is Mg3Si4O10(OH)2: 379.27. mL of the stock solution, add 50 mL of 0.5 mol/L
Talc may contain related mineral substances consist- hydrochloric acid VS, then add water to make exactly 100
ing chiefly of chlorite (hydrous magnesium aluminum mL, and use this solution as the sample solution. Separately,
silicate), magnesite (magnesium carbonate), calcite to 50 mL each of 0.5 mol/L hydrochloric acid VS add exact-
(calcium carbonate) and dolomite (calcium magnesi- ly 2 mL, 2.5 mL, 3 mL and 4 mL of Standard Iron Solution
um carbonate). for Atomic Absorption Spectrophotometry, add water to
It contains no asbestos. make them exactly 100 mL, and use these solutions as the
It contains not less than 17.0z and not more than standard solutions. Perform the test with the sample solu-
19.5z of magnesium (Mg: 24.31). tion and standard solutions as directed under Atomic Ab-
Description Talc occurs as a white to grayish white, fine, sorption Spectrophotometry <2.23> according to the follow-
crystalline powder. ing conditions, and calculate the amount of iron from the
calibration curve prepared from the absorbances of the stan-
Supplement I, JP XV Official Monographs 1929

dard solutions: not more than 0.25z. sulfuric acid, and heat gently to boiling with shaking. Cool
Gas: Combustible gas—Acetylene. immediately, filter, and wash the residue with 5 mL of dilute
Supporting gas—Air. sulfuric acid, then with 10 mL of water. Combine the filtrate
Lamp: Iron hollow-cathode lamp. and the washings, evaporate to 5 mL on a water bath, and
Wavelength: 248.3 nm. perform the test with this solution as the test solution (not
(5) Aluminum—Pipet 5 mL of the sample stock solution more than 4 ppm).
obtained in the Assay, add 10 mL of cesium chloride TS and
Loss on ignition <2.43> Not more than 7.0z (1 g, 1050 –
10 mL of hydrochloric acid, then add water to make exactly
11009C, constant mass).
100 mL, and use this solution as the sample solution.
Separately, to 10 mL of hydrochloric acid and 10 mL of cesi- Assay Weigh accurately about 0.5 g of Talc in a poly-
um chloride TS add exactly 5 mL, 10 mL, 15 mL and 20 mL tetrafluoroethylene dish, add 5 mL of hydrochloric acid, 5
of Standard Aluminum Solution for Atomic Absorption mL of nitric acid and 5 mL of perchloric acid, then add 35
Spectrophotometry, add water to make them exactly 100 mL of hydrofluoric acid while mixing gently, and evaporate
mL, and use these solutions as the standard solutions. Per- to dryness on a hot plate by heating gradually. Add 5 mL of
form the test with the sample solution and standard solu- hydrochloric acid to the residue, cover the dish with a watch
tions as directed under Atomic Absorption Spectrophoto- glass, and heat to boil. After cooling, transfer the content to
metry <2.23> according to the following conditions, and cal- a volumetric flask while washing the watch glass and dish
culate the amount of aluminum from the calibration curve with water, further wash the dish with water, transfer the
prepared from the absorbances of the standard solutions: washings to the flask, then add water to make exactly 50
not more than 2.0z. mL, and use this solution as the sample stock solution. Pipet
Gas: Combustible gas—Acetylene. 0.5 mL of the sample stock solution, and add water to make
Supporting gas—Nitrous oxide. exactly 100 mL. Pipet 4 mL of this solution, add 10 mL of
Lamp: Aluminum hollow-cathode lamp. hydrochloric acid and 10 mL of lanthanum chloride TS,
Wavelength: 309.3 nm. then add water to make exactly 100 mL, and use this
(6) Lead—Use the sample stock solution obtained in (4) solution as the sample solution. Separately, to 10 mL of
as the sample solution. Separately, to 50 mL of 0.5 mol/L hydrochloric acid and 10 mL of lanthanum chloride TS add
hydrochloric acid VS add exactly 5 mL, 7.5 mL, 10 mL and exactly 2.5 mL, 3 mL, 4 mL and 5 mL of Standard Magnesi-
12 mL of Standard Lead Solution, add water to make them um Solution for Atomic Absorption Spectrophotometry,
exactly 100 mL, and use these solutions as the standard solu- add water to make them exactly 100 mL, and use these solu-
tions. Perform the test with the sample solution and stan- tions as the standard solutions. Perform the test with the
dard solutions as directed under Atomic Absorption Spec- sample solution and standard solutions as directed under
trophotometry <2.23> according to the following conditions, Atomic Absorption Spectrophotometry <2.23> according to
and calculate the amount of lead from the calibration curve the following conditions, and calculate the amount of mag-
prepared from the absorbances of the standard solutions: nesium from the calibration curve prepared from the absor-
not more than 10 ppm. bances of the standard solutions.
Gas: Combustible gas—Acetylene. Gas: Combustible gas—Acetylene.
Supporting gas—Air. Supporting gas—Air.
Lamp: Lead hollow-cathode lamp. Lamp: Magnesium hollow-cathode lamp.
Wavelength: 217.0 nm. Wavelength: 285.2 nm.
(7) Calcium—Pipet 2.5 mL of the sample stock solution
Containers and storage Containers—Well-closed contain-
obtained in the Assay, add 10 mL of hydrochloric acid and
ers.
10 mL of lanthanum chloride TS, then add water to make
exactly 100 mL, and use this solution as the sample solution.
Separately, to 10 mL of hydrochloric acid and 10 mL of lan-
thanum chloride TS add exactly 1 mL, 2 mL, 3 mL and 4 Teceleukin for Injection
mL of Standard Calcium Solution, add water to make them (Genetical Recombination)
exactly 100 mL, and use these solutions as the standard solu-
tions. Perform the test with the sample solution and stan- 注射用テセロイキン(遺伝子組換え)
dard solutions as directed under Atomic Absorption Spec-
trophotometry <2.23> according to the following conditions, Change the Loss on drying to read:
and calculate the amount of calcium from the calibration Loss on drying Transfer the content of the vial of Teceleu-
curve prepared from the absorbances of the standard solu- kin for Injection (Genetical Recombination) to a weighing
tions: not more than 0.9z. bottle under the atmosphere not exceeding 10z relative hu-
Gas: Combustible gas—Acetylene. midity, and perform the test as directed in the Water content
Supporting gas—Nitrous oxide. determination described in the Minimum Requirements for
Lamp: Calcium hollow-cathode lamp. Biological Products: not more than 5z.
Wavelength: 422.7 nm.
(8) Arsenic <1.11>—To 0.5 g of Talc add 5 mL of dilute
1930 Official Monographs Supplement I, JP XV

Amount (mg) of tipepidine hibenzate (C15H17NS2.C14H10O4)


Thiamine Chloride Hydrochloride =WS×(QT/QS)×(V/50)
Injection WS: Amount (mg) of tipepidine hibenzate for assay
チアミン塩化物塩酸塩注射液 Internal standard solution—A solution of dibucaine hydro-
chloride in methanol (1 in 2000).
Add the following next to Identification:
Bacterial endotoxins <4.01> Less than 6.0 EU/mg.
Add the following:
Add the following next to Extractable volume:
Tobramycin Injection
Foreign insoluble matter <6.06> Perform the test according
to Method 1: it meets the requirement. トブラマイシン注射液
Insoluble particulate matter <6.07> It meets the require-
ment.
Tobramycin Injection is an aqueous injection.
It contains not less than 90.0z and not more than
Sterility <4.06> Perform the test according to the Mem- 110.0z of the labeled amount of tobramycin
brane filtration method: it meets the requirement. (C18H37N5O9: 467.51).
Method of preparation Prepare as directed under Injec-
tions, with Tobramycin.
Thiopental Sodium for Injection
Description Tobramycin Injection occurs as a colorless or
注射用チオペンタールナトリウム very pale yellow clear liquid.

Identification To a volume of Tobramycin Injection,


Add the following next to Loss on drying:
equivalent to 10 mg (potency) of Tobramycin according to
Bacterial endotoxins <4.01> Less than 0.30 EU/mg. the labeled amount, add water to make 1 mL, and use this
Uniformity of dosage units <6.02> It meets the requirement solution as the sample solution. Separately, dissolve 10 mg
of the Mass variation test. (potency) of Tobramycin Reference Standard in 1 mL of
water, and use this solution as the standard solution. Then,
Foreign insoluble matter <6.06> Perform the test according proceed as directed in the Identification (2) under Tobramy-
to Method 2: it meets the requirement. cin.
Insoluble particulate matter <6.07> It meets the require- Osmotic pressure ratio Being specified separately.
ment.
pH <2.54> 5.0 – 7.0

Bacterial endotoxins <4.01> Less than 0.50 EU/mg (poten-


Tipepidine Hibenzate Tablets cy).

チペピジンヒベンズ酸塩錠 Extractable volume <6.05> It meets the requirement.

Foreign insoluble matter <6.06> Perform the test according


Add the following next to Identification: to Method 1: it meets the requirement.
Uniformity of dosage units <6.02> Perform the test accord- Insoluble particulate matter <6.07> It meets the require-
ing to the following method: it meets the requirement of the ment.
Content uniformity test.
To 1 tablet of Tipepidine Hibenzate Tablets add 5 mL of Sterility <4.06> Perform the test according to the Mem-
diluted acetic acid (100) (1 in 2) and 15 mL of methanol per brane filtration method: it meets the requirement.
11 mg of tipepidine hibenzate (C15H17NS2.C14H10O4), and Assay Perform the test according to the Cylinder-plate
warm for 15 minutes with occasional shaking. After cooling, method as directed under Microbial Assay for Antibiotics
add diluted methanol (1 in 2) to make exactly V mL so that <4.02> according to the following conditions.
each mL contains about 0.44 mg of tipepidine hibenzate (i) Test organism, culture medium, and standard
(C15H17NS2.C14H10O4), and filter. Discard the first 10 mL of solutions—Proceed as directed in the Assay under Tobramy-
the filtrate, pipet 5 mL of the subsequent filtrate, add exact- cin.
ly 5 mL of the internal standard solution, then add diluted (ii) Sample solutions—To exactly 5 mL of Tobramycin
methanol (1 in 2) to make 25 mL, and use this solution as the Injection add 0.1 mol/L phosphate buffer solution, pH 8.0
sample solution. Then, proceed as directed in the Assay. so that each mL contains 1 mg (potency) of Tobramycin.
Take exactly a suitable amount of this solution, add 0.1
mol/L phosphate buffer solution, pH 8.0 to make solutions
Supplement I, JP XV Official Monographs 1931

so that each mL contains 8 mg (potency) and 2 mg (potency), that the peak area of trimetazidine obtained from 10 mL of
and use these solutions as the high and low concentration this solution is equivalent to 18 to 32z of that from 10 mL of
sample solutions, respectively. the standard solution.
System performance: When the procedure is run with 10
Containers and storage Containers—Hermetic containers.
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
factor of the peak of trimetazidine are not less than 15,000
Trimetazidine Hydrochloride and not more than 1.5, respectively.
System repeatability: When the test is repeated 6 times
トリメタジジン塩酸塩
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
Change the Purity (2) to read:
area of trimetazidine is not more than 2.0z.
Purity (2) Related substances—Dissolve 0.2 g of
Trimetazidine Hydrochloride in 50 mL of water, and use
this solution as the sample solution. Pipet 2 mL of the sam- Delete the following two Monographs:
ple solution, add water to make exactly 20 mL. Pipet 2 mL
of this solution, add water to make exactly 100 mL, and use Tubocurarine Chloride Hydrochloride Hydrate
this solution as the standard solution. Perform the test with
ツボクラリン塩化物塩酸塩水和物
exactly 10 mL each of the sample solution and standard solu-
tion as directed under Liquid Chromatography <2.01> ac- Tubocurarine Chloride Hydrochloride Injection
cording to the following conditions, and determine each
ツボクラリン塩化物塩酸塩注射液
peak area by the automatic integration method: the area of
the peak other than trimetazidine obtained from the sample
solution is not larger than 1.5 times that of trimetazidine
Add the following:
from the standard solution, and the total area of the peaks
other than trimetazidine from the sample solution is not
larger than 2.5 times the peak area of trimetazidine from the
L-Tyrosine
standard solution. L-チロジン
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 240 nm).
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized C9H11NO3: 181.19
silica gel for liquid chromatography (5 mm in particle di- (2S)-2-Amino-3-(4-hydroxyphenyl)propanoic acid
ameter). [60-18-4]
Column temperature: A constant temperature of about
409C. L-Tyrosine, when dried, contains not less than 99.0
Mobile phase A: Dissolve 2.87 g of sodium 1-heptanesul- z and not more than 101.0z of C9H11NO3.
fonate in water to make 1000 mL, and adjust the pH to 3.0
Description L-Tyrosine occurs as white crystals or a crys-
with diluted phosphoric acid (1 in 10). Mix 3 volumes of this
talline powder.
solution and 2 volumes of methanol.
It is freely soluble in formic acid, and practically insoluble
Mobile phase B: Methanol.
in water and in ethanol (99.5).
Flowing of the mobile phase: Control the gradient by mix-
It dissolves in dilute hydrochloric acid and in ammonia
ing the mobile phases A and B as directed in the following
TS.
table.
Identification (1) Determine the absorption spectrum of
Time after injection Mobile phase Mobile phase a solution of L-Tyrosine in 0.1 mol/L hydrochloric acid (1 in
of sample (min) A (volz) B (volz)
10,000) as directed under Ultraviolet-visible Spectrophoto-
0 – 50 95ª 75 5 ª25 metry <2.24>, and compare the spectrum with the Reference
Spectrum: both spectra exhibit similar intensities of absorp-
Flow rate: Adjust the flow rate so that the retention time
tion at the same wavelengths.
of trimetazidine is about 25 minutes.
(2) Determine the infrared absorption spectrum of
Time span of measurement: About 2 times as long as the
L-Tyrosine as directed in the potassium bromide disk
retention time of trimetazidine, beginning after the solvent
method under Infrared Spectrophotometry <2.25>, and com-
peak.
pare the spectrum with the Reference Spectrum: both spec-
System suitability—
tra exhibit similar intensities of absorption at the same wave
Test for required detectability: Pipet 5 mL of the standard
numbers.
solution, and add water to make exactly 20 mL. Confirm
1932 Official Monographs Supplement I, JP XV

Optical rotation <2.49> [a]20


D :-10.5 – -12.59 (after dry- rection.
ing, 2.5 g, 1 mol/L hydrochloric acid TS, 50 mL, 100 mm).
Each mL of 0.1 mol/L perchloric acid VS
Purity (1) Clarity and color of solution—Dissolve 1.0 g =18.12 mg of C9H11NO3
of L-Tyrosine in 20 mL of 1 mol/L hydrochloric acid TS: the
Containers and storage Containers—Tight containers.
solution is clear and colorless.
(2) Chloride <1.03>—Dissolve 0.5 g of L-Tyrosine in 12
mL of dilute nitric acid and 20 mL of water, and add water
Add the following:
to make 50 mL. Perform the test using this solution as the
test solution. Prepare the control solution as follows: To
0.30 mL of 0.01 mol/L hydrochloric acid VS add 12 mL of Ubenimex
dilute nitric acid and water to make 50 mL (not more than
ウベニメクス
0.021z).
(3) Sulfate <1.14>—Dissolve 0.6 g of L-Tyrosine in 5 mL
of dilute hydrochloric acid, and add water to make 45 mL.
Perform the test using this solution as the test solution. Pre-
pare the control solution as follows: To 0.35 mL of 0.005
mol/L sulfuric acid VS add 5 mL of dilute hydrochloric acid
and water to make 45 mL. To the test solution and the con- C16H24N2O4: 308.37
trol solution add 5 mL of barium chloride TS (not more than (2S)-2-[(2S,3R)-3-Amino-2-hydroxy-
0.028z). 4-phenylbutanoylamino]-4-methylpentanoic acid
(4) Ammonium <1.02>—Perform the test with 0.25 g of [58970-76-6]
L-Tyrosine. Prepare the control solution with 5.0 mL of
Standard Ammonium Solution (not more than 0.02z). Ubenimex, when dried, contains not less than 98.5
(5) Heavy metals <1.07>—Proceed with 1.0 g of L-Tyro- z and not more than 101.0z of C16H24N2O4.
sine according to Method 4, and perform the test. Prepare
Description Ubenimex occurs as a white crystalline pow-
the control solution with 1.0 mL of Standard Lead Solution
der.
(not more than 10 ppm).
It is freely soluble in acetic acid (100), slightly soluble in
(6) Iron <1.10>—Prepare the test solution with 1.0 g of
water, and very slightly soluble in ethanol (99.5).
L-Tyrosine according to Method 3, and perform the test ac-
It dissolves in 1 mol/L hydrochloric acid TS.
cording to Method A. Prepare the control solution with 1.0
Melting point: about 2309 C (with decomposition).
mL of Standard Iron Solution (not more than 10 ppm).
(7) Related substances—Dissolve 0.20 g of L-Tyrosine in Identification (1) Determine the absorption spectrum of
10 mL of diluted ammonia solution (28) (1 in 2), add water a solution of Ubenimex (1 in 2000) as directed under Ultrav-
to make 20 mL, and use this solution as the sample solution. iolet-visible Spectrophotometry <2.24>, and compare the
Pipet 1 mL of the sample solution, add water to make exact- spectrum with the Reference Spectrum: both spectra exhibit
ly 10 mL, pipet 1 mL of this solution, add water to make similar intensities of absorption at the same wavelengths.
exactly 50 mL, and use this solution as the standard solu- (2) Determine the infrared absorption spectrum as
tion. Perform the test with these solutions as directed under directed in the potassium bromide disk method under In-
Thin-layer Chromatography <2.03>. Spot 5 mL each of the frared Spectrophotometry <2.25>, and compare the spectrum
sample solution and standard solution on a plate of silica gel with the Reference Spectrum: both spectra exhibit similar
for thin-layer chromatography. Then develop with a mixture intensities of absorption at the same wave numbers.
of 1-propanol and ammonia solution (28) (67:33) to a dis-
Optical rotation <2.49> [a]20
D :-15.5 – -17.59 (after dry-
tance of about 10 cm, and dry the plate at 809 C for 30
ing, 0.5 g, 1 mol/L hydrochloric acid TS, 50 mL, 100 mm).
minutes. Spray evenly a solution of ninhydrin in a mixture
of methanol and acetic acid (100) (97:3) (1 in 100) on the Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of
plate, and then heat at 809 C for 10 minutes: the spot other Ubenimex according to Method 2, and perform the test. Pre-
than the principal spot obtained from the sample solution is pare the control solution with 2.0 mL of Standard Lead
not more intense than the spot from the standard solution. Solution (not more than 10 ppm).
(2) Related substances—Dissolve 30 mg of Ubenimex in
Loss on drying <2.41> Not more than 0.3z (1 g, 1059C, 3
10 mL of the mobile phase A, and use this solution as the
hours).
sample solution. Pipet 2 mL of the sample solution, add the
Residue on ignition <2.44> Not more than 0.1z (1 g). mobile phase A to make exactly 200 mL, and use this solu-
tion as the standard solution. Perform the test with exactly
Assay Weigh accurately about 0.18 g of L-Tyrosine previ-
20 mL each of the sample solution and standard solution as
ously dried, dissolve in 6 mL of formic acid, add 50 mL of
directed under Liquid Chromatography <2.01> according to
acetic acid (100), and titrate <2.50> with 0.1 mol/L perchlor-
the following conditions, and determine each peak area of
ic acid VS (potentiometric titration). Perform a blank deter-
these solutions by the automatic integration method: the
mination in the same manner, and make any necessary cor-
Supplement I, JP XV Official Monographs 1933

area of the peak other than ubenimex obtained from the titration). Perform a blank determination in the same man-
sample solution is not larger than 1/2 times the peak area of ner, and make any necessary correction.
ubenimex from the standard solution. Furthermore, the
Each mL of 0.1 mol/L perchloric acid VS
total area of the peaks other than ubenimex from the sample
=30.84 mg of C16H24N2O4
solution is not larger than the peak area of ubenimex from
the standard solution. Containers and storage Containers—Tight containers.
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 220 nm). Vincristine Sulfate
Column: A stainless steel column 4.6 mm in inside di-
ameter and 25 cm in length, packed with octadecylsilanized ビンクリスチン硫酸塩
silica gel for liquid chromatography (5 mm in particle di-
ameter). Change the Description to read:
Column temperature: A constant temperature of about
Description Vincristine Sulfate occurs as a white to light
259C.
yellowish white powder.
Mobile phase A: A mixture of diluted 0.1 mol/L potassi-
It is very soluble in water, and practically insoluble in
um dihydrogen phosphate TS (13 in 20) and acetonitrile for
ethanol (99.5).
liquid chromatography (17:3).
It is hygroscopic.
Mobile phase B: A mixture of acetonitrile for liquid chro-
Optical rotation [a]20D : +28.5 – +35.59(0.2 g, calculat-
matography and diluted 0.1 mol/L potassium dihydrogen
ed on the dried basis, water, 10 mL, 100 mm).
phosphate TS (13 in 20) (2:1).
Flowing of the mobile phase: Control the gradient by mix-
Change the Identification to read:
ing the mobile phases A and B as directed in the following
table. Identification
(1) Determine the absorption spectrum of a solution of
Time after injection Mobile phase Mobile phase Vincristine Sulfate (1 in 50,000) as directed under Ultrav-
of sample (min) A (volz) B (volz)
iolet-visible Spectrophotometry <2.24>, and compare the
0 – 20 100 0 spectrum with the Reference Spectrum or the spectrum of a
20 – 60 100ª0 0ª100 solution of Vincristine Sulfate Reference Standard prepared
60 – 70 0 100
in the same manner as the sample solution: both spectra
Flow rate: Adjust the flow rate so that the retention time exhibit similar intensities of absorption at the same
of ubenimex is about 14 minutes. wavelengths.
Time span of measurement: About 5 times as long as the (2) Determine the infrared absorption spectrum of Vin-
retention time of ubenimex, beginning after the solvent cristine Sulfate as directed in the potassium bromide disk
peak. method under Infrared Spectrophotometry <2.25>, and
System suitability— compare the spectrum with the Reference Spectrum or the
Test for required detectability: Pipet 1 mL of the standard spectrum of Vincristine Sulfate Reference Standard: both
solution, and add the mobile phase A to make exactly 10 spectra exhibit similar intensities of absorption at the same
mL. Confirm that the peak area of ubenimex obtained from wave numbers.
20 mL of this solution is equivalent to 7 to 13z of that from (3) A solution of Vincristine Sulfate (1 in 100) responds
20 mL of the standard solution. to the Qualitative Tests <1.09> for sulfate.
System performance: When the procedure is run with 20
mL of the standard solution under the above operating con- Change the Purity to read:
ditions, the number of theoretical plates and the symmetry
Purity (1) Clarity and color of solution—Dissolve 50 mg
factor of the peak of ubenimex are not less than 5000 and
of Vincristine Sulfate in 10 mL of water: the solution is clear
not more than 2.0, respectively.
and colorless.
System repeatability: When the test is repeated 6 times
(2) Related substances—Dissolve 10 mg of Vincristine
with 20 mL of the standard solution under the above operat-
Sulfate in 10 mL of water, and use this solution as the sam-
ing conditions, the relative standard deviation of the peak
ple solution. Pipet 2 mL of the sample solution, add water to
area of ubenimex is not more than 2.0z.
make exactly 50 mL, and use this solution as the standard
Loss on drying <2.41> Not more than 0.5z (0.5 g, in vacu- solution. Perform the test with exactly 200 mL each of the
um, 809 C, 4 hours). sample solution and standard solution as directed under Liq-
uid Chromatography <2.01> according to the following con-
Residue on ignition <2.44> Not more than 0.1z (1 g).
ditions. Determine each peak area by the automatic integra-
Assay Weigh accurately about 0.5 g of Ubenimex, previ- tion method: the peak area of desacetyl vincristine and vin-
ously dried, dissolve in 60 mL of acetic acid (100), and titrate blastine, having the relative retention times of about 0.9 and
<2.50> with 0.1 mol/L perchloric acid VS (potentiometric about 1.6 with respect to vincristine, respectively, obtained
1934 Official Monographs Supplement I, JP XV

from the sample solution are not larger than 1/8 times and Temperature range: room temperature to 2009C.
3/20 times, respectively, the peak area of vincristine from Atmospheric gas: dried nitrogen.
the standard solution, and the area of the peak other than Flow rate of atmospheric gas: 40 mL/minute.
vincristine, desacetyl vincristine and vinblastine from the
sample solution is not larger than 1/4 times the peak area of Change the Assay to read:
vincristine from standard solution. Furthermore, the total
Assay Weigh accurately about 10 mg each of Vincristine
area of the peaks other than vincristine from the sample so-
Sulfate and Vincristine Sulfate Reference Standard
lution is not larger than the peak area of vincristine from the
(separately determine the loss on drying in the same manner
standard solution.
as Vincristine Sulfate), dissolve each in water to make exact-
Operating conditions—
ly 10 mL, and use these solutions as the sample solution and
Detector: An ultraviolet absorption photometer (wave-
the standard solution, respectively. Perform the test with ex-
length: 297 nm).
actly 10 mL each of the sample solution and standard solu-
Column: A stainless steel column 4.6 mm in inside di-
tion as directed under Liquid Chromatography <2.01> ac-
ameter and 25 cm in length, packed with octylsilanized silica
cording to the following conditions, and determine the vin-
gel for liquid chromatography (5 mm in particle diameter).
cristine peak areas, AT and AS, of both solutions.
Column temperature: A constant temperature of about
409C. Amount (mg) of C46H56N4O10.H2SO4=WS×(AT/AS)
Mobile phase A: methanol.
WS: Amount (mg) of Vincristine Sulfate Reference Stan-
Mobile phase B: A mixture of water and diethylamine
dard, calculated on the dried basis
(197:3), adjusted the pH to 7.5 with phosphoric acid.
Flowing of the mobile phase: Control the gradient by mix- Operating conditions—
ing the mobile phases A and B as directed in the following Detector: An ultraviolet absorption photometer (wave-
table. length: 297 nm).
Column: A stainless steel column 4.6 mm in inside di-
Time after injection Mobile phase Mobile phase ameter and 25 cm in length, packed with octylsilanized silica
of sample (min) A (volz) B (volz)
gel for liquid chromatography (5 mm in particle diameter).
0 – 12 62 38 Column temperature: A constant temperature of about
12 – 27 62ª 92 38ª8 259C.
Flow rate: Adjust the flow rate so that the retention time Mobile phase: Adjust the pH to 7.5 of a mixture of water
of vincristine is about 15 minutes. and diethylamine (59:1) with phosphoric acid. To 300 mL of
Time span of measurement: About 1.7 times as long as the this solution add 700 mL of methanol.
retention time of vincristine, beginning after the solvent Flow rate: Adjust the flow rate so that the retention time
peak. of vincristine is about 7 minutes.
System suitability— System suitability—
Test for required detectability: Pipet 5 mL of the standard System performance: Dissolve 5 mg each of Vincristine
solution, and add the mobile phase to make exactly 200 mL. Sulfate and vinblastine sulfate in 5 mL of water. When the
Confirm that the peak area of vincristine obtained from 200 procedure is run with 10 mL of this solution under the above
mL of this solution is equivalent to 1.75 to 3.25z of that operating conditions, vincristine and vinblastine are eluted
from 200 mL of the standard solution. in this order with the resolution between these peaks being
System performance: Dissolve 15 mg each of Vincristine not less than 4.
Sulfate and vinblastine sulfate in 100 mL of water. When the System repeatability: When the test is repeated 6 times
procedure is run with 200 mL of this solution under the with 10 mL of the standard solution under the above operat-
above operating conditions, vincristine and vinblastine are ing conditions, the relative standard deviation of the peak
eluted in this order with the resolution between these peaks area of vincristine is not more than 1.0z.
being not less than 4.
System repeatability: When the test is repeated 6 times Change the Containers and storage to read:
with 200 mL of the standard solution under the above oper- Containers and storage Containers—Tight containers.
ating conditions, the relative standard deviation of the peak Storage—Light-resistant, and at not exceeding -209
C.
area of vincristine is not more than 1.5z.

Change the Loss on drying to read: Water for Injection


Loss on drying Perform the test with about 10 mg of Vin-
cristine Sulfate as directed in Method 2 under Thermal Anal- 注射用水
ysis <2.52> according to the following conditions: not more
than 12.0z. Add the following next to Extractable volume:
Operating conditions— Foreign insoluble matter <6.06> Perform the test according
Heating rate: 59C/minute. to Method 1: it meets the requirement.
Supplement I, JP XV Official Monographs 1935

Insoluble particulate matter <6.07> It meets the require- mm).


ment.
Purity (1) Heavy metals <1.07>— Proceed with 1.0 g of
Zidovudine according to Method 4, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead
Xylitol Injection Solution (not more than 20 ppm).
(2) 1-[(2R,5S)-2,5-Dihydro-5-(hydroxymethyl)-2-furyl]
キシリトール注射液
thymine, triphenylmethanol, and other related substan-
ces—Dissolve 0.20 g of Zidovudine in methanol to make ex-
Add the following next to Extractable volume:
actly 10 mL, and use this solution as the sample solution.
Foreign insoluble matter <6.06> Perform the test according Separately, add 1 mL of the sample solution to 20 mg each
to Method 1: it meets the requirement. of thymine for liquid chromatography, 1-[(2R,5S)-2,5-di-
hydro-5-(hydroxymethyl)-2-furyl]thymine for thin-layer
Insoluble particulate matter <6.07> It meets the require-
chromatography, and triphenylmethanol for thin-layer
ment.
chromatography, and add methanol to dissolve to make ex-
Sterility <4.06> Perform the test according to the Mem- actly 100 mL. Pipet 5 mL of this solution, add methanol to
brane filtration method: it meets the requirement. make exactly 10 mL, and use this solution as the standard
solution. Perform the test with these solutions as directed
under Thin-layer Chromatography <2.03>. Spot 10 mL each
Add the following: of the sample solution and standard solution on a plate of
silica gel with fluorescent indicator for thin-layer chro-
Zidovudine matography. Develop the plate with a mixture of chlo-
roform and methanol (9:1) to a distance of about 12 cm, and
ジドブジン air-dry the plate. Examine under ultraviolet light (main
wavelength: 254 nm): the spot obtained from the sample so-
lution that corresponds to the position of the 1-[(2R,5S)-2,5-
dihydro-5-(hydroxymethyl)-2-furyl]thymine obtained from
the standard solution is not more intense than the spot from
the standard solution, and the spot other than the principal
spot and spots other than thymine and 1-[(2R,5S)-2,5-di-
hydro-5-(hydroxymethyl)-2-furyl]thymine from the sample
solution is not more intense than zidovudine spot from the
C10H13N5O4: 267.24 standard solution. However, the 3 spots from the standard
3?-Azido-3?-deoxythymidine [30516-87-1] solution appear in ascending order of Rf value thymine,
1-[(2R,5S)-2,5-dihydro-5-(hydroxymethyl)-2-furyl]thymine,
Zidovudine contains not less than 97.0z and not and zidovudine. Furthermore, spray evenly on the plate a so-
more than 102.0z of C10H13N5O4, calculated on the lution of vanillin in sulfuric acid (1 in 100): the spot from the
anhydrous basis. sample solution corresponding to the spot of
triphenylmethanol from the standard solution is not more
Description Zidovudine occurs as a white to pale yellowish
intense than the spot from the standard solution.
white powder.
(3) Thymine, 3?-chloro-3?-deoxythymidine, and other
It is freely soluble in methanol, soluble in ethanol (99.5),
related substances—Use the sample solution obtained in the
and sparingly soluble in water.
Assay as the sample solution. Separately, weigh accurately
It gradually turns yellow-brown on exposure to light.
about 20 mg of thymine for liquid chromatography, dissolve
Melting point: about 1249 C.
in 100 mL of methanol, and add the mobile phase to make
Identification Determine the infrared absorption spectrum exactly 250 mL. Pipet 5 mL of this solution, add the mobile
of Zidovudine as directed in the potassium bromide disc phase to make exactly 50 mL, and use this solution as the
method under Infrared Spectrophotometry <2.25>, and com- standard solution. Perform the test with exactly 10 mL each
pare the spectrum with the Reference Spectrum or the spec- of the sample solution and standard solution as directed un-
trum of Zidovudine Reference Standard: both spectra ex- der Liquid Chromatography <2.01> according to the follow-
hibit similar intensities of absorption at the same wave num- ing conditions. Determine the thymine peak areas, AT and
bers. If any difference appears between the spectra, dissolve AS, of the sample and standard solutions, and calculate the
the sample and the Reference Standard separately in a small amount of thymine using the following formula: the amount
amount of water and dry them in a desiccator (in vacuum, is not more than 2.0z. Also, determine the peak area of
phosphorus (V) oxide), and perform the test with the each peak obtained from the sample solution by the auto-
residues. matic integration method, and calculate the amounts of
related substances other than thymine by the area percentage
Optical rotation <2.49> [a]25
D : +60.5 – +63.09(0.5 g calcu-
method: the amount of 3?-chloro-3?-deoxythymidine, whose
lated on the anhydrous basis, ethanol (99.5), 50 mL, 100
1936 Official Monographs Supplement I, JP XV

relative retention time to zidovudine is 1.2, is not more than mL each of the sample solution and standard solution as
1.0z, and is not more than 0.5z for all other related sub- directed under Liquid Chromatography <2.01> according to
stances. Finally, the total amount of thymine, 3?-chloro-3?- the following conditions. Determine the peak areas of
deoxythymidine, and all related substances obtained above zidovudine, AT and AS of both solutions.
is not more than 3.0z.
Amount (mg) of C10H13N5O4=WS×(AT/AS)
Amount (z) of thymine=(WS/WT)×(AT/AS)×10
WS: Amount (mg) of Zidovudine Reference Standard,
WS: Amount (mg) of thymine for liquid chromatography calculated on the anhydrous basis
WT: Amount (mg) of Zidovudine
Operating conditions—
Operating conditions— Detector: An ultraviolet absorption photometer (wave-
Detector, column, column temperature, mobile phase, length: 265 nm).
and flow rate: Proceed as directed in the operating condi- Column: A stainless steel column 4.6 mm in inside di-
tions in the Assay. ameter and 25 cm in length, packed with octadecylsilanized
Time span of measurement: About 2 times as long as the silica gel for liquid chromatography (particle diameter: 5
retention time of zidovudine, beginning after the solvent mm).
peak. Column temperature: A constant temperature of about
System suitability— 259C.
Test for required detectability: Pipet 2 mL of the sample Mobile phase: A mixture of water and methanol (4:1).
solution, add the mobile phase to make exactly 100 mL, and Flow rate: Adjust the flow rate so that the retention time
use this solution as the solution for system suitability test. of zidovudine is about 15 minutes.
Pipet 1 mL of the solution for system suitability test, and System suitability—
add the mobile phase to make exactly 20 mL. Confirm that System performance: Dissolve 50 mg of Zidovudine in 50
the peak area of zidovudine obtained from 10 mL of this so- mL of the mobile phase. Separately, dissolve 5 mg of 3?-
lution is equivalent to 3.5 to 6.5z of that from 10 mL of the chloro-3?-deoxythymidine for liquid chromatography in 50
solution for system suitability test. mL of the mobile phase. Mix 10 mL and 1 mL of these solu-
System performance and system repeatability: Proceed as tions, respectively, and add the mobile phase to make 50
directed in the system suitability in the Assay. mL. When the procedure is run with 10 mL of this solution
under the above conditions, zidovudine and 3?-chloro-3?-
Water <2.48> Not more than 1.0z (0.25 g, coulometric
deoxythymidine are eluted in this order with the resolution
titration).
between these peaks being not less than 1.4, and the symmet-
Residue on ignition <2.44> Not more than 0.2z (0.5 g). ry factor of the peak of zidovudine is not more than 1.5.
System repeatability: When the test is repeated 6 times
Assay Weigh accurately about 50 mg of Zidovudine and
with 10 mL of the standard solution under the above condi-
Zidovudine Reference Standard (separately determine the
tions, the relative standard deviation of the peak area of
water <2.48> using the same manner as Zidovudine), and dis-
zidovudine is not more than 2.0z.
solve in the mobile phase to make exactly 50 mL. Pipet 10
mL of each solution, add the mobile phase to make them ex- Containers and storage Containers—Tight containers.
actly 50 mL, and use these solutions as the sample and stan- Storage—Light-resistant.
dard solutions, respectively. Perform the test with exactly 10
Crude Drugs

Alpinia Officinarum Rhizome Apricot Kernel


リョウキョウ キョウニン

Add the following next to Identification: Change the Identification to read:


Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of Identification To 1.0 g of ground Apricot Kernel add 10
pulverized Alpinia Officinarum Rhizome according to mL of methanol, immediately heat under a reflux condenser
Method 3, and perform the test. Prepare the control solution on a water bath for 10 minutes, cool, filter, and use the
with 3.0 mL of Standard Lead Solution (not more than 10 filtrate as the sample solution. Separately, dissolve 2 mg of
ppm). amygdalin for thin-layer chromatography in 1 mL of
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g methanol, and use this solution as the standard solution.
of pulverized Alpinia Officinarum Rhizome according to Perform the test with these solutions as directed under Thin-
Method 4, and perform the test (not more than 5 ppm). layer Chromatography <2.03>. Spot 20 mL each of the sam-
ple solution and standard solution on a plate of silica gel for
thin-layer chromatography. Develop the plate with a mix-
Anemarrhena Rhizome ture of ethyl acetate, methanol and water (20:5:4) to a dis-
tance of about 10 cm, and air-dry the plate. Examine under
チモ ultraviolet light (main wavelength: 365 nm): a spot with a
blueish white fluorescence appears at around Rf 0.7. Spray
Change the Purity to read: evenly thymol-sulfuric acid-methanol TS for spraying upon
the plate, and heat at 1059C for 5 minutes: one of the spot
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
among the several spots from the sample solution has the
pulverized Anemarrhena Rhizome according to Method 3,
same color tone and Rf value with the red-brown spot from
and perform the test. Prepare the control solution with 3.0
the standard solution.
mL of Standard Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of pulverized Anemarrhena Rhizome according to Method
4, and perform the test (not more than 5 ppm).
Add the following:
(3) Foreign matter <5.01>—The amount of fiber,
originating from the dead leaves, and other foreign matters Aralia Rhizome
contained in Anemarrhena Rhizome is not more than 3.0z.
Araliae Cordatae Rhizoma
ドクカツ
Angelica Dahurica Root
Aralia Rhizome is usually the rhizome of Aralia cor-
ビャクシ data Thunberg (Araliaceae).
Description Aralia Rhizome is curved, irregular cylindrical
Change the Purity to read:
to masses occasionally with remains of short roots. 4 to 12
Purity (1) Leaf sheath—The amount of leaf sheath con- cm in length, 2.5 to 7 cm in diameter, often cut crosswise or
tained in Angelica Dahurica Root does not exceed 3.0z. lengthwise. 1 to several, enlarged dents by remains of stems
(2) Heavy metals <1.07>—Proceed with 3.0 g of pulver- on the upper part or rarely 1.5 to 2.5 cm in diameter,
ized Angelica Dahurica Root according to Method 3, and remains of short stem. The outer surface is dark brown to
perform the test. Prepare the control solution with 3.0 mL yellowish brown, with longitudinally wrinkles, bases or
of Standard Lead Solution (not more than 10 ppm). dents of root. The transverse section of rhizome reveals dark
(3) Arsenic <1.11>—Prepare the test solution with 0.40 g brown to yellowish brown, scattered brownish small spots
of pulverized Angelica Dahurica Root according to Method with oil canals, and with numerous splits.
4, and perform the test (not more than 5 ppm). Odor, characteristic; taste, slightly bitter.
(4) Foreign matter <5.01>—The amount of foreign mat- Under a microscope <5.01>, a transverse section of rhi-
ter other than leaf sheath contained in Angelica Dahurica zome reveals the outermost layer to be cork layer, rarely
Root is not more than 1.0z. composed of cork stone cells, followed these appeared sever-

1937
1938 Crude Drugs Supplement I, JP XV

al layers of collenchyma. Vascular bundle and medullary aristolochic acid I.


rays is distinct, pith broad. Phloem fibre bundles are some- Operating conditions—
times observed at the outer portion of phloem. Oil canals Detector: An ultraviolet or visible absorption photometer
composed of schizogenous intercellular space in cortex and (wavelength: 400 nm).
pith. Cortex composed of vessels, xylem fibres, and oc- Column: A stainless steel column 4.6 mm in inside di-
casionally thick-wall xylem parenchyma. Vascular bundles ameter and 25 cm in length, packed with octadecylsilanized
scattered on the pith. And, parenchymatous cells observed silica gel for liquid chromatography (5 mm in particle di-
rosette aggregates of calcium oxalate. Starch grains com- ameter).
posed of simple grains, 2- to 6- compound grains. Column temperature: A constant temperature of about
409C.
Identification To 1 g of pulverized Aralia Rhizome add 10
Mobile phase: A mixture of a solution prepared by dis-
mL of methanol, shake for 5 minutes, filter, and use the
solving 7.8 g of sodium dihydrogen phosphate dihydrate and
filtrate as the sample solution. Perform the test with the
2 mL of phosphoric acid in water to make 1000 mL and
sample solution as directed under Thin-layer Chro-
acetonitrile (11:9).
matography <2.03>. Spot 5 mL of the sample solution on a
Flow rate: Adjust the flow rate so that the retention time
plate of silica gel for thin-layer chromatography, develop the
of aristolochic acid I is about 15 minutes.
plate with a mixture of hexane, ethyl acetate and acetic acid
System suitability—
(100) (30:10:1) to a distance of about 10 cm, and air-dry the
Test for required detectability: Measure exactly 1 mL of
plate. Spray evenly vanillin-sulfuric acid TS on the plate,
the standard solution, and add diluted methanol (3 in 4) to
and heat at 1059 C for 5 minutes: a purple spot appears at
make exactly 10 mL. Confirm that the ratio, S/N, of the sig-
around Rf 0.6.
nal (S) and noise (N) of aristolochic acid I obtained from 20
Loss on drying <5.01> Not more than 12.0z. mL of this solution is not less than 3. In this case, S means
the peak height on the chromatogram not including noise
Total ash <5.01> Not more than 9.0z.
obtained by drawing an average line of the detector output,
Acid-insoluble ash <5.01> Not more than 1.5z. and N is 1/2 of the difference between the maximum and
minimum output signals of the baseline around the peak in
Extract content <5.01> Dilute ethanol-soluble extract: not
the range of 20 times the width at half-height of the peak.
less than 15.0z.
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
Asiasarum Root area of aristolochic acid I is not more than 5.0z.
(5) Total BHC's and total DDT's <5.01>—Not more
サイシン
than 0.2 ppm, respectively.
Change the Purity to read:
Purity (1) Terrestrial part—Any terrestrial parts are not Asparagus Tuber
found.
(2) Arsenic <1.11>—Prepare the test solution with 0.4 g テンモンドウ
of pulverized Asiasarum Root according to Method 4, and
perform the test (not more than 5 ppm). Add the following next to Identification:
(3) Foreign matter <5.01>—The amount of foreign mat-
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
ter other than terrestrial part contained in Asiasarum Root is
pulverized Asparagus Tuber according to Method 3, and
not more than 1.0z.
perform the test. Prepare the control solution with 3.0 mL
(4) Aristolochic acid I—To exactly 2.0 g of pulverized
of Standard Lead Solution (not more than 10 ppm).
Asiasarum Root add exactly 50 mL of diluted methanol (3 in
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
4), shake for 15 minutes, filter, and use the filtrate as the
of pulverized Asparagus Tuber according to Method 4, and
sample solution. Separately, dissolve exactly 1.0 mg of
perform the test (not more than 5 ppm).
aristolochic acid I for crude drugs purity test in diluted
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 Atractylodes Rhizome
the test with exactly 20 mL each of the sample solution and
ビャクジュツ
standard solution as directed under Liquid Chromatography
<2.01>, according to the following conditions: the sample
Change the Purity to read:
solution shows no peak at the retention time corresponding
to aristolochic acid I from the standard solution. If the sam- Purity (1) Arsenic <1.11>—Prepare the test solution with
ple solution shows such a peak, repeat the test under differ- 0.40 g of pulverized Atractylodes Rhizome according to
ent conditions to confirm that the peak in question is not Method 4, and perform the test (not more than 5 ppm).
Supplement I, JP XV Crude Drugs 1939

(2) Atractylodes lancea rhizome—To 2.0 g of pulverized


Atractylodes Rhizome add exactly 5 mL of hexane, shake Powdered Calumba
for 5 minutes, filter, and use this filtrate as the sample solu-
tion. Perform the test with the sample solution as directed コロンボ末
under Thin-layer Chromatography <2.03>. Spot 10 mL of the
solution on a plate of silica gel for thin-layer chro- Add the following next to Identification:
matography. Develop the plate with a mixture of hexane and
Purity Arsenic <1.11>—Prepare the test solution with 0.40
acetone (7:1) to a distance of about 10 cm, and air-dry the
g of Powdered Calumba according to Method 4, and per-
plate. Spray evenly 4-dimethylaminobenzaldehyde TS for
form the test (not more than 5 ppm).
spraying on the plate, and heat at 1009 C for 5 minutes: no
green to grayish green spot appears at the Rf value of be-
tween 0.3 and 0.6.
Cimicifuga Rhizome
ショウマ
Powdered Atractylodes Rhizome
Change the Purity to read:
ビャクジュツ末
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
Change the Purity to read: pulverized Cimicifuga Rhizome according to Method 3, and
perform the test. Prepare the control solution with 3.0 mL
Purity (1) Arsenic <1.11>—Prepare the test solution with
of Standard Lead Solution (not more than 10 ppm).
0.40 g of Powdered Atractylodes Rhizome according to
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
Method 4, and perform the test (not more than 5 ppm).
of pulverized Cimicifuga Rhizome according to Method 4,
(2) Atractylodes lancea rhizome—To 2.0 g of Powdered
and perform the test (not more than 5 ppm).
Atractylodes Rhizome add exactly 5 mL of hexane, shake
(3) Rhizome of Astilbe thunbergii Miquel—Under a
for 5 minutes, filter, and use this filtrate as the sample solu-
microscope <5.01>, pulverized Cimicifuga Rhizome does not
tion. Perform the test with the sample solution as directed
contain crystal druses in the parenchyma.
under Thin-layer Chromatography <2.03>. Spot 10 mL of the
solution on a plate of silica gel for thin-layer chro-
matography. Develop the plate with a mixture of hexane and
acetone (7:1) to a distance of about 10 cm, and air-dry the
Clematis Root
plate. Spray evenly 4-dimethylaminobenzaldehyde TS for イレイセン
spraying on the plate, and heat at 1009 C for 5 minutes: no
green to grayish green spot appears at the Rf value of be- Add the following next to Identification:
tween 0.3 and 0.6.
Purity Arsenic <1.11>—Prepare the test solution with 0.40
g of pulverized Clematis Root according to Method 4, and
Belladonna Extract perform the test (not more than 5 ppm).

ベラドンナエキス
Cnidium Rhizome
Add the following next to Identification:
センキュウ
Purity Heavy metals <1.07>—Prepare the test solution with
1.0 g of Belladonna Extract as directed in the Extracts (4)
Add the following next to Description:
under General Rules for Preparations, and perform the test
(not more than 30 ppm). Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
pulverized Cnidium Rhizome according to Method 3, and
perform the test. Prepare the control solution with 3.0 mL
Calumba of Standard Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
コロンボ of pulverized Cnidium Rhizome according to Method 4, and
perform the test (not more than 5 ppm).
Add the following next to Identification:
Purity Arsenic <1.11>—Prepare the test solution with 0.40
g of pulverized Calumba according to Method 4, and per-
form the test (not more than 5 ppm).
1940 Crude Drugs Supplement I, JP XV

(3) Arsenic <1.11>—Prepare the test solution with 0.40 g


Powdered Cnidium Rhizome of Powdered Coptis Rhizome according to Method 4, and
perform the test (not more than 5 ppm).
センキュウ末

Change the Purity to read: Corydalis Tuber


Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
エンゴサク
Powdered Cnidium Rhizome according to Method 3, and
perform the test. Prepare the control solution with 3.0 mL
Change the Identification to read:
of Standard Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g Identification To 2 g of pulverized Corydalis Tuber add 10
of Powdered Cnidium Rhizome according to Method 4, and mL of methanol, shake for 15 minutes, filter, and use the
perform the test (not more than 5 ppm). filtrate as the sample solution. Perform the test with the
(3) Foreign matter—Under a microscope <5.01>, Pow- sample solution as directed under Thin-layer Chro-
dered Cnidium Rhizome does not contain a large quantity of matography <2.03>. Spot 10 mL of the sample solution on a
starch grains, stone cells, crystals of calcium oxalate or other plate of silica gel for thin-layer chromatography. Develop
foreign matter. with a mixture of methanol, a solution of ammonium
acetate (3 in 10) and acetic acid (100) (20:1:1) to a distance of
about 10 cm, and air-dry the plate. Examine under ultravio-
Condurango Fluidextract let light (main wavelength: 365 nm): a yellow-green fluores-
cent spot at around Rf 0.4 and a yellow fluorescent spot at
コンズランゴ流エキス around Rf 0.35 appear. When spray evenly Dragendorff's
TS for spraying on the plate, air-dry, and then spray sodium
Add the following next to Identification: nitrite TS: a brown spot appears at around Rf 0.6.
Purity Heavy metals <1.07>—Prepare the test solution with
1.0 g of Condurango Fluidextract as direct in the Fluidex-
Add the following next to Identification:
tracts (4) under General Rules for Preparations, and per- Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
form the test (not more than 30 ppm). pulverized Corydalis Tuber according to Method 3, and per-
form the test. Prepare the control solution with 3.0 mL of
Standard Lead Solution (not more than 10 ppm).
Coptis Rhizome (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of pulverized Corydalis Tuber according to Method 4, and
オウレン perform the test (not more than 5 ppm).

Add the following next to Identification:


Purity Arsenic <1.11>—Prepare the test solution with 0.40 Add the following:
g of pulverized Coptis Rhizome according to Method 4, and
perform the test (not more than 5 ppm). Powdered Corydalis Tuber
Corydalis Tuber Pulveratum
Powdered Coptis Rhizome エンゴサク末

オウレン末 Powdered Corydalis Tuber is the powder of Coryda-


lis Tuber.
Change the Purity to read: It contains not less than 0.08z of dehydrocoryda-
Purity (1) Phellodendron bark—Under a microscope line (as dehydrocorydaline nitrate), calculated on the
<5.01>, crystal cell rows or mucilage masses are not observa- basis of dried material.
ble. Stir 0.5 g of Powdered Coptis Rhizome with 2 mL of Description Powdered Corydalis Tuber occurs as a green-
water: the solution does not become gelatinous. ish yellow to grayish yellow powder. Almost odorless; taste,
(2) Curcuma—Place Powdered Coptis Rhizome on a bitter.
filter paper, drop diethyl ether on it, and allow to stand. Re- Under a microscope <5.01>, Powdered Corydalis Tuber
move the powder from the filter paper, and drop 1 drop of reveals mainly, masses of gelatinized starch or light yellow to
potassium hydroxide TS: no red-purple color develops. Un- colorless parenchymatous cells containing starch grains,
der a microscope <5.01>, Powdered Coptis Rhizome does not fragments of cork layers, light yellow stone cells, scleren-
contain gelatinized starch or secretory cells containing yel- chymatous cells, reticulate vessels, spiral vessels and ring
low-red resin. vessels; starch grains observed simple grains and 2- to 3-
Supplement I, JP XV Crude Drugs 1941

compound grains. silica gel for liquid chromatography (5 mm in particle di-


ameter).
Identification To 2 g of Powdered Corydalis Tuber add 10
Column temperature: A constant temperature of about
mL of methanol, shake for 15 minutes, filter, and use the
409C.
filtrate as the sample solution. Perform the test with the
Mobile phase: Dissolve 17.91 g of disodium hydrogen
sample solution as directed under Thin-layer Chro-
phosphate dodecahydrate in 970 mL of water, and adjust to
matography <2.03>. Spot 10 mL of the sample solution on a
pH 2.2 with phosphoric acid. Dissolve 14.05 g of sodium
plate of silica gel for thin-layer chromatography, develop the
perchlorate monohydrate in this solution, and add water to
plate with a mixture of methanol, ammonium acetate solu-
make exactly 1000 mL. Add 450 mL of acetonitrile, and dis-
tion (3 in 10) and acetic acid (100) (20:1:1) to a distance of
solve 0.20 g of sodium lauryl sulfate in this solution.
about 10 cm, and air-dry the plate. Examine under ultravio-
Flow rate: Adjust the flow rate so that the retention time
let light (main wavelength: 365 nm): a yellow-green fluores-
of dehydrocorydaline is about 24 minutes.
cent spot and a yellow fluorescent spot appear at around Rf
System suitability—
0.4 and at around Rf 0.35, respectively. Separately, spray
System performance: Dissolve 1 mg of dehydrocorydaline
evenly Dragendorff's TS for spraying on the plate, air-dry,
nitrate for component determination and 1 mg of berberine
and then spray sodium nitrite TS: a brown spot appears at
chloride in 20 mL of a mixture of water and acetonitrile
around Rf 0.6.
(20:9). When the procedure is run with 5 mL of this solution
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of under the above operating conditions, berberine and de-
Powdered Corydalis Tuber according to Method 3, and per- hydrocorydaline are eluted in this order with the resolution
form the test. Prepare the control solution with 3.0 mL of between these peaks being not less than 1.5.
Standard Lead Solution (not more than 10 ppm). System repeatability: When the test is repeated 6 times
(2) Arsenic <1.11>—Prepare the test solution with 0.4 g with 5 mL of the standard solution under the above operat-
of Powdered Corydalis Tuber according to Method 4, and ing conditions, the relative standard deviation of the peak
perform the test (not more than 5 ppm). area of dehydrocorydaline is not more than 1.5z.

Loss on drying <5.01> Not more than 15.0z.

Total ash <5.01> Not more than 3.0z. Add the following:
Component determination To about 1 g of Powdered
Corydalis Tuber, accurately weighed, add 30 mL of a mix- Crataegus Fruit
ture of methanol and dilute hydrochloric acid (3:1), heat un-
Crataegi Fructus
der a reflux condenser on a water bath for 30 minutes, and
filter after cooling. To the residue add 15 mL of the mixture サンザシ
of methanol and dilute hydrochloric acid (3:1), and proceed
in the same way as above. Combine the filtrate, add the mix- Crataegus Fruit is the pseudocarp of 1) Crataegus
ture of methanol and dilute hydrochloric acid (3:1) to make cuneata Siebold et Zuccarini or 2) Crataegus pinnatifi-
exactly 50 mL, and use this solution as the sample solution. da Bunge var. major N. E. Brown (Rosaceae) without
Separately, weigh accurately about 10 mg of de- any treatment or cut crosswise or lengthwise.
hydrocorydaline nitrate for component determination,
Description
previously dried in a desiccator (silica gel) for not less than 1
1) Crataegus cuneata Siebold et Zuccarini Nearly
hour, dissolve in the mixture of methanol and dilute
sphaerical fruits, 8 to 14 mm in diameter; externally yellow-
hydrochloric acid (3:1) to make exactly 200 mL, and use this
ish brown to grayish brown, with fine reticulated wrinkles,
solution as the standard solution. Perform the test with ex-
remained dent of 4 to 6 mm in diameter at one end, often the
actly 5 mL each of the sample solution and standard solution
base of calyx around the dent, short peduncle or scar at the
as directed under Liquid Chromatography <2.01> according
other end.True fruits, usually five loculus, often split five,
to the following conditions, and determine the peak areas, A
mericarp, 5 to 8 mm in length, light brown, usually, contain-
T and AS, of dehydrocorydaline.
ing one seed into each mericarp.
Amount (mg) of dehydrocorydaline [as dehydrocorydaline Almost odorless; taste, slightly acid.
nitrate (C22H24N2O7)] Under a microscope <5.01>, a transverse section of central
=WS×(AT/AS)×(1/4) parts reveals in the outermost layer composed of epidermis
to be covered with comparatively thick cuticle layer, cuticle
WS: Amount (mg) of dehydrocorydaline nitrate for com-
intrude into lateral cell walls of epidermis, and reveal wedge-
ponent determination
like. Cell of the epidermis or 2- to 3-layer of parenchyma-
Operating conditions— tous cells beneath these observed contents of yellowish
Detector: An ultraviolet absorption photometer (wave- brown to red brown in color followed these appeared paren-
length: 340 nm). chyma. Vascular bundles and numerous stone cells appear
Column: A stainless steel column 4.6 mm in inside di- single or gathered 2 to several cells scattered on the paren-
ameter and 15 cm in length, packed with octadecylsilanized chyma, and observed solitary crystals and rosette aggregates
1942 Crude Drugs Supplement I, JP XV

of calcium oxalate. Pericarp of true fruits composed of


mainly sclerenchymatous cells, seed covered with seed coats, Powdered Cyperus Rhizome
perisperm, endosperm, cotyledon observed inside seed
coats; sclerenchymatous cells of true fruits and cells of seed コウブシ末
coats containing solitary crystals of calcium oxalate.
2) Crataegus pinnatifida Bunge var. major N. E. Brown Change the Purity to read:
Approximate to Crataegus Fruits 1), but it is a large size,
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
17 to 23 mm in diameter, the outer surface red brown and
Powdered Cyperus Rhizome according to Method 3, and
lustrous, spot-like scars of hairs are distinct. At one end
perform the test. Prepare the control solution with 3.0 mL
remained dent, 7 to 9 mm in diameter, mericarp, 10 to 12
of Standard Lead Solution (not more than 10 ppm).
mm in length, yellowish brown in color, usually ripe seeds
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
are absent.
of Powdered Cyperus Rhizome according to Method 4, and
Odor, characteristic; taste, acid.
perform the test (not more than 5 ppm).
Under a microscope <5.01>, a transverse section of the
(3) Foreign matter—Under a microscope <5.01>, Pow-
central parts apploximate to 1), but it is a few stone cells on
dered Cyperus Rhizome does not show extremely lignified
parenchyma.
cells, except stone cells, and crystals.
Identification To 1 g of pulverized Crataegus Fruit add 5
mL of methanol, shake for 30 minutes, centrifuge, and use
the supernatant liquid as the sample solution. Separately, Dioscorea Rhizome
dissolve 1 mg of hyperoside for thin-layer chromatography
in 20 mL of methanol, and use this solution as the standard サンヤク
solution. Perform the test with these solutions as directed
under Thin-layer Chromatography <2.03>. Spot 10 mL each Add the following next to Identification:
of the sample solution and standard solution on a plate of Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
silica gel for thin-layer chromatography, develop the plate pulverized Dioscorea Rhizome according to Method 3, and
with a mixture of ethyl acetate, 2-butanone, water and for- perform the test. Prepare the control solution with 3.0 mL
mic acid (5:3:1:1) to a distance of about 10 cm, and air-dry of Standard Lead Solution (not more than 10 ppm).
the plate. Spray evenly dilute sulfuric acid on the plate, heat (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
at 1059 C for 5 minutes, and examine under ultraviolet light of pulverized Dioscorea Rhizome according to Method 4,
(main wavelength: 365 nm): one of the spot among the sever- and perform the test (not more than 5 ppm).
al spots from the sample solution has the same color tone
and Rf value with the green fluorescent spot from the stan-
dard solution. This spot disappears gradually by allowing to
cool, and appears again by heating.
Powdered Dioscorea Rhizome
Loss on drying <5.01> Not more than 17.0z. サンヤク末

Total ash <5.01> Not more than 4.0z. Add the following next to Identification:
Extract content <5.01> Dilute ethanol-soluble extract: not Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
less than 8.0z. Powdered Dioscorea Rhizome according to Method 3, and
perform the test. Prepare the control solution with 3.0 mL
of Standard Lead Solution (not more than 10 ppm).
Cyperus Rhizome (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of Powdered Dioscorea Rhizome according to Method 4,
コウブシ and perform the test (not more than 5 ppm).

Add the following next to Description:


Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of Fritillaria Bulb
pulverized Cyperus Rhizome according to Method 3, and
perform the test. Prepare the control solution with 3.0 mL バイモ
of Standard Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g Add the following next to Identification:
of pulverized Cyperus Rhizome according to Method 4, and Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
perform the test (not more than 5 ppm). pulverized Fritillaria Bulb according to Method 3, and per-
form the test. Prepare the control solution with 3.0 mL of
Standard Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
Supplement I, JP XV Crude Drugs 1943

of pulverized Fritillaria Bulb according to Method 4, and


perform the test (not more than 5 ppm). Glycyrrhiza Extract
カンゾウエキス
Gastrodia Tuber
Change the Purity to read:
テンマ
Purity (1) Heavy metals <1.07>—Prepare the test solution
with 1.0 g of Glycyrrhiza Extract as directed in the Extracts
Add the following next to Identification:
(4) under General Rules for Preparations, and perform the
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of test (not more than 30 ppm).
pulverized Gastrodia Tuber according to Method 3, and per- (2) Insoluble matter—Dissolve 2.0 g of Glycyrrhiza Ex-
form the test. Prepare the control solution with 3.0 mL of tract in 18 mL of water, and filter. To 10 mL of the filtrate
Standard Lead Solution (not more than 10 ppm). add 5 mL of ethanol (95): a clear solution results.
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of pulverized Gastrodia Tuber according to Method 4, and
perform the test (not more than 5 ppm). Crude Glycyrrhiza Extract
カンゾウ粗エキス
Gentian
Change the Purity to read:
ゲンチアナ
Purity (1) Heavy metals <1.07>—Prepare the test solution
with 1.0 g of Crude Glycyrrhiza Extract as directed in the
Add the following next to Identification:
Extracts (4) under General Rules for Preparations, and per-
Purity Arsenic <1.11>—Prepare the test solution with 0.40 form the test (not more than 30 ppm).
g of pulverized Gentian according to Method 4, and perform (2) Water-insoluble substances—Boil 5.0 g of pulverized
the test (not more than 5 ppm). Crude Glycyrrhiza Extract with 100 mL of water. After
cooling, filter the mixture through tared filter paper, wash
with water, and dry the residue at 1059C for 5 hours: the
Powdered Gentian mass of the residue is not more than 1.25 g.
(3) Foreign matter—The filtrate obtained in (2) does not
ゲンチアナ末 have a strong bitter taste.
(4) Starch—To about 1 g of pulverized Crude Glycyrrhi-
Change the Purity to read: za Extract add water to make 20 mL, shake the mixture
thoroughly, and filter. Examine the insoluble substance on
Purity (1) Arsenic <1.11>—Prepare the test solution with
the filter paper under a microscope: the residue contains no
0.40 g of Powdered Gentian according to Method 4, and
starch grains.
perform the test (not more than 5 ppm).
(2) Foreign matter—Under a microscope <5.01>, stone
cell and fiber are not observed.
Add the following:

Glehnia Root Hangekobokuto Extract


半夏厚朴湯エキス
ハマボウフウ

Add the following next to Description: Hangekobokuto Extract contains not less than 2 mg
and not more than 6 mg of magnolol, not less than
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of 4 mg (for preparation prescribed 2 g of Perilla Herb)
pulverized Glehnia Root according to Method 3, and per- or not less than 6 mg (for preparation prescribed 3 g of
form the test. Prepare the control solution with 3.0 mL of Perilla Herb) of rosmarinic acid, and not less than
Standard Lead Solution (not more than 10 ppm). 0.6 mg and not more than 2.4 mg (for preparation
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g prescribed 1 g of Ginger) or not less than 0.8 mg and
of pulverized Glehnia Root according to Method 4, and per- not more than 3.2 mg (for preparation prescribed
form the test (not more than 5 ppm). 1.3 g of Ginger) or not less than 0.9 mg and not more
than 3.6 mg (for preparation prescribed 1.5 g of Gin-
ger) of [6]-gingerol per amount specified in the
Method of preparation.
1944 Crude Drugs Supplement I, JP XV

Method of preparation Prepare a dry extract or viscous the standard solution. Perform the test with these solutions
extract as directed under Extracts, with 6 g of Pinellia as directed under Thin-layer Chromatography <2.03>. Spot 5
Tuber, 5 g of Poria Sclerotium, 3 g of Magnolia Bark, 2 g of mL each of the sample solution and standard solution on a
Perilla Herb and 1 g of Ginger, or with 6 g of Pinellia Tuber, plate of silica gel for thin-layer chromatography. Develop
5 g of Poria Sclerotium, 3 g of Magnolia Bark, 3 g of Perilla the plate with a mixture of hexane and acetone (2:1) to a dis-
Herb and 1 g of Ginger, or with 6 g of Pinellia Tuber, 5 g of tance of about 10 cm, and air-dry the plate. Spray evenly 4-
Poria Sclerotium, 3 g of Magnolia Bark, 2 g of Perilla Herb dimethylaminobenzaldehyde TS for spraying on the plate,
and 1.3 g of Ginger, or with 6 g of Pinellia Tuber, 5 g of heat at 1059 C for 5 minutes, and allow to cool: one of the
Poria Sclerotium, 3 g of Magnolia Bark, 2 g of Perilla Herb spot among the several spots from the sample solution has
and 1.5 g of Ginger. the same color tone and Rf value with the blue-green spot
from the standard solution (Ginger).
Description Hangekobokuto Extract is a light brown to
black-brown, powder or viscous extract. It has a characteris- Purity (1) Heavy metals <1.07>—Prepare the test solution
tic odor and has a bitter and astringent taste first then pun- with 1.0 g of the dry extract (or an amount of the viscous ex-
gent later. tract, equivalent to 1.0 g of the dried substance) as directed
in the Extracts (4) under General Rules for Preparations,
Identification (1) Shake 1.0 g of the dry extract (or 3.0 g
and perform the test (not more than 30 ppm).
of the viscous extract) with 10 mL of water, add 25 mL of
(2) Arsenic <1.11>—Prepare the test solution with 0.67 g
diethyl ether, and shake. Take the diethyl ether layer,
of the dry extract (or an amount of the viscous extract,
evaporate the layer under reduced pressure, dissolve the
equivalent to 0.67 g of the dried substance) according to
residue in 2 mL of diethyl ether, and use this solution as the
Method 3, and perform the test (not more than 3 ppm).
sample solution. Separately, dissolve 1 mg of magnolol for
thin-layer chromatography in 1 mL of methanol, and use Loss on drying <2.41> The dry extract: Not more than 11.0
this solution as the standard solution. Perform the test with z (1 g, 1059C, 5 hours).
these solutions as directed under Thin-layer Chro- The viscous extract: Not more than 66.7z (1 g, 1059C, 5
matography <2.03>. Spot 5 mL each of the sample solution hours).
and standard solution on a plate of silica gel with fluorescent
Total ash <5.01> Not more than 14.0z, calculated on the
indicator for thin-layer chromatography. Develop the plate
dried basis.
with a mixture of ethyl acetate and hexane (1:1) to a distance
of about 10 cm, and air-dry the plate. Examine under ultrav- Assay (1) Magnolol—Weigh accurately about 0.5 g of
iolet light (main wavelength: 254 nm): one of the spot among the dry extract (or an amount of the viscous extract, equiva-
the several spots from the sample solution has the same color lent to about 0.5 g of the dried substance), add exactly 50
tone and Rf value with the dark purple spot from the stan- mL of diluted methanol (7 in 10), shake for 15 minutes,
dard solution (Magnolia Bark). filter, and use the filtrate as the sample solution. Separately,
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous weigh accurately about 10 mg of magnolol for component
extract) with 10 mL of 0.1 mol/L hydrochloric acid TS, add determination, previously dried in a desiccator (silica gel) for
25 mL of diethyl ether, and shake. Take the diethyl ether not less than 1 hour, and dissolve in diluted methanol (7 in
layer, evaporate the layer under reduced pressure, dissolve 10) to make exactly 100 mL. Pipet 5 mL of this solution, add
the residue in 1 mL of methanol, and use this solution as the diluted methanol (7 in 10) to make exactly 20 mL, and use
sample solution. Separately, dissolve 1 mg of rosmarinic this solution as the standard solution. Perform the test with
acid for thin-layer chromatography in 1 mL of methanol, exactly 10 mL each of the sample solution and standard solu-
and use this solution as the standard solution. Perform the tion as directed under Liquid Chromatography <2.01> ac-
test with these solutions as directed under Thin-layer Chro- cording to the following conditions, and determine the peak
matography <2.03>. Spot 5 mL each of the sample solution areas, AT and AS, of magnolol.
and standard solution on a plate of silica gel for thin-layer
Amount (mg) of magnolol=WS×(AT/AS)×(1/8)
chromatography. Develop the plate with a mixture of ethyl
acetate, water and formic acid (60:1:1) to a distance of about WS: Amount (mg) of magnolol for component determina-
10 cm, and air-dry the plate. Spray evenly iron (III) chloride tion
TS on the plate: one of the spot among the several spots
Operating conditions—
from the sample solution has the same color tone and Rf
Detector: An ultraviolet absorption photometer (wave-
value with the dark purple spot from the standard solution
length: 289 nm).
(Perilla Herb).
Column: A stainless steel column 4.6 mm in inside di-
(3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
ameter and 15 cm in length, packed with octadecylsilanized
extract) with 10 mL of water, add 25 mL of diethyl ether,
silica gel for liquid chromatography (5 mm in particle di-
and shake. Take the diethyl ether layer, evaporate the layer
ameter).
under reduced pressure, dissolve the residue in 2 mL of
Column temperature: A constant temperature of about
diethyl ether, and use this solution as the sample solution.
409C.
Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro-
Mobile phase: A mixture of water, acetonitrile and acetic
matography in 1 mL of methanol, and use this solution as
Supplement I, JP XV Crude Drugs 1945

acid (100) (50:50:1). diluted methanol (7 in 10), shake for 15 minutes, filter, and
Flow rate: 1.0 mL per minute. (the retention time of mag- use the filtrate as the sample solution. Separately, weigh ac-
nolol is about 15 minutes.) curately about 10 mg of [6]-gingerol for component determi-
System suitability— nation, dissolve in diluted methanol (7 in 10) to make exactly
System performance: Dissolve 1 mg each of magnolol for 100 mL. Pipet 5 mL of this solution, add methanol to make
component determination and honokiol in diluted methanol exactly 50 mL, and use this solution as the standard solu-
(7 in 10) to make 10 mL. When the procedure is run with 10 tion. Perform the test with exactly 10 mL each of the sample
mL of this solution under the above operating conditions, solution and standard solution as directed under Liquid
honokiol and magnolol are eluted in this order with the reso- Chromatography <2.01> according to the following condi-
lution between these peaks being not less than 2.5. tions, and determine the peak areas, AT and AS, of [6]-gin-
System repeatability: When the test is repeated 6 times gerol.
with 10 mL of the standard solution under the above operat-
Amount (mg) of [6]-gingerol=WS×(AT/AS)×(1/20)
ing conditions, the relative standard deviation of the peak
area of magnolol is not more than 1.5z. WS: Amount (mg) of [6]-gingerol for component determi-
(2) Rosmarinic acid—Weigh accurately about 0.5 g of nation
the dry extract (or an amount of the viscous extract, equiva-
Operating conditions—
lent to about 0.5 g of the dried substance), add exactly 50
Detector: An ultraviolet absorption photometer (wave-
mL of diluted methanol (7 in 10), shake for 15 minutes,
length: 282 nm).
filter, and use the filtrate as the sample solution. Separately,
Column: A stainless steel column 4.6 mm in inside di-
weigh accurately about 10 mg of rosmarinic acid for compo-
ameter and 15 cm in length, packed with octadecylsilanized
nent determination, dissolve in diluted methanol (7 in 10) to
silica gel for liquid chromatography (5 mm in particle di-
make exactly 200 mL, and use this solution as the standard
ameter).
solution. Perform the test with exactly 10 mL each of the
Column temperature: A constant temperature of about
sample solution and standard solution as directed under
309C.
Liquid Chromatography <2.01> according to the following
Mobile phase: A mixture of water, acetonitrile and phos-
conditions, and determine the peak areas, AT and AS, of
phoric acid (620:380:1).
rosmarinic acid.
Flow rate: 1.0 mL per minute. (the retention time of [6]-
Amount (mg) of rosmarinic acid =WS×(AT/AS)×(1/4) gingerol is about 15 minutes.)
System suitability—
WS: Amount (mg) of rosmarinic acid for component
System performance: When the procedure is run with 10
determination
mL of the standard solution under the above operating con-
Operating conditions— ditions, the number of theoretical plates and the symmetry
Detector: An ultraviolet absorption photometer (wave- factor of the peak of [6]-gingerol are not less than 5000 and
length: 330 nm). not more than 1.5, respectively.
Column: A stainless steel column 4.6 mm in inside di- System repeatability: When the test is repeated 6 times
ameter and 15 cm in length, packed with octadecylsilanized with 10 mL of the standard solution under the above operat-
silica gel for liquid chromatography (5 mm in particle di- ing conditions, the relative standard deviation of the peak
ameter). area of [6]-gingerol is not more than 1.5z.
Column temperature: A constant temperature of about
Containers and storage Containers—Tight containers.
309C.
Mobile phase: A mixture of water, acetonitrile and phos-
phoric acid (800:200:1).
Change to read:
Flow rate: 1.0 mL per minute. (the retention time of ros-
marinic acid is about 11 minutes.)
System suitability— Hochuekkito Extract
System performance: When the procedure is run with 10
補中益気湯エキス
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
Hochuekkito Extract contains not less than 16 mg
factor of the peak of rosmarinic acid are not less than 5000
and not more than 48 mg of hesperidin, not less than
and not more than 1.5, respectively.
0.3 mg and not more than 1.2 mg (for preparation
System repeatability: When the test is repeated 6 times
prescribed 1 g of Bupleurum Root) or not less than 0.6
with 10 mL of the standard solution under the above operat-
mg and not more than 2.4 mg (for preparation
ing conditions, the relative standard deviation of the peak
prescribed 2 g of Bupleurum Root) of saikosaponin
area of rosmarinic acid is not more than 1.5z.
b2, and not less than 12 mg and not more than 36 mg
(3) [6]-Gingerol—Weigh accurately about 0.5 g of the
of glycyrrhizic acid (C42H62O16: 822.93) per a dried ex-
dry extract (or an amount of the viscous extract, equivalent
tract prepared as directed in the Method of prepara-
to about 0.5 g of the dried substance), add exactly 50 mL of
tion.
1946 Crude Drugs Supplement I, JP XV

Method of preparation Prepare a dry extract or viscous on the plate, heat at 1059C for 5 minutes, and allow to cool:
extract as directed under Extracts, with 4 g of Ginseng, 4 g one of the spot among the sevelral spots from the sample so-
of Atractylodes Rhizome or Atractylodes Lancea Rhizome, lution has the same color tone and Rf value with the red spot
4 g of Astragalus Root, 3 g of Japanese Angelica Root, 2 g from the standard solution (Atractylodes Rhizome).
of Citrus Unshiu Peel, 2 g of Jujube, 2 g of Bupleurum (3) For preparation prescribed Atractylodes Lancea
Root, 1.5 g of Glycyrrhiza, 0.5 g of Ginger and 1 g of Rhizome—To 2.0 g of the dry extract (or 6.0 g of the viscous
Cimicifuga Rhizome, or with 4 g of Ginseng, 4 g of Atrac- extract) add 10 mL of water, shake, then add 25 mL of
tylodes Rhizome or Atractylodes Lancea Rhizome, 4 g of hexane, shake, and take the hexane layer. To the hexane
Astragalus Root, 3 g of Japanese Angelica Root, 2 g of layer add anhydrous sodium sulfate to dry, filter, evaporate
Citrus Unshiu Peel, 2 g of Jujube, 1 g of Bupleurum Root, the filtrate under reduced pressure, add 2 mL of hexane to
1.5 g of Glycyrrhiza, 0.5 g of Ginger and 0.5 g of Cimicifuga the residue, and use this solution as the sample solution. Per-
Rhizome, or with 4 g of Ginseng, 4 g of Atractylodes Rhi- form the test with the sample solution as directed under
zome, 3 g of Astragalus Root, 3 g of Japanese Angelica Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Root, 2 g of Citrus Unshiu Peel, 2 g of Jujube, 2 g of ple solution on a plate of silica gel with fluorescent indicator
Bupleurum Root, 1.5 g of Glycyrrhiza, 0.5 g of Ginger and 1 for thin-layer chromatography. Develop the plate with a
g of Cimicifuga Rhizome, or with 4 g of Ginseng, 4 g of mixture of hexane and acetone (7:1) to a distance of about
Atractylodes Rhizome, 4 g of Astragalus Root, 3 g of 10 cm, and air-dry the plate. Examine under ultraviolet light
Japanese Angelica Root, 2 g of Citrus Unshiu Peel, 2 g of (main wavelength: 254 nm): a dark purple spot appears
Jujube, 1 g of Bupleurum Root, 1.5 g of Glycyrrhiza, 0.5 g around Rf 0.4, which shows a greenish brown color after
of Processed Ginger and 0.5 g of Cimicifuga Rhizome. spraying 4-dimethylaminobenzaldehyde TS for spraying,
heating at 1059 C for 5 minutes and allowing to cool (Atrac-
Description Hochuekkito Extract occurs as a light brown
tylodes Lancea Rhizome).
to black-brown powder or viscous extract. It has a slight
(4) To 3.0 g of the dry extract (or 9.0 g of the viscous
odor, and a sweet and bitter taste.
extract) add 40 mL of a solution of potassium hydroxide in
Identification (1) To 2.0 g of the dry extract (or 6.0 g of methanol (1 in 50), shake for 15 minutes, centrifuge, and
the viscous extract) add 30 mL of water, shake, then add 50 evaporate the supernatant liquid under reduced pressure.
mL of 1-butanol, and shake. Take the 1-butanol layer, Add 30 mL of water to the residue, then add 20 mL of
evaporate the layer under reduced pressure, add 3 mL of diethyl ether, shake, and take the water layer. To the water
methanol to the residue, and use this solution as the sample layer add 20 mL of 1-butanol, shake, and take the 1-butanol
solution. Separately, dissolve 1 mg of Ginsenoside Rb1 layer. To the 1-butanol layer add 20 mL of water, shake,
Reference Standard in 1 mL of methanol, and use this solu- take the 1-butanol layer, evaporate the layer under reduced
tion as the standard solution. Perform the test with these so- pressure, add 1 mL of methanol to the residue, and use this
lutions as directed under Thin-layer Chromatography solution as the sample solution. Separately, dissolve 1 mg of
<2.03>. Spot 5 mL each of the sample solution and standard astragaloside IV for thin-layer chromatography in 1 mL of
solution on a plate of silica gel for thin-layer chro- methanol, and use this solution as the standard solution.
matography, develop the plate with a mixture of ethyl Perform the test with these solutions as directed under Thin-
acetate, 1-propanol, water and acetic acid (100) (7:5:4:1) to a layer Chromatography <2.03>. Spot 5 mL each of the sample
distance of about 10 cm, and air-dry the plate. Spray evenly solution and standard solution on a plate of octadecyl-
vanillin-sulfuric acid TS on the plate, heat at 1059 C for 5 silanized silica gel for thin-layer chromatography. Develop
minutes, and allow to cool: one of the spot among the sever- the plate with a mixture of methanol, water, 1-butanol and
al spots from the sample solution has the same color tone acetic acid (100) (60:30:10:1) to a distance of about 10 cm,
and Rf value with the purple spot from the standard solution and air-dry the plate. Spray evenly 4-dimethylaminobenzal-
(Ginseng). dehyde TS for spraying on the plate, and heat at 1059C for 5
(2) For preparation prescribed Atractylodes Rhizome— minutes: one of the spot among the several spots from the
To 3.0 g of the dry extract (or 9.0 g of the viscous extract) sample solution has the same color tone and Rf value with
add 30 mL of water, shake, then add 50 mL of diethyl ether, the red-brown spot from the standard solution (Astragalus
shake, and take the diethyl ether layer. Evaporate the layer Root).
under reduced pressure, add 1 mL of diethyl ether to the (5) To 3.0 g of the dry extract (or 9.0 g of the viscous ex-
residue, and use this solution as the sample solution. tract) add 30 mL of water, shake, then add 50 mL of diethyl
Separately, dissolve 1 mg of atractylenolide III for thin-layer ether, shake, and take the diethyl ether layer. Evaporate the
chromatography in 1 mL of methanol, and use this solution layer under reduced pressure, add 1 mL of diethyl ether to
as the standard solution. Perform the test with these solu- the residue, and use this solution as the sample solution.
tions as directed under Thin-layer Chromatography <2.03>. Separately, dissolve 1 mg of (Z)-ligustilide for thin-layer
Spot 5 mL of the sample solution and 10 mL of the standard chromatography in 10 mL of methanol, and use this solu-
solution on a plate of silica gel for thin-layer chro- tion as the standard solution. Perform the test with these so-
matography. Develop the plate with a mixture of ethyl lutions as directed under Thin-layer Chromatography
acetate and hexane (1:1) to a distance of about 10 cm, and <2.03>. Spot 10 mL each of the sample solution and standard
air-dry the plate. Spray evenly 1-naphthol-sulfuric acid TS solution on a plate of silica gel for thin-layer chro-
Supplement I, JP XV Crude Drugs 1947

matography. Develop the plate with a mixture of ethyl 1059 C for 5 minutes: one of the spot among the several spots
acetate and hexane (1:1) to a distance of about 10 cm, and from the sample solution has the same color tone and Rf
air-dry the plate. Examine under ultraviolet light (main value with the yellow-brown spot from the standard solution
wavelength: 365 nm): one of the spot among the several (Glycyrrhiza).
spots from the sample solution has the same color tone and (9) For preparation prescribed Ginger—To 3.0 g of the
Rf value with the bluish white fluorescent spot from the dry extract (or 9.0 g of the viscous extract) add 30 mL of
standard solution (Japanese Angelica Root). water, shake, then add 50 mL of diethyl ether, shake, and
(6) To 2.0 g of the dry extract (or 6.0 g of the viscous take the diethyl ether layer. Evaporate the layer under
extract) add 30 mL of water, shake, then add 50 mL of 1- reduced pressure, add 1 mL of diethyl ether to the residue,
butanol, shake, and take the 1-butanol layer. Evaporate the and use this solution as the sample solution. Separately, dis-
layer under reduced pressure, add 3 mL of methanol to the solve 1 mg of [6]-gingerol for thin-layer chromatography in
residue, and use this solution as the sample solution. 1 mL of methanol, and use this solution as the standard so-
Separately, dissolve 1 mg of hesperidin for thin-layer chro- lution. Perform the test with these solutions as directed un-
matography in 2 mL of methanol, and use this solution as der Thin-layer Chromatography <2.03>. Spot 5 mL each of
the standard solution. Perform the test with these solutions the sample solution and standard solution on a plate of silica
as directed under Thin-layer Chromatography <2.03>. Spot 2 gel for thin-layer chromatography. Develop the plate with a
mL of the sample solution and 20 mL of the standard solution mixture of ethyl acetate and hexane (1:1) to a distance of
on a plate of silica gel for thin-layer chromatography. De- about 10 cm, and air-dry the plate. Spray evenly 4-
velop the plate with a mixture of ethyl acetate, acetone, dimethylaminobenzaldehyde TS for spraying on the plate,
water and acetic acid (100) (10:6:3:1) to a distance of about heat at 1059 C for 5 minutes, and allow to cool: one of the
10 cm, and air-dry the plate. Spray evenly 2,6-dibromo-N- spot among the several spots from the sample solution has
chloro-1,4-benzoquinone monoimine TS on the plate, and the same color tone and Rf value with the blue-green spot
expose to ammonia vapor: one of the spot among the several from the standard solution (Ginger).
spots from the sample solution has the same color tone and (10) For preparation prescribed Processed Ginger—Put
Rf value with the blue spot from the standard solution 10 g of the dry extract (or 30 g of the viscous extract) in a
(Citrus Unshiu Peel). 300-mL hard-glass flask, add 100 mL of water and 1 mL of
(7) To 2.0 g of the dry extract (or 6.0 g of the viscous silicone resin, connect an apparatus for essential oil determi-
extract) add 30 mL of water, shake, then add 50 mL of 1- nation, and heat to boil under a reflux condenser. The
butanol, shake, and take the 1-butanol layer. Evaporate the graduated tube of the apparatus is to be previously filled
layer under reduced pressure, add 3 mL of methanol to the with water to the standard line, and 2 mL of hexane is added
residue, and use this solution as the sample solution. to the graduated tube. After heating under reflux for about
Separately, dissolve 1 mg of saikosaponin b2 for thin-layer 1 hour, separate the hexane layer, and use this as the sample
chromatography in 1 mL of methanol, and use this solution solution. Separately, dissolve 1 mg of [6]-shogaol for thin-
as the standard solution. Perform the test with these layer chromatography in 1 mL of methanol, and use this so-
solutions as directed under Thin-layer Chromatography lution as the standard solution. Perform the test with these
<2.03>. Spot 5 mL of the sample solution and 2 mL of the solutions as directed under Thin-layer Chromatography
standard solution on a plate of silica gel for thin-layer <2.03>. Spot 60 mL of the sample solution and 10 mL of the
chromatography. Develop the plate with a mixture of ethyl standard solution on a plate of silica gel for thin-layer chro-
acetate, ethanol (99.5) and water (8:2:1) to a distance of matography. Develop the plate with a mixture of cyclo-
about 10 cm, and air-dry the plate. Spray evenly 4- hexane and ethyl acetate (2:1) to a distance of about 10 cm,
dimethylaminobenzaldehyde TS on the plate: one of the spot and air-dry the plate. Spray evenly 4-dimethylaminobenzal-
among the several spots from the sample solution has the dehyde TS for spraying on the plate, heat at 1059C for 5
same color tone and Rf value with the red spot from the minutes, and allow to cool: one of the spot among the sever-
standard solution (Bupleurum Root). al spots from the sample solution has the same color tone
(8) To 2.0 g of the dry extract (or 6.0 g of the viscous ex- and Rf value with the blue-green spot from the standard
tract) add 30 mL of water, shake, then add 50 mL of 1- solution (Processed Ginger).
butanol, and take the 1-butanol layer. Evaporate the layer (11) To 2.0 g of the dry extract (or 6.0 g of the viscous
under reduced pressure, add 3 mL of methanol to the extract) add 30 mL of water, shake, then add 50 mL of 1-
residue, and use this solution as the sample solution. butanol, and take the 1-butanol layer. Evaporate the layer
Separately, dissolve 1 mg of liquiritin for thin-layer chro- under reduced pressure, add 3 mL of methanol to the
matography in 1 mL of methanol, and use this solution as residue, and use this solution as the sample solution. Use 3-
the standard solution. Perform the test with these solutions (3-hydroxy-4-methoxyphenyl)-2-(E)-propenic acid-(E)-ferul-
as directed under Thin-layer Chromatography <2.03>. Spot ic acid TS for thin-layer chromatography as the standard so-
5 mL each of the sample solution and standard solution on a lution. Perform the test with these solutions as directed un-
plate of silica gel for thin-layer chromatography. Develop der Thin-layer Chromatography <2.03>. Spot 5 mL of the
the plate with a mixture of ethyl acetate, methanol and water sample solution and 2 mL of the standard solution on a plate
(20:3:2) to a distance of about 10 cm, and air-dry the plate. of silica gel for thin-layer chromatography. Develop the
Spray evenly dilute sulfuric acid on the plate, and heat at plate with a mixture of ethyl acetate, acetone and water
1948 Crude Drugs Supplement I, JP XV

(20:12:3) to a distance of about 10 cm, and air-dry the plate. System suitability—
Spray evenly sulfuric acid on the plate, heat at 1059 C for 5 System performance: Dissolve 1 mg each of hesperidin for
minutes, and examine under ultraviolet light (main component determination and naringin for thin-layer chro-
wavelength: 365 nm): one of the spot among the several matography in diluted methanol (1 in 2) to make 100 mL.
spots from the sample solution has the same color tone and When the procedure is run with 10 mL of this solution under
Rf value with the yellow fluorescent spot from the standard the above operating conditions, naringin and hesperidin are
solution (Cimicifuga Rhizome). eluted in this order with the resolution between these peaks
being not less than 1.5.
Purity (1) Heavy metals <1.07>—Prepare the test solution
System repeatability: When the test is repeated 6 times
with 1.0 g of the dry extract (or an amount of the viscous ex-
with 10 mL of the standard solution under the above operat-
tract, equivalent to 1.0 g of the dried substance) as directed
ing conditions, the relative standard deviation of the peak
in the Extracts (4) under General Rules for Preparations,
area of hespiridin is not more than 1.5z.
and perform the test (not more than 30 ppm).
(2) Saikosaponin b2—Weigh accurately about 0.5 g of
(2) Arsenic <1.11>—Prepare the test solution with 0.67 g
the dry extract (or an amount of the viscous extract, equiva-
of the dry extract (or an amount of the viscous extract,
lent to about 0.5 g of the dried substance), add exactly 50
equivalent to 0.67 g of the dried substance) according to
mL of diluted methanol (1 in 2), shake for 15 minutes, filter,
Method 3, and perform the test (not more than 3 ppm).
and use the filtrate as the sample solution. Separately, weigh
Loss on drying <2.41> The dry extract: Not more than 11.5 accurately about 10 mg of saikosaponin b2 for component
z (1 g, 1059C, 5 hours). determination, previously dried in a desiccator (silica gel) for
The viscous extract: Not more than 66.7z (1g, 1059C, 5 not less than 24 hours, and dissolve in diluted methanol (1 in
hours). 2) to make exactly 100 mL. Pipet 10 mL of this solution, add
diluted methanol (1 in 2) to make exactly 100 mL, and use
Total ash <5.01> Not more than 9.0z, calculated on the
this solution as the standard solution. Perform the test with
dried basis.
exactly 10 mL each of the sample solution and standard solu-
Assay (1) Hesperidin—Weigh accurately about 0.1 g of tion as directed under Liquid Chromatography <2.01> ac-
the dry extract (or an amount of the viscous extract, equiva- cording to the following conditions, and determine the peak
lent to about 0.1 g of the dried substance), add exactly 50 areas, AT and AS, of saikosaponin b2.
mL of diluted tetrahydrofuran (1 in 4), shake for 30
Amount (mg) of saikosaponin b2=WS×(AT/AS)×(1/20)
minutes, centrifuge, and use the supernatant liquid as the
sample solution. Separately, weigh accurately about 10 mg WS: Amount (mg) of saikosaponin b2 for component de-
of hesperidin for component determination, previously termination
dried in a desiccator (silica gel) for not less than 24 hours,
Operating conditions—
and dissolve in methanol to make exactly 100 mL. Pipet 10
Detector: An ultraviolet absorption photometer (wave-
mL of this solution, add diluted tetrahydrofuran (1 in 4) to
length: 254 nm).
make exactly 100 mL, and use this solution as the standard
Column: A stainless steel column 4.6 mm in inside di-
solution. Perform the test with exactly 10 mL each of the
ameter and 15 cm in length, packed with octadecylsilanized
sample solution and standard solution as directed under
silica gel for liquid chromatography (5 mm in particle di-
Liquid Chromatography <2.01> according to the following
ameter).
conditions, and determine the peak areas, AT and AS, of
Column temperature: A constant temperature of about
hesperidin.
409C.
Amount (mg) of hesperidin=WS×(AT/AS)×(1/20) Mobile phase: A mixture of 0.05 mol/L sodium dihydro-
gen phosphate TS and acetonitrile (5:3).
WS: Amount (mg) of hesperidin for component determi-
Flow rate: 1.0 mL/min. (the retention time of saikosapo-
nation
nin b2 is about 12 minutes.)
Operating conditions— System suitability—
Detector: An ultraviolet absorption photometer (wave- System performance: When the procedure is run with 10
length: 285 nm). mL of the standard solution under the above operating con-
Column: A stainless steel column 4.6 mm in inside di- ditions, the number of theoretical plates and the symmetry
ameter and 15 cm in length, packed with octadecylsilanized factor of the peak of saikosaponin b2 are not less than 5000
silica gel for liquid chromatography (5 mm in particle di- and not more than 1.5, respectively.
ameter). System repeatability: When the test is repeated 6 times
Column temperature: A constant temperature of about with 10 mL of the standard solution under the above operat-
409C. ing conditions, the relative standard deviation of the peak
Mobile phase: A mixture of water, acetonitrile and acetic area of saikosaponin b2 is not more than 1.5z.
acid (100) (82:18:1). (3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
Flow rate: 1.0 mL/min. (the retention time of hesperidin the dry extract (or an amount of the viscous extract, equiva-
is about 15 minutes.) lent to about 0.5 g of the dried substance), add exactly 50
Supplement I, JP XV Crude Drugs 1949

mL of diluted methanol (1 in 2), shake for 15 minutes, filter, (4) Foreign matter <5.01>—The amount of foreign mat-
and use the filtrate as the sample solution. Separately, weigh ter other than rootlets and scaly leaves is not more than
accurately about 10 mg of Glycyrrhizic Acid Reference Stan- 1.0z.
dard (separately determine the water), 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 Ipecac
10 mL each of the sample solution and standard solution as
directed under Liquid Chromatography <2.01> according to トコン
the following conditions, and determine the peak areas, AT
and AS, of glycyrrhizic acid. Add the following next to Identification:
Amount (mg) of glycyrrhizic acid (C42H62O16) Purity Arsenic <1.11>—Prepare the test solution with 0.40
=WS×(AT/AS)×(1/2) g of pulverized Ipecac according to Method 4, and perform
the test (not more than 5 ppm).
WS: Amount (mg) of Glycyrrhizic Acid Reference Stan-
dard, calculated on the anhydrous basis

Operating conditions— Powdered Ipecac


Detector: An ultraviolet absorption photometer (wave-
length: 254 nm). トコン末
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized Change the Purity to read:
silica gel for liquid chromatography (5 mm in particle di- Purity (1) Arsenic <1.11>—Prepare the test solution with
ameter). 0.40 g of Powdered Ipecac according to Method 4, and per-
Column temperature: A constant temperature of about form the test (not more than 5 ppm).
409C. (2) Foreign matter—Under a microscope <5.01>, groups
Mobile phase: A mixture of diluted acetic acid (31) (1 in of stone cells and thick-walled fibers are not observed.
15) and acetonitrile (13:7).
Flow rate: 1.0 mL/min. (the retention time of glycyrrhizic
acid is about 12 minutes.)
System suitability—
Japanese Gentian
System performance: When the procedure is run with 10 リュウタン
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry Add the following next to Identification:
factor of the peak of glycyrrhizic acid are not less than 5000
and not more than 1.5, respectively. Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
System repeatability: When the test is repeated 6 times pulverized Japanese Gentian according to Method 3, and
with 10 mL of the standard solution under the above operat- perform the test. Prepare the control solution with 3.0 mL
ing conditions, the relative standard deviation of the peak of Standard Lead Solution (not more than 10 ppm).
area of glycyrrhizic acid is not more than 1.5z. (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of pulverized Japanese Gentian according to Method 4, and
Containers and storage Containers—Tight containers. perform the test (not more than 5 ppm).

Imperata Rhizome Powdered Japanese Gentian


ボウコン リュウタン末

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


Purity (1) Rootlet and scaly leaf—The amount of the Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
rootlets and scaly leaves contained in Imperata Rhizome is Powdered Japanese Gentian according to Method 3, and
not more than 3.0z. perform the test. Prepare the control solution with 3.0 mL
(2) Heavy metals <1.07>—Proceed with 3.0 g of pulver- of Standard Lead Solution (not more than 10 ppm).
ized Imperata Rhizome according to Method 3, and perform (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
the test. Prepare the control solution with 3.0 mL of Stan- of Powdered Japanese Gentian according to Method 4, and
dard Lead Solution (not more than 10 ppm). perform the test (not more than 5 ppm).
(3) Arsenic <1.11>—Prepare the test solution with 0.40 g (3) Foreign matter—Under a microscope <5.01>, Pow-
of pulverized Imperata Rhizome according to Method 4, and dered Japanese Gentian usually reveals no stone cells and
perform the test (not more than 5 ppm). fibers. No starch grains; if any, very few.
1950 Crude Drugs Supplement I, JP XV

Identification (1) To 1.0 g of the dry extract (or 3.0 g of


Japanese Valerian the viscous extract) add 10 mL of water, shake, then add 10
mL of 1-butanol, shake, centrifuge, and use the supernatant
カノコソウ liquid as the sample solution. Separately, dissolve 1 mg of
Puerarin Reference Standard in 1 mL of methanol, and use
Add the following next to Description: this solution as the standard solution. Perform the test with
these solutions as directed under Thin-layer Chromato-
Purity Arsenic <1.11>—Prepare the test solution with 0.40
graphy <2.03>. Spot 5 mL each of the sample solution and
g of pulverized Japanese Valerian according to Method 4,
standard solution on a plate of silica gel for thin-layer chro-
and perform the test (not more than 5 ppm).
matography. Develop the plate with a mixture of ethyl
acetate, methanol and water (20:3:2) to a distance of about
10 cm, and air-dry the plate. Examine under ultraviolet light
Powdered Japanese Valerian (main wavelength: 365 nm): one of the spot among the sever-
カノコソウ末 al spots from the sample solution has the same color tone
and Rf value with the bluish white fluorescent spot from the
standard solution (Pueraria Root).
Add the following next to Description:
(2) To 1.0 g of the dry extract (or 3.0 g of the viscous ex-
Purity Arsenic <1.11>—Prepare the test solution with 0.40 tract) add 10 mL of water, shake, then add 10 mL of 1-
g of Powdered Japanese Valerian according to Method 4, butanol, shake, centrifuge, and use the supernatant liquid as
and perform the test (not more than 5 ppm). the sample solution. Separately, dissolve 1 mg of ephedrine
hydrochloride in 1 mL of methanol, and use this solution as
the standard solution. Perform the test with these solutions
Kakkonto Extract as directed under Thin-layer Chromatography <2.03>. Spot 5
mL each of the sample solution and standard solution on a
葛根湯エキス plate of silica gel for thin-layer chromatography. Develop
the plate with a mixture of 1-butanol, water and acetic acid
Change to read: (100) (7:2:1) to a distance of about 10 cm, and air-dry the
plate. Spray evenly ninhydrin TS on the plate, and heat at
Kakkonto Extract contains not less than 9 mg and 1059 C for 5 minutes: one of the spot among the several spots
not more than 27 mg (for preparation prescribed 3 g from the sample solution has the same color tone and Rf
of Ephedra Herb) or not less than 12 mg and not more value with the red-purple spot from the standard solution
than 36 mg (for preparation prescribed 4 g of Ephedra (Ephedra Herb).
Herb) of total alkaloids [ephedrine (C10H15NO: (3) Put 10 g of the dry extract (or 30 g of the viscous ex-
165.23) and pseudoephedrine (C10H15NO: 165.23)], tract) in a 300-mL hard-glass flask, add 100 mL of water and
not less than 14 mg and not less than 42 mg (for prepa- 1 mL of silicone resin, connect the apparatus for essential oil
ration prescribed 2 g of Peony Root) or not less than determination, and heat to boil under a reflux condenser.
21 mg and not more than 63 mg (for preparation The graduated tube of the apparatus is to be previously filled
prescribed 3 g of Peony Root) of peoniflorin with water to the standard line, and 2 mL of hexane is added
(C23H28O11: 480.46), and not less than 19 mg and not to the graduated tube. After heating under reflux for 1 hour,
more than 57 mg of glycyrrhizic acid (C42H62O16: separate the hexane layer, and use the layer as the sample so-
822.93) per the extract prepared as directed in the lution. Separately, dissolve 1 mg of (E)-cinnamaldehyde for
Method of preparation. thin-layer chromatography in 1 mL of methanol, and use
Method of preparation Prepare a dry extract or viscous ex- this solution as the standard solution. Perform the test with
tract as directed under Extracts, with 8 g of Pueraria Root, 4 these solutions as directed under Thin-layer Chromato-
g of Ephedra Herb, 4 g of Jujube, 3 g of Cinnamon Bark, 3 graphy <2.03>. Spot 20 mL of the sample solution and 2 mL
g of Peony Root, 2 g of Glycyrrhiza and 1 g of Ginger, or of the standard solution on a plate of silica gel for thin-layer
with 4 g of Pueraria Root, 4 g of Ephedra Herb, 3 g of Ju- chromatography. Develop the plate with a mixture of
jube, 2 g of Cinnamon Bark, 2 g of Peony Root, 2 g of hexane and ethyl acetate (2:1) to a distance of about 10 cm,
Glycyrrhiza and 1 g of Ginger, or with 4 g of Pueraria Root, and air-dry the plate. Spray evenly 2,4-dinitrophenylhydra-
3 g of Ephedra Herb, 3 g of Jujube, 2 g of Cinnamon Bark, zine TS on the plate: one of the spot among the several spots
2 g of Peony Root, 2 g of Glycyrrhiza and 1 g of Ginger, or from the sample solution has the same color tone and Rf
with 4 g of Pueraria Root, 3 g of Ephedra Herb, 3 g of Ju- value with the yellow-orange spot from the standard solu-
jube, 2 g of Cinnamon Bark, 2 g of Peony Root, 2 g of tion (Cinnamon Bark).
Glycyrrhiza and 2 g of Ginger. (4) To 1.0 g of the dry extract (or 3.0 g of the viscous
extract) add 10 mL of water, shake, then add 10 mL of 1-
Description Kakkonto Extract occurs as a light brown to butanol, shake, centrifuge, and use the supernatant liquid as
black-brown powder or viscous extract. It has a characteris- the sample solution. Separately, dissolve 1 mg of Peoniflo-
tic odor, and a sweet first, then hot, and slightly bitter taste. rin Reference Standard in 1 mL of methanol, and use this
Supplement I, JP XV Crude Drugs 1951

solution as the standard solution. Perform the test with these The viscous extract: Not more than 66.7z (1 g, 1059C, 5
solutions as directed under Thin-layer Chromatography hours).
<2.03>. Spot 5 mL each of the sample solution and standard
Total ash <5.01> Not more than 10.0z, calculated on the
solution on a plate of silica gel for thin-layer chro-
dried basis.
matography. Develop the plate with a mixture of ethyl
acetate, methanol and water (20:3:2) to a distance of about Assay (1) Total alkaloids (ephedrine and pseudoephe-
10 cm, and air-dry the plate. Spray evenly 4-methoxybezal- drine)—Weigh accurately about 0.5 g of the dry extract (or
dehyde-sulfuric acid TS on the plate, and heat at 1059C for 5 an amount of the viscous extract, equivalent to about 0.5 g
minutes: one of the spot among the several spots from the of the dried substance), add exactly 50 mL of diluted
sample solution has the same color tone and Rf value with methanol (1 in 2), shake for 15 minutes, filter, and use the
the purple spot from the standard solution (Peony Root). filtrate as the sample solution. Separately, weigh accurately
(5) To 1.0 g of the dry extract (or 3.0 g of the viscous about 10 mg of ephedrine hydrochloride for assay, previous-
extract) add 10 mL of water, shake, then add 10 mL of 1- ly dried at 1059 C for 3 hours, and dissolve in diluted
butanol, shake, centrifuge, and use the supernatant liquid as methanol (1 in 2) to make exactly 100 mL. Pipet 10 mL of
the sample solution. Separately, dissolve 1 mg of liquiritin this solution, add diluted methanol (1 in 2) to make exactly
for thin-layer chromatography in 1 mL of methanol, and use 50 mL, and use this solution as the standard solution. Per-
this solution as the standard solution. Perform the test with form the test with exactly 10 mL each of the sample solution
these solutions as directed under Thin-layer Chro- and standard solution as directed under Liquid Chro-
matography <2.03>. Spot 5 mL each of the sample solution matography <2.01> according to the following conditions.
and standard solution on a plate of silica gel for thin-layer Determine the peak areas, ATE and ATP, of ephedrine and
chromatography. Develop the plate with a mixture of ethyl pseudoephedrine with the sample solution, and the peak
acetate, methanol and water (20:3:2) to a distance of about area, AS, of ephedrine with the standard solution.
10 cm, and air-dry the plate. Spray evenly dilute sulfuric acid
Amount (mg) of total alkaloids [ephedrine (C10H15NO) and
on the plate, and heat at 1059 C for 5 minutes: one of the
pseudoephedrine (C10H15NO)]
spot among the several spots from the sample solution has
=WS×{(ATE+ATP)/AS}×0.819×(1/10)
the same color tone and Rf value with the yellow-brown spot
from the standard solution (Glycyrrhiza). WS: Amount (mg) of ephedrine hydrochloride for assay
(6) To 1.0 g of the dry extract (or 3.0 g of the viscous
Operating conditions—
extract) add 10 mL of water, shake, then add 25 mL of
Detector: An ultraviolet absorption photometer (wave-
diethyl ether, shake, and take the diethyl ether layer.
length: 210 nm).
Evaporate the layer under reduced pressure, dissolve the
Column: A stainless steel column 4.6 mm in inside di-
residue in 2 mL of diethyl ether, and use the solution as the
ameter and 15 cm in length, packed with octadecylsilanized
sample solution. Separately, dissolve 1 mg of [6]-gingerol
silica gel for liquid chromatography (5 mm in particle di-
for thin-layer chromatography in 1 mL of methanol, and use
ameter).
this solution as the standard solution. Perform the test with
Column temperature: A constant temperature of about
these solutions as directed under Thin-layer Chro-
409C.
matography <2.03>. Spot 10 mL of the sample solution and 5
Mobile phase: A mixture of a solution of sodium lauryl
mL of the standard solution on a plate of silica gel for thin-
sulfate (1 in 130), acetonitrile and phosphoric acid
layer chromatography. Develop the plate with a mixture of
(650:350:1).
ethyl acetate and hexane (1:1) to a distance of about 10 cm,
Flow rate: 1.0 mL/min. (the retention time of ephedrine is
and air-dry the plate. Spray evenly 4-dimethylaminobenzal-
about 27 minutes.)
dehyde TS for spraying on the plate, heat at 1059C for 5
System suitability—
minutes, and allow to cool: one of the spot among the sever-
System performance: Dissolve 1 mg each of ephedrine
al spots from the sample solution has the same color tone
hydrochloride for assay and pseudoephedrine hydrochloride
and Rf value with the blue-green spot from the standard so-
in diluted methanol (1 in 2) to make 10 mL. When the proce-
lution (Ginger).
dure is run with 10 mL of this solution under the above oper-
Purity (1) Heavy metals <1.07>—Prepare the test solution ating conditions, pseudoephedrine and ephedrine are eluted
with 1.0 g of the dry extract (or an amount of the viscous in this order with the resolution between these peaks being
extract, equivalent to 1.0 g of the dried substance) as direct- not less than 1.5.
ed in the Extracts (4) under General Rules for Preparations, System repeatability: When the test is repeated 6 times
and perform the test (not more than 30 ppm). with 10 mL of the standard solution under the above operat-
(2) Arsenic <1.11>—Prepare the test solution with 0.67 g ing conditions, the relative standard deviation of the peak
of the dry extract (or an amount of the viscous extract, area of ephedrine is not more than 1.5z.
equivalent to 0.67 g of the dried substance) according to (2) Peoniflorin—Weigh accurately about 0.5 g of the
Method 3, and perform the test (not more than 3 ppm). dry extract (or an amount of the viscous extract, equivalent
to about 0.5 g of the dried substance), add exactly 50 mL of
Loss on drying <2.41> The dry extract: Not more than 10.0
diluted methanol (1 in 2), shake for 15 minutes, and filter.
z (1 g, 1059C, 5 hours).
1952 Crude Drugs Supplement I, JP XV

Pipet 5 mL of the filtrate, flow through in a column packed


Amount (mg) of glycyrrhizic acid (C42H62O16)
with 2 g of polyamide for column chromatography, elute
=WS×(AT/AS)×(1/2)
with water to make exactly 20 mL of eluate, and use this as
the sample solution. Separately, weigh accurately about 10 WS: Amount (mg) of Glycyrrhizic Acid Reference Stan-
mg of Peoniflorin Reference Standard (separately determine dard, calculated on the anhydrous basis
the water), and dissolve in diluted methanol (1 in 2) to make
Operating conditions—
exactly 100 mL. Pipet 5 mL of this solution, add diluted
Detector: An ultraviolet absorption photometer (wave-
methanol (1 in 2) to make exactly 20 mL, and use this solu-
length: 254 nm).
tion as the standard solution. Perform the test with exactly
Column: A stainless steel column 4.6 mm in inside di-
10 mL each of the sample solution and standard solution as
ameter and 15 cm in length, packed with octadecylsilanized
directed under Liquid Chromatography <2.01> according to
silica gel for liquid chromatography (5 mm in particle di-
the following conditions, and determine the peak areas, AT
ameter).
and AS, of peoniflorin.
Column temperature: A constant temperature of about
Amount (mg) of peoniflorin (C23H28O11) 409C.
=WS×(AT/AS)×(1/2) Mobile phase: A mixture of diluted acetic acid (31) (1 in
15) and acetonitrile (13:7).
WS: Amount (mg) of Peoniflorin Reference Standard,
Flow rate: 1.0 mL/min. (the retention time of glycyrrhizic
calculated on the anhydrous basis
acid is about 12 minutes.)
Operating conditions— System suitability—
Detector: An ultraviolet absorption photometer (wave- System performance: When the procedure is run with 10
length: 232 nm). mL of the standard solution under the above operating con-
Column: A stainless steel column 4.6 mm in inside di- ditions, the number of theoretical plates and the symmetry
ameter and 15 cm in length, packed with octadecylsilanized factor of the peak of glycyrrhizic acid are not less than 5000
silica gel for liquid chromatography (5 mm in particle di- and not more than 1.5, respectively.
ameter). System repeatability: When the test is repeated 6 times
Column temperature: A constant temperature of about with 10 mL of the standard solution under the above operat-
209C. ing conditions, the relative standard deviation of the peak
Mobile phase: A mixture of water, acetonitrile and phos- area of glycyrrhizic acid is not more than 1.5z.
phoric acid (850:150:1).
Containers and storage Containers—Tight containers.
Flow rate: 1.0 mL/min. (the retention time of peoniflorin
is about 9 minutes.)
System suitability—
System performance: Dissolve 1 mg each of Peoniflorin Kamishoyosan Extract
Reference Standard and albiflorin in diluted methanol (1 in
加味逍遙散エキス
2) to make 10 mL. When the procedure is run with 10 mL of
this solution under the above operating conditions, albiflo-
Change to read:
rin and peoniflorin are eluted in this order with the resolu-
tion between these peaks being not less than 2.5.
Kamishoyosan Extract contains not less than 28 mg
System repeatability: When the test is repeated 6 times
and not more than 84 mg of peoniflorin (C23H28O11:
with 10 mL of the standard solution under the above operat-
480.46), not less than 25 mg and not more than 75 mg
ing conditions, the relative standard deviation of the peak
of geniposide, and not less than 12 mg and not more
area of peoniflorin is not more than 1.5z.
than 36 mg (for preparation prescribed 1.5 g of
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
Glycyrrhiza) or not less than 16 mg and not more than
the dry extract (or an amount of the viscous extract, equiva-
48 mg (for preparation prescribed 2 g of Glycyrrhiza)
lent to about 0.5 g of the dried substance), add exactly 50
of glycyrrhizic acid (C42H62O16: 822.93) per the extract
mL of diluted methanol (1 in 2), shake for 15 minutes, filter,
prepared as directed in the Method of preparation.
and use the filtrate as the sample solution. Separately, weigh
accurately about 10 mg of Glycyrrhizic Acid Reference Stan- Method of preparation Prepare a dry extract or viscous
dard (separately determine the water) dissolve in diluted extract as directed under Extracts, with 3 g of Japanese An-
methanol (1 in 2) to make exactly 100 mL, and use this solu- gelica Root, 3 g of Peony Root, 3 g of Atractylodes Rhizome
tion as the standard solution. Perform the test with exactly or Atractylodes Lancea Rhizome, 3 g of Poria Sclerotium, 3
10 mL each of the sample solution and standard solution as g of Bupleurum Root, 2 g of Moutan Bark, 2 g of Gardenia
directed under Liquid Chromatography <2.01> according to Fruit, 2 g of Glycyrrhiza, 1 g of Ginger and 1 g of Mentha
the following conditions, and determine the peak areas, AT Herb, or with 3 g of Japanese Angelica Root, 3 g of Peony
and AS, of glycyrrhizic acid. Root, 3 g of Atractylodes Rhizome or Atractylodes Lancea
Rhizome, 3 g of Poria Sclerotium, 3 g of Bupleurum Root,
2 g of Moutan Bark, 2 g of Gardenia Fruit, 1.5 g of Glycyr-
Supplement I, JP XV Crude Drugs 1953

rhiza, 1 g of Ginger and 1 g of Mentha Herb, or with 3 g of chromatography. Develop the plate with a mixture of ethyl
Japanese Angelica Root, 3 g of Peony Root, 3 g of Atrac- acetate and hexane (1:1) to a distance of about 10 cm, and
tylodes Rhizome, 3 g of Poria Sclerotium, 3 g of Bupleurum air-dry the plate. Spray evenly 1-naphthol-sulfuric acid TS
Root, 2 g of Moutan Bark, 2 g of Gardenia Fruit, 1.5 g of on the plate, heat at 1059C for 5 minutes, and allow to cool:
Glycyrrhiza, 1.5 g of Ginger and 1 g of Mentha Herb, or one of the spot among the several spots from the sample so-
with 3 g of Japanese Angelica Root, 3 g of Peony Root, 3 g lution has the same color tone and Rf value with the red spot
of Atractylodes Rhizome, 3 g of Poria Sclerotium, 3 g of from the standard solution (Atractylodes Rhizome).
Bupleurum Root, 2 g of Moutan Bark, 2 g of Gardenia (4) For preparation prescribed Atractylodes Lancea
Fruit, 1.5 g of Glycyrrhiza, 0.5 g of Ginger and 1 g of Men- Rhizome—To 2.0 g of the dry extract (or 6.0 g of the viscous
tha Herb. extract) add 10 mL of water, shake, then add 25 mL of
hexane, and shake. Take the hexane layer, add anhydrous
Description Kamishoyosan Extract occurs as a yellow-
sodium sulfate to dry, and filter. Evaporate the filtrate un-
brown to black-brown powder or viscous extract. It has
der reduced pressure, add 2 mL of hexane to the residue, and
slightly a characteristic odor, and a sweet, slightly hot, then
use this solution as the sample solution. Perform the test
bitter taste.
with the sample solution as directed under Thin-layer Chro-
Identification (1) To 2.0 g of the dry extract (or 6.0 g of matography <2.03>. Spot 20 mL of the sample solution on a
the viscous extract) add 10 mL of water, shake, then add 5 plate of silica gel with fluorescent indicator for thin-layer
mL of diethyl ether, shake, centrifuge, and use the super- chromatography, develop the plate with a mixture of hexane
natant liquid as the sample solution. Separately, dissolve 1 and acetone (7:1) to a distance of about 10 cm, and air-dry
mg of (Z)-ligustilide for thin-layer chromatography in 10 the plate. Examine under ultraviolet light (main wavelength:
mL of methanol, and use this solution as the standard solu- 254 nm): a dark purple spot is observed at around Rf 0.4.
tion. Perform the test with these solutions as directed under The spot shows a greenish brown color after being sprayed
Thin-layer Chromatography <2.03>. Spot 10 mL each of the 4-dimethylaminobenzaldehyde TS for spraying, heated at
sample solution and standard solution on a plate of silica gel 1059 C for 5 minutes, and allowed to cool (Atractylodes Lan-
for thin-layer chromatography. Develop the plate with a cea Rhizome).
mixture of ethyl acetate and hexane (1:1) to a distance of (5) To 2.0 g of the dry extract (or 6.0 g of the viscous
about 10 cm, and air-dry the plate. Examine under ultravio- extract) add 10 mL of sodium hydroxide TS, shake, then
let light (main wavelength: 365 nm): one of the spot among add 5 mL of 1-butanol, shake, centrifuge, and use the super-
the several spots from the sample solution has the same color natant liquid as the sample solution. Separately, dissolve 1
tone and Rf value with the bluish white fluorescent spot mg of saikosaponin b2 for thin-layer chromatography in 1
from the standard solution (Japanese Angelica Root). mL of methanol, and use this solution as the standard solu-
(2) To 2.0 g of the dry extract (or 6.0 g of the viscous tion. Perform the test with these solutions as directed under
extract) add 10 mL of water, shake, then add 5 mL of 1- Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
butanol, shake, centrifuge, and use the supernatant liquid as ple solution and 2 mL of the standard solution on a plate of
the sample solution. Separately, dissolve 1 mg of albiflorin silica gel for thin-layer chromatography. Develop the plate
in 1 mL of methanol, and use this solution as the standard with a mixture of ethyl acetate, ethanol (99.5) and water
solution. Perform the test with these solutions as directed (8:2:1) to a distance of about 10 cm, and air-dry the plate.
under Thin-layer Chromatography <2.03>. Spot 10 mL each Spray evenly 4-dimethylaminobenzaldehyde TS on the plate:
of the sample solution and standard solution on a plate of one of the spot among the several spots from the sample
silica gel for thin-layer chromatography. Develop the plate solution has the same color tone and Rf value with the red
with a mixture of ethyl acetate, methanol and ammonia spot from the standard solution (Bupleurum Root).
solution (28) (6:3:2) to a distance of about 10 cm, and air- (6) To 2.0 g of the dry extract (or 6.0 g of the viscous
dry the plate. Spray evenly 4-methoxybenzaldehyde-sulfuric extract) add 10 mL of water, shake, then add 15 mL of
acid TS on the plate, heat at 1059C for 5 minutes, and exa- diethyl ether, and shake. Take the diethyl ether layer,
mine under ultraviolet light (main wavelength: 365 nm): one evaporate the layer under reduced pressure, add 1 mL of
of the spot among the several spots from the sample solution diethyl ether to the residue, and use this solution as the
has the same color tone and Rf value with the orange sample solution. Separately, dissolve 1 mg of peonol for
fluorescent spot from the standard solution (Peony Root). thin-layer chromatography in 1 mL of methanol, and use
(3) For preparation prescribed Atractylodes Rhizome— this solution as the standard solution. Perform the test with
To 2.0 g of the dry extract (or 6.0 g of the viscous extract) these solutions as directed under Thin-layer Chro-
add 10 mL of water, shake, then add 5 mL of diethyl ether, matography <2.03>. Spot 10 mL each of the sample solution
shake, centrifuge, and use the supernatant liquid as the sam- and standard solution on a plate of silica gel for thin-layer
ple solution. Separately, dissolve 1 mg of atractylenolide III chromatography, develop the plate with a mixture of hexane
for thin-layer chromatography in 1 mL of methanol, and use and diethyl ether (5:3) to a distance of about 10 cm, and air-
this solution as the standard solution. Perform the test with dry the plate. Spray evenly 4-methoxybenzaldehyde-sulfuric
these solutions as directed under Thin-layer Chro- acid TS on the plate, and heat at 1059 C for 5 minutes: one of
matography <2.03>. Spot 10 mL each of the sample solution the spot among the several spots from the sample solution
and standard solution on a plate of silica gel for thin-layer has the same color tone and Rf value with the orange spot
1954 Crude Drugs Supplement I, JP XV

from the standard solution (Moutan Bark). the standard solution. Perform the test with these solutions
(7) To 2.0 g of the dry extract (or 6.0 g of the viscous as directed under Thin-layer Chromatography <2.03>. Spot
extract) add 10 mL of water, shake, then add 5 mL of 1- 10 mL each of the sample solution and standard solution on
butanol, shake, centrifuge, and use the supernatant liquid as a plate of silica gel for thin-layer chromatography. Develop
the sample solution. Separately, dissolve 1 mg of geniposide the plate with a mixture of ethyl acetate, water and formic
for thin-layer chromatography in 1 mL of methanol, and use acid (10:1:1) to a distance of about 10 cm, and air-dry the
this solution as the standard solution. Perform the test with plate. Spray evenly vanillin-sulfuric acid TS on the plate,
these solutions as directed under Thin-layer Chromato- heat at 1059 C for 5 minutes, and allow to cool: one of the
graphy <2.03>. Spot 10 mL each of the sample solution and spot among the several spots from the sample solution has
standard solution on a plate of silica gel for thin-layer chro- the same color tone and Rf value with the red-purple spot
matography. Develop the plate with a mixture of ethyl (around Rf 0.6) from the standard solution (Mentha Herb).
acetate, methanol and ammonia solution (28) (6:3:2) to a
Purity (1) Heavy metals <1.07>—Prepare the test solution
distance of about 10 cm, and air-dry the plate. Spray evenly
with 1.0 g of the dry extract (or an amount of the viscous
4-methoxybenzaldehide-sulfric acid TS on the plate, and
extract, equivalent to 1.0 g of the dried substance) as direct-
heat at 1059 C for 5 minutes: one of the spot among the
ed in the Extracts (4) under General Rules for Preparations,
several spots from the sample solution has the same color
and perform the test (not more than 30 ppm).
tone and Rf value with the purple spot from the standard so-
(2) Arsenic <1.11>—Prepare the test solution with 0.67 g
lution (Gardenia Fruit).
of the dry extract (or an amount of the viscous extract,
(8) To 2.0 g of the dry extract (or 6.0 g of the viscous
equivalent to 0.67 g of the dried substance) according to
extract) add 10 mL of water, shake, then add 5 mL of 1-
Method 3, and perform the test (not more than 3 ppm).
butanol, centrifuge, and use the supernatant liquid as the
sample solution. Separately, dissolve 1 mg of liquiritin for Loss on drying <2.41> The dry extract: Not more than 9.0
thin-layer chromatography in 1 mL of methanol, and use z (1 g, 1059C, 5 hours).
this solution as the standard solution. Perform the test with The viscous extract: Not more than 66.7z (1 g, 1059C, 5
these solutions as directed under Thin-layer Chro- hours).
matography <2.03>. Spot 10 mL of the sample solution and 5
Total ash <5.01> Not more than 10.0z, calculated on the
mL of the standard solution on a plate of silica gel for thin-
dried basis.
layer chromatography. Develop the plate with a mixture of
ethyl acetate, methanol and water (20:3:2) to a distance of Assay (1) Peoniflorin—Weigh accurately about 0.5 g of
about 10 cm, and air-dry the plate. Spray evenly dilute sul- the dry extract (or an amount of the viscous extract, equiva-
furic acid on the plate, and heat at 1059C for 5 minutes: one lent to about 0.5 g of the dried substance), add exactly 50
of the spot among the several spots from the sample solution mL of diluted methanol (1 in 2), shake for 15 minutes, filter,
has the same color tone and Rf value with the yellow-brown and use the filtrate as the sample solution. Separately, weigh
spot from the standard solution (Glycyrrhiza). accurately about 10 mg of Peoniflorin Reference Standard
(9) To 2.0 g of the dry extract (or 6.0 g of the viscous (separately determine the water), and dissolve in diluted
extract) add 10 mL of water, shake, then add 5 mL of methanol (1 in 2) to make exactly 100 mL, and use this solu-
diethyl ether, centrifuge, and use the supernatant liquid as tion as the standard solution. Perform the test with exactly
the sample solution. Separately, dissolve 1 mg of [6]-gin- 10 mL each of the sample solution and standard solution as
gerol for thin-layer chromatography in 1 mL of methanol, directed under Liquid Chromatography <2.01> according to
and use this solution as the standard solution. Perform the the following conditions, and determine the peak areas, AT
test with these solutions as directed under Thin-layer Chro- and AS, of peoniflorin.
matography <2.03>. Spot 10 mL each of the sample solution
Amount (mg) of peoniflorin (C23H28O11)
and standard solution on a plate of silica gel for thin-layer
=WS×(AT/AS)×(1/2)
chromatography. Develop the plate with a mixture of ethyl
acetate and hexane (1:1) to a distance of about 10 cm, and WS: Amount (mg) of Peoniflorin Reference Standard,
air-dry the plate. Spray evenly 4-dimethylaminobenzalde- calculated on the anhydrous basis
hyde TS for spraying on the plate, heat at 1059C for 5
Operating conditions—
minutes, and allow to cool: one of the spot among the sever-
Detector: An ultraviolet absorption photometer (wave-
al spots from the sample solution has the same color tone
length: 232 nm).
and Rf value with the blue-green spot from the standard
Column: A stainless steel column 4.6 mm in inside di-
solution (Ginger).
ameter and 15 cm in length, packed with octadecylsilanized
(10) To 2.0 g of the dry extract (or 6.0 g of the viscous
silica gel for liquid chromatography (5 mm in particle di-
extract) add 10 mL of diluted phosphoric acid (1 in 30),
ameter).
shake, then add 15 mL of ethyl acetate, shake, centrifuge,
Column temperature: A constant temperature of about
and use the supernatant liquid as the sample solution.
209C.
Separately, shake 0.2 g of pulverized Mentha Herb with 10
Mobile phase: A mixture of water, acetonitrile and phos-
mL of diluted phosphoric acid (1 in 30), add 15 mL of ethyl
phoric acid (850:150:1)‚
acetate, shake, centrifuge, and use the supernatant liquid as
Supplement I, JP XV Crude Drugs 1955

Flow rate: 1.0 mL/min. (the retention time of peoniflorin mL of diluted methanol (1 in 2), shake for 15 minutes, filter,
is about 9 minutes.) and use the filtrate as the sample solution. Separately, weigh
System suitability— accurately about 10 mg of Glycyrrhizic Acid Reference Stan-
System performance: Dissolve 1 mg each of Peoniflorin dard (separately determine the water), dissolve in diluted
Reference Standard and albiflorin in diluted methanol (1 in methanol (1 in 2) to make exactly 100 mL, and use this solu-
2) to make 10 mL. When the procedure is run with 10 mL of tion as the standard solution. Perform the test with exactly
this solution under the above operating conditions, albiflo- 10 mL each of the sample solution and standard solution as
rin and peoniflorin are eluted in this order with the resolu- directed under Liquid Chromatography <2.01> according to
tion between these peaks being not less than 2.5. the following conditions, and determine the peak areas, AT
System repeatability: When the test is repeated 6 times and AS, of glycyrrhizic acid.
with 10 mL of the standard solution under the above operat-
Amount (mg) of glycyrrhizic acid (C42H62O16)
ing conditions, the relative standard deviation of the peak
=WS×(AT/AS)×(1/2)
area of peoniflorin is not more than 1.5z.
(2) Geniposide—Weigh accurately about 0.5 g of the dry WS: Amount (mg) of Glycyrrhizic Acid Reference Stan-
extract (or an amount of the viscous extract, equivalent to dard, calculated on the anhydrous basis
about 0.5 g of the dried substance), add exactly 50 mL of
Operating conditions—
diluted methanol (1 in 2), shake for 15 minutes, filter, and
Detector: An ultraviolet absorption photometer (wave-
use the filtrate as the sample solution. Separately, weigh
length: 254 nm).
accurately about 10 mg of geniposide for component deter-
Column: A stainless steel column 4.6 mm in inside di-
mination, previously dried in a desiccator (in vacuum, phos-
ameter and 15 cm in length, packed with octadecylsilanized
phorous (V) oxide) for 24 hours, dissolve in diluted
silica gel for liquid chromatography (5 mm in particle di-
methanol (1 in 2) to make exactly 100 mL, and use this solu-
ameter).
tion as the standard solution. Perform the test with exactly
Column temperature: A constant temperature of about
10 mL each of the sample solution and standard solution as
409C.
directed under Liquid Chromatography <2.01> according to
Mobile phase: A mixture of diluted acetic acid (31) (1 in
the following conditions, and determine the peak areas, AT
15) and acetonitrile (13:7).
and AS, of geniposide.
Flow rate: 1.0 mL/min. (the retention time of glycyrrhizic
Amount (mg) of geniposide=WS×(AT/AS)×(1/2) acid is about 12 minutes.)
System suitability—
WS: Amount (mg) of geniposide for component determi-
System performance: When the procedure is run with 10
nation
mL of the standard solution under the above operating con-
Operating conditions— ditions, the number of theoretical plates and the symmetry
Detector: An ultraviolet absorption photometer (wave- factor of the peak of glycyrrhizic acid are not less than 5000
length: 240 nm). and not more than 1.5, respectively.
Column: A stainless steel column 4.6 mm in inside di- System repeatability: When the test is repeated 6 times
ameter and 15 cm in length, packed with octadecylsilanized with 10 mL of the standard solution under the above operat-
silica gel for liquid chromatography (5 mm in particle di- ing conditions, the relative standard deviation of the peak
ameter). area of glycyrrhizic acid is not more than 1.5z.
Column temperature: A constant temperature of about
Containers and storage Containers—Tight containers.
409C.
Mobile phase: A mixture of water, acetonitrile and phos-
phoric acid (900:100:1).
Add the following:
Flow rate: 1.0 mL/min. (the retention time of geniposide
is about 10 minutes.)
System suitability— Keishibukuryogan Extract
System performance: When the procedure is run with 10
桂枝茯苓丸エキス
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
Keishibukuryogan Extract contains not less than 0.6
factor of the peak of geniposide are not less than 5000 and
mg and not more than 2.4 mg (for preparation
not more than 1.5, respectively.
prescribed 3 g of Cinnamon Bark) or not less than 0.8
System repeatability: When the test is repeated 6 times
mg and not more than 3.2 mg (for preparation
with 10 mL of the standard solution under the above operat-
prescribed 4 g of Cinnamon Bark) of (E)-cinnamic
ing conditions, the relative standard deviation of the peak
acid, not less than 30 mg and not more than 90 mg (for
area of geniposide is not more than 1.5z.
preparation prescribed 3 g each of Moutan Bark and
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
Peony Root) or not less than 40 mg and not more than
the dry extract (or an amount of the viscous extract, equiva-
120 mg (for preparation prescribed 4 g each of Mou-
lent to about 0.5 g of the dried substance), add exactly 50
tan Bark and Peony Root) of peoniflorin (C23H28O11:
1956 Crude Drugs Supplement I, JP XV

480.46), and not less than 21 mg and not more than 63 as the sample solution. Separately, dissolve 2 mg of amygda-
mg (for preparation prescribed 3 g of Peach Kernel) or lin for thin-layer chromatography in 1 mL of methanol, and
not less than 28 mg and not more than 84 mg (for use this solution as the standard solution. Perform the test
preparation prescribed 4 g of Peach Kernel) of amyg- with these solutions as directed under Thin-layer Chro-
dalin per amount specified in the Method of prepara- matography <2.03>. Spot 5 mL each of the sample solution
tion. and standard solution on a plate of silica gel for thin-layer
chromatography. Develop the plate with a mixture of 1-
Method of preparation Prepare a dry extract or viscous ex-
propanol, ethyl acetate and water (4:4:3) to a distance of
tract as directed under Extracts, with 4 g of Cinnamon Bark,
about 10 cm, and air-dry the plate. Spray evenly 4-me-
4 g of Poria Sclerotium, 4 g of Moutan Bark, 4 g of Peach
thoxybenzaldehyde-sulfuric acid TS on the plate, and heat at
Kernel and 4 g of Peony Root, or prepare a dry extract by
1059 C for 10 minutes: one of the spot among the several
adding Light Anhydrous Silicic Acid to an extractive, pre-
spots from the sample solution has the same color tone and
pared as directed under Extracts with 3 g of Cinnamon Bark,
Rf value with the green-brown spot from the standard solu-
3 g of Poria Sclerotium, 3 g of Moutan Bark, 3 g of Peach
tion (Peach Kernel).
Kernel and 3 g of Peony Root.
(4) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
Description Keishibukuryogan Extract is a light brown to extract) with 10 mL of water, add 5 mL of 1-butanol, shake,
black-brown, powder or viscous extract. It has a characteris- centrifuge, and use the supernatant liquid as the sample
tic odor and has a taste slightly sweet first then bitter later. solution. Separately, dissolve 1 mg of albiflorin in 1 mL of
methanol, and use this solution as the standard solution.
Identification (1) Shake 1.0 g of the dry extract (or 3.0 g
Perform the test with these solutions as directed under Thin-
of the viscous extract) with 10 mL of water, add 25 mL of
layer Chromatography <2.03>. Spot 5 mL each of the sample
diethyl ether, and shake. Take the diethyl ether layer,
solution and standard solution on a plate of silica gel for
evaporate the layer under reduced pressure, dissolve the
thin-layer chromatography. Develop the plate with a mix-
residue in 2 mL of diethyl ether, and use this solution as the
ture of ethyl acetate, methanol and ammonia water (28)
sample solution. Separately, dissolve 1 mg of (E)-cinnamic
(6:3:2) to a distance of about 10 cm, and air-dry the plate.
acid for thin-layer chromatography in 1 mL of methanol,
Spray evenly 4-methoxybenzaldehyde-sulfuric acid TS on
and use this solution as the standard solution. Perform the
the plate, heat at 1059 C for 5 minutes, and examine under
test with these solutions as directed under Thin-layer Chro-
ultraviolet light (main wavelength: 365 nm): one of the spot
matography <2.03>. Spot 5 mL each of the sample solution
among the several spots from the sample solution has the
and standard solution on a plate of silica gel with fluorescent
same color tone and Rf value with the orange fluorescent
indicator for thin-layer chromatography. Develop the plate
spot from the standard solution (Peony Root).
with a mixture of hexane, ethyl acetate, formic acid and
water (60:40:4:1) to a distance of about 10 cm, and air-dry Purity (1) Heavy metals <1.07>—Prepare the test solution
the plate. Examine under ultraviolet light (main wavelength: with 1.0 g of the dry extract (or an amount of the viscous
254 nm): one of the spot among the several spots from the extract, equivalent to 1.0 g of the dried substance) as direct-
sample solution has the same color tone and Rf value with ed in the Extracts (4) under General Rules for Preparations,
the blue-purple spot from the standard solution (Cinnamon and perform the test (not more than 30 ppm).
Bark). (2) Arsenic <1.11>—Prepare the test solution with 0.67 g
(2) Shake 2.0 g of the dry extract (or 6.0 g of the viscous of the dry extract (or an amount of the viscous extract,
extract) with 10 mL of water, add 25 mL of diethyl ether, equivalent to 0.67 g of the dried substance) according to
and shake. Take the diethyl ether layer, evaporate the layer Method 3, and perform the test (not more than 3 ppm).
under reduced pressure, dissolve the residue in 1 mL of
Loss on drying <2.41> The dry extract: Not more than 10.0
diethyl ether, and use this solution as the sample solution.
z (1 g, 1059C, 5 hours).
Separately, dissolve 1 mg of peonol for thin-layer chro-
The viscous extract: Not more than 66.7z (1 g, 1059C, 5
matography in 1 mL of methanol, and use this solution as
hours).
the standard solution. Perform the test with these solutions
as directed under Thin-layer Chromatography <2.03>. Spot Total ash <5.01> Not more than 10.0z, calculated on the
10 mL each of the sample solution and standard solution on dried basis. However, for the dry extract prepared by adding
a plate of silica gel for thin-layer chromatography. Develop Light Anhydrous Silicic Acid, between 9.0z and 18.0z.
the plate with a mixture of hexane and diethyl ether (5:3) to a
Assay (1) (E)-Cinnamic acid—Conduct this procedure
distance of about 10 cm, and air-dry the plate. Spray evenly
without exposure to light, using light-resistant vessels.
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
Weigh accurately about 0.5 g of the dry extract (or an
heat at 1059 C for 5 minutes: one of the spot among the
amount of the viscous extract, equivalent to about 0.5 g of
several spots from the sample solution has the same color
the dried substance), add exactly 50 mL of diluted methanol
tone and Rf value with the orange spot from the standard so-
(1 in 2), shake for 15 minutes, filter, and use the filtrate as
lution (Moutan Bark).
the sample solution. Separately, weigh accurately about 10
(3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
mg of (E)-cinnamic acid for component determination,
extract) with 10 mL of methanol, filter, and use the filtrate
previously dried in a desiccator (silica gel) for not less than
Supplement I, JP XV Crude Drugs 1957

24 hours, and dissolve in diluted methanol (1 in 2) to make Column: A stainless steel column 4.6 mm in inside di-
exactly 100 mL. Pipet 10 mL of this solution, add diluted ameter and 15 cm in length, packed with octadecylsilanized
methanol (1 in 2) to make exactly 100 mL, and use this solu- silica gel for liquid chromatography (5 mm in particle di-
tion as the standard solution. Perform the test with exactly ameter).
10 mL each of the sample solution and standard solution as Column temperature: A constant temperature of about
directed under Liquid Chromatography <2.01> according to 209C.
the following conditions, and determine the peak areas, AT Mobile phase: A mixture of water, acetonitrile and phos-
and AS, of (E)-cinnamic acid. phoric acid (850:150:1).
Flow rate: 1.0 mL per minute. (the retention time of
Amount (mg) of (E)-cinnamic acid
paeoniflorin is about 9 minutes.)
=WS×(AT/AS)×(1/20)
System suitability—
WS: Amount (mg) of (E)-cinnamic acid for component System performance: Dissolve 1 mg each of Paeoniflorin
determination Reference Standard and albiflorin in diluted methanol (1 in
2) to make 10 mL. When the procedure is run with 10 mL of
Operating conditions—
this solution under the above operating conditions, albiflo-
Detector: An ultraviolet absorption photometer (wave-
rin and paeoniflorin are eluted in this order with the resolu-
length: 273 nm).
tion between these peaks being not less than 2.5.
Column: A stainless steel column 4.6 mm in inside di-
System repeatability: When the test is repeated 6 times
ameter and 15 cm in length, packed with octadecylsilanized
with 10 mL of the standard solution under the above operat-
silica gel for liquid chromatography (5 mm in particle di-
ing conditions, the relative standard deviation of the peak
ameter).
area of paeoniflorin is not more than 1.5z.
Column temperature: A constant temperature of about
(3) Amygdalin—Weigh accurately about 0.5 g of the dry
409C.
extract (or an amount of the viscous extract, equivalent to
Mobile phase: A mixture of water, acetonitrile and phos-
about 0.5 g of the dried substance), add exactly 50 mL of
phoric acid (750:250:1).
diluted methanol (1 in 2), shake for 15 minutes, filter, and
Flow rate: 1.0 mL per minute. (the retention time of (E)-
use the filtrate as the sample solution. Separately, weigh ac-
cinnamic acid is about 12 minutes.)
curately about 10 mg of amygdalin for component determi-
System suitability—
nation, previously dried in a desiccator (silica gel) for not
System performance: When the procedure is run with 10
less than 24 hours, dissolve in diluted methanol (1 in 2) to
mL of the standard solution under the above operating con-
make exactly 50 mL, and use this solution as the standard
ditions, the number of theoretical plates and the symmetry
solution. Perform the test with exactly 10 mL each of the
factor of the peak of (E)-cinnamic acid are not less than 5000
sample solution and standard solution as directed under Liq-
and not more than 1.5, respectively.
uid Chromatography <2.01> according to the following con-
System repeatability: When the test is repeated 6 times
ditions, and determine the peak areas, AT and AS, of amyg-
with 10 mL of the standard solution under the above operat-
dalin.
ing conditions, the relative standard deviation of the peak
area of (E)-cinnamic acid is not more than 1.5z. Amount (mg) of amygdalin=WS×(AT/AS)
(2) Paeoniflorin—Weigh accurately about 0.5 g of the
WS: Amount (mg) of amygdalin for component determi-
dry extract (or an amount of the viscous extract, equivalent
nation
to about 0.5 g of the dried substance), add exactly 50 mL of
diluted methanol (1 in 2), shake for 15 minutes, filter, and Operating conditions—
use the filtrate as the sample solution. Separately, weigh Detector: An ultraviolet absorption photometer (wave-
accurately about 10 mg of Paeoniflorin Reference Standard length: 210 nm).
(separately determine the water), dissolve in diluted Column: A stainless steel column 4.6 mm in inside di-
methanol (1 in 2) to make exactly 50 mL, and use this solu- ameter and 15 cm in length, packed with octadecylsilanized
tion as the standard solution. Perform the test with exactly silica gel for liquid chromatography (5 mm in particle di-
10 mL each of the sample solution and standard solution as ameter).
directed under Liquid Chromatography <2.01> according to Column temperature: A constant temperature of about
the following conditions, and determine the peak areas, AT 459C.
and AS, of paeoniflorin. Mobile phase: A mixture of 0.05 mol/L sodium dihydro-
gen phosphate TS and methanol (5:1).
Amount (mg) of paeoniflorin (C23H28O11)
Flow rate: 0.8 mL per minute. (the retention time of
=WS×(AT/AS)
amygdalin is about 12 minutes.)
WS: Amount (mg) of Paeoniflorin Reference Standard, System suitability—
calculated on the anhydrous basis System performance: When the procedure is run with 10
mL of the standard solution under the above operating con-
Operating conditions—
ditions, the number of theoretical plates and the symmetry
Detector: An ultraviolet absorption photometer (wave-
factor of the peak of amygdalin are not less than 5000 and
length: 232 nm).
1958 Crude Drugs Supplement I, JP XV

not more than 1.5, respectively. Total ash <5.01> Not more than 10.0z.
System repeatability: When the test is repeated 6 times
Acid-insoluble ash <5.01> Not more than 2.0z.
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak Extract content <5.01> Dilute ethanol-soluble extract: not
area of amygdalin is not more than 1.5z. less than 12.0z.

Containers and storage Containers—Tight containers.

Add the following:


Add the following:
Lilium Bulb
Leonurus Herb Lilii Bulbus
Leonuri Herba ビャクゴウ

ヤクモソウ
Lilium Bulb is the scaly leaves of Lilium lancifolium
Thunberg, Lilium brownii F.E.Brown var. colchesteri
Leonurus Herb is the aerial part of Leonurus
Wilson, Lilium brownii F.E.Brown or Lilium pumi-
japonicus Houttuyn or Leonurus sibiricus Linn áe
lum De Candolle (Liliaceae), usually with the applica-
(Labiatae), collected during the flowering season.
tion of steaming.
Description Stem, leaves, and flowers usually cross sec-
Description Lilium Bulb reveals oblong with narrowed
tioned, stems squre, 0.2 to 3 cm in diameter, yellow-green to
apex, lanceolate, or narrowly triangular boat-shaped, trans-
green-brown in color, covered densely with white short
lucent, 1.3 to 6 cm in length, 0.5 to 2.0 cm in diameter, ex-
hairs; the pith white, a great parts of central of sections.
ternally milky white to light yellowish brown occasionally
Light in texture. Leaves opposite, petiolated, 3-dissected to
purplish in color, nearly smooth; central portion somewhat
3-incised, each lobes split pinnately, and end lobes reveals
thickend, circumferential portion thin, slightly waved, oc-
linear-lanceolate, acute or acuminate, the upper surface light
casionally rolled inside; usually several lines of vascular bun-
green, the lower surface bristle with white short hairs,
dles longitudinally in parallel are seen through parenchyma;
grayish green. Flower, verticillate; sepal, tubular, and the
hard in texture, easy to break; fractured surface horny and
upper end acerate with five lobes; light green to light green-
flat.
brown in color, corolla labiate, light red-purple to light
Odorless; taste, slightly acid and bitter.
brown.
Under a microscope <5.01>, the surface reveals epidermal
Odor, slightly; taste, slightly bitter, astringent.
cells rectangular to almost square, stomata nearly circular,
Under a microscope <5.01>, a transverse section of stem
the cells adjacent to stomata mostly 4 in number. Under a
reveals four ridge, a parts of the ridge of Leonurus sibiricus
microscope <5.01>, a transverse section reveals the outermost
Linn áe protruding knobby. Epidermis, observed non-glandu-
layer composed of epidermal cells covered with smooth
lar hairs from 1 to 3 cells, glandular hairs with head of 1 to 4
cuticle; beneath epidermis circular to quadrangular paren-
celled or glandular scale with 8 cells. Each ridge parts,
chymatous cells distributed evenly, palisade tissue not ob-
beneath epidermis, collenchyma developed, development of
served; in parenchyma of mesophyll collateral vascular bun-
xylem fibres remarkably. Cortex composed of several layers
dles extended from adaxial side to abaxial side of scaly
parenchymatous cells. Collateral vascular bundle arranged
leaves are arranged almost in a transverse line; starch grains
in a circle. Phloem fibres observed at the outer portion of
contained in parenchymatous cells, usually gelatinized.
phloem. Parenchymatous cells of cortex and pith observe
needle crystals or plate-like crystals of calcium oxalate. Identification To 3 g of pulverized Lilium Bulb add 10 mL
of 1-butanol, shake, add 10 mL of water, shake for 30
Identification To 1 g of pulverized Leonurus Herb add 10
minutes, and centrifuge. Evaporate the supernatant liquid
mL of methanol, shake for 10 minutes, centrifuge, and use
under reduced pressure, add 1 mL of methanol to the
the supernatant liquid as the sample solution. Perform the
residue, shake gently, and use the supernatant liquid so
test with the sample solution as directed under Thin-layer
obtained as the sample solution. Perform the test with the
Chromatography <2.03>. Spot 10 mL of the sample solution
sample solution as directed under Thin-layer Chro-
on a plate of silica gel for thin-layer chromatography, de-
matography <2.03>. Spot 10 mL of the sample solution on a
velop the plate with a mixture of water and methanol (1:1) to
plate of silica gel with fluorescent indicator for thin-layer
a distance of about 10 cm, and air-dry the plate. Spray even-
chromatography. Develop the plate with a mixture of ethyl
ly Dragendorff's TS for spraying followed by immediate
acetate, methanol and water (12:2:1) to a distance of about
spraying of sodium nitrite TS on the plate: a grayish brown
10 cm, and air-dry the plate. Examine under ultraviolet light
spot appears at around Rf 0.5, which color fades soon and
(main wavelength: 254 nm): two spots appear at around Rf
then disappears after air-drying the plate.
0.3. When examine these spots under ultraviolet light (main
Loss on drying <5.01> Not more than 12.0z. wavelength: 365 nm) after spraying with sodium carbonate
Supplement I, JP XV Crude Drugs 1959

TS, they appear as blue-purple fluorescent spots. procedure with the residue, using 40 mL of diluted methanol
(7 in 10). Combine the whole filtrates, add diluted methanol
Loss on drying <5.01> Not more than 16.0z.
(7 in 10) to make exactly 100 mL, and use this solution as the
Total ash <5.01> Not more than 4.5z. sample solution. Separately, dry magnolol for component
determination in a desiccator (silica gel) for 1 hour or more.
Extract content <5.01> Dilute ethanol-soluble extract: not
Weigh accurately about 10 mg of it, dissolve in diluted
less than 8.0z.
methanol (7 in 10) to make exactly 100 mL, and use this
solution as the standard solution. Perform the test with ex-
actly 10 mL each of the sample solution and standard solu-
Lindera Root tion as directed under Liquid Chromatography <2.01> ac-
cording to the following conditions, and determine the peak
ウヤク
areas, AT and AS, of magnolol in each solution.
Add the following next to Identification: Amount (mg) of magnolol=WS×(AT/AS)
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of WS: Amount (mg) of magnolol for component determina-
pulverized Lindera Root according to Method 3, and per- tion
form the test. Prepare the control solution with 3.0 mL of
Operating conditions—
Standard Lead Solution (not more than 10 ppm).
Detector: An ultraviolet absorption photometer (wave-
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
length: 289 nm).
of pulverized Lindera Root according to Method 4, and per-
Column: A stainless steel column 4 to 6 mm in inside di-
form the test (not more than 5 ppm).
ameter and 15 to 25 cm in length, packed with octadecyl-
silanized silica gel (5 to 10 mm in particle diameter).
Column temperature: A constant temperature of about
Lithospermum Root 209C.
シコン Mobile phase: A mixture of water, acetonitrile and acetic
acid (100) (50:50:1).
Add the following next to Identification: Flow rate: Adjust the flow rate so that the retention time
of magnolol is about 14 minutes.
Purity Arsenic <1.11>—Prepare the test solution with 0.40 System suitability—
g of pulverized Lithospermum Root according to Method 4, System performance: Dissolve 1 mg each of magnolol for
and perform the test (not more than 5 ppm). component determination and honokiol in diluted methanol
(7 in 10) to make 10 mL. When the procedure is run with 10
mL of this solution under the above operating conditions,
Lycium Bark honokiol and magnolol are eluted in this order with the reso-
lution between these peaks being not less than 5.
ジコッピ System repeatability: When the test is repeated 6 times
with 10 mL of the standard solution under the above operat-
Add the following next to Identification: ing conditions, the relative standard deviation of the peak
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of area of magnolol is not more than 1.5z.
pulverized Lycium Bark according to Method 3, and per-
form the test. Prepare the control solution with 3.0 mL of
Standard Lead Solution (not more than 10 ppm). Powdered Magnolia Bark
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of pulverized Lycium Bark according to Method 4, and per- コウボク末
form the test (not more than 5 ppm).
Change the Component determination to read:
Component determination Weigh accurately about 0.5 g
Magnolia Bark of Powdered Magnolia Bark, add 40 mL of diluted
methanol (7 in 10), heat under a reflux condenser on a water
コウボク bath for 20 minutes, cool, and filter. Repeat the above
procedure with the residue, using 40 mL of diluted methanol
Change the Component determination to read: (7 in 10). Combine the whole filtrates, add diluted methanol
Component determination Weigh accurately about 0.5 g (7 in 10) to make exactly 100 mL, and use this solution as the
of pulverized Magnolia Bark, add 40 mL of diluted sample solution. Separately, dry magnolol for component
methanol (7 in 10), heat under a reflux condenser on a water determination in a desiccator (silica gel) for 1 hour or more.
bath for 20 minutes, cool, and filter. Repeat the above Weigh accurately about 10 mg of it, dissolve in diluted
methanol (7 in 10) to make exactly 100 mL, and use this
1960 Crude Drugs Supplement I, JP XV

solution as the standard solution. Perform the test with ex- pulverized Notopterygium Rhizome according to Method 3,
actly 10 mL each of the sample solution and standard solu- and perform the test. Prepare the control solution with 3.0
tion as directed under Liquid Chromatography <2.01> ac- mL of Standard Lead Solution (not more than 10 ppm).
cording to the following conditions, and determine the peak (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
areas, AT and AS, of magnolol in each solution. of pulverized Notopterygium Rhizome according to Method
4, and perform the test (not more than 5 ppm).
Amount (mg) of magnolol=WS×(AT/AS)

WS: Amount (mg) of magnolol for component determina-


tion Nuphar Rhizome
Operating conditions—
センコツ
Detector: An ultraviolet absorption photometer (wave-
length: 289 nm).
Change the Purity to read:
Column: A stainless steel column 4 to 6 mm in inside di-
ameter and 15 to 25 cm in length, packed with octadecyl- Purity (1) Petiole—The amount of its petioles contained
silanized silica gel (5 to 10 mm in particle diameter). in Nuphar Rhizome does not exceed 3.0z.
Column temperature: A constant temperature of about (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
209C. of pulverized Nuphar Rhizome according to Method 4, and
Mobile phase: A mixture of water, acetonitrile and acetic perform the test (not more than 5 ppm).
acid (100) (50:50:1). (3) Foreign matter <5.01>—The amount of foreign mat-
Flow rate: Adjust the flow rate so that the retention time ter other than petiole is not more than 1.0z.
of magnolol is about 14 minutes.
System suitability—
System performance: Dissolve 1 mg each of magnolol for Nux Vomica Extract
component determination and honokiol in diluted methanol
(7 in 10) to make 10 mL. When the procedure is run with 10 ホミカエキス
mL of this solution under the above operating conditions,
honokiol and magnolol are eluted in this order with the reso- Add the following next to Identification:
lution between these peaks being not less than 5. Purity Heavy metals <1.07>—Prepare the test solution with
System repeatability: When the test is repeated 6 times 1.0 g of Nux Vomica Extract as directed in the Extracts (4)
with 10 mL of the standard solution under the above operat- under General Rules for Preparations, and perform the test
ing conditions, the relative standard deviation of the peak (not more than 30 ppm).
area of magnolol is not more than 1.5z.

Panax Japonicus Rhizome


Mulberry Bark
チクセツニンジン
ソウハクヒ
Add the following next to Identification:
Change the Purity to read:
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of pulverized Panax Japonicus Rhizome according to Method
pulverized Mulberry Bark according to Method 3, and per- 3, and perform the test. Prepare the control solution with
form the test. Prepare the control solution with 3.0 mL of 3.0 mL of Standard Lead Solution (not more than 10 ppm).
Standard Lead Solution (not more than 10 ppm). (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g of pulverized Panax Japonicus Rhizome according to
of pulverized Mulberry Bark according to Method 4, and Method 4, and perform the test (not more than 5 ppm).
perform the test (not more than 5 ppm).
(3) Foreign matter <5.01>—The amount of the root xy-
lem and other foreign matter is not more than 1.0z.
Powdered Panax Japonicus
Rhizome
Notopterygium Rhizome チクセツニンジン末
キョウカツ
Add the following next to Identification:
Add the following next to Identification: Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of Powdered Panax Japonicus Rhizome according to Method
Supplement I, JP XV Crude Drugs 1961

3, and perform the test. Prepare the control solution with


3.0 mL of Standard Lead Solution (not more than 10 ppm). Perilla Herb
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
of Powdered Panax Japonicus Rhizome according to ソヨウ
Method 4, and perform the test (not more than 5 ppm).
Change the Purity (1) to read:
Purity (1) Stem—Perilla Herb does not contain its stems
Peach Kernel equal to or greater than 3 mm in diameter.

トウニン

Add the following:


Change the Identification to read:
Identification To 1.0 g of ground Peach Kernel add 10 mL Peucedanum Root
of methanol, immediately heat under a reflux condenser on
a water bath for 10 minutes, cool, filter, and use the filtrate Peucedani Radix
as the sample solution. Separately, dissolve 2 mg of amygda-
ゼンコ
lin for thin-layer chromatography in 1 mL of methanol, and
use this solution as the standard solution. Perform the test
Peucedanum Root is the root of 1) Peucedanum
with these solutions as directed under Thin-layer Chro-
praeruptorum Dunn or 2) Angelica decursiva Franchet
matography <2.03>. Spot 10 mL each of the sample solution
et Savatier (Peucedanum decursivum Maximowicz)
and standard solution on a plate of silica gel for thin-layer
(Umbelliferae).
chromatography. Develop the plate with a mixture of ethyl
acetate, methanol and water (20:5:4) to a distance of about Description 1) Peucedanum praeruptorum Dunn
10 cm, and air-dry the plate. Spray evenly thymol-sulfuric Slender obconical to cylindrical root, occasionally
acid-methanol TS for spraying upon the plate, and heat at dichotomized at the lower part 3 to 15 cm in length, 0.8 to
1059 C for 5 minutes: one of the spot among the several spots 1.8 cm in diameter at the crown; externally light brown to
from the sample solution has the same color tone and Rf dark brown; ring-node-like wrinkles numerous at the crown,
value with the red-brown spot from the standard solution. sometimes with hair-like remains of petioles; the root having
somewhat deep longitudinal wrinkles and scars of cutting
off of lateral roots; cross section surface light brown to whit-
Powdered Peach Kernel ish in color; brittle in texture.
Odor, characteristic; taste, slightly bitter.
トウニン末 Under a microscope <5.01>, a transverse section reveals
the outermost layer composed of a cork layer, inner tangen-
Change the Identification to read: tial walls of some cork cells thickened; collenchyma just in-
side of the cork layer; in cortex numerous oil canals scat-
Identification (1) Grind Powdered Peach Kernel with
tered and intercellular air spaces observed; occasionally
water: the odor of benzaldehyde is perceptible.
phloem fibers observed at the terminal portion of phloem;
(2) To 1.0 g of Powdered Peach Kernel add 10 mL of
vessels and scattered oil canals in xylem; starch grains in
methanol, and immediately heat under a reflux condenser on
parenchyma, 2 to 10 several-compound grains.
a water bath for 10 minutes. After cooling, filter, and use
2) Angelica decursiva Franchet et Savatier
the filtrate as the sample solution. Separately, dissolve 2 mg
Similar to 1), but without hair-like remains of petioles at
of amygdalin for thin-layer chromatography in 1 mL of
the crown.
methanol, and use this solution as the standard solution.
Under a microscope <5.01>, a transverse section reveals,
Perform the test with these solutions as directed under Thin-
similar to 1), but cell wall of cork cells not thickened,
layer Chromatography <2.03>. Spot 10 mL each of the
phloem fibers not observed at the terminal portion of
sample solution and standard solution on a plate of silica gel
phloem, nor oil canals observed in xylem.
for thin-layer chromatography. Develop the plate with a
mixture of ethyl acetate, methanol and water (20:5:4) to a Identification (1) Peucedanum praeruptorum Dunn—To
distance of about 10 cm, and air-dry the plate. Spray evenly 1 g of pulverized Peucedanum Root add 10 mL of methanol,
thymol-sulfuric acid-methanol TS for spraying on the plate, shake for 10 minutes, centrifuge, and use the supernatant
and heat at 1059 C for 5 minutes: one of the spot among the liquid as the sample solution. Separately, dissolve 1 mg of
several spots from the sample solution has the same color (±)-praeruptorin A for thin-layer chromatography in 1 mL
tone and Rf value with the red-brown spot from the stan- of methanol, and use this solution as the standard solution.
dard solution. Perform the test with these solutions as directed under Thin-
layer Chromatography <2.03>. Spot 10 mL each of the sam-
ple solution and standard solution on a plate of silica gel for
thin-layer chromatography. Develop the plate with a mix-
1962 Crude Drugs Supplement I, JP XV

ture of diethyl ether and hexane (3:1) to a distance of about of pulverized Polygala Root according to Method 4, and
10 cm, and air-dry the plate. Examine under ultraviolet light perform the test (not more than 5 ppm).
(main wavelength: 365 nm): one of the spot among the sever- (4) Foreign matter <5.01>—The amount of foreign mat-
al spots from the sample solution has the same color tone ter other than the stems is not more than 1.0z.
and Rf value with the blue-purple fluorescent spot from the (5) Total BHC's and total DDT's <5.01>—Not more
standard solution. than 0.2 ppm, respectively.
(2) Angelica decursiva Franchet et Savatier—To 1 g of
pulverized Peucedanum Root add 10 mL of methanol, shake
for 10 minutes, centrifuge, and use the supernatant liquid as Powdered Polygala Root
the sample solution. Separately, dissolve 1 mg of nodakenin
for thin-layer chromatography in 1 mL of methanol, and use オンジ末
this solution as the standard solution. Perform the test with
these solutions as directed under Thin-layer Chro- Change the Purity to read:
matography <2.03>. Spot 10 mL each of the sample solution
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
and standard solution on a plate of silica gel for thin-layer
Powdered Polygala Root according to Method 3, and per-
chromatography. Develop the plate with a mixture of ethyl
form the test. Prepare the control solution with 3.0 mL of
acetate, methanol and water (12:2:1) to a distance of about
Standard Lead Solution (not more than 10 ppm).
10 cm, and air-dry the plate. Examine under ultraviolet light
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
(main wavelength: 365 nm): one of the spot among the sever-
of Powdered Polygala Root according to Method 4, and
al spots from the sample solution has the same color tone
perform the test (not more than 5 ppm).
and Rf value with the purple fluorescent spot from the stan-
(3) Foreign matter—Under a microscope <5.01>, Pow-
dard solution.
dered Polygala Root does not show stone cells or starch
Loss on drying <5.01> Not more than 13.0z. grains.
(4) Total BHC's and total DDT's <5.01>—Not more
Total ash <5.01> Not more than 7.0z.
than 0.2 ppm, respectively.
Acid-insoluble ash <5.01> Not more than 2.0z.

Extract content <5.01> Dilute ethanol-soluble extract: not


less than 20.0z.
Polygonum Root
カシュウ

Platycodon Fluidextract Add the following next to Identification:


キキョウ流エキス Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
pulverized Polygonum Root according to Method 3, and
Change the Purity to read: perform the test. Prepare the control solution with 3.0 mL
of Standard Lead Solution (not more than 10 ppm).
Purity (1) Heavy metals <1.07>—Prepare the test solution
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
with 1.0 g of Platycodon Fluidextract as directed in the
of pulverized Polygonum Root according to Method 4, and
Fluidextracts (4) under General Rules for Preparations, and
perform the test (not more than 5 ppm).
perform the test (not more than 30 ppm).
(2) Starch—Mix 1 mL of Platycodon Fluidextract with 4
mL of water, and add 1 drop of dilute iodine TS: no purple
or blue color develops.
Polyporus Sclerotium
チョレイ

Polygala Root Add the following next to Identification:


オンジ Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
pulverized Polyporus Sclerotium according to Method 3,
Change the Purity to read: and perform the test. Prepare the control solution with 3.0
mL of Standard Lead Solution (not more than 10 ppm).
Purity (1) Stem—The amount of the stems contained in
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
Polygala Root does not exceed 10.0z.
of pulverized Polyporus Sclerotium according to Method 4,
(2) Heavy metals <1.07>—Proceed with 3.0 g of pulver-
and perform the test (not more than 5 ppm).
ized Polygala Root according to Method 3, and perform the
test. Prepare the control solution with 3.0 mL of Standard
Lead Solution (not more than 10 ppm).
(3) Arsenic <1.11>—Prepare the test solution with 0.40 g
Supplement I, JP XV Crude Drugs 1963

Amount (mg) of jesaconitine (C35H49NO12)


Powdered Polyporus Sclerotium =(CSJ/W)×(HTJ/HSJ)×10
チョレイ末 Amount (mg) of hypaconitine (C33H45NO10)
=(CSH/W)×(HTH/HSH)×10
Add the following next to Identification:
Amount (mg) of mesaconitine (C33H45NO11)
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of =(CSM/W)×(HTM/HSM)×10
Powdered Polyporus Sclerotium according to Method 3,
CSA: Concentration (mg/mL) of aconitine for purity in
and perform the test. Prepare the control solution with 3.0
aconitum diester alkaloids standard solution for
mL of Standard Lead Solution (not more than 10 ppm).
purity
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
CSJ: Concentration (mg/mL) of jesaconitine for purity in
of Powdered Polyporus Sclerotium according to Method 4,
aconitum diester alkaloids standard solution for puri-
and perform the test (not more than 5 ppm).
ty
CSH: Concentration (mg/mL) of hypaconitine for purity in
aconitum diester alkaloids standard solution for
Processed Aconite Root purity
ブシ CSM: Concentration (mg/mL) of mesaconitine for purity
in aconitum diester alkaloids standard solution for
Change the Purity to read: purity
W: Amount (g) of sample, calculated on the dried basis
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
pulverized Processed Aconite Root according to Method 3, Operating conditions—
and perform the test. Prepare the control solution with 3.0 Detector: An ultraviolet absorption photometer (wave-
mL of Standard Lead Solution (not more than 10 ppm). length: 231 nm for aconitine, hypaconitine and mesaconi-
(2) Arsenic <1.11>—Prepare the test solution with 0.4 g tine; 254 nm for jesaconitine).
of pulverized Processed Aconite Root according to Method Column: A stainless steel column 4.6 mm in inside di-
4, and perform the test (not more than 5 ppm). ameter and 15 cm in length, packed with octadecylsilanized
(3) Aconitum diester alkaloids (aconitine, jesaconitine, silica gel for liquid chromatography (5 mm in particle di-
hypaconitine and mesaconitine)—Weigh accurately about ameter).
0.5 g of pulverized Processed Aconite Root, put in a glass- Column temperature: A constant temperature of about
stoppered centrifuge tube, suspend in 3.0 mL of water by 409C.
shaking, and add 1.0 mL of ammonia TS and 20 mL of Mobile phase: A mixture of phosphate buffer solution for
diethyl ether. Stopper tightly the tube, shake for 30 minutes, processed aconite root and tetrahydrofuran (183:17).
centrifuge, and separate the ether layer. To the residue add Flow rate: Adjust the flow rate so that the retention time
1.0 mL of ammonia TS and 20 mL of diethyl ether, and of mesaconitine is about 31 minutes.
repeat the above process twice more. Combine all extracts, System suitability—
evaporate to dryness under reduced pressure below 409C, System performance: When the procedure is run with 20
and dissolve the residue with exactly 10 mL of a mixture of mL of aconitum diester alkaloids standard solution for puri-
phosphate buffer solution for processed aconite root and ty under the above operating conditions, using 254 nm,
acetonitrile (1:1). Centrifuge this solution, and use the su- mesaconitine, hypaconitine, aconitine and jesaconitine are
pernatant liquid as the sample solution. Perform the test eluted in this order, and each resolution between their peaks
with exactly 20 mL each of the sample solution and aconitum is not less than 1.5, respectively.
diester alkaloids standard solution for purity as directed un- System repeatability: To 1 mL of aconitum diester
der Liquid Chromatography <2.01> according to the follow- alkaloids standard solution for purity add a mixture of phos-
ing conditions, and determine the heights of the peaks cor- phate buffer solution for processed aconite root and acetoni-
responding to aconitine, HTA and HSA, jesaconitine, HTJ and trile (1:1) to make 10 mL. When the test is repeated 6 times
HSJ, hypaconitine, HTH and HSH, and mesaconitine, HTM with 20 mL of this solution under the above operating condi-
and HSM, respectively, and calculate the amounts of them by tions, using 231 nm, the relative standard deviation of the
the following formulae: the amounts of aconitine, jesaconi- peak height of mesaconitine is not more than 1.5z.
tine, hypaconitine and mesaconitine per g calculated on the
dried basis are not more than 60 mg, 60 mg, 280 mg and 140
mg, respectively, and the total amount of them is not more Powdered Processed Aconite Root
than 450 mg.
ブシ末
Amount (mg) of aconitine (C34H47NO11)
=(CSA/W)×(HTA/HSA)×10 Change the Purity to read:
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
1964 Crude Drugs Supplement I, JP XV

Powdered Processed Aconite Root according to Method 3, Operating conditions—


and perform the test. Prepare the control solution with 3.0 Detector: An ultraviolet absorption photometer (wave-
mL of Standard Lead Solution (not more than 10 ppm). length: 231 nm for aconitine, hypaconitine and mesaconi-
(2) Arsenic <1.11>—Prepare the test solution with 0.4 g tine; 254 nm for jesaconitine).
of Powdered Processed Aconite Root according to Method Column: A stainless steel column 4.6 mm in inside di-
4, and perform the test (not more than 5 ppm). ameter and 15 cm in length, packed with octadecylsilanized
(3) Aconitum diester alkaloids (aconitine, jesaconitine, silica gel for liquid chromatography (5 mm in particle di-
hypaconitine and mesaconitine)—Weigh accurately about ameter).
0.5 g of Powdered Processed Aconite Root, put in a glass- Column temperature: A constant temperature of about
stoppered centrifuge tube, suspend in 3.0 mL of water by 409C.
shaking, and add 1.0 mL of ammonia TS and 20 mL of Mobile phase: A mixture of phosphate buffer solution for
diethyl ether. Stopper tightly the tube, shake for 30 minutes, processed aconite root and tetrahydrofuran (183:17).
centrifuge, and separate the ether layer. To the residue add Flow rate: Adjust the flow rate so that the retention time
1.0 mL of ammonia TS and 20 mL of diethyl ether, and of mesaconitine is about 31 minutes.
repeat the above process two times. Combine all extracts, System suitability—
evaporate to dryness under reduced pressure below 409C, System performance: When the procedure is run with 20
and dissolve the residue with exactly 10 mL of a mixture of mL of aconitum diester alkaloids standard solution for puri-
phosphate buffer solution for processed aconite root and ty under the above operating conditions, using 254 nm,
acetonitrile (1:1). Centrifuge this solution, and use the su- mesaconitine, hypaconitine, aconitine and jesaconitine are
pernatant liquid as the sample solution. Perform the test eluted in this order, and each resolution between their peaks
with exactly 20 mL each of the sample solution and aconitum is not less than 1.5, respectively.
diester alkaloids standard solution for purity as directed un- System repeatability: To 1 mL of aconitum diester
der Liquid Chromatography <2.01> according to the follow- alkaloids standard solution for purity add a mixture of phos-
ing conditions, and determine the heights of the peaks cor- phate buffer solution for processed aconite root and acetoni-
responding to aconitine, HTA and HSA, jesaconitine, HTJ and trile (1:1) to make 10 mL. When the test is repeated 6 times
HSJ, hypaconitine, HTH and HSH, and mesaconitine, HTM with 20 mL of this solution under the above operating condi-
and HSM, respectively, and calculate the amounts of them by tions, using 231 nm, the relative standard deviation of the
the following formulae: the amounts of aconitine, jesaconi- peak height of mesaconitine is not more than 1.5z.
tine, hypaconitine and mesaconitine per g calculated on the
dried basis are not more than 55 mg, 40 mg, 55 mg and 120 mg,
respectively, and the total amount of them is not more than Processed Ginger
230 mg.
カンキョウ
Amount (mg) of aconitine (C34H47NO11)
=(CSA/W)×(HTA/HSA)×10
Add the following next to Identification:
Amount (mg) of jesaconitine (C35H49NO12)
Purity Arsenic <1.11>—Prepare the test solution with 0.40
=(CSJ/W)×(HTJ/HSJ)×10
g of pulverized Processed Ginger according to Method 4,
Amount (mg) of hypaconitine (C33H45NO10) and perform the test (not more than 5 ppm).
=(CSH/W)×(HTH/HSH)×10

Amount (mg) of mesaconitine (C33H45NO11)


=(CSM/W)×(HTM/HSM)×10
Rehmannia Root
CSA: Concentration (mg/mL) of aconitine for purity in ジオウ
aconitum diester alkaloids standard solution for
purity Change the Description to read:
CSJ: Concentration (mg/mL) of jesaconitine for purity in Description Thin and long, usually, fusiform root, 5 – 10
aconitum diester alkaloids standard solution for puri- cm in length, 0.5 – 3.0 cm in diameter, often broken or mar-
ty kedly deformed in shape; externally yellow-brown to black-
CSH: Concentration (mg/mL) of hypaconitine for purity in ish brown, with deep, longitudinal wrinkles and constric-
aconitum diester alkaloids standard solution for tions; soft in texture and mucous; cross section yellow-
purity brown to blackish brown, and cortex darker than xylem in
CSM: Concentration (mg/mL) of mesaconitine for purity color; pith hardly observable.
in aconitum diester alkaloids standard solution for Odor, characteristic; taste, slightly sweet at first, followed
purity by a slight bitterness.
W: Amount (g) of the sample, calculated on the dried ba- Under a microscope <5.01>, a transverse section reveals 7
sis to 15 layers of cork; cortex composed entirely of parenchy-
ma cells; outer region of cortex with scattered cells contain-
Supplement I, JP XV Crude Drugs 1965

ing brown secretes; xylem practically filled with parenchy- cm, and air-dry the plate. Spray evenly dilute sulfuric acid
ma; vessels radially lined, mainly reticulate vessels. on the plat, heat at 1059 C for 5 minutes, and examine under
ultraviolet light (main wavelength: 365 nm): one of the spot
among the several spots from the sample solution has the
Change to read: same color tone and Rf value with the bluish white fluores-
cent spot from the standard solution (Atractylodes Rhizome).
Ryokeijutsukanto Extract (3) For preparation prescribed Atractylodes Lancea
Rhizome—To 2.0 g of dry extract (6.0 g for viscous extract)
苓桂朮甘湯エキス of Ryokeijutsukanto Extract add 10 mL of water, shake,
then add 25 mL of hexane, and shake. Take the hexane lay-
Ryokeijutsukanto Extract contains not less than 1 er, add anhydrous sodium sulfate to dry, and filter.
mg and not more than 4 mg of (E)-cinnamic acid, and Evaporate the filtrate under reduced pressure, add 2 mL of
not less than 21 mg and not more than 63 mg of hexane to the residue, and use this solution as the sample so-
glycyrrhizic acid (C42H62O16: 822.93) per a dried ex- lution. Perform the test with the sample solution as directed
tract prepared as directed in the Method of prepara- under Thin-layer Chromatography <2.03>. Spot 20 mL of the
tion. sample solution on a plate of silica gel with fluorescent indi-
cator for thin-layer chromatography, develop the plate with
Method of preparation Prepare a dry or viscous extract as
a mixture of hexane and acetone (7:1) to a distance of about
directed under Extracts, with 6 g of Poria Sclerotium, 4 g of
10 cm, and air-dry the plate. Examine under ultraviolet light
Cinnamon Bark, 3 g of Atractylodes Rhizome or Atrac-
(main wavelength: 254 nm): a dark purple spot is observed at
tylodes Lancea Rhizome and 2 g of Glycyrrhiza.
around Rf 0.4. The spot shows a greenish brown color after
Description Ryokeijutsukanto Extract occurs as a brown being sprayed 4-dimethylaminobenzaldehyde TS for
to black-brown powder or viscous extract. It has an odor, spraying, heated at 1059 C for 5 minutes, and allowed to cool
and a sweet first then bitter taste. (Atractylodes Lancea Rhizome).
(4) To 1.0 g of dry extract (3.0 g for viscous extract) of
Identification (1) To 1.0 g of dry extract (3.0 g for vis-
Ryokeijutsukanto Extract add 10 mL of water, shake, then
cous extract) of Ryokeijutsukanto Extract add 10 mL of
add 10 mL of 1-butanol, centrifuge, and use the supernatant
water, shake, then add 25 mL of diethyl ether, and shake.
liquid as the sample solution. Separately, dissolve 1 mg of li-
Take the diethyl ether layer, evaporate the layer under
quiritin for thin-layer chromatography in 1 mL of methanol,
reduced pressure, add 2 mL of diethyl ether to the residue,
and use this solution as the standard solution. Perform the
and use this solution as the sample solution. Separately, dis-
test with these solutions as directed under Thin-layer Chro-
solve 1 mg of (E)-cinnamic acid for thin-layer chro-
matography <2.03>. Spot 5 mL each of the sample solution
matography in 1 mL of methanol, and use this solution as
and standard solution on a plate of silica gel for thin-layer
the standard solution. Perform the test with these solutions
chromatography. Develop the plate with a mixture of ethyl
as directed under Thin-layer Chromatography <2.03>. Spot 5
acetate, methanol and water (20:3:2) to a distance of about
mL each of the sample solution and standard solution on a
10 cm, and air-dry the plate. Spray evenly dilute sulfuric acid
plate of silica gel with fluorescent indicator for thin-layer
on the plate, and heat at 1059 C for 5 minutes: one of the
chromatography, develop the plate with a mixture of
spot among the several spots from the sample solution has
hexane, ethyl acetate, formic acid and water (60:40:4:1) to a
the same color tone and Rf value with the yellow-brown spot
distance of about 10 cm, and air-dry the plate. Examine un-
from the standard solution (Glycyrrhiza).
der ultraviolet light (main wavelength: 254 nm): one of the
spot among the several spots from the sample solution has Purity (1) Heavy metals <1.07>—Prepare the test solution
the same color tone and Rf value with the blue-purple spot with 1.0 g of dry extract (or an amount of the viscous ex-
from the standard solution (Cinnamon Bark). tract, equivalent to 1.0 g of the dried substance) of
(2) For preparation prescribed Atractylodes Rhizome— Ryokeijutsukanto Extract as directed in the Extracts (4) un-
To 1.0 g of dry extract (3.0 g for viscous extract) of der General Rules for Preparations, and perform the test
Ryokeijutsukanto Extract add 10 mL of water, shake, then (not more than 30 ppm).
add 25 mL of diethyl ether, and shake. Take the diethyl (2) Arsenic <1.11>—Prepare the test solution with 0.67 g
ether layer, evaporate the layer under reduced pressure, add of dry extract (or an amount of the viscous extract, equiva-
2 mL of diethyl ether to the residue, and use this solution as lent to 0.67 g of the dried substance) of Ryokeijutsukanto
the sample solution. Separately, dissolve 1 mg of atrac- Extract according to Method 3, and perform the test (not
tylenolide III for thin-layer chromatography in 2 mL of more than 3 ppm).
methanol, and use this solution as the standard solution.
Loss on drying <2.41> Dry extract: Not more than 8.5z
Perform the test with these solutions as directed under Thin-
(1 g, 1059C, 5 hours).
layer Chromatography <2.03>. Spot 5 mL each of the sample
Viscous extract: Not more than 66.7z (1 g 1059C, 5
solution and standard solution on a plate of silica gel for
hours).
thin-layer chromatography, develop the plate with a mixture
of ethyl acetate and hexane (1:1) to a distance of about 10 Total ash <5.01> Not more than 8.0z, calculated on the
1966 Crude Drugs Supplement I, JP XV

dried basis. matography <2.01> according to the following conditions,


and determine the peak areas, AT and AS, of glycyrrhizic
Assay (1) (E)-Cinnamic acid—Conduct this procedure
acid.
without exposure to light, using light-resistant vessels.
Weigh accurately about 0.5 g of dry extract (for viscous ex- Amount (mg) of glycyrrhizic acid (C42H62O16)
tract an amount equivalent to about 0.5 g as dried sub- =WS×(AT/AS)×(1/2)
stance) of Ryokeijutsukanto Extract, add exactly 50 mL of
WS: Amount (mg) of Glycyrrhizic Acid Reference Stan-
diluted methanol (1 in 2), shake for 15 minutes, filter, and
dard, calculated on the anhydrous basis
use the filtrate as the sample solution. Separately, weigh ac-
curately about 10 mg of (E)-cinnamic acid for component Operating conditions—
determination, previously dried in a desiccator (silica gel) for Detector: An ultraviolet absorption photometer (wave-
more than 24 hours, dissolve in diluted methanol (1 in 2) to length: 254 nm).
make exactly 100 mL. Pipet 10 mL of this solution, add Column: A stainless steel column 4.6 mm in inside di-
diluted methanol (1 in 2) to make exactly 100 mL, and use ameter and 15 cm in length, packed with octadecylsilanized
this solution as the standard solution. Perform the test with silica gel for liquid chromatography (5 mm in particle di-
exactly 10 mL each of the sample solution and standard solu- ameter).
tion as directed under Liquid Chromatography <2.01> ac- Column temperature: A constant temperature of about
cording to the following conditions, and determine the peak 409C.
areas, AT and AS, of (E)-cinnamic acid. Mobile phase: A mixture of diluted acetic acid (31) (1 in
15) and acetonitrile (13:7).
Amount (mg) of (E)-cinnamic acid
Flow rate: 1.0 mL per minute. (the retention time of
=WS×(AT/AS)×(1/20)
glycyrrhizic acid is about 12 minutes.)
WS: Amount (mg) of (E)-cinnamic acid for component System suitability—
determination System performance: When the procedure is run with 10
mL of the standard solution under the above operating con-
Operating conditions—
ditions, the number of theoretical plates and the symmetry
Detector: An ultraviolet absorption photometer (wave-
factor of the peak of glycyrrhizic acid are not less than 5000
length: 273 nm).
and not more than 1.5, respectively.
Column: A stainless steel column 4.6 mm in inside di-
System repeatability: When the test is repeated 6 times
ameter and 15 cm in length, packed with octadecylsilanized
with 10 mL of the standard solution under the above operat-
silica gel for liquid chromatography (5 mm in particle di-
ing conditions, the relative standard deviation of the peak
ameter).
area of glycyrrhizic acid is not more than 1.5z.
Column temperature: A constant temperature of about
409C. Containers and storage Containers—Tight containers.
Mobile phase: A mixture of water, acetonitrile and phos-
phoric acid (750:250:1).
Flow rate: 1.0 mL per minute. (the retention time of (E)- Saposhnikovia Root
cinnamic acid is about 12 minutes.)
System suitability— ボウフウ
System performance: When the procedure is run with 10
mL of the standard solution under the above operating con- Change the Purity to read:
ditions, the number of theoretical plates and the symmetry
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
factor of the peak of (E)-cinnamic acid are not less than 5000
pulverized Saposhnikovia Root according to Method 3, and
and not more than 1.5, respectively.
perform the test. Prepare the control solution with 3.0 mL
System repeatability: When the test is repeated 6 times
of Standard Lead Solution (not more than 10 ppm).
with 10 mL of the standard solution under the above operat-
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
ing conditions, the relative standard deviation of the peak
of pulverized Saposhnikovia Root according to Method 4,
area of (E)-cinnamic acid is not more than 1.5z.
and perform the test (not more than 5 ppm).
(2) Glycyrrhizic acid—Weigh accurately about 0.5 g of
(3) Foreign matter <5.01>—The amount of stems and
dry extract (for viscous extract an amount equivalent to
other foreign matter is not more than 2.0z.
about 0.5 g as dried substance) of Ryokeijutsukanto Ex-
tract, add exactly 50 mL of diluted methanol (1 in 2), shake
for 15 minutes, filter, and use the filtrate as the sample solu-
tion. Separately, weigh accurately about 10 mg of Glycyr-
Saussurea Root
rhizic Acid Reference Standard (separately determin the モッコウ
water), dissolve in diluted methanol (1 in 2) to make exactly
100 mL, and use this solution as the standard solution. Per-
Change the Purity to read:
form the test with exactly 10 mL each of the sample solution
and standard solution as directed under Liquid Chro- Purity (1) Arsenic <1.11>—Prepare the test solution with
Supplement I, JP XV Crude Drugs 1967

0.40 g of pulverized Saussurea Root according to Method 4,


Amount (mg) of baicalin (C21H18O11)=WS×(AT/AS)×5
and perform the test (not more than 5 ppm).
(2) Foreign matter—Add iodine TS dropwise to a trans- WS: Amount (mg) of Baicalin Reference Standard, calcu-
verse section: no blue-purple color develops. lated on the anhydrous basis

Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Scopolia Extract length: 277 nm).
Column: A stainless steel column 4.6 mm in inside di-
ロートエキス
ameter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle di-
Add the following next to Identification:
ameter).
Purity Heavy metals <1.07>—Prepare the test solution with Column temperature: A constant temperature of about
1.0 g of Scopolia Extract as directed in the Extracts (4) under 509C.
General Rules for Preparations, and perform the test (not Mobile phase: A mixture of diluted phosphoric acid (1 in
more than 30 ppm). 146) and acetonitrile (18:7).
Flow rate: Adjust the flow rate so that the retention time
of baicalin is about 6 minutes.
Scopolia Rhizome System suitability—
System performance: Dissolve 1 mg of Baicalin Reference
ロートコン Standard and 2 mg of methyl parahydroxybenzoate in
methanol to make 100 mL. When the procedure is run with
Add the following next to Identification: 10 mL of this solution under the above operating conditions,
Purity Arsenic <1.11>—Prepare the test solution with 0.40 baicalin and methyl parahydroxybenzoate are eluted in this
g of pulverized Scopolia Rhizome according to Method 4, order with the resolution between these peaks being not less
and perform the test (not more than 5 ppm). than 3.
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 peak
Scutellaria Root area of baicalin is not more than 1.5z.
オウゴン

Change the Assay to read: Powdered Scutellaria Root


Assay Weigh accurately about 0.5 g of pulverized Scutel- オウゴン末
laria Root, add 30 mL of diluted methanol (7 in 10), heat
under a reflux condenser on a water bath for 30 minutes, Change the Assay to read:
and cool. Transfer the mixture to a glass-stoppered cen-
trifuge tube, centrifuge, and separate the supernatant liquid. Assay Weigh accurately about 0.5 g of Powdered Scutel-
Wash the vessel for the reflux extraction with 30 mL of dilut- laria Root, add 30 mL of diluted methanol (7 in 10), heat
ed methanol (7 in 10), transfer the washings to the glass- under a reflux condenser on a water bath for 30 minutes,
stoppered centrifuge tube, centrifuge after shaking for 5 and cool. Transfer the mixture to a glass-stoppered cen-
minutes, and separate the supernatant liquid. To the residue trifuge tube, centrifuge, and separate the supernatant liquid.
add 30 mL of diluted methanol (7 in 10), shake for 5 Wash the vessel for the reflux extraction with 30 mL of dilut-
minutes, centrifuge, and separate the supernatant liquid. ed methanol (7 in 10), transfer the washings to the glass-
Combine all the extracts, add diluted methanol (7 in 10) to stoppered centrifuge tube, centrifuge after shaking for 5
make exactly 100 mL, then pipet 2 mL of the extract, add minutes, and separate the supernatant liquid. To the residue
diluted methanol (7 in 10) to make exactly 20 mL, and use add 30 mL of diluted methanol (7 in 10), shake for 5
this solution as the sample solution. Separately, weigh ac- minutes, centrifuge, and separate the supernatant liquid.
curately about 10 mg of Baicalin Reference Standard Combine all the extracts, add diluted methanol (7 in 10) to
(separately determine the water), and dissolve in methanol to make exactly 100 mL, then pipet 2 mL of the extract, add
make exactly 20 mL. Pipet 2 mL of the solution, add diluted diluted methanol (7 in 10) to make exactly 20 mL, and use
methanol (7 in 10) to make exactly 20 mL, and use this solu- this solution as the sample solution. Separately, weigh ac-
tion as the standard solution. Perform the test with exactly curately about 10 mg of Baicalin Reference Standard
10 mL each of the sample solution and standard solution as (separately determine the water), and dissolve in methanol to
directed under Liquid Chromatography <2.01> according to make exactly 20 mL. Pipet 2 mL of the solution, add diluted
the following conditions. Determine the peak areas, AT and methanol (7 in 10) to make exactly 20 mL, and use this solu-
AS, of baicalin in each solution. tion as the standard solution. Perform the test with exactly
10 mL each of the sample solution and standard solution as
1968 Crude Drugs Supplement I, JP XV

directed under Liquid Chromatography <2.01> according to form the test (not more than 5 ppm).
the following conditions. Determine the peak areas, AT and (2) Foreign matter—Under a microscope <5.01>, stone
AS, of baicalin in each solution. cells, starch grains or crystals of calcium oxalate are not ob-
servable.
Amount (mg) of baicalin (C21H18O11)=WS×(AT/AS)×5

WS: Amount (mg) of Baicalin Reference Standard, calcu-


lated on the anhydrous basis Smilax Rhizome
Operating conditions—
サンキライ
Detector: An ultraviolet absorption photometer (wave-
length: 277 nm).
Add the following next to Description:
Column: A stainless steel column 4.6 mm in inside di-
ameter and 15 cm in length, packed with octadecylsilanized Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
silica gel for liquid chromatography (5 mm in particle di- pulverized Smilax Rhizome according to Method 3, and per-
ameter). form the test. Prepare the control solution with 3.0 mL of
Column temperature: A constant temperature of about Standard Lead Solution (not more than 10 ppm).
509C. (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
Mobile phase: A mixture of diluted phosphoric acid (1 in of pulverized Smilax Rhizome according to Method 4, and
146) and acetonitrile (18:7). perform the test (not more than 5 ppm).
Flow rate: Adjust the flow rate so that the retention time
of baicalin is about 6 minutes.
System suitability— Powdered Smilax Rhizome
System performance: Dissolve 1 mg of Baicalin Reference
Standard and 2 mg of methyl parahydroxybenzoate in サンキライ末
methanol to make 100 mL. When the procedure is run with
10 mL of this solution under the above operating conditions, Change the Purity to read:
baicalin and methyl parahydroxybenzoate are eluted in this Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
order with the resolution between these peaks being not less Powdered Smilax Rhizome according to Method 3, and per-
than 3. form the test. Prepare the control solution with 3.0 mL of
System repeatability: When the test is repeated 6 times Standard Lead Solution (not more than 10 ppm).
with 10 mL of the standard solution under the above operat- (2) Arsenic <1.11>—Prepare the test solution with 0.40 g
ing conditions, the relative standard deviation of the peak of Powdered Smilax Rhizome according to Method 4, and
area of baicalin is not more than 1.5z. perform the test (not more than 5 ppm).
(3) Foreign matter—Under a microscope <5.01>, Pow-
dered Smilax Rhizome does not show a large quantity of
Senega stone cells or thick-walled fibers.

セネガ

Change the Purity to read:


Sophora Root
Purity (1) Stem—The amount of stems contained in クジン
Senega is not more than 2.0z.
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g Change the Purity to read:
of pulverized Senega according to Method 4, and perform Purity (1) Stem—The amount of its stems contained in
the test (not more than 5 ppm). Sophora Root does not exceed 10.0z.
(3) Foreign matter <5.01>—The amount of foreign mat- (2) Heavy metals <1.07>—Proceed with 3.0 g of pulver-
ter other than stems contained in Senega is not more than 1.0 ized Sophora Root according to Method 3, and perform the
z. test. Prepare the control solution with 3.0 mL of Standard
Lead Solution (not more than 10 ppm).
(3) Arsenic <1.11>—Prepare the test solution with 0.40 g
Powdered Senega of pulverized Sophora Root according to Method 4, and per-
form the test (not more than 5 ppm).
セネガ末
(4) Foreign matter <5.01>—The amount of foreign mat-
ter other than stems is not more than 1.0z.
Change the Purity to read:
Purity (1) Arsenic <1.11>—Prepare the test solution with
0.40 g of Powdered Senega according to Method 4, and per-
Supplement I, JP XV Crude Drugs 1969

spot appears at around Rf 0.4.


Powdered Sophora Root Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
Powdered Turmeric according to Method 3, and perform
クジン末
the test. Prepare the control solution with 3.0 mL of Stan-
dard Lead Solution (not more than 10 ppm).
Add the following next to Identification:
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of of Powdered Turmeric according to Method 4, and perform
Powdered Sophora Root according to Method 3, and per- the test (not more than 5 ppm).
form the test. Prepare the control solution with 3.0 mL of
Loss on drying <5.01> Not more than 17.0z (6 hours).
Standard Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g Total ash <5.01> Not more than 7.5z.
of Powdered Sophora Root according to Method 4, and per-
Acid-insoluble ash <5.01> Not more than 1.0z.
form the test (not more than 5 ppm).
Extract content <5.01> Dilute ethanol-soluble extract: not
less than 9.0z.
Turmeric
ウコン
Uva Ursi Fluidextract
Add the following next to Identification: ウワウルシ流エキス
Purity (1) Heavy metals <1.07>—Proceed with 3.0 g of
pulverized Turmeric according to Method 3, and perform
Add the following next to Identification:
the test. Prepare the control solution with 3.0 mL of Stan- Purity Heavy metals <1.07>—Prepare the test solution with
dard Lead Solution (not more than 10 ppm). 1.0 g of Uva Ursi Fluidextract as direct in the Fluidextracts
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g (4) under General Rules for Preparations, and perform the
of pulverized Turmeric according to Method 4, and perform test (not more than 30 ppm).
the test (not more than 5 ppm).

Zedoary
Add the following:
ガジュツ
Powdered Turmeric
Add the following next to Description:
Curcumae Rhizoma Purveratum
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
ウコン末 pulverized Zedoary according to Method 3, and perform the
test. Prepare the control solution with 1.0 mL of Standard
Powdered Turmeric is the powder of Turmeric. Lead Solution (not more than 10 ppm).
(2) Arsenic <1.11>—Prepare the test solution with 0.40 g
Description Powdered Turmeric occurs as a yellow-brown
of pulverized Zedoary according to Method 4, and perform
to dark yellow-brown powder. It has a characteristic odor
the test (not more than 5 ppm).
and a bitter, stimulant taste, and colors the saliva yellow on
chewing.
Under a microscope <5.01>, all elements are yellow in
color; it reveals parenchymatous cells containing mainly
masses of gelatinized starch or yellow substances, also frag-
ments of scalariform vessels; fragments of cork layers,
epidermis, thick-walled xylem parenchymatous cells, and
non-glandular hairs are occasionally observed.

Identification To 0.5 g of Powdered Turmeric add 20 mL


of methanol, shake for 15 minutes, filter, and use the filtrate
as the sample solution. Perform the test with the sample so-
lution as directed under Thin-layer Chromatography <2.03>.
Spot 5 mL of the sample solution on a plate of silica gel for
thin-layer chromatography. Develop the plate with a mix-
ture of ethyl acetate, hexane and acetic acid (100) (70:30:1)
to a distance of about 10 cm, and air-dry the plate: a yellow
Supplement I, JP XV Infrared Reference Spectra 1971

Delete the following Infrared Reference Spectra:

Fosfestrol
Sulˆnpyrazone
1972 Infrared Reference Spectra Supplement I, JP XV

Change to read the following 2 spectra:

Calcium Folinate

Vincristine Sulfate
Supplement I, JP XV Infrared Reference Spectra 1973

Add the following 42 spectra:

Acemetacin

Alminoprofen

Amlexanox
1974 Infrared Reference Spectra Supplement I, JP XV

Amlodipine Besilate

Amosulalol Hydrochloride

Azelastine Hydrochloride
Supplement I, JP XV Infrared Reference Spectra 1975

Biotin

Bisoprolol Fumarate

Buformin Hydrochloride
1976 Infrared Reference Spectra Supplement I, JP XV

Buprenorphine Hydrochloride

Cetirizine Hydrochloride

Cibenzoline Succinate
Supplement I, JP XV Infrared Reference Spectra 1977

Cilazapril Hydrate

Clobetasol Propionate

Clorazepate Dipotassium
1978 Infrared Reference Spectra Supplement I, JP XV

L-Cysteine

L-Cysteine Hydrochloride Hydrate

Domperidone
Supplement I, JP XV Infrared Reference Spectra 1979

Emorfazone

Enalapril Maleate

Felbinac
1980 Infrared Reference Spectra Supplement I, JP XV

L-Glutamine

Ibudilast

Isoxsuprine Hydrochloride
Supplement I, JP XV Infrared Reference Spectra 1981

Itraconazole

Labetalol Hydrochloride

Manidipine Hydrochloride
1982 Infrared Reference Spectra Supplement I, JP XV

Medazepam

Mizoribine

Nabumetone
Supplement I, JP XV Infrared Reference Spectra 1983

Nafamostat Mesilate

Nizatidine

Omeprazole
1984 Infrared Reference Spectra Supplement I, JP XV

Ozagrel Sodium

Piperacillin Hydrate

Salicylic Acid
Supplement I, JP XV Infrared Reference Spectra 1985

L-Serine

Sodium Starch Glycolate, Type A

Sodium Starch Glycolate, Type B


1986 Infrared Reference Spectra Supplement I, JP XV

L-Tyrosine

Ubenimex

Zidovudine
Supplement I, JP XV Ultraviolet-visible Reference Spectra 1987

Delete the following Ultraviolet-visible Reference Spectra:

Sulˆnpyrazone
Tubocurarine Chloride Hydrochloride Hydrate
1988 Ultraviolet-visible Reference Spectra Supplement I, JP XV

Add the following 30 spectra:

Acemetacin

Alminoprofen

Amlexanox
Supplement I, JP XV Ultraviolet-visible Reference Spectra 1989

Amlodipine Besilate

Amosulalol Hydrochloride

Azelastine Hydrochloride
1990 Ultraviolet-visible Reference Spectra Supplement I, JP XV

Bisoprolol Fumarate

Buformin Hydrochloride

Buprenorphine Hydrochloride
Supplement I, JP XV Ultraviolet-visible Reference Spectra 1991

Cetirizine Hydrochloride

Cibenzoline Succinate

Clorazepate Dipotassium
1992 Ultraviolet-visible Reference Spectra Supplement I, JP XV

Domperidone

Emorfazone

Felbinac
Supplement I, JP XV Ultraviolet-visible Reference Spectra 1993

Ibudilast

Isoxsuprine Hydrochloride

Itraconazole
1994 Ultraviolet-visible Reference Spectra Supplement I, JP XV

Labetalol Hydrochloride

Manidipine Hydrochloride

Mizoribine
Supplement I, JP XV Ultraviolet-visible Reference Spectra 1995

Nabumetone

Nafamostat Mesilate

Nicorandil
1996 Ultraviolet-visible Reference Spectra Supplement I, JP XV

Nizatidine

Omeprazole

Ozagrel Sodium
Supplement I, JP XV Ultraviolet-visible Reference Spectra 1997

Salicylic Acid

L-Tyrosine

Ubenimex
GENERAL INFORMATION

8. International Harmonization
Implemented in the Japanese
Pharmacopoeia Fifteenth Edition

Add the following:


May 2005 (Rev.1)

Harmonized items JP15 (Supplement I) Remarks

Sodium Starch Glycolate Sodium Starch Glycolate


Definition origin, limit of sodium
Identification A Identification (1)
Identification B Identification (3)
pH pH
Loss on drying Loss on drying
Limit of iron Purity (2) Iron
Limit of sodium chloride Purity (4) Sodium chloride
Limit of sodium glycolate Purity (3) Sodium glycolate
Assay Assay

Add the following:


June 2006

Harmonized items JP15 (Supplement I) Remarks

Hypromellose Phthalate Hypromellose Phthalate


Definition origin, limit of carboxybenzoyl
Packaging and storage Containers and storage
Viscosity Viscosity
Water Water
Residue on ignition Residue on ignition
Chloride Purity (1) Chloride
Limit of free phthalic acid Purity (3) Phthalic acid
Phthalyl content Assay

1999
2000 General Information Supplement I, JP XV

Add the following:


Nov. 2005

Harmonized items JP15 (Supplement I) Remarks

Anhydrous Dibasic Calcium Phosphate Anhydrous Dibasic Calcium Phosphate


Definition limit of content
Identification (1) Identification (1)
Identification (2) Identification (2)
Acid-insoluble substances Purity (1) Acid-insoluble substance
Chloride Purity (2) Chloride
Sulfate Purity (3) Sulfate
Carbonate Purity (4) Carbonate
Barium Purity (6) Barium
Loss on ignition Loss on drying
Assay Assay

Add the following:


Nov. 2005

Harmonized items JP15 (Supplement I) Remarks

Dibasic Calcium Phosphate Dibasic Calcium Phosphate Hydrate


Definition limit of content
Identification (1) Identification (1)
Identification (2) Identification (2)
Acid-insoluble substances Purity (1) Acid-insoluble substances
Chloride Purity (2) Chloride
Sulfate Purity (3) Sulfate
Carbonate Purity (4) Carbonate
Barium Purity (6) Barium
Loss on ignition Loss on drying
Assay Assay

Add the following:


Nov. 2005

Harmonized items JP15 (Supplement I) Remarks

Microbiological Examination of Non- 4.05 Microbiological Examination of


sterile Products: Non-sterile Produds
Microbial Enumeration Tests I. Microbiological Examination of
Non-sterile Products: Microbial
Enumeration Tests
Introduction 1. Introduction
General procedures 2. General Procedures
Enumeration methods 3. Enumeration Methods
Growth promotion test and suitability 4. Growth Promotion Test and
of the counting method Suitability of the Counting Method
Supplement I, JP XV General Information 2001

General considerations 4.1. General considerations


Preparation of test strains 4.2. Preparation of test strains
Negative control 4.3. Negative control
Growth promotion of the media 4.4. Growth promotion of the me-
dia
Suitability of the counting method 4.5. Suitability of the counting
in the presence of product method in the presence of
product
Results and interpretation 4.6. Results and interpretation
Testing of products 5. Testing of Products
Amount used for the test 5.1. Amount used for the test
Examination of the product 5.2. Examination of the product
Interpretation of the results 5.3. Interpretation of the results
Microbiological Examination of Non-
sterile Products: II. Microbiological Examination of
Tests for Specified Micro-organisms Non-sterile Products: Tests for
Specified Micro-organisms
Introduction 1. Introduction
General procedures 2. General Procedures
Growth promoting and inhibitory 3. Growth Promoting and Inhibitory
properties of the media and suitability Properties of the Media and
of the test Suitability of the Test
Preparation of test strains 3.1. Preparation of test strains
Negative control 3.2. Negative control
Growth promotion and inhibitory 3.3. Growth promotion and inhibi-
properties of the media tory properties of the media
Suitability of the test method 3.4. Suitability of the test method
Testing of products 4. Testing of Products
Bile-tolerant gram-negative bacteria 4.1. Bile-tolerant gram-negative
bacteria
Escherichia coli 4.2. Escherichia coli
Salmonella 4.3. Salmonella
Pseudomonas aeruginosa 4.4. Pseudomonas aeruginosa
Staphylococcus aureus 4.5. Staphylococcus aureus
Clostridia 4.6. Clostridia
Candida albicans 4.7. Candida albicans
Recommended solutions and culture 5. Recommended Solutions and Cul-
media ture Media

Add the following:


Nov. 2005

Harmonized items JP15 (Supplement I) Remarks

Microbiological Examination of Non- General Information


sterile Products: 12. Microbial Attributes of Non-sterile
Pharmaceutical Products
JP's particular description:
1. Definitions
2. Scope
2002 General Information Supplement I, JP XV

3. Sampling plan and frequency of


testing
4. Microbial control program
Acceptance Criteria for Pharmaceuti- 5. Microbial acceptance criteria for
cal Preparations and Substances for non-sterile pharmaceutical products
Pharmaceutical use
JP's particular description:
Explanation of the microbial accep-
tance criteria
JP's particular description:
6. Acceptance criteria for herbal drugs
and herbal drug containing prepara-
tions
Table 2. Acceptance Criteria for Table 1. Acceptance criteria for
Microbiological Quality of Non-sterile microbiological quality of non-sterile
Substances for Pharmaceutical use substances for pharmaceutical use
Table 1. Acceptance Criteria for Table 2. Acceptance criteria for
Microbiological Quality of Non-sterile microbiological quality of non-sterile
Dosage Forms dosage forms

Change to read:
Jan. 2000

Harmonized items JP 15 (Supplement I) Remarks

Bacterial Endotoxins Test 4.01 Bacterial Endotoxins Test


(Introduction) (Introduction)
Apparatus Apparatus
Preparation of standard endotoxin Preparation of standard endotoxin
stock solution stock solution
Preparation of standard endotoxin so- Preparation of standard endotoxin so-
lution lution
Preparation of sample solutions Preparation of sample solutions JP's particular description: Addition
of the preparation of sample solutions
for containers, and deletion of that for
medical devices.
Determination of maximum valid di- Determination of maximum valid di- JP's particular description: Addition
lution lution of the concentration unit of sample so-
lutions in the case where the endotoxin
limit per equivalent is specified.
Gel-clot technique Gel-clot technique
(1) Preparatory testing (1) Preparatory testing
(i) Test for confirmation of labeled (i) Test for confirmation of labeled
lysate sensitivity lysate sensitivity
(ii) Test for interfering factors (ii) Test for interfering factors
(2) Limit test (2) Limit test
(i) Procedure (i) Procedure
(ii) Interpretation (ii) Interpretation
(3) Assay (3) Assay
(i) Procedure (i) Procedure
Supplement I, JP XV General Information 2003

(ii) Calculation and interpretation (ii) Calculation and interpretation


Photometric techniques Photometric techniques
(1) Turbidimetric techniques (1) Turbidimetric techniques
(2) Chromogenic technique (2) Chromogenic technique
(3) Preparatory testing (3) Preparatory testing
(i) Assurance of criteria for the (i) Assurance of criteria for the JP's particular description: The test for
standard curve standard curve assurance of criteria for the standard
curve must be carried out for each lot
of lysate reagent.
(ii) Test for interfering factors (ii) Test for interfering factors JP's particular description:
Two conditions which the test must
meet are specified.
Explanation of the test method
where the interfering action is found.
Explanation of the usual methods
for eliminating the interference.
(4) Assay (4) Assay
(i) Procedure (i) Procedure
(ii) Calculation (ii) Calculation JP's particular description: Addition
of the alternative requirement to which
solution D must meet for valid test.
(iii) Interpretation (iii) Interpretation
Reagents, test solutions deletion
Amebocyte lysate deletion
Lysate TS deletion
Water for bacterial endotoxins test deletion
(BET)

Change to read:
Nov. 2005 (Rev. 2)

Harmonized items JP15 (Supplement I) Remarks

Anhydrous Lactose Anhydrous Lactose


Definition origin
Specific rotation Optical rotation
Clarity and color of solution Purity (1) Clarity and color of solu-
tion
Acidity or alkalinity Purity (2) Acidity or alkalinity
Protein and light-absorbing Purity (4) Proteins and light absorb-
impurities ing substances
Loss on drying Loss on drying
Water Water
Residue on ignition Residue on ignition
Content of alpha and beta anomers Isomer ratio
2004 General Information Supplement I, JP XV

applied to drugs containing viable micro-organisms as an


Change to read: active ingredient.

3. Sampling plan and frequency of testing


12. Microbial Attributes of 3.1 Sampling methods
Non-sterile Pharmaceutical Microbial contaminants are usually not uniformly dis-
tributed throughout the batches of non-sterile pharmaceuti-
Products cal products or raw materials. A biased sampling plan,
This chapter is harmonized with the European Phar- therefore, cannot be used to estimate the real bioburden in
macopoeia and the U.S. Pharmacopeia. The parts of the text the product. A sampling plan which can properly reflect the
that are not harmonized are marked with symbols ( ). status of the product batch should be established on the
basis of the bioburden data obtained by retrospective valida-
The presence of certain micro-organisms in non-sterile tion and/or concurrent validation. In general, a mixture of
preparations may have the potential to reduce or even inacti- samples randomly taken from at least different three por-
vate the therapeutic activity of the product and has a poten- tions, almost the same amount for each portion, is used for
tial to adversely affect the health of the patient. Manufac- the tests of the product. When the sampling is difficult in a
turers have therefore to ensure a low bioburden of finished clean area, special care is required during sampling to avoid
dosage forms by implementing current guidelines on Good introducing microbial contamination into the product or
Manufacturing Practice during the manufacture, storage affecting the nature of the product bioburden. If it is con-
and distribution of pharmaceutical preparations. This firmed that the product bioburden is stable for a certain
chapter provides guidelines for acceptable limits of viable period, as in the case of non-aqueous or dried products, it is
micro-organisms (bacteria and fungi) existing in raw materi- not necessary to do the tests, immediately after the sam-
als and non-sterile pharmaceutical products. Microbial pling.
examination of non-sterile products is performed according 3.2 Testing frequency
to the methods given in the Microbial Limit Test <4.05> on The frequency of the tests should be established on the
Microbiological Examination of Non-sterile Products: basis of a variety of factors unless otherwise specified. These
Microbial Enumeration Tests and Tests for Specified Micro- factors include:
organisms. When these tests are carried out, a microbial a) Dosage forms of non-sterile pharmaceutical products
control program must be established as an important part of (usage);
the quality management system of the product. Personnel b) Manufacturing processes;
responsible for conducting the tests should have specialized c) Manufacturing frequency;
training in microbiology, biosafety measures and in the d) Characteristics of raw materials (natural raw material,
interpretation of the testing results. synthetic compound, etc.);
1. Definitions e) Batch sizes;
1.1 Non-sterile pharmaceutical products: Non-sterile drugs f) Variations in bioburden estimates (changes in batches,
shown in monographs of the JP and non-sterile finished seasonal variations, etc.);
dosage forms. g) Changes affecting the product bioburden (changes in
1.2 Raw materials: All materials, including raw ingredients manufacturing process, supplier of raw materials, batch
and excipients, used for the preparation of drugs, except for number of raw materials, etc.);
water and gases. h) Others.
1.3 Bioburden: Number and type of viable micro-organ- In general, the tests may be performed at a high frequency
isms existing in non-sterile pharmaceutical products. during the initial production of a drug to get information on
1.4 Action levels: Established bioburden levels that require the microbiological attributes of the product or raw materi-
immediate follow-up and corrective action if they are als used for the production. However, this frequency may be
exceeded. reduced as bioburden data are accumulated through
1.5 Alert levels: Established bioburden levels that give retrospective validation and/or concurrent validation. For
early warning of a potential drift from normal bioburden example, the tests may be performed at a frequency based on
level, but which are not necessary grounds for definitive time (e.g., weekly, monthly or seasonally), or on alternate
corrective action, though they may require follow-up investi- batches.
gation. 4. Microbial control program
1.6 Quality management system: The procedures, opera- When the ``Microbial Limit Test <4.05>'' is applied to a
tion methods and organizational structure of a manufac- non-sterile pharmaceutical product, the methods for the
turer (including responsibilities, authorities and relation- recovery, cultivation and estimation of the bioburden from
ships between these) needed to implement quality manage- the product must be validated and a ``Microbial control pro-
ment. gram'' covering the items listed below must be prepared.
2. Scope a) Subject pharmaceutical name (product name);
In general, the test for total viable aerobic count is not b) Frequency of sampling and testing;
c) Sampling methods (including responsible person, quan-
Supplement I, JP XV General Information 2005

tity, environment, etc. for sampling); levels due to the high temperatures, organic solvents, etc.,
d) Transfer methods of the samples to the testing area used in their manufacturing processes. Raw materials origi-
(including storage condition until the tests); nated from plants and animals in general have higher bio-
e) Treatment of the samples (recovery methods of microbi- burdens than synthetic compounds.
al contaminants); The microbial quality of the water used in the processing
f) Enumeration of viable micro-organisms (including test- of active ingredients or non-sterile pharmaceuticals may
ing quantity, culture media, growth-supporting test of the have a direct effect on the quality of the finished dosage
media, culturing methods, etc.); form. This means it is necessary to keep the level of microbi-
g) Detection of specified micro-organisms (including test- al contaminants in the water as low as possible.
ing quantity, culture media, growth-supporting test of the Acceptance criteria for microbiological quality for non-
media, culturing methods, etc.); sterile finished dosage forms are shown in Table 2. These
h) Estimation of the number of and characterization of microbial limits are based primarily on the type of dosage
microbial contaminants; form, water activity, and so on. For oral liquids and phar-
i) Establishment of ``Microbial acceptance criteria'' maceutical products having a high water activity, in general,
(including alert level and action level); low microbial acceptance criteria are given.
j) Actions to be taken when the levels exceed ``Microbial Table 2 includes a list of specified micro-organisms for
acceptance criteria''; which acceptance criteria are set. The list is not necessarily
k) Persons responsible for the testing and evaluation, etc.; exhaustive and for a given preparation it may be necessary to
l) Other necessary items. test for other micro-organisms depending on the nature of
the starting materials and the manufacturing process.
5. Microbial acceptance criteria for non-sterile pharmaceu-
If it has been shown that none of the prescribed tests will
tical products
allow valid enumeration of micro-organisms at the level
By establishing ``Microbial acceptance criteria'' for non-
sterile pharmaceutical products based upon the total aerobic
microbial count (TAMC) and the total combined Table 1. Acceptance criteria for microbiological quality of
yeasts/moulds count (TYMC), it is possible to evaluate at non-sterile substances for pharmaceutical use
the initial processing stage of the product whether the Total Combined
microbiological quality of the raw materials is adequate or Total Aerobic
Yeasts/Moulds
not. Furthermore, it is then possible to implement appropri- Microbial Count
Count
ate corrective action as needed to maintain or improve the (CFU/g or CFU/mL)
(CFU/g or CFU/mL)
microbiological quality of the product. The target limits of
Substances for
microbial levels for raw materials (synthetic compounds and
pharmaceutical 103 102
minerals) are shown in Table 1.
In general, synthetic compounds have low bioburden
use

Table 2. Acceptance criteria for microbiological quality of non-sterile dosage forms

Total Aerobic Total Combined


Route of administration Microbial Count Yeasts/Moulds Count Specified Micro-organism
(CFU/g or CFU/mL) (CFU/g or CFU/mL)

Non-aqueous preparations 103 102 Absence of Escherichia coli


for oral use (1 g or 1 mL)

Aqueous preparations 102 101 Absence of Escherichia coli


for oral use (1 g or 1 mL)

Rectal use 103 102 —

Oromucosal use 102 101 Absence of Staphylococcus aureus


Gingival use (1 g or 1 mL)
Cutaneous use Absence of Pseudomonas aeruginosa
Nasal use (1 g or 1 mL)
Auricular use

Vaginal use 102 101 Absence of Pseudomonas aeruginosa


(1 g or 1 mL)
Absence of Staphylococcus aureus
(1 g or 1 mL)
Absence of Candida albicans
(1 g or 1 mL)
2006 General Information Supplement I, JP XV

Transdermal patches (limits 102 101 Absence of Staphylococcus aureus


for one patch including ad- (1 patch)
hesive layer and backing) Absence of Pseudomonas aeruginosa
(1 patch)

Inhalation use (more rigo- 102 101 Absence of Staphylococcus aureus


rous requirements apply to (1 g or 1 mL)
liquid preparations for Absence of Pseudomonas aeruginosa
nebulization) (1 g or 1 mL)
Absence of bile-tolerant gram-negative
bacteria
(1g or 1 mL)

prescribed, a validated method with a limit of detection as ration method of the preparations.
close as possible to the indicated acceptance criterion is used.
In addition to the micro-organisms listed in Table 2, the 
Table 3. Acceptance criteria for herbal drugs and their
significance of other micro-organisms recovered should be
preparations
evaluated in terms of:
the use of the product: hazard varies according to the Category 1 Category 2
Micro-organisms
route of administration (eye, nose, respiratory tract); (CFU/g or CFU/mL) (CFU/g or CFU/mL)
the nature of the product: does the product support Aerobic bacteria 107 105
growth, does it have adequate antimicrobial preserva- Molds and yeasts 104 103
tion? Enterobacteria
the method of application; and other
※ 103
the intended recipient: risk may differ for neonates, gram-negative
infants, the debilitated; bacteria
use of immunosuppressive agents, corticosteroids; Escherichia coli 102 Not detected
presence of disease, wounds, organ damage. Salmonella Not detected Not detected
Where warranted, a risk-based assessment of the relevant Staphylococcus
※ ※
aureus
factors is conducted by personnel with specialized training in
microbiology and the interpretation of microbiological data. ※ : The limits are not specified.
For raw materials, the assessment takes account of
processing to which the product is subjected, the current
technology of testing and the availability of materials of the 21. Quality Control of Water
desired quality. Acceptance criteria are based on individual
results or on the average of replicate counts when replicate
for Pharmaceutical Use
counts are performed (e.g. direct plating methods).
When an acceptance criterion for microbiological quality Change the 3.4.1 Media and Incubation Condi-
is prescribed it is interpreted as follows: tions to read:
—101 CFU: maximum acceptable count=20, 3.4.1 Media and Incubation Conditions
—102 CFU: maximum acceptable count=200, There are many mesophilic bacteria of heterotrophic type
—103 CFU: maximum acceptable count=2000, and so that are adaptable to poor nutrient water environments. In
forth. many pharmaceutical water systems, heterotrophic bacteria
6. Acceptance criteria for herbal drugs may form bio-films and cause water quality deterioration.
Target limits of microbial contamination for herbal drugs It, therefore, is useful to monitor the water quality by use of
and herbal drug containing preparations are shown in Table R2A Agar Medium, which is excellent for growing bacteria
3. Category 1 indicates herbal drugs and their preparations of oligotrophic type. On the other hand, in routine microbial
to which boiling water is added before use, and category 2 monitoring, an approach that identifies the trend in
indicates other herbal drugs and their preparations. In this microbiological quality change is widely employed; a stan-
guideline, enterobacteria and other gram-negative bacteria, dard agar plate is used for counting the total number of via-
Escherichia coli, Salmonella, and Staphylococcus aureus are ble microorganisms capable of proliferating at 30–359C in a
mentioned as specified micro-organisms, but other micro- comparatively short period of time.
organisms such as certain species of Bacillus cereus, Clos- Table 2 shows examples of measurement methods, mini-
tridia, Pseudomonas, Burkholderia, Asperigillus and En- mum sample sizes, media, and incubation periods for es-
terobacter species are also necessary to be tested depending timating viable counts.
on the origin of the herbal drug raw materials or the prepa- The media shown in Table 2 are as follows.
Supplement I, JP XV General Information 2007

Table 2. Methods for Assessment of Viable Counts in Pharmaceutical Water

Method Pharmaceutical Water

Water Purified Water in Bulk Water for Injection in Bulk

Measurement Pour Plate Method or Pour Plate Method or Membrane


Membrane Filtration
Method Membrane Filtration Filtration

Minimum
1.0 mL 1.0 mL 100 mL
Sample Size

R2A Agar Medium, Standard R2A Agar Medium, Standard


Media Standard Agar Medium
Agar Medium Agar Medium

R2A Agar Medium: 4 – 7 days R2A Agar Medium: 4 – 7 days


Incubation Standard Agar Medium: (or longer) (or longer)
Period 48 – 72 hours (or longer) Standard Agar Medium: 48 – 72 Standard Agar Medium: 48 – 72
hours (or longer) hours (or longer)

R2A Agar Medium: 20 – 259 C or R2A Agar Medium: 20 – 259C or


Incubation Standard Agar Medium:
30 – 359C 30 – 359
C
Temperature 30 – 359C
Standard Agar Medium: 30 – 359
C Standard Agar Medium: 30 – 359
C

Standard Agar Medium Sodium pyruvate CH3COCOONa White to pale yel-


Casein peptone 5.0 g low crystalline powder. Freely soluble in water, and slight-
Yeast extract 2.5 g ly soluble in ethanol (99.5) and in acetone.
Glucose 1.0 g Identification (1) Determine the infrared absorption
Agar 15.0 g spectrum as directed in the potassium bromide disk
Water 1000 mL method under Infrared Spectrophotometry <2.25>: it
Mix all the ingredients, and sterilize by heating in an exhibits absorption at the wave numbers of about 1710
autoclave at 1219 C for 15 to 20 minutes. pH after cm-1, 1630 cm-1, 1410 cm-1, 1360 cm-1, 1190 cm-1,
sterilization: 6.9–7.1. 1020 cm-1, 980 cm-1, 830 cm-1, 750 cm-1, 630 cm-1 and
430 cm-1.
R2A Agar Medium (2) A solution (1 in 20) responds to the Qualitative
Peptone (casein and animal tissue) 0.5 g Tests <1.09> (1) for sodium salt.
Casamino acid 0.5 g Content: not less than 97.0z. Assay—Dissolve about
Yeast extract 0.5 g 0.4 g of sodium pyruvate, accurately weighed, in 200 mL
Sodium pyruvate 0.3 g of water. Transfer 20 mL of this solution into a ground
Glucose 0.5 g joint iodine bottle, and cool to 109
C or lower. Add 40 mL
Magnesium sulfate heptahydrate 0.05 g of 0.05 mol/L iodine VS and 20 mL of 4 mol/L sodium
Soluble starch 0.5 g hydroxide solution, then allow to stand in a dark place for
Dipotassium hydrogen phosphate 0.3 g 2 hours, and add 15 mL of diluted sulfuric acid (1 in 6).
Agar 15 g Titrate <2.50> with 0.1 mol/L sodium thiosulfate VS (indi-
Water 1000 mL cator: starch TS). Perform a blank determination in the
Mix all the ingredients, and sterilize by heating in an same manner, and make any necessary correction.
autoclave at 1219 C for 15 to 20 minutes. pH after
Each mL of 0.05 mol/L iodine VS
sterilization: 7.1 – 7.3.
=1.854 mg of C3H3NaO3
For the ingredients, which are not specified in the
Japanese Pharmacopoeia, use the following reagents.
Casamino acid Prepared for microbial test, by the acid
Add the following:
hydrolysis of casein.
Loss on drying <2.41>: Not more than 8z (0.5 g,
1059C, constant mass). 31. Purity Tests on Crude Drugs
Residue on ignition <2.44>: Not more than 55z (0.5 g). Using Genetic Information
Nitrogen content <1.08>: Not less than 7z (1059C, con-
stant mass, after drying). The first step in the quality assurance of natural products
is the use of raw materials from the right part of the right
2008 General Information Supplement I, JP XV

origin (the right source). Therefore, it is clearly stated in Ar- shown that these 4 types of plants can be clearly classified by
ticle 4 of the General Rules For Crude Drugs that the source comparing the base sequences of the ITS mentioned above,
of a crude drug is the approval or rejection criteria. There and that the species can be easily classified without perform-
are various methods for differentiating the sources of crude ing base sequence analysis by performing PCR using a spe-
drugs, such as morphological methods, organoleptic tests, cies specific primer set and determining the presence or ab-
and chemical methods, and appropriate methods for each sence of an amplification band.
are described in the individual monographs. Morphological In cooperative studies, the degree of simplicity of a study
methods, organoleptic tests, and chemical methods are dis- is given maximum consideration. We examined methods
crimination methods for species that are based on the ex- that observe PCR amplification bands using species specific
pression characteristics of the crude drugs. On the other primer sets and do not involve base sequence analyses. Test
hand, together with recent progress in molecular biology methods based on PCR which uses species specific primer
techniques and the accumulation of genetic information on sets are analytical methods with very high degrees of
plants, methods for differentiating species have been estab- sensitivity. Therefore, when using them as identification
lished by confirming the genotype of a crude drug. Methods tests for powdered crude drugs, amplification bands can be
such as these are different from morphological and other observed even if the vast majority of the crude drug for
methods that differentiate based on phenotype in that they analysis is not suitable and there is only a minute amount of
are not affected by the environment. Also, the methods have powder from a crude drug derived from a suitable plant.
several advantages, such as specialized expertise and skill for Consequently, in identification tests, either a cut or a whole
classification are not needed, and objective results are easily crude drug must be used, as long as one is careful to avoid
obtained. contamination by powder originating from other crude
The evolution of living organisms is accomplished by drugs. On the other hand, when used as a purity test, the
genetic mutation, and differences among the base sequences form of the crude drug is irrelevant as long as the gene am-
of genes of closely related species reflect the strain relation- plification is performed properly and the target gene is not
ships between the species. Based on this theory, in recent polymorphic, so if amplification bands of an inappropriate
years methods that classify species phylogenetically using the plant are confirmed in the purity test, regardless of the form
base sequence of rDNA that codes for ribosomal RNA of the crude drug, it becomes clear there is contamination by
(rRNA) on the nuclear genome have been adopted. In the an inappropriate crude drug.
same way, the base sequence of this rDNA is also the most The methods shown here are reference information and at
often used in the classification of higher plants using the the present stage results obtained using the methods do not
genotype. In particular, it has become very easy to classify affect the approval or rejection of the crude drug in each
closely related species using the intergenic transcriber space monograph. Furthermore, by performing the sequence
(ITS) region of the rDNA region, since by comparison with outlined in the previous paper, it goes without saying that a
the coded gene region base substitution is easily undertaken. more accurate decision concerning the source species can be
Furthermore, since the genes on the nuclear genome made.
originate from the parents, an advantage is interspecies
DNA Amplification Equipment
hybrids can be confirmed. Higher plants also have
DNA amplification equipment is used to amplify the
mitochondrial genes and chloroplastic genes. Although the
DNA which is extracted from a crude drug and then puri-
genes on these genomes are also often used for classification,
fied. Since there are slight differences in the methods of tem-
interspecies hybrids cannot be confirmed because the genes
perature control, and so on depending on the equipment
are normally monogenic.
used, there may be differences in the intensity, etc. of the
The methods presented here have been developed based
PCR amplification bands even if PCR is carried out under
on the reported identification methods of Atractylodes Lan-
the stipulated conditions. Therefore, when judging results
cea Rhizome and Atractylodes Rhizome (Y. Guo, et al, J.
based solely on the presence or absence of PCR amplifica-
Nat. Med. 60, 149–156, 2006) utilizing the gene sequence of
tion bands, confirm that only proper amplification bands
the ITS of rDNA. Cooperative studies on the validation of
are obtained when performing PCR using DNA obtained us-
purity tests related to Atractylodes Lancea Rhizome in
ing samples confirmed beforehand to be the source species.
Atractylodes Rhizome, have been completed. The plant
If proper amplification bands are not obtained, the PCR
sources for Atractylodes Lancea Rhizome stipulated in the
temperature conditions should be slightly adjusted.
individual monographs are Atractylodes lancea De Candolle
or A. chinensis Koidzumi (Compositae), while those for Procedure
Atractylodes Rhizome are A. japonica Koidzumi ex The following is a sample procedure.
Kitamura or A. ovata De Candolle (Compositae). The ap- 1. Preparation of template DNA
proval or rejection of the source of Atractylodes Lancea Crude drugs are different from fresh plants in that they
Rhizome is, in principle, determined by the description of are dried products and a certain amount of time has passed
the crude drug, including microscopy, while that of Atrac- since they were harvested. Therefore, in many cases the
tylodes Rhizome is determined by the description of the DNA has undergone fragmentation. Furthermore, various
crude drug, including microscopy, together with color reac- substances that can block or interfere with the PCR reaction
tion, which is an identification test. In the manuscript, it was may be present in the plant. For these reasons, the extraction
Supplement I, JP XV General Information 2009

and purification of template DNA is the process that should tive with A. lancea, D is positive with A. chinensis) as
receive the greatest amount of attention. In the case of described in the paper mentioned above (J. Nat. Med. 60,
Atractylodes crude drugs, the periderm should be removed 149–156, 2006), however, when a combination of primer A
using a clean scalpel or other clean instrument before pul- and B is used, it is possible to confirm the source species of
verizing the sample because very often there are inhibitory each of the respective specimens. In order to confirm that
substances present in the periderm. the DNA has been extracted correctly, the reaction solution
There are various methods with which to extract and puri- containing the positive control primer (Pf and Pr) as shown
fy DNA from the samples. It is recommended that commer- below should be prepared. In addition, the negative control
cially available DNA extraction kits be used if one takes into solutions which are not containing DNA sample or either of
consideration their advantages of not using any noxious the primer sets should be prepared and simultaneously con-
reagents and not requiring any complicated purification duct PCR.
procedures. In this case, attention should be paid to the final
amount (concentration) of DNA obtained, and the amount Pf: 5'–CAT TGT CGA AGC CTG CAC AGC A–3'
of initial sample and the volume of liquid to elute the DNA Pr: 5'–CGA TGC GTG AGC CGA GAT ATC C–3'
need to be controlled. When extraction and purification are
performed using silica gel membrane type kits stipulated in The PCR reaction is performed under the following con-
notifications (Notification No. 110, Director of Food Health ditions. After starting the reaction at 959C for 10 minutes,
Department, March 2001; Partial Amendment: Notification followed by one cycle of 0.5 minutes at 959C and 0.75
No. 0629002, 2.2.1.2, Director of Food Safety Department, minutes at 689 C (699 C only when using the primer set C),
June 2006) related to inspection methods for recombinant and 30 cycles of PCR amplification. Then terminate reaction
DNA foods, it is appropriate to use 200 mg of sample, 1 mL at 729C for 7 minutes, store at 49C, and use the reaction so-
of AP1 buffer solution, 2 mL of RNase A, and 325 mL of lution obtained as the PCR amplification reaction solution.
AP2 buffer solution. Also, the most important things are 4. Gel electrophoresis
that the supernatant loaded on the first column is clear and After completion of the reaction, mix 5 mL of the PCR
that there is no need to load 1 mL unreasonably. Further- amplification reaction solution with an appropriate volume
more, 50 mL is an appropriate volume used in the final elu- of gel loading buffer solution, add the mixture to the wells
tion of the DNA, and normally the initial eluate is used as of 2z agarose gel, and then perform electrophoresis using 1-
the DNA sample stock solution. fold TAE buffer solution (refer to General Information,
2. Confirmation of purity of DNA in DNA sample stock Rapid Identification of Microorganisms Based on Molecular
solution and assay of DNA Biological Method). Run in parallel an appropriate DNA
The purity of the DNA in the stock solution can be con- molecular mass standard. Electrophoresis is terminated
firmed by the OD260 nm/OD280 nm ratio using a spec- when the bromophenol blue dye in the gel loading buffer has
trophotometer. A ratio of 1.5 indicates that the DNA has advanced to a point corresponding to 1/2 to 2/3 the length
been adequately purified. The amount of DNA is calculated of the gel.
using 1 OD260 nm=50 mg/mL. The measurement mentioned 5. Detection and evaluation of PCR products
above is performed using appropriately diluted DNA sample Counterstain the gel after electrophoresis when not using
stock solution. Based on the results obtained, dilute with gel stained in advance with ethidium bromide. Place the gel
water to the concentration needed for the subsequent PCR that has undergone electrophoresis and staining in a gel
reactions, use this solution as the DNA sample solution, image analyzer, irradiate with ultraviolet light (312 nm), and
pipet into micro test tubes, and if necessary store frozen at determine its electrophoresis pattern. Compare this to the
not over -209C. The pipeted DNA sample is used immedi- DNA molecular mass standard and determine the absence or
ately after thawing and any remaining solution should be presence of the target amplification band. In the case of
discarded and not refrozen. If the concentration of the DNA purity tests on Atractylodes Lancea Rhizome in Atrac-
sample stock solution does not reach the concentration tylodes Rhizome, first confirm a 305 bp band with the
stipulated in PCR, it is used as a DNA sample solution. reaction solution to which the positive control primer set has
3. PCR been added, and confirm there are no bands in a solution
When a commercially available enzyme is used with the with no primer sets and a solution with no DNA sample so-
qualitative PCR method mentioned in the above notification lution. Next, if a 226 bp band is confirmed when the primer
(Notification No. 0629002, 2.1.3.1.1, Director of Food Safe- set C is added or if a 200 bp band is confirmed when the
ty Department), to a solution consisting of 2.5 mL of the primer set D is added, the sample is judged to be Atrac-
PCR buffer solution containing magnesium that comes with tylodes Lancea Rhizome (in the case of cut crude drug, con-
the enzyme, dNTP (0.2 mmol/L) that also comes with the tamination of Atractylodes Lancea Rhizome is observed)
enzyme, 5' and 3' primer (0.4 mmol/L), and Taq DNA poly- and it is rejected. The sample is judged not to be Atrac-
merase (1.25 units), add 5 mL of 10 ng/mL DNA sample so- tylodes Lancea Rhizome (in the case of cut crude drug, there
lution (50 ng of DNA) on ice. It is appropriate to perform is no contamination of Atractylodes Lancea Rhizome) and
the reaction at a total volume of 25 mL. When conducting the purity test is acceptable if a 305 bp band is confirmed
purity tests on Atractylodes Lancea Rhizome in Atrac- with the positive control primer set, bands are not observed
tylodes Rhizome, the primer sets used are C and D (C is posi- in reaction solution without primer and reaction solution
2010 General Information Supplement I, JP XV

without DNA sample solution, and a 226 bp band is not ob-


served with the primer set C and a 200 bp band is not ob-
served with the primer set D. If a band is not observed with
the positive control primer, it is to be concluded that the
DNA extraction failed and the procedure should be started
over again from the DNA extraction step. If bands are con-
firmed in reaction solutions without primer sets or without
DNA sample solution, it should be assumed that there was
an error in the PCR procedure and therefore the procedure
should be repeated again from the step 3. PCR.
INDEX

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


the JP XV main volume, including those where the text being revised in
the Supplement originally appeared. This Supplement I commences with
page 1789 and succeeding Supplements will continue to be paged in se-
quence.

1„1788 Main Volume of JP XV


1789„2040 Supplement I

Tablets, 279, 1826 Aminobenzylpenicillin, 307


A Akebia Stem, 1253 Anhydrous, 306
Alacepril, 279 Hydrate, 307
Absorptive Ointment, 265 Tablets, 280 Sodium, 308
Acacia, 1251 Albumin Tannate, 282 Aminophylline
Powdered, 1251 Alcohol, 638 Hydrate, 297
Acebutolol Hydrochloride, 265 Dehydrated, 639 Injection, 297, 1831
Aceglutamide Aluminum, 266 for Disinfection, 640 Amitriptyline Hydrochloride, 298
Acemetacin, 1825 Aldioxa, 282 Tablets, 299, 1831
Acetaminophen, 267 Alimemazine Tartrate, 283 Amlexanox, 1831
Acetazolamide, 268 Alisma Rhizome, 1253 Tablets, 1833
Acetic Acid, 268 Powdered, 1253 Amlodipine Besilate, 1834
Glacial, 269 Allopurinol, 284 Ammonia Water, 299
Acetohexamide, 269 Alminoprofen, 1826 Amobarbital, 300
Acetylcholine Chloride for Injection, Tablets, 1827 Sodium for Injection, 300
271, 1825 Aloe, 1254 Amomum Seed, 1256
Acetylsalicylic Acid, 318 Powdered, 1255 Powdered, 1256
Tablets, 319 Alpinia Officinarum Rhizome, 1256, Amorphous Insulin Zinc Injection
Achyranthes Root, 1252 1937 (Aqueous Suspension), 764
Aclarubicin Hydrochloride, 272 Alprazolam, 284 Amosulalol Hydrochloride, 1835
Acrinol Alprenolol Hydrochloride, 285 Tablets, 1836
and Zinc Oxide Oil, 273 Alprostadil, 286 Amoxapine, 301
and Zinc Oxide Ointment, 274 Alfadex, 287 Amoxicillin Hydrate, 302
Hydrate, 274 Injection, 1828 Amphotericin B, 303
Actinomycin D, 275 Alum, 292 for Injection, 304
Adrenaline, 276 Solution, 293 Syrup, 305
Injection, 276 Aluminum Tablets, 305
Solution, 277 Acetylsalicylate, 319 Ampicillin
Adsorbed Hydroxide Gel, Dried, 288 Anhydrous, 306
Diphtheria-Purified Pertussis-Teta- Hydroxide Gel Fine Granules, Dried, Ethoxycarbonyloxyethyl Hydro-
nus Combined Vaccine, 596 289 chloride, 329
Diphtheria-Tetanus Combined Monostearate, 291 Hydrate, 307
Toxoid, 596 Potassium Sulfate, Dried, 292 Sodium, 308
Diphtheria Toxoid for Adult Use, Potassium Sulfate Hydrate, 292 Sodium for Injection, 1838
596 Silicate, Natural, 289 Ampicillinphthalidyl Hydrochloride,
Habu-venom Toxoid, 716 Silicate, Synthetic, 290 1138
Hepatitis B Vaccine, 716 Sucrose Sulfate Ester, 1118 Amyl Nitrite, 309
Purified Pertussis Vaccine, 980 Amantadine Hydrochloride, 293 Anemarrhena Rhizome, 1256, 1937
Tetanus Toxoid, 1156 Ambenonium Chloride, 294 Anesthamine, 644
Afloqualone, 277 Amidotrizoic Acid, 295 Anesthetic Ether, 641
Agar, 1252 Amikacin Sulfate, 296 Angelica Dahurica Root, 1257, 1937
Powdered, 1253 Injection, 1830 Anhydrous
Ajmaline, 278 Aminoacetic Acid, 704 Aminobenzylpenicillin, 306

2011
2012 Index Supplement I, JP XV

Ampicillin, 306 Beef Tallow, 336 Sulfate, 372


Caffeine, 386 Beeswax Boric Acid, 374
Citric Acid, 514 White, 336 Bromazepam, 374
Dibasic Calcium Phosphate, 396, Yellow, 336 Bromhexine Hydrochloride, 375
1874 Bekanamycin Sulfate, 337 Bromocriptine Mesilate, 376
Ethanol, 639 Belladonna Bromovalerylurea, 377
Lactose, 802, 1900 Extract, 1263, 1939 Bucillamine, 377
Antipyrine, 310 Root, 1263 Tablets, 1845
Apricot Kernel, 1257, 1937 Benidipine Hydrochloride, 338 Bucumolol Hydrochloride, 378
Water, 1257 Tablets, 339 Bufetolol Hydrochloride, 379
Aralia Rhizome, 1937 Benincasa Seed, 1264 Bufexamac, 380
Arbekacin Sulfate, 311 Benoxinate Hydrochloride, 951 Cream, 380
Injection, 312 Benserazide Hydrochloride, 340 Ointment, 381
Areca, 1258 Bentonite, 341 Buformin Hydrochloride, 1846
L-Arginine, 313 Benzalkonium Chloride, 342 Enteric-coated Tablets, 1847
Hydrochloride, 313 Concentrated Solution 50, 343 Tablets, 1849
Hydrochloride Injection, 314, 1839 Solution, 342 Bumetanide, 382
Aromatic Castor Oil, 415 Benzbromarone, 343 Bunazosin Hydrochloride, 383
Arotinolol Hydrochloride, 314 Benzethonium Chloride, 344 Bupleurum Root, 1266
Arsenical Paste, 315 Solution, 345 Bupranolol Hydrochloride, 383
Arsenic Trioxide, 316 Benzocaine, 644 Buprenorphine Hydrochloride, 1850
Arsenous Acid, 316 Benzoic Acid, 345 Burdock Fruit, 1267
Artemisia Capillaris Flower, 1258 Benzoin, 1265 Burnt Alum, 292
Ascorbic Acid, 316 Benzyl Busulfan, 384
Injection, 317, 1839 Alcohol, 346 Butropium Bromide, 385
Powder, 317 Benzoate, 347 Butyl Parahydroxybenzoate, 386
Asiasarum Root, 1259, 1938 Benzylpenicillin
Asparagus Tuber, 1259, 1938 Benzathine Hydrate, 348 C
L-Aspartic Acid, 318 Potassium, 349
Aspirin, 318 Potassium for Injection, 1841 Cacao Butter, 386
Aluminum, 319 Berberine Caffeine
Tablets, 319 Chloride Hydrate, 351 and Sodium Benzoate, 388
Aspoxicillin Hydrate, 320 Tannate, 352 Anhydrous, 386
Astragalus Root, 1260 Betahistine Mesilate, 353, 1842 Hydrate, 387
Astromicin Sulfate, 322 Tablets, 354 Calciferol, 624
Atenolol, 323 Betamethasone, 355 Calcium
Atractylodes Dipropionate, 358 Carbonate, Precipitated, 389
Lancea Rhizome, 1260 Sodium Phosphate, 359 Chloride Hydrate, 390
Lancea Rhizome, Powdered, 1260 Tablets, 356 Chloride Injection, 390, 1851
Rhizome, 1261, 1938 Valerate, 360 Folinate, 391, 1851
Rhizome, Powdered, 1261, 1939 Valerate and Gentamicin Sulfate Gluconate Hydrate, 391
Atropine Sulfate Cream, 361 Hydroxide, 392
Hydrate, 324 Valerate and Gentamicin Sulfate Lactate Hydrate, 393
Injection, 324 Ointment, 362 Leucovorin, 391
Azathioprine, 325 Bethanechol Chloride, 363 Oxide, 393
Tablets, 326 Bezafibrate, 364 Pantothenate, 394
Azelastine Hydrochloride, 1839 Sustained Release Tablets, 365 Paraaminosalicylate Granules, 394
Azithromycin Hydrate, 327 Bifonazole, 366 Paraaminosalicylate Hydrate, 395
Aztreonam, 328 Biotin, 1842 Polystyrene Sulfonate, 398
for Injection, 1840 Biperiden Hydrochloride, 366 Stearate, 400
Bisacodyl, 367 Calumba, 1267, 1939
B Suppositories, 368, 1843 Powdered, 1268, 1939
Bismuth Camellia Oil, 400
Bacampicillin Hydrochloride, 329 Subgallate, 368 Camostat Mesilate, 400, 1852
Bacitracin, 330 Subnitrate, 369 d-Camphor, 401
Baclofen, 331 Bisoprolol Fumarate, 1843 dl-Camphor, 402
Tablets, 332, 1841 Tablets, 1844 Capsicum, 1268
Bamethan Sulfate, 333 Bitter and Salicylic Acid Spirit, 1270
Barbital, 333 Cardamon, 1265 Powdered, 1269
Barium Sulfate, 334 Orange Peel, 1265 Tincture, 1269
Bear Bile, 1262 Tincture, 1266 Capsules, 403
Bearberry Leaf, 1262 Bleomycin Capsules
Beclometasone Dipropionate, 335 Hydrochloride, 370 Cefaclor, 416
Supplement I, JP XV Index 2013

Cefadroxil, 1852 Fine Granules, 432 Hydrochloride, 493


Cefdinir, 435 Hydrate, 430 Chlorinated Lime, 494
Clindamycin Hydrochloride, 520 Tablets, 433 Chlormadinone Acetate, 494
Clofibrate, 526 Cefdinir, 434 Chlorobutanol, 495
Clorazepate Dipotassium, 1869 Capsules, 435 Chlorphenesin Carbamate, 496, 1858
Doxifluridine, 604 Fine Granules, 436 Tablets, 1859
Flopropione, 667 Cefditoren Pivoxil, 437 Chlorpheniramine
Flurazepam, 678 Fine Granules, 438 and Calcium Powder, 497
Indometacin, 756, 1893 Tablets, 438 Maleate, 498
Nizatidine, 1913 Cefepime Dihydrochloride Maleate Injection, 499
Rifampicin, 1065 for Injection, 441 Maleate Powder, 499
Roxatidine Acetate Hydrochloride Hydrate, 439 Maleate Tablets, 500
Extended-release, 1071 Cefixime, 442 d-Chlorpheniramine Maleate, 501
Sodium Iodide (123I), 1103 Cefmenoxime Hydrochloride, 443 Chlorpromazine Hydrochloride, 502
Sodium Iodide (131I), 1103 Cefmetazole Sodium, 445 Injection, 503, 1860
Sulpiride, 1132 for Injection, 1855 Tablets, 503, 1861
Tranexamic Acid, 1192 Cefminox Sodium Hydrate, 446 Chlorpropamide, 504
Vitamin A, 1230 Cefodizime Sodium, 447 Tablets, 505, 1861
Vitamin A Oil, 1230 Cefoperazone Sodium, 448 Cholecalciferol, 506
Captopril, 403 Cefotaxime Sodium, 449 Cholera Vaccine, 506
Carbamazepine, 404 Cefotetan, 451 Cholesterol, 507
Carbazochrome Sodium Sulfonate Cefotiam Chorionic Gonadotrophin, 707
Hydrate, 405 Hexetil Hydrochloride, 453 for Injection, 708
Carbetapentane Citrate, 974 Hydrochloride, 455 Chrysanthemum Flower, 1271
Carbetapentene Citrate, 974 Hydrochloride for Injection, 456 Cibenzoline Succinate, 1861
Carbidopa Hydrate, 406 Cefozopran Hydrochloride, 457 Tablets, 1862
L-Carbocisteine, 407 for Injection, 458 Ciclacillin, 507
Carbolic Acid, 985 Cefpiramide Sodium, 458 Ciclosporin, 509
for Disinfection, 986 Cefpirome Sulfate, 460 A, 509
Liquefied, 986 Cefpodoxime Proxetil, 461 Cilastatin Sodium, 509
Carbon Dioxide, 407 Cefroxadine Hydrate, 462 Cilazapril
Carboxymethylcellulose, 408 Cefsulodin Sodium, 464 Hydrate, 1863
Calcium, 409 Ceftazidime Tablets, 1864
Sodium, 410 for Injection, 1856 Cilostazol, 511
Cardamon, 1271 Hydrate, 466 Tablets, 512, 1866
Carmellose, 408 Cefteram Pivoxil, 468 Cimetidine, 513
Calcium, 409 Fine Granules, 469 Cimicifuga Rhizome, 1272, 1939
Sodium, 410 Ceftibuten Hydrate, 470 Cinchocaine Hydrochloride, 569
Carmofur, 411 Ceftizoxime Sodium, 471 Cinnamon
Carnauba Wax, 411 Ceftriaxone Sodium Hydrate, 472 Bark, 1272
Carteolol Hydrochloride, 412 Cefuroxime Bark, Powdered, 1273
Carumonam Sodium, 412 Axetil, 474 Oil, 1273
Cassia Seed, 1271 Sodium, 476 Cisplatin, 513
Castor Oil, 414 Cellacefate, 480 Citric Acid
Aromatic, 415 Cellulose Anhydrous, 514
Catalpa Fruit, 1271 Microcrystalline, 477 Hydrate, 515
Cefaclor, 415 Powdered, 480 Citrus Unshiu Peel, 1273
Capsules, 416 Celmoleukin (Genetical Recombina- Clarithromycin, 516
Compound Granules, 419 tion), 481 Tablets, 517
Fine Granules, 417 Cetanol, 484 Clemastine Fumarate, 519
Cefadroxil, 421 Cetirizine Hydrochloride, 1856 Clematis Root, 1274, 1939
Capsules, 1852 Tablets, 1857 Clindamycin Hydrochloride, 519
for Syrup, 1853 Cetraxate Hydrochloride, 485 Capsules, 520
Cefalexin, 422 Chenodeoxycholic Acid, 486 Clindamycin Phosphate, 521
Cefalotin Sodium, 423, 1854 Chloral Hydrate, 487 Injection, 1866
Cefapirin Sodium, 424 Chloramphenicol, 487 Clinofibrate, 522
Cefatrizine Propylene Glycolate, 425, Palmitate, 488 Clobetasol Propionate, 1867
1854 Sodium Succinate, 489 Clocapramine Hydrochloride Hydrate,
Cefazolin Sodium, 426 Chlordiazepoxide, 490 523
for Injection, 1854 Powder, 491 Clofedanol Hydrochloride, 524
Hydrate, 428 Tablets, 492, 1858 Clofibrate, 525
Cefbuperazone Sodium, 429 Chlorhexidine Capsules, 526
Cefcapene Pivoxil Hydrochloride Gluconate Solution, 493 Clomifene Citrate, 526
2014 Index Supplement I, JP XV

Tablets, 527 Oil, 542 Dextromethorphan Hydrobromide


Clomipramine Hydrochloride, 528 Starch, 542 Hydrate, 566
Clonazepam, 528 Cornus Fruit, 1279 Diagnostic Sodium Citrate Solution,
Clonidine Hydrochloride, 529 Cortisone Acetate, 543 1100
Cloperastine Hydrochloride, 530 Corydalis Tuber, 1279, 1940 Diastase, 567
Clorazepate Dipotassium, 1868 Powdered, 1940 and Sodium Bicarbonate Powder,
Capsules, 1869 Crataegus Fruit, 1941 567
Clotiazepam, 531 Creosote, 544, 1870 Diazepam, 567
Clotrimazole, 532 Cresol, 544 Dibasic
Clove, 1274 Solution, Saponated, 545 Calcium Phosphate, Anhydrous,
Oil, 1275 Croconazole Hydrochloride, 546 396, 1874
Powdered, 1274 Croscarmellose Sodium, 546 Calcium Phosphate Hydrate, 396,
Cloxacillin Sodium Hydrate, 533 Crude Glycyrrhiza Extract, 1297, 1875
Cloxazolam, 534 1943 Sodium Phosphate Hydrate, 568
CMC, 408 Crystalline Insulin Zinc Injection Dibekacin Sulfate, 569
Calcium, 409 (Aqueous Suspension), 765 Dibucaine Hydrochloride, 569
Sodium, 410 Crystal Violet, 881 Dichlorphenamide, 571
Cnidium Cyanamide, 548 Tablets, 572
Monnieri Fruit, 1275 Cyanocobalamin, 548, 1870 Diclofenac Sodium, 570
Rhizome, 1275, 1939 Injection, 549, 1871 Diclofenamide, 571
Rhizome, Powdered, 1276, 1940 Cyclopentolate Hydrochloride, 549 Tablets, 572
Cocaine Hydrochloride, 535 Cyclophosphamide Hydrate, 550 Dicloxacillin Sodium Hydrate, 573
Coconut Oil, 535 Cycloserine, 551 Diethylcarbamazine Citrate, 574
Codeine Phosphate Cyperus Rhizome, 1280, 1942 Tablets, 574
Hydrate, 536 Powdered, 1280, 1942 Diethylstilbestrol Diphosphate, 683
Powder, 1, 536 Cyproheptadine Hydrochloride Hy- Tablets, 684
Powder, 10, 537 drate, 551 Difenidol Hydrochloride, 575
Tablets, 537 L-Cysteine, 1871 Digenea, 1280
Cod Liver Oil, 538 Hydrochloride Hydrate, 1872 Digitoxin, 576
Coix Seed, 1276 Cytarabine, 552 Tablets, 576
Powdered, 1276 Digoxin, 578
Colchicine, 538 D Injection, 579
Colistin Tablets, 580
Sodium Methanesulfonate, 540 Daiokanzoto Extract, 1280 Dihydrocodeine Phosphate, 581
Sulfate, 541 Dantrolene Sodium Hydrate, 553 Powder, 1, 582
Colophonium, 1347 Daunorubicin Hydrochloride, 553 Powder, 10, 582
Compound Deferoxamine Mesilate, 554, 1873 Dihydroergotamine Mesilate, 583
Acrinol and Zinc Oxide Oil, 273 Dehydrated Alcohol, 639 Dihydroergotoxine Mesilate, 584
Diastase and Sodium Bicarbonate Dehydrocholate Sodium Injection, Dilazep Hydrochloride Hydrate, 586
Powder, 567 557 Diltiazem Hydrochloride, 586
Hycodenone Injection, 952 Dehydrocholic Acid, 556 Dilute
Iodine Glycerin, 770 Injection, 557, 1873 Hydrochloric Acid, 726
Methyl Salicylate Spirit, 879 Purified, 556 Iodine Tincture, 769
Oxycodone and Atropine Injection, Demethylchlortetracycline Hydro- Diluted Opium Powder, 940
953 chloride, 558 Dimemorfan Phosphate, 588
Oxycodone Injection, 952 Dental Dimenhydrinate, 588
Phellodendron Powder for Antiformin, 310 Tablets, 589
Cataplasm, 1332 Iodine Glycerin, 771 Dimercaprol, 590
Rhubarb and Senna Powder, 1346 Paraformaldehyde Paste, 969 Injection, 590
Salicylic Acid Spirit, 1080 Phenol with Camphor, 987 Dimorpholamine, 591
Scopolia Extract and Diastase Pow- Sodium Hypochlorite Solution, 310 Injection, 591
der, 1355 Triozinc Paste, 1210 Dinoprost, 592
Thianthol and Salicylic Acid Solu- Dermatol, 368 Dionin, 648
tion, 1165 Deslanoside, 559 Dioscorea Rhizome, 1282, 1942
Vitamin B Powder, 1230 Injection, 560, 1873 Powdered, 1282, 1942
Concentrated Dexamethasone, 560 Diphenhydramine, 593
Glycerin, 703 Dextran and Bromovalerylurea Powder, 593
Glycerol, 703 40, 561, 1873 Hydrochloride, 594
Condurango, 1276 40 Injection, 562 ,Phenol and Zinc Oxide Liniment,
Fluidextract, 1277, 1940 70, 563 595
Coptis Rhizome, 1277, 1940 Sulfate Sodium Sulfur 5, 564 Tannate, 595
Powdered, 1278, 1940 Sulfate Sodium Sulfur 18, 565 Diphenylhydantoin, 991
Corn Dextrin, 565 Powder, 992
Supplement I, JP XV Index 2015

Sodium for Injection, 992 Injection, 276 Glycyrrhiza, 1296, 1943


Tablets, 992 Solution, 277 Crude Glycyrrhiza, 1297, 1943
Diphtheria Epirizole, 621 Hangekobokuto, 1943
Tetanus Combined Toxoid, 596 Epirubicin Hydrochloride, 622 Hochuekkito, 1298, 1945
Toxoid, 596 Ergocalciferol, 624 Kakkonto, 1308, 1950
Dipyridamole, 597 Ergometrine Maleate, 625 Kamishoyosan, 1310, 1952
Disodium Edetate Hydrate, 598 Injection, 625 Keishibukuryogan, 1955
Disopyramide, 598 Tablets, 626 Nux Vomica, 1322, 1960
Distigmine Bromide, 599 Ergotamine Tartrate, 627 Ryokeijutsukanto, 1347, 1965
Tablets, 600 Erythromycin, 627 Saireito, 1349
Disulfiram, 600 Enteric-Coated Tablets, 1882 Scopolia, 1353, 1967
Dobutamine Hydrochloride, 601 Ethylsuccinate, 629
Dolichos Seed, 1282 Lactobionate, 629 F
Domperidone, 1876 Stearate, 630
Dopamine Hydrochloride, 602 Estazolam, 631 Famotidine, 655
Injection, 602, 1877 Estradiol Benzoate, 631 for Injection, 656, 1885
Dover's Powder, 1324 Injection, 632 Powder, 657
Doxapram Hydrochloride Hydrate, Injection (Aqueous Suspension), Tablets, 657
603 633 Faropenem Sodium
Doxifluridine, 604 Estriol, 633 for Syrup, 659, 1886
Capsules, 604 Injection (Aqueous Suspension), Hydrate, 658, 1885
Doxorubicin Hydrochloride, 605 634 Tablets, 660, 1886
for Injection, 1877 Tablets, 635 Felbinac, 1887
Doxycycline Hydrochloride Hydrate, Etacrynic Acid, 636 Fenbufen, 660
606 Tablets, 636 Fennel, 1285
Dried Ethacridine Lactate, 274 Oil, 1286
Aluminum Hydroxide Gel, 288 Ethambutol Hydrochloride, 637 Powdered, 1285
Aluminum Hydroxide Gel Fine Ethanol, 638 Fentanyl Citrate, 661
Granules, 289 Anhydrous, 639 Ferrous Sulfate Hydrate, 661
Aluminum Potassium Sulfate, 292 for Disinfection, 640 Fine Granules
Sodium Carbonate, 1096 Ethenzamide, 640 Cefaclor, 417
Sodium Sulfite, 1109 Ether, 641 Cefcapene Pivoxil Hydrochloride,
Thyroid, 1171 Anesthetic, 641 432
Yeast, 1241 Ethinylestradiol, 642 Cefdinir, 436
Droperidol, 608 Tablets, 642 Cefditoren Pivoxil, 438
Dydrogesterone, 609 Ethionamide, 643 Cefteram Pivoxil, 469
Tablets, 610 Ethosuximide, 644 Dried Aluminum Hydroxide Gel,
Ethoxybenzamide, 640 289
E Ethyl Etizolam, 1883
Aminobenzoate, 644 Flavin Adenine Dinucleotide Sodium,
Ecarazine Hydrochloride, 1185 Cysteine Hydrochloride, 645 662
Ecothiopate Iodide, 610 L-Cysteine Hydrochloride, 645 Flavoxate Hydrochloride, 664
Edrophonium Chloride, 611 Icosapentate, 646 Flomoxef Sodium, 664
Injection, 612, 1878 Parahydroxybenzoate, 647 for Injection, 666
EDTA Sodium Hydrate, 598 Ethylenediamine, 648 Flopropione, 667
Elcatonin, 612 Ethylmorphine Hydrochloride Hy- Capsules, 667
Eleutherococcus Senticosus Rhizome, drate, 648 Flucytosine, 668
1283 Etidronate Disodium, 649 Fludiazepam, 669
Emorfazone, 1878 Tablets, 650 Fluidextract
Enalapril Maleate, 1879 Etilefrine Hydrochloride, 650 Condurango, 1277, 1940
Tablets, 1880 Tablets, 651 Platycodon, 1334, 1962
Enflurane, 615 Etizolam, 652 Uva Ursi, 1371, 1969
Enoxacin Hydrate, 616 Fine Granules, 1883 Flunitrazepam, 670
Enviomycin Sulfate, 616 Tablets, 1884 Fluocinolone Acetonide, 670
Eperisone Hydrochloride, 618 Etodolac, 653 Fluocinonide, 672
Ephedra Herb, 1283 Etoposide, 653 Fluorescein Sodium, 673
Ephedrine Hydrochloride, 619 Eucalyptus Oil, 654 Fluorometholone, 673
Injection, 619, 1882 Eucommia Bark, 1284 Fluorouracil, 674
Powder, 620 Evodia Fruit, 1285 Fluoxymesterone, 675
Powder, 10, 620 Exsiccated Gypsum, 1298 Fluphenazine Enanthate, 676
Tablets, 621, 1882 Extract Flurazepam, 677
Epimedium Herb, 1284 Belladonna, 1263, 1939 Capsules, 678
Epinephrine, 276 Daiokanzoto, 1280 Hydrochloride, 678
2016 Index Supplement I, JP XV

Flurbiprofen, 679 Powdered, 1290, 1943 Honey, 1301


Foeniculated Ammonia Spirit, 1286 Powdered, Japanese, 1306, 1949 Houttuynia Herb, 1302
Folic Acid, 680 Geranium Herb, 1291 Human
Injection, 681, 1888 Powdered, 1291 Chorionic Gonadotrophin, 707
Tablets, 681, 1888 Ginger, 1291 Chorionic Gonadotrophin for
Formalin, 681 Powdered, 1292 Injection, 708
Water, 682 Ginseng, 1292 Menopausal Gonadotrophin, 708
Formoterol Fumarate Hydrate, 682 Powdered, 1293 Normal Immunoglobulin, 724
Forsythia Fruit, 1286 Glacial Acetic Acid, 269 Hycoato Injection, 953
Fosfestrol, 683, 1888 Glehnia Root, 1294, 1943 Hydralazine Hydrochloride, 724
Tablets, 684, 1888 Glibenclamide, 699 for Injection, 724
Fosfomycin Glucose, 700 Powder, 725
Calcium Hydrate, 685 Injection, 701, 1889 Tablets, 725, 1890
Sodium, 686 L-Glutamine, 1889 Hydrochloric Acid, 725
Sodium for Injection, 687 Glutathione, 701 Dilute, 726
Fradiomycin Sulfate, 688 Glycerin, 702 Lemonade, 727
Freeze-dried and Potash Solution, 704 Hydrochlorothiazide, 727
BCG Vaccine (for Percutaneous Concentrated, 703 Hydrocortisone, 728
Use), 335 Glycerol, 702 Acetate, 729
Botulism Antitoxin, Equine, 374 Concentrated, 703 and Diphenhydramine Ointment,
Diphtheria Antitoxin, Equine, 596 Glyceryl Monostearate, 704 730
Habu Antivenom, Equine, 716 Glycine, 704 Butyrate, 730
Inactivated Tissue Culture Rabies Glycyrrhiza, 1295 Sodium Phosphate, 731
Vaccine, 1054 Extract, 1296, 1943 Sodium Succinate, 732
Japanese Encephalitis Vaccine, 786 Extract, Crude, 1297, 1943 Succinate, 733
Live Attenuated Measles Vaccine, Powdered, 1296 Hydrocotarnine Hydrochloride
843 Gonadorelin Acetate, 705 Hydrate, 734
Live Attenuated Mumps Vaccine, Gonadotrophin Hydrogenated Oil, 735
902 Chorionic, 707 Hydrophilic
Live Attenuated Rubella Vaccine, for Injection, Chorionic, 708 Ointment, 735
1074 for Injection, Human Chorionic, Petrolatum, 981
Mamushi Antivenom, Equine, 841 708 Hydrous Lanolin, 807
Smallpox Vaccine, 1091 Human Chorionic, 707 Hydroxocobalamin Acetate, 736
Smallpox Vaccine Prepared in Cell Human Menopausal, 708 Hydroxypropylcellulose, 736
Culture, 1091 Serum, 710 Hydroxypropylmethylcellulose, 741
Tetanus Antitoxin, Equine, 1156 for Injection, Serum, 711 Hydroxyzine
Fritillaria Bulb, 1287, 1942 Gramicidin, 712 Hydrochloride, 739
Fructose, 689 Granules Pamoate, 739
Injection, 689, 1888 Calcium Paraaminosalicylate, 394 Hymecromone, 740
Furosemide, 690 Cefaclor Compound, 419 Hypromellose, 741
Tablets, 691 Pas-calcium Granules, 394 Phthalate, 743, 1891
Fursultiamine Hydrochloride, 692 Griseofulvin, 713
Tablets, 1890 I
G Guaiacol Glyceryl Ether, 714
Guaifenesin, 714 Ibudilast, 1892
Gabexate Mesilate, 693, 1889 Guanabenz Acetate, 715 Ibuprofen, 744
b-Galactosidase Guanethidine Sulfate, 716 Ichthammol, 744
(Aspergillus), 694 Gypsum, 1297 Idarubicin Hydrochloride, 745
(Penicillium), 695 for Injection, 746
Gallium (67Ga) Citrate Injection, 696 H Idoxuridine, 747
Gambir, 1287 Ophthalmic Solution, 747, 1893
Powdered, 1287 Haloperidol, 717 Ifenprodil Tartrate, 748
Gardenia Fruit, 1288 Tablets, 718 Imipenem
Powdered, 1288 Halothane, 719 and Cilastatin for Injection, 750
Gas Gangrene Antitoxin, Equine, 696 Haloxazolam, 719 Hydrate, 749
Gastrodia Tuber, 1289, 1943 Hangekobokuto Extract, 1943 Imipramine Hydrochloride, 751
Gelatin, 696 Hemp Fruit, 1298 Tablets, 752, 1893
Purified, 697 Heparin Sodium, 721 Immature Orange, 1302
Gentamicin Sulfate, 698 Injection, 722 Imperata Rhizome, 1302, 1949
Gentian, 1290, 1943 Hochuekkito Extract, 1298, 1945 Indenolol Hydrochloride, 753
and Sodium Bicarbonate Powder, Homatropine Hydrobromide, 722 Indigocarmine, 754
1290 Homochlorcyclizine Hydrochloride, Injection, 755
Japanese, 1306, 1949 723 Indium (111In) Chloride Injection, 755
Supplement I, JP XV Index 2017

Indometacin, 755 1882 pound, 953


Capsules, 756, 1893 Epinephrine, 276 Oxycodone, Compound, 952
Suppositories, 757 Ergometrine Maleate, 625 Oxytocin, 960
Influenza HA Vaccine, 758 Estradiol Benzoate, 632 Ozagrel Sodium for, 1917
Injection Famotidine for, 656, 1885 Papaverine Hydrochloride, 965,
Acetylcholine Chloride for, 271, Flomoxef Sodium for, 666 1918
1825 Folic Acid, 681, 1888 Peplomycin Sulfate for, 1918
Adrenaline, 276 Fosfomycin Sodium for, 687 Pethidine Hydrochloride, 981, 1918
Alprostadil, 1828 Fructose, 689, 1888 Phenolsulfonphthalein, 988
Amikacin Sulfate, 1830 Gallium (67Ga) Citrate, 696 Phenytoin Sodium for, 992
Aminophylline, 297, 1831 Glucose, 701, 1889 Piperacillin Sodium for, 998
Amobarbital Sodium for, 300 Heparin Sodium, 722 Prednisolone Sodium Succinate for,
Amphotericin B for, 304 Human Chorionic Gonadotrophin 1025, 1921
Ampicillin Sodium for, 1838 for, 708 Procainamide Hydrochloride, 1029
Estradiol Benzoate, (Aqueous Sus- Hycoato, 953 Procaine Hydrochloride, 1030
pension), 633 Hycodenone, Compound, 952 Progesterone, 1034
Estriol, (Aqueous Suspension), 634 Hydralazine Hydrochloride for, Protamine Sulfate, 1042, 1922
Insulin Zinc, (Aqueous Suspension), 724 Pyridoxine Hydrochloride, 1048,
763 Idarubicin Hydrochloride for, 746 1922
Insulin Zinc, Amorphous, (Aqueous Imipenem and Cilastatin for, 750 Reserpine, 1056, 1922
Suspension), 764 Indigocarmine, 755 Riboflavin Phosphate, 1062
Insulin Zinc, Crystalline, (Aqueous Indium (111In) Chloride, 755 Riboflavin Sodium Phosphate,
Suspension), 765 Insulin, 762 1062, 1922
Insulin Zinc Protamine, (Aqueous Iodinated (131I) Human Serum Albu- Serum Gonadotrophin for, 711
Suspension), 766 min, 768 Sodium Bicarbonate, 1094, 1925
Isophane Insulin, (Aqueous Suspen- Isoniazid, 781 0.9 Sodium Chloride, 1098
sion), 762 Isotonic Sodium Chloride, 1098 10 Sodium Chloride, 1098, 1925
Arbekacin Sulfate, 312 Levallorphan Tartrate, 814, 1900 Sodium Chromate (51Cr), 1099
L-Arginine Hydrochloride, 314, Lidocaine, 818 Sodium Citrate, for Transfusion,
1839 Lidocaine Hydrochloride, 818 1099, 1926
Ascorbic Acid, 317, 1839 Magnesium Sulfate, 839, 1900 Sodium Iodohippurate (131I), 1103
Atropine Sulfate, 324 D-Mannite, 842 Sodium Iotalamate, 1103
Aztreonam for, 1840 D-Mannitol, 842, 1903 Sodium Pertechnetate (99mTc), 1105
Benzylpenicillin Potassium for, Meglumine Amidotrizoate, 851 Sodium Thiosulfate, 1110, 1927
1841 Meglumine Iotalamate, 852 Sulfobromophthalein Sodium,
Calcium Chloride, 390, 1851 Meglumine Sodium Amidotrizoate, 1130
Cefazolin Sodium for, 1854 853 Sulpyrine, 1134, 1927
Cefepime Dihydrochloride for, 441 Meglumione Sodium Iodamide, 854 Suxamethonium Chloride for,
Cefmetazole Sodium for, 1855 Mepivacaine Hydrochloride, 860 1137, 1928
Cefotiam Hydrochloride for, 456 Metenolone Enanthate, 866 Suxamethonium Chloride, 1138,
Cefozopran Hydrochloride for, 458 Minocycline Hydrochloride for, 1928
Ceftazidime for, 1856 1904 Teceleukin for, (Genetical Recombi-
Chlorpheniramine Maleate, 499 Mitomycin C for, 1905 nation), 1148, 1929
Chlorpromazine Hydrochloride, Morphine and Atropine, 899 Testosterone Enanthate, 1154
503, 1860 Morphine Hydrochloride, 901 Testosterone Propionate, 1155
Chorionic Gonadotrophin for, 708 Neostigmine Methylsulfate, 910, Thallium (201Tl) Chloride, 1158
Clindamycin Phosphate, 1866 1912 Thiamine Chloride Hydrochloride,
Cyanocobalamin, 549, 1871 Nicardipine Hydrochloride, 912, 1161, 1930
Dehydrocholate Sodium, 557 1912 Thiamylal Sodium for, 1164
Dehydrocholic Acid, 557, 1873 Nicotinic Acid, 921, 1912 Thiopental Sodium for, 1167, 1930
Deslanoside, 560, 1873 Noradrenaline, 931, 1915 Tobramycin, 1930
Dextran 40, 562 Noradrenaline Hydrochloride, 931 Tranexamic Acid, 1193
Digoxin, 579 Norepinephrine, 931 Tubocurarine Chloride Hydrochlo-
Dimercaprol, 590 Norepinephrine Hydrochloride, ride, 1213, 1931
Dimorpholamine, 591 931 Tubocurarine Hydrochloride, 1213
Diphenylhydantoin Sodium for, Operidine, 981 Vancomycin Hydrochloride for,
992 Opium Alkaloids and Atropine, 1223
Dopamine Hydrochloride, 602, 942 Vasopressin, 1223
1877 Opium Alkaloids and Scopolamine, Vinblastine Sulfate for, 1227
Doxorubicin Hydrochloride for, 943 Vitamin B1 Hydrochloride, 1161
1877 Opium Alkaloids Hydrochlorides, Vitamin B2 Phosphate Ester, 1062
Edrophonium Chloride, 612, 1878 942 Vitamin B6, 1048
Ephedrine Hydrochloride, 619, Oxycodone and Atropine, Com- Vitamin B12, 549
2018 Index Supplement I, JP XV

Vitamin C, 317 Gentian, 1306, 1949 Hydrochloride Injection, 818


Water for, 1235, 1934 Gentian, Powdered, 1306, 1949 Injection, 818
Weak Opium Alkaloids and Scopola- Valerian, 1306, 1950 Light
mine, 945 Valerian, Powdered, 1307, 1950 Anhydrous Silicic Acid, 1087
Xylitol, 1241, 1935 Josamycin, 786 Liquid Paraffin, 967
Insulin, 758 Propionate, 788 Lilium Bulb, 1958
Human (Genetical Recombination), Tablets, 1897 Limaprost Alfadex, 819
760 Jujube, 1307 Liniment
Injection, 762 Seed, 1307 Diphenhydramine, Phenol and Zinc
Zinc Injection (Aqueous Suspen- Oxide, 595
sion), 763 K Lincomycin Hydrochloride Hydrate,
Amorphous, 764 820
Crystalline, 765 Kainic Acid Lindera Root, 1313, 1959
Zinc Protamine Injection (Aqueous and Santonin Powder, 790 Liothyronine Sodium, 821
Suspension), 766 Hydrate, 789 Tablets, 822
Iodamide, 767 Kakkonto Extract, 1308, 1950 Liquefied
Iodinated (131I) Human Serum Albumin Kallidinogenase, 790 Carbolic Acid, 986
Injection, 768 Kamishoyosan Extract, 1310, 1952 Phenol, 986
Iodine, 768 Kanamycin Liquid Paraffin, 966
Salicylic Acid and Phenol Spirit, Monosulfate, 793 Lisinopril
772 Sulfate, 794 Hydrate, 823
Tincture, 769 Kaolin, 795 Tablets, 824
Iodoform, 773 Keishibukuryogan Extract, 1955 Lithium Carbonate, 826
Iopamidol, 773 Ketamine Hydrochloride, 795 Lithospermum Root, 1313, 1959
Iotalamic Acid, 774 Ketoprofen, 796 Live Oral Poliomyelitis Vaccine, 1005
Iotroxic Acid, 775 Ketotifen Fumarate, 797 Longgu, 1313
Ipecac, 1303, 1949 Kitasamycin, 798 Lonicera Leaf and Stem, 1314
Powdered, 1303, 1949 Acetate, 799 Loquat Leaf, 1314
Syrup, 1304 Tartrate, 800 Lorazepam, 827
Ipratropium Bromide Hydrate, 776 Lotion
Iproveratril Hydrochloride, 1225 L Sulfur and Camphor, 1130
Isepamicin Sulfate, 777 Low Substituted Hydroxypropylcellu-
Isoflurane, 778 Labetalol Hydrochloride, 1898 lose, 738
L-Isoleucine, 780 Tablets, 1899 Loxoprofen Sodium Hydrate, 828
Isoniazid, 780 Lactic Acid, 801 Lycium
Injection, 781 Lactose, 803 Bark, 1315, 1959
Tablets, 781 Anhydrous, 802, 1900 Fruit, 1315
Isophane Insulin Injection (Aqueous Hydrate, 803 Lysine Hydrochloride, 829
Suspension), 762 Lactulose, 804 L-Lysine Hydrochloride, 829
l-Isoprenaline Hydrochloride, 782 Lanatoside C, 805 Lysozyme Hydrochloride, 830
Isopropanol, 783 Tablets, 806
Isopropyl Alcohol, 783 Lanolin M
Isopropylantipyrine, 783 Hydrous, 807
Isosorbide, 784 Purified, 808 Macrogol
Dinitrate, 785 Lard, 808 400, 830
Dinitrate Tablets, 786 Latamoxef Sodium, 809 1500, 831
Isotonic Lauromacrogol, 810 4000, 832
Salt Solution, 1098 Lemonade 6000, 832
Sodium Chloride Injection, 1098 Hydrochloric Acid, 727 20000, 833
Sodium Chloride Solution, 1098, Lenampicillin Hydrochloride, 810 Ointment, 833
1893 Leonurus Herb, 1958 Magnesium
Isoxsuprine Hydrochloride, 1894 L-Leucine, 812 Carbonate, 834
Tablets, 1895 Leucomycin, 798 Oxide, 835
Itraconazole, 1896 Acetate, 799 Silicate, 836
Tartrate, 800 Stearate, 837
J Levallorphan Tartrate, 813 Sulfate Hydrate, 838
Injection, 814, 1900 Sulfate Injection, 839, 1900
Japanese Levodopa, 814 Sulfate Mixture, 839
Angelica Root, 1305 Levomepromazine Maleate, 815 Magnolia
Angelica Root, Powdered, 1305 Levothyroxine Sodium Bark, 1315, 1959
Encephalitis Vaccine, 786 Hydrate, 816 Bark, Powderd, 1316, 1959
Encephalitis Vaccine, Freeze-dried, Tablets, 817 Flower, 1317
786 Lidocaine, 818 Mallotus Bark, 1317
Supplement I, JP XV Index 2019

Maltose Hydrate, 840 Powder, 10, 876 Narcotine, 936


Manidipine Hydrochloride, 1900 Methylergometrine Maleate, 876 Hydrochloride, 937
Tablets, 1902 Tablets, 877 Natamycin, 994
D-Mannite Injection, 842 Methyl Natural Aluminum Silicate, 289
D-Mannitol, 841 Parahydroxybenzoate, 878 Nelumbo Seed, 1320
Injection, 842, 1903 Salicylate, 881 Neomycin Sulfate, 688
Maprotiline Hydrochloride, 842 Methylprednisolone, 879 Neostigmine Methylsulfate, 909
Meclofenoxate Hydrochloride, 843 Succinate, 880 Injection, 910, 1912
Mecobalamin, 844 Methylrosanilinium Chloride, 881 Netilmicin Sulfate, 910
Medazepam, 845, 1903 Methyltestosterone, 882 Nicardipine Hydrochloride, 911
Medicinal Tablets, 883 Injection, 912, 1912
Carbon, 845 Meticrane, 884 Nicergoline, 913
Soap, 846 Metildigoxin, 885 Powder, 914
Mefenamic Acid, 847 Metoclopramide, 886 Tablets, 915
Mefloquine Hydrochloride, 848 Tablets, 886 Niceritrol, 916
Mefruside, 849 Metoprolol Tartrate, 887 Nicomol, 917
Tablets, 849, 1903 Tablets, 888 Tablets, 918
Meglumine, 850 Metronidazole, 889 Nicorandil, 919, 1912
Amidotrizoate Injection, 851 Tablets, 889 Nicotinamide, 919
Iotalamate Injection, 852 Metyrapone, 890 Nicotinic Acid, 920
Sodium Amidotrizoate Injection, Mexiletine Hydrochloride, 891 Injection, 921, 1912
853 Miconazole, 892 Nifedipine, 922
Sodium Iodamide Injection, 854 Nitrate, 893 Nilvadipine, 923
Melphalan, 855 Microcrystalline Cellulose, 477 Tablets, 924
Menatetrenone, 855 Micronomicin Sulfate, 893 Nitrazepam, 925
Mentha Midecamycin, 894 Nitrendipine, 926
Herb, 1317 Acetate, 895 Tablets, 927
Oil, 1318 Migrenin, 896 Nitrogen, 928
Water, 1318 Minocycline Hydrochloride, 897 Nitroglycerin Tablets, 928
dl-Menthol, 856 for Injection, 1904 Nitrous Oxide, 929
l-Menthol, 857 Mitomycin C, 898 Nizatidine, 1912
Mepenzolate Bromide, 857 for Injection, 1905 Capsules, 1913
Mepirizole, 621 Mizoribine, 1906 Noradrenaline, 930
Mepitiostane, 858 Tablets, 1907 Hydrochloride Injection, 931
Mepivacaine Hydrochloride, 859 Monobasic Calcium Phosphate Injection, 931, 1915
Injection, 860 Hydrate, 397 Norepinephrine, 930
Mequitazine, 861 Monosodium Trichloroethyl Phos- Hydrochloride Injection, 931
Merbromin, 862 phate, 1202 Injection, 931
Solution, 863 Syrup, 1203 Norethisterone, 932
Mercaptopurine Hydrate, 861 Morphine Norfloxacin, 932
Mercurochrome, 862 and Atropine Injection, 899 Norgestrel, 933
Solution, 863 Hydrochloride Hydrate, 900 and Ethinylestradiol Tablets, 934
Meropenem Hydrate, 863 Hydrochloride Injection, 901 Nortriptyline Hydrochloride, 935
Mestranol, 864 Hydrochloride Tablets, 902, 1908 Noscapine, 936
Metenolone Moutan Bark, 1318 Hydrochloride Hydrate, 937
Acetate, 865 Powdered, 1319 Notopterygium Rhizome, 1321, 1960
Enanthate, 866 Mulberry Bark, 1320, 1960 Nuphar Rhizome, 1321, 1960
Enanthate Injection, 866 Mupirocin Calcium Hydrate, 902 Nux Vomica, 1321
Metformin Hydrochloride, 867 Extract, 1322, 1960
Tablets, 867 N Extract Powder, 1323
Methamphetamine Hydrochloride, Tincture, 1323
868 Nabumetone, 1908 Nystatin, 937
L-Methionine, 869 Tablets, 1910
Methotrexate, 869 Nadolol, 904 O
Methoxsalen, 870 Nafamostat Mesilate, 1911
Methylbenactyzium Bromide, 871 Nalidixic Acid, 905 Ofloxacin, 938
Methylcellulose, 871 Naloxone Hydrochloride, 906 Ointment
Methyldopa Naphazoline Absorptive, 265
Hydrate, 873 and Chlorpheniramine Solution, Acrinol and Zinc Oxide, 274
Tablets, 874, 1903 906 Betamethasone Valerate and Gen-
dl-Methylephedrine Hydrochloride, Hydrochloride, 907 tamicin Sulfate, 362
875 Nitrate, 908 Bufexamac, 381
Powder, 876 Naproxen, 908
2020 Index Supplement I, JP XV

Ointment (continued) Pantethine, 964 Phenytoin, 991


Hydrocortisone and Diphenhydra- Papaverine Hydrochloride, 965 Powder, 992
mine, 730 Injection, 965, 1918 Sodium for Injection, 992
Hydrophilic, 735 Paracetamol, 267 Tablets, 992
Macrogol, 833 Paraffin, 966 Phytomenadione, 993
Polyethylene Glycol, 833 Light Liquid, 967 Phytonadione, 993
Simple, 1089 Liquid, 966 Picrasma Wood, 1333
Sulfur, Salicylic Acid and Thianthol, Paraformaldehyde, 968 Powdered, 1333
1131 Parnaparin Sodium, 969 Pilocarpine Hydrochloride, 994
White, 1238 Pas-calcium Pimaricin, 994
Zinc Oxide, 1246 Granules, 394 Pindolol, 995
Olive Oil, 939 Hydrate, 395 Pinellia Tuber, 1333
Omeprazole, 1915 Paste Pipemidic Acid Hydrate, 996
Operidine, 980 Arsenical, 315 Piperacillin
Injection, 981 Paraformaldehyde, Dental, 969 Hydrate, 1919
Ophiopogon Tuber, 1324 Triozinc, Dental, 1210 Sodium, 997
Ophthalmic Solution Peach Kernel, 1327, 1961 Sodium for Injection, 998
Idoxuridine, 747, 1893 Powdered, 1327, 1961 Piperazine
Silver Nitrate, 1088 Peanut Oil, 971 Adipate, 999
Zinc Sulfate, 1247 Penbutolol Sulfate, 972 Phosphate Hydrate, 999
Opium Penicillin G Potassium, 349 Phosphate Tablets, 1000
Ipecac Powder, 1324 Pentazocine, 972 Pirarubicin, 1000
Powdered, 939 Pentobarbital Calcium, 973 Pirenoxine, 1001
Tincture, 940 Pentoxyverine Citrate, 974 Pirenzepine Hydrochloride Hydrate,
Opium Alkaloids Peony Root, 1328 1002
and Atropine Injection, 942 Powdered, 1329 Piroxicam, 1003
and Scopolamine Injection, 943 Peplomycin Sulfate, 975 Pivmecillinam Hydrochloride, 1004
Hydrochlorides, 941 for Injection, 1918 Plantago
Hydrochlorides Injection, 942 Perilla Herb, 1329, 1961 Herb, 1334
Orange Perphenazine, 977 Seed, 1334
Oil, 946 Maleate, 978 Platycodon
Peel Syrup, 1325 Maleate Tablets, 979 Fluidextract, 1334, 1962
Peel Tincture, 1325 Tablets, 977 Root, 1334
Orciprenaline Sulfate, 946 Pethidine Hydrochloride, 980 Root, Powdered, 1335
Oriental Bezoar, 1325 Injection, 981, 1918 Polyethylene Glycol
Oxapium Iodide, 947 Petrolatum 400, 830
Oxaprozin, 948 Hydrophilic, 981 1500, 831
Oxazolam, 948 White, 981 4000, 832
Oxetacaine, 949 Yellow, 982 6000, 832
Oxethazaine, 949 Petroleum Benzin, 982 20000, 833
Oxprenolol Hydrochloride, 950 Peucedanum Root, 1961 Ointment, 833
Oxybuprocaine Hydrochloride, 951 Pharbitis Seed, 1330 Polygala Root, 1335, 1962
Oxycodone Hydrochloride Hydrate, Phellodendron, Powdered, 1335, 1962
951 Albumin Tannate and Bismuth Sub- Polygonatum Rhizome, 1336
Oxydol, 954 nitrate Powder, 1332 Polygonum Root, 1336, 1962
Oxygen, 955 Bark, 1330 Polymixin B Sulfate, 1006
Oxymetholone, 956 Bark, Powdered, 1331 Polyoxyethylene Lauryl Alcohol Ether,
Oxytetracycline Hydrochloride, 956 Phenazone, 310 810
Oxytocin, 958 Phenethicillin Potassium, 983 Polyoxyl 40 Stearate, 1007
Injection, 960 Phenobarbital, 984 Polyporus Sclerotium, 1336, 1962
Oyster Shell, 1326 Powder, 985 Powdered, 1337, 1963
Powdered, 1326 Powder, 10, 985 Polysorbate 80, 1007
Ozagrel Sodium, 1916 Phenol, 985 Polyvidone, 1015
for Injection, 1917 and Zinc Oxide Liniment, 987 Polyvinylpyrrolidone, 1015
for Disinfection, 986 Poria Sclerotium, 1337
P Liquefied, 986 Powdered, 1337
Phenolated Water, 987 Potash Soap, 1007
Panax Japonicus Rhizome, 1326, for Disinfection, 987 Potassium
1960 Phenolsulfonphthalein, 988 Bromide, 1008
Powdered, 1327, 1960 Injection, 988 Canrenoate, 1008
Pancreatin, 961 L-Phenylalanine, 989 Carbonate, 1009
Pancuronium Bromide, 962 Phenylbutazone, 990 Chloride, 1009
Panipenem, 962 Phenylephrine Hydrochloride, 990 Clavulanate, 1010
Supplement I, JP XV Index 2021

Guaiacolsulfonate, 1011 Swertia and Sodium Bicarbonate, Sophora Root, 1364, 1969
Hydroxide, 1012 1367 Sweet Hydrangea Leaf, 1365
Iodide, 1013 Thiamine Chloride Hydrochloride, Swertia Herb, 1366
Permanganate, 1013 1162 Tragacanth, 1369
Sulfate, 1014 Vitamin B, Compound, 1230 Turmeric, 1969
Potato Starch, 1014 Vitamin B1 Hydrochloride, 1162 Zanthoxylum Fruit, 1371
Povidone, 1015 Vitamin B2, 1060 Pranoprofen, 1018
Iodine, 1017 Vitamin C, 317 Pravastatin Sodium, 1018
Powder Zinc Oxide Starch, 1246 Prazepam, 1020
Ascorbic Acid, 317 Powdered Tablets, 1021
Chlordiazepoxide, 491 Acacia, 1251 Precipitated Calcium Carbonate, 389
Chlorpheniramine and Calcium, Agar, 1253 Prednisolone, 1021
497 Alisma Rhizome, 1253 Acetate, 1023
Chlorpheniramine Maleate, 499 Aloe, 1255 Sodium Succinate for Injection,
Codeine Phosphate, 1, 536 Amomum Seed, 1256 1025, 1921
Codeine Phosphate, 10, 537 Atractylodes Lancea Rhizome, Succinate, 1024
Diastase and Sodium Bicarbonate, 1260 Tablets, 1022
567 Atractylodes Rhizome, 1261, 1939 Primidone, 1026
Diastase and Sodium Bicarbonate, Calumba, 1268, 1939 Probenecid, 1027
Compound, 567 Capsicum, 1269 Tablets, 1027
Dihydrocodeine Phosphate, 1, Cellulose, 480 Procainamide Hydrochloride, 1028
582 Cinnamon Bark, 1273 Injection, 1029
Dihydrocodeine Phosphate, 10, Clove, 1274 Tablets, 1029
582 Cnidium Rhizome, 1276, 1940 Procaine Hydrochloride, 1030
Diluted Opium, 940 Coix Seed, 1276 Injection, 1030
Diphenhydramine and Coptis Rhizome, 1278, 1940 Procarbazine Hydrochloride, 1031
Bromovalerylurea, 593 Corydalis Tuber, 1940 Procaterol Hydrochloride Hydrate,
Diphenylhydantoin, 992 Cyperus Rhizome, 1280, 1942 1032
Dover's, 1324 Dioscorea Rhizome, 1282, 1942 Processed
Ephedrine Hydrochloride, 620 Fennel, 1285 Aconite Root, 1338, 1963
Ephedrine Hydrochloride, 10, Gambir, 1287 Aconite Root, Powdered, 1339,
620 Gardenia Fruit, 1288 1963
Famotidine, 657 Gentian, 1290, 1943 Ginger, 1341, 1964
Gentian and Sodium Bicarbonate, Geranium Herb, 1291 Prochlorperazine Maleate, 1033
1290 Ginger, 1292 Tablets, 1033
Hydralazine Hydrochloride, 725 Ginseng, 1293 Progesterone, 1034
Kainic Acid and Santonin, 790 Glycyrrhiza, 1296 Injection, 1034
dl-Methylephedrine Hydrochloride, Ipecac, 1303, 1949 Proglumide, 1035
876 Japanese Angelica Root, 1305 Promethazine Hydrochloride, 1036
dl-Methylephedrine Hydrochloride, Japanese Gentian, 1306, 1949 Propantheline Bromide, 1036
10, 876 Japanese Valerian, 1307, 1950 Propranolol Hydrochloride, 1037
Nicergoline, 914 Magnolia Bark, 1316, 1959 Tablets, 1038
Nux Vomica Extract, 1323 Moutan Bark, 1319 Propyl Parahydroxybenzoate, 1039
Opium Ipecac, 1324 Opium, 939 Propylene Glycol, 1039
Phellodendron, Albumin Tannate Oyster Shell, 1326 Propylthiouracil, 1040
and Bismuth Subnitrate, 1332 Panax Japonicus Rhizome, 1327, Tablets, 1040
Phenobarbital, 985 1960 Propyphenazone, 783
Phenobarbital, 10, 985 Peach Kernel, 1327, 1961 Prostaglandin
Phenytoin, 992 Peony Root, 1329 E1, 286
Reserpine, 1057 Phellodendron Bark, 1331 E1 a-Cyclodextrin Clathrate Com-
Reserpine, 0.1, 1057 Picrasma Wood, 1333 pound, 287
Rhubarb and Senna, Compound, Platycodon Root, 1335 F2a, 592
1346 Polygala Root, 1335, 1962 Protamine Sulfate, 1041, 1921
Riboflavin, 1060 Polyporus Sclerotium, 1337, 1963 Injection, 1042, 1922
Salicylated Alum, 1078 Poria Sclerotium, 1337 Prothionamide, 1042
Scopolia Extract, 1354 Processed Aconite Root, 1339, Protirelin, 1043
Scopolia Extract and Carbon, 1355 1963 Tartrate Hydrate, 1044
Scopolia Extract and Diastase, Com- Rhubarb, 1345 Prunella Spike, 1341
pound, 1355 Rose Fruit, 1346 Pueraria Root, 1341
Scopolia Extract and Ethyl Scutellaria Root, 1359, 1967 Pullulan, 1045
Aminobenzoate, 1355 Senega, 1361, 1968 Purified
Scopolia Extract, Paparerine and Senna Leaf, 1362 Dehydrocholic Acid, 556
Ethyl Aminobenzoate, 1356 Smilax Rhizome, 1364, 1968 Gelatin, 697
2022 Index Supplement I, JP XV

Purified (continued) Powdered, 1364, 1968


Lanolin, 808 S Sodium
Shellac, 1085 Acetate Hydrate, 1091
Water, 1236 Saccharated Pepsin, 1074 Aurothiomalate, 1092
Water, Sterile, 1236 Saccharin, 1075 Benzoate, 1093
Pyrantel Pamoate, 1045 Sodium, 1076 Bicarbonate, 1093
Pyrazinamide, 1046 Sodium Hydrate, 1076 Bicarbonate and Bitter Tincture Mix-
Pyridostigmine Bromide, 1047 Safflower, 1348 ture, 1364
Pyridoxine Hydrochloride, 1047 Saffron, 1349 Bicarbonate Injection, 1094, 1925
Injection, 1048, 1922 Saireito Extract, 1349 Bisulfite, 1094
Pyroxylin, 1049 Salazosulfapyridine, 1077 Borate, 1095
Pyrrolnitrin, 1049 Salbutamol Sulfate, 1078 Bromide, 1095
Salicylated Alum Powder, 1078 Carbonate, Dried, 1096
Q Salicylic Acid, 1079, 1924 Carbonate Hydrate, 1096
Plaster, Adhesive, 1080 Chloride, 1097
Quick Lime, 393 Spirit, 1080 Chloride Injection, 0.9, 1098
Quinidine Sulfate Hydrate, 1050 Santonin, 1081 Chloride Injection, 10, 1098,
Quinine Saponated Cresol Solution, 545 1925
Ethyl Carbonate, 1051 Saposhnikovia Root, 1352, 1966 Chloride Solution, Isotonic, 1098,
Hydrochloride Hydrate, 1052 Sappan Wood, 1352 1893
Sulfate Hydrate, 1053 Saussurea Root, 1352, 1966 Chromate (51Cr) Injection, 1099
Schisandra Fruit, 1352 Citrate Hydrate, 1099
R Schizonepeta Spike, 1353 Citrate Injection for Transfusion,
Scopolamine 1099, 1926
Ranitidine Hydrochloride, 1054 Butylbromide, 1082 Citrate Solution, Diagnostic, 1100
Rape Seed Oil, 1055 Hydrobromide Hydrate, 1083 Cromoglicate, 1100
Red Ginseng, 1342 Scopolia Extract, 1353, 1967 Fusidate, 1101
Rehmannia Root, 1343, 1964 and Carbon Powder, 1355 Hydrogen Sulfite, 1094
Reserpine, 1055 and Ethyl Aminobenzoate Powder, Hydroxide, 1101
Injection, 1056, 1922 1355 Iodide, 1102
Powder, 1057 and Tannic Acid Suppositories, Iodide (123I) Capsules, 1103
Powder, 0.1, 1057 1357 Iodide (131I) Capsules, 1103
Tablets, 1057 ,Papaverine and Ethyl Aminobenzo- Iodide (131I) Solution, 1103
Retinol ate Powder, 1356 Iodohippurate (131I) Injection, 1103
Acetate, 1058 Powder, 1354 Iotalamate Injection, 1103
Palmitate, 1059 Rhizome, 1357, 1967 Lauryl Sulfate, 1104
Rhubarb, 1344 Scutellaria Root, 1358, 1967 Metabisulfite, 1108
Powdered, 1345 Powdered, 1359, 1967 Pertechnetate (99mTc) Injection,
Riboflavin, 1059 Senega, 1360, 1968 1105
Butyrate, 1060 Powdered, 1361, 1968 Picosulfate Hydrate, 1105
Phosphate, 1061 Syrup, 1361 Polystyrene Sulfonate, 1106
Phosphate Injection, 1062 Senna Leaf, 1361 Prasterone Sulfate Hydrate, 1107
Powder, 1060 Powdered, 1362 Pyrosulfate, 1108
Sodium Phosphate, 1061 L-Serine, 1925 Salicylate, 1108
Sodium Phosphate Injection, 1062, Serrapeptase, 1083 Starch Glycolate, 1926
1922 Serum Gonadotrophin, 710 Sulfite, Dried, 1109
Ribostamycin Sulfate, 1063 for Injection, 711 Thiosulfate Hydrate, 1109
Rice Starch, 1346 Sesame Oil, 1084 Thiosulfate Injection, 1110, 1927
Rifampicin, 1064 Shellac Valproate, 1110
Capsules, 1065 Purified, 1085 Solution
Ringer's Solution, 1066, 1922 Siccanin, 1086 Adrenaline, 277
Ritodrine Hydrochloride, 1067 Silver Alum, 293
Tablets, 1068 Nitrate, 1088 Benzalkonium Chloride, 342
Rokitamycin, 1069 Nitrate Ophthalmic Solution, 1088 Benzethonium Chloride, 345
Tablets, 1923 Protein, 1088 Chlorhexidine Gluconate, 493
Rose Fruit, 1346 Protein Solution, 1089 Cresol, 545
Powdered, 1346 Simple Dental Sodium Hypochlorite, 310
Rosin, 1347 Ointment, 1089 Diagnostic Sodium Citrate, 1100
Roxatidine Acetate Hydrochloride, Syrup, 1089 Epinephrine, 277
1070 Sinomenium Stem, 1363 Glycerin and Potash, 704
Extended-release Capsules, 1071 Sisomicin Sulfate, 1090 Isotonic Salt, 1098
Roxithromycin, 1073, 1923 Slaked Lime, 392 Isotonic Sodium Chloride, 1098,
Ryokeijutsukanto Extract, 1347, 1965 Smilax Rhizome, 1364, 1968 1893
Supplement I, JP XV Index 2023

Merbromin, 863 Tablets, 1132 433


Mercurochrome, 863 Sulpyrine Cefditoren Pivoxil, 438
Naphazoline and Chlorpheniramine, Hydrate, 1133 Cetirizine Hydrochloride, 1857
906 Injection, 1134, 1927 Chlordiazepoxide, 492, 1858
Ringer's, 1066, 1922 Sultamicillin Tosilate Hydrate, 1134, Chlorphenesin Carbamate, 1859
Saponated Cresol, 545 1927 Chlorpheniramine Maleate, 500
Silver Protein, 1089 Sultiame, 1136 Chlorpromazine Hydrochloride,
Sodium Citrate, Diagnostic, 1100 Suppositories 503, 1861
Sodium Iodide (131I), 1103 Bisacodyl, 368, 1843 Chlorpropamide, 505, 1861
D-Sorbitol, 1113 Indometacin, 757 Cibenzoline Succinate, 1862
Thianthol and Salicylic Acid, Com- Scopolia Extract and Tannic Acid, Cilazapril, 1864
pound, 1165 1357 Cilostazol, 512, 1866
Tolnaftate, 1189 Suxamethonium Chloride Clarithromycin, 517
Sophora Root, 1364, 1968 for Injection, 1137, 1928 Clomifene Citrate, 527
Powdered, 1364, 1969 Hydrate, 1137 Codeine Phosphate, 537
Sorbitan Sesquioleate, 1111 Injection, 1138, 1928 Dichlorphenamide, 572
D-Sorbitol, 1112 Sweet Hydrangea Leaf, 1365 Diclofenamide, 572
Solution, 1113 Powdered, 1365 Diethylcarbamazine Citrate, 574
Soybean Oil, 1113 Swertia Diethylstilbestrol Diphosphate, 684
Spectinomycin Hydrochloride Hydrate, and Sodium Bicarbonate Powder, Digitoxin, 576
1114 1367 Digoxin, 580
Spiramycin Acetate, 1114 Herb, 1365 Dimenhydrinate, 589
Spirit Herb, Powdered, 1366 Diphenylhydantoin, 992
Capsicum and Salicylic Acid, 1270 Synthetic Distigmine Bromide, 600
Foeniculated Ammonia, 1286 Aluminum Silicate, 290 Dydrogesterone, 610
Iodine Salicylic Acid and Phenol, Camphor, 402 Enalapril Maleate, 1880
772 Syrup Ephedrine Hydrochloride, 621,
Methyl Salicylate, Compound, 879 Amphotericin B, 305 1882
Salicylic Acid, 1080 Cefadroxil for, 1853 Ergometrine Maleate, 626
Salicylic Acid, Compound, 1080 Faropenem Sodium for, 659, 1886 Erythromycin Enteric-Coated, 1882
Spironolactone, 1116 Ipecac, 1304 Estriol, 635
Starch Monosodium Trichloroethyl Phos- Etacrynic Acid, 636
Corn, 542 phate, 1203 Ethinylestradiol, 642
Glycolate, Sodium, 1926 Orange Peel, 1325 Etidronate Disodium, 650
Potato, 1014 Senega, 1361 Etilefrine Hydrochloride, 651
Rice, 1346 Simple, 1089 Etizolam, 1884
Wheat, 1237 Triclofos Sodium, 1203 Famotidine, 657
Stearic Acid, 1116 Faropenem Sodium, 660, 1886
Stearyl Alcohol, 1117 T Folic Acid, 681, 1888
Sterile Purified Water, 1236 Fosfestrol, 684, 1888
Streptomycin Sulfate, 1117 Tablets Furosemide, 691
Sucralfate Hydrate, 1118 Acetylsalicylic Acid, 319 Griseofulvin, 1890
Sucrose, 1120 Ajmaline, 279, 1826 Haloperidol, 718
Sulbactam Sodium, 1122, 1927 Alacepril, 280 Hydralazine Hydrochloride, 725,
Sulbenicillin Sodium, 1123 Alminoprofen, 1827 1890
Sulfadiazine Silver, 1124 Amitriptyline Hydrochloride, 299, Imipramine Hydrochloride, 752,
Sulfafurazole, 1128 1831 1893
Sulfamethizole, 1125 Amlexanox, 1833 Isoniazid, 781
Sulfamethoxazole, 1126 Amosulalol Hydrochloride, 1836 Isosorbide Dinitrate, 786
Sulfamonomethoxine Hydrate, 1126 Amphotericin B, 305 Isoxsuprine Hydrochloride, 1895
Sulfasalazine, 1077 Aspirin, 319 Josamycin, 1897
Sulfinpyrazone, 1127, 1927 Azathioprine, 326 Labetalol Hydrochloride, 1899
Tablets, 1128, 1927 Baclofen, 332, 1841 Lanatoside C, 806
Sulfisomezole, 1126 Benidipine Hydrochloride, 339 Levothyroxine Sodium, 817
Sulfisoxazole, 1128 Betahistine Mesilate, 354 Liothyronine Sodium, 822
Sulfobromophthalein Sodium, 1129 Betamethasone, 356 Lisinopril, 824
Injection, 1130 Bezafibrate Sustained Release, 365 Manidipine Hydrochloride, 1902
Sulfur, 1130 Bisoprolol Fumarate, 1844 Mefruside, 849, 1903
and Camphor Lotion, 1130 Bucillamine, 1845 Metformin Hydrochloride, 867
,Salicylic Acid and Thianthol Oint- Buformine Hydrochloride, 1849 Methyldopa, 874, 1903
ment, 1131 Buformine Hydrochloride Enteric- Methylergometrine Maleate, 877
Sulpiride, 1131 coated, 1847 Methyltestosterone, 883
Capsules, 1132 Cefcapene Pivoxil Hydrochloride, Metoclopramide, 886
2024 Index Supplement I, JP XV

Tablets (continued) Tetracycline Hydrochloride, 1157 Capsules, 1192


Metoprolol Tartrate, 888 Thallium (201Tl) Chloride Injection, Injection, 1193
Metronidazole, 889 1158 Tablets, 1193
Mizoribine, 1907 Theophylline, 1158 Trapidil, 1194
Morphine Hydrochloride, 902, Thiamazole, 1159 Trepibutone, 1195
1908 Tablets, 1160 Tretoquinol Hydrochloride, 1209
Nabumetone, 1910 Thiamine Triamcinolone, 1195
Nicergoline, 915 Chloride Hydrochloride, 1160 Acetonide, 1196
Nicomol, 918 Chloride Hydrochloride Injection, Triamterene, 1197
Nilvadipine, 924 1161, 1930 Tribulus Fruit, 1369
Nitrendipine, 927 Chloride Hydrochloride Powder, Trichlormethiazide, 1198
Nitroglycerin, 928 1162 Tablets, 1199
Norgestrel and Ethinylestradiol, Nitrate, 1162 Trichomycin, 1201
934 Thiamylal Sodium, 1163 Trichosanthes Root, 1369
Perphenazine, 977 for Injection, 1164 Triclofos Sodium, 1202
Perphenazine Maleate, 979 Thianthol, 1165 Syrup, 1203
Phenytoin, 992 Thiopental Sodium, 1166 Trihexyphenidyl Hydrochloride, 1204
Piperazine Phosphate, 1000 for Injection, 1167, 1930 Tablets, 1204
Prazepam, 1021 Thioridazine Hydrochloride, 1168 Trimebutine Maleate, 1206
Prednisolone, 1022 Thiotepa, 1168 Trimetazidine Hydrochloride, 1207,
Probenecid, 1027 L-Threonine, 1169 1931
Procainamide Hydrochloride, 1029 Thrombin, 1170 Tablets, 1207
Prochlorperazine Maleate, 1033 Thymol, 1170 Trimethadione, 1208
Propranolol Hydrochloride, 1038 Tiaramide Hydrochloride, 1171 Tablets, 1209
Propylthiouracil, 1040 Tablets, 1172 Trimetoquinol Hydrochloride Hydrate,
Reserpine, 1057 Ticlopidine Hydrochloride, 1173 1209
Ritodrine Hydrochloride, 1068 Timepidium Bromide Hydrate, 1174 Tropicamide, 1211
Rokitamycin, 1923 Timolol Maleate, 1174 L-Tryptophan, 1211
Sulfinpyrazone, 1128, 1927 Tincture Tubocurarine Chloride Hydrochloride
Sulpiride, 1132 Bitter, 1266 Hydrate, 1212, 1931
Thiamazole, 1160 Capsicum, 1269 Injection, 1213, 1931
Tiaramide Hydrochloride, 1172 Iodine, 769 Tubocurarine Hydrochloride, 1212
Tipepidine Hibenzate, 1177, 1930 Iodine, Dilute, 769 Injection, 1213
Tolbutamide, 1188 Nux Vomica, 1323 Tulobuterol Hydrochloride, 1213
Tranexamic Acid, 1193 Opium, 940 Turmeric, 1367, 1969
Trichlormethiazide, 1199 Orange Peel, 1325 Powdered, 1969
Trihexyphenidyl Hydrochloride, Tinidazole, 1175 Turpentine Oil, 1214
1204 Tipepidine Hibenzate, 1176 L-Tyrosine, 1931
Trimetazidine Hydrochloride, 1207 Tablets, 1177, 1930
Trimethadione, 1209 Titanium Oxide, 1178 U
Verapamil Hydrochloride, 1226 Tizanidine Hydrochloride, 1179
Voglibose, 1232 Toad Venom, 1368 Ubenimex, 1932
Warfarin Potassium, 1234 Tobramycin, 1180 Ubidecarenone, 1214
Zaltoprofen, 1243 Injection, 1930 Ulinastatin, 1215
Talampicillin Hydrochloride, 1138 Tocopherol, 1181 Uncaria Hook, 1370
Talc, 1139, 1928 Acetate, 1182 Urapidil, 1217
Tamsulosin Hydrochloride, 1140 Calcium Succinate, 1183 Urea, 1218
Tannic Acid, 1141 Nicotinate, 1184 Urokinase, 1219
Tartaric Acid, 1141 dl-a-Tocopherol, 1181 Ursodeoxycholic Acid, 1220
Taurine, 1142 Acetate, 1182 Uva Ursi Fluidextract, 1371, 1969
Teceleukin Nicotinate, 1184
for Injection (Genetical Recombina- Todralazine Hydrochloride Hydrate, V
tion), 1148, 1929 1185
(Genetical Recombination), 1143 Tofisopam, 1186 Vaccine
Tegafur, 1149 Tolazamide, 1187 BCG, Freeze-dried, (for Percutane-
Teicoplanin, 1150 Tolbutamide, 1188 ous Use), 335
Terbutaline Sulfate, 1153 Tablets, 1188 Cholera, 506
Testosterone Tolnaftate, 1189 Diphtheria-Purified Pertussis-Teta-
Enanthate, 1154 Solution, 1189 nus Combined, Adsorbed, 596
Enanthate Injection, 1154 Tolperisone Hydrochloride, 1190 Hepatitis B, Adsorbed, 716
Propionate, 1155 Tragacanth, 1369 Inactivated Tissue Culture Rabies,
Propionate Injection, 1155 Powdered, 1369 Freeze-dried, 1054
Tetracaine Hydrochloride, 1156 Tranexamic Acid, 1191 Influenza HA, 758
Supplement I, JP XV Index 2025

Japanese Encephalitis, 786 Vitamin B2, 1059 Ointment, 1238


Japanese Encephalitis, Freeze-dried, Phosphate Ester, 1061 Petrolatum, 981
786 Phosphate Ester Injection, 1062 Shellac, 1085
Live Attenuated Measles, Freeze-d- Powder, 1060 Soft Sugar, 1121
ried, 843 Vitamin B6, 1047 Whole Human Blood, 1238
Live Attenuated Mumps, Freeze-d- Injection, 1048 Wine, 1238
ried, 902 Vitamin B12, 548 Wood Creosote, 1870
Live Attenuated Rubella, Freeze-d- Injection, 549
ried, 1074 Vitamin C, 316 X
Live Oral Poliomyelitis, 1005 Injection, 317
Purified Pertussis, Adsorbed, 980 Powder, 317 Xylitol, 1240
Smallpox, Freeze-dried, 1091 Vitamin D2, 624 Injection, 1241, 1935
Smallpox, Freeze-dried, Prepared in Vitamin D3, 506
Cell Culture, 1091 Vitamin E, 1181 Y
Weil's Disease and Akiyami Com- Acetate, 1182
bined, 1237 Calcium Succinate, 1183 Yellow
L-Valine, 1221 Nicotinate, 1184 Beeswax, 336
Vancomycin Hydrochloride, 1221 Vitamin K1, 993 Petrolatum, 982
for Injection, 1223 Voglibose, 1231
Vasopressin Injection, 1223 Tablets, 1232 Z
Verapamil Hydrochloride, 1225
Tablets, 1226 W Zaltoprofen, 1242
Vinblastine Sulfate, 1226 Tablets, 1243
for Injection, 1227 Warfarin Potassium, 1233 Zanthoxylum Fruit, 1371
Vincristine Sulfate, 1228, 1933 Tablets, 1234 Powdered, 1371
Vitamin A Water, 1235 Zedoary, 1372, 1969
Acetate, 1058 for Injection, 1235, 1934 Zidovudine, 1935
Capsules, 1230 Purified, 1236 Zinc
Oil, 1229 Sterile Purified, 1236 Chloride, 1244
Oil Capsules, 1230 Weak Opium Alkaloids and Scopola- Oxide, 1245
Palmitate, 1059 mine Injection, 945 Oxide Oil, 1245
Vitamin B1 Weil's Disease and Akiyami Combined Oxide Ointment, 1246
Hydrochloride, 1160 Vaccine, 1237 Oxide Starch Powder, 1246
Hydrochloride Injection, 1161 Wheat Starch, 1237 Sulfate Hydrate, 1246
Hydrochloride Powder, 1162 White Sulfate Ophthalmic Solution, 1247
Nitrate, 1162 Beeswax, 336 Zinostatin Stimalamer, 1247
INDEX IN LATIN NAME
Eucommiae Cortex, 1284
A C Evodiae Fructus, 1285
Extractum Belladonnae, 1263
Achyranthis Radix, 1252 Calumbae Radix, 1267
Adeps Pulverata, 1268 F
Lanae Rurificatus, 808 Cannabis Fructus, 1298
Suillus, 808 Capsici Fructus, 1268 Fel Ursi, 1262
Agar, 1252 Pulveratus, 1269 Foeniculi Fructus, 1285
Pulveratum, 1253 Cardamomi Fructus, 1271 Pulveratus, 1285
Akebiae Caulis, 1253 Carthami Flos, 1348 Forsythiae Fructus, 1286
Alismatis Rhizoma, 1253 Caryophylli Flos, 1274 Fossilia Ossis Mastodi, 1313
Pulveratum, 1253 Pulveratus, 1274 Fritillariae Bulbus, 1287
Aloe, 1254 Cassiae Semen, 1271
Pulverata, 1255 Catalpae Fructus, 1271 G
Alpiniae Cera
Fructus, 1265 Alba, 336 Gambir, 1287
Officinari Rhizoma, 1256 Carnauba, 411 Pulveratum, 1287
Amomi Semen, 1256 Flava, 336 Gardeniae Fructus, 1288
Pulveratum, 1256 Chrysanthemi Flos, 1271 Pulveratus, 1288
Amylum Cimicifugae Rhizoma, 1272 Gastrodiae Tuber, 1289
Maydis, 542 Cinnamomi Cortex, 1272 Gentianae
Oryzae, 1346 Pulveratus, 1273 Radix, 1290
Solani, 1014 Clematidis Radix, 1274 Radix Pulverata, 1290
Tritici, 1237 Cnidii Scabrae Radix, 1306
Anemarrhenae Rhizoma, 1256 Monnieris Fructus, 1275 Scabrae Radix Pulverata, 1306
Angelicae Radix, 1305 Rhizoma, 1275 Geranii Herba, 1291
Pulverata, 1305 Rhizoma Pulveratum, 1276 Pulverata, 1291
Araliae Cordatae Rhizoma, 1937 Coicis Semen, 1276 Ginseng Radix, 1292
Arctii Fructus, 1267 Pulveratum, 1276 Pulverata, 1293
Arecae Semen, 1258 Condurango Cortex, 1276 Rubra, 1342
Armeniacae Semen, 1257 Coptidis Rhizoma, 1277 Glehniae Radix cum Rhizoma, 1294
Artemisiae Capillaris Flos, 1258 Pulveratum, 1278 Glycyrrhizae Radix, 1295
Asiasari Radix, 1259 Corni Fructus, 1279 Pulverata, 1296
Asparagi Tuber, 1259 Corydalis Tuber, 1279 Gummi Arabicum, 1251
Astragali Radix, 1260 Pulveratum, 1940 Pulveratum, 1251
Atractylodis Crataegi Fructus, 1941 Gypsum
Lanceae Rhizoma, 1260 Crocus, 1349 Exsiccatum, 1298
Lanceae Rhizoma Pulveratum, Curcumae Rhizoma, 1367 Fibrosum, 1297
1260 Purveratum, 1969
Rhizoma, 1261 Cyperi Rhizoma, 1280 H
Rhizoma Pulveratum, 1261 Pulveratum, 1280
Aurantii Houttuyniae Herba, 1302
Bobilis Pericarpium, 1273 D Hydrangeae Dulcis Folium, 1365
Fructus Immaturus, 1302 Pulveratum, 1365
Pericarpium, 1265 Digenea, 1280
Dioscoreae Rhizoma, 1282 I
B Pulveratum, 1282
Dolichi Semen, 1282 Imperatae Rhizoma, 1302
Belladonnae Radix, 1263 Ipecacuanhae Radix, 1303
Benincasae Semen, 1264 E Pulverata, 1303
Benzoinum, 1265
Bezoar Bovis, 1325 Eleutherococci senticosi Rhizoma, L
Bufonis Venenum, 1368 1283
Bupleuri Radix, 1266 Ephdrae Herba, 1283 Leonuri Herba, 1958
Epimedii Herba, 1284 Lilii Bulbus, 1958
Eriobotryae Folium, 1314 Linderae Radix, 1313

2027
2028 Index in Latin name Supplement I, JP XV

Lithospermi Radix, 1313 Scopoliae Rhizoma, 1357


Lonicerae Folium Cum Caulis, 1314 P Scutellariae Radix, 1358
Lycii Pulverata, 1359
Cortex, 1315 Paeoniae Radix, 1328 Senegae Radix, 1360
Fructus, 1315 Pulverata, 1329 Pulverata, 1361
Panacis Japonici Rhizoma, 1326 Sennae Folium, 1361
M Pulveratum, 1327 Pulveratum, 1362
Perillae Herba, 1329 Sevum Bovinum, 336
Magnoliae Persicae Semen, 1327 Sinomeni Caulis et Rhizoma, 1363
Cortex, 1315 Pulveratum, 1327 Smilacis Rhizoma, 1364
Cortex Pulveratus, 1316 Peucedani Radix, 1961 Pulveratum, 1364
Flos, 1317 Pharbitidis Semen, 1330 Sophorae Radix, 1364
Malloti Cortex, 1317 Phellodendri Cortex, 1330 Pulverata, 1364
Mel, 1301 Pulveratus, 1331 Strychni Semen, 1321
Menthae Herba, 1317 Picrasmae Lignum, 1333 Swertiae Herba, 1365
Mori Cortex, 1320 Pulveratum, 1333 Pulverata, 1366
Moutan Cortex, 1318 Pinelliae Tuber, 1333 Syrupus
Pulveratus, 1319 Plantaginis Ipecacuanha, 1304
Herba, 1334 Senegae, 1361
N Semen, 1334
Platycodi Radix, 1334 T
Nelumbis Semen, 1320 Pulverata, 1335
Notopterygii Rhizoma, 1321 Polygalae Radix, 1335 Tinctura Amara, 1266
Nupharis Rhizoma, 1321 Pulverata, 1335 Tragacantha, 1369
Polygonati Rhizoma, 1336 Pulverata, 1369
O Polygoni Multiflori Radix, 1336 Tribuli Fructus, 1369
Polyporus, 1336 Trichosanthis Radix, 1369
Oleum Pulveratus, 1337
Arachidis, 971 Poria, 1337 U
Aurantii, 946 Pulveratum, 1337
Cacao, 386 Processi Aconiti Radix, 1338 Uncariae Uncis Cum Ramulus, 1370
Camelliae, 400 Pulverata, 1339 Uvae Ursi Folium, 1262
Caryophylli, 1275 Prunellae Spica, 1341
Cinnamomi, 1273 Puerariae Radix, 1341 V
Cocois, 535
Eucalypti, 654 R Valerianae Radix, 1306
Foeniculi, 1286 Pulverata, 1307
Maydis, 542 Rehmanniae Radix, 1343
Menthae Japonicae, 1318 Resina Pini, 1347 Z
Olivae, 939 Rhei Rhizoma, 1344
Rapae, 1055 Pulveratum, 1345 Zanthoxyli Fructus, 1371
Ricini, 414 Rosae Fructus, 1346 Pulveratus, 1371
Sesami, 1084 Pulveratus, 1346 Zedoariae Rhizoma, 1372
Sojae, 1113 Zingiberis
Terebinthinae, 1214 S Processum Rhizoma, 1341
Ophiopogonis Tuber, 1324 Rhizoma, 1291
Opium Pulveratum, 939 Saposhnikoviae Radix, 1352 Rhizoma Pulveratum, 1292
Ostreae Testa, 1326 Sappan Lignum, 1352 Zizyphi
Pulverata, 1326 Saussureae Radix, 1352 Fructus, 1307
Schisandrae Fructus, 1352 Semen, 1307
Schizonepetae Spica, 1353
INDEX IN JAPANESE
アマチャ 1365 アンピシリン水和物 307
ア アマチャ末 1365 アンピシリンナトリウム 308
アマンタジン塩酸塩 293 アンベノニウム塩化物 294
亜鉛華デンプン 1246 アミカシン硫酸塩 296 アンモニア・ウイキョウ精 1286
亜鉛華軟膏 1246 アミカシン硫酸塩注射液 1830 アンモニア水 299
アカメガシワ 1317 アミドトリゾ酸 295 アンレキサノクス 1831
アクチノマイシン D 275 アミドトリゾ酸ナトリウムメグルミン アンレキサノクス錠 1833
アクラルビシン塩酸塩 272 注射液 853
アクリノール・亜鉛華軟膏 274 アミドトリゾ酸メグルミン注射液 イ
アクリノール水和物 274 851
アクリノール・チンク油 273 アミトリプチリン塩酸塩 298 イオウ 1130
アザチオプリン 325 アミトリプチリン塩酸塩錠 299, イオウ・カンフルローション 1130
アザチオプリン錠 326 1831 イオウ・サリチル酸・チアントール軟膏
亜酸化窒素 929 アミノ安息香酸エチル 644 1131
アジスロマイシン水和物 327 アミノフィリン水和物 297 イオタラム酸 774
アジマリン 278 アミノフィリン注射液 297, 1831 イオタラム酸ナトリウム注射液
アジマリン錠 279, 1826 アムホテリシン B 303 1103
亜硝酸アミル 309 アムホテリシン B 錠 305 イオタラム酸メグルミン注射液 852
アスコルビン酸 316 アムホテリシン B シロップ 305 イオトロクス酸 775
アスコルビン酸散 317 アムロジピンベシル酸塩 1834 イオパミドール 773
アスコルビン酸注射液 317, 1839 アモキサピン 301 イクタモール 744
アズトレオナム 328 アモキシシリン水和物 302 イコサペント酸エチル 646
アストロマイシン硫酸塩 322 アモスラロール塩酸塩 1835 イセパマイシン硫酸塩 777
L-アスパラギン酸 318 アモスラロール塩酸塩錠 1836 イソクスプリン塩酸塩 1894
アスピリン 318 アモバルビタール 300 イソクスプリン塩酸塩錠 1895
アスピリンアルミニウム 319 アラセプリル 279 イソソルビド 784
アスピリン錠 319 アラセプリル錠 280 イソニアジド 780
アスポキシシリン水和物 320 アラビアゴム 1251 イソニアジド錠 781
アセグルタミドアルミニウム 266 アラビアゴム末 1251 イソニアジド注射液 781
アセタゾラミド 268 アリメマジン酒石酸塩 283 イソフェンインスリン水性懸濁注射液
アセトアミノフェン 267 亜硫酸水素ナトリウム 1094 762
アセトヘキサミド 269 L-アルギニン 313 イソフルラン 778
アセブトロール塩酸塩 265 L-アルギニン塩酸塩 313 l-イソプレナリン塩酸塩 782
アセメタシン 1825 L-アルギニン塩酸塩注射液 314, イソプロパノール 783
アゼラスチン塩酸塩 1839 1839 イソプロピルアンチピリン 783
アセンヤク 1287 アルジオキサ 282 L-イソロイシン 780
アセンヤク末 1287 アルプラゾラム 284 イダルビシン塩酸塩 745
アテノロール 323 アルプレノロール塩酸塩 285 イドクスウリジン 747
アドレナリン 276 アルプロスタジル 286 イドクスウリジン点眼液 747, 1893
アドレナリン液 277 アルプロスタジルアルファデクス イトラコナゾール 1896
アドレナリン注射液 276 287 イフェンプロジル酒石酸塩 748
アトロピン硫酸塩水和物 324 アルプロスタジル注射液 1828 イブジラスト 1892
アトロピン硫酸塩注射液 324 アルベカシン硫酸塩 311 イブプロフェン 744
亜ヒ酸パスタ 315 アルベカシン硫酸塩注射液 312 イプラトロピウム臭化物水和物 776
アフロクアロン 277 アルミノプロフェン 1826 イミプラミン塩酸塩 751
アヘンアルカロイド・アトロピン注射 アルミノプロフェン錠 1827 イミプラミン塩酸塩錠 752, 1893
液 942 アロエ 1254 イミペネム水和物 749
アヘンアルカロイド塩酸塩 941 アロエ末 1255 イレイセン 1274, 1939
アヘンアルカロイド塩酸塩注射液 アロチノロール塩酸塩 314 インジゴカルミン 754
942 アロプリノール 284 インジゴカルミン注射液 755
アヘンアルカロイド・スコポラミン注 安息香酸 345 インスリン 758
射液 943 安息香酸ナトリウム 1093 インスリン亜鉛水性懸濁注射液 763
アヘン散 940 安息香酸ナトリウムカフェイン 388 インスリン注射液 762
アヘンチンキ 940 安息香酸ベンジル 347 インチンコウ 1258
アヘン・トコン散 1324 アンソッコウ 1265 インデノロール塩酸塩 753
アヘン末 939 アンチピリン 310 インドメタシン 755

2029
2030 Index in Japanese Supplement I, JP XV

インドメタシンカプセル 756, 1893 エナラプリルマレイン酸塩 1879 オキシドール 954


インドメタシン坐剤 757 エナラプリルマレイン酸塩錠 1880 オキシブプロカイン塩酸塩 951
インフルエンザ HA ワクチン 758 エノキサシン水和物 616 オキシメトロン 956
インヨウカク 1284 エピリゾール 621 オキセサゼイン 949
エピルビシン塩酸塩 622 オクスプレノロール塩酸塩 950
ウ エフェドリン塩酸塩 619 オザグレルナトリウム 1916
エフェドリン塩酸塩散 10 620 オフロキサシン 938
ウイキョウ 1285 エフェドリン塩酸塩錠 621, 1882 オメプラゾール 1915
ウイキョウ末 1285 エフェドリン塩酸塩注射液 619, オリブ油 939
ウイキョウ油 1286 1882 オルシプレナリン硫酸塩 946
ウコン 1367, 1969 エペリゾン塩酸塩 618 オレンジ油 946
ウコン末 1969 エモルファゾン 1878 オンジ 1335, 1962
ウベニメクス 1932 エリスロマイシン 627 オンジ末 1335, 1962
ウヤク 1313, 1959 エリスロマイシンエチルコハク酸エス
ウラピジル 1217 テル 629 カ
ウリナスタチン 1215 エリスロマイシンステアリン酸塩
ウルソデオキシコール酸 1220 630 カイニン酸・サントニン散 790
ウロキナーゼ 1219 エリスロマイシン腸溶錠 1882 カイニン酸水和物 789
ウワウルシ 1262 エリスロマイシンラクトビオン酸塩 カオリン 795
ウワウルシ流エキス 1371, 1969 629 カカオ脂 386
エルカトニン 612 加香ヒマシ油 415
エ エルゴカルシフェロール 624 カゴソウ 1341
エルゴタミン酒石酸塩 627 カシュウ 1336, 1962
エイジツ 1346 エルゴメトリンマレイン酸塩 625 ガジュツ 1372, 1969
エイジツ末 1346 エルゴメトリンマレイン酸塩錠 626 加水ラノリン 807
液状フェノール 986 エルゴメトリンマレイン酸塩注射液 ガスえそウマ抗毒素 696
エコチオパートヨウ化物 610 625 カッコン 1341
エスタゾラム 631 塩化亜鉛 1244 葛根湯エキス 1308, 1950
エストラジオール安息香酸エステル 塩化インジウム (111In) 注射液 755 過テクネチウム酸ナトリウム (99mTc)
631 塩化カリウム 1009 注射液 1105
エストラジオール安息香酸エステル水 塩化カルシウム水和物 390 果糖 689
性懸濁注射液 633 塩化カルシウム注射液 390, 1851 果糖注射液 689, 1888
エストラジオール安息香酸エステル注 塩化タリウム (201Tl) 注射液 1158 カナマイシン一硫酸塩 793
射液 632 塩化ナトリウム 1097 カナマイシン硫酸塩 794
エストリオール 633 10 塩化ナトリウム注射液 1098, カノコソウ 1306, 1950
エストリオール錠 635 1925 カノコソウ末 1307, 1950
エストリオール水性懸濁注射液 634 エンゴサク 1279, 1940 カフェイン水和物 387
エタクリン酸 636 エンゴサク末 1940 カプセル 403
エタクリン酸錠 636 塩酸 725 カプトプリル 403
エタノール 638 塩酸リモナーデ 727 ガベキサートメシル酸塩 693, 1889
エタンブトール塩酸塩 637 エンビオマイシン硫酸塩 616 過マンガン酸カリウム 1013
エチオナミド 643 エンフルラン 615 加味逍遙散エキス 1310, 1952
エチゾラム 652 カモスタットメシル酸塩 400, 1852
エチゾラム細粒 1883 オ b-ガラクトシダーゼ(アスペルギルス)
エチゾラム錠 1884 694
エチドロン酸二ナトリウム 649 オウギ 1260 b-ガラクトシダーゼ(ペニシリウム)
エチドロン酸二ナトリウム錠 650 オウゴン 1358, 1967 695
エチニルエストラジオール 642 オウゴン末 1359, 1967 カリジノゲナーゼ 790
エチニルエストラジオール錠 642 黄色ワセリン 982 カリ石ケン 1007
L-エチルシステイン塩酸塩 645 オウセイ 1336 カルテオロール塩酸塩 412
エチルモルヒネ塩酸塩水和物 648 オウバク 1330 カルナウバロウ 411
エチレフリン塩酸塩 650 オウバク・タンナルビン・ビスマス散 カルバゾクロムスルホン酸ナトリウム
エチレフリン塩酸塩錠 651 1332 水和物 405
エチレンジアミン 648 オウバク末 1331 カルバマゼピン 404
エデト酸ナトリウム水和物 598 オウレン 1277, 1940 カルビドパ水和物 406
エーテル 641 オウレン末 1278, 1940 L-カルボシステイン 407
エテンザミド 640 オキサゾラム 948 カルメロース 408
エトスクシミド 644 オキサピウムヨウ化物 947 カルメロースカルシウム 409
エトドラク 653 オキサプロジン 948 カルメロースナトリウム 410
エトポシド 653 オキシコドン塩酸塩水和物 951 カルモナムナトリウム 412
エドロホニウム塩化物 611 オキシテトラサイクリン塩酸塩 956 カルモフール 411
エドロホニウム塩化物注射液 612, オキシトシン 958 カロコン 1369
1878 オキシトシン注射液 960 カンキョウ 1341, 1964
Supplement I, JP XV Index in Japanese 2031

乾燥亜硫酸ナトリウム 1109 クロモグリク酸ナトリウム 1100


乾燥甲状腺 1171 ク クロラゼプ酸二カリウム 1868
乾燥酵母 1241 クロラゼプ酸二カリウムカプセル
乾燥細胞培養痘そうワクチン 1091 グアイフェネシン 714 1869
乾燥ジフテリアウマ抗毒素 596 グアナベンズ酢酸塩 715 クロラムフェニコール 487
乾燥弱毒生おたふくかぜワクチン グアネチジン硫酸塩 716 クロラムフェニコールコハク酸エステ
902 グアヤコールスルホン酸カリウム ルナトリウム 489
乾燥弱毒生風しんワクチン 1074 1011 クロラムフェニコールパルミチン酸エ
乾燥弱毒生麻しんワクチン 843 クエン酸ガリウム (67Ga) 注射液 696 ステル 488
乾燥水酸化アルミニウムゲル 288 クエン酸水和物 515 クロルジアゼポキシド 490
乾燥水酸化アルミニウムゲル細粒 クエン酸ナトリウム水和物 1099 クロルジアゼポキシド散 491
289 クコシ 1315 クロルジアゼポキシド錠 492, 1858
乾燥組織培養不活化狂犬病ワクチン クジン 1364, 1968 クロルフェニラミン・カルシウム散
1054 クジン末 1364, 1969 497
乾燥炭酸ナトリウム 1096 苦味重曹水 1364 クロルフェニラミンマレイン酸塩
乾燥痘そうワクチン 1091 苦味チンキ 1266 498
乾燥日本脳炎ワクチン 786 クラブラン酸カリウム 1010 d-クロルフェニラミンマレイン酸塩
乾燥破傷風ウマ抗毒素 1156 グラミシジン 712 501
乾燥はぶウマ抗毒素 716 クラリスロマイシン 516 クロルフェニラミンマレイン酸塩散
乾燥 BCG ワクチン 335 クラリスロマイシン錠 517 499
乾燥ボツリヌスウマ抗毒素 374 グリシン 704 クロルフェニラミンマレイン酸塩錠
乾燥まむしウマ抗毒素 841 グリセオフルビン 713 500
乾燥硫酸アルミニウムカリウム 292 グリセオフルビン錠 1890 クロルフェニラミンマレイン酸塩注射
カンゾウ 1295 グリセリン 702 液 499
カンゾウエキス 1296, 1943 グリセリンカリ液 704 クロルフェネシンカルバミン酸エステ
カンゾウ粗エキス 1297, 1943 クリノフィブラート 522 ル 496, 1858
カンゾウ末 1296 グリベンクラミド 699 クロルフェネシンカルバミン酸エステ
カンテン 1252 クリンダマイシン塩酸塩 519 ル錠 1859
カンテン末 1253 クリンダマイシン塩酸塩カプセル クロルプロパミド 504
含糖ペプシン 1074 520 クロルプロパミド錠 505, 1861
d-カンフル 401 クリンダマイシンリン酸エステル クロルプロマジン塩酸塩 502
dl-カンフル 402 521 クロルプロマジン塩酸塩錠 503,
肝油 538 クリンダマイシンリン酸エステル注射 1861
カンレノ酸カリウム 1008 液 1866 クロルプロマジン塩酸塩注射液 503,
グルコン酸カルシウム水和物 391 1860
キ グルタチオン 701 クロルヘキシジン塩酸塩 493
L-グルタミン 1889 クロルヘキシジングルコン酸塩液
希塩酸 726 クレオソート 544 493
キキョウ 1334 クレゾール 544 クロルマジノン酢酸エステル 494
キキョウ末 1335 クレゾール水 545 クロロブタノール 495
キキョウ流エキス 1334, 1962 クレゾール石ケン液 545
キクカ 1271 クレマスチンフマル酸塩 519 ケ
キササゲ 1271 クロカプラミン塩酸塩水和物 523
キジツ 1302 クロキサシリンナトリウム水和物 ケイガイ 1353
キシリトール 1240 533 経口生ポリオワクチン 1005
キシリトール注射液 1241, 1935 クロキサゾラム 534 ケイ酸マグネシウム 836
キタサマイシン 798 クロコナゾール塩酸塩 546 軽質無水ケイ酸 1087
キタサマイシン酢酸エステル 799 クロスカルメロースナトリウム 546 軽質流動パラフィン 967
キタサマイシン酒石酸塩 800 クロチアゼパム 531 桂枝茯苓丸エキス 1955
キニジン硫酸塩水和物 1050 クロトリマゾール 532 ケイヒ 1272
キニーネエチル炭酸エステル 1051 クロナゼパム 528 ケイヒ末 1273
キニーネ塩酸塩水和物 1052 クロニジン塩酸塩 529 ケイヒ油 1273
キニーネ硫酸塩水和物 1053 クロフィブラート 525 ケタミン塩酸塩 795
牛脂 336 クロフィブラートカプセル 526 結晶性インスリン亜鉛水性懸濁注射液
吸水軟膏 265 クロフェダノール塩酸塩 524 765
キョウカツ 1321, 1960 クロベタゾールプロピオン酸エステル 結晶セルロース 477
キョウニン 1257, 1937 1867 血清性性腺刺激ホルモン 710
キョウニン水 1257 クロペラスチン塩酸塩 530 ケツメイシ 1271
希ヨードチンキ 769 クロミフェンクエン酸塩 526 ケトチフェンフマル酸塩 797
金チオリンゴ酸ナトリウム 1092 クロミフェンクエン酸塩錠 527 ケトプロフェン 796
クロミプラミン塩酸塩 528 ケノデオキシコール酸 486
クロム酸ナトリウム (51Cr) 注射液 ケンゴシ 1330
1099 ゲンタマイシン硫酸塩 698
2032 Index in Japanese Supplement I, JP XV

ゲンチアナ 1290, 1943 ザルトプロフェン 1242 ジスルフィラム 600


ゲンチアナ・重曹散 1290 ザルトプロフェン錠 1243 ジソピラミド 598
ゲンチアナ末 1290, 1943 サルブタモール硫酸塩 1078 シソマイシン硫酸塩 1090
ゲンノショウコ 1291 酸化亜鉛 1245 シタラビン 552
ゲンノショウコ末 1291 酸化カルシウム 393 シッカニン 1086
酸化チタン 1178 シツリシ 1369
コ 酸化マグネシウム 835 ジドブジン 1935
サンキライ 1364, 1968 ジドロゲステロン 609
コウカ 1348 サンキライ末 1364, 1968 ジドロゲステロン錠 610
硬化油 735 サンザシ 1941 ジノスタチン スチマラマー 1247
コウジン 1342 三酸化ヒ素 316 ジノプロスト 592
合成ケイ酸アルミニウム 290 サンシシ 1288 ジヒドロエルゴタミンメシル酸塩
コウブシ 1280, 1942 サンシシ末 1288 583
コウブシ末 1280, 1942 サンシュユ 1279 ジヒドロエルゴトキシンメシル酸塩
コウボク 1315, 1959 サンショウ 1371 584
コウボク末 1316, 1959 サンショウ末 1371 ジヒドロコデインリン酸塩 581
ゴオウ 1325 酸素 955 ジヒドロコデインリン酸塩散 1
コカイン塩酸塩 535 サンソウニン 1307 582
ゴシツ 1252 サントニン 1081 ジヒドロコデインリン酸塩散 10
ゴシュユ 1285 サンヤク 1282, 1942 582
コデインリン酸塩散 1 536 サンヤク末 1282, 1942 ジピリダモール 597
コデインリン酸塩散 10 537 ジフェニドール塩酸塩 575
コデインリン酸塩錠 537 シ ジフェンヒドラミン 593
コデインリン酸塩水和物 536 ジフェンヒドラミン塩酸塩 594
ゴナドレリン酢酸塩 705 ジアスターゼ 567 ジフェンヒドラミン・バレリル尿素散
ゴボウシ 1267 ジアスターゼ・重曹散 567 593
ゴマ油 1084 ジアゼパム 567 ジフェンヒドラミン・フェノール・亜鉛
ゴミシ 1352 シアナミド 548 華リニメント 595
コムギデンプン 1237 シアノコバラミン 548, 1870 ジブカイン塩酸塩 569
コメデンプン 1346 シアノコバラミン注射液 549, 1871 ジフテリアトキソイド 596
コリスチンメタンスルホン酸ナトリウ ジエチルカルバマジンクエン酸塩 ジフテリア破傷風混合トキソイド
ム 540 574 596
コリスチン硫酸塩 541 ジエチルカルバマジンクエン酸塩錠 シプロヘプタジン塩酸塩水和物 551
コルチゾン酢酸エステル 543 574 ジベカシン硫酸塩 569
コルヒチン 538 ジオウ 1343, 1964 シベンゾリンコハク酸塩 1861
コレカルシフェロール 506 歯科用アンチホルミン 310 シベンゾリンコハク酸塩錠 1862
コレステロール 507 歯科用トリオジンクパスタ 1210 シメチジン 513
コレラワクチン 506 歯科用パラホルムパスタ 969 ジメモルファンリン酸塩 588
コロンボ 1267, 1939 歯科用フェノール・カンフル 987 ジメルカプロール 590
コロンボ末 1268, 1939 歯科用ヨード・グリセリン 771 ジメルカプロール注射液 590
コンズランゴ 1276 ジギトキシン 576 ジメンヒドリナート 588
コンズランゴ流エキス 1277, 1940 ジギトキシン錠 576 ジメンヒドリナート錠 589
シクラシリン 507 次没食子酸ビスマス 368
サ ジクロキサシリンナトリウム水和物 ジモルホラミン 591
573 ジモルホラミン注射液 591
サイクロセリン 551 シクロスポリン 508 弱アヘンアルカロイド・スコポラミン
サイコ 1266 ジクロフェナクナトリウム 570 注射液 945
サイシン 1259, 1938 ジクロフェナミド 571 シャクヤク 1328
柴苓湯エキス 1349 ジクロフェナミド錠 572 シャクヤク末 1329
酢酸 268 シクロペントラート塩酸塩 549 ジャショウシ 1275
酢酸ナトリウム水和物 1091 シクロホスファミド水和物 550 シャゼンシ 1334
サッカリン 1075 シゴカ 1283 シャゼンソウ 1334
サッカリンナトリウム水和物 1076 ジゴキシン 578 臭化カリウム 1008
サフラン 1349 ジゴキシン錠 580 臭化ナトリウム 1095
サラシ粉 494 ジゴキシン注射液 579 ジュウヤク 1302
サラシミツロウ 336 ジコッピ 1315, 1959 シュクシャ 1256
サラゾスルファピリジン 1077 シコン 1313, 1959 シュクシャ末 1256
サリチル酸 1079, 1924 次硝酸ビスマス 369 酒石酸 1141
サリチル酸精 1080 ジスチグミン臭化物 599 ショウキョウ 1291
サリチル酸ナトリウム 1108 ジスチグミン臭化物錠 600 ショウキョウ末 1292
サリチル酸絆創膏 1080 L-システイン 1871 硝酸イソソルビド 785
サリチル酸メチル 881 L-システイン塩酸塩水和物 1872 硝酸イソソルビド錠 786
サリチル・ミョウバン散 1078 シスプラチン 513 硝酸銀 1088
Supplement I, JP XV Index in Japanese 2033

硝酸銀点眼液 1088 スルフイソキサゾール 1128 セフタジジム水和物 466


常水 1235 スルフィンピラゾン 1127, 1927 セフチゾキシムナトリウム 471
ショウズク 1271 スルフィンピラゾン錠 1128, 1927 セフチブテン水和物 470
焼セッコウ 1298 スルベニシリンナトリウム 1123 セフテラム ピボキシル 468
消毒用エタノール 640 スルホブロモフタレインナトリウム セフテラム ピボキシル細粒 469
消毒用フェノール 986 1129 セフトリアキソンナトリウム水和物
消毒用フェノール水 987 スルホブロモフタレインナトリウム注 472
ショウマ 1272, 1939 射液 1130 セフピラミドナトリウム 458
ジョサマイシン 786 セフピロム硫酸塩 460
ジョサマイシン錠 1897 セ セフブペラゾンナトリウム 429
ジョサマイシンプロピオン酸エステル セフポドキシム プロキセチル 461
788 成人用沈降ジフテリアトキソイド セフミノクスナトリウム水和物 446
シラザプリル錠 1864 596 セフメタゾールナトリウム 445
シラザプリル水和物 1863 精製水 1236 セフメノキシム塩酸塩 443
シラスタチンナトリウム 509 精製ゼラチン 697 セフロキサジン水和物 462
ジラゼプ塩酸塩水和物 586 精製セラック 1085 セフロキシム アキセチル 474
ジルチアゼム塩酸塩 586 精製デヒドロコール酸 556 セフロキシムナトリウム 476
シロスタゾール 511 精製白糖 1120 セラセフェート 480
シロスタゾール錠 512, 1866 精製ラノリン 808 ゼラチン 696
シロップ用セファドロキシル 1853 生理食塩液 1098, 1893 セラペプターゼ 1083
シロップ用ファロペネムナトリウム 石油ベンジン 982 L-セリン 1925
659, 1886 セタノール 484 セルモロイキン(遺伝子組換え) 481
シンイ 1317 セチリジン塩酸塩 1856 センキュウ 1275, 1939
親水軟膏 735 セチリジン塩酸塩錠 1857 センキュウ末 1276, 1940
親水ワセリン 981 セッコウ 1297 ゼンコ 1961
診断用クエン酸ナトリウム液 1100 セトラキサート塩酸塩 485 センコツ 1321, 1960
セネガ 1360, 1968 センソ 1368
ス セネガシロップ 1361 センナ 1361
セネガ末 1361, 1968 センナ末 1362
水酸化カリウム 1012 セファクロル 415 センブリ 1365
水酸化カルシウム 392 セファクロルカプセル 416 センブリ・重曹散 1367
水酸化ナトリウム 1101 セファクロル細粒 417 センブリ末 1366
スキサメトニウム塩化物水和物 セファクロル複合顆粒 419
1137 セファゾリンナトリウム 426 ソ
スキサメトニウム塩化物注射液 セファゾリンナトリウム水和物 428
1138, 1928 セファトリジンプロピレングリコール ソウジュツ 1260
スクラルファート水和物 1118 425, 1854 ソウジュツ末 1260
スコポラミン臭化水素酸塩水和物 セファドロキシル 421 ソウハクヒ 1320, 1960
1083 セファドロキシルカプセル 1852 ソボク 1352
ステアリルアルコール 1117 セファピリンナトリウム 424 ソヨウ 1329, 1961
ステアリン酸 1116 セファレキシン 422 ソルビタンセスキオレイン酸エステル
ステアリン酸カルシウム 400 セファロチンナトリウム 423, 1854 1111
ステアリン酸ポリオキシル 40 1007 セフィキシム 442 D-ソルビトール 1112
ステアリン酸マグネシウム 837 セフェピム塩酸塩水和物 439 D-ソルビトール液 1112
ストレプトマイシン硫酸塩 1117 セフォジジムナトリウム 447
スピラマイシン酢酸エステル 1114 セフォゾプラン塩酸塩 457 タ
スピロノラクトン 1116 セフォタキシムナトリウム 449
スペクチノマイシン塩酸塩水和物 セフォチアム塩酸塩 455 ダイオウ 1344
1114 セフォチアム ヘキセチル塩酸塩 453 大黄甘草湯エキス 1280
スルタミシリントシル酸塩水和物 セフォテタン 451 ダイオウ末 1345
1134, 1927 セフォペラゾンナトリウム 448 ダイズ油 1113
スルチアム 1136 セフカペン ピボキシル塩酸塩細粒 タイソウ 1307
スルバクタムナトリウム 1122, 1927 432 ダウノルビシン塩酸塩 553
スルピリド 1131 セフカペン ピボキシル塩酸塩錠 433 タウリン 1142
スルピリドカプセル 1132 セフカペン ピボキシル塩酸塩水和物 タクシャ 1253
スルピリド錠 1132 430 タクシャ末 1253
スルピリン水和物 1133 セフジトレン ピボキシル 437 タムスロシン塩酸塩 1140
スルピリン注射液 1134, 1927 セフジトレン ピボキシル細粒 438 タランピシリン塩酸塩 1138
スルファジアジン銀 1124 セフジトレン ピボキシル錠 438 タルク 1139, 1928
スルファメチゾール 1125 セフジニル 434 炭酸カリウム 1009
スルファメトキサゾール 1126 セフジニルカプセル 435 炭酸水素ナトリウム 1093
スルファモノメトキシン水和物 セフジニル細粒 436 炭酸水素ナトリウム注射液 1094,
1126 セフスロジンナトリウム 464 1925
2034 Index in Japanese Supplement I, JP XV

炭酸ナトリウム水和物 1096 注射用セフォチアム塩酸塩 456 テガフール 1149


炭酸マグネシウム 834 注射用セフタジジム 1856 デキサメタゾン 560
炭酸リチウム 826 注射用セフメタゾールナトリウム デキストラン 40 561, 1873
単シロップ 1089 1855 デキストラン 40 注射液 562
ダントロレンナトリウム水和物 553 注射用チアミラールナトリウム デキストラン 70 563
単軟膏 1089 1164 デキストラン硫酸エステルナトリウム
タンニン酸 1141 注射用チオペンタールナトリウム イオウ 5 564
タンニン酸アルブミン 282 1167, 1930 デキストラン硫酸ナトリウム イオウ
タンニン酸ジフェンヒドラミン 595 注射用テセロイキン(遺伝子組換え) 18 565
タンニン酸ベルベリン 352 1148, 1929 デキストリン 565
注射用ドキソルビシン塩酸塩 1877 デキストロメトルファン臭化水素酸塩
チ 注射用バンコマイシン塩酸塩 1223 水和物 566
注射用ヒト絨毛性性腺刺激ホルモン テストステロンエナント酸エステル
チアマゾール 1159 708 1154
チアマゾール錠 1160 注射用ヒドララジン塩酸塩 724 テストステロンエナント酸エステル注
チアミラールナトリウム 1163 注射用ピペラシリンナトリウム 998 射液 1154
チアミン塩化物塩酸塩 1160 注射用ビンブラスチン硫酸塩 1227 テストステロンプロピオン酸エステル
チアミン塩化物塩酸塩散 1162 注射用ファモチジン 656, 1885 1155
チアミン塩化物塩酸塩注射液 1161, 注射用フェニトインナトリウム 992 テストステロンプロピオン酸エステル
1930 注射用プレドニゾロンコハク酸エステ 注射液 1155
チアミン硝化物 1162 ルナトリウム 1025, 1921 デスラノシド 559
チアラミド塩酸塩 1171 注射用フロモキセフナトリウム 666 デスラノシド注射液 560, 1873
チアラミド塩酸塩錠 1172 注射用ペプロマイシン硫酸塩 1918 テセロイキン(遺伝子組換え) 1143
チアントール 1165 注射用ベンジルペニシリンカリウム テトラカイン塩酸塩 1156
チオテパ 1168 1841 テトラサイクリン塩酸塩 1157
チオペンタールナトリウム 1166 注射用ホスホマイシンナトリウム デヒドロコール酸 556
チオリダジン塩酸塩 1168 687 デヒドロコール酸注射液 557, 1873
チオ硫酸ナトリウム水和物 1109 注射用マイトマイシンC 1905 デフェロキサミンメシル酸塩 554,
チオ硫酸ナトリウム注射液 1110, 注射用ミノサイクリン塩酸塩 1904 1873
1927 チョウジ 1274 デメチルクロルテトラサイクリン塩酸
チクセツニンジン 1326, 1960 チョウジ末 1274 塩 558
チクセツニンジン末 1327, 1960 チョウジ油 1275 テルブタリン硫酸塩 1153
チクロピジン塩酸塩 1173 チョウトウコウ 1370 テレビン油 1214
チザニジン塩酸塩 1179 チョレイ 1336, 1962 天然ケイ酸アルミニウム 289
窒素 928 チョレイ末 1337, 1963 デンプングリコール酸ナトリウム
チニダゾール 1175 L-チロジン 1931 1926
チペピジンヒベンズ酸塩 1176 チンク油 1245 テンマ 1289, 1943
チペピジンヒベンズ酸塩錠 1177, 沈降ジフテリア破傷風混合トキソイド テンモンドウ 1259, 1938
1930 596
チメピジウム臭化物水和物 1174 沈降精製百日せきジフテリア破傷風混 ト
チモ 1256, 1937 合ワクチン 596
チモール 1170 沈降精製百日せきワクチン 980 トウガシ 1264
チモロールマレイン酸塩 1174 沈降炭酸カルシウム 389 トウガラシ 1268
注射用アズトレオナム 1840 沈降破傷風トキソイド 1156 トウガラシ・サリチル酸精 1270
注射用アセチルコリン塩化物 271, 沈降はぶトキソイド 716 トウガラシチンキ 1269
1825 沈降 B 型肝炎ワクチン 716 トウガラシ末 1269
注射用アムホテリシン B 304 チンピ 1273 トウキ 1305
注射用アモバルビタールナトリウム トウキ末 1305
300 ツ トウニン 1327, 1961
注射用アンピシリンナトリウム トウニン末 1327, 1961
1838 ツバキ油 400 トウヒ 1265
注射用イダルビシン塩酸塩 746 ツボクラリン塩化物塩酸塩水和物 トウヒシロップ 1325
注射用イミペネム・シラスタチンナト 1212, 1931 トウヒチンキ 1325
リウム 750 ツボクラリン塩化物塩酸塩注射液 トウモロコシデンプン 542
注射用オザグレルナトリウム 1917 1213, 1931 トウモロコシ油 542
注射用血清性性腺刺激ホルモン 711 ツロブテロール塩酸塩 1213 ドキサプラム塩酸塩水和物 603
注射用水 1235, 1934 ドキシサイクリン塩酸塩水和物 606
注射用スキサメトニウム塩化物 テ ドキシフルリジン 604
1137, 1928 ドキシフルリジンカプセル 604
注射用セファゾリンナトリウム テイコプラニン 1150 ドキソルビシン塩酸塩 605
1854 低置換度ヒドロキシプロピルセルロー ドクカツ 1937
注射用セフェピム塩酸塩 441 ス 738 トコフェロール 1181
注射用セフォゾプラン塩酸塩 458 テオフィリン 1158 トコフェロールコハク酸エステルカル
Supplement I, JP XV Index in Japanese 2035

シウム 1183 906 オール錠 934


トコフェロール酢酸エステル 1182 ナファゾリン硝酸塩 908 ノルトリプチリン塩酸塩 935
トコフェロールニコチン酸エステル ナファモスタットメシル酸塩 1911 ノルフロキサシン 932
1184 ナブメトン 1908
トコン 1303, 1949 ナブメトン錠 1910 ハ
トコンシロップ 1304 ナプロキセン 908
トコン末 1303, 1949 ナリジクス酸 905 バイモ 1287, 1942
トチュウ 1284 ナロキソン塩酸塩 906 バカンピシリン塩酸塩 329
トドララジン塩酸塩水和物 1185 白色セラック 1085
ドパミン塩酸塩 602 ニ 白色軟膏 1238
ドパミン塩酸塩注射液 602, 1877 白色ワセリン 981
トフィソパム 1186 ニガキ 1333 白糖 1121
ドブタミン塩酸塩 601 ニガキ末 1333 バクモンドウ 1324
トブラマイシン 1180 ニカルジピン塩酸塩 911 バクロフェン 331
トブラマイシン注射液 1930 ニカルジピン塩酸塩注射液 912, バクロフェン錠 332, 1841
トラガント 1369 1912 バシトラシン 330
トラガント末 1369 ニコチン酸 920 バソプレシン注射液 1223
トラザミド 1187 ニコチン酸アミド 919 ハチミツ 1301
トラネキサム酸 1191 ニコチン酸注射液 921, 1912 ハッカ 1317
トラネキサム酸カプセル 1192 ニコモール 917 ハッカ水 1318
トラネキサム酸錠 1193 ニコモール錠 918 ハッカ油 1318
トラネキサム酸注射液 1193 ニコランジル 919, 1912 パップ用複方オウバク散 1332
トラピジル 1194 ニザチジン 1912 パニペネム 962
トリアムシノロン 1195 ニザチジンカプセル 1913 パパベリン塩酸塩 965
トリアムシノロンアセトニド 1196 二酸化炭素 407 パパベリン塩酸塩注射液 965, 1918
トリアムテレン 1197 ニセリトロール 916 ハマボウフウ 1294, 1943
トリクロホスナトリウム 1202 ニセルゴリン 913 バメタン硫酸塩 333
トリクロホスナトリウムシロップ ニセルゴリン散 914 パラアミノサリチル酸カルシウム顆粒
1203 ニセルゴリン錠 915 394
トリクロルメチアジド 1198 ニトラゼパム 925 パラアミノサリチル酸カルシウム水和
トリクロルメチアジド錠 1199 ニトレンジピン 926 物 395
トリコマイシン 1201 ニトレンジピン錠 927 パラオキシ安息香酸エチル 647
L-トリプトファン 1211 ニトログリセリン錠 928 パラオキシ安息香酸ブチル 386
トリヘキシフェニジル塩酸塩 1204 ニフェジピン 922 パラオキシ安息香酸プロピル 1039
トリヘキシフェニジル塩酸塩錠 日本脳炎ワクチン 786 パラオキシ安息香酸メチル 878
1204 乳酸 801 パラフィン 966
トリメタジオン 1208 乳酸カルシウム水和物 393 パラホルムアルデヒド 968
トリメタジオン錠 1209 乳糖水和物 803 L-バリン 1221
トリメタジジン塩酸塩 1207, 1931 尿素 1218 パルナパリンナトリウム 969
トリメタジジン塩酸塩錠 1207 ニルバジピン 923 バルビタール 333
トリメトキノール塩酸塩水和物 ニルバジピン錠 924 バルプロ酸ナトリウム 1110
1209 ニンジン 1292 バレイショデンプン 1014
トリメブチンマレイン酸塩 1206 ニンジン末 1293 ハロキサゾラム 719
トルナフタート 1189 ニンドウ 1314 ハロタン 719
トルナフタート液 1189 ハロペリドール 717
トルブタミド 1188 ネ ハロペリドール錠 718
トルブタミド錠 1188 パンクレアチン 961
トルペリゾン塩酸塩 1190 ネオスチグミンメチル硫酸塩 909 パンクロニウム臭化物 962
L-トレオニン 1169 ネオスチグミンメチル硫酸塩注射液 ハンゲ 1333
トレピブトン 1195 910, 1912 半夏厚朴湯エキス 1943
トロピカミド 1211 ネチルマイシン硫酸塩 910 バンコマイシン塩酸塩 1221
ドロペリドール 608 パンテチン 964
トロンビン 1170 ノ パントテン酸カルシウム 394
豚脂 808
ドンペリドン 1876 濃グリセリン 703 ヒ
濃ベンザルコニウム塩化物液 50 343
ナ ノスカピン 936 ビオチン 1842
ノスカピン塩酸塩水和物 937 ピコスルファートナトリウム水和物
ナイスタチン 937 ノルアドレナリン 930 1105
ナタネ油 1055 ノルアドレナリン注射液 931, 1915 ビサコジル 367
ナドロール 904 ノルエチステロン 932 ビサコジル坐剤 368, 1843
ナファゾリン塩酸塩 907 ノルゲストレル 933 ビソプロロールフマル酸塩 1843
ナファゾリン・クロルフェニラミン液 ノルゲストレル・エチニルエストラジ ビソプロロールフマル酸塩錠 1844
2036 Index in Japanese Supplement I, JP XV

ビタミン A 油 1229 ピンドロール 995 ブフェキサマク軟膏 381


ビタミン A 油カプセル 1230 ビンブラスチン硫酸塩 1226 ブフェトロール塩酸塩 379
ヒトインスリン(遺伝子組換え) 760 ビンロウジ 1258 ブプラノロール塩酸塩 383
ヒト下垂体性性腺刺激ホルモン 708 ブプレノルフィン塩酸塩 1850
ヒト絨毛性性腺刺激ホルモン 707 フ ブホルミン塩酸塩 1846
人全血液 1238 ブホルミン塩酸塩錠 1849
人免疫グロブリン 724 ファモチジン 655 ブホルミン塩酸塩腸溶錠 1847
ヒドララジン塩酸塩 724 ファモチジン散 657 ブメタニド 382
ヒドララジン塩酸塩散 725 ファモチジン錠 657 フラジオマイシン硫酸塩 688
ヒドララジン塩酸塩錠 725, 1890 ファロペネムナトリウム錠 660, プラステロン硫酸エステルナトリウム
ヒドロキシジン塩酸塩 739 1886 水和物 1107
ヒドロキシジンパモ酸塩 739 ファロペネムナトリウム水和物 658, プラゼパム 1020
ヒドロキシプロピルセルロース 736 1885 プラゼパム錠 1021
ヒドロキソコバラミン酢酸塩 736 フィトナジオン 993 プラノプロフェン 1018
ヒドロクロロチアジド 727 フェニトイン 991 プラバスタチンナトリウム 1018
ヒドロコタルニン塩酸塩水和物 734 フェニトイン散 992 フラビンアデニンジヌクレオチドナト
ヒドロコルチゾン 728 フェニトイン錠 992 リウム 662
ヒドロコルチゾンコハク酸エステル L-フェニルアラニン 989 フラボキサート塩酸塩 664
733 フェニルブタゾン 990 プリミドン 1026
ヒドロコルチゾンコハク酸エステルナ フェニレフリン塩酸塩 990 フルオキシメステロン 675
トリウム 732 フェネチシリンカリウム 983 フルオシノニド 672
ヒドロコルチゾン酢酸エステル 729 フェノバルビタール 984 フルオシノロンアセトニド 670
ヒドロコルチゾン・ジフェンヒドラミ フェノバルビタール散 10 985 フルオレセインナトリウム 673
ン軟膏 730 フェノール 985 フルオロウラシル 674
ヒドロコルチゾン酪酸エステル 730 フェノール・亜鉛華リニメント 987 フルオロメトロン 673
ヒドロコルチゾンリン酸エステルナト フェノール水 987 フルジアゼパム 669
リウム 731 フェノールスルホンフタレイン 988 フルシトシン 668
ピブメシリナム塩酸塩 1004 フェノールスルホンフタレイン注射液 フルスルチアミン塩酸塩 692
ヒプロメロース 741 988 フルニトラゼパム 670
ヒプロメロースフタル酸エステル フェルビナク 1887 フルフェナジンエナント酸エステル
743, 1891 フェンタニルクエン酸塩 661 676
ピペミド酸水和物 996 フェンブフェン 660 フルラゼパム 677
ピペラシリン水和物 1919 複方アクリノール・チンク油 273 フルラゼパム塩酸塩 678
ピペラシリンナトリウム 997 複方オキシコドン・アトロピン注射液 フルラゼパムカプセル 678
ピペラジンアジピン酸塩 999 953 プルラン 1045
ピペラジンリン酸塩錠 1000 複方オキシコドン注射液 952 フルルビプロフェン 679
ピペラジンリン酸塩水和物 999 複方サリチル酸精 1080 ブレオマイシン塩酸塩 370
ビペリデン塩酸塩 366 複方サリチル酸メチル精 879 ブレオマイシン硫酸塩 372
ビホナゾール 366 複方ジアスターゼ・重曹散 567 プレドニゾロン 1021
ヒマシ油 414 複方ダイオウ・センナ散 1346 プレドニゾロンコハク酸エステル
ピマリシン 994 複方チアントール・サリチル酸液 1024
ヒメクロモン 740 1165 プレドニゾロン酢酸エステル 1023
ビャクゴウ 1958 複方ビタミン B 散 1230 プレドニゾロン錠 1022
ビャクシ 1257, 1937 複方ヨード・グリセリン 770 プロカインアミド塩酸塩 1028
ビャクジュツ 1261, 1938 複方ロートエキス・ジアスターゼ散 プロカインアミド塩酸塩錠 1029
ビャクジュツ末 1261, 1939 1355 プロカインアミド塩酸注射液 1029
氷酢酸 269 ブクモロール塩酸塩 378 プロカイン塩酸塩 1030
ピラジナミド 1046 ブクリョウ 1337 プロカイン塩酸塩注射液 1030
ピラルビシン 1000 ブクリョウ末 1337 プロカテロール塩酸水和物 1032
ピランテルパモ酸塩 1045 ブシ 1338, 1963 プロカルバジン塩酸塩 1031
ピリドキシン塩酸塩 1047 フシジン酸ナトリウム 1101 プログルミド 1035
ピリドキシン塩酸塩注射液 1048, ブシ末 1339, 1963 プロクロルペラジンマレイン酸塩
1922 ブシラミン 377 1033
ピリドスチグミン臭化物 1047 ブシラミン錠 1845 プロクロルペラジンマレイン酸塩錠
ピレノキシン 1001 ブスルファン 384 1033
ピレンゼピン塩酸塩水和物 1002 ブチルスコポラミン臭化物 1082 プロゲステロン 1034
ピロ亜硫酸ナトリウム 1108 ブドウ酒 1238 プロゲステロン注射液 1034
ピロカルピン塩酸塩 994 ブドウ糖 700 フロセミド 690
ピロキシカム 1003 ブドウ糖注射液 701, 1889 フロセミド錠 691
ピロキシリン 1049 ブトロピウム臭化物 385 プロタミンインスリン亜鉛水性懸濁注
ピロールニトリン 1049 ブナゾシン塩酸塩 383 射液 766
ビワヨウ 1314 ブフェキサマク 380 プロタミン硫酸塩 1041, 1921
ビンクリスチン硫酸塩 1228, 1933 ブフェキサマククリーム 380 プロタミン硫酸塩注射液 1042, 1922
Supplement I, JP XV Index in Japanese 2037

プロチオナミド 1042 ベンジルペニシリンカリウム 349 マクロゴール 4000 832


プロチレリン 1043 ベンジルペニシリンベンザチン水和物 マクロゴール 6000 832
プロチレリン酒石酸塩水和物 1044 348 マクロゴール 20000 833
プロテイン銀 1088 ヘンズ 1282 マクロゴール軟膏 833
プロテイン銀液 1089 ベンズブロマロン 343 マシニン 1298
プロパンテリン臭化物 1036 ベンゼトニウム塩化物 344 麻酔用エーテル 641
プロピルチオウラシル 1040 ベンゼトニウム塩化物液 345 マニジピン塩酸塩 1900
プロピルチオウラシル錠 1040 ベンセラジド塩酸塩 340 マニジピン塩酸塩錠 1902
プロピレングリコール 1039 ペンタゾシン 972 マプロチリン塩酸塩 842
プロプラノロール塩酸塩 1037 ペントキシベリンクエン酸塩 974 マルトース水和物 840
プロプラノロール塩酸塩錠 1038 ベントナイト 341 D-マンニトール 841
フロプロピオン 667 ペントバルビタールカルシウム 973 D-マンニトール注射液 842, 1903
フロプロピオンカプセル 667 ペンブトロール硫酸塩 972
プロベネシド 1027 ミ
プロベネシド錠 1027 ホ
ブロマゼパム 374 ミグレニン 896
ブロムヘキシン塩酸塩 375 ボウイ 1363 ミクロノマイシン硫酸塩 893
プロメタジン塩酸塩 1036 ボウコン 1302, 1949 ミコナゾール 892
フロモキセフナトリウム 664 ホウ酸 374 ミコナゾール硝酸塩 893
ブロモクリプチンメシル酸塩 376 ホウ砂 1095 ミゾリビン 1906
ブロモバレリル尿素 377 抱水クロラール 487 ミゾリビン錠 1907
粉末セルロース 480 ボウフウ 1352, 1966 ミツロウ 336
ボグリボース 1231 ミデカマイシン 894
ヘ ボグリボース錠 1232 ミデカマイシン酢酸エステル 895
ホスフェストロール 683, 1888 ミノサイクリン塩酸塩 897
ベカナマイシン硫酸塩 337 ホスフェストロール錠 684, 1888 ミョウバン水 293
ベクロメタゾンプロピオン酸エステル ホスホマイシンカルシウム水和物
335 685 ム
ベザフィブラート 364 ホスホマイシンナトリウム 686
ベザフィブラート徐放錠 365 ボタンピ 1318 無晶性インスリン亜鉛水性懸濁注射液
ベタネコール塩化物 363 ボタンピ末 1319 764
ベタヒスチンメシル酸塩 353, 1842 補中益気湯エキス 1298, 1945 無水アンピシリン 306
ベタヒスチンメシル酸塩錠 354 ポビドン 1015 無水エタノール 639
ベタメタゾン 355 ポビドンヨード 1017 無水カフェイン 386
ベタメタゾン吉草酸エステル 360 ホマトロピン臭化水素酸塩 722 無水クエン酸 514
ベタメタゾン吉草酸エステル・ゲンタ ホミカ 1321 無水乳糖 802, 1900
マイシン硫酸塩クリーム 361 ホミカエキス 1322, 1960 無水リン酸水素カルシウム 396,
ベタメタゾン吉草酸エステル・ゲンタ ホミカエキス散 1323 1874
マイシン硫酸塩軟膏 362 ホミカチンキ 1323 ムピロシンカルシウム 水和物 902
ベタメタゾンジプロピオン酸エステル ホモクロルシクリジン塩酸塩 723
358 ポリスチレンスルホン酸カルシウム メ
ベタメタゾン錠 356 398
ベタメタゾンリン酸エステルナトリウ ポリスチレンスルホン酸ナトリウム メキシレチン塩酸塩 891
ム 359 1106 メキタジン 861
ペチジン塩酸塩 980 ポリソルベート 80 1007 メグルミン 850
ペチジン塩酸塩注射液 981, 1918 ホリナートカルシウム 391, 1851 メクロフェノキサート塩酸塩 843
ベニジピン塩酸塩 338 ポリミキシン B 硫酸塩 1006 メコバラミン 844
ベニジピン塩酸塩錠 339 ホルマリン 681 メシル酸塩ガベキサート 693
ヘパリンナトリウム 721 ホルマリン水 682 メストラノール 864
ヘパリンナトリウム注射液 722 ホルモテロールフマル酸塩水和物 メダゼパム 845, 1903
ペプロマイシン硫酸塩 975 682 メタンフェタミン塩酸塩 868
ベラドンナエキス 1263, 1939 ボレイ 1326 L-メチオニン 869
ベラドンナコン 1263 ボレイ末 1326 メチクラン 884
ベラパミル塩酸塩 1225 メチラポン 890
ベラパミル塩酸塩錠 1226 マ dl-メチルエフェドリン塩酸塩 875
ペルフェナジン 977 dl-メチルエフェドリン塩酸塩散 10
ペルフェナジン錠 977 マイトマイシン C 898 876
ペルフェナジンマレイン酸塩 978 マオウ 1283 メチルエルゴメトリンマレイン酸塩
ペルフェナジンマレイン酸塩錠 979 マーキュロクロム 862 876
ベルベリン塩化物水和物 351 マーキュロクロム液 863 メチルエルゴメトリンマレイン酸塩錠
ベンザルコニウム塩化物 342 マクリ 1280 877
ベンザルコニウム塩化物液 342 マクロゴール 400 830 メチルジゴキシン 885
ベンジルアルコール 346 マクロゴール 1500 831 メチルセルロース 871
2038 Index in Japanese Supplement I, JP XV

メチルテストステロン 882 輸血用クエン酸ナトリウム注射液 ム 1061


メチルテストステロン錠 883 1099, 1926 リボフラビンリン酸エステルナトリウ
メチルドパ錠 874, 1903 ユビデカレノン 1214 ム注射液 1062, 1922
メチルドパ水和物 873 リマプロスト アルファデクス 819
メチルプレドニゾロン 879 ヨ リュウコツ 1313
メチルプレドニゾロンコハク酸エステ 硫酸亜鉛水和物 1246
ル 880 ヨウ化カリウム 1013 硫酸亜鉛点眼液 1247
メチルベナクチジウム臭化物 871 ヨウ化ナトリウム 1102 硫酸アルミニウムカリウム水和物
メチルロザニリン塩化物 881 ヨウ化ナトリウム (123I) カプセル 292
滅菌精製水 1236 1103 硫酸カリウム 1014
メテノロンエナント酸エステル 866 ヨウ化ナトリウム (131I) 液 1103 硫酸鉄水和物 661
メテノロンエナント酸エステル注射液 ヨウ化ナトリウム (131I) カプセル 硫酸バリウム 334
866 1103 硫酸マグネシウム水 839
メテノロン酢酸エステル 865 ヨウ化人血清アルブミン (131I) 注射液 硫酸マグネシウム水和物 838
メトキサレン 870 768 硫酸マグネシウム注射液 839, 1900
メトクロプラミド 886 ヨウ化ヒプル酸ナトリウム (131I) 注射 リュウタン 1306, 1949
メトクロプラミド錠 886 液 1103 リュウタン末 1306, 1949
メトトレキサート 869 葉酸 680 流動パラフィン 966
メトプロロール酒石酸塩 887 葉酸錠 681, 1888 リョウキョウ 1256, 1937
メトプロロール酒石酸塩錠 888 葉酸注射液 681, 1888 苓桂朮甘湯エキス 1347, 1965
メトホルミン塩酸塩 867 ヨウ素 768 リンゲル液 1066, 1922
メトホルミン塩酸塩錠 867 ヨクイニン 1276 リンコマイシン塩酸塩水和物 820
メトロニダゾール 889 ヨクイニン末 1276 リン酸水素カルシウム水和物 396,
メトロニダゾール錠 889 ヨーダミド 767 1875
メナテトレノン 855 ヨーダミドナトリウムメグルミン注射 リン酸水素ナトリウム水和物 568
メピチオスタン 858 液 854 リン酸二水素カルシウム水和物 397
メピバカイン塩酸塩 859 ヨード・サリチル酸・フェノール精
メピバカイン塩酸塩注射液 860 772 レ
メフェナム酸 847 ヨードチンキ 769
メフルシド 849 ヨードホルム 773 レセルピン 1055
メフルシド錠 849, 1903 レセルピン散 0.1 1057
メフロキン塩酸塩 848 ラ レセルピン錠 1057
メペンゾラート臭化物 857 レセルピン注射液 1056, 1922
メルカプトプリン水和物 861 ラウリル硫酸ナトリウム 1104 レチノール酢酸エステル 1058
メルファラン 855 ラウロマクロゴール 810 レチノールパルミチン酸エステル
メロペネム 水和物 863 ラクツロース 804 1059
dl-メントール 856 ラタモキセフナトリウム 809 レナンピシリン塩酸塩 810
l-メントール 857 ラッカセイ油 971 レバロルファン酒石酸塩 813
ラナトシド C 805 レバロルファン酒石酸塩注射液 814,
モ ラナトシド C 錠 806 1900
ラニチジン塩酸塩 1054 レボチロキシンナトリウム錠 817
木クレオソート 1870 ラベタロール塩酸塩 1898 レボチロキシンナトリウム水和物
モクツウ 1253 ラベタロール塩酸塩錠 1899 816
モッコウ 1352, 1966 レボドパ 814
モノステアリン酸アルミニウム 291 リ レボメプロマジンマレイン酸塩 815
モノステアリン酸グリセリン 704 レンギョウ 1286
モルヒネ・アトロピン注射液 899 リオチロニンナトリウム 821 レンニク 1320
モルヒネ塩酸塩錠 902, 1908 リオチロニンナトリウム錠 822
モルヒネ塩酸塩水和物 900 リシノプリル錠 824 ロ
モルヒネ塩酸塩注射液 901 リシノプリル水和物 823
L-リジン塩酸塩 829 L-ロイシン 812
ヤ リゾチーム塩酸塩 830 ロキサチジン酢酸エステル塩酸塩
リドカイン 818 1070
ヤクチ 1265 リドカイン注射液 818 ロキサチジン酢酸エステル塩酸塩徐放
ヤクモソウ 1958 リトドリン塩酸塩 1067 カプセル 1071
薬用石ケン 846 リトドリン塩酸塩錠 1068 ロキシスロマイシン 1073, 1923
薬用炭 845 リファンピシン 1064 ロキソプロフェンナトリウム水和物
ヤシ油 535 リファンピシンカプセル 1065 828
リボスタマイシン硫酸塩 1063 ロキタマイシン 1069
ユ リボフラビン 1059 ロキタマイシン錠 1923
リボフラビン散 1060 ロジン 1347
ユウタン 1262 リボフラビン酪酸エステル 1060 ロートエキス 1353, 1967
ユーカリ油 654 リボフラビンリン酸エステルナトリウ ロートエキス・アネスタミン散 1355
Supplement I, JP XV Index in Japanese 2039

ロートエキス・カーボン散 1355 ン散 1356


ロートエキス散 1353 ロートコン 1357, 1967 ワ
ロートエキス・タンニン坐剤 1357 ロラゼパム 827
ロートエキス・パパベリン・アネスタミ ワイル病秋やみ混合ワクチン 1237
ワルファリンカリウム 1233
ワルファリンカリウム錠 1234

You might also like