You are on page 1of 254

SUPPLEMENT I

TO
THE JAPANESE
PHARMACOPOEIA
SEVENTEENTH EDITION
O‹cial from December 1, 2017

English Version

THE MINISTRY OF HEALTH, LABOUR AND WELFARE


Notice: This English Version of the Japanese Pharmacopoeia is published
for the convenience of users unfamiliar with the Japanese language. When
and if any discrepancy arises between the Japanese original and its English
translation, the former is authentic.

Printed in Japan
The Ministry of Health, Labour and
Welfare Ministerial Notification No. 348

Pursuant to Paragraph 1, Article 41 of the Law on Securing Quality, Efficacy and


Safety of Products including Pharmaceuticals and Medical Devices (Law No. 145,
1960), we hereby revise a part of the Japanese Pharmacopoeia (Ministerial Notifica-
tion No. 64, 2016) as follows*. However, in the case of drugs which are listed in the
Pharmacopoeia (hereinafter referred to as ``previous Pharmacopoeia'') [limited to
those listed in the Japanese Pharmacopoeia whose standards are changed in accor-
dance with this notification (hereinafter referred to as ``new Pharmacopoeia'')] and
drugs which have been approved as of December 1, 2017 as prescribed under Para-
graph 1, Article 14 of the same law [including drugs the Minister of Health, Labour
and Welfare specifies (the Ministry of Health and Welfare Ministerial Notification
No. 104, 1994) as of November 30, 2017 as those exempted from marketing approval
pursuant to Paragraph 1, Article 14 of the same law (hereinafter referred to as ``drugs
exempted from approval'')], the Name and Standards established 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 established in the new Phar-
macopoeia before and on May 31, 2019. 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 December 1, 2017 as prescribed under the
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 Pharmacopoeia before and
on May 31, 2019.

Katsunobu Kato
The Minister of Health, Labour and Welfare

December 1, 2017

(The text referred to by the term ``as follows'' are omitted here. All of them are made
available for public exhibition at the Pharmaceutical Evaluation Division, Phar-
maceutical Safety and Environmental Health 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 content of Supplement I to the Japanese Pharmacopoeia
Seventeenth Edition from General Notice to Ultraviolet-visible Reference Spectra (pp. 2631 – 2811).
CONTENTS
Preface ...................................................... i General Information
Supplement I to The Japanese Pharmacopoeia, G2 Solid-state Properties
Seventeenth Edition ......................... 2631–2811 Laser Diffraction Measurement of Par-
General Rules for Preparations ............... 2631 ticle Size....................................... 2813
General Tests, Processes and Apparatus ... 2633 Powder Fineness ............................... 2813
2.24 Ultraviolet-visible Spectrophotometry Powder Flow.................................... 2813
................................................. 2633 Solid and Particle Densities ................. 2814
2.46 Residual Solvents .......................... 2634 G3 Biotechnological/Biological Products
3.06 Laser Diffraction Measurement of Amino Acid Analysis ......................... 2814
Particle Size ................................. 2641 Enzyme-linked Immunosorbent Assay
4.03 Digestion Test .............................. 2645 (ELISA) ....................................... 2814
6.02 Uniformity of Dosage Units ............ 2645 G5 Crude Drugs
6.04 Test for Acid-neutralizing Capacity Purity Tests on Crude Drugs using
of Gastrointestinal Medicines........... 2648 Genetic Information ....................... 2818
6.14 Uniformity of Delivered Dose for On the Scientific Names of Crude Drugs
Inhalations .................................. 2648 listed in the JP .............................. 2819
6.15 Aerodynamic Particle Size Measure- G6 Drug Formulation
ment for Inhalations ...................... 2651 Aerodynamic Particle Size Measure-
9.01 Reference Standards ...................... 2661 ment for Inhalations by Glass Impin-
9.21 Standard Solutions for Volumetric gers ............................................. 2820
Analysis ...................................... 2661 G7 Containers and Package
9.41 Reagents, Test Solutions................. 2662 Glass Containers for Pharmaceutical
9.42 Solid Supports/Column Packings for Products ...................................... 2822
Chromatography........................... 2670 Moisture Permeability Test for Blister
Packaging of Solid Preparations ....... 2824
O‹cial Monographs ................................ 2671 G10 Others
Crude Drugs and Related Drugs.............. 2747 Basic Concepts for Quality Assurance
of Drug Substances and Drug
Infrared Reference Spectra ................ 2801–2805 Products ...................................... 2826
Criteria for Content Uniformity in Real
Ultraviolet-visible Reference Spectra .... 2807–2811 Time Release Testing by Process
Analytical Technology..................... 2826
International Harmonization Imple-
mented in the Japanese Pharma-
copoeia Seventeenth Edition ............. 2829
Stability Testing of Drug Substances
and Drug Products ......................... 2835

Index.................................................... 2841
Index in Latin Name................................ 2861
Index in Japanese.................................... 2863
PREFACE
The 17th Edition of the Japanese Pharmacopoeia It was also agreed that JP articles should cover
(JP) was promulgated by Ministerial Notification drugs, which are important from the viewpoint of
No.64 of the Ministry of Health, Labour and Welfare health care and medical treatment, clinical perfor-
(MHLW) on March 7, 2016. mance or merits and frequency of use, as soon as pos-
In July 2016, the Committee on JP established the sible after they reach the market.
basic principles for the preparation of the JP 18th Edi- The target date for the publication of JP 18th Edi-
tion, setting out the roles and characteristics of the JP, tion (the Japanese edition) was set as April 2021.
the definite measures for the revision, and the date of JP drafts are discussed in the following committees
the revision. that were established in the Pharmaceuticals and
At the Committee, the five basic principles of JP, Medical Devices Agency: Expert Committee; Sub-
which we refer to as the ``five pillars'', were estab- expert Committee; Sub-committee on Manufacturing
lished as follows: 1) Including all drugs which are im- Process-related Matters; Committee on Chemicals;
portant from the viewpoint of health care and medical Committee on Antibiotics; Committee on Biologicals;
treatment; 2) Making qualitative improvement by in- Committee on Crude Drugs; Committee on Pharma-
troducing the latest science and technology; 3) Further ceutical Excipients; Committee on Physico-Chemical
promoting internationalization in response to globali- Methods; Committee on Drug Formulation; Commit-
zation of drug market; 4) Making prompt partial tee on Physical Methods; Committee on Biological
revision as necessary and facilitating smooth adminis- Methods; Committee on Nomenclature for Pharma-
trative operation; and 5) Ensuring transparency re- ceuticals; Committee on International Harmoniza-
garding the revision, and disseminating the JP to the tion; and Committee on Reference Standards. Fur-
public. It was agreed that the Committee on JP should thermore, working groups are established under the
make efforts, on the basis of these principles, to Committee on Pharmaceutical Excipients, Committee
ensure that the JP is used more effectively in the fields on Physico-Chemical Methods and Committee on
of health care and medical treatment by taking ap- Drug Formulation.
propriate measurements, including getting the under- In the Committee on JP, Mitsuru Hashida took the
standing and cooperation of other parties concerned. role of chairman from January 2011 to November
It was agreed that the JP should provide an official 2017.
standard, being required to assure the quality of medi- In addition to the regular revision every five years in
cines in Japan in response to the progress of science line with the basic principles for the preparation of the
and technology and medical demands at the time. It JP it was agreed that partial revision should be done as
should define the standards for specifications, as well necessary to take account of recent progress of science
as the methods of testing to assure overall quality of and in the interests of international harmonization.
all drugs in principle, and it should have a role in In accordance with the above principles, the com-
clarifying the criteria for quality assurance of drugs mittees initiated deliberations on selection of articles
that are recognized to be essential for public health and on revisions for General Notices, General Rules
and medical treatment. for Crude Drugs, General Rules for Preparations,
The JP has been prepared with the aid of the General Tests, Monographs and so on.
knowledge and experience of many professionals in Draft revisions covering subjects in General No-
the pharmaceutical field. Therefore, the JP should tices, General Rules for Crude Drugs, General Rules
have the characteristics of an official standard, which for Preparations, General Tests and Monographs, for
might be widely used by all parties concerned, and it which discussions were finished between August 2015
should play an appropriate role of providing informa- and March 2017, were prepared for a supplement to
tion and understanding about the quality of drugs to the JP 17. They were examined by the Committee on
the public. Moreover, as a pharmaceutical quality JP in April 2017, followed by the Pharmaceutical Af-
standard, it should contribute promoting and main- fairs and Food Sanitation Council (PAFSC) in June
taining of advancedness as well as international con- 2017, and then submitted to the Minister of Health,
sistency and harmonization of technical requirements Labour and Welfare.
in the international community. Numbers of discussions in the committees to pre-
i
ii Preface Supplement I, JP XVII

pare the supplement drafts were as follows: Expert 3. The following items in each monograph are put
Committee (8); Sub-committee on Manufacturing in the order shown below, except that unnecessary i-
Process-related Matters (9), Committee on Chemicals tems are omitted depending on the nature of the drug:
(20), Committee on Antibiotics (5); Committee on (1) English title
Biologicals (8); Committee on Crude Drugs (17); (2) Commonly used name(s)
Committee on Pharmaceutical Excipients (10); Com- (3) Latin title (only for crude drugs)
mittee on Physico-Chemical Methods (14, including a (4) Title in Japanese
working group); Committee on Drug Formulation (5) Structural formula or empirical formula
(27, including working groups); Committee on Physi- (6) Molecular formula and molecular mass
cal Methods (8); Committee on Biological Methods (7) Chemical name
(6); Committee on Nomenclature for Pharmaceuticals (8) Chemical Abstracts Service (CAS) Registry
(7); Committee on International Harmonization (6); Number
and Committee on Reference Standards (4). (9) Origin
It should be noted that in the preparation of the (10) Limits of the content of the ingredient(s) and/or
drafts for the supplement, generous cooperation was the unit of potency
given by the Pharmaceutical Technology Committee (11) Labeling requirements
of the Osaka Pharmaceutical Manufacturers Associa- (12) Method of preparation
tion, the Pharmacopeia and CMC Committee of the (13) Manufacture
Pharmaceutical Manufacturer's Association of (14) Description
Tokyo, the Tokyo Crude Drugs Association, the In- (15) Identification tests
ternational Pharmaceutical Excipients Council Japan, (16) Specific physical and/or chemical values
the Home Medicine Association of Japan, the Japan (17) Purity tests
Kampo Medicines Manufacturers Association, the (18) Potential adulteration
Japan Flavor and Fragrance Materials Association, (19) Loss on drying or Ignition, or Water
the Japan Medical Plants Federation, the Japan Phar- (20) Residue on ignition, Total ash or Acid-insoluble
maceutical Manufacturers Association, the Federation ash
of Pharmaceutical Manufacturers' Association of (21) Tests being required for pharmaceutical prepa-
Japan, the Parenteral Drug Association Japan Chap- rations
ter, the Japan Reagent Association, the Japan Oil- (22) Other special tests
seeds Processors Association, the Japan Analytical (23) Assay
Instruments Manufacturers' Association, and the Asi- (24) Containers and storage
an Society of Innovative Packaging Technology. (25) Shelf life
In consequence of this revision, the JP 17th Edition (26) Others
carries 1977 articles, owing to the addition of 32 arti-
cles and the deletion of 17 article. 4. In each monograph, the following physical and
chemical values representing the properties and quali-
The principles of description and the salient points ty of the drug are given in the order indicated below,
of the revision in this volume are as follows: except that unnecessary items are omitted depending
1. The Supplement I to JP 17th Edition comprises on the nature of drug:
the following items, in order: Notification of MHLW; (1) Alcohol number
Contents; Preface; General Rules for Preparations; (2) Absorbance
General Tests, Processes and Apparatus; Official (3) Congealing point
Monographs; then followed by Infrared Reference (4) Refractive index
Spectra and Ultraviolet-visible Reference Spectra; (5) Osmolar ratio
General Information; and as an appendix a Cumula- (6) Optical rotation
tive Index containing references to the main volume (7) Constituent amino acids
and the Supplement I. (8) Viscosity
(9) pH
2. The articles in Official Monographs, Infrared (10) Content ratio of the active ingredients
Reference Spectra and Ultraviolet-visible Reference (11) Specific gravity
Spectra are respectively placed in alphabetical order in (12) Boiling point
principle. (13) Melting point
(14) Acid value
Supplement I, JP XVII Preface iii

(15) Saponification value (31) Lead


(16) Ester value (32) Silver
(17) Hydroxyl value (33) Alkaline earth metals
(18) Iodine value (34) Arsenic
(35) Free phosphoric acid
5. Identification tests comprise the following i- (36) Foreign matters
tems, which are generally put in the order given below: (37) Related substances
(1) Coloration reactions (38) Isomer
(2) Precipitation reactions (39) Optical isomer
(3) Decomposition reactions (40) Polymer
(4) Derivatives (41) Residual solvent
(5) Infrared and/or ultraviolet-visible absorption (42) Other impurities
spectrometry (43) Residue on evaporation
(6) Nuclear magnetic resonance spectrometry (44) Readily carbonizable substances
(7) Chromatography
(8) Special reactions 7. The following paragraphs were newly added to
(9) Cations the General Rules for Preparations:
(10) Anions (1) The following items were newly added to the
[3] Monographs for Preparations according to
6. Purity tests comprise the following items, which increase of commercial products:
are generally put in the order given below, except that 1.8. Films for Oral Administration
unnecessary items are omitted depending on the na- 1.8.1 Orally Disintegrating Films
ture of drug:
(1) Color 8. The General Rules for Preparations was revised
(2) Odor as follows in general:
(3) Clarity and/or color of solution (1) [1] General Notices for Preparations (9):
(4) Acidity or alkalinity Microbial Limit Test for Crude Drugs and
(5) Acidity Preparations containing Crude Drugs as Main
(6) Alkalinity Ingredient <5.02> was added as a test to be ap-
(7) Chloride plied as necessary for non-sterile preparations
(8) Sulfate to prevent contamination.
(9) Sulfite (2) [3] Monographs for Preparations: ``5-1-1. Dry
(10) Nitrate Powder Inhalers'' and ``5-1-3. Metered-dose
(11) Nitrite Inhalers'': the requirements for general tests of
(12) Carbonate Uniformity of Delivered Dose for Inhalations
(13) Bromide <6.14> and Aerodynamic Particle Size Meas-
(14) Iodide urement for Inhalations <6.15> were pre-
(15) Soluble halide scribed.
(16) Thiocyanate (3) [3] Monographs for Preparations, ``9-1. Sup-
(17) Selenium positories for Rectal Application'' and ``10-2.
(18) Cationic salts Suppositories for Vaginal Use'': suppositories
(19) Ammonium for rectal application and vaginal use using
(20) Heavy metals lipophilic bases were revised to be able to apply
(21) Iron the evaluation of meltability instead of active
(22) Manganese substance release tests to assure appropriate
(23) Chromium drug release.
(24) Bismuth (4) [3] Monographs for Preparations, ``11-2. Liq-
(25) Tin uids and Solutions for Cutaneous Applica-
(26) Aluminum tion'': the specification that Transdermal Sys-
(27) Zinc tems in single-dose packages among Liquids
(28) Cadmium and Solutions for Cutaneous Application is re-
(29) Mercury quired to meet the requirements of Uniformity
(30) Copper of Dosage Units <6.02> was added.
iv Preface Supplement I, JP XVII

9. The following items were newly added to Diclofenamide RS


General Tests, Processes and Apparatus: Digitoxin RS
(1) 3.06 Laser Diffraction Measurement of Par- Fluoxymesterone RS
ticle Size Gramicidin RS
(2) 6.14 Uniformity of Delivered Dose for Inha- Lanatoside C RS
lations Rokitamycin RS
(3) 6.15 Aerodynamic Particle Size Measure- Tolazamide RS
ment for Inhalations Zinostatin Stimalamer RS

10. The following items in General Tests, Proc- 14. The following substances were newly added to
esses and Apparatus were revised: the Official Monographs:
(1) 2.24 Ultraviolet-visible Spectrophotometry Azosemide
(2) 2.46 Residual Solvents Azosemide Tablets
(3) 4.03 Digestion Test Calcium Levofolinate Hydrate
(4) 6.02 Uniformity of Dosage Units Cefoperazone Sodium for Injection
(5) 6.04 Test for Acid-neutralizing Capacity of Chloramphenicol and Colistin Sodium Methanesul-
Gastrointestinal Medicines fonate Ophthalmic Solution
(6) 9.01 Reference Standards Clomipramine Hydrochloride Tablets
(7) 9.21 Standard Solutions for Volumetric Clotiazepam Tablets
Analysis Entacapone
(8) 9.41 Reagents, Test Solutions Entacapone Tablets
(9) 9.42 Solid Supports/Column Packings for Glucose Hydrate
Chromatography Purified Glucose
Isophane Insulin Human (Genetical Recombina-
11. The following Reference Standards were new- tion) Injectable Aqueous Suspension
ly added: Biphasic Isophane Insulin Human (Genetical
Entacapone RS Recombination) Injectable Aqueous Suspension
Entacapone Related Substance A RS for System Insulin Aspart (Genetical Recombination)
Suitability Irbesartan Tablets
Glucose RS Irbesartan and Amlodipine Besilate Tablets
Insulin Aspart RS Magnesium Aluminosilicate
Pazufloxacin Mesilate RS Magnesium Aluminometasilicate
Pyridoxal Phosphate RS Mesalazine
Saccharin Sodium RS for Identification Mesalazine Extended-release Tablets
Zonisamide RS Methotrexate Tablets
Montelukast Sodium Granules
12. The following Reference Standards were Pazufloxacin Mesilate
revised the name: Pazufloxacin Mesilate Injection
Adrenaline Bitartrate RS for Purity Pentobarbital Calcium Tablets
p-Aminobenzoyl Glutamic Acid RS for Purity Pyridoxal Phosphate Hydrate
Anhydrous Lactose RS for Identification Roxithromycin Tablets
Cellacefate RS for Identification Tramadol Hydrochloride
Gitoxin RS for Purity Voriconazole for Injection
Heparin Sodium RS for Identification Zonisamide
Lactose RS for Identification Zonisamide Tablets
Over-sulfated Chondroitin Sulfate RS for System Goreisan Extract
Suitability
Povidone RS for Identification 15. The following monographs were revised:
Tyrosine RS for Digestion Test Amoxicillin Hydrate
Ampicillin Hydrate
13. The following Reference Standards were delet- Bacitracin
ed from the list of 9.01 Reference Standards: Benzylpenicillin Potassium
Aceglutamide RS Cefixime Hydrate
Supplement I, JP XVII Preface v

Ceftizoxime Sodium Verapamil Hydrochloride Tablets


Cellacefate Vinblastine Sulfate
Chloramphenicol Sodium Succinate Vincristine Sulfate
Clarithromycin Powdered Alisma Tuber
Cloxacillin Sodium Hydrate Artemisia Capillaris Flower
Colistin Sodium Methanesulfonate Bakumondoto Extract
Demethylchlortetracycline Hydrochloride Bofutsushosan Extract
Dextran 40 Boiogito Extract
Digoxin Chotosan Extract
Doxorubicin Hydrochloride Cornus Fruit
Doxycycline Hydrochloride Hydrate Curcuma Rhizome
Edaravone Injection Cyperus Rhizome
Epalrestat Powdered Cyperus Rhizome
Erythromycin Daiokanzoto Extract
Ethanol Daisaikoto Extract
Anhydrous Ethanol Euodia Fruit
Folic Acid Gardenia Fruit
Fosfomycin Calcium Hydrate Glycyrrhiza Extract
Fosfomycin Sodium Crude Glycyrrhiza Extract
Gentamicin Sulfate Goshajinkigan Extract
Glucose Injection Hachimijiogan Extract
Heparin Calcium Hangekobokuto Extract
Heparin Sodium Hangeshashinto Extract
Hydrocortisone Acetate Hochuekkito Extract
Hydrocortisone Butyrate Jujube
Hydroxocobalamin Acetate Jujube Seed
Hydroxypropylcellulose Juzentaihoto Extract
Low Substituted Hydroxypropylcellulose Kakkonto Extract
Hypromellose Kakkontokasenkyushin'i Extract
Iohexol Injection Kamikihito Extract
Isosorbide Mononitrate 70z/Lactose 30z Kamishoyosan Extract
Anhydrous Lactose Keishibukuryogan Extract
Lactose Hydrate Maoto Extract
D-Mannitol Mukoi-Daikenchuto Extract
Methylcellulose Orengedokuto Extract
Noradrenaline Oriental Bezoar
Oxytetracycline Hydrochloride Otsujito Extract
Pentobarbital Calcium Platycodon Root
Polymixin B Sulfate Rape Seed Oil
Polysorbate 80 Rikkunshito Extract
Povidone Ryokeijutsukanto Extract
Saccharin Sodium Hydrate Saibokuto Extract
Sodium Lauryl Sulfate Saikokeishito Extract
Spiramycin Acetate Saireito Extract
Sulbactam Sodium Saposhnikovia Root and Rhizome
Sultamicillin Tosilate Hydrate Shakuyakukanzoto Extract
Suxamethonium Chloride Injection Shimbuto Extract
Teicoplanin Shosaikoto Extract
Tetracycline Hydrochloride Shoseiryuto Extract
Thrombin Powdered Sweet Hydrangea Leaf
Tobramycin Tokakujokito Extract
Vasopressin Injection Tokishakuyakusan Extract
Verapamil Hydrochloride Trichosanthes Root
vi Preface Supplement I, JP XVII

Turmeric Pazufloxacin Mesilate


Powdered Turmeric Pyridoxal Phosphate Hydrate
Yokukansan Extract Tramadol Hydrochloride
Zonisamide
16. The following monographs were deleted:
Aceglutamide Aluminum 20. The following articles in Infrared Reference
Diclofenamide Spectra were deleted:
Diclofenamide Tablets Diclofenamide
Digitoxin Fluoxymesterone
Digitoxin Tablets Rokitamycin
Fluoxymesterone Saccharin Sodium Hydrate
Gramicidin Tolazamide
Lanatoside C
Lanatoside C Tablets Those who were engaged in the preparation of the
Mercurochrome Supplement I to JP 17 are as follows:
Mercurochrome Solution Youhei Aita Koji Ichinose
Rokitamycin Kenichi Akao Nobukazu Igoshi
Rokitamycin Tablets Teruyo Arato Yoshiaki Ikarashi
Serrapeptase Keiko Arimoto Shingo Ikedo
Tolazamide Naoki Aruga Kazuhiko Ikegami
Zinostatin Stimalamer Hiroshi Asama Masato Ishida
Scopolia Extract, Papaverine and Ethyl Aminoben- Kazuhide Ashizawa Akiko Ishii
zoate Powder Shinichiro Aso Shigeru Itai
Yukio Aso Michiho Ito
17. The following articles were newly added to Yosuke Demizu Ryoichi Ito
Ultraviolet-visible Reference Spectra: Makoto Emura Yuji Ito
Azosemide Hiroyuki Fuchino Yusuke Izutani
Calcium Levofolinate Hydrate Makiko Fujii Kenichi Izutsu
Doxycycline Hydrochloride Hydrate Yuzo Fujimoto Makoto Kanebako
Entacapone Shinji Fukuda Hirohito Katayama
Mesalazine Kiyoshi Fukuhara Kumiko Kato
Pazufloxacin Mesilate Harumi Furuichi Noriko Katori
Pyridoxal Phosphate Hydrate Hiromitsu Furukawa Nobuo Kawahara
Sultamicillin Tosilate Hydrate Yukihiro Goda Tomomi Kawamata
Tramadol Hydrochloride Takashi Goto Toru Kawanishi**
Zonisamide Tamami Goto Yoshihiko Kawarasaki
Yuji Haishima Nana Kawasaki
18. The following articles in Ultraviolet-visible Hideki Hakamata Keiji Kijima
Reference Spectra were deleted: Takashi Hakamatsuka Yutaka Kikuchi
Diclofenamide Kentaro Hanada Yuuichi Kikuchi
Fluoxymesterone Ruri Hanajiri Yasuhiro Kishimoto
Gramicidin Akira Harazono Mitsukazu Kitada
Rokitamycin Mitsuru Hashida* Akihito Kitajima
Tolazamide Noritaka Hashii Atsushi Kittaka
Rika Hatano Fumiyuki Kiuchi
19. The following articles were newly added to In- Ai Hayashi Takashi Kobayashi
frared Reference Spectra: Yoshinori Hayashi Rika Kochi
Azosemide Taro Higano Tatsuo Koide
Calcium Levofolinate Hydrate Yasuhiko Higuchi Takashi Kojima
Entacapone Kenichi Hirai Hiroyasu Kokubo
Low Substituted Hydroxypropylcellulose Yukio Hiyama Katsuko Komatsu
Mesalazine Masatoshi Hori Toshifumi Konda
Oxytetracycline Hydrochloride Masashi Hyuga Seizo Kondo
Supplement I, JP XVII Preface vii

Atsutaka Kubosaki Satoshi Sasaki Takumi Watanabe Hitoshi Yamauchi


Kiyoshi Kubota Yuko Sasaki Rumiko Yamada Takeshi Yamazaki
Kenichi Kumasaka Kazumichi Sasaoki Tetsuji Yamaguchi Masato Yasuhara
Masaaki Kurihara Koji Sato Hiroki Yamaji Shiho Yasuo
Hideki Kusunoki Kyoko Sato Keiji Yamamoto Chikako Yomota
Toshinobu Makiura Norihisa Sato Tosuke Yamamoto Etsuo Yonemochi
Takuro Maruyama Fujio Sekigawa Chikamasa Yamashita Hiroyuki Yoshida
Sayaka Masada Michiko Sekiguchi
Kazuhiro Matsumoto Hiroko Shibata *: Chairman, the Committee on JP
Makoto Matsumoto Keigo Shibayama **: Acting Chairman, the Committee on JP
Hajime Matsumura Shizuka Shida
Eiichi Mikami Yasuo Shimada
Takao Mitsuhashi Masaro Shimizu
Naoki Miyata Katsuaki Shinohara
Tamaki Miyazaki Osamu Shirota
Takeshi Mizuno Takuji Shoda
Mitsuo Mori Hirotaka Sudo
Kunikazu Moribe Keiichi Sudo
Seiki Morimoto Naoki Sugimoto
Takashi Morimoto Mikio Suzuki
Takahito Morisaki Noriyuki Suzuki
Osamu Morita Shigeo Suzuki
Toshimi Murai Tomoyuki Suzuki
Masashi Muroi Katsumi Tachibana
Nobuyuki Naka Minoru Tada
Shinsaku Nakagawa Shinichi Tadaki
Yukari Nakagawa Nobuo Taguchi
Toru Nakai Yoshikazu Takahashi
Emi Nakajima Akihito Takano
Tatsuya Nakano Masaki Takao
Tatsumi Nakashima Osami Takeda
Mitsuo Nanaura Tomoko Takeda
Masao Nasu Hirohumi Takeuchi
Yutaka Nishihara Hisashi Takeuchi
Noriaki Nishikawa Toyoshige Tanabe
Sumiaki Oba Masakazu Tanaka
Toru Ogawa Satoshi Tanaka
Yasumitsu Ogura Setsuko Tanaka
Yasutaka Ohgami Tomohide Tanaka
Tsuneo Okubo Tsuyoshi Tanimoto
Akihiro Okuda Susumu Terabayashi
Haruhiro Okuda Katsuhide Terada
Makoto Ono Reiko Teraoka
Hiroshi Onoda Hiroshi Tokunaga
Yuko Osumi Shogo Tokuoka
Masami Otsuka Kiyoshi Tomioka
Tadashi Ouchi Hiroyuki Tomitsuka
Hideyuki Saito Taichi Toyoda
Eiji Sakai Shigeki Tsuda
Tomoaki Sakamoto Eriko Uchida
Tomofumi Santa Nahoko Uchiyama
Hiroshi Sasaki Atsuhiro Uetake
Kunio Sasaki Yoshio Wada
Supplement I to
The Japanese
Pharmacopoeia
Seventeenth Edition
GENERAL RULES
FOR PREPARATIONS
(4) Dry Powder Inhalers meet the requirements of Aero-
[1] General Notices for Preparations dynamic Particle Size Measurement for Inhalations <6.15>,
unless otherwise specified.

Change the paragraphs (9) as follows:


Change the paragraphs (3) and (4) under 5-1-3.
(9) Even non-sterile preparations should be prepared Metered-dose Inhalers as follows:
with precautions to prevent contamination and growth of
5-1-3. Metered-dose Inhalers
microorganisms, and the test of Microbiological Examina-
(3) Metered-dose Inhalers meet the requirements of
tion of Non-sterile Products <4.05> or Microbial Limit Test
Uniformity of Delivered Dose for Inhalations <6.14>, unless
for Crude Drugs and Preparations containing Crude Drugs
otherwise specified.
as Main Ingredient <5.02> is applied to them, if necessary.
(4) Metered-dose Inhalers meet the requirements of
Aerodynamic Particle Size Measurement for Inhalations
<6.15>, unless otherwise specified.
[3] Monographs for Preparations
Change the paragraphs (5) under 9-1. Supposito-
Add the following next to section 1-7. Jellies for ries for Rectal Application as follows:
Oral Administration:
9-1. Suppositories for Rectal Application
1-8. Films for Oral Administration (5) Suppositories for Rectal Application show an ap-
(1) Films for Oral Administration are preparations in propriate release. Release of Suppositories for Rectal Appli-
film form, intended for oral administration. cation prepared using a lipophilic base can be evaluated by
(2) Films for Oral Administration are usually prepared melting behavior of suppositories in place of release of ac-
by spreading to dry a solution, composed of active sub- tive substances. When the melting behavior of Suppositories
stance(s) and a mixture of water-soluble polymer and other for Rectal Application is measured according to Method 2
additives as a base, or by melting the mixture of active sub- under Melting Point Determination <2.60> unless otherwise
stances(s) and the base to form. Layers different in additive specified, it shows an appropriate melting temperature.
compositions may be stacked in appropriate manner to
form the films. Change the paragraphs (5) under 10-2. Supposi-
(3) Unless otherwise specified, Films for Oral Adminis- tories for Vaginal Use as follows:
tration meet the requirement of Uniformity of Dosage Units
10-2. Suppositories for Vaginal Use
<6.02>.
(5) Suppositories for Vaginal Use show an appropriate
(4) Unless otherwise specified, Films for Oral Adminis-
release. Release of Suppositories for Vaginal Use prepared
tration meet the requirement of Dissolution Test <6.10> or
using a lipophilic base can be evaluated by melting behavior
show an appropriate disintegration.
of suppositories in place of release of active substances.
(5) Well-closed containers are usually used for Films for
When the melting behavior of Suppositories for Vaginal
Oral Administration. For the preparations susceptible to
Use is measured according to Method 2 under Melting Point
degradation by moisture, a moisture-proof container or
Determination <2.60> unless otherwise specified, it shows an
packaging may be used.
appropriate melting temperature.
1-8-1. Orally Disintegrating Films
(1) Orally Disintegrating Films are films which are
Change the paragraphs (3) under 11-2. Liquids
quickly dissolved or disintegrated in the oral cavity.
and Solutions for Cutaneous Application as
(2) Orally Disintegrating Films show an appropriate dis-
follows:
integration.
11-2. Liquids and Solutions for Cutaneous Application
Change the paragraphs (3) and (4) under 5-1-1. (3) Unless otherwise specified, Liquids and Solutions
Dry Powder Inhalers as follows: for Cutaneous Application in single-dose packages such as
Transdermal Systems meet the requirements of Uniformity
5-1-1. Dry Powder Inhalers
of Dosage Units <6.02>.
(3) Metered-dose types among Dry Powder Inhalers
meet the requirements of Uniformity of Delivered Dose for
Inhalations <6.14>, unless otherwise specified.

2631
The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
GENERAL TESTS, PROCESSES
AND APPARATUS
assay, water determination, and X-ray powder diffraction
Change the introduction to read: are performed as directed in the corresponding articles
under the General Tests, Processes and Apparatus. The
General Tests, Processes and Apparatus includes com-
tests for melting point of fats, congealing point of fatty
mon methods for tests, useful test methods for quality
acids, specific gravity, acid value, saponification value,
recognition of drugs and other articles related to them.
ester value, hydroxyl value, unsaponifiable matter and
Unless otherwise specified, acid-neutralizing capacity deter-
iodine value of fats and fatty oils are performed as directed
mination of gastrointestinal medicines, aerodynamic par-
in the corresponding items under Fats and Fatty Oils Test,
ticle size measurement for inhalations, alcohol number
and sampling, preparation of sample for analysis, micro-
determination, amino acid analysis of proteins, ammonium
scopic examination, purity test, loss on drying, total ash,
determination, arsenic determination, atomic absorption
acid-insoluble ash, extract content, essential oil content of
spectrophotometry, boiling point determination, chloride
crude drugs and assay of marker compounds for the assay
determination, conductivity measurement, congealing point
of crude drugs and extracts of Kampo Formulations utiliz-
determination, determination of bulk and tapped densities,
ing nuclear magnetic resonance (NMR) spectroscopy are
digestion test, disintegration test, dissolution test, distilling
performed as directed in the corresponding items under the
range determination, endpoint determination in titrimetry,
Crude Drugs Test.
flame coloration, fluorometry, foreign insoluble matter test
The number of each test method is a category number
for injections, foreign insoluble matter test for ophthalmic
given individually. The number in blackets (< >) appeared
liquids and solutions, gas chromatography, glycosylation
in monograph indicates the number corresponding to the
analysis of glycoprotein, heavy metal determination, induc-
general test method.
tively coupled plasma-atomic emission spectrometry and
inductively coupled plasma-mass spectrometry, infrared
spectrophotometry, insoluble particulate matter test for
injections, insoluble particulate matter test for ophthalmic 2.24 Ultraviolet-visible
liquids and solutions, iron determination, laser diffraction Spectrophotometry
measurement of particle size, liquid chromatography, loss
on drying determination, loss on ignition determination,
Change the following as follows:
mass spectrometry, melting point determination, methanol
determination, methods for color matching, methods of 1. Apparatus and adjustment
adhesion testing, microbial assay for antibiotics, mineral oil A spectrophotometer or a photoelectric photometer is
determination, nitrogen determination, nuclear magnetic used for the measurement of absorbance.
resonance spectroscopy, optical rotation determination, os- After adjusting the spectrophotometer or photoelectric
molarity determination, oxygen flask combustion method, photometer based on the operation manual of the appa-
particle size determination, particle size distribution test for ratus, it should be confirmed that the wavelength and the
preparations, pH determination, powder particle density de- transmission rate meet the specifications of the tests de-
termination, qualitative test, refractive index determination, scribed below.
release test for preparations for cutaneous application, The calibration of wavelength should be carried out as
residual solvents, residue on ignition determination, specific follows. Using an optical filter for wavelength calibration,
gravity and density determination, specific surface area de- measure the transmission rate in the vicinity of the standard
termination, sulfate determination, test for bacterial endo- wavelength value shown in the test results form, under the
toxins, test for glass containers for injections, test for metal test conditions given in the test results form attached to each
particles in ophthalmic ointments, test for microbial limit, of the filters. When performing a test to determine the
test for microbial limit for crude drugs, test for plastic con- wavelength which shows minimal transmission rate, the
tainers, test for pyrogen, test for readily carbonizable sub- difference between the measured wavelength and the stand-
stances, test for rubber closure for aqueous infusions, test ard wavelength value should be within ± 0.5 nm. When the
for sterility, test for total organic carbon, test of extractable measurement is repeated three times, each value obtained
volume for injection, thermal analysis, thin-layer chroma- should be within the mean ± 0.2 nm. It is also possible to
tography, turbidity measurement, ultravioletvisible spectro- carry out the test using a deuterium discharge lamp at bright
photometry, uniformity of delivered dose for inhalations, line wavelengths of 486.00 nm and 656.10 nm. In the case of
uniformity of dosage units (test for content uniformity, these tests, the difference between the measured wavelength
mass variation test), viscosity determination, vitamin A and the wavelength of the bright line should be within ±

2633
The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2634 General Tests, Processes and Apparatus Supplement I, JP XVII

0.3 nm. When the measurement is repeated three times, potential adverse effects. Ideally, less toxic solvents (Class
each value obtained should be within the mean ± 0.2 nm. 3, Table 2.46-3) should be used where practical.
The calibration of transmission rate or absorbance should Testing should be performed for residual solvents when
be carried out as follows. Using an optical filter for trans- production or purification processes are known to result in
mission rate calibration, determine the transmission rate at the presence of such solvents. It is only necessary to test for
the standard wavelength value under the test conditions solvents that are used or produced in the manufacture or
given in the test results form attached to each of the filters. purification of drug substances, excipients, or drug prod-
The difference between the measured transmission rate and ucts. Although manufacturers may choose to test the drug
the standard transmission rate value should be within the product, a cumulative method may be used to calculate the
range of from 1z larger of the upper limit to 1z smaller of residual solvent levels in the drug product from the levels in
the lower limit for the relative accuracy shown in the test the ingredients used to produce the drug product. If the
results form. When the measurement is repeated three calculation results in a level equal to or below that recom-
times, each absorbance obtained (or calculated from the mended in this chapter, no testing of the drug product for
transmission rate) should be within the mean ± 0.002 when residual solvents needs to be considered. If, however, the
the absorbance is not more than 0.500, and within the mean calculated level is above the recommended level, the drug
± 0.004 when the absorbance is more than 0.500. In addi- product should be tested to ascertain whether the formula-
tion, it will be desirable to confirm the linearity of transmis- tion process has reduced the relevant solvent level to within
sion rate at the same wavelength using several optical filters the acceptable amount. Drug product should also be tested
for calibration of transmission rate with different transmis- if a solvent is used during its manufacture.
sion rates. The limit applies to all dosage forms and routes of admin-
istration. Higher levels of residual solvents may be accepta-
ble in certain cases such as short term (30 days or less) or
2.46 Residual Solvents topical application. Justification for these levels should be
made on a case by case basis.

Change the following as follows: 2. General principles


2.1. Classification of residual solvents by risk assessment
The chapter of residual solvents describes the control,
The term ``PDE'' (Permitted Daily Exposure) is defined
identification and quantification of organic solvents
in this chapter as a pharmaceutically acceptable daily intake
remaining in drug substances, excipients and drug products.
of residual solvents. Residual solvents assessed in this chap-
I. Control of residual solvents ter were evaluated for their possible risk to human health
1. Introduction and placed into one of three classes as follows:
Residual solvents in pharmaceuticals (except for crude (i) Class 1 solvents: Solvents to be avoided in manufac-
drugs and their preparations) are defined here as organic ture of pharmaceuticals
volatile chemicals that are used or produced in the manufac- Known human carcinogens, strongly suspected human
ture of drug substances or excipients, or in the preparation carcinogens, and environmental hazards. Class 1 solvents
of drug products. The solvents are not completely removed are listed in Table 2.46-1.
by practical manufacturing techniques. Appropriate selec- (ii) Class 2 solvents: Solvents to be limited in phar-
tion of the solvent for the synthesis of drug substance may maceuticals
enhance the yield, or determine characteristics such as crys- Non-genotoxic animal carcinogens or possible causative
tal form, purity, and solubility. Therefore, the solvent may agents of other irreversible toxicity such as neurotoxicity or
sometimes be a critical parameter in the synthetic process. teratogenicity. Solvents suspected of other significant but
The test method described in this chapter does not address reversible toxicities. Class 2 solvents are listed in Table
solvents deliberately used as excipients nor does it address 2.46-2.
solvates. However, the content of solvents in such products (iii) Class 3 solvents: Solvents with low toxic potential
should be evaluated and justified. Solvents with low toxic potential to man; no health-based
Since there is no therapeutic benefit from residual sol- exposure limit is needed. Class 3 solvents are listed in Table
vents, all residual solvents should be reduced to the extent 2.46-3 and have PDEs of 50 mg or more per day.
possible to meet product specifications, good manufactur- 2.2. Option for describing limits of Class 2 solvents
ing practices, or other quality-based requirements. Drug Two options are available when setting limits for Class 2
products should contain no higher levels of residual solvents solvents.
than can be supported by safety data. Some solvents that 2.2.1. Option 1
are known to cause unacceptable toxicities (Class 1, Table The concentration limits in ppm can be calculated using
2.46-1) should be avoided in the production of drug sub- equation (1) below by assuming a product mass of 10 g
stances, excipients, or drug products unless their use can be administered daily.
strongly justified in a risk-benefit assessment. Some sol- 1000 × PDE
Concentration limit (ppm) = (1)
vents associated with less severe toxicity (Class 2, Table dose
2.46-2) should be limited in order to protect patients from Here, PDE is given in terms of mg per day and dose is

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2635

given in g per day. 5. Limits of residual solvents


These limits are considered acceptable for all substances, 5.1. Solvents to be avoided in manufacture of pharmaceu-
excipients, or products. Therefore this option may be ap- ticals
plied if the daily dose is not known or fixed. If all excipients Solvents in Class 1 should not be employed in the
and drug substances in a formulation meet the limits given manufacture of drug substances, excipients, and drug prod-
in Option 1, then these components may be used in any pro- ucts because of their unacceptable toxicity or their deleteri-
portion. No further calculation is necessary provided the ous environmental effect. However, if their use is unavoida-
daily dose does not exceed 10 g. Products that are ad- ble in order to produce a drug product with a significant
ministered in doses greater than 10 g per day should be con- therapeutic advance, then their levels should be restricted as
sidered under Option 2. shown in Table 2.46-1, unless otherwise justified. 1,1,1-
2.2.2. Option 2 Trichloroethane is included in Table 2.46-1 because it is an
It is not considered necessary for each component of the environmental hazard. The stated limit of 1500 ppm shown
drug product to comply with the limits given in Option 1. in Table 2.46-1 is based on a review of the safety data.
The PDE in terms of mg per day as stated in Table 2.46-2 5.2. Solvents to be limited in pharmaceuticals
can be used with the known maximum daily dose and equa- Solvents in Table 2.46-2 should be limited in drug prod-
tion (1) above to determine the concentration of residual ucts because of their inherent toxicity. PDEs are given to
solvent allowed in drug product. Such limits are considered the nearest 0.1 mg per day, and concentrations are given to
acceptable provided that it has been demonstrated that the the nearest 10 ppm. The stated values do not reflect the nec-
residual solvent has been reduced to the practical minimum. essary analytical precision of determination. Precision
The limits should be realistic in relation to analytical preci- should be determined as part of the validation of the
sion, manufacturing capability, reasonable variation in the method.
manufacturing process, and the limits should reflect con- 5.3. Solvents with low toxic potential
temporary manufacturing standards. Solvents in Class 3 shown in Table 2.46-3 may be re-
Option 2 may be applied by adding the amounts of a garded as less toxic and of lower risk to human health.
residual solvent present in each of the components of the Class 3 includes no solvent known as a human health hazard
drug product. The sum of the amounts of solvent per day at levels normally accepted in pharmaceuticals. The am-
should be less than that given by the PDE. ounts of these residual solvents of 50 mg per day or less
(corresponding to 5000 ppm or 0.5z under Option 1)
3. Analytical procedures
would be acceptable without justification. Higher amounts
Residual solvents are typically determined using chro-
may also be acceptable provided they are realistic in relation
matographic techniques such as gas chromatography. The
to manufacturing capability and good manufacturing prac-
analytical method should be validated adequately. If only
tice.
Class 3 solvents are present, a nonspecific method such as
5.4 Solvents for which no adequate toxicological data was
loss on drying may be used.
found
4. Reporting levels of residual solvents The following solvents (Table 2.46-4) may also be of in-
Manufacturers of drug products need certain information terest to manufacturers of excipients, drug substances, or
about the content of residual solvents in excipients or drug drug products. However, no adequate toxicological data on
substances. The following statements are given as accepta- which to base a PDE was found. Manufacturers should
ble examples of the information. supply justification for residual levels of these solvents in
(i) Only Class 3 solvents are likely to be present. Loss on drug products.
drying is not more than 0.5z.
(ii) Only Class 2 solvents are likely to be present. Name
the Class 2 solvents that are present. All are not more than
the Option 1 limit.
(iii) Only Class 2 solvents and Class 3 solvents are likely
to be present. Residual Class 2 solvents are not more than
the Option 1 limit and residual Class 3 solvents are not more Table 2.46-1 Class 1 solvents in drug products (solvents
than 0.5z. that should be avoided).
If Class 1 solvents are likely to be present, they should be Concentration
Solvent Concern
identified and quantified. ``Likely to be present'' refers to Limit (ppm)
the solvents that were used in the final manufacturing step
and to the solvents that were used in earlier manufacturing Benzene 2 Carcinogen
steps and not always possible to be excluded even in a vali- Carbon tetrachloride 4 Toxic and environ-
mental hazard
dated process.
1,2-Dichloroethane 5 Toxic
If solvents of Class 2 or Class 3 are present at greater than
1,1-Dichloroethene 8 Toxic
their Option 1 limits or 0.5z, respectively, they should be
identified and quantified. 1,1,1-Trichloroethane 1500 Environmental
hazard

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2636 General Tests, Processes and Apparatus Supplement I, JP XVII

Table 2.46-2 Class 2 Solvents which residual amount Table 2.46-4 Solvents for which no adequate toxicological
should be limited in drug products data was found.

PDE Concentration 1,1-Diethoxypropane Methyl isopropyl ketone


Solvent
(mg/day) limit (ppm) 1,1-Dimethoxymethane Methyltetrahydrofuran
2,2-Dimethoxypropane Petroleum ether
Acetonitrile 4.1 410
Isooctane Trichloroacetic acid
Chlorobenzene 3.6 360
Isopropyl ether Trifluoroacetic acid
Chloroform 0.6 60
Cumene 0.7 70
Cyclohexane 38.8 3880
1,2-Dichloroethene 18.7 1870 II. Identification and quantification of residual solvents
Dichloromethane 6.0 600 Whenever possible, the substance under test needs to be
1,2-Dimethoxyethane 1.0 100 dissolved to release the residual solvent. Because drug prod-
N, N-Dimethylacetamide 10.9 1090 ucts, as well as active ingredients and excipients are treated,
N, N-Dimethylformamide 8.8 880 it may be acceptable that in some cases, some of the compo-
1,4-Dioxane 3.8 380 nents of the formulation will not dissolve completely. In
2-Ethoxyethanol 1.6 160 those cases, the drug product may first need to be pulver-
Ethylene glycol 6.2 620 ized into a fine powder so that any residual solvent that may
Formamide 2.2 220 be present can be released. This operation should be per-
Hexane 2.9 290 formed as fast as possible to prevent the loss of volatile sol-
Methanol 30.0 3000 vents during the procedure.
2-Methoxyethanol 0.5 50 In the operating conditions of gas chromatography and
Methyl butyl ketone 0.5 50 headspace described below, parameters to be set and their
Methylcyclohexane 11.8 1180 description may be different depending on the apparatus.
N-Methylpyrrolidone 5.3 530 When setting these conditions, it is necessary to change
Nitromethane 0.5 50 them according to the apparatus used, if it is confirmed that
Pyridine 2.0 200 they meet the system suitability.
Sulfolane 1.6 160 In addition to the reagents specified to be used for the
Tetrahydrofuran 7.2 720 test, those that meet the purpose of the test can be used.
Tetralin 1.0 100
1. Class 1 and Class 2 residual solvents
Toluene 8.9 890
The following procedures are useful to identify and quan-
1,1,2-Trichloroethene 0.8 80
tify residual solvents when the information regarding which
Xylene* 21.7 2170
solvents are likely to be present in the material is not availa-
* Usually 60z m-xylene, 14z p-xylene, 9z o-xylene with ble. When the information about the presence of specific
17z ethylbenzene residual solvents are available, it is not necessary to perform
Procedure A and Procedure B, and only Procedure C or
other appropriate procedure is needed to quantify the
Table 2.46-3 Class 3 solvents which should be limited by amount of residual solvents.
GMP or other quality-based requirements. A flow chart for the identification of residual solvents
Acetic acid Heptane and the application of limit and quantitative tests is shown
Acetone Isobutyl acetate in Fig. 2.46-1.
Anisole Isopropyl acetate 1.1. Water-soluble articles
1-Butanol Methyl acetate 1.1.1. Procedure A
2-Butanol 3-Methyl-1-butanol The test is performed by gas chromatography <2.02> ac-
n-Butyl acetate Methyl ethyl ketone cording to the following conditions.
tert-Butyl methyl ether Methyl isobutyl ketone Class 1 standard stock solution: Pipet 1 mL of Residual
Dimethylsulfoxide 2-Methyl-1-propanol Solvents Class 1 RS, dissolve in about 9 mL of dimethylsul-
Ethanol Pentane foxide, and add water to make exactly 100 mL. Pipet 1 mL
Ethyl acetate 1-Pentanol of this solution in a volumetric flask, previously filled with
Diethyl ether 1-Propanol about 50 mL of water and add water to make exactly 100
Ethyl formate 2-Propanol mL. Pipet 10 mL of this solution in a volumetric flask, pre-
Formic acid Propyl acetate viously filled with about 50 mL of water and add water to
make exactly 100 mL.
Class 1 standard solution: Pipet 1 mL of Class 1 standard
stock solution in an appropriate head space vial containing
exactly 5 mL of water, apply the stopper, cap, and mix.
Class 2 standard stock solution A: Pipet 1 mL of Residual

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2637

silicon polymer for gas chromatography in a thickness of


1.8 mm (or 3.0 mm).
Column temperature: Maintain at 409 C for 20 minutes
after injection, then raise the temperature to 2409C at a rate
of 109C per minute, and maintain at 2409C for 20 minutes.
Injection port temperature: 1409C.
Detector temperature: 2509C.
Carrier gas: Nitrogen or Helium.
Flow rate: About 35 cm per second.
Split ratio: 1:5. (Note: The split ratio can be modified in
order to optimize sensitivity.)
System suitability—
Test for required detectability: When the procedure is run
with Class 1 standard solution and Class 1 system suitability
solution under the above operating conditions, the signal to
noise (SN) ratio of 1,1,1-trichloroethane in Class 1 standard
solution is not less than 5, and the SN ratio of each peak in
Class 1 system suitability solution is not less than 3.
System performance: When the procedure is run with
Class 2 standard solution A or the solution for system
suitability under the above operating conditions, the resolu-
tion between acetonitrile and dichloromethane is not less
than 1.0. Pipet 1 mL of a solution of Residual Solvents RS
for System Suitability (1 in 100) in an appropriate head-
space vial, add exactly 5 mL of water, and apply the stop-
per, cap, and mix, and use this solution as the solution for
system suitability.
System repeatability: When the test is repeated 6 times
with Class 1 standard solution under the above operating
conditions, the relative standard deviation of each peak area
Fig. 2.46-1 Flow chart for the identification of residual is not more than 15z.
solvents and the application of limit and qualification Separately inject (following one of the headspace operat-
tests. ing parameter sets described in Table 2.46-5) equal volumes
of headspace (about 1.0 mL) of Class 1 standard solution,
Class 2 standard solution A, Class 2 standard solution B,
Solvents Class 2A RS, add water to make exactly 100 mL. and test solution into the chromatograph, record the chro-
Class 2 standard stock solution B: Pipet 1 mL of Residual matograms, and measure the responses for the major peaks.
Solvents Class 2B RS, add water to make exactly 100 mL. If a peak response of any peak, other than a peak for 1,1,1-
Class 2 standard solution A: Pipet 1 mL of Class 2 stand- trichloroethane, in the test solution is greater than or equal
ard stock solution A in an appropriate headspace vial, add to a corresponding peak in either Class 1 standard solution,
exactly 5 mL of water, and apply the stopper, cap, and mix. Class 2 standard solution A or Class 2 standard solution B,
Class 2 standard solution B: Pipet 5 mL of Class 2 stand- or a peak response of 1,1,1-trichloroethane is greater than
ard stock solution B in an appropriate headspace vial, add or equal to 150 times the peak response corresponding to
exactly 1 mL of water, and apply the stopper, cap, and mix. 1,1,1-trichloroethane in Class 1 standard solution, proceed
Test stock solution: Dissolve 0.25 g of the article under to Procedure B to verify the identity of the peak; otherwise
test in water, and add water to make exactly 25 mL. the article meets the requirements of this test.
Test solution: Pipet 5 mL of test stock solution in an 1.1.2. Procedure B
appropriate headspace vial, add exactly 1 mL of water, and The test is performed by gas chromatography <2.02> ac-
apply the stopper, cap, and mix. cording to the following conditions.
Class 1 system suitability solution: Pipet 1 mL of Class 1 Class 1 standard stock solution, Class 1 standard solu-
standard stock solution in an appropriate headspace vial, tion, Class 1 system suitability solution, Class 2 standard
add exactly 5 mL of test stock solution, and apply the stop- stock solutions A and B, Class 2 standard solutions A and
per, cap, and mix. B, test stock solution and test solution: Prepare as directed
Operating conditions— for Procedure A.
Detector: A hydrogen flame-ionization detector. Operating conditions—
Column: A fused silica column (or a wide-bore column) Detector: A hydrogen flame-ionization detector.
0.32 mm (or 0.53 mm) in inside diameter and 30 m in Column: A fused silica column (or a wide-bore column)
length, coated inside with 6z cyanopropylphenylmethyl 0.32 mm (or 0.53 mm) in inside diameter and 30 m in

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2638 General Tests, Processes and Apparatus Supplement I, JP XVII

length, coated inside with polyethylene glycol for gas chro- a suitable container, and dilute quantitatively and stepwise
matography in a thickness of 0.25 mm. if necessary, with water to obtain a solution having a final
Column temperature: Maintain at 509C for 20 minutes concentration of 1/20 of the value stated in Table 2.46-1 or
after injection, then raise the temperature to 1659C at a rate Table 2.46-2.
of 69C per minute, and maintain at 1659C for 20 minutes. Standard solution: Pipet 1 mL of standard stock solution
Injection port temperature: 1409C. in an appropriate headspace vial, add exactly 5 mL of
Detector temperature: 2509C. water, and apply the stopper, cap, and mix.
Carrier gas: Nitrogen or Helium. Test stock solution: Weigh accurately about 0.25 g of the
Flow rate: About 35 cm per second. article under test, dissolve in water, and add water to make
Split ratio: 1:5. (Note: The split ratio can be modified in exactly 25 mL.
order to optimize sensitivity.) Test solution: Pipet 5 mL of test stock solution in an ap-
System suitability— propriate headspace vial, add exactly 1 mL of water, and
Test for required detectability: When the procedure is run apply the stopper, cap, and mix.
with Class 1 standard solution and Class 1 system suitability Spiked test solution (Note: prepare a separate spiked test
solution under the above operating conditions, the SN ratio solution for each peak identified and verified by Procedure
of benzene in Class 1 standard solution is not less than 5, A and B): Pipet 5 mL of test stock solution in an appropri-
and the SN ratio of each peak in Class 1 system suitability ate headspace vial, add exactly 1 mL of standard stock solu-
solution is not less than 3. tion, and apply the stopper, cap, and mix.
System performance: When the procedure is run with Operating conditions and system suitability fundamen-
Class 2 standard solution A or the solution for system tally follow the procedure A. If the results of the chroma-
suitability under the above operating conditions, the resolu- tography from Procedure A are found to be inferior to
tion between acetonitrile and cis-1,2-dichloroethene is not those found with Procedure B, the operating conditions
less than 1.0. Pipet 1 mL of a solution of Residual Solvents from Procedure B may be substituted.
RS for System Suitability (1 in 100) in an appropriate head- Separately inject (following one of the headspace operat-
space vial, add exactly 5 mL of water, and apply the stop- ing parameters described in Table 2.46-5) equal volumes of
per, cap, and mix, and use this solution as the solution for headspace (about 1.0 mL) of the standard solution, test so-
system suitability. lution, and spiked test solution into the chromatograph,
System repeatability: When the test is repeated 6 times record the chromatograms, and measure the responses for
with Class 1 standard solution under the above operating the major peaks. Calculate the amount of each residual sol-
conditions, the relative standard deviation of each peak area vent found in the article under test by the formula:
is not more than 15z.
Residual solvent (ppm) = 5(C/M) {AT/(AS - AT)}
Separately inject (following one of the headspace operat-
ing parameter sets described in Table 2.46-5) equal volumes C: Concentration (mg/mL) of the appropriate Reference
of headspace (about 1.0 mL) of Class 1 standard solution, Standard in the standard stock solution
Class 2 standard solution A, Class 2 standard solution B, M: Amount (g) of the article under test taken to prepare
and test solution into the chromatograph, record the chro- the test stock solution
matograms, and measure the responses for the major peaks. AT: Peak responses of each residual solvent obtained
If the peak response(s) in the test solution of the peak(s) from the test solution
identified in Procedure A is/are greater than or equal to a AS: Peak responses of each residual solvent obtained
corresponding peak(s) in either Class 1 standard solution, from the spiked test solution
Class 2 standard solution A or Class 2 standard solution B,
1.2. Water-insoluble articles
proceed to Procedure C to quantify the peak(s); otherwise
1.2.1. Procedure A
the article meets the requirements of this test.
The test is performed by gas chromatography <2.02> ac-
1.1.3. Procedure C
cording to the following conditions. Dimethylsulfoxide may
The test is performed by gas chromatography <2.02> ac-
be substituted as an alternative solvent to N,N-dimethylfor-
cording to the following conditions.
mamide.
Class 1 standard stock solution, Class 1 standard solu-
Class 1 standard stock solution: Pipet 1 mL of Residual
tion, Class 2 standard stock solution A, Class 2 standard so-
Solvents Class 1 RS, dissolve in about 80 mL of N,N-
lution A and Class 1 system suitability solution: Prepare as
dimethylformamide, and add N,N-dimethylformamide to
directed for Procedure A.
make exactly 100 mL. Pipet 1 mL of this solution in a volu-
Standard stock solution (Note: Prepare a separate stand-
metric flask, previously filled with about 80 mL of N,N-
ard stock solution for each peak identified and verified by
dimethylformamide and add N,N-dimethylformamide to
Procedures A and B. For Class 1 solvents other than 1,1,1-
make exactly 100 mL (this solution is the intermediate dilu-
trichloroethane, prepare the first dilution as directed for the
tion prepared from Residual Solvents Class 1 RS and use it
first dilution under Class 1 standard stock solution in
for preparation of Class 1 system suitability solution). Pipet
Procedure A.): Transfer an accurately measured volume of
1 mL of this solution, and add N,N-dimethylformamide to
each individual solvent corresponding to each residual sol-
make exactly 10 mL.
vent peak identified and verified by Procedures A and B to

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2639

Class 1 standard solution: Pipet 1 mL of Class 1 standard System repeatability: When the test is repeated 6 times
stock solution in an appropriate head space vial containing with Class 1 standard solution under the above operating
exactly 5 mL of water, apply the stopper, cap, and mix. conditions, the relative standard deviation of each peak
Class 2 standard stock solution A: Pipet 1 mL of Residual areas is not more than 15z.
Solvents Class 2A RS, dissolve in about 80 mL of N,N- Separately inject (use headspace operating parameters in
dimethylformamide, and add N,N-dimethylformamide to column 3 of Table 2.46-5) equal volumes of headspace
make exactly 100 mL. (about 1.0 mL) of Class 1 standard solution, Class 2 stand-
Class 2 standard stock solution B: Pipet 0.5 mL of Resid- ard solution A, Class 2 standard solution B, and test solu-
ual Solvents Class 2B RS, add N,N-dimethylformamide to tion into the chromatograph, record the chromatograms,
make exactly 10 mL. and measure the responses for the major peaks. If a peak
Class 2 standard solution A: Pipet 1 mL of Class 2 stand- response of any peak, other than a peak for 1,1,1-
ard stock solution A in an appropriate headspace vial, add trichloroethane, in the test solution is greater than or equal
exactly 5 mL of water, and apply the stopper, cap, and mix. to a corresponding peak in either Class 1 standard solution,
Class 2 standard solution B: Pipet 1 mL of Class 2 stand- Class 2 standard solution A or Class 2 standard solution B,
ard stock solution B in an appropriate headspace vial, add or a peak response of 1,1,1-trichloroethane is greater than
exactly 5 mL of water, and apply the stopper, cap, and mix. or equal to 150 times the peak response corresponding to
Test stock solution: Dissolve 0.5 g of the article under test 1,1,1-trichloroethane in Class 1 standard solution, proceed
in N,N-dimethylformamide, and add N,N-dimethylfor- to Procedure B to verify the identity of the peak; otherwise,
mamide to make exactly 10 mL. the article meets the requirements of this test.
Test solution: Pipet 1 mL of test stock solution in an ap- 1.2.2. Procedure B
propriate headspace vial, add exactly 5 mL of water, and The test is performed by gas chromatography <2.02> ac-
apply the stopper, cap, and mix. cording to the following conditions.
Class 1 system suitability solution: Pipet 5 mL of test Class 1 standard stock solution, Class 1 standard solu-
stock solution and 0.5 mL of the intermediate dilution pre- tion, Class 1 system suitability solution, Class 2 standard
pared from Residual Solvents Class 1 RS, and mix. Pipet 1 stock solutions A and B, Class 2 standard solutions A and
mL of this solution in an appropriate headspace vial, add B, test stock solution, and test solution: Proceed as directed
exactly 5 mL of water, and apply the stopper, cap, and mix. for Procedure A.
Operating conditions— Proceed as directed for Procedure B under Water-soluble
Detector: A hydrogen flame-ionization detector. articles with a split ratio of 1:3. (Note: The split ratio can be
Column: A wide-bore column 0.53 mm in inside diameter modified in order to optimize sensitivity.) The solution for
and 30 m in length, coated inside with 6z cyanopropyl- system suitability: Proceed as directed for Procedure A.
phenylmethyl silicon polymer for gas chromatography in a Separately inject (use headspace operating parameters in
thickness of 3.0 mm. Table 2.46-5) equal volumes of headspace (about 1.0 mL)
Column temperature: Maintain at 409C for 20 minutes of Class 1 standard solution, Class 2 standard solution A,
after injection, then raise the temperature to 2409C at a rate Class 2 standard solution B, and test solution into the chro-
of 109C per minute, and maintain at 2409C for 20 minutes. matograph, record the chromatograms, and measure the
Injection port temperature: 1409C. responses for the major peaks. If the peak response(s) in
Detector temperature: 2509C. test solution of the peak(s) identified in Procedure A is/are
Carrier gas: Helium. greater than or equal to a corresponding peak(s) in either
Flow rate: About 35 cm per second. Class 1 standard solution, Class 2 standard solution A or
Split ratio: 1:3. (Note: The split ratio can be modified in Class 2 standard solution B, proceed to Procedure C to
order to optimize sensitivity.) quantify the peak; otherwise, the article meets the require-
System suitability— ments of this test.
Test for required detectability: When the procedure is run 1.2.3 Procedure C
with Class 1 standard solution and Class 1 system suitability The test is performed by gas chromatography <2.02> ac-
solution under the above operating conditions, the SN ratio cording to the following conditions.
of 1,1,1-trichloroethane in Class 1 standard solution is not Class 1 standard stock solution, Class 1 standard solu-
less than 5, and the SN ratio of each peak in Class 1 system tion, Class 1 system suitability solution, Class 2 standard
suitability solution is not less than 3. stock solution A, and Class 2 standard solution A: Proceed
System performance: When the procedure is run with as directed for Procedure A.
Class 2 standard solution A or the solution for system Standard stock solution (Note: Prepare a separate stand-
suitability under the above operating conditions, the resolu- ard stock solution for each peak identified and verified by
tion between acetonitrile and dichloromethane is not less Procedures A and B. For Class 1 solvents other than 1,1,1-
than 1.0. Pipet 1 mL of the N,N-dimethylformamide solu- trichloroethane, prepare the first dilution as directed for the
tion of Residual Solvents RS for System Suitability (1 in first dilution under Class 1 standard stock solution in
100) in an appropriate headspace vial, add exactly 5 mL of Procedure A.): Transfer an accurately measured volume of
water, and apply the stopper, cap, and mix, and use this so- each individual solvent corresponding to each residual sol-
lution as the solution for system suitability. vent peak identified and verified by Procedures A and B to

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2640 General Tests, Processes and Apparatus Supplement I, JP XVII

a suitable container, and dilute quantitatively and stepwise Table 2.46-5 Headspace operating parameters
if necessary, with water to obtain a solution having a final
Headspace Operating
concentration of 1/20 of the value stated in Table 2.46-1 or
Parameter Sets
Table 2.46-2.
Standard solution: Pipet 1 mL of standard stock solution 1 2 3
in an appropriate headspace vial, add exactly 5 mL of
Equilibration temperature (9C) 80 105 80
water, and apply the stopper, cap, and mix.
Equilibration time (min.) 60 45 45
Test stock solution: Weigh accurately about 0.5 g of the
Transfer-line temperature (9C) 85 110 105
article under test, and add N,N-dimethylformamide to
Syringe temperature (9C) 80 – 90 105 – 115 80 – 90
make exactly 10 mL.
Test solution: Pipet 1 mL of test stock solution in an ap- Carrier gas: nitrogen or helium at an appropriate pressure
propriate headspace vial, add exactly 5 mL of water, and
Pressurization time (s) ≧ 60 ≧ 60 ≧ 60
apply the stopper, cap, and mix.
Injection volume (mL)* 1 1 1
Spiked test solution (Note: prepare a separate spiked test
solution for each peak identified and verified by Procedure * Or follow the instrument manufacture's recommenda-
A and B): Pipet 1 mL of test stock solution in an appropri- tions, as long as the method criteria are met. Injecting less
ate headspace vial, add exactly 4 mL of water, and apply than this amount is allowed as long as adequate sensi-
the stopper, cap, and mix. tivity is achieved.
Operating conditions and system suitability fundamental-
ly follow the procedure A. If the results of the chromatogra-
phy from Procedure A are found to be inferior to those necessary.
found with Procedure B, the operating conditions from
2. Class 3 residual solvents
Procedure B may be substituted.
Perform the test according to 1. Otherwise an appropriate
Separately inject (use headspace operating parameters in
validated procedure is to be employed. Prepare appropriate-
Table 2.46-5) equal volumes of headspace (about 1.0 mL)
ly standard solutions, etc. according to the residual solvent
of the standard solution, test solution, and spiked test solu-
under test.
tion into the chromatograph, record the chromatograms,
If only Class 3 solvents are present, the level of residual
and measure the responses for the major peaks. Calculate
solvents may be determined by loss on drying <2.41>.
the amount of each residual solvent found in the article
However when the value of the loss on drying is more than
under test by the formula:
0.5z, or other solvents exist, the individual Class 3 residual
Residual solvent (ppm) = 10(C/M) {AT/(AS - AT)} solvent or solvents present in the article under test should be
identified using the procedures as described above or other
C: Concentration (mg/mL) of the appropriate Reference
appropriate procedure, and quantified as necessary.
Standard in the standard stock solution
M: Amount (g) of the article under test taken to prepare 3. Reference Standards
the test stock solution (i) Residual Solvents Class 1 RS (A mixture of benzene,
AT: Peak responses of each residual solvent obtained carbon tetrachloride, 1,2-dichloroethane, 1,1-dichloro-
from the test solution ethene and 1,1,1-trichloroethane)
AS: Peak responses of each residual solvent obtained (ii) Residual Solvents Class 2A RS (A mixture of aceto-
from the spiked test solution nitrile, chlorobenzene, cumene, cyclohexane, 1,2-dichloro-
ethene (cis-1,2-dichloroethene, trans-1,2-dichloroethene),
1.3. Headspace operating parameters and other considera-
dichloromethane, 1,4-dioxane, methanol, methylcyclo-
tions
hexane, tetrahydrofuran, toluene and xylene (m-xylene,
Examples of headspace operating parameters are shown
p-xylene, o-xylene, ethylbenzene))
in Table 2.46-5.
(iii) Residual Solvents Class 2B RS (A mixture of chlo-
These test methods describe the analytical methods using
roform, 1,2-dimethoxyethane, hexane, methyl butyl ketone,
the headspace gas chromatography. The following Class 2
nitromethane, pyridine, tetralin and 1,1,2-trichloroethene)
residual solvents are not readily detected by the headspace
(iv) Residual Solvents RS for System Suitability (A
injection conditions: 2-ethoxyethanol, ethylene glycol, for-
mixture of acetonitrile, cis-1,2-dichloroethene and dichloro-
mamide, 2-methoxyethanol, N-methylpyrrolidone, and sul-
methane)
folane. Other appropriate validated procedures are to be
employed for the quantification of these residual solvents.
In the headspace methods, N,N-dimethylformamide and
N,N-dimethylacetoamide are often used as solvents. As not
only 6 solvents described above but these two solvents are
not included in the Residual Solvents Class 2A RS and/or
the Residual Solvents Class 2B RS, appropriate validated
procedures are to be employed for these residual solvents as

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2641

Add the following:

3.06 Laser Diffraction


Measurement of Particle Size
This test is harmonized with the European Pharma-
copoeia and the U.S. Pharmacopoeia.

The laser light diffraction technique used for the determi-


nation of particle-size distribution is based on the analysis
of the diffraction pattern produced when particles are ex-
posed to a beam of monochromatic light. Historically, the
early laser diffraction instruments only used scattering at
small angles. However, the technique has since been broa-
dened to include laser light scattering in a wider range and
application of the Mie theory, in addition to the Fraunhofer
approximation and anomalous diffraction.
The technique cannot distinguish between scattering by
primary particles and scattering by clusters of primary parti-
cles, i.e. by agglomerates or aggregates. As most particulate Fig. 3.06-1 Example of a set-up of a laser light diffraction
samples contain agglomerates or aggregates and as the focus instrument
of interest is generally on the size distribution of primary
particles, the clusters are usually dispersed into primary par-
ticles before measurement. of the lens. The second set-up allows only small path lengths
For non-spherical particles, an equivalent sphere-size dis- but enables measurement of scattered light at larger angles,
tribution is obtained because the technique assumes spheri- which is useful when submicron particles are present.
cal particles in its optical model. The resulting particle-size The interaction of the incident light beam and the ensem-
distribution may differ from those obtained by methods ble of dispersed particles results in a scattering pattern with
based on other physical principles (e.g. sedimentation, siev- different light intensities at various angles. The total angu-
ing). lar intensity distribution, consisting of both direct and scat-
This chapter provides guidance for the measurement of tered light, is then focused onto a multi-element detector by
size distributions of particles in different dispersed systems, a lens or a series of lenses. These lenses create a scattering
for example, powders, sprays, aerosols, suspensions, emul- pattern that, within limits, does not depend on the location
sions, and gas bubbles in liquids, through analysis of their of the particles in the light beam. Hence, the continuous
angular light-scattering patterns. It does not address specific angular intensity distribution is converted into a discrete
requirements of particle-size measurement of specific prod- spatial intensity distribution on a set of detector elements.
ucts. This technique complies with ISO13320-1 (1999) and It is assumed that the measured scattering pattern of the
9276-1 (1998). particle ensemble is identical to the sum of the patterns
from all individual single scattering particles presented in
1. Instrument
random relative positions. Note that only a limited angular
The instrument is located in an environment where it is
range of scattered light is collected by the lens(es) and,
not affected by electrical noise, mechanical vibrations, tem-
therefore, by the detector.
perature fluctuations, humidity or direct bright light. An
example of a set-up of a laser light diffraction instrument is 2. Development of the method
given in Fig. 3.06-1. Other equipment may be used. The measurement of particle size by laser diffraction can
The instrument comprises a laser light source, beam proc- give reproducible data, even in the sub-micron region, pro-
essing optics, a sample measurement region (or cell), a vided the instrument used and the sample tested are care-
Fourier lens, and a multi-element detector for measuring the fully controlled to limit variability of the test conditions
scattered light pattern. A data system is also required for (e.g. dispersion medium, method of preparation of the sam-
deconvolution of the scattering data into a volumetric size ple dispersion).
distribution and associated data analysis and reporting. Traditionally, the measurement of particle size using laser
The particles can enter the laser beam in 2 positions. In diffraction has been limited to particles in the range of ap-
the conventional case the particles enter the parallel beam proximately 0.1 mm to 3 mm. Because of recent advances in
before the collecting lens and within its working distance. In lens and equipment design, newer instruments are capable
socalled reversed Fourier optics the particles enter behind of exceeding this range routinely. With the validation report
the collecting lens and thus, in a converging beam. The ad- the user demonstrates the applicability of the method for its
vantage of the conventional set-up is that a reasonable path intended use.
length for the sample is allowed within the working distance

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2642 General Tests, Processes and Apparatus Supplement I, JP XVII

2.1. Sampling 2.3. Optimization of the liquid dispersion


The sampling technique must be adequate to obtain a Liquids, surfactants, and dispersing aids used to disperse
representative sample of a suitable volume for the particle powders must:
size measurement. Sample splitting techniques such as rotat- (i) be transparent at the laser wavelength and practically
ing riffler or the cone and quartering method may be ap- free from air bubbles or particles;
plied. (ii) have a refractive index that differs from that of the
2.2. Evaluation of the dispersion procedure test material;
Inspect the sample to be analyzed, visually or with the aid (iii) be non-solvent of the test material (pure liquid or
of a microscope, to estimate its size range and particle pre-filtered, saturated solution);
shape. The dispersion procedure must be adjusted to the (iv) not alter the size of the test materials (e.g. by solu-
purpose of the measurement. The purpose may be such that bility, solubility enhancement, or recrystallization effects);
it is preferable to deagglomerate clusters into primary parti- (v) favor easy formation and stability of the dispersion;
cles as far as possible, or it may be desirable to retain (vi) be compatible with the materials used in the instru-
clusters as intact as possible. In this sense, the particles of ment (such as O-rings, gaskets, tubing, etc.);
interest may be either primary particles or clusters. (vii) possess a suitable viscosity to facilitate recircula-
For the development of a method it is highly advisable to tion, stirring and filtration.
check that comminution of the particles does not occur, and Surfactants and/or dispersing aids are often used to wet
conversely, that dispersion of particles or clusters is satisfac- the particles and to stabilize the dispersion. For weak acids
tory. This can usually be done by changing the dispersing and weak bases, buffering of the dispersing medium at low
energy and monitoring the change of the particle-size distri- or high pH respectively can assist in identifying a suitable
bution. The measured size distribution must not change dispersant.
significantly when the sample is well dispersed and the A preliminary check of the dispersion quality can be per-
particles are neither fragile nor soluble. Moreover, if the formed by visual or microscopic inspection. It is also possi-
manufacturing process (e.g. crystallization, milling) of the ble to take fractional samples out of a well-mixed stock dis-
material has changed, the applicability of the method must persion. Such stock dispersions are formed by adding a liq-
by verified (e.g. by microscopic comparison). uid to the sample while mixing it with, for example, a glass
Sprays, aerosols and gas bubbles in a liquid should be rod, a spatula or a vortex mixer. Care must be taken to en-
measured directly, provided that their concentration is ade- sure the transfer of a representative sample and that settling
quate, because sampling or dilution generally alters the par- of larger particles does not occur. Therefore a sample paste
ticle-size distribution. is prepared or sampling is carried out quickly from a sus-
In other cases (such as emulsions, pastes and powders), pension maintained under agitation.
representative samples may be dispersed in suitable liquids. 2.4. Optimization of the gas dispersion
Dispersing aids (wetting agents, stabilizers) and/or mechan- For sprays and dry powder dispersions, a compressed gas
ical forces (e.g. agitation, sonication) are often applied for free from oil, water and particles may be used. To remove
deagglomeration or deaggregation of clusters and stabiliza- such materials from the compressed gas, a dryer with a filter
tion of the dispersion. For these liquid dispersions, a recir- can be used. Any vacuum unit should be located away from
culating system is most commonly used, consisting of an the measurement zone, so that its output does not disturb
optical measuring cell, a dispersion bath usually equipped the measurement.
with stirrer and ultrasonic elements, a pump, and tubing. 2.5. Determination of the concentration range
Non-recirculating, stirred cells are useful when only small In order to produce an acceptable SN ratio in the detec-
amounts of a sample are available or when special disper- tor, the particle concentration in the dispersion must exceed
sion liquids are used. a minimum level. Likewise, it must be below a maximum
Dry powders can also be converted into aerosols through level in order to avoid multiple scattering. The concentra-
the use of suitable dry powder dispersers, which apply tion range is influenced by the width of the laser beam, the
mechanical force for deagglomeration or deaggregation. path length of the measurement zone, the optical properties
Generally, the dispersers use the energy of compressed gas of the particles, and the sensitivity of the detector elements.
or the differential pressure of a vacuum to disperse the par- In view of the above, measurements must be performed
ticles to an aerosol, which is blown through the measuring at different particle concentrations to determine the ap-
zone, usually into the inlet of a vacuum unit that collects the propriate concentration range for any typical sample of ma-
particles. However, for free flowing, coarser particles or terial. (Note: in different instruments, particle concentra-
granules the effect of gravity may be sufficient to disperse tions are usually represented by differently scaled and
the particles adequately. differently named numbers, e.g. obscuration, optical con-
If the maximum particle size of the sample exceeds the centration, proportional number of total mass).
measuring range of the instrument, the material that is too 2.6. Determination of the measuring time
coarse can be removed by sieving and the mass and percen- The time of measurement, the reading time of the detec-
tage of removed material are reported. However, after tor and the acquisition frequency is determined experimen-
presieving, note that the sample is no longer representative, tally in accordance with the required precision. Generally,
unless otherwise proven. the time for measurement permits a large number of detec-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2643

tor scans or sweeps at short time intervals. tions) may vary appreciably from one substance to another.
2.7. Selection of an appropriate optical model However, it is good practice to aim at acceptance criteria
Most instruments use either the Fraunhofer or the Mie for repeatability such as RSD (z) ≦ 10z [n=6] for any
theory, though other approximation theories are sometimes central value of the distribution (e.g. for x50). Values at the
applied for calculation of the scattering matrix. The choice sides of the distribution (e.g. x10 and x90) are oriented
of the theoretical model depends on the intended applica- towards less stringent acceptance criteria such as RSD ≦
tion and the different assumptions (size, absorbance, refrac- 15z [n=6]. Below 10 mm, these values must be doubled.
tive index, roughness, crystal orientation, mixture, etc.) Robustness may be tested during the selection and optimiza-
made for the test material. If the refractive index values tion of the dispersion media and forces. The change of the
(real and imaginary parts for the used wavelength) are not dispersing energy may be monitored by the change in the
exactly known, then the Fraunhofer approximation or the particle-size distribution.
Mie theory with a realistic estimate of the refractive index
3. Measurement
can be used. The former has the advantages that it is simple
A representative sample, dispersed at an adequate con-
and it does not need refractive index values; the latter usu-
centration in a suitable liquid or gas, is passed through a
ally provides less-biased particle-size distributions for small
beam of monochromatic light, usually a laser. The light
particles. For instance, if the Fraunhofer model is used for
scattered by the particles at various angles is measured by a
samples containing an appreciable amount of small, trans-
multi-element detector. Numerical values representing the
parent particles, a significantly large amount of small parti-
scattering pattern are then recorded for analysis. These scat-
cles may be calculated. In order to obtain traceable results,
tering pattern values are then transformed, using an ap-
it is essential to document the refractive index values used,
propriate optical model and mathematical procedure, to
since small differences in the values assumed for the real
yield the proportion of total volume to a discrete number of
and imaginary part of the complex refractive index may
size classes, forming a volumetric particle-size distribution.
cause significant differences in the resulting particle-size dis-
3.1. Precautions
tributions. Small values of the imaginary part of the refrac-
(i) never look into the direct path of the laser beam or
tive index (about 0.01 – 0.1i ) are often applied to allow the
its reflections;
correction of the absorbance for the surface roughness of
(ii) earth all instrument components to prevent ignition
the particles. It should be noted, in general, that the optical
of solvents or dust explosions;
properties of the substance to be tested, as well as the struc-
(iii) check the instrument set-up (e.g. warm-up, required
ture (e.g. shape, surface roughness and porosity) bear upon
measuring range and lens, appropriate working distance,
the final result.
position of the detector, no direct bright daylight);
2.8. Validation
(iv) in the case of wet dispersions, avoid air bubbles,
Typically, the validity of a procedure may be assessed by
evaporation of liquid, schlieren or other inhomogeneities in
the evaluation of its specificity, linearity, range, accuracy,
the dispersion; similarly, avoid improper mass-flow from
precision and robustness. In particle-size analysis by laser
the disperser or turbulent air-flow in the case of dry disper-
light diffraction, specificity as defined by ICH (Validation
sions; such effects can cause erroneous particle-size distri-
of Analytical Procedures) is not applicable as it is not possi-
butions.
ble to discriminate different components into a sample, as is
3.2. Measurement of the light scattering of dispersed sam-
neither possible to discriminate between agglomerates from
ple(s)
dispersed particles unless properly complemented by
After proper alignment of the optical part of the instru-
microscopic techniques. Exploring a linear relationship be-
ment, a blank measurement of the particle-free dispersion
tween concentration and response, or a mathematical model
medium must be performed using the same method as that
for interpolation, is not applicable to this procedure. Rather
used for the measurement of the sample. The background
than evaluating linearity, this method requires the definition
signal must be below an appropriate threshold. The detector
of a concentration range within which the result of the mea-
data are saved in order to substract them later from the data
surements does not vary significantly. Concentrations below
obtained with the sample. The sample dispersion is meas-
that range produce an error due to a poor SN ratio, while
ured according to the developed method.
concentrations above that range produce an error due to
For each detector element, an average signal is calculated,
multiple scattering. The range depends mostly in the instru-
sometimes together with its standard deviation. The magni-
ment hardware. Accuracy should be confirmed through an
tude of the signal from each detector element depends upon
appropriate instrument qualification and comparison with
the detection area, the light intensity and the quantum ef-
microscopy, while precision may be assessed by means of a
ficiency. The co-ordinates (size and position) of the detector
repeatability determination.
elements together with the focal distance of the lens deter-
The attainable repeatability of the method mainly de-
mine the range of scattering angles for each element. Most
pends on the characteristics of the material (milled/not
instruments also measure the intensity of the central (un-
milled, robust/fragile, width of its size distribution, etc.),
scattered) laser beam. The ratio of the intensity of a dis-
whereas the required repeatability depends on the purpose
persed sample to that in its absence (the blank meas-
of the measurement. Mandatory limits cannot be specified
urement) indicates the proportion of scattered light and
in this chapter, as repeatabilities (different sample prepara-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2644 General Tests, Processes and Apparatus Supplement I, JP XVII

hence the particle concentration. taken using any certified reference material that is accepta-
3.3. Conversion of scattering pattern into particle-size dis- ble in industrial practice. The entire measurement procedure
tribution is examined, including sample collection, sample dispersion,
This deconvolution step is the inverse of the calculation sample transport through the measuring zone, measure-
of a scattering pattern for a given particle-size distribution. ment, and the deconvolution procedure. It is essential that
The assumption of spherical particle shape is particularly the total operational procedure is fully described.
important as most algorithms use the mathematical solution The preferred certified reference materials consist of
for scattering from spherical particles. Furthermore, the spherical particles of a known distribution. They must be
measured data always contain some random and systematic certified as to the mass-percentage size distribution by an
errors, which may vitiate the size distributions. Several absolute technique, if available, and used in conjunction
mathematical procedures have been developed for use in the with an agreed, detailed operation procedure. It is essential
available instruments. They contain some weighting of that the real and imaginary parts of the complex refractive
deviations between measured and calculated scattering index of the material are indicated if the Mie theory is ap-
patterns (e.g. least squares), some constraints (e.g. non- plied in data analysis. The representation of the particle-size
negativity for amounts of particles), and/or some smooth- distribution by volume will equal that of the distribution by
ing of the size distribution curve. mass, provided that the density of the particles is the same
The algorithms used are specific to each maker and model for all size fractions.
of equipment, and are proprietary. The differences in the The response of a laser diffraction instrument is consi-
algorithms between different instruments may give rise to dered to meet the requirements if the mean value of x50 from
differences in the calculated particle-size distributions. at least 3 independent measurements does not deviate by
3.4. Replicates more than 3z from the certified range of values of the cer-
The number of replicate measurements (with individual tified reference material. The mean values for x10 and x90
sample preparations) to be performed, depends on the re- must not deviate by more than 5z from the certified range
quired measurement precision. It is recommended to set this of values. Below 10 mm, these values must be doubled.
number in a substance-specific method. Although the use of materials consisting of spherical par-
ticles is preferable, non-spherical particles may also be em-
4. Reporting of results
ployed. Preferably, these particles have certified or typical
The particle-size distribution data are usually reported as
values from laser diffraction analysis performed according
cumulative undersize distribution and/or as density distri-
to an agreed, detailed operating procedure. The use of ref-
bution by volume. The symbol x is used to denote the parti-
erence values from methods other than laser diffraction
cle size, which in turn is defined as the diameter of a
may cause a significant bias. The reason for this bias is that
volume-equivalent sphere. Q3(x) denotes the volume frac-
the different principles inherent in the various methods may
tion undersize at the particle size x. In a graphical represen-
lead to different sphere-equivalent diameters for the same
tation, x is plotted on the abscissa and the dependent varia-
non-spherical particle.
ble Q3(x) on the ordinate. Most common characteristic
Although the use of certified reference materials is
values are calculated from the particle-size distribution by
preferred, other well-defined reference materials may also
interpolation. The particle sizes at the undersize values of
be employed. They consist of substances of typical composi-
10z, 50z, and 90z (denoted as x10, x50, and x90 respec-
tion and particle-size distribution for a specified class of
tively) are frequently used. x50 is also known as the median
substances. Their particle-size distribution has proven to be
particle size. It is recognized that the symbol d is also widely
stable over time. The results must comply with previously
used to designate the particle size, thus the symbol x may be
determined data, with the same precision and bias as for the
replaced by d.
certified reference material.
Moreover, sufficient information must be documented
5.2. Qualification of the system
about the sample, the sample preparation, the dispersion
In addition to the calibration, the performance of the
conditions, and the cell type. As the results depend on the
instrument must be qualified at regular time intervals or as
particular instrument, data analysis program, and optical
frequently as appropriate. This can be undertaken using any
model used, these details must also be documented.
suitable reference material as mentioned in the previous
5. Control of the instrument performance paragraph.
Use the instrument according to the manufacturer's in- The qualification of the system is based on the concept
structions and carry out the prescribed qualifications at an that the equipment, electronics, software and analytical
appropriate frequency, according to the use of the instru- operations constitute an integral system, which can be eval-
ment and substances to be tested. uated as an entity. Thus the entire measurement procedure
5.1. Calibration is examined, including sample collection, sample dispersion,
Laser diffraction systems, although assuming idealized sample transport through the measuring zone, and the
properties of the particles, are based on first principles of measurement and deconvolution procedure. It is essential
laser light scattering. Thus, calibration in the strict sense is that the total operational procedure is fully described.
not required. However, it is still necessary to confirm that In general, unless otherwise specified in the individual
the instrument is operating correctly. This can be under- monograph, the response of a laser diffraction instrument is

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2645

considered to meet the requirements if the x50 value does not capsules, packets of powders or granules, ampoules, con-
deviate by more than 10z from the range of values of the tain a single dose or a part of a dose of a drug substance in
reference material. If optionally the values at the sides of each dosage unit. The Uniformity of Dosage Units specifi-
the distribution are evaluated (e.g. x10 and x90), then these cation is not intended to apply to suspensions, emulsions, or
values must not deviate by more than 15z from the certi- gels in unit-dose containers intended for external, cutaneous
fied range of values. Below 10 mm, these values must be administration.
doubled. For calibration of the instrument stricter require- The uniformity of dosage units can be demonstrated by
ments are laid down in 5.1. Calibration. either of two methods, Content uniformity or Mass varia-
tion (see Table 6.02-1). The test for Content Uniformity of
preparations presented in dosage units is based on the assay
4.03 Digestion Test of the individual contents of drug substance(s) of a number
of dosage units to determine whether the individual contents
are within the limits set. The Content Uniformity method
Change the 2.2. Tyrosine Calibration Curve as
may be applied in all cases.
follows:
The test for Mass Variation is applicable for the follow-
2.2. Tyrosine Calibration Curve ing dosage forms:
Weigh exactly 50 mg of Tyrosine RS for Digestion Test, (i) solutions enclosed in unit-dose containers and into
previously dried at 1059C for 3 hours, and dissolve in 0.2 soft capsules ◇in which all components are perfectly dis-
mol/L hydrochloric acid TS to make exactly 50 mL. Pipet 1 solved◇;
mL, 2 mL, 3 mL and 4 mL of this solution separately, and (ii) solids (including powders, granules and sterile
add 0.2 mol/L hydrochloric acid TS to each solution to solids) that are packaged in single-dose packages and con-
make exactly 100 mL. Pipet 2 mL of each solution, and add tain no active or inactive added substances;
exactly 5 mL of 0.55 mol/L sodium carbonate TS and 1 mL (iii) solids (including sterile solids) that are packaged in
of diluted Folin's TS (1 in 3) to each solution, shake imme- single-dose packages, with or without active or inactive
diately, then stand them at 37 ± 0.59 C for 30 minutes. added substances, that have been prepared from true solu-
Determine the absorbances, A1, A2, A3 and A4, of these tions ◇in which all components are perfectly dissolved◇ and
solutions at 660 nm as directed under Ultraviolet-visible freeze-dried in the final packages and are labeled to indicate
Spectrophotometry <2.24>, using a solution prepared with this method of preparation; and
exactly 2 mL of 0.2 mol/L hydrochloric acid TS in the same (iv) hard capsules, uncoated tablets, or film-coated
manner as the blank. Then, draw a calibration curve with tablets, containing 25 mg or more of a drug substance com-
the absorbances, A1, A2, A3 and A4 as the ordinate, and prising 25z or more, by weight, of the dosage unit or, in
with the amount (mg) of tyrosine in 2 mL of each solution as the case of hard capsules, the capsule contents, ◇or in the
the abscissa. Obtain the amount (mg) of tyrosine for the ab- case of film-coated tablets, the pre-coated tablets,◇ except
sorbance difference of 1. that uniformity of other drug substances present in lesser
proportions is demonstrated by meeting Content Uniformi-
ty requirements.
6.02 Uniformity of Dosage Units The test for Content Uniformity is required for all dosage
forms not meeting the above conditions for the Mass Varia-
tion test. Alternatively, products listed in item (iv) above
Change as follows:
that do not meet the 25 mg/25z threshold limit may be
This test is harmonized with the European Phar- tested for uniformity of dosage units by Mass Variation
macopoeia and the U.S. Pharmacopeia. instead of the Content Uniformity test if the concentration
The corresponding part of the attributes/provisions relative standard deviation (RSD) of the drug substance in
which are agreed as non-harmonized within the scope of the the final dosage units is not more than 2z, based on proc-
harmonization is marked with symbols (◆ ◆), and the cor- ess validation data and development data, and if there has
responding parts which are agreed as the JP local require- been regulatory approval of such a change. The concentra-
ment other than the scope of the harmonization are marked tion RSD is the RSD of the concentration per dosage unit
with symbols (◇ ◇). (w/w or w/v), where concentration per dosage unit equals
the assay result per dosage unit divided by the individual
The term ``Uniformity of dosage unit'' is defined as the
dosage unit weight. See the RSD formula in Table 6.02-2.
degree of uniformity in the amount of the drug substance
among dosage units. Therefore, the requirements of this 1. Content Uniformity
chapter apply to each drug substance being comprised in Select not less than 30 units, and proceed as follows for
dosage units containing one or more drug substances, unless the dosage form designated.
otherwise specified elsewhere in this Pharmacopoeia. Where different procedures are used for assay of the
To ensure the consistency of dosage units, each unit in a preparation and for the content uniformity test, it may be
batch should have a drug substance content within a narrow necessary to establish a correction factor to be applied to
range around the label claim. Dosage forms such as tablets, the results of the latter.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2646 General Tests, Processes and Apparatus Supplement I, JP XVII

Table 6.02-1 Application of Content Uniformity (CU) and Mass Variation (MV) Test for dosage forms
Dose and ratio of drug substance
Dosage Forms Type Sub-type
 25 mg &  25z < 25 mg or < 25z

Tablets uncoated MV CU
coated Film MV CU

Others CU CU

Capsules hard MV CU
soft Sus., eml., gel CU CU

Solutions MV MV
Solids in single-dose packages ◇(divided forms, Single component MV MV
lyophilized forms, et al.)◇
Multiple components Freeze-dried from
solutions in final MV MV
container

Others CU CU

Solutions (perfectly dissolved)◇ enclosed in MV MV
unit-dose containers

Others—Among the preparations not classified as CU CU


the above dosage forms in this table, supposito-
ries, percutaneous absorption type preparations
(patches), semi-solid dosage forms intended for
application of active pharmaceutical ingredients
to the skin for the purpose of systemic action,
and the like.

Sus.: suspension; eml.: emulsion;

(i) Solid dosage forms: Assay 10 units individually using dividual tablets and the result of the assay. Calculate the ac-
an appropriate analytical method. Calculate the acceptance ceptance value.
value (see Table 6.02-2). (ii) Hard Capsules: Accurately weigh 10 capsules indi-
(ii) Liquid or Semi-Solid dosage forms: Assay 10 units vidually, taking care to preserve the identity of each cap-
individually using an appropriate analytical method. Carry sule. Remove the contents of each capsule by suitable
out the assay on the amount of well-mixed material that is means. Accurately weigh the emptied shells individually,
removed from an individual container in conditions of nor- and calculate for each capsule the net mass of its contents
mal use and express the results as delivered dose. Calculate by subtracting the mass of the shell from the respective
the acceptance value (see Table 6.02-2). gross mass. Calculate the drug substance content of each
1.1. Calculation of Acceptance Value capsule from the mass of the individual capsules and the
Calculate the acceptance value by the formula: result of the assay. Calculate the acceptance value.
(iii) Soft Capsules: Accurately weigh the 10 intact cap-
|M - X̃| + ks,
sules individually to obtain their gross masses, taking care
in which the terms are as defined in Table 6.02-2. to preserve the identity of each capsule. Then cut open the
capsules by means of a suitable clean, dry cutting instru-
2. Mass Variation

ment such as scissors or a sharp open blade, and remove the
Mass Variation is carried out based on the assumption
contents by washing with a suitable solvent. Allow the oc-
that the concentration (mass of drug substance per mass of
cluded solvent to evaporate from the shells at room temper-
dosage unit) is uniform in a lot.◇
ature over a period of about 30 minutes, taking precautions
Carry out an assay for the drug substance(s) on a
to avoid uptake or loss of moisture. Weigh the individual
representative sample of the batch using an appropriate
shells, and calculate the net contents. Calculate the drug
analytical method. This value is result A, expressed as z of
substance content in each capsule from the mass of product
label claim (see Calculation of the Acceptance Value). Select
removed from the individual capsules and the result of the
not less than 30 dosage units, and proceed as follows for the
assay. Calculate the acceptance value.
dosage form designated.
(iv) Solid dosage forms other than tablets and capsules:
(i) Uncoated or Film-coated Tablets: Accurately weigh
Proceed as directed for Hard Capsules, treating each dosage
10 tablets individually. Calculate the content, expressed as
unit as described therein. Calculate the acceptance value.
z of label claim, of each tablet from the mass of the in-
(v) Liquid dosage forms: Accurately weigh the amount

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2647

Table 6.02-2

Variable Definition Conditions Value


X̃ Mean of individual contents (x1, x2, ..., xn)
expressed as a percentage of the label claim

x1, x2, ..., xn Individual contents of the dosage units tested,


expressed as a percentage of the label claim
n Sample size (number of dosage units in a sample)

k Acceptability constant If n = 10, then 2.4


If n = 30, then 2.0
n
s Sample standard deviation

i=
(xi - X̃ )2
1

n-1

RSD Relative standard deviation (the sample standard 100s


deviation expressed as a percentage of the mean) X̃
M (case 1) Reference value If 98.5z ≦ X̃ ≦ 101.5z, M = X̃
then (AV = ks)

To be applied If X̃ < 98.5z, then M = 98.5z


when T ≦ 101.5 (AV = 98.5 - X̃ + ks)

If X̃ > 101.5z, then M = 101.5z


(AV = X̃ - 101.5 + ks)
M (case 2) Reference value If 98.5z ≦ X̃ ≦ T, then M = X̃
(AV = ks)

To be applied If X̃ < 98.5z, then M = 98.5z


when T > 101.5 (AV = 98.5 - X̃ + ks)
If X̃ > T, then M = Tz
(AV = X̃ - T + ks)

Acceptance General formula:


Value (AV ) |M - X̃| + ks
[Calculations are specified above for
the different cases.]

L1 Maximum allowed acceptance value L1 = 15.0 unless otherwise specified.

L2 Maximum allowed range for deviation of each On the low side, no dosage L2 = 25.0 unless otherwise specified.
dosage unit tested from the calculated value of M unit result can be less than
0.75M while on the high
side, no dosage unit result
can be greater than 1.25M
(This is based on an L2
value of 25.0.)

T Target content per dosage unit at time of manu-


facture, expressed as the percentage of the label
claim. Unless otherwise stated, T is 100.0z, or T
is the manufacturer's approved target content per
dosage unit.

of liquid that is removed from each of 10 individual con- units tested


tainers in conditions of normal use. If necessary, compute
A
the equivalent volume after determining the density. Calcu- xi = wi ×

late the drug substance content in each container from the
mass of product removed from the individual containers w1, w2 ... wn: Individual masses of the dosage units tested
and the result of the assay. Calculate the acceptance value. A: Content of drug substance (z of label claim) obtained
2.1. Calculation of Acceptance Value using an appropriate analytical method
Calculate the acceptance value as shown in Content W̃: Mean of individual masses (w1, w2 ... wn)
Uniformity, except that ◇the value of X̃ is replaced with A,
3. Criteria
and that◇ the individual contents of the dosage units are
Apply the following criteria, unless otherwise specified.
replaced with the individual estimated contents defined
(i) Solid, Semi-Solid and Liquid dosage forms: The re-
below.
quirements for dosage uniformity are met if the acceptance
x1, x2 ... xn: individual estimated contents of the dosage value of the first 10 dosage units is less than or equal to

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2648 General Tests, Processes and Apparatus Supplement I, JP XVII

L1z. If the acceptance value is greater than L1z, test the Add the following:
next 20 dosage units and calculate the acceptance value. The
requirements are met if the final acceptance value of the 30 6.14 Uniformity of Delivered
dosage units is less than or equal to L1z and no individual
content of the dosage unit is less than (1 - L2 × 0.01)M Dose for Inhalations
nor more than (1 + L2 × 0.01)M in Calculation of Accep-
This test is used to quantitatively evaluate the uniformity
tance Value under Content Uniformity or under Mass Vari-
of the amount of active substances sprayed or discharged
ation. Unless otherwise specified, L1 is 15.0 and L2 is 25.0.
from inhalations (metered-dose inhalers and dry powder in-
halers). Uniformity of the amount of active substances
which are administered to patients from these preparations
6.04 Test for Acid-neutralizing is necessary, and is confirmed by this test. Examples for the
Capacity of Gastrointestinal evaluation is shown as follows. Select a suitable test method
from the following, according to the characteristic of prepa-
Medicines rations. Original methods are able to be set, including the
test that can evaluate intra and inter-inhalers dose uniformi-
Change the following as follows: ty simultaneously.

2. Procedure 1. Test methods for metered-dose inhalers


Take an amount of the sample so that `a' in the equation Metered-dose inhalers usually operate in a valve-down
falls between 20 mL and 30 mL, and perform the test. position. For inhalers that operate in a valve-up position, an
Accurately weigh the sample of the crude material or equivalent test is applied using methods that ensure the
preparation, and place it in a glass-stoppered 200-mL flask. complete collection of the delivered dose.
Add exactly 100 mL of 0.1 mol/L hydrochloric acid VS, The dose collection apparatus must be capable of quan-
stopper tightly, shake at 37 ± 29 C for 1 hour, and filter. titatively capturing the delivered dose.
Take precaution against gas to be generated on the addition The following apparatus (Fig. 6.14-1) and procedure may
of 0.1 mol/L hydrochloric acid VS, and stopper tightly. be used.
After cooling, filter the solution again, if necessary. Pipet The apparatus consists of a filter-support base with an
50 mL of the filtrate, and titrate <2.50> the excess hydro-
chloric acid with 0.1 mol/L sodium hydroxide VS (pH
Determination <2.54>, end point: pH 3.5). Perform a blank
determination in the same manner.
For liquid preparations, pipet the sample in a 100-mL
volumetric flask, add water to make 45 mL, then add ex-
actly 50 mL of 0.2 mol/L hydrochloric acid VS while shak-
ing. Add water again to make the solution 100 mL. Transfer
the solution to a glass-stoppered 200-mL flask, wash the
residue with 20.0 mL of water, stopper tightly, shake at 37
± 29C for 1 hour, and filter. Pipet 60 mL of the filtrate,
and titrate <2.50> the excess hydrochloric acid with 0.1
mol/L sodium hydroxide VS (pH Determination <2.54>, end
point: pH 3.5). Perform a blank determination in the same
manner.

Acid-neutralizing capacity (amount of 0.1 mol/L hydro-


chloric acid VS consumed per g or daily dose) (mL)
= (b - a) × 2 × (t/s)

a: Amount (mL) of 0.1 mol/L sodium hydroxide VS con-


sumed
b: Amount (mL) of 0.1 mol/L sodium hydroxide VS con-
sumed in the blank determination
t: 1000 mg of crude material, or daily dose of preparation
(in mg of solid preparation, mL of liquid preparation)
s: Amount of the sample taken (in mg of crude material
and solid preparation, mL of liquid preparation)

Fig. 6.14-1 Dose collection apparatus for metered-dose in-


halers

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2649

open-mesh filter-support, such as a stainless steel screen, a value lies outside the limits of 65 to 135z.
collection tube that is clamped or screwed to the filter-sup- In justified cases, these ranges may be extended. But no
port base, and a mouthpiece adapter to ensure an airtight value should be less than 50z or more than 150z of the
seal between the collection tube and the mouthpiece. Use a limit.
mouthpiece adapter that ensures that the front face of the The mean value must be within 85 to 115z of the label
inhaler's mouthpiece is flush with the front face or the 2.5 claim for delivered dose.
mm indented shoulder of the sample collection tube, as ap- 1.2. Test method 2: evaluation of inter-inhaler dose
propriate. The vacuum connector is connected to a system uniformity
comprising a vacuum source and a flow regulator. The Take one inhaler, and perform the test. Unless otherwise
source is adjusted to draw air through the complete assem- prescribed in the instructions to the patient, shake the inhal-
bly, including the filter and the inhaler to be tested, at 28.3 er for 5 seconds and discharge one delivery to waste. Dis-
L per minute (±5z). Air should be drawn continuously charge the inverted inhaler into the apparatus, depressing
through the apparatus to avoid loss of the active substance the valve for a sufficient time to ensure complete discharge.
into the atmosphere. The filter-support base is designed to Repeat the procedure until the number of deliveries that
accommodate 25 mm diameter filter disks. The filter disk constitute the minimum recommended dose have been sam-
and other materials used in the construction of the appa- pled. Quantitatively collect the contents of the apparatus
ratus must be compatible with the active substance and sol- and determine the amount of active substance as the deli-
vents that are used to extract the active substance from the vered dose.
filter. One end of the collection tube is designed to hold the Repeat the procedure for a further 9 inhalers. Determine
filter disk tightly against the filter-support base. When as- 10 delivered doses, each 1 dose at the beginning for 10 in-
sembled, the joints between the components of the appa- halers, by the above process.
ratus are airtight so that when a vacuum is applied to the For preparations containing more than one active sub-
base of the filter, all of the air drawn through the collection stance, carry out the test for uniformity of delivered dose
tube passes through the inhaler. for each active substance.
1.1. Test method 1: evaluation of intra-inhaler dose The mean of the delivered doses or the delivered dose
uniformity stated on the label is used as the limit for judgement.
Take one inhaler, and perform the test. Unless otherwise The preparation complies with the test if 9 out of 10
prescribed in the instructions to the patient, shake the inhal- results lie between 75z and 125z of the limit and all lie be-
er for 5 seconds and discharge one delivery to waste. Dis- tween 65z and 135z of the limit. If 2 or 3 values lie out-
charge the inverted inhaler into the apparatus, depressing side the limits of 75 to 125z, repeat the above procedure
the valve for a sufficient time to ensure complete discharge. for 20 more inhalers, and obtain 30 values as the total. Not
Repeat the procedure until the number of deliveries that more than 3 of the 30 values lie outside the limits of 75 to
constitute the minimum recommended dose have been sam- 125z and no value lies outside the limits of 65 to 135z.
pled. Quantitatively collect the contents of the apparatus In justified cases, these ranges may be extended. But no
and determine the amount of active substance as the deli- value should be less than 50z or more than 150z of the
vered dose. limit.
Repeat the procedure for a further 2 doses. The mean value must be within 85 to 115z of the label
Discharge the inhaler to waste, waiting not less than 5 sec- claim for delivered dose.
onds between actuations, until (n/2) + 1 deliveries remain,
2. Test method for dry powder inhalers
where n is the number of deliveries stated on the label. Col-
The dose collection apparatus must be capable of quan-
lect 4 doses using the procedure described above.
titatively capturing the delivered dose. A dose collection
Discharge the inhaler to waste, waiting not less than 5 sec-
apparatus similar to that described for the valuation of
onds between actuations, until 3 doses remain. Collect these
metered-dose inhalers may be used provided that the dimen-
3 doses using the procedure described above. Determine 10
sions of the tube and the filter can accommodate the meas-
delivered doses per one inhaler, i.e., 3 doses at the begin-
ured flow rate. A suitable tube is defined in Table 6.14-1.
ning, 4 doses at the middle and 3 doses at the end by the
Connect the tube to a flow system according to the scheme
above process.
specified in Table 6.14-1 and Fig. 6.14-2.
For preparations containing more than one active sub-
Unless otherwise specified, determine the test flow rate
stance, carry out the test for uniformity of delivered dose
and duration using the dose collection tube, the associated
for each active substance.
flow system, a suitable differential pressure meter and a
The mean of the delivered doses or the delivered dose
suitable volumetric flowmeter, calibrated for the flow leav-
stated on the label is used as the limit for judgement.
ing the meter, according to the following procedure.
The preparation complies with the test if 9 out of 10
Prepare the inhaler for use according to the instructions
results lie between 75z and 125z of the limit and all lie
to the patient and connect it to the inlet of the apparatus
between 65z and 135z of the limit. If 2 or 3 values lie
using a mouthpiece adapter to ensure an airtight seal. Use a
outside the limits of 75 to 125z, repeat the test for 2 more
mouthpiece adapter that ensures that the front face of the
inhalers, and obtain 30 values as the total. Not more than 3
inhaler mouthpiece is flush with the front face of the sample
of the 30 values lie outside the limits of 75 to 125z and no

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2650 General Tests, Processes and Apparatus Supplement I, JP XVII

Table 6.14-1 Specifications of the apparatus described in Fig. 6.14-2

Code Component Description

A Sample collection tube Dimensions of 34.85 mm internal diameter × 12 cm length

B Filter 47 mm glass fiber filter

C Connector Internal diameter ≧ 8 mm (e.g., short metal coupling, with low-diameter branch to P3)

D Vacuum tubing A length of suitable tubing having an internal diameter ≧ 8 mm and an internal volume
of 25 ± 5 mL.

E Two-way solenoid valve A 2-way, 2-port solenoid valve having a minimum airflow resistance orifice with internal
diameter ≧ 8 mm and an opening time ≦ 100 ms

F Vacuum pump Pump must be capable of drawing the required flow rate through the assembled appa-
ratus with the dry powder inhaler in the mouthpiece adapter. Connect the pump to the
2-way solenoid valve using short and/or wide (≧ 10 mm internal diameter) vacuum tub-
ing and connectors to minimize pump capacity requirements.

G Timer Timer capable of driving the solenoid valve for the required time period.

P1 Pressure tap 2.2 mm internal diameter, 3.1 mm outer diameter, flush with internal surface of the
sample collection tube, centered and burr-free, 59 mm from its inlet. The pressure tap
P1 must never be open to the atmosphere. Differential pressure to the atmosphere is
measured at P1.

P2, P3 Pressure measurements Absolute pressure

H Flow control valve Adjustable regulating valve with maximum Cv ≧ 1.

Qin × P0
Qout =
P0 - D P
P0: atmospheric pressure
DP: pressure drop over the meter

If the flow rate is above 100 L per minute adjust the flow
control valve to obtain a flow rate of 100 L per minute
(±5z). Note the volumetric airflow rate exiting the meter
and define this as the test flow rate, Qout', in L per minute.
Fig. 6.14-2 Apparatus suitable for measuring the unifor- Define the test flow duration, T, in seconds so that a
mity of delivered dose for dry powder inhalers volume of 4 L of air is drawn from the mouthpiece of the
inhaler at the test flow rate, Qout'. Ensure that critical flow
occurs in the flow control valve by the following procedure:
with the inhaler in place and the test flow rate Qout', meas-
collection tube. Connect one port of a differential pressure ure the absolute pressure on both sides of the control valve
meter to the pressure reading point P1 in Fig. 6.14-2, and (pressure reading points P2 and P3 in Fig. 6.14-2); a ratio
let the other be open to the atmosphere. Switch on the P3/P2 of less than or equal to 0.5 indicates critical flow;
pump, open the 2-way solenoid valve and adjust the flow switch to a more powerful pump and re-measure the test
control valve until the pressure drop across the inhaler is 4.0 flow rate if critical flow is not indicated.
kPa (40.8 cm H2O) as indicated by the differential pressure Dry powder inhalers contain two types of inhalers, pre-
meter. Remove the inhaler from the mouthpiece adapter metered inhalers where powders for one emission are pre-
and, without touching the flow control valve, connect a dispensed in capsules or other suitable dosage forms and
flowmeter to the inlet of the sampling apparatus. Use a device-metered inhalers where powders for one emission are
flowmeter calibrated for the volumetric flow leaving the metered within the inhalers. Perform the test by the follow-
meter, or calculate the volumetric flow leaving the meter ing methods depending on each function of the pre-metered
(Qout) using the ideal gas law. For a meter calibrated for the inhalers or device-metered inhalers.
entering volumetric flow (Qin), use the following expression. 2.1. Pre-metered inhalers
Connect the inhaler to the apparatus using an adapter
that ensures a good seal. Draw air through the inhaler under
the predetermined conditions. Repeat the procedure until

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2651

the number of deliveries that constitute the minimum halers, and obtain 30 values as the total. Not more than 3 of
recommended dose have been sampled. Quantitatively col- the 30 values lie outside the limits of 75 to 125z and no
lect the contents of the apparatus and determine the amount value lies outside the limits of 65 to 135z.
of active substance as the delivered dose. In justified cases, these ranges may be extended. But no
Repeat the procedure for a further 9 doses. The sampling value should be less than 50z or more than 150z of the
procedure to obtain 10 values of delivered doses is limit.
prescribed individually in considering the discharge mechan- The mean value must be within 85 to 115z of the label
ism of each preparation. claim for delivered dose.
For preparations containing more than one active sub- 2.2.2. Test method 2: evaluation of inter-inhaler dose
stance, carry out the test for uniformity of delivered dose uniformity
for each active substance. Take one inhaler, and perform the test. Connect the in-
The mean of the delivered doses or the delivered dose haler to the apparatus using an adapter that ensures a good
stated on the label is used as the limit for judgement. seal. Draw air through the inhaler under the predetermined
The preparation complies with the test if 9 out of 10 conditions. Repeat the procedure until the number of deliv-
results lie between 75z and 125z of the limit and all lie be- eries that constitute the minimum recommended dose have
tween 65z and 135z of the limit. If 2 or 3 values lie out- been sampled. Quantitatively collect the contents of the ap-
side the limits of 75 to 125z, repeat the above procedure paratus and determine the amount of active substance as the
for 20 more delivered doses, and obtain 30 values as the delivered dose.
total. Not more than 3 of the 30 values lie outside the limits Repeat the procedure for a further 9 inhalers. Determine
of 75 to 125z and no value lies outside the limits of 65 to 10 delivered doses, each 1 dose at the beginning for 10 in-
135z. halers, by the above process.
In justified cases, these ranges may be extended. But no For preparations containing more than one active sub-
value should be less than 50z or more than 150z of the stance, carry out the test for uniformity of delivered dose
limit. for each active substance.
The mean value must be within 85 to 115z of the label The mean of the delivered doses or the delivered dose
claim for delivered dose. stated on the label is used as the limit for judgement.
2.2. Device-metered inhalers The preparation complies with the test if 9 out of 10
2.2.1. Test method 1: evaluation of intra-inhaler dose results lie between 75z and 125z of the limit and all lie be-
uniformity tween 65z and 135z of the limit. If 2 or 3 values lie out-
Take one inhaler, and perform the test. Connect the in- side the limits of 75 to 125z, repeat the above procedure
haler to the apparatus using an adapter that ensures a good for 20 more inhalers, and obtain 30 values as the total. Not
seal. Draw air through the inhaler under the predetermined more than 3 of the 30 values lie outside the limits of 75 to
conditions. Repeat the procedure until the number of deliv- 125z and no value lies outside the limits of 65 to 135z.
eries that constitute the minimum recommended dose have In justified cases, these ranges may be extended. But no
been sampled. Quantitatively collect the contents of the ap- value should be less than 50z or more than 150z of the
paratus and determine the amount of active substance as the limit.
delivered dose. The mean value must be within 85 to 115z of the label
Repeat the procedure for a further 2 doses. claim for delivered dose.
Discharge the inhaler to waste until (n/2) + 1 deliveries
remain, where n is the number of deliveries stated on the
label. If necessary, store the inhaler to discharge electrostat- Add the following:
ic charges. Collect 4 doses using the procedure described
above. 6.15 Aerodynamic Particle Size
Discharge the inhaler to waste until 3 doses remain. If
necessary, store the inhaler to discharge electrostatic Measurement for Inhalations
charges. Collect 3 doses using the procedure described
This test is used to evaluate the fine particle characteris-
above. Determine 10 delivered doses per one inhaler, i.e., 3
tics of the aerosol clouds generated by preparations for
doses at the beginning, 4 doses at the middle and 3 doses at
inhalation, and is performed using one of the following
the end by the above process.
apparatuses and test procedures. If justified, modified eq-
For preparations containing more than one active sub-
uipment or test procedure may be used.
stance, carry out the test for uniformity of delivered dose
for each active substance. 1. Stage mensuration
The mean of the delivered doses or the delivered dose The most reliable calibration for the separation character-
stated on the label is used as the limit for judgement. istic of each impaction stage is performed in terms of the
The preparation complies with the test if 9 out of 10 relationship between the stage collection efficiency and the
results lie between 75z and 125z of the limit and all lie be- aerodynamic diameter of particles and droplets passing
tween 65z and 135z of the limit. If 2 or 3 values lie out- through it as an aerosol.
side the limits of 75 to 125z, repeat the test for 2 more in- Calibration is usually performed by examination of the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2652 General Tests, Processes and Apparatus Supplement I, JP XVII

jet nozzle dimensions, the spatial arrangement of the jet method (apparatus 1) is shown in Fig. 6.15-1. The appa-
nozzle and its collection part, and the airflow rate passing ratus 1 consists of impaction stages 1 (pre-separator), 2, 3
through it. and 4 and an integral filter stage (stage 5), see Figures
Because jet nozzles can corrode and wear over time, the 6.15-1 to 6.15-3. An impaction stage comprises an upper
critical dimensions of each stage must be measured on a horizontal metal partition wall (B) through which a metal
regular basis to confirm them being within required ranges. inlet jet tube (A) with its collection plate (D) is protruding.
Only apparatuses that conform to specifications are used A glass cylinder (E) with sampling port (F) forms the verti-
for aerodynamic particle size measurement for inhalations. cal wall of the stage, and the stage is connected to the next
An alternate validated and justified method of mensuration lower stage by the tube (H) through a lower horizontal
may be used. metal partition wall (G). The tube into stage 4 (U) ends in a
multi-jet arrangement. The collection plate (D) is secured in
2. Re-entrainment
a metal frame (J) which is fastened by two wires (K) to a
When using the apparatuses 2 and 3, the selected tech-
sleeve (L) secured on the jet tube. The horizontal face of the
nique should seek to minimize particle re-entrainment (from
collection plate is perpendicular to the axis of the jet tube
an upper to a lower impaction stage) where this may affect
and centrally aligned. The upper surface of the collection
the amounts of drug collected. For example, minimizing the
plate is slightly raised above the edge of the metal frame. A
number of sampled doses and coating the particle collection
recess around the perimeter of the horizontal partition wall
surfaces are used to minimize particle re-entrainment.
guides the position of the glass cylinder. The glass cylinders
Glycerol, silicone oil or similar high viscosity liquid are used
are sealed against the horizontal partition walls with gaskets
to coat particle collection surfaces. Plate coating must be
(M) and clamped together by six bolts (N). The sampling
part of method validation and may be omitted where it is
ports are sealed by stoppers. The bottom-side (back) of the
demonstrated that the aerodynamic particle size is not in-
lower partition wall of stage 4 has a concentrical protrusion
fluenced by the coating.
fitted with a rubber O-ring (P) which seals against the edge
3. Inter-stage drug losses (wall losses) of a filter placed in the filter holder. The filter holder (R) is
Wall losses should be considered in method development constructed as a basin with a concentrical recess in which a
and validation. If the wall losses affect the recovery rate perforated filter support (S) is flush-fitted. The filter holder
(mass balance) of drugs, they should be controlled. Wall is dimensioned for 76 mm diameter filters. The assembly of
losses will be dependent upon a number of factors including impaction stages is clamped onto the filter holder by two
the impactor type, operating conditions, formulation type snap-locks (T). Connect an induction port (see Fig. 6.15-4)
and discharged amount to an impactor. How the wall loss is
reflected within the calculation of the aerodynamic diameter
of particles should be judged based up on the level and
variability of the wall loss. For example, in cases where wall
losses are low or have a low level of variability, the aer-
odynamic particle size is calculated by the assay of the drug
recovered from the collection plate. In cases where wall
losses are high or variable, it may be necessary to collect the
wall loss drug separately and take it into account for calcu-
lation of the aerodynamic particle size.

4. Recovery rate of drugs (mass balance)


In addition to the size distribution, good analytical prac-
tice dictates that a mass balance be performed in order to
confirm that the amount of the drug discharged from the in-
haler, which is collected in the mouthpiece adapter and the
apparatus, is within an acceptable range around the ex-
pected value. The total mass of drug collected in all of the
components of the mouthpiece adapter and the apparatus
divided by the minimum recommended dose described in
the dosage and administration is not less than 75z and not
more than 125z of the average delivered dose determined
under Uniformity of Delivered Dose for Inhalations <6.14>.
This mass balance is necessary to ensure that the test results
of particle size distributions are valid.

5. Measurement of fine particle dose and particle size dis-


tribution
5.1. Multi-stage liquid impinger method (Apparatus 1)
The apparatus used for the multi-stage liquid impinger Fig. 6.15-1 Multi-stage liquid impinger (Apparatus 1)

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2653

Table 6.15-1 Component specification for apparatus 1 in


Figures 6.15-1, 2 and 3
Code* Item Description Dimensions**
A, H Jet tube Metal tube screwed onto parti- see Fig. 6.15-2
tion wall sealed by gasket (C),
polished inner surface
B, G Partition Circular metal plate
wall —diameter 120
—thickness see Fig. 6.15-2

C Gasket e.g. polytetrafluoroethylene to fit jet tube

D Collection Porosity 0 sintered-glass disk


plate —diameter see Fig. 6.15-2

E Glass Plane polished cut glass tube


cylinder —height, including gaskets 46
—outer diameter 100
—wall thickness 3.5
—sampling port (F) diameter 18
—stopper in sampling port ISO24/25

J Metal L-profiled circular frame with slit


frame —inner diameter to fit collec-
tion plate
—height 4
Fig. 6.15-2 Apparatus 1: Details of jet tube and collection —thickness of horizontal section 0.5
plate —thickness of vertical section 2
K Wire Steel wire interconnecting metal
frame and sleeve (2 for each
frame)
—diameter 1

L Sleeve Metal sleeve secured on jet tube


by screw
—inner diameter to fit jet tube
—height 6
—thickness 5

M Gasket e.g. silicone to fit glass


cylinder

N Bolt Metal bolt with nut (6 pairs)


—length 205
—diameter 4
Fig. 6.15-3 Apparatus 1: Details of the filter stage (stage P O-ring Rubber O-ring
5) —diameter × thickness 66.34 × 2.62

Q O-ring Rubber O-ring


—diameter × thickness 29.1 × 1.6
onto the stage 1 inlet jet tube of the impinger. A rubber O-
R Filter Metal housing with stand and see Fig. 6.15-3
ring on the jet tube provides an airtight connection to the
holder outlet
induction port. A suitable mouthpiece adapter is used to
provide an airtight seal between the inhaler and the induc- S Filter Perforated sheet metal
support —diameter 65
tion port.
—hole diameter 3
5.1.1. Procedure for metered-dose inhalers
—distance between holes 4
Dispense 20 mL of a solvent, capable of dissolving the
(center-points)
active substance, into each of stages 1 to 4 and replace the
stoppers. Tilt the apparatus to wet the stoppers, thereby T Snap-locks
neutralizing electrostatic charge. Place a suitable filter capa- U Multi-jet Jet tube (H) ending in multi-jet see Fig. 6.15-2
ble of quantitatively collecting the active substance in stage tube arrangement
5 and assemble the apparatus. Place a suitable mouthpiece * Refers to Fig. 6.15-1.
adapter in position at the end of the induction port. The ** Measures in mm with tolerances according to JIS B 0405 unless
mouthpiece end of the inhaler, when inserted to the otherwise stated.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2654 General Tests, Processes and Apparatus Supplement I, JP XVII

Table 6.15-2 Dimensions(1) of jet tube with collection plate


of apparatus 1
Filter
Type Code(2) Stage 1 Stage 2 Stage 3 Stage 4
(Stage 5)

Distance 1 9.5 5.5 4.0 6.0 n.a.


(Length) (- .0 + .5) (- .0 + .5) (- .0 + .5) (- .0 + .5)
Distance 2 26 31 33 30.5 0
(Length)
Distance 3 8 5 5 5 5
(Length)
Distance 4 3 3 3 3 n.a.
(Length)
Distance 5 0 3 3 3 3
(Length)
Distance 6(3) 20 25 25 25 25
(Length)
Distance 7 n.a. n.a. n.a. 8.5 n.a.
(Length)

Diameter c 25 14 8.0 (± .1) 21 14


Diameter d 50 30 20 30 n.a.
Diameter e 27.9 16.5 10.5 23.9 n.a.
Diameter f 31.75 22 14 31 22
(- .0 + .5)
Diameter g 25.4 21 13 30 21
Diameter h n.a. n.a. n.a 2.70 (± .5) n.a.
Diameter l n.a. n.a. n.a 6.3 n.a.
Diameter k n.a. n.a. n.a 12.6 n.a.

Radius(4) r 16 22 27 28.5 0
Radius s 46 46 46 46 n.a.
Radius t n.a. 50 50 50 50

Angle w 109 539 539 539 539


Angle u n.a. n.a. n.a. 459 n.a.
Angle v n.a. n.a. n.a. 609 n.a.
Fig. 6.15-4 Induction port
(1) Measures in mm with tolerances according to JIS B 0405 unless
otherwise stated.
(2) Refer to Fig. 6.15-2
(3) Including gasket
mouthpiece adapter, lines up along the horizontal axis of
(4) Relative centerline of stage compartment
the induction port. The front face of the inhaler mouthpiece
n.a.: not applicable
must be flush with the front face of the induction port.
When attached to the mouthpiece adapter, the inhaler is
positioned in the same orientation as intended for use. Con-
nect a suitable vacuum pump to the outlet of the apparatus Dismantle the filter stage of the apparatus. Carefully
and adjust the air flow through the apparatus, as measured remove the filter and extract the active substance into an
at the inlet to the induction port, to 30 L per minute aliquot of the solvent. Remove the induction port and
(±5z). Switch off the pump. mouthpiece adapter from the apparatus and extract the ac-
Unless otherwise prescribed in the patient instructions, tive substance into an aliquot of the solvent. If necessary,
shake the inhaler for 5 seconds and discharge one delivery rinse the inside of the inlet jet tube to stage 1 with the sol-
to waste. Switch on the pump to the apparatus, locate the vent, allowing the solvent to flow into the stage. Extract the
mouthpiece end of the inhaler in the adapter and discharge active substance from the inner walls and the collection
the inhaler into the apparatus, actuating the inhaler for a plate of each of the 4 upper stages of the apparatus into the
sufficient time to ensure complete discharge. Wait for 5 sec- solution in the respective stage by carefully tilting and rotat-
onds before removing the assembled inhaler from the adap- ing the apparatus, observing that no liquid transfer occurs
ter. Repeat the procedure. The number of discharges should between the stages.
be minimized and typically would not be greater than 10. Using a suitable method of analysis, determine the
The number of discharges is sufficient to ensure an accurate amount of active substance contained in each of the ali-
and precise determination of the fine particle dose. After quots of solvent. Calculate the fine particle dose (see 6. Cal-
the final discharge, wait for 5 seconds and then switch off culations).
the pump.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2655

5.1.2. Procedure for dry powder inhalers for use.


Place a suitable low resistance filter capable of quantita- Prepare the dry powder inhaler for use according to the
tively collecting the active substance in stage 5 and assemble patient instructions. With the pump running and the 2-way
the apparatus. Connect the apparatus to a flow system ac- solenoid valve closed, locate the mouthpiece of the inhaler
cording to the scheme specified in Fig. 6.15-5 and Table in the mouthpiece adapter. Discharge the powder into the
6.15-3. Unless otherwise prescribed, conduct the test at the apparatus by opening the valve for the required time, T
flow rate, Qout, used in Uniformity of Delivered Dose for (±5z). Repeat the discharge procedure. The number of
Inhalations <6.14>, drawing 4 L of air from the mouthpiece discharges should be minimized and typically would not be
of the inhaler and through the apparatus. greater than 10. The number of discharges is sufficient to
Connect a flowmeter to the induction port. Use a flow- ensure an accurate and precise determination of fine particle
meter calibrated for the volumetric flow leaving the meter, dose.
or calculate the volumetric flow leaving the meter (Qout) Dismantle the filter stage of the apparatus. Carefully
using the ideal gas law. For a meter calibrated for the enter- remove the filter and extract the active substance into an
ing volumetric flow (Qin), use the following expression: aliquot of the solvent. Remove the induction port and
mouthpiece adapter from the apparatus and extract the ac-
Qin × P0
Qout = tive substance into an aliquot of the solvent. If necessary,
P0 - D P
rinse the inside of the inlet jet tube to stage 1 with the sol-
P0: atmospheric pressure vent, allowing the solvent to flow into the stage. Extract the
DP: pressure drop over the meter active substance from the inner walls and the collection
plate of each of the 4 upper stages of the apparatus into the
Adjust the flow control valve to achieve steady flow
solution in the respective stage by carefully tilting and rotat-
through the system at the required rate, Qout (±5z). En-
ing the apparatus, observing that no liquid transfer occurs
sure that critical flow occurs in the flow control valve by the
between the stages.
following procedure. Switch off the pump.
Using a suitable method of analysis, determine the
With the inhaler in place and the test flow rate estab-
amount of active substance contained in each of the ali-
lished, measure the absolute pressure on both sides of the
quots of solvent. Calculate the fine particle dose (see 6. Cal-
control valve (pressure reading points P2 and P3 in Fig.
culations).
6.15-5). A ratio P3/P2 of less than or equal to 0.5 indicates
5.2. Andersen cascade impactor method (Apparatus 2)
critical flow. Switch to a more powerful pump and re-meas-
The apparatus used for Andersen cascade impactor
ure the test flow rate if critical flow is not indicated.
Dispense 20 mL of a solvent, capable of dissolving the ac-
tive substance, into each of the 4 upper stages of the appa-
ratus and replace the stoppers. Tilt the apparatus to wet the Table 6.15-3 Component specification for Fig. 6.15-5
stoppers, thereby neutralizing electrostatic charge. Place a Code* Item Description
suitable mouthpiece adapter in position at the end of the
A Connector ID ≧ 8 mm, e.g., short metal coupling, with
induction port. The mouthpiece end of the inhaler, when low-diameter branch to P3.
inserted to the mouthpiece adapter, lines up along the
horizontal axis of the induction port. The front face of the B Vacuum A length of suitable tubing having an ID ≧ 8
tubing mm and an internal volume of 25 ± 5 mL.
inhaler mouthpiece must be flush with the front face of the
induction port. When attached to the mouthpiece adapter, C Two-way A 2-way, 2-port solenoid valve having a mini-
the inhaler is positioned in the same orientation as intended solenoid mum airflow resistance orifice with ID ≧ 8
valve mm and an opening time ≦ 100 ms.
D Vacuum Pump must be capable of drawing the required
pump flow rate through the assembled apparatus
with the inhaler in the mouthpiece adapter, or
equivalent. Connect the pump to the 2-way
solenoid valve using short and/or wide (ID ≧
10 mm) vacuum tubing and connectors to
minimize pump capacity requirements.

E Timer Timer capable to drive the 2-way solenoid


valve for the required duration, or equivalent.

P2, P3 Pressure Determine under steady-state flow condition


measure- with an absolute pressure transducer.
ments

F Flow con- Adjustable regulating valve with maximum Cv


trol valve ≧ 1.
Fig. 6.15-5 Experimental set-up for testing dry powder in- * Refer to Fig. 6.15-5.
halers ID: inner diameter

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2656 General Tests, Processes and Apparatus Supplement I, JP XVII

method (apparatus 2) is shown in Fig. 6.15-6. The appa- along the horizontal axis of the induction port. The front
ratus 2 consists of 8 stages together with a final filter. face of the inhaler mouthpiece must be flush with the front
Material of construction may be aluminium, stainless steel face of the induction port. When attached to the
or other suitable material. The stages are clamped together mouthpiece adapter, the inhaler unit is positioned in the
and sealed with O-rings. Critical dimensions of apparatus 2 same orientation as the intended use. Connect a suitable
are provided in Table 6.15-4. In use, some occlusions and pump to the outlet of the apparatus and adjust the air flow
wear of nozzles will occur. In-use mensuration tolerances through the apparatus, as measured at the inlet to the induc-
need to be justified. tion port, to 28.3 L per minute (±5z). Switch off the
The configuration used for metered-dose inhalers is pump.
shown in Fig. 6.15-6. The entry cone (see Fig. 6.15-7b) of Unless otherwise prescribed in the patient instructions,
the impactor is connected to an induction port (see Fig. shake the inhaler for 5 seconds and discharge one delivery
6.15-4). A suitable mouthpiece adapter is used to provide an to waste. Switch on the pump to the apparatus, locate the
airtight seal between the inhaler and the induction port. mouthpiece end of the inhaler in the adapter and discharge
In the configuration for dry powder inhalers, a pre-sepa- the inhaler into the apparatus, actuating the inhaler for a
rator is placed above the top stage to collect large masses of sufficient time to ensure complete discharge. Wait for 5 sec-
non-respirable powder. The top of the pre-separator shown onds before removing the assembled inhaler from the adap-
in Fig. 6.15-7a is used to adapt the pre-separator to the in- ter. Repeat the procedure. The number of discharges should
duction port. To accommodate high flow rates through the be minimized and typically would not be greater than 10.
impactor, the outlet nipple, used to connect the impactor to The number of discharges is sufficient to ensure an accurate
the vacuum system, is enlarged to have an internal diameter
greater than or equal to 8 mm.
5.2.1. Procedure for metered-dose inhalers
Table 6.15-4 Critical dimensions for apparatus 2
Assemble the Andersen cascade impactor with a suitable
filter in place. Ensure that the system is airtight by a suita- Number Dimension
Description
ble method. Place a suitable mouthpiece adapter in position of nozzle (mm)
at the end of the induction port. The mouthpiece end of the
Stage 0 nozzle diameter 96 2.55 ± 0.025
inhaler, when inserted to the mouthpiece adapter, lines up
Stage 1 nozzle diameter 96 1.89 ± 0.025
Stage 2 nozzle diameter 400 0.914 ± 0.0127
Stage 3 nozzle diameter 400 0.711 ± 0.0127
Stage 4 nozzle diameter 400 0.533 ± 0.0127
Stage 5 nozzle diameter 400 0.343 ± 0.0127
Stage 6 nozzle diameter 400 0.254 ± 0.0127
Stage 7 nozzle diameter 201 0.254 ± 0.0127

Fig. 6.15-6 Andersen cascade impactor used for metered- Fig. 6.15-7a Expanded view of top for the Andersen pre-
dose inhalers (Apparatus 2) separator adapted to the induction port

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2657

or calculate the volumetric flow leaving the meter (Qout)


using the ideal gas law. For a meter calibrated for the enter-
ing volumetric flow (Qin), use the following expression:
Qin × P0
Qout =
P0 - D P
P0: atmospheric pressure
DP: pressure drop over the meter

Adjust the flow control valve to achieve steady flow


through the system at the required rate, Qout (±5z). En-
sure that critical flow occurs in the flow control valve by the
procedure described in 5.1.2. Procedure for dry powder in-
halers. Switch off the pump.
Place a suitable mouthpiece adapter in position at the end
of the induction port. The mouthpiece end of the inhaler,
when inserted to the mouthpiece adapter, lines up along the
Fig. 6.15-7b Expanded view of the entry cone for mount- horizontal axis of the induction port. The front face of the
ing induction port on the Andersen cascade impactor inhaler mouthpiece must be flush with the front face of the
without pre-separator. induction port. When attached to the mouthpiece adapter,
the inhaler is positioned in the same orientation as intended
for use.
Prepare the dry powder inhaler for use according to the
and precise determination of the fine particle dose. After
patient instructions. With the pump running and the 2-way
the final discharge, wait for 5 seconds and then switch off
solenoid valve closed, locate the mouthpiece of the inhaler
the pump.
in the mouthpiece adapter. Discharge the powder into the
Dismantle the apparatus. Carefully remove the filter and
apparatus by opening the valve for the required time, T
extract the active substance into an aliquot of the solvent.
(±5z). Repeat the discharge sequence. The number of dis-
Remove the induction port and mouthpiece adapter from
charges should be minimized and typically would not be
the apparatus and extract the active substance into an ali-
greater than 10. The number of discharges is sufficient to
quot of the solvent. Extract the active substance from the
ensure an accurate and precise determination of fine particle
inner walls and the collection plate of each of the stages of
dose.
the apparatus into aliquots of the solvent.
Dismantle the apparatus. Carefully remove the filter and
Using a suitable method of analysis, determine the
extract the active substance into an aliquot of the solvent.
amount of active substance contained in each of the ali-
Remove the pre-separator, induction port and mouthpiece
quots of solvent. Calculate the fine particle dose (see 6. Cal-
adapter from the apparatus and extract the active substance
culations).
into an aliquot of the solvent. Extract the active substance
5.2.2. Procedure for dry powder inhalers
from the inner walls and the collection plate of each of the
The aerodynamic cut-off diameters of the individual
stages of the apparatus into aliquots of the solvent.
stages of this apparatus are currently not well-established at
Using a suitable method of analysis, determine the
flow rates other than 28.3 L per minute. Users must justify
amount of active substance contained in each of the
and validate the use of the impactor in the chosen condi-
aliquots of solvent. Calculate the fine particle dose (see 6.
tions, when flow rates different from 28.3 L per minute are
Calculations).
selected.
5.3. Next generation impactor method (Apparatus 3)
Assemble the Andersen cascade impactor with the pre-
The apparatus used for next generation impactor method
separator and a suitable filter in place and ensure that the
(apparatus 3) is shown in Fig. 6.15-8. The apparatus 3 is a
system is airtight. Depending on the product characteristics,
cascade impactor with 7 stages and a micro-orifice collector
the pre-separator may be omitted, where justified. Stages 6
(MOC). Over the flow rate range of 30 to 100 L per minute
and 7 may also be omitted at high flow rates, if justified.
the 50z-collection efficiency cut-off diameters (D50 values)
The pre-separator may be coated in the same way as the col-
range between 0.24 mm and 11.7 mm, evenly spaced on a
lection plates or may contain 10 mL of a suitable solvent.
logarithmic scale. In this flow range, there are always at
Connect the apparatus to a flow system according to the
least 5 stages with D50 values between 0.5 mm and 6.5 mm.
scheme specified in Fig. 6.15-5 and Table 6.15-3.
The collection efficiency curves for each stage are sharp and
Unless otherwise prescribed, conduct the test at the flow
minimize overlap between stages.
rate, Qout, used in Uniformity of Delivered Dose for Inhal-
Material of construction may be aluminium, stainless
ers <6.14> drawing 4 L of air from the mouthpiece of the in-
steel or other suitable material.
haler and through the apparatus.
The impactor configuration has removable impaction
Connect a flowmeter to the induction port. Use a flow-
cups with all the cups in one plane (Figures 6.15-8 to
meter calibrated for the volumetric flow leaving the meter,

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2658 General Tests, Processes and Apparatus Supplement I, JP XVII

Table 6.15-5 Critical dimensions for apparatus 3

Description Dimension (mm)

Pre-separator 12.8 ± 0.05


(dimension a—see Fig. 6.15-12)

Stage 1* nozzle diameter 14.3 ± 0.05

Stage 2* nozzle diameter 4.88 ± 0.04

Stage 3* nozzle diameter 2.185 ± 0.02

Stage 4* nozzle diameter 1.207 ± 0.01

Stage 5* nozzle diameter 0.608 ± 0.01

Stage 6* nozzle diameter 0.323 ± 0.01

Stage 7* nozzle diameter 0.206 ± 0.01

MOC* aprox. 0.070

Cup depth 14.625 ± 0.10


(dimension b—see Fig. 6.15-11)

Collection cup surface roughness (Ra) 0.5 to 2 mm


Fig. 6.15-8 Next generation impactor (shown with the pre-
Stage 1 nozzle to seal body distance** 0 ± 1.18
separator in place) (Apparatus 3)
—dimension c

Stage 2 nozzle to seal body distance** 5.236 ± 0.736


—dimension c

Stage 3 nozzle to seal body distance** 8.445 ± 0.410


—dimension c

Stage 4 nozzle to seal body distance** 11.379 ± 0.237


—dimension c

Stage 5 nozzle to seal body distance** 13.176 ± 0.341


—dimension c

Stage 6 nozzle to seal body distance** 13.999 ± 0.071


—dimension c

Stage 7 nozzle to seal body distance** 14.000 ± 0.071


—dimension c
Fig. 6.15-9 Apparatus 3 showing component parts MOC nozzle to seal body distance** 14.429 to 14.571
—dimension c
* See Fig. 6.15-10
6.15-11). There are three main sections to the impactor; the
** See Fig. 6.15-11
bottom frame that holds the impaction cups, the seal body
that holds the jets and the lid that contains the interstage
passageways (Figures 6.15-8 and 6.15-9). Multiple nozzles
are used at all but the first stage (Fig. 6.15-10). The flow inhalers, and connects between the induction port and the
passes through the impactor in a saw-tooth pattern. impactor. A suitable mouthpiece adapter is used to provide
Critical dimensions are provided in Table 6.15-5. an airtight seal between the inhaler and the induction port.
In routine operation, the seal body and lid are held Apparatus 3 contains a terminal MOC that for most
together as a single assembly. The impaction cups are acces- preparations will eliminate the need for a final filter as de-
sible when this assembly is opened at the end of an inhaler termined by method validation. The MOC is a collection
test. The cups are held in a support tray, so that all cups can plate with nominally 4032 holes, each approximately 70 mm
be removed from the impactor simultaneously by lifting out in diameter. Most particles not captured on stage 7 of the
the tray. impactor will be captured on the cup surface below the
An induction port with internal dimensions defined in MOC. For impactors operated at 60 L per minute, the MOC
Fig. 6.15-4 connects to the impactor inlet. A pre-separator is capable of collecting 80z of 0.14 mm particles. For
can be added when required, typically with dry powder preparations with a significant fraction of particles not cap-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2659

Fig. 6.15-10 Apparatus 3: nozzle configuration

Fig. 6.15-12 Apparatus 3: pre-separator configuration

and then switch off the pump.


Dismantle the apparatus and recover the active substance
as follows. Remove the induction port and mouthpiece
adapter from the apparatus and extract the deposited active
Fig. 6.15-11 Apparatus 3: configuration of interstage pas- substance into an aliquot of the solvent. Open the impactor
sageways by releasing the handle and lifting the lid. Remove the cup
tray, with the collection cups, and extract the active sub-
stance in each cup into an aliquot of the solvent.
tured by the MOC, there is an optional filter holder that can Using a suitable method of analysis, determine the
replace the MOC or be placed downstream of the MOC (a amount of active substance contained in each of the ali-
glass fiber filter is suitable). quots of solvent. Calculate the fine particle dose (see 6. Cal-
5.3.1. Procedure for metered-dose inhalers culations).
Place cups into the apertures in the cup tray. Insert the 5.3.2. Procedure for dry powder inhalers
cup tray into the bottom frame, and lower into place. Close Assemble the apparatus with the pre-separator (Fig.
the impactor lid with the seal body attached and operate the 6.15-12). Depending on the product characteristics, the pre-
handle to lock the impactor together so that the system is separator may be omitted, where justified.
airtight. Place cups into the apertures in the cup tray. Insert the
Connect an induction port with internal dimensions de- cup tray into the bottom frame, and lower into place. Close
fined in Fig. 6.15-4 to the impactor inlet. Place a suitable the impactor lid with the seal body attached and operate the
mouthpiece adapter in position at the end of the induction handle to lock the impactor together so that the system is
port. The mouthpiece end of the inhaler to the mouthpiece airtight.
adapter, when inserted, lines up along the horizontal axis of When used, the pre-separator should be assembled as
the induction port. The front face of the inhaler mouthpiece follows. Assemble the pre-separator insert into the pre-sepa-
must be flush with the front face of the induction port. rator base. Fit the pre-separator base to the impactor inlet.
When attached to the mouthpiece adapter, the inhaler is Add 15 mL of the solvent used for active substance recovery
positioned in the same orientation as intended for use. Con- to the central cup of the pre-separator insert. Place the pre-
nect a suitable pump to the outlet of the apparatus and separator body on top of this assembly and close the two
adjust the air flow through the apparatus, as measured at catches.
the inlet to the induction port, to 30 L per minute (±5z). Connect an induction port with internal dimensions de-
Switch off the pump. fined in Fig. 6.15-4 to the impactor inlet or pre-separator
Unless otherwise prescribed in the patient instructions, inlet. Connect the apparatus to a flow system according to
shake the inhaler for 5 seconds and discharge one delivery the scheme specified in Fig. 6.15-5 and Table 6.15-3.
to waste. Switch on the pump to the apparatus, locate the Unless otherwise prescribed, conduct the test at the flow
mouthpiece end of the inhaler in the adapter and discharge rate, Qout, used in Uniformity of Delivered Dose for Inhala-
the inhaler into the apparatus, actuating the inhaler for a tions <6.14> drawing 4 L of air from the mouthpiece of the
sufficient time to ensure a complete discharge. inhaler and through the apparatus. Connect a flowmeter to
Wait for 5 seconds before removing the assembled inhaler the induction port. Use a flowmeter calibrated for the volu-
from the adapter. Repeat the procedure. The number of dis- metric flow leaving the meter, or calculate the volumetric
charges should be minimized, and typically would not be flow leaving the meter (Qout) using the ideal gas law. For a
greater than 10. The number of discharges is sufficient to meter calibrated for the entering volumetric flow (Qin), use
ensure an accurate and precise determination of the fine the following expression:
particle dose. After the final discharge, wait for 5 seconds

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2660 General Tests, Processes and Apparatus Supplement I, JP XVII

Qin × P0 Table 6.15-6 Calculations for Apparatus 1


Qout =
P0 - D P Cut-off Mass of active sub- Cumulative mass Cumulative fraction
P0: atmospheric pressure diameter stance deposited on of active substance of active substance
DP: pressure drop over the meter ( mm) stage per discharge per discharge (z)
d4 = 1.7 × q Mass from filter c4 = m 5 f4 = (c4/c) × 100
Adjust the flow control valve to achieve steady flow
stage (m5*)
through the system at the required rate, Qout (±5z). En-
sure that critical flow occurs in the flow control valve by the d3 = 3.1 × q Mass from stage 4 c3 = c4 + m4 f3 = (c3/c) × 100
procedure described in 5.1.2. Procedure for dry powder in- ( m 4)
halers. Switch off the pump. d2 = 6.8 × q Mass from stage 3 c2 = c3 + m3 f2 = (c2/c) × 100
Place a suitable mouthpiece adapter in position at the end ( m 3)
of the induction port. The mouthpiece end of the inhaler, Mass from stage 2 c = c2 + m2 100
when inserted to the mouthpiece adapter, lines up along the ( m 2)
horizontal axis of the induction port. The front face of the
* Stage 5 is the filter stage.
inhaler mouthpiece must be flush with the front face of the
q = (60/Q), Q: the test flow rate in L per minute (Qout for dry
induction port. When attached to the mouthpiece adapter,
powder inhalers).
the inhaler is positioned in the same orientation as intended
for use.
Prepare the dry powder inhaler for use according to the Table 6.15-7 Calculations for Apparatus 2 when used at a
patient instructions. With the pump running and the 2-way flow rate of 28.3 L per minute
solenoid valve closed, locate the mouthpiece of the inhaler
Cut-off Mass of active sub- Cumulative mass Cumulative fraction
in the mouthpiece adapter. Discharge the powder into the
diameter stance deposited on of active substance of active substance
apparatus by opening the valve for the required time, T
( mm) stage per discharge per discharge (z)
(±5z). Repeat the discharge sequence. The number of dis-
charges should be minimized and typically would not be d7 = 0.4 Mass from filter c7 = m 8 f7 = (c7/c) × 100
stage (m8)
greater than 10. The number of discharges is sufficient to
ensure an accurate and precise determination of fine particle d6 = 0.7 Mass from stage 7 c6 = c7 + m7 f6 = (c6/c) × 100
dose. ( m 7)
Dismantle the apparatus and recover the active substance d5 = 1.1 Mass from stage 6 c5 = c6 + m6 f5 = (c5/c) × 100
as follows. ( m 6)
Remove the induction port and mouthpiece adapter from
d4 = 2.1 Mass from stage 5 c4 = c5 + m5 f4 = (c4/c) × 100
the pre-separator, when used, and extract the deposited ac-
( m 5)
tive substance into an aliquot of the solvent. When used, re-
move the pre-separator from the impactor, being careful to d3 = 3.3 Mass from stage 4 c3 = c4 + m4 f3 = (c3/c) × 100
( m 4)
avoid spilling the cup liquid into the impactor. Recover the
active substance from the pre-separator. d2 = 4.7 Mass from stage 3 c2 = c3 + m3 f2 = (c2/c) × 100
Open the impactor by releasing the handle and lifting the ( m 3)
lid. Remove the cup tray, with the collection cups, and ex- d1 = 5.8 Mass from stage 2 c1 = c2 + m2 f1 = (c1/c) × 100
tract the active substance in each cup into an aliquot of the ( m 2)
solvent.
d0 = 9.0 Mass from stage 1 c0 = c1 + m1 f0 = (c0/c) × 100
Using a suitable method of analysis, determine the
( m 1)
amount of active substance contained in each of the ali-
quots of solvent. Calculate the fine particle dose (see 6. Cal- Mass from stage 0 c = c0 + m0 100
( m 0)
culations).

6. Calculations
From the analysis of the solutions, calculate the mass of
culate the FPD as the mass of the active substance deposited
active substance deposited on each stage per discharge and
on the stages corresponding to the cut-off diameter of 5 mm
the mass of active substance per discharge deposited in the
and less.
induction port, mouthpiece adapter and when used, the pre-
If necessary, and where appropriate (e.g., where there is a
separator.
log-normal distribution), determine values for the Mass
Starting at the collection site (filter or MOC) close to the
Median Aerodynamic Diameter (MMAD) and Geometric
airflow outlet of the apparatus, derive a table of cumulative
Standard Deviation (GSD) from the cumulative fraction of
mass versus cut-off diameter of the respective stage (see
active substance versus cut-off diameter (see Tables 6.15-6
Table 6.15-6 for Apparatus 1, Table 6.15-7 for Apparatus
to 6.15-8). Appropriate computational methods may also be
2, Table 6.15-8 for Apparatus 3). Calculate the Fine Parti-
used.
cle Dose (FPD) by interpolation of the mass of the active
substance less than or equal to 5 mm. Or it is possible to cal-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2661

Table 6.15-8 Calculations for Apparatus 3 Insulin Aspart RS


Mass of active Cumulative Pazufloxacin Mesilate RS
Cut-off Cumulative
substance depos- mass of active
diameter x fraction of active Pyridoxal Phosphate RS
ited on stage per substance per
( mm) substance (z)
discharge discharge
Saccharin Sodium RS for Identification
d7 = 0.34 × q 0.67 Mass from MOC c7 = m8 F7 = (c7/c) × 100
Zonisamide RS
or terminal filter
(m8)

d6 = 0.55 × q 0.60 Mass from stage c6 = c7 + m7 F6 = (c6/c) × 100


Delete the following under section (1):
7 (m7)
Aceglutamide RS
d5 = 0.94 × q 0.53 Mass from stage c5 = c6 + m6 F5 = (c5/c) × 100
6 (m6) Diclofenamide RS
d4 = 1.66 × q 0.47 Mass from stage c4 = c5 + m5 F4 = (c4/c) × 100
Digitoxin RS
5 (m5)
Fluoxymesterone RS
d3 = 2.82 × q 0.50 Mass from stage c3 = c4 + m4 F3 = (c3/c) × 100
4 (m4) Lanatoside C RS
d2 = 4.46 × q 0.52 Mass from stage c2 = c3 + m3 F2 = (c2/c) × 100
Tolazamide RS
3 (m3)

d1 = 8.06 × q 0.54 Mass from stage c1 = c2 + m2 F1 = (c1/c) × 100


2 (m2) Delete the following under section (2):
Mass from stage c = c1 + m1 100
Gramicidin RS
1 (m1)
Rokitamycin RS
q = (60/Q) x, Q: the test flow rate in L per minute, x: listed in the table
Zinostatin Stimalamer RS

9.01 Reference Standards 9.21 Standard Solutions for


Volumetric Analysis
Change the following under section (1) as
follows: Add the following:
Adrenaline Bitartrate RS for Purity Benzethonium chloride, 0.004 mol/L
1000 mL of this solution contains 1.7923 g of benzethoni-
p-Aminobenzoyl Glutamic Acid RS for Purity
um chloride (C27H42ClNO2: 448.08).
Anhydrous Lactose RS for Identification Preparation—Dissolve 1.792 g of benzethonium chloride
for assay, previously dried at 1059C for 4 hours, in water to
Cellacefate RS for Identification
make exactly 1000 mL, and standardize the solution as
Gitoxin RS for Purity follows:
Standardization—Pipet 10 mL of the prepared benzetho-
Heparin Sodium RS for Identification
nium chloride solution, adjust to pH between 2.6 and 3.4 by
Lactose RS for Identification adding dropwise diluted dilute hydrochloric acid (1 in 2),
add 1 drop of methyl orange TS, and titrate <2.50> with 0.02
Over-sulfated Chondroitin Sulfate RS for System
mol/L sodium tetraphenylboron VS until the color of the
Suitability
solution becomes red. Calculate the molarity factor.
Povidone RS for Identification
Each mL of 0.02 mol/L sodium tetraphenylboron VS
Tyrosine RS for Digestion Test = 8.962 mg of C27H42ClNO2

Zinc sulfate, 0.05 mol/L


1000 mL of this solution contains 14.378 g of zinc sulfate
Add the following to section (1):
heptahydrate (ZnSO4.7H2O: 287.55).
Entacapone RS Preparation—Before use, dilute 0.1 mol/L zinc sulfate
VS with water to make exactly twice the initial volume.
Entacapone Related Substance A RS for System Suitabil-
ity

Glucose RS

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2662 General Tests, Processes and Apparatus Supplement I, JP XVII

[Same as the monograph Clomipramine Hydrochloride.


9.41 Reagents, Test Solutions When dried, it contains not less than 99.0z of clomipra-
mine hydrochloride (C19H23ClN2.HCl).]

Add the following: Clotiazepam for assay C16H15ClN2OS [Same as the


monograph Clotiazepam. When dried, it contains not less
Aniline sulfate (C6H5NH2)2.H2SO4 A white to grayish
than 99.0z of clotiazepam (C16H15ClN2OS).]
white crystalline powder.
Purity Clarity and color of solution—Dissolve 1.0 g of Deoxycholic acid for thin-layer chromatography
aniline sulfate in 50 mL of water: the solution is clear and C24H40O4 A white powder. Soluble in methanol and in
colorless. ethanol (99.5), and practically insoluble in water. Melting
point: about 1759C (with decomposition).
3-Aminobenzoic acid C7H7NO2 White crystals.
Identification—Determine the infrared absorption spec-
Melting point <2.60>: About 1749C
trum of deoxycholic acid for thin-layer chromatography as
2-Aminophenol C6H7NO Pale yellow-brown crystals. directed in the potassium bromide disk method under In-
Soluble in ethanol (99.5), and sparingly soluble in water. frared Spectrophotometry <2.25>: it exhibits absorption at
Melting point <2.60>: About 1729C the wave numbers of about 2930 cm-1, 1716 cm-1, 1447
cm-1 and 1042 cm-1.
4-Aminophenol C6H7NO A white to yellowish white
Purity Related substances—Dissolve 20 mg of deoxy-
crystalline powder. Soluble in ethanol (99.5), and sparingly
cholic acid for thin-layer chromatography in 10 mL of
soluble in water.
methanol, and use this solution as the sample solution.
Melting point <2.60>: About 1869C
Pipet 1 mL of the sample solution, add methanol to make
Azosemide for assay C12H11ClN6O2S2 [Same as the exactly 100 mL, and use this solution as the standard solu-
monograph Azosemide] tion. Perform the test with these solutions as directed under
Thin-Layer Chromatography <2.03>. Perform the test with
Baicalein for resolution check C15H10O5 Yellow, crys-
5 mL each of the sample solution and standard solution as
tals or crystalline powder. Slightly soluble in methanol and
directed in the Identification under Oriental Bezoar: the
in ethanol (99.5), and practically insoluble in water.
spots other than the principal spot at an Rf value of about
Identification—Determine the absorption spectrum of a
0.5 obtained from the sample solution are not more intense
solution of baicalein for resolution check in methanol (1 in
than the spot from the standard solution.
200,000) as directed under Ultraviolet-visible Spectropho-
tometry <2.24>: it exhibits maxima between 213 nm and 217 Dimidium bromide C20H18BrN3 Red to dark brown,
nm, between 273 nm and 277 nm, and between 321 nm and crystalline powder or powder.
325 nm. Identification—(1) Determine the infrared absorption
Purity Related substances—Dissolve 1 mg of baicalein spectrum of dimidium bromide as directed in the potassium
for resolution check in 50 mL of methanol, and use this so- bromide disk method under Infrared Spectrophotometry
lution as the sample solution. Perform the test with 10 mL <2.25>: it exhibits absorption at the wave numbers of about
of the sample solution as directed under Liquid Chromatog- 3300 cm-1, 1619 cm-1, 1489 cm-1, 1470 cm-1, 1422 cm-1
raphy <2.01> according to the following conditions: the total and 1316 cm-1.
area of the peaks other than baicalein from the sample solu- (2) A solution of dimidium bromide (1 in 1000) re-
tion is not larger than 1/10 times the total area of all peaks sponds to Qualitative Tests <1.09> (1) for bromide.
other than the solvent peak.
Dimidium bromide-patent blue TS Dissolve each 0.5 g
Operating conditions
of dimidium bromide and 0.25 g of patent blue in 30 mL of
Detector, column, column temperature, mobile phase,
a warmed mixture of water and ethanol (99.5) (9:1), com-
and flow rate: Proceed as directed in the operating condi-
bine the solutions, and add a mixture of water and ethanol
tions in the Assay (4) i) under Saikokeishito Extract.
(99.5) (9:1) to make 250 mL. To 20 mL of this solution add
Time span of measurement: About 2 times as long as the
270 mL of diluted sulfuric acid (7 in 675) and water to make
retention time of baicalein.
500 mL.
System Suitability
Storage—Preserve in light-resistant containers.
System performance: When the procedure is run with 10
mL of the sample solution under the above operating condi- Gentisic acid C7H6O4 Light yellow crystals.
tions, the number of theoretical plates and the symmetry Melting point <2.60>: About 2009C
factor of the peak of baicalein are not less than 5000 and
Imidazole hydrobromide C3H4N2.HBr White to pale
not more than 1.5, respectively.
yellow crystals. Melting point: about 2219C.
System repeatability: When the test is repeated 6 times
with 10 mL of the sample solution under the above operating Irbesartan for assay C25H28N6O [Same as the mono-
conditions, the relative standard deviation of the peak area graph Irbesartan]
of baicalein is not more than 1.5z.
Mesalazine for assay C7H7NO3 [Same as the mono-
Clomipramine hydrochloride for assay C19H23ClN2.HCl graph Mesalazine. When dried, it contains not less than

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2663

99.0z of mesalazine (C7H7NO3).] reaction reservoir, adjust the pH of the solution to 8.00 by
adding dropwise 0.1 mol/L sodium hydroxide VS while stir-
Patent blue C27H31N2NaO6S2 Red-purple-brown to
ring and passing a current of nitrogen, add exactly 1 mL of
dark red-brown, crystalline powder to powder, or masses.
the test solution I previously allowed to stand at 25 ± 0.19C
Identification—(1) To 5 mg of patent blue add 20 mL
for 10 minutes, then immediately add dropwise 0.1 mol/L
of ethanol (99.5): a dark blue color develops.
sodium hydroxide VS by a 50-mL micropipet (minimum
(2) Determine the infrared absorption spectrum of pa-
graduation of 1 mL), while stirring, to keep the reaction
tent blue as directed in the potassium bromide disk method
solution at pH 8.00, and read the amount of 0.1 mol/L
under Infrared Spectrophotometry <2.25>: it exhibits ab-
sodium hydroxide VS consumed and the reaction time when
sorption at the wave numbers of about 1580 cm-1, 1420
the pH reached 8.00. Continue this procedure up to 6
cm-1, 1340 cm-1, 1180 cm-1, 1150 cm-1, 1070 cm-1, 1030
minutes. Separately, pipet 2 mL of the trypsin solution and
cm-1, 910 cm-1, 790 cm-1, 700 cm-1 and 620 cm-1.
1 mL of the sample solution, add sodium tetraborate-
Phthalate buffer solution (pH 5.8) Dissolve 100.0 g of calcium chloride buffer solution (pH 8.0) to make exactly
potassium hydrogen phthalate in about 800 mL of water, 10 mL, and use this solution as the test solution II. Transfer
adjust to pH 5.8 with a solution of sodium hydroxide (1 in 10.0 mL of the substrate solution to the reaction reservoir,
2), and add water to make 1000 mL. adjust the pH of the solution to 8.00, while stirring and
passing a current of nitrogen, add exactly 1 mL of the test
0.01 mol/L sodium dihydrogen phosphate TS (pH 7.5)
solution II, previously allowed to stand at 25 ± 0.19 C for
Dissolve 1.56 g of sodium dihydrogen phosphate dihydrate
10 minutes, and proceed in the same manner. Separately,
in 900 mL of water, adjust to pH 7.5 with sodium hydrox-
transfer 10.0 mL of the substrate solution to the reaction
ide TS, and add water to make 1000 mL.
reservoir, adjust the pH of the solution to 8.00, while stir-
0.2 mol/L Tris buffer solution (pH 8.1) Dissolve 24.2 g ring and passing a current of nitrogen, add 1.0 mL of so-
of 2-amino-2-hydroxymethyl-1,3-propanediol in water to dium tetraborate-calcium chloride buffer solution (pH 8.0),
make 1000 mL, and adjust to pH 8.1 with hydrochloric previously allowed to stand at 25 ± 0.19C for 10 minutes,
acid. and perform a blank determination in the same manner.
(v) Calculation: Plot the amount of consumption (mL) of
Voriconazole C16H14F3N5O [Same as the namesake
0.1 mol/L sodium hydroxide VS against the reaction time
monograph]
(minutes), select linear reaction times, t1 and t2, designate
the corresponding consumption amount of 0.1 mol/L so-
Change the following as follows:
dium hydroxide VS as v1 and v2, respectively, and designate
Aprotinin A clear and colorless liquid containing mmol of sodium hydroxide consumed per minute as D.
aprotinin extracted from the lung or parotid gland of a
v2 - v1 1
healthy cattle. The pH is between 5.0 and 7.0. D (mmol NaOH/minute) = ×
t2 - t1 10
Content: not less than 15,000 KIE Units and not more
than 25,000 KIE Units of aprotinin per mL. Assay—(i) KIE Units per mL of aprotinin
Trypsin solution: Weigh an amount of crystalline trypsin 2 (DA - D0) - (DB - D0)
= × n × 32.5
equivalent to about 250 FIP Units of trypsin according to L
the labeled FIP Units, and dissolve in 0.001 mol/L hydro-
L: Amount (mL) of the sample solution added to the test
chloric acid TS to make exactly 10 mL. Prepare before use,
solution II
and preserve in ice.
n: Dilution coefficient of aprotinin
(ii) Sample solution: To a suitable quantity of aprotinin add
DA: mmol of sodium hydroxide consumed in 1 minute
sodium tetraborate-calcium chloride buffer solution (pH
when the test solution I is used
8.0) so that each mL contains 800 KIE Units of aprotinin,
DB: mmol of sodium hydroxide consumed in 1 minute
and use this solution as the sample solution.
when the test solution II is used
(iii) Apparatus: Use a glass bottle as a reaction reservoir, 20
D0: mmol of sodium hydroxide consumed in 1 minute
mm in inside diameter and 50 mm in height, stopper with a
when the solution for blank determination is used
rubber stopper equipped with a glass/silver-silver chloride
32.5: Equivalent coefficient for calculation of KIE Units
electrode for pH determination, a nitrogen-induction tube
from FIP Units
and an exhaust port. Fix the reaction reservoir in a ther-
mostat, and keep the temperature of the bath at 25 ± 0.19C One KIE Unit means an amount of aprotinin making a
by means of a precise thermoregulator. reduction of 50z off the potency of 2 Units of kallidino-
(iv) Procedure: To 5.0 mL of N-a-benzoyl-L-arginine ethyl genase at pH 8.0 and room temperature for 2 hours.
ester TS add 45.0 mL of sodium tetraborate-calcium chlo- Storage—Preserve in a light-resistant, hermetic contain-
ride buffer solution (pH 8.0), and use this solution as the ers and in a cold place.
substrate solution. Pipet 1 mL of the trypsin solution, add
Crystalline trypsin To trypsin obtained from bovine
sodium tetraborate-calcium chloride buffer solution (pH
pancreas add an appropriate amount of trichloroacetic acid
8.0) to make exactly 10 mL, and use this solution as the test
to precipitate the trypsin, and recrystallize in ethanol (95).
solution I. Transfer 10.0 mL of the substrate solution to the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2664 General Tests, Processes and Apparatus Supplement I, JP XVII

White to yellow-white, crystals or powder. It is odorless. under the conditions described in the Assay.
Freely soluble in water and in sodium tetraborate-calcium Storage—Preserve in a cold place.
chloride buffer solution (pH 8.0).
[6]-Gingerol for assay C17H26O4 [6]-Gingerol for thin-
Content: not less than 45 FIP Units of trypsin per mg.
layer chromatography. It meets the requirements of the fol-
Assay-(i) Sample solution: Weigh accurately an appropri-
lowing 1) [6]-Gingerol for assay 1 or 2) [6]-Gingerol for
ate amount of crystalline trypsin, dissolve in 0.001 mol/L
assay 2 (Purity value by quantitative NMR). The latter is
hydrochloric acid TS to prepare a solution containing 50
used with correction for its amount based on the result ob-
FIP Units per mL, and use this solution as the sample solu-
tained in the Assay.
tion. Prepare before use, and preserve in ice.
1) [6]-Gingerol for assay 1
(ii) Apparatus: Use a glass bottle as a reaction reservoir, 20 z
Absorbance <2.24> E 11cm (281 nm): 101 – 112 [7 mg,
mm in inside diameter and 50 mm in height, stopper with a
ethanol (99.5), 200 mL].
rubber stopper equipped with a glass/silver-silver chloride
Purity Related substances—Dissolve 5 mg of [6]-gin-
electrode for pH determination, a nitrogen-induction tube
gerol for assay 1 in 5 mL of methanol, and use this solution
and an exhaust port. Fix the reaction reservoir in a ther-
as the sample solution. Pipet 1 mL of the sample solution,
mostat, and keep the temperature of the both at 25 ± 0.19 C
add methanol to make exactly 50 mL, and use this solution
by means of a precise thermoregulator.
as the standard solution. Perform the test with exactly 10
(iii) Procedure: Pipet 1.0 mL of N-a-benzoyl-L-arginine
mL each of the sample solution and standard solution as di-
ethyl ester TS, transfer to the reaction reservoir, and add
rected under Liquid Chromatography <2.01> according to
9.0 mL of sodium tetraborate-calcium chloride buffer solu-
the following conditions, and determine each peak area by
tion (pH 8.0). Allow to stand in the thermostat for 10
the automatic integration method: the total area of the
minutes to make the temperature of the contents reach to 25
peaks other than [6]-gingerol from the sample solution is
± 0.19 C, adjust the pH of the solution to 8.00 by adding
not larger than the peak area of [6]-gingerol from the stand-
dropwise 0.1 mol/L sodium hydroxide VS while stirring and
ard solution.
passing a current of nitrogen, add exactly 0.05 mL of the
Operating conditions
sample solution previously allowed to stand at 25 ± 0.19C,
Detector, column, column temperature, mobile phase and
then immediately add dropwise 0.1 mol/L sodium hydrox-
flow rate: Proceed as directed in the operating conditions in
ide VS by a 50 mL-micropipet (minimum graduation of 1
the Assay (3) under Hangekobokuto Extract.
mL) while stirring, to keep the reaction solution at pH 8.00,
Time span of measurement: About 6 times as long as the
and read the amount of 0.1 mol/L sodium hydroxide VS
retention time of [6]-gingerol.
consumed and the reaction time when the pH reached 8.00.
System suitability
Continue this procedure up to 8 minutes. Separately, trans-
System performance: Proceed as directed in the system
fer 10 mL of sodium tetraborate-calcium chloride buffer so-
suitability in the Assay (3) under Hangekobokuto Extract.
lution (pH 8.0), and perform a blank determination in the
Test for required detectability: Pipet 1 mL of the stand-
same manner.
ard solution, and add methanol to make exactly 20 mL.
(iv) Calculation: Plot the amount of consumption (mL) of
Confirm that the peak area of [6]-gingerol obtained with 10
0.1 mol/L sodium hydroxide VS against the reaction time
mL of this solution is equivalent to 3.5 to 6.5z of that with
(minutes), select linear reaction times, t1 and t2, designate
10 mL of the standard solution.
the corresponding consumption amount of 0.1 mol/L so-
System repeatability: When the test is repeated 6 times
dium hydroxide VS as v1 and v2, respectively, and designate
with 10 mL of the standard solution under the above operat-
mmol of sodium hydroxide consumed per minute as D (FIP
ing conditions, the relative standard deviation of the peak
Unit).
area of [6]-gingerol is not more than 1.5z.
v2 - v1 1 2) [6]-Gingerol for assay 2 (Purity value by quantitative
D (mmol NaOH/min) = ×
t2 - t1 10 NMR)
Unity of peak—Dissolve 5 mg of [6]-gingerol for assay 2
FIP Units per mL of crystalline trypsin
in 5 mL of methanol, and use this solution as the sample so-
( D1 - D0 ) × T
= lution. Perform the test with 10 mL of the sample solution
L×M
as directed under Liquid Chromatography <2.01> according
D1: mmol of sodium hydroxide consumed in 1 minute to the following conditions, and compare the absorption
when the sample solution is used spectra of at least 3 points including the top of [6]-gingerol
D0: mmol of sodium hydroxide consumed in 1 minute peak and around the two middle peak heights of before and
when the solution for blank determination is used after the top: no difference in form is observed among their
M: Amount (mg) of crystalline trypsin taken spectra.
L: Amount (mL) of the sample solution put in the reac- Operating conditions
tion reservoir Column, column temperature, mobile phase, and flow
T: Total volume (mL) of the sample solution rate: Proceed as directed in the operating conditions in the
Assay (3) under Hangekobokuto Extract.
One FIP Unit is an amount of enzyme which decomposes
Detector: A photodiode array detector (wavelength: 282
1 mmol of N-a-benzoyl-L-arginine ethyl ester per minute

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2665

nm, measuring range of spectrum: 220 – 400 nm). lapped with any signal of obvious foreign substance, and
System suitability the ratio of the resonance intensities, (A1/3)/A2, of each
System performance: Proceed as directed in the system signal around d 3.56 ppm and d 6.52 ppm is between 0.99
suitability in the Assay (3) under Hangekobokuto Extract. and 1.01.
Assay—Weigh accurately 5 mg of [6]-gingerol for assay 2 System repeatability: When the test is repeated 6 times
and 1 mg of 1,4-BTMSB-d4 for nuclear magnetic resonance with the sample solution under the above operating condi-
spectroscopy using an ultramicrobalance, dissolve both in tions, the relative standard deviation of the ratio of the
1 mL of deuterated methanol for nuclear magnetic reso- resonance intensity, A1 or A2, to that of the internal refer-
nance spectroscopy, and use this solution as the sample ence is not more than 1.0z.
solution. Transfer the sample solution into an NMR tube
[6]-Gingerol for thin-layer chromatography
5 mm in outer diameter, and measure 1H-NMR as directed
C17H26O4 A yellow-white to yellow, liquid or solid. Freely
under Nuclear Magnetic Resonance Spectroscopy <2.21>
soluble in methanol and in ethanol (99.5), and practically
and Crude Drugs Test <5.01> according to the following
insoluble in water.
conditions, using 1,4-BTMSB-d4 for nuclear magnetic reso-
Identification—Determine the absorption spectrum of a
nance spectroscopy as the internal reference compound.
solution of [6]-gingerol for thin-layer chromatography in
Calculate the resonance intensities, A1 (equivalent to 3
ethanol (99.5) (7 in 200,000) as directed under Ultraviolet-
hydrogens) and A2 (equivalent to 1 hydrogen), of the sig-
visible Spectrophotometry <2.24>: it exhibits a maximum be-
nals around d 3.56 ppm and d 6.52 ppm assuming the signal
tween 279 nm and 283 nm.
of the internal reference compound as d 0 ppm.
Purity Related substances—Dissolve 1.0 mg of [6]-gin-
Amount (z) of [6]-gingerol (C17H26O4) gerol for thin-layer chromatography in exactly 2 mL of
= MS × I × P/(M × N ) × 1.2997 methanol. Perform the test with 10 mL of this solution as di-
rected in the Identification under Ginger: any spot other
M: Amount (mg) of [6]-gingerol for assay 2 taken
than the principal spot at the Rf value of about 0.3 does not
MS: Amount (mg) of 1,4-BTMSB-d4 for nuclear magnetic
appear.
resonance spectroscopy taken
I: Sum of the signal resonance intensities, A1 and A2, Glycerin for gas chromatography C3H8O3 [K 8295,
based on the signal resonance intensity of 1,4-BTMSB- Special class or for gas chromatography] When perform the
d4 for nuclear magnetic resonance spectroscopy as test as directed in the Purity (11) under Concentrated Glyce-
18.000 rin, it does not show any peak at the retention times corre-
N: Sum of the numbers of the hydrogen derived from A1 sponding to ethylene glycol and diethylene glycol.
and A2
Hexyl parahydroxybenzoate C13H18O3 White, crystals
P: Purity (z) of 1,4-BTMSB-d4 for nuclear magnetic
or crystalline powder.
resonance spectroscopy
Melting point <2.60>: 49 – 539C
Operating conditions Content: not less than 98.0z. Assay—Weigh accurately
Apparatus: A nuclear magnetic resonance spectrometer about 0.3 g of hexyl parahydroxybenzoate, dissolve in 50
having 1H resonance frequency of not less than 400 MHz. mL of diluted N,N-dimethylformamide (4 in 5), and titrate
Target nucleus: 1H. <2.50> with 0.1 mol/L sodium hydroxide VS (potentiometric
Digital resolution: 0.25 Hz or lower. titration). Perform a blank determination in the same man-
Measuring spectrum range: 20 ppm or upper, including ner, and make any necessary correction.
between -5 ppm and 15 ppm.
Each mL of 0.1 mol/L sodium hydroxide VS
Spinning: off.
= 22.23 mg of C13H18O3
Pulse angle: 909.
13C decoupling: on. Isopromethazine hydrochloride for thin-layer chromatog-
Delay time: Repeating pulse waiting time not less than 60 raphy C17H20N2S.HCl White, crystalline powder. Odor-
seconds. less. Freely soluble in water, in ethanol (95) and in chlo-
Integrating times: 8 or more times. roform, and practically insoluble in diethyl ether.
Dummy scanning: 2 or more times. Melting point <2.60>: 186 – 1959C
Measuring temperature: A constant temperature between Purity Related substances—Dissolve 5.0 mg of isopro-
209C and 309C. methazine hydrochloride for thin-layer chromatography in
System suitability exactly 25 mL of ethanol (95), and perform the test with this
Test for required detectability: When the procedure is run solution as directed in the Purity (3) under Promethazine
with the sample solution under the above operating condi- Hydrochloride: any spot other than the principal spot at the
tions, the SN ratio of each signal around d 3.56 ppm and d Rf value of about 0.65 does not appear.
6.52 ppm is not less than 100.
Loganin for assay C17H26O10 Loganin for thin-layer
System performance: When the procedure is run with the
chromatography. It meets the requirement of the following
sample solution under the above operating conditions, the
1) Loganin for assay 1 or 2) Loganin for assay 2 (Purity
two signals around d 3.56 ppm and d 6.52 ppm are not over-
value by quantitative NMR). The former is used after dry-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2666 General Tests, Processes and Apparatus Supplement I, JP XVII

ing in a desiccator (silica gel) for 24 hours, and the latter is spectroscopy, and use this solution as the sample solution.
used with correction for its amount based on the result ob- Transfer the sample solution into an NMR tube 5 mm in
tained in the Assay. outer diameter, and measure 1H-NMR as directed under
1) Loganin for assay 1 Nuclear Magnetic Resonance Spectroscopy <2.21> and
z
Absorbance <2.24> E 11cm (235 nm): 275 – 303 [5 mg after Crude Drugs Test <5.01> according to the following condi-
drying in a desiccator (silica gel) for 24 hours, methanol, tions, using 1,4-BTMSB-d4 for nuclear magnetic resonance
500 mL] spectroscopy as the internal reference compound. Calculate
Purity Related substances—Dissolve 2 mg of loganin the resonance intensity A (equivalent to 1 hydrogen) of the
for assay 1 in 5 mL of the mobile phase, and use this solu- signal around d 7.14 ppm assuming the signal of the internal
tion as the sample solution. Pipet 1 mL of the sample solu- reference compound as d 0 ppm.
tion, add the mobile phase to make exactly 100 mL, and use
Amount (z) of loganin (C17H26O10)
this solution as the standard solution. Perform the test with
= MS × I × P/(M × N ) × 1.7235
exactly 10 mL each of the sample solution and standard so-
lution as directed under Liquid Chromatography <2.01> ac- M: Amount (mg) of loganin for assay 2 taken
cording to the following conditions, and determine each MS: Amount (mg) of 1,4-BTMSB-d4 for nuclear magnetic
peak area by the automatic integration method: the total resonance spectroscopy taken
area of the peaks other than loganin from the sample solu- I: Signal resonance intensity A based on the signal reso-
tion is not larger than the peak area of loganin from the nance intensity of 1,4-BTMSB-d4 for nuclear magnetic
standard solution. resonance spectroscopy as 18.000
Operating conditions N: Number of the hydrogen derived from A
Detector, column, column temperature, mobile phase and P: Purity (z) of 1,4-BTMSB-d4 for nuclear magnetic
flow rate: Proceed as directed in the operating conditions in resonance spectroscopy
the Assay (1) under Goshajinkigan Extract.
Operating conditions
Time span of measurement: About 3 times as long as the
Apparatus: A nuclear magnetic resonance spectrometer
retention time of loganin.
having 1H resonance frequency of not less than 400 MHz.
System suitability
Target nucleus: 1H.
System performance and system repeatability: Proceed as
Digital resolution: 0.25 Hz or lower.
directed in the system suitability in the Assay (1) under
Measuring spectrum range: 20 ppm or upper, including
Goshajinkigan Extract.
between -5 ppm and 15 ppm.
Test for required detectability: Pipet 1 mL of the stand-
Spinning: off.
ard solution, and add the mobile phase to make exactly 20
Pulse angle: 909.
mL. Confirm that the peak area of loganin obtained with 10 13C decoupling: on.
mL of this solution is equivalent to 3.5 to 6.5z of that with
Delay time: Repeating pulse waiting time not less than 60
10 mL of the standard solution.
seconds.
2) Loganin for assay 2 (Purity value by quantitative
Integrating times: 8 or more times.
NMR)
Dummy scanning: 2 or more times.
Unity of peak—Dissolve 2 mg of loganin for assay 2 in 5
Measuring temperature: A constant temperature between
mL of the mobile phase, and use this solution as the sample
209C and 309C.
solution. Perform the test with 10 mL of the sample solution
System suitability
as directed under Liquid Chromatography <2.01> according
Test for required detectability: When the procedure is run
to the following conditions, and compare the absorption
with the sample solution under the above operating condi-
spectra of at least 3 points including the top of loganin peak
tions, the SN ratio of each signal around d 5.02 ppm and d
and around the two middle peak heights of before and after
7.14 ppm is not less than 100.
the top: no difference in form is observed among their spec-
System performance: When the procedure is run with the
tra.
sample solution under the above operating conditions, the
Operating conditions
two signals around d 5.02 ppm and d 7.14 ppm are not
Column, column temperature, mobile phase, and flow
overlapped with any signal of obvious foreign substance.
rate: Proceed as directed in the operating conditions in the
Furthermore, when determined the resonance intensities
Assay (1) under Goshajinkigan Extract.
A1 and A, both equivalent to 1 hydrogen, of each signal
Detector: A photodiode array detector (wavelength: 238
around d 5.02 ppm and d 7.14 ppm, the ratio of them,
nm, measuring range of spectrum: 220 – 400 nm).
A1/A, is between 0.99 and 1.01.
System suitability
System repeatability: When the test is repeated 6 times
System performance: Proceed as directed in the system
with the sample solution under the above operating condi-
suitability in the Assay (1) under Goshajinkigan Extract.
tions, the relative standard deviation of the ratio of the
Assay—Weigh accurately 5 mg of loganin for assay 2 and
resonance intensity A to that of the internal reference is not
1 mg of 1,4-BTMSB-d4 for nuclear magnetic resonance
more than 1.0z.
spectroscopy using an ultramicrobalance, dissolve in 1 mL
of deuterated methanol for nuclear magnetic resonance Methanol, anhydrous CH4O To 1000 mL of methanol

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2667

add 5 g of magnesium powder. After the evolving of a gas is fluorescent light for 60 minutes. After polymerization, re-
stopped, distillate the solution, and preserve the distillate move the water from the upper surface of the gel.
protecting from moisture. Water content per mL is not iii) Sample solution Dissolve 3.0 mg of the substance
more than 0.3 mg. to be examined in the buffer solution for sample to make 10
mL.
Neocarzinostatin-styrene-maleic acid alternating copoly-
(iv) Procedure Mount the gel in electrophoresis appa-
mer partial butyl ester condensate (2:3) Condensate of ne-
ratus. Add a mixture of 200 mL of Solution F and 2 mL of
ocarzinostatin and styrene-maleic acid alternating copoly-
bromophenol blue solution (1 in 100,000) to the upper
mer partial butyl ester in a rate of 2:3 by amide bond.
reservoir (cathode) and 300 mL of Solution F to the lower
Average molecular mass: about 28,400. A pale yellow pow-
reservoir (anode). Introduce carefully exactly 100 mL of the
der.
sample solution onto the surface of the gel, and allow
Identification—Dissolve 4 mg of the substance to be
electrophoresis at room temperature to take place with a
examined in 0.05 mol/L phosphate buffer solution (pH 7.0)
current of 2 mA per tube as a bromophenol blue band is
to make 10 mL. Determine the absorption spectrum of this
passing in the stacking gel and then increase the current to 4
solution as directed under Ultraviolet-visible Spectropho-
mA per tube as the bromophenol blue band is passing in the
tometry <2.24>: it exhibits a maximum between 266 nm and
resolving gel, and stop the current when the band reached 5
270 nm, and shoulders between 257 nm and 262 nm, be-
cm from the upper end of the gel.
tween 286 nm and 291 nm and between 318 nm and 348 nm.
(v) Staining and decolorization Dissolve 0.1 g of
Absorbance <2.24> E 11cmz
(268 nm): 13.0 – 17.5 [4 mg cal-
Coomassie brilliant blue G-250 in 100 mL of trichloroacetic
culated on the anhydrous basis, 0.05 mol/L phosphate
acid solution (1 in 2), and mix 1 volume of this solution and
buffer solution (pH 7.0), 10 mL].
2 volumes of water before using. Immerse the gels for 15
Purity (i) Test solutions
hours in this mixture, and transfer into about 20 mL of
Solution A: Dissolve 36.6 g of 2-amino-2-hydroxymethyl-
acetic acid (100) solution (7 in 100) to remove the excess of
1,3-propanediol in 48 mL of 1 mol/L hydrochloric acid TS,
dye. Replace the acetic acid (100) solution until the back
0.23 mL of N,N,N?,N?-tetramethylethylenediamine and
ground of the gel becomes colorless.
water to make 100 mL.
(vi) Determination Determine the peak area, AT, of
Solution B: Dissolve 33.3 g of acrylamide and 0.89 g of
neocarzinostatin-styrene-maleic acid alternating copolymer
N,N?-methylenebisacrylamide in water to make 100 mL.
partial butyl ester condensate (2:3) and the total area, A, of
Preserve in a cold place, avoiding exposure to light.
the peaks other than neocarzinostatin-styrene-maleic acid
Solution C: Dissolve 5.98 g of 2-amino-2-hydroxymethyl-
alternating copolymer partial butyl ester condensate (2:3),
1,3-propanediol in 48 mL of 1 mol/L hydrochloric acid TS,
based on the absorbance at 600 nm of the gel determined by
0.46 mL of N,N,N?,N?-tetramethylethylenediamine and
using a densitometer. Calculate the amount of neocar-
water to make 100 mL.
zinostatin-styrene-maleic acid alternating copolymer partial
Solution D: Dissolve 10.0 g of acrylamide and 2.5 g of
butyl ester condensate (2:3) by the following formula: not
N,N?-methylenebisacrylamide in water to make 100 mL.
less than 90.0z.
Preserve in a cold place, avoiding exposure to light.
Solution E: Dissolve 4 mg of riboflavin in water to make Amount (z) of neocarzinostatin-styrene-maleic acid
100 mL. Preserve in a cold place, avoiding exposure to alternating copolymer partial butyl ester condensate (2:3)
light. = AT/(AT + A) × 100
Solution F: Dissolve 3.0 g of 2-amino-2-hydroxymethyl-
Water <2.48> Not more than 12.0z (10 mg, coulometric
1,3-propanediol and 14.4 g of glycine in water to make 500
titration).
mL.
Buffer solution for sample: To 50 mL of Solution C add Polyvinyl alcohol I Colorless to white or pale yellow,
20 mL of water and 10 mL of glycerin solution (3 in 5). granules or powder. It is odorless, or has a faint odor of
(ii) Gels acetic acid. It is tasteless. Practically insoluble in ethanol
Resolving gel: Mix 2.5 mL of Solution A and 7.5 mL of (95) and in diethyl ether. To polyvinyl alcohol I add water,
Solution B. Mix the mixture with 10 mL of freshly prepared and heat: a clear, viscous solution is obtained. Polyvinyl
ammonium peroxodisulfate solution (7 in 5000) after alcohol I is hygroscopic.
degassing under reduced pressure. Pour this mixture into a Viscosity <2.53> 25.0 – 31.0 mm2/s. Weigh 4.000 g of
glass tube, 5 mm in inside diameter and 10 cm in length, to polyvinyl alcohol I, previously dried, add 95 mL of water,
make 7 cm height, put water gently on the upper surface of allow to stand for 30 minutes, and heat to dissolve on a
the mixture, and allow to polymerize for 60 minutes. After water bath under a reflux condenser for 2 hours while stir-
polymerization, remove the water from the upper surface of ring. After cooling, add water to make 100.0 g, and mix.
the gel. Allow to stand still to remove bubbles, and perform the test
Stacking gel: Mix 1 mL of Solution C, 2 mL of Solution at 20 ± 0.19C as directed in Method 1.
D, 1 mL of Solution E and 4 mL of water, pour 0.2 mL of pH <2.54>—The pH of a solution of 1.0 g of polyvinyl
the mixture on the resolving gel, put water gently on the up- alcohol I in 25 mL of water is between 5.0 and 8.0.
per surface of the mixture, and allow to polymerize under a Purity Clarity and color of solution—To 20 mL of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2668 General Tests, Processes and Apparatus Supplement I, JP XVII

water add 1.0 g of polyvinyl alcohol I, disperse by thorough


3.0025 × (a - b)
stirring, warm between 609 C and 809 C for 2 hours, and A=
amount (g) of polyvinyl alcohol II taken
cool: the solution is colorless and clear.
Saponification value 98.0 – 99.0 molz. Weigh accu- a: Volume (mL) of 0.5 mol/L sodium hydroxide VS con-
rately about 3.0 g of polyvinyl alcohol I, previously dried, sumed in the test
transfer to a glass-stoppered conical flask, add 100 mL of b: Volume (mL) of 0.5 mol/L sodium hydroxide VS con-
water, and dissolve by heating on a water bath. After cool- sumed in the blank test
ing, add exactly 25 mL of 0.1 mol/L sodium hydroxide VS,
[6]-Shogaol for assay C17H24O3 [6]-Shogaol for thin-
stopper tightly, and allow to stand for 2 hours. Then add
layer chromatography. It meets the requirement of the fol-
exactly 30 mL of 0.05 mol/L sulfuric acid VS, shake thor-
lowing 1) [6]-Shogaol for assay 1 or 2) [6]-Shogaol for assay
oughly, and titrate <2.50> with 0.1 mol/L sodium hydroxide
2 (Purity value by quantitative NMR). The latter is used
VS (indicator: 3 drops of phenolphthalein TS). Perform a
with correction for its amount based on the result obtained
blank determination in the same manner, and make any
in the Assay.
necessary correction. However, when the volume of 0.1
1) [6]-Shogaol for assay 1
mol/L sodium hydroxide VS consumed in the test is 25 mL z
Absorbance <2.24> E 11cm (225 nm): 727 – 781 [5 mg,
or more, use about 2.0 g of the sample.
ethanol (99.5), 500 mL].
44.05A Purity Related substances—Dissolve 5 mg of [6]-
Saponification value (molz) = 100 -
60.05 - 0.42A shogaol for assay 1 in 10 mL of a mixture of acetonitrile
and water (2:1), and use this solution as the sample solu-
0.6005 × (a - b)
A= tion. Pipet 1 mL of the sample solution, add a mixture of
amount (g) of polyvinyl alcohol I taken
acetonitrile and water (2:1) to make exactly 100 mL, and use
a: Volume (mL) of 0.1 mol/L sodium hydroxide VS con- this solution as the standard solution. Perform the test with
sumed in the test exactly 10 mL each of the sample solution and standard
b: Volume (mL) of 0.1 mol/L sodium hydroxide VS con- solution as directed under Liquid Chromatography <2.01>
sumed in the blank test according to the following conditions, and determine each
peak area by the automatic integration method: the total
Polyvinyl alcohol II Colorless to white or pale yellow,
area of the peaks other than [6]-shogaol from the sample
granules or powder. It is odorless, or has a faint odor of
solution is not larger than the peak area of [6]-shogaol from
acetic acid. It is tasteless. Practically insoluble in ethanol
the standard solution.
(95) and in diethyl ether. To polyvinyl alcohol II add water,
Operating conditions
and heat: a clear, viscous solution is obtained. Polyvinyl
Detector, column, column temperature, mobile phase and
alcohol II is hygroscopic.
flow rate: Proceed as directed in the operating conditions in
Viscosity <2.53> 4.6 – 5.4 mm2/s. Weigh 4.000 g of
the Assay (2) under Mukoi-Daikenchuto Extract.
polyvinyl alcohol II, previously dried, add 95 mL of water,
Time span of measurement: 3 times as long as the reten-
allow to stand for 30 minutes, and dissolve by stirring on a
tion time of [6]-shogaol, beginning after the solvent peak.
water bath between 609 C and 809 C for 2 hours. After cool-
System suitability
ing, add water to make 100.0 g, and mix. Allow to stand
Test for required detectability: Pipet 1 mL of the stand-
still to remove bubbles, and perform the test at 20 ± 0.19C
ard solution, and add a mixture of acetonitorile and water
as directed in Method 1.
(2:1) to make exactly 20 mL. Confirm that the peak area of
pH <2.54>—The pH of a solution of 1.0 g of polyvinyl al-
[6]-shogaol obtained with 10 mL of this solution is equiva-
cohol II in 25 mL of water is between 5.0 and 8.0.
lent to 3.5 to 6.5z of that with 10 mL of the standard solu-
Purity Clarity and color of solution—To 20 mL of
tion.
water add 1.0 g of polyvinyl alcohol II, disperse by
System performance: When the procedure is run with 10
thorough stirring, heat on a water bath for 2 hours, and
mL of the standard solution under the above operating con-
cool: the solution is clear and colorless.
ditions, the number of theoretical plates and the symmetry
Saponification value 86.5 – 89.5 molz. Weigh accu-
factor of the peak of [6]-shogaol are not less than 5000 and
rately about 2 g of polyvinyl alcohol II, previously dried,
not more than 1.5z, respectively.
transfer to a glass-stoppered conical flask, add 100 mL of
System repeatability: When the test is repeated 6 times
water, and warm while stirring for 2 hours. After cooling,
with 10 mL of the standard solution under the above operat-
add exactly 25 mL of 0.5 mol/L sodium hydroxide VS,
ing conditions, the relative standard deviation of the peak
stopper tightly, and allow to stand for 2 hours. Then add
area of [6]-shogaol is not more than 1.5z.
exactly 30 mL of 0.25 mol/L sulfuric acid VS, shake thor-
2) [6]-Shogaol for assay 2 (Purity value by quantitative
oughly, and titrate <2.50> with 0.5 mol/L sodium hydroxide
NMR)
VS (indicator: 3 drops of phenolphthalein TS). Perform a
Unity of peak—Dissolve 5 mg of [6]-shogaol for assay 2
blank determination in the same manner, and make any
in 10 mL of a mixture of acetonitrile and water (2:1), and
necessary correction.
use this solution as the sample solution. Perform the test
44.05A
Saponification value (molz) = 100 - with 10 mL of the sample solution as directed under Liquid
60.05 - 0.42A

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII General Tests, Processes and Apparatus 2669

Chromatography <2.01> according to the following condi- System suitability


tions, and compare the absorption spectra of at least 3 Test for required detectability: When the procedure is run
points including the top of [6]-shogaol peak and around the with the sample solution under the above operating condi-
two middle peak heights of before and after the top: no tions, the SN ratio of each signal around d 3.57 ppm and d
difference in form is observed among their spectra. 6.37 – 6.43 ppm is not less than 100.
Operating conditions System performance: When the procedure is run with the
Column, column temperature, mobile phase, and flow sample solution under the above operating conditions, the
rate: Proceed as directed in the operating conditions in the two signals around d 3.57 ppm and d 6.37 – 6.43 ppm are
Assay (2) under Mukoi-Daikenchuto Extract. not overlapped with any signal of obvious foreign sub-
Detector: A photodiode array detector (wavelength: 225 stance. Furthermore, when determined the resonance inten-
nm, measuring range of spectrum: 220 – 400 nm). sities, A (equivalent to 3 hydrogens) and A1 (equivalent to 2
System suitability hydrogens) of each signal around d 3.57 ppm and d 6.37 –
System performance: Proceed as directed in the system 6.43 ppm, the ratio of the resonance intensities, (A1/2)/
suitability in the Assay (2) under Mukoi-Daikenchuto Ex- (A/3), of each signal around d 3.57 ppm and d 6.37 – 6.43
tract. ppm is between 0.99 and 1.01.
Assay—Weigh accurately 5 mg of [6]-shogaol for assay 2 System repeatability: When the test is repeated 6 times
and 1 mg of 1,4-BTMSB-d4 for nuclear magnetic resonance with the sample solution under the above operating condi-
spectroscopy using an ultramicrobalance, dissolve in 1 mL tions, the relative standard deviation of the ratio of the
of deuterated methanol for nuclear magnetic resonance resonance intensity A to that of the internal reference com-
spectroscopy, and use this solution as the sample solution. pound is not more than 1.0z.
Transfer the sample solution into an NMR tube 5 mm in
[6]-Shogaol for thin-layer chromatography C17H24O3
outer diameter, and measure 1H-NMR as directed under
A pale yellow, clear liquid. Miscible with methanol and with
Nuclear Magnetic Resonance Spectroscopy <2.21> and
ethanol (99.5), and practically insoluble in water.
Crude Drugs Test <5.01> according to the following condi-
Purity Related substances—Dissolve 1.0 mg of [6]-
tions, using 1,4-BTMSB-d4 for nuclear magnetic resonance
shogaol for thin-layer chromatography in 2 mL of metha-
spectroscopy as the internal reference compound. Calculate
nol, and use this solution as the sample solution. Perform
the resonance intensity A (equivalent to 3 hydrogens) of the
the test with the sample solution as directed under Thin-
signal around d 3.57 ppm assuming the signal of the internal
layer Chromatography <2.03>. Spot 10 mL of the sample so-
reference compound as d 0 ppm.
lution on a plate of silica gel for thin-layer chromatography.
Amount (z) of [6]-shogaol (C17H24O3) Develop the plate with a mixture of ethyl acetate and hexane
= MS × I × P/(M × N) × 1.2202 (1:1) to a distance of about 10 cm, and air-dry the plate.
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
M: Amount (mg) of [6]-shogaol for assay 2 taken
on the plate, heat at 1059C for 5 minutes, and allow to cool:
MS: Amount (mg) of 1,4-BTMSB-d4 for nuclear magnetic
any spot other than the principal spot at an Rf value of
resonance spectroscopy taken
about 0.5 does not appear.
I: Signal resonance intensity A based on the signal reso-
nance intensity of 1,4-BTMSB-d4 for nuclear magnetic Styrene-maleic acid alternating copolymer partial butyl
resonance spectroscopy as 18.000 ester Polymerize styrene and maleic anhydride using cu-
N: Number of the hydrogen derived from A mene as solvent, and add 1-butanol or water to the maleic
P: Purity (z) of 1,4-BTMSB-d4 for nuclear magnetic anhydride groups. Average molecular mass: about 1600. A
resonance spectroscopy white to pale yellowish white powder.
Identification—Dissolve 5 mg of the substance to be ex-
Operating conditions
amined in sodium hydrogen carbonate solution (1 in 15) to
Apparatus: A nuclear magnetic resonance spectrometer
make 10 mL. Determine the absorption spectrum of this so-
having 1H resonance frequency of not less than 400 MHz.
lution as directed under Ultraviolet-visible Spectrophotome-
Target nucleus: 1H.
try <2.24>: it exhibits a maximum between 256 nm and 260
Digital resolution: 0.25 Hz or lower.
nm, and a shoulder between 251 nm and 256 nm.
Measuring spectrum range: 20 ppm or upper, including z
Absorbance <2.24> E 11cm (258 nm): 6.3 – 7.3 [5 mg calcu-
between -5 ppm and 15 ppm.
lated on the anhydrous basis, sodium hydrogen carbonate
Spinning: off.
solution (1 in 15), 10 mL].
Pulse angle: 909.
13C decoupling: on.
Purity (i) Test solutions
Solution A: Dissolve 36.6 g of 2-amino-2-hydroxymethyl-
Delay time: Repeating pulse waiting time not less than 60
1,3-propanediol in 48 mL of 1 mol/L hydrochloric acid TS,
seconds.
0.23 mL of N,N,N?,N?-tetramethylethylenediamine and
Integrating times: 8 or more times.
water to make 100 mL.
Dummy scanning: 2 or more times.
Solution B: Dissolve 33.3 g of acrylamide and 0.89 g of
Measuring temperature: A constant temperature between
N,N?-methylenebisacrylamide in water to make 100 mL.
209C and 309C.
Preserve in a cold place, avoiding exposure to light.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2670 General Tests, Processes and Apparatus Supplement I, JP XVII

Solution C: Dissolve 5.98 g of 2-amino-2-hydroxymethyl- absorbance at 600 nm of the gel determined by using a
1,3-propanediol in 48 mL of 1 mol/L hydrochloric acid TS, densitometer. Calculate the amount of styrene-maleic acid
0.46 mL of N,N,N?,N?-tetramethylethylenediamine and alternating copolymer partial butyl ester by the following
water to make 100 mL. formula: not less than 98.0z.
Solution D: Dissolve 10.0 g of acrylamide and 2.5 g of
Amount (z) of styrene-maleic acid alternating
N,N?-methylenebisacrylamide in water to make 100 mL.
copolymer partial butyl ester
Preserve in a cold place, avoiding exposure to light.
= AT/(AT + A) × 100
Solution E: Dissolve 4 mg of riboflavin in water to make
100 mL. Preserve in a cold place, avoiding exposure to Water <2.48>: Not more than 10.0z (10 mg, coulometric
light. titration).
Solution F: Dissolve 3.0 g of 2-amino-2-hydroxymethyl-
p-Toluenesulfonamide CH3C6H4SO2NH2 White, crys-
1,3-propanediol and 14.4 g of glycine in water to make 500
tals or crystalline powder. Melting point: about 1379 C.
mL.
Purity Related substances—Dissolve 30 mg of p-tol-
Buffer solution for sample: To 50 mL of Solution C add
uenesulfonamide in acetone to make exactly 200 mL. Spot
20 mL of water and 10 mL of glycerin solution (3 in 5).
10 mL of this solution on a plate of silica gel for thin-layer
(ii) Gels
chromatography. Develop the plate with a mixture of chlo-
Resolving gel: Mix 2.5 mL of Solution A and 7.5 mL of
roform, methanol, cyclohexane and diluted ammonia solu-
Solution B. Mix the mixture with 10 mL of freshly prepared
tion (28) (10 in 11) (200:100:60:23) to a distance of about 12
ammonium peroxodisulfate solution (7 in 5000) after
cm, and air-dry the plate. Heat the plate at 1109C for 10
degassing under reduced pressure. Pour this mixture into a
minutes, and immediately expose to chlorine for 2 minutes.
glass tube, 5 mm in inside diameter and 10 cm in length, to
Expose the plate to cold wind until a very pale blue color de-
make 7 cm height, put water gently on the upper surface of
velops when 1 drop of potassium iodide-starch TS is placed
the mixture, and allow to polymerize for 60 minutes. After
on a site below the starting line on the plate. Spray evenly
polymerization, remove the water from the upper surface of
potassium iodide-starch TS on the plate: no spot other than
the gel.
the principal spot at an Rf value of about 0.6 appears.
Stacking gel: Mix 1 mL of Solution C, 2 mL of Solution
D, 1 mL of Solution E and 4 mL of water, pour 0.2 mL of
Delete the following:
the mixture on the resolving gel, put water gently on the
upper surface of the mixture, and allow to polymerize under 2-Acetamidoglutarimide
a fluorescent light for 60 minutes. After polymerization,
Mercury (II) chloride TS
remove the water from the upper surface of the gel.
(iii) Sample solution Dissolve 3.0 mg of the substance PBS containing bovine serum
to be examined in the buffer solution for sample to make 20
PBS containing bovine serum albumin
mL.
(iv) Procedure Mount the gel in an electrophoresis ap- Peroxidase-labeled rabbit anti-ECP antibody Fab? TS
paratus. Add a mixture of 200 mL of Solution F and 2 mL
Thimerosal
of bromophenol blue solution (1 in 100,000) to the upper
reservoir (cathode) and 300 mL of Solution F to the lower Thymine
reservoir (anode). Introduce carefully exactly 100 mL of the
Trichloroacetic acid TS for serrapeptase
sample solution onto the surface of the gel, and allow elec-
trophoresis at room temperature to take place with a cur-
rent of 2 mA per tube as a bromophenol blue band is pass-
ing in the stacking gel and then increase the current to 4 mA 9.42 Solid Supports/Column
per tube as the bromophenol blue band is passing in the Packings for Chromatography
resolving gel, and stop the current when the band reached 5
cm from the upper end of the gel.
Add the following:
(v) Staining and decolorization Dissolve 0.1 g of
Coomassie brilliant blue G-250 in 100 mL of trichloroacetic 14z Cyanopropylphenyl-86z dimethyl silicone polymer
acid solution (1 in 2), and mix 1 volume of this solution and for gas chromatography Prepared for gas chromatogra-
2 volumes of water before using. Immerse the gels for 15 phy.
hours in this mixture, and transfer into about 20 mL of
Octadecylsilanized monolithic silica for liquid chroma-
acetic acid (100) solution (7 in 100) to remove the excess of
tography Prepared for liquid chromatography.
dye. Replace the acetic acid (100) solution until the back
ground of the gel becomes colorless. Human albumin chemically bonded silica gel for liquid
(vi) Determination Determine the peak area, AT, of chromatography Prepared for liquid chromatography.
styrene-maleic acid alternating copolymer partial butyl ester
and the total area, A, of the peaks other than styrene-maleic
acid alternating copolymer partial butyl ester, based on the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Official Monographs
Delete the following Monograph: to 13z of that with 10 mL of the standard solution.
System performance: When the procedure is run with 10
Aceglutamide Aluminum mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
アセグルタミドアルミニウム factor of the peak of amoxicillin are not less than 2500 and
not more than 1.5, respectively.
System repeatability: When the test is repeated 6 times
Amoxicillin Hydrate with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
アモキシシリン水和物 area of amoxicillin is not more than 1.0z.

Change the Purity (3) as follows:


Purity Ampicillin Hydrate
(3) Related substances—Dissolve 0.10 g of Amoxicillin
アンピシリン水和物
Hydrate in 50 mL of a solution of boric acid (1 in 200), and
use this solution as the sample solution. Pipet 1 mL of the
Change the Purity (3) as follows:
sample solution, add a solution of boric acid (1 in 200) to
make exactly 100 mL, and use this solution as the standard Purity
solution. Perform the test with exactly 10 mL each of the (3) Related substances—Dissolve 50 mg of Ampicillin
sample solution and standard solution as directed under Hydrate in 50 mL of the mobile phase, and use this solution
Liquid Chromatography <2.01> according to the following as the sample solution. Pipet 1 mL of the sample solution,
conditions. Determine each peak area of both solutions by add the mobile phase to make exactly 100 mL, and use this
the automatic integration method: the area of the peak solution as the standard solution. Perform the test with ex-
other than amoxicillin obtained from the sample solution is actly 10 mL each of the sample solution and standard solu-
not larger than the peak area of amoxicillin from the stand- tion as directed under Liquid Chromatography <2.01> ac-
ard solution. Furthermore, the total area of the peaks other cording to the following conditions, and determine each
than amoxicillin from the sample solution is not larger than peak area by the automatic integration method: the area of
3 times the peak area of amoxicillin from the standard solu- the peak other than ampicillin obtained from the sample so-
tion. lution is not larger than the peak area of ampicillin from the
Operating conditions— standard solution, and the total area of the peaks other than
Detector: An ultraviolet absorption photometer (wave- ampicillin from the sample solution is not larger than 2
length: 254 nm). times the peak area of ampicillin from the standard solu-
Column: A stainless steel column 4 mm in inside diameter tion.
and 30 cm in length, packed with octadecylsilanized silica Operating conditions—
gel for liquid chromatography (10 mm in particle diameter). Detector, column, column temperature, mobile phase,
Column temperature: A constant temperature of about and flow rate: Proceed as directed in the operating condi-
259C. tions in the Assay.
Mobile phase: Dissolve 1.36 g of sodium acetate trihy- Time span of measurement: About 10 times as long as the
drate in 750 mL of water, adjust to pH 4.5 with acetic acid retention time of ampicillin.
(31), and add water to make 1000 mL. To 950 mL of this System suitability—
solution add 50 mL of methanol. Test for required detectability: Pipet 1 mL of the stand-
Flow rate: Adjust so that the retention time of amoxicillin ard solution, and add the mobile phase to make exactly 10
is about 8 minutes. mL. Confirm that the peak area of ampicillin obtained with
Time span of measurement: About 4 times as long as the 10 mL of this solution is equivalent to 7 to 13z of that with
retention time of amoxicillin, beginning after the solvent 10 mL of the standard solution.
peak. System performance: When the procedure is run with 10
System suitability— mL of the standard solution under the above operating con-
Test for required detectability: To exactly 1 mL of the ditions, the number of theoretical plates and the symmetry
standard solution add a solution of boric acid (1 in 200) to factor of the peak of ampicillin are not less than 5000 and
make exactly 10 mL. Confirm that the peak area of amoxi- not more than 1.5, respectively.
cillin obtained with 10 mL of this solution is equivalent to 7 System repeatability: When the test is repeated 6 times

2671
The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2672 Official Monographs Supplement I, JP XVII

with 10 mL of the standard solution under the above operat- semide in 5 mL of N,N-dimethylformamide, add 12 mL of
ing conditions, the relative standard deviation of the peak water, 1.0 mL of a solution of sodium nitrite (1 in 200) and
area of ampicillin is not more than 1.0z. 2.0 mL of diluted hydrochloric acid (1 in 10) under ice-cool-
ing, shake, and allow to stand for 3 minutes. Add 1.0 mL of
ammonium amidosulfate TS, shake thoroughly, allow to
Add the following: stand for 3 minutes, and add 1.0 mL of a solution of N-1-
naphthylethylenediamine dihydrochloride (1 in 200). Shake
Azosemide this solution, and add N,N-dimethylformamide to make ex-
actly 50 mL. Determine the absorbance of this solution at
アゾセミド 540 nm as directed under Ultraviolet-visible Spectropho-
tometry <2.24>, using a solution prepared in the same man-
ner with 5 mL of N,N-dimethylformamide as the blank: the
absorbance is not more than 0.15.

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


C, 3
hours).

C12H11ClN6O2S2: 370.84 Residue on ignition <2.44> Not more than 0.1z (1 g).
2-Chloro-5-(1H-tetrazol-5-yl)-4-[(thien-2-ylmethyl)amino]
Assay Weigh accurately about 0.6 g of Azosemide, previ-
benzenesulfonamide
ously dried, dissolve in 50 mL of N,N-dimethylformamide,
[27589-33-9]
and titrate <2.50> with 0.1 mol/L potassium hydroxide-
ethanol VS until the color of the solution changes from yel-
Azosemide, when dried, contains not less than
low to yellow-green (indicator: 10 drops of thymol blue-
99.0z and not more than 101.0z of azosemide
N,N-dimethylformamide TS). Perform a blank determina-
(C12H11ClN6O2S2).
tion with a solution prepared by adding 15 mL of ethanol
Description Azosemide occurs as a white to yellow-white (95) to 50 mL of N,N-dimethylformamide, and make any
crystalline powder. necessary correction.
It is freely soluble in N,N-dimethylformamide, slightly
Each mL of 0.1 mol/L potassium hydroxide-ethanol VS
soluble in methanol and in ethanol (99.5), and practically
= 37.08 mg of C12H11ClN6O2S2
insoluble in water.
It dissolves in dilute sodium hydroxide TS. Containers and storage Containers—Tight containers.
It is gradually colored to yellow by light. Storage—Light-resistant.
Melting point: about 2269 C (with decomposition).

Identification (1) Determine the absorption spectrum of


Add the following:
a solution of Azosemide in dilute sodium hydroxide TS (3 in
500,000) as directed under Ultraviolet-visible Spectropho-
tometry <2.24>, and compare the spectrum with the Refer- Azosemide Tablets
ence Spectrum: both spectra exhibit similar intensities of ab-
アゾセミド錠
sorption at the same wavelengths.
(2) Determine the infrared absorption spectrum of
Azosemide Tablets contain not less than 95.0z and
Azosemide, previously dried, as directed in the potassium
not more than 105.0z of the labeled amount of
bromide disk method under Infrared Spectrophotometry
azosemide (C12H11ClN6O2S2: 370.84).
<2.25>, and compare the spectrum with the Reference Spec-
trum: both spectra exhibit similar intensities of absorption Method of preparation Prepare as directed under Tablets,
at the same wave numbers. with Azosemide.

Purity (1) Chloride <1.03>—To 1.0 g of Azosemide add Identification To a quantity of powdered Azosemide
60 mL of dilute sodium hydroxide TS, dissolve by warming. Tablets, equivalent to 60 mg of Azosemide, add dilute so-
After cooling, add 0.5 mL of nitric acid and filter. To 30 dium hydroxide TS to make 100 mL, shake, and filter. To 1
mL of the filtrate add 6 mL of dilute nitric acid and water mL of the filtrate add dilute sodium hydroxide TS to make
to make 50 mL. Perform the test using this solution as the 100 mL. Determine the absorption spectrum of this solution
test solution. Prepare the control solution with 0.45 mL of as directed under Ultraviolet-visible Spectrophotometry
0.01 mol/L hydrochloric acid VS (not more than 0.032z). <2.24>: it exhibits maxima between 234 nm and 238 nm, be-
(2) Heavy metal <1.07>—Proceed with 1.0 g of Azo- tween 272 nm and 276 nm and between 324 nm and 330 nm.
semide according to Method 2, and perform the test. Pre-
Purity Primary aromatic amines—To a quantity of pow-
pare the control solution with 2.0 mL of Standard Lead So-
dered Azosemide Tablets, equivalent to 20 mg of Azo-
lution (not more than 20 ppm).
semide, add 5 mL of N,N-dimethylformamide, and allow to
(3) Primary aromatic amines—Dissolve 20 mg of Azo-
stand with occasional shaking. Add 12 mL of water, 1.0 mL

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2673

of a solution of sodium nitrite (1 in 200) and 2.0 mL of droxide TS to make exactly 50 mL. Pipet 15 mL of this so-
diluted hydrochloric acid (1 in 10) under ice-cooling, shake, lution, add the dissolution medium to make exactly 25 mL,
and allow to stand for 3 minutes. Add 1.0 mL of ammo- and use this solution as the standard solution. Determine
nium amidosulfate TS, shake thoroughly, and allow to the absorbances, AT and AS, of the sample solution and
stand for 3 minutes. Add 1.0 mL of a solution of N-1- standard solution at 274 nm as directed under Ultraviolet-
naphthylethylenediamine dihydrochloride (1 in 200), and visible Spectrophotometry <2.24>, using a solution prepared
shake. Add N,N-dimethylformamide to make exactly 50 by adding 0.2 mol/L sodium hydroxide TS to 8 mL of the
mL, centrifuge, and use the supernatant liquid as the sam- dissolution medium to make 20 mL as the blank.
ple solution. Determine the absorbance of the sample solu-
Dissolution rate (z) with respect to the labeled amount
tion at 540 nm as directed under Ultraviolet-visible Spectro-
of azosemide (C12H11ClN6O2S2)
photometry <2.24>, using a solution prepared in the same
= MS × AT/AS × V?/V × 1/C × 135
manner with 5 mL of N,N-dimethylformamide as the blank:
the absorbance is not more than 0.15. MS: Amount (mg) of azosemide for assay taken
C: Labeled amount (mg) of azosemide (C12H11ClN6O2S2)
Uniformity of dosage unit <6.02> Perform the Mass varia-
in 1 tablet
tion test, or the Content uniformity test according to the
following method: it meets the requirement. Assay Weigh accurately the mass of not less than 20
To 1 tablet of Azosemide Tablets add dilute sodium hy- tablets of Azosemide Tablets, and powder. Weigh accu-
droxide TS to make exactly V mL so that each mL contains rately a portion of the powder, equivalent to about 60 mg of
about 0.6 mg of azosemide (C12H11ClN6O2S2), shake thor- azosemide (C12H11ClN6O2S2), add dilute sodium hydroxide
oughly, and centrifuge. Pipet 10 mL of the supernatant liq- TS to make exactly 100 mL, shake thoroughly, and filter.
uid, and add dilute sodium hydroxide TS to make exactly Discard the first 5 mL of the filtrate, pipet 5 mL of the sub-
100 mL. Pipet 10 mL of this solution, add dilute sodium hy- sequent filtrate, add exactly 10 mL of the internal standard
droxide TS to make exactly 50 mL, and use this solution as solution, add the mobile phase to make 100 mL, and use
the sample solution. Separately, weigh accurately about 60 this solution as the sample solution. Separately, weigh accu-
mg of azosemide for assay, previously dried at 1059C for 3 rately about 60 mg of azosemide for assay, previously dried
hours, and dissolve in dilute sodium hydroxide TS to make at 1059 C for 3 hours, and dissolve in dilute sodium hydrox-
exactly 100 mL. Pipet 10 mL of this solution, and add dilute ide TS to make exactly 100 mL. Pipet 5 mL of this solution,
sodium hydroxide TS to make exactly 100 mL. Pipet 10 mL add exactly 10 mL of the internal standard solution, add the
of this solution, add dilute sodium hydroxide TS to make mobile phase to make 100 mL, and use this solution as the
exactly 50 mL, and use this solution as the standard solu- standard solution. Perform the test with 10 mL each of the
tion. Determine the absorbances, AT and AS, of the sample sample solution and standard solution as directed under
solution and standard solution at 274 nm as directed under Liquid Chromatography <2.01> according to the following
Ultraviolet-visible Spectrophotometry <2.24>. conditions, and calculate the ratios, QT and QS, of the peak
area of azosemide to that of the internal standard.
Amount (mg) of azosemide (C12H11ClN6O2S2)
= MS × AT/AS × V/100 Amount (mg) of azosemide (C12H11ClN6O2S2)
= M S × QT / QS
MS: Amount (mg) of azosemide for assay taken
MS: Amount (mg) of azosemide for assay taken
Dissolution <6.10> When the test is performed at 50 revo-
lutions per minute according to the Paddle method, using Internal standard solution—A solution of propyl parahy-
900 mL of 2nd fluid for dissolution test as the dissolution droxybenzoate in the mobile phase (3 in 5000).
medium, the dissolution rate in 60 minutes of 30-mg tablet Operating conditions—
and in 90 minutes of 60-mg tablet are not less than 70z, re- Detector: An ultraviolet absorption photometer (wave-
spectively. length: 280 nm).
Start the test with 1 tablet of Azosemide Tablets, with- Column: A stainless steel column 4.6 mm in inside diame-
draw not less than 20 mL of the medium at the specified ter and 15 cm in length, packed with octadecylsilanized
minute after starting the test, and filter through a mem- silica gel for liquid chromatography (5 mm in particle diame-
brane filter with a pore size not exceeding 0.5 mm. Discard ter).
the first 10 mL or more of the filtrate, pipet V mL of the Column temperature: A constant temperature of about
subsequent filtrate, and add the dissolution medium to 259C.
make exactly V? mL so that each mL contains about 33 mg Mobile phase: A mixture of 0.03 mol/L potassium dihy-
of azosemide (C12H11ClN6O2S2). Pipet 8 mL of this solu- drogen phosphate solution, acetonitrile and methanol
tion, add 0.2 mol/L sodium hydroxide TS to make exactly (55:27:18).
20 mL, and use this solution as the sample solution. Sepa- Flow rate: Adjust so that the retention time of azosemide
rately, weigh accurately about 22 mg of azosemide for is about 5 minutes.
assay, previously dried at 1059C for 3 hours, and dissolve in System suitability—
0.2 mol/L sodium hydroxide TS to make exactly 100 mL. System performance: When the procedure is run with 10
Pipet 5 mL of this solution, and add 0.2 mol/L sodium hy- mL of the standard solution under the above operating con-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2674 Official Monographs Supplement I, JP XVII

ditions, azosemide and the internal standard are eluted in retention time of benzylpenicillin, beginning after the sol-
this order with the resolution between these peaks being not vent peak.
less than 8. System suitability—
System repeatability: When the test is repeated 6 times Test for required detectability: Pipet 10 mL of the stand-
with 10 mL of the standard solution under the above operat- ard solution, and add water to make exactly 100 mL. Con-
ing conditions, the relative standard deviation of the ratio firm that the peak area of benzylpenicillin obtained with 20
of the peak area of azosemide to that of the internal stand- mL of this solution is equivalent to 7 to 13z of that with 20
ard is not more than 1.0z. mL of the standard solution.
System performance: When the procedure is run with 20
Containers and storage Containers—Tight 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 benzylpenicillin are not less than 4000
Bacitracin and 0.7 to 1.2, respectively.
System repeatability: When the test is repeated 6 times
バシトラシン
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
Add the following next to Japanese name:
area of benzylpenicillin is not more than 1.0z.

Assay Weigh accurately amounts of Benzylpenicillin


Potassium and Benzylpenicillin Potassium RS, equivalent to
about 6 × 104 Units, dissolve each in water to make exactly
20 mL, and use these solutions as the sample solution and
the standard solution, respectively. Perform the test with
Bacitracin A
exactly 20 mL each of the sample solution and standard so-
C66H103N17O16S: 1422.69
lution as directed under Liquid Chromatography <2.01> ac-
[22601-59-8]
cording to the following conditions, and determine the peak
areas, AT and AS, of benzylpenicillin in each solution.

Change the CAS No. as follows: Amount (unit) of benzylpenicillin potassium


(C16H17KN2O4S)
[1405-87-4, Bacitracin]
= M S × AT / AS

MS: Amount (unit) of benzylpenicillin potassium RS


Benzylpenicillin Potassium taken

Operating conditions—
ベンジルペニシリンカリウム
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Change the Purity (4) and Assay as follows:
Column: A stainless steel column 4.6 mm in inside diame-
Purity ter and 25 cm in length, packed with octadecylsilanized
(4) Related substances—Dissolve 40 mg of Benzylpeni- silica gel for liquid chromatography (7 mm in particle diame-
cillin Potassium in 20 mL of water, and use this solution as ter).
the sample solution. Pipet 1 mL of the sample solution, add Column temperature: A constant temperature of about
water to make exactly 100 mL, and use this solution as the 259C.
standard solution. Perform the test with exactly 20 mL each Mobile phase: A mixture of diammonium hydrogen phos-
of the sample solution and standard solution as directed phate solution (33 in 5000) and acetonitrile (19:6), adjusted
under Liquid Chromatography <2.01> according to the fol- to pH 8.0 with phosphoric acid.
lowing conditions, and determine each peak area by the au- Flow rate: Adjust so that the retention time of benzyl-
tomatic integration method: the area of the peak other than penicillin is about 7.5 minutes.
benzylpenicillin obtained from the sample solution is not System suitability—
larger than the peak area of benzylpenicillin from the stand- System performance: When the procedure is run with 20
ard solution, and the total area of the peaks other than ben- mL of the standard solution under the above operating con-
zylpenicillin from the sample solution is not larger than 3 ditions, the number of theoretical plates and the symmetry
times the peak area of benzylpenicillin from the standard factor of the peak of benzylpenicillin are not less than 2000
solution. and not more than 3.0, respectively.
Operating conditions— System repeatability: When the test is repeated 6 times
Detector, column, column temperature, mobile phase, with 20 mL of the standard solution under the above operat-
and flow rate: Proceed as directed in the operating condi- ing conditions, the relative standard deviation of the peak
tions in the Assay. area of benzylpenicillin is not more than 1.0z.
Time span of measurement: About 5 times as long as the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2675

Add the following: dissolve, add 10 mL of 2 mol/L nitric acid TS, and titrate
<2.50> with 0.005 mol/L silver nitrate VS (potentiometric
Calcium Levofolinate Hydrate titration) (not more than 0.5z).

Each mL of 0.005 mol/L silver nitrate VS


レボホリナートカルシウム水和物
= 0.177 mg of Cl

(3) Heavy metals <1.07>—Proceed with 1.0 g of Calcium


Levofolinate Hydrate according to Method 2, and perform
the test. Prepare the control solution with 2.0 mL of Stand-
ard Lead Solution (not more than 20 ppm).
(4) Platinum—Being specified separately when the drug
C20H21CaN7O7.5H2O: 601.58 is granted approval based on the Law (not more than 5
Monocalcium N-[4-({[(6S )-2-amino-5-formyl-4-oxo- ppm).
1,4,5,6,7,8-hexahydropteridin-6-yl]methyl}amino)benzoyl]- (5) Related substances—Dissolve 20 mg of Calcium
L-glutamate pentahydrate Levofolinate Hydrate in 25 mL of water, and use this solu-
[419573-16-3] tion as the sample solution. Pipet 1 mL of the sample solu-
tion, add water to make exactly 200 mL, and use this solu-
Calcium Levofolinate Hydrate contains not less tion as the standard solution. Perform the test with exactly
than 97.0z and not more than 102.0z of calcium 20 mL each of the sample solution and standard solution as
levofolinate (C20H21CaN7O7: 511.50), calculated on directed under Liquid Chromatography <2.01> according to
the anhydrous and residual solvent-free basis. the following conditions. Determine each peak area by the
automatic integration method: the area of the peak other
Description Calcium Levofolinate Hydrate occurs as a
than levofolinate obtained from the sample solution is not
white to light yellow crystalline powder.
larger than the peak area of levofolinate from the standard
It is sparingly soluble in water, and practically insoluble
solution, and the total area of the peaks other than
in methanol and in ethanol (99.5).
levofolinate from the sample solution is not larger than 5
It is hygroscopic.
times the peak area of levofolinate from the standard solu-
Optical rotation [a]25 D : -10 – -159 (0.25 g calculated
tion.
on the anhydrous and residual solvent-free basis, 0.2 mol/L
Operating conditions—
tris buffer solution (pH 8.1), 25 mL, 100 mm).
Detector, column, column temperature, mobile phase,
Identification (1) Determine the absorption spectrum of and flow rate: Proceed as directed in the operating condi-
a solution of Calcium Levofolinate Hydrate (1 in 100,000) tions in the Assay.
as directed under Ultraviolet-visible Spectrophotometry Time span of measurement: About 3 times as long as the
<2.24>, and compare the spectrum with the Reference Spec- retention time of levofolinate, beginning after the solvent
trum: both spectra exhibit similar intensities of absorption peak.
at the same wavelengths. System suitability—
(2) Determine the infrared absorption spectrum of Cal- Test for required detectability: To exactly 5 mL of the
cium Levofolinate Hydrate as directed in the potassium bro- standard solution add water to make exactly 25 mL. Con-
mide disk method under Infrared Spectrophotometry firm that the peak area of levofolinate obtained with 20 mL
<2.25>, and compare the spectrum with the Reference Spec- of this solution is equivalent to 14 to 26z of that with 20
trum: both spectra exhibit similar intensities of absorption mL of the standard solution.
at the same wave numbers. System performance: When the procedure is run with 20
(3) A solution of Calcium Levofolinate Hydrate (1 in mL of the standard solution under the above operating con-
200) responds to Qualitative Tests <1.09> (2) and (3) for cal- ditions, the number of theoretical plates and the symmetry
cium salt. factor of the peak of levofolinate are not less than 1500 and
not more than 1.5, respectively.
pH <2.54> To 0.4 g of Calcium Levofolinate Hydrate add
System repeatability: When the test is repeated 6 times
50 mL of freshly boiled and cooled water, and warm to
with 20 mL of the standard solution under the above operat-
409C, if necessary, to dissolve: the pH of the solution is be-
ing conditions, the relative standard deviation of the peak
tween 7.0 and 8.5.
area of levofolinate is not more than 2.0z.
Purity (1) Clarity and color of solution—To 0.4 g of (6) Diastereomer—Dissolve 50 mg of Calcium Levofo-
Calcium Levofolinate Hydrate add 50 mL of water, and linate Hydrate in 100 mL of water, and use this solution as
warm to 409 C, if necessary, to dissolve: the solution is the sample solution. Perform the test with 10 mL of the sam-
clear, and its absorbance at 420 nm determined as directed ple solution as directed under Liquid Chromatography
under Ultraviolet-visible Spectrophotometry <2.24> is not <2.01> according to the following conditions. Determine
more than 0.25. each peak area by the automatic integration method, and
(2) Chloride—To 0.300 g of Calcium Levofolinate Hy- calculate their amounts by the area percentage method: the
drate add 50 mL of water, warm to 409 C, if necessary, to amount of the peak of the diastereomer, having the relative

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2676 Official Monographs Supplement I, JP XVII

retention time of about 2.0 to levofolinate, is not more than Operating conditions—
0.3z. Detector: An ultraviolet absorption photometer (wave-
Operating conditions— length: 254 nm).
Detector: An ultraviolet absorption photometer (wave- Column: A stainless steel column 4.6 mm in inside diame-
length: 286 nm). ter and 15 cm in length, packed with octadecylsilanized
Column: A stainless steel column 4 mm in inside diameter silica gel for liquid chromatography (5 mm in particle diame-
and 15 cm in length, packed with human albumin chemi- ter).
cally bonded silica gel for liquid chromatography (5 mm in Column temperature: A constant temperature of about
particle diameter). 459C.
Column temperature: A constant temperature of about Mobile phase: Adjust the pH of a mixture of diluted 0.05
409C. mol/L disodium hydrogen phosphate TS (4 in 25), metha-
Mobile phase: Dissolve 3.4 g of sodium dihydrogen phos- nol and tetrabutylammonium hydroxide TS (385:110:4) to
phate dihydrate in 870 mL of water, adjust to pH 4.9 with 7.5 with phosphoric acid.
sodium hydroxide TS or phosphoric acid, and add 110 mL Flow rate: Adjust so that the retention time of folinate is
of 2-propanol and 20 mL of acetonitrile. about 10 minutes.
Flow rate: Adjust so that the retention time of System suitability—
levofolinate is about 16 minutes. System performance: Dissolve 10 mg of folic acid in 50
System suitability— mL of the mobile phase. To 5 mL of this solution add 5 mL
Test for required detectability: Dissolve 10 mg of Calcium of the standard solution. When the procedure is run with 20
Folinate RS in water to make 50 mL. To 1 mL of this solu- mL of this solution under the above operating conditions,
tion add the sample solution to make 20 mL, and use this folinate and folic acid are eluted in this order with the reso-
solution as the solution for system suitability test. Pipet 1 lution between these peaks being not less than 10.
mL of the solution for system suitability test, and add water System repeatability: When the test is repeated 6 times
to make exactly 10 mL. Confirm that the peak area of the with 20 mL of the standard solution under the above operat-
diastereomer obtained with 10 mL of this solution is equiva- ing conditions, the relative standard deviation of the peak
lent to 7 to 13z of that with 10 mL of the solution for sys- area of folinate is not more than 1.0z.
tem suitability test.
Containers and storage Containers—Tight containers.
System performance: When the procedure is run with 10
Storage—Light-resistant.
mL of the solution for system suitability test under the above
operating conditions, levofolinate and the diastereomer are
eluted in this order with the resolution between these peaks
being not less than 5. Cefixime Hydrate
System repeatability: When the test is repeated 6 times
セフィキシム水和物
with 10 mL of the solution for system suitability test under
the above operating conditions, the relative standard devia-
Change the Identification (3), Purity and Assay
tion of the peak area of the diastereomer is not more than
as follows:
2.0z.
Identification
Water <2.48> 12.0 – 17.0z (0.2 g, volumetric titration,
(3) Dissolve 50 mg of Cefixime Hydrate in 0.5 mL of a
direct titration ).
mixture of deuterated dimethylsulfoxide for nuclear mag-
Assay Weigh accurately about 10 mg each of Calcium netic resonance spectroscopy and heavy water for nuclear
Levofolinate Hydrate and Calcium Folinate RS (separately magnetic resonance spectroscopy (4:1). Determine the 1H
determine the water <2.48> in the same manner as Calcium spectrum of this solution, as directed under Nuclear Mag-
Folinate), and dissolve each in water to make exactly 25 netic Resonance Spectroscopy <2.21>, using tetramethylsi-
mL, and use these solutions as the sample solution and the lane for nuclear magnetic resonance spectroscopy as an in-
standard solution, respectively. Perform the test with ex- ternal reference compound: it exhibits a singlet signal A at
actly 20 mL each of the sample solution and standard solu- around d 4.7 ppm, and a multiplet signal B between d 6.5
tion as directed under Liquid Chromatography <2.01> ac- ppm and d 7.4 ppm. The ratio of integrated intensity of
cording to the following conditions, and determine the peak these signals, A:B, is about 1:1.
area, AT, of levofolinate with the sample solution, and the
Purity Dissolve 0.1 g of Cefixime Hydrate in 100 mL of
peak area, AS, of folinate with the standard solution.
0.1 mol/L phosphate buffer solution (pH 7.0), and use this
Amount (mg) of calcium levofolinate (C20H21CaN7O7) solution as the sample solution. Perform the test with 10 mL
= M S × AT / AS of the sample solution as directed under Liquid Chromatog-
raphy <2.01> according to the following conditions. Deter-
MS: Amount (mg) of Calcium Folinate RS taken, calcu-
mine each peak area by the automatic integration method,
lated on the anhydrous basis
and calculate their amounts by the area percentage method:
the amount of the peak other than cefixime is not more than

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2677

1.0z, and the total amount of the peaks other than mL, and adjust to pH 6.5 with diluted phosphoric acid (1 in
cefixime is not more than 2.5z. 10). To 300 mL of this solution add 100 mL of acetonitrile.
Operating conditions— Flow rate: Adjust so that the retention time of cefixime is
Detector, column, column temperature, mobile phase, about 10 minutes.
and flow rate: Proceed as directed in the operating condi- System suitability—
tions in the Assay. System performance: When the procedure is run with 10
Time span of measurement: About 3 times as long as the mL of the standard solution under the above operating con-
retention time of cefixime, beginning after the solvent peak. ditions, the number of theoretical plates and the symmetry
System suitability— factor of the peak of cefixime are not less than 4000 and not
Test for required detectability: To 1 mL of the sample so- more than 2.0, respectively.
lution add 0.1 mol/L phosphate buffer solution (pH 7.0) to System repeatability: When the test is repeated 6 times
make 100 mL, and use this solution as the solution for sys- with 10 mL of the standard solution under the above operat-
tem suitability test. Pipet 1 mL of the solution for system ing conditions, the relative standard deviation of peak area
suitability test, add 0.1 mol/L phosphate buffer solution of cefixime is not more than 2.0z.
(pH 7.0) to make exactly 10 mL. Confirm that the peak area
of cefixime obtained with 10 mL of this solution is equiva-
lent to 7 to 13z of that with 10 mL of the solution for sys- Add the following:
tem suitability test.
System performance: When the procedure is run with 10 Cefoperazone Sodium for Injection
mL of the solution for system suitability test under the above
operating conditions, the number of theoretical plates and 注射用セフォペラゾンナトリウム
the symmetry factor of the peak of cefixime are not less
than 4000 and not more than 2.0, respectively. Cefoperazone Sodium for Injection is a preparation
System repeatability: When the test is repeated 6 times for injection which is dissolved before use.
with 10 mL of the solution for system suitability test under It contains not less than 93.0z and not more than
the above operating conditions, the relative standard devia- 107.0z of the labeled potency of cefoperazone
tion of the peak areas of cefixime is not more than 2.0z. (C25H27N9O8S2: 645.67).
Assay Weigh accurately an amount of Cefixime Hydrate, Method of preparation Prepare as directed under Injec-
equivalent to about 0.1 g (potency), and dissolve in 0.1 tions, with Cefoperazone Sodium.
mol/L phosphate buffer solution (pH 7.0) to make exactly
Description Cefoperazone Sodium for Injection occurs as
100 mL. Pipet 10 mL of this solution, add 0.1 mol/L phos-
a white to yellowish white, crystalline powder or masses.
phate buffer solution (pH 7.0) to make exactly 50 mL, and
use this solution as the sample solution. Separately, weigh Identification Determine the absorption spectrum of a so-
accurately an amount of Cefixime RS, equivalent to about lution of Cefoperazone Sodium for Injection (1 in 50,000)
20 mg (potency), and dissolve in 0.1 mol/L phosphate as directed under Ultraviolet-visible Spectrophotometry
buffer solution (pH 7.0) to make exactly 20 mL. Pipet 10 <2.24>: it exhibits maxima between 226 nm and 230 nm, and
mL of this solution, add 0.1 mol/L phosphate buffer solu- between 263 nm and 267 nm.
tion (pH 7.0) to make exactly 50 mL, and use this solution
pH <2.54> The pH of a solution prepared by dissolving an
as the standard solution. Perform the test with exactly 10
amount of Cefoperazone Sodium for Injection, equivalent
mL each of the sample solution and standard solution as di-
to 1.0 g (potency) of Cefoperazone Sodium, in 4 mL of
rected under Liquid Chromatography <2.01> according to
water is between 4.5 and 6.5.
the following conditions, and determine the peak areas, AT
and AS, of cefixime in each solution. Purity (1) Clarity and color of solution—Dissolve an
amount of Cefoperazone Sodium for Injection, equivalent
Amount [mg (potency)] of cefixime (C16H15N5O7S2)
to 1.0 g (potency) of Cefoperazone Sodium, in 10 mL of
= MS × AT/AS × 5000
water: the solution is clear, and its absorbance at 400 nm,
MS: Amount [mg (potency)] of Cefixime RS taken determined as directed under Ultraviolet-visible Spectro-
photometry <2.24>, is not more than 0.22.
Operating conditions—
(2) Related substances—Dissolve an amount of Cefo-
Detector: An ultraviolet absorption photometer (wave-
perazone Sodium for Injection, equivalent to 0.1 g (po-
length: 254 nm).
tency) of Cefoperazone Sodium, in 100 mL of water, and
Column: A stainless steel column 4 mm in inside diameter
use this solution as the sample solution. Pipet 1 mL of the
and 125 mm in length, packed with octadecylsilanized silica
sample solution, add water to make exactly 50 mL, and use
gel for liquid chromatography (4 mm in particle diameter).
this solution as the standard solution. Perform the test with
Column temperature: A constant temperature of about
exactly 25 mL each of the sample solution and standard so-
409C.
lution as directed under Liquid Chromatography <2.01> ac-
Mobile phase: To 25 mL of a solution of tetrabutylam-
cording to the following conditions, and determine each
monium hydroxide TS (10 in 13) add water to make 1000

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2678 Official Monographs Supplement I, JP XVII

peak area by the automatic integration method: the peak


area of related substance I, having the relative retention Ceftizoxime Sodium
time of about 0.8 to cefoperazone, obtained from the sam-
ple solution is not larger than 2.5 times the peak area of セフチゾキシムナトリウム
cefoperazone from the standard solution, the peak area of
related substance II, having the relative retention time of Change the Purity (1) and Assay as follows:
about 1.7, from the sample solution are not larger than 3/4
Purity (1) Clarity and color of solution—Dissolve 1.0 g
times the peak area of cefoperazone from the standard solu-
of Ceftizoxime Sodium in 10 mL of water: the solution is
tion. Furthermore, the total area of the peaks other than
clear. Perform the test with this solution as directed under
cefoperazone from the sample solution is not larger than 3.5
Method for Color Matching <2.65>: the color is not more
times the peak area of cefoperazone from the standard solu-
colored than Matching Fluid M.
tion. For the peak areas of the related substances I and II,
multiply their relative response factors, 0.90 and 0.75, re- Assay Weigh accurately an amount of Ceftizoxime So-
spectively. dium and Ceftizoxime RS, equivalent to about 25 mg (po-
Operating conditions— tency), dissolve each in 0.1 mol/L phosphate buffer solu-
Detector, column, column temperature, mobile phase and tion (pH 7.0), add exactly 20 mL of the internal standard
flow rate: Proceed as directed in the operating conditions in solution, then add 0.1 mol/L phosphate buffer solution (pH
the Assay under Cefoperazone Sodium. 7.0) to make 50 mL, and use these solutions as the sample
Time span of measurement: About 3 times as long as the solution and the standard solution, respectively. Perform
retention time of cefoperazone, beginning after the solvent the test with 5 mL each of these solutions as directed under
peak. Liquid Chromatography <2.01> according to the following
System suitability— conditions, and calculate the ratios, QT and QS, of the peak
Proceed as directed in the system suitability in the Purity area of ceftizoxime to that of the internal standard.
(4) under Cefoperazone Sodium.
Amount [mg (potency)] of ceftizoxime (C13H13N5O5S2)
Water <2.48> Not more than 1.0z (3 g, volumetric titra- = MS × QT/QS × 1000
tion, direct titration).
MS: Amount [mg (potency)] of Ceftizoxime RS taken
Bacterial endotoxins <4.01> Less than 0.05 EU/mg (po-
Internal standard solution—A solution of 3-hydroxybenzoic
tency).
acid in 0.1 mol/L phosphate buffer solution (pH 7.0) (3 in
Uniformity of dosage units <6.02> It meets the require- 400).
ment of the Mass variation test. Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Foreign insoluble matter <6.06> Perform the test accord-
length: 254 nm).
ing to Method 2: it meets the requirement.
Column: A stainless steel column 4.6 mm in inside diame-
Insoluble particulate matter <6.07> It meets the require- ter and 25 cm in length, packed with octadecylsilanized
ment. silica gel for liquid chromatography (10 mm in particle di-
ameter).
Sterility <4.06> Perform the test according to the Mem-
Column temperature: A constant temperature of about
brane filtration method: it meets the requirement.
359C.
Assay Weigh accurately the mass of the contents of not Mobile phase: Dissolve 2.31 g of disodium hydrogen
less than 10 Cefoperazone Sodium for Injection. Weigh ac- phosphate dodecahydrate and 1.42 g of citric acid monohy-
curately a portion of the content, equivalent to about 0.1 g drate in 1000 mL of water, and adjust to pH 3.6 with
(potency) of Cefoperazone Sodium, and dissolve in water to diluted phosphoric acid (1 in 10) or dilute sodium hydroxide
make exactly 100 mL. Pipet 5 mL of this solution, add ex- TS. To 450 mL of this solution add 50 mL of acetonitrile.
actly 5 mL of the internal standard solution, and use this so- Flow rate: Adjust so that the retention time of
lution as the sample solution. Then, proceed as directed in ceftizoxime is about 4 minutes.
the Assay under Cefoperazone Sodium. System suitability—
System performance: When the procedure is run with 5
Amount [mg (potency)] of cefoperazone (C25H27N9O8S2)
mL of the standard solution under the above operating con-
= M S × QT / QS × 5
ditions, ceftizoxime and the internal standard are eluted in
MS: Amount [mg (potency)] of Cefoperazone RS taken this order with the resolution between these peaks being not
less than 7.0 and the symmetry factor of each peak is not
Internal standard solution—A solution of acetanilide in a
more than 1.5.
mixture of water and acetonitrile (43:7) (3 in 8000).
System repeatability: When the test is repeated 6 times
Containers and storage Containers—Hermetic containers. with 5 mL of the standard solution under the above operat-
Storage—In a cold place. ing conditions, the relative standard deviation of the ratio
of the peak area of ceftizoxime to that of the internal stand-
Shelf life 24 months after preparation.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2679

ard is not more than 1.0z. Identification (1) To a volume of Chloramphenicol and
Colistin Sodium Methanesulfonate Ophthalmic Solution,
equivalent to about 2.5 mg (potency) of Chloramphenicol,
Cellacefate and add water to make 100 mL. Determine the absorption
spectrum of this solution as directed under Ultraviolet-
セラセフェート visible Spectrophotometry <2.24>, using water as a blank: it
exhibits a maximum between 276 nm and 280 nm.
Change the Identification as follows: (2) To a volume of Chloramphenicol and Colistin So-
dium Methanesulfonate Ophthalmic Solution, equivalent to
Identification Determine the infrared absorption spectrum
about 5 × 105 Units of Colistin Sodium Methanesulfonate
of Cellacefate as directed in the potassium bromide disk
add 0.5 mL of ninhydrin TS, boil for 1 minute, and cool: a
method under Infrared Spectrophotometry <2.25>, and
blue color develops.
compare the spectrum with the Reference Spectrum or the
spectrum of Cellacefate RS for Identification: both spectra Osmotic pressure ratio Being specified separately when the
exhibit similar intensities of absorption at the same wave drug is granted approval based on the Law.
numbers.
pH <2.54> 6.0 – 8.0

Foreign insoluble matter <6.11> It meets the requirement.


Chloramphenicol Sodium Succinate Insoluble particulate matter <6.08> It meets the require-
ment.
クロラムフェニコールコハク酸エステルナトリウム
Sterility <4.06> Perform the test according to the Mem-
Change the Purity as follows: brane filtration method: it meets the requirement.

Purity (1) Clarity and color of solution—Dissolve 1.0 g Assay Perform the test according to the Cylinder-plate
of Chloramphenicol Sodium Succinate in 10 mL of water: method as directed under Microbial Assay for Antibiotics
the solution is clear, and the absorbance at 420 nm of the <4.02> according to the following conditions.
solution determined as directed under Ultraviolet-visible (1) Chloramphenicol
Spectrophotometry <2.24> is not more than 0.30. (i) Test organism—Kocuria rhizophila ATCC 9341
(2) Heavy metals <1.07>—Proceed with 1.0 g of Chlo- (ii) Agar media for base layer and seed layer—Use the
ramphenicol Sodium Succinate according to Method 2, and medium ii in 3) under (1) Agar media for seed and base
perform the test. Prepare the control solution with 2.0 mL layer.
of Standard Lead Solution (not more than 20 ppm). (iii) Agar medium for transferring test organisms—Use
the medium i in 2) under (2) Agar media for transferring
test organisms.
Add the following: (iv) Liquid medium for suspending test organisms—Use
the medium (2) Liquid media for suspending test organisms
Chloramphenicol and Colistin of 3.2. Culture media.
(v) Standard solutions—Weigh accurately an amount of
Sodium Methanesulfonate Chloramphenicol RS, equivalent to about 20 mg (potency),
Ophthalmic Solution dissolve in 2 mL of ethanol (95), add phosphate buffer solu-
tion (pH 6.0) to make exactly 20 mL, and use this solution
クロラムフェニコール・コリスチンメタンスルホン酸ナト as the standard stock solution. Keep the standard stock so-
リウム点眼液 lution at 159C or below, and use within 30 days. Take ex-
actly a suitable amount of the standard stock solution be-
Chloramphenicol and Colistin Sodium Methanesul- fore use, add phosphate buffer solution (pH 6.0) to make
fonate Ophthalmic Solution is an aqueous ophthalmic solutions so that each mL contains 100 mg (potency) and 25
preparation. mg (potency), and use these solutions as the high concentra-
It contains not less than 90.0z and not more than tion standard solution and the low concentration standard
120.0z of the labeled potency of chloramphenicol solution, respectively.
(C11H12Cl2N2O5: 323.13) and labeled Units of colistin (vi) Sample solutions—Weigh accurately an amount of
A (C53H100N16O13: 1169.46). Chloramphenicol and Colistin Sodium Methanesulfonate
Ophthalmic Solution, equivalent to about 10 mg (potency)
Method of preparation Prepare as directed under Oph-
of Chloramphenicol, add phosphate buffer solution (pH
thalmic Liquids and Solutions, with Chloramphenicol and
6.0) to make exactly 100 mL, and filter, if necessary. Take
Colistin Sodium Methanesulfonate.
exactly a suitable amount of this solution, add phosphate
Description Chloramphenicol and Colistin Sodium buffer solution (pH 6.0) to make solutions so that each mL
Methanesulfonate Ophthalmic Solution is a clear, colorless contains 100 mg (potency) and 25 mg (potency), and use
to pale yellow liquid. these solutions as the high concentration sample solution

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2680 Official Monographs Supplement I, JP XVII

and the low concentration sample solution, respectively. Chloramphenicol and Colistin Sodium Methanesulfonate
(2) Colistin Sodium Methanesulfonate Ophthalmic Solution, equivalent to about 1 × 105 Units of
(i) Test organism—Bordetella bronchiseptica ATCC Colistin Sodium Methanesulfonate, add phosphate buffer
4617 solution (pH 6.0) to make a solution so that each mL con-
(ii) Agar medium for base layer— tains 1000 Units, and use this solution as the high concen-
Casein peptone 17.0 g tration sample solution. Pipet 5 mL of the high concentra-
Sodium chloride 5.0 g tion sample solution, add phosphate buffer solution (pH
Glucose 2.5 g 6.0) to make a solution so that each mL contains 250 Units,
Soybean peptone 3.0 g and use this solution as the low concentration sample solu-
Dipotassium hydrogen phosphate 2.0 g tion.
Agar 20.0 g
Containers and storage Containers—Tight containers.
Water 1000 mL
Storage—At a temperature between 29
C and 89C.
Mix all the ingredients, then add a suitable amount of
sodium hydroxide TS so that the pH of the medium
will be 7.2 to 7.3 after sterilization, and sterile.
(iii) Agar medium for seed layer— Clarithromycin
Casein peptone 17.0 g
クラリスロマイシン
Glucose 2.5 g
Soybean peptone 3.0 g
Delete the Identification (4) and change the Op-
Sodium chloride 5.0 g
tical rotation, Purity, and Assay as follows:
Polysorbate 80 10.0 g
Dipotassium hydrogen phosphate 2.5 g Optical rotation <2.49> [a]20
D : -96 – -1069(0.25 g calcu-
Agar 12.0 g lated on the anhydrous basis, acetone, 25 mL, 100 mm).
Water 1000 mL
Purity (1) Heavy metals <1.07>—Proceed with 2.0 g of
Mix all the ingredients, then add a suitable amount of
Clarithromycin according to Method 4, and perform the
sodium hydroxide TS so that the pH of the medium
test. Prepare the control solution with 2.0 mL of Standard
will be 7.2 to 7.3 after sterilization, and sterile.
Lead Solution (not more than 10 ppm).
(iv) Agar medium for transferring test organisms—Use
(2) Related substances—Weigh accurately about 0.1 g
the medium i in 2) under (2) Agar media for transferring
of Clarithromycin, dissolve in the mobile phase to make ex-
test organisms.
actly 20 mL, and use this solution as the sample solution.
(v) Preparation of test organism and seeded agar
Separately, weigh accurately about 10 mg of Clarithromycin
layer—Cultivate the test organism on the slant of the agar
RS, dissolve in the mobile phase to make exactly 20 mL,
medium for transferring test organism at 32 to 379C for 16
and use this solution as the standard solution. Perform the
to 24 hours. Subcultures at least three times. Cultivate the
test with exactly 10 mL each of the sample solution and
grown organism on the slant of the agar medium for trans-
standard solution as directed under Liquid Chromatogra-
ferring test organism at 32 to 379C for 16 to 24 hours, add a
phy <2.01> according to the following conditions, and deter-
suitable amount of water to the grown organism, and sus-
mine the each peak area by the automatic integration
pend. Adjust the suspension so that the transmittance at 660
method: the amount of each related substance calculated on
nm is 60z as directed under Ultraviolet-visible Spectropho-
the anhydrous basis is not more than 2.0z, and the total
tometry <2.24> using a spectrophotometer or a photoelectric
amount of them is not more than 5.0z. For these calcula-
photometer, and use this suspension as the test organism
tions, exclude any peak with an area of less than 0.05z.
suspension. Keep the test organism suspension at 159C or
below, and use within 3 days. Before use, dissolve 0.13 mL Amount (z) of each related substance calculated
of the test organism suspension, add it to 100 mL of agar on the anhydrous basis
medium for seed previously cooled at 489C, mix thor- = MS/MT × AT/AS × 100
oughly, and use this as the seeded agar layer.
Total amount (z) of the related substances calculated
(vi) Standard solutions—Weigh accurately an amount
on the anhydrous basis
of Colistin Sodium Methanesulfonate RS, equivalent to
= MS/MT × SAT/AS × 100
about 1 × 106 Units, dissolve in phosphate buffer solution
(pH 6.0) to make exactly 100 mL, and use this solution as MS: Amount (mg) of Clarithromycin RS taken
the standard stock solution. Keep the standard stock solu- MT: Amount (mg) of Clarithromycin taken, calculated on
tion at 109 C or below, and use within 7 days. Take exactly a the anhydrous basis
suitable amount of the standard stock solution before use, AS: Peak area of clarithromycin obtained with the stand-
add phosphate buffer solution (pH 6.0) to make solutions ard solution
so that each mL contains 1000 Units and 250 Units, and use AT: Peak area of each related substance obtained with the
these solutions as the high concentration standard solution sample solution
and the low concentration standard solution, respectively. SAT: Total area of the peaks other than clarithromycin
(vii) Sample solutions—Weigh accurately an amount of obtained with the sample solution

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2681

Operating conditions— System suitability—


Detector, column, column temperature, mobile phase, System performance: When the procedure is run with 10
and flow rate: Proceed as directed in the operating condi- mL of the standard solution under the above operating con-
tions in the Assay. ditions, clarithromycin and the internal standard are eluted
Time span of measurement: About 5 times as long as the in this order with the resolution between these peaks being
retention time of the main peak, beginning from 2 minutes not less than 3.
after injection of the sample solution. System repeatability: When the test is repeated 6 times
System suitability— with 10 mL of the standard solution under the above operat-
Test for required detectability: To exactly 2 mL of the ing conditions, the relative standard deviation of the ratios
standard solution add the mobile phase to make exactly 10 of the peak area of clarithromycin to that of the internal
mL, and use this solution as the solution for system suitabil- standard is not more than 2.0z.
ity test. Pipet 1 mL of the solution for system suitability
test, add the mobile phase to make exactly 10 mL. Pipet 2.5
mL of this solution, add the mobile phase to make exactly Add the following:
10 mL. Confirm that the peak area of clarithromycin ob-
tained with 10 mL of this solution is equivalent to 0.25 – Clomipramine Hydrochloride
0.75z of that with 10 mL of the standard solution.
System performance: When the procedure is run with 10 Tablets
mL of the solution for system suitability test under the above
クロミプラミン塩酸塩錠
operating conditions, the number of theoretical plates and
the symmetry factor of the peak of clarithromycin are not
Clomipramine Hydrochloride Tablets contain not
less than 2500 and not more than 2.5, respectively.
less than 92.0z and not more than 108.0z of the
System repeatability: When the test is repeated 6 times
labeled amount of clomipramine hydrochloride
with 10 mL of the solution for system suitability test under
(C19H23ClN2.HCl: 351.31).
the above operating conditions, the relative standard devia-
tion of the peak area of clarithromycin is not more than Method of preparation Prepare as directed under Tablets,
3.0z. with Clomipramine Hydrochloride.

Assay Weigh accurately an amount of Clarithromycin and Identification To a portion of powdered Clomipramine
Clarithromycin RS, equivalent to about 50 mg (potency), Hydrochloride Tablets, equivalent to 50 mg of Clomipra-
and dissolve each in the mobile phase to make exactly 10 mine Hydrochloride, add a suitable amount of 0.1 mol/L
mL. Pipet 2 mL each of these solutions, add exactly 2 mL hydrochloric acid TS, shake thoroughly, and add 0.1 mol/L
of the internal standard solution, add the mobile phase to hydrochloric acid TS to make 250 mL. Centrifuge this solu-
make 20 mL, and use these solutions as the sample solution tion, and to 10 mL of the supernatant liquid add 0.1 mol/L
and the standard solution. Perform the test with 10 mL each hydrochloric acid TS to make 100 mL. Determine the ab-
of the sample solution and standard solution as directed sorption spectrum of this solution as directed under Ultravi-
under Liquid Chromatography <2.01> according to the fol- olet-visible Spectrophotometry <2.24>: it exhibits a maxi-
lowing conditions, and calculate the ratios, QT and QS, of mum between 250 nm and 254 nm.
the peak area of clarithromycin to that of the internal stand-
Uniformity of dosage unit <6.02> Perform the test accord-
ard.
ing to the following method: it meets the requirement of the
Amount [mg (potency)] of clarithromycin (C38H69NO13) Content uniformity test.
= MS × QT/QS × 1000 To 1 tablet of Clomipramine Hydrochloride Tablets add
V/5 mL of a mixture of methanol and 0.1 mol/L hydro-
MS: Amount [mg (potency)] of Clarithromycin RS taken
chloric acid TS (3:1), sonicate to disintegrate the tablet, and
Internal standard solution—A solution of butyl parahy- shake thoroughly for 30 minutes. To this solution add 3V/5
droxybenzoate in the mobile phase (1 in 20,000). mL of methanol, shake for 15 minutes, and add methanol
Operating conditions— to make exactly V mL so that each mL contains about
Detector: An ultraviolet absorption photometer (wave- 0.1 mg of clomipramine hydrochloride (C19H23ClN2.HCl).
length: 210 nm). Centrifuge this solution, and use the supernatant liquid as
Column: A stainless steel column 4 mm in inside diameter the sample solution. Then, proceed as directed in the Assay.
and 15 cm in length, packed with octadecylsilanized silica
Amount (mg) of clomipramine hydrochloride
gel for liquid chromatography (5 mm in particle diameter).
(C19H23ClN2.HCl)
Column temperature: A constant temperature of about
= MS × AT/AS × V/250
509C.
Mobile phase: A mixture of diluted 0.2 mol/L potassium MS: Amount (mg) of clomipramine hydrochloride for
dihydrogen phosphate TS (1 in 3) and acetonitrile (13:7). assay taken
Flow rate: Adjust so that the retention time of
Dissolution <6.10> When the test is performed at 50 revo-
clarithromycin is about 8 minutes.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2682 Official Monographs Supplement I, JP XVII

lutions per minute according to the Paddle method, using Column: A stainless steel column 4.6 mm in inside diame-
900 mL of water as the dissolution medium, the dissolution ter and 25 cm in length, packed with octadecylsilanized
rates in 45 minutes of 10-mg tablet and in 90 minutes of silica gel for liquid chromatography (10 mm in particle di-
25-mg tablet are not less than 80z, respectively. ameter).
Start the test with 1 tablet of Clomipramine Hydrochlo- Column temperature: A constant temperature of about
ride Tablets, withdraw not less than 20 mL of the medium 259C.
at the specified minute after starting the test, and filter Mobile phase: Dissolve 2 g of sodium 1-octanesulfonate
through a membrane filter with a pore size not exceeding in 300 mL of water, and add 450 mL of methanol, 250 mL
0.45 mm. Discard the first 10 mL or more of the filtrate, of acetonitrile and 1 mL of 0.5 mol/L sulfuric acid TS.
pipet V mL of the subsequent filtrate, add water to make Flow rate: Adjust so that the retention time of clomipra-
exactly V? mL so that each mL contains about 11 mg of mine is about 13 minutes.
clomipramine hydrochloride (C19H23ClN2.HCl), and use System suitability—
this solution as the sample solution. Separately, weigh accu- System performance: When the procedure is run with 20
rately about 28 mg of clomipramine hydrochloride for mL of the standard solution under the above operating con-
assay, previously dried at 1059C for 3 hours, and dissolve in ditions, the number of theoretical plates and the symmetry
water to make exactly 100 mL. Pipet 4 mL of this solution, factor of the peak of clomipramine are not less than 3000
add water to make exactly 100 mL, and use this solution as and not more than 1.5, respectively.
the standard solution. Determine the absorbances, AT and System repeatability: When the test is repeated 6 times
AS, of the sample solution and standard solution at 252 nm with 20 mL of the standard solution under the above operat-
as directed under Ultraviolet-visible Spectrophotometry ing conditions, the relative standard deviation of the peak
<2.24>. area of clomipramine is not more than 1.0z.

Dissolution rate (z) with respect to the labeled amount Containers and storage Containers—Tight containers.
of clomipramine hydrochloride (C19H23ClN2.HCl)
= MS × AT/AS × V?/V × 1/C × 36
Add the following:
MS: Amount (mg) of clomipramine hydrochloride for
assay taken
C: Labeled amount (mg) of clomipramine hydrochloride Clotiazepam Tablets
(C19H23ClN2.HCl) in 1 tablet
クロチアゼパム錠
Assay Weigh accurately the mass of not less than 20
Clomipramine Hydrochloride Tablets, and powder. Weigh Clotiazepam Tablets contain not less than 95.0z
accurately a portion of the powder, equivalent to about 25 and not more than 105.0z of the labeled amount of
mg of clomipramine hydrochloride (C19H23ClN2.HCl), add clotiazepam (C16H15ClN2OS: 318.82).
50 mL of a mixture of methanol and 0.1 mol/L hydrochlo-
Method of preparation Prepare as directed under Tablets,
ric acid TS (3:1), sonicate, and shake thoroughly for 30
with Clotiazepam.
minutes. To this solution add 150 mL of methanol, shake
for 15 minutes, and add methanol to make exactly 250 mL. Identification Determine the absorption spectrum of the
Centrifuge this solution, and use the supernatant liquid as sample solution obtained in the Assay as directed under
the sample solution. Separately, weigh accurately about 25 Ultraviolet-visible Spectrophotometry <2.24>: it exhibits a
mg of clomipramine hydrochloride for assay, previously maximum between 260 nm and 264 nm.
dried at 1059C for 3 hours, dissolve in 50 mL of a mixture
Uniformity of dosage unit <6.02> Perform the test accord-
of methanol and 0.1 mol/L hydrochloric acid TS (3:1), add
ing to the following method: it meets the requirement of the
methanol to make exactly 250 mL, and use this solution as
Content uniformity test.
the standard solution. Perform the test with exactly 20 mL
To 1 tablet of Clotiazepam Tablets add 35 mL of 0.1
each of the sample solution and standard solution as di-
mol/L hydrochloric acid TS, stir until the tablet is com-
rected under Liquid Chromatography <2.01> according to
pletely disintegrated, stir for a further 10 minutes, and add
the following conditions, and determine the peak areas, AT
0.1 mol/L hydrochloric acid TS to make exactly 50 mL.
and AS, of clomipramine in each solution.
Centrifuge this solution, pipet V mL of the supernatant liq-
Amount (mg) of clomipramine hydrochloride uid, add 0.1 mol/L hydrochloric acid TS to make exactly
(C19H23ClN2.HCl) V? mL so that each mL contains about 10 mg of clotiazepam
= MS × AT/AS (C16H15ClN2OS), and use this solution as the sample solu-
tion. Then, proceed as directed in the Assay.
MS: Amount (mg) of clomipramine hydrochloride for
assay taken Amount (mg) of clotiazepam (C16H15ClN2OS)
= MS × AT/AS × V?/V × 1/50
Operating conditions—
Detector: An ultraviolet absorption photometer (wave- MS: Amount (mg) of clotiazepam for assay taken
length: 254 nm).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2683

Dissolution <6.10> When the test is performed at 50 revo-


lutions per minute according to the Paddle method, using Cloxacillin Sodium Hydrate
900 mL of 1st fluid for dissolution test as the dissolution
medium, the dissolution rate in 45 minutes of Clotiazepam クロキサシリンナトリウム水和物
Tablets is not less than 80z.
Start the test with 1 tablet of Clotiazepam Tablets, with- Change the Purity (4) as follows:
draw not less than 20 mL of the medium at the specified
Purity
minute after starting the test, and filter through a mem-
(4) Related substances—Dissolve 50 mg of Cloxacillin
brane filter with a pore size not exceeding 0.45 mm. Discard
Sodium Hydrate in 50 mL of the mobile phase, and use this
the first 10 mL or more of the filtrate, pipet V mL of the
solution as the sample solution. Pipet 1 mL of the sample
subsequent filtrate, add the dissolution medium to make ex-
solution, add the mobile phase to make exactly 100 mL, and
actly V? mL so that each mL contains about 5.6 mg of clotia-
use this solution as the standard solution. Perform the test
zepam (C16H15ClN2OS), and use this solution as the sample
with exactly 10 mL each of the sample solution and standard
solution. Separately, weigh accurately about 28 mg of
solution as directed under Liquid Chromatography <2.01>
clotiazepam for assay, previously dried at 809C for 3 hours,
according to the following conditions, and determine each
and dissolve in ethanol (95) to make exactly 25 mL. Pipet 5
peak area by the automatic integration method: the area of
mL of this solution, and add the dissolution medium to
the peak other than cloxacillin obtained from the sample so-
make exactly 100 mL. Pipet 5 mL of this solution, add the
lution is not larger than the peak area of cloxacillin from
dissolution medium to make exactly 50 mL, and use this so-
the standard solution, and the total area of the peaks other
lution as the standard solution. Determine the absorbances,
than chloxacillin from the sample solution is not larger than
AT and AS, of the sample solution and standard solution at
3 times the peak area of cloxacillin from the standard solu-
262 nm as directed under Ultraviolet-visible Spectropho-
tion.
tometry <2.24>, using the dissolution medium as the blank.
Operating conditions—
Dissolution rate (z) with respect to the labeled amount Detector, column, column temperature, mobile phase,
of clotiazepam (C16H15ClN2OS) and flow rate: Proceed as directed in the operating condi-
= MS × AT/AS × V?/V × 1/C × 18 tions in the Assay.
Time span of measurement: About 3 times as long as the
MS: Amount (mg) of clotiazepam for assay taken
retention time of cloxacillin.
C: Labeled amount (mg) of clotiazepam (C16H15ClN2OS)
System suitability—
in 1 tablet
Test for required detectability: Pipet 1 mL of the stand-
Assay To 20 Clotiazepam Tablets add 350 mL of 0.1 ard solution, and add the mobile phase to make exactly 10
mol/L hydrochloric acid TS, stir until the tablets are com- mL. Confirm that the peak area of cloxacillin obtained with
pletely disintegrated, stir for a further 10 minutes, and add 10 mL of this solution is equivalent to 7 to 13z of that with
0.1 mol/L hydrochloric acid TS to make exactly 500 mL. 10 mL of the standard solution.
Centrifuge this solution, pipet V mL of the supernatant liq- System performance: When the procedure is run with 10
uid, add 0.1 mol/L hydrochloric acid TS to make exactly mL of the standard solution under the above operating con-
V? mL so that each mL contains about 10 mg of clotiazepam ditions, the number of theoretical plates and the symmetry
(C16H15ClN2OS), and use this solution as the sample solu- factor of the peak of cloxacillin are not less than 5000 and
tion. Separately, weigh accurately about 25 mg of clotiazep- not more than 1.3, respectively.
am for assay, previously dried at 809C for 3 hours, and dis- System repeatability: When the test is repeated 6 times
solve in 0.1 mol/L hydrochloric acid TS to make exactly 50 with 10 mL of the standard solution under the above operat-
mL. Pipet 2 mL of this solution, add 0.1 mol/L hydrochlo- ing conditions, the relative standard deviation of the peak
ric acid TS to make exactly 100 mL, and use this solution as area of cloxacillin is not more than 1.0z.
the standard solution. Determine the absorbances, AT and
AS, of the sample solution and standard solution at 261 nm
as directed under Ultraviolet-visible Spectrophotometry Colistin Sodium Methanesulfonate
<2.24>.
コリスチンメタンスルホン酸ナトリウム
Amount (mg) of clotiazepam (C16H15ClN2OS) in 1 tablet
= MS × AT/AS × V?/V × 1/100
Change the origin/limits of content as follows:
MS: Amount (mg) of clotiazepam for assay taken
Colistin Sodium Methanesulfonate is the sodium
Containers and storage Containers—Tight containers.
salt of colistin derivatives.
Storage—Light-resistant.
It is a mixture of colistin A sodium methanesul-
fonate and colistin B sodium methanesulfonate.
It, when dried, contains not less than 11,500 Units
and not more than 15,500 Units per mg. The unit of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2684 Official Monographs Supplement I, JP XVII

Colistin Sodium Methanesulfonate is expressed as


mass of colistin A (R = 6-methyloctanic acid, R? = Dextran 40
H; C53H100N16O13: 1169.46).
デキストラン40

Demethylchlortetracycline Add the following next to the Origin/limits of


content:
Hydrochloride
Manufacture Dextran 40 is produced by the manufactur-
デメチルクロルテトラサイクリン塩酸塩 ing method to eliminate or minimize impurities having a
possible antigenicity. The manufacturing method is verified
Change the Purity as follows: to meet the antigenicity test.
Antigenicity Dissolve 10.0 g of Dextran 40 in isotonic
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
sodium chloride solution to make 100 mL, sterilize, and use
Demethylchlortetracycline Hydrochloride according to
this solution as the sample solution. Inject 1.0 mL of the
Method 2, and perform the test. Prepare the control solu-
sample solution on 3 occasions at intervals of 2 days
tion with 2.0 mL of Standard Lead Solution (not more than
intraperitoneally to each of 4 well-nourished, healthy guinea
20 ppm).
pigs weighing 250 to 300 g. Inject 0.10 mL of horse serum
(2) Related substances—Dissolve 25 mg of Demethyl-
intraperitoneally to each of 4 guinea pigs of another group
chlortetracycline Hydrochloride in 50 mL of 0.01 mol/L
as a control. Inject 0.20 mL of the sample solution intrave-
hydrochloric acid TS, and use this solution as the sample
nously to each of 2 guinea pigs of the first group 14 days
solution. Pipet 5 mL of the sample solution, add 0.01
after the first intraperitoneal injection and into each of the
mol/L hydrochloric acid TS to make exactly 100 mL, and
remaining 2 guinea pigs 21 days after the injection, and in-
use this solution as the standard solution. Perform the test
ject 0.20 mL of horse serum intravenously in the same man-
with exactly 20 mL each of the sample solution and standard
ner into each guinea pig of the second group. Observe the
solution as directed under Liquid Chromatography <2.01>
signs of respiratory distress, collapse or death of the
according to the following conditions. Determine each peak
animals for 30 minutes after each intravenous injection and
area by the automatic integration method: the area of the
24 hours later: the animals of the first group exhibit no
peak other than demethylchlortetracycline obtained from
signs mentioned above.
the sample solution is not larger than 1.2 times the peak
All the animals of the second group exhibit symptoms of
area of demethylchlortetracycline from the standard solu-
respiratory distress or collapse and not less than 3 animals
tion, and the total area of the peaks other than demethyl-
are killed.
chlortetracycline from the sample solution is not larger than
2 times the peak area of demethylchlortetracycline from the
Delete the Antigenicity:
standard solution.
Operating conditions—
Detector, column, column temperature, mobile phase,
Delete the following Monographs:
and flow rate: Proceed as directed in the operating condi-
tions in the Assay.
Time span of measurement: About 2 times as long as the Diclofenamide
retention time of demethylchlortetracycline, beginning after
ジクロフェナミド
the solvent peak.
System suitability—
System performance: Proceed as directed in the system
suitability in the Assay. Diclofenamide Tablets
Test for required detectability: Measure 10 mL of the
ジクロフェナミド錠
standard solution, add 0.01 mol/L hydrochloric acid TS to
make 50 mL, and use this solution as the solution for sys-
tem suitability test. Pipet 5 mL of the solution for system
suitability test, and add 0.01 mol/L hydrochloric acid TS to Digitoxin
make exactly 50 mL. Confirm that the peak area of
ジギトキシン
demethylchlortetracycline obtained with 20 mL of this solu-
tion is equivalent to 7 to 13z of that with 20 mL of the solu-
tion for system suitability test.
System repeatability: When the test is repeated 6 times Digitoxin Tablets
with 20 mL of the standard solution under the above operat-
ジギトキシン錠
ing conditions, the relative standard deviation of the peak
area of demethylchlortetracycline is not more than 1.0z.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2685

Digoxin Doxorubicin Hydrochloride


ジゴキシン ドキソルビシン塩酸塩

Change the Purity (2) as follows: Change the Purity (2) and Assay as follows:
Purity Purity
(2) Related substances—Dissolve 25.0 mg of Digoxin in (2) Related substances—Dissolve 25 mg of Doxorubicin
50 mL of warm ethanol (95), cool, and add ethanol (95) to Hydrochloride in 100 mL of the mobile phase, and use this
make exactly 100 mL. Pipet 10 mL of this solution, add 10 solution as the sample solution. Pipet 2 mL of the sample
mL of water and dilute ethanol to make exactly 50 mL, and solution, add the mobile phase to make exactly 100 mL, and
use this solution as the sample solution. Separately, dissolve use this solution as the standard solution. Perform the test
exactly 5.0 mg of Gitoxin RS for Purity, previously dried with exactly 20 mL each of the sample solution and standard
under reduced pressure at 1059C for 1 hour, in a mixture of solution as directed under Liquid Chromatography <2.01>
acetonitrile and water (7:3) to make exactly 200 mL. Pipet 2 according to the following conditions, and determine each
mL of this solution, add dilute ethanol to make exactly 50 peak area by the automatic integration method: the area of
mL, and use this solution as the standard solution. Perform the peak other than doxorubicin obtained from the sample
the test with exactly 10 mL each of the sample solution and solution is not larger than 1/4 times the peak area of
standard solution as directed under Liquid Chromatogra- doxorubicin from the standard solution, and the total area
phy <2.01> according to the following conditions, and deter- of the peaks other than doxorubicin from the sample solu-
mine the peak areas, AT and AS, of gitoxin: AT is not larger tion is not larger than the peak area of doxorubicin from
than AS, and the total area of the peaks other than digoxin the standard solution.
and gitoxin from the sample solution, obtained by the area Operating conditions—
percentage method, is not more than 3z. Detector, column, column temperature, mobile phase,
Operating conditions— and flow rate: Proceed as directed in the operating condi-
Detector, column, column temperature, mobile phase, tions in the Assay.
and flow rate: Proceed as directed in the operating condi- Time span of measurement: About 3 times as long as the
tions in the Assay. retention time of doxorubicin.
Time span of measurement: About 4 times as long as the System suitability—
retention time of digoxin, beginning after the solvent peak. Test for required detectability: Pipet 1 mL of the stand-
System suitability— ard solution, and add the mobile phase to make exactly 20
Test for required detectability: Dissolve 25 mg of Digoxin mL. Confirm that the peak area of doxorubicin obtained
in 50 mL of warm ethanol (95), cool, and add ethanol (95) with 20 mL of this solution is equivalent to 3.5 to 6.5z of
to make 100 mL. To 10 mL of this solution add 10 mL of that with 20 mL of the standard solution.
water and dilute ethanol to make 50 mL, and use this solu- System performance: Dissolve 5 mg of Doxorubicin Hy-
tion as the solution for system suitability test. Pipet 2 mL of drochloride in 20 mL of water, add 1.5 mL of phosphoric
the solution for system suitability test, and add dilute acid, and allow to stand at room temperature for 30
ethanol to make exactly 100 mL. Pipet 5 mL of this solu- minutes. Adjust the pH of this solution to 2.5 with 2 mol/L
tion, and add dilute ethanol to make exactly 100 mL. Con- sodium hydroxide TS. When the procedure is run with 20
firm that the peak area of digoxin obtained with 10 mL of mL of this solution under the above operating conditions,
this solution is equivalent to 0.07 to 0.13z of that with 10 doxorubicinone, having the relative retention time of about
mL of the solution for system suitability test. 0.6 to doxorubicin, and doxorubicin are eluted in this order
System performance: Dissolve 25 mg of Digoxin in 50 mL with the resolution between these peaks being not less than
of warm ethanol (95), cool, and add ethanol (95) to make 5.
100 mL. To 10 mL of this solution add 5 mL of a solution System repeatability: When the test is repeated 6 times
of propyl parahydroxybenzoate in ethanol (95) (1 in 4000), with 20 mL of the standard solution under the above operat-
10 mL of water and dilute ethanol to make 50 mL. When ing conditions, the relative standard deviation of the peak
the procedure is run with 10 mL of this solution under the area of doxorubicin is not more than 2.0z.
above operating conditions, digoxin and propyl parahy-
Assay Weigh accurately amounts of Doxorubicin Hydro-
droxybenzoate are eluted in this order with the resolution
chloride and Doxorubicin Hydrochloride RS, equivalent to
between these peaks being not less than 5.
about 10 mg (potency), add exactly 5 mL of the internal
System repeatability: When the test is repeated 6 times
standard solution to each, dissolve each in the mobile phase
with 10 mL of the solution for system suitability test under
to make 100 mL, and use these solutions as the sample solu-
the above operating conditions, the relative standard devia-
tion and the standard solution, respectively. Perform the
tion of the peak area of digoxin is not more than 2.5z.
test with 10 mL each of the sample solution and standard so-
lution as directed under Liquid Chromatography <2.01> ac-
cording to the following conditions, and calculate the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2686 Official Monographs Supplement I, JP XVII

ratios, QT and QS, of the peak area of doxorubicin to that sium bromide disk method under Infrared Spectrophotome-
of the internal standard. try <2.25>, and compare the spectrum with the Reference
Spectrum or the spectrum of Doxycycline Hydrochloride
Amount [mg (potency)] of doxorubicin hydrochloride
RS: both spectra exhibit similar intensities of absorption at
(C27H29NO11.HCl)
the same wave numbers.
= MS × QT/QS × 1000
(3) Dissolve 10 mg of Doxycycline Hydrochloride Hy-
MS: Amount [mg (potency)] of Doxorubicin Hydrochlo- drate in 10 mL of water, and add silver nitrate TS: a white
ride RS taken turbidity is produced.

Internal standard solution—A solution of butyl parahy- Purity


droxybenzoate in the mobile phase (1 in 1000). (2) Related substance—Dissolve 20 mg of Doxycycline
Operating conditions— Hydrochloride Hydrate in 0.01 mol/L hydrochloric acid TS
Detector: An ultraviolet absorption photometer (wave- to make exactly 25 mL, and use this solution as the sample
length: 254 nm). solution. Separately, dissolve 20 mg of 6-epidoxycycline hy-
Column: A stainless steel column 4.6 mm in inside diame- drochloride in 0.01 mol/L hydrochloric acid TS to make ex-
ter and 25 cm in length, packed with octadecylsilanized actly 25 mL, and use this solution as the 6-epidoxycycline
silica gel for liquid chromatography (5 mm in particle diame- hydrochloride stock solution. Separately, dissolve 20 mg of
ter). metacycline hydrochloride in 0.01 mol/L hydrochloric acid
Column temperature: A constant temperature of about TS to make exactly 25 mL, and use this solution as the
259C. metacycline hydrochloride stock solution. Pipet 2 mL each
Mobile phase: Dissolve 3 g of sodium lauryl sulfate in of the 6-epidoxycycline hydrochloride stock solution and
1000 mL of diluted phosphoric acid (7 in 5000), and add the metacycline hydrochloride stock solution, add 0.01
1000 mL of acetonitrile. mol/L hydrochloric acid TS to make exactly 100 mL, and
Flow rate: Adjust so that the retention time of doxoru- use this solution as the standard solution. Perform the test
bicin is about 8 minutes. with exactly 20 mL each of the sample solution and standard
System suitability— solution as directed under Liquid Chromatography <2.01>
System performance: When the procedure is run with 10 according to the following conditions, and determine each
mL of the standard solution under the above operating con- peak area by the automatic integration method: the peak
ditions, doxorubicin and the internal standard are eluted in areas of metacycline and 6-epidoxycycline obtained from
this order with the resolution between these peaks being not the sample solution are not larger than the peak areas of
less than 5, and the symmetry factor of the peak of doxoru- them from the standard solution, respectively, and the areas
bicin is 0.8 to 1.2. of the two peaks, appeared between the solvent peak and
System repeatability: When the test is repeated 6 times metacycline and behind of doxycycline, from the sample so-
with 10 mL of the standard solution under the above operat- lution are not larger than 1/4 times the peak area of 6-
ing conditions, the relative standard deviation of the ratio epidoxycycline from the standard solution, and the total
of the peak area of doxorubicin to that of the internal area of the peaks other than doxycycline from the sample
standard is not more than 1.0z. solution is not larger than 1.5 times the peak area of 6-
epidoxycycline from the standard solution.
Operating conditions—
Doxycycline Hydrochloride Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Hydrate Column: A stainless steel column 4.6 mm in inside diame-
ter and 25 cm in length, packed with styrene-divinylbenzene
ドキシサイクリン塩酸塩水和物
copolymer for liquid chromatography (8 mm in particle di-
ameter).
Change the Identification and Purity (2) as
Column temperature: A constant temperature of about
follows:
609C.
Identification (1) Determine the absorption spectrum of Mobile phase: Take 125 mL of 0.2 mol/L potassium di-
a solution of Doxycycline Hydrochloride Hydrate in 0.01 hydrogen phosphate TS and 117 mL of 0.2 mol/L sodium
mol/L hydrochloric acid-methanol TS (1 in 74,000) as di- hydroxide TS, and add water to make 500 mL. To 400 mL
rected under Ultraviolet-visible Spectrophotometry <2.24>, of this solution add 50 mL of a solution of tetrabutylammo-
and compare the spectrum with the Reference Spectrum or nium hydrogensulfate (1 in 100), 10 mL of a solution of dis-
the spectrum of a solution of Doxycycline Hydrochloride odium dihydrogen ethylenediamine tetraacetate dihydrate (1
RS prepared in the same manner as the sample solution: in 25), 60 g of t-butyl alcohol and 200 mL of water, adjust
both spectra exhibit similar intensities of absorption at the to pH 8.0 with 2 mol/L sodium hydroxide TS, and add
same wavelengths. water to make 1000 mL.
(2) Determine the infrared absorption Spectrum of Dox- Flow rate: Adjust so that the retention time of doxycy-
ycycline Hydrochloride Hydrate as directed in the potas- cline is about 19 minutes.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2687

Time span of measurement: About 2.4 times as long as edaravone.


the retention time of doxycycline, beginning after the sol- System suitability—
vent peak. System performance: When the procedure is run with 50
System suitability— mL of the standard solution under the above operating con-
Test for required detectability: Pipet 1 mL of the stand- ditions, the number of theoretical plates and the symmetry
ard solution, and add 0.01 mol/L hydrochloric acid TS to factor of the peak of edaravone are not less than 1500 and
make exactly 20 mL. Confirm that the peak areas of 6- not more than 1.4, respectively.
epidoxycycline and metacycline obtained with 20 mL of this System repeatability: When the test is repeated 6 times
solution are equivalent to 3.5 to 6.5z of them with 20 mL of with 50 mL of the standard solution under the above operat-
the standard solution, respectively. ing conditions, the relative standard deviation of the peak
System performance: To 8 mL of the sample solution, 3 area of edaravone is not more than 2.0z.
mL of the 6-epidoxycycline hydrochloride stock solution 2) Use Edaravone Injection as the sample solution.
and 2 mL of the metacycline hydrochloride stock solution Pipet 1 mL of the sample solution, and add the mobile
add 0.01 mol/L hydrochloric acid TS to make 50 mL. When phase to make exactly 50 mL. Pipet 1 mL of this solution,
the procedure is run with 20 mL of this solution under the add the mobile phase to make exactly 20 mL, and use this
above operating conditions, metacycline, 6-epidoxycycline solution as the standard solution. Perform the test with ex-
and doxycycline are eluted in this order with the resolutions actly 10 mL each of the sample solution and standard solu-
between the peaks, metacycline and 6-epidoxycycline, and tion as directed under Liquid Chromatography <2.01> ac-
6-epidoxycycline and doxycycline, being not less than 1.3 cording to the following conditions. Determine each peak
and not less than 2.0, respectively, and the symmetry factor area by the automatic integration method: the area of the
of the peak of doxycycline is not more than 1.3. peak other than edaravone obtained from the sample solu-
System repeatability: When the test is repeated 6 times tion is not larger than 2 times the peak area of edaravone
with 20 mL of the standard solution under the above operat- obtained from the standard solution.
ing conditions, the relative standard deviations of the peak Operating conditions—
areas of metacycline and 6-epidoxycycline are not more Detector, column, column temperature, mobile phase,
than 3.0z and not more than 2.0z, respectively. and flow rate: Proceed as directed in the operating condi-
tions in the Purity (2) under Edaravone.
Time span of measurement: About 7 times as long as the
Edaravone Injection retention time of edaravone, beginning after the peak of
edaravone.
エダラボン注射液 System suitability—
System performance: When the procedure is run with 10
Change the Purity, Assay, and Containers and mL of the standard solution under the above operating con-
storage as follows: ditions, the number of theoretical plates and the symmetry
factor of the peak of edaravone are not less than 1500 and
Purity Related substance—(i) Perform the following test
not more than 1.4, respectively.
1). For preparations to which the test 2) can be applied, the
System repeatability: When the test is repeated 6 times
test 2) may be performed instead of 1).
with 10 mL of the standard solution under the above operat-
1) To a suitable amount of Edaravone Injection add the
ing conditions, the relative standard deviation of the peak
mobile phase so that each mL contains 0.3 mg of edaravone
area of edaravone is not more than 2.0z.
(C10H10N2O), and use this solution as the sample solution.
(ii) Perform the following test 1). For preparations to
Pipet 1 mL of the sample solution, and add the mobile
which the test 2) can be applied, the test 2) may be per-
phase to make exactly 50 mL. Pipet 1 mL of this solution,
formed instead of 1).
add the mobile phase to make exactly 20 mL, and use this
1) To a suitable amount of Edaravone Injection add the
solution as the standard solution. Perform the test with ex-
mobile phase so that each mL contains 0.3 mg of edaravone
actly 50 mL each of the sample solution and standard solu-
(C10H10N2O), and use this solution as the sample solution.
tion as directed under Liquid Chromatography <2.01> ac-
Pipet 1 mL of the sample solution, and add the mobile
cording to the following conditions. Determine each peak
phase to make exactly 50 mL. Pipet 1 mL of this solution,
area by the automatic integration method: the area of the
add the mobile phase to make exactly 20 mL, and use this
peak other than edaravone obtained from the sample solu-
solution as the standard solution. Perform the test with ex-
tion is not larger than 2 times the peak area of edaravone
actly 50 mL each of the sample solution and standard solu-
obtained from the standard solution.
tion as directed under Liquid Chromatography <2.01> ac-
Operating conditions—
cording to the following conditions. Determine each peak
Detector, column, column temperature, mobile phase,
area by the automatic integration method: the area of the
and flow rate: Proceed as directed in the operating condi-
peak, having the relative retention time of about 0.3 to
tions in the Purity (2) under Edaravone.
edaravone, obtained from the sample solution is not larger
Time span of measurement: About 7 times as long as the
than 4 times the peak area of edaravone obtained from the
retention time of edaravone, beginning after the peak of
standard solution, the area of the peak, having the relative

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2688 Official Monographs Supplement I, JP XVII

retention time of about 0.4 to edaravone obtained from the ditions, the number of theoretical plates and the symmetry
sample solution, is not larger than the peak area of edara- factor of the peak of edaravone are not less than 2000 and
vone from the standard solution, and the area of the peak not more than 1.4, respectively.
other than edaravone and the peaks mentioned above from System repeatability: When the test is repeated 6 times
the sample solution is not larger than 2 times the peak area with 10 mL of the standard solution under the above operat-
of edaravone from the standard solution. ing conditions, the relative standard deviation of the peak
Operating conditions— area of edaravone is not more than 2.0z.
Detector, column, and mobile phase: Proceed as directed
Assay Perform the following test 1). For preparations to
in the operating conditions in the Assay 1).
which the test 2) can be applied, the test 2) may be per-
Column temperature: A constant temperature of about
formed instead of 1).
409C.
1) To exactly V mL of Edaravone Injection add metha-
Flow rate: Adjust so that the retention time of edaravone
nol to make exactly V? mL so that each mL contains about
is about 11 minutes.
0.3 mg of edaravone (C10H10N2O). Pipet 2 mL of this solu-
Time span of measurement: About 2.5 times as long as
tion, add exactly 10 mL of the internal standard solution,
the retention time of edaravone, beginning after the solvent
then add methanol to make 20 ml, and use this solution as
peak.
the sample solution. Separately, weigh accurately about 30
System suitability—
mg of edaravone for assay, previously dried in vacuum
System performance: When the procedure is run with 50
using phosphorus (V) oxide as a desiccant for 3 hours, and
mL of the standard solution under the above operating con-
dissolve in methanol to make exactly 100 mL. Pipet 2 mL of
ditions, the number of theoretical plates and the symmetry
this solution, add exactly 10 mL of the internal standard so-
factor of the peak of edaravone are not less than 2000 and
lution, then add methanol to make 20 mL, and use this so-
not more than 1.4, respectively.
lution as the standard solution. Perform the test with 10 mL
System repeatability: When the test is repeated 6 times
each of the sample solution and standard solution as di-
with 50 mL of the standard solution under the above operat-
rected under Liquid Chromatography <2.01> according to
ing conditions, the relative standard deviation of the peak
the following conditions, and calculate the ratios, QT and
area of edaravone is not more than 2.0z.
QS, of the peak area of edaravone to that of the internal
2) Use Edaravone Injection as the sample solution.
standard.
Pipet 1 mL of the sample solution, and add the mobile
phase to make exactly 50 mL. Pipet 1 mL of this solution, Amount (mg) of edaravone (C10H10N2O)
add the mobile phase to make exactly 20 mL, and use this = MS × QT/QS × V?/V × 1/100
solution as the standard solution. Perform the test with ex-
MS: Amount (mg) of edaravone for assay taken
actly 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 ethyl aminoben-
cording to the following conditions. Determine each peak zoate in methanol (1 in 2500).
area by the automatic integration method: the area of the Operating conditions—
peak, having the relative retention time of about 0.3 to Detector: An ultraviolet absorption photometer (wave-
edaravone, obtained from the sample solution is not larger length: 240 nm).
than 4 times the peak area of edaravone obtained from the Column: A stainless steel column 4.6 mm in inside diame-
standard solution, the area of the peak, having the relative ter and 15 cm in length, packed with octadecylsilanized
retention time of about 0.4 to edaravone obtained from the silica gel for liquid chromatography (5 mm in particle diame-
sample solution , is not larger than the peak area of edara- ter).
vone from the standard solution, and the area of the peak Column temperature: A constant temperature of about
other than edaravone and the peaks mentioned above from 509C.
the sample solution is not larger than 2 times the peak area Mobile phase: A mixture of diluted dilute acetic acid (1 in
of edaravone from the standard solution. 100) and methanol (3:1), adjusted to pH 5.5 with diluted
Operating conditions— ammonia solution (28) (1 in 20).
Detector, column, and mobile phase: Proceed as directed Flow rate: Adjust so that the retention time of edaravone
in the operating conditions in the Assay 1). is about 8 minutes.
Column temperature: A constant temperature of about System suitability—
409C. System performance: When the procedure is run with 10
Flow rate: Adjust so that the retention time of edaravone mL of the standard solution under the above operating con-
is about 11 minutes. ditions, edaravone and the internal standard are eluted in
Time span of measurement: About 2.5 times as long as this order with the resolution between these peaks being not
the retention time of edaravone, beginning after the solvent less than 7.
peak. System repeatability: When the test is repeated 6 times
System suitability— with 10 mL of the standard solution under the above operat-
System performance: When the procedure is run with 10 ing conditions, the relative standard deviation of the ratio
mL of the standard solution under the above operating con- of the peak area of edaravone to that of the internal stand-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2689

ard is not more than 1.0z. more than 102.0z of entacapone (C14H15N3O5), cal-
2) To an exact volume of Edaravone Injection, equiva- culated on the dried basis.
lent to about 3 mg of edaravone (C10H10N2O) add exactly 10
Description Entacapone occurs as a yellow to greenish yel-
mL of the internal standard solution, add methanol to make
low crystalline powder.
20 mL, and use this solution as the sample solution. Sepa-
It is sparingly soluble in methanol, slightly soluble in
rately, weigh accurately about 75 mg of edaravone for
ethanol (99.5), and practically insoluble in water.
assay, previously dried in vacuum using phosphorus (V)
It shows crystal polymorphism.
oxide as a desiccant for 3 hours, and dissolve in methanol to
make exactly 50 mL. Pipet 2 mL of this solution, add ex- Identification (1) Dissolve 35 mg of Entacapone in 200
actly 10 mL of the internal standard solution, add methanol mL of methanol. To 7 mL of this solution add 0.1 mol/L
to make 20 mL, and use this solution as the standard solu- hydrochloric acid TS to make 100 mL. Determine the ab-
tion. Perform the test with 2 mL each of the sample solution sorption spectrum of this solution as directed under Ultravi-
and standard solution as directed under Liquid Chromatog- olet-visible Spectrophotometry <2.24>, using a solution pre-
raphy <2.01> according to the following conditions, and cal- pared by adding 0.1 mol/L hydrochloric acid TS to 7 mL of
culate the ratios, QT and QS, of the peak area of edaravone methanol to make 100 mL as the blank, and compare the
to that of the internal standard. spectrum with the Reference Spectrum or the spectrum of a
solution of Entacapone RS prepared in the same manner as
Amount (mg) of edaravone (C10H10N2O)
the sample solution: both spectra exhibit similar intensities
= MS × QT/QS × 1/25
of absorption at the same wavelengths.
MS: Amount (mg) of edaravone for assay taken (2) Determine the infrared absorption spectrum of En-
tacapone as directed in the potassium bromide disk method
Internal standard solution—A solution of ethyl aminoben-
under Infrared Spectrophotometry <2.25>, and compare the
zoate in methanol (1 in 500).
spectrum with the Reference Spectrum or the spectrum of
Operating conditions—
Entacapone RS: both spectra exhibit similar intensities of
Proceed as directed in the operating conditions in the
absorption at the same wave numbers.
Assay 1).
System suitability— Purity (1) Heavy metals—Dissolve 1.0 g of Entacapone
System performance: When the procedure is run with 2 in 20 mL of a mixture of methanol and N,N-dimethylfor-
mL of the standard solution under the above operating con- mamide (3:1), and use this solution as the sample solution.
ditions, edaravone and the internal standard are eluted in Separately, weigh exactly 0.400 g of lead (II) nitrate, dis-
this order with the resolution between these peaks being not solve in water to make exactly 250 mL. Before use, dilute
less than 7. this solution with water to make exactly 10 times the initial
System repeatability: When the test is repeated 6 times volume, then dilute this solution with water to make exactly
with 2 mL of the standard solution under the above operat- 10 times the initial volume. Pipet 1 mL of this solution, add
ing conditions, the relative standard deviation of the ratio a mixture of methanol and N,N-dimethylformamide (3:1) to
of the peak area of edaravone to that of the internal stand- make exactly 20 mL, and use this solution as the standard
ard is not more than 1.0z. solution. To each of the sample solution and standard solu-
tion add 2 mL of acetate buffer solution (pH 3.5), mix, add
Containers and storage Containers—Hermetic containers.
1.2 mL of thioacetamide TS, and mix immediately. Allow
Plastic containers for aqueous injections may be used.
them to stand for 2 minutes, filter separately all the amount
of each solution through a membrane filter with a pore size
of 0.45 mm, wash the membrane filters with not less than 20
Add the following:
mL of methanol, and compare the colors on the membrane
filters: the color obtained from the sample solution is not
Entacapone darker than that obtained from the standard solution (not
more than 10 ppm).
エンタカポン
(2) Halide—Being specified separately when the drug is
granted approval based on the Law.
(3) Related substances—Dissolve 50 mg of Entacapone
in 50 mL of a mixture of methanol and tetrahydrofuran
(7:3), and use this solution as the sample solution. Pipet 5
mL of the sample solution, and add a mixture of methanol
C14H15N3O5: 305.29 and tetrahydrofuran (7:3) to make exactly 50 mL. Pipet 5
(2E )-2-Cyano-3-(3,4-dihydroxy-5-nitrophenyl)-N,N-diethylprop-2- mL of this solution, and add a mixture of methanol and
enamide tetrahydrofuran (7:3) to make exactly 50 mL. Pipet 1 mL of
[130929-57-6] this solution, add a mixture of methanol and tetrahydrofu-
ran (7:3) to make exactly 10 mL, and use this solution as the
Entacapone contains not less than 98.0z and not standard solution. Perform the test with exactly 10 mL each

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2690 Official Monographs Supplement I, JP XVII

of the sample solution and standard solution as directed Amount (mg) of entacapone (C14H15N3O5)
under Liquid Chromatography <2.01> according to the fol- = M S × A T / AS
lowing conditions. Determine each peak area by the auto-
MS: Amount (mg) of Entacapone RS taken, calculated on
matic integration method: the peak area of the related sub-
the dried basis
stance A, having the relative retention time of about 0.8 to
entacapone, obtained from the sample solution is not larger Operating conditions—
than 1.5 times the peak area of entacapone obtained from Detector: An ultraviolet absorption photometer (wave-
the standard solution, the area of the peak other than en- length: 300 nm).
tacapone and the peak mentioned above from the sample Column: A stainless steel column 4.6 mm in inside diame-
solution is not larger than the peak area of entacapone from ter and 25 cm in length, packed with phenylated silica gel
the standard solution, and the total area of the peaks other for liquid chromatography (5 mm in particle diameter).
than entacapone and the related substance A, having the Column temperature: A constant temperature of about
relative retention time of about 0.8 to entacapone, from the 259C.
sample solution is not larger than 2 times the peak area of Mobile phase: Dissolve 2.34 g of sodium dihydrogen
entacapone from the standard solution. For the areas of the phosphate dihydrate in water to make 1000 mL, and adjust
peaks of related substances B and C, having the relative to pH 2.1 with phosphoric acid. To 540 mL of this solution
retention times of about 0.6 and about 1.4 to entacapone, add 440 mL of methanol and 20 mL of tetrahydrofuran.
multiply their relative response factors, 1.7 and 2.5, respec- Flow rate: 1 mL per minute.
tively. System suitability—
Operating conditions— System performance: Dissolve 5 mg of Entacapone Relat-
Detector, column, column temperature, mobile phase and ed Substance A RS for System Suitability in a mixture of
flow rate: Proceed as directed in the operating conditions in methanol and tetrahydrofuran (7:3) to make 25 mL. To 1
the Assay. mL of this solution add a mixture of methanol and tetra-
Time span of measurement: About 2.5 times as long as hydrofuran (7:3) to make 20 mL, and use this solution as
the retention time of entacapone, beginning after the sol- the solution for system suitability test. Separately, to 5 mL
vent peak. of the standard solution add a mixture of methanol and
System suitability— tetrahydrofuran (7:3) to make 50 mL. To 1 mL of this solu-
System performance: Proceed as directed in the system tion and 1 mL of the solution for system suitability test add
suitability in the Assay. a mixture of methanol and tetrahydrofuran (7:3) to make 10
Test for required detectability: Pipet 5 mL of the stand- mL. When the procedure is run with 10 mL of this solution
ard solution, add a mixture of methanol and tetrahydrofu- under the above operating conditions, the related substance
ran (7:3) to make exactly 10 mL. Confirm that the peak A and entacapone are eluted in this order with the resolu-
area of entacapone obtained with 10 mL of this solution is tion between these peaks being not less than 3.
equivalent to 35 to 65z of that obtained with 10 mL of the System repeatability: When the test is repeated 6 times
standard solution. with 10 mL of the standard solution under the above operat-
System repeatability: When the test is repeated 5 times ing conditions, the relative standard deviation of the peak
with 10 mL of the standard solution under the above operat- area of entacapone is not more than 1.0z.
ing conditions, the relative standard deviation of the peak
Containers and storage Containers—Well-closed contain-
area of entacapone is not more than 5z.
ers.
Loss on drying <2.41> Not more than 0.5z (1 g, in vacu-
Others
um, 609C, 3 hours).
Related substance A:
Residue on ignition <2.44> Not more than 0.1z (1 g). (2Z )-2-Cyano-3-(3,4-dihydroxy-5-nitrophenyl)-N,N-
diethylprop-2-enamide
Assay Weigh accurately about 50 mg each of Entacapone
and Entacapone RS (separately determine the loss on drying
<2.41> under the same conditions as Entacapone), dissolve
each in a mixture of methanol and tetrahydrofuran (7:3) to
make exactly 50 mL. Pipet 5 mL each of these solutions,
add a mixture of methanol and tetrahydrofuran (7:3) to
Related substance B:
make exactly 50 mL, and use these solutions as the sample
(2E )-2-Cyano-3-(3,4-dihydroxyphenyl)-N,N-diethylprop-2-
solution and the standard solution, respectively. Perform
enamide
the test with exactly 10 mL each of the sample solution and
standard solution as directed under Liquid Chromatogra-
phy <2.01> according to the following conditions, and deter-
mine the peak areas, AT and AS, of entacapone in each solu-
tion.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2691

Related substance C: actly V? mL so that each mL contains about 11 mg of en-


(2E )-3-(3-Bromo-4,5-dihydroxyphenyl)-2-cyano-N,N- tacapone (C14H15N3O5), and use this solution as the sample
diethylprop-2-enamide solution. Separately, weigh accurately about 22 mg of En-
tacapone RS (separately determine the loss on drying <2.41>
under the same conditions as Entacapone), add 4 mL of
methanol, dissolve by sonicating, and add the dissolution
medium to make exactly 200 mL. Pipet 5 mL of this solu-
tion, add the dissolution medium to make exactly 50 mL,
and use this solution as the standard solution. Determine
the absorbances, AT and AS, of the sample solution and
standard solution at 313 nm as directed under Ultraviolet-
Add the following:
visible Spectrophotometry <2.24>, using the dissolution me-
dium as the blank.
Entacapone Tablets
Dissolution rate (z) with respect to the labeled amount
エンタカポン錠 of entacapone (C14H15N3O5)
= MS × AT/AS × V?/V × 1/C × 45
Entacapone Tablets contain not less than 95.0z
MS: Amount (mg) of Entacapone RS taken, calculated on
and not more than 105.0z of the labeled amount of
the dried basis
entacapone (C14H15N3O5: 305.29).
C: Labeled amount (mg) of entacapone (C14H15N3O5) in 1
Method of preparation Prepare as directed under Tablets, tablet
with Entacapone.
Assay Conduct this procedure using light-resistant vessels.
Identification To 1 mL of the sample solution obtained in Weigh accurately the mass of not less than 20 Entacapone
the Assay add methanol to make 50 mL. Determine the ab- Tablets, and powder. Weigh accurately a portion of the
sorption spectrum of this solution as directed under Ultravi- powder, equivalent to about 0.1 g of entacapone
olet-visible Spectrophotometry <2.24>: it exhibits a maxi- (C14H15N3O5), add 60 mL of tetrahydrofuran, and sonicate
mum between 301 nm and 305 nm. for 3 minutes. Add 60 mL of methanol, shake for 5
minutes, and add methanol to make exactly 200 mL. Centri-
Uniformity of dosage units <6.02> Perform the Mass vari-
fuge this solution, and use the supernatant liquid as the
ation test, or the Content uniformity test according to the
sample solution. Separately, weigh accurately about 50 mg
following method: it meets the requirement.
of Entacapone RS (separately determine the loss on drying
Conduct this procedure using light-resistant vessels. To 1
<2.41> under the same conditions as Entacapone), dissolve
tablet of Entacapone Tablets add 70 mL of methanol, shake
in 30 mL of tetrahydrofuran, add methanol to make exactly
for 5 minutes, and add 60 mL of tetrahydrofuran. Sonicate
100 mL, and use this solution as the standard solution. Per-
for 3 minutes, shake for 5 minutes, and add methanol to
form the test with exactly 10 mL each of the sample solution
make exactly 200 mL. Centrifuge this solution, pipet V mL
and standard solution as directed under Liquid Chromatog-
of the supernatant liquid, add methanol to make exactly
raphy <2.01> according to the following conditions, and de-
V? mL so that each mL contains about 0.5 mg of entaca-
termine the peak areas, AT and AS, of entacapone in each
pone (C14H15N3O5), and use this solution as the sample solu-
solution.
tion. Then, proceed as directed in the Assay.
Amount (mg) of entacapone (C14H15N3O5)
Amount (mg) of entacapone (C14H15N3O5)
= M S × A T / AS × 2
= MS × AT/AS × V?/V × 2
MS: Amount (mg) of Entacapone RS taken, calculated on
MS: Amount (mg) of Entacapone RS taken, calculated on
the dried basis
the dried basis
Operating conditions—
Dissolution <6.10> When the test is performed at 50 revo-
Detector: An ultraviolet absorption photometer (wave-
lutions per minute according to the Paddle method, using
length: 300 nm).
900 mL of 0.05 mol/L potassium dihydrogen phosphate TS,
Column: A stainless steel column 4.6 mm in inside diame-
adjusted to pH 5.5 with sodium hydroxide TS, as the disso-
ter and 25 cm in length, packed with phenylated silica gel
lution medium, the dissolution rate in 30 minutes of Entaca-
for liquid chromatography (5 mm in particle diameter).
pone Tablets is not less than 80z.
Column temperature: A constant temperature of about
Start the test with 1 tablet of Entacapone Tablets, with-
259C.
draw not less than 15 mL of the medium at the specified
Mobile phase: Dissolve 2.34 g of sodium dihydrogen
minute after starting the test, and filter through a mem-
phosphate dihydrate in water, add 2 mL of phosphoric acid,
brane filter with a pore size not exceeding 0.45 mm. Discard
and add water to make 1000 mL. To 540 mL of this solution
the first 5 mL or more of the filtrate, pipet V mL of the sub-
add 440 mL of methanol and 20 mL of tetrahydrofuran.
sequent filtrate, add the dissolution medium to make ex-
Flow rate: Adjust so that the retention time of entaca-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2692 Official Monographs Supplement I, JP XVII

pone is about 12 minutes. lution as the standard stock solution. Dissolve 5 mg each of
System suitability— erythromycin B and erythromycin C in 2 mL of methanol,
System performance: To 20 mL of the standard solution add exactly 2 mL of the standard stock solution, add a mix-
add a mixture of methanol and tetrahydrofuran (7:3) to ture of phosphate buffer solution (pH 7.0) and methanol
make 50 mL, and use this solution as the solution for sys- (15:1) to make exactly 25 mL, and use this solution as the
tem suitability test. Separately, dissolve 5 mg of Entacapone standard solution. Perform the test with exactly 100 mL
Related Substance A RS for System Suitability in a mixture each of the sample solution and standard solution as di-
of methanol and tetrahydrofuran (7:3) to make 25 mL. To rected under Liquid Chromatography <2.01> according to
15 mL of this solution and 15 mL of the solution for system the following conditions, and determine each peak area by
suitability test add a mixture of methanol and tetrahydrofu- the automatic integration method: the peak areas of
ran (7:3) to make 100 mL. When the procedure is run with erythromycin B and erythromycin C from the sample solu-
10 mL of this solution under the above operating conditions, tion are not larger than those of erythromycin B and
the related substance A, having the relative retention time erythromycin C from the standard solution, respectively,
of about 0.8 to entacapone, and entacapone are eluted in and the area of the peaks other than erythromycin,
this order with the resolution between these peaks being not erythromycin B and erythromycin C from the sample solu-
less than 2.0. tion is not larger than the peak area of erythromycin from
System repeatability: When the test is repeated 6 times the standard solution.
with 10 mL of the standard solution under the above operat- Operating conditions—
ing conditions, the relative standard deviation of the peak Detector: An ultraviolet absorption photometer (wave-
area of entacapone is not more than 1.0z. length: 215 nm).
Column: A stainless steel column 4.6 mm in inside diame-
Containers and storage Containers—Tight containers.
ter and 25 cm in length, packed with styrene-divinylbenzene
Others copolymer for liquid chromatography (8 mm in particle di-
Related substance A: refer to it described in Entacapone. ameter).
Column temperature: A constant temperature of about
709C.
Epalrestat Mobile phase: Dissolve 3.5 g of dipotassium hydrogen
phosphate in water to make 100 mL, and adjust the pH to
エパルレスタット 9.0 with diluted phosphoric acid (1 in 10). To 50 mL of this
solution add 190 mL of t-butyl alcohol, 30 mL of aceto-
Change the origin/limits of content as follows: nitrile and water to make 1000 mL.
Flow rate: Adjust so that the retention time of
Epalrestat, when dried, contains not less than erythromycin is about 20 minutes.
98.0z and not more than 102.0z of epalrestat Time span of measurement: About 4 times as long as the
(C15H13NO3S2). retention time of erythromycin, beginning after the solvent
peak.
System suitability—
Erythromycin System performance: Dissolve 2 mg of N-demethylery-
thromycin in 10 mL of the standard solution. When the
エリスロマイシン procedure is run with 100 mL of this solution under the
above operating conditions, N-demethylerythromycin,
Change the Description and Purity as follows: erythromycin C, erythromycin and erythromycin B are
eluted in this order, with the resolution between the peaks
Description Erythromycin occurs as a white to light yel-
of N-demethylerythromycin and erythromycin C being not
low-white powder.
less than 0.8, and with the resolution between the peaks of
It is freely soluble in methanol and in ethanol (95), and
N-demethylerythromycin and erythromycin being not less
very slightly soluble in water.
than 5.5.
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of System repeatability: When the test is repeated 3 times
Erythromycin according to Method 4, and perform the test. with 100 mL of the standard solution under the above oper-
Prepare the control solution with 2.0 mL of Standard Lead ating conditions, the relative standard deviation of the peak
Solution (not more than 20 ppm). area of erythromycin is not more than 3.0z.
(2) Related substances—Dissolve 40 mg of Erythromy-
cin in 2 mL of methanol, add a mixture of phosphate buffer
solution (pH 7.0) and methanol (15:1) to make exactly 10
mL, and use this solution as the sample solution. Sepa-
rately, dissolve 16 mg of Erythromycin RS in 2 mL of meth-
anol, add a mixture of phosphate buffer solution (pH 7.0)
and methanol (15:1) to make exactly 10 mL, and use this so-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2693

1.8 mm thickness.
Ethanol Column temperature: Inject at a constant temperature of
about 409C, maintain the temperature for 12 minutes, then
エタノール raise to 2409C at a rate of 109C per minute, and maintain at
a constant temperature of about 2409C for 10 minutes.
Change the Purity (3) as follows: Injection port temperature: 2009C.
Detector temperature: 2809C.
Purity
Carrier gas: Helium.
(3) Volatile impurities—Pipet 500 mL of Ethanol, add
Flow rate: 35 cm per second.
150 mL of 4-methylpentan-2-ol, and use this solution as the
Split ratio: 1:20.
sample solution. Separately, to 100 mL of anhydrous metha-
System suitability—
nol add Ethanol to make exactly 50 mL. Pipet 5 mL of this
System performance: When the procedure is run with 1
solution, add Ethanol to make exactly 50 mL, and use this
mL of the standard solution (2) under the above operating
solution as the standard solution (1). Separately, to 50 mL
conditions, acetaldehyde and methanol are eluted in this
each of anhydrous methanol and acetaldehyde add Ethanol
order with the resolution between these peaks being not less
to make exactly 50 mL. To 100 mL of this solution add
than 1.5.
Ethanol to make exactly 10 mL, and use this solution as the
standard solution (2). Separately, to 150 mL of acetal add
Ethanol to make exactly 50 mL. To 100 mL of this solution
add Ethanol to make exactly 10 mL, and use this solution as Anhydrous Ethanol
the standard solution (3). Separately, to 100 mL of benzene
無水エタノール
add Ethanol to make exactly 100 mL. To 100 mL of this
solution add Ethanol to make exactly 50 mL, and use this
Change the Purity (3) as follows:
solution as the standard solution (4). Perform the test with
exactly 1 mL each of Ethanol, the sample solution and Purity
standard solutions (1), (2), (3) and (4) as directed under Gas (3) Volatile impurities—Pipet 500 mL of Anhydrous
Chromatography <2.02> according to the following condi- Ethanol, add 150 mL of 4-methylpentan-2-ol, and use this
tions, and determine the peak areas of acetaldehyde, AE, solution as the sample solution. Separately, to 100 mL of an-
benzene, BE and acetal, CE obtained with Ethanol, and the hydrous methanol add Anhydrous Ethanol to make exactly
peak area of methanol with the standard solution (1), the 50 mL. Pipet 5 mL of this solution, add Anhydrous Ethanol
peak area of acetaldehyde, AT with the standard solution to make exactly 50 mL, and use this solution as the standard
(2), the peak area of acetal, CT with the standard solution solution (1). Separately, to 50 mL each of anhydrous metha-
(3) and the peak area of benzene, BT with the standard solu- nol and acetaldehyde add Anhydrous Ethanol to make ex-
tion (4) by the automatic integration method: the peak area actly 50 mL. To 100 mL of this solution add Anhydrous
of methanol obtained with Ethanol is not larger than 1/2 Ethanol to make exactly 10 mL, and use this solution as the
times the peak area of methanol with the standard solution standard solution (2). Separately, to 150 mL of acetal add
(1). When calculate the amounts of the volatile impurities Anhydrous Ethanol to make exactly 50 mL. To 100 mL of
by the following equation, the total amount of acetaldehyde this solution add Anhydrous Ethanol to make exactly 10
and acetal is not more than 10 vol ppm as acetaldehyde, and mL, and use this solution as the standard solution (3). Sepa-
the amount of benzene is not more than 2 vol ppm. The rately, to 100 mL of benzene add Anhydrous Ethanol to
total area of the peaks other than the peak mentioned above make exactly 100 mL. To 100 mL of this solution add Anhy-
with the sample solution is not larger than the peak area of drous Ethanol to make exactly 50 mL, and use this solution
4-methylpentan-2-ol. For this calculation the peak having as the standard solution (4). Perform the test with exactly 1
the area not more than 3z that of 4-methylpentan-2-ol is mL each of Anhydrous Ethanol, the sample solution and
excluded. standard solutions (1), (2), (3) and (4) as directed under Gas
Chromatography <2.02> according to the following condi-
Total amount (vol ppm) of acetaldehyde and acetal
tions, and determine the peak areas of acetaldehyde, AE,
= (10 × AE)/(AT - AE)
benzene, BE and acetal, CE obtained with Anhydrous
+ (30×CE × 44.05)/{(CT - CE) × 118.2}
Ethanol, and the peak area of methanol with the standard
Amount (vol ppm) of benzene = 2BE/(BT - BE) solution (1), the peak area of acetaldehyde, AT with the
standard solution (2), the peak area of acetal, CT with the
If necessary, identify the peak of benzene by using a
standard solution (3) and the peak area of benzene, BT with
different stationary liquid phase and suitable chromato-
the standard solution (4) by the automatic integration
graphic conditions.
method: the peak area of methanol obtained with Anhy-
Operating conditions—
drous Ethanol is not larger than 1/2 times the peak area of
Detector: A hydrogen flame-ionization detector.
methanol with the standard solution (1). When calculate the
Column: A fused silica column 0.32 mm in inside diame-
amounts of the volatile impurities by the following equa-
ter and 30 m in length, coated with 6z cyanopropyl phenyl-
tion, the total amount of acetaldehyde and acetal is not
94z dimethyl silicone polymer for gas chromatography in

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2694 Official Monographs Supplement I, JP XVII

more than 10 vol ppm as acetaldehyde, and the amount of exactly 100 mL. Pipet 3 mL of this solution, add water to
benzene is not more than 2 vol ppm. The total area of the make exactly 1000 mL, and use this solution as the standard
peaks other than the peak mentioned above with the sample solution. Pipet 4 mL each of the sample solution and stand-
solution is not larger than the peak area of 4-methylpentan- ard solution, proceed as directed in the Assay, and perform
2-ol. For this calculation the peak having the area not more the test as directed under Ultraviolet-visible Spectropho-
than 3z that of 4-methylpentan-2-ol is excluded. tometry <2.24>. Determine the absorbances, AT and AS, of
subsequent solutions of the sample solution and standard
Total amount (vol ppm) of acetaldehyde and acetal
solution at 550 nm: the content of free amines is not more
= (10 × AE)/(AT - AE)
than 1.0z.
+ (30 × CE × 44.05)/{(CT - CE) × 118.2}
Content (z) of free amines = MS/MT × AT/AS
Amount (vol ppm) of benzene = 2BE/(BT - BE)
MS: Amount (mg) of p-Aminobenzoyl Glutamic Acid RS
If necessary, identify the peak of benzene by using a
for Purity taken
different stationary liquid phase and suitable chromato-
MT: Amount (mg) of Folic Acid taken, calculated on the
graphic conditions.
anhydrous basis
Operating conditions—
Detector: A hydrogen flame-ionization detector.
Column: A fused silica column 0.32 mm in inside diame-
ter and 30 m in length, coated with 6z cyanopropyl phenyl- Fosfomycin Calcium Hydrate
94z dimethyl silicone polymer for gas chromatography in
ホスホマイシンカルシウム水和物
1.8 mm thickness.
Column temperature: Inject at a constant temperature of
Add the following next to the Purity (2):
about 409 C, maintain the temperature for 12 minutes, then
raise to 2409C at a rate of 109C per minute, and maintain at Purity
a constant temperature of about 2409C for 10 minutes. (3) Glycol substance—Weigh accurately about 0.2 g of
Injection port temperature: 2009C. Fosfomycin Calcium Hydrate, transfer into a 250-mL
Detector temperature: 2809C. iodine flask, add 100 mL of water, and dissolve by
Carrier gas: Helium. sonicating while cooling in ice. Add exactly 50 mL of phtha-
Flow rate: 35 cm per second. late buffer solution (pH 5.8) and exactly 5 mL of sodium
Split ratio: 1:20. periodate solution (107 in 100,000), stopper, stir, and add 1
System suitability— mL of water in the receiving part. Avoid exposure to light,
System performance: When the procedure is run with 1 allow to stand in a water bath at 309C for 60 minutes, add
mL of the standard solution (2) under the above operating exactly 10 mL of a solution of potassium iodide (2 in 5)
conditions, acetaldehyde and methanol are eluted in this without haste, and titrate <2.50> with 0.01 mol/L sodium
order with the resolution between these peaks being not less thiosulfate VS (indicator: 2 mL of starch TS). Perform a
than 1.5. blank determination in the same manner, and make any
necessary correction: amount of glycol substance
(C3H7CaO5P) is not more than 1.5z.
Delete the following Monograph:
Each mL of 0.01 mol/L sodium thiosulfate VS
= 0.4854 mg of C3H7CaO5P
Fluoxymesterone
フルオキシメステロン
Fosfomycin Sodium
ホスホマイシンナトリウム
Folic Acid
Add the following next to the Purity (3):
葉酸
Purity
Change the Purity (2) as follows: (4) Glycol substance—Weigh accurately about 0.2 g of
Fosfomycin Sodium, and dissolve in 100 mL of water in a
Purity
250-mL iodine flask. Add exactly 50 mL of phthalate buffer
(2) Free amines—Pipet 30 mL of the sample solution
solution (pH 5.8) and exactly 5 mL of sodium periodate so-
obtained in the Assay, add 20 mL of dilute hydrochloric
lution (107 in 100,000), stopper, stir, and add 1 mL of water
acid and water to make exactly 100 mL, and use this solu-
in the receiving part. Allow to stand in a dark place for 90
tion as the sample solution. Separately, weigh accurately
minutes, add exactly 10 mL of a solution of potassium
about 50 mg of p-Aminobenzoyl Glutamic Acid RS for
iodide (2 in 5) without haste, and titrate <2.50> with 0.01
Purity, previously dried in a desiccator (in vacuum, silica
mol/L sodium thiosulfate VS (indicator: 2 mL of starch
gel) for 4 hours, dissolve in diluted ethanol (2 in 5) to make

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2695

TS). Perform a blank determination in the same manner, Glucose Hydrate (1 in 20) to 5 mL of boiling Fehling's TS: a
and make any necessary correction: amount of glycol sub- red precipitate is produced.◇
stance (C3H7Na2O5P) is not more than 0.5z. (2) Perform the test with 20 mL each of the sample solu-
tion and standard solution obtained in the Assay as directed
Each mL of 0.01 mol/L sodium thiosulfate VS
under Liquid Chromatography <2.01> according to the fol-
= 0.5001 mg of C3H7Na2O5P
lowing conditions: the principal peak in the chromatogram
obtained from the sample solution is similar in retention
time and size to the principal peak in the chromatogram
Gentamicin Sulfate from the standard solution.
Operating conditions—
ゲンタマイシン硫酸塩
Proceed as directed in the operating conditions in the
Assay.
Change the Purity (1) as follows:
System suitability—
Purity (1) Clarity and color of solution—Dissolve 1.0 g Proceed as directed in the system suitability in the Assay.
of Gentamicin Sulfate in 10 mL of water: the solution is
Purity (1) Clarity and color of solution—Dissolve 10.0 g
clear and its absorbance at 400 nm determined as directed
of Glucose Hydrate in 15 mL of water, and use this solution
under Ultraviolet-visible Spectrophotometry <2.24> is not
as the test solution. Perform the test with the test solution
more than 0.08.
as directed under Turbidity Measurement <2.61>: the solu-
tion is clear. Perform the test with the test solution accord-
ing to Method 2 under Methods for Color Matching <2.65>:
Add the following:
the solution is not more colored than Matching Fluid BY7.
◆(2) Heavy metals <1.07>—Proceed with 5.0 g of Glu-
Glucose Hydrate cose Hydrate according to Method 2, and perform the test.
Prepare the control solution with 2.0 mL of Standard Lead
ブドウ糖水和物
Solution (not more than 4 ppm).◆
(3) Related substances—Use the sample solution ob-
tained in the Assay as the sample solution. Pipet 1 mL of
the sample solution, add water to make exactly 250 mL, and
use this solution as the standard solution (1). Pipet 25 mL
of the standard solution (1), add water to make exactly 200
a-D-glucopyranose monohydrate: R1=H, R2=OH mL, and use this solution as the standard solution (2). Per-
b-D-glucopyranose monohydrate: R1=OH, R2=H form the test with exactly 20 mL each of the sample solu-
tion, the standard solution (1) and the standard solution (2)
C6H12O6.H2O: 198.17 as directed under Liquid Chromatography <2.01> according
D-Glucopyranose monohydrate to the following conditions. Determine each peak area by
[77938-63-7] the automatic integration method: the total area of the
peaks of maltose and isomaltose, having the relative reten-
This monograph is harmonized with the European Phar- tion time of about 0.8 to glucose, obtained from the sample
macopoeia and the U.S. Pharmacopeia. solution, is not larger than the peak area of glucose from
The corresponding part of the attributes/provisions the standard solution (1) (not more than 0.4z), and the
which are agreed as non-harmonized within the scope of the peak area of maltotriose, having the relative retention time
harmonization is marked with symbols (◆ ◆), and the cor- of about 0.7 from the sample solution, is not larger than
responding parts which are agreed as the JP local require- 1/2 times the peak area of glucose from the standard solu-
ment other than the scope of the harmonization are marked tion (1) (not more than 0.2z), and the peak area of fruc-
with symbols (◇ ◇). tose, having the relative retention time of about 1.3 from
the sample solution, is not larger than 3 times the peak area
Glucose Hydrate is the monohydrate of D- of glucose from the standard solution (2) (not more than
glucopyranose derived from starch. 0.15z), and the area of the peak other than glucose and the
It contains not less than 97.5z and not more than peaks mentioned above from the sample solution is not
102.0z of glucose [D-glucopyranose (C6H12O6: larger than 2 times the peak area of glucose from the stand-
180.16)], calculated on the anhydrous basis. ard solution (2) (not more than 0.10z). Furthermore, the
◆Description
total area of the peaks other than glucose from the sample
Glucose Hydrate occurs as white, crystals or
solution is not larger than 1.25 times the peak area of glu-
crystalline powder, and has a sweet taste.
cose from the standard solution (1) (not more than 0.5z).
It is freely soluble in water, sparingly soluble in metha-
For these calculations the peak areas not larger than the
nol, and slightly soluble in ethanol (95).◆
peak area of glucose from the standard solution (2) are ex-
Identification ◇(1) Add 2 to 3 drops of a solution of cluded (disregard limit: 0.05z).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2696 Official Monographs Supplement I, JP XVII

Operating conditions— 859C.


Detector, column, column temperature, mobile phase and Mobile phase: Water.
flow rate: Proceed as directed in the operating conditions in Flow rate: 0.3 mL per minute (the retention time of glu-
the Assay. cose is about 21 minutes).
Time span of measurement: About 1.5 times as long as System suitability—
the retention time of glucose. System performance: Dissolve 5 mg of maltose, 5 mg of
System suitability— maltotriose and 5 mg of fructose in 50 mL of water, and use
System performance: Proceed as directed in the system this solution as the solution for system suitability test.
suitability in the Assay. When the procedure is run with 20 mL each of the solution

Test for required detectability: Confirm that the peak for system suitability test and the standard solution (2) in
area of glucose obtained with 20 mL of the standard solution Purity (3) under the above operating conditions, mal-
(2) is equivalent to 8.8 to 16.3z of that with 20 mL of the totriose, maltose, glucose and fructose are eluted in this
standard solution (1). order, the relative retention times of maltotriose, maltose,
System repeatability: When the test is repeated 6 times isomaltose and fructose to glucose are about 0.7, about 0.8,
with 20 mL of the standard solution (1) under the above op- about 0.8 and about 1.3, respectively, and the resolution be-
erating conditions, the relative standard deviation of the tween the peaks of maltotriose and maltose is not less than
peak area of glucose is not more than 1.0z.◇ 1.3.
(4) Dextrin—To 1.0 g of powdered Glucose Hydrate ◇System repeatability: When the test is repeated 6 times

add 20 mL of ethanol (95), and boil under a reflux con- with 20 mL of the standard solution under the above operat-
denser: the solution is clear. ing conditions, the relative standard deviation of the peak
(5) Soluble starch and sulfite—To 7.4 g of Glucose Hy- area of glucose is not more than 1.0z.◇
drate add 15 mL of water, dissolve by heating on a water ◆Containers and storage Containers—Tight containers.◆
bath, cool, and add 25 mL of 0.05 mol/L iodine VS: a yel-
low color develops (not more than 15 ppm as SO3).

Conductivity <2.51> Dissolve 20.0 g of Glucose Hydrate in Add the following:


a fleshly boiled and cooled distilled water to make 100 mL,
and use this solution as the sample solution. Measure the Purified Glucose
conductivity of the sample solution at 25 ± 0.19 C while
gently stirring with a magnetic stirrer: not more than 20 mS・ 精製ブドウ糖
cm-1.

Water <2.48> 7.5 – 9.5z (0.25 g, volumetric titration,


direct titration).

Assay Weigh accurately about 0.33 g of Glucose Hydrate


and 0.3 g of ◆Glucose RS◆ (separately determine the water a-D-glucopyranose: R1=H, R2=OH
<2.48> in the same manner as Purified Glucose), dissolve b-D-glucopyranose: R1=OH, R2=H
separately in water to make exactly 10 mL, and use these so-
lutions as the sample solution and the standard solution, re- C6H12O6: 180.16
spectively. Perform the test with exactly 20 mL each of the D-Glucopyranose
sample solution and standard solution as directed under [50-99-7]
Liquid Chromatography <2.01> according to the following
conditions, and determine the peak areas, AT and AS, of This monograph is harmonized with the European Phar-
glucose in each solution. macopoeia and the U.S. Pharmacopeia.
The corresponding part of the attributes/provisions
Amount (g) of glucose (C6H12O6) = MS × AT/AS
which are agreed as non-harmonized within the scope of the
MS: Amount (g) of Glucose RS taken, calculated on the harmonization is marked with symbols (◆ ◆), and the cor-
anhydrous basis responding parts which are agreed as the JP local require-
ment other than the scope of the harmonization are marked
Operating conditions—
with symbols (◇ ◇).
Detector: A differential refractometer maintained at a
constant temperature (409C for example).
Purified Glucose is D-glucopyranose derived from
Column: A stainless steel column 7.8 mm in inside diame-
starch.
ter and 30 cm in length, packed with strongly acidic ion-ex-
It contains not less than 97.5z and not more than
change resin for liquid chromatography (Ca type) composed
102.0z of glucose [D-glucopyranose (C6H12O6)], cal-
with a sulfonated polystyrene cross-linked with divinylben-
culated on the anhydrous basis.
zene (degree of cross-linkage: 8z) (9 mm in particle diame-
ter). ◆Description Purified Glucose occurs as white, crystals or
Column temperature: A constant temperature of about crystalline powder, and has a sweet taste.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2697

It is freely soluble in water, and slightly soluble in metha- solution is not larger than 1.25 times the peak area of glu-
nol and in ethanol (95).◆ cose from the standard solution (1) (not more than 0.5z).
For these calculations the peak areas not larger than the
Identification ◇(1) Add 2 to 3 drops of a solution of
peak area of glucose from the standard solution (2) are ex-
Purified Glucose (1 in 20) to 5 mL of boiling Fehling's TS: a
cluded (disregard limit: 0.05z).
red precipitate is produced.◇
Operating conditions—
(2) Perform the test with 20 mL each of the sample solu-
Detector, column, column temperature, mobile phase and
tion and standard solution obtained in the Assay as directed
flow rate: Proceed as directed in the operating conditions in
under Liquid Chromatography <2.01> according to the fol-
the Assay.
lowing conditions: the principal peak in the chromatogram
Time span of measurement: About 1.5 times as long as
obtained from the sample solution is similar in retention
the retention time of glucose.
time and size to the principal peak in the chromatogram
System suitability—
from the standard solution.
System performance: Proceed as directed in the system
Operating conditions—
suitability in the Assay.
Proceed as directed in the operating conditions in the ◇
Test for required detectability: Confirm that the peak
Assay.
area of glucose obtained with 20 mL of the standard solution
System suitability—
(2) is equivalent to 8.8 to 16.3z of that with 20 mL of the
Proceed as directed in the system suitability in the Assay.
standard solution (1).
Purity (1) Clarity and color of solution—Dissolve 10.0 g System repeatability: When the test is repeated 6 times
of Purified Glucose in 15 mL of water by heating on a water with 20 mL of the standard solution (1) under the above op-
bath, and allow to cool to room temperature, and use this erating conditions, the relative standard deviation of the
solution as the test solution. Perform the test with the test peak area of glucose is not more than 1.0z.◇
solution as directed under Turbidity Measurement <2.61>: (4) Dextrin—To 1.0 g of powdered Purified Glucose
the solution is clear. Perform the test with the test solution add 20 mL of ethanol (95), and boil under a reflux con-
according to Method 2 under Methods for Color Matching denser: the solution is clear.
<2.65>: the solution is not more colored than Matching (5) Soluble starch and sulfite—To 6.7 g of Purified Glu-
Fluid BY7. cose add 15 mL of water, dissolve by heating on a water
◆(2) Heavy metals <1.07>—Proceed with 5.0 g of Puri- bath, cool, and add 25 mL of 0.05 mol/L iodine VS: a yel-
fied Glucose according to Method 2, and perform the test. low color develops (not more than 15 ppm as SO3).
Prepare the control solution with 2.0 mL of Standard Lead
Conductivity <2.51> Dissolve 20.0 g of Purified Glucose in
Solution (not more than 4 ppm).◆
a fleshly boiled and cooled distilled water to make 100 mL,
(3) Related substances—Use the sample solution ob-
and use this solution as the sample solution. Measure the
tained in the Assay as the sample solution. Pipet 1 mL of
conductivity of the sample solution at 25 ± 0.19 C while
the sample solution, add water to make exactly 250 mL, and
gently stirring with a magnetic stirrer: not more than 20 mS・
use this solution as the standard solution (1). Pipet 25 mL
cm-1.
of the standard solution (1), add water to make exactly 200
mL, and use this solution as the standard solution (2). Per- Water <2.48> Not more than 1.0z (0.5 g, volumetric titra-
form the test with exactly 20 mL each of the sample solu- tion, direct titration).
tion, the standard solution (1) and the standard solution (2)
Assay Weigh accurately about 0.3 g each of Purified Glu-
as directed under Liquid Chromatography <2.01> according
cose and ◆Glucose RS◆ (separately determine the water
to the following conditions. Determine each peak area by
<2.48> in the same manner as Purified Glucose), dissolve
the automatic integration method: the total area of the
separately in water to make exactly 10 mL, and use these so-
peaks of maltose and isomaltose, having the relative reten-
lutions as the sample solution and the standard solution, re-
tion time of about 0.8 to glucose, obtained from the sample
spectively. Perform the test with exactly 20 mL each of the
solution, is not larger than the peak area of glucose from
sample solution and standard solution as directed under
the standard solution (1) (not more than 0.4z), and the
Liquid Chromatography <2.01> according to the following
peak area of maltotriose, having the relative retention time
conditions, and determine the peak areas, AT and AS, of
of about 0.7 from the sample solution, is not larger than
glucose in each solution.
1/2 times the peak area of glucose from the standard solu-
tion (1) (not more than 0.2z), and the peak area of fruc- Amount (g) of glucose (C6H12O6) = MS × AT/AS
tose, having the relative retention time of about 1.3 from
MS: Amount (g) of Glucose RS taken, calculated on the
the sample solution, is not larger than 3 times the peak area
anhydrous basis
of glucose from the standard solution (2) (not more than
0.15z), and the area of the peak other than glucose and the Operating conditions—
peaks mentioned above from the sample solution is not Detector: A differential refractometer maintained at a
larger than 2 times the peak area of glucose from the stand- constant temperature (409 C for example).
ard solution (2) (not more than 0.10z). Furthermore, the Column: A stainless steel column 7.8 mm in inside diame-
total area of the peaks other than glucose from the sample ter and 30 cm in length, packed with strongly acidic ion-ex-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2698 Official Monographs Supplement I, JP XVII

change resin for liquid chromatography (Ca type) composed Delete the following Monograph:
with a sulfonated polystyrene cross-linked with divinylben-
zene (degree of cross-linkage: 8z) (9 mm in particle diame- Gramicidin
ter).
Column temperature: A constant temperature of about グラミシジン
859C.
Mobile phase: Water.
Flow rate: 0.3 mL per minute (the retention time of glu- Heparin Calcium
cose is about 21 minutes).
System suitability— ヘパリンカルシウム
System performance: Dissolve 5 mg of maltose, 5 mg of
maltotriose and 5 mg of fructose in 50 mL of water, and use Change the Identification (2), the Purity (8) and
this solution as the solution for system suitability test. (9) as follows:
When the procedure is run with 20 mL each of the solution
Identification
for system suitability test and the standard solution (2) in
(2) Dissolve 1 mg each of Heparin Calcium and Heparin
Purity (3) under the above operating conditions, mal-
Sodium RS for Identification in 1 mL of water, and use
totriose, maltose, glucose and fructose are eluted in this
these solutions as the sample solution and the standard solu-
order, the relative retention times of maltotriose, maltose,
tion, respectively. Perform the test with 20 mL each of the
isomaltose and fructose to glucose are about 0.7, about 0.8,
sample solution and standard solution as directed under
about 0.8 and about 1.3, respectively, and the resolution be-
Liquid Chromatography <2.01> according to the following
tween the peaks of maltotriose and maltose is not less than
conditions: the retention time for the major peak from the
1.3.
◇System repeatability: When the test is repeated 6 times
sample solution and standard solution is identical.
Operating conditions—
with 20 mL of the standard solution under the above operat-
Detector, column, column temperature, mobile phases A
ing conditions, the relative standard deviation of the peak
and B, flowing of mobile phase and flow rate: Proceed as
area of glucose is not more than 1.0z.◇
directed under the operating conditions in Purity (9).
◆Containers and storage Containers—Tight containers.◆ System suitability—
System performance: Dissolve 1.0 mg of Heparin Sodium
RS for Identification in 0.60 mL of water. Dissolve 0.10 mg
Glucose Injection of Over-sulfated Chondroitin Sulfate RS for System
Suitability in 0.20 mL of water. Dissolve 1.0 mg of derma-
ブドウ糖注射液 tan sulfate in 2.0 mL of water. To 90 mL of the solution of
Heparin Sodium RS for Identification add 30 mL each of
Change the Method of preparation, Identifica- the solutions of Over-sulfated Chondroitin Sulfate RS for
tion and Purity as follows: System Suitability and dermatan sulfate, and mix. When the
procedure is run with 20 mL of the mixture under the above
Method of preparation Prepare as directed under Injec-
operating conditions, dermatan sulfate, heparin and over-
tions, with Purified Glucose.
sulfated chondroitin sulfate are eluted in this order with the
No preservative is added.
resolution between the peaks of dermatan sulfate and hepa-
Identification Measure a volume of Glucose Injection, rin being not less than 1.0 and that between the peaks of
equivalent to 0.1 g of Purified Glucose, and, if necessary, heparin and over-sulfated chondroitin sulfate being not less
add water or evaporate on a water bath to make 2 mL. Add than 1.5.
2 to 3 drops of the solution to 5 mL of boiling Fehling’s TS:
Purity
a red precipitate is produced.
(8) Over-sulfated chondroitin sulfate—Dissolve 20 mg
Purity 5-Hydroxymethylfurfural and related substances— of Heparin Calcium in 0.60 mL of a solution of sodium 3-
Measure exactly a volume of Glucose Injection, equivalent trimethylsilylpropionate-d4 for nuclear magnetic resonance
to 2.5 g of Purified Glucose, and add water to make exactly spectroscopy in heavy water for nuclear magnetic resonance
100 mL. Determine the absorbance of this solution at 284 spectroscopy (1 in 10,000). Determine the spectrum of this
nm as directed under Ultraviolet-visible Spectrophotometry solution as directed under Nuclear Magnetic Resonance
<2.24>: it is not more than 0.80. Spectroscopy <2.21> (1H) in accordance with the following
conditions, using sodium 3-trimethylsilylpropionate-d4 for
nuclear magnetic resonance spectroscopy as an internal ref-
erence compound: it exhibits no signal corresponding to N-
acetyl proton of over-sulfated chondroitin sulfate at d 2.18
± 0.05 ppm, or the signal disappears when determining the
spectrum of the sample solutions as directed under 1H with
13C-decoupling.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2699

Operating conditions—
Spectrometer: 1.1. FT-NMR, Not less than 400 MHz. Time after injection Mobile phase A Mobile phase B
Temperature: 259C. of sample (min) (volz) (volz)
Spinning: off. 0–3 90 10
Number of data points: 32,768. 3 – 15 90 → 0 10 → 100
Spectral range: Signal of DHO ± 6.0 ppm.
Flip angle: 909.
Flow rate: 0.2 mL per minute.
Delay time: 20 seconds.
Time span of measurement: About 2 times as long as the
Dummy scans: 4.
retention time of heparin, beginning after the solvent peak.
Number of scans: SN ratio of the signal of N-acetyl pro-
System suitability—
ton of heparin is not less than 1000.
Test for required detectability: Dissolve 10 mg of Heparin
Window function: Exponential function (Line broaden-
Sodium RS for Identification in 0.40 mL of water, and use
ing factor = 0.2 Hz).
this solution as the heparin sodium standard stock solution.
System suitability—
Separately, dissolve 0.10 mg of Over-sulfated Chondroitin
System performance: Dissolve 20 mg of Heparin Calcium
Sulfate RS for System Suitability in 0.20 mL of water, and
in 0.40 mL of a solution of sodium 3-trimethylsilyl-
use this solution as the over-sulfated chondroitin sulfate
propionate-d4 for nuclear magnetic resonance spectroscopy
standard solution. To 60 mL of the heparin sodium standard
in heavy water for nuclear magnetic resonance spectroscopy
stock solution add 3 mL of the over-sulfated chondroitin
(1 in 10,000). Dissolve 0.10 mg of Over-sulfated Chondroi-
sulfate standard solution and 12 mL of water, and mix.
tin Sulfate RS for System Suitability in 1.0 mL of a solution
When the procedure is run with 20 mL of the mixture under
of sodium 3-trimethylsilylpropionate-d4 for nuclear mag-
the above operating conditions, it exhibits an over-sulfated
netic resonance spectroscopy in heavy water for nuclear
chondroitin sulfate peak.
magnetic resonance spectroscopy (1 in 10,000). To the solu-
System performance: To 120 mL of the heparin sodium
tion of heparin calcium add 0.20 mL of the solution of
standard stock solution add 30 mL of the over-sulfated
Over-sulfated Chondroitin Sulfate RS for System Suitabil-
chondroitin sulfate standard solution, mix and use this solu-
ity. When determining the spectrum of this solution under
tion as the solution for system suitability test. When the
the above operating conditions, it exhibits the signal of N-
procedure is run with 20 mL of the solution for system
acetyl proton of heparin and the signal of N-acetyl proton
suitability test under the above operating conditions, hepa-
of over-sulfated chondroitin sulfate at d 2.04 ± 0.02 ppm
rin and oversulfated chondroitin sulfate are eluted in this
and d 2.18 ± 0.05 ppm, respectively.
order with the resolution between these peaks being not less
(9) Related substances—Dissolve 2.0 mg of Heparin
than 1.5.
Calcium in 0.1 mL of water, and perform the test with ex-
System repeatability: When the test is repeated 6 times
actly 20 mL of this solution as directed under Liquid Chro-
with 20 mL of the solution for system suitability test under
matography <2.01> according to the following conditions: it
the above operating conditions, the relative standard devia-
exhibits no peaks after the heparin peak.
tion of the peak area of over-sulfated chondroitin sulfate is
Operating conditions—
not more than 2.0z.
Detector: An ultraviolet absorption photometer (wave-
length: 202 nm).
Column: A stainless steel column 2.0 mm in inside diame-
ter and 7.5 cm in length, packed with synthetic polymer for Heparin Sodium
liquid chromatography to which diethylaminoethyl group
ヘパリンナトリウム
binds (10 mm in particle diameter).
Column temperature: A constant temperature of about
Change the Identification, the Purity (6) and (7)
359C.
as follows:
Mobile phase A: Dissolve 0.4 g of sodium dihydrogen
phosphate dihydrate in 1000 mL of water, and adjust to pH Identification Dissolve 1 mg each of Heparin Sodium and
3.0 with diluted phosphoric acid (1 in 10). Heparin Sodium RS for Identification in 1 mL of water,
Mobile phase B: Dissolve 0.4 g of sodium dihydrogen and use these solutions as the sample solution and the stand-
phosphate dihydrate and 106.4 g of lithium perchlorate in ard solution, respectively. Perform the test with 20 mL each
1000 mL of water, and adjust to pH 3.0 with diluted phos- of the sample solution and standard solution as directed
phoric acid (1 in 10). under Liquid Chromatography <2.01> according to the fol-
Flowing of mobile phase: Control the gradient by mixing lowing conditions: the retention time for the major peak
the mobile phases A and B as directed in the following from the sample solution and standard solution is identical.
table. Operating conditions—
Detector, column, column temperature, mobile phases A
and B, flowing of mobile phase and flow rate: Proceed as
directed under the operating conditions in Purity (7).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2700 Official Monographs Supplement I, JP XVII

System suitability— trum of this solution under the above operating conditions,
System performance: Dissolve 1.0 mg of Heparin Sodium it exhibits the signal of N-acetyl proton of heparin and the
RS for Identification in 0.60 mL of water. Dissolve 0.10 mg signal of N-acetyl proton of over-sulfated chondroitin sul-
of Over-sulfated Chondroitin Sulfate RS for System fate at d 2.04 ± 0.02 ppm and d 2.15 ± 0.02 ppm, respec-
Suitability in 0.20 mL of water. Dissolve 1.0 mg of derma- tively.
tan sulfate in 2.0 mL of water. To 90 mL of the solution of (7) Related substances—Dissolve 2.0 mg of Heparin So-
Heparin Sodium RS for Identification add 30 mL each of dium in 0.1 mL of water and perform the test with exactly
the solutions of Over-sulfated Chondroitin Sulfate RS for 20 mL of this solution as directed under Liquid Chromatog-
System Suitability and dermatan sulfate, and mix. When the raphy <2.01> according to the following conditions: it exhib-
procedure is run with 20 mL of the mixture under the above its no peaks after the heparin peak.
operating conditions, dermatan sulfate, heparin and over- Operating conditions—
sulfated chondroitin sulfate are eluted in this order with the Detector: An ultraviolet absorption photometer (wave-
resolution between the peaks of dermatan sulfate and hepa- length: 202 nm).
rin being not less than 1.0 and that between the peaks of Column: A stainless steel column 2.0 mm in inside diame-
heparin and over-sulfated chondroitin sulfate being not less ter and 7.5 cm in length, packed with synthetic polymer for
than 1.5. liquid chromatography to which diethylaminoethyl group
binds (10 mm in particle diameter).
Purity
Column temperature: A constant temperature of about
(6) Over-sulfated chondroitin sulfate—Dissolve 20 mg
359C.
of Heparin Sodium in 0.60 mL of a solution of sodium 3-
Mobile phase A: Dissolve 0.4 g of sodium dihydrogen
trimethylsilylpropionate-d4 for nuclear magnetic resonance
phosphate dihydrate in 1000 mL of water, and adjust to pH
spectroscopy in heavy water for nuclear magnetic resonance
3.0 with diluted phosphoric acid (1 in 10).
spectroscopy (1 in 10,000). Determine the spectrum of this
Mobile phase B: Dissolve 0.4 g of sodium dihydrogen
solution as directed under Nuclear Magnetic Resonance
phosphate dihydrate and 106.4 g of lithium perchlorate in
Spectroscopy <2.21> (1H) in accordance with the following
1000 mL of water, and adjust to a pH of 3.0 with diluted
conditions, using sodium 3-trimethylsilylpropionate-d4 for
phosphoric acid (1 in 10).
nuclear magnetic resonance spectroscopy as an internal ref-
Flowing of mobile phase: Control the gradient by mixing
erence compound: it exhibits no signal corresponding to N-
the mobile phases A and B as directed in the following
acetyl proton of over-sulfated chondroitin sulfate at d 2.15
table.
± 0.02 ppm, or the signal disappears when determining the
spectrum of the sample solutions as directed under 1H with
13C-decoupling. Time after injection Mobile phase A Mobile phase B
Operating conditions— of sample (min) (volz) (volz)
Spectrometer: 1.1. FT-NMR, Not less than 400 MHz. 0–3 90 10
Temperature: 259C. 3 – 15 90 → 0 10 → 100
Spinning: off.
Number of data points: 32,768.
Flow rate: 0.2 mL per minute.
Spectral range: Signal of DHO ± 6.0 ppm. Time span of measurement: About 2 times as long as the
Flip angle: 909. retention time of heparin, beginning after the solvent peak.
Delay time: 20 seconds. System suitability—
Dummy scans: 4. Test for required detectability: Dissolve 10 mg of Heparin
Number of scans: SN ratio of the signal of N-acetyl pro- Sodium RS for Identification in 0.40 mL of water, and use
ton of heparin is not less 1000. this solution as the heparin sodium standard stock solution.
Window function: Exponential function (Line broaden- Separately, dissolve 0.10 mg of Over-sulfated Chondroitin
ing factor = 0.2 Hz). Sulfate RS for System Suitability in 0.20 mL of water, and
System suitability— use this solution as the over-sulfated chondroitin sulfate
standard solution. To 60 mL of the heparin sodium standard
System performance: Dissolve 20 mg of Heparin Sodium
stock solution add 3 mL of the over-sulfated chondroitin
RS for Identification in 0.40 mL of a solution of sodium 3-
sulfate standard solution and 12 mL of water, and mix.
trimethylsilylpropionate-d4 for nuclear magnetic resonance When the procedure is run with 20 mL of the mixture under
spectroscopy in heavy water for nuclear magnetic resonance the above operating conditions, it exhibits a peak for over-
spectroscopy (1 in 10,000). Dissolve 0.10 mg of Over-sulfat- sulfated chondroitin sulfate.
ed Chondroitin Sulfate RS for System Suitability in 1.0 mL System performance: To 120 mL of the heparin sodium
of a solution of sodium 3-trimethylsilylpropionate-d4 for standard stock solution add 30 mL of the over-sulfated
nuclear magnetic resonance spectroscopy in heavy water for chondroitin sulfate standard solution, mix and use this solu-
nuclear magnetic resonance spectroscopy (1 in 10,000). To tion as the solution for system suitability test. When the
the solution of Heparin Sodium RS for Identification add procedure is run with 20 mL of the solution for system
0.20 mL of the solution of Over-sulfated Chondroitin Sul- suitability test under the above operating conditions, hepa-
rin and over-sulfated chondroitin sulfate are eluted in this
fate RS for System Suitability. When determining the spec-
order with the resolution between these peaks being not less

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2701

than 1.5. 259C.


System repeatability: When the test is repeated 6 times Mobile phase A: Dissolve 1 g of potassium dihydrogen
with 20 mL of the solution for system suitability test under phosphate in 1000 mL of water, and adjust to pH 5.5 with
the above operating conditions, the relative standard devia- potassium hydroxide TS.
tion of the peak area of over-sulfated chondroitin sulfate is Mobile phase B: Acetonitrile.
not more than 2.0z.
Flowing of mobile phase: Control the gradient by mixing
the mobile phases A and B as directed in the following
table.
Hydrocortisone Acetate
ヒドロコルチゾン酢酸エステル Time after injection Mobile phase A Mobile phase B
of sample (min) (volz) (volz)
Change the Optical rotation as follows: 0 – 12.5 80 → 35 20 → 65
Optical rotation <2.49> [a]20
+158 – +1679(after drying,
D: 12.5 – 15.5 35 65
50 mg, 1,4-dioxane, 10 mL, 100 mm).
Flow rate: 2.0 mL per minute.
Time span of measurement: For 15.5 minutes after injec-
Hydrocortisone Butyrate tion, beginning after the solvent peak.
System suitability—
ヒドロコルチゾン酪酸エステル Test for required detectability: Pipet 1 mL of the stand-
ard solution, and add a mixture of acetonitrile and the mo-
Change the Description and Purity as follows: bile phase A (4:1) to make exactly 20 mL. Confirm that the
Description Hydrocortisone Butyrate occurs as a white peak area of hydrocortisone butyrate obtained with 5 mL of
powder. It is odorless. this solution is equivalent to 3.5 to 6.5z of that with 5 mL
It is freely soluble in chloroform, soluble in methanol, of the standard solution.
sparingly soluble in ethanol (99.5) and practically insoluble System performance: When the procedure is run with 5
in water. mL of the standard solution under the above operating con-
Melting point: about 2009 C (with decomposition). ditions, the number of theoretical plates and the symmetry
factor of the peak of hydrocortisone butyrate are not less
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of than 10,000 and not more than 1.5, respectively.
Hydrocortisone Butyrate according to Method 2, and per- System repeatability: When the test is repeated 6 times
form the test. Prepare the control solution with 2.0 mL of with 5 mL of the standard solution under the above operat-
Standard Lead Solution (not more than 20 ppm). ing conditions, the relative standard deviation of the peak
(2) Related substances—Dissolve 50 mg of Hydrocorti- area of hydrocortisone butyrate is not more than 2.0z.
sone Butyrate in 50 mL of a mixture of acetonitrile and the
mobile phase A (4:1), and use this solution as the sample so-
lution. Pipet 1 mL of the sample solution, add a mixture of Hydroxocobalamin Acetate
acetonitrile and the mobile phase A (4:1) to make exactly
100 mL, and use this solution as the standard solution. Per- ヒドロキソコバラミン酢酸塩
form the test with exactly 5 mL each of the sample solution
and standard solution as directed under Liquid Chromatog- Change the Origin/limits of content, Description
raphy <2.01> according to the following conditions. Deter- and Purity as follows:
mine each peak area by the automatic integration method:
the area of the peak other than hydrocortisone butyrate ob- Hydroxocobalamin Acetate contains not less than
tained from the sample solution is not larger than the peak 96.0z and not more than 101.0z of hydroxocobala-
area of hydrocortisone butyrate from the standard solution, min acetate (C62H89CoN13O15P.C2H4O2), calculated
and the total area of the peaks other than hydrocortisone on the anhydrous and residual solvent-free basis.
butyrate from the sample solution is not larger than 2 times
the peak area of hydrocortisone butyrate from the standard Description Hydroxocobalamin Acetate occurs as dark
solution. red, crystals or powder. It is odorless.
Operating conditions— It is freely soluble in water and in methanol, slightly solu-
Detector: An ultraviolet absorption photometer (wave- ble in ethanol (95), and practically insoluble in diethyl ether.
length: 254 nm). It is hygroscopic.
Column: A stainless steel column 4.6 mm in inside diame- Purity Conduct this procedure using light-resistant ves-
ter and 10 cm in length, packed with octadecylsilanized sels. Dissolve 75 mg of Hydroxocobalamin Acetate in 100
silica gel for liquid chromatography (3 mm in particle diame- mL of the dissolving solution, and use this solution as the
ter). sample solution. Pipet 1 mL of the sample solution, add the
Column temperature: A constant temperature of about dissolving solution to make exactly 20 mL, and use this so-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2702 Official Monographs Supplement I, JP XVII

lution as the standard solution. Perform the test with ex- Delete the Loss on drying:
actly 20 mL each of the sample solution and standard solu-
tion as directed under Liquid Chromatography <2.01> ac- Add the following next to the purity:
cording to the following conditions, and determine each
Water <2.48> 8.0 – 12.0z (50 mg, volumetric titration,
peak area by the automatic integration method: the total
direct titration).
area of the peaks other than hydroxocobalamin from the
sample solution is not larger than the peak area of hydrox-
Change the Assay as follows:
ocobalamin from the standard solution.
Dissolving solution: A mixture of water, mobile phase C Assay Weigh accurately about 0.1 g of Hydroxocobalamin
and methanol (41:5:4). Acetate, and dissolve in water to make exactly 500 mL.
Operating conditions— Pipet 5 mL of this solution, and add acetic acid-sodium ace-
Detector: An ultraviolet absorption photometer (wave- tate buffer solution (pH 4.5) to make exactly 25 mL. Deter-
length: 351 nm). mine the absorbance, A, of this solution at 351 nm as di-
Column: Connect two columns which are 4.6 mm in in- rected under Ultraviolet-visible Spectrophotometry <2.24>.
side diameter and 10 cm in length, composed of octadecyl-
Amount (mg) of hydroxocobalamin acetate
silanized monolithic silica for liquid chromatography, hav-
(C62H89CoN13O15P.C2H4O2)
ing a bimodal pore structure with 2 mm macropore and 13
= A/187 × 25,000
nm mesopore, coated with polyether ether ketone.
Column temperature: A constant temperature of about
309C.
Mobile phase A: Water. Hydroxypropylcellulose
Mobile phase B: Methanol.
ヒドロキシプロピルセルロース
Mobile phase C: Dissolve 15.6 g of sodium dihydrogen
phosphate dihydrate in 1000 mL of water, and adjust to pH
Change the Purity (2) as follows:
3 with diluted phosphoric acid (1 in 100).
Flowing of mobile phase: Control the gradient by mixing Purity
the mobile phase A, B and C as directed in the following (2) Silicon dioxide—Apply to Hydroxypropylcellulose,
table. if the addition of silicon dioxide is stated on the label and if
more than 0.2z residue is found in the Residue on ignition.
Time after Mobile Mobile Mobile Weigh accurately the crucible containing the residue tested
injection of phase phase phase in the Residue on ignition of Hydroxypropylcellulose (a
sample (min) A (volz) B (volz) C (volz) (g)). Moisten the residue with water, and add 5 mL of
hydrofluoric acid, in small portions. Evaporate it on a
0 – 20 82 8 10 steam bath to dryness and cool. Add 5 mL of hydrofluoric
20 – 40 82 → 50 8 → 40 10 acid and 0.5 mL of sulfuric acid, and evaporate to dryness.
Slowly increase the temperature until all the acids have been
Flow rate: 2 mL per minute. volatilized, and ignite at 1000 ± 259C. Cool the crucible in
Time span of measurement: For 40 minutes after injec- a desiccator, and weigh (b (g)). Calculate the amount of sili-
tion of the sample solution. con dioxide by the following equation: not more than 0.6z.
System suitability—
Amount (z) of silicon dioxide (SiO2)
Test for required detectability: Pipet 1 mL of the stand-
= (a – b)/M × 100
ard solution, add the dissolving solution to make exactly 50
mL. Confirm that the peak area of hydroxocobalamin ob- M: Amount (g) of Hydroxypropylcellulose used for
tained with 20 mL of this solution is equivalent to 1.4 to Residue on ignition
2.6z of that with 20 mL of the standard solution.
System performance: When the procedure is run with 20
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry
factor of the peak of hydroxocobalamin are not less than
4000 and not more than 2.4, respectively.
System repeatability: When the test is repeated 6 times
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
area of hydroxocobalamin is not more than 2.0z.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2703

Residue on ignition <2.44> Not more than 0.8z (1 g).


Low Substituted Assay (i) Apparatus—Reaction vial: A 5-mL pressure-
Hydroxypropylcellulose tight serum vial, 20 mm in outside diameter and 50 mm in
height, 20 mm in outside diameter and 13 mm in inside di-
低置換度ヒドロキシプロピルセルロース ameter at the neck, equipped with a septum made of butyl-
rubber whose surface is processed with fluoroplastics,
Change as follows: which can be fixed to the vial and stopper tightly with an
aluminum cap or another system providing an equivalent
[9004-64-2, Hydroxypropylcellulose] air-tightness.
Heater: A square-shaped aluminum block, having holes
This monograph is harmonized with the European Phar- 20 mm in diameter and 32 mm in depth, adopted to the
macopoeia and the U.S. Pharmacopeia. reaction vial. Capable of stirring the content of the reaction
The corresponding part of the attributes/provisions vial by means of a magnetic stirrer or of a reciprocal shaker
which are agreed as non-harmonized within the scope of the about 100 times per minute.
harmonization is marked with symbols (◆ ◆), and the cor- (ii) Procedure—Weigh accurately about 65 mg of Low
responding parts which are agreed as the JP local require- Substituted Hydroxypropylcellulose, transfer to the reac-
ment other than the scope of the harmonization are marked tion vial, add 0.06 to 0.10 g of adipic acid, 2.0 mL of the
with symbols (◇ ◇). internal standard solution and 2.0 mL of hydroiodic acid,
immediately stopper the vial tightly, and weigh accurately.
Low Substituted Hydroxypropylcellulose is a low Using a magnetic stirrer equipped in the heating module, or
substituted hydroxypropyl ether of cellulose. using a shaker, mix for 60 minutes while heating so that the
It contains not less than 5.0z and not more than temperature of the vial content is 130 ± 29C. In the case
16.0z of hydroxypropoxy group (-OC3H6OH: when a magnetic stirrer or shaker is not available, heat for
75.09), calculated on the dried basis. 30 minutes with repeated shaking at 5-minute intervals by
◆Description
hand, and continue heating for an additional 30 minutes.
Low Substituted Hydroxypropylcellulose
Allow the vial to cool, and again weigh accurately. If the
occurs as a white to yellowish white, powder or granules.
mass loss is less than 26 mg and there is no evidence of a
It is practically insoluble in ethanol (99.5).
leak of the content, use the upper layer of the mixture as the
It dissolves in a solution of sodium hydroxide (1 in 10),
sample solution. Separately, put 0.06 to 0.10 g of adipic
and produces a viscous solution.
acid, 2.0 mL of the internal standard solution and 2.0 mL
It swells in water, in sodium carbonate TS and in 2 mol/L
of hydroiodic acid in a reaction vial, immediately stopper
hydrochloric acid TS.◆
the vial tightly, and weigh accurately. Add 15 to 22 mL of
Identification isopropyl iodide for assay through the septum using a
(1) Shake thoroughly 0.1 g of Low Substituted Hydrox- micro-syringe, and weigh accurately. Shake the reaction vial
ypropylcellulose with 10 mL of water: it does not dissolve. thoroughly, and use the upper layer of the content as the
(2) To the dispersed solution obtained in (1) add 1 g of standard solution. Perform the test with 1 to 2 mL each of
sodium hydroxide, and shake until the solution becomes the sample solution and standard solution as directed under
uniform. Transfer 5 mL of this solution to a suitable vessel, Gas Chromatography <2.02> according to the following
add 10 mL of a mixture of acetone and methanol (4:1), and conditions, and calculate the ratios, QT and QS, of the peak
shake: a white, flocculent precipitate is formed. area of isopropyl iodide to that of the internal standard.
(3) Determine the infrared absorption spectrum of Low
Amount (z) of hydroxypropoxy group (C3H7O2)
Substituted Hydroxypropylcellulose as directed in the potas-
= MS/MT × QT/QS × 44.17
sium bromide disk method under Infrared Spectrophotome-
try <2.25>, and compare the spectrum with the Reference MS: Amount (mg) of isopropyl iodide for assay taken
Spectrum: both spectra exhibit similar intensities of absorp- MT: Amount (mg) of Low Substituted Hydroxypropylcel-
tion at the same wave numbers. lulose taken, calculated on the dried basis

pH <2.54> To 1.0 g of Low Substituted Hydroxypropylcel- Internal standard solution—A solution of n-octane in o-xy-
lulose add 100 mL of freshly boiled and cooled water, and lene (3 in 100).
shake: the pH of the solution is between 5.0 and 7.5. Operating conditions—
Detector: A thermal conductivity detector or hydrogen
Purity ◇Heavy metals <1.07>—Proceed with 2.0 g of Low
flame-ionization detector.
Substituted Hydroxypropylcellulose according to Method 2,
Column: A fused silica column 0.53 mm in inside diame-
and perform the test. Prepare the control solution with 2.0
ter and 30 m in length, coated with dimethylpolysiloxane
mL of Standard Lead Solution (not more than 10 ppm).◇
for gas chromatography 3 mm in thickness. Use a guard
Loss on drying <2.41> Not more than 5.0z (1 g, 1059C, 1 column if necessary.
hour). Column temperature: Maintain the temperature at 509C
for 3 minutes after injection, raise to 1009C at a rate of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2704 Official Monographs Supplement I, JP XVII

109C per minute, then to 2509 C at a rate of 359 C per equivalent apparatus.
minute and maintain at 2509C for 8 minutes. Rotor No., rotation frequency, and calculation multipli-
Injection port temperature: 2509C. er: According to the following table, depending on the
Detector temperature: 2809C. labeled viscosity.
Carrier gas: Helium.
Flow rate: Adjust so that the retention time of the inter- Rotation
Labeled viscosity Rotor Calculation
nal standard is about 10 minutes (4.3 mL per minute). frequency
(mPa・s) No. multiplier
Split ratio: 1:40. /min
System suitability— Not less than 600 and less than 1400 3 60 20
System performance: When the procedure is run with 1 to 〃 1400 〃 3500 3 12 100
2 mL of the standard solution under the above operating 〃 3500 〃 9500 4 60 100
〃 9500 〃 99,500 4 6 1000
conditions, isopropyl iodide and n-octane are eluted in this
〃 99,500 4 3 2000
order with the resolution between these peaks being not less
than 5.
System repeatability: When the test is repeated 6 times Procedure of apparatus: Read the value after 2 minutes
with 1 to 2 mL of the standard solution under the above op- of rotation, and stop the rotation for at least 2 minutes.
erating conditions, the relative standard deviation of the Repeat this procedure more two times, and average the
ratio of the peak area of isopropyl iodide to that of the in- three observed values.
ternal standard is not more than 2.0z. Assay
Containers and storage Containers—Tight containers. (ii) Procedure—Weigh accurately about 65 mg of
Hypromellose, transfer to the reaction vial, add 0.06 to
0.10 g of adipic acid, 2.0 mL of the internal standard solu-
tion and 2.0 mL of hydroiodic acid, immediately stopper
Hypromellose the vial tightly, and weigh accurately. Using a magnetic stir-
ヒプロメロース rer equipped in the heating module, or using a shaker, stir
for 60 minutes while heating so that the temperature of the
Change the Viscosity and Assay (ii) as follows: vial content is 130 ± 29C. In the case when a magnetic stir-
rer or shaker is not available, heat for 30 minutes with
Viscosity <2.53> (i) Method I: Apply to Hypromellose repeated shaking at 5-minute intervals by hand, and con-
having a labeled viscosity of less than 600 mPa・s. Put an ex- tinue heating for an additional 30 minutes. Allow the vial to
act amount of Hypromellose, equivalent to 4.000 g calcu- cool, and again weigh accurately. If the mass loss is less
lated on the dried basis, in a tared, wide-mouth bottle, add than 0.50z and there is no evidence of a leak of the con-
hot water (between 909C and 999C) to make 200 g, stopper tent, use the upper layer of the mixture as the sample solu-
the bottle, stir by mechanical means at 350 to 450 revolu- tion. Separately, put 0.06 to 0.10 g of adipic acid, 2.0 mL of
tions per minute for 10 to 20 minutes to get a homogeneous the internal standard solution and 2.0 mL of hydroiodic
dispersion. If necessary, take off the sample attached on the acid in a reaction vial, immediately stopper the vial tightly,
walls of the bottle, put them in the dispersed solution, and and weigh accurately. Add 45 mL of iodomethane for assay
dissolve by continuing the stirring in a water bath at not and 15 to 22 mL of isopropyl iodide for assay through the
exceeding 109 C for 20 to 40 minutes. Add cold water, if septum using a micro-syringe with weighing accurately
necessary, to make 200 g, and use this solution as the sam- every time, shake thoroughly, and use the upper layer of the
ple solution. Centrifuge the solution if necessary to expel content as the standard solution. Perform the test with 1 to
any entrapped air bubbles. Perform the test with the sample 2 mL each of the sample solution and standard solution as
solution at 20 ± 0.19C as directed in Method I under Vis- directed under Gas Chromatography <2.02> according to the
cosity Determination: not less than 80z and not more than following conditions, and calculate the ratios, QTa and QTb,
120z of the labeled viscosity. of the peak area of iodomethane and isopropyl iodide to
(ii) Method II: Apply to Hypromellose having a labeled that of the internal standard obtained from the sample solu-
viscosity of not less than 600 mPa・s. Put an exact amount tion, and QSa and QSb, of the peak area of iodomethane and
of Hypromellose, equivalent to 10.00 g calculated on the isopropyl iodide to that of the internal standard from the
dried basis, in a tared, wide-mouth bottle, add hot water standard solution.
(between 909 C and 999C) to make 500 g, and prepare the
sample solution in the same manner as directed in Method I. Content (z) of methoxy group (CH3O)
Perform the test with the sample solution at 20 ± 0.19C as = QTa/QSa × MSa/M × 21.86
directed in Method II under Viscosity Determination, using Content (z) of hydroxypropoxy group (C3H7O2)
a single cylinder-type rotational viscometer, according to = QTb/QSb × MSb/M × 44.17
the following operating conditions: not less than 75z and
not more than 140z of the labeled viscosity. MSa: Amount (mg) of iodomethane for assay taken
Operating conditions— MSb: Amount (mg) of isopropyl iodide for assay taken
Apparatus: Brookfield type viscometer LV model or an M: Amount (mg) of Hypromellose taken, calculated on

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2705

the dried basis Identification Adjust Isophane Insulin Human (Genetical


Recombination) Injectable Aqueous Suspension to pH be-
Internal standard solution—A solution of n-octane in o-xy-
tween 2.5 and 3.0 with dilute hydrochloric acid: the precipi-
lene (3 in 100).
tate dissolves, and the solution is clear and colorless.
Operating conditions—
Detector: A thermal conductivity detector or hydrogen pH Being specified separately when the drug is granted ap-
flame-ionization detector. proval based on the Law.
Column: A glass column 3 – 4 mm in inside diameter and
Purity (1) Desamido substance—Perform the test with
1.8 – 3 m in length, packed with siliceous earth for gas chro-
20 mL of the sample solution obtained in the Assay (1) as di-
matography, 125 to 150 mm in diameter, coated with methyl
rected under Liquid Chromatography <2.01> according to
silicone polymer for gas chromatography at the ratio of 10 –
the following conditions. Determine each peak area by the
20z.
automatic integration method, and calculate the amount of
Column temperature: A constant temperature of about
them by the area percentage method: the amount of the
1009C.
peak, having the relative retention time of about 1.3 to insu-
Carrier gas: Helium for thermal conductivity detector, or
lin human, is not more than 1.5z.
Helium or Nitrogen for hydrogen flame-ionization detector.
Operating conditions—
Flow rate: Adjust so that the retention time of the inter-
Proceed as directed in the operating conditions in the
nal standard is about 10 minutes.
Assay (1).
System suitability—
System suitability—
System performance: When the procedure is run with
System performance: Proceed as directed in the system
1 – 2 mL of the standard solution under the above operating
suitability in the Assay (1).
conditions, iodomethane, isopropyl iodide and the internal
Test for required detectability: Pipet 1 mL of the sample
standard are eluted in this order, with complete separation
solution, add 0.01 mol/L hydrochloric acid TS to make ex-
of these peaks.
actly 50 mL. Confirm that the peak area of insulin human
obtained with 20 mL of this solution is equivalent to 1.4 to
2.6z of that with 20 mL of the sample solution.
Add the following:
System repeatability: Dissolve Insulin Human RS in 0.01
mol/L hydrochloric acid TS so that each mL contains about
Isophane Insulin Human (Genetical 4 Insulin Units. When the test is repeated 6 times with 20 mL
Recombination) Injectable of this solution under the above operating conditions, the
relative standard deviation of the peak area of insulin hu-
Aqueous Suspension man is not more than 2.0z.
(2) Dissolved insulin human—Centrifuge Isophane In-
イソフェンインスリン ヒト(遺伝子組換え)水性懸濁注
sulin Human (Genetical Recombination) Injectable Aque-
射液
ous Suspension, and use the supernatant liquid as the sam-
ple solution. Separately, dissolve exactly Insulin Human RS
Isophane Insulin Human (Genetical Recombina-
in 0.01 mol/L hydrochloric acid TS to make a solution so
tion) Injectable Aqueous Suspension is an aqueous
that each mL contains about 1.0 Insulin Units, and use this
suspension for injection.
solution as the standard solution. Perform the test with ex-
It contains not less than 95.0z and not more than
actly 20 mL each of the sample solution and standard solu-
105.0z of the labeled Insulin Unit of insulin human
tion as directed under Liquid Chromatography <2.01> ac-
(genetical recombination) (C257H383N65O77S6:
cording to the following conditions. Determine the peak
5807.57). It contains not less than 10 mg and not more
areas, AT and AS, of insulin human by the automatic in-
than 40 mg of zinc (Zn: 65.38) per the labeled 100 In-
tegration method, and calculate the amount of dissolved in-
sulin Units.
sulin human by the following equation: not more than 0.5
Method of preparation Prepare as directed under Injec- Insulin Units per mL.
tions, with Insulin Human (Genetical Recombination) and
Amount (mg) of dissolved insulin human
Protamine Sulfate.
(Insulin Unit /mL)
Description Isophane Insulin Human (Genetical Recombi- = (MS × F )/D × AT/AS
nation) Injectable Aqueous Suspension is a white aqueous
MS: Amount (mg) of Insulin Human RS taken
suspension. When allowed to stand, it separates into a white
F: Labeled unit (Insulin Unit /mg) of Insulin Human RS
precipitate and colorless supernatant liquid, and the precipi-
D: Volume (mL) of 0.01 mol/L hydrochloric acid TS
tate returns to the suspension state on gentle shaking.
used to dissolve Insulin Human RS
When it is examined microscopically, the precipitate
mostly consists of fine, oblong crystals of 1 to 30 mm in Operating conditions—
major axis, and does not contain amorphous substances or Proceed as directed in the operating conditions in the
large aggregates. Assay (1).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2706 Official Monographs Supplement I, JP XVII

System suitability— Amount (Insulin Unit) of insulin human (C257H383N65O77S6)


System performance: When the procedure is run with 20 in 1 mL
mL of insulin human desamido substance-containing TS = (MS × F )/D × (ATI + ATD)/(ASI + ASD)
under the above operating conditions, insulin human and × 1.004 × 5/2
insulin human desamido substance are eluted in this order
MS: Amount (mg) of Insulin Human RS taken
with the resolution between these peaks being not less than
F: Labeled unit (Insulin Unit /mg) of Insulin Human RS
2.0, and the symmetry factor of the peak of insulin human
D: Volume (mL) of 0.01 mol/L hydrochloric acid TS
is not more than 1.6.
used to dissolve Insulin Human RS
System repeatability: When the test is repeated 4 times
with 20 mL of the standard solution under the above operat- (2) Zinc—Pipet a volume of gently shaken Isophane In-
ing conditions, the relative standard deviation of the peak sulin Human (Genetical Recombination) Injectable
area of insulin human is not more than 6.0z. Aqueous Suspension, equivalent to 300 Insulin Units, and
(3) High-molecular mass protein—Take a suitable add 0.01 mol/L hydrochloric acid TS to make exactly 50
volume of gently shaken Isophane Insulin Human (Geneti- mL. If necessary, further add 0.01 mol/L hydrochloric acid
cal Recombination) Injectable Aqueous Suspension, add 4 TS to make exactly 100 mL, and use this solution as the
mL of 6 mol/L hydrochloric acid TS for each mL of the sus- sample solution. Separately, pipet a suitable volume of
pension, and mix until the solution becomes clear. Perform Standard Zinc Solution for Atomic Absorption Spec-
the test with 100 mL of this solution as directed under Liq- troscopy, dilute with 0.01 mol/L hydrochloric acid TS to
uid Chromatography <2.01> according to the following con- make three solutions containing 0.20 mg, 0.60 mg and 1.20
ditions. Determine each peak area by the automatic integra- mg of zinc (Zn: 65.38) in each mL, respectively, and use
tion method, and calculate the amount of them by the area these solutions as the standard solutions. Perform the test
percentage method: the total amount of the peaks other with the sample solution and standard solutions as directed
than insulin human is not more than 2.5z. under Atomic Absorption Spectroscopy <2.23> according to
Operating conditions— the following conditions, using 0.01 mol/L hydrochloric
Detector, column temperature, mobile phase and flow acid TS as the blank, and calculate the content of zinc in the
rate: Proceed as directed in the operating conditions in the sample solution by using the calibration curve obtained
Purity (2) under Insulin Human (Genetical Recombination). from the absorbances of the standard solutions.
Column: A stainless steel column 7.8 mm in inside diame- Gas: Combustible gas—Acetylene.
ter and 30 cm in length, packed with hydrophilic silica gel Supporting gas—Air.
for liquid chromatography. Lamp: Zinc hollow cathode lamp.
Time span of measurement: From the retention time cor- Wavelenghth: 213.9 nm.
responding to the exclusion volume of the size-exclusion
Containers and storage Containers—Hermetic containers.
column to the completion of the elution of insulin human.
Storage—Light-resistant, at a temperature between 29C
System suitability—
and 89C avoiding freezing.
System performance: Proceed as directed in the system
suitability in the Purity (2) under Insulin Human (Genetical
Recombination).
Add the following:
Test for required detectability: Pipet 1 mL of the sample
solution, and add 0.01 mol/L hydrochloric acid TS to make
exactly 50 mL. Confirm that the peak area of insulin human Biphasic Isophane Insulin Human
obtained with 100 mL of this solution is equivalent to 1.4 to (Genetical Recombination)
2.6z of that with 100 mL of the sample solution.
Injectable Aqueous Suspension
Extractable volume <6.05> It meets the requirement.
二相性イソフェンインスリン ヒト(遺伝子組換え)水性
Foreign insoluble matter <6.06> Perform the test accord-
懸濁注射液
ing to Method 1: it meets the requirement.

Sterility <4.06> Perform the test according to the Mem- Biphasic Isophane Insulin Human (Genetical
brane filtration method: it meets the requirement. Recombination) Injectable Aqueous Suspension is an
aqueous suspension for injection.
Assay (1) Insulin human—Pipet 10 mL of gently shaken
It contains not less than 95.0z and not more
Isophane Insulin Human (Genetical Recombination) Inject-
than 105.0z of the labeled Insulin Unit of insulin
able Aqueous Suspension, and add exactly 40 mL of 6
human (genetical recombination) (C257H383N65O77S6:
mol/L hydrochloric acid TS. Pipet 2 mL of this solution,
5807.57). It contains not less than 10 mg and not more
add 0.01 mol/L hydrochloric acid TS to make exactly 5 mL,
than 40 mg of zinc (Zn: 65.38) per the labeled 100 In-
and use this solution as the sample solution. Then, proceed
sulin Units.
as directed in the Assay under Insulin Human (Genetical
Recombination). Method of preparation Prepare as directed under Injec-
tions, with Insulin Human (Genetical Recombination) In-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2707

jection and Isophane Insulin Human (Genetical Recombina- rate: Proceed as directed in the operating conditions in the
tion) Injectable Aqueous Suspension. Purity (2) under Insulin Human (Genetical Recombination).
Column: A stainless steel column 7.8 mm in inside diame-
Description Biphasic Isophane Insulin Human (Genetical
ter and 30 cm in length, packed with hydrophilic silica gel
Recombination) Injectable Aqueous Suspension is a white
for liquid chromatography.
aqueous suspension. When allowed to stand, it separates
Time span of measurement: From the retention time cor-
into a white precipitate and colorless supernatant liquid,
responding to the exclusion volume of the size-exclusion
and the precipitate returns to the suspension state on gentle
column to the completion of the elution of insulin human.
shaking.
System suitability—
When it is examined microscopically, the precipitate
System performance: Proceed as directed in the system
mostly consists of fine, oblong crystals of 1 to 30 mm in
suitability in the Purity (2) under Insulin Human (Genetical
major axis, and does not contain amorphous substances or
Recombination).
large aggregates.
Test for required detectability: Pipet 1 mL of the sample
Identification Adjust Biphasic Isophane Insulin Human solution, and add 0.01 mol/L hydrochloric acid TS to make
(Genetical Recombination) Injectable Aqueous Suspension exactly 50 mL. Confirm that the peak area of insulin human
to pH between 2.5 and 3.0 with dilute hydrochloric acid: the obtained with 100 mL of this solution is equivalent to 1.4 to
precipitate dissolves, and the solution is clear and colorless. 2.6z of that with 100 mL of the sample solution.

pH Being specified separately when the drug is granted ap- Extractable volume <6.05> It meets the requirement.
proval based on the Law.
Foreign insoluble matter <6.06> Perform the test accord-
Purity (1) Desamido substance—Perform the test with ing to Method 1: it meets the requirement.
20 mL of the sample solution obtained in the Assay (1) as di-
Sterility <4.06> Perform the test according to the Mem-
rected under Liquid Chromatography <2.01> according to
brane filtration method: it meets the requirement.
the following conditions. Determine each peak area by the
automatic integration method, and calculate the amount of Soluble Insulin Human Being specified separately when
them by the area percentage method: the amount of the the drug is granted approval based on the Law.
peak, having the relative retention time of about 1.3 to insu-
Assay (1) Insulin human—Pipet 10 mL of gently shaken
lin human, is not more than 1.5z.
Biphasic Isophane Insulin Human (Genetical Recombina-
Operating conditions—
tion) Injectable Aqueous Suspension, and add exactly 40 mL
Proceed as directed in the operating conditions in the
of 6 mol/L hydrochloric acid TS. Pipet 2 mL of this solu-
Assay (1).
tion, add 0.01 mol/L hydrochloric acid TS to make exactly
System suitability—
5 mL, and use this solution as the sample solution. Then,
System performance: Proceed as directed in the system
proceed as directed in the Assay under Insulin Human (Ge-
suitability in the Assay (1).
netical Recombination).
Test for required detectability: Pipet 1 mL of the sample
solution, add 0.01 mol/L hydrochloric acid TS to make ex- Amount (Insulin Unit) of insulin human (C257H383N65O77S6)
actly 50 mL. Confirm that the peak area of insulin human in 1 mL
obtained with 20 mL of this solution is equivalent to 1.4 to = (MS × F )/D × (ATI + ATD)/(ASI + ASD)
2.6z of that with 20 mL of the sample solution. × 1.004 × 5/2
System repeatability: Dissolve Insulin Human RS in 0.01
MS: Amount (mg) of Insulin Human RS taken
mol/L hydrochloric acid TS so that each mL contains about
F: Labeled unit (Insulin Unit /mg) of Insulin Human RS
4 Insulin Units. When the test is repeated 6 times with 20 mL
D: Volume (mL) of 0.01 mol/L hydrochloric acid TS
of this solution under the above operating conditions, the
used to dissolve Insulin Human RS
relative standard deviation of the peak area of insulin hu-
man is not more than 2.0z. (2) Zinc—Pipet a volume of gently shaken Biphasic
(2) High-molecular mass protein—Take a suitable Isophane Insulin Human (Genetical Recombination) Inject-
volume of gently shaken Biphasic Isophane Insulin Human able Aqueous Suspension, equivalent to 300 Insulin Units,
(Genetical Recombination) Injectable Aqueous Suspension, and add 0.01 mol/L hydrochloric acid TS to make exactly
add 4 mL of 6 mol/L hydrochloric acid TS for each mL of 50 mL. If necessary, further add 0.01 mol/L hydrochloric
the suspension, and mix until the solution becomes clear. acid TS to make exactly 100 mL, and use this solution as the
Perform the test with 100 mL of this solution as directed sample solution. Separately, pipet a suitable volume of
under Liquid Chromatography <2.01> according to the fol- Standard Zinc Solution for Atomic Absorption Spec-
lowing conditions. Determine each peak area by the auto- troscopy, dilute with 0.01 mol/L hydrochloric acid TS to
matic integration method, and calculate the amount of them make three solutions containing 0.20 mg, 0.60 mg and 1.20
by the area percentage method: the total amount of the mg of zinc (Zn: 65.38) in each mL, respectively, and use
peaks other than insulin human is not more than 2.0z. these solutions as the standard solutions. Perform the test
Operating conditions— with the sample solution and standard solutions as directed
Detector, column temperature, mobile phase and flow under Atomic Absorption Spectroscopy <2.23> according to

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2708 Official Monographs Supplement I, JP XVII

the following conditions, using 0.01 mol/L hydrochloric tions, and compare the peak (peak 1) eluted just after the
acid TS as the blank, and calculate the content of zinc in the peak of the solvent and the succeeding three peaks (peaks 2,
sample solution by using the calibration curve obtained 3 and 4) with apparently higher peak height in the chro-
from the absorbances of the standard solutions. matograms obtained from these solutions: the similar peaks
Gas: Combustible gas—Acetylene. are observed at the same retention times.
Supporting gas—Air. Operating conditions—
Lamp: Zinc hollow cathode lamp. Detector: An ultraviolet absorption photometer (wave-
Wavelenghth: 213.9 nm. length: 214 nm).
Column: A stainless steel column 4.6 mm in inside diame-
Containers and storage Containers—Hermetic containers.
ter and 10 cm in length, packed with octadecylsilanized
Storage—Light-resistant, at a temperature between 29C
silica gel for liquid chromatography (not exceeding 5 mm in
and 89C avoiding freezing.
particle diameter).
Column temperature: A constant temperature of about
409C.
Add the following:
Mobile phase A: A mixture of water, ammonium sulfate
buffer solution and acetonitrile for liquid chromatography
Insulin Aspart (Genetical (7:2:1).
Recombination) Mobile phase B: A mixture of water, acetonitrile for liq-
uid chromatography and ammonium sulfate buffer solution
インスリン アスパルト(遺伝子組換え) (2:2:1).
Flowing of mobile phase: Control the gradient by mixing
the mobile phases A and B as directed in the following
table.
C256H381N65O79S6: 5825.54
[116094-23-6] Time after injection Mobile phase A Mobile phase B
of sample (min) (volz) (volz)
Insulin Aspart (Genetical Recombination) is an
analogue of human insulin (genetical recombination), 0 – 60 90 → 30 10 → 70
in which proline residue at 28th of B chain is substit- 60 – 65 30 → 0 70 → 100
uted with aspartic acid. It is a peptide composed of A 65 – 70 0 100
chain consisting of 21 amino acid residues and B
chain consisting of 30 amino acid residues. Flow rate: 1 mL per minute.
It contains not less than 92.6z and not more than System suitability—
109.5z of insulin aspart (genetical recombination) System performance: When the procedure is run with 50
(C256H381N65O79S6), calculated on the dried and mL of the standard solution under the above operating con-
residue on ignition-free basis. ditions, the symmetry factors of the peaks 2 and 3 are not
0.0350 mg of Insulin Aspart (Genetical Recombina- more than 1.5, respectively, and the resolution between
tion) is equivalent to 1 Insulin Unit. these peaks is not less than 8.

Description Insulin Aspart (Genetical Recombination) oc- Purity (1) Related substances—Perform the test with 10
curs as a white powder. mL of the sample solution obtained in the Assay as directed
It is practically insoluble in water and in ethanol (95). under Liquid Chromatography <2.01> according to the fol-
It dissolves in 0.01 mol/L hydrochloric acid TS. lowing conditions. Determine each peak area by the auto-
It is hygroscopic. matic integration method, and calculate the amounts of
them by the area percentage method: the amount of the
Identification Weigh a suitable amount of Insulin Aspart
peak of B28isoAsp insulin aspart, having the relative reten-
(Genetical Recombination), and dissolve in 0.01 mol/L hy-
tion time of about 0.9 to insulin aspart, is not more than
drochloric acid TS to make a solution so that each mL con-
0.3z, the total amount of the peak of A21Asp insulin
tains 2.0 mg. Separately, dissolve Insulin Aspart RS in 0.01
aspart and B3Asp insulin aspart, having the relative reten-
mol/L hydrochloric acid TS to make a solution so that each
tion time of about 1.3, and the peak of B3isoAsp insulin
mL contains 2.0 mg. Transfer 25 mL each of these solutions
aspart, having the relative retention time of about 1.5, is not
into clean test tubes, add 100 mL of HEPES buffer solution
more than 1.0z, and the total amount of the peaks other
(pH 7.5) and 20 mL of V8-protease TS, and allow to react at
than insulin aspart and the peaks mentioned above is not
259C for 6 hours. Then add 145 mL of ammonium sulfate
more than 0.5z.
buffer solution to stop the reaction, and use these solutions
Operating conditions—
as the sample solution and the standard solution, respec-
Detector, column, column temperature, mobile phase A,
tively. Perform the test with exactly 50 mL each of the sam-
mobile phase B, flowing of mobile phase and flow rate:
ple solution and standard solution as directed under Liquid
Proceed as directed in the operating conditions in the
Chromatography <2.01> according to the following condi-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2709

Assay. time: about 17.5 minutes) and insulin aspart (retention time:
Time span of measurement: From 4 minutes to 50 18 to 20 minutes) are eluted in this order, and determine the
minutes after injection of the sample solution. peak height of the dimer and the height of the bottom be-
System suitability— tween the peaks of the dimer and the monomer: the peak-
System performance and system repeatability: Proceed as valley ratio is not less than 2.0.
directed in the system suitability in the Assay. System repeatability: When the test is repeated 6 times
Test for required detectability: Pipet 5 mL of the solution with 100 mL of the solution for system suitability test under
for system suitability test obtained in the Assay, add 0.01 the above operating conditions, the relative standard devia-
mol/L hydrochloric acid TS to make exactly 10 mL. Con- tion of the peak area of insulin aspart is not more than
firm that the area percentage of the peak of B28isoAsp insu- 2.0z.
lin aspart obtained with 10 mL of this solution is equivalent (3) Host cell proteins—Being specified separately when
to 80 to 120z of that with 10 mL of the solution for system the drug is granted approval based on the Law.
suitability test. (4) DNA—Being specified separately when the drug is
(2) High-molecular proteins—Store the sample solution granted approval based on the Law.
at a temperature between 29C and 89C, and use within 48
Loss on drying <2.41> Not more than 10.0z (0.2 g,
hours after preparation. Dissolve 4 mg of Insulin Aspart
1059C, 24 hours).
(Genetical Recombination) in 1 mL of 0.01 mol/L hydro-
chloric acid TS, and use this solution as the sample solution. Residue on ignition <2.44> Not more than 6.0z (0.2 g).
Perform the test with 100 mL of the sample solution as di-
Assay Store the sample solution and the standard solution
rected under Liquid Chromatography <2.01> according to
at a temperature between 29C and 89 C, use the sample solu-
the following conditions, determine each peak area by the
tion within 24 hours after preparation, and use the standard
automatic integration method, and calculate the amounts of
solution within 48 hours after preparation. Weigh accu-
them by the area percentage method: the total amount of
rately a suitable amount of Insulin Aspart (Genetical
the peaks other than insulin aspart is not more than 0.3z.
Recombination), dissolve in 0.01 mol/L hydrochloric acid
Operating conditions—
TS so that each mL contains 4.0 mg, and use this solution as
Detector: An ultraviolet absorption photometer (wave-
the sample solution. Separately, dissolve Insulin Aspart RS
length: 276 nm).
in 0.01 mol/L hydrochloric acid TS so that each mL con-
Column: A stainless steel column 7.8 mm in inside diame-
tains 4.0 mg, and use this solution as the standard solution.
ter and 30 cm in length, packed with hydrophilic silica gel
Perform the test with exactly 10 mL each of the sample solu-
for liquid chromatography (5 to 10 mm in particle diameter).
tion and standard solution as directed under Liquid Chro-
Column temperature: A constant temperature of about
matography <2.01> according to the following conditions,
209C.
and determine the total areas, AT and AS, of the peak of
Mobile phase: A mixture of a solution of L-arginine (1 in
B28isoAsp insulin aspart (relative retention time to insulin
1000), acetonitrile for liquid chromatography and acetic
aspart: about 0.9), the peak of insulin aspart (retention
acid (100) (13:4:3).
time: 20 to 24 minutes), the peak of A21Asp insulin aspart
Flow rate: 0.5 mL per minute.
and B3Asp insulin aspart (usually eluted together having the
Time span of measurement: From the retention time cor-
relative retention time of about 1.3) and the peak of
responding to the exclusion volume of the size-exclusion
B3isoAsp insulin aspart (relative retention time: about 1.5)
column to the completion of the elution of insulin aspart.
in each solution.
System suitability—
Test for required detectability: Allow Insulin Aspart Amount (mg) of insulin aspart (C256H381N65O79S6)
(Genetic Recombination) to stand at ordinary temperature = M S × AT / AS
for about 10 days, which results in containing about 0.4z
MS: Total amount (mg) of insulin aspart, B28isoAsp in-
of high-molecular proteins, dissolve in 0.01 mol/L hydro-
sulin aspart, A21Asp insulin aspart and B3Asp insu-
chloride TS so that each mL contains about 4 mg of insulin
lin aspart, and B3isoAsp insulin aspart in 1 mL of
aspart, and use this solution as the solution for system
the standard solution
suitability test. Store the solution for system suitability test
at a temperature between 29 C and 89C, and use within 7 Operating conditions—
days. Pipet 5 mL of the solution for system suitability test, Detector: An ultraviolet absorption photometer (wave-
and add 0.01 mol/L hydrochloric acid TS to make exactly length: 214 nm).
10 mL. Confirm that the area percentage of the peak of in- Column: A stainless steel column 4.0 mm in inside diame-
sulin aspart dimer obtained with 100 mL of this solution is ter and 25 cm in length, packed with octadecylsilanized
equivalent to 80 to 120z of that with 100 mL of the solution silica gel for liquid chromatography (not exceeding 5 mm in
for system suitability test. particle diameter).
System performance: When the procedure is run with 100 Column temperature: A constant temperature of about
mL of the solution for system suitability test under the above 409C.
operating conditions, insulin aspart polymer (retention Mobile phase A: Dissolve 142.0 g of anhydrous sodium
time: 13 to 17 minutes), insulin aspart dimer (retention sulfate in water, add 13.5 mL of phosphoric acid, and add

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2710 Official Monographs Supplement I, JP XVII

water to make 5 L. Adjust to pH 3.6 with sodium hydroxide Add the following:
TS. To 4500 mL of this solution add 500 mL of acetonitrile
for liquid chromatography. Irbesartan Tablets
Mobile phase B: A mixture of water and acetonitrile for
liquid chromatography (1:1). イルベサルタン錠
Flowing of mobile phase: Control the gradient by mixing
the mobile phases A and B as directed in the following Irbesartan Tablets contain not less than 95.0z and
table. not more than 105.0z of the labeled amount of ir-
besartan (C25H28N6O: 428.53).
Time after injection Mobile phase A Mobile phase B Method of preparation Prepare as directed under Tablets,
of sample (min) (volz) (volz) with Irbesartan.
0 – 35 58 42 Identification To a quantity of powdered Irbesartan
35 – 40 58 → 20 42 → 80 Tablets, equivalent to about 25 mg of Irbesartan, add 2 mL
40 – 45 20 80 of acetone, shake, and filter through a membrane filter with
45 – 46 20 → 58 80 → 42 a pore size not exceeding 0.45 mm. Evaporate the filtrate to
46 – 60 58 42 dryness, and determine the infrared absorption spectrum of
the residue as directed in the potassium bromide disk
Flow rate: 1 mL per minute. method under Infrared Spectrophotometry <2.25>: it exhib-
System suitability— its absorptions at the wave numbers of about 1733 cm-1,
System performance: Dissolve Insulin Aspart (Genetical 1617 cm-1, 1435 cm-1 and 758 cm-1.
Recombination) in 0.01 mol/L sodium dihydrogen phos-
Uniformity of dosage unit <6.02> Perform the Mass varia-
phate TS (pH 7.5) so that each mL contains 8 mg, and allow
tion test, or the Content uniformity test according to the
to stand at ordinary temperature for 10 to 15 days. To 1 mL
following method: it meets the requirement.
of this solution add 1 mL of 0.01 mol/L hydrochloric acid
To 1 tablet of Irbesartan Tablets add 1.5 mL of water,
TS, allow to stand at ordinary temperature for 1 to 3 days,
shake vigorously to disintegrate, and add 15 mL of metha-
and use this solution as the solution for system suitability
nol. Shake vigorously for 15 minutes, add methanol to
test. The solution for system suitability test contains 0.1 to
make exactly 20 mL, and centrifuge. Pipet V mL of the su-
2.2z of B28isoAsp insulin aspart, and not less than 1z of
pernatant liquid, equivalent to about 20 mg of irbesartan
B3Asp insulin aspart and A21Asp insulin aspart. Store the
(C25H28N6O), and add a mixture of water and acetonitrile
solution for system suitability test at a temperature between
for liquid chromatography (3:2) to make exactly 20 mL.
29 C and 89 C, and use within 72 hours. When the procedure
Pipet 2.5 mL of this solution, add a mixture of water and
is run with 10 mL of the solution for system suitability test
acetonitrile for liquid chromatography (3:2) to make exactly
under the above operating conditions, B28isoAsp insulin
20 mL, and use this solution as the sample solution. Then,
aspart, insulin aspart, A21Asp insulin aspart and B3Asp in-
proceed as directed in the Assay.
sulin aspart, and B3isoAsp insulin aspart are eluted in this
order with the resolution between the peak of insulin aspart Amount (mg) of irbesartan (C25H28N6O)
and the peak of A21Asp insulin aspart and B3Asp insulin = MS × AT/AS × 16/V
aspart being not less than 2.0.
MS: Amount (mg) of irbesartan for assay taken, calcu-
System repeatability: When the test is repeated 5 times
lated on the anhydrous basis
with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of AS is not Dissolution <6.10> When the test is performed at 50 revo-
more than 1.5z. lutions per minute according to the Paddle method, using
900 mL of 2nd fluid for dissolution test as the dissolution
Containers and storage Containers—Tight containers.
medium, the dissolution rate in 45 minutes of 50-mg and
Storage—Not exceeding -189C.
100-mg tablets is not less than 85z, respectively, and that in
60 minutes of 200-mg tablet is not less than 70z.
Start the test with 1 tablet of Irbesartan Tablets, with-
Iohexol Injection draw not less than 10 mL of the medium at the specified
minute after starting the test, and filter through a mem-
イオヘキソール注射液
brane filter with a pore size not exceeding 0.45 mm. Discard
the first 3 mL of the filtrate, pipet V mL of the subsequent
Change the Containers and storage as follows:
filtrate, add the dissolution medium to make exactly V? mL
Containers and storage Containers—Hermetic containers. so that each mL contains about 22 mg of irbesartan
Colored containers and plastic containers for aqueous injec- (C25H28N6O), and use this solution as the sample solution.
tions may be used. Separately, weigh accurately about 44 mg of irbesartan for
assay (separately determine the water <2.48> in the same
manner as Irbesartan), and dissolve in methanol to make

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2711

exactly 20 mL. Pipet 2 mL of this solution, add the dissolu- mL of the standard solution under the above operating con-
tion medium to make exactly 200 mL, and use this solution ditions, the number of theoretical plates and the symmetry
as the standard solution. Determine the absorbances, AT factor of the peak of irbesartan are not less than 10,000 and
and AS, of the sample solution and standard solution at 244 not more than 1.5, respectively.
nm as directed under Ultraviolet-visible Spectrophotometry System repeatability: When the test is repeated 6 times
<2.24>, using the dissolution medium as the control. with 15 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
Dissolution rate (z) with respect to the labeled amount
area of irbesartan is not more than 1.0z.
of irbesartan (C25H28N6O)
= MS × AT/AS × V?/V × 1/C × 45 Containers and storage Containers—Tight containers.

MS: Amount (mg) of irbesartan for assay taken, calcu-


lated on the anhydrous basis
Add the following:
C: Labeled amount (mg) of irbesartan (C25H28N6O) in 1
tablet
Irbesartan and Amlodipine Besilate
Assay To 10 Irbesartan Tablets add 15 mL of water, shake
vigorously to disintegrate, and add 150 mL of methanol. Tablets
Shake vigorously for 15 minutes, add methanol to make
イルベサルタン・アムロジピンベシル酸塩錠
exactly 200 mL, and centrifuge. Pipet V mL of the super-
natant liquid, equivalent to about 20 mg of irbesartan
Irbesartan and Amlodipine Besilate Tablets contain
(C25H28N6O), and add a mixture of water and acetonitrile
not less than 95.0z and not more than 105.0z of the
for liquid chromatography (3:2) to make exactly 20 mL.
labeled amount of irbesartan (C25H28N6O: 428.53)
Pipet 2.5 mL of this solution, add a mixture of water and
and amlodipine besilate (C20H25ClN2O5.C6H6O3S:
acetonitrile for liquid chromatography (3:2) to make exactly
567.05).
20 mL, and use this solution as the sample solution. Sepa-
rately, weigh accurately about 25 mg of irbesartan for assay Method of preparation Prepare as directed under Tablets,
(separately determine the water <2.48> in the same manner with Irbesartan and Amlodipine Besilate.
as Irbesartan), and dissolve in methanol to make exactly 10
Identification (1) Perform the test with 5 mL each of the
mL. Pipet 2.5 mL of this solution, add a mixture of water
sample solution and standard solution obtained in the Assay
and acetonitrile for liquid chromatography (3:2) to make
(1) as directed under Liquid Chromatography <2.01> ac-
exactly 50 mL, and use this solution as the standard solu-
cording to the following conditions: the retention time of
tion. Perform the test with exactly 15 mL each of the sample
the peak of irbesartan in the chromatogram from the sam-
solution and standard solution as directed under Liquid
ple solution is the same with that of the principal peak in the
Chromatography <2.01> according to the following condi-
chromatogram from the standard solution, and both ab-
tions, and determine the peak areas, AT and AS, of irbesar-
sorption spectra of these peaks exhibit similar intensities of
tan in each solution.
absorption at the same wavelengths.
Amount (mg) of irbesartan (C25H28N6O) in 1 tablet Operating conditions—
= MS × AT/AS × 16/V Column, column temperature, mobile phase and flow
rate: Proceed as directed in the operating conditions in the
MS: Amount (mg) of irbesartan for assay taken, calcu-
Assay (1).
lated on the anhydrous basis
Detector: A photodiode array detector (wavelength: 237
Operating conditions— nm, spectrum range of measurement: 210 – 400 nm).
Detector: An ultraviolet absorption photometer (wave- System suitability—
length: 220 nm). System performance: Proceed as directed in the system
Column: A stainless steel column 4.6 mm in inside diame- suitability in the Assay (1).
ter and 25 cm in length, packed with octadecylsilanized (2) Perform the test with 5 mL each of the sample solu-
silica gel for liquid chromatography (5 mm in particle diame- tion and standard solution obtained in the Assay (2) as di-
ter). rected under Liquid Chromatography <2.01> according to
Column temperature: A constant temperature of about the following conditions: the retention time of the peak of
259C. amlodipine in the chromatogram from the sample solution
Mobile phase: To 5.5 mL of phosphoric acid add 950 mL is the same with that of the principal peak in the chromato-
of water, adjust to pH 3.0 with triethylamine, and add gram from the standard solution, and both absorption spec-
water to make 1000 mL. To 3 volume of this solution add 2 tra of these peaks exhibit similar intensities of absorption at
volume of acetonitrile for liquid chromatography. the same wavelengths.
Flow rate: Adjust so that the retention time of irbesartan Operating conditions—
is about 13 minutes. Column, column temperature, mobile phase and flow
System suitability— rate: Proceed as directed in the operating conditions in the
System performance: When the procedure is run with 15 Assay (1).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2712 Official Monographs Supplement I, JP XVII

Detector: A photodiode array detector (wavelength: 237 0.45 mm. Discard the first 10 mL or more of the filtrate,
nm, spectrum range of measurement: 210 – 400 nm). pipet V mL of the subsequent filtrate, and add the mobile
System suitability— phase to make exactly V? mL so that each mL contains
System performance: Proceed as directed in the system about 0.11 mg of irbesartan (C25H28N6O). Pipet 2 mL of
suitability in the Assay (2). this solution, add exactly 2 mL of the mobile phase, and use
this solution as the sample solution. Separately, weigh accu-
Uniformity of dosage unit <6.02> (1) Irbesartan—Per-
rately about 20 mg of irbesartan for assay (separately deter-
form the Mass variation test, or the Content uniformity test
mine the water <2.48> in the same manner as Irbesartan),
according to the following method: it meets the require-
dissolve in the mobile phase to make exactly 25 mL, and use
ment.
this solution as the irbesartan standard stock solution. Pipet
To 1 tablet of Irbesartan and Amlodipine Besilate Tablets
7 mL of the irbesartan standard stock solution, and add the
add 4 mL of 0.02 mol/L phosphate buffer solution (pH
mobile phase to make exactly 50 mL. Pipet 5 mL of this so-
3.0), and sonicate. Add 16 mL of methanol, shake vigor-
lution, add exactly 5 mL of the dissolution medium, and use
ously until the tablet is disintegrated completely, and add
this solution as the standard solution. Perform the test with
the mobile phase to make exactly 100 mL. Pipet V mL of
exactly 10 mL each of the sample solution and standard so-
this solution, add the mobile phase to make exactly V? mL
lution as directed under Liquid Chromatography <2.01> ac-
so that each mL contains about 1 mg of irbesartan
cording to the following conditions, and determine the peak
(C25H28N6O), and filter through a membrane filter with a
areas, AT and AS, of irbesartan in each solution.
pore size not exceeding 0.45 mm. Discard the first 5 mL of
the filtrate, and use the subsequent filtrate as the sample so- Dissolution rate (z) with respect to the labeled amount
lution. Then, proceed as directed under the Assay (1). of irbesartan (C25H28N6O)
= MS × AT/AS × V?/V × 1/C × 504
Amount (mg) of irbesartan (C25H28N6O)
= MS × AT/AS × V?/V × 2 MS: Amount (mg) of irbesartan for assay taken, calcu-
lated on the anhydrous basis
MS: Amount (mg) of irbesartan for assay taken, calcu-
C: Labeled amount (mg) of irbesartan (C25H28N6O) in 1
lated on the anhydrous basis
tablet
(2) Amlodipine besilate—Perform the test according to
Operating conditions—
the following method: it meets the requirement of the Con-
Proceed as directed in the operating conditions in the
tent uniformity test.
Assay (1).
To 1 tablet of Irbesartan and Amlodipine Besilate Tablets
System suitability—
add 4 mL of 0.02 mol/L phosphate buffer solution (pH
System performance: To 7 mL of the irbesartan standard
3.0), and sonicate. Add 16 mL of methanol, shake vigor-
stock solution and 5 mL of the amlodipine besilate standard
ously until the tablet is disintegrated completely, and add
stock solution obtained in (2) add the mobile phase to make
the mobile phase to make exactly 100 mL. Pipet V mL of
50 mL. To 5 mL of this solution add 5 mL of the dissolu-
this solution, add the mobile phase to make exactly V? mL
tion medium. When the procedure is run with 10 mL of this
so that each mL contains about 69 mg of amlodipine besilate
solution under the above operating conditions, amlodipine
(C20H25ClN2O5.C6H6O3S), and filter through a membrane
and irbesartan are eluted in this order with the resolution
filter with a pore size not exceeding 0.45 mm. Discard the
between these peaks being not less than 5.
first 5 mL of the filtrate, and use the subsequent filtrate as
System repeatability: When the test is repeated 6 times
the sample solution. Then, proceed as directed under the
with 10 mL of the standard solution under the above operat-
Assay (2).
ing conditions, the relative standard deviation of the peak
Amount (mg) of amlodipine besilate area of irbesartan is not more than 2.0z.
(C20H25ClN2O5.C6H6O3S) (2) Amlodipine besilate—When the test is performed at
= MS × AT/AS × V?/V × 1/5 50 revolutions per minute according to the Paddle method,
using 900 mL of 2nd fluid for dissolution test as the dissolu-
MS: Amount (mg) of Amlodipine Besilate RS taken, cal-
tion medium, the dissolution rate in 30 minutes of Irbesar-
culated on the anhydrous basis
tan and Amlodipine Besilate Tablets is not less than 75z.
Dissolution <6.10> (1) Irbesartan—When the test is per- Start the test with 1 tablet of Irbesartan and Amlodipine
formed at 50 revolutions per minute according to the Pad- Besilate Tablets, withdraw not less than 15 mL of the me-
dle method, using 900 mL of 2nd fluid for dissolution test dium at the specified minute after starting the test, and filter
as the dissolution medium, the dissolution rate in 30 through a membrane filter with a pore size not exceeding
minutes of Irbesartan and Amlodipine Besilate Tablets is 0.45 mm. Discard the first 10 mL of the filtrate, pipet V mL
not less than 70z. of the subsequent filtrate, and add the mobile phase to
Start the test with 1 tablet of Irbesartan and Amlodipine make exactly V? mL so that each mL contains about 7.7 mg
Besilate Tablets, withdraw not less than 15 mL of the me- of amlodipine besilate (C20H25ClN2O5.C6H6O3S). Pipet 2
dium at the specified minute after starting the test, and filter mL of this solution, add exactly 2 mL of the mobile phase,
through a membrane filter with a pore size not exceeding and use this solution as the sample solution. Separately,

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2713

weigh accurately about 26 mg of Amlodipine Besilate RS tion. Perform the test with exactly 5 mL each of the sample
(separately determine the water <2.48> in the same manner solution and standard solution as directed under Liquid
as Amlodipine Besilate), and dissolve in the mobile phase to Chromatography <2.01> according to the following condi-
make exactly 50 mL. Pipet 15 mL of this solution, add the tions, and determine the peak areas, AT and AS, of irbesar-
mobile phase to make exactly 100 mL, and use this solution tan in each solution.
as the amlodipine besilate standard stock solution. Pipet 5
Amount (mg) of irbesartan (C25H28N6O) in 1 tablet
mL of the amlodipine besilate standard stock solution, and
= MS × AT/AS × V?/V × 2/5
add the mobile phase to make exactly 50 mL. Pipet 5 mL of
this solution, add exactly 5 mL of the dissolution medium, MS: Amount (mg) of irbesartan for assay taken, calcu-
and use this solution as the standard solution. Perform the lated on the anhydrous basis
test with exactly 10 mL each of the sample solution and
Operating conditions—
standard solution as directed under Liquid Chromatogra-
Detector: An ultraviolet absorption photometer (wave-
phy <2.01> according to the following conditions, and deter-
length: 237 nm).
mine the peak areas, AT and AS, of amlodipine in each solu-
Column: A stainless steel column 3.0 mm in inside diame-
tion.
ter and 75 mm in length, packed with octadecylsilanized
Dissolution rate (z) with respect to the labeled amount silica gel for liquid chromatography (2.2 mm in particle di-
of amlodipine besilate (C20H25ClN2O5.C6H6O3S) ameter).
= MS × AT/AS × V?/V × 1/C × 27 Column temperature: A constant temperature of about
409C.
MS: Amount (mg) of Amlodipine Besilate RS taken, cal-
Mobile phase: A mixture of methanol and 0.02 mol/L
culated on the anhydrous basis
phosphate buffer solution (pH 3.0) (3:2).
C: Labeled amount (mg) of amlodipine besilate
Flow rate: Adjust so that the retention time of irbesartan
(C20H25ClN2O5.C6H6O3S) in 1 tablet
is about 3 minutes.
Operating conditions— System suitability—
Proceed as directed in the operating conditions in the System performance: To 10 mL of the irbesartan stand-
Assay (1). ard stock solution and 2 mL of the amlodipine besilate
System suitability— standard stock solution obtained in (2) add 0.02 mol/L
System performance: To 7 mL of the irbesartan standard phosphate buffer solution (pH 3.0) to make 20 mL. When
stock solution obtained in (1) and 5 mL of the amlodipine the procedure is run with 5 mL of this solution under the
besilate standard stock solution add the mobile phase to above operating conditions, amlodipine and irbesartan are
make 50 mL. To 5 mL of this solution add 5 mL of the dis- eluted in this order with the resolution between these peaks
solution medium. When the procedure is run with 10 mL of being not less than 5.
this solution under the above operating conditions, amlodi- System repeatability: When the test is repeated 6 times
pine and irbesartan are eluted in this order with the resolu- with 5 mL of the standard solution under the above operat-
tion between these peaks being not less than 5. ing conditions, the relative standard deviation of the peak
System repeatability: When the test is repeated 6 times area of irbesartan is not more than 1.0z.
with 10 mL of the standard solution under the above operat- (2) Amlodipine besilate—To 10 tablets of Irbesartan
ing conditions, the relative standard deviation of the peak and Amlodipine Besilate Tablets add 20 mL of 0.02 mol/L
area of amlodipine is not more than 2.0z. phosphate buffer solution (pH 3.0), and sonicate. Add 120
mL of methanol, shake vigorously until the tablets are dis-
Assay (1) Irbesartan—To 10 tablets of Irbesartan and
integrated completely, and add the mobile phase to make
Amlodipine Besilate Tablets add 20 mL of 0.02 mol/L
exactly 200 mL. Pipet V mL of this solution, add the mobile
phosphate buffer solution (pH 3.0), and sonicate. Add 120
phase to make exactly V? mL so that each mL contains
mL of methanol, shake vigorously until the tablets are dis-
about 69 mg of amlodipine besilate (C20H25ClN2O5.
integrated completely, and add the mobile phase to make
C6H6O3S), and filter through a membrane filter with a pore
exactly 200 mL. Pipet V mL of this solution, add the mobile
size not exceeding 0.45 mm. Discard the first 5 mL of the fil-
phase to make exactly V? mL so that each mL contains
trate, and use the subsequent filtrate as the sample solution.
about 1 mg of irbesartan (C25H28N6O), and filter through a
Separately, weigh accurately about 35 mg of Amlodipine
membrane filter with a pore size not exceeding 0.45 mm.
Besilate RS (separately determine the water <2.48> in the
Discard the first 5 mL of the filtrate, and use the subsequent
same manner as Amlodipine Besilate), dissolve in methanol
filtrate as the sample solution. Separately, weigh accurately
to make exactly 50 mL, and use this solution as the amlodi-
about 50 mg of irbesartan for assay (separately determine
pine besilate standard stock solution. Pipet 2 mL of the am-
the water <2.48> in the same manner as Irbesartan), dissolve
lodipine besilate standard stock solution, add 10 mL of
in methanol to make exactly 25 mL, and use this solution as
methanol, add 0.02 mol/L phosphate buffer solution (pH
the irbesartan standard stock solution. Pipet 10 mL of the
3.0) to make exactly 20 mL, and use this solution as the
irbesartan standard stock solution, add 2 mL of methanol,
standard solution. Perform the test with exactly 5 mL each
add 0.02 mol/L phosphate buffer solution (pH 3.0) to make
of the sample solution and standard solution as directed
exactly 20 mL, and use this solution as the standard solu-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2714 Official Monographs Supplement I, JP XVII

under Liquid Chromatography <2.01> according to the fol- for Identification: both spectra exhibit similar intensities of
lowing conditions, and determine the peak areas, AT and absorption at the same wave numbers.
AS, of amlodipine in each solution.
Isomer ratio Place 10 mg of Anhydrous Lactose in a screw
Amount (mg) of amlodipine besilate capped reaction vial for gas chromatography, add 4 mL of a
(C20H25ClN2O5.C6H6O3S) in 1 tablet mixture of pyridine, trimethylsilylimidazole and dimethyl-
= MS × AT/AS × V?/V × 1/25 sulfoxide (117:44:39), stopper, and sonicate at room tem-
perature for 20 minutes. After cooling, transfer 400 mL of
MS: Amount (mg) of Amlodipine Besilate RS taken, cal-
this solution into a vial for injection, add 1 mL of pyridine,
culated on the anhydrous basis
stopper tightly, mix, and use this fluid as the sample solu-
Operating conditions— tion. Perform the test with 0.5 mL of the sample solution as
Proceed as directed in the operating conditions in (1). directed under Gas Chromatography <2.02> according to the
System suitability— following conditions. Determine the peak areas of a-lactose
System performance: To 10 mL of the irbesartan stand- and b-lactose, Aa and Ab, and calculate the contents (z) of
ard stock solution obtained in (1) and 2 mL of the amlodi- a-lactose and b-lactose in Anhydrous Lactose by the follow-
pine besilate standard stock solution add 0.02 mol/L phos- ing equations.
phate buffer solution (pH 3.0) to make 20 mL. When the
Content (z) of a-lactose = Aa/(Aa + Ab) × 100
procedure is run with 5 mL of this solution under the above
operating conditions, amlodipine and irbesartan are eluted Content (z) of b-lactose = Ab/(Aa + Ab) × 100
in this order with the resolution between these peaks being
Operating conditions—
not less than 5.
Detector: A hydrogen flame-ionization detector.
System repeatability: When the test is repeated 6 times
Column: A fused silica column 0.25 mm in inside diame-
with 5 mL of the standard solution under the above operat-
ter and 15 m in length, coated with 5z diphenyl-95z
ing conditions, the relative standard deviation of the peak
dimethylpolysiloxane in 0.25 mm thickness. Use a middle
area of amlodipine is not more than 1.0z.
polar inertness fused silica column 0.53 mm in inside diame-
Containers and storage Containers—Tight containers. ter and 2 m in length as a guard column.
Column temperature: Maintain the temperature at 809C
for 1 minute after injection, raise to 1509C at a rate of 359C
Isosorbide Mononitrate 70z/ per minute, then raise to 3009C at a rate of 129C per
minute, and maintain at 3009C for 2 minutes.
Lactose 30z Injection port temperature: A constant temperature of
about 2759 C, or use cold-on column injection.
70z一硝酸イソソルビド乳糖末
Detector temperature: A constant temperature of about
3259C.
Change the Identification (2) as follows:
Carrier gas: Helium.
Identification Flow rate: 2.8 mL per minute (Retention time of b-lactose
(2) Dry the residue obtained in (1) at 809 C for 2 hours. is about 12 minutes).
Determine the infrared absorption spectrum of the residue Sprit ratio: Spritless.
as directed in the potassium bromide disk method under In- System suitability—
frared Spectrophotometry <2.25>, and compare the spec- System performance: Prepare a solution with 10 mg of a
trum with the Reference Spectrum of Lactose Hydrate or mixture of a-lactose and b-lactose (1:1) in the same manner
the spectrum of Lactose RS for Identification: both spectra as for preparing the sample solution, and proceed with 0.5
exhibit similar intensities of absorption at the same wave mL of this solution under the above operating conditions,
numbers. and determine the retention times of the peaks of a-lactose
and b-lactose: the relative retention time of a-lactose to b-
lactose is about 0.9 with the resolution between these peaks
Anhydrous Lactose being not less than 3.0.
◆System repeatability: When the test is repeated 6 times

無水乳糖 with 0.5 mL of the solution used in the system performance


under the above operating conditions, the relative standard
Change the Identification and Isomer ratio as deviation of the peak area of b-lactose is not more than
follows: 5.0z.◆

Identification Determine the infrared absorption spectrum


of Anhydrous Lactose, previously dried, as directed in the
potassium bromide disk method under Infrared Spectro-
photometry <2.25>, and compare the spectrum with the Ref-
erence Spectrum or the spectrum of Anhydrous Lactose RS

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2715

(1 in 10,000), and wash with 30 mL of water: the precipitate


Lactose Hydrate has a blue color.

Purity (1) Soluble salts—To 10.0 g of Magnesium


乳糖水和物
Aluminosilicate add 150 mL of water, boil gently for 15
minutes while shaking thoroughly. After cooling, add water
Change the Identification as follows:
to make 150 mL, and centrifuge. To 75 mL of the superna-
Identification Determine the infrared absorption spectrum tant liquid add water to make 100 mL, and use this solution
of Lactose Hydrate, previously dried, as directed in the po- as the sample solution. Evaporate 25 mL of the sample solu-
tassium bromide disk method under Infrared Spectropho- tion on a water bath to dryness, then ignite the residue at
tometry <2.25>, and compare the spectrum with ◆the Refer- 7009C for 2 hours: the mass of the residue is not more than
ence Spectrum or◆ the spectrum of Lactose RS for Identifi- 20 mg.
cation: both spectra exhibit similar intensities of absorption (2) Alkalinity—To 20 mL of the sample solution ob-
at the same wave numbers. tained in (1), add 2 drops of phenolphthalein TS, and add
0.1 mol/L hydrochloric acid VS until the solution becomes
colorless: the consumed volume is not more than 0.50 mL.
Delete the following Monographs: (3) Chloride <1.03>—To 10 mL of the sample solution
obtained in (1), add 6 mL of dilute nitric acid and water to
Lanatoside C make 50 mL. Perform the test using this solution as the test
solution. Prepare the control solution with 0.75 mL of 0.01
ラナトシドC mol/L hydrochloric acid VS (not more than 0.053z).
(4) Sulfate <1.14>—To 2 mL of the sample solution ob-
tained in (1), add 1 mL of dilute hydrochloric acid and
Lanatoside C Tablets water to make 50 mL. Perform the test using this solution
as the test solution. Prepare the control solution with 1.0
ラナトシドC錠 mL of 0.005 mol/L sulfuric acid VS (not more than
0.480z).
(5) Heavy metals <1.07>—To 2.67 g of Magnesium
Add the following: Aluminosilicate add 20 mL of water and 8 mL of hydro-
chloric acid, evaporate to dryness on a water bath. To the
Magnesium Aluminosilicate residue add 5 mL of dilute acetic acid and 20 mL of water,
boil for 2 minutes, add 0.4 g of hydroxylammonium chlo-
ケイ酸アルミン酸マグネシウム ride, and heat to boiling. After cooling, add water to make
exactly 100 mL, and filter. Pipet 25 mL of the filtrate,
Magnesium Aluminosilicate contains not less than adjust to pH 3.0 with dilute acetic acid or ammonia TS, and
27.0z and not more than 34.3z of aluminum oxide add water to make 50 mL. Perform the test using this solu-
(Al2O3: 101.96), not less than 20.5z and not more tion as the test solution. Prepare the control solution as
than 27.7z of magnesium oxide (MgO: 40.30), and follows: evaporate 2 mL of hydrochloric acid to dryness on
not less than 14.4z and not more than 21.7z of sili- a water bath, add 2.0 mL of Standard Lead Solution, 0.1 g
con dioxide (SiO2: 60.08), calculated on the dried of hydroxylammonium chloride and water to make 25 mL,
basis. adjust to pH 3.0 with dilute acetic acid or ammonia TS, and
add water to make 50 mL (not more than 30 ppm).
Description Magnesium Aluminosilicate occurs as a white,
(6) Iron—To 0.11 g of Magnesium Aluminosilicate add
powder or grain.
8 mL of 2 mol/L nitric acid TS, boil for 1 minute, cool, add
It is practically insoluble in water and in ethanol (99.5).
water to make exactly 100 mL, and centrifuge. Pipet 30 mL
When heat 1 g of Magnesium Aluminosilicate with 10 mL
of the supernatant liquid, add water to make 45 mL, add 2
of dilute hydrochloric acid, most of it dissolves.
mL of hydrochloric acid, and shake. Add 50 mg of ammo-
Identification (1) To 0.5 g of Magnesium Aluminosili- nium peroxodisulfate and 3 mL of a solution of ammonium
cate add 5 mL of diluted sulfuric acid (1 in 3), heat until thiocyanate (3 in 10), and shake: the solution is not more
white fumes are evolved, cool, add 20 mL of water, and fil- colored than the following control solution (not more than
trate. Neutralize the filtrate with ammonia TS, and filter the 0.03z).
precipitate produced. Dissolve the residue in dilute hydro- Control solution: Pipet 1 mL of Standard Iron Solution,
chloric acid: the solution responds to Qualitative Tests add water to make 45 mL, add 2 mL of hydrochloric acid,
<1.09> for aluminum salt. shake, and proceed in the same manner.
(2) The filtrate obtained in (1) responds to Qualitative (7) Arsenic < 1.11 > —To 1.0 g of Magnesium
Tests <1.09> (2) for magnesium salt. Aluminosilicate add 10 mL of water and 1 mL of sulfuric
(3) Wash the residue obtained in (1) with 30 mL of acid, and shake thoroughly. After cooling, perform the test
water, add 2 mL of a solution of methylene blue trihydrate using this solution as the test solution (not more than 2

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2716 Official Monographs Supplement I, JP XVII

ppm). through a filter paper for quantitative analysis, add 10 mL


of hot water to the residue, stir, and decant the supernatant
Loss on drying <2.41> Not more than 20.0z (1 g, 1109C,
liquid on a filter paper to filter. Then wash the residue with
7 hours).
three 10-mL portions of hot water, add 50 mL of water to
Acid-consuming capacity <6.04> Weigh accurately about the residue, and heat on a water bath for 15 minutes. Trans-
0.2 g of Magnesium Aluminosilicate, transfer to a glass- fer the residue onto the filter paper, wash the residue with
stoppered flask, add exactly 100 mL of 0.1 mol/L hydro- hot water until the last 5 mL of washing yields no precipi-
chloric acid VS, stopper the flask tightly, shake at 37 ± 29 C tate on addition of 1 mL of silver nitrate TS, place the
for 1 hour, and filter. Pipet 50 mL of the filtrate, and titrate residue and the filter paper in a platinum crucible, ignite to
<2.50> the excess hydrochloric acid, while stirring thor- ash, and then ignite at 800 ± 259C for 1 hour. After cool-
oughly, with 0.1 mol/L sodium hydroxide VS until the pH ing, weigh the crucible, and designate the mass as a (g).
of the solution becomes 3.5. Perform a blank determination Then add 6 mL of hydrofluoric acid, evaporate to dryness,
in the same manner. The consumed volume of 0.1 mol/L ignite for 5 minutes, weigh the crucible after cooling, and
hydrochloric acid VS is not less than 250 mL per g of Mag- designate the mass as b (g).
nesium Aluminosilicate calculated on the dried basis.
Amount (g) of silicon dioxide (SiO2) = a – b
Assay (1) Aluminum oxide—Weigh accurately about
Containers and storage Containers—Well-closed contain-
1.25 g of Magnesium Aluminosilicate, transfer to a conical
ers.
flask, add 10 mL of 3 mol/L hydrochloric acid TS and 50
mL of water, and heat on a water bath for 15 minutes. To
the solution add 8 mL of hydrochloric acid, heat on a water
Add the following:
bath for 10 minutes. After cooling, transfer to a 250-mL
volumetric flask, rinse the flask with water, and add water
to make 250 mL. Centrifuge the solution, and use the super- Magnesium Aluminometasilicate
natant liquid as the sample solution. Pipet 20 mL of the
メタケイ酸アルミン酸マグネシウム
sample solution, add exactly 20 mL of 0.05 mol/L disodium
dihydrogen ethylenediamine tetraacetate VS. To this solu-
Magnesium Aluminometasilicate contains not less
tion add 15 mL of acetic acid–ammonium acetate buffer so-
than 29.1z and not more than 35.5z of aluminum
lution (pH 4.8) and 20 mL of water, and boil for 5 minutes.
oxide (Al2O3: 101.96), not less than 11.4z and not
After cooling, add 50 mL of ethanol (95), and titrate <2.50>
more than 14.0z of magnesium oxide (MgO: 40.30),
with 0.05 mol/L zinc sulfate VS until the color of the solu-
and not less than 29.2z and not more than 35.6z of
tion changes from light dark green to light red (indicator: 2
silicon dioxide (SiO2: 60.08), calculated on the dried
mL of dithizone TS). Perform a blank determination in the
basis.
same manner.
Description Magnesium Aluminometasilicate occurs as a
Each mL of 0.05 mol/L disodium dihydrogen
white, powder or grain.
ethylenediamine tetraacetate VS
It is practically insoluble in water and in ethanol (99.5).
= 2.549 mg of Al2O3
When heat 1 g of Magnesium Aluminometasilicate with
(2) Magnesium oxide—Pipet 50 mL of the sample solu- 10 mL of dilute hydrochloric acid, most of it dissolves.
tion obtained in (1), add 50 mL of water and 25 mL of a so-
Identification (1) To 0.5 g of Magnesium Alumino-
lution of 2,2?,2!-nitrilotriethanol (1 in 2), shake thoroughly,
metasilicate add 5 mL of diluted sulfuric acid (1 in 3), heat
then add 25 mL of ammonia-ammonium chloride buffer so-
until white fumes are evolved, cool, add 20 mL of water,
lution (pH 10.7), and titrate <2.50> with 0.05 mol/L disodi-
and filtrate. Neutralize the filtrate with ammonia TS, and
um dihydrogen ethylenediamine tetraacetate VS until the
filter the precipitate produced. Dissolve the residue in dilute
color of the solution changes from red-purple to blue lasting
hydrochloric acid: the solution responds to Qualitative
for 30 seconds (indicator: 40 mg of eriochrome black T-so-
Tests <1.09> for aluminum salt.
dium chloride indicator).
(2) The filtrate obtained in (1) responds to Qualitative
Each mL of 0.05 mol/L disodium dihydrogen Tests <1.09> (2) for magnesium salt.
ethylenediamine tetraacetate VS (3) Wash the residue obtained in (1) with 30 mL of
= 2.015 mg of MgO water, add 2 mL of a solution of methylene blue trihydrate
(1 in 10,000), and wash with 30 mL of water: the precipitate
(3) Silicon dioxide—Weigh accurately about 1 g of
has a blue color.
Magnesium Aluminosilicate, add 30 mL of dilute hydro-
chloric acid, and evaporate to dryness on a water bath. Purity (1) Soluble salts—To 10.0 g of Magnesium Alu-
Moisten the residue with hydrochloric acid, evaporate to minometasilicate add 150 mL of water, boil gently for 15
dryness on a water bath. To the residue add 8 mL of hydro- minutes while shaking thoroughly. After cooling, add water
chloric acid, stir, then add 25 mL of hot water, and stir to make 150 mL, and centrifuge. To 75 mL of the superna-
again. After allowing to stand, filter the supernatant liquid tant liquid add water to make 100 mL, and use this solution

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2717

as the sample solution. Evaporate 25 mL of the sample solu- titrate <2.50> the excess hydrochloric acid, while stirring
tion on a water bath to dryness, then ignite the residue at thoroughly, with 0.1 mol/L sodium hydroxide VS until the
7009C for 2 hours: the mass of the residue is not more than pH of the solution becomes 3.5. Perform a blank determi-
20 mg. nation in the same manner. The consumed volume of 0.1
(2) Alkalinity—To 20 mL of the sample solution ob- mol/L hydrochloric acid VS is not less than 210 mL per g of
tained in (1), add 2 drops of phenolphthalein TS, and add Magnesium Aluminometasilicate calculated on the dried
0.1 mol/L hydrochloric acid VS until the solution becomes basis.
colorless: the consumed volume is not more than 0.50 mL.
Assay (1) Aluminum oxide—Weigh accurately about
(3) Chloride <1.03>—To 10 mL of the sample solution
1.25 g of Magnesium Aluminometasilicate, transfer to a
obtained in (1), add 6 mL of dilute nitric acid and water to
conical flask, add 10 mL of 3 mol/L hydrochloric acid TS
make 50 mL. Perform the test using this solution as the test
and 50 mL of water, and heat on a water bath for 15
solution. Prepare the control solution with 0.75 mL of 0.01
minutes. To the solution add 8 mL of hydrochloric acid,
mol/L hydrochloric acid VS (not more than 0.053z).
heat on a water bath for 10 minutes. After cooling, transfer
(4) Sulfate <1.14>—To 2 mL of the sample solution ob-
to a 250-mL volumetric flask, rinse the flask with water,
tained in (1), add 1 mL of dilute hydrochloric acid and
and add water to make 250 mL. Centrifuge the solution,
water to make 50 mL. Perform the test using this solution
and use the supernatant liquid as the sample solution. Pipet
as the test solution. Prepare the control solution with 1.0
20 mL of the sample solution, add exactly 20 mL of 0.05
mL of 0.005 mol/L sulfuric acid VS (not more than
mol/L disodium dihydrogen ethylenediamine tetraacetate
0.480z).
VS. To this solution add 15 mL of acetic acid–ammonium
(5) Heavy metals <1.07>—To 2.67 g of Magnesium Alu-
acetate buffer solution (pH 4.8) and 20 mL of water, and
minometasilicate add 20 mL of water and 8 mL of hydro-
boil for 5 minutes. After cooling, add 50 mL of ethanol
chloric acid, evaporate to dryness on a water bath. To the
(95), and titrate <2.50> with 0.05 mol/L zinc sulfate VS until
residue add 5 mL of dilute acetic acid and 20 mL of water,
the color of the solution changes from light dark green to
boil for 2 minutes, add 0.4 g of hydroxylammonium chlo-
light red (indicator: 2 mL of dithizone TS). Perform a blank
ride, and heat to boiling. After cooling, add water to make
determination in the same manner.
exactly 100 mL, and filter. Pipet 25 mL of the filtrate,
adjust to pH 3.0 with dilute acetic acid or ammonia TS, and Each mL of 0.05 mol/L disodium dihydrogen
add water to make 50 mL. Perform the test using this solu- ethylenediamine tetraacetate VS
tion as the test solution. Prepare the control solution as = 2.549 mg of Al2O3
follows: evaporate 2 mL of hydrochloric acid to dryness on
(2) Magnesium oxide—Pipet 50 mL of the sample solu-
a water bath, add 2.0 mL of Standard Lead Solution, 0.1 g
tion obtained in (1), add 50 mL of water and 25 mL of a so-
of hydroxylammonium chloride and water to make 25 mL,
lution of 2,2?,2!-nitrilotriethanol (1 in 2), shake thoroughly,
adjust to pH 3.0 with dilute acetic acid or ammonia TS, and
then add 25 mL of ammonia-ammonium chloride buffer so-
add water to make 50 mL (not more than 30 ppm).
lution (pH 10.7), and titrate <2.50> with 0.05 mol/L disodi-
(6) Iron—To 0.11 g of Magnesium Aluminometasilicate
um dihydrogen ethylenediamine tetraacetate VS until the
add 8 mL of 2 mol/L nitric acid TS, boil for 1 minute, cool,
color of the solution changes from red-purple to blue lasting
add water to make exactly 100 mL, and centrifuge. Pipet 30
for 30 seconds (indicator: 40 mg of eriochrome black T-so-
mL of the supernatant liquid, add water to make 45 mL,
dium chloride indicator).
add 2 mL of hydrochloric acid, and shake. Add 50 mg of
ammonium peroxodisulfate and 3 mL of a solution of am- Each mL of 0.05 mol/L disodium dihydrogen
monium thiocyanate (3 in 10), and shake: the solution is not ethylenediamine tetraacetate VS
more colored than the following control solution (not more = 2.015 mg of MgO
than 0.03z).
(3) Silicon dioxide—Weigh accurately about 1 g of
Control solution: Pipet 1 mL of Standard Iron Solution,
Magnesium Aluminometasilicate, add 30 mL of dilute hy-
add water to make 45 mL, add 2 mL of hydrochloric acid,
drochloric acid, and evaporate to dryness on a water bath.
shake, and proceed in the same manner.
Moisten the residue with hydrochloric acid, evaporate to
(7) Arsenic <1.11>—To 1.0 g of Magnesium Alumino-
dryness on a water bath. To the residue add 8 mL of hydro-
metasilicate add 10 mL of water and 1 mL of sulfuric acid,
chloric acid, stir, then add 25 mL of hot water, and stir
and shake thoroughly. After cooling, perform the test using
again. After allowing to stand, filter the supernatant liquid
this solution as the test solution (not more than 2 ppm).
through a filter paper for quantitative analysis, add 10 mL
Loss on drying <2.41> Not more than 20.0z (1 g, 1109C, of hot water to the residue, stir, and decant the supernatant
7 hours). liquid on a filter paper to filter. Then wash the residue with
three 10-mL portions of hot water, add 50 mL of water to
Acid-consuming capacity <6.04> Weigh accurately about
the residue, and heat on a water bath for 15 minutes. Trans-
0.2 g of Magnesium Aluminometasilicate, transfer to a
fer the residue onto the filter paper, wash the residue with
glass-stoppered flask, add exactly 100 mL of 0.1 mol/L hy-
hot water until the last 5 mL of washing yields no precipi-
drochloric acid VS, stopper the flask tightly, shake at 37 ±
tate on addition of 1 mL of silver nitrate TS, place the
29C for 1 hour, and filter. Pipet 50 mL of the filtrate, and

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2718 Official Monographs Supplement I, JP XVII

residue and the filter paper in a platinum crucible, ignite to ble in ethanol (99.5).
ash, and then ignite at 800 ± 259C for 1 hour. After cool- It dissolves in dilute hydrochloric acid.
ing, weigh the crucible, and designate the mass as a (g).
Identification (1) Determine the absorption spectrum of
Then add 6 mL of hydrofluoric acid, evaporate to dryness,
a solution of Mesalazine in 0.1 mol/L hydrochloric acid TS
ignite for 5 minutes, weigh the crucible after cooling, and
(1 in 80,000) as directed under Ultraviolet-visible Spectro-
designate the mass as b (g).
photometry <2.24>, and compare the spectrum with the Ref-
Amount (g) of silicon dioxide (SiO2) = a – b erence Spectrum: both spectra exhibit similar intensities of
absorption at the same wavelengths.
Containers and storage Containers—Well-closed contain-
(2) Determine the infrared absorption spectrum of
ers.
Mesalazine, previously dried, as directed in the potassium
bromide disk method under Infrared Spectrophotometry
<2.25>, and compare the spectrum with the Reference Spec-
D-Mannitol trum: both spectra exhibit similar intensities of absorption
at the same wave numbers.
D-マンニトール
Purity (1) Clarity and color of solution—Perform this
Change the Purity (1) as follows: procedure while keeping the solution at 409 C. A solution
obtained by dissolving 0.5 g of Mesalazine in 20 mL of 1
Purity (1) Clarity and color of solution—Dissolve 5.0 g
mol/L hydrochloric acid TS is clear, and its absorbance at
of D-Mannitol in water to make 50 mL, and use this solu-
440 nm and 650 nm, determined immediately as directed
tion as the test solution. Perform the test with the test solu-
under Ultraviolet-visible Spectrophotometry <2.24>, is not
tion as directed under Turbidity Measurement <2.61>: the
more than 0.15 and not more than 0.10, respectively.
solution is clear. Perform the test with the test solution ac-
(2) Chloride <1.03>—Dissolve 0.30 g of Mesalazine in 6
cording to Method 2 under Methods for Color Matching
mL of dilute nitric acid and 40 mL of water, and add water
<2.65>: the solution is colorless.
to make 50 mL. Perform the test using this solution as the
test solution. Prepare the control solution with 0.80 mL of
0.01 mol/L hydrochloric acid VS (not more than 0.095z).
Delete the following Monographs:
(3) Sulfate—To 1.0 g of Mesalazine add 20 mL of
water, shake for 1 minute, and filter. To 15 mL of the fil-
Mercurochrome trate add 0.5 mL of acetic acid (31), then add 2.5 mL of the
following solution A, and use this solution as the test solu-
マーキュロクロム
tion. Solution A: To 3 mL of barium chloride TS add 4.5
mL of a solution of potassium sulfate in diluted ethanol (3
in 10) (181 in 10,000,000), shake, and allow to stand for 1
Mercurochrome Solution minute. Prepare the control solution by adding 14.7 mL of
water and 0.5 mL of acetic acid (31) to 0.31 mL of 0.005
マーキュロクロム液
mol/L sulfuric acid VS, and then proceeding in the same
manner for the test solution. Compare the test solution and
the control solution after allowing to stand for 5 minutes:
Add the following:
the turbidity of the test solution is not more intense than
that of the control solution (not more than 0.02z).
Mesalazine (4) Heavy metals <1.07>—Proceed with 0.5 g of Mesala-
zine according to Method 2, and perform the test. Prepare
メサラジン
the control solution with 1.0 mL of Standard Lead Solution
(not more than 20 ppm).
(5) Reducing substances—Dissolve 0.10 g of Mesalazine
in dilute hydrochloric acid to make 25 mL, add 0.2 mL of
C7H7NO3: 153.14 starch TS and 0.25 mL of dilute iodine TS, and allow to
5-Amino-2-hydroxybenzoic acid stand for 2 minutes: a blue or purple-brown color is pro-
[89-57-6] duced.
(6) 2-Aminophenol and 4-aminophenol—Weigh exactly
Mesalazine, when dried, contains not less than 50 mg of Mesalazine, dissolve in the mobile phase A to
98.5z and not more than 101.0z of mesalazine make exactly 50 mL, and use this solution as the sample so-
(C7H7NO3). lution. Separately, weigh exactly 5.0 mg of 2-aminophenol,
and dissolve in the mobile phase A to make exactly 100 mL.
Description Mesalazine occurs as white, light gray or red-
Pipet 10 mL of this solution, add the mobile phase A to
dish-white, crystals or crystalline powder.
make exactly 100 mL, and use this solution as the 2-
It is very slightly soluble in water, and practically insolu-
aminophenol standard stock solution. Weigh exactly 5.0 mg

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2719

of 4-aminophenol, dissolve in the mobile phase A to make 100 mL. Pipet 1 mL of this solution, add the mobile phase
exactly 250 mL, and use this solution as the 4-aminophenol to make exactly 100 mL. Pipet 1 mL of this solution, add
standard stock solution. Pipet 1 mL each of the 2- the mobile phase to make exactly 100 mL, and use this solu-
aminophenol standard stock solution and 4-aminophenol tion as the standard solution. Perform the test with exactly
standard stock solution, add the mobile phase A to make 100 mL each of the sample solution and standard solution as
exactly 100 mL, and use this solution as the standard solu- directed under Liquid Chromatography <2.01> according to
tion. Perform the test with exactly 20 mL each of the sample the following conditions, and determine the peak area of
solution and standard solution as directed under Liquid aniline in each solution: the peak area of aniline obtained
Chromatography <2.01> according to the following condi- from the sample solution is not larger than that of aniline
tions, and determine the peak areas of 4-aminophenol and from the standard solution (not more than 10 ppm).
2-aminophenol: the peak area of 4-aminophenol obtained Operating conditions—
from the sample solution is not larger than that of 4- Detector: A fluorophotometer (excitation wavelength:
aminophenol from the standard solution (not more than 280 nm, fluorescence wavelength: 340 nm).
0.02z), and the peak area of 2-aminophenol from the sam- Column: A stainless steel column 4.6 mm in inside diame-
ple solution is not larger than 4 times that of 2-aminophenol ter and 25 cm in length, packed with octadecylsilanized
from the standard solution (not more than 0.02z). silica gel for liquid chromatography (5 mm in particle diame-
Operating conditions— ter).
Detector: An ultraviolet absorption photometer (wave- Column temperature: A constant temperature of about
length: 220 nm). 409C.
Column: A stainless steel column 4.6 mm in inside diame- Mobile phase: Dissolve 9.52 g of sodium acetate trihy-
ter and 25 cm in length, packed with octadecylsilanized drate in a suitable amount of water, add 1.72 mL of acetic
silica gel for liquid chromatography (3 mm in particle diame- acid (100), then add water to make 1000 mL, and adjust to
ter). pH 5.0 with acetic acid (100) or dilute sodium hydroxide
Column temperature: A constant temperature of about TS. To 500 mL of this solution add 500 mL of acetonitrile
259C. for liquid chromatography.
Mobile phase A: Mix 2.2 g of perchloric acid and 1.0 g of Flow rate: Adjust so that the retention time of aniline is
phosphoric acid with water to make 1000 mL. about 5 minutes.
Mobile phase B: Mix 1.7 g of perchloric acid and 1.0 g of System suitability—
phosphoric acid with acetonitrile for liquid chromatography System performance: When the procedure is run with 100
to make 1000 mL. mL of the standard solution under the above operating con-
Flowing of mobile phase: Control the gradient by mixing ditions, the number of theoretical plates and the symmetry
the mobile phases A and B as directed in the following factor of the peak of aniline are not less than 3000 and not
table. more than 2.0, respectively.
System repeatability: When the test is repeated 6 times
Time after injection Mobile phase A Mobile phase B with 100 mL of the standard solution under the above oper-
of sample (min) (volz) (volz) ating conditions, the relative standard deviation of the peak
area of aniline is not more than 2.0z.
0 – 10 100 0 (8) 3-Aminophenol, 3-aminobenzoic acid, gentisic acid,
10 – 25 100 → 40 0 → 60 salicylic acid and other related substances—Weigh exactly
50 mg of Mesalazine, dissolve in the mobile phase A to
Flow rate: About 0.8 mL per minute (the retention time make exactly 50 mL, and use this solution as the sample so-
of mesalazine is about 16 minutes). lution. Pipet 1 mL of the sample solution, add the mobile
System suitability— phase A to make exactly 100 mL, and use this solution as
System performance: To 1 mL of the sample solution add the standard solution. Separately, weigh exactly 10 mg of 3-
the mobile phase A to make 200 mL. To 5 mL of this solu- aminophenol, and dissolve in the mobile phase A to make
tion add 5 mL of the 2-aminophenol standard stock solu- exactly 100 mL. Pipet 1 mL of this solution, add the mobile
tion. When the procedure is run with 20 mL of this solution phase A to make exactly 50 mL, and use this solution as the
under the above operating conditions, 2-aminophenol and 3-aminophenol standard solution. Weigh exactly 5.0 mg of
mesalazine are eluted in this order with the resolution be- 3-aminobenzoic acid, dissolve in the mobile phase A to
tween these peaks being not less than 3. make exactly 100 mL. Pipet 1 mL of this solution, add the
System repeatability: When the test is repeated 6 times mobile phase A to make exactly 50 mL, and use this solu-
with 20 mL of the standard solution under the above operat- tion as the 3-aminobezoic acid standard solution. Weigh ex-
ing conditions, the relative standard deviation of the peak actly 5.0 mg of gentisic acid, dissolve in the mobile phase A
area of 2-aminophenol is not more than 2.5z. to make exactly 100 mL. Pipet 1 mL of this solution, add
(7) Aniline—Dissolve exactly 0.10 g of Mesalazine in the mobile phase A to make exactly 50 mL, and use this so-
the mobile phase to make exactly 50 mL, and use this solu- lution as the gentisic acid standard solution. Weigh exactly
tion as the sample solution. Separately, dissolve exactly 30.5 15 mg of salicylic acid, dissolve in the mobile phase A to
mg of aniline sulfate in the mobile phase to make exactly make exactly 100 mL. Pipet 1 mL of this solution, add the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2720 Official Monographs Supplement I, JP XVII

mobile phase A to make exactly 50 mL, and use this solu- that with 10 mL of the standard solution.
tion as the salicylic acid standard solution. Perform the test System performance: To 1 mL of the sample solution and
with exactly 10 mL each of the sample solution, standard so- 2 mL of a solution of 3-aminobenzoic acid in the mobile
lution, 3-aminophenol standard solution, 3-aminobezoic phase A (1 in 20,000) add the mobile phase A to make 100
acid standard solution, gentisic acid standard solution and mL. When the procedure is run with 10 mL of this solution
salicylic acid standard solution as directed under Liquid under the above operating conditions, mesalazine and 3-
Chromatography <2.01> according to the following condi- aminobenzoic acid are eluted in this order with the resolu-
tions, and determine each peak area by the automatic in- tion between these peaks being not less than 3.
tegration method: the peak area of 3-aminophenol obtained System repeatability: When the test is repeated 6 times
from the sample solution is not larger than that from of 3- with 10 mL of the standard solution under the above operat-
aminophenol standard solution (not more than 0.2z), the ing conditions, the relative standard deviation of the peak
peak area of 3-aminobenzoic acid from the sample solution area of mesalazine is not more than 2.0z.
is not larger than that from 3-aminobenzoic acid standard
Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 2
solution (not more than 0.1z), the peak area of gentisic
hours).
acid from the sample solution is not larger than that from
gentisic acid standard solution (not more than 0.1z), and Residue on ignition <2.44> Not more than 0.2z (1 g).
the peak area of salicylic acid from the sample solution is
Assay Weigh accurately about 50 mg of Mesalazine, previ-
not larger than that from salicylic acid standard solution
ously dried, dissolve in 100 mL of hot water, cool to room
(not more than 0.3z). The area of the peak other than 3-
temperature quickly, and titrate <2.50> with 0.1 mol/L so-
aminophenol, mesalazine, 3-aminobenzoic acid, gentisic
dium hydroxide VS (potentiometric titration). Perform a
acid and salicylic acid from the sample solution is not larger
blank determination in the same manner, and make any
than 1/10 times the peak area of mesalazine from the stand-
necessary correction.
ard solution (not more than 0.1z), and the total area of the
peaks other than mesalazine from the sample solution is not Each mL of 0.1 mol/L sodium hydroxide VS
larger than the peak area of mesalazine from the standard = 15.31 mg of C7H7NO3
solution (not more than 1.0z).
Containers and storage Containers—Well-closed contain-
Operating conditions—
ers.
Detector: An ultraviolet absorption photometer (wave-
Storage—Light-resistant.
length: 220 nm).
Column: A stainless steel column 4.6 mm in inside diame-
ter and 25 cm in length, packed with octylsilanized silica gel
Add the following:
for liquid chromatography (5 mm in particle diameter).
Column temperature: A constant temperature of about
259C. Mesalazine Extended-release
Mobile phase A: Mix 2.2 g of perchloric acid and 1.0 g of Tablets
phosphoric acid with water to make 1000 mL.
Mobile phase B: Mix 1.7 g of perchloric acid and 1.0 g of メサラジン徐放錠
phosphoric acid with acetonitrile for liquid chromatography
to make 1000 mL. Mesalazine Extended-release Tablets contain not
Flowing of mobile phase: Control the gradient by mixing less than 95.0z and not more than 105.0z of the
the mobile phases A and B as directed in the following labeled amount of mesalazine (C7H7NO3: 153.14).
table.
Method of preparation Prepare as directed under Tablets,
with Mesalazine.
Time after injection Mobile phase A Mobile phase B
of sample (min) (volz) (volz) Identification Powder Mesalazine Extended-release
Tablets. To a portion of the powder, equivalent to 20 mg of
0–7 100 0 Mesalazine, add 100 mL of diluted phosphoric acid (1 in
7 – 25 100 → 40 0 → 60 1000) and shake vigorously. To 5 mL of this solution add
diluted phosphoric acid (1 in 1000) to make 50 mL, filter,
Flow rate: About 1.8 mL per minute (the retention time and determine the absorbance spectrum of the filtrate as di-
of mesalazine is about 5 minutes). rected under Ultraviolet-visible Spectrophotometry <2.24>:
Time span of measurement: For 25 minutes after injec- it exhibits maxima between 227 nm and 231 nm, and be-
tion of the sample solution. tween 298 nm and 302 nm.
System suitability—
Uniformity of dosage units <6.02> Perform the Mass vari-
Test for required detectability: To exactly 1 mL of the
ation test, or the Content uniformity test according to the
standard solution add the mobile phase A to make exactly
following method: it meets the requirement.
20 mL. Confirm that the peak area of mesalazine obtained
To 1 tablet of Mesalazine Extended-release Tablets add
with 10 mL of this solution is equivalent to 3.5 to 6.5z of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2721

6V/25 mL of diluted phosphoric acid (1 in 1000), shake tion, then add 90 mL of methanol and diluted phosphoric
until the tablet is disintegrated, then add 3V/5 mL of meth- acid (1 in 1000) to make 250 mL. Filter this solution
anol, and sonicate for 30 minutes. Add diluted phosphoric through a membrane filter with a pore size 0.45 mm. Discard
acid (1 in 1000) to make exactly V mL so that each mL con- the first 5 mL of the filtrate, and use the subsequent filtrate
tains about 1 mg of mesalazine (C7H7NO3), and centrifuge. as the sample solution. Separately, weigh accurately about
Pipet 8 mL of the supernatant liquid, add exactly 2 mL of 40 mg of mesalazine for assay, previously dried at 1059 C
the internal standard solution and 13 mL of methanol, then for 2 hours, add 100 mL of diluted phosphoric acid (1 in
add diluted phosphoric acid (1 in 1000) to make 50 mL, and 1000), shake vigorously, and sonicate for 5 minutes to dis-
use this solution as the sample solution. Then, proceed as solve. Add exactly 10 mL of the internal standard solution,
directed in the Assay. then add 90 mL of methanol and diluted phosphoric acid (1
in 1000) to make 250 mL, and use this solution as the stand-
Amount (mg) of mesalazine (C7H7NO3)
ard solution. Perform the test with 10 mL each of the sample
= MS × QT/QS × V/40
solution and standard solution as directed under Liquid
MS: Amount (mg) of mesalazine for assay taken Chromatography <2.01> according to the following condi-
tions, and calculate the ratios, QT and QS, of the peak area
Internal standard solution—A solution of ethyl aminoben-
of mesalazine to that of the internal standard.
zoate in methanol (1 in 800).
Amount (mg) of mesalazine (C7H7NO3) = MS × QT/QS
Dissolution <6.10> When the test is performed at 50 revo-
lutions per minute according to the Paddle method, using MS: Amount (mg) of mesalazine for assay taken
900 mL of 2nd fluid for dissolution test as the dissolution
Internal standard solution—A solution of ethyl aminoben-
medium, the dissolution rates in 3 hours, in 6 hours and in
zoate in methanol (1 in 800).
24 hours of Mesalazine Extended-release Tablets are 10 to
Operating conditions—
40z, 30 to 60z, and not less than 80z, respectively.
Detector: An ultraviolet absorption photometer (wave-
Start the test with 1 tablet of Mesalazine Extended-release
length: 300 nm).
Tablets, withdraw exactly 20 mL of the medium at the spe-
Column: A stainless steel column 4.0 mm in inside diame-
cified minutes after starting the test and supply exactly 20
ter and 10 cm in length, packed with octadecylsilanized
mL of dissolution medium warmed to 37 ± 0.59C immedi-
silica gel for liquid chromatography (5 mm in particle diame-
ately after withdrawing of the medium every time. Filter the
ter).
withdrawn media through a membrane filter with a pore
Column temperature: A constant temperature of about
size not exceeding 0.45 mm. Discard the first 10 mL or more
409C.
of the filtrate, pipet V mL of the subsequent filtrate, add
Mobile phase: Dissolve 400 mL of methanol, 1 mL of
the dissolution medium to make exactly V? mL so that each
phosphoric acid, 0.865 g of sodium lauryl sulfate and
mL contains about 56 mg of mesalazine (C7H7NO3), and use
0.679 g of tetrabutylammonium hydrogensulfate in water to
these solutions as the sample solutions. Separately, weigh
make 1000 mL.
accurately about 28 mg of mesalazine for assay, previously
Flow rate: Adjust so that the retention time of mesalazine
dried at 1059C for 2 hours, and dissolve in the dissolution
is about 10 minutes.
medium to make exactly 100 mL. Pipet 5 mL of this solu-
System suitability—
tion, add the dissolution medium to make exactly 25 mL,
System performance: When the procedure is run with 10
and use this solution as the standard solution. Determine
mL of the standard solution under the above operating con-
the absorbances, AT(n) and AS, of the sample solutions and
ditions, mesalazine and the internal standard are eluted in
standard solution at 330 nm as directed under Ultraviolet-
this order with the resolution between these peaks being not
visible Spectrophotometry <2.24>.
less than 3.
Dissolution rate (z) with respect to the labeled amount of System repeatability: When the test is repeated 6 times
mesalazine (C7H7NO3) on the nth medium withdrawing with 10 mL of the standard solution under the above operat-
(n = 1, 2, 3) ing conditions, the relative standard deviation of the ratio
AT(n) + ∑
Ø 1  V?
»
n-1 A 1
T(i ) of the peak area of mesalazine to that of the internal stand-
= MS × × × × × 180
 AS i=1 AS 45  V C ard is not more than 1.0z.

MS: Amount (mg) of mesalazine for assay taken Containers and storage Containers—Well-closed contain-
C: Labeled amount (mg) of mesalazine (C7H7NO3) in 1 ers.
tablet Storage—Light-resistant.

Assay Weigh accurately the mass of not less than 20


Mesalazine Extended-release Tablets, and powder. Weigh
accurately a portion of the powder, equivalent to about 40
mg of mesalazine (C7H7NO3), add 100 mL of diluted phos-
phoric acid (1 in 1000), shake vigorously, and sonicate for 5
minutes. Add exactly 10 mL of the internal standard solu-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2722 Official Monographs Supplement I, JP XVII

Add the following: Dissolution rate (z) with respect to the labeled amount
of methotrexate (C20H22N8O5)
Methotrexate Tablets = MS × AT/AS × V?/V × 1/C × 9

MS: Amount (mg) of Methotrexate RS taken, calculated


メトトレキサート錠
on the anhydrous basis
C: Labeled amount (mg) of methotrexate (C20H22N8O5) in
Methotrexate Tablets contain not less than 95.0z
1 tablet
and not more than 105.0z of the labeled amount of
methotrexate (C20H22N8O5: 454.44). (This monograph Operating conditions—
is applied to only 2.5-mg tablets.) Detector: An ultraviolet absorption photometer (wave-
length: 302 nm).
Method of preparation Prepare as directed under Tablets,
Column: A stainless steel column 4 mm in inside diameter
with Methotrexate.
and 15 cm in length, packed with octadecylsilanized silica
Identification To a quantity of powdered Methotrexate gel for liquid chromatography (5 mm in particle diameter).
Tablets, equivalent to 2.5 mg of Methotrexate, add 100 mL Column temperature: A constant temperature of about
of diluted hydrochloric acid (1 in 100), shake, and filter or 259C.
centrifuge. Determine the absorption spectrum of this solu- Mobile phase: To 250 mL of 0.2 mol/L of potassium di-
tion as directed under Ultraviolet-visible Spectrophotometry hydrogen phosphate TS add 29 mL of 0.2 mol/L sodium
<2.24>: it exhibits maxima between 241 nm and 245 nm and hydroxide TS and water to make 1000 mL. To 890 mL of
between 305 nm and 309 nm. this solution add 110 mL of acetonitrile.
Flow rate: Adjust so that the retention time of
Uniformity of dosage unit <6.02> Perform the test accord-
methotrexate is about 4 minutes.
ing to the following method: it meets the requirement of the
System suitability—
Content uniformity test.
System performance: When the procedure is run with 50
To 1 tablet of Methotrexate Tablets add the mobile
mL of the standard solution under the above operating con-
phase, stir, and add the mobile phase to make exactly V mL
ditions, the number of theoretical plates and the symmetry
so that each mL contains about 0.1 mg of methotrexate
factor of the peak of methotrexate are not less than 3000
(C20H22N8O5). Centrifuge this solution, and use the superna-
and not more than 1.5, respectively.
tant liquid as the sample solution. Then, proceed as directed
System repeatability: When the test is repeated 6 times
in the Assay.
with 50 mL of the standard solution under the above operat-
Amount (mg) of methotrexate (C20H22N8O5) ing conditions, the relative standard deviation of the peak
= MS × AT/AS × V/250 area of methotrexate is not more than 1.0z.

MS: Amount (mg) of Methotrexate RS taken, calculated Assay Weigh accurately the mass of not less than 20
on the anhydrous basis Methotrexate Tablets, and powder. Weigh accurately a
portion of the powder, equivalent to about 10 mg of
Dissolution <6.10> When the test is performed at 50 revo-
methotrexate (C20H22N8O5), add 50 mL of the mobile phase,
lutions per minute according to the Paddle method, using
shake, and add the mobile phase to make exactly 100 mL.
900 mL of water as the dissolution medium, the dissolution
Centrifuge this solution, and use the supernatant liquid as
rate in 45 minutes of Methotrexate Tablets is not less than
the sample solution. Separately, weigh accurately about 25
85z.
mg of Methotrexate RS (separately determine the water
Start the test with 1 tablet of Methotrexate Tablets, with-
<2.48> in the same manner as Methotrexate), dissolve in the
draw not less than 20 mL of the medium at the specified
mobile phase to make exactly 250 mL, and use this solution
minute after starting the test, and filter through a mem-
as the standard solution. Perform the test with exactly 20
brane filter with a pore size not exceeding 0.45 mm. Discard
mL each of the sample solution and standard solution as di-
the first 10 mL or more of the filtrate, pipet V mL of the
rected under Liquid Chromatography <2.01> according to
subsequent filtrate, add water to make exactly V? mL so
the following conditions, and determine the peak areas, AT
that each mL contains about 2.8 mg of methotrexate
and AS, of methotrexate in each solution.
(C20H22N8O5), and use this solution as the sample solution.
Separately, weigh accurately about 25 mg of Methotrexate Amount (mg) of methotrexate (C20H22N8O5)
RS (separately determine the water <2.48> in the same man- = MS × AT/AS × 2/5
ner as Methotrexate), and dissolve in the mobile phase to
MS: Amount (mg) of Methotrexate RS taken, calculated
make exactly 100 mL. Pipet 1 mL of this solution, add
on the anhydrous basis
water to make exactly 100 mL, and use this solution as the
standard solution. Perform the test with exactly 50 mL each Operating conditions—
of the sample solution and standard solution as directed Detector: An ultraviolet absorption photometer (wave-
under Liquid Chromatography <2.01>, and determine the length: 302 nm).
peak areas, AT and AS, of methotrexate in each solution. Column: A stainless steel column 4.6 mm in inside diame-
ter and 25 cm in length, packed with octadecylsilanized

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2723

silica gel for liquid chromatography (5 mm in particle diame- the following operating conditions: not less than 75z and
ter). not more than 140z of the labeled viscosity.
Column temperature: A constant temperature of about Operating conditions—
259C. Apparatus: Brookfield type viscometer LV model or an
Mobile phase: A mixture of disodium hydrogen phos- equivalent apparatus.
phate-citric acid buffer solution (pH 6.0) and acetonitrile Rotor No., rotation frequency, and calculation multi-
(89:11). plier: According to the following table, depending on the
Flow rate: Adjust so that the retention time of metho- labeled viscosity.
trexate is about 8 minutes.
System suitability— Rotation
Labeled viscosity Rotor Calculation
System performance: Dissolve 10 mg each of metho- frequency
(mPa・s) No. multiplier
trexate and folic acid in 100 mL of the mobile phase. When /min
the procedure is run with 20 mL of this solution under the Not less than 600 and less than 1400 3 60 20
above operating conditions, folic acid and methotrexate are 〃 1400 〃 3500 3 12 100
eluted in this order with the resolution between these peaks 〃 3500 〃 9500 4 60 100
〃 9500 〃 99,500 4 6 1000
being not less than 8.
〃 99,500 4 3 2000
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 Procedure of apparatus: Read the value after 2 minutes
area of methotrexate is not more than 1.0z. of rotation, and stop the rotation for at least 2 minutes.
Repeat this procedure more two times, and average the
Containers and storage Containers—Well-closed contain- three observed values.
ers.
Storage—Light-resistant. Assay
(ii) Procedure—Weigh accurately about 65 mg of
Methylcellulose, transfer to the reaction vial, add 0.06 to
0.10 g of adipic acid, 2.0 mL of the internal standard solu-
Methylcellulose tion and 2.0 mL of hydroiodic acid, immediately stopper
メチルセルロース the vial tightly, and weigh accurately. Using a magnetic stir-
rer equipped in the heating module, or using a shaker, stir
Change the Viscosity and Assay (ii) as follows: for 60 minutes while heating so that the temperature of the
vial content is 130 ± 29C. In the case when a magnetic stir-
Viscosity <2.53> rer or shaker is not available, heat for 30 minutes with
(i) Method I: Apply to Methylcellulose having a labeled repeated shaking at 5-minute intervals by hand, and con-
viscosity of less than 600 mPa・s. Put an exact amount of tinue heating for an additional 30 minutes. Allow the vial to
Methylcellulose, equivalent to 4.000 g on the dried basis, in cool, and again weigh accurately. If the mass loss is less
a tared, wide-mouth bottle, add hot water (between 909C than 0.50z and there is no evidence of a leak of the con-
and 999C) to make 200 g, stopper the bottle, stir by tent, use the upper layer of the mixture as the sample solu-
mechanical means at 350 to 450 revolutions per minute for tion. Separately, put 0.06 to 0.10 g of adipic acid, 2.0 mL of
10 to 20 minutes to get a homogeneous dispersion. If neces- the internal standard solution and 2.0 mL of hydroiodic
sary, take off the sample attached on the walls of the bottle, acid in a reaction vial, stopper the vial immediately, and
put them in the dispersed solution, and dissolve by con- weigh accurately. Add 45 mL of iodomethane for assay
tinuing the stirring in a water bath at not exceeding 59C for through the septum using a micro-syringe, weigh accurately,
20 to 40 minutes. Add cold water, if necessary, to make shake, and use the upper layer of the mixture as the stand-
200 g, and use this solution as the sample solution. Centri- ard solution. Perform the test with 1 to 2 mL each of the
fuge the solution if necessary to expel any entrapped air sample solution and standard solution as directed under Gas
bubbles. Perform the test with the sample solution at 20 ± Chromatography <2.02> according to the following condi-
0.19C as directed in Method I under Viscosity Determina- tions, and calculate the ratios, QT and QS, of the peak area
tion: not less than 80z and not more than 120z of the of iodomethane to that of the internal standard.
labeled viscosity.
(ii) Method II: Apply to Methylcellulose having a Content (z) of methoxy group (CH3O)
labeled viscosity of not less than 600 mPa・s. Put an exact = MS/M × QT/QS × 21.86
amount of Methylcellulose, equivalent to 10.00 g on the MS: Amount (mg) of iodomethane for assay taken
dried basis, in a tared, wide-mouth bottle, add hot water M: Amount (mg) of Methylcellulose taken, calculated on
(between 909 C and 999C) to make 500 g, and prepare the the dried basis
sample solution in the same manner as directed in Method I.
Perform the test with the sample solution at 20 ± 0.19C as Internal standard solution—A solution of n-octane in o-xy-
directed in Method II under Viscosity Determination, using lene (3 in 100).
a single cylinder-type rotational viscometer, according to

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2724 Official Monographs Supplement I, JP XVII

Operating conditions— from the sample solution is not larger than 3/20 times the
Detector: A thermal conductivity detector or hydrogen peak area of montelukast from the standard solution, and
flame-ionization detector. the area of the peak other than montelukast and the peaks
Column: A glass column 3 – 4 mm in inside diameter and mentioned above from the sample solution is not larger
1.8 – 3 m in length, packed with siliceous earth for gas chro- than 1/10 times the peak area of montelukast from the
matography, 125 to 150 mm in diameter, coated with methyl standard solution. Furthermore, the total area of the peaks
silicone polymer for gas chromatography at the ratio of 10 – other than montelukast from the sample solution is not
20z. larger than 1.2 times the peak area of montelukast from the
Column temperature: A constant temperature of about standard solution. However, the peaks of the related sub-
1009C. stances derived from Montelukast Sodium [having the rela-
Carrier gas: Helium for thermal conductivity detector, or tive retention time of about 1.04 (related substance E),
Helium or Nitrogen for hydrogen flame-ionization detector. about 1.16 (related substance C), about 1.18 (related sub-
Flow rate: Adjust so that the retention time of the inter- stance D), about 1.24 and about 1.55 (related substance F)
nal standard is about 10 minutes. to montelukast] are excluded. For the area of the peak, hav-
System suitability— ing the relative retention time of about 0.71 to montelukast,
System performance: When the procedure is run with multiply the relative response factor 0.6.
1 – 2 mL of the standard solution under the above operating Operating conditions—
conditions, iodomethane and the internal standard are Detector, column, column temperature, mobile phase and
eluted in this order, with complete separation of these flow rate: Proceed as directed in the operating conditions in
peaks. the Assay.
Time span of measurement: About 1.5 times as long as
the retention time of montelukast, beginning after the sol-
Add the following: vent peak.
System suitability—
Montelukast Sodium Granules System performance: Proceed as directed in the system
suitability in the Assay.
モンテルカストナトリウム顆粒 Test for required detectability: Pipet 10 mL of the stand-
ard solution, and add a mixture of methanol and water (3:1)
Montelukast Sodium Granules contain not less than to make exactly 100 mL. When the procedure is run with 20
95.0z and not more than 105.0z of the labeled mL of this solution under the above operating conditions,
amount of montelukast (C35H36ClNO3S: 586.18). the SN ratio of the peak of montelukast is not less than 10.
System repeatability: When the test is repeated 5 times
Method of preparation Prepare as directed under Gran-
with 20 mL of the standard solution under the above operat-
ules, with Montelukast Sodium.
ing conditions, the relative standard deviation of the peak
Identification To an amount of Montelukast Sodium area of montelukast is not more than 2.0z.
Granules, equivalent to 5 mg of montelukast
Uniformity of dosage unit <6.02> Perform the test accord-
(C35H36ClNO3S), add 500 mL of a mixture of methanol and
ing to the following method: Montelukast Sodium Granules
water (3:1), shake, and centrifuge. Determine the absorp-
in single-dose packages meet the requirement of the Content
tion spectrum of the supernatant liquid as directed under
uniformity test.
Ultraviolet-visible Spectrophotometry <2.24>: it exhibits
Conduct this procedure using light-resistant vessels. To
maxima between 281 nm and 285 nm, between 325 nm and
the total content of 1 package of Montelukast Sodium
329 nm, between 343 nm and 347 nm, and between 357 nm
Granules add 130 mL of methanol, disperse the fine parti-
and 361 nm.
cles by sonicating, and add methanol to make exactly V mL
Purity Related substances—Use the sample solution ob- so that each mL contains about 20 mg of montelukast
tained in the Assay as the sample solution. Pipet 1 mL of (C35H36ClNO3S). Centrifuge, and use the supernatant liquid
the sample solution, add a mixture of methanol and water as the sample solution. Separately, weigh accurately about
(3:1) to make exactly 100 mL, and use this solution as the 33 mg of Montelukast Dicyclohexylamine RS, and dissolve
standard solution. Perform the test with exactly 20 mL each in methanol to make exactly 100 mL. Pipet 8 mL of this so-
of the sample solution and standard solution as directed lution, add methanol to make exactly 100 mL, and use this
under Liquid Chromatography <2.01> according to the fol- solution as the standard solution. Perform the test with ex-
lowing conditions, and determine each peak area by the au- actly 5 mL each of the sample solution and standard solution
tomatic integration method: the total area of the two peaks as directed under Liquid Chromatography <2.01> according
of related substance A, having the relative retention time of to the following conditions, and determine the peak areas,
about 0.45 to montelukast, obtained from the sample solu- AT and AS, of montelukast in each solution.
tion is not larger than the peak area of montelukast from
Amount (mg) of montelukast (C35H36ClNO3S)
the standard solution, and the peak area of related sub-
= MS × AT/AS × V/1250 × 0.764
stance B, having the relative retention time of about 0.92,

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2725

MS: Amount (mg) of Montelukast Dicyclohexylamine RS Operating conditions—


taken Proceed as directed in the operating conditions in the
Uniformity of dosage units.
Operating conditions—
System suitability—
Detector: An ultraviolet absorption photometer (wave-
System performance: When the procedure is run with 25
length: 389 nm).
mL of the standard solution under the above operating con-
Column: A stainless steel column 3.0 mm in inside diame-
ditions, the number of theoretical plates and the symmetry
ter and 10 cm in length, packed with phenylated silica gel
factor of the peak of montelukast are not less than 2000 and
for liquid chromatography (5 mm in particle diameter).
not more than 1.5, respectively.
Column temperature: A constant temperature of about
System repeatability: When the test is repeated 6 times
509C.
with 25 mL of the standard solution under the above operat-
Mobile phase: A solution of trifluoroacetic acid in a mix-
ing conditions, the relative standard deviation of the peak
ture of water and acetonitrile (1:1) (1 in 500).
area of montelukast is not more than 1.0z.
Flow rate: Adjust so that the retention time of mon-
telukast is about 2 minutes. Assay Conduct this procedure using light-resistant vessels.
System suitability— Weigh accurately an amount of Montelukast Sodium Gran-
System performance: When the procedure is run with 5 ules, equivalent to about 48 mg of montelukast
mL of the standard solution under the above operating con- (C35H36ClNO3S), and add exactly 200 mL of a mixture of
ditions, the number of theoretical plates and the symmetry methanol and water (3:1). Disperse the fine particles by
factor of the peak of montelukast are not less than 1500 and sonicating, centrifuge, and use the supernatant liquid as the
not more than 1.5, respectively. sample solution. Separately, weigh accurately about 33 mg
System repeatability: When the test is repeated 5 times of Montelukast Dicyclohexylamine RS, and dissolve in a
with 5 mL of the standard solution under the above operat- mixture of methanol and water (3:1) to make exactly 100
ing conditions, the relative standard deviation of the peak mL, and use this solution as the standard solution. Perform
area of montelukast is not more than 1.0z. the test with exactly 20 mL each of the sample solution and
standard solution as directed under Liquid Chromatogra-
Dissolution <6.10> When the test is performed at 50 revo-
phy <2.01> according to the following conditions, and deter-
lutions per minute according to the Paddle method, using
mine the peak areas, AT and AS, of montelukast in each so-
900 mL of a solution of sodium lauryl sulfate (1 in 200) as
lution.
the dissolution medium, the dissolution rate in 15 minutes
of Montelukast Sodium Granules is not less than 85z. Amount (mg) of montelukast (C35H36ClNO3S)
Conduct this procedure using light-resistant vessels. Start = MS × AT/AS × 2 × 0.764
the test with an accurately weighed amount of Montelukast
MS: Amount (mg) of Montelukast Dicyclohexylamine RS
Sodium Granules, equivalent to about 4 mg of montelukast
taken
(C35H36ClNO3S), withdraw not less than 15 mL of the me-
dium at the specified minute after starting the test, and filter Operating conditions—
through a membrane filter with a pore size not exceeding Detector: An ultraviolet absorption photometer (wave-
0.45 mm. Discard the first 10 mL or more of the filtrate, and length: 255 nm).
use the subsequent filtrate as the sample solution. Sepa- Column: A stainless steel column 4.6 mm in inside diame-
rately, weigh accurately about 27 mg of Montelukast Dicy- ter and 10 cm in length, packed with phenylhexylsilanized
clohexylamine RS, and dissolve in methanol to make exactly silica gel for liquid chromatography (3 mm in particle diame-
100 mL. Pipet 2 mL of this solution, add the dissolution ter).
medium to make exactly 100 mL, and use this solution as Column temperature: A constant temperature of about
the standard solution. Perform the test with exactly 25 mL 509C.
each of the sample solution and standard solution as di- Mobile phase A: A solution of trifluoroacetic acid (1 in
rected under Liquid Chromatography <2.01> according to 500).
the following conditions, and determine the peak areas, AT Mobile phase B: A mixture of methanol and acetonitrile
and AS, of montelukast in each solution. (3:2).
Flowing of mobile phase: Control the gradient by mixing
Dissolution rate (z) with respect to the labeled amount
the mobile phases A and B as directed in the following
of montelukast (C35H36ClNO3S)
table.
= MS/MT × AT/AS × 1/C × 18 × 0.764

MS: Amount (mg) of Montelukast Dicyclohexylamine RS


taken
MT: Amount (g) of Montelukast Sodium Granules taken
C: Labeled amount (mg) of montelukast (C35H36ClNO3S)
in 1 g

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2726 Official Monographs Supplement I, JP XVII

Time after injection Mobile phase A Mobile phase B


of sample (min) (volz) (volz)
Oxytetracycline Hydrochloride
0–5 48 → 45 52 → 55 オキシテトラサイクリン塩酸塩
5 – 12 45 55
12 – 22 45 → 25 55 → 75 Change the Description and Identification as
22 – 23 25 75 follows:
Description Oxytetracycline Hydrochloride occurs as yel-
Flow rate: 1.5 mL per minute (the retention time of mon- low, crystals or crystalline powder.
telukast is about 14 minutes). It is freely soluble in water, and slightly soluble in ethanol
System suitability— (99.5).
System performance: Take 10 mL of the standard solu- It shows crystal polymorphism.
tion in a transparent vessel, add 4 mL of hydrogen peroxide
Identification (1) Determine the absorption spectrum of
(30), and allow to stand under 4000 lx white light for 10
a solution of Oxytetracycline Hydrochloride in 0.1 mol/L
minutes. When the procedure is run with 20 mL of this solu-
hydrochloric acid TS (1 in 50,000) as directed under Ultravi-
tion under the above operating conditions, the resolution
olet-visible Spectrophotometry <2.24>, and compare the
between the peak of related substance B, having the relative
spectrum with the Reference Spectrum or the spectrum of a
retention time of about 0.92 to montelukast, and the peak
solution of Oxytetracycline Hydrochloride RS prepared in
of montelukast is not less than 1.5. And proceed with 20 mL
the same manner as the sample solution: both spectra ex-
of the standard solution under the above operating condi-
hibit similar intensities of absorption at the same wave-
tions, the number of the theoretical plates and the symmetry
lengths.
factor of the peak of montelukast are not less than 5000 and
(2) Determine the infrared absorption spectrum of
not more than 2.5, respectively.
Oxytetracycline Hydrochloride, previously dried, as di-
System repeatability: When the test is repeated 5 times
rected in the potassium chloride disk method under Infrared
with 20 mL of the standard solution under the above operat-
Spectrophotometry <2.25>, and compare the spectrum with
ing conditions, the relative standard deviation of the peak
the Reference Spectrum or the spectrum of Oxytetracycline
area of montelukast is not more than 1.0z.
Hydrochloride RS: both spectra exhibit similar intensities of
Containers and storage Containers—Tight containers. absorption at the same wave numbers. If any difference ap-
Storage—Light-resistant. pears between the spectra, dissolve the sample and the Ref-
erence Standard separately in methanol, evaporate the sol-
Others
vent, and perform the test with the residues.
Related substances A, B, C, D, E and F: Refer to them
(3) Dissolve 20 mg of Oxytetracycline Hydrochloride in
described in Montelukast Sodium.
3 mL of water, and add 1 drop of silver nitrate TS: a white
turbidity is produced.

Noradrenaline
Add the following:
ノルアドレナリン

Change the Purity (3) as follows: Pazufloxacin Mesilate


Purity パズフロキサシンメシル酸塩
(3) Adrenaline—Dissolve 10.0 mg of Noradrenaline in
2.0 mL of diluted acetic acid (100) (1 in 2). Pipet 1 mL of
this solution, add water to make 10 mL, then mix with 0.3
mL of a solution of sodium nitrite (1 in 100), and observe
after 1 minute: the solution has no more color than the fol-
lowing control solution.
Control solution: Dissolve 2.0 mg of Adrenaline Bitar- C16H15FN2O4.CH4O3S: 414.41
trate RS for Purity and 90 mg of Noradrenaline Bitartrate (3S )-10-(1-Aminocyclopropyl)-9-fluoro-3-methyl-7-oxo-2,3-dihydro-
RS in water to make exactly 10 mL. Measure exactly 1 mL 7H-pyrido[1,2,3-de][1,4]benzoxazine-6-carboxylic acid
of this solution, add 1.0 mL of diluted acetic acid (100) (1 in monomethanesulfonate
2) and water to make 10 mL, and proceed in the same man- [163680-77-1]
ner.
Pazufloxacin Mesilate, when dried, contains not
less than 98.0z and not more than 102.0z of
pazufloxacin mesilate (C16H15FN2O4.CH4O3S).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2727

Description Pazufloxacin Mesilate occurs as a white to (1 in 1000) and acetonitrile (39:11).


light yellow crystalline powder. Flow rate: Adjust so that the retention time of pazufloxa-
It is freely soluble in water, and slightly soluble in ethanol cin is about 8 minutes.
(99.5). Time span of measurement: About 6 times as long as the
It dissolves in sodium hydroxide TS. retention time of pazufloxacin, beginning after the solvent
The pH of a solution prepared by dissolving 0.4 g of peak.
Pazufloxacin Mesilate in 10 mL of water is between 3.0 and System suitability—
4.0. Test for required detectability: To 1 mL of the sample so-
Melting point: about 2589 C (with decomposition). lution add the mobile phase to make 100 mL, and use this
It shows crystal polymorphism. solution as the solution for system suitability test. Pipet 1
mL of the solution for system suitability test, and add the
Identification (1) Determine the absorption spectrum of
mobile phase to make exactly 10 mL. Confirm that the peak
a solution of Pazufloxacin Mesilate in a mixture of metha-
area of pazufloxacin obtained with 20 mL of this solution is
nol and 1 mol/L hydrochloric acid TS (49:1) (1 in 100,000)
equivalent to 7 to 13z of that with 20 mL of the solution for
as directed under Ultraviolet-visible Spectrophotometry
system suitability test.
<2.24>, and compare the spectrum with the Reference Spec-
System performance: When the procedure is run with 20
trum or the spectrum of a solution of Pazufloxacin Mesilate
mL of the solution for system suitability test under the above
RS prepared in the same manner as the sample solution:
operating conditions, the number of theoretical plates and
both spectra exhibit similar intensities of absorption at the
the symmetry factor of the peak of pazufloxacin are not less
same wavelengths.
than 2500 and not more than 2.0, respectively.
(2) Determine the infrared absorption spectrum of
System repeatability: When the test is repeated 6 times
Pazufloxacin Mesilate, previously dried, as directed in the
with 20 mL of the solution for system suitability test under
paste method under Infrared Spectrophotometry <2.25>,
the above operating conditions, the relative standard devia-
and compare the spectrum with the Reference Spectrum or
tion of the peak area of pazufloxacin is not more than
the spectrum of dried Pazufloxacin Mesilate RS: both spec-
2.0z.
tra exhibit similar intensities of absorption at the same wave
numbers. Loss on drying <2.41> Not more than 0.5z (1 g, 1059
C, 3
(3) Pazufloxacin Mesilate responds to Qualitative Tests hours).
<1.09> for mesilate.
Residue on ignition <2.44> Not more than 0.1z (0.2 g,
Optical rotation <2.49> [a]20
D : -61 – -659(after drying, platinum crucible).
0.2 g, sodium hydroxide TS, 20 mL, 100 mm).
Assay Weigh accurately about 26 mg each of Pazufloxacin
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of Mesilate and Pazufloxacin Mesilate RS, both previously
Pazufloxacin Mesilate according to Method 2, and perform dried, dissolve each in water to make exactly 100 mL. Pipet
the test. Prepare the control solution with 2.0 mL of Stand- 5 mL each of these solutions, add exactly 5 mL of the inter-
ard Lead Solution (not more than 20 ppm). nal standard solution, and use these solutions as the sample
(2) Related substances—Dissolve 26 mg of Pazufloxacin solution and the standard solution, respectively. Perform
Mesilate in 100 mL of the mobile phase, and use this solu- the test with 10 mL each of the sample solution and standard
tion as the sample solution. Perform the test with 20 mL of solution as directed under Liquid Chromatography <2.01>
the sample solution as directed under Liquid Chromatogra- according to the following conditions, and calculate the
phy <2.01> according to the following conditions. Determine ratios, QT and QS, of the peak area of pazufloxacin to that
each peak area by the automatic integration method, and of the internal standard.
calculate the amounts of them by the area percentage
Amount (mg) of pazufloxacin mesilate
method: the amount of the peak other than pazufloxacin is
(C16H15FN2O4.CH4O3S)
not more than 0.10z. For the area of the peak, having the
= MS × QT/QS
relative retention time of about 2.7 to pazufloxacin, multi-
ply the relative response factor, 1.6. MS: Amount (mg) of Pazufloxacin Mesilate RS taken
Operating conditions—
Internal standard solution—A solution of acetanilide in the
Detector: An ultraviolet absorption photometer (wave-
mobile phase (3 in 10,000).
length: 254 nm).
Operating conditions—
Column: A stainless steel column 4.6 mm in inside diame-
Detector: An ultraviolet absorption photometer (wave-
ter and 15 cm in length, packed with octadecylsilanized
length: 254 nm).
silica gel for liquid chromatography (5 mm in particle diame-
Column: A stainless steel column 4 mm in inside diameter
ter).
and 25 cm in length, packed with octadecylsilanized silica
Column temperature: A constant temperature of about
gel for liquid chromatography (10 mm in particle diameter).
409C.
Column temperature: A constant temperature of about
Mobile phase: Dissolve 1.08 g of sodium 1-octanesul-
259C.
fonate in 1000 mL of a mixture of diluted phosphoric acid
Mobile phase: To 200 mL of water add gradually 30 mL

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2728 Official Monographs Supplement I, JP XVII

of methanesulfonic acid while ice-cooling, then add 30 mL ment.


of triethylamine in the same manner, and add water to
Sterility <4.06> Perform the test according to the Mem-
make 300 mL. To 50 mL of this solution add 150 mL of
brane filtration method: it meets the requirement.
acetonitrile, 35 mL of 1 mol/L dipotassium hydrogen phos-
phate TS for buffer solution and water to make 1000 mL. Assay Pipet a volume of Pazufloxacin Mesilate Injection,
Flow rate: Adjust so that the retention time of pazufloxa- equivalent to about 12 mg of pazufloxacin mesilate
cin is about 5 minutes. (C16H15FN2O4.CH4O3S), and add water to make exactly
System suitability— 50 mL. Pipet 5 mL of this solution, add exactly 5 mL of the
System performance: When the procedure is run with 10 internal standard solution, and use this solution as the
mL of the standard solution under the above operating con- sample solution. Separately, weigh accurately about 23 mg
ditions, pazufloxacin and the internal standard are eluted in of Pazufloxacin Mesilate RS, previously dried at 1059C for
this order with the resolution between these peaks being not 3 hours, and add water to make exactly 100 mL. Pipet 5 mL
less than 3. of this solution, add exactly 5 mL of the internal standard
System repeatability: When the test is repeated 6 times solution, and use this solution as the standard solution. Per-
with 10 mL of the standard solution under the above operat- form the test with 10 mL each of the sample solution and
ing conditions, the relative standard deviation of the ratio standard solution as directed under Liquid Chromatogra-
of the peak area of pazufloxacin to that of the internal phy <2.01> according to the following conditions, and calcu-
standard is not more than 1.0z. late the ratios, QT and QS, of the peak area of pazufloxacin
to that of the internal standard.
Containers and storage Containers—Tight containers.
Amount (mg) of pazufloxacin mesilate
(C16H15FN2O4.CH4O3S)
Add the following: = MS × QT/QS × 1/2

MS: Amount (mg) of Pazufloxacin Mesilate RS taken


Pazufloxacin Mesilate Injection
Internal standard solution—A solution of acetanilide in the
パズフロキサシンメシル酸塩注射液 mobile phase (3 in 10,000).
Operating conditions—
Pazufloxacin Mesilate Injection is an aqueous in- Proceed as directed in the operating conditions in the
jection. Assay under Pazufloxacin Mesilate.
It contains not less than 95.0z and not more than System suitability—
105.0z of the labeled amount of pazufloxacin mesi- System performance: When the procedure is run with 10
late (C16H15FN2O4.CH4O3S: 414.41). mL of the standard solution under the above operating con-
ditions, pazufloxacin and acetanilide are eluted in this order
Method of preparation Prepare as directed under Injec-
with the resolution between these peaks being not less than
tions, with Pazufloxacin Mesilate.
3.
Description Pazufloxacin Mesilate Injection is a clear, col- System repeatability: When the test is repeated 6 times
orless liquid. with 10 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the ratio
Identification To a volume of Pazufloxacin Mesilate In-
of the peak area of pazufloxacin to that of the internal
jection, equivalent to 20 mg of Pazufloxacin Mesilate, add a
standard is not more than 1.0z.
mixture of methanol and 1 mol/L hydrochloric acid TS
(49:1) to make 100 mL. To 5 mL of this solution add a mix- Containers and storage Containers—Hermetic containers.
ture of methanol and 1 mol/L hydrochloric acid TS (49:1) Plastic containers for aqueous injections may be used.
to make 100 mL. Determine the absorption spectrum of this Storage—Light-resistant.
solution as directed under Ultraviolet-visible Spectropho-
tometry <2.24>: it exhibits maxima between 237 nm and 241
nm, between 314 nm and 324 nm, between 328 nm and 332 Pentobarbital Calcium
nm, and between 343 nm and 347 nm.
ペントバルビタールカルシウム
pH Being specified separately when the drug is granted ap-
proval based on the Law.
Change the Assay as follows:
Bacterial endotoxins <4.01> Less than 0.30 EU/mg.
Assay Weigh accurately about 20 mg of Pentobarbital
Extractable volume <6.05> It meets the requirement. Calcium, dissolve in 5 mL of water, add exactly 5 mL of the
internal standard solution and water to make 50 mL. To 5
Foreign insoluble matter <6.06> Perform the test accord-
mL of this solution add water to make 20 mL. To 2 mL of
ing to Method 1: it meets the requirement.
this solution add water to make 20 mL, and use this solu-
Insoluble particulate matter <6.07> It meets the require- tion as the sample solution. Separately, weigh accurately

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2729

about 18 mg of Pentobarbital RS, previously dried at 1059C with Pentobarbital Calcium.


for 2 hours, dissolve in 10 mL of acetonitrile for liquid
Identification To a quantity of powdered Pentobarbital
chromatography, add exactly 5 mL of the internal standard
Calcium Tablets, equivalent to 5.6 mg of Pentobarbital Cal-
solution, and add water to make 50 mL. To 5 mL of this so-
cium, add 60 mL of water, shake thoroughly, then add
lution add water to make 20 mL. To 2 mL of this solution
water to make 100 mL, and filter. To 6 mL of the filtrate
add water to make 20 mL, and use this solution as the
add dilute sodium hydroxide TS to make 20 mL, and deter-
standard solution. Perform the test with 20 mL each of the
mine the absorption spectrum of this solution as directed
sample solution and standard solution as directed under
under Ultraviolet-visible Spectrophotometry <2.24>: it ex-
Liquid Chromatography <2.01> according to the following
hibits a maximum between 240 nm and 244 nm.
conditions, and calculate the ratios, QT and QS, of the peak
area of pentobarbital to that of the internal standard. Uniformity of dosage unit <6.02> Perform the Mass varia-
tion test, or the Content uniformity test according to the
Amount (mg) of pentobarbital calcium (C22H34CaN4O6)
following method: it meets the requirement.
= MS × QT/QS × 1.084
To 1 tablet of Pentobarbital Calcium Tablets add exactly
MS: Amount (mg) of Pentobarbital RS taken V/10 mL of the internal standard solution, add 60 mL of
water, shake vigorously until the tablet is completely disin-
Internal standard solution—Dissolve 0.2 g of isopropyl par-
tegrated, then add water to make 100 mL, and filter
ahydroxybenzoate in 20 mL of acetonitrile for liquid chro-
through a membrane filter with a pore size not exceeding
matography, and add water to make 100 mL.
0.45 mm. Discard the first 10 mL of the filtrate, to 2 mL
Operating conditions—
of the subsequent filtrate add water to make V mL so that
Detector: An ultraviolet absorption photometer (wave-
each mL contains about 10 mg of pentobarbital calcium
length: 210 nm).
(C22H34CaN4O6), and use this solution as the sample solu-
Column: A stainless steel column 4.6 mm in inside diame-
tion. Then, proceed as directed in the Assay.
ter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle diame- Amount (mg) of pentobarbital calcium (C22H34CaN4O6)
ter). = MS × QT/QS × V/50 × 1.084
Column temperature: A constant temperature of about
MS: Amount (mg) of Pentobarbital Calcium RS taken
409C.
Mobile phase: Dissolve 1.36 g of potassium dihydrogen Internal standard solution—Dissolve 0.5 g of isopropyl par-
phosphate in 1000 mL of water, and adjust to pH 4.0 with ahydroxybenzoate in 20 mL of acetonitrile for liquid chro-
diluted phosphoric acid (1 in 10). To 650 mL of this solu- matography, and add water to make 200 mL.
tion add 350 mL of acetonitrile for liquid chromatography.
Dissolution <6.10> When the test is performed at 50 revo-
Flow rate: Adjust so that the retention time of pentobar-
lutions per minute according to the Paddle method, using
bital is about 7 minutes.
900 mL of 2nd fluid for dissolution test as the dissolution
System suitability—
medium, the dissolution rate in 15 minutes of Pentobarbital
System performance: When the procedure is run with 20
Calcium Tablets is not less than 80z.
mL of the standard solution under the above operating con-
Start the test with 1 tablet of Pentobarbital Calcium
ditions, pentobarbital and the internal standard are eluted
Tablets, withdraw not less than 20 mL of the medium at the
in this order with the resolution between these peaks being
specified minute after starting the test, and filter through a
not less than 5.
membrane filter with a pore size not exceeding 0.45 mm.
System repeatability: When the test is repeated 6 times
Discard the first 10 mL or more of the filtrate, pipet V mL
with 20 mL of the standard solution under the above operat-
of the subsequent filtrate, add the dissolution medium to
ing conditions, the relative standard deviation of the ratios
make exactly V? mL so that each mL contains about 56 mg
of the peak area of pentobarbital to that of the internal
of pentobarbital calcium (C22H34CaN4O6). Pipet 3 mL of
standard is not more than 1.0z.
this solution, add dilute sodium hydroxide TS to make ex-
actly 10 mL, and use this solution as the sample solution.
Separately, weigh accurately about 26 mg of Pentobarbital
Add the following:
RS, previously dried at 1059 C for 2 hours, dissolve in 2 mL
of ethanol (99.5), and add water to make exactly 100 mL.
Pentobarbital Calcium Tablets Pipet 4 mL of this solution, and add the dissolution medium
to make exactly 20 mL. Pipet 3 mL of this solution, add
ペントバルビタールカルシウム錠
dilute sodium hydroxide TS to make exactly 10 mL, and use
this solution as the standard solution. Determine the absor-
Pentobarbital Calcium Tablets contain not less
bances, AT and AS, of the sample solution and standard so-
than 95.0z and not more than 105.0z of the labeled
lution at 241 nm as directed under Ultraviolet-visible Spec-
amount of pentobarbital calcium (C22H34CaN4O6:
trophotometry <2.24>, using a solution, prepared by adding
490.61).
dilute sodium hydroxide TS to 3 mL of the dissolution me-
Method of preparation Prepare as directed under Tablets, dium to make 10 mL, as the blank.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2730 Official Monographs Supplement I, JP XVII

Dissolution rate (z) with respect to the labeled amount System repeatability: When the test is repeated 6 times
of pentobarbital calcium (C22H34CaN4O6) with 20 mL of the standard solution under the above operat-
= MS × AT/AS × V?/V × 1/C × 180 × 1.084 ing conditions, the relative standard deviation of the ratio
of the peak area of pentobarbital to that of the internal
MS: Amount (mg) of Pentobarbital RS taken
standard is not more than 1.0z.
C: Labeled amount (mg) of pentobarbital calcium
(C22H34CaN4O6) in 1 tablet Containers and storage Containers—Tight containers.

Assay To 20 Pentobarbital Calcium Tablets add 120 mL


of water, shake vigorously for 10 minutes, then add water
to make exactly 200 mL, and filter through a membrane Polymixin B Sulfate
filter with a pore size not exceeding 0.45 mm. Discard the
ポリミキシン B 硫酸塩
first 10 mL of the filtrate, pipet 5 mL of the subsequent fil-
trate, add exactly V/10 mL of the internal standard solu-
Change the Origin/limits of content and
tion, and add water to make V mL so that each mL contains
Description as follows:
about 0.5 mg of pentobarbital calcium (C22H34CaN4O6). To
2 mL of this solution, add water to make 100 mL, and use
Polymixin B Sulfate is the sulfate of a mixture of
this solution as the sample solution. Separately, weigh accu-
peptide substances having antibacterial activity pro-
rately about 23 mg of Pentobarbital RS, previously dried at
duced by the growth of Bacillus polymyxa.
1059C for 2 hours, dissolve in 10 mL of acetonitrile for liq-
It contains not less than 6500 units and not more
uid chromatography, add exactly 5 mL of the internal
than 10,500 units per mg, calculated on the dried
standard solution, and add water to make 50 mL. To 2 mL
basis. The potency of Polymixin B Sulfate is
of this solution add water to make 100 mL, and use this so-
expressed as mass unit of polymixin B
lution as the standard solution. Perform the test with 20 mL
(C55–56H96–98N16O13). One unit of Polymixin B Sulfate
each of the sample solution and standard solution as di-
is equivalent to 0.129 mg of polymixin B sulfate
rected under Liquid Chromatography <2.01> according to
(C55–56H96–98N16O13.1–2H2SO4).
the following conditions, and calculate the ratios, QT and
QS, of the peak area of pentobarbital to that of the internal Description Polymixin B Sulfate occurs as a white pow-
standard. der.
It is freely soluble in water, and practically insoluble in
Amount (mg) of pentobarbital calcium (C22H34CaN4O6)
ethanol (99.5).
in 1 tablet
= MS × QT/QS × V/25 × 1.084

MS: Amount (mg) of Pentobarbital RS taken Polysorbate 80


Internal standard solution—Dissolve 0.5 g of isopropyl par-
ポリソルベート80
ahydroxybenzoate in 20 mL of acetonitrile for liquid chro-
matography, and add water to make 200 mL.
Change the Composition of fatty acids as
Operating conditions—
follows:
Detector: An ultraviolet absorption photometer (wave-
length: 210 nm). Composition of fatty acids Dissolve 0.10 g of Polysorbate
Column: A stainless steel column 4.6 mm in inside diame- 80 in 2 mL of a solution of sodium hydroxide in methanol
ter and 15 cm in length, packed with octadecylsilanized (1 in 50) in a 25-mL conical flask, and boil under a reflux
silica gel for liquid chromatography (5 mm in particle diame- condenser for 30 minutes. Add 2.0 mL of boron trifluoride-
ter). methanol TS through the condenser, and boil for 30
Column temperature: A constant temperature of about minutes. Add 4 mL of heptane through the condenser, and
409C. boil for 5 minutes. After cooling, add 10.0 mL of saturated
Mobile phase: Dissolve 1.36 g of potassium dihydrogen sodium chloride solution, shake for about 15 seconds, and
phosphate in 1000 mL of water, and adjust to pH 4.0 with add a quantity of saturated sodium chloride solution such
diluted phosphoric acid (1 in 10). To 650 mL of this solu- that the upper layer is brought into the neck of the flask.
tion add 350 mL of acetonitrile for liquid chromatography. Collect 2 mL of the upper layer, wash with three 2-mL por-
Flow rate: Adjust so that the retention time of pentobar- tions of water, dry with anhydrous sodium sulfate, and use
bital is about 7 minutes. this solution as the sample solution. Perform the test with 1
System suitability— mL each of the sample solution and fatty acid methyl esters
System performance: When the procedure is run with 20 mixture TS as directed under Gas Chromatography <2.02>
mL of the standard solution under the above operating con- according to the following conditions. Identify each peak
ditions, pentobarbital and the internal standard are eluted obtained with the sample solution using the chromatogram
in this order with the resolution between these peaks being with fatty acid methyl esters mixture TS. Determine each
not less than 5. peak area with the sample solution by the automatic in-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2731

tegration method, and calculate the composition of fatty


acids by the area percentage method: myristic acid is not Povidone
more than 5.0z, palmitic acid is not more than 16.0z,
palmitoleic acid is not more than 8.0z, stearic acid is not ポビドン
more than 6.0z, oleic acid is not less than 58.0z, linoleic
acid is not more than 18.0z and linolenic acid is not more Change the Identification (2) as follows:
than 4.0z.
Identification
Operating conditions—
(2) Determine the infrared absorption spectrum of Povi-
Detector: A hydrogen flame-ionization detector.
done, previously dried at 1059 C for 6 hours, as directed in
Column: A fused silica column 0.32 mm in inside diame-
the potassium bromide disk method under Infrared Spectro-
ter and 30 m in length, coated with polyethylene glycol 20 M
photometry <2.25>, and compare the spectrum with the
for gas chromatography 0.5 mm in thickness.
Reference Spectrum or the spectrum of Povidone RS for
Column temperature: Inject at a constant temperature of
Identification (previously dried at 1059C for 6 hours): both
about 809C, raise the temperature to 2209 C at a rate of
spectra exhibit similar intensities of absorption at the same
109C per minute, and maintain at 2209 C for 40 minutes.
wave numbers.
Injection port temperature: A constant temperature of
about 2509 C.
Detector temperature: A constant temperature of about
Add the following:
2509C.
Carrier gas: Helium.
Flow rate: 50 cm per second. Pyridoxal Phosphate Hydrate
Split ratio: 1: 50.
ピリドキサールリン酸エステル水和物
System suitability—
Test for required detectability: Dissolve 0.50 g of the mix-
ture of fatty acid methyl esters described in the following
table in heptane to make exactly 50 mL, and use this solu-
tion as the solution for system suitability test. Pipet 1 mL of
the solution for system suitability test add heptane to make C8H10NO6P.H2O: 265.16
exactly 10 mL. When the procedure is run with 1 mL of this (4-Formyl-5-hydroxy-6-methylpyridin-
solution under the above operating conditions, the SN ratio 3-yl)methyl dihydrogenphosphate monohydrate
of methyl myristate is not less than 5. [41468-25-1]

Mixture of fatty acid methyl esters Composition (z) Pyridoxal Phosphate Hydrate contains not less
than 98.0z and not more than 101.0z of pyridoxal
Methyl myristate for gas chromatography 5 phosphate (C8H10NO6P: 247.14), calculated on the
Methyl palmitate for gas chromatography 10 anhydrous basis.
Methyl stearate for gas chromatography 15
Methyl arachidate for gas chromatography 20 Description Pyridoxal Phosphate Hydrate occurs as a pale
Methyl oleate for gas chromatography 20 yellow-white to light yellow crystalline powder.
Methyl eicosenoate for gas chromatography 10 It is slightly soluble in water, and practically insoluble in
Methyl behenate 10 ethanol (99.5).
Methyl behenate 10 It dissolves in dilute hydrochloric acid and in sodium hy-
droxide TS.
The pH of a solution prepared by dissolving 0.1 g of
System performance: When the procedure is run with 1
Pyridoxal Phosphate Hydrate in 200 mL of water is be-
mL of the solution for system suitability test under the above
tween 3.0 and 3.5.
operating conditions, ◆methyl stearate and methyl oleate
Pyridoxal Phosphate Hydrate is colored to light red by
are eluted in this order,◆ the resolution between these peaks
light.
is not less than 1.8, and the number of theoretical plates of
the peak of methyl stearate is not less than 30,000. Identification (1) Determine the absorption spectrum of
a solution of Pyridoxal Phosphate Hydrate in phosphate
buffer solution (pH 6.8) (1 in 50,000) as directed under
Ultraviolet-visible Spectrophotometry <2.24>, and compare
the spectrum with the Reference Spectrum or the spectrum
of a solution of Pyridoxal Phosphate RS prepared in the
same manner as the sample solution: both spectra exhibit
similar intensities of absorption at the same wavelengths.
(2) Determine the infrared absorption spectrum of
Pyridoxal Phosphate Hydrate as directed in the potassium

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2732 Official Monographs Supplement I, JP XVII

bromide disk method under Infrared Spectrophotometry Mobile phase: Dissolve 3.63 g of potassium dihydrogen
<2.25>, and compare the spectrum with the Reference Spec- phosphate and 5.68 g of anhydrous disodium hydrogen
trum or the spectrum of Pyridoxal Phosphate RS: both phosphate in water to make 1000 mL.
spectra exhibit similar intensities of absorption at the same Flow rate: Adjust so that the retention time of pyridoxal
wave numbers. phosphate is about 6 minutes.
Time span of measurement: About 2.5 times as long as
Purity (1) Heavy metals <1.07>—Proceed with 4.0 g of
the retention time of pyridoxal phosphate, beginning after
Pyridoxal Phosphate Hydrate according to Method 2, and
the solvent peak.
perform the test. Prepare the control solution with 2.0 mL
System suitability—
of Standard Lead Solution (not more than 5 ppm).
Test for required detectability: Pipet 2 mL of the stand-
(2) Arsenic <1.11>—Dissolve 1.0 g of Pyridoxal Phos-
ard solution, and add the mobile phase to make exactly 20
phate Hydrate in 5 mL of dilute hydrochloric acid. Use this
mL. Confirm that the peak area of pyridoxal phosphate ob-
solution as the test solution, and perform the test (not more
tained with 5 mL of this solution is equivalent to 7 to 13z of
than 2 ppm).
that of pyridoxal phosphate with 5 mL of the standard solu-
(3) Free phosphoric acid—Weigh accurately about 0.1 g
tion.
of Pyridoxal Phosphate Hydrate, dissolve in water to make
System performance: When the procedure is run with 5
exactly 100 mL, and use this solution as the sample solu-
mL of the standard solution under the above operating con-
tion. Pipet 5 mL each of the sample solution and Standard
ditions, the number of theoretical plates and the symmetry
Phosphoric Acid Solution, to each add 2.5 mL of hexaam-
factor of the peak of pyridoxal phosphate are not less than
monium heptamolybdate-sulfuric acid TS and 1 mL of 1-
3000 and not more than 1.5, respectively.
amino-2-naphthol-4-sulfonic acid TS, and shake. Add water
System repeatability: When the test is repeated 6 times
to make exactly 25 mL, and allow to stand at 20 ± 19 C for
with 5 mL of the standard solution under the above operat-
30 minutes. Perform the test with these solutions as directed
ing conditions, the relative standard deviation of the peak
under Ultraviolet-visible Spectrophotometry <2.24>, using a
area of pyridoxal phosphate is not more than 2.0z.
solution prepared with 5 mL of water in the same manner as
the blank. Determine the absorbances, AT and AS, of each Water <2.48> 6.0 – 9.0z (0.1 g, volumetric titration,
solution from the sample solution and Standard Phosphoric direct titration. Use a solution prepared by dissolving 50 g
Acid Solution at 740 nm: the amount of free phosphoric of imidazole for water determination in 100 mL of the dis-
acid is not more than 0.5z. solving solution instead of methanol for water determina-
tion).
Content (z) of free phosphoric acid (H3PO4)
Dissolving solution: A solution containing 80z of 1-
= 1/M × AT/AS × 258.0
methoxy-2-propanol, 18z of ethanol (99.5), 1z of imida-
M: Amount (mg) of Pyridoxal Phosphate Hydrate taken, zole and 1z of imidazole hydrobromide.
calculated on the anhydrous basis
Assay Weigh accurately about 45 mg each of Pyridoxal
(4) Related substances—Dissolve 50 mg of Pyridoxal Phosphate Hydrate and Pyridoxal Phosphate RS (separate-
Phosphate Hydrate in 20 mL of the mobile phase, and use ly determine the water <2.48> in the same manner as
this solution as the sample solution. Pipet 1 mL of the sam- Pyridoxal Phosphate Hydrate), and dissolve each in phos-
ple solution, add the mobile phase to make exactly 100 mL, phate buffer solution (pH 6.8) to make exactly 250 mL.
and use this solution as the standard solution. Perform the Pipet 10 mL each of these solutions, add phosphate buffer
test with exactly 5 mL each of the sample solution and stand- solution (pH 6.8) to make exactly 100 mL, and use these so-
ard solution as directed under Liquid Chromatography lutions as the sample solution and the standard solution.
<2.01> according to the following conditions. Determine Determine the absorbances, AT and AS, of the sample solu-
each peak area by the automatic integration method: the tion and standard solution at 388 nm as directed under Ul-
area of the peak other than pyridoxal phosphate obtained traviolet-visible Spectrophotometry <2.24> using phosphate
from the sample solution is not larger than the peak area of buffer solution (pH 6.8) as the blank.
pyridoxal phosphate from the standard solution, and the
Amount (mg) of pyridoxal phosphate (C8H10NO6P)
total area of the peaks other than pyridoxal phosphate from
= M S × A T / AS
the sample solution is not larger than 2 times the peak area
of pyridoxal phosphate from the standard solution. MS: Amount (mg) of Pyridoxal Phosphate RS taken, cal-
Operating conditions— culated on the anhydrous basis
Detector: An ultraviolet absorption photometer (wave-
Containers and storage Containers—Well-closed contain-
length: 254 nm).
ers.
Column: A stainless steel column 4.6 mm in inside diame-
Storage—Light-resistant.
ter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle diame-
ter).
Column temperature: A constant temperature of about
309C.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2733

Delete the following Monographs: cin Tablets is not less than 80z.
Start the test with 1 tablet of Roxithromycin Tablets,
Rokitamycin withdraw not less than 20 mL of the medium at the speci-
fied minute after starting the test, and filter through a mem-
ロキタマイシン brane filter with a pore size not exceeding 0.45 mm. Discard
the first 10 mL or more of the filtrate, pipet V mL of the
subsequent filtrate, add the dissolution medium to make ex-
Rokitamycin Tablets actly V? mL so that each mL contains about 0.17 mg (po-
tency) of roxithromycin (C41H76N2O15), and use this solu-
ロキタマイシン錠 tion as the sample solution. Separately, weigh accurately
about 33 mg (potency) of Roxithromycin RS, dissolve in the
dissolution medium to make exactly 200 mL, and use this
Add the following: solution as the standard solution. Perform the test with ex-
actly 50 mL each of the sample solution and standard solu-
Roxithromycin Tablets tion as directed under Liquid Chromatography <2.01> ac-
cording to the following conditions, and determine the peak
ロキシスロマイシン錠 areas, AT and AS, of roxithromycin in each solution.

Dissolution rate (z) with respect to the labeled amount


Roxithromycin Tablets contain not less than 95.0z
[mg (potency)] of roxithromycin (C41H76N2O15)
and not more than 110.0z of the labeled potency of
= MS × AT/AS × V?/V × 1/C × 450
roxithromycin (C41H76N2O15: 837.05).
MS: Amount [mg (potency)] of Roxithromycin RS taken
Method of preparation Prepare as directed under Tablets,
C: Labeled amount [mg (potency)] of roxithromycin
with Roxithromycin.
(C41H76N2O15) in 1 tablet
Identification To a quantity of powdered Roxithromycin
Operating conditions—
Tablets, equivalent to 0.3 g (potency) of Roxithromycin,
Detector, column temperature and mobile phase: Proceed
add 10 mL of acetonitrile, shake, and centrifuge. Evaporate
as directed in the operating conditions in the Assay.
the supernatant liquid on a water bath under reduced pres-
Column: A stainless steel column 4.6 mm in inside diame-
sure, dry the residue at 609C under reduced pressure for 1
ter and 15 cm in length, packed with octadecylsilanized
hour, and determine the infrared absorption spectrum of
silica gel for liquid chromatography (5 mm in particle diame-
the residue as directed in the potassium bromide disk
ter).
method under Infrared Spectrophotometry <2.25>: it exhib-
Flow rate: Adjust so that the retention time of rox-
its absorption at the wave numbers of about 3460 cm-1,
ithromycin is about 5 minutes.
2940 cm-1, 1728 cm-1, 1633 cm-1 and 1464 cm-1.
System suitability—
Uniformity of dosage unit <6.02> Perform the Mass varia- System performance: When the procedure is run with 50
tion test, or the Content uniformity test according to the mL of the standard solution under the above operating con-
following method: it meets the requirement. ditions, the number of theoretical plates and the symmetry
To 1 tablet of Roxithromycin Tablets add 7V/10 mL of factor of the peak of roxithromycin are not less than 2300
the mobile phase, sonicate to disintegrate the tablet, shake, and not more than 2.0, respectively.
add exactly V/25 mL of the internal standard solution, and System repeatability: When the test is repeated 6 times
add the mobile phase to make V mL so that each mL with 50 mL of the standard solution under the above operat-
contains about 1.5 mg (potency) of roxithromycin ing conditions, the relative standard deviation of the peak
(C41H76N2O15). Filter this solution through a membrane area of roxithromycin is not more than 1.0z.
filter with a pore size not exceeding 0.45 mm, discard the
Assay Weigh accurately the mass of not less than 20 Rox-
first 5 mL of the filtrate, and use the subsequent filtrate as
ithromycin Tablets, and powder. Weigh accurately a por-
the sample solution. Then, proceed as directed in the Assay.
tion of the powder, equivalent to about 38 mg (potency) of
Amount [mg (potency)] of roxithromycin (C41H76N2O15) roxithromycin (C41H76N2O15), add 20 mL of the mobile
= MS × QT/QS × V/25 phase, shake vigorously, add exactly 1 mL of the internal
standard solution, and then add the mobile phase to make
MS: Amount [mg (potency)] of Roxithromycin RS taken
25 mL. Filter the solution through a membrane filter with a
Internal standard solution—A solution of isopropyl parahy- pore size not exceeding 0.45 mm, discard the first 5 mL of
droxybenzoate in the mobile phase (1 in 800). the filtrate, and use the subsequent filtrate as the sample so-
lution. Separately, weigh accurately about 38 mg (potency)
Dissolution <6.10> When the test is performed at 50 revo-
of Roxithromycin RS, dissolve in the mobile phase, add ex-
lutions per minute according to the Paddle method, using
actly 1 mL of the internal standard solution, then add the
900 mL of 2nd fluid for dissolution test as the dissolution
mobile phase to make 25 mL, and use this solution as the
medium, the dissolution rate in 30 minutes of Roxithromy-
standard solution. Perform the test with 10 mL each of the

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2734 Official Monographs Supplement I, JP XVII

sample solution and standard solution as directed under <2.61>: the solution is clear. Perform the test with the test
Liquid Chromatography <2.01> according to the following solution according to Method 2 under Methods for Color
conditions, and calculate the ratios, QT and QS, of the peak Matching <2.65>: the solution is colorless.
area of roxithromycin to that of the internal standard.

Amount [mg (potency)] of roxithromycin (C41H76N2O15)


Delete the following Monograph:
= M S × QT / QS

MS: Amount [mg (potency)] of Roxithromycin RS taken Serrapeptase


Internal standard solution—A solution of isopropyl parahy-
セラペプターゼ
droxybenzoate in the mobile phase (1 in 800).
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 230 nm). Sodium Lauryl Sulfate
Column: A stainless steel column 4.6 mm in inside diame-
ラウリル硫酸ナトリウム
ter and 25 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle diame-
Change to read:
ter).
Column temperature: A constant temperature of about
C12H25NaO4S: 288.38
259C.
Monosodium monododecyl sulfate
Mobile phase: Dissolve 49.1 g of ammonium dihydrogen
[151-21-3]
phosphate in water to make 1000 mL, and adjust to pH 5.3
with 2 mol/L sodium hydroxide TS. To 690 mL of this solu-
This monograph is harmonized with the European Phar-
tion add 310 mL of acetonitrile.
macopoeia and the U.S. Pharmacopeia.
Flow rate: Adjust so that the retention time of rox-
The corresponding part of the attributes/provisions
ithromycin is about 12 minutes.
which are agreed as non-harmonized within the scope of the
System suitability—
harmonization is marked with symbols (◆ ◆), and the cor-
System performance: When the procedure is run with 10
responding parts which are agreed as the JP local require-
mL of the standard solution under the above operating con-
ment other than the scope of the harmonization are marked
ditions, roxithromycin and the internal standard are eluted
with symbols (◇ ◇).
in this order with the resolution between these peaks being
not less than 10.
Sodium Lauryl Sulfate is a mixture of sodium alkyl
System repeatability: When the test is repeated 6 times
sulfate consisting chiefly of sodium lauryl sulfate.
with 10 mL of the standard solution under the above operat-
It contains not less than 85.0z of sodium alkyl sul-
ing conditions, the relative standard deviation of the ratio
fate [as sodium lauryl sulfate (C12H25NaO4S)].
of the peak area of roxithromycin to that of the internal

standard is not more than 1.0z. Description Sodium Lauryl Sulfate occurs as white to
light yellow, crystals or powder. It has a slightly characteris-
Containers and storage Containers—Tight containers.
tic odor.
It is sparingly soluble in ethanol (95).
A solution of 1 g of Sodium Lauryl Sulfate in 10 mL of
Saccharin Sodium Hydrate water is a clear or an opalescent solution.◆
サッカリンナトリウム水和物 Identification (1) Put 2.5 g of Sodium Lauryl Sulfate in
a platinum or quartz crucible, and add 2 mL of 5 mol/L
Change the Identification (1) and Purity (1): surfuric acid TS. Heat on a water bath, cautiously raise the
temperature gradually with a burner, and ignite. Ignite,
Identification (1) Determine the infrared absorption
preferably in an electric furnace, at 600 ± 259 C and incin-
spectrum of Saccharin Sodium Hydrate, previously dried at
erate the residue completely. After cooling, add a few drops
1059C to constant mass, as directed in the potassium bro-
of 1 mol/L sulfuric acid TS, and heat and ignite as above.
mide disk method under Infrared Spectrophotometry
After cooling, add a few drops of ammonium carbonate
<2.25>, and compare the spectrum with the spectrum of Sac-
TS, evaporate to dryness, and further ignite as above. After
charin Sodium RS for Identification dried in the same man-
cooling, dissolve the residue in 50 mL of water, and stir. To
ner as Saccharin Sodium Hydrate: both spectra exhibit simi-
2 mL of this solution add 4 mL of potassium hexahydrox-
lar intensities of absorption at the same wave numbers.
oantimonate (V) TS. If necessary, rub the inside wall of the
Purity (1) Clarity and color of solution—Dissolve 2.0 g vessel with a glass rod: a white, crystalline precipitate is
of Saccharin Sodium Hydrate in water to make 10 mL, and formed.
use this solution as the test solution. Perform the test with (2) Acidify a solution of Sodium Lauryl Sulfate (1 in 10)
the test solution as directed under Turbidity Measurement with hydrochloric acid, and boil for 20 minutes: no precipi-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2735

tate is formed. To this solution add barium chloride TS: a titration, direct titration).◇
white precipitate is formed. ◇
Total alcohol content Dissolve about 5 g of Sodium
(3) Dissolve 0.1 g of Sodium Lauryl Sulfate in 10 mL of
Lauryl Sulfate, accurately weighed, in 150 mL of water and
water, and shake: the solution foams strongly.
50 mL of hydrochloric acid, and boil under a reflux con-
(4) To 0.1 mL of the solution obtained in (3) add 0.1
denser for 4 hours. Cool, extract with two 75-mL portions
mL of methylene blue TS and 2 mL of dilute sulfuric acid,
of diethyl ether, and evaporate the combined diethyl ether
then add 2 mL of dichloromethane, and shake: a deep blue
extracts on a water bath. Dry the residue at 1059C for 30
color develops in the dichloromethane layer.
minutes, and weigh: the mass of the residue is not less than
Purity (1) Alkalinity—Dissolve 1.0 g of Sodium Lauryl 59.0z.◇
Sulfate in 100 mL of water, add 0.1 mL of phenol red TS,
Assay Weigh accurately about 1.15 g of Sodium Lauryl
and titrate <2.50> with 0.1 mol/L hydrochloric acid VS: the
Sulfate, and dissolve in water to make exactly 1000 mL, by
consumed volume is not more than 0.5 mL.
warming if necessary. Transfer exactly 20 mL of this solu-
(2) Sodium chloride—Dissolve about 5 g of Sodium
tion to a 100-mL stoppered graduated cylinder, add 15 mL
Lauryl Sulfate, accurately weighed, in 50 mL of water, neu-
of dichloromethane and 10 mL of dimidium bromide-patent
tralize the solution with dilute nitric acid, if necessary, add
blue TS, and shake. Titrate <2.50> with 0.004 mol/L ben-
exactly 5 mL of 0.1 mol/L sodium chloride TS, and titrate
zethonium chloride VS until the color of the dichlorometh-
<2.50> with 0.1 mol/L silver nitrate VS until the color of the
ane layer changes from light red to grayish blue, while shak-
solution changes from yellow-green through yellow to
ing vigorously. Allow the layers to separate before each
orange (indicator: 2 drops of fluorescein sodium TS). Per-
titration.
form a blank determination in the same manner, and make
any necessary correction. Each mL of 0.004 mol/L benzethonium chloride VS
= 1.154 mg of C12H25NaO4S
Each mL of 0.1 mol/L silver nitrate VS
= 5.844 mg of NaCl ◆Containers and storage Containers—Well-closed con-
tainers.◆
The combined content of sodium chloride (NaCl: 58.44)
and sodium sulfate (Na2SO4: 142.04) obtained in (3) is not
more than 8.0z.
(3) Sodium sulfate—Dissolve about 1 g of Sodium Spiramycin Acetate
Lauryl Sulfate, accurately weighed, in 10 mL of water, add
スピラマイシン酢酸エステル
100 mL of ethanol (95), and heat at a temperature just
below the boiling point for 2 hours. Filter through a glass
Change the Purity as follows:
filter (G4) while hot, and wash with 100 mL of boiling
ethanol (95). Dissolve the residue on the glass filter by wash- Purity Heavy metals <1.07>—Proceed with 1.0 g of
ing with 150 mL of water, collecting the washings in a Spiramycin Acetate according to Method 2, and perform
beaker. Add 10 mL of dilute hydrochloric acid, heat to boil- the test. Prepare the control solution with 1.0 mL of Stand-
ing, add 25 mL of barium chloride TS, and allow to stand ard Lead Solution (not more than 10 ppm).
overnight. Collect the precipitate, and wash with water until
the last washing produces no opalescence with silver nitrate
TS. Dry the precipitate together with the filter paper, ignite Sulbactam Sodium
to a constant mass between 5009C and 6009C by raising the
temperature gradually, and weigh as barium sulfate (BaSO4: スルバクタムナトリウム
233.39).
Change the Purity and Assay as follows:
Amount (mg) of sodium sulfate (Na2SO4)
= amount (mg) of barium sulfate (BaSO4) × 0.6086 Purity (1) Clarity and color of solution—Dissolve 1.0 g
of Sulbactam Sodium in 20 mL of water: the solution is
(4) Unsulfated alcohols—Dissolve about 10 g of Sodium
clear, and its absorbance at 430 nm determined as directed
Lauryl Sulfate, accurately weighed, in 100 mL of water, add
under Ultraviolet-visible Spectrophotometry <2.24> is not
100 mL of ethanol (95), and transfer to a separator. Extract
more than 0.10.
the solution with three 50-mL portions of pentane. If an
(2) Heavy metals <1.07>—Proceed with 1.0 g of Sulbac-
emulsion forms, sodium chloride may be added to promote
tam Sodium according to Method 2, and perform the test.
separation of the two layers. Combine the pentane extracts,
Prepare the control solution with 2.0 mL of Standard Lead
wash with three 50-mL portions of water, dehydrate with
Solution (not more than 20 ppm).
anhydrous sodium sulfate, and filter. Put the filtrate to a
(3) Sulbactam penicillamine—Weigh accurately about
tared beaker, and evaporate the pentane on a water bath.
0.2 g of Sulbactam Sodium, dissolve in the mobile phase to
Dry the residue at 1059C for 30 minutes, cool, and weigh:
make exactly 50 mL, and use this solution as the sample so-
the mass of the residue is not more than 4.0z.
lution. Separately, weigh accurately about 40 mg of sulbac-
◇Water <2.48> Not more than 5.0z (0.5 g, volumetric tam sodium for sulbactam penicillamine, dissolve in 2 mL

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2736 Official Monographs Supplement I, JP XVII

of water, and add 0.5 mL of sodium hydroxide TS. Allow 359C.


to stand at room temperature for 10 minutes, add 0.5 mL of Mobile phase: To 750 mL of 0.005 mol/L tetrabutylam-
1 mol/L hydrochloric acid TS, and then add the mobile monium hydroxide TS add 250 mL of acetonitrile for liquid
phase to make exactly 100 mL. Pipet 5 mL of this solution, chromatography.
add the mobile phase to make exactly 50 mL, and use this Flow rate: Adjust so that the retention time of sulbactam
solution as the standard solution. Perform the test with ex- is about 6 minutes.
actly 10 mL each of the sample solution and standard solu- System suitability—
tion as directed under Liquid Chromatography <2.01> ac- System performance: When the procedure is run with 10
cording to the following conditions, and determine the peak mL of the standard solution under the above operating con-
areas, AT and AS, of sulbactam penicillamine by the auto- ditions, sulbactam and the internal standard are eluted in
matic integration method: the amount of sulbactam penicil- this order with the resolution between these peaks being not
lamine is not more than 1.0z. less than 1.5.
System repeatability: When the test is repeated 6 times
Amount (z) of sulbactam penicillamine
with 10 mL of the standard solution under the above operat-
= MS/MT × AT/AS × 5
ing conditions, the relative standard deviation of the peak
MS: Amount (mg) of sulbactam sodium for sulbactam area of sulbactam is not more than 1.0z.
penicillamine taken
MT: Amount (mg) of Sulbactam Sodium taken

Operating conditions— Sultamicillin Tosilate Hydrate


Column, column temperature, mobile phase, and flow
スルタミシリントシル酸塩水和物
rate: Proceed as directed in the operating conditions in the
Assay.
Change the Identification and Purity as follows:
Detector: An ultraviolet absorption photometer (wave-
length: 230 nm). Identification (1) Determine the absorption spectrum of
System suitability— a solution of Sultamicillin Tosilate Hydrate in methanol (1
System performance: Proceed as directed in the system in 1000) as directed under Ultraviolet-visible Spectropho-
suitability in the Assay. tometry <2.24>, and compare the spectrum with the Refer-
System repeatability: When the test is repeated 6 times ence Spectrum or the spectrum of a solution of Sultamicillin
with 10 mL of the standard solution under the above operat- Tosilate RS prepared in the same manner as the sample so-
ing conditions, the relative standard deviation of the peak lution: both spectra exhibit similar intensities of absorption
area of sulbactam penicillamine is not more than 2.0z. at the same wavelengths.
(2) Determine the infrared absorption spectrum of Sul-
Assay Weigh accurately amounts of Sulbactam Sodium
tamicillin Tosilate Hydrate as directed in the paste method
and Sulbactam RS, equivalent to about 50 mg (potency),
under Infrared Spectrophotometry <2.25>, and compare the
dissolve each in a suitable amount of the mobile phase, add
spectrum with the Reference Spectrum or the spectrum of
exactly 5 mL of the internal standard solution, then add the
Sultamicillin Tosilate RS: both spectra exhibit similar inten-
mobile phase to make 50 mL, and use these solutions as the
sities of absorption at the same wave numbers.
sample solution and the standard solution, respectively.
Perform the test with 10 mL each of the sample solution and Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of
standard solution as directed under Liquid Chromatogra- Sultamicillin Tosilate Hydrate according to Method 2, and
phy <2.01> according to the following conditions, and calcu- perform the test. Prepare the control solution with 2.0 mL
late the ratios, QT and QS, of the peak area of sulbactam to Standard Lead Solution (not more than 20 ppm).
that of the internal standard. (2) Ampicillin—Perform the procedure rapidly. Weigh
accurately about 20 mg of Sultamicillin Tosilate Hydrate,
Amount [mg (potency)] of sulbactam (C8H11NO5S)
dissolve in the mobile phase to make exactly 50 mL, and use
= MS × QT/QS × 1000
this solution as the sample solution. Separately, weigh accu-
MS: Amount [mg (potency)] of Sulbactam RS taken rately an amount of Ampicillin RS, equivalent to about 20
mg (potency), dissolve in the mobile phase to make exactly
Internal standard solution—A solution of ethyl parahy-
100 mL. Pipet 6 mL of this solution, add the mobile phase
droxybenzoate in the mobile phase (7 in 1000).
to make exactly 100 mL, and use this solution as the stand-
Operating conditions—
ard solution. Perform the test with exactly 25 mL each of the
Detector: An ultraviolet absorption photometer (wave-
sample solution and standard solution as directed under
length: 220 nm).
Liquid Chromatography <2.01> according to the following
Column: A stainless steel column 3.9 mm in inside diame-
conditions, and determine the peak area of ampicillin by the
ter and 30 cm in length, packed with octadecylsilanized
automatic integration method: the peak area from the sam-
silica gel for liquid chromatography (10 mm in particle di-
ple solution is not larger than that from the standard solu-
ameter).
tion.
Column temperature: A constant temperature of about

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2737

Operating conditions— 0.1 g of Sultamicillin Tosilate Hydrate, dissolve in 2 mL of


Detector, column, and column temperature: Proceed as methanol, add water to make exactly 20 mL, and use this
directed in the operating conditions in the Assay. solution as the sample solution. Separately, weigh accu-
Mobile phase: Dissolve 3.12 g of sodium dihydrogen rately about 1 g of ethyl acetate, and mix with water to
phosphate dihydrate in about 750 mL of water, adjust to make exactly 200 mL. Pipet 2 mL of this solution, add 10
pH 3.0 with diluted phosphoric acid (1 in 10), and add mL of methanol, then add water to make exactly 100 mL,
water to make 1000 mL. To 80 mL of acetonitrile for liquid and use this solution as the standard solution. Perform the
chromatography add this solution to make 1000 mL. test with exactly 5 mL each of the sample solution and stand-
Flow rate: Adjust so that the retention time of ampicillin ard solution as directed under Gas Chromatography <2.02>
is about 14 minutes. according to the following conditions, and determine the
System suitability— peak areas, AT and AS, of ethyl acetate in each solution.
System performance: Dissolve 12 mg of Ampicillin RS, 4 Calculate the amount of ethyl acetate by the following
mg of Sulbactam RS and 4 mg of p-toluenesulfonic acid equation: not more than 2.0z.
monohydrate in 1000 mL of the mobile phase. When the
Amount (z) of ethyl acetate = MS/MT × AT/AS × 1/5
procedure is run with 25 mL of this solution under the above
operating conditions, sulbactam, p-toluenesulfonic acid and MS: Amount (mg) of ethyl acetate taken
ampicillin are eluted in this order, and the resolutions be- MT: Amount (mg) of the Sultamicillin Tosilate Hydrate
tween these peaks are not less than 2.0, respectively. taken
System repeatability: When the test is repeated 6 times
Operating conditions—
with 25 mL of the standard solution under the above operat-
Detector: A hydrogen flame-ionization detector.
ing conditions, the relative standard deviation of the peak
Column: A column 3 mm in inside diameter and 1 m in
area of ampicillin is not more than 2.0z.
length, packed with porous styrene-divinylbenzene copoly-
(3) Sulbactam—Perform the procedure rapidly. Weigh
mer for gas chromatography (average pore diameter: 0.0085
accurately about 20 mg of Sultamicillin Tosilate Hydrate,
mm, 300 – 400 m2/g) (150 to 180 mm in particle diameter).
dissolve in the mobile phase to make exactly 50 mL, and use
Column temperature: A constant temperature of about
this solution as the sample solution. Separately, weigh accu-
1559C.
rately an amount of Sulbactam RS, equivalent to about 20
Carrier gas: Nitrogen.
mg (potency), dissolve in the mobile phase to make exactly
Flow rate: Adjust so that the retention time of ethyl ace-
100 mL. Pipet 2 mL of this solution, add the mobile phase
tate is about 6 minutes.
to make exactly 100 mL, and use this solution as the stand-
System suitability—
ard solution. Perform the test with exactly 25 mL each of the
System performance: When the procedure is run with 5
sample solution and standard solution as directed under
mL of the standard solution under the above operating con-
Liquid Chromatography <2.01> according to the following
ditions, the number of theoretical plates and the symmetry
conditions, and determine the peak area of sulbactam by
factor of the peak of ethyl acetate are not less than 500 and
the automatic integration method: the peak area from the
not more than 1.5, respectively.
sample solution is not larger than that from the standard so-
System repeatability: When the test is repeated 6 times
lution.
with 5 mL of the standard solution under the above operat-
Operating conditions—
ing conditions, the relative standard deviation of the peak
Proceed as directed in the operating conditions in the
area of ethyl acetate is not more than 5z.
Purity (2).
System suitability—
Proceed as directed in the system suitability in the Purity
(2). Suxamethonium Chloride Injection
(4) Penicilloic acids—Weigh accurately about 25 mg of
スキサメトニウム塩化物注射液
Sultamicillin Tosilate Hydrate, dissolve in 1 mL of aceto-
nitrile, and add 25 mL of 0.02 mol/L phosphate buffer so-
Change the Expiration date as follows:
lution (pH 3.0) in a 100-mL glass-stopperd flask. Add ex-
actly 5 mL of 0.005 mol/L iodine VS, and allow to stand Shelf life 12 months after preparation.
the stoppered flask for 5 minutes. Titrate <2.50> with 0.005
mol/L sodium thiosulfate VS (indicator: 1.0 mL of starch
TS). Perform a blank determination in the same manner, Teicoplanin
and make any necessary correction. Calculate the amount
of penicilloic acid (C25H34N4O11S2: 630.69) by using the fol- テイコプラニン
lowing equation: it is not more than 3.0z.
Change the Purity (1) and (3) as follows:
Each mL of 0.005 mol/L sodium thiosulfate VS
= 0.2585 mg of C25H34N4O11S2 Purity (1) Clarity and color of solution—Dissolve 0.8 g
of Teicoplanin in 10 mL of water: the solution is clear. Per-
(5) Residual solvent <2.46>—Weigh accurately about

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2738 Official Monographs Supplement I, JP XVII

form the test with this solution according to Method 1 tions in the Assay.
under Methods for Color Matching <2.65>: the color is not Time span of measurement: About 7 times as long as the
more colored than Matching Fluids BY3 and B4. retention time of tetracycline, beginning after the solvent
(3) Heavy metals <1.07>—Place 2.0 g of Teicoplanin in peak.
a quartz or porcelain crucible, cover loosely with a lid, and System suitability—
heat gently to carbonize. After cooling, add 2 mL of nitric System performance: Proceed as directed in the system
acid and 5 drops of sulfuric acid, heat cautiously until white suitability in the Assay.
fumes are no longer evolved, and incinerate by ignition be- Test for required detectability: Pipet 3 mL of the stand-
tween 5009 C and 6009C. If a carbonized substance remains, ard solution, add 0.1 mol/L hydrochloric acid TS to make
add 2 mL of nitric acid and 5 drops of sulfuric acid, heat in exactly 100 mL, and confirm that the peak area of tetracy-
the same manner as above, and incinerate by ignition be- cline obtained with 20 mL of this solution is equivalent to 1
tween 5009C and 6009C. Cool, then proceed according to to 5z of that with 20 mL of the standard solution.
Method 2, and perform the test. The control solution is pre- System repeatability: When the test is repeated 6 times
pared as follows: Evaporate a mixture of 4 mL of nitric with 20 mL of the standard solution under the above operat-
acid, 10 drops of sulfuric acid and 2 mL of hydrochloric ing conditions, the relative standard deviation of the peak
acid on a water bath, further evaporate to dryness on a sand area of tetracycline is not more than 1.0z.
bath, and moisten the residue with 3 drops of hydrochloric
acid. Then proceed in the same manner as the test solution,
and add 1.0 mL of Standard Lead Solution and water to Thrombin
make 50 mL (not more than 5 ppm).
トロンビン
Delete the Bacterial endotoxins and Blood pres-
sure depressant: Change the Expiration date as follows:
Shelf life 36 months after preparation.
Change the Containers and storage as follows:
Containers and storage Containers—Tight containers.
Storage—Light-resistant, and at a temperature between Tobramycin
C and 89C.
29
トブラマイシン

Tetracycline Hydrochloride Change the Purity (1) as follows:


Purity (1) Clarity and color of solution—Dissolve 1.0 g
テトラサイクリン塩酸塩
of Tobramycin in 10 mL of water: the solution is clear, and
its absorbance at 400 nm, determined as directed under
Change the Purity as follows:
Ultraviolet-visible Spectrophotometry <2.24>, is not more
Purity (1) Heavy metals <1.07>—Proceed with 1.0 g of than 0.05.
Tetracycline Hydrochloride according to Method 2, and
perform the test. Prepare the control solution with 1.0 mL
of Standard Lead Solution (not more than 10 ppm). Delete the following Monograph:
(2) Related substances—Dissolve 25 mg of Tetracycline
Hydrochloride in 50 mL of 0.01 mol/L hydrochloric acid Tolazamide
TS, and use this solution as the sample solution. Pipet 3 mL
of the sample solution, add 0.01 mol/L hydrochloric acid トラザミド
TS 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
under Liquid Chromatography <2.01> according to the fol-
lowing conditions, and determine each peak area by the au-
tomatic integration method: the area of the peak other than
tetracycline from the sample solution is not larger than the
peak area of tetracycline from the standard solution, and
the total area of the peaks other than tetracycline from the
sample solution is not larger than 3 times the peak area of
tetracycline from the standard solution.
Operating conditions—
Detector, column, column temperature, mobile phase,
and flow rate: Proceed as directed in the operating condi-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2739

Add the following: (3) Related substances—(i) Dissolve 0.10 g of


Tramadol Hydrochloride in 2 mL of methanol, and use this
Tramadol Hydrochloride solution as the sample solution. Pipet 1 mL of the sample
solution, add methanol to make exactly 500 mL, and use
トラマドール塩酸塩 this solution as the standard solution. Perform the test with
these solutions as directed under Thin-layer Chromatogra-
phy <2.03>. Spot 10 mL each of the sample solution and
standard solution on a plate of silica gel with fluorescent in-
dicator for thin-layer chromatography. Allow the plate to
stand in ammonia vapor for 20 minutes, develop with a
mixture of toluene, isopropanol and ammonia solution (28)
(80:19:1) to a distance of about 15 cm, and air-dry the plate.
Allow the plate to stand in iodine vapor for 1 hour, and exa-
C16H25NO2.HCl: 299.84 mine under ultraviolet light (main wavelength: 254 nm): the
(1RS,2RS )-2-[(Dimethylamino)methyl]-1-(3- spot at the Rf value of about 0.5 obtained from the sample
methoxyphenyl)cyclohexanol monohydrochloride solution is not more intense than the spot from the standard
[36282-47-0] solution.
(ii) Dissolve 0.15 g of Tramadol Hydrochloride in 100
Tramadol Hydrochloride contains not less than mL of the mobile phase, and use this solution as the sample
99.0z and not more than 101.0z of tramadol hydro- solution. Pipet 1 mL of the sample solution, add the mobile
chloride (C16H25NO2.HCl), calculated on the anhy- phase to make exactly 100 mL, and use this solution as the
drous basis. standard solution. Perform the test with exactly 20 mL each
of the sample solution and standard solution as directed
Description Tramadol Hydrochloride occurs as a white
under Liquid Chromatography <2.01> according to the fol-
crystalline powder.
lowing conditions. Determine each peak area by the auto-
It is very soluble in water, and freely soluble in methanol,
matic integration method: the area of the peak having the
in ethanol (95) and in acetic acid (100).
relative retention time of about 0.9 to tramadol obtained
A solution of Tramadol Hydrochloride (1 in 20) shows no
from the sample solution is not larger than 1/5 times the
optical rotation.
peak area of tramadol from the standard solution, the area
Melting point: 180 – 1849C
of the peak other than tramadol and the peak mentioned
Tramadol Hydrochloride shows crystal polymorphism.
above from the sample solution is not larger than 1/10 times
Identification (1) Determine the absorption spectrum of the peak area of tramadol from the standard solution, and
a solution of Tramadol Hydrochloride in ethanol (95) (1 in the total area of the peaks other than tramadol from the
10,000) as directed under Ultraviolet-visible Spectropho- sample solution is not larger than 2/5 times the peak area of
tometry <2.24>, and compare the spectrum with the Refer- tramadol from the standard solution.
ence Spectrum: both spectra exhibit similar intensities of ab- Operating conditions—
sorption at the same wavelengths. Detector: An ultraviolet absorption photometer (wave-
(2) Determine the infrared absorption spectrum of length: 270 nm).
Tramadol Hydrochloride as directed in the potassium chlo- Column: A stainless steel column 4.0 mm in inside diame-
ride disk method under Infrared Spectrophotometry <2.25>, ter and 25 cm in length, packed with octylsilanized silica gel
and compare the spectrum with the Reference Spectrum: for liquid chromatography (5 mm in particle diameter).
both spectra exhibit similar intensities of absorption at the Column temperature: A constant temperature of about
same wave numbers. 259C.
(3) A solution of Tramadol Hydrochloride (1 in 100) re- Mobile phase: A mixture of diluted trifluoroacetic acid (1
sponds to Qualitative Tests <1.09> (2) for chloride. in 500) and acetonitrile (141:59).
Flow rate: Adjust so that the retention time of tramadol
Purity (1) Acidity or alkalinity—Dissolve 1.0 g of
is about 5 minutes.
Tramadol Hydrochloride in water to make 20 mL. To 10
Time span of measurement: About 4 times as long as the
mL of this solution add 0.2 mL of methyl red TS for acidity
retention time of tramadol, beginning after the solvent
or alkalinity test and 0.2 mL of 0.01 mol/L hydrochloric
peak.
acid VS: a red color develops. To this solution add 0.01
System suitability—
mol/L sodium hydroxide VS until the color of the solution
Test for required detectability: Pipet 1 mL of the stand-
changes from red to yellow: the consumed volume is not
ard solution, add the mobile phase to make exactly 20 mL.
more than 0.4 mL.
Confirm that the peak area of tramadol obtained with 20
(2) Heavy metals <1.07>—Proceed with 1.0 g of
mL of this solution is equivalent to 3.5 to 6.5z of that with
Tramadol Hydrochloride according to Method 1, and per-
20 mL of the standard solution.
form the test. Prepare the control solution with 2.0 mL of
System performance: When the procedure is run with 20
Standard Lead Solution (not more than 20 ppm).
mL of the standard solution under the above operating con-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2740 Official Monographs Supplement I, JP XVII

ditions, the number of theoretical plates and the symmetry not more than 10.0z.
factor of the peak of tramadol are not less than 5000 and Operating conditions—
not more than 1.5, respectively. Detector: An ultraviolet absorption photometer (wave-
System repeatability: When the test is repeated 6 times length: 220 nm).
with 20 mL of the standard solution under the above operat- Column: A stainless steel column 4.6 mm in inside diame-
ing conditions, the relative standard deviation of the peak ter and 15 cm in length, packed with octadecylsilanized
area of tramadol is not more than 2.0z. silica gel for liquid chromatography (3 mm in particle diame-
ter).
Water <2.48> Not more than 0.5z (1 g, volumetric titra-
Column temperature: A constant temperature of about
tion, direct titration).
409C.
Residue on ignition <2.44> Not more than 0.1z (1 g). Mobile phase A: Dissolve 6.6 g of diammonium hydrogen
phosphate in 950 mL of water, adjust to pH 3.0 with phos-
Assay Weigh accurately about 0.18 g of Tramadol Hydro-
phoric acid, and add water to make 1000 mL. To 950 mL of
chloride, dissolve in 25 mL of acetic acid(100), add 10 mL
this solution add 50 mL of acetonitrile.
of acetic anhydride, and titrate <2.50> with 0.1 mol/L per-
Mobile phase B: Dissolve 6.6 g of diammonium hydrogen
chloric acid VS (potentiometric titration). Perform a blank
phosphate in 950 mL of water, adjust to pH 3.0 with phos-
determination in the same manner, and make any necessary
phoric acid, and add water to make 1000 mL. To 450 mL of
correction.
this solution add 550 mL of acetonitrile.
Each mL of 0.1 mol/L perchloric acid VS Flowing of mobile phase: Control the gradient by mixing
= 29.98 mg of C16H25NO2.HCl the mobile phases A and B as directed in the following
table.
Containers and storage Containers—Tight containers.

Time after injection Mobile phase A Mobile phase B


Vasopressin Injection of sample (min) (volz) (volz)

0 – 45 90 10
バソプレシン注射液 45 – 90 90 → 30 10 → 70
90 – 100 30 70
Change as follows:
Flow rate: About 0.6 mL per minute.
Time span of measurement: About 3 times as long as the
C46H65N15O12S2: 1084.23 retention time of vasopressin.
[113-79-1] System suitability—
Test for required detectability: To 1 mL of the sample so-
Vasopressin Injection is an aqueous injection. lution add diluted acetic acid (100) (1 in 400) to make 100
It is a synthetic vasopressin consisting of 9 amino mL, and use this solution as the solution for system suitabil-
acid residues. ity test. Pipet 1 mL of the solution for system suitability
It contains not less than 90.0z and not more test, and add diluted acetic acid (100) (1 in 400) to make ex-
than 120.0z of the labeled Units of vasopressin actly 10 mL. Confirm that the peak area of vasopressin ob-
(C46H65N15O12S2). tained with 20 mL of this solution is equivalent to 7 to 13z
of that with 20 mL of the solution for system suitability test.
Method of preparation Prepare as directed under Injec-
System performance: When the procedure is run with 20
tions, with vasopressin.
mL of the solution for system suitability test under the above
Description Vasopressin Injection is a clear and colorless operating conditions, the number of theoretical plates and
liquid. the symmetry factor of the peak of vasopressin are not less
than 17,500 and not more than 1.5, respectively.
pH <2.54> 3.0 – 4.0
System repeatability: When the test is repeated 6 times
Purity Related substances—To a suitable amount of with 20 mL of the solution for system suitability test under
Vasopressin Injection add diluted acetic acid (100) (1 in 400) the above operating conditions, the relative standard devia-
so that each mL contains 20 Units of vasopressin tion of the peak area of vasopressin is not more than 2.0z.
(C46H65N15O12S2), and use this solution as the sample solu-
Bacterial endotoxins <4.01> Less than 15 EU/ Unit.
tion. Perform the test with 20 mL of the sample solution as
directed under Liquid Chromatography <2.01> according to Extractable volume <6.05> It meets the requirement.
the following conditions. Determine each peak area by the
Foreign insoluble matter <6.06> Perform the test accord-
automatic integration method, and calculate the amount of
ing to Method 1: it meets the requirement.
them by the area percentage method: the amount of the
peak eluted before the vasopressin is not more than 2.0z, Insoluble particulate matter <6.07> It meets the require-
and the total amount of the peaks other than vasopressin is ment.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2741

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


brane filtration method: it meets the requirement. Verapamil Hydrochloride
Assay Pipet V mL of Vasopressin Injection, equivalent to
ベラパミル塩酸塩
about 40 Units of vasopressin, add diluted acetic acid (100)
(1 in 400) to make exactly 25 mL, and use this solution as
Change the Origin/limits of content, Description
the sample solution. Separately, weigh accurately about 4
and Purity (4) as follows:
mg of Vasopressin RS, and dissolve in diluted acetic acid
(100) (1 in 400) to make exactly 20 mL. Pipet 4 mL of this
Verapamil Hydrochloride, when dried, contains
solution, and add diluted acetic acid (100) (1 in 400) to
not less than 98.5z and not more than 101.0z of
make exactly 50 mL. Pipet 10 mL of this solution, add
verapamil hydrochloride (C27H38N2O4.HCl).
diluted acetic acid (100) (1 in 400) to make exactly 50 mL,
and use this solution as the standard solution. Perform the Description Verapamil Hydrochloride occurs as a white
test with exactly 20 mL each of the sample solution and crystalline powder.
standard solution as directed under Liquid Chromatogra- It is freely soluble in methanol and in acetic acid (100),
phy <2.01> according to the following conditions, and deter- soluble in ethanol (95) and in acetic anhydride, and spar-
mine the peak areas, AT and AS, of vasopressin in each solu- ingly soluble in water.
tion.
Purity
Amount (Vasopressin Unit) of vasopressin in 1 mL (4) Related substances—Dissolve 0.50 g of Verapamil
= MS × AT/AS × F × 1/V × 2 Hydrochloride in 10 mL of methanol, and use this solution
as the sample solution. Pipet 1 mL of the sample solution,
MS: Amount (mg) of Vasopressin RS taken
add methanol to make exactly 100 mL, and use this solution
F: Content (Unit/mg) of Vasopressin RS
as the standard stock solution. Pipet 5 mL of the standard
Operating conditions— stock solution, add methanol to make exactly 100 mL, and
Detector: An ultraviolet absorption photometer (wave- use this solution as the standard solution (1). Separately,
length: 220 nm). pipet 5 mL of the standard stock solution, add methanol to
Column: A stainless steel column 4.6 mm in inside diame- make exactly 50 mL, and use this solution as the standard
ter and 25 cm in length, packed with octadecylsilanized solution (2). Perform the test with these solutions as di-
silica gel for liquid chromatography (5 mm in particle diame- rected under Thin-layer Chromatography <2.03>. Spot 10
ter). mL each of the sample solution and standard solutions (1)
Column temperature: A constant temperature of about and (2) on two plates of silica gel for thin-layer chromatog-
409C. raphy. With the one plate, develop the plate with a mixture
Mobile phase: Dissolve 6.6 g of diammonium hydrogen of cyclohexane and diethylamine (17:3) to a distance of
phosphate in 950 mL of water, adjust to pH 3.0 with phos- about 15 cm, air-dry the plate, heat at 1109C for 1 hour,
phoric acid, and add water to make 1000 mL. To 870 mL of and cool. Examine immediately after spraying evenly iron
this solution add 130 mL of acetonitrile. (III) chloride-iodine TS on the plate: the spots other than
Flow rate: About 1 mL per minute. the principal spot and the spot on the original point from
System suitability— the sample solution, are not more intense than the spot
System performance: When the procedure is run with 20 from the standard solution (2), and the number of them
mL of the standard solution under the above operating con- which are more intense than the spot from the standard so-
ditions, the number of theoretical plates and the symmetry lution (1) is not more than 3. With another plate, develop
factor of the peak of vasopressin are not less than 9500 and the plate with a mixture of toluene, methanol, acetone and
not more than 1.5, respectively. acetic acid (100) (14:4:1:1), and perform the test in the same
System repeatability: When the test is repeated 6 times manner.
with 20 mL of the standard solution under the above operat-
ing conditions, the relative standard deviation of the peak
area of vasopressin is not more than 2.0z. Verapamil Hydrochloride Tablets
Containers and storage Containers—Hermetic containers.
ベラパミル塩酸塩錠
Storage—In a cold place, and avoid freezing.

Change the Identification, Uniformity of dosage


unit and Assay, and add the Disintegration as
follows:
Identification To 2.5 mL of the sample solution obtained
in the Assay add the mixture of methanol and 0.1 mol/L
hydrochloric acid TS (3:1) to make 100 mL, and determine
the absorption spectrum of this solution as directed under

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2742 Official Monographs Supplement I, JP XVII

Ultraviolet-visible Spectrophotometry <2.24>: it exhibits 409C.


maxima between 228 nm and 232 nm, and between 277 nm Mobile phase: A mixture of methanol, water and perchlo-
and 281 nm. ric acid (550:450:1).
Flow rate: Adjust so that the retention time of verapamil
Uniformity of dosage units <6.02> Perform the Mass vari-
is about 5 minutes.
ation test, or the Content uniformity test according to the
System suitability—
following method: it meets the requirement.
System performance: When the procedure is run with 10
To 1 tablet of Verapamil Hydrochloride Tablets add
mL of the standard solution under the above operating con-
7V/10 mL of a mixture of methanol and 0.1 mol/L hydro-
ditions, the number of theoretical plates and the symmetry
chloric acid TS (3:1), and sonicate until the tablet is disin-
factor of the peak of verapamil are not less than 2000 and
tegrated. After cooling, add a mixture of methanol and 0.1
not more than 2.0, respectively.
mol/L hydrochloric acid TS (3:1) to make exactly V mL so
System repeatability: When the test is repeated 6 times
that each mL contains about 0.8 mg of verapamil hydro-
with 10 mL of the standard solution under the above operat-
chloride (C27H38N2O4.HCl). Centrifuge this solution, and
ing conditions, the relative standard deviation of the peak
use the supernatant liquid as the sample solution. Then,
area of verapamil is not more than 1.0z.
proceed as directed in the Assay.

Amount (mg) of verapamil hydrochloride


(C27H38N2O4.HCl) Vinblastine Sulfate
= MS × AT/AS × V/50
ビンブラスチン硫酸塩
MS: Amount (mg) of verapamil hydrochloride for assay
taken
Change the Structural formula as follows:
Disintegration <6.09> It meets the requirement.

Assay To 25 Verapamil Hydrochloride Tablets, add


7V/10 mL a mixture of methanol and 0.1 mol/L hydrochlo-
ric acid TS (3:1), and sonicate until the tablets are disin-
tegrated. Further, sonicate for about 5 minutes. After cool-
ing, add a mixture of methanol and 0.1 mol/L hydrochloric
acid TS (3:1) to make exactly V mL so that each mL con-
tains about 2 mg of verapamil hydrochloride (C27H38N2O4.
HCl). Centrifuge this solution, pipet 10 mL of the superna- Methyl (3aR,4R,5S,5aR,10bR,13aR)-4-acetoxy-3a-ethyl-
tant liquid, add a mixture of methanol and 0.1 mol/L hy- 9-[(5S,7R,9S )-5-ethyl-5-hydroxy-9-methoxycarbonyl-
drochloric acid TS (3:1) to make exactly 25 mL, and use this 1,4,5,6,7,8,9,10-octahydro-3,7-methano-
solution as the sample solution. Separately, weigh accu- 3-azacycloundecino[5,4-b]indol-9-yl]-5-hydroxy-8-methoxy-
rately about 40 mg of verapamil hydrochloride for assay, 6-methyl-3a,4,5,5a,6,11,12,13a-octahydro-
previously dried at 1059C for 2 hours, dissolve in a mixture 1H-indolizino[8,1-cd]carbazole-5-carboxylate monosulfate
of methanol and 0.1 mol/L hydrochloric acid TS (3:1) to
make exactly 50 mL, and use this solution as the standard
solution. Perform the test with exactly 10 mL each of the Vincristine Sulfate
sample solution and standard solution as directed under
Liquid Chromatography <2.01> according to the following ビンクリスチン硫酸塩
conditions, and determine the peak areas, AT and AS, of
verapamil in each solution. Change the Structural formula as follows:
Amount (mg) of verapamil hydrochloride
(C27H38N2O4.HCl) in 1 tablet
= MS × AT/AS × V/500

MS: Amount (mg) of verapamil hydrochloride for assay


taken

Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 280 nm). Methyl (3aR,4R,5S,5aR,10bR,13aR)-4-acetoxy-3a-ethyl-
Column: A stainless steel column 4.6 mm in inside diame- 9-[(5S,7R,9S )-5-ethyl-5-hydroxy-9-methoxycarbonyl-
ter and 15 cm in length, packed with octadecylsilanized 1,4,5,6,7,8,9,10-octahydro-3,7-methano-3-
silica gel for liquid chromatography (5 mm in particle diame- azacycloundecino[5,4-b]indol-9-yl]-6-formyl-5-hydroxy-
ter). 8-methoxy-3a,4,5,5a,6,11,12,13a-octahydro-
Column temperature: A constant temperature of about 1H-indolizino[8,1-cd]carbazole-5-carboxylate monosulfate

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2743

Add the following: and about 0.5, multiply their relative response factors, 0.7,
0.7 and 1.2, respectively.
Voriconazole for Injection Operating conditions—
Detector, column, column temperature, mobile phase and
注射用ボリコナゾール flow rate: Proceed as directed in the operating conditions in
the Assay.
Voriconazole for Injection is a preparation for in- Time span of measurement: About 1.3 times as long as
jection which is dissolved before use. the retention time of voriconazole.
It contains not less than 93.0z and not more than System suitability—
105.0z of the labeled amount of voriconazole System performance: Suspend 0.1 g of voriconazole in 10
(C16H14F3N5O: 349.31). Correct the amount obtained mL of a solution of sodium hydroxide (1 in 25), add the mo-
in the Assay with T value. bile phase to make 20 mL, and allow to stand for 30
minutes. To 1 mL of this solution add the mobile phase to
Method of preparation Prepare as directed under Injec-
make 100 mL. When the procedure is run with 20 mL of this
tions, with Voriconazole.
solution under the above operating conditions, the resolu-
Description Voriconazole for Injection is white, masses or tion between the peaks, having the relative retention times
powder. about 0.26 and about 0.32 to voriconazole, is not less than
1.5.
Identification To 5 mL of the sample solution obtained in
System repeatability: To 5 mL of the standard solution
the Assay add the mobile phase in the Assay to make 25
add the mobile phase to make 10 mL. When the test is
mL. Determine the absorption spectrum of this solution as
repeated 6 times with 20 mL of this solution under the above
directed under Ultraviolet-visible Spectrophotometry
operating conditions, the relative standard deviation of the
<2.24>: it exhibits a maximum between 254 nm and 258 nm.
peak area of voriconazole is not more than 5.0z.
pH Being specified separately when the drug is granted ap- (2) Optical isomer—Dissolve the content of 1 container
proval based on the Law. of Voriconazole for Injection in the mobile phase so
that each mL contains about 1 mg of voriconazole
Purity (1) Related substances—Dissolve the content of 1
(C16H14F3N5O). To 5 mL of this solution add the mobile
container of Voriconazole for Injection in water so that
phase to make 10 mL, and use this solution as the sample
each mL contains about 10 mg of voriconazole
solution. Pipet 1 mL of the sample solution, and add the
(C16H14F3N5O). To 5 mL of this solution add the mobile
mobile phase to make exactly 100 mL. Pipet 1 mL of this
phase to make 100 mL, and use this solution as the sample
solution, add the mobile phase to make exactly 10 mL, and
solution. Pipet 5 mL of the sample solution, and add the
use this solution as the standard solution. Perform the test
mobile phase to make exactly 50 mL. Pipet 2 mL of this so-
with exactly 20 mL each of the sample solution and standard
lution, add the mobile phase to make exactly 100 mL, and
solution as directed under Liquid Chromatography <2.01>
use this solution as the standard solution. Perform the test
according to the following conditions. Determine each peak
with exactly 20 mL each of the sample solution and standard
area by the automatic integration method: the area of the
solution as directed under Liquid Chromatography <2.01>
peak, having the relative retention time of about 1.3 to
according to the following conditions. Determine each peak
voriconazole, obtained from the sample solution is not
area by the automatic integration method: the area of the
larger than 4 times the peak area of voriconazole from the
peak, having the relative retention time of about 0.26 to
standard solution.
voriconazole, obtained from the sample solution is not
Operating conditions—
larger than 2.5 times the peak area of voriconazole from the
Proceed as directed in the operating conditions in the
standard solution, the area of the peak, having the relative
Purity (3) under Voriconazole.
retention time of about 0.32, from the sample solution is
System suitability—
not larger than the peak area of voriconazole from the
Proceed as directed in the system suitability in the Purity
standard solution, the area of the peak, having the relative
(3) under Voriconazole.
retention time of about 0.5, from the sample solution is not
larger than 2 times the peak area of voriconazole from the Bacterial endotoxins <4.01> Less than 1.5 EU/mg.
standard solution, and the area of peak other than
Uniformity of dosage units <6.02> It meets the require-
voriconazole, the peak having the relative retention time of
ment of the Mass variation test (T: 106.0z).
about 0.61 and the peaks mentioned above from the sample
solution is not larger than the peak area of voriconazole Foreign insoluble matter <6.06> Perform the test accord-
from the standard solution. Furthermore, the total area of ing to Method 2: it meets the requirement.
the peaks other than voriconazole and the peak having the
Insoluble particulate matter <6.07> It meets the require-
relative retention time of about 0.61 from the sample solu-
ment.
tion is not larger than 7 times the peak area of voriconazole
from the standard solution. For the areas of the peaks, hav- Sterility <4.06> Perform the test according to the Mem-
ing the relative retention times of about 0.26, about 0.32 brane filtration method: it meets the requirement.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2744 Official Monographs Supplement I, JP XVII

Assay Take 10 containers of Voriconazole for Injection, Add the following:


dissolve the contents of each in the mobile phase, combine
the solutions, and add the mobile phase to make exactly Zonisamide
1000 mL. Pipet 5 mL of this solution, add the mobile phase
to make exactly 100 mL, and use this solution as the sample ゾニサミド
solution. Separately, weigh accurately about 50 mg of
Voriconazole RS (separately determine the water <2.48> in
the same manner as Voriconazole), and dissolve in the mo-
bile phase to make exactly 50 mL. Pipet 5 mL of this solu-
tion, add the mobile phase to make exactly 50 mL, and use
C8H8N2O3S: 212.23
this solution as the standard solution. Perform the test with
1,2-Benzisoxazol-3-ylmethanesulfonamide
exactly 20 mL each of the sample solution and standard so-
[68291-97-4]
lution as directed under Liquid Chromatography <2.01> ac-
cording to the following conditions, and determine the peak
Zonisamide, when dried, contains not less than
areas, AT and AS, of voriconazole in each solution.
98.0z and not more than 101.0z of zonisamide
Amount (mg) of voriconazole (C16H14F3N5O) in 1 container (C8H8N2O3S).
of Voriconazole for Injection
Description Zonisamide occurs as white to pale yellow,
= M S × AT / AS × 4
crystals or crystalline powder.
MS: Amount (mg) of Voriconazole RS taken, calculated It is freely soluble in acetone and in tetrahydrofuran,
on the anhydrous basis sparingly soluble in methanol, slightly soluble in ethanol
(99.5), and very slightly soluble in water.
Operating conditions—
Detector: An ultraviolet absorption photometer (wave- Identification (1) Determine the absorption spectrum of
length: 256 nm). a solution of Zonisamide in methanol (3 in 200,000) as di-
Column: A stainless steel column 3.9 mm in inside diame- rected under Ultraviolet-visible Spectrophotometry <2.24>,
ter and 15 cm in length, packed with octadecylsilanized and compare the spectrum with the Reference Spectrum or
silica gel for liquid chromatography (4 mm in particle diame- the spectrum of a solution of Zonisamide RS prepared in
ter). the same manner as the sample solution: both spectra ex-
Column temperature: A constant temperature of about hibit similar intensities of absorption at the same wave-
359C. lengths.
Mobile phase: Dissolve 1.9 g of ammonium formate in (2) Determine the infrared absorption spectrum of
1000 mL of water, and adjust to pH 4.0 with formic acid. Zonisamide, previously dried, as directed in the potassium
To 550 mL of this solution add 300 mL of methanol and bromide disk method under Infrared Spectrophotometry
150 mL of acetonitrile. <2.25>, and compare the spectrum with the Reference Spec-
Flow rate: Adjust so that the retention time of voricona- trum or the spectrum of dried Zonisamide RS: both spectra
zole is about 9 minutes. exhibit similar intensities of absorption at the same wave
System suitability— numbers.
System performance: When the procedure is run with 20
Melting point <2.60> 164 – 1689C
mL of the standard solution under the above operating con-
ditions, the number of theoretical plates and the symmetry Purity (1) Chloride <1.03>—Dissolve 1.0 g of Zonisa-
factor of the peak of voriconazole are not less than 5000 mide in 30 mL of acetone, and add 6 mL of dilute nitric
and not more than 1.7, respectively. acid and water to make 50 mL. Perform the test using this
System repeatability: When the test is repeated 6 times solution as the test solution. Prepare the control solution as
with 20 mL of the standard solution under the above operat- follows: to 1.0 mL of 0.01 mol/L hydrochloric acid VS add
ing conditions, the relative standard deviation of the peak 30 mL of acetone, 6 mL of dilute nitric acid and water to
area of voriconazole is not more than 1.0z. make 50 mL (not more than 0.036z).
(2) Sulfate <1.14>—Dissolve 1.0 g of Zonisamide in 30
Containers and storage Containers—Hermetic containers.
mL of acetone, and add 1 mL of dilute hydrochloric acid
and water to make 50 mL. Perform the test using this solu-
tion as the test solution. Prepare the control solution as
Delete the following Monograph:
follows: to 1.0 mL of 0.005 mol/L sulfuric acid VS add 30
mL of acetone, 1 mL of dilute hydrochloric acid and water
Zinostatin Stimalamer to make 50 mL (not more than 0.048z).
(3) Heavy metals <1.07>—Proceed with 2.0 g of Zonisa-
ジノスタチン スチマラマー
mide according to Method 4, and perform the test. Prepare
the control solution with 2.0 mL of Standard Lead Solution
(not more than 10 ppm).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Official Monographs 2745

(4) Related substances—Dissolve 25 mg of Zonisamide Internal standard solution—A solution of 4-aminoa-


in 8 mL of tetrahydrofuran, add water to make 50 mL, and cetophenone in methanol (1 in 1000).
use this solution as the sample solution. Pipet 1 mL of the Operating conditions—
sample solution, add the mobile phase to make exactly 200 Detector: An ultraviolet absorption photometer (wave-
mL, and use this solution as the standard solution. Perform length: 239 nm).
the test with exactly 10 mL each of the sample solution and Column: A stainless steel column 5 mm in inside diameter
standard solution as directed under Liquid Chromatogra- and 15 cm in length, packed with octadecylsilanized silica
phy <2.01> according to the following conditions. Determine gel for liquid chromatography (5 mm in particle diameter).
each peak area by the automatic integration method: the Column temperature: A constant temperature of about
area of the peak other than zonisamide obtained from the 409C.
sample solution is not larger than 1/5 times the peak area of Mobile phase: A mixture of water and tetrahydrofuran
zonisamide from the standard solution. (5:1).
Operating conditions— Flow rate: Adjust so that the retention time of
Detector, column, column temperature, mobile phase, zonisamide is about 11 minutes.
and flow rate: Proceed as directed in the operating condi- System suitability—
tions in the Assay. System performance: When the procedure is run with 10
Time span of measurement: About 2 times as long as the mL of the standard solution under the above operating con-
retention time of zonisamide, beginning after the solvent ditions, the internal standard and zonisamide are eluted in
peak. this order with the resolution between these peaks being not
System suitability— less than 5.
Test for required detectability: Pipet 3 mL of the stand- System repeatability: When the test is repeated 6 times
ard solution, add the mobile phase to make exactly 50 mL. with 10 mL of the standard solution under the above operat-
Confirm that the peak area of zonisamide obtained with 10 ing conditions, the relative standard deviation of the ratio
mL of this solution is equivalent to 4.2 to 7.8z of that with of the peak area of zonisamide to that of the internal stand-
10 mL of the standard solution. ard is not more than 1.0z.
System performance: When the procedure is run with 10
Containers and storage Containers—Tight 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 zonisamide are not less than 8000 and
Add the following:
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- Zonisamide Tablets
ing conditions, the relative standard deviation of the peak
ゾニサミド錠
area of zonisamide is not more than 2.0z.

Loss on drying <2.41> Not more than 0.5z (1 g, 1059C, 3 Zonisamide Tablets contain not less than 95.0z
hours). and not more than 105.0z of the labeled amount of
zonisamide (C8H8N2O3S: 212.23).
Residue on ignition <2.44> Not more than 0.1z (1 g).
Method of preparation Prepare as directed under Tablets,
Assay Weigh accurately about 0.1 g of Zonisamide, previ-
with Zonisamide.
ously dried, and dissolve in methanol to make exactly 100
mL. Pipet 5 mL of this solution, add exactly 5 mL of the in- Identification To 5 mL of the sample solution obtained in
ternal standard solution, add the mobile phase to make 100 the Assay add 5 mL of methanol. Determine the absorption
mL, and use this solution as the sample solution. Sepa- spectrum of this solution as directed under Ultraviolet-
rately, weigh accurately about 50 mg of Zonisamide RS, visible Spectrophotometry <2.24>: it exhibits maxima be-
previously dried, and dissolve in methanol to make exactly tween 237 nm and 241 nm, between 243 nm and 247 nm,
50 mL. Pipet 5 mL of this solution, add exactly 5 mL of the and between 282 nm and 286 nm.
internal standard solution, add the mobile phase to make
Uniformity of dosage unit <6.02> Perform the Mass varia-
100 mL, and use this solution as the standard solution. Per-
tion test, or the Content uniformity test according to the
form the test with 10 mL each of the sample solution and
following method: it meets the requirement.
standard solution as directed under Liquid Chromatogra-
To 1 tablet of Zonisamide Tablets add V/25 mL of water,
phy <2.01> according to the following conditions, and calcu-
disintegrate completely by sonicating, add 7V/10 mL of
late the ratios, QT and QS, of the peak area of zonisamide to
methanol, and shake for 15 minutes. Add methanol to
that of the internal standard.
make exactly V mL so that each mL contains about 0.5 mg
Amount (mg) of zonisamide (C8H8N2O3S) of zonisamide (C8H8N2O3S). Centrifuge this solution, pipet
= M S × QT / QS × 2 3 mL of the supernatant liquid, add methanol to make ex-
actly 50 mL, and use this solution as the sample solution.
MS: Amount (mg) of Zonisamide RS taken
Then, proceed as directed in the Assay.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2746 Official Monographs Supplement I, JP XVII

Amount (mg) of zonisamide (C8H8N2O3S) MS: Amount (mg) of Zonisamide RS taken


= MS × AT/AS × V/75
Containers and storage Containers—Tight containers.
MS: Amount (mg) of Zonisamide RS taken

Dissolution <6.10> When the test is performed at 50 revo-


lutions per minute according to the Paddle method, using
900 mL of water as the dissolution medium, the dissolution
rate in 45 minutes of 25-mg tablet is not less than 75z, and
those in 10 minutes and 45 minutes of 100-mg tablet are not
more than 65z and not less than 70z, respectively.
Start the test with 1 tablet of Zonisamide Tablets. In the
case of 25-mg tablets, withdraw not less than 20 mL of the
medium at the specified minutes after starting the test. In
the case of 100-mg tablets, withdraw exactly 20 mL of the
medium at the specified minutes after starting the test, and
supply exactly 20 mL of water warmed to 37 ± 0.59 C im-
mediately after withdrawing of the medium every time.
Filter these media through a membrane filter with a pore
size not exceeding 0.45 mm. Discard the first 10 mL or more
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 of zonisamide (C8H8N2O3S), and use this solu-
tion as the sample solution. Separately, weigh accurately
about 22 mg of Zonisamide RS, previously dried at 1059C
for 3 hours, and dissolve in water to make exactly 100 mL.
Pipet 5 mL of this solution, add water to make exactly 50
mL, and use this solution as the standard solution. Deter-
mine the absorbances, AT(n) and AS, of the sample solution
and standard solution at 285 nm as directed under Ultravio-
let-visible Spectrophotometry <2.24>.

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


zonisamide (C8H8N2O3S) on the nth medium withdrawing
(n = 1, 2)
AT(n) + ∑
Ø 1  V?
»
n-1 A 1
T(i )
= MS × × × × × 90
 S i=1 S
A A 45  V C
MS: Amount (mg) of Zonisamide RS taken
C: Labeled amount (mg) of zonisamide (C8H8N2O3S) in 1
tablet

Assay Weigh accurately the mass of not less than 20


Zonisamide Tablets, and powder. Weigh accurately a por-
tion of the powder, equivalent to about 75 mg of zoni-
samide (C8H8N2O3S), and moisten with 2 mL of water. Add
70 mL of methanol, shake for 15 minutes, and add metha-
nol to make exactly 100 mL. Centrifuge this solution, pipet
2 mL of the supernatant liquid, add methanol to make ex-
actly 50 mL, and use this solution as the sample solution.
Separately, weigh accurately about 38 mg of Zonisamide
RS, previously dried at 1059 C for 3 hours, dissolve in 1 mL
of water and methanol to make exactly 50 mL. Pipet 2 mL
of this solution, add methanol to make exactly 50 mL, and
use this solution as the standard solution. Determine the ab-
sorbances, AT and AS, of the sample solution and standard
solution at 284 nm as directed under Ultraviolet-visible
Spectrophotometry <2.24>.

Amount (mg) of zonisamide (C8H8N2O3S)


= M S × AT / AS × 2

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Crude Drugs and Related Drugs
under a reflux condenser for 1 hour. After cooling, shake 20
Powdered Alisma Tuber mL of the extract with 5 mL of 1-butanol, centrifuge, re-
move the 1-butanol layer, and use the aqueous layer as the
タクシャ末 standard solution. Perform the test with these solutions as
directed under Thin-layer Chromatography <2.03>. Spot 2
Add the following next to the Description: mL of the sample solution and 5 mL of the standard solution
as bands on the original line on a plate of silica gel for thin-
Identification To 1.0 g of Powdered Alisma Tuber add 10
layer chromatography. Develop the plate with a mixture of
mL of diethyl ether, shake for 10 minutes, centrifuge, and
ethanol (99.5), water and acetic acid (100) (120:80:1) to a
use the supernatant liquid as the sample solution. Use
distance of about 7 cm, and air-dry the plate. Spray evenly
alisma tuber triterpenes TS for identification as the stand-
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
ard solution. Perform the test with these solutions as di-
heat the plate at 1059C for 5 minutes: one of the several
rected under Thin-layer Chromatography <2.03>. Spot 5 mL
spots obtained from the sample solution has the same color
of the sample solution and 1 mL of the standard solution on
tone and Rf value with the dark blue-green spot (Rf value:
a plate of silica gel for thin-layer chromatography. Develop
about 0.3) from the standard solution (Ophiopogon Root).
the plate with a mixture of ethyl acetate, hexane and acetic
(2) Shake 5.0 g of the dry extract (or 15 g of the viscous
acid (100) (10:10:3) to a distance of about 7 cm, and air-dry
extract) with 15 mL of water, add 5 mL of diethyl ether,
the plate. Spray evenly vanillin-sulfuric acid-ethanol TS for
shake, centrifuge, and use the supernatant liquid as the
spraying on the plate, and heat at 1059 C for 5 minutes: one
sample solution. Separately, dissolve 1 mg of cycloartenyl
of the spot among the several spots obtained from the sam-
ferulate for thin-layer chromatography in 1 mL of ethyl ace-
ple solution has the same color tone and Rf value with a
tate, and use this solution as the standard solution. Perform
spot among the three spots obtained from the standard
the test with these solutions as directed under Thin-layer
solution.
Chromatography <2.03>. Spot 30 mL of the sample solution
and 5 mL of the standard solution on a plate of silica gel for
thin-layer chromatography. Develop the plate with a mix-
Artemisia Capillaris Flower ture of hexane, acetone and acetic acid (100) (50:20:1) to a
distance of about 7 cm, and air-dry the plate. Examine
インチンコウ
under ultraviolet light (main wavelength: 365 nm): one of
the several spots obtained from the sample solution has the
Change the alias in Japanese as follows:
same color tone and Rf value with the blue-white fluores-
cent spot from the standard solution. Or examine under ul-
茵 蒿
traviolet light (main wavelength: 365 nm) after spraying
evenly a mixture of sulfuric acid and ethanol (99.5) (1:1)
on the plate, and heating the plate at 1059C for 5 minutes:
Bakumondoto Extract one of the several spots from the sample solution has the
same color tone and Rf value with the yellow fluorescent
麦門冬湯エキス
spot from the standard solution (Brown Rice).
(3) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
Change the Origin/limits of content, Identifica-
extract) with 10 mL of sodium hydroxide TS, add 5 mL of
tion and Assay (2) as follows:
1-butanol, shake, centrifuge, and use the supernatant liquid
as the sample solution. Separately, dissolve 1 mg of Gin-
Bakumondoto Extract contains not less than 1.2
senoside Rb1 RS or ginsenoside Rb1 for thin-layer chroma-
mg of ginesenoside Rb1 (C54H92O23: 1109.29), and not
tography in 1 mL of methanol, and use this solution as the
less than 14 mg and not more than 42 mg of glycyr-
standard solution. Perform the test with these solutions as
rhizic acid (C42H62O16: 822.93), per extract prepared
directed under Thin-layer Chromatography <2.03>. Spot 10
with the amount specified in the Method of prepara-
mL of the sample solution and 2 mL of the standard solution
tion.
on a plate of silica gel for thin-layer chromatography. De-
Identification (1) Shake 2.0 g of the dry extract (or 6.0 g velop the plate with a mixture of ethyl acetate, 1-propanol,
of the viscous extract) with 10 mL of water, add 5 mL of 1- water and acetic acid (100) (7:5:4:1) to a distance of about 7
butanol, shake, centrifuge, remove the 1-butanol layer, and cm, and air-dry the plate. Spray evenly vanillin-sulfuric
use the aqueous layer as the sample solution. Separately, acid-ethanol TS for spraying on the plate, heat the plate at
heat 3.0 g of pulverized ophiopogon root in 50 mL of water 1059C for 5 minutes, and allow to cool: one of the several
2747
The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2748 Crude Drugs and Related Drugs Supplement I, JP XVII

spots obtained from the sample solution has the same color ethanol. When the procedure is run with 10 mL of this solu-
tone and Rf value with the blue-purple spot from the stand- tion under the above operating conditions, the resolution
ard solution (Ginseng). between the peak having the relative retention time of about
(4) Shake 1.0 g of the dry extract (or 3.0 g of the viscous 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
extract) with 10 mL of water, add 10 mL of 1-butanol, not less than 1.5.
shake, centrifuge, and use the supernatant liquid as the System repeatability: When the test is repeated 6 times
sample solution. Separately, dissolve 1 mg of liquiritin for with 10 mL of the standard solution under the above operat-
thin-layer chromatography in 1 mL of methanol, and use ing conditions, the relative standard deviation of the peak
this solution as the standard solution. Perform the test with area of glycyrrhizic acid is not more than 1.5z.
these solutions as directed under Thin-layer Chromatogra-
phy <2.03>. Spot 1 mL each of the sample solution and
standard solution on a plate of silica gel for thin-layer chro- Bofutsushosan Extract
matography. Develop the plate with a mixture of ethyl ace-
tate, methanol and water (20:3:2) to a distance of about 7 防風通聖散エキス
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
on the plate, heat the plate at 1059C for 5 minutes, and exa- Change the Origin/limits of content as follows:
mine under ultraviolet light (main wavelength: 365 nm): one
of the several spots obtained from the sample solution has Bofutsushosan Extract contains not less than 9 mg
the same color tone and Rf value with the yellow-green fluo- and not more than 36 mg of paeoniflorin (C23H28O11:
rescent spot from the standard solution (Glycyrrhiza). 480.46), not less than 4 mg and not more than 12 mg
of total alkaloids [ephedrine (C10H15NO: 165.23) and
Assay
pseudoephedrine (C10H15NO: 165.23)], not less than
(2) Glycyrrhizic acid—Weigh accurately about 0.5 g of
54 mg and not more than 162 mg of baicalin
the dry extract (or an amount of the viscous extract, equiva-
(C21H18O11: 446.36), and not less than 13 mg and not
lent to about 0.5 g of the dried substance), add exactly 50
more than 39 mg of glycyrrhizic acid (C42H62O16:
mL of diluted methanol (1 in 2), shake for 15 minutes,
822.93), per extract prepared with the amount speci-
filter, and use the filtrate as the sample solution. Separately,
fied in the Method of preparation.
weigh accurately about 10 mg of Glycyrrhizic Acid RS
(separately determine the water <2.48> by coulometric titra-
Change the Identification (7), (8), (9) and (15) as
tion, using 10 mg), dissolve in diluted methanol (1 in 2) to
follows:
make exactly 100 mL, and use this solution as the standard
solution. Perform the test with exactly 10 mL each of the Identification
sample solution and standard solution as directed under (7) To 1.0 g of the dry extract (or 3.0 g of the viscous
Liquid Chromatography <2.01> according to the following extract) add 10 mL of 0.1 mol/L hydrochloric acid TS,
conditions, and determine the peak areas, AT and AS, of shake, then add 25 mL of diethyl ether, and shake. Separate
glycyrrhizic acid in each solution. the diethyl ether layer, evaporate the solvent under reduced
pressure, add 1 mL of methanol to the residue, and use the
Amount (mg) of glycyrrhizic acid (C42H62O16)
solution as the sample solution. Separately, dissolve 1 mg of
= MS × AT/AS × 1/2
rosmarinic acid for thin-layer chromatography in 1 mL of
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- methanol, and use this solution as the standard solution.
lated on the anhydrous basis Perform the test with these solutions as directed under
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
Operating conditions—
sample solution and standard solution on a plate of silica
Detector: An ultraviolet absorption photometer (wave-
gel for thin-layer chromatography. Develop the plate with a
length: 254 nm).
mixture of ethyl acetate, water and acetic acid (100) (60:1:1)
Column: A stainless steel column 4.6 mm in inside diame-
to a distance of about 10 cm, and air-dry the plate. Spray
ter and 15 cm in length, packed with octadecylsilanized
evenly iron (III) chloride-methanol TS on the plate: one of
silica gel for liquid chromatography (5 mm in particle diame-
the several spots obtained from the sample solution has the
ter).
same color tone and Rf value with the greenish brown spot
Column temperature: A constant temperature of about
from the standard solution (Schizonepeta Spike; Mentha
409C.
Herb).
Mobile phase: Dissolve 3.85 g of ammonium acetate in
(8) For preparation prescribed Saposhnikovia Root and
720 mL of water, and add 5 mL of acetic acid (100) and 280
Rhizome—To 2.0 g of the dry extract (or 6.0 g of the vis-
mL of acetonitrile.
cous extract) add 10 mL of sodium hydroxide TS, shake,
Flow rate: 1.0 mL per minute (the retention time of
then add 5 mL of 1-butanol, shake, centrifuge, and use the
glycyrrhizic acid is about 15 minutes).
supernatant liquid as the sample solution. Separately, dis-
System suitability—
solve 1 mg of 4?-O-glycosyl-5-O-methylvisamminol for thin-
System performance: Dissolve 5 mg of monoammonium
layer chromatography in 1 mL of methanol, and use this so-
glycyrrhizinate for resolution check in 20 mL of dilute

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2749

lution as the standard solution. Perform the test with these cooling, add 20 mL of water, shake, and filter. To 5 mL of
solutions as directed under Thin-layer Chromatography the filtrate add ammonia TS until a white gelatinous pre-
<2.03>. Spot 10 mL of the sample solution and 5 mL of the cipitate is formed, centrifuge, and remove the supernatant
standard solution on a plate of silica gel for thin-layer chro- liquid. To the residue add 5 mL of water, shake, centrifuge,
matography. Develop the plate with a mixture of ethyl ace- and remove the supernatant liquid. Then, to the residue add
tate, 1-propanol, water and acetic acid (100) (7:5:4:1) to a 5 mL of water, shake, centrifuge, and remove the superna-
distance of about 7 cm, and air-dry the plate. Spray evenly tant liquid. To the obtained residue add 5 drops of alizarin
dilute sulfuric acid on the plate, heat the plate at 1059C for red S TS, and shake occasionally in lukewarm water: the
2 minutes, then examine under ultraviolet light (main wave- residue is red to red-brown in color (Kasseki).
length: 365 nm): one of the several spots obtained from the
sample solution has the same color tone and Rf value with Change the Assay (4) as follows:
the blue-white fluorescent spot from the standard solution
Assay
(Saposhnikovia Root and Rhizome).
(4) Glycyrrhizic acid—Weigh accurately about 0.5 g of
(9) For preparation prescribed Glehnia Root and Rhi-
the dry extract (or an amount of the viscous extract, equiva-
zome—To 0.5 g of the dry extract (or 1.5 g of the viscous
lent to about 0.5 g of the dried substance), add 20 mL of
extract) add 5 mL of ethyl acetate, and heat on a water bath
ethyl acetate and 10 mL of water, and shake for 10 minutes.
under a reflux condenser for 30 minutes. After cooling,
After centrifugation, remove the upper layer, add 20 mL of
filter, and use the filtrate as the sample solution. Separately,
ethyl acetate, proceed in the same manner as described
dissolve 1 mg of scopoletin for thin-layer chromatography
above, and remove the upper layer. To the resultant aque-
in 10 mL of methanol, and use this solution as the standard
ous layer add 10 mL of methanol, shake for 30 minutes,
solution. Perform the test with these solutions as directed
centrifuge, and take the supernatant liquid. To the residue
under Thin-layer Chromatography <2.03>. Spot 20 mL of
add 20 mL of diluted methanol (1 in 2), shake for 5
the sample solution and 2 mL of the standard solution on a
minutes, centrifuge, and take the supernatant liquid. Com-
plate of silica gel for thin-layer chromatography. Develop
bine these supernatant liquids, add diluted methanol (1 in 2)
the plate with a mixture of ethyl acetate and hexane (3:1) to
to make exactly 50 mL, and use this solution as the sample
a distance of about 7 cm, and air-dry the plate. Spray evenly
solution. Separately, weigh accurately about 10 mg of
dilute sulfuric acid on the plate, heat the plate at 1059C for
Glycyrrhizic Acid RS (separately determine the water <2.48>
5 minutes, and examine under ultraviolet light (main wave-
by coulometric titration, using 10 mg), dissolve in diluted
length: 365 nm): one of the several spots obtained from the
methanol (1 in 2) to make exactly 100 mL, and use this solu-
sample solution has the same color tone and Rf value with
tion as the standard solution. Perform the test with exactly
the blue-white fluorescent spot from the standard solution
10 mL each of the sample solution and standard solution as
(Glehnia Root and Rhizome).
directed under Liquid Chromatography <2.01> according to
(15) To 1.0 g of the dry extract (or 3.0 g of the viscous
the following conditions, and determine the peak areas, AT
extract) add 10 mL of water, shake, then add 10 mL of 1-
and AS, of glycyrrhizic acid in each solution.
butanol, shake, centrifuge, and use the supernatant liquid
as the sample solution. Separately, dissolve 1 mg of liquiri- Amount (mg) of glycyrrhizic acid (C42H62O16)
tin for thin-layer chromatography in 1 mL of methanol, and = MS × AT/AS × 1/2
use this solution as the standard solution. Perform the test
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
with these solutions as directed under Thin-layer Chroma-
lated on the anhydrous basis
tography <2.03>. Spot 1 mL each of the sample solution and
standard solution on a plate of silica gel for thin-layer chro- Operating conditions—
matography. Develop the plate with a mixture of ethyl ace- Detector: An ultraviolet absorption photometer (wave-
tate, methanol and water (20:3:2) to a distance of about 7 length: 254 nm).
cm, and air-dry the plate. Spray evenly dilute sulfuric acid Column: A stainless steel column 4.6 mm in inside diame-
on the plate, heat the plate at 1059 C for 5 minutes, and exa- ter and 15 cm in length, packed with octadecylsilanized
mine under ultraviolet light (main wavelength: 365 nm): one silica gel for liquid chromatography (5 mm in particle diame-
of the several spots obtained from the sample solution has ter).
the same color tone and Rf value with the yellow-green fluo- Column temperature: A constant temperature of about
rescent spot from the standard solution (Glycyrrhiza). 409C.
Mobile phase: Dissolve 3.85 g of ammonium acetate in
Add the following next to the Identification (17) 720 mL of water, and add 5 mL of acetic acid (100) and 280
as follows: mL of acetonitrile.
Flow rate: 1.0 mL per minute (the retention time of
Identification
glycyrrhizic acid is about 15 minutes).
(18) Place 2.0 g of the dry extract (or 6.0 g of the vis-
System suitability—
cous extract) in a crucible, and ignite at 5509C for 5 hours
System performance: Dissolve 5 mg of monoammonium
to incinerate. To the residue add 3 mL of diluted sulfuric
glycyrrhizinate for resolution check in 20 mL of dilute
acid (1 in 3), and heat until white fumes are evolved. After
ethanol. When the procedure is run with 10 mL of this solu-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2750 Crude Drugs and Related Drugs Supplement I, JP XVII

tion under the above operating conditions, the resolution rescent spot from the standard solution (Glycyrrhiza).
between the peak having the relative retention time of about
Assay
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
(2) Glycyrrhizic acid—Weigh accurately about 0.5 g of
not less than 1.5.
the dry extract (or an amount of the viscous extract, equiva-
System repeatability: When the test is repeated 6 times
lent to about 0.5 g of the dried substance), add exactly 50
with 10 mL of the standard solution under the above operat-
mL of diluted methanol (1 in 2), shake for 15 minutes,
ing conditions, the relative standard deviation of the peak
filter, and use the filtrate as the sample solution. Separately,
area of glycyrrhizic acid is not more than 1.5z.
weigh accurately about 10 mg of Glycyrrhizic Acid RS
(separately determine the water <2.48> by coulometric titra-
tion, using 10 mg), dissolve in diluted methanol (1 in 2) to
Boiogito Extract make exactly 100 mL, and use this solution as the standard
solution. Perform the test with exactly 10 mL each of the
防已黄耆湯エキス
sample solution and standard solution as directed under
Liquid Chromatography <2.01> according to the following
Change the Origin/limits of content, Identifica-
conditions, and determine the peak areas, AT and AS, of
tion (4), (6) and Assay (2) as follows:
glycyrrhizic acid in each solution.

Boiogito Extract contains not less than 4 mg and Amount (mg) of glycyrrhizic acid (C42H62O16)
not more than 16 mg of shinomenine, and not less = MS × AT/AS × 1/2
than 10 mg and not more than 30 mg of glycyrrhizic
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
acid (C42H62O16: 822.93), per extract prepared with
lated on the anhydrous basis
the amount specified in the Method of preparation.
Operating conditions—
Identification
Detector: An ultraviolet absorption photometer (wave-
(4) For preparation prescribed Atractylodes Lancea
length: 254 nm).
Rhizome—To 2.0 g of the dry extract (or 6.0 g of the vis-
Column: A stainless steel column 4.6 mm in inside diame-
cous extract) add 10 mL of water, shake, then add 25 mL of
ter and 15 cm in length, packed with octadecylsilanized
hexane, and shake. Separate the hexane layer, evaporate the
silica gel for liquid chromatography (5 mm in particle diame-
solvent under reduced pressure, then add 0.5 mL of hexane
ter).
to the residue, and use this solution as the sample solution.
Column temperature: A constant temperature of about
Perform the test with the sample solution as directed under
409C.
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
Mobile phase: Dissolve 3.85 g of ammonium acetate in
ple solution on a plate of silica gel with fluorescent indicator
720 mL of water, and add 5 mL of acetic acid (100) and 280
for thin-layer chromatography. Develop the plate with a
mL of acetonitrile.
mixture of hexane and acetone (7:1) to a distance of about 7
Flow rate: 1.0 mL per minute (the retention time of
cm, and air-dry the plate. Examine under ultraviolet light
glycyrrhizic acid is about 15 minutes).
(main wavelength: 254 nm): a dark purple spot is observed
System suitability—
at an Rf value of about 0.5. The spot shows a greenish
System performance: Dissolve 5 mg of monoammonium
brown color after being sprayed evenly 4-dimethylamino-
glycyrrhizinate for resolution check in 20 mL of dilute
benzaldehyde TS for spraying on the plate, heated at 1059C
ethanol. When the procedure is run with 10 mL of this solu-
for 5 minutes, and allowed to cool (Atractylodes Lancea
tion under the above operating conditions, the resolution
Rhizome).
between the peak having the relative retention time of about
(6) To 1.0 g of the dry extract (or 3.0 g of the viscous
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
extract) add 10 mL of water, shake, then add 10 mL of 1-
not less than 1.5.
butanol, shake, centrifuge, and use the supernatant liquid
System repeatability: When the test is repeated 6 times
as the sample solution. Separately, dissolve 1 mg of liquiri-
with 10 mL of the standard solution under the above operat-
tin for thin-layer chromatography in 1 mL of methanol, and
ing conditions, the relative standard deviation of the peak
use this solution as the standard solution. Perform the test
area of glycyrrhizic acid is not more than 1.5z.
with these solutions as directed under Thin-layer Chroma-
tography <2.03>. Spot 1 mL each of the sample solution and
standard solution on a plate of silica gel for thin-layer chro-
matography. Develop the plate with a mixture of ethyl ace-
tate, methanol and water (20:3:2) to a distance of about 7
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
on the plate, heat the plate at 1059 C for 5 minutes, and exa-
mine under ultraviolet light (main wavelength: 365 nm): one
of the several spots obtained from the sample solution has
the same color tone and Rf value with the yellow-green fluo-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2751

reflux condenser for 1 hour. After cooling, shake 20 mL of


Chotosan Extract the extract with 5 mL of 1-butanol, centrifuge, remove the
1-butanol layer, and use the aqueous layer as the standard
釣藤散エキス solution. Perform the test with these solutions as directed
under Thin-layer Chromatography <2.03>. Spot 2 mL of the
Change the Origin/limits of content, Identifica- sample solution and 5 mL of the standard solution as bands
tion (1) to (8) and Assay (2) as follows: on the original line on a plate of silica gel for thin-layer
chromatography. Develop the plate with a mixture of
Chotosan Extract contains not less than 24 mg and ethanol (99.5), water and acetic acid (100) (120:80:1) to a
not more than 72 mg of hesperidin, not less than 6 mg distance of about 7 cm, and air-dry the plate. Spray evenly
and not more than 18 mg of glycyrrhizic acid 4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
(C42H62O16: 822.93), and not less than 0.3 mg of the heat the plate at 1059C for 5 minutes: one of the several
total alkaloid (rhyncophylline and hirsutine), per spots obtained from the sample solution has the same color
extract prepared with the amount specified in the tone and Rf value with the dark blue-green spot (around Rf
Method of preparation. value 0.3) from the standard solution (Ophiopogon Root).
(4) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
Identification (1) Shake 2.0 g of the dry extract (or 6.0 g
extract) with 10 mL of sodium hydroxide TS, add 5 mL of
of the viscous extract) with 20 mL of water and 2 mL of am-
1-butanol, shake, centrifuge, and use the supernatant liquid
monia TS, add 20 mL of diethyl ether, and shake. Separate
as the sample solution. Separately, dissolve 1 mg of Gin-
the diethyl ether layer, evaporate the layer under reduced
senoside Rb1 RS or ginsenoside Rb1 for thin-layer chroma-
pressure, add 1 mL of methanol to the residue, and use this
tography in 1 mL of methanol, and use this solution as the
solution as the sample solution. Separately, dissolve 1 mg
standard solution. Perform the test with these solutions as
each of rhyncophylline for thin-layer chromatography and
directed under Thin-layer Chromatography <2.03>. Spot 10
hirsutine for thin-layer chromatography in 1 mL of metha-
mL of the sample solution and 2 mL of the standard solution
nol, and use this solution as the standard solution. Perform
on a plate of silica gel for thin-layer chromatography. De-
the test with these solutions as directed under Thin-layer
velop the plate with a mixture of ethyl acetate, 1-propanol,
Chromatography <2.03>. Spot 10 mL of the sample solution
water and acetic acid (100) (7:5:4:1) to a distance of about 7
and 2 mL of the standard solution on a plate of silica gel
cm, and air-dry the plate. Spray evenly vanillin-sulfuric
with fluorescent indicator for thin-layer chromatography.
acid-ethanol TS for spraying on the plate, heat the plate at
Develop the plate with a mixture of ethyl acetate, 1-
1059C for 5 minutes, and allow to cool: one of the several
propanol, water and acetic acid (100) (7:5:4:1) to a distance
spots obtained from the sample solution has the same color
of about 7 cm, and air-dry the plate. Examine under ultravi-
tone and Rf value with the blue-purple spot from the stand-
olet light (main wavelength: 254 nm): one of the several
ard solution (Ginseng).
spots obtained from the sample solution has the same color
(5) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
tone and Rf value with one of the two dark purple spots
extract) with 10 mL of sodium hydroxide TS, add 5 mL of
from the standard solution (Uncaria Hook).
1-butanol, shake, centrifuge, and use the supernatant liquid
(2) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
as the sample solution. Separately, dissolve 1 mg of 4?-O-
extract) with 10 mL of water, add 10 mL of 1-butanol,
glycosyl-5-O-methylvisamminol for thin-layer chromatogra-
shake, centrifuge, and use the supernatant liquid as the
phy in 1 mL of methanol, and use this solution as the stand-
sample solution. Separately, dissolve 1 mg of hesperidin for
ard solution. Perform the test with these solutions as di-
thin-layer chromatography in 1 mL of methanol, and use
rected under Thin-layer Chromatography <2.03>. Spot 5 mL
this solution as the standard solution. Perform the test with
each of the sample solution and standard solution on a plate
these solutions as directed under Thin-layer Chromatogra-
of silica gel with fluorescent indicator for thin-layer chro-
phy <2.03>. Spot 20 mL of the sample solution and 10 mL of
matography. Develop the plate with a mixture of 1-butanol,
the standard solution on a plate of silica gel for thin-layer
water and acetic acid (100) (7:2:1) to a distance of about 7
chromatography. Develop the plate with a mixture of ethyl
cm, and air-dry the plate. Examine under ultraviolet light
acetate, acetone, water and acetic acid (100) (10:6:3:1) to a
(main wavelength: 254 nm): one of the several spots ob-
distance of about 7 cm, and air-dry the plate. Spray evenly
tained from the sample solution has the same color tone and
2,6-dibromo-N-chloro-1,4-benzoquinone monoimine TS on
Rf value with the blue spot from the standard solution
the plate, and allow to stand in an ammonia gas: one of the
(Saposhnikovia Root and Rhizome).
several spots obtained from the sample solution has the
(6) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
same color tone and Rf value with the blue spot from the
extract) with 10 mL of water, add 20 mL of diethyl ether,
standard solution (Citrus Unshiu Peel).
and shake. Separate the diethyl ether layer, evaporate the
(3) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
layer under reduced pressure, add 1 mL of methanol to the
extract) with 10 mL of water, add 5 mL of 1-butanol,
residue, and use this solution as the sample solution. Sepa-
shake, centrifuge, remove the 1-butanol layer, and use the
rately, dissolve 1 mg of luteolin for thin-layer chromatogra-
aqueous layer as the sample solution. Separately, heat 3.0 g
phy in 1 mL of methanol, and use this solution as the stand-
of pulverized ophiopogon root in 50 mL of water under a
ard solution. Perform the test with these solutions as di-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2752 Crude Drugs and Related Drugs Supplement I, JP XVII

rected under Thin-layer Chromatography <2.03>. Spot 10 bine these supernatant liquids, add diluted methanol (1 in 2)
mL of the sample solution and 3 mL of the standard solution to make exactly 50 mL, and use this solution as the sample
on a plate of silica gel for thin-layer chromatography. De- solution. Separately, weigh accurately about 10 mg of
velop the plate with a mixture of ethyl acetate, hexane and Glycyrrhizic Acid RS (separately determine the water <2.48>
formic acid (5:5:1) to a distance of about 7 cm, and air-dry by coulometric titration, using 10 mg), dissolve in diluted
the plate. Spray evenly iron (III) chloride-methanol TS on methanol (1 in 2) to make exactly 100 mL, and use this solu-
the plate: one of the several spots obtained from the sample tion as the standard solution. Perform the test with exactly
solution has the same color tone and Rf value with the yel- 10 mL each of the sample solution and standard solution as
low-brown spot from the standard solution (Chrysanthe- directed under Liquid Chromatography <2.01> according to
mum Flower). the following conditions, and determine the peak areas, AT
(7) Shake 2.0 g of the dry extract (or 6.0 g of the viscous and AS, of glycyrrhizic acid in each solution.
extract) with 10 mL of water, add 10 mL of 1-butanol,
Amount (mg) of glycyrrhizic acid (C42H62O16)
shake, centrifuge, and use the supernatant liquid as the
= MS × AT/AS × 1/2
sample solution. Separately, dissolve 1 mg of liquiritin for
thin-layer chromatography in 1 mL of methanol, and use MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
this solution as the standard solution. Perform the test with lated on the anhydrous basis
these solutions as directed under Thin-layer Chromatogra-
Operating conditions—
phy <2.03>. Spot 1 mL each of the sample solution and
Detector: An ultraviolet absorption photometer (wave-
standard solution on a plate of silica gel for thin-layer chro-
length: 254 nm).
matography. Develop the plate with a mixture of ethyl ace-
Column: A stainless steel column 4.6 mm in inside diame-
tate, methanol and water (20:3:2) to a distance of about 7
ter and 15 cm in length, packed with octadecylsilanized
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
silica gel for liquid chromatography (5 mm in particle diame-
on the plate, heat the plate at 1059C for 5 minutes, and exa-
ter).
mine under ultraviolet light (main wavelength: 365 nm): one
Column temperature: A constant temperature of about
of the several spots obtained from the sample solution has
409C.
the same color tone and Rf value with the yellow-green fluo-
Mobile phase: Dissolve 3.85 g of ammonium acetate in
rescent spot from the standard solution (Glycyrrhiza).
720 mL of water, and add 5 mL of acetic acid (100) and 280
(8) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
mL of acetonitrile.
extract) with 10 mL of water, add 25 mL of diethyl ether,
Flow rate: 1.0 mL per minute (the retention time of
and shake. Separate the diethyl ether layer, evaporate the
glycyrrhizic acid is about 15 minutes).
layer under reduced pressure, add 2 mL of diethyl ether to
System suitability—
the residue, and use this solution as the sample solution.
System performance: Dissolve 5 mg of monoammonium
Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro-
glycyrrhizinate for resolution check in 20 mL of dilute
matography in 1 mL of methanol, and use this solution as
ethanol. When the procedure is run with 10 mL of this solu-
the standard solution. Perform the test with these solutions
tion under the above operating conditions, the resolution
as directed under Thin-layer Chromatography <2.03>. Spot
between the peak having the relative retention time of about
10 mL of the sample solution and 5 mL of the standard solu-
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
tion on a plate of silica gel for thin-layer chromatography.
not less than 1.5.
Develop the plate with a mixture of ethyl acetate and hexane
System repeatability: When the test is repeated 6 times
(1:1) to a distance of about 7 cm, and air-dry the plate.
with 10 mL of the standard solution under the above operat-
Spray evenly vanillin-sulfuric acid TS on the plate, heat the
ing conditions, the relative standard deviation of the peak
plate at 1059 C for 5 minutes: one of the several spots ob-
area of glycyrrhizic acid is not more than 1.5z.
tained from the sample solution has the same color tone and
Rf value with the red-purple spot from the standard solu-
tion (Ginger).
Cornus Fruit
Assay
(2) Glycyrrhizic acid—Weigh accurately about 0.5 g of サンシュユ
the dry extract (or an amount of the viscous extract, equiva-
lent to about 0.5 g of the dried substance), add 20 mL of Change the Assay as follows:
ethyl acetate and 10 mL of water, and shake for 10 minutes.
Assay Weigh accurately about 1 g of fine cuttings of Cor-
After centrifugation, remove the upper layer, add 20 mL of
nus Fruit (separately determine the loss on drying <5.01>),
ethyl acetate, proceed in the same manner as described
put in a glass-stoppered centrifuge tube, add 30 mL of
above, and remove the upper layer. To the resultant aque-
diluted methanol (1 in 2), shake for 20 minutes, centrifuge,
ous layer add 10 mL of methanol, shake for 30 minutes,
and take the supernatant liquid. To the residue add 30 mL
centrifuge, and take the supernatant liquid. To the residue
of diluted methanol (1 in 2), and repeat the above process
add 20 mL of diluted methanol (1 in 2), shake for 5
twice more. Combine all the supernatant liquids, add
minutes, centrifuge, and take the supernatant liquid. Com-
diluted methanol (1 in 2) to make exactly 100 mL, and use

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2753

this solution as the sample solution. Separately, weigh accu- grayish yellow-brown to grayish brown; nodes protruded as
rately about 10 mg of loganin for assay, dissolve in diluted rings; internode of 0.3 – 0.8 cm, with scars of roots, and
methanol (1 in 2) to make exactly 100 mL, and use this solu- small protrusions consisting of scars of branched rhizomes;
tion as the standard solution. Perform the test with exactly hard in texture; a transverse section reveals cortex and stele
10 mL each of the sample solution and standard solution as distinctly; cortex 2 – 5 mm in thickness; a transverse section,
directed under Liquid Chromatography <2.01> according to grayish brown in rhizome of 1) Curcuma zedoria origin,
the following conditions, and determine the peak areas, AT light yellow to grayish yellow or light yellow-green to
and AS, of loganin in each solution. grayish yellow-green in 2) Curcuma phaeocaulis origin and
purplish brown to dark purple-brown in 3) Curcuma kwan-
Amount (mg) of loganin = MS × AT/AS
gsiensis origin, and sometimes lustrous.
MS: Amount (mg) of loganin for assay taken Odor, characteristic; taste, pungent, bitter and cool feel-
ing on chewing.
Operating conditions—
Under a microscope <5.01>, a transverse section of central
Detector: An ultraviolet absorption photometer (wave-
part reveals the outermost layer usually consisting of a cork
length: 238 nm).
layer 4 – 10 cells thick; cortex and stele divided by endoder-
Column: A stainless steel column 4.6 mm in inside diame-
mis, composed of parenchyma cells, vascular bundles scat-
ter and 15 cm in length, packed with octadecylsilianized
tered; small sized vascular bundles line up beneath the
silica gel for liquid chromatography (5 mm in particle diame-
endodermis; oil cells contain yellow-brown to dark brown
ter).
oily substances, scattered in parenchyma; parenchyma
Column temperature: A constant temperature of about
contains gelatinized starch and rarely crystals of calcium
509C.
oxalate.
Mobile phase: A mixture of water, acetonitrile and meth-
anol (55:4:1).
Flow rate: Adjust so that the retention time of loganin is
about 25 minutes. Cyperus Rhizome
System suitability—
コウブシ
System performance: When the procedure is run with 10
mL of the standard solution under the above operating con-
Add the following next to the Description:
ditions, the number of theoretical plates and the symmetry
factor of the peak of loganin are not less than 5000 and not Identification To 2.0 g of pulverized Cyperus Rhizome
more than 1.5, respectively. add 10 mL of diethyl ether, shake for 5 minutes, filter, and
System repeatability: When the test is repeated 6 times use the filtrate as the sample solution. Perform the test with
with 10 mL of the standard solution under the above operat- the sample solution as directed under Thin-layer Chroma-
ing conditions, the relative standard deviation of the peak tography <2.03>. Spot 5 mL of the sample solution on a plate
area of loganin is not more than 1.5z. of silica gel for thin-layer chromatography. Develop the
plate with a mixture of diethyl ether, cyclohexane and for-
mic acid (10:10:1) to a distance of about 7 cm, and air-dry
Zedoary the plate. Spray evenly 4-dimethylaminobenzaldehyde TS
for spraying on the plate, and heat at 1059C for 5 minutes:
ガジュツ a red-purple spot appears at an Rf value of about 0.35.

Change the Title of the monograph, Latin name,


Origin/limits of content and Description as Powdered Cyperus Rhizome
follows:
コウブシ末

Curcuma Rhizome Add the following next to the Description:


Identification To 2.0 g of Powdered Cyperus Rhizome
Curcumae Rhizoma
add 10 mL of diethyl ether, shake for 5 minutes, filter, and
ガジュツ use the filtrate as the sample solution. Perform the test with
the sample solution as directed under Thin-layer Chroma-
Curcuma Rhizome is the rhizome of 1) Curcuma
tography <2.03>. Spot 5 mL of the sample solution on a plate
zedoaria Roscoe, 2) Curcuma phaeocaulis Valeton or
of silica gel for thin-layer chromatography. Develop the
3) Curcuma kwangsiensis S. G. Lee et C. F. Liang
plate with a mixture of diethyl ether, cyclohexane and for-
(Zingiberaceae), usually after being passed through
mic acid (10:10:1) to a distance of about 7 cm, and air-dry
hot water.
the plate. Spray evenly 4-dimethylaminobenzaldehyde TS
Description Nearly ovoid to oblong-ovoid or conical rhi- for spraying on the plate, and heat at 1059C for 5 minutes:
zome, 2 – 8 cm in length, 1.5 – 4 cm in diameter; externally a red-purple spot appears at an Rf value of about 0.35.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2754 Crude Drugs and Related Drugs Supplement I, JP XVII

supernatant liquid. To the residue add 20 mL of diluted


Daiokanzoto Extract methanol (1 in 2), shake for 5 minutes, centrifuge, and take
the supernatant liquid. Combine these supernatant liquids,
大黄甘草湯エキス add diluted methanol (1 in 2) to make exactly 50 mL, and
use this solution as the sample solution. Separately, weigh
Change the Origin/limits of content, Identifica- accurately about 10 mg of Glycyrrhizic Acid RS (separately
tion and Assay (2) as follows: determine the water <2.48> by coulometric titration, using
10 mg), dissolve in diluted methanol (1 in 2) to make exactly
Daiokanzoto Extract contains not less than 3.5 mg 100 mL, and use this solution as the standard solution. Per-
of sennoside A (C42H38O20: 862.74), and not less than form the test with exactly 10 mL each of the sample solution
7 mg and not more than 21 mg (for preparation and standard solution as directed under Liquid Chromatog-
prescribed 1 g of Glycyrrhiza) or not less than 14 mg raphy <2.01> according to the following conditions, and de-
and not more than 42 mg (for preparation prescribed termine the peak areas, AT and AS, of glycyrrhizic acid in
2 g of Glycyrrhiza) of glycyrrhizic acid (C42H62O16: each solution.
822.93), per extract prepared with the amount speci-
Amount (mg) of glycyrrhizic acid (C42H62O16)
fied in the Method of preparation.
= MS × AT/AS × 1/2
Identification (1) To 1.0 g of Daiokanzoto Extract add
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
10 mL of water, shake, then add 10 mL of diethyl ether,
lated on the anhydrous basis
shake, centrifuge, and use the supernatant liquid as the
sample solution. Separately, dissolve 1 mg of rhein for thin- Operating conditions—
layer chromatography in 10 mL of acetone, and use this so- Detector: An ultraviolet absorption photometer (wave-
lution as the standard solution. Perform the test with these length: 254 nm).
solutions as directed under Thin-layer Chromatography Column: A stainless steel column 4.6 mm in inside diame-
<2.03>. Spot 5 mL each of the sample solution and standard ter and 15 cm in length, packed with octadecylsilanized
solution on a plate of silica gel for thin-layer chromatogra- silica gel for liquid chromatography (5 mm in particle diame-
phy. Develop the plate with a mixture of ethyl acetate, ter).
methanol and water (20:3:2) to a distance of about 7 cm, Column temperature: A constant temperature of about
and air-dry the plate. Examine under ultraviolet light (main 409C.
wavelength: 365 nm): one of the several spots obtained Mobile phase: Dissolve 3.85 g of ammonium acetate in
from the sample solution has the same color tone and Rf 720 mL of water, and add 5 mL of acetic acid (100) and 280
value with the orange fluorescent spot from the standard so- mL of acetonitrile.
lution (Rhubarb). Flow rate: 1.0 mL per minute (the retention time of
(2) To 0.5 g of Daiokanzoto Extract add 10 mL of glycyrrhizic acid is about 15 minutes).
water, shake, then add 10 mL of 1-butanol, shake, centri- System suitability—
fuge, and use the supernatant liquid as the sample solution. System performance: Dissolve 5 mg of monoammonium
Separately, dissolve 1 mg of liquiritin for thin-layer chroma- glycyrrhizinate for resolution check in 20 mL of dilute
tography in 1 mL of methanol, and use this solution as the ethanol. When the procedure is run with 10 mL of this solu-
standard solution. Perform the test with these solutions as tion under the above operating conditions, the resolution
directed under Thin-layer Chromatography <2.03>. Spot 1 between the peak having the relative retention time of about
mL each of the sample solution and standard solution on a 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
plate of silica gel for thin-layer chromatography. Develop not less than 1.5.
the plate with a mixture of ethyl acetate, methanol and System repeatability: When the test is repeated 6 times
water (20:3:2) to a distance of about 7 cm, and air-dry the with 10 mL of the standard solution under the above operat-
plate. Spray evenly dilute sulfuric acid on the plate, heat the ing conditions, the relative standard deviation of the peak
plate at 1059 C for 5 minutes, and examine under ultraviolet area of glycyrrhizic acid is not more than 1.5z.
light (main wavelength: 365 nm): one of the several spots
obtained from the sample solution has the same color tone
and Rf value with the yellow-green fluorescent spot from Daisaikoto Extract
the standard solution (Glycyrrhiza).
大柴胡湯エキス
Assay
(2) Glycyrrhizic acid—Weigh accurately about 0.2 g of
Change the Identification (5) as follows:
Daiokanzoto Extract, add 20 mL of ethyl acetate and 10 mL
of water, and shake for 10 minutes. After centrifugation, Identification
remove the upper layer, add 20 mL of ethyl acetate, proceed (5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
in the same manner as described above, and remove the extract) with 10 mL of water, add 25 mL of diethyl ether,
upper layer. To the resultant aqueous layer add 10 mL of and shake. Separate the diethyl ether layer, evaporate the
methanol, shake for 30 minutes, centrifuge, and take the solvent under reduced pressure, add 2 mL of diethyl ether to

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2755

the residue, and use this solution as the sample solution.


Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro- Glycyrrhiza Extract
matography in 1 mL of methanol, and use this solution as
the standard solution. Perform the test with these solutions カンゾウエキス
as directed under Thin-layer Chromatography <2.03>. Spot
10 mL of the sample solution and 5 mL of the standard solu- Change the Origin/limits of content and Assay
tion on a plate of silica gel for thin-layer chromatography. as follows:
Develop the plate with a mixture of ethyl acetate and hexane
(1:1) to a distance of about 7 cm, and air-dry the plate. Glycyrrhiza Extract contains not less than 3.6z of
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying glycyrrhizic acid (C42H62O16: 822.93).
on the plate, heat the plate at 1059C for 5 minutes, allow to
Assay Weigh accurately about 0.15 g of Glycyrrhiza Ex-
cool, and spray water: one of the several spots obtained
tract, place in a glass-stoppered centrifuge tube, add 25 mL
from the sample solution has the same color tone and Rf
of dilute ethanol, and heat at 509C for 30 minutes with oc-
value with the blue-green to grayish green spot from the
casional shaking. Cool, centrifuge, and take the superna-
standard solution (Ginger).
tant liquid. To the residue add 20 mL of dilute ethanol, and
proceed in the same manner. Combine the supernatant
liquids, add dilute ethanol to make exactly 100 mL, and use
Euodia Fruit this solution as the sample solution. Separately, weigh accu-
rately about 20 mg of Glycyrrhizic Acid RS (separately de-
ゴシュユ
termine the water <2.48> by coulometric titration, using 10
mg), dissolve in dilute ethanol to make exactly 100 mL, and
Change the Identification as follows:
use this solution as the standard solution. Perform the test
Identification To 1.0 g of pulverized Euodia Fruit add 10 with exactly 10 mL each of the sample solution and standard
mL of methanol, shake for 10 minutes, centrifuge, and use solution as directed under Liquid Chromatography <2.01>
the supernatant liquid as the sample solution. Perform the according to the following conditions, and determine the
test with the sample solution as directed under Thin-layer peak areas, AT and AS, of glycyrrhizic acid in each solution.
Chromatography <2.03>. Spot 10 mL of the sample solution
Amount (mg) of glycyrrhizic acid (C42H62O16)
on a plate of silica gel for thin-layer chromatography. De-
= MS × AT/AS
velop the plate with a mixture of acetone, 2-propanol, water
and formic acid (7:7:1:1) to a distance of about 7 cm, and MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
air-dry the plate. Examine under ultraviolet light (main lated on the anhydrous basis
wavelength: 365 nm): a blue-white fluorescent spot is ob-
Operating conditions—
served at an Rf value of about 0.6. The spot shows a yellow-
Detector: An ultraviolet absorption photometer (wave-
red color after being sprayed evenly Dragendorff's TS for
length: 254 nm).
spraying.
Column: A stainless steel column 4.6 mm in inside diame-
ter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle diame-
Gardenia Fruit ter).
Column temperature: A constant temperature of about
サンシシ
409C.
Mobile phase: Dissolve 3.85 g of ammonium acetate in
Change the origin/limits of content as follows:
720 mL of water, and add 5 mL of acetic acid (100) and 280
mL of acetonitrile.
Gardenia Fruit is the fruit of Gardenia jasminoides
Flow rate: Adjust so that the retention time of glycyrrhiz-
Ellis (Rubiaceae), sometimes after being passed
ic acid is about 15 minutes.
through hot water or steamed.
System suitability—
It contains not less than 3.0z of geniposide, calcu-
System performance: Dissolve 5 mg of monoammonium
lated on the basis of dried material.
glycyrrhizinate for resolution check in 20 mL of dilute
ethanol. When the procedure is run with 10 mL of this solu-
tion under the above operating conditions, the resolution
between the peak with the relative retention time of about
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
not less than 1.5.
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
area of glycyrrhizic acid is not more than 1.5z.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2756 Crude Drugs and Related Drugs Supplement I, JP XVII

area of glycyrrhizic acid is not more than 1.5z.


Crude Glycyrrhiza Extract
カンゾウ粗エキス Add the following:

Change the Origin/limits of content and Assay Goreisan Extract


as follows:
五苓散エキス
Crude Glycyrrhiza Extract contains not less than
4.8z of glycyrrhizic acid (C42H62O16: 822.93). Goreisan Extract contains not less than 0.3 mg and
not more than 1.2 mg (for preparation prescribed
Assay Weigh accurately about 0.15 g of Crude Glycyr-
1.5 g of Cinnamon Bark) or not less than 0.4 mg and
rhiza Extract, place in a glass-stoppered centrifuge tube,
not more than 1.6 mg (for preparation prescribed 2 g
add 25 mL of dilute ethanol, and heat at 509C for 30
of Cinnamon Bark) or not less than 0.5 mg and not
minutes with occasional shaking. Cool, centrifuge, and take
more than 2.0 mg (for preparation prescribed 2.5 g of
the supernatant liquid. To the residue add 20 mL of dilute
Cinnamon Bark) or not less than 0.6 mg and not more
ethanol, and proceed in the same manner. Combine the
than 2.4 mg (for preparation prescribed 3 g of Cinna-
supernatant liquids, add dilute ethanol to make exactly 100
mon Bark) of (E )-cinnamic acid, per extract prepared
mL, and use this solution as the sample solution. Sepa-
with the amount specified in the Method of prepara-
rately, weigh accurately about 20 mg of Glycyrrhizic Acid
tion.
RS (separately determine the water <2.48> by coulometric
titration, using 10 mg), dissolve in dilute ethanol to make Method of preparation
exactly 100 mL, and use this solution as the standard solu- 1) 2) 3) 4) 5)
tion. Perform the test with exactly 10 mL each of the sample
Alisma Tuber 5g 6g 6g 4g 6g
solution and standard solution as directed under Liquid
Polyporus Sclerotium 3g 4.5 g 4.5 g 3g 4.5 g
Chromatography <2.01> according to the following condi-
Poria Sclerotium 3g 4.5 g 4.5 g 3g 4.5 g
tions, and determine the peak areas, AT and AS, of glycyr-
Atractylodes Rhizome 3g 4.5 g 4.5 g — —
rhizic acid in each solution.
Atractylodes Lancea
Amount (mg) of glycyrrhizic acid (C42H62O16) Rhizome — — — 3 g 4.5 g
= M S × AT / AS Cinnamon Bark 2g 2.5 g 3g 1.5 g 3 g

MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-


lated on the anhydrous basis Prepare a dry extract or viscous extract as directed under
Extracts, according to the prescription 1) to 5), using the
Operating conditions— crude drugs shown above.
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm). Description Goreisan Extract occurs as a light red-brown
Column: A stainless steel column 4.6 mm in inside diame- to light brown powder, or a black-brown viscous extract. It
ter and 15 cm in length, packed with octadecylsilanized has a characteristic odor, and a slightly sweet first, bitter,
silica gel for liquid chromatography (5 mm in particle diame- then acrid taste.
ter). Identification (1) Weigh exactly 2.0 g of the dry extract
Column temperature: A constant temperature of about (or 6.0 g of the viscous extract), add 20 mL of water and 2
409C. mL of ammonia solution (28), and shake. Add 20 mL of a
Mobile phase: Dissolve 3.85 g of ammonium acetate in mixture of hexane and ethyl acetate (20:1), shake, centri-
720 mL of water, and add 5 mL of acetic acid (100) and 280 fuge, and separate the supernatant liquid. Add 20 mL of a
mL of acetonitrile. mixture of hexane and ethyl acetate (20:1) to the residue,
Flow rate: Adjust so that the retention time of glycyr- shake, centrifuge, and separate the supernatant liquid.
rhizic acid is about 15 minutes. Combine these supernatant liquids, evaporate the solvent
System suitability— under reduced pressure, add exactly 2 mL of methanol to
System performance: Dissolve 5 mg of monoammonium the residue, and use this solution as the sample solution.
glycyrrhizinate for resolution check in 20 mL of dilute Separately, weigh exactly 10 mg of alisol A for thin-layer
ethanol. When the procedure is run with 10 mL of this solu- chromatography, and dissolve in exactly 10 mL of metha-
tion under the above operating conditions, the resolution nol. Pipet 1 mL of this solution, add methanol to make ex-
between the peak with the relative retention time of about actly 50 mL, and use this solution as the standard solution.
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is Perform the test with these solutions as directed under
not less than 1.5. Thin-layer Chromatography <2.03>. Spot 2 mL each of the
System repeatability: When the test is repeated 6 times sample solution and standard solution on a plate of silica
with 10 mL of the standard solution under the above operat- gel for thin-layer chromatography. Develop the plate with a
ing conditions, the relative standard deviation of the peak

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2757

mixture of ethyl formate, water and formic acid (30:1:1) to layer as the sample solution. Separately, dissolve 1 mg of
a distance of about 7 cm, and air-dry the plate. Spray evenly (E )-cinnamaldehyde for thin-layer chromatography in 1 mL
4-methoxybenzaldehyde-sulfuric acid-acetic acid TS on the of methanol, and use this solution as the standard solution.
plate, heat the plate at 1059C for 5 minutes, allow to cool, Perform the test with these solutions as directed under
and examine under ultraviolet light (main wavelength: 365 Thin-layer Chromatography <2.03>. Spot 50 mL of the sam-
nm): one of the several spots obtained from the sample so- ple solution and 2 mL of the standard solution on a plate of
lution has the same color tone and Rf value with the yellow silica gel for thin-layer chromatography. Develop the plate
fluorescent spot from the standard solution, and it is larger with a mixture of hexane, diethyl ether and methanol
and more intense than the spot from the standard solution (15:5:1) to a distance of about 7 cm, and air-dry the plate.
(Alisma Tuber). Spray evenly 2,4-dinitrophenylhydradine TS on the plate:
(2) For preparation prescribed Atractylodes Rhizome— one of the several spots obtained from the sample solution
Shake 1.0 g of the dry extract (or 3.0 g of the viscous ex- has the same color tone and Rf value with the yellow-orange
tract) with 10 mL of water, add 25 mL of diethyl ether, and spot from the standard solution.
shake. Separate the diethyl ether layer, evaporate the sol- ii) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
vent under reduced pressure, add 2 mL of diethyl ether to extract) with 10 mL of water, add 5 mL of hexane, and
the residue, and use this solution as the sample solution. shake. Centrifuge this solution, and use the supernatant
Separately, dissolve 1 mg of atractylenolide III for thin- liquid as the sample solution. Separately, dissolve 1 mg of
layer chromatography in 2 mL of methanol, and use this so- (E )-2-methoxycinnamaldehyde for thin-layer chromatogra-
lution as the standard solution. Perform the test with these phy in 1 mL of methanol, and use this solution as the stand-
solutions as directed under Thin-layer Chromatography ard solution. Perform the test with these solutions as di-
<2.03>. Spot 5 mL each of the sample solution and standard rected under Thin-layer Chromatography <2.03>. Spot 20
solution on a plate of silica gel for thin-layer chromatogra- mL of the sample solution and 2 mL of the standard solution
phy. Develop the plate with a mixture of hexane and ethyl on a plate of silica gel for thin-layer chromatography. De-
acetate (2:1) to a distance of about 7 cm, and air-dry the velop the plate with a mixture of hexane and ethyl acetate
plate. Spray evenly 1-naphthol-sulfuric acid TS on the plate, (2:1) to a distance of about 7 cm, and air-dry the plate. Exa-
heat the plate at 1059C for 5 minutes, and allow to cool: mine under ultraviolet light (main wavelength: 365 nm): one
one of the several spots obtained from the sample solution of the several spots obtained from the sample solution has
has the same color tone and Rf value with the red to red- the same color tone and Rf value with the blue-white fluo-
purple spot from the standard solution (Atractylodes Rhi- rescent spot from the standard solution.
zome).
Purity (1) Heavy metals <1.07>—Prepare the test solu-
(3) For preparation prescribed Atractylodes Lancea
tion with 1.0 g of the dry extract (or an amount of the vis-
Rhizome—Shake 2.0 g of the dry extract (or 6.0 g of the vis-
cous extract, equivalent to 1.0 g of the dried substance) as
cous extract) with 10 mL of water, add 25 mL of hexane,
directed under Extracts (4), and perform the test (not more
and shake. Separate the hexane layer, and evaporate the sol-
than 30 ppm).
vent under reduce pressure, add 0.5 mL of hexane to the
(2) Arsenic <1.11>—Prepare the test solution with 0.67 g
residue, and use this solution as the sample solution. Per-
of the dry extract (or an amount of the viscous extract,
form the test with the sample solution as directed under
equivalent to 0.67 g of the dried substance) according to
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Method 3, and perform the test (not more than 3 ppm).
ple solution on a plate of silica gel with fluorescent indicator
for thin-layer chromatography. Develop the plate with a Loss on drying <2.41> The dry extract: Not more than
mixture of hexane and acetone (7:1) to a distance of about 10.0z (1 g, 1059C, 5 hours).
7 cm, and air-dry the plate. Examine under ultraviolet light The viscous extract: Not more than 66.7z (1 g, 1059C, 5
(main wavelength: 254 nm): a dark purple spot is observed hours).
at an Rf value of about 0.5. The spot shows a greenish
Total ash <5.01> Not more than 10.0z, calculated on the
brown color after being sprayed evenly 4-dimethylamino-
dried basis.
benzaldehyde TS for spraying, heated at 1059C for 5
minutes, and allowed to cool (Atractylodes Lancea Rhi- Assay Conduct this procedure using light-resistant vessels.
zome). Weigh accurately about 0.5 g of the dry extract (or an
(4) Perform the test according to the following i) or ii) amount of the viscous extract, equivalent to about 0.5 g of
(Cinnamon Bark). the dried substance), add exactly 50 mL of diluted methanol
i) Put 10 g of the dry extract (or 30 g of the viscous ex- (1 in 2), shake for 15 minutes, filter, and use the filtrate as
tract) in a 300-mL hard-glass flask, add 100 mL of water the sample solution. Separately, weigh accurately about 10
and 1 mL of silicone resin, connect an apparatus for essen- mg of (E )-cinnamic acid for assay, and dissolve in diluted
tial oil determination, and heat to boil under a reflux con- methanol (1 in 2) to make exactly 100 mL. Pipet 10 mL of
denser. The graduated tube of the apparatus is to be previ- this solution, add diluted methanol (1 in 2) to make exactly
ously filled with water to the standard line, and 2 mL of 100 mL, and use this solution as the standard solution. Per-
hexane is added to the graduated tube. After heating under form the test with exactly 10 mL each of the sample solution
reflux for 1 hour, separate the hexane layer, and use the and standard solution as directed under Liquid Chromatog-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2758 Crude Drugs and Related Drugs Supplement I, JP XVII

raphy <2.01> according to the following conditions, and de- solve 1 mg of alisol A for thin-layer chromatography in 1
termine the peak areas, AT and AS, of (E )-cinnamic acid in mL of methanol, and use this solution as the standard solu-
each solution. tion. Perform the test with these solutions as directed under
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Amount (mg) of (E )-cinnamic acid
ple solution and 2 mL of the standard solution on a plate of
= MS × AT/AS × 1/20
silica gel for thin-layer chromatography. Develop the plate
MS: Amount (mg) of (E )-cinnamic acid for assay taken with a mixture of ethyl acetate, hexane and acetic acid (100)
(10:10:3) to a distance of about 7 cm, and air-dry the plate.
Operating conditions—
Spray evenly 4-methoxybenzaldehyde-sulfuric acid-acetic
Detector: An ultraviolet absorption photometer (wave-
acid TS on the plate, heat the plate at 1059C for 5 minutes,
length: 273 nm).
and allow to cool, and examine under ultraviolet light (main
Column: A stainless steel column 4.6 mm in inside diame-
wavelength: 365 nm): one of the several spots obtained
ter and 15 cm in length, packed with octadecylsilanized
from the sample solution has the same color tone and Rf
silica gel for liquid chromatography (5 mm in particle diame-
value with the yellow fluorescent spot from the standard so-
ter).
lution (Alisma Tuber).
Column temperature: A constant temperature of about
(4) To 2.0 g of the dry extract (or 6.0 g of the viscous
409C.
extract), add 10 mL of water, shake, then add 5 mL of
Mobile phase: A mixture of water, acetonitrile and phos-
diethyl ether, shake, centrifuge, and use the supernatant
phoric acid (750:250:1).
liquid as the sample solution. Separately, dissolve 1 mg of
Flow rate: 1.0 mL per minute (the retention time of (E )-
paeonol for thin-layer chromatography in 1 mL of metha-
cinnamic acid is about 12 minutes).
nol, and use this solution as the standard solution. Perform
System suitability—
the test with these solutions as directed under Thin-layer
System performance: When the procedure is run with 10
Chromatography <2.03>. Spot 20 mL of the sample solution
mL of the standard solution under the above operating con-
and 2 mL of the standard solution on a plate of silica gel for
ditions, the number of theoretical plates and the symmetry
thin-layer chromatography. Develop the plate with a mix-
factor of the peak of (E )-cinnamic acid are not less than
ture of hexane and diethyl ether (5:3) to a distance of about
5000 and not more than1.5, respectively.
7 cm, and air-dry the plate. Spray evenly 4-methoxybenzal-
System repeatability: When the test is repeated 6 times
dehyde-sulfuric acid TS on the plate, and heat the plate at
with 10 mL of the standard solution under the above operat-
1059C for 5 minutes: one of the several spots obtained from
ing conditions, the relative standard deviation of the peak
the sample solution has the same color tone and Rf value
area of (E )-cinnamic acid is not more than 1.5z.
with the orange spot from the standard solution (Moutan
Containers and storage Containers—Tight containers. Bark).
(5) Perform the test according to the following i) or ii)
(Cinnamon Bark).
Goshajinkigan Extract i) Put 10 g of the dry extract (or 30 g of the viscous ex-
tract) in a 300-mL hard-glass flask, add 100 mL of water
牛車腎気丸エキス and 1 mL of silicone resin, connect an apparatus for essen-
tial oil determination, and heat to boil under a reflux con-
Change the Identification (1), (3) to (7) as denser. The graduated tube of the apparatus is to be previ-
follows: ously filled with water to the standard line, and 2 mL of
hexane is added to the graduated tube. After heating under
Identification (1) To 1.0 g of the dry extract (or 3.0 g of
reflux for 1 hour, separate 1 mL of the hexane layer, add
the viscous extract), add 10 mL of water, shake, then add 30
0.5 mL of sodium hydroxide TS, shake, centrifuge, and use
mL of methanol, shake, centrifuge, and use the supernatant
the supernatant liquid as the sample solution. Separately,
liquid as the sample solution. Perform the test with the sam-
dissolve 1 mg of (E )-cinnamaldehyde for thin-layer chroma-
ple solution as directed under Thin-layer Chromatography
tography in 1 mL of methanol, and use this solution as the
<2.03>. Spot 5 mL of the sample solution on a plate of silica
standard solution. Perform the test with these solutions as
gel for thin-layer chromatography. Develop the plate with a
directed under Thin-layer Chromatography <2.03>. Spot 50
mixture of water, methanol and 1-butanol (1:1:1) to a dis-
mL of the sample solution and 2 mL of the standard solution
tance of about 7 cm, and air-dry the plate. Spray evenly 4-
on a plate of silica gel for thin-layer chromatography.
methoxybenzaldehyde-sulfuric acid TS on the plate, heat
Develop the plate with a mixture of hexane, diethyl ether
the plate at 1059 C for 5 minutes, and allow to cool; a dark-
and methanol (15:5:1) to a distance of about 7 cm, and air-
green spot is observed at an Rf value of about 0.6 (Rehman-
dry the plate. Spray evenly 2,4-dinitrophenylhydrazine TS
nia Root).
on the plate: one of the several spots obtained from the
(3) To 2.0 g of the dry extract (or 6.0 g of the viscous
sample solution has the same color tone and Rf value with
extract), add 10 mL of sodium carbonate TS, shake, then
the yellow-orange spot from the standard solution.
add 10 mL of diethyl ether, shake, centrifuge, and use the
ii) To 2.0 g of the dry extract (or 6.0 g of the viscous
supernatant liquid as the sample solution. Separately, dis-
extract), add 10 mL of water, shake, then add 5 mL of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2759

hexane, shake, centrifuge, and use the supernatant liquid as to about 0.5 g of the dried substance), add exactly 50 mL of
the sample solution. Separately, dissolve 1 mg of (E )-2- diluted methanol (1 in 2), shake for 15 minutes, filter, and
methoxycinnamaldehyde for thin-layer chromatography in use the filtrate as the sample solution. Separately, weigh
1 mL of methanol, and use this solution as the standard so- accurately about 10 mg of loganin for assay, dissolve in
lution. Perform the test with these solutions as directed diluted methanol (1 in 2) to make exactly 100 mL, and use
under Thin-layer Chromatography <2.03>. Spot 20 mL of this solution as the standard solution. Perform the test with
the sample solution and 2 mL of the standard solution on a exactly 10 mL each of the sample solution and standard so-
plate of silica gel for thin-layer chromatography. Develop lution as directed under Liquid Chromatography <2.01> ac-
the plate with a mixture of hexane and ethyl acetate (2:1) to cording to the following conditions, and determine the peak
a distance of about 7 cm, and air-dry the plate. Examine areas, AT and AS, of loganin in each solution.
under ultraviolet light (main wavelength: 365 nm): one of
Amount (mg) of loganin = MS × AT/AS × 1/2
the several spots obtained from the sample solution has the
same color tone and Rf value with the blue-white fluores- MS: Amount (mg) of loganin for assay taken
cent spot from the standard solution.
Operating conditions—
(6) To 3.0 g of the dry extract (or 9.0 g of the viscous
Detector: An ultraviolet absorption photometer (wave-
extract), add 20 mL of diethyl ether and 2 mL of ammonia
length: 238 nm).
TS, shake for 10 minutes, centrifuge, and evaporate the
Column: A stainless steel column 4.6 mm in inside diame-
supernatant liquid under reduced pressure. Add 1 mL of
ter and 15 cm in length, packed with octadecylsilanized
acetonitrile to the residue, and use this solution as the sam-
silica gel for liquid chromatography (5 mm in particle diame-
ple solution. Separately, dissolve 1 mg of benzoylmesaco-
ter).
nine hydrochloride for thin-layer chromatography in 10 mL
Column temperature: A constant temperature of about
of ethanol (99.5), and use this solution as the standard solu-
509C.
tion. Perform the test with these solutions as directed under
Mobile phase: A mixture of water, acetonitrile and meth-
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
anol (55:4:1).
ple solution and 10 mL of the standard solution on a plate of
Flow rate: 1.2 mL per minute (the retention time of loga-
silica gel for thin-layer chromatography. Develop the plate
nin is about 25 minutes).
with a mixture of 1-butanol, water and acetic acid (100)
System suitability—
(4:2:1) to a distance of about 7 cm, and air-dry the plate.
System performance: When the procedure is run with 10
Spray evenly Dragendorff's TS for spraying on the plate,
mL of the standard solution under the above operating con-
and air-dry the plate. Then spray evenly sodium nitrite TS
ditions, the number of theoretical plates and symmetry fac-
on the plate: one of the several spots obtained from the
tor of the peak of loganin are not less than 5000 and not
sample solution has the same color tone and Rf value with
more than 1.5, respectively.
the yellow-brown spot from the standard solution (Pow-
System repeatability: When the test is repeated 6 times
dered Processed Aconite Root).
with 10 mL of the standard solution under the above operat-
(7) To 2.0 g of the dry extract (or 6.0 g of the viscous
ing conditions, the relative standard deviation of the peak
extract), add 10 mL of water, shake, then add 5 mL of 1-
area of loganin is not more than 1.5z.
butanol, shake, centrifuge, and use the supernatant liquid
as the sample solution. Separately, to 0.3 g of pulverized
plantago seed for thin-layer chromatography, add 1 mL of
methanol, warm on a water bath for 3 minutes, centrifuge Hachimijiogan Extract
after cooling, and use the supernatant liquid as the standard
八味地黄丸エキス
solution. Perform the test with these solutions as directed
under Thin-layer Chromatography <2.03>. Spot 10 mL each
Change the Identification (1), (3) to (6) as
of the sample solution and standard solution on a plate of
follows:
silica gel for thin-layer chromatography. Develop the plate
with a mixture of acetone, ethyl acetate, water and acetic Identification (1) To 1.0 g of the dry extract (or 3.0 g of
acid (100) (10:10:3:1) to a distance of about 7 cm, and air- the viscous extract), add 10 mL of water, shake, then add 30
dry the plate. Spray evenly 4-methoxybenzaldehyde-sulfuric mL of methanol, shake, centrifuge, and use the supernatant
acid TS on the plate, and heat the plate at 1059C for 5 liquid as the sample solution. Perform the test with the sam-
minutes: one of the several spots obtained from the sample ple solution as directed under Thin-layer Chromatography
solution has the same color tone and Rf value with the deep <2.03>. Spot 5 mL of the sample solution on a plate of silica
blue spot (Rf value: about 0.3) from the standard solution gel for thin-layer chromatography. Develop the plate with a
(Plantago Seed). mixture of water, methanol and 1-butanol (1:1:1) to a dis-
tance of about 7 cm, and air-dry the plate. Spray evenly 4-
Change the Assay (1) as follows: methoxybenzaldehyde-sulfuric acid TS on the plate, heat
the plate at 1059C for 5 minutes, and allow to cool; a dark
Assay (1) Loganin—Weigh accurately about 0.5 g of the
green spot is observed at an Rf value of about 0.6 (Rehman-
dry extract (or an amount of the viscous extract, equivalent
nia Root).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2760 Crude Drugs and Related Drugs Supplement I, JP XVII

(3) To 2.0 g of the dry extract (or 6.0 g of the viscous sample solution has the same color tone and Rf value with
extract), add 10 mL of sodium carbonate TS, shake, then the yellow-orange spot from the standard solution.
add 10 mL of diethyl ether, shake, centrifuge, and use the ii) To 2.0 g of the dry extract (or 6.0 g of the viscous
supernatant liquid as the sample solution. Separately, dis- extract), add 10 mL of water, shake, then add 5 mL of
solve 1 mg of alisol A for thin-layer chromatography in 1 hexane, shake, centrifuge, and use the supernatant liquid as
mL of methanol, and use this solution as the standard solu- the sample solution. Separately, dissolve 1 mg of (E )-2-
tion. Perform the test with these solutions as directed under methoxycinnamaldehyde for thin-layer chromatography in
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam- 1 mL of methanol, and use this solution as the standard
ple solution and 2 mL of the standard solution on a plate of solution. Perform the test with these solutions as directed
silica gel for thin-layer chromatography. Develop the plate under Thin-layer Chromatography <2.03>. Spot 20 mL of
with a mixture of ethyl acetate, hexane and acetic acid (100) the sample solution and 2 mL of the standard solution on a
(10:10:3) to a distance of about 7 cm, and air-dry the plate. plate of silica gel for thin-layer chromatography. Develop
Spray evenly 4-methoxybenzaldehyde-sulfuric acid-acetic the plate with a mixture of hexane and ethyl acetate (2:1) to
acid TS on the plate, heat the plate at 1059C for 5 minutes, a distance of about 7 cm, and air-dry the plate. Examine
allow to cool, and examine under ultraviolet light (main under ultraviolet light (main wavelength: 365 nm): one of
wavelength: 365 nm): one of the several spots obtained the several spots obtained from the sample solution has the
from the sample solution has the same color tone and Rf same color tone and Rf value with the blue-white fluores-
value with the yellow fluorescent spot from the standard cent spot from the standard solution.
solution (Alisma Tuber). (6) To 3.0 g of the dry extract (or 9.0 g of the viscous
(4) To 2.0 g of the dry extract (or 6.0 g of the viscous extract), add 20 mL of diethyl ether and 2 mL of ammonia
extract), add 10 mL of water, shake, then add 5 mL of TS, shake for 10 minutes, centrifuge, and evaporate the
diethyl ether, shake, centrifuge, and use the supernatant supernatant liquid under reduced pressure. Add 1 mL of
liquid as the sample solution. Separately, dissolve 1 mg of acetonitrile to the residue, and use this solution as the sam-
paeonol for thin-layer chromatography in 1 mL of metha- ple solution. Separately, dissolve 1 mg of benzoylmesaco-
nol, and use this solution as the standard solution. Perform nine hydrochloride for thin-layer chromatography in 10 mL
the test with these solutions as directed under Thin-layer of ethanol (99.5), and use this solution as the standard solu-
Chromatography <2.03>. Spot 20 mL of the sample solution tion. Perform the test with these solutions as directed under
and 2 mL of the standard solution on a plate of silica gel for Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
thin-layer chromatography. Develop the plate with a mix- ple solution and 10 mL of the standard solution on a plate of
ture of hexane and diethyl ether (5:3) to a distance of about silica gel for thin-layer chromatography. Develop the plate
7 cm, and air-dry the plate. Spray evenly 4-methoxybenzal- with a mixture of 1-butanol, water and acetic acid (100)
dehyde-sulfuric acid TS on the plate, and heat the plate at (4:2:1) to a distance of about 7 cm, and air-dry the plate.
1059C for 5 minutes: one of the several spots obtained from Spray evenly Dragendorff's TS for spraying on the plate,
the sample solution has the same color tone and Rf value and air-dry the plate. Then spray evenly sodium nitrite TS
with the orange spot from the standard solution (Moutan on the plate: one of the several spots obtained from the
Bark). sample solution has the same color tone and Rf value with
(5) Perform the test according to the following i) or ii) the yellow-brown spot from the standard solution (Proc-
(Cinnamon Bark). essed Aconite Root or Powdered Processed Aconite Root).
i) Put 10 g of the dry extract (or 30 g of the viscous ex-
tract) in a 300-mL hard-glass flask, add 100 mL of water Change the Assay (1) as follows:
and 1 mL of silicone resin, connect an apparatus for essen-
Assay (1) Loganin—Weigh accurately about 0.5 g of the
tial oil determination, and heat to boil under a reflux con-
dry extract (or an amount of the viscous extract, equivalent
denser. The graduated tube of the apparatus is to be previ-
to about 0.5 g of the dried substance), add exactly 50 mL of
ously filled with water to the standard line, and 2 mL of
diluted methanol (1 in 2), shake for 15 minutes, filter, and
hexane is added to the graduated tube. After heating under
use the filtrate as the sample solution. Separately, weigh
reflux for 1 hour, separate 1 mL of the hexane layer, add
accurately about 10 mg of loganin for assay, dissolve in
0.5 mL of sodium hydroxide TS, shake, centrifuge, and use
diluted methanol (1 in 2) to make exactly 100 mL, and use
the supernatant liquid as the sample solution. Separately,
this solution as the standard solution. Perform the test with
dissolve 1 mg of (E )-cinnamaldehyde for thin-layer chroma-
exactly 10 mL each of the sample solution and standard so-
tography in 1 mL of methanol, and use this solution as the
lution as directed under Liquid Chromatography <2.01> ac-
standard solution. Perform the test with these solutions as
cording to the following conditions, and determine the peak
directed under Thin-layer Chromatography <2.03>. Spot 50
areas, AT and AS, of loganin in each solution.
mL of the sample solution and 2 mL of the standard solution
on a plate of silica gel for thin-layer chromatography. Amount (mg) of loganin = MS × AT/AS × 1/2
Develop the plate with a mixture of hexane, diethyl ether
MS: Amount (mg) of loganin for assay taken
and methanol (15:5:1) to a distance of about 7 cm, and air-
dry the plate. Spray evenly 2,4-dinitrophenylhydrazine TS Operating conditions—
on the plate: one of the several spots obtained from the Detector: An ultraviolet absorption photometer (wave-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2761

length: 238 nm). chromatography. Develop the plate with a mixture of ethyl
Column: A stainless steel column 4.6 mm in inside diame- acetate, water and formic acid (60:1:1) to a distance of
ter and 15 cm in length, packed with octadecylsilanized about 7 cm, and air-dry the plate. Spray evenly iron (III)
silica gel for liquid chromatography (5 mm in particle diame- chloride-methanol TS on the plate: one of the several spots
ter). obtained from the sample solution has the same color tone
Column temperature: A constant temperature of about and Rf value with the dark purple spot from the standard
509C. solution (Perilla Herb).
Mobile phase: A mixture of water, acetonitrile and meth- (3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
anol (55:4:1). extract) with 10 mL of water, add 25 mL of diethyl ether,
Flow rate: 1.2 mL per minute (the retention time of loga- and shake. Separate the diethyl ether layer, evaporate the
nin is about 25 minutes). layer under reduced pressure, add 2 mL of diethyl ether to
System suitability— the residue, and use this solution as the sample solution.
System performance: When the procedure is run with 10 Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro-
mL of the standard solution under the above operating con- matography in 1 mL of methanol, and use this solution as
ditions, the number of theoretical plates and symmetry fac- the standard solution. Perform the test with these solutions
tor of the peak of loganin are not less than 5000 and not as directed under Thin-layer Chromatography <2.03>. Spot
more than 1.5, respectively. 5 mL each of the sample solution and standard solution on a
System repeatability: When the test is repeated 6 times plate of silica gel for thin-layer chromatography. Develop
with 10 mL of the standard solution under the above operat- the plate with a mixture of hexane and acetone (2:1) to a
ing conditions, the relative standard deviation of the peak distance of about 7 cm, and air-dry the plate. Spray evenly
area of loganin is not more than 1.5z. 4-dimethylaminobenzaldehyde TS for spraying on the plate,
heat the plate at 1059C for 5 minutes, allow to cool, and
spray water: one of the several spots obtained from the sam-
Hangekobokuto Extract ple solution has the same color tone and Rf value with the
blue-green to grayish green spot from the standard solution
半夏厚朴湯エキス (Ginger).

Change the Identification as follows:


Identification (1) Shake 1.0 g of the dry extract (or 3.0 g Hangeshashinto Extract
of the viscous extract) with 10 mL of water, add 25 mL of
半夏瀉心湯エキス
diethyl ether, and shake. Separate the diethyl ether layer,
evaporate the layer under reduced pressure, add 2 mL of
Change the Origin/limits of content, Identifica-
diethyl ether to the residue, and use this solution as the sam-
tion (2), (3), (5) and Assay (2) as follows:
ple solution. Separately, dissolve 1 mg of magnolol for thin-
layer chromatography in 1 mL of methanol, and use this so-
Hangeshashinto Extract contains not less than 70
lution as the standard solution. Perform the test with these
mg and not more than 210 mg (for preparation
solutions as directed under Thin-layer Chromatography
prescribed 2.5 g of Scutellaria Root) or not less than
<2.03>. Spot 5 mL each of the sample solution and standard
80 mg and not more than 240 mg (for preparation
solution on a plate of silica gel with fluorescent indicator
prescribed 3 g of Scutellaria Root) of baicalin
for thin-layer chromatography. Develop the plate with a
(C21H18O11: 446.36), not less than 18 mg and not more
mixture of ethyl acetate and hexane (1:1) to a distance of
than 54 mg (for preparation prescribed 2.5 g of
about 7 cm, and air-dry the plate. Examine under ultravio-
Glycyrrhiza) or not less than 20 mg and not more than
let light (main wavelength: 254 nm): one of the several spots
60 mg (for preparation prescribed 3 g of Glycyrrhiza)
obtained from the sample solution has the same color tone
of glycyrrhizic acid (C42H62O16: 822.93), and not less
and Rf value with the dark purple spot from the standard
than 7 mg and not more than 21 mg of berberine [as
solution (Magnolia Bark).
berberine chloride (C20H18ClNO4: 371.81)], per ex-
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
tract prepared with the amount specified in the
extract) with 10 mL of 0.1 mol/L hydrochloric acid TS, add
Method of preparation.
25 mL of diethyl ether, and shake. Separate the diethyl
ether layer, evaporate the layer under reduced pressure, add Identification
1 mL of methanol to the residue, and use this solution as the (2) For preparation prescribed Processed Ginger—
sample solution. Separately, dissolve 1 mg of rosmarinic Shake 1.0 g of the dry extract (or 3.0 g of the viscous ex-
acid for thin-layer chromatography in 1 mL of methanol, tract) with 10 mL of water, add 25 mL of diethyl ether, and
and use this solution as the standard solution. Perform the shake. Separate the diethyl ether layer, evaporate the layer
test with these solutions as directed under Thin-layer Chro- under reduced pressure, add 2 mL of diethyl ether to the
matography <2.03>. Spot 5 mL each of the sample solution residue, and use this solution as the sample solution. Sepa-
and standard solution on a plate of silica gel for thin-layer rately, dissolve 1 mg of [6]-shogaol for thin-layer chroma-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2762 Crude Drugs and Related Drugs Supplement I, JP XVII

tography in 1 mL of methanol, and use this solution as the the sample solution. Separately, weigh accurately about 10
standard solution. Perform the test with these solutions as mg of Glycyrrhizic Acid RS (separately determine the water
directed under Thin-layer Chromatography <2.03>. Spot 20 <2.48> by coulometric titration, using 10 mg), dissolve in
mL of the sample solution and 1 mL of the standard solution diluted methanol (1 in 2) to make exactly 100 mL, and use
on a plate of silica gel for thin-layer chromatography. De- this solution as the standard solution. Perform the test with
velop the plate with a mixture of cyclohexane and ethyl ace- exactly 10 mL each of the sample solution and standard so-
tate (2:1) to a distance of about 7 cm, and air-dry the plate. lution as directed under Liquid Chromatography <2.01> ac-
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying cording to the following conditions, and determine the peak
on the plate, heat the plate at 1059C for 5 minutes, allow to areas, AT and AS, of glycyrrhizic acid in each solution.
cool, and spray water: one of the several spots obtained
Amount (mg) of glycyrrhizic acid (C42H62O16)
from the sample solution has the same color tone and Rf
= MS × AT/AS × 1/2
value with the blue-green to grayish green spot from the
standard solution (Processed Ginger). MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
(3) For preparation prescribed Ginger—Shake 1.0 g of lated on the anhydrous basis
the dry extract (or 3.0 g of the viscous extract) with 10 mL
Operating conditions—
of water, add 25 mL of diethyl ether, and shake. Separate
Detector: An ultraviolet absorption photometer (wave-
the diethyl ether layer, evaporate the layer under reduced
length: 254 nm).
pressure, add 2 mL of diethyl ether to the residue, and use
Column: A stainless steel column 4.6 mm in inside diame-
this solution as the sample solution. Separately, dissolve 1
ter and 15 cm in length, packed with octadecylsilanized
mg of [6]-gingerol for thin-layer chromatography in 1 mL
silica gel for liquid chromatography (5 mm in particle diame-
of methanol, and use this solution as the standard solution.
ter).
Perform the test with these solutions as directed under
Column temperature: A constant temperature of about
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
409C.
ple solution and 5 mL of the standard solution on a plate of
Mobile phase: Dissolve 3.85 g of ammonium acetate in
silica gel for thin-layer chromatography. Develop the plate
720 mL of water, and add 5 mL of acetic acid (100) and 280
with a mixture of ethyl acetate and hexane (1:1) to a dis-
mL of acetonitrile.
tance of about 7 cm, and air-dry the plate. Spray evenly 4-
Flow rate: 1.0 mL per minute (the retention time of
dimethylaminobenzaldehyde TS for spraying on the plate,
glycyrrhizic acid is about 15 minutes).
heat the plate at 1059C for 5 minutes, allow to cool, and
System suitability—
spray water: one of the several spots obtained from the sam-
System performance: Dissolve 5 mg of monoammonium
ple solution has the same color tone and Rf value with the
glycyrrhizinate for resolution check in 20 mL of dilute
blue-green to grayish green spot from the standard solution
ethanol. When the procedure is run with 10 mL of this solu-
(Ginger).
tion under the above operating conditions, the resolution
(5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
between the peak having the relative retention time of about
extract) with 10 mL of water, add 5 mL of 1-butanol,
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
shake, centrifuge, and use the supernatant liquid as the
not less than 1.5. Dissolve 1 mg of baicalein for resolution
sample solution. Separately, dissolve 1 mg of liquiritin for
check in 50 mL of methanol. To 2 mL of this solution add 2
thin-layer chromatography in 1 mL of methanol, and use
mL of the standard solution. When the procedure is run
this solution as the standard solution. Perform the test with
with 10 mL of this solution under the above operating condi-
these solutions as directed under Thin-layer Chromatogra-
tions, the resolution between the peaks of glycyrrhizic acid
phy <2.03>. Spot 1 mL each of the sample solution and
and baicalein is not less than 1.5.
standard solution on a plate of silica gel for thin-layer chro-
System repeatability: When the test is repeated 6 times
matography. Develop the plate with a mixture of ethyl ace-
with 10 mL of the standard solution under the above operat-
tate, methanol and water (20:3:2) to a distance of about 7
ing conditions, the relative standard deviation of the peak
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
area of glycyrrhizic acid is not more than 1.5z.
on the plate, heat the plate at 1059C for 5 minutes, and exa-
ii) Weigh accurately about 0.5 g of the dry extract (or an
mine under ultraviolet light (main wavelength: 365 nm): one
amount of the viscous extract, equivalent to about 0.5 g of
of the several spots obtained from the sample solution has
the dried substance), add 20 mL of ethyl acetate and 10 mL
the same color tone and Rf value with the yellow-green fluo-
of water, and shake for 10 minutes. After centrifugation,
rescent spot from the standard solution (Glycyrrhiza).
remove the upper layer, add 20 mL of ethyl acetate, proceed
Assay in the same manner as described above, and remove the
(2) Glycyrrhizic acid—Perform the test according to the upper layer. To the resultant aqueous layer add 10 mL of
following i) or ii). methanol, shake for 30 minutes, centrifuge, and take the su-
i) Weigh accurately about 0.5 g of the dry extract (or an pernatant liquid. To the residue add 20 mL of diluted meth-
amount of the viscous extract, equivalent to about 0.5 g of anol (1 in 2), shake for 5 minutes, centrifuge, and take the
the dried substance), add exactly 50 mL of diluted methanol supernatant liquid. Combine these supernatant liquids, add
(1 in 2), shake for 15 minutes, filter, and use the filtrate as diluted methanol (1 in 2) to make exactly 50 mL, and use

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2763

this solution as the sample solution. Separately, weigh accu- mixture of ethyl acetate, 1-propanol, water and acetic acid
rately about 10 mg of Glycyrrhizic Acid RS (separately de- (100) (7:5:4:1) to a distance of about 7 cm, and air-dry the
termine the water <2.48> by coulometric titration, using 10 plate. Spray evenly vanillin-sulfuric acid-ethanol TS for
mg), dissolve in diluted methanol (1 in 2) to make exactly spraying on the plate, heat the plate at 1059 C for 5 minutes,
100 mL, and use this solution as the standard solution. Per- and allow to cool: one of the several spots obtained from
form the test with exactly 10 mL each of the sample solution the sample solution has the same color tone and Rf value
and standard solution as directed under Liquid Chromatog- with the blue-purple spot from the standard solution (Gin-
raphy <2.01> according to the following conditions, and de- seng).
termine the peak areas, AT and AS, of glycyrrhizic acid in (2) For preparation prescribed Atractylodes Rhizome—
each solution. To 3.0 g of the dry extract (or 9.0 g of the viscous extract)
add 30 mL of water, shake, then add 50 mL of diethyl
Amount (mg) of glycyrrhizic acid (C42H62O16)
ether, shake, and separate the diethyl ether layer. Evaporate
= MS × AT/AS × 1/2
the layer under reduced pressure, add 1 mL of diethyl ether
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- to the residue, and use this solution as the sample solution.
lated on the anhydrous basis Separately, dissolve 1 mg of atractylenolide III for thin-
layer chromatography in 1 mL of methanol, and use this so-
Operating conditions—
lution as the standard solution. Perform the test with these
Proceed as directed in the operating conditions in i).
solutions as directed under Thin-layer Chromatography
System suitability—
<2.03>. Spot 5 mL of the sample solution and 10 mL of the
System repeatability: Proceed as directed in the system
standard solution on a plate of silica gel for thin-layer chro-
suitability in i).
matography. Develop the plate with a mixture of ethyl ace-
System performance: Dissolve 5 mg of monoammonium
tate and hexane (1:1) to a distance of about 7 cm, and air-
glycyrrhizinate for resolution check in 20 mL of dilute
dry the plate. Spray evenly 1-naphthol-sulfuric acid TS on
ethanol. When the procedure is run with 10 mL of this solu-
the plate, heat the plate at 1059C for 5 minutes, and allow
tion under the above operating conditions, the resolution
to cool: one of the several spots obtained from the sample
between the peak having the relative retention time of about
solution has the same color tone and Rf value with the red
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
spot from the standard solution (Atractylodes Rhizome).
not less than 1.5.
(3) For preparation prescribed Atractylodes Lancea
Rhizome—To 2.0 g of the dry extract (or 6.0 g of the vis-
cous extract) add 10 mL of water, shake, then add 25 mL of
Hochuekkito Extract hexane, shake, and separate the hexane layer. Evaporate the
layer under reduced pressure, add 2 mL of hexane to the
補中益気湯エキス
residue, and use this solution as the sample solution. Per-
form the test with the sample solution as directed under
Change the Origin/limits of content, Identifica-
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
tion and Assay (3) as follows:
ple solution on a plate of silica gel with fluorescent indicator
for thin-layer chromatography. Develop the plate with a
Hochuekkito Extract contains not less than 16 mg
mixture of hexane and acetone (7:1) to a distance of about 7
and not more than 64 mg of hesperidin, not less than
cm, and air-dry the plate. Examine under ultraviolet light
0.3 mg and not more than 1.2 mg (for preparation
(main wavelength: 254 nm): a dark purple spot is observed
prescribed 1 g of Bupleurum Root) or not less than
at an Rf value of about 0.5. The spot shows a greenish
0.6 mg and not more than 2.4 mg (for preparation
brown color after being sprayed evenly 4-dimethylamino-
prescribed 2 g of Bupleurum Root) of saikosaponin
benzaldehyde TS for spraying, heated at 1059C for 5
b2, and not less than 10 mg and not more than 30 mg
minutes, and allowed to cool (Atractylodes Lancea Rhi-
of glycyrrhizic acid (C42H62O16: 822.93), per extract
zome).
prepared with the amount specified in the Method of
(4) To 3.0 g of the dry extract (or 9.0 g of the viscous
preparation.
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 10 mL of sodium hydroxide TS, evaporate the supernatant liquid under reduced pressure.
shake, then add 5 mL of 1-butanol, and shake. Centrifuge Add 30 mL of water and 20 mL of diethyl ether to the
this solution, and use the supernatant liquid as the sample residue, shake, remove the diethyl ether layer, and separate
solution. Separately, dissolve 1 mg of Ginsenoside Rb1 RS the aqueous layer. To the aqueous layer add 20 mL of 1-
or ginsenoside Rb1 for thin-layer chromatography in 1 mL butanol, shake, and separate the 1-butanol layer. To the 1-
of methanol, and use this solution as the standard solution. butanol layer add 20 mL of water, shake, separate the 1-
Perform the test with these solutions as directed under butanol layer, evaporate the layer under reduced pressure,
Thin-layer Chromatography <2.03>. Spot 5 mL each of the add 1 mL of methanol to the residue, and use this solution
sample solution and standard solution on a plate of silica as the sample solution. Separately, dissolve 1 mg of as-
gel for thin-layer chromatography. Develop the plate with a tragaloside IV for thin-layer chromatography in 1 mL of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2764 Crude Drugs and Related Drugs Supplement I, JP XVII

methanol, and use this solution as the standard solution. 5 mL of the sample solution and 2 mL of the standard solu-
Perform the test with these solutions as directed under tion on a plate of silica gel for thin-layer chromatography.
Thin-layer Chromatography <2.03>. Spot 5 mL each of the Develop the plate with a mixture of ethyl acetate, ethanol
sample solution and standard solution on a plate of oc- (99.5) and water (8:2:1) to a distance of about 7 cm, and air-
tadecylsilanized silica gel for thin-layer chromatography. dry the plate. Spray evenly 4-dimethylaminobenzaldehyde
Develop the plate with a mixture of methanol, water, 1- TS for spraying on the plate, heat the plate at 1059C for 5
butanol and acetic acid (100) (60:30:10:1) to a distance of minutes, and examine under ultraviolet light (main wave-
about 7 cm, and air-dry the plate. Spray evenly 4-dimethyl- length: 365 nm): one of the several spots obtained from the
aminobenzaldehyde TS for spraying on the plate, and heat sample solution has the same color tone and Rf value with
the plate at 1059 C for 5 minutes: one of the several spots the yellow fluorescent spot from the standard solution
obtained from the sample solution has the same color tone (Bupleurum Root).
and Rf value with the red-brown spot from the standard (8) To 2.0 g of the dry extract (or 6.0 g of the viscous
solution (Astragalus Root). extract) add 30 mL of water, shake, then add 50 mL of 1-
(5) To 3.0 g of the dry extract (or 9.0 g of the viscous butanol, shake, and separate the 1-butanol layer. Evaporate
extract) add 30 mL of water, shake, then add 50 mL of the layer under reduced pressure, add 3 mL of methanol to
diethyl ether, shake, and separate the diethyl ether layer. the residue, and use this solution as the sample solution.
Evaporate the layer under reduced pressure, add 1 mL of Separately, dissolve 1 mg of liquiritin for thin-layer chroma-
diethyl ether to the residue, and use this solution as the sam- tography in 1 mL of methanol, and use this solution as the
ple solution. Separately, dissolve 1 mg of (Z )-ligustilide for standard solution. Perform the test with these solutions as
thin-layer chromatography in 10 mL of methanol, and use directed under Thin-layer Chromatography <2.03>. Spot 1
this solution as the standard solution. Perform the test with mL each of the sample solution and standard solution on a
these solutions as directed under Thin-layer Chromatogra- plate of silica gel for thin-layer chromatography. Develop
phy <2.03>. Spot 10 mL each of the sample solution and the plate with a mixture of ethyl acetate, methanol and
standard solution on a plate of silica gel for thin-layer chro- water (20:3:2) to a distance of about 7 cm, and air-dry the
matography. Develop the plate with a mixture of ethyl ace- plate. Spray evenly dilute sulfuric acid on the plate, heat the
tate and hexane (1:1) to a distance of about 7 cm, and air- plate at 1059 C for 5 minutes, and examine under ultraviolet
dry the plate. Examine under ultraviolet light (main wave- light (main wavelength: 365 nm): one of the several spots
length: 365 nm): one of the several spots obtained from the obtained from the sample solution has the same color tone
sample solution has the same color tone and Rf value with and Rf value with the yellow-green fluorescent spot from
the blue-white fluorescent spot from the standard solution the standard solution (Glycyrrhiza).
(Japanese Angelica Root). (9) For preparation prescribed Ginger—To 3.0 g of the
(6) To 2.0 g of the dry extract (or 6.0 g of the viscous dry extract (or 9.0 g of the viscous extract) add 30 mL of
extract) add 30 mL of water, shake, then add 50 mL of 1- water, shake, then add 50 mL of diethyl ether, shake, and
butanol, shake, and separate the 1-butanol layer. Evaporate separate the diethyl ether layer. Evaporate the layer under
the layer under reduced pressure, add 3 mL of methanol to reduced pressure, add 1 mL of diethyl ether to the residue,
the residue, and use this solution as the sample solution. and use this solution as the sample solution. Separately, dis-
Separately, dissolve 1 mg of hesperidin for thin-layer chro- solve 1 mg of [6]-gingerol for thin-layer chromatography in
matography in 2 mL of methanol, and use this solution as 1 mL of methanol, and use this solution as the standard so-
the standard solution. Perform the test with these solutions lution. Perform the test with these solutions as directed
as directed under Thin-layer Chromatography <2.03>. Spot under Thin-layer Chromatography <2.03>. Spot 5 mL each
2 mL of the sample solution and 20 mL of the standard solu- of the sample solution and standard solution on a plate of
tion on a plate of silica gel for thin-layer chromatography. silica gel for thin-layer chromatography. Develop the plate
Develop the plate with a mixture of ethyl acetate, acetone, with a mixture of ethyl acetate and hexane (1:1) to a dis-
water and acetic acid (100) (10:6:3:1) to a distance of about tance of about 7 cm, and air-dry the plate. Spray evenly 4-
7 cm, and air-dry the plate. Spray evenly 2,6-dibromo-N- dimethylaminobenzaldehyde TS for spraying on the plate,
chloro-1,4-benzoquinone monoimine TS on the plate, and heat the plate at 1059C for 5 minutes, allow to cool, and
expose to ammonia vapor: one of the several spots obtained spray water: one of the several spots obtained from the sam-
from the sample solution has the same color tone and Rf ple solution has the same color tone and Rf value with the
value with the blue spot from the standard solution (Citrus blue-green to grayish green spot from the standard solution
Unshiu Peel). (Ginger).
(7) To 2.0 g of the dry extract (or 6.0 g of the viscous (10) For preparation prescribed Processed Ginger—Put
extract) add 10 mL of sodium hydroxide TS, shake, then 10 g of the dry extract (or 30 g of the viscous extract) in a
add 5 mL of 1-butanol, and shake. Centrifuge this solution, 300-mL hard-glass flask, add 100 mL of water and 1 mL of
and use the supernatant liquid as the sample solution. Sepa- silicone resin, connect an apparatus for essential oil deter-
rately, dissolve 1 mg of saikosaponin b2 for thin-layer chro- mination, and heat to boil under a reflux condenser. The
matography in 1 mL of methanol, and use this solution as graduated tube of the apparatus is previously filled with
the standard solution. Perform the test with these solutions water to the standard line, and 2 mL of hexane is added to
as directed under Thin-layer Chromatography <2.03>. Spot the graduated tube. After heating under reflux for 1 hour,

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2765

separate the hexane layer, and use the layer as the sample Amount (mg) of glycyrrhizic acid (C42H62O16)
solution. Separately, dissolve 1 mg of [6]-shogaol for thin- = MS × AT/AS × 1/2
layer chromatography in 1 mL of methanol, and use this so-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
lution as the standard solution. Perform the test with these
lated on the anhydrous basis
solutions as directed under Thin-layer Chromatography
<2.03>. Spot 60 mL of the sample solution and 10 mL of the Operating conditions—
standard solution on a plate of silica gel for thin-layer chro- Detector: An ultraviolet absorption photometer (wave-
matography. Develop the plate with a mixture of cyclo- length: 254 nm).
hexane and ethyl acetate (2:1) to a distance of about 7 cm, Column: A stainless steel column 4.6 mm in inside diame-
and air-dry the plate. Spray evenly 4-dimethylamino- ter and 15 cm in length, packed with octadecylsilanized
benzaldehyde TS for spraying on the plate, heat the plate at silica gel for liquid chromatography (5 mm in particle diame-
1059C for 5 minutes, allow to cool, and spray water: one of ter).
the several spots obtained from the sample solution has the Column temperature: A constant temperature of about
same color tone and Rf value with the blue-green to grayish 409C.
green spot from the standard solution (Processed Ginger). Mobile phase: Dissolve 3.85 g of ammonium acetate in
(11) To 2.0 g of the dry extract (or 6.0 g of the viscous 720 mL of water, and add 5 mL of acetic acid (100) and 280
extract) add 30 mL of water, shake, then add 50 mL of 1- mL of acetonitrile.
butanol, shake, and separate the 1-butanol layer. Evaporate Flow rate: 1.0 mL per minute (the retention time of
the layer under reduced pressure, add 3 mL of methanol to glycyrrhizic acid is about 15 minutes).
the residue, and use this solution as the sample solution. System suitability—
Use (E )-isoferulic acid-(E )-ferulic acid TS for thin-layer System performance: Dissolve 5 mg of monoammonium
chromatography as the standard solution. Perform the test glycyrrhizinate for resolution check in 20 mL of dilute
with these solutions as directed under Thin-layer Chroma- ethanol. When the procedure is run with 10 mL of this solu-
tography <2.03>. Spot 5 mL of the sample solution and 2 mL tion under the above operating conditions, the resolution
of the standard solution on a plate of silica gel for thin-layer between the peak having the relative retention time of about
chromatography. Develop the plate with a mixture of ethyl 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
acetate, acetone and water (20:12:3) to a distance of about 7 not less than 1.5.
cm, and air-dry the plate. Spray evenly sulfuric acid on the System repeatability: When the test is repeated 6 times
plate, heat the plate at 1059C for 5 minutes, and examine with 10 mL of the standard solution under the above operat-
under ultraviolet light (main wavelength: 365 nm): one of ing conditions, the relative standard deviation of the peak
the several spots obtained from the sample solution has the area of glycyrrhizic acid is not more than 1.5z.
same color tone and Rf value with the light yellow-white
fluorescent spot from the standard solution (Cimicifuga
Rhizome). Jujube
Assay
タイソウ
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
the dry extract (or an amount of the viscous extract, equiva-
Change the latin name and origin/limits of con-
lent to about 0.5 g of the dried substance), add 20 mL of
tent as follows:
ethyl acetate and 10 mL of water, and shake for 10 minutes.
After centrifugation, remove the upper layer, add 20 mL of Ziziphi Fructus
ethyl acetate, proceed in the same manner as described
above, and remove the upper layer. To the resultant aque- Jujube is the fruit of Ziziphus jujuba Miller var.
ous layer add 10 mL of methanol, shake for 30 minutes, inermis Rehder (Rhamnaceae).
centrifuge, and take the supernatant liquid. To the residue
add 20 mL of diluted methanol (1 in 2), shake for 5
minutes, centrifuge, and take the supernatant liquid. Com- Jujube Seed
bine these supernatant liquids, add diluted methanol (1 in 2)
to make exactly 50 mL, and use this solution as the sample サンソウニン
solution. Separately, weigh accurately about 10 mg of
Glycyrrhizic Acid RS (separately determine the water <2.48> Change the latin name and origin/limits of con-
by coulometric titration, using 10 mg), dissolve in diluted tent as follows:
methanol (1 in 2) to make exactly 100 mL, and use this solu-
Ziziphi Semen
tion as the standard solution. Perform the test with exactly
10 mL each of the sample solution and standard solution as
Jujube Seed is the seed of Ziziphus jujuba Miller
directed under Liquid Chromatography <2.01> according to
var. spinosa Hu ex H. F. Chou (Rhamnaceae).
the following conditions, and determine the peak areas, AT
and AS, of glycyrrhizic acid in each solution.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2766 Crude Drugs and Related Drugs Supplement I, JP XVII

Develop the plate with a mixture of ethyl acetate, 1-


Juzentaihoto Extract propanol, water and acetic acid (100) (7:5:4:1) to a distance
of about 7 cm, and air-dry the plate. Spray evenly 4-
十全大補湯エキス dimethylaminobenzaldehyde TS for spraying on the plate,
heat the plate at 1059C for 5 minutes, and allow to cool:
Change the Origin/limits of content, Identifica- one of the several spots obtained from the sample solution
tion and Assay (3) as follows: has the same color tone and Rf value with the red-brown
spot from the standard solution (Astragalus Root).
Juzentaihoto Extract contains not less than 1.5 mg (3) For preparation prescribed Atractylodes Rhizome—
(for preparation prescribed 2.5 g of Ginseng) or not Shake 1.0 g of the dry extract (or 3.0 g of the viscous
less than 1.8 mg (for preparation prescribed 3 g of extract) with 10 mL of water, add 5 mL of diethyl ether,
Ginseng) of ginsenoside Rb1 (C54H92O23: 1109.29), shake, and centrifuge. Use the supernatant liquid as the
not less than 26 mg and not more than 78 mg of sample solution. Separately, dissolve 1 mg of atrac-
paeonifrolin (C23H28O11: 480.46), and not less than tylenolide III for thin-layer chromatography in 1 mL of
6 mg and not more than 18 mg (for preparation methanol, and use this solution as the standard solution.
prescribed 1 g of Glycyrrhiza) or not less than 10 mg Perform the test with these solutions as directed under
and not more than 30 mg (for preparation prescribed Thin-layer Chromatography <2.03>. Spot 10 mL each of the
1.5 g of Glycyrrhiza) of glycyrrhizic acid (C42H62O16: sample solution and standard solution on a plate of silica
822.93), per extract prepared with the amount speci- gel for thin-layer chromatography. Develop the plate with a
fied in the Method of preparation. mixture of ethyl acetate and hexane (1:1) to a distance of
about 7 cm, and air-dry the plate. Spray evenly 1-naphthol-
Identification (1) Shake 2.0 g of the dry extract (or 6.0 g
sulfuric acid TS on the plate, heat the plate at 1059C for
of the viscous extract) with 15 mL of sodium hydroxide TS,
5 minutes, and allow to cool: one of the several spots
centrifuge, and take the supernatant liquid. To the liquid
obtained from the sample solution has the same color tone
add 10 mL of 1-butanol, shake, centrifuge, and separate the
and Rf value with the red spot from the standard solution
1-butanol layer. To the 1-butanol layer add 10 mL of water,
(Atractylodes Rhizome).
shake, centrifuge, and separate the 1-butanol layer.
(4) For preparation prescribed Atractylodes Lancea
Evaporate the layer under reduced pressure, add 1 mL of
Rhizome—Shake 5.0 g of the dry extract (or 15.0 g of the
methanol to the residue, and use this solution as the sample
viscous extract) with 10 mL of water, add 25 mL of hexane ,
solution. Separately, dissolve 1 mg of Ginsenoside Rb1 RS
and shake. Separate the hexane layer, evaporate the layer
or ginsenoside Rb1 for thin-layer chromatography in 1 mL
under reduced pressure, add 2 mL of hexane to the residue,
of methanol, and use this solution as the standard solution.
and use this solution as the sample solution. Perform the
Perform the test with these solutions as directed under
test with the sample solution as directed under Thin-layer
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
Chromatography <2.03>. Spot 40 mL of the sample solution
ple solution and 2 mL of the standard solution on a plate of
on a plate of silica gel with fluorescent indicator for thin-
silica gel for thin-layer chromatography. Develop the plate
layer chromatography. Develop the plate with a mixture of
with a mixture of ethyl acetate, 1-propanol, water and
hexane and acetone (7:1) to a distance of about 7 cm, and
acetic acid (100) (7:5:4:1) to a distance of about 7 cm, and
air-dry the plate. Examine under ultraviolet light (main
air-dry the plate. Spray evenly 4-dimethylaminobenzalde-
wavelength: 254 nm): a dark purple spot is observed at an
hyde TS for spraying on the plate, heat the plate at 1059 C
Rf value of about 0.5. The spot shows a greenish brown
for 5 minutes, and allow to cool: one of the several spots
color after being sprayed evenly 4-dimethylaminobenzalde-
obtained from the sample solution has the same color tone
hyde TS for spraying, heated at 1059C for 5 minutes, and
and Rf value with the dark brown spot from the standard
allowed to cool (Atractylodes Lancea Rhizome).
solution (Ginseng).
(5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
(2) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
extract) with 15 mL of water and 5 mL of 0.1 mol/L hydro-
extract) with 15 mL of sodium hydroxide TS, centrifuge,
chloric acid TS, add 25 mL of diethyl ether, and shake.
and take the supernatant liquid. To the liquid add 10 mL of
Separate the diethyl ether layer, evaporate the layer under
1-butanol, shake, centrifuge, and separate the 1-butanol
reduced pressure, then add 2 mL of diethyl ether to the
layer. To the 1-butanol layer add 10 mL of water, shake,
residue, and use this solution as the sample solution. Sepa-
centrifuge, and separate the 1-butanol layer. Evaporate the
rately, dissolve 1 mg of (Z )-ligustilide for thin-layer chro-
layer under reduced pressure, add 1 mL of methanol to the
matography in 10 mL of methanol, and use this solution as
residue, and use this solution as the sample solution. Sepa-
the standard solution. Perform the test with these solutions
rately, dissolve 1 mg of astragaloside IV for thin-layer chro-
as directed under Thin-layer Chromatography <2.03>. Spot
matography in 1 mL of methanol, and use this solution as
10 mL each of the sample solution and standard solution on
the standard solution. Perform the test with these solutions
a plate of silica gel for thin-layer chromatography. Develop
as directed under Thin-layer Chromatography <2.03>. Spot
the plate with a mixture of ethyl acetate and hexane (1:1) to
10 mL of the sample solution and 2 mL of the standard solu-
a distance of about 7 cm, and air-dry the plate. Examine
tion on a plate of silica gel for thin-layer chromatography.
under ultraviolet light (main wavelength: 365 nm): one of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2767

the several spots obtained from the sample solution has the centrifuge, and use the supernatant liquid as the sample
same color tone and Rf value with the blue-white fluores- solution. Separately, dissolve 1 mg of (E )-2-methoxycin-
cent spot from the standard solution (Cnidium Rhizome; namaldehyde for thin-layer chromatography in 1 mL of
Japanese Angelica Root). methanol, and use this solution as the standard solution.
(6) Shake 1.0 g of the dry extract (or 3.0 g of the viscous Perform the test with these solutions as directed under
extract) with 10 mL of water, add 10 mL of 1-butanol, Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
shake, centrifuge, and use the supernatant liquid as the ple solution and 2 mL of the standard solution on a plate of
sample solution. Separately, dissolve 1 mg of Paeoniflorin silica gel for thin-layer chromatography. Develop the plate
RS or paeoniflorine for thin-layer chromatography in 1 mL with a mixture of hexane and ethyl acetate (2:1) to a dis-
of methanol, and use this solution as the standard solution. tance of about 7 cm, and air-dry the plate. Examine under
Perform the test with these solutions as directed under ultraviolet light (main wavelength: 365 nm): one of the
Thin-layer Chromatography <2.03>. Spot 5 mL each of the several spots obtained from the sample solution has the
sample solution and standard solution on a plate of silica same color tone and Rf value with the blue-white fluores-
gel for thin-layer chromatography. Develop the plate with a cent spot from the standard solution.
mixture of ethyl acetate, methanol and water (20:3:2) to a (9) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
distance of about 7 cm, and air-dry the plate. Spray evenly extract) with 10 mL of water, add 10 mL of 1-butanol,
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and shake, centrifuge, and use the supernatant liquid as the
heat the plate at 1059C for 5 minutes: one of the several sample solution. Separately, dissolve 1 mg of liquiritin for
spots obtained from the sample solution has the same color thin-layer chromatography in 1 mL of methanol, and use
tone and Rf value with the purple spot from the standard this solution as the standard solution. Perform the test with
solution (Peony Root). these solutions as directed under Thin-layer Chromatogra-
(7) Shake 1.0 g of the dry extract (or 3.0 g of the viscous phy <2.03>. Spot 1 mL each of the sample solution and
extract) with 10 mL of water, add 30 mL of methanol, standard solution on a plate of silica gel for thin-layer chro-
shake, centrifuge, and use the supernatant liquid as the matography. Develop the plate with a mixture of ethyl ace-
sample solution. Perform the test with the sample solution tate, methanol and water (20:3:2) to a distance of about 7
as directed under Thin-layer Chromatography <2.03>. Spot cm, and air-dry the plate. Spray evenly dilute sulfuric acid
5 mL of the sample solution on a plate of silica gel for thin- on the plate, heat the plate at 1059C for 5 minutes, and exa-
layer chromatography. Develop the plate with a mixture of mine under ultraviolet light (main wavelength: 365 nm): one
water, methanol and 1-butanol (1:1:1) to a distance of of the several spots obtained from the sample solution has
about 7 cm, and air-dry the plate. Spray evenly 4-methox- the same color tone and Rf value with the yellow-green fluo-
ybenzaldehyde-sulfuric acid TS on the plate, heat the plate rescent spot from the standard solution (Glycyrrhiza).
at 1059C for 5 minutes, and allow to cool: a dark green spot
Assay
is observed at an Rf value of about 0.6 (Rehmannia Root).
(3) Glycyrrhizic acid—Perform the test according to the
(8) Perform the test according to the following i) or ii)
following i) or ii).
(Cinnamon Bark).
i) Weigh accurately about 0.5 g of the dry extract (or an
i) Put 10 g of the dry extract (or 30 g of the viscous ex-
amount of the viscous extract, equivalent to about 0.5 g of
tract) in a 300-mL hard-glass flask, add 100 mL of water
the dried substance), add exactly 50 mL of diluted methanol
and 1 mL of silicone resin, connect the apparatus for essen-
(1 in 2), shake for 15 minutes, filter, and use the filtrate as
tial oil determination, and heat to boil under a reflux con-
the sample solution. Separately, weigh accurately about 10
denser. The graduated tube of the apparatus is to be previ-
mg of Glycyrrhizic Acid RS (separately determine the water
ously filled with water to the standard line, and 2 mL of
<2.48> by coulometric titration, using 10 mg), dissolve in
hexane is added to the graduated tube. After heating under
diluted methanol (1 in 2) to make exactly 100 mL, and use
reflux for 1 hour, separate the hexane layer, and use the
this solution as the standard solution. Perform the test with
layer as the sample solution. Separately, dissolve 1 mg of
exactly 10 mL each of the sample solution and standard so-
(E )-cinnamaldehyde for thin-layer chromatography in 1 mL
lution as directed under Liquid Chromatography <2.01> ac-
of methanol, and use this solution as the standard solution.
cording to the following conditions, and determine the peak
Perform the test with these solutions as directed under
areas, AT and AS, of glycyrrhizic acid in each solution.
Thin-layer Chromatography <2.03>. Spot 50 mL of the sam-
ple solution and 2 mL of the standard solution on a plate of Amount (mg) of glycyrrhizic acid (C42H62O16)
silica gel for thin-layer chromatography. Develop the plate = MS × AT/AS × 1/2
with a mixture of hexane, diethyl ether and methanol
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
(15:5:1) to a distance of about 7 cm, and air-dry the plate.
lated on the anhydrous basis
Spray evenly 2,4-dinitrophenylhydrazine TS on the plate:
one of the several spots obtained from the sample solution Operating conditions—
has the same color tone and Rf value with the yellow-orange Detector: An ultraviolet absorption photometer (wave-
spot from the standard solution. length: 254 nm).
ii) Shake 2.0 g of the dry extract (or 6.0 g of the viscous Column: A stainless steel column 4.6 mm in inside diame-
extract) with 10 mL of water, add 5 mL of hexane, shake, ter and 15 cm in length, packed with octadecylsilanized

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2768 Crude Drugs and Related Drugs Supplement I, JP XVII

silica gel for liquid chromatography (5 mm in particle diame- System suitability—


ter). System repeatability: Proceed as directed in the system
Column temperature: A constant temperature of about suitability in i).
409C. System performance: Dissolve 5 mg of monoammonium
Mobile phase: Dissolve 3.85 g of ammonium acetate in glycyrrhizinate for resolution check in 20 mL of dilute
720 mL of water, and add 5 mL of acetic acid (100) and 280 ethanol. When the procedure is run with 10 mL of this solu-
mL of acetonitrile. tion under the above operating conditions, the resolution
Flow rate: 1.0 mL per minute (the retention time of between the peak having the relative retention time of about
glycyrrhizic acid is about 15 minutes). 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
System suitability— not less than 1.5.
System performance: Dissolve 5 mg of monoammonium
glycyrrhizinate for resolution check in 20 mL of dilute
ethanol. When the procedure is run with 10 mL of this solu- Kakkonto Extract
tion under the above operating conditions, the resolution
between the peak having the relative retention time of about 葛根湯エキス
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for Change the Origin/limits of content, Identifica-
thin-layer chromatography in 50 mL of methanol. To 2 mL tion (1), (3) to (6) and Assay as follows:
of this solution add 2 mL of the standard solution. When
the procedure is run with 10 mL of this solution under the Kakkonto Extract contains not less than 7 mg and
above operating conditions, the resolution between the not more than 21 mg (for preparation prescribed 3 g
peaks of glycyrrhizic acid and (E )-cinnamaldehyde is not of Ephedra Herb) or not less than 10 mg and not
less than 1.5. more than 30 mg (for preparation prescribed 4 g of
System repeatability: When the test is repeated 6 times Ephedra Herb) of total alkaloids [ephedrine
with 10 mL of the standard solution under the above operat- (C10H15NO: 165.23) and pseudoephedrine (C10H15NO:
ing conditions, the relative standard deviation of the peak 165.23)], not less than 14 mg and not more than 56
area of glycyrrhizic acid is not more than 1.5z. mg (for preparation prescribed 2 g of Peony Root) or
ii) Weigh accurately about 0.5 g of the dry extract (or an not less than 21 mg and not more than 84 mg (for
amount of the viscous extract, equivalent to about 0.5 g of preparation prescribed 3 g of Peony Root) of
the dried substance), add 20 mL of ethyl acetate and 10 mL paeoniflorin (C23H28O11: 480.46), and not less than 15
of water, and shake for 10 minutes. After centrifugation, mg and not more than 45 mg of glycyrrhizic acid
remove the upper layer, add 20 mL of ethyl acetate, proceed (C42H62O16: 822.93), per extract prepared with the
in the same manner as described above, and remove the up- amount specified in the Method of preparation.
per layer. To the resultant aqueous layer add 10 mL of
Identification (1) To 1.0 g of the dry extract (or 3.0 g of
methanol, shake for 30 minutes, centrifuge, and take the su-
the viscous extract) add 10 mL of water, shake, then add 10
pernatant liquid. To the residue add 20 mL of diluted meth-
mL of 1-butanol, shake, centrifuge, and use the supernatant
anol (1 in 2), shake for 5 minutes, centrifuge, and take the
liquid as the sample solution. Separately, dissolve 1 mg of
supernatant liquid. Combine these supernatant liquids, add
Puerarin RS or puerarin for thin-layer chromatography in 1
diluted methanol (1 in 2) to make exactly 50 mL, and use
mL of methanol, and use this solution as the standard solu-
this solution as the sample solution. Separately, weigh accu-
tion. Perform the test with these solutions as directed under
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
termine the water <2.48> by coulometric titration, using 10
sample solution and standard solution on a plate of silica
mg), dissolve in diluted methanol (1 in 2) to make exactly
gel for thin-layer chromatography. Develop the plate with a
100 mL, and use this solution as the standard solution. Per-
mixture of ethyl acetate, methanol and water (20:3:2) to a
form the test with exactly 10 mL each of the sample solution
distance of about 7 cm, and air-dry the plate. Examine
and standard solution as directed under Liquid Chromatog-
under ultraviolet light (main wavelength: 365 nm): one of
raphy <2.01> according to the following conditions, and de-
the several spots obtained from the sample solution has the
termine the peak areas, AT and AS, of glycyrrhizic acid in
same color tone and Rf value with the blue-white fluores-
each solution.
cent spot from the standard solution (Pueraria Root).
Amount (mg) of glycyrrhizic acid (C42H62O16) (3) Perform the test according to the following i) or ii)
= MS × AT/AS × 1/2 (Cinnamon Bark).
i) Put 10 g of the dry extract (or 30 g of the viscous ex-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
tract) in a 300-mL hard-glass flask, add 100 mL of water
lated on the anhydrous basis
and 1 mL of silicone resin, connect an apparatus for essen-
Operating conditions— tial oil determination, and heat to boil under a reflux con-
Proceed as directed in the operating conditions in i). denser. The graduated tube of the apparatus is to be previ-
ously filled with water to the standard line, and 2 mL of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2769

hexane is added to the graduated tube. After heating under tate, methanol and water (20:3:2) to a distance of about 7
reflux for 1 hour, separate the hexane layer, and use the cm, and air-dry the plate. Spray evenly dilute sulfuric acid
layer as the sample solution. Separately, dissolve 1 mg of on the plate, heat the plate at 1059C for 5 minutes, and exa-
(E )-cinnamaldehyde for thin-layer chromatography in 1 mL mine under ultraviolet light (main wavelength: 365 nm): one
of methanol, and use this solution as the standard solution. of the several spots obtained from the sample solution has
Perform the test with these solutions as directed under the same color tone and Rf value with the yellow-green fluo-
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam- rescent spot from the standard solution (Glycyrrhiza).
ple solution and 2 mL of the standard solution on a plate of (6) To 1.0 g of the dry extract (or 3.0 g of the viscous
silica gel for thin-layer chromatography. Develop the plate extract) add 10 mL of water, shake, then add 25 mL of
with a mixture of hexane and ethyl acetate (2:1) to a dis- diethyl ether, shake, and separate the diethyl ether layer.
tance of about 7 cm, and air-dry the plate. Spray evenly 2,4- Evaporate the layer under reduced pressure, add 2 mL of
dinitrophenylhydrazine TS on the plate: one of the several diethyl ether to the residue, and use this solution as the sam-
spots obtained from the sample solution has the same color ple solution. Separately, dissolve 1 mg of [6]-gingerol for
tone and Rf value with the yellow-orange spot from the thin-layer chromatography in 1 mL of methanol, and use
standard solution. this solution as the standard solution. Perform the test with
ii) To 2.0 g of the dry extract (or 6.0 g of the viscous ex- these solutions as directed under Thin-layer Chromatogra-
tract), add 10 mL of water, shake, then add 5 mL of phy <2.03>. Spot 10 mL of the sample solution and 5 mL of
hexane, shake, centrifuge, and use the supernatant liquid as the standard solution on a plate of silica gel for thin-layer
the sample solution. Separately, dissolve 1 mg of (E )-2- chromatography. Develop the plate with a mixture of ethyl
methoxycinnamaldehyde for thin-layer chromatography in acetate and hexane (1:1) to a distance of about 7 cm, and
1 mL of methanol, and use this solution as the standard air-dry the plate. Spray evenly 4-dimethylaminobenzalde-
solution. Perform the test with these solutions as directed hyde TS for spraying on the plate, heat the plate at 1059 C
under Thin-layer Chromatography <2.03>. Spot 40 mL of for 5 minutes, allow to cool, and spray water: one of the
the sample solution and 2 mL of the standard solution on a several spots obtained from the sample solution has the
plate of silica gel for thin-layer chromatography. Develop same color tone and Rf value with the blue-green to grayish
the plate with a mixture of hexane and ethyl acetate (2:1) to green spot from the standard solution (Ginger).
a distance of about 7 cm, and air-dry the plate. Examine
Assay (1) Total alkaloids (ephedrine and pseudoephe-
under ultraviolet light (main wavelength: 365 nm): one of
drine)—Weigh accurately about 0.5 g of the dry extract (or
the several spots obtained from the sample solution has the
an amount of the viscous extract, equivalent to about 0.5 g
same color tone and Rf value with the blue-white fluores-
of the dried substance), add 20 mL of diethyl ether, shake,
cent spot from the standard solution.
then add 3.0 mL of 0.1 mol/L hydrochloric acid TS, and
(4) To 1.0 g of the dry extract (or 3.0 g of the viscous
shake for 10 minutes. After centrifugation, remove the up-
extract) add 10 mL of water, shake, then add 10 mL of 1-
per layer, add 20 mL of diethyl ether, proceed in the same
butanol, shake, centrifuge, and use the supernatant liquid
manner as described above, and remove the upper layer. To
as the sample solution. Separately, dissolve 1 mg of
the aqueous layer add 1.0 mL of ammonia TS and 20 mL of
Paeoniflorin RS or paeoniflorin for thin-layer chromatog-
diethyl ether, shake for 30 minutes, centrifuge, and separate
raphy in 1 mL of methanol, and use this solution as the
the supernatant liquid. In addition, repeat twice in the same
standard solution. Perform the test with these solutions as
manner for the aqueous layer using 1.0 mL of ammonia TS
directed under Thin-layer Chromatography <2.03>. Spot 5
and 20 mL of diethyl ether. Combine all the supernatant liq-
mL each of the sample solution and standard solution on a
uids, evaporate the solvent under reduced pressure, dissolve
plate of silica gel for thin-layer chromatography. Develop
the residue in diluted methanol (1 in 2) to make exactly 50
the plate with a mixture of ethyl acetate, methanol and
mL. Centrifuge this solution, and use the supernatant liquid
water (20:3:2) to a distance of about 7 cm, and air-dry the
as the sample solution. Separately, weigh accurately about
plate. Spray evenly 4-methoxybenzaldehyde-sulfuric acid
10 mg of ephedrine hydrochloride for assay of crude drugs,
TS on the plate, and heat the plate at 1059C for 5 minutes:
previously dried at 1059 C for 3 hours, and dissolve in
one of the several spots obtained from the sample solution
diluted methanol (1 in 2) to make exactly 100 mL. Pipet 10
has the same color tone and Rf value with the purple spot
mL of this solution, add diluted methanol (1 in 2) to make
from the standard solution (Peony Root).
exactly 50 mL, and use this solution as the standard solu-
(5) To 1.0 g of the dry extract (or 3.0 g of the viscous
tion. Perform the test with exactly 10 mL each of the sample
extract) add 10 mL of water, shake, then add 10 mL of 1-
solution and standard solution as directed under Liquid
butanol, shake, centrifuge, and use the supernatant liquid
Chromatography <2.01> according to the following condi-
as the sample solution. Separately, dissolve 1 mg of liquiri-
tions. Determine the peak areas, ATE and ATP, of ephedrine
tin for thin-layer chromatography in 1 mL of methanol, and
and pseudoephedrine obtained with the sample solution,
use this solution as the standard solution. Perform the test
and the peak area, AS, of ephedrine with the standard solu-
with these solutions as directed under Thin-layer Chroma-
tion.
tography <2.03>. Spot1 mL each of the sample solution and
standard solution on a plate of silica gel for thin-layer chro-
matography. Develop the plate with a mixture of ethyl ace-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2770 Crude Drugs and Related Drugs Supplement I, JP XVII

Amount (mg) of total alkaloids [ephedrine (C10H15NO) Operating conditions—


and pseudoephedrine (C10H15NO)] Detector: An ultraviolet absorption photometer (wave-
= MS × (ATE + ATP)/AS × 1/10 × 0.819 length: 232 nm).
Column: A stainless steel column 4.6 mm in inside diame-
MS: Amount (mg) of ephedrine hydrochloride for assay
ter and 15 cm in length, packed with octadecylsilanized
of crude drugs taken
silica gel for liquid chromatography (5 mm in particle diame-
Operating conditions— ter).
Detector: An ultraviolet absorption photometer (wave- Column temperature: A constant temperature of about
length: 210 nm). 209C.
Column: A stainless steel column 4.6 mm in inside diame- Mobile phase: A mixture of water, acetonitrile and phos-
ter and 15 cm in length, packed with octadecylsilanized phoric acid (850:150:1).
silica gel for liquid chromatography (5 mm in particle diame- Flow rate: 1.0 mL per minute (the retention time of
ter). paeoniflorin is about 9 minutes).
Column temperature: A constant temperature of about System suitability—
409C. System performance: Dissolve 1 mg each of Paeoniflorin
Mobile phase: To 5 g of sodium lauryl sulfate add 350 RS and albiflorin in diluted methanol (1 in 2) to make 10
mL of acetonitrile, shake, and add 650 mL of water and 1 mL. When the procedure is run with 10 mL of this solution
mL of phosphoric acid to dissolve sodium lauryl sulfate. under the above operating conditions, albiflorin and
Flow rate: 1.0 mL per minute (the retention time of ephe- paeoniflorin are eluted in this order with the resolution be-
drine is about 27 minutes). tween these peaks being not less than 2.5.
System suitability— System repeatability: When the test is repeated 6 times
System performance: Dissolve 1 mg each of ephedrine hy- with 10 mL of the standard solution under the above operat-
drochloride for assay of crude drugs and pseudoephedrine ing conditions, the relative standard deviation of the peak
hydrochloride in diluted methanol (1 in 2) to make 10 mL. area of paeoniflorin is not more than 1.5z.
When the procedure is run with 10 mL of this solution under (3) Glycyrrhizic acid—Perform the test according to the
the above operating conditions, pseudoephedrine and ephe- following i) or ii).
drine are eluted in this order with the resolution between i) Weigh accurately about 0.5 g of the dry extract (or an
these peaks being not less than 1.5. amount of the viscous extract, equivalent to about 0.5 g of
System repeatability: When the test is repeated 6 times the dried substance), add exactly 50 mL of diluted methanol
with 10 mL of the standard solution under the above operat- (1 in 2), shake for 15 minutes, filter, and use the filtrate as
ing conditions, the relative standard deviation of the peak the sample solution. Separately, weigh accurately about 10
area of ephedrine is not more than 1.5z. mg of Glycyrrhizic Acid RS (separately determine the water
(2) Paeoniflorin—Weigh accurately about 0.5 g of the <2.48> by coulometric titration, using 10 mg), dissolve in
dry extract (or an amount of the viscous extract, equivalent diluted methanol (1 in 2) to make exactly 100 mL, and use
to about 0.5 g of the dried substance), add exactly 50 mL of this solution as the standard solution. Perform the test with
diluted methanol (1 in 2), shake for 15 minutes, and filter. exactly 10 mL each of the sample solution and standard so-
Pipet 5 mL of the filtrate, flow through in a column packed lution as directed under Liquid Chromatography <2.01> ac-
with 2 g of polyamide for column chromatography, elute cording to the following conditions, and determine the peak
with 20 mL of water, add 1 mL of acetic acid (100) and areas, AT and AS, of glycyrrhizic acid in each solution.
water to make exactly 25 mL, and use this solution as the
Amount (mg) of glycyrrhizic acid (C42H62O16)
sample solution. Separately, weigh accurately about 10 mg
= MS × AT/AS × 1/2
of Paeoniflorin RS (separately determine the water <2.48>
by coulometric titration, using 10 mg), and dissolve in MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
diluted methanol (1 in 2) to make exactly 100 mL. Pipet 5 lated on the anhydrous basis
mL of this solution, add diluted methanol (1 in 2) to make
Operating conditions—
exactly 20 mL, and use this solution as the standard solu-
Detector: An ultraviolet absorption photometer (wave-
tion. Perform the test with exactly 10 mL each of the sample
length: 254 nm).
solution and standard solution as directed under Liquid
Column: A stainless steel column 4.6 mm in inside diame-
Chromatography <2.01> according to the following condi-
ter and 15 cm in length, packed with octadecylsilanized
tions, and determine the peak areas, AT and AS, of
silica gel for liquid chromatography (5 mm in particle diame-
paeoniflorin in each solution.
ter).
Amount (mg) of paeoniflorin (C23H28O11) Column temperature: A constant temperature of about
= MS × AT/AS × 5/8 409C.
Mobile phase: Dissolve 3.85 g of ammonium acetate in
MS: Amount (mg) of Paeoniflorin RS taken, calculated
720 mL of water, and add 5 mL of acetic acid (100) and 280
on the anhydrous basis
mL of acetonitrile.
Flow rate: 1.0 mL per minute (the retention time of

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2771

glycyrrhizic acid is about 15 minutes). not less than 1.5.


System suitability—
System performance: Dissolve 5 mg of monoammonium
glycyrrhizinate for resolution check in 20 mL of dilute Kakkontokasenkyushin'i Extract
ethanol. When the procedure is run with 10 mL of this solu-
tion under the above operating conditions, the resolution 葛根湯加川 辛夷エキス
between the peak having the relative retention time of about
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is Change the Origin/limits of content, Identifica-
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for tion (5), (6) and Assay (3) as follows:
thin-layer chromatography in 50 mL of methanol. To 2 mL
of this solution add 2 mL of the standard solution. When Kakkontokasenkyushin'i Extract contains not less
the procedure is run with 10 mL of this solution under the than 9.5 mg and not more than 28.5 mg (for prepara-
above operating conditions, the resolution between the tion prescribed 3 g of Ephedra Herb) or not less than
peaks of glycyrrhizic acid and (E )-cinnamaldehyde is not 13 mg and not more than 39 mg (for preparation
less than 1.5. prescribed 4 g of Ephedra Herb) of total alkaloids
System repeatability: When the test is repeated 6 times [ephedrine (C10H15NO: 165.23) and pseudoephedrine
with 10 mL of the standard solution under the above operat- (C10H15NO: 165.23)], not less than 17 mg and not
ing conditions, the relative standard deviation of the peak more than 51 mg of paeoniflorin (C23H28O11: 480.46),
area of glycyrrhizic acid is not more than 1.5z. not less than 14 mg and not more than 42 mg of
ii) Weigh accurately about 0.5 g of the dry extract (or an glycyrrhizic acid (C42H62O16: 822.93), and not less
amount of the viscous extract, equivalent to about 0.5 g of than 1.5 mg and not more than 6 mg (for preparation
the dried substance), add 20 mL of ethyl acetate and 10 mL prescribed 2 g of Magnolia Flower) or not less than 2
of water, and shake for 10 minutes. After centrifugation, mg and not more than 8 mg (for preparation
remove the upper layer, add 20 mL of ethyl acetate, proceed prescribed 3 g of Magnolia Flower) of magnoflorine
in the same manner as described above, and remove the up- [as magnoflorine iodide (C20H24INO4: 469.31)], per
per layer. To the resultant aqueous layer add 10 mL of extract prepared with the amount specified in the
methanol, shake for 30 minutes, centrifuge, and take the su- Method of preparation.
pernatant liquid. To the residue add 20 mL of diluted meth-
Identification
anol (1 in 2), shake for 5 minutes, centrifuge, and take the
(5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
supernatant liquid. Combine these supernatant liquids, add
extract) with 10 mL of water, add 10 mL of 1-butanol,
diluted methanol (1 in 2) to make exactly 50 mL, and use
shake, centrifuge, and use the supernatant liquid as the
this solution as the sample solution. Separately, weigh accu-
sample solution. Separately, dissolve 1 mg of liquiritin for
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
thin-layer chromatography in 1 mL of methanol, and use
termine the water <2.48> by coulometric titration, using 10
this solution as the standard solution. Perform the test with
mg), dissolve in diluted methanol (1 in 2) to make exactly
these solutions as directed under Thin-layer Chromatogra-
100 mL, and use this solution as the standard solution. Per-
phy <2.03>. Spot 1 mL each of the sample solution and
form the test with exactly 10 mL each of the sample solution
standard solution on a plate of silica gel for thin-layer chro-
and standard solution as directed under Liquid Chromatog-
matography. Develop the plate with a mixture of ethyl ace-
raphy <2.01> according to the following conditions, and de-
tate, methanol and water (20:3:2) to a distance of about 7
termine the peak areas, AT and AS, of glycyrrhizic acid in
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
each solution.
on the plate, heat the plate at 1059C for 5 minutes, and exa-
Amount (mg) of glycyrrhizic acid (C42H62O16) mine under ultraviolet light (main wavelength: 365 nm): one
= MS × AT/AS × 1/2 of the several spots obtained from the sample solution has
the same color tone and Rf value with the yellow-green fluo-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
rescent spot from the standard solution (Glycyrrhiza).
lated on the anhydrous basis
(6) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
Operating conditions— extract) with 10 mL of water, add 25 mL of diethyl ether,
Proceed as directed in the operating conditions in i). and shake. Separate the diethyl ether layer, evaporate the
System suitability— layer under reduced pressure, add 2 mL of diethyl ether to
System repeatability: Proceed as directed in the system the residue, and use this solution as the sample solution.
suitability in i). Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro-
System performance: Dissolve 5 mg of monoammonium matography in 1 mL of methanol, and use this solution as
glycyrrhizinate for resolution check in 20 mL of dilute the standard solution. Perform the test with these solutions
ethanol. When the procedure is run with 10 mL of this solu- as directed under Thin-layer Chromatography <2.03>. Spot
tion under the above operating conditions, the resolution 10 mL of the sample solution and 5 mL of the standard solu-
between the peak having the relative retention time of about tion on a plate of silica gel for thin-layer chromatography.
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is Develop the plate with a mixture of ethyl acetate and hexane

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2772 Crude Drugs and Related Drugs Supplement I, JP XVII

(1:1) to a distance of about 7 cm, and air-dry the plate. with 10 mL of the standard solution under the above operat-
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying ing conditions, the relative standard deviation of the peak
on the plate, heat the plate at 1059C for 5 minutes, allow to area of glycyrrhizic acid is not more than 1.5z.
cool, and spray water: one of the several spots obtained ii) Weigh accurately about 0.5 g of the dry extract (or an
from the sample solution has the same color tone and Rf amount of the viscous extract, equivalent to about 0.5 g of
value with the blue-green to grayish green spot from the the dried substance), add 20 mL of ethyl acetate and 10 mL
standard solution (Ginger). of water, and shake for 10 minutes. After centrifugation,
remove the upper layer, add 20 mL of ethyl acetate, proceed
Assay
in the same manner as described above, and remove the
(3) Glycyrrhizic acid—Perform the test according to the
upper layer. To the resultant aqueous layer add 10 mL of
following i) or ii).
methanol, shake for 30 minutes, centrifuge, and take the su-
i) Weigh accurately about 0.5 g of the dry extract (or an
pernatant liquid. To the residue add 20 mL of diluted meth-
amount of the viscous extract, equivalent to about 0.5 g of
anol (1 in 2), shake for 5 minutes, centrifuge, and take the
the dried substance), add exactly 50 mL of diluted methanol
supernatant liquid. Combine these supernatant liquids, add
(1 in 2), shake for 15 minutes, filter, and use the filtrate as
diluted methanol (1 in 2) to make exactly 50 mL, and use
the sample solution. Separately, weigh accurately about 10
this solution as the sample solution. Separately, weigh accu-
mg of Glycyrrhizic Acid RS (separately determine the water
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
<2.48> by coulometric titration, using 10 mg), dissolve in
termine the water <2.48> by coulometric titration, using 10
diluted methanol (1 in 2) to make exactly 100 mL, and use
mg), dissolve in diluted methanol (1 in 2) to make exactly
this solution as the standard solution. Perform the test with
100 mL, and use this solution as the standard solution. Per-
exactly 10 mL each of the sample solution and standard so-
form the test with exactly 10 mL each of the sample solution
lution as directed under Liquid Chromatography <2.01> ac-
and standard solution as directed under Liquid Chromatog-
cording to the following conditions, and determine the peak
raphy <2.01> according to the following conditions, and de-
areas, AT and AS, of glycyrrhizic acid in each solution.
termine the peak areas, AT and AS, of glycyrrhizic acid in
Amount (mg) of glycyrrhizic acid (C42H62O16) each solution.
= MS × AT/AS × 1/2
Amount (mg) of glycyrrhizic acid (C42H62O16)
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- = MS × AT/AS × 1/2
lated on the anhydrous basis
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
Operating conditions— lated on the anhydrous basis
Detector: An ultraviolet absorption photometer (wave-
Operating conditions—
length: 254 nm).
Proceed as directed in the operating conditions in i).
Column: A stainless steel column 4.6 mm in inside diame-
System suitability—
ter and 15 cm in length, packed with octadecylsilanized
System repeatability: Proceed as directed in the system
silica gel for liquid chromatography (5 mm in particle diame-
suitability in i).
ter).
System performance: Dissolve 5 mg of monoammonium
Column temperature: A constant temperature of about
glycyrrhizinate for resolution check in 20 mL of dilute
409C.
ethanol. When the procedure is run with 10 mL of this solu-
Mobile phase: Dissolve 3.85 g of ammonium acetate in
tion under the above operating conditions, the resolution
720 mL of water, and add 5 mL of acetic acid (100) and 280
between the peak having the relative retention time of about
mL of acetonitrile.
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
Flow rate: 1.0 mL per minute (the retention time of
not less than 1.5.
glycyrrhizic acid is about 15 minutes).
System suitability—
System performance: Dissolve 5 mg of monoammonium
glycyrrhizinate for resolution check in 20 mL of dilute Kamikihito Extract
ethanol. When the procedure is run with 10 mL of this solu-
加味帰脾湯エキス
tion under the above operating conditions, the resolution
between the peak having the relative retention time of about
Change the Origin/limits of content, Identifica-
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
tion (9) and Assay (3) as follows:
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for
thin-layer chromatography in 50 mL of methanol. To 2 mL
Kamikihito Extract contains not less than 0.8 mg
of this solution add 2 mL of the standard solution. When
and not more than 3.2 mg of saikosaponin b2, not less
the procedure is run with 10 mL of this solution under the
than 27 mg and not more than 81 mg of geniposide,
above operating conditions, the resolution between the
and not less than 6 mg and not more than 18 mg of
peaks of glycyrrhizic acid and (E )-cinnamaldehyde is not
glycyrrhizic acid (C42H62O16: 822.93), per extract pre-
less than 1.5.
pared with the amount specified in the Method of
System repeatability: When the test is repeated 6 times

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2773

preparation. Mobile phase: Dissolve 3.85 g of ammonium acetate in


720 mL of water, and add 5 mL of acetic acid (100) and 280
Identification
mL of acetonitrile.
(9) To 1.0 g of the dry extract (or 3.0 g of the viscous
Flow rate: 1.0 mL per minute (the retention time of
extract) add 10 mL of water, shake, add 10 mL of 1-
glycyrrhizic acid is about 15 minutes).
butanol, shake, centrifuge, and use the supernatant liquid
System suitability—
as the sample solution. Separately, dissolve 1 mg of liquiri-
System performance: Dissolve 5 mg of monoammonium
tin for thin-layer chromatography in 1 mL of methanol, and
glycyrrhizinate for resolution check in 20 mL of dilute
use this solution as the standard solution. Perform the test
ethanol. When the procedure is run with 10 mL of this solu-
with these solutions as directed under Thin-layer Chroma-
tion under the above operating conditions, the resolution
tography <2.03>. Spot 1 mL each of the sample solution and
between the peak having the relative retention time of about
standard solution on a plate of silica gel for thin-layer chro-
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
matography. Develop the plate with a mixture of ethyl ace-
not less than 1.5.
tate, methanol and water (20:3:2) to a distance of about 7
System repeatability: When the test is repeated 6 times
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
with 10 mL of the standard solution under the above operat-
on the plate, heat the plate at 1059C for 5 minutes, and exa-
ing conditions, the relative standard deviation of the peak
mine under ultraviolet light (main wavelength: 365 nm): one
area of glycyrrhizic acid is not more than 1.5z.
of the several spots obtained from the sample solution has
the same color tone and Rf value with the yellow-green fluo-
rescent spot from the standard solution (Glycyrrhiza).
Kamishoyosan Extract
Assay
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of 加味逍遙散エキス
the dry extract (or an amount of the viscous extract, equiva-
lent to about 0.5 g of the dried substance), add 20 mL of Change the Origin/limits of content, Identifica-
diethyl ether and 10 mL of water, and shake for 10 minutes. tion and Assay (3) as follows:
After centrifugation, remove the upper layer, add 20 mL of
diethyl ether, proceed in the same manner as described Kamishoyosan Extract contains not less than 28 mg
above, and remove the upper layer. To the resultant aque- and not more than 84 mg of paeoniflorin (C23H28O11:
ous layer add 10 mL of methanol, shake for 30 minutes, 480.46), not less than 25 mg and not more than 75 mg
centrifuge, and take the supernatant liquid. To the residue of geniposide, and not less than 10 mg and not more
add 20 mL of diluted methanol (1 in 2), shake for 5 than 30 mg (for preparation prescribed 1.5 g of
minutes, centrifuge, and take the supernatant liquid. Com- Glycyrrhiza) or not less than 13 mg and not more than
bine these supernatant liquids, add diluted methanol (1 in 2) 39 mg (for preparation prescribed 2 g of Glycyrrhiza)
to make exactly 50 mL, and use this solution as the sample of glycyrrhizic acid (C42H62O16: 822.93), per extract
solution. Separately, weigh accurately about 10 mg of prepared with the amount specified in the Method of
Glycyrrhizic Acid RS (separately determine the water <2.48> preparation.
by coulometric titration, using 10 mg), dissolve in diluted
Identification (1) To 2.0 g of the dry extract (or 6.0 g of
methanol (1 in 2) to make exactly 100 mL, and use this solu-
the viscous extract) add 10 mL of water, shake, then add 5
tion as the standard solution. Perform the test with exactly
mL of diethyl ether, shake, centrifuge, and use the superna-
10 mL each of the sample solution and standard solution as
tant liquid as the sample solution. Separately, dissolve 1 mg
directed under Liquid Chromatography <2.01> according to
of (Z )-ligustilide for thin-layer chromatography in 10 mL of
the following conditions, and determine the peak areas, AT
methanol, and use this solution as the standard solution.
and AS, of glycyrrhizic acid in each solution.
Perform the test with these solutions as directed under
Amount (mg) of glycyrrhizic acid (C42H62O16) Thin-layer Chromatography <2.03>. Spot 10 mL each of the
= MS × AT/AS × 1/2 sample solution and standard solution on a plate of silica
gel for thin-layer chromatography. Develop the plate with a
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
mixture of ethyl acetate and hexane (1:1) to a distance of
lated on the anhydrous basis
about 7 cm, and air-dry the plate. Examine under ultravio-
Operating conditions— let light (main wavelength: 365 nm): one of the several spots
Detector: An ultraviolet absorption photometer (wave- obtained from the sample solution has the same color tone
length: 254 nm). and Rf value with the blue-white fluorescent spot from the
Column: A stainless steel column 4.6 mm in inside diame- standard solution (Japanese Angelica Root).
ter and 15 cm in length, packed with octadecylsilanized (2) To 1.0 g of the dry extract (or 3.0 g of the viscous
silica gel for liquid chromatography (5 mm in particle diame- extract) add 10 mL of water, shake, then add 10 mL of
ter). methanol, shake, centrifuge, and use the supernatant liquid
Column temperature: A constant temperature of about as the sample solution. Separately, dissolve 1 mg of albiflo-
409C. rin in 1 mL of methanol, and use this solution as the stand-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2774 Crude Drugs and Related Drugs Supplement I, JP XVII

ard solution. Perform the test with these solutions as di- silica gel for thin-layer chromatography. Develop the plate
rected under Thin-layer Chromatography <2.03>. Spot 10 with a mixture of ethyl acetate, ethanol (99.5) and water
mL each of the sample solution and standard solution on a (8:2:1) to a distance of about 7 cm, and air-dry the plate.
plate of silica gel for thin-layer chromatography. Develop Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
the plate with a mixture of ethyl acetate, methanol and am- on the plate, heat the plate at 1059C for 5 minutes, and exa-
monia solution (28) (6:3:2) to a distance of about 10 cm, mine under ultraviolet light (main wavelength: 365 nm): one
and air-dry the plate. Spray evenly 4-methoxybenzaldehyde- of the several spots obtained from the sample solution has
sulfuric acid TS on the plate, heat the plate at 1059C for 5 the same color tone and Rf value with the yellow fluorescent
minutes, allow to cool for more than 30 minutes, and exa- spot from the standard solution (Bupleurum Root).
mine under ultraviolet light (main wavelength: 365 nm): one (6) To 2.0 g of the dry extract (or 6.0 g of the viscous
of the several spots obtained from the sample solution has extract) add 10 mL of water, shake, then add 15 mL of
the same color tone and Rf value with the orange fluores- diethyl ether, and shake. Separate the diethyl ether layer,
cent spot from the standard solution (Peony Root). evaporate the layer under reduced pressure, add 1 mL of
(3) For preparation prescribed Atractylodes Rhizome— diethyl ether to the residue, and use this solution as the sam-
To 2.0 g of the dry extract (or 6.0 g of the viscous extract) ple solution. Separately, dissolve 1 mg of paeonol for thin-
add 10 mL of water, shake, then add 5 mL of diethyl ether, layer chromatography in 1 mL of methanol, and use this so-
shake, centrifuge, and use the supernatant liquid as the lution as the standard solution. Perform the test with these
sample solution. Separately, dissolve 1 mg of atrac- solutions as directed under Thin-layer Chromatography
tylenolide III for thin-layer chromatography in 1 mL of <2.03>. Spot 10 mL each of the sample solution and standard
methanol, and use this solution as the standard solution. solution on a plate of silica gel for thin-layer chromatogra-
Perform the test with these solutions as directed under phy. Develop the plate with a mixture of hexane and diethyl
Thin-layer Chromatography <2.03>. Spot 10 mL each of the ether (5:3) to a distance of about 7 cm, and air-dry the
sample solution and standard solution on a plate of silica plate. Spray evenly 4-methoxybenzaldehyde-sulfuric acid
gel for thin-layer chromatography. Develop the plate with a TS on the plate, and heat the plate at 1059C for 5 minutes:
mixture of ethyl acetate and hexane (1:1) to a distance of one of the several spots obtained from the sample solution
about 7 cm, and air-dry the plate. Spray evenly 1-naphthol- has the same color tone and Rf value with the orange spot
sulfuric acid TS on the plate, heat the plate at 1059C for from the standard solution (Moutan Bark).
5 minutes, and allow to cool: one of the several spots (7) To 2.0 g of the dry extract (or 6.0 g of the viscous
obtained from the sample solution has the same color tone extract) add 10 mL of water, shake, then add 5 mL of 1-
and Rf value with the red spot from the standard solution butanol, shake, centrifuge, and use the supernatant liquid
(Atractylodes Rhizome). as the sample solution. Separately, dissolve 1 mg of genipo-
(4) For preparation prescribed Atractylodes Lancea side for thin-layer chromatography in 1 mL of methanol,
Rhizome—To 2.0 g of the dry extract (or 6.0 g of the vis- and use this solution as the standard solution. Perform the
cous extract) add 10 mL of water, shake, then add 25 mL of test with these solutions as directed under Thin-layer Chro-
hexane, and shake. Separate the hexane layer, evaporate the matography <2.03>. Spot 10 mL each of the sample solution
layer under reduced pressure, add 2 mL of hexane to the and standard solution on a plate of silica gel for thin-layer
residue, and use this solution as the sample solution. Per- chromatography. Develop the plate with a mixture of ethyl
form the test with the sample solution as directed under acetate, methanol and ammonia solution (28) (6:3:2) to a
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam- distance of about 7 cm, and air-dry the plate. Spray evenly
ple solution on a plate of silica gel with fluorescent indicator 4-methoxybenzaldehyde-sulfric acid TS on the plate, and
for thin-layer chromatography. Develop the plate with a heat the plate at 1059C for 5 minutes: one of the several
mixture of hexane and acetone (7:1) to a distance of about 7 spots obtained from the sample solution has the same color
cm, and air-dry the plate. Examine under ultraviolet light tone and Rf value with the purple spot from the standard
(main wavelength: 254 nm): a dark purple spot is observed solution (Gardenia Fruit).
at an Rf value of about 0.5. The spot shows a greenish (8) To 2.0 g of the dry extract (or 6.0 g of the viscous
brown color after being sprayed evenly 4-dimethylamino- extract) add 10 mL of water, shake, then add 5 mL of 1-
benzaldehyde TS for spraying, heated at 1059C for 5 butanol, shake, centrifuge, and use the supernatant liquid
minutes, and allowed to cool (Atractylodes Lancea Rhi- as the sample solution. Separately, dissolve 1 mg of liquiri-
zome). tin for thin-layer chromatography in 1 mL of methanol, and
(5) To 2.0 g of the dry extract (or 6.0 g of the viscous use this solution as the standard solution. Perform the test
extract) add 10 mL of sodium hydroxide TS, shake, then with these solutions as directed under Thin-layer Chroma-
add 5 mL of 1-butanol, shake, centrifuge, and use the su- tography <2.03>. Spot 1 mL each of the sample solution and
pernatant liquid as the sample solution. Separately, dissolve standard solution on a plate of silica gel for thin-layer chro-
1 mg of saikosaponin b2 for thin-layer chromatography in 1 matography. Develop the plate with a mixture of ethyl ace-
mL of methanol, and use this solution as the standard solu- tate, methanol and water (20:3:2) to a distance of about 7
tion. Perform the test with these solutions as directed under cm, and air-dry the plate. Spray evenly dilute sulfuric acid
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam- on the plate, heat the plate at 1059C for 5 minutes, and exa-
ple solution and 2 mL of the standard solution on a plate of mine under ultraviolet light (main wavelength: 365 nm): one

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2775

of the several spots obtained from the sample solution has tion as the standard solution. Perform the test with exactly
the same color tone and Rf value with the yellow-green fluo- 10 mL each of the sample solution and standard solution as
rescent spot from the standard solution (Glycyrrhiza). directed under Liquid Chromatography <2.01> according to
(9) To 2.0 g of the dry extract (or 6.0 g of the viscous the following conditions, and determine the peak areas, AT
extract) add 10 mL of water, shake, then add 5 mL of and AS, of glycyrrhizic acid in each solution.
diethyl ether, shake, centrifuge, and use the supernatant liq-
Amount (mg) of glycyrrhizic acid (C42H62O16)
uid as the sample solution. Separately, dissolve 1 mg of [6]-
= MS × AT/AS × 1/2
gingerol for thin-layer chromatography in 1 mL of metha-
nol, and use this solution as the standard solution. Perform MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
the test with these solutions as directed under Thin-layer lated on the anhydrous basis
Chromatography <2.03>. Spot 10 mL each of the sample so-
Operating conditions—
lution and standard solution on a plate of silica gel for thin-
Detector: An ultraviolet absorption photometer (wave-
layer chromatography. Develop the plate with a mixture of
length: 254 nm).
ethyl acetate and hexane (1:1) to a distance of about 7 cm,
Column: A stainless steel column 4.6 mm in inside diame-
and air-dry the plate. Spray evenly 4-dimethylaminobenzal-
ter and 15 cm in length, packed with octadecylsilanized
dehyde TS for spraying on the plate, heat the plate at 1059C
silica gel for liquid chromatography (5 mm in particle diame-
for 5 minutes, allow to cool, and spray water: one of the
ter).
several spots obtained from the sample solution has the
Column temperature: A constant temperature of about
same color tone and Rf value with the blue-green to grayish
409C.
green spot from the standard solution (Ginger).
Mobile phase: Dissolve 3.85 g of ammonium acetate in
(10) To 2.0 g of the dry extract (or 6.0 g of the viscous
720 mL of water, and add 5 mL of acetic acid (100) and 280
extract) add 10 mL of diluted phosphoric acid (1 in 30),
mL of acetonitrile.
shake, then add 15 mL of ethyl acetate, shake, centrifuge,
Flow rate: 1.0 mL per minute (the retention time of
and use the supernatant liquid as the sample solution. Sepa-
glycyrrhizic acid is about 15 minutes).
rately, shake 0.2 g of pulverized mentha herb with 10 mL of
System suitability—
diluted phosphoric acid (1 in 30), add 15 mL of ethyl ace-
System performance: Dissolve 5 mg of monoammonium
tate, shake, centrifuge, and use the supernatant liquid as the
glycyrrhizinate for resolution check in 20 mL of dilute
standard solution. Perform the test with these solutions as
ethanol. When the procedure is run with 10 mL of this solu-
directed under Thin-layer Chromatography <2.03>. Spot 20
tion under the above operating conditions, the resolution
mL each of the sample solution and standard solution on a
between the peak having the relative retention time of about
plate of silica gel for thin-layer chromatography. Develop
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
the plate with a mixture of acetone, ethyl acetate, water and
not less than 1.5.
acetic acid (100) (10:10:3:1) to a distance of about 7 cm, and
System repeatability: When the test is repeated 6 times
air-dry the plate. Spray evenly 2,6-dibromo-N-chloro-1,4-
with 10 mL of the standard solution under the above operat-
benzoquinone monoimine TS on the plate, heat the plate at
ing conditions, the relative standard deviation of the peak
1059C for 5 minutes: one of the several spots obtained from
area of glycyrrhizic acid is not more than 1.5z.
the sample solution has the same color tone and Rf value
with the red-brown spot (around Rf value 0.4) from the
standard solution (Mentha Herb).
Keishibukuryogan Extract
Assay
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of 桂枝茯苓丸エキス
the dry extract (or an amount of the viscous extract, equiva-
lent to about 0.5 g of the dried substance), add 20 mL of Change the Identification as follows:
ethyl acetate and 10 mL of water, and shake for 10 minutes.
Identification (1) Shake 1.0 g of the dry extract (or 3.0 g
After centrifugation, remove the upper layer, add 20 mL of
of the viscous extract) with 10 mL of water, add 25 mL of
ethyl acetate, proceed in the same manner as described
diethyl ether, and shake. Separate the diethyl ether layer,
above, and remove the upper layer. To the resultant aque-
evaporate the layer under reduced pressure, dissolve the
ous layer add 10 mL of methanol, shake for 30 minutes,
residue in 2 mL of diethyl ether, and use this solution as the
centrifuge, and take the supernatant liquid. To the residue
sample solution. Separately, dissolve 1 mg of (E )-cinnamic
add 20 mL of diluted methanol (1 in 2), shake for 5
acid for thin-layer chromatography in 1 mL of methanol,
minutes, centrifuge, and take the supernatant liquid. Com-
and use this solution as the standard solution. Perform the
bine these supernatant liquids, add diluted methanol (1 in 2)
test with these solutions as directed under Thin-layer Chro-
to make exactly 50 mL, and use this solution as the sample
matography <2.03>. Spot 5 mL each of the sample solution
solution. Separately, weigh accurately about 10 mg of
and standard solution on a plate of silica gel with fluores-
Glycyrrhizic Acid RS (separately determine the water <2.48>
cent indicator for thin-layer chromatography. Develop the
by coulometric titration, using 10 mg), dissolve in diluted
plate with a mixture of hexane, ethyl acetate, formic acid
methanol (1 in 2) to make exactly 100 mL, and use this solu-
and water (60:40:4:1) to a distance of about 7 cm, and air-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2776 Crude Drugs and Related Drugs Supplement I, JP XVII

dry the plate. Examine under ultraviolet light (main wave-


length: 254 nm): one of the several spots obtained from the Maoto Extract
sample solution has the same color tone and Rf value with
the blue-purple spot from the standard solution (Cinnamon 麻黄湯エキス
Bark).
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous Change the Origin/limits of content, Identifica-
extract) with 10 mL of water, add 25 mL of diethyl ether, tion (4) and Assay (3) as follows:
and shake. Separate the diethyl ether layer, evaporate the
layer under reduced pressure, dissolve the residue in 2 mL Maoto Extract contains not less than 15 mg and
of diethyl ether, and use this solution as the sample solu- not more than 45 mg of total alkaloids [ephedrine
tion. Separately, dissolve 1 mg of paeonol for thin-layer (C10H15NO: 165.23) and pseudoephedrine (C10H15NO:
chromatography in 1 mL of methanol, and use this solution 165.23)], not less than 48 mg and not more than 192
as the standard solution. Perform the test with these solu- mg of amygdalin, and not less than 11 mg and not
tions as directed under Thin-layer Chromatography <2.03>. more than 33 mg of glycyrrhizic acid (C42H62O16:
Spot 10 mL each of the sample solution and standard solu- 822.93), per extract prepared with the amount speci-
tion on a plate of silica gel for thin-layer chromatography. fied in the Method of preparation.
Develop the plate with a mixture of hexane and diethyl ether
Identification
(5:3) to a distance of about 7 cm, and air-dry the plate.
(4) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
Spray evenly 4-methoxybenzaldehyde-sulfuric acid TS on
extract) with 10 mL of water, add 10 mL of 1-butanol,
the plate, and heat the plate at 1059C for 5 minutes: one of
shake, centrifuge, and use the supernatant liquid as the
the several spots obtained from the sample solution has the
sample solution. Separately, dissolve 1 mg of liquiritin for
same color tone and Rf value with the orange spot from the
thin-layer chromatography in 1 mL of methanol, and use
standard solution (Moutan Bark).
this solution as the standard solution. Perform the test with
(3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
these solutions as directed under Thin-layer Chromatogra-
extract) with 10 mL of methanol, filter, and use the filtrate
phy <2.03>. Spot 1 mL each of the sample solution and
as the sample solution. Separately, dissolve 2 mg of amyg-
standard solution on a plate of silica gel for thin-layer chro-
dalin for thin-layer chromatography in 1 mL of methanol,
matography. Develop the plate with a mixture of ethyl ace-
and use this solution as the standard solution. Perform the
tate, methanol and water (20:3:2) to a distance of about 7
test with these solutions as directed under Thin-layer Chro-
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
matography <2.03>. Spot 5 mL each of the sample solution
on the plate, heat the plate at 1059C for 5 minutes, and exa-
and standard solution on a plate of silica gel for thin-layer
mine under ultraviolet light (main wavelength: 365 nm): one
chromatography. Develop the plate with a mixture of 1-
of the several spots obtained from the sample solution has
propanol, ethyl acetate and water (4:4:3) to a distance of
the same color tone and Rf value with the yellow-green fluo-
about 7 cm, and air-dry the plate. Spray evenly 4-methox-
rescent spot from the standard solution (Glycyrrhiza).
ybenzaldehyde-sulfuric acid TS on the plate, and heat the
plate at 1059C for 10 minutes: one of the several spots Assay
obtained from the sample solution has the same color tone (3) Glycyrrhizic acid—Perform the test according to the
and Rf value with the green-brown spot from the standard following i) or ii).
solution (Peach Kernel). i) Weigh accurately about 0.5 g of the dry extract (or an
(4) Shake 1.0 g of the dry extract (or 3.0 g of the viscous amount of the viscous extract, equivalent to about 0.5 g of
extract) with 10 mL of water, add 10 mL of methanol, the dried substance), add exactly 50 mL of diluted methanol
shake, centrifuge, and use the supernatant liquid as the (1 in 2), shake for 15 minutes, filter, and use the filtrate as
sample solution. Separately, dissolve 1 mg of albiflorin in 1 the sample solution. Separately, weigh accurately about 10
mL of methanol, and use this solution as the standard solu- mg of Glycyrrhizic Acid RS (separately determine the water
tion. Perform the test with these solutions as directed under <2.48> by coulometric titration, using 10 mg), dissolve in
Thin-layer Chromatography <2.03>. Spot 5 mL each of the diluted methanol (1 in 2) to make exactly 100 mL, and use
sample solution and standard solution on a plate of silica this solution as the standard solution. Perform the test with
gel for thin-layer chromatography. Develop the plate with a exactly 10 mL each of the sample solution and standard so-
mixture of ethyl acetate, methanol and ammonia solution lution as directed under Liquid Chromatography <2.01> ac-
(28) (6:3:2) to a distance of about 10 cm, and air-dry the cording to the following conditions, and determine the peak
plate. Spray evenly 4-methoxybenzaldehyde-sulfuric acid areas, AT and AS, of glycyrrhizic acid in each solution.
TS on the plate, heat the plate at 1059 C for 5 minutes, allow
Amount (mg) of glycyrrhizic acid (C42H62O16)
to cool for more than 30 minutes, and examine under ultra-
= MS × AT/AS × 1/2
violet light (main wavelength: 365 nm): one of the several
spots obtained from the sample solution has the same color MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
tone and Rf value with the orange fluorescent spot from the lated on the anhydrous basis
standard solution (Peony Root).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2777

Operating conditions— lated on the anhydrous basis


Detector: An ultraviolet absorption photometer (wave-
Operating conditions—
length: 254 nm).
Proceed as directed in the operating conditions in i).
Column: A stainless steel column 4.6 mm in inside diame-
System suitability—
ter and 15 cm in length, packed with octadecylsilanized
System repeatability: Proceed as directed in the system
silica gel for liquid chromatography (5 mm in particle diame-
suitability in i).
ter).
System performance: Dissolve 5 mg of monoammonium
Column temperature: A constant temperature of about
glycyrrhizinate for resolution check in 20 mL of dilute
409C.
ethanol. When the procedure is run with 10 mL of this solu-
Mobile phase: Dissolve 3.85 g of ammonium acetate in
tion under the above operating conditions, the resolution
720 mL of water, and add 5 mL of acetic acid (100) and 280
between the peak having the relative retention time of about
mL of acetonitrile.
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
Flow rate: 1.0 mL per minute (the retention time of
not less than 1.5.
glycyrrhizic acid is about 15 minutes).
System suitability—
System performance: Dissolve 5 mg of monoammonium
glycyrrhizinate for resolution check in 20 mL of dilute Mukoi-Daikenchuto Extract
ethanol. When the procedure is run with 10 mL of this solu-
無コウイ大建中湯エキス
tion under the above operating conditions, the resolution
between the peak having the relative retention time of about
Change the Identification as follows:
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for Identification (1) Shake 2.0 g of Mukoi-Daikenchuto
thin-layer chromatography in 50 mL of methanol. To 2 mL Extract with 10 mL of water, add 10 mL of diethyl ether,
of this solution add 2 mL of the standard solution. When shake, centrifuge, and use the supernatant liquid as the
the procedure is run with 10 mL of this solution under the sample solution. Separately, shake 2.0 g of pulverized
above operating conditions, the resolution between the japanese zanthoxylum peel with 10 mL of water, add 5 mL
peaks of glycyrrhizic acid and (E )-cinnamaldehyde is not of diethyl ether, shake, centrifuge, and use the supernatant
less than 1.5. liquid as the standard solution. Perform the test with these
System repeatability: When the test is repeated 6 times solutions as directed under Thin-layer Chromatography
with 10 mL of the standard solution under the above operat- <2.03>. Spot 10 mL each of the sample solution and standard
ing conditions, the relative standard deviation of the peak solution on a plate of silica gel with fluorescent indicator
area of glycyrrhizic acid is not more than 1.5z. for thin-layer chromatography. Develop the plate with a
ii) Weigh accurately about 0.5 g of the dry extract (or an mixture of ethyl acetate, hexane, methanol and acetic acid
amount of the viscous extract, equivalent to about 0.5 g of (100) (20:20:1:1) to a distance of about 7 cm, and air-dry
the dried substance), add 20 mL of ethyl acetate and 10 mL the plate. Examine under ultraviolet light (main wavelength:
of water, and shake for 10 minutes. After centrifugation, 254 nm): one of the several spots obtained from the sample
remove the upper layer, add 20 mL of ethyl acetate, proceed solution has the same color tone and Rf value with the dark
in the same manner as described above, and remove the up- purple spot (Rf value: about 0.3) from the standard solution
per layer. To the resultant aqueous layer add 10 mL of (Japanese Zanthoxylum Peel).
methanol, shake for 30 minutes, centrifuge, and take the su- (2) Shake 2.0 g of Mukoi-Daikenchuto Extract with 10
pernatant liquid. To the residue add 20 mL of diluted meth- mL of water, add 10 mL of 1-butanol, shake, centrifuge,
anol (1 in 2), shake for 5 minutes, centrifuge, and take the and use the supernatant liquid as the sample solution. Sepa-
supernatant liquid. Combine these supernatant liquids, add rately, dissolve 1 mg of Ginsenoside Rb1 RS or ginsenoside
diluted methanol (1 in 2) to make exactly 50 mL, and use Rb1 for thin-layer chromatography in 1 mL of methanol,
this solution as the sample solution. Separately, weigh accu- and use this solution as the standard solution. Perform the
rately about 10 mg of Glycyrrhizic Acid RS (separately de- test with these solutions as directed under Thin-layer Chro-
termine the water <2.48> by coulometric titration, using 10 matography <2.03>. Spot 10 mL of the sample solution and 2
mg), dissolve in diluted methanol (1 in 2) to make exactly mL of the standard solution on a plate of silica gel for thin-
100 mL, and use this solution as the standard solution. Per- layer chromatography. Develop the plate with a mixture of
form the test with exactly 10 mL each of the sample solution ethyl acetate, 1-propanol, water and acetic acid (100)
and standard solution as directed under Liquid Chromatog- (7:5:4:1) to a distance of about 7 cm, and air-dry the plate.
raphy <2.01> according to the following conditions, and de- Spray evenly vanillin-sulfuric acid-ethanol TS for spraying
termine the peak areas, AT and AS, of glycyrrhizic acid in on the plate, heat the plate at 1059C for 5 minutes, and
each solution. allow to cool: one of the several spots obtained from the
sample solution has the same color tone and Rf value with
Amount (mg) of glycyrrhizic acid (C42H62O16)
the blue-purple spot from the standard solution (Ginseng).
= MS × AT/AS × 1/2
(3) Shake 2.0 g of Mukoi-Daikenchuto Extract with 10
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- mL of water, add 10 mL of diethyl ether, shake, centrifuge,

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2778 Crude Drugs and Related Drugs Supplement I, JP XVII

and use the supernatant liquid as the sample solution. Sepa- (3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
rately, dissolve 1 mg of [6]-shogaol for thin-layer chroma- extract) with 10 mL of water, then add 10 mL of diethyl
tography in 1 mL of methanol, and use this solution as the ether, shake, centrifuge, and use the supernatant liquid as
standard solution. Perform the test with these solutions as the sample solution. Separately, dissolve 1 mg of wogonin
directed under Thin-layer Chromatography <2.03>. Spot 10 for thin-layer chromatography in 1 mL of methanol, and
mL of the sample solution and 2 mL of the standard solution use this solution as the standard solution. Perform the test
on a plate of silica gel for thin-layer chromatography. De- with these solutions as directed under Thin-layer Chroma-
velop the plate with a mixture of ethyl acetate and hexane tography <2.03>. Spot 20 mL of the sample solution and 5
(1:1) to a distance of about 7 cm, and air-dry the plate. mL of the standard solution on a plate of silica gel for thin-
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying layer chromatography. Develop the plate with a mixture of
on the plate, heat the plate at 1059C for 5 minutes, allow to ethyl acetate, hexane and acetic acid (100) (10:10:1) to a
cool, and spray water: one of the several spots obtained distance of about 7 cm, and air-dry the plate. Spray evenly
from the sample solution has the same color tone and Rf iron (III) chloride-methanol TS on the plate: one of the
value with the blue-green to grayish green spot from the several spots obtained from the sample solution has the
standard solution (Processed ginger). same color tone and Rf value with the yellow-brown spot
from the standard solution (Scutellaria Root).
(4) Shake 0.5 g of the dry extract (or 1.5 g of the viscous
Orengedokuto Extract extract) with 10 mL of methanol, centrifuge, and use the su-
pernatant liquid as the sample solution. Separately, dissolve
黄連解毒湯エキス 1 mg of geniposide for thin-layer chromatography in 1 mL
of methanol, and use this solution as the standard solution.
Change the Identification as follows: Perform the test with these solutions as directed under
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
Identification (1) Shake 0.5 g of the dry extract (or 1.5 g
sample solution and standard solution on a plate of silica
of the viscous extract) with 10 mL of methanol, centrifuge,
gel for thin-layer chromatography. Develop the plate with a
and use the supernatant liquid as the sample solution. Sepa-
mixture of ethyl acetate, methanol and water (20:3:2) to a
rately, dissolve 1 mg of coptisine chloride for thin-layer
distance of about 7 cm, and air-dry the plate. Spray evenly
chromatography in 5 mL of methanol, and use this solution
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
as the standard solution. Perform the test with these solu-
heat the plate at 1059C for 5 minutes: one of the several
tions as directed under Thin-layer Chromatography <2.03>.
spots obtained from the sample solution has the same color
Spot 5 mL each of the sample solution and standard solution
tone and Rf value with the dark purple spot from the stand-
on a plate of silica gel for thin-layer chromatography. De-
ard solution (Gardenia Fruit).
velop the plate with a mixture of ethyl acetate, ammonia so-
lution (28) and methanol (15:1:1) to a distance of about 7
cm, and air-dry the plate. Examine under ultraviolet light
(main wavelength: 365 nm): one of the several spots ob- Oriental Bezoar
tained from the sample solution has the same color tone and
ゴオウ
Rf value with the yellow fluorescent spot from the standard
solution (Coptis Rhizome).
Change the Identification (1) as follows:
(2) Shake 0.5 g of the dry extract (or 1.5 g of the viscous
extract) with 5 mL of water, then add 25 mL of ethyl ace- Identification (1) To 25 mg of pulverized Oriental
tate, and shake. Separate the ethyl acetate layer, evaporate Bezoar add 10 mL of methanol, shake for 5 minutes, and
the solvent under reduced pressure, add 1 mL of methanol centrifuge. Take the supernatant liquid, evaporate the sol-
to the residue, and use this solution as the sample solution. vent under reduced pressure, dissolve the residue in 0.5 mL
Separately, dissolve 1 mg of limonin for thin-layer chroma- of methanol, and use this solution as the sample solution.
tography in 1 mL of methanol, and use this solution as the Separately, dissolve 5 mg each of cholic acid for thin-layer
standard solution. Perform the test with these solutions as chromatography and deoxycholic acid for thin-layer chro-
directed under Thin-layer Chromatography <2.03>. Spot 10 matography in 5 mL of methanol, respectively, and use
mL of the sample solution and 5 mL of the standard solution these solutions as the standard solution (1) and the standard
on a plate of silica gel for thin-layer chromatography. De- solution (2). Perform the test with these solutions as di-
velop the plate with a mixture of ethyl acetate and hexane rected under Thin-layer Chromatography <2.03>. Spot 5 mL
(5:1) to a distance of about 7 cm, and air-dry the plate. each of the sample solution, standard solutions (1) and (2)
Spray evenly vanillin-sulfuric acid-ethanol TS for spraying on a plate of silica gel for thin-layer chromatography. De-
on the plate, heat the plate at 1059C for 5 minutes, and velop the plate with a mixture of ethyl acetate, formic acid
allow to cool: one of the several spots obtained from the and methanol (30:1:1) to a distance of about 7 cm, and air-
sample solution has the same color tone and Rf value with dry the plate. Spray evenly vanillin-sulfuric acid-ethanol TS
the purple spot from the standard solution (Phellodendron for spraying on the plate, and heat at 1059C for 10 minutes:
Bark). two of the several spots obtained from the sample solution

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2779

have the same color tone and Rf value with each spot from to make exactly 50 mL, and use this solution as the sample
the standard solutions (1) and (2). solution. Separately, weigh accurately about 10 mg of
Glycyrrhizic Acid RS (separately determine the water <2.48>
by coulometric titration, using 10 mg), dissolve in diluted
Otsujito Extract methanol (1 in 2) to make exactly 100 mL, and use this solu-
tion as the standard solution. Perform the test with exactly
乙字湯エキス 10 mL each of the sample solution and standard solution as
directed under Liquid Chromatography <2.01> according to
Change the Origin/limits of content, Identifica- the following conditions, and determine the peak areas, AT
tion (4) and Assay (3) as follows: and AS, of glycyrrhizic acid in each solution.

Amount (mg) of glycyrrhizic acid (C42H62O16)


Otsujito Extract contains not less than 1.2 mg and
= MS × AT/AS × 1/2
not more than 4.8 mg of saikosaponin b2, not less
than 80 mg and not more than 240 mg of baicalin MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
(C21H18O11: 446.36), not less than 14 mg and not more lated on the anhydrous basis
than 42 mg (for preparation prescribed 2 g of Glycyr-
Operating conditions—
rhiza) or not less than 20 mg and not more than 60 mg
Detector: An ultraviolet absorption photometer (wave-
(for preparation prescribed 3 g of Glycyrrhiza) of
length: 254 nm).
glycyrrhizic acid (C42H62O16: 822.93), and not less
Column: A stainless steel column 4.6 mm in inside diame-
than 0.5 mg of sennoside A (C42H38O20: 862.74) or not
ter and 15 cm in length, packed with octadecylsilanized
less than 1.5 mg of rhein (for preparation prescribed
silica gel for liquid chromatography (5 mm in particle diame-
0.5 g of Rhubarb) or not less than 1 mg of sennoside
ter).
A (C42H38O20: 862.74) or not less than 3 mg of rhein
Column temperature: A constant temperature of about
(for preparation prescribed 1 g of Rhubarb), per
409C.
extract prepared with the amount specified in the
Mobile phase: Dissolve 3.85 g of ammonium acetate in
Method of preparation.
720 mL of water, and add 5 mL of acetic acid (100) and 280
Identification mL of acetonitrile.
(4) Shake 1.0 g of the dry extract (or 3.0 g of the viscous Flow rate: 1.0 mL per minute (the retention time of
extract) with 10 mL of water, add 10 mL of 1-butanol, glycyrrhizic acid is about 15 minutes).
shake, centrifuge, and use the supernatant liquid as the System suitability—
sample solution. Separately, dissolve 1 mg of liquiritin for System performance: Dissolve 5 mg of monoammonium
thin-layer chromatography in 1 mL of methanol, and use glycyrrhizinate for resolution check in 20 mL of dilute
this solution as the standard solution. Perform the test with ethanol. When the procedure is run with 10 mL of this solu-
these solutions as directed under Thin-layer Chromatogra- tion under the above operating conditions, the resolution
phy <2.03>. Spot 1 mL each of the sample solution and between the peak having the relative retention time of about
standard solution on a plate of silica gel for thin-layer chro- 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
matography. Develop the plate with a mixture of ethyl ace- not less than 1.5.
tate, methanol and water (20:3:2) to a distance of about 7 System repeatability: When the test is repeated 6 times
cm, and air-dry the plate. Spray evenly dilute sulfuric acid with 10 mL of the standard solution under the above operat-
on the plate, heat the plate at 1059C for 5 minutes, and exa- ing conditions, the relative standard deviation of the peak
mine under ultraviolet light (main wavelength: 365 nm): one area of glycyrrhizic acid is not more than 1.5z.
of the several spots obtained from the sample solution has
the same color tone and Rf value with the yellow-green fluo-
rescent spot from the standard solution (Glycyrrhiza). Platycodon Root
Assay
キキョウ
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
the dry extract (or an amount of the viscous extract, equiva-
Change the origin/limits of content as follows:
lent to about 0.5 g of the dried substance), add 20 mL of
diethyl ether and 10 mL of water, and shake for 10 minutes.
Platycodon Root is the root of Platycodon gran-
After centrifugation, remove the upper layer, add 20 mL of
diflorus A. De Candolle (Campanulaceae).
diethyl ether, proceed in the same manner as described
above, and remove the upper layer. To the resultant aque-
ous layer add 10 mL of methanol, shake for 30 minutes,
centrifuge, and take the supernatant liquid. To the residue
add 20 mL of diluted methanol (1 in 2), shake for 5
minutes, centrifuge, and take the supernatant liquid. Com-
bine these supernatant liquids, add diluted methanol (1 in 2)

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2780 Crude Drugs and Related Drugs Supplement I, JP XVII

(1:1) to a distance of about 7 cm, and air-dry the plate.


Rape Seed Oil Spray evenly dilute sulfuric acid on the plate, heat the plate
at 1059 C for 5 minutes, and examine under ultraviolet light
ナタネ油 (main wavelength: 365 nm): one of the several spots ob-
tained from the sample solution has the same color tone and
Change the origin/limits of content as follows: Rf value with the blue-white fluorescent spot from the
standard solution (Atractylodes Rhizome).
Rape Seed Oil is the fixed oil obtained from the (3) For preparation prescribed Atractylodes Lancea
seed of Brassica napus Linn áe or Brassica rapa Linn áe Rhizome—Shake 2.0 g of the dry extract (or 6.0 g of the vis-
var. oleifera De Candolle (Cruciferae). cous extract) with 10 mL of water, add 25 mL of hexane,
and shake. Separate the hexane layer, evaporate the layer
under reduced pressure, add 2 mL of hexane to the residue,
Rikkunshito Extract and use this solution as the sample solution. Perform the
test with the sample solution as directed under Thin-layer
六君子湯エキス Chromatography <2.03>. Spot 20 mL of the sample solution
on a plate of silica gel with fluorescent indicator for thin-
Change the Origin/limits of content, Identifica- layer chromatography. Develop the plate with a mixture of
tion and Assay (3) as follows: hexane and acetone (7:1) to a distance of about 7 cm, and
air-dry the plate. Examine under ultraviolet light (main
Rikkunshito Extract contains not less than 2.4 mg wavelength: 254 nm): a dark purple spot is observed at an
of ginsenoside Rb1 (C54H92O23: 1109.29), not less than Rf value of about 0.5. The spot shows a greenish brown
16 mg and not more than 48 mg of hesperidin, and color after being sprayed evenly 4-dimethylaminobenzalde-
not less than 6 mg and not more than 18 mg of glycyr- hyde TS for spraying, heated at 1059C for 5 minutes, and
rhizic acid (C42H62O16: 822.93), per extract prepared allowed to cool (Atractylodes Lancea Rhizome).
with the amount specified in the Method of prepara- (4) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
tion. extract) with 10 mL of water, add 10 mL of 1-butanol,
shake, centrifuge, and use the supernatant liquid as the
Identification (1) Shake 2.0 g of the dry extract (or 6.0 g
sample solution. Separately, dissolve 1 mg of hesperidin for
of the viscous extract) with 10 mL of sodium hydroxide TS,
thin-layer chromatography in 1 mL of methanol, and use
add 5 mL of 1-butanol, shake, centrifuge, and use the su-
this solution as the standard solution. Perform the test with
pernatant liquid as the sample solution. Separately, dissolve
these solutions as directed under Thin-layer Chromatogra-
1 mg of Ginsenoside Rb1 RS or ginsenoside Rb1 for thin-
phy <2.03>. Spot 20 mL of the sample solution and 10 mL of
layer chromatography in 1 mL of methanol, and use this so-
the standard solution on a plate of silica gel for thin-layer
lution as the standard solution. Perform the test with these
chromatography. Develop the plate with a mixture of ethyl
solutions as directed under Thin-layer Chromatography
acetate, acetone, water and acetic acid (100) (10:6:3:1) to a
<2.03>. Spot 10 mL of the sample solution and 2 mL of the
distance of about 7 cm, and air-dry the plate. Spray evenly
standard solution on a plate of silica gel for thin-layer chro-
2,6-dibromo-N-chloro-1,4-benzoquinone monoimine TS on
matography. Develop the plate with a mixture of ethyl ace-
the plate, and allow to stand in an ammonia gas: one of the
tate, 1-propanol, water and acetic acid (100) (7:5:4:1) to a
several spots obtained from the sample solution has the
distance of about 7 cm, and air-dry the plate. Spray evenly
same color tone and Rf value with the blue spot from the
vanillin-sulfuric acid-ethanol TS for spraying on the plate,
standard solution (Citrus Unshiu Peel).
heat the plate at 1059C for 5 minutes, and allow to cool:
(5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
one of the several spots obtained from the sample solution
extract) with 10 mL of water, add 10 mL of 1-butanol,
has the same color tone and Rf value with the blue-purple
shake, centrifuge, and use the supernatant liquid as the
spot from the standard solution (Ginseng).
sample solution. Separately, dissolve 1 mg of liquiritin for
(2) For preparation prescribed Atractylodes Rhizome—
thin-layer chromatography in 1 mL of methanol, and use
Shake 1.0 g of the dry extract (or 3.0 g of the viscous ex-
this solution as the standard solution. Perform the test with
tract) with 10 mL of water, add 25 mL of diethyl ether, and
these solutions as directed under Thin-layer Chromatogra-
shake. Separate the diethyl ether layer, evaporate the layer
phy <2.03>. Spot 1 mL each of the sample solution and
under reduced pressure, add 2 mL of diethyl ether to the
standard solution on a plate of silica gel for thin-layer chro-
residue, and use this solution as the sample solution. Sepa-
matography. Develop the plate with a mixture of ethyl ace-
rately, dissolve 1 mg of atractylenolide III for thin-layer
tate, methanol and water (20:3:2) to a distance of about 7
chromatography in 2 mL of methanol, and use this solution
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
as the standard solution. Perform the test with these solu-
on the plate, heat the plate at 1059C for 5 minutes, and exa-
tions as directed under Thin-layer Chromatography <2.03>.
mine under ultraviolet light (main wavelength: 365 nm): one
Spot 5 mL each of the sample solution and standard solution
of the several spots obtained from the sample solution has
on a plate of silica gel for thin-layer chromatography. De-
the same color tone and Rf value with the yellow-green fluo-
velop the plate with a mixture of ethyl acetate and hexane
rescent spot from the standard solution (Glycyrrhiza).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2781

(6) Shake 1.0 g of the dry extract (or 3.0 g of the viscous tion under the above operating conditions, the resolution
extract) with 10 mL of water, add 25 mL of diethyl ether, between the peak having the relative retention time of about
and shake. Separate the diethyl ether layer, evaporate the 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
layer under reduced pressure, add 2 mL of diethyl ether to not less than 1.5.
the residue, and use this solution as the sample solution. System repeatability: When the test is repeated 6 times
Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro- with 10 mL of the standard solution under the above operat-
matography in 1 mL of methanol, and use this solution as ing conditions, the relative standard deviation of the peak
the standard solution. Perform the test with these solutions area of glycyrrhizic acid is not more than 1.5z.
as directed under Thin-layer Chromatography <2.03>. Spot
30 mL of the sample solution and 5 mL of the standard solu-
tion on a plate of silica gel for thin-layer chromatography. Ryokeijutsukanto Extract
Develop the plate with a mixture of ethyl acetate and hexane
(1:1) to a distance of about 7 cm, and air-dry the plate. 苓桂朮甘湯エキス
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
on the plate, heat the plate at 1059C for 5 minutes, allow to Change the Origin/limits of content, Identifica-
cool, and spray water: one of the several spots obtained tion and Assay (2) as follows:
from the sample solution has the same color tone and Rf
value with the blue-green to grayish green spot from the Ryokeijutsukanto Extract contains not less than 1
standard solution (Ginger). mg and not more than 4 mg of (E )-cinnamic acid, and
not less than 17 mg and not more than 51 mg of
Assay
glycyrrhizic acid (C42H62O16: 822.93), per extract pre-
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
pared with the amount specified in the Method of
the dry extract (or an amount of the viscous extract, equiva-
preparation.
lent to about 0.5 g of the dried substance), add exactly 50
mL of diluted methanol (1 in 2), shake for 15 minutes, Identification (1) To 1.0 g of the dry extract (or 3.0 g of
filter, and use the filtrate as the sample solution. Separately, the viscous extract) add 10 mL of water, shake, then add 25
weigh accurately about 10 mg of Glycyrrhizic Acid RS mL of diethyl ether, and shake. Separate the diethyl ether
(separately determine the water <2.48> by coulometric titra- layer, evaporate the layer under reduced pressure, add 2 mL
tion, using 10 mg), dissolve in diluted methanol (1 in 2) to of diethyl ether to the residue, and use this solution as the
make exactly 100 mL, and use this solution as the standard sample solution. Separately, dissolve 1 mg of (E )-cinnamic
solution. Perform the test with exactly 10 mL each of the acid for thin-layer chromatography in 1 mL of methanol,
sample solution and standard solution as directed under and use this solution as the standard solution. Perform the
Liquid Chromatography <2.01> according to the following test with these solutions as directed under Thin-layer Chro-
conditions, and determine the peak areas, AT and AS, of matography <2.03>. Spot 5 mL each of the sample solution
glycyrrhizic acid in each solution. and standard solution on a plate of silica gel with fluores-
cent indicator for thin-layer chromatography. Develop the
Amount (mg) of glycyrrhizic acid (C42H62O16)
plate with a mixture of hexane, ethyl acetate, formic acid
= MS × AT/AS × 1/2
and water (60:40:4:1) to a distance of about 7 cm, and air-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- dry the plate. Examine under ultraviolet light (main wave-
lated on the anhydrous basis length: 254 nm): one of the several spots obtained from the
sample solution has the same color tone and Rf value with
Operating conditions—
the blue-purple spot from the standard solution (Cinnamon
Detector: An ultraviolet absorption photometer (wave-
Bark).
length: 254 nm).
(2) For preparation prescribed Atractylodes Rhizome—
Column: A stainless steel column 4.6 mm in inside diame-
To 1.0 g of the dry extract (or 3.0 g of the viscous extract)
ter and 15 cm in length, packed with octadecylsilanized
add 10 mL of water, shake, then add 25 mL of diethyl
silica gel for liquid chromatography (5 mm in particle diame-
ether, and shake. Separate the diethyl ether layer, evaporate
ter).
the layer under reduced pressure, add 2 mL of diethyl ether
Column temperature: A constant temperature of about
to the residue, and use this solution as the sample solution.
409C.
Separately, dissolve 1 mg of atractylenolide III for thin-
Mobile phase: Dissolve 3.85 g of ammonium acetate in
layer chromatography in 2 mL of methanol, and use this so-
720 mL of water, and add 5 mL of acetic acid (100) and 280
lution as the standard solution. Perform the test with these
mL of acetonitrile.
solutions as directed under Thin-layer Chromatography
Flow rate: 1.0 mL per minute (the retention time of
<2.03>. Spot 5 mL each of the sample solution and standard
glycyrrhizic acid is about 15 minutes).
solution on a plate of silica gel for thin-layer chromatogra-
System suitability—
phy. Develop the plate with a mixture of ethyl acetate and
System performance: Dissolve 5 mg of monoammonium
hexane (1:1) to a distance of about 7 cm, and air-dry the
glycyrrhizinate for resolution check in 20 mL of dilute
plate. Spray evenly dilute sulfuric acid on the plate, heat the
ethanol. When the procedure is run with 10 mL of this solu-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2782 Crude Drugs and Related Drugs Supplement I, JP XVII

plate at 1059 C for 5 minutes, and examine under ultraviolet Amount (mg) of glycyrrhizic acid (C42H62O16)
light (main wavelength: 365 nm): one of the several spots = MS × AT/AS × 1/2
obtained from the sample solution has the same color tone
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
and Rf value with the blue-white fluorescent spot from the
lated on the anhydrous basis
standard solution (Atractylodes Rhizome).
(3) For preparation prescribed Atractylodes Lancea Operating conditions—
Rhizome—To 2.0 g of the dry extract (or 6.0 g of the vis- Detector: An ultraviolet absorption photometer (wave-
cous extract) add 10 mL of water, shake, then add 25 mL of length: 254 nm).
hexane, and shake. Separate the hexane layer, evaporate the Column: A stainless steel column 4.6 mm in inside diame-
layer under reduced pressure, add 2 mL of hexane to the ter and 15 cm in length, packed with octadecylsilanized
residue, and use this solution as the sample solution. Per- silica gel for liquid chromatography (5 mm in particle diame-
form the test with the sample solution as directed under ter).
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam- Column temperature: A constant temperature of about
ple solution on a plate of silica gel with fluorescent indicator 409C.
for thin-layer chromatography. Develop the plate with a Mobile phase: Dissolve 3.85 g of ammonium acetate in
mixture of hexane and acetone (7:1) to a distance of about 7 720 mL of water, and add 5 mL of acetic acid (100) and 280
cm, and air-dry the plate. Examine under ultraviolet light mL of acetonitrile.
(main wavelength: 254 nm): a dark purple spot is observed Flow rate: 1.0 mL per minute (the retention time of
at an Rf value of about 0.5. The spot shows a greenish glycyrrhizic acid is about 15 minutes).
brown color after being sprayed evenly 4-dimethylamino- System suitability—
benzaldehyde TS for spraying, heated at 1059C for 5 System performance: Dissolve 5 mg of monoammonium
minutes, and allowed to cool (Atractylodes Lancea Rhi- glycyrrhizinate for resolution check in 20 mL of dilute
zome). ethanol. When the procedure is run with 10 mL of this solu-
(4) To 1.0 g of the dry extract (or 3.0 g of the viscous tion under the above operating conditions, the resolution
extract) add 10 mL of water, shake, then add 10 mL of 1- between the peak having the relative retention time of about
butanol, and shake. Centrifuge, and use the supernatant 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
liquid as the sample solution. Separately, dissolve 1 mg of not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for
liquiritin for thin-layer chromatography in 1 mL of metha- thin-layer chromatography in 50 mL of methanol. To 2 mL
nol, and use this solution as the standard solution. Perform of this solution add 2 mL of the standard solution. When
the test with these solutions as directed under Thin-layer the procedure is run with 10 mL of this solution under the
Chromatography <2.03>. Spot 1 mL each of the sample solu- above operating conditions, the resolution between the
tion and standard solution on a plate of silica gel for thin- peaks of glycyrrhizic acid and (E )-cinnamaldehyde is not
layer chromatography. Develop the plate with a mixture of less than 1.5.
ethyl acetate, methanol and water (20:3:2) to a distance of System repeatability: When the test is repeated 6 times
about 7 cm, and air-dry the plate. Spray evenly dilute sulfu- with 10 mL of the standard solution under the above operat-
ric acid on the plate, heat the plate at 1059C for 5 minutes, ing conditions, the relative standard deviation of the peak
and examine under ultraviolet light (main wavelength: 365 area of glycyrrhizic acid is not more than 1.5z.
nm): one of the several spots obtained from the sample so- ii) Weigh accurately about 0.5 g of the dry extract (or an
lution has the same color tone and Rf value with the yellow- amount of the viscous extract, equivalent to about 0.5 g of
green fluorescent spot from the standard solution (Glycyr- the dried substance), add 20 mL of ethyl acetate and 10 mL
rhiza). of water, and shake for 10 minutes. After centrifugation,
remove the upper layer, add 20 mL of ethyl acetate, proceed
Assay
in the same manner as described above, and remove the up-
(2) Glycyrrhizic acid—Perform the test according to the
per layer. To the resultant aqueous layer add 10 mL of
following i) or ii).
methanol, shake for 30 minutes, centrifuge, and take the su-
i) Weigh accurately about 0.5 g of the dry extract (or an
pernatant liquid. To the residue add 20 mL of diluted meth-
amount of the viscous extract, equivalent to about 0.5 g of
anol (1 in 2), shake for 5 minutes, centrifuge, and take the
the dried substance), add exactly 50 mL of diluted methanol
supernatant liquid. Combine these supernatant liquids, add
(1 in 2), shake for 15 minutes, filter, and use the filtrate as
diluted methanol (1 in 2) to make exactly 50 mL, and use
the sample solution. Separately, weigh accurately about 10
this solution as the sample solution. Separately, weigh accu-
mg of Glycyrrhizic Acid RS (separately determine the water
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
<2.48> by coulometric titration, using 10 mg), dissolve in
termine the water <2.48> by coulometric titration, using 10
diluted methanol (1 in 2) to make exactly 100 mL, and use
mg), dissolve in diluted methanol (1 in 2) to make exactly
this solution as the standard solution. Perform the test with
100 mL, and use this solution as the standard solution. Per-
exactly 10 mL each of the sample solution and standard so-
form the test with exactly 10 mL each of the sample solution
lution as directed under Liquid Chromatography <2.01> ac-
and standard solution as directed under Liquid Chromatog-
cording to the following conditions, and determine the peak
raphy <2.01> according to the following conditions, and de-
areas, AT and AS, of glycyrrhizic acid in each solution.
termine the peak areas, AT and AS, of glycyrrhizic acid in

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2783

each solution. Separately, dissolve 1 mg of wogonin for thin-layer chroma-


tography in 1 mL of methanol, and use this solution as the
Amount (mg) of glycyrrhizic acid (C42H62O16)
standard solution. Perform the test with these solutions as
= MS × AT/AS × 1/2
directed under Thin-layer Chromatography <2.03>. Spot 20
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- mL of the sample solution and 2 mL of the standard solution
lated on the anhydrous basis on a plate of silica gel for thin-layer chromatography. De-
velop the plate with a mixture of ethyl acetate, hexane and
Operating conditions—
acetic acid (100) (10:10:1) to a distance of about 7 cm, and
Proceed as directed in the operating conditions in i).
air-dry the plate. Spray evenly iron (III) chloride-methanol
System suitability—
TS on the plate: one of the several spots obtained from the
System repeatability: Proceed as directed in the system
sample solution has the same color tone and Rf value with
suitability in i).
the yellow-brown spot from the standard solution (Scutel-
System performance: Dissolve 5 mg of monoammonium
laria Root).
glycyrrhizinate for resolution check in 20 mL of dilute
(3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
ethanol. When the procedure is run with 10 mL of this solu-
extract) with 10 mL of water, add 25 mL of diethyl ether,
tion under the above operating conditions, the resolution
shake, and separate the diethyl ether layer. Evaporate the
between the peak having the relative retention time of about
layer under reduced pressure, add 2 mL of diethyl ether to
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
the residue, and use this solution as the sample solution.
not less than 1.5.
Separately, dissolve 1 mg of magnolol for thin-layer chro-
matography in 1 mL of methanol, and use this solution as
the standard solution. Perform the test with these solutions
Saibokuto Extract as directed under Thin-layer Chromatography <2.03>. Spot
5 mL each of the sample solution and standard solution on a
柴朴湯エキス
plate of silica gel with fluorescent indicator for thin layer
chromatography. Develop the plate with a mixture of ethyl
Change the Origin/limits of content, Identifica-
acetate and hexane (1:1) to a distance of about 7 cm, and
tion and Assay (3) as follows:
air-dry the plate. Examine under ultraviolet light (main
wavelength: 254 nm): one of the several spots obtained
Saibokuto Extract contains not less than 2 mg and
from the sample solution has the same color tone and Rf
not more than 8 mg of saikosaponin b2, not less than
value with the dark purple spot from the standard solution
90 mg and not more than 270 mg of baicalin
(Magnolia Bark).
(C21H18O11: 446.36), and not less than 14 mg and not
(4) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
more than 42 mg of glycyrrhizic acid (C42H62O16:
extract) with 10 mL of sodium hydroxide TS, add 5 mL of
822.93), per extract prepared with the amount speci-
1-butanol, shake, centrifuge, and use the supernatant liquid
fied in the Method of preparation.
as the sample solution. Separately, dissolve 1 mg of Gin-
Identification (1) Shake 2.0 g of the dry extract (or 6.0 g senoside Rb1 RS or ginsenoside Rb1 for thin-layer chroma-
of the viscous extract) with 10 mL of sodium hydroxide TS, tography in 1 mL of methanol, and use this solution as the
add 5 mL of 1-butanol, shake, centrifuge, and use the su- standard solution. Perform the test with these solutions as
pernatant liquid as the sample solution. Separately, dissolve directed under Thin-layer Chromatography <2.03>. Spot 10
1 mg of saikosaponin b2 for thin-layer chromatography in 1 mL of the sample solution and 2 mL of the standard solution
mL of methanol, and use this solution as the standard solu- on a plate of silica gel for thin-layer chromatography. De-
tion. Perform the test with these solutions as directed under velop the plate with a mixture of ethyl acetate, 1-propanol,
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam- water and acetic acid (100) (7:5:4:1) to a distance of about 7
ple solution and 2 mL of the standard solution on a plate of cm, and air-dry the plate. Spray evenly vanillin-sulfuric
silica gel for thin-layer chromatography. Develop the plate acid-ethanol TS for spraying on the plate, heat the plate at
with a mixture of ethyl acetate, ethanol (99.5) and water 1059C for 5 minutes, and allow to cool: one of the several
(8:2:1) to a distance of about 7 cm, and air-dry the plate. spots obtained from the sample solution has the same color
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying tone and Rf value with the blue-purple spot from the stand-
on the plate, heat the plate at 1059
C for 5 minutes, and exa- ard solution (Ginseng).
mine under ultraviolet light (main wavelength: 365 nm): one (5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
of the several spots obtained from the sample solution has extract) with 10 mL of water, add 10 mL of 1-butanol,
the same color tone and Rf value with the yellow fluorescent shake, centrifuge, and use the supernatant liquid as the
spot from the standard solution (Bupleurum Root). sample solution. Separately, dissolve 1 mg of liquiritin for
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous thin-layer chromatography in 1 mL of methanol, and use
extract) with 10 mL of water, add 25 mL of diethyl ether, this solution as the standard solution. Perform the test with
shake, and separate the diethyl ether layer. Evaporate the these solutions as directed under Thin-layer Chromatogra-
layer under reduced pressure, add 2 mL of diethyl ether to phy <2.03>. Spot 1 mL each of the sample solution and
the residue, and use this solution as the sample solution. standard solution on a plate of silica gel for thin-layer chro-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2784 Crude Drugs and Related Drugs Supplement I, JP XVII

matography. Develop the plate with a mixture of ethyl ace- exactly 10 mL each of the sample solution and standard so-
tate, methanol and water (20:3:2) to a distance of about 7 lution as directed under Liquid Chromatography <2.01> ac-
cm, and air-dry the plate. Spray evenly dilute sulfuric acid cording to the following conditions, and determine the peak
on the plate, heat the plate at 1059C for 5 minutes, and exa- areas, AT and AS, of glycyrrhizic acid in each solution.
mine under ultraviolet light (main wavelength: 365 nm): one
Amount (mg) of glycyrrhizic acid (C42H62O16)
of the several spots obtained from the sample solution has
= MS × AT/AS × 1/2
the same color tone and Rf value with the yellow-green fluo-
rescent spot from the standard solution (Glycyrrhiza). MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
(6) Shake 1.0 g of the dry extract (or 3.0 g of the viscous lated on the anhydrous basis
extract) with 10 mL of 0.1 mol/L hydrochloric acid TS, add
Operating conditions—
25 mL of diethyl ether, shake, and separate the diethyl ether
Detector: An ultraviolet absorption photometer (wave-
layer. Evaporate the layer under reduced pressure, add 1
length: 254 nm).
mL of methanol to the residue, and use this solution as the
Column: A stainless steel column 4.6 mm in inside diame-
sample solution. Separately, dissolve 1 mg of rosmarinic
ter and 15 cm in length, packed with octadecylsilanized
acid for thin-layer chromatography in 1 mL of methanol,
silica gel for liquid chromatography (5 mm in particle diame-
and use this solution as the standard solution. Perform the
ter).
test with these solutions as directed under Thin-layer Chro-
Column temperature: A constant temperature of about
matography <2.03>. Spot 5 mL each of the sample solution
409C.
and standard solution on a plate of silica gel for thin-layer
Mobile phase: Dissolve 3.85 g of ammonium acetate in
chromatography. Develop the plate with a mixture of ethyl
720 mL of water, and add 5 mL of acetic acid (100) and 280
acetate, water and formic acid (60:1:1) to a distance of
mL of acetonitrile.
about 7 cm, and air-dry the plate. Spray evenly iron (III)
Flow rate: 1.0 mL per minute (the retention time of
chloride-methanol TS on the plate: one of the several spots
glycyrrhizic acid is about 15 minutes).
obtained from the sample solution has the same color tone
System suitability—
and Rf value with the dark purple spot from the standard
System performance: Dissolve 5 mg of monoammonium
solution (Perilla Herb).
glycyrrhizinate for resolution check in 20 mL of dilute
(7) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
ethanol. When the procedure is run with 10 mL of this solu-
extract) with 10 mL of water, add 25 mL of diethyl ether,
tion under the above operating conditions, the resolution
and shake. Separate the diethyl ether layer, evaporate the
between the peak having the relative retention time of about
layer under reduced pressure, add 2 mL of diethyl ether to
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
the residue, and use this solution as the sample solution.
not less than 1.5. Dissolve 1 mg of baicalein for resolution
Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro-
check in 50 mL of methanol. To 2 mL of this solution add 2
matography in 1 mL of methanol, and use this solution as
mL of the standard solution. When the procedure is run
the standard solution. Perform the test with these solutions
with 10 mL of this solution under the above operating condi-
as directed under Thin-layer Chromatography <2.03>. Spot
tions, the resolution between the peaks of glycyrrhizic acid
10 mL of the sample solution and 5 mL of the standard solu-
and baicalein is not less than 1.5.
tion on a plate of silica gel for thin-layer chromatography.
System repeatability: When the test is repeated 6 times
Develop the plate with a mixture of ethyl acetate and hexane
with 10 mL of the standard solution under the above operat-
(1:1) to a distance of about 7 cm, and air-dry the plate.
ing conditions, the relative standard deviation of the peak
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
area of glycyrrhizic acid is not more than 1.5z.
on the plate, heat the plate at 1059C for 5 minutes, allow to
ii) Weigh accurately about 0.5 g of the dry extract (or an
cool, and spray water: one of the several spots obtained
amount of the viscous extract, equivalent to about 0.5 g of
from the sample solution has the same color tone and Rf
the dried substance), add 20 mL of ethyl acetate and 10 mL
value with the blue-green to grayish green spot from the
of water, and shake for 10 minutes. After centrifugation,
standard solution (Ginger).
remove the upper layer, add 20 mL of ethyl acetate, proceed
Assay in the same manner as described above, and remove the
(3) Glycyrrhizic acid—Perform the test according to the upper layer. To the resultant aqueous layer add 10 mL of
following i) or ii). methanol, shake for 30 minutes, centrifuge, and take the su-
i) Weigh accurately about 0.5 g of the dry extract (or an pernatant liquid. To the residue add 20 mL of diluted meth-
amount of the viscous extract, equivalent to about 0.5 g of anol (1 in 2), shake for 5 minutes, centrifuge, and take the
the dried substance), add exactly 50 mL of diluted methanol supernatant liquid. Combine these supernatant liquids, add
(1 in 2), shake for 15 minutes, filter, and use the filtrate as diluted methanol (1 in 2) to make exactly 50 mL, and use
the sample solution. Separately, weigh accurately about 10 this solution as the sample solution. Separately, weigh accu-
mg of Glycyrrhizic Acid RS (separately determine the water rately about 10 mg of Glycyrrhizic Acid RS (separately de-
<2.48> by coulometric titration, using 10 mg), dissolve in termine the water <2.48> by coulometric titration, using 10
diluted methanol (1 in 2) to make exactly 100 mL, and use mg), dissolve in diluted methanol (1 in 2) to make exactly
this solution as the standard solution. Perform the test with 100 mL, and use this solution as the standard solution. Per-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2785

form the test with exactly 10 mL each of the sample solution Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
and standard solution as directed under Liquid Chromatog- on the plate, heat the plate at 1059C for 5 minutes, and exa-
raphy <2.01> according to the following conditions, and de- mine under ultraviolet light (main wavelength: 365 nm): one
termine the peak areas, AT and AS, of glycyrrhizic acid in of the several spots obtained from the sample solution has
each solution. the same color tone and Rf value with the yellow fluorescent
spot from the standard solution (Bupleurum Root).
Amount (mg) of glycyrrhizic acid (C42H62O16)
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
= MS × AT/AS × 1/2
extract) with 10 mL of water, add 25 mL of diethyl ether,
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- shake, and separate the diethyl ether layer. Evaporate the
lated on the anhydrous basis layer under reduced pressure, add 2 mL of diethyl ether to
the residue, and use this solution as the sample solution.
Operating conditions—
Separately, dissolve 1 mg of wogonin for thin-layer chroma-
Proceed as directed in the operating conditions in i).
tography in 1 mL of methanol, and use this solution as the
System suitability—
standard solution. Perform the test with these solutions as
System repeatability: Proceed as directed in the system
directed under Thin-layer Chromatography <2.03>. Spot 20
suitability in i).
mL of the sample solution and 2 mL of the standard solution
System performance: Dissolve 5 mg of monoammonium
on a plate of silica gel for thin-layer chromatography. De-
glycyrrhizinate for resolution check in 20 mL of dilute
velop the plate with a mixture of ethyl acetate, hexane and
ethanol. When the procedure is run with 10 mL of this solu-
acetic acid (100) (10:10:1) to a distance of about 7 cm, and
tion under the above operating conditions, the resolution
air-dry the plate. Spray evenly iron (III) chloride-methanol
between the peak having the relative retention time of about
TS on the plate: one of the several spots obtained from the
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
sample solution has the same color tone and Rf value with
not less than 1.5.
the yellow-brown spot from the standard solution (Scutel-
laria Root).
(3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
Saikokeishito Extract extract) with 10 mL of water, add 10 mL of 1-butanol,
shake, centrifuge, and use the supernatant liquid as the
柴胡桂枝湯エキス
sample solution. Separately, dissolve 1 mg of Paeoniflorin
RS or paeoniflorin for thin-layer chromatography in 1 mL
Change the Origin/limits of content, Identifica-
of methanol, and use this solution as the standard solution.
tion and Assay (4) as follows:
Perform the test with these solutions as directed under
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
Saikokeishito Extract contains not less than 1.5 mg
sample solution and standard solution on a plate of silica
and not more than 6 mg of saikosaponin b2, not less
gel for thin-layer chromatography. Develop the plate with a
than 60 mg and not more than 180 mg of baicalin
mixture of ethyl acetate, methanol and water (20:3:2) to a
(C21H18O11: 446.36), not less than 17 mg and not more
distance of about 7 cm, and air-dry the plate. Spray evenly
than 51 mg (for preparation prescribed 2 g of Peony
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
Root) or not less than 21 mg and not more than 63 mg
heat the plate at 1059C for 5 minutes: one of the several
(for preparation prescribed 2.5 g of Peony Root) of
spots obtained from the sample solution has the same color
paeoniflorin (C23H28O11: 480.46), and not less than
tone and Rf value with the purple spot from the standard
10 mg and not more than 30 mg (for preparation
solution (Peony Root).
prescribed 1.5 g of Glycyrrhiza) or not less than 14 mg
(4) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
and not more than 42 mg (for preparation prescribed
extract) with 10 mL of sodium hydroxide TS, add 5 mL of
2 g of Glycyrrhiza) of glycyrrhizic acid (C42H62O16:
1-butanol, shake, centrifuge, and use the supernatant liquid
822.93), per extract prepared with the amount speci-
as the sample solution. Separately, dissolve 1 mg of Gin-
fied in the Method of preparation.
senoside Rb1 RS or ginsenoside Rb1 for thin-layer chroma-
Identification (1) Shake 2.0 g of the dry extract (or 6.0 g tography in 1 mL of methanol, and use this solution as the
of the viscous extract) with 10 mL of sodium hydroxide TS, standard solution. Perform the test with these solutions as
add 5 mL of 1-butanol, shake, centrifuge, and use the su- directed under Thin-layer Chromatography <2.03>. Spot 10
pernatant liquid as the sample solution. Separately, dissolve mL of the sample solution and 2 mL of the standard solution
1 mg of saikosaponin b2 for thin-layer chromatography in 1 on a plate of silica gel for thin-layer chromatography. De-
mL of methanol, and use this solution as the standard solu- velop the plate with a mixture of ethyl acetate, 1-propanol,
tion. Perform the test with these solutions as directed under water and acetic acid (100) (7:5:4:1) to a distance of about 7
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam- cm, and air-dry the plate. Spray evenly vanillin-sulfuric
ple solution and 2 mL of the standard solution on a plate of acid-ethanol TS for spraying on the plate, heat the plate at
silica gel for thin-layer chromatography. Develop the plate 1059C for 5 minutes, and allow to cool: one of the several
with a mixture of ethyl acetate, ethanol (99.5) and water spots obtained from the sample solution has the same color
(8:2:1) to a distance of about 7 cm, and air-dry the plate. tone and Rf value with the blue-purple spot from the stand-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2786 Crude Drugs and Related Drugs Supplement I, JP XVII

ard solution (Ginseng). extract) with 10 mL of water, add 25 mL of diethyl ether,


(5) Perform the test according to the following i) or ii) and shake. Separate the diethyl ether layer, evaporate the
(Cinnamon Bark). layer under reduced pressure, add 2 mL of diethyl ether to
i) Put 10 g of the dry extract (or 30 g of the viscous ex- the residue, and use this solution as the sample solution.
tract) in a 300-mL hard-glass flask, add 100 mL of water Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro-
and 1 mL of silicone resin, connect an apparatus for essen- matography in 1 mL of methanol, and use this solution as
tial oil determination, and heat to boil under a reflux con- the standard solution. Perform the test with these solutions
denser. The graduated tube of the apparatus is to be previ- as directed under Thin-layer Chromatography <2.03>. Spot
ously filled with water to the standard line, and 2 mL of 10 mL of the sample solution and 5 mL of the standard solu-
hexane is added to the graduated tube. After heating under tion on a plate of silica gel for thin-layer chromatography.
reflux for 1 hour, separate the hexane layer, and use this so- Develop the plate with a mixture of ethyl acetate and hexane
lution as the sample solution. Separately, dissolve 1 mg of (1:1) to a distance of about 7 cm, and air-dry the plate.
(E )-cinnamaldehyde for thin-layer chromatography in 1 mL Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
of methanol, and use this solution as the standard solution. on the plate, heat the plate at 1059 C for 5 minutes, allow to
Perform the test with these solutions as directed under cool, and spray water: one of the several spots obtained
Thin-layer Chromatography <2.03>. Spot 50 mL of the sam- from the sample solution has the same color tone and Rf
ple solution and 2 mL of the standard solution on a plate of value with the blue-green to grayish green spot from the
silica gel for thin-layer chromatography. Develop the plate standard solution (Ginger).
with a mixture of hexane, diethyl ether and methanol
Assay
(15:5:1) to a distance of about 7 cm, and air-dry the plate.
(4) Glycyrrhizic acid—Perform the test according to the
Spray evenly 2,4-dinitrophenylhydradine TS on the plate:
following i) or ii).
one of the several spots obtained from the sample solution
i) Weigh accurately about 0.5 g of the dry extract (or an
has the same color tone and Rf value with the yellow-orange
amount of the viscous extract, equivalent to about 0.5 g of
spot from the standard solution.
the dried substance), add exactly 50 mL of diluted methanol
ii) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
(1 in 2), shake for 15 minutes, filter, and use the filtrate as
extract) with 10 mL of water, add 5 mL of hexane, shake,
the sample solution. Separately, weigh accurately about 10
centrifuge, and use the supernatant liquid as the sample
mg of Glycyrrhizic Acid RS (separately determine the water
solution. Separately, dissolve 1 mg of (E )-2-methoxycin-
<2.48> by coulometric titration, using 10 mg), dissolve in
namaldehyde for thin-layer chromatography in 1 mL of
diluted methanol (1 in 2) to make exactly 100 mL, and use
methanol, and use this solution as the standard solution.
this solution as the standard solution. Perform the test with
Perform the test with these solutions as directed under
exactly 10 mL each of the sample solution and standard so-
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
lution as directed under Liquid Chromatography <2.01> ac-
ple solution and 2 mL of the standard solution on a plate of
cording to the following conditions, and determine the peak
silica gel for thin-layer chromatography. Develop the plate
areas, AT and AS, of glycyrrhizic acid in each solution.
with a mixture of hexane and ethyl acetate (2:1) to a dis-
tance of about 7 cm, and air-dry the plate. Examine under Amount (mg) of glycyrrhizic acid (C42H62O16)
ultraviolet light (main wavelength: 365 nm): one of the = MS × AT/AS × 1/2
several spots obtained from the sample solution has the
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
same color tone and Rf value with the blue-white fluores-
lated on the anhydrous basis
cent spot from the standard solution.
(6) Shake 1.0 g of the dry extract (or 3.0 g of the viscous Operating conditions—
extract) with 10 mL of water, add 10 mL of 1-butanol, Detector: An ultraviolet absorption photometer (wave-
shake, centrifuge, and use the supernatant liquid as the length: 254 nm).
sample solution. Separately, dissolve 1 mg of liquiritin for Column: A stainless steel column 4.6 mm in inside diame-
thin-layer chromatography in 1 mL of methanol, and use ter 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 diame-
these solutions as directed under Thin-layer Chromatogra- ter).
phy <2.03>. Spot 1 mL each of the sample solution and Column temperature: A constant temperature of about
standard solution on a plate of silica gel for thin-layer chro- 409C.
matography. Develop the plate with a mixture of ethyl ace- Mobile phase: Dissolve 3.85 g of ammonium acetate in
tate, methanol and water (20:3:2) to a distance of about 7 720 mL of water, and add 5 mL of acetic acid (100) and 280
cm, and air-dry the plate. Spray evenly dilute sulfuric acid mL of acetonitrile.
on the plate, heat the plate at 1059C for 5 minutes, and exa- Flow rate: 1.0 mL per minute (the retention time of
mine under ultraviolet light (main wavelength: 365 nm): one glycyrrhizic acid is about 15 minutes).
of the several spots obtained from the sample solution has System suitability—
the same color tone and Rf value with the yellow-green fluo- System performance: Dissolve 5 mg of monoammonium
rescent spot from the standard solution (Glycyrrhiza). glycyrrhizinate for resolution check in 20 mL of dilute
(7) Shake 1.0 g of the dry extract (or 3.0 g of the viscous ethanol. When the procedure is run with 10 mL of this solu-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2787

tion under the above operating conditions, the resolution


between the peak having the relative retention time of about Saireito Extract
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for 柴苓湯エキス
thin-layer chromatography and 1 mg of baicalein for resolu-
tion check in 50 mL of methanol. To 2 mL of this solution Change the Origin/limits of content, Identifica-
add 2 mL of the standard solution. When the procedure is tion and Assay (3) as follows:
run with 10 mL of this solution under the above operating
conditions, two peaks other than glycyrrhizic acid are ob- Saireito Extract contains not less than 2 mg and not
served with the resolutions between the peak of glycyrrhizic more than 8 mg of saikosaponin b2, not less than 80
acid and each of the two peaks being not less than 1.5. mg and not more than 240 mg of baicalin (C21H18O11:
System repeatability: When the test is repeated 6 times 446.36), and not less than 14 mg and not more than
with 10 mL of the standard solution under the above operat- 42 mg of glycyrrhizic acid (C42H62O16: 822.93), per
ing conditions, the relative standard deviation of the peak extract prepared with the amount specified in the
area of glycyrrhizic acid is not more than 1.5z. Method of preparation.
ii) Weigh accurately about 0.5 g of the dry extract (or an
Identification (1) To 2.0 g of Saireito Extract add 10 mL
amount of the viscous extract, equivalent to about 0.5 g of
of sodium hydroxide TS, shake, then add 5 mL of 1-
the dried substance), add 20 mL of ethyl acetate and 10 mL
butanol, shake, centrifuge, and use the supernatant liquid
of water, and shake for 10 minutes. After centrifugation,
as the sample solution. Separately, dissolve 1 mg of sai-
remove the upper layer, add 20 mL of ethyl acetate, proceed
kosaponin b2 for thin-layer chromatography in 1 mL of
in the same manner as described above, and remove the
methanol, and use this solution as the standard solution.
upper layer. To the resultant aqueous layer add 10 mL of
Perform the test with these solutions as directed under
methanol, shake for 30 minutes, centrifuge, and take the su-
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
pernatant liquid. To the residue add 20 mL of diluted meth-
ple solution and 2 mL of the standard solution on a plate of
anol (1 in 2), shake for 5 minutes, centrifuge, and take the
silica gel for thin-layer chromatography. Develop the plate
supernatant liquid. Combine these supernatant liquids, add
with a mixture of ethyl acetate, ethanol (99.5) and water
diluted methanol (1 in 2) to make exactly 50 mL, and use
(8:2:1) to a distance of about 7 cm, and air-dry the plate.
this solution as the sample solution. Separately, weigh accu-
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
C for 5 minutes, and exa-
on the plate, heat the plate at 1059
termine the water <2.48> by coulometric titration, using 10
mine under ultraviolet light (main wavelength: 365 nm): one
mg), dissolve in diluted methanol (1 in 2) to make exactly
of the several spots obtained from the sample solution has
100 mL, and use this solution as the standard solution. Per-
the same color tone and Rf value with the yellow fluorescent
form the test with exactly 10 mL each of the sample solution
spot from the standard solution (Bupleurum Root).
and standard solution as directed under Liquid Chromatog-
(2) To 1.0 g of Saireito Extract add 10 mL of water,
raphy <2.01> according to the following conditions, and de-
shake, then add 25 mL of diethyl ether, and shake. Separate
termine the peak areas, AT and AS, of glycyrrhizic acid in
the diethyl ether layer, evaporate the layer under reduced
each solution.
pressure, add 2 mL of diethyl ether to the residue, and use
Amount (mg) of glycyrrhizic acid (C42H62O16) this solution as the sample solution. Separately, dissolve 1
= MS × AT/AS × 1/2 mg of [6]-gingerol for thin-layer chromatography in 1 mL
of methanol, and use this solution as the standard solution.
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
Perform the test with these solutions as directed under
lated on the anhydrous basis
Thin-layer Chromatography <2.03>. Spot 15 mL of the sam-
Operating conditions— ple solution and 5 mL of the standard solution on a plate of
Proceed as directed in the operating conditions in i). silica gel for thin-layer chromatography. Develop the plate
System suitability— with a mixture of ethyl acetate and hexane (1:1) to a dis-
System repeatability: Proceed as directed in the system tance of about 7 cm, and air-dry the plate. Spray evenly 4-
suitability in i). dimethylaminobenzaldehyde TS for spraying on the plate,
System performance: Dissolve 5 mg of monoammonium heat the plate at 1059C for 5 minutes, allow to cool, and
glycyrrhizinate for resolution check in 20 mL of dilute spray water: one of the several spots obtained from the sam-
ethanol. When the procedure is run with 10 mL of this solu- ple solution has the same color tone and Rf value with the
tion under the above operating conditions, the resolution blue-green to grayish green spot from the standard solution
between the peak having the relative retention time of about (Ginger).
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is (3) To 1.0 g of Saireito Extract add 10 mL of water,
not less than 1.5. shake, then add 25 mL of diethyl ether, and shake. Separate
the diethyl ether layer, evaporate the layer under reduced
pressure, add 2 mL of diethyl ether to the residue, and use
this solution as the sample solution. Separately, dissolve 1

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2788 Crude Drugs and Related Drugs Supplement I, JP XVII

mg of wogonin for thin-layer chromatography in 1 mL of of about 7 cm, and air-dry the plate. Spray evenly 4-
methanol, and use this solution as the standard solution. methoxybenzaldehyde-sulfuric acid-acetic acid TS on the
Perform the test with these solutions as directed under plate, heat the plate at 1059C for 5 minutes, allow to cool,
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam- and examine under ultraviolet light (main wavelength: 365
ple solution and 2 mL of the standard solution on a plate of nm): one of the several spots obtained from the sample so-
silica gel for thin-layer chromatography. Develop the plate lution has the same color tone and Rf value with the yellow
with a mixture of ethyl acetate, hexane and acetic acid (100) fluorescent spot from the standard solution (Alisma Tuber).
(10:10:1) to a distance of about 7 cm, air-dry the plate. (7) For preparation prescribed Atractylodes Rhizome—
Spray evenly iron (III) chloride-methanol TS on the plate: To 1.0 g of Saireito Extract add 10 mL of water, shake,
one of the several spots obtained from the sample solution then add 25 mL of diethyl ether, and shake. Separate the
has the same color tone and Rf value with the yellow-brown diethyl ether layer, evaporate the layer under reduced pres-
spot from the standard solution (Scutellaria Root). sure, add 2 mL of diethyl ether to the residue, and use this
(4) To 2.0 g of Saireito Extract add 10 mL of sodium solution as the sample solution. Separately, dissolve 1 mg of
hydroxide TS, shake, then add 5 mL of 1-butanol, shake, atractylenolide III for thin-layer chromatography in 2 mL
centrifuge, and use the supernatant liquid as the sample so- of methanol, and use this solution as the standard solution.
lution. Separately, dissolve 1 mg of Ginsenoside Rb1 RS or Perform the test with these solutions as directed under
ginsenoside Rb1 for thin-layer chromatography in 1 mL of Thin-layer Chromatography <2.03>. Spot 5 mL each of the
methanol, and use this solution as the standard solution. sample solution and standard solution on a plate of silica
Perform the test with these solutions as directed under gel for thin-layer chromatography. Develop the plate with a
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam- mixture of ethyl acetate and hexane (1:1) to a distance of
ple solution and 2 mL of the standard solution on a plate of about 7 cm, and air-dry the plate. Spray evenly dilute sulfu-
silica gel for thin-layer chromatography. Develop the plate ric acid on the plate, heat the plate at 1059C for 5 minutes,
with a mixture of ethyl acetate, 1-propanol, water and and examine under ultraviolet light (main wavelength: 365
acetic acid (100) (7:5:4:1) to a distance of about 7 cm, and nm): one of the several spots obtained from the sample
air-dry the plate. Spray evenly vanillin-sulfuric acid-ethanol solution has the same color tone and Rf value with the blue-
TS for spraying on the plate, heat the plate at 1059C for 5 white fluorescent spot from the standard solution (Atrac-
minutes, and allow to cool: one of the several spots ob- tylodes Rhizome).
tained from the sample solution has the same color tone and (8) For preparation prescribed Atractylodes Lancea
Rf value with the blue-purple spot from the standard solu- Rhizome—To 2.0 g of Saireito Extract add 10 mL of water,
tion (Ginseng). shake, then add 25 mL of hexane, and shake. Separate the
(5) To 2.0 g of Saireito Extract add 10 mL of water, hexane layer, evaporate the layer under reduced pressure,
shake, then add 5 mL of 1-butanol, shake, centrifuge, and add 2 mL of hexane to the residue, and use this solution as
use the supernatant liquid as the sample solution. Sepa- the sample solution. Perform the test with the sample solu-
rately, dissolve 1 mg of liquiritin for thin-layer chromatog- tion as directed under Thin-layer Chromatography <2.03>.
raphy in 1 mL of methanol, and use this solution as the Spot 20 mL of the sample solution on a plate of silica gel
standard solution. Perform the test with these solutions as with fluorescent indicator for thin-layer chromatography.
directed under Thin-layer Chromatography <2.03>. Spot 1 Develop the plate with a mixture of hexane and acetone
mL each of the sample solution and standard solution on a (7:1) to a distance of about 7 cm, and air-dry the plate. Exa-
plate of silica gel for thin-layer chromatography. Develop mine under ultraviolet light (main wavelength: 254 nm): a
the plate with a mixture of ethyl acetate, methanol and dark purple spot is observed at an Rf value of about 0.5.
water (20:3:2) to a distance of about 7 cm, and air-dry the The spot shows a greenish brown color after being sprayed
plate. Spray evenly dilute sulfuric acid on the plate, heat the evenly 4-dimethylaminobenzaldehyde TS for spraying, heat-
plate at 1059 C for 5 minutes, and examine under ultraviolet ed at 1059C for 5 minutes, and allowed to cool (Atrac-
light (main wavelength: 365 nm): one of the several spots tylodes Lancea Rhizome).
obtained from the sample solution has the same color tone (9) To 1.0 g of Saireito Extract add 10 mL of water,
and Rf value with the yellow-green fluorescent spot from shake, then add 25 mL of diethyl ether, and shake. Separate
the standard solution (Glycyrrhiza). the diethyl ether layer, evaporate the layer under reduced
(6) To 2.0 g of Saireito Extract add 10 mL of sodium pressure, add 2 mL of diethyl ether to the residue, and use
carbonate TS, shake, then add 10 mL of diethyl ether, this solution as the sample solution. Separately, dissolve 1
shake, centrifuge, and use the supernatant liquid as the mg of (E )-cinnamic acid for thin-layer chromatography in 1
sample solution. Separately, dissolve 1 mg of alisol A for mL of methanol, and use this solution as the standard solu-
thin-layer chromatography in 1 mL of methanol, and use tion. Perform the test with these solutions as directed under
this solution as the standard solution. Perform the test with Thin-layer Chromatography <2.03>. Spot 40 mL of the sam-
these solutions as directed under Thin-layer Chromatogra- ple solution and 2 mL of the standard solution on a plate of
phy <2.03>. Spot 20 mL of the sample solution and 2 mL of silica gel with fluorescent indicator for thin-layer chroma-
the standard solution on a plate of silica gel for thin-layer tography. Develop the plate with a mixture of hexane, ethyl
chromatography. Develop the plate with a mixture of ethyl acetate, formic acid and water (60:40:4:1) to a distance of
acetate, hexane and acetic acid (100) (10:10:3) to a distance about 7 cm, and air-dry the plate. Examine under ultravio-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2789

let light (main wavelength: 254 nm): one of the several spots ii) Weigh accurately about 0.5 g of Saireito Extract, add
obtained from the sample solution has the same color tone 20 mL of ethyl acetate and 10 mL of water, and shake for
and Rf value with the dark purple spot from the standard 10 minutes. After centrifugation, remove the upper layer,
solution (Cinnamon Bark). add 20 mL of ethyl acetate, proceed in the same manner as
described above, and remove the upper layer. To the
Assay
resultant aqueous layer add 10 mL of methanol, shake for
(3) Glycyrrhizic acid—Perform the test according to the
30 minutes, centrifuge, and take the supernatant liquid. To
following i) or ii).
the residue add 20 mL of diluted methanol (1 in 2), shake
i) Weigh accurately about 0.5 g of Saireito Extract, add
for 5 minutes, centrifuge, and take the supernatant liquid.
exactly 50 mL of diluted methanol (1 in 2), shake for 15
Combine these supernatant liquids, add diluted methanol (1
minutes, filter, and use the filtrate as the sample solution.
in 2) to make exactly 50 mL, and use this solution as the
Separately, weigh accurately about 10 mg of Glycyrrhizic
sample solution. Separately, weigh accurately about 10 mg
Acid RS (separately determine the water <2.48> by coulo-
of Glycyrrhizic Acid RS (separately determine the water
metric titration, using 10 mg), dissolve in diluted methanol
<2.48> by coulometric titration, using 10 mg), dissolve in
(1 in 2) to make exactly 100 mL, and use this solution as the
diluted methanol (1 in 2) to make exactly 100 mL, and use
standard solution. Perform the test with exactly 10 mL each
this solution as the standard solution. Perform the test with
of the sample solution and standard solution as directed
exactly 10 mL each of the sample solution and standard so-
under Liquid Chromatography <2.01> according to the fol-
lution as directed under Liquid Chromatography <2.01> ac-
lowing conditions, and determine the peak areas, AT and
cording to the following conditions, and determine the peak
AS, of glycyrrhizic acid in each solution.
areas, AT and AS, of glycyrrhizic acid in each solution.
Amount (mg) of glycyrrhizic acid (C42H62O16)
Amount (mg) of glycyrrhizic acid (C42H62O16)
= MS × AT/AS × 1/2
= MS × AT/AS × 1/2
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
lated on the anhydrous basis
lated on the anhydrous basis
Operating conditions—
Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
Proceed as directed in the operating conditions in i).
length: 254 nm).
System suitability—
Column: A stainless steel column 4.6 mm in inside diame-
System repeatability: Proceed as directed in the system
ter and 15 cm in length, packed with octadecylsilanized
suitability in i).
silica gel for liquid chromatography (5 mm in particle diame-
System performance: Dissolve 5 mg of monoammonium
ter).
glycyrrhizinate for resolution check in 20 mL of dilute
Column temperature: A constant temperature of about
ethanol. When the procedure is run with 10 mL of this solu-
409C.
tion under the above operating conditions, the resolution
Mobile phase: Dissolve 3.85 g of ammonium acetate in
between the peak having the relative retention time of about
720 mL of water, and add 5 mL of acetic acid (100) and 280
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
mL of acetonitrile.
not less than 1.5.
Flow rate: 1.0 mL per minute (the retention time of
glycyrrhizic acid is about 15 minutes).
System suitability—
System performance: Dissolve 5 mg of monoammonium Saposhnikovia Root and Rhizome
glycyrrhizinate for resolution check in 20 mL of dilute
ボウフウ
ethanol. When the procedure is run with 10 mL of this solu-
tion under the above operating conditions, the resolution
Change the Identification as follows:
between the peak having the relative retention time of about
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is Identification To 1.0 g of pulverized Saposhnikovia Root
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for and Rhizome, add 5 mL of methanol, shake for 10 minutes,
thin-layer chromatography and 1 mg of baicalein for resolu- filter, and use the filtrate as the sample solution. Separately,
tion check in 50 mL of methanol. To 2 mL of this solution dissolve 1 mg of 4?-O-glucosyl-5-O-methylvisamminol for
add 2 mL of the standard solution. When the procedure is thin-layer chromatography in 1 mL of methanol, and use
run with 10 mL of this solution under the above operating this solution as the standard solution. Perform the test with
conditions, two peaks other than glycyrrhizic acid are ob- these solutions as directed under Thin-layer Chromatogra-
served with the resolutions between the peak of glycyrrhizic phy <2.03>. Spot 4 mL of the sample solution and 1 mL of
acid and each of the two peaks being not less than 1.5. the standard solution on a plate of silica gel with fluorescent
System repeatability: When the test is repeated 6 times indicator for thin-layer chromatography. Develop the plate
with 10 mL of the standard solution under the above operat- with a mixture of ethyl formate, formic acid, 2-butanone,
ing conditions, the relative standard deviation of the peak and water (20:5:5:1) to a distance of about 7 cm, and air-
area of glycyrrhizic acid is not more than 1.5z. dry the plate. Examine under ultraviolet light (main wave-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2790 Crude Drugs and Related Drugs Supplement I, JP XVII

length: 254 nm): one of the several spots from the sample mine under ultraviolet light (main wavelength: 365 nm): one
solution has the same color tone and Rf value with the spot of the several spots obtained from the sample solution has
from the standard solution. the same color tone and Rf value with the yellow-green fluo-
rescent spot from the standard solution (Glycyrrhiza).

Assay
Delete the following monograph:
(2) Glycyrrhizic acid—Weigh accurately about 0.2 g of
the dry extract (or an amount of the viscous extract, equiva-
Scopolia Extract, Papaverine and lent to about 0.2 g of the dried substance), add exactly 50
Ethyl Aminobenzoate Powder mL of diluted methanol (1 in 2), shake for 15 minutes,
filter, and use the filtrate as the sample solution. Separately,
ロートエキス・パパベリン・アネスタミン散 weigh accurately about 10 mg of Glycyrrhizic Acid RS
(separately determine the water <2.48> by coulometric titra-
tion, using 10 mg), dissolve in diluted methanol (1 in 2) to
make exactly 100 mL, and use this solution as the standard
Shakuyakukanzoto Extract solution. Perform the test with exactly 10 mL each of the
sample solution and standard solution as directed under
芍薬甘草湯エキス Liquid Chromatography <2.01> according to the following
conditions, and determine the peak areas, AT and AS, of
Change the Origin/limits of content, Identifica- glycyrrhizic acid in each solution.
tion and Assay (2) as follows:
Amount (mg) of glycyrrhizic acid (C42H62O16)
= MS × AT/AS × 1/2
Shakuyakukanzoto Extract contains not less than
50 mg and not more than 150 mg of paeoniflorin MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
(C23H28O11: 480.46), and not less than 40 mg and not lated on the anhydrous basis
more than 120 mg of glycyrrhizic acid (C42H62O16:
Operating conditions—
822.93), per extract prepared with the amount speci-
Detector: An ultraviolet absorption photometer (wave-
fied in the Method of preparation.
length: 254 nm).
Identification (1) Shake 0.5 g of the dry extract (or 1.5 g Column: A stainless steel column 4.6 mm in inside diame-
of the viscous extract) with 10 mL of water, then add 10 mL ter and 15 cm in length, packed with octadecylsilanized
of 1-butanol, shake, centrifuge, and use the supernatant liq- silica gel for liquid chromatography (5 mm in particle diame-
uid as the sample solution. Separately, dissolve 1 mg of ter).
Paeoniflorin RS or paeoniflorin for thin-layer chromatogra- Column temperature: A constant temperature of about
phy in 1 mL of methanol, and use this solution as the stand- 409C.
ard solution. Perform the test with these solutions as di- Mobile phase: Dissolve 3.85 g of ammonium acetate in
rected under Thin-layer Chromatography <2.03>. Spot 5 mL 720 mL of water, and add 5 mL of acetic acid (100) and 280
each of the sample solution and standard solution on a plate mL of acetonitrile.
of silica gel for thin-layer chromatography. Develop the Flow rate: 1.0 mL per minute (the retention time of
plate with a mixture of ethyl acetate, methanol and water glycyrrhizic acid is about 15 minutes).
(20:3:2) to a distance of about 7 cm, and air-dry the plate. System suitability—
Spray evenly 4-methoxybenzaldehyde-sulfuric acid TS on System performance: Dissolve 5 mg of monoammonium
the plate, and heat the plate at 1059C for 5 minutes: one of glycyrrhizinate for resolution check in 20 mL of dilute
the several spots obtained from the sample solution has the ethanol. When the procedure is run with 10 mL of this solu-
same color tone and Rf value with the purple spot from the tion under the above operating conditions, the resolution
standard solution (Peony Root). between the peak having the relative retention time of about
(2) Shake 0.5 g of the dry extract (or 1.5 g of the viscous 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
extract) with 10 mL of water, then add 10 mL of 1-butanol, not less than 1.5.
shake, centrifuge, and use the supernatant liquid as the System repeatability: When the test is repeated 6 times
sample solution. Separately, dissolve 1 mg of liquiritin for with 10 mL of the standard solution under the above operat-
thin-layer chromatography in 1 mL of methanol, and use ing conditions, the relative standard deviation of the peak
this solution as the standard solution. Perform the test with area of glycyrrhizic acid is not more than 1.5z.
these solutions as directed under Thin-layer Chromatogra-
phy <2.03>. Spot 1 mL each of the sample solution and
standard solution on a plate of silica gel for thin-layer chro-
matography. Develop the plate with a mixture of ethyl ace-
tate, methanol and water (20:3:2) to a distance of about 7
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
on the plate, heat the plate at 1059C for 5 minutes, and exa-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2791

color after being sprayed evenly 4-dimethylaminobenzalde-


Shimbuto Extract hyde TS for spraying, heated at 1059C for 5 minutes, and
allowed to cool (Atractylodes Lancea Rhizome).
真武湯エキス (4) To 1.0 g of Shimbuto Extract, add 10 mL of water,
shake, then add 25 mL of diethyl ether, and shake. Separate
Change the Identification as follows: the diethyl ether layer, evaporate the layer under reduced
pressure, add 2 mL of diethyl ether to the residue, and use
Identification (1) To 2.0 g of Shimbuto Extract, add 10
this solution as the sample solution. Separately, dissolve 1
mL of water, shake, then add 5 mL of 1-butanol, shake,
mg of [6]-gingerol for thin-layer chromatography in 1 mL
centrifuge, and use the supernatant liquid as the sample
of methanol, and use this solution as the standard solution.
solution. Separately, dissolve 1 mg of Paeoniflorin RS or
Perform the test with these solutions as directed under
paeoniflorin for thin-layer chromatography in 1 mL of
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
methanol, and use this solution as the standard solution.
ple solution and 5 mL of the standard solution on a plate of
Perform the test with these solutions as directed under
silica gel for thin-layer chromatography. Develop the plate
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
with a mixture of ethyl acetate and hexane (1:1) to a dis-
sample solution and standard solution on a plate of silica
tance of about 7 cm, and air-dry the plate. Spray evenly 4-
gel for thin-layer chromatography. Develop the plate with a
dimethylaminobenzaldehyde TS for spraying on the plate,
mixture of ethyl acetate, methanol and water (20:3:2) to a
heat the plate at 1059C for 5 minutes, allow to cool, and
distance of about 7 cm, and air-dry the plate. Spray evenly
spray water: one of the several spots obtained from the sam-
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
ple solution has the same color tone and Rf value with the
heat the plate at 1059C for 5 minutes: one of the several
blue-green to grayish green spot from the standard solution
spots obtained from the sample solution has the same color
(Ginger).
tone and Rf value with the purple spot from the standard
(5) To 3.0 g of Shimbuto Extract, add 20 mL of diethyl
solution (Peony Root).
ether and 2 mL of ammonia TS, shake for 10 minutes,
(2) For preparation prescribed Atractylodes Rhizome—
centrifuge, and take the supernatant liquid. Evaporate the
To 1.0 g of Shimbuto Extract, add 10 mL of water, shake,
supernatant liquid under reduced pressure, add 1 mL of
then add 25 mL of diethyl ether, and shake. Separate the
acetonitrile to the residue, and use this solution as the sam-
diethyl ether layer, evaporate the layer under reduced pres-
ple solution. Separately, dissolve 1 mg of benzoylmesaco-
sure, add 2 mL of diethyl ether to the residue, and use this
nine hydrochloride for thin-layer chromatography in 10 mL
solution as the sample solution. Separately, dissolve 1 mg of
of ethanol (99.5), and use this solution as the standard solu-
atractylenolide III for thin-layer chromatography in 2 mL
tion. Perform the test with these solutions as directed under
of methanol, and use this solution as the standard solution.
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Perform the test with these solutions as directed under
ple solution and 10 mL of the standard solution on a plate of
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
silica gel for thin-layer chromatography. Develop the plate
sample solution and standard solution on a plate of silica
with a mixture of 1-butanol, water and acetic acid (100)
gel for thin-layer chromatography. Develop the plate with a
(4:2:1) to a distance of about 7 cm, and air-dry the plate.
mixture of ethyl acetate and hexane (1:1) to a distance of
Spray evenly Dragendorff's TS for spraying on the plate,
about 7 cm, and air-dry the plate. Spray evenly diluted sul-
and air-dry the plate. Then spray evenly sodium nitrite TS
furic acid on the plate, heat the plate at 1059C for 5
on the plate: one of the several spots obtained from the
minutes, and examine under ultraviolet light (main wave-
sample solution has the same color tone and Rf value with
length: 365 nm): one of the several spots obtained from the
the yellow-brown spot from the standard solution (Proc-
sample solution has the same color tone and Rf value with
essed Aconite Root or Powdered Processed Aconite Root).
the blue-white fluorescent spot from the standard solution
(Atractylodes Rhizome).
(3) For preparation prescribed Atractylodes Lancea
Rhizome—To 2.0 g of Shimbuto Extract, add 10 mL of Shosaikoto Extract
water, shake, then add 25 mL of hexane, and shake.
小柴胡湯エキス
Separate the hexane layer, evaporate the layer under
reduced pressure, add 2 mL of hexane to the residue, and
Change the Origin/limits of content, Identifica-
use this solution as the sample solution. Perform the test
tion and Assay (3) as follows:
with the sample solution as directed under Thin-layer Chro-
matography <2.03>. Spot 20 mL of the sample solution on a
Shosaikoto Extract contains not less than 2 mg and
plate of silica gel with fluorescent indicator for thin-layer
not more than 8 mg of saikosaponin b2, not less than
chromatography. Develop the plate with a mixture of
80 mg and not more than 240 mg of baicalin
hexane and acetone (7:1) to a distance of about 7 cm, and
(C21H18O11: 446.36), and not less than 14 mg and not
air-dry the plate. Examine under ultraviolet light (main
more than 42 mg of glycyrrhizic acid (C42H62O16:
wavelength: 254 nm): a dark purple spot is observed at an
822.93), per extract prepared with the amount speci-
Rf value of about 0.5. The spot shows a greenish brown
fied in the Method of preparation.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2792 Crude Drugs and Related Drugs Supplement I, JP XVII

Identification (1) Shake 2.0 g of the dry extract (or 6.0 g mL of 1-butanol, shake, centrifuge, and use the supernatant
of the viscous extract) with 10 mL of sodium hydroxide TS, liquid as the sample solution. Separately, dissolve 1 mg of
then add 5 mL of 1-butanol, shake, centrifuge, and use the Ginsenoside Rb1 RS or ginsenoside Rb1 for thin-layer chro-
supernatant liquid as the sample solution. Separately, dis- matography in 1 mL of methanol, and use this solution as
solve 1 mg of saikosaponin b2 for thin-layer chromatog- the standard solution. Perform the test with these solutions
raphy in 1 mL of methanol, and use this solution as the as directed under Thin-layer Chromatography <2.03>. Spot
standard solution. Perform the test with these solutions as 10 mL of the sample solution and 2 mL of the standard solu-
directed under Thin-layer Chromatography <2.03>. Spot 10 tion on a plate of silica gel for thin-layer chromatography.
mL of the sample solution and 2 mL of the standard solution Develop the plate with a mixture of ethyl acetate, 1-
on a plate of silica gel for thin-layer chromatography. De- propanol, water and acetic acid (100) (7:5:4:1) to a distance
velop the plate with a mixture of ethyl acetate, ethanol of about 7 cm, and air-dry the plate. Spray evenly vanillin-
(99.5) and water (8:2:1) to a distance of about 7 cm, and air- sulfuric acid-ethanol TS for spraying on the plate, heat the
dry the plate. Spray evenly 4-dimethylaminobenzaldehyde plate at 1059C for 5 minutes, and allow to cool: one of the
TS for spraying on the plate, heat the plate at 1059C for 5 several spots obtained from the sample solution has the
minutes, and examine under ultraviolet light (main wave- same color tone and Rf value with the blue-purple spot
length: 365 nm): one of the several spots obtained from the from the standard solution (Ginseng).
sample solution has the same color tone and Rf value with (5) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
the yellow fluorescent spot from the standard solution extract) with 10 mL of water, then add 5 mL of 1-butanol,
(Bupleurum Root). shake, centrifuge, and use the supernatant liquid as the
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous sample solution. Separately, dissolve 1 mg of liquiritin for
extract) with 10 mL of water, then add 25 mL of diethyl thin-layer chromatography in 1 mL of methanol, and use
ether, and shake. Separate the diethyl ether layer, evaporate this solution as the standard solution. Perform the test with
the layer under reduced pressure, add 2 mL of diethyl ether these solutions as directed under Thin-layer Chromatogra-
to the residue, and use this solution as the sample solution. phy <2.03>. Spot 1 mL each of the sample solution and
Separately, dissolve 1 mg of [6]-gingerol for thin-layer chro- standard solution on a plate of silica gel for thin-layer chro-
matography in 1 mL of methanol, and use this solution as matography. Develop the plate with a mixture of ethyl ace-
the standard solution. Perform the test with these solutions tate, methanol and water (20:3:2) to a distance of about 7
as directed under Thin-layer Chromatography <2.03>. Spot cm, and air-dry the plate. Spray evenly dilute sulfuric acid
15 mL of the sample solution and 5 mL of the standard solu- on the plate, heat the plate at 1059C for 5 minutes, and exa-
tion on a plate of silica gel for thin-layer chromatography. mine under ultraviolet light (main wavelength: 365 nm): one
Develop the plate with a mixture of ethyl acetate and hexane of the several spots obtained from the sample solution has
(1:1) to a distance of about 7 cm, and air-dry the plate. the same color tone and Rf value with the yellow-green fluo-
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying rescent spot from the standard solution (Glycyrrhiza).
on the plate, heat the plate at 1059C for 5 minutes, allow to
Assay
cool, and spray water: one of the several spots obtained
(3) Glycyrrhizic acid—Perform the test according to the
from the sample solution has the same color tone and Rf
following i) or ii).
value with the blue-green to grayish green spot from the
i) Weigh accurately about 0.5 g of the dry extract (or an
standard solution (Ginger).
amount of the viscous extract, equivalent to about 0.5 g of
(3) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
dried substance), add exactly 50 mL of diluted methanol (1
extract) with 10 mL of water, then add 25 mL of diethyl
in 2), shake for 15 minutes, filter, and use the filtrate as the
ether, and shake. Separate the diethyl ether layer, evaporate
sample solution. Separately, weigh accurately about 10 mg
the layer under reduced pressure, add 2 mL of diethyl ether
of Glycyrrhizic Acid RS (separately determine the water
to the residue, and use this solution as the sample solution.
<2.48> by coulometric titration, using 10 mg), dissolve in
Separately, dissolve 1 mg of wogonin for thin-layer chroma-
diluted methanol (1 in 2) to make exactly 100 mL, and use
tography in 1 mL of methanol, and use this solution as the
this solution as the standard solution. Perform the test with
standard solution. Perform the test with these solutions as
exactly 10 mL each of the sample solution and standard so-
directed under Thin-layer Chromatography <2.03>. Spot 20
lution as directed under Liquid Chromatography <2.01> ac-
mL of the sample solution and 2 mL of the standard solution
cording to the following conditions, and determine the peak
on a plate of silica gel for thin-layer chromatography. De-
areas, AT and AS, of glycyrrhizic acid in each solution.
velop the plate with a mixture of ethyl acetate, hexane and
acetic acid (100) (10:10:1) to a distance of about 7 cm, air- Amount (mg) of glycyrrhizic acid (C42H62O16)
dry the plate. Spray evenly iron (III) chloride-methanol TS = MS × AT/AS × 1/2
on the plate: one of the several spots obtained from the
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
sample solution has the same color tone and Rf value with
lated on the anhydrous basis
the yellow-brown spot from the standard solution (Scutel-
laria Root). Operating conditions—
(4) Shake 2.0 g of the dry extract (or 6.0 g of the viscous Detector: An ultraviolet absorption photometer (wave-
extract) with 10 mL of sodium hydroxide TS, then add 5 length: 254 nm).

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2793

Column: A stainless steel column 4.6 mm in inside diame- System suitability—


ter and 15 cm in length, packed with octadecylsilanized System repeatability: Proceed as directed in the system
silica gel for liquid chromatography (5 mm in particle diame- suitability in i).
ter). System performance: Dissolve 5 mg of monoammonium
Column temperature: A constant temperature of about glycyrrhizinate for resolution check in 20 mL of dilute
409C. ethanol. When the procedure is run with 10 mL of this solu-
Mobile phase: Dissolve 3.85 g of ammonium acetate in tion under the above operating conditions, the resolution
720 mL of water, and add 5 mL of acetic acid (100) and 280 between the peak having the relative retention time of about
mL of acetonitrile. 0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
Flow rate: 1.0 mL per minute (the retention time of not less than 1.5.
glycyrrhizic acid is about 15 minutes).
System suitability—
System performance: Dissolve 5 mg of monoammonium Shoseiryuto Extract
glycyrrhizinate for resolution check in 20 mL of dilute
ethanol. When the procedure is run with 10 mL of this solu- 小青竜湯エキス
tion under the above operating conditions, the resolution
between the peak having the relative retention time of about Change the Origin/limits of content, Identifica-
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is tion (2)-(8) and Assay (1), (3) as follows:
not less than 1.5. Dissolve 1 mg of baicalein for resolution
check in 50 mL of methanol. To 2 mL of this solution add 2 Shoseiryuto Extract contains not less than 8 mg and
mL of the standard solution. When the procedure is run not more than 24 mg of the total alkaloids [ephedrine
with 10 mL of this solution under the above operating condi- (C10H15NO: 165.23) and pseudoephedrine (C10H15NO:
tions, the resolution between the peaks of glycyrrhizic acid 165.23)], not less than 26 mg and not more than 78
and baicalein is not less than 1.5. mg of paeoniflorin (C23H28O11: 480.46), and not less
System repeatability: When the test is repeated 6 times than 14 mg and not more than 42 mg of glycyrrhizic
with 10 mL of the standard solution under the above operat- acid (C42H62O16: 822.93), per extract prepared with
ing conditions, the relative standard deviation of the peak the amount specified in the Method of preparation.
area of glycyrrhizic acid is not more than 1.5z.
Identification
ii) Weigh accurately about 0.5 g of the dry extract (or an
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
amount of the viscous extract, equivalent to about 0.5 g of
extract) with 10 mL of water, add 10 mL of 1-butanol,
the dried substance), add 20 mL of ethyl acetate and 10 mL
shake, centrifuge, and use the supernatant liquid as the
of water, and shake for 10 minutes. After centrifugation,
sample solution. Separately, dissolve 1 mg of Paeoniflorin
remove the upper layer, add 20 mL of ethyl acetate, proceed
RS or paeoniflorin for thin-layer chromatography in 1 mL
in the same manner as described above, and remove the up-
of methanol, and use this solution as the standard solution.
per layer. To the resultant aqueous layer add 10 mL of
Perform the test with these solutions as directed under
methanol, shake for 30 minutes, centrifuge, and take the su-
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
pernatant liquid. To the residue add 20 mL of diluted meth-
sample solution and standard solution on a plate of silica
anol (1 in 2), shake for 5 minutes, centrifuge, and take the
gel for thin-layer chromatography. Develop the plate with a
supernatant liquid. Combine these supernatant liquids, add
mixture of ethyl acetate, methanol and water (20:3:2) to a
diluted methanol (1 in 2) to make exactly 50 mL, and use
distance of about 7 cm, and air-dry the plate. Spray evenly
this solution as the sample solution. Separately, weigh accu-
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
heat the plate at 1059C for 5 minutes: one of the several
termine the water <2.48> by coulometric titration, using 10
spots obtained from the sample solution has the same color
mg), dissolve in diluted methanol (1 in 2) to make exactly
tone and Rf value with the purple spot from the standard
100 mL, and use this solution as the standard solution. Per-
solution (Peony Root).
form the test with exactly 10 mL each of the sample solution
(3) For preparation prescribed Processed Ginger—
and standard solution as directed under Liquid Chromatog-
Shake 1.0 g of the dry extract (or 3.0 g of the viscous ex-
raphy <2.01> according to the following conditions, and de-
tract) with 10 mL of water, add 25 mL of diethyl ether, and
termine the peak areas, AT and AS, of glycyrrhizic acid in
shake. Separate the diethyl ether layer, evaporate the layer
each solution.
under reduced pressure, add 2 mL of diethyl ether to the
Amount (mg) of glycyrrhizic acid (C42H62O16) residue, and use this solution as the sample solution. Sepa-
= MS × AT/AS × 1/2 rately, dissolve 1 mg of [6]-shogaol for thin-layer chroma-
tography in 1 mL of methanol, and use this solution as the
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
standard solution. Perform the test with these solutions as
lated on the anhydrous basis
directed under Thin-layer Chromatography <2.03>. Spot 20
Operating conditions— mL of the sample solution and 1 mL of the standard solution
Proceed as directed in the operating conditions in i). on a plate of silica gel for thin-layer chromatography. De-

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2794 Crude Drugs and Related Drugs Supplement I, JP XVII

velop the plate with a mixture of cyclohexane and ethyl ace- Perform the test with these solutions as directed under
tate (2:1) to a distance of about 7 cm, and air-dry the plate. Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Spray evenly 4-dimethylaminobenzaldehyde TS for spraying ple solution and 2 mL the standard solution on a plate of
on the plate, heat the plate at 1059C for 5 minutes, allow to silica gel for thin-layer chromatography. Develop the plate
cool, and spray water: one of the several spots obtained with a mixture of hexane and ethyl acetate (2:1) to a dis-
from the sample solution has the same color tone and Rf tance of about 7 cm, and air-dry the plate. Spray evenly 2,4-
value with the blue-green to grayish green spot from the dinitrophenylhydrazine TS on the plate: one of the several
standard solution (Processed Ginger). spots obtained from the sample solution has the same color
(4) For preparation prescribed Ginger—Shake 1.0 g of tone and Rf value with the yellow-orange spot from the
the dry extract (or 3.0 g of the viscous extract) with 10 mL standard solution.
of water, add 25 mL of diethyl ether, and shake. Separate ii) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
the diethyl ether layer, evaporate the layer under reduced extract) with 10 mL of water, then add 5 mL of hexane,
pressure, add 2 mL of diethyl ether to the residue, and use shake, centrifuge, and use the supernatant liquid as the
this solution as the sample solution. Separately, dissolve 1 sample solution. Separately, dissolve 1 mg of (E )-2-methox-
mg of [6]-gingerol for thin-layer chromatography in 1 mL ycinnamaldehyde for thin-layer chromatography in 1 mL of
of methanol, and use this solution as the standard solution. methanol, and use this solution as the standard solution.
Perform the test with these solutions as directed under Perform the test with these solutions as directed under
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam- Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
ple solution and 5 mL of the standard solution on a plate of ple solution and 2 mL the standard solution on a plate of
silica gel for thin-layer chromatography. Develop the plate silica gel for thin-layer chromatography. Develop the plate
with a mixture of ethyl acetate and hexane (1:1) to a dis- with a mixture of hexane and ethyl acetate (2:1) to a dis-
tance of about 7 cm, and air-dry the plate. Spray evenly 4- tance of about 7 cm, and air-dry the plate. Examine under
dimethylaminobenzaldehyde TS for spraying on the plate, ultraviolet light (main wavelength: 365 nm): one of the
heat the plate at 1059C for 5 minutes, allow to cool, and several spots obtained from the sample solution has the
spray water: one of the several spots obtained from the sam- same color tone and Rf value with the blue-white fluores-
ple solution has the same color tone and Rf value with the cent spot from the standard solution.
blue-green to grayish green spot from the standard solution (7) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
(Ginger). extract) with 10 mL of water, then add 25 mL of diethyl
(5) Shake 1.0 g of the dry extract (or 3.0 g of the viscous ether, and shake. Separate the diethyl ether layer, evaporate
extract) with 10 mL of water, add 10 mL of 1-butanol, the layer under reduced pressure, add 2 mL of diethyl ether
shake, centrifuge, and use the supernatant liquid as the to the residue, and use this solution as the sample solution.
sample solution. Separately, dissolve 1 mg of liquiritin for Separately, dissolve 1 mg of asarinin for thin-layer chroma-
thin-layer chromatography in 1 mL of methanol, and use tography in 1 mL of methanol, and use this solution as the
this solution as the standard solution. Perform the test with standard solution. Perform the test with these solutions as
these solutions as directed under Thin-layer Chromatogra- directed under Thin-layer Chromatography <2.03>. Spot 20
phy <2.03>. Spot 1 mL each of the sample solution and mL of the sample solution and 5 mL of the standard solution
standard solution on a plate of silica gel for thin-layer chro- on a plate of silica gel for thin-layer chromatography. De-
matography. Develop the plate with a mixture of ethyl ace- velop the plate with a mixture of hexane and ethyl acetate
tate, methanol and water (20:3:2) to a distance of about 7 (2:1) to a distance of about 7 cm, and air-dry the plate.
cm, and air-dry the plate. Spray evenly dilute sulfuric acid Spray evenly dilute sulfuric acid on the plate, and heat the
on the plate, heat the plate at 1059C for 5 minutes, and exa- plate at 1059C for 5 minutes: one of the several spots ob-
mine under ultraviolet light (main wavelength: 365 nm): one tained from the sample solution has the same color tone and
of the several spots obtained from the sample solution has Rf value with the yellow-brown spot from the standard so-
the same color tone and Rf value with the yellow-green fluo- lution (Asiasarum Root).
rescent spot from the standard solution (Glycyrrhiza). (8) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
(6) Perform the test according to the following i) or ii) extract) with 10 mL of sodium hydroxide TS, then add 25
(Cinnamon Bark). mL of diethyl ether, and shake. Separate the diethyl ether
i) Put 10 g of the dry extract (or 30 g of the viscous ex- layer, evaporate the layer under reduced pressure, add 2 mL
tract) in a 300-mL hard-glass flask, add 100 mL of water of diethyl ether to the residue, and use this solution as the
and 1 mL of silicone resin, connect the apparatus for essen- sample solution. Separately, dissolve 1 mg of schisandrin
tial oil determination, and heat to boil under a reflux con- for thin-layer chromatography in 1 mL of methanol, and
denser. The graduated tube of the apparatus is to be previ- use this solution as the standard solution. Perform the test
ously filled with water to the standard line, and 2 mL of with these solutions as directed under Thin-layer Chroma-
hexane is added to the graduated tube. After heating under tography <2.03>. Spot 10 mL of the sample solution and 5
reflux for 1 hour, separate the hexane layer, and use the mL of the standard solution on a plate of silica gel with fluo-
layer as the sample solution. Separately, dissolve 1 mg of rescent indicator for thin-layer chromatography. Develop
(E )-cinnamaldehyde for thin-layer chromatography in 1 mL the plate with a mixture of ethyl acetate, hexane and acetic
of methanol, and use this solution as the standard solution. acid (100) (10:10:1) to a distance of about 7 cm, and air-dry

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2795

the plate. Examine under ultraviolet light (main wavelength: hydrochloride in diluted methanol (1 in 2) to make 10 mL.
254 nm): one of the several spots obtained from the sample When the procedure is run with 10 mL of this solution under
solution has the same color tone and Rf value with the blue- the above operating conditions, pseudoephedrine and ephe-
purple spot from the standard solution (Schisandra Fruit). drine are eluted in this order with the resolution between
these peaks being not less than 1.5.
Assay
System repeatability: When the test is repeated 6 times
(1) Total alkaloids (ephedrine and pseudoephedrine)—
with 10 mL of the standard solution under the above operat-
Weigh accurately about 0.5 g of the dry extract (or an
ing conditions, the relative standard deviation of the peak
amount of the viscous extract, equivalent to about 0.5 g of
area of ephedrine is not more than 1.5z.
dried substance), add 20 mL of diethyl ether, shake, then
(3) Glycyrrhizic acid—Perform the test according to the
add 3.0 mL of 0.1 mol/L hydrochloric acid TS, and shake
following i) or ii).
for 10 minutes. After centrifugation, remove the upper
i) Weigh accurately about 0.5 g of the dry extract (or an
layer, add 20 mL of diethyl ether, proceed in the same man-
amount of the viscous extract, equivalent to about 0.5 g of
ner as described above, and remove the upper layer. To the
the dried substance), add exactly 50 mL of diluted methanol
aqueous layer add 1.0 mL of ammonia TS and 20 mL of
(1 in 2), shake for 15 minutes, filter, and use the filtrate as
diethyl ether, shake for 30 minutes, centrifuge, and separate
the sample solution. Separately, weigh accurately about 10
the supernatant liquid. In addition, repeat twice in the same
mg of Glycyrrhizic Acid RS, (separately determine the water
manner for the aqueous layer using 1.0 mL of ammonia TS
<2.48> by coulometric titration, using 10 mg), dissolve in
and 20 mL of diethyl ether. Combine all the supernatant liq-
diluted methanol (1 in 2) to make exactly 100 mL, and use
uids, evaporate the solvent under reduced pressure, dissolve
this solution as the standard solution. Perform the test with
the residue in diluted methanol (1 in 2) to make exactly 50
exactly 10 mL each of the sample solution and standard so-
mL. Centrifuge this solution, and use the supernatant liquid
lution as directed under Liquid Chromatography <2.01> ac-
as the sample solution. Separately, weigh accurately about
cording to the following conditions, and determine the peak
10 mg of ephedrine hydrochloride for assay of crude drugs,
areas, AT and AS, of glycyrrhizic acid in each solution.
previously dried at 1059 C for 3 hours, and dissolve in
diluted methanol (1 in 2) to make exactly 100 mL. Pipet 10 Amount (mg) of glycyrrhizic acid (C42H62O16)
mL of this solution, add diluted methanol (1 in 2) to make = MS × AT/AS × 1/2
exactly 50 mL, and use this solution as the standard solu-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
tion. Perform the test with exactly 10 mL each of the sample
lated on the anhydrous basis
solution and standard solution as directed under Liquid
Chromatography <2.01> according to the following condi- Operating conditions—
tions, and determine the peak areas, ATE and ATP, of ephe- Detector: An ultraviolet absorption photometer (wave-
drine and pseudoephedrine obtained from the sample solu- length: 254 nm).
tion, and the peak area, AS, of ephedrine from the standard Column: A stainless steel column 4.6 mm in inside diame-
solution. ter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle diame-
Amount (mg) of total alkaloids [ephedrine (C10H15NO)
ter).
and pseudoephedrine (C10H15NO)]
Column temperature: A constant temperature of about
= MS × (ATE + ATP)/AS × 1/10 × 0.819
409C.
MS: Amount (mg) of ephedrine hydrochloride for assay Mobile phase: Dissolve 3.85 g of ammonium acetate in
of crude drugs taken 720 mL of water, and add 5 mL of acetic acid (100) and 280
mL of acetonitrile.
Operating conditions—
Flow rate: 1.0 mL per minute (the retention time of
Detector: An ultraviolet absorption photometer (wave-
glycyrrhizic acid is about 15 minutes).
length: 210 nm).
System suitability—
Column: A stainless steel column 4.6 mm in inside diame-
System performance: Dissolve 5 mg of monoammonium
ter and 15 cm in length, packed with octadecylsilanized
glycyrrhizinate for resolution check in 20 mL of dilute
silica gel for liquid chromatography (5 mm in particle diame-
ethanol. When the procedure is run with 10 mL of this solu-
ter).
tion under the above operating conditions, the resolution
Column temperature: A constant temperature of about
between the peak having the relative retention time of about
409C.
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
Mobile phase: To 5 g of sodium lauryl sulfate add 350
not less than 1.5. Dissolve 1 mg of (E )-cinnamaldehyde for
mL of acetonitrile, shake, and add 650 mL of water and 1
thin-layer chromatography in 50 mL of methanol. To 2 mL
mL of phosphoric acid to dissolve lauryl sulfate.
of this solution add 2 mL of the standard solution. When
Flow rate: 1.0 mL per minute (the retention time of ephe-
the procedure is run with 10 mL of this solution under the
drine is about 27 minutes).
above operating conditions, the resolution between the
System suitability—
peaks of glycyrrhizic acid and (E )-cinnamaldehyde is not
System performance: Dissolve 1 mg each of ephedrine hy-
less than 1.5.
drochloride for assay of crude drugs and pseudoephedrine

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2796 Crude Drugs and Related Drugs Supplement I, JP XVII

System repeatability: When the test is repeated 6 times Thin-layer Chromatography <2.03>. Spot 5 mL each of the
with 10 mL of the standard solution under the above operat- sample solution and standard solution on a plate of silica
ing conditions, the relative standard deviation of the peak gel with fluorescent indicator for thin-layer chromatogra-
area of glycyrrhizic acid is not more than 1.5z. phy. Develop the plate with a mixture of diethyl ether,
ii) Weigh accurately about 0.5 g of the dry extract (or an hexane and formic acid (5:5:1) to a distance of about 7 cm,
amount of the viscous extract, equivalent to about 0.5 g of and air-dry the plate. Examine under ultraviolet light (main
the dried substance), add 20 mL of diethyl ether and 10 mL wavelength: 254 nm): two of the several spots obtained
of water, and shake for 10 minutes. After centrifugation, from the sample solution have the same color tone and Rf
remove the upper layer, add 20 mL of diethyl ether, proceed value with the spots from the standard solution.
in the same manner as described above, and remove the
upper layer. To the resultant aqueous layer add 10 mL of
methanol, shake for 30 minutes, centrifuge, and take the su- Tokakujokito Extract
pernatant liquid. To the residue add 20 mL of diluted meth-
anol (1 in 2), shake for 5 minutes, centrifuge, and take the 桃核承気湯エキス
supernatant liquid. Combine these supernatant liquids, add
diluted methanol (1 in 2) to make exactly 50 mL, and use Change the Origin/limits of content, Identifica-
this solution as the sample solution. Separately, weigh accu- tion (4) and Assay (5) as follows:
rately about 10 mg of Glycyrrhizic Acid RS (separately de-
termine the water <2.48> by coulometric titration, using 10 Tokakujokito Extract contains not less than 38 mg
mg), dissolve in diluted methanol (1 in 2) to make exactly and not more than 152 mg of amygdalin, not less than
100 mL, and use this solution as the standard solution. Per- 1 mg and not more than 4 mg of (E )-cinnamic acid,
form the test with exactly 10 mL each of the sample solution not less than 3 mg of sennosides A (C42H38O20:
and standard solution as directed under Liquid Chromatog- 862.74) or not less than 9 mg of rhein, and not less
raphy <2.01> according to the following conditions, and de- than 10 mg and not more than 30 mg of glycyrrhizic
termine the peak areas, AT and AS, of glycyrrhizic acid in acid (C42H62O16: 822.93), per extract prepared with
each solution. the amount specified in the Method of preparation.
Amount (mg) of glycyrrhizic acid (C42H62O16) Identification
= MS × AT/AS × 1/2 (4) To 1.0 g of Tokakujokito Extract add 10 mL of
water, shake, then add 10 mL of 1-butanol, shake, centri-
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-
fuge, and use the supernatant liquid as the sample solution.
lated on the anhydrous basis
Separately, dissolve 1 mg of liquiritin for thin-layer chroma-
Operating conditions— tography in 1 mL of methanol, and use this solution as the
Proceed as directed in the operating conditions in i). standard solution. Perform the test with these solutions as
System suitability— directed under Thin-layer Chromatography <2.03>. Spot 1
System repeatability: Proceed as directed in the system mL each of the sample solution and standard solution on a
suitability in i). plate of silica gel for thin-layer chromatography. Develop
System performance: Dissolve 5 mg of monoammonium the plate with a mixture of ethyl acetate, methanol and
glycyrrhizinate for resolution check in 20 mL of dilute water (20:3:2) to a distance of about 7 cm, and air-dry the
ethanol. When the procedure is run with 10 mL of this solu- plate. Spray evenly dilute sulfuric acid on the plate, heat the
tion under the above operating conditions, the resolution plate at 1059 C for 5 minutes, and examine under ultraviolet
between the peak having the relative retention time of about light (main wavelength: 365 nm): one of the several spots
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is obtained from the sample solution has the same color tone
not less than 1.5. and Rf value with the yellow-green fluorescent spot from
the standard solution (Glycyrrhiza).

Assay
Powdered Sweet Hydrangea Leaf (5) Glycyrrhizic acid—Weigh accurately about 0.5 g of
Tokakujokito Extract, add 20 mL of ethyl acetate and 10
アマチャ末
mL of water, and shake for 10 minutes. After centrifuga-
tion remove the upper layer, add 20 mL of ethyl acetate,
Change the Identification as follows:
proceed in the same manner as described above, and remove
Identification To 1.0 g of Powdered Sweet Hydrangea the upper layer. To the resultant aqueous layer add 10 mL
Leaf add 10 mL of methanol, shake for 10 minutes, centri- of methanol, shake for 30 minutes, centrifuge, and take the
fuge, and use the supernatant liquid as the sample solution. supernatant liquid. To the residue add 20 mL of diluted
Separately, dissolve 2 mg of sweet hydrangea leaf dihydro- methanol (1 in 2), shake for 5 minutes, centrifuge, and take
isocoumarin for thin-layer chromatography in 1 mL of the supernatant liquid. Combine these supernatant liquids,
methanol, and use this solution as the standard solution. add diluted methanol (1 in 2) to make exactly 50 mL, and
Perform the test with these solutions as directed under use this solution as the sample solution. Separately, weigh

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2797

accurately about 10 mg of Glycyrrhizic Acid RS (separately <2.03>. Spot 10 mL each of the sample solution and standard
determine the water <2.48> by coulometric titration, using solution on a plate of silica gel for thin-layer chromatogra-
10 mg), dissolve in diluted methanol (1 in 2) to make exactly phy. Develop the plate with a mixture of ethyl acetate and
100 mL, and use this solution as the standard solution. Per- hexane (1:1) to a distance of about 7 cm, and air-dry the
form the test with exactly 10 mL each of the sample solution plate. Examine under ultraviolet light (main wavelength:
and standard solution as directed under Liquid Chromatog- 365 nm): one of the several spots obtained from the sample
raphy <2.01> according to the following conditions, and de- solution has the same color tone and Rf value with the blue-
termine the peak areas, AT and AS, of glycyrrhizic acid in white fluorescent spot from the standard solution (Japanese
each solution. Angelica Root and Cnidium Rhizome).
(2) Shake 1.0 g of the dry extract (or 3.0 g of the viscous
Amount (mg) of glycyrrhizic acid (C42H62O16)
extract) with 10 mL of water, add 10 mL of 1-butanol,
= MS × AT/AS × 1/2
shake, centrifuge, and use the supernatant liquid as the
MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu- sample solution. Separately, dissolve 1 mg of Paeoniflorin
lated on the anhydrous basis RS or paeoniflorin for thin-layer chromatography in 1 mL
of methanol, and use this solution as the standard solution.
Operating conditions—
Perform the test with these solutions as directed under
Detector: An ultraviolet absorption photometer (wave-
Thin-layer Chromatography <2.03>. Spot 5 mL each of the
length: 254 nm).
sample solution and standard solution on a plate of silica
Column: A stainless steel column 4.6 mm in inside diame-
gel for thin-layer chromatography. Develop the plate with a
ter and 15 cm in length, packed with octadecylsilanized
mixture of ethyl acetate, methanol and water (20:3:2) to a
silica gel for liquid chromatography (5 mm in particle diame-
distance of about 7 cm, and air-dry the plate. Spray evenly
ter).
4-methoxybenzaldehyde-sulfuric acid TS on the plate, and
Column temperature: A constant temperature of about
heat the plate at 1059C for 5 minutes: one of the several
409C.
spots obtained from the sample solution has the same color
Mobile phase: Dissolve 3.85 g of ammonium acetate in
tone and Rf value with the purple spot from the standard
720 mL of water, and add 5 mL of acetic acid (100) and 280
solution (Peony Root).
mL of acetonitrile.
(3) For preparation prescribed Atractylodes Rhizome—
Flow rate: 1.0 mL per minute (the retention time of
Shake 1.0 g of the dry extract (or 3.0 g of the viscous ex-
glycyrrhizic acid is about 15 minutes).
tract) with 10 mL of water, add 25 mL of diethyl ether, and
System suitability—
shake. Separate the diethyl ether layer, evaporate the layer
System performance: Dissolve 5 mg of monoammonium
under reduced pressure, add 2 mL of diethyl ether to the
glycyrrhizinate for resolution check in 20 mL of dilute
residue, and use this solution as the sample solution. Sepa-
ethanol. When the procedure is run with 10 mL of this solu-
rately, dissolve 1 mg of atractylenolide III for thin-layer
tion under the above operating conditions, the resolution
chromatography in 2 mL of methanol, and use this solution
between the peak having the relative retention time of about
as the standard solution. Perform the test with these solu-
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
tions as directed under Thin-layer Chromatography <2.03>.
not less than 1.5.
Spot 5 mL each of the sample solution and standard solution
System repeatability: When the test is repeated 6 times
on a plate of silica gel for thin-layer chromatography. De-
with 10 mL of the standard solution under the above operat-
velop the plate with a mixture of ethyl acetate and hexane
ing conditions, the relative standard deviation of the peak
(1:1) to a distance of about 7 cm, and air-dry the plate.
area of glycyrrhizic acid is not more than 1.5z.
Spray evenly dilute sulfuric acid on the plate, heat the plate
at 1059 C for 5 minutes, and examine under ultraviolet light
(main wavelength: 365 nm): one of the several spots ob-
Tokishakuyakusan Extract tained from the sample solution has the same color tone and
Rf value with the blue-white fluorescent spot from the
当帰芍薬散エキス
standard solution (Atractylodes Rhizome).
(4) For preparation prescribed Atractylodes Lancea
Change the Identification as follows:
Rhizome—Shake 2.0 g of the dry extract (or 6.0 g of the vis-
Identification (1) Shake 1.0 g of the dry extract (or 3.0 g cous extract) with 10 mL of water, add 25 mL of hexane,
of the viscous extract) with 15 mL of water and 5 mL of 0.1 and shake. Separate the hexane layer, evaporate the layer
mol/L hydrochloric acid TS, add 25 mL of diethyl ether, under reduced pressure, add 0.5 mL of hexane to the
and shake. Separate the diethyl ether layer, evaporate the residue, and use this solution as the sample solution. Per-
layer under reduced pressure, add 2 mL of diethyl ether to form the test with the sample solution as directed under
the residue, and use this solution as the sample solution. Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Separately, dissolve 1 mg of (Z )-ligustilide for thin-layer ple solution on a plate of silica gel with fluorescent indicator
chromatography in 10 mL of methanol, and use this solu- for thin-layer chromatography. Develop the plate with a
tion as the standard solution. Perform the test with these mixture of hexane and acetone (7:1) to a distance of about 7
solutions as directed under Thin-layer Chromatography cm, and air-dry the plate. Examine under ultraviolet light

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2798 Crude Drugs and Related Drugs Supplement I, JP XVII

(main wavelength: 254 nm): a dark purple spot is observed


at an Rf value of about 0.5. The spot shows a greenish Powdered Turmeric
brown color after being sprayed evenly 4-dimethylamino-
benzaldehyde TS for spraying, heated at 1059C for 5 ウコン末
minutes, and allowed to cool (Atractylodes Lancea Rhi-
zome). Change the latin name as follows:
(5) Shake 2.0 g of the dry extract (or 6.0 g of the viscous
Curcumae Longae Rhizoma Purveratum
extract) with 20 mL of water and 2 mL of ammonia solution
(28), add 20 mL of a mixture of hexane and ethyl acetate
(20:1), shake, and centrifuge. Separate the upper layer,
evaporate the layer under reduced pressure, add 2 mL of Yokukansan Extract
methanol to the residue, and use this solution as the sample
抑肝散エキス
solution. Separately, dissolve 1 mg of alisol A for thin-layer
chromatography in 1 mL of methanol, and use this solution
Change the Origin/limits of content, Identifica-
as the standard solution. Perform the test with these solu-
tion (4) and (6), and Assay (3) as follows:
tions as directed under Thin-layer Chromatography <2.03>.
Spot 10 mL each of the sample solution and standard solu-
Yokukansan Extract contains not less than 0.15 mg
tion on a plate of silica gel for thin-layer chromatography.
of total alkaloids (rhyncophylline and hirsutine), not
Develop the plate with a mixture of ethyl formate, water
less than 0.6 mg and not more than 2.4 mg of sai-
and formic acid (30:1:1) to a distance of about 7 cm, and
kosaponin b2, and not less than 10 mg and not more
air-dry the plate. Spray evenly 4-methoxybenzaldehyde-sul-
than 30 mg of glycyrrhizic acid (C42H62O16: 822.93),
furic acid-acetic acid TS on the plate, heat the plate at
per extract prepared with the amount specified in the
1059C for 5 minutes, allow to cool, and examine under ul-
Method of preparation.
traviolet light (main wavelength: 365 nm): one of the several
spots obtained from the sample solution has the same color Identification
tone and Rf value with the yellow fluorescent spot from the (4) For preparation prescribed Atractylodes Lancea
standard solution (Alisma Tuber). Rhizome—To 2.0 g of the dry extract (or 6.0 g of the vis-
cous extract) add 10 mL of water, shake, then add 25 mL of
hexane, and shake. Separate the hexane layer, evaporate the
Trichosanthes Root solvent under reduced pressure, add 2 mL of hexane to the
residue, and use this solution as the sample solution. Per-
カロコン form the test with the sample solution as directed under
Thin-layer Chromatography <2.03>. Spot 20 mL of the sam-
Add the following next to the Description: ple solution on a plate of silica gel with fluorescent indicator
for thin-layer chromatography. Develop the plate with a
Identification To 2.0 g of pulverized Trichosanthes Root
mixture of hexane and acetone (7:1) to a distance of about 7
add 5 mL of methanol, shake for 10 minutes, centrifuge,
cm, and air-dry the plate. Examine under ultraviolet light
and use the supernatant liquid as the sample solution. Per-
(main wavelength: 254 nm): a dark purple spot is observed
form the test with the sample solution as directed under
at an Rf value of about 0.5. The spot shows a greenish
Thin-layer Chromatography <2.03>. Spot 10 mL of the sam-
brown color after being sprayed evenly 4-dimethylamino-
ple solution on a plate of silica gel for thin-layer chromatog-
benzaldehyde TS for spraying on the plate, heated at 1059C
raphy. Develop the plate with a mixture of hexane, ethyl
for 5 minutes, and allowed to cool (Atractylodes Lancea
acetate and acetic acid (100) (20:10:1) to a distance of about
Rhizome).
7 cm, and air-dry the plate. Spray evenly dilute sulfuric acid
(6) To 1.0 g of the dry extract (or 3.0 g of the viscous
on the plate, heat at 1059C for 10 minutes, and examine
extract) add 10 mL of water, shake, then add 10 mL of 1-
under ultraviolet light (main wavelength: 365 nm): a light
butanol, shake, centrifuge, and use the supernatant liquid
yellow to light yellow-green fluorescent spot appears at an
as the sample solution. Separately, dissolve 1 mg of liquiri-
Rf value of about 0.4.
tin for thin-layer chromatography in 1 mL of methanol, and
use this solution as the standard solution. Perform the test
with these solutions as directed under Thin-layer Chroma-
Turmeric tography <2.03>. Spot 1 mL each of the sample solution and
standard solution on a plate of silica gel for thin-layer chro-
ウコン
matography. Develop the plate with a mixture of ethyl ace-
tate, methanol and water (20:3:2) to a distance of about 7
Change the latin name as follows:
cm, and air-dry the plate. Spray evenly dilute sulfuric acid
Curcumae Longae Rhizoma on the plate, heat the plate at 1059 C for 5 minutes, and exa-
mine under ultraviolet light (main wavelength: 365 nm): one
of the several spots obtained from the sample solution has

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Crude Drugs and Related Drugs 2799

the same color tone and Rf value with the yellow-green fluo-
rescent spot from the standard solution (Glycyrrhiza).

Assay
(3) Glycyrrhizic acid—Weigh accurately about 0.5 g of
the dry extract (or an amount of the viscous extract, equiva-
lent to about 0.5 g of the dried substance), add 20 mL of
diethyl ether and 10 mL of water, and shake for 10 minutes.
After centrifugation, remove the upper layer, add 20 mL of
diethyl ether, proceed in the same manner as described
above, and remove the upper layer. To the resultant aque-
ous layer add 10 mL of methanol, shake for 30 minutes,
centrifuge, and take the supernatant liquid. To the residue
add 20 mL of diluted methanol (1 in 2), shake for 5
minutes, centrifuge, and take the supernatant liquid. Com-
bine these supernatant liquids, add diluted methanol (1 in 2)
to make exactly 50 mL, and use this solution as the sample
solution. Separately, weigh accurately about 10 mg of
Glycyrrhizic Acid RS (separately determine the water <2.48>
by coulometric titration, using 10 mg), 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
10 mL each of the sample solution and standard solution as
directed under Liquid Chromatography <2.01> according to
the following conditions, and determine the peak areas, AT
and AS, of glycyrrhizic acid in each solution.

Amount (mg) of glycyrrhizic acid (C42H62O16)


= MS × AT/AS × 1/2

MS: Amount (mg) of Glycyrrhizic Acid RS taken, calcu-


lated on the anhydrous basis

Operating conditions—
Detector: An ultraviolet absorption photometer (wave-
length: 254 nm).
Column: A stainless steel column 4.6 mm in inside diame-
ter and 15 cm in length, packed with octadecylsilanized
silica gel for liquid chromatography (5 mm in particle diame-
ter).
Column temperature: A constant temperature of about
409C.
Mobile phase: Dissolve 3.85 g of ammonium acetate in
720 mL of water, and add 5 mL of acetic acid (100) and 280
mL of acetonitrile.
Flow rate: 1.0 mL per minute (the retention time of
glycyrrhizic acid is about 15 minutes).
System suitability—
System performance: Dissolve 5 mg of monoammonium
glycyrrhizinate for resolution check in 20 mL of dilute
ethanol. When the procedure is run with 10 mL of this solu-
tion under the above operating conditions, the resolution
between the peak having the relative retention time of about
0.9 to glycyrrhizic acid and the peak of glycyrrhizic acid is
not less than 1.5.
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
area of glycyrrhizic acid is not more than 1.5z.

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Infrared
Reference Spectra
Supplement I, JP XVII Infrared Reference Spectra 2801

Delete the following spectra:

Diclofenamide
Fluoxymesterone
Rokitamycin
Saccharin Sodium Hydrate
Tolazamide

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2802 Infrared Reference Spectra Supplement I, JP XVII

Add the following spectra:

Azosemide

Calcium Levofolinate Hydrate

Entacapone

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Infrared Reference Spectra 2803

Low Substituted Hydroxypropylcellulose

Mesalazine

Oxytetracycline Hydrochloride

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2804 Infrared Reference Spectra Supplement I, JP XVII

Pazufloxacin Mesilate

Pyridoxal Phosphate Hydrate

Tramadol Hydrochloride

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Infrared Reference Spectra 2805

Zonisamide

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Ultraviolet-visible
Reference Spectra
Supplement I, JP XVII Ultraviolet-visible Reference Spectra 2807

Delete the following spectra:

Diclofenamide
Fluoxymesterone
Gramicidin
Rokitamycin
Tolazamide

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2808 Ultraviolet-visible Reference Spectra Supplement I, JP XVII

Add the following spectra:

Azosemide

Calcium Levofolinate Hydrate

Doxycycline Hydrochloride Hydrate

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Ultraviolet-visible Reference Spectra 2809

Entacapone

Mesalazine

Pazufloxacin Mesilate

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
2810 Ultraviolet-visible Reference Spectra Supplement I, JP XVII

Pyridoxal Phosphate Hydrate

Sultamicillin Tosilate Hydrate

Tramadol Hydrochloride

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
Supplement I, JP XVII Ultraviolet-visible Reference Spectra 2811

Zonisamide

The JP Drugs are to be tested according to the provisions given in the pertinent monographs, General Notices, General Rules for Crude Drugs,
General Rules for Preparations, and General Tests for their conformity to the Japanese Pharmacopoeia. (See the General Notices 5.)
General Information
GENERAL INFORMATION
Therefore, by definition:
G2 Solid-state Properties Qr(x) = 0.90 when x = x90
Qr(x) = 0.50 when x = x50
Delete the following item: Qr(x) = 0.10 when x = x10

An alternative but less informative method of classifying


Laser Diffraction Measurement powder fineness is by use of the descriptive terms in the fol-
of Particle Size lowing table.

Classification of powders by fineness

Descriptive Cumulative distribution by volume


Powder Fineness term
x50 ( mm)
basis, Q 3 (x )

Coarse > 355 Q 3 (355) < 0.50


Change the following:
Moderately
This classification is harmonized with the European 180 – 355 Q 3 (180) < 0.50, Q 3 (355) ≧ 0.50
fine
Pharmacopoeia and the U.S. Pharmacopeia.
Fine 125 – 180 Q 3 (125) < 0.50, Q 3 (180) ≧ 0.50
A simple descriptive classification of powder fineness is
provided in this chapter. Sieving is most suitable where a Very fine ≦ 125 Q 3 (125) ≧ 0.50
majority of the particles are larger than about 75 mm,
although it can be used for some powders having smaller
particle sizes where the method can be validated. Light
diffraction is also a widely used technique for measuring the Powder Flow
size of a wide range of particles. Where the cumulative dis-
tribution has been determined by analytical sieving or by
Change the 4.1. Basic methods for shear cell as
application of other methods, particle size may be charac-
follows:
terized in the following manner:
4.1. Basic methods for shear cell
x90: Particle size corresponding to 90z of the cumulative
One type of shear cell is the cylindrical shear cell which is
undersize distribution
split horizontally, forming a shear plane between the lower
x50: Median particle size (i.e. 50z of the particles are
stationary base and the upper movable portion of the shear
smaller and 50z of the particles are larger)
cell ring. After powder bed consolidation in the shear cell
x10: Particle size corresponding to 10z of the cumulative
(using a well-defined procedure), the force necessary to
undersize distribution
shear the powder bed by moving the upper ring is deter-
It is recognized that the symbol d is also widely used to mined. Annular shear cell designs offer some advantages
designate these values. Therefore, the symbols d90, d50 and over the cylindrical shear cell design, including the need for
d10 may be used. less material. A disadvantage, however, is that because of
The following parameters may be defined based on the its design, the powder bed is not sheared as uniformly be-
cumulative distribution. cause material on the outside of the annulus is sheared more
Qr(x): cumulative distribution of particles with a dimen- than material in the inner region. A third type of shear cell
sion less than or equal to x where the subscript r reflects the (parallel-plate type) consists of a thin sandwich of powder
distribution type between a lower stationary rough surface and an upper
rough surface that is moveable.
r Distribution type All of the shear cell methods have their advantages and
disadvantages, but a detailed review is beyond the scope of
0 Number this chapter. As with the other methods for characterizing
1 Length powder flow, many variations are described in the litera-
ture. A significant advantage of shear cell methodology in
2 Area general is a greater degree of experimental control. The
3 Volume methodology generally is rather time-consuming and re-
quires significant amounts of material and a well-trained
operator.
2813
2814 General Information Supplement I, JP XVII

mol of protein. The response factors from protein/peptide


Solid and Particle Densities hydrolysates are typically about 30z lower than those for
nonhydrolyzed cysteic acid standards. Because histidine,
methionine, tyrosine, and tryptophan are also modified, a
Change the Particle Density as follows:
complete compositional analysis is not obtained with this
Particle Density technique.
The particle density takes account both the crystal density
Method 7
and the intraparticulate porosity (sealed and/or experimen-
Cysteine-cystine reduction and alkylation is accomplished
tally non-accessible open pores) as a part of the particle
by a vapor phase pyridylethylation reaction.
volume. The particle density depends on the value of the
Reducing Solution Transfer 83.3 mL of pyridine, 16.7
volume determined, and the volume in turn depends on the
mL of 4-vinylpyridine, 16.7 mL of tributylphosphine, and
method of measurement. Concerning the determination of
83.3 mL of water to a suitable container, and mix.
particle density , the Japanese Pharmacopoeia specifies the
Procedure Add the protein/peptide (between 1 mg and
pycnometry as the ``Powder Particle Density Determina-
100 mg) to a hydrolysis tube, and place in a larger tube.
tion''.
Transfer the Reducing Solution to the large tube, seal in
The pycnometric density is obtained by assuming that the
vacuum (about 50 mm of mercury or 6.7 Pa), and incubate
volume of the gas displaced, which is measured with the gas
at about 1009C for 5 minutes. Then remove the inner hy-
displacement pycnometer, is equivalent to that of a known
drolysis tube, and dry it in a vacuum desiccator for 15
mass of the powder. In pycnometric density measurements,
minutes to remove residual reagents. The pyridylethylated
any volume with the open pores accessible to the gas is not
protein/peptide can then be acid hydrolyzed using previ-
included as a part of volume of the powder, but the sealed
ously described procedures. The pyridylethylation reaction
pores or pores inaccessible to the gas is included as a part of
is performed simultaneously with a protein standard sample
the volume of the powder. Due to the high diffusivity of
containing 1 to 8 mol of cysteine per mol of protein to im-
helium which can penetrate to most open pores, it is recom-
prove accuracy in the pyridylethylcysteine recovery. Longer
mendable as the measurement gas of particle density. There-
incubation times for the pyridylethylation reaction can
fore, the pycnometric particle density of a finely milled
cause modifications to the a-amino terminal group and the
powder is generally not very different from the crystal den-
e-amino group of lysine in the protein.
sity. Hence, the particle density by this method is the best
estimate of the true density of an amorphous or partially
crystalline sample, and can be widely used for manufactur-
Add the following:
ing control of the processed pharmaceutical powder sam-
ples.
Enzyme-linked Immunosorbent
Assay (ELISA)
G3 Biotechnological/Biological
ELISA (Enzyme-linked immunosorbent assay) is one of
Products the immunological assay methods to detect analytes by
antigen-antibody reaction, in which an enzyme-labeled rea-
gent is used as a detection reagent. In general, 96-well-plates
Amino Acid Analysis and such are used, on which capture molecules specifically
bound to the analyte are immobilized. A test sample, an
Change the Method 6 and 7 in Protein Hydroly- enzyme-labeled reagent and other required reagents are
sis as follows: sequentially added and washed to have the enzyme-labeled
reagent bind onto a plate. After reaction by adding a sub-
Method 6
strate for the labeled enzyme, the response (e.g., absor-
Cysteine-cystine oxidation is accomplished with dimethyl
bance) by the enzymatic reaction is measured to determine
sulfoxide (DMSO).
the concentration of the analyte or binding activity in the
Hydrolysis Solution 6 mol/L hydrochloric acid contain-
test sample. ELISA is also used as a qualitative test to detect
ing 0.1z to 1.0z of phenol, to which DMSO is added to
the presence or absence of binding of the analyte with spe-
obtain a final concentration of 2z (v/v).
cific molecules.
Vapor Phase Hydrolysis The protein/peptide hydrolysis
In tests for biotechnological/biological products, ELISA
is conducted at about 1109C for 24 hours. During the
is mainly used for two different purposes. One is to quan-
hydrolysis, the cysteine-cystine present in the sample is
titate the target product or process-related impurities usu-
converted to cysteic acid by the DMSO present in the
ally by measuring the concentration of the analyte with anti-
Hydrolysis Solution. As an approach to limit variability and
bodies which specifically bind to the analyte. Another is to
compensate for partial destruction, it is recommended to
evaluate the biological activity of products such as ther-
evaluate the cysteic acid recovery from oxidative hydrolyses
apeutic antibodies. For the latter purpose, ELISA is used to
of standard proteins containing 1 to 8 mol of cysteine per
evaluate the binding activity of the target product with
Supplement I, JP XVII General Information 2815

molecules related to its pharmacological action, or to evalu- capture molecule without competing with other molecules
ate the cell response based on the amount of the endogenous (Fig. 1). This method can be used when the analyte pos-
protein secreted from the cells treated with the test samples sesses rather high molecular mass and has binding sites for
containing the target product. the capture molecule as well as for the molecule used for
detection.
1. Analytical methods
1.2. Competitive method
ELISA is broadly classified into competitive and non-
The competitive method has two approaches: the first is
competitive methods, and also classified into direct and in-
to immobilize a capture molecule, then has an analyte and
direct detection methods based on the detection procedures
an enzyme-labeled antibody compete to each other for bind-
(Fig. 1). In addition, ELISA is also classified into direct and
ing with the capture molecule (Fig. 1a), and the second is to
indirect immobilization methods by the method for im-
use the analyte prepared as the reagent which is immobilized
mobilizing capture molecules (Fig. 2).
onto a plate, then have the immobilized analyte and the
An analyte bound to a solid phase is detected by the anti-
analyte in test samples compete with each other for binding
body against the analyte or other reagents (Fig. 1). In the
with an enzyme-labeled antibody (Fig. 1b). The competitive
direct detection method, an enzyme-labelled antibody
method is used when the molecular mass of the analyte is
against the analyte is used. In the indirect detection method,
rather low, and it is difficult to prepare two molecules
a molecule indirectly bound to the analyte such as an anti-
which bind to the analyte specifically.
body (secondary antibody) against the antibody binding to
the analyte (primary antibody), is used. The procedure of 2. Analytical procedures
the direct detection method is simple, but the enzyme- 2.1. Procedure
labeled antibody against the analyte is required for each General procedures for both noncompetitive and com-
analyte. Compared to the direct detection method, the petitive methods are shown below. As for a quantitative
procedure of the indirect detection method is more com- test, prepare reference material solutions diluted serially in
plex, however, it allows for using a common secondary anti- order to obtain a dose-response curve or a calibration curve.
body such as an anti-IgG antibody even if the analyte is 2.1.1. Noncompetitive method
different. 1) Add a solution containing capture molecules onto a
When ELISA is used for measuring analyte concentra- plate, and incubate to immobilize the capture molecules on
tion, an antibody against the analyte is typically used as a a solid phase, then wash off the unbound capture molec-
capture molecule. When ELISA is used to evaluate biologi- ules.
cal activity by measuring binding activity, the target molec- 2) Add a blocking reagent, and have the reagent bind on
ule of a drug involved in its pharmacologic action is used as the surface not occupied by the capture molecules. Wash
a capture molecule. off the unbound blocking reagent.
1.1. Noncompetitive method 3) Add a reference material or a test sample onto each well
In the noncompetitive method, an analyte is bound to a of the plate, and have the analyte bind on the solid phase.
Wash off the unbound analyte.
4) When the direct detection method is used, add an en-
zyme-labeled antibody to bind to the analyte. When the in-
direct detection method is used, add an antibody against the
analyte, then wash and add the enzyme-labeled antibody
which binds to the antibody against the analyte in order to
bind it to the solid phase. Wash off the unbound enzyme-
labeled antibody.
5) Add a substrate solution, incubate and add a stopping
solution if required. Then measure the absorbance, lumines-
cent intensity, or fluorescent intensity, which reflects the
amount of substrate converted by the enzyme reaction.
6) Determine the binding activity or concentration of the
analyte with reference to the dose-response curve (calibra-
Fig. 1 Classification of ELISA by analytical method tion curve) of the reference material.
2.1.2. Competitive method
1) Competitive method (a): Add a solution containing
capture molecules onto a plate, then incubate so that the
capture molecules bind to a solid phase. Wash off the un-
bound capture molecules.
Competitive method (b): Add an analyte prepared for im-
mobilizing onto a plate, and incubate so that the analyte
bind to the solid phase. Wash off the unbound analyte.
Fig. 2 Examples of direct immobilization method and in-
2) Add a blocking reagent to bind on the solid phase sur-
direct immobilization method
2816 General Information Supplement I, JP XVII

face that is not occupied by the operation of 1). Wash off the dose-response curve (the calibration curve) of the refer-
the unbound blocking reagent. ence material.
3) Competitive method (a): Add a solution containing a 2) Obtain the dose-response curves of the reference mate-
reference material and an enzyme-labeled analyte, or a test rial and the test sample, respectively. Calculate the relative
sample and an enzyme-labeled analyte onto each well of the activity of the test sample against the reference material
plate. Then have the analyte and the enzyme-labeled analyte from the ratio of the concentration corresponding to 50z
bind on the solid phase. Wash off the unbound molecules. of the maximum response (EC50 for the noncompetitive
Competitive method (b): In the direct detection method, method, and IC50 for the competitive method).
add a solution containing a reference material and an en- 3) Use the range that can be approximated linear regres-
zyme-labeled antibody, or a test sample and an enzyme- sion in the dose-response curve. Calculate the relative ac-
labeled antibody onto each well of the plate, and then have tivity of the test sample against the reference material based
the enzyme-labeled antibody bind to the solid phase. Wash on the ratio of the dose that arise the same response.
off the unbound molecules. In the indirect detection 1) uses the same method as 2.2.1. and calculate the rela-
method, add a solution containing a reference material and tive concentration against the reference material. 2) uses the
an antibody against the analyte, or a test sample and an same method as 2.2.1. to lead the regression equation on the
antibody against the analyte onto each well of the plate. reference material and the test sample. Better regression can
After washing, add the enzyme-labeled antibody which be obtained by weighting to equalize the contribution of
binds to the antibody against the analyte. Wash off the un- each concentration response in leading the regression equa-
bound enzyme-labeled antibody. tion. The methods of weighting are to use 1/y 2, 1/y and
4) Add the substrate of the enzyme, incubate and then add 1/x. Upon the establishment of the test method, choose a
a stopping solution if required. Measure the amount of the regression method to obtain a better result based on the ac-
substrate converted by the enzyme reaction by measuring curacy and precision. 3) uses the concentration region near
absorbance, luminescent intensity or fluorescent intensity. EC50 or IC50 that can be approximated as a straight line for
5) Calculate the binding activity or the concentration of analysis.
the analyte from the dose-response curve (calibration curve) 2.3. Reagents, test Solutions
of the reference material. 2.3.1. Capture reagents
2.2. Data analysis Use molecules (antigen, antibody, etc.) which can spe-
2.2.1. Quantitation cifically bind to the analyte. Physical adsorption is fre-
When ELISA is applied to determine the concentration of quently used for immobilizing a capture reagent on a plate,
analyte, use an appropriately diluted test sample and calcu- and covalent binding is also possible to use for its binding to
late the concentration of the analyte in the test sample from the plate which is covered by materials having the binding
the calibration curve obtained from the reference material. activity with an amino or sulfhydryl functional group. Note
Usually, the calibration curve is prepared by using an equa- that there is a case that binding activity with an analyte may
tion of such as 4-parameter logistic regression, setting the be changed due to the conformational change by the bind-
log concentrations of the target molecule on the x-axis and ing onto the plate.
responses obtained on the y-axis. The capture reagent is a critical reagent that affects assay
4-parameter logistic model performance, and therefore, its quality should be controlled
A-D by setting necessary specifications. Establish the procedure
y=D+
1+ Ø »
C
x B for lot renewal as well.
2.3.2. Blocking reagents
A buffer solution containing protein such as albumin,
A: Lower asymptote
gelatin, or casein, which is supplemented with surfactants
B: Slope parameter
such as polysorbate 20 as required, is used as a blocking rea-
C: EC50 (IC50)
gent.
D: Upper asymptote
2.3.3. Detection reagents
x: Concentration of test sample
As enzymes for detection, peroxidase, alkaline phos-
y: Response
phatase, and b-galactosidase are typically used. As a label-
When the calibration curve is not bilateral symmetric as a ing method for an enzyme, covalent binding with a target
sigmoid curve, applying 5-parameter logistic regression may protein is used; A N-hydroxysuccinimide ester group intro-
improve the analytical result. As for the noncompetitive duced into an enzyme binds to the amino group of the
method, a calibration curve may be obtained by linear labeled protein, and a maleimide group introduced into the
regression by limiting the concentration at the lower range. enzyme binds to the sulfhydryl group of the labeled protein.
2.2.2. Biological activity As a method used for enzyme labeling of antibodies, cova-
For evaluating biological activity, the methods such as 1) lent binding with which the maleimide group introduced
to 3) described below are used. into the enzyme is bound to the sulfhydryl group of the
1) Use the test sample diluted with an appropriate dilution antibody is often used.
ratio. Determine the relative activity against the reference Detection Reagents are the critical reagents affecting
material by calculating the relative concentration based on assay performance, and therefore, their quality should be
Supplement I, JP XVII General Information 2817

Table 1 Examples of substrates sample by using calibration curves. When it is used as a


limit test, confirm the test sample response is not higher
Chromogenic Chemiluminescent Fluorescent
Enzyme than that of the control containing the impurities equal to
substrate substrates substrates
the upper limits of the acceptance criteria.
peroxidase TMB Luminol In general, samples include much more amount of a
OPD target product than impurities, and therefore, the target
ABTS product may disturb the detection of the impurities. Espe-
cially when ligands of affinity column are analytes, pay at-
alkaline pNPP
tention to the disturbance by the target product as the target
phosphatase
product binds to the ligands. Consider a recovery rate when
b-galactosidase MG sample pretreatment is performed.
NG 3.3. Biological assay
ELISA is used as a test to determine the binding activity
TMB: 3,3?,5,5?-Tetramethylbenzidine
of a therapeutic antibody as the target product with its tar-
OPD: o-Phenylenediamine
get molecule, and used to quantitate bioactive proteins
ABTS: 2,2?-Azino-bis[3-ethylbenzothiazoline-6-sulfonate]
secreted from the cells treated with test samples containing
pNPP: p-Nitrophenyl phosphate
the target product in cell-based assay.
MG: 4-Methylumbelliferyl galactoside
Determine relative activity by the method indicated in
NG: Nitrophenyl galactoside
2.2.2. 1) to 3).
3.4. Assay
ELISA is used for measuring the amount of target prod-
controlled by setting necessary specifications. Establish the ucts. Obtain the calibration curve of reference materials and
procedures for lot renewal as well. As for the indirect detec- calculate the concentration of the target products.
tion method, unlabeled antibodies against an analyte are
4. Validity of test
also used as a detection reagent, and therefore, it is neces-
In general, the validity of the test can be set as follows;
sary to control by setting necessary specifications.
use those in combination as necessary.
2.3.4. Substrates
4.1. Identification
Use substrates which are appropriate for each enzyme.
Confirm the results of reference materials and a negative
There are chromogenic, chemiluminescent and fluorescent
control pass the acceptance criteria specified in the mono-
substrates. Chemiluminescent substrates or fluorescent sub-
graph.
strates are suitable when high sensitivity is required.
4.2. Purity test
2.4. Points to consider
As for a quantitative test, confirm the reliability of
Since types of plates, amount of immobilized capture
calibration curves. Accuracy and/or precision of each con-
molecule, and incubation time as well as incubation temper-
centration of material solutions for the calibration curve
ature may affect test results, determine these procedures in-
and the coefficient of determination (R2 Value) calculated
cluding materials and reagents of use. Also determine the
from a regression equation are used to confirm the reliabil-
test conditions and the sample placement in plates to pre-
ity. Precision of test samples or accuracy of control samples
vent that the sample placement on the plates (the position of
prepared from the known concentration of a reference ma-
the well where the test is performed) affects the test results.
terial (Quality Control Sample: QC sample) could be set as
3. Application on specifications the test suitability. As for a limit test, confirm the response
3.1. Identification of the control sample containing the analyte at a concentra-
In monographs of biotechnological/biological products, tion equal to the upper limit of the acceptance criteria satis-
ELISA is used as an identification test which uses specific fies the criterion specified in the monograph.
antibodies against the target product to evaluate the binding 4.3. Biological assay
with the antibodies. As to therapeutic antibodies ELISA is When determining biological activity by using the method
also used as an identification test which evaluates the bind- of 1) of 2.2.2., confirm the reliability of the dose-response
ing of the antibodies with antigen. Usually it is used as a curve (the calibration curve) of the reference material . To
qualitative test. In the meantime, acceptance criteria can confirm the reliability of the dose-response curve, accuracy
also be set regarding the binding activity compared with a and/or precision of each concentration of the reference ma-
reference material when used as an identification test which terial and R2 value calculated from the regression equation
evaluates the binding of therapeutic antibody with antigen. or each parameter value of the regression equation obtained
3.2. Purity test from the dose-response curve of the reference material can
ELISA is used mainly as a purity test for process-related be used. Magnitude of the response of test samples, preci-
impurities such as host cell proteins, impurities derived sion of the relative activity calculated from the response or
from culture media and ligands eluted from affinity column accuracy of the concentration of QC samples can also be
resin. When ELISA is used as a test to determine the used to confirm the validity of the test.
amount of impurities, calculate the concentrations in a test When determining biological activity by using the method
2818 General Information Supplement I, JP XVII

of 2) of 2.2.2., confirm the parallelism of the two regression with recent progress in molecular biology techniques and
curves obtained from a reference material and a test sample. the accumulation of genetic information on plants,
As for the parallelism confirmation, following methods are differentiating methods of crude drugs based on genotypes
the examples. Obtain the ration of the difference between is being established. Unlike morphological and other
the upper asymptote and the lower asymptote (D - A of methods that are based on phenotypic characteristics, the
the 4-parameter regression equation in 2.2.1.) of the test genotypic methods are not affected by environmental fac-
sample to that of reference material or the ratio of the slope tors. Also, the methods have several advantages, such as
parameter (B of the 4-parameter regression equation in specialized expertise and skill for classification are not need-
2.2.1.), then confirm that those ratios are within the ed, and objective results are easily obtained.
predetermined range. R2 value of the dose-response curves The evolution of living organisms is accomplished by
of the reference material and the test sample, and accuracy genetic mutation, and differences among the nucleotide
of the QC samples are also used to confirm the validity of sequences of genes of closely related species reflect the
the test. strain relationships between the species. Based on this
When determining biological activity by using the method theory, methods that classify species phylogenetically using
of 3) of 2.2.2., confirm the linearity of the dose-response the nucleotide sequence of rDNA that codes for ribosomal
lines of a reference material and a test sample as well as the RNA (rRNA) on the nuclear genome have recently been
parallelism of these lines. adopted for the classification of microorganisms. In the
As for 2) and 3) of 2.2.2., there is a method to confirm same way, the sequence of this rDNA is also most often
the parallelism by comparing the residual variances of two used in the classification of higher plants based on the geno-
regression curves, using the constrained model for control type. In particular, it is very easy to classify closely related
and sample data and using unconstrained models for the species using the internal transcribed spacer (ITS) region of
control and sample data, and determining the parallelism of the rDNA region, since nucleotide substitution is more
the two regression curves by the method of analysis of vari- often undertaken by comparison with the coded gene
ance. However, it should be noted that if the precision of region. Furthermore, since the genes on the nuclear genome
the data is low, then the determination can be unrigorous. originate from the parents' genom, there is an advantage
4.4. Assay that interspecies hybrids can be detected. Higher plants also
Confirm the reliability of calibration curves obtained have mitochondrial genes and chloroplastic genes. Although
from the dose-response curves of reference materials. To the genes on these genomes are also often used for classifi-
confirm the reliability of the calibration curve, accuracy cation, interspecies hybrids cannot be confirmed because
and/or precision of each concentration of the reference the genes are normally uniparental inheritance.
material calculated from the regression equation, each The two methods presented here have been developed
parameter value of the regression equation and R2 value can based on the reported identification methods of Atrac-
be used. Precision of the measured results of test samples or tylodes Lancea Rhizome and Atractylodes Rhizome1,2)
accuracy of QC samples is also used to confirm the validity utilizing the gene sequence of the ITS region of rDNA.
of the test. Inter-laboratory validation study for the purity test of
Atractylodes Rhizome targeted for Atractylodes Lancea
Rhizome have been completed. The plant sources for Atrac-
G5 Crude Drugs tylodes Lancea Rhizome stipulated in the individual mono-
graphs are Atractylodes lancea De Candolle and A. chinen-
sis Koidzumi (Compositae), while those for Atractylodes
Purity Tests on Crude Drugs Rhizome are A. japonica Koidzumi ex Kitamura and A.
macrocephala Koidzumi (Compositae). The approval or
using Genetic Information rejection of the both sources is, in principle, determined by
the description of each crude drug, including microscopy,
Change the introduction as follows: together with thin-layer chromatography in identification
test. In the above scientific paper, it was shown that these 4
The first step in the quality assurance of natural products
plant species can be clearly classified by comparing the
is the use of raw materials from the right part of the right
nucleotide sequences of the ITS region mentioned above,
origin. Therefore, it is clearly stated in Article 4 of the
and that the species can be easily classified without per-
General Rules For Crude Drugs that the source of a crude
forming sequence analysis by performing PCR using a
drug is an approval or rejection criterion. There are various
species-specific primer set or by using a restriction enzyme
methods for differentiating the sources of crude drugs, such
which recognizes species-specific sequence.
as morphological methods, organoleptic tests, and chemical
In collaborative studies, the simplicity of the test is given
methods, and appropriate methods for each are described in
maximum consideration. We examined methods that ob-
the individual monographs. Morphological methods, or-
serve PCR amplification bands using species-specific primer
ganoleptic tests, and chemical methods are discrimination
sets (Mutant Allele Specific Amplification: Method 1) and
methods for species that are based on the phenotypic char-
methods that observe DNA fragments produced by restric-
acteristics of the crude drugs. On the other hand, together
tion enzyme treatment of the PCR products, which are pre-
Supplement I, JP XVII General Information 2819

pared using a primer set common to each plant source test, regardless of the form of the crude drug, it becomes
(PCR-Restriction Fragment Length Polymorphism: Method clear there is contamination by an inappropriate crude drug
2), without nucleotide sequence analyses. In these methods to be examined.
based on PCR, an extremely small amount of template The methods shown here are general information and at
DNA is amplified to billions to hundreds of billions times. the present stage results obtained using the methods do not
Therefore, when using them as identification tests for affect the approval or rejection of the crude drug in each
powdered crude drugs, the target DNA fragment can be ob- monograph. Furthermore, by performing the sequence
served even if the vast majority of the crude drug for analy- analysis outlined in the previous paper, it goes without
sis is not appropriate plant species and there is only a saying that more accurate decision concerning the source
minute amount of powder from a crude drug derived from a species can be made.
suitable plant. (Consequently, in identification tests, either
a cut or a whole crude drug must be used, as long as one is
careful to avoid contamination by powder originating from On the Scientific Names of Crude
other crude drugs.) On the other hand, when used as a puri-
ty test, the form of the crude drug is irrelevant as long as the Drugs listed in the JP
gene amplification is performed properly and the target
gene is not polymorphic, so if DNA fragments of an inap- Change the following as follows:
propriate plant to be examined are confirmed in the purity

Scientific names used in the JP and those being used taxonomically

Scientific names used in the JP


= Scientific names being used taxonomically (Combined notation,
Standard form for author or authors)
Crude Drug Scientific names that are different from those written in JP but identical Family
to them taxonomically or being regarded as identical, and typical sub-
classified groups belonging to their species. The names marked with
``*'' are those being written together in JP.

Curcuma Rhizome Curcuma zedoaria Roscoe


ガジュツ
Curcuma phaeocaulis Valeton Zingiberaceae

Curcuma kwangsiensis S. G. Lee et C. F. Liang

Jujube Seed Ziziphus jujuba Miller var. spinosa Hu ex H. F. Chou


Rhamnaceae
サンソウニン = Ziziphus jujuba Mill. var. spinosa (Bunge) Hu ex H. F. Chou

Jujube Ziziphus jujuba Miller var. inermis Rehder


Rhamnaceae
タイソウ = Ziziphus jujuba Mill. var. inermis (Bunge) Rehder

Platycodon Root Platycodon grandiflorus A. De Candolle


Campanulaceae
キキョウ = Platycodon grandiflorus (Jacq.) A. DC.

Rape Seed Oil Brassica napus Linn áe


ナタネ油 = Brassica napus L.
Cruciferae
Brassica rapa Linn áe var. oleifera De Candolle
= Brassica rapa L. var. oleifera DC.
2820 General Information Supplement I, JP XVII

G6 Drug Formulation
Add the following:

Aerodynamic Particle Size


Measurement for Inhalations
by Glass Impingers
This test is used to evaluate the fine particle characteris-
tics of the aerosol clouds generated by preparations for in-
halation, and is performed using the following apparatus
and test procedures. If justified, modified apparatus or test
procedure may be used.

1. Stage mensuration
The most reliable calibration for the separation character-
istics of each impaction stage is performed in terms of the
relationship between the aerodynamic diameter of particles
and droplets passing through it and the stage collection ef-
ficiency as an aerosol.
Calibration is usually performed by examination of a
property of the jet dimensions, the spatial arrangement of
the jet and its collection surface, and the airflow rate pass-
ing through it.
Because jets can corrode and wear over time, the critical
dimensions of each stage must be measured on a regular
basis to confirm them being within required ranges.
Only apparatuses that conform to specifications are used Fig. 1 Glass impinger
for the aerodynamic particle size measurement for inhala-
tions by glass impingers. An alternate validated and justi-
fied method of mensuration may be used. divided by the minimum recommended dose described in
the dosage and administration is not less than 75z and not
2. Inter-stage drug losses (wall losses)
more than 125z of the average delivered dose determined
Wall losses should be considered in method development
under Uniformity of Delivered Dose for Inhalations <6.14>.
and validation. If the wall losses affect the recovery rate
This mass balance is necessary to ensure that the test results
(mass balance) of drugs, they should be controlled. Wall
of particle size distributions of inhalations are valid.
losses will be dependent upon a number of factors including
the impactor type, operating conditions, formulation type 4. Glass impinger method
and discharged amount to an impactor. How the wall loss is The apparatus used for the glass impinger method is
reflected within the calculation of the aerodynamic diameter shown in Fig. 1. The apparatus consists of glass parts from
of particles should be judged based up on the level and the throat (B) to the lower impingement chamber (H) and
variability of the wall loss. For example, in the cases where plastic clips to hold them.
wall losses that are low or have a low level of variability, the This apparatus is operated based on a collision to a liquid
aerodynamic particle size is calculated by the assay of the surface and separate the drug discharged from the inhaler to
drug collected on the stage. In cases where wall losses are an inhalation part and a non-inhalation part. The drug in
high or variable, it may be necessary to collect the wall loss the non-inhalation part, which collides with an oral cavity
drug separately and take it into account for the calculation and a pharyngeal region to result in being swallowed, is
of the aerodynamic particle size. recovered in the rear of the throat and the upper impinge-
ment chamber (collectively stage 1). The drug in the inhala-
3. Recovery rate of drugs (mass balance)
tion part, which reaches lungs, is recovered in the lower
In addition to the size distribution, good analytical prac-
impingement chamber (stage 2). Because the upper impinge-
tice dictates that a mass balance be performed in order to
ment chamber is designed so that the cut-off diameter is 6.4
confirm that the amount of the drug discharged from the
mm when the test flow rate is 60 L per minute, particles with
inhaler, which is collected in the mouthpiece adapter and
a diameter of 6.4 mm or less flow down to the lower impin-
the apparatus, is within an acceptable range around the
gement chamber.
expected value. The total mass of drug collected in all of the
components of the mouthpiece adapter and the apparatus
Supplement I, JP XVII General Information 2821

Table 1 Component specification for apparatus shown in Fig. 1

Code Item Description Dimensions*

A Mouthpiece adapter Moulded rubber adapter for actuator mouthpiece.

B Throat Modified round-bottomed flask: 50 mL


—ground-glass inlet socket 29/32
—ground-glass outlet cone 24/29

C Neck Modified glass adapter:


—ground-glass inlet socket 24/29
—ground-glass outlet cone 24/29
Lower outlet section of precision-bore glass tubing:
—bore diameter 14
Selected bore light-wall glass tubing:
—external diameter 17

D Upper impingement Modified round-bottomed flask: 100 mL


chamber —ground-glass inlet socket 24/29
—ground-glass outlet cone 14/23

E Coupling tube Medium-wall glass tubing:


—ground-glass cone 14/23
Bent section and upper vertical section:
—external diameter 13
Lower vertical section:
—external diameter 8

F Screwthread, side-arm, Plastic screw cap 28/13


adapter Silicone rubber ring 28/11
Polytetrafluoroethylene (PTFE) washer 28/11
Glass screwthread:
—thread size 28
Side-arm outlet to vacuum pump:
—minimum bore diameter 5

G Lower jet assembly Modified polypropylene filter holder connected to lower vertical section of see Fig. 1
coupling tube by PTFE tubing
Acetal circular disc with the centres of four jets arranged on a projected cir- 10
cle of diameter 5.3 mm with an integral jet spacer peg:
—peg diameter 2
—peg protrusion 2

H Lower impingement Conical flask 250 mL


chamber —ground-glass inlet socket 24/29
* Dimensions in mm, unless otherwise stated.

4.1. Procedure for nebulizers by means of a mouthpiece adapter to the device.


Introduce 7 mL and 30 mL of a suitable solvent into the Switch on the pump of the apparatus and after 10 seconds
upper and lower impingement chambers, respectively. switch on the nebulizer.
Connect all the component parts. Ensure that the assem- After 60 seconds, unless otherwise justified, switch off
bly is vertical and adequately supported and that the jet the nebulizer, wait for 5 seconds and then switch off the
spacer peg of the lower jet assembly just touches the bottom pump of the apparatus.
of the lower impingement chamber. Connect a suitable Dismantle the apparatus and wash the inner wall surface
pump fitted with a filter (of suitable pore size) to the outlet of the upper impingement chamber collecting the washings
of the apparatus. Adjust the air flow through the appa- in a volumetric flask. Wash the inner wall surface of the
ratus, as measured at the inlet to the throat, to 60 ± 5 L per lower impingement chamber collecting the washings in a
minute. second volumetric flask. Finally, wash the filter preceding
Introduce the inhalation liquids and solutions into the the pump and its connections to the lower impingement
reservoir of the nebulizer. Fit the mouthpiece and connect it chamber and combine the washings with those obtained
2822 General Information Supplement I, JP XVII

from the lower impingement chamber. Determine the repeat the discharge sequence. The number of discharges
amount of active substance collected in each of the two should be minimized and typically would not be greater
flasks. Express the results for each of the two parts of the than 10.
apparatus as a percentage of the total amount of active sub- Dismantle the apparatus. Wash the inner wall surface of
stance. the coupling tube to the lower impingement chamber and its
4.2. Procedure for metered-dose inhalers outer wall surface that projects into the chamber with a suit-
Install a suitable mouthpiece adapter in position at the able solvent, collecting the washings in the lower impinge-
end of the throat. When the mouthpiece end of the inhaler ment chamber. Determine the content of active substance in
is inserted in the mouthpiece adapter to a depth of about 10 this solution. Calculate the amount of active substance col-
mm, the mouthpiece end of the inhaler lines up along the lected in the lower impingement chamber per discharge and
horizontal axis of the throat. The open end of the inhaler, express the results as a percentage of the active substance
which accepts the pressurized container, is uppermost and stated on the label.
in the same vertical plane as the rest of the apparatus.
Introduce 7 mL and 30 mL of a suitable solvent into the
upper and lower impingement chambers, respectively. G7 Containers and Package
Connect all the component parts. Ensure that the assem-
bly is vertical and adequately supported and that the jet
Add the following:
spacer peg of the lower jet assembly just touches the bottom
of the lower impingement chamber. Connect a suitable
pump to the outlet of the apparatus. Adjust the air flow Glass Containers for
through the apparatus, as measured at the inlet to the Pharmaceutical Products
throat, to 60 ± 5 L per minute.
Unless otherwise prescribed in the patient instruction, Glass containers for pharmaceutical products are widely
shake for 5 seconds and discharge once to waste. After not used. Glass bottles are used for tight and well-closed con-
less than 5 seconds, shake and discharge again to waste. tainers for bulk packaging of solid preparations for oral ad-
Repeat the procedure a further three times. ministration such as tablets and capsules etc., and ampules,
Shake for about 5 seconds, switch on the pump to the vials or glass syringes are for hermetic containers of injec-
apparatus and locate the mouthpiece end of the inhaler in tions etc.
the adapter, discharge once immediately in the apparatus. Glass containers used as a primary packaging have char-
Remove the assembled inhaler from the adapter, shake for acteristics of high chemical durability etc. in addition to
not less than 5 seconds, relocate the mouthpiece end of the high strength, high transparency, no air permeability and no
inhaler in the adapter and discharge again. Repeat the dis- moisture permeability. On the other hand, they are heavy,
charge sequence. The number of discharges should be bulky, fragile and easy to be broken by a physical shock
minimized and typically would not be greater than 10. After during manufacturing or transportation, so they require
the final discharge wait for not less than 5 seconds and then attention on handling.
switch off the pump. This chapter provides basic information about glass con-
Dismantle the apparatus. Wash the inner wall surface of tainers, items to be confirmed for the selection of glass con-
the coupling tube to the lower impingement chamber and its tainers and for the proper performance of a quality evalua-
outer wall surface that projects into the chamber with a suit- tion that comes along with the selection, and information
able solvent, collecting the washings in the lower impinge- about the quality control at the manufacturing stage of
ment chamber. Determine the content of active substance in preparations.
this solution. Calculate the amount of active substance col-
1. Basic information about glass containers for phar-
lected in the lower impingement chamber per discharge and
maceutical products
express the results as a percentage of the active substance
Glass containers for pharmaceutical products do not in-
stated on the label.
teract physically or chemically with the contained medica-
4.3. Procedure for dry powder inhalers
ments to alter any property or quality. Glass containers for
Introduce 7 mL and 30 mL of a suitable solvent into the
injections can protect the contained medicaments from the
upper and lower impingement chambers, respectively.
invasion of microbes by means of perfect sealing or other
Connect all the component parts. Ensure that the assem-
suitable process.
bly is vertical and adequately supported and that the jet
To ensure the quality of contained medicaments over the
spacer peg of the lower jet assembly just touches the bottom
shelf life, it is necessary to select a suitable glass container.
of the lower impingement chamber. Connect a suitable
In the selection of container, it is necessary to consider the
pump to the outlet of the apparatus. Adjust the air flow
physicochemical condition of the contained medicaments,
through the apparatus, as measured at the inlet to the
i.e., solid or liquid and the adoption of a well-closed con-
throat, to 60 ± 5 L per minute.
tainer, a tight container, a hermetic container or a colored
Prepare an inhaler and connect it to the throat using a
container to ensure the chemical stability of the contained
suitable adapter. Switch on the pump of the apparatus,
medicaments. Furthermore, it is necessary to consider sur-
after 5 seconds switch off the pump of the apparatus, and
Supplement I, JP XVII General Information 2823

face treatment on the inner surface of containers in the case pected to suppress the elution of glass components and the
where it is assumed that foreign substances occur by interac- occurrence of flakes because the drug solution does not con-
tions with the preparation ingredients. tact directly with the inner surface of the glass.
1.1 Glass composition and molding Fluorine resin processing is a method to form a thin film
Composition of the glass used for primary packaging of of fluorine resin on an inner surface by coating fluorine
pharmaceutical products is either borosilicate glass or so- resin using coupling agents and baking. This treatment en-
dalime glass. hance water repellency and prevent a drug solution from
Borosilicate glass has a reticulated network made of remaining to the inner surface of the glass. Also it is ex-
silicon dioxide (silica: SiO2) and diboron trioxide (B2O3). pected to suppress the elution of glass components and the
Borosilicate glass has a small coefficient of thermal occurrence of flakes because the drug solution does not con-
expansion, relatively high hardness and high hydrolytic tact directly with the inner surface of the glass.
resistance1). Containers made of this chemical composition
2. Quality evaluation of glass containers for pharmaceuti-
are classified as Type I glass in the USP and the EP.
cal products at the design stage of preparations
Cylinder-shaped and long material glass tubes made of
At the design stage of preparations it is necessary to per-
borosilicate glass are cut and undergo secondary processing
form the quality evaluation of a glass container used and
to mold ampules, vials or syringes, which are mostly used
the compatibility of it with the contained medicaments.
for containers of small amount of injections or lyophilized
Since each glass container for pharmaceutical products
preparations.
has characteristic properties and properties of pharmaceuti-
Sodalime glass is composed of silicon dioxide (silica:
cal products packed in the glass containers are diverse, the
SiO2), sodium oxide (Na2O) and calcium oxide (CaO) as the
compatibility of glass containers with pharmaceutical prod-
principal components. It has low hydrolytic resistance as a
ucts should be judged by considering the combination of the
drawback, but it is easy to manufacture and process1). Con-
both.
tainers made of this chemical composition are classified as
When evaluated, refer to General Rules for Preparations
Type II or III glass in the USP and the EP.
[2] General Notices for Packaging of Preparations, ``Basic
A glass container made of sodalime glass is called a blown
Requirements and Terms for the Packaging of Pharmaceu-
bottle or a molded bottle because it is molded by pouring
tical Products'' and ``Basic Requirements for Plastic Con-
melted glass into a mold and blowing air. Also it is called a
tainers for Pharmaceutical Use and Rubber Closures for
standard bottle or an automatic bottle because of its mass
Containers for Aqueous Infusions'' under General Infor-
production at low cost. It is widely used not only for glass
mation, and verify that the glass container used for prepara-
bottles of solid preparations for oral administration but also
tions conform to the basic requirements, i.e., the design
as containers for injections such as large volume vials of
specifications, based on tests and literatures.2,3) The com-
parenteral infusions or vials of powder injections for an-
patibility must be maintained based on an appropriate qual-
tibiotics etc.
ity assurance plan.
1.2 Surface treatment of inner surface of glass containers
2.1 Glass containers for pharmaceutical products
for pharmaceutical products
equipped with closures
Surface treatments are performed to modify the nature of
In the case of solid preparations for oral administration,
the inner surface of glass containers. The treatments are
glass containers with closures consist of a glass bottle and a
such as dealkalization treatment and coating, etc.
resin cap with a packing or a metal cap with a compound,
The dealkalization treatment is a method to neutralize the
and in the case of lyophilized injections they consist of a
surface layer of the glass by selectively extracting and
vial and a rubber closure. In the case of syringe prepara-
removing alkali components using sulfur compounds at
tions they consist of a glass outer (barrel, some has a nee-
high temperature above the glass-transition, which results in
dle), a gasket and a top cap.
exposure of the surface rich silica. This treatment reduce the
In the case of pharmaceutical products susceptible to be
elution of alkali components. The coating includes methods
oxidized, it is unsuitable to select the closure material that
using silica (SiO2), silicon resin and fluorine resin, etc.
permeate oxygen easily. In the case of aqueous pharmaceu-
Silica processing is a method to form a thin film on an
tical products and hygroscopic pharmaceutical products, it
inner surface by melt coating of silica (SiO2) on the inner
is unsuitable to select the closure material that permeate
surface of the glass at a high temperature. It is expected to
water vapor easily. Closures must not be deformed, deterio-
suppress the elution of glass components and the occurrence
rated and degenerated by contained medicaments. Unac-
of flakes, because the thin film is high purity silica with no
ceptable loss of function of containers must not be caused
water-soluble component such as alkali, weld to the inner
by a possible high temperature or low temperature or their
surface of the glass container and the drug solution does not
repetitions during storage and transportation and vibrations
contact directly with the inner surface of the glass.
during transportation. Glass containers for multiple-dose
Silicone processing is a method to form a thin film of sili-
pharmaceutical products equipped with closures are re-
cone resin on a glass surface by immersing the glass in
quired to have an appropriate stability after opening.
dimethylpolysiloxane solution and baking. This treatment
The compatibility (fitting compatibility) of closures with
enhance water repellency and prevent a drug solution from
glass containers for pharmaceutical products must be main-
remaining to the inner surface of the glass. Also it is ex-
2824 General Information Supplement I, JP XVII

tained based on an appropriate quality assurance plan. 1) Glass bottles used for solid preparations for oral ad-
2.2 Transparency of glass containers for pharmaceutical ministration etc.:
products and colored glass containers (i) Appearance4): Shape and dimensions are correct,
In the case of pharmaceutical products such as injections and there must not be failures of wall thickness,
where foreign matters and turbidity must be examined failures of color tone, breakage, lacks, cracks, in-
visually, glass containers for pharmaceutical products ternal cracks, scratches, bubbles, foreign matters,
should have the required level of transparency that enables striae, streaks, rough surfaces, burrs, stains and
inspection. insoluble matters, which cause a hindrance in
On the other hand, the quality of contained medicaments usage.
unstable to light must not be lowered during storage be- (ii) Quality tests: Soluble alkali test for a container,
cause of a high transparency of glass containers for phar- heat resistance and distortion.
maceutical products. A sufficient level of light shielding is (iii) Others: Items to be necessary.
required to ensure light stability, and the select of colored 2) Ampules or vials used for injections etc.:
glass containers must be considered. (i) Appearance4): Shape and dimensions are correct,
When colored glass containers are used for injections, it and there must not be failures of wall thickness,
must meet the requirements of the light transmission test for failures of color tone, breakage, lacks, cracks, in-
light-resistant containers under Test for Glass Containers ternal cracks, scratches, foreign matters, striae,
for Injections <7.01>. streaks, stains and insoluble matters, which cause
2.3 Glass containers for pharmaceutical products required a hindrance in usage.
to be sterile (ii) Quality tests: Tests prescribed under Test for
In selecting suitable glass containers (ampules) or glass Glass Containers for Injections <7.01>, heat
containers with closures (vials, syringes) as a primary pack- resistance (only for sodalime glass) and distortion.
aging for injections, it is desirable to obtain information on (iii) Others: Items to be necessary.
the manufacturing processes of the glass container including 4. References
substances added. 1) Glossary of terms relating to fine ceramics, JIS R 1600
For pharmaceutical products that require terminal sterili- (2011), Japanese Industrial Standards
zation, it is required for glass containers to satisfy the basic 2) Containers-Glass, US Pharmacopeia 40 (2017) <660>
requirements even after the sterilization. There must be no 3) Evaluation of the inner surface durability of glass con-
residue or generation of new toxic substances of more than tainers, US Pharmacopeia 40 (2017) <1660>
a certain quantity after the sterilization. Structures and ma- 4) Glass bottles for drug, JIS R 3522 (1955), Japanese In-
terials of glass containers must cause no microbial contami- dustrial Standards
nation to contained medicaments during storage and trans-
portation after the sterilization.
2.4 Foreign matters derived from glass containers for Add the following:
pharmaceutical products for injections
In the case of glass containers for injections, glass frag- Moisture Permeability Test for
ments generated at cutting ampules, flakes generated by
peeling of inner surfaces of glass and insoluble foreign mat- Blister Packaging of Solid
ters generated by elution of glass components or by stains Preparations
on inner surfaces of glass should be examined.
Eluates and flakes etc. derived from glass containers must The test is the method to measure the moisture transmis-
be sufficiently small from the viewpoint of safety. They sion rate of the blister packaging represented by PTP pack-
must not damage the efficacy and safety of the contained aging. It can be used for the following studies to evaluate
medicaments. moisture transmission through a packaging of drug prepa-
Foreign matters derived from glass containers must be ration.
sufficiently evaluated at the design stage of preparations. It (i) Screening of the material and/or thickness for plastic
must be also evaluated, when the molding process or suppli- sheets, forming conditions and/or size of pockets, etc. in
er is changed. the development phase.
Scanning Electron Microscopy–Energy Dispersive X-ray (ii) Comparison of the moisture transmission rate of a
Spectroscopy (SEM-EDX) is useful to analyze flakes de- plastic sheet before and after the change in material, thick-
rived from glass containers and inorganic foreign matters, ness, forming conditions, and/or size of pockets, etc. in the
for example aerosol of reaction products etc. development or production phase.
3. Test results to be recorded for each management unit Note that when a sufficient amount of desiccant cannot
At the manufacturing stage of glass containers for phar- be filled up in the pockets due to the minute pockets a relia-
maceutical products the specification of the following test ble result might be not obtained. The test is intended to de-
items should be set, and the test results should be recorded termine the moisture transmission rate of successfully pre-
for each management unit of glass containers for phar- pared blister packaging, but not to detect the leakage due to
maceutical products. pinholes and the like.
Supplement I, JP XVII General Information 2825

1. Terms the control is at least 2 sheets, however, desirable to be the


(i) Molding materials: Materials forming pockets and same as the sample number.
sealing areas. Usually, a single or double layer plastic sheet
5. Method
or that laminated with aluminum foil is used.
5.1. Storage conditions
(ii) Sealing materials: Materials to seal tightly pockets
The following conditions are desirable, though other con-
packed with drug preparations. Usually, an aluminum foil
ditions may be used.
is used.
(i) 25 ± 29 C/60 ± 5zRH
(iii) Pockets: Places where the molding material is in-
(ii) 40 ± 29C/75 ± 5zRH
flated in a convex shape to put drug preparations.
5.2. Storage
(iv) Moisture transmission rate: An amount of water
Place the samples and controls in a constant temperature
transmitted into the pockets of a blister packaging per unit
and humidity chamber without overlapping each other of
time (mg/day/pocket).
the sheets, not in standing position, as the pocket facing up-
2. Apparatus wards, as not intercepting the air circulation and avoiding
(i) Constant temperature and humidity chamber: An ap- exposure to the air flow from the outlet.
paratus which can maintain a temperature and humidity 5.3. Mass measuring
storage condition. Take out the samples and controls from the chamber,
(ii) Balance: A chemical balance. allow cooling to room temperature, measure the mass of
each sheet quickly, and place them back to the chamber.
3. Desiccants
Weigh exactly the masses of them to a degree of 0.1 mg.
It may be chosen from the following.
5.4. Measuring intervals
(i) Calcium chloride for water determination
Intervals of the measurement are adjusted depending on
Pretreatment before use: Put the desiccant taking out of
the moisture transmission rate and avoiding large change in
fine powder in a depthless vessel, dry at 1109C for 1 hour,
the temperature and humidity inside of the chamber (for ex-
then allow to cool in a desiccator [phosphorus (V) oxide].
ample, 0, 1, 3, 7, 14, 21 and 28 days).
(ii) Synthetic zeolite for drying
5.5. Termination of measurements
Moisture adsorption ability: Not less than 15z. Weigh
Measure the mass of each sheet of the sample and control
accurately about 10 g of the desiccant, allow to stand at
at each measuring point, and calculate the differences in
409C and 75z relative humidity for 24 hours, then weigh
their average values (the increasing amount of the sample
the mass, and calculate the gain in weight.
mass). Prepare a linear regression equation by the least-
Pretreatment before use: Put the desiccant in a depthless
squares method by plotting the increasing amount (mg) of
vessel, dry at 350 „ 6009C for 2 hours, then allow to cool
the sample mass on the vertical axis against the time (day)
in a desiccator [phosphorus (V) oxide].
on the horizontal axis. The measurement should be finished
4. Samples when the increase in the mass shows linearity in at least
4.1. Preparation of sample three sequenced points (expect for the starting point) and
Amount of the desiccant filled in a pocket is determined before the desiccant absorbs moisture of 10z amount of
appropriately depending on the form or size of the pocket, the mass of packed desiccant. The correlation coefficient of
however, it should be about 80z of the capacity of the the linearity is desirable to be not less than 0.98.
pocket for avoiding the deforming or impairing of the 5.6. Others
covering material. For preparation of the sample, carefully Data of samples showing extremely larger mass increase
prepare the sample avoiding moisture adsorption of the as compared to the others should be excluded since the
desiccant. Fill the desiccant in all the pockets as evenly as package may have some leakage due to pinholes or the like.
possible, seal with a sealing material, and cut out to a suita- Appropriate statistical tests are performed, as needed.
ble size. Separately, prepare a control in the same manner
6. Calculation of moisture transmission rate
by packing with the similar mass of glass beads. The form
The moisture transmission rate (mg/day/pocket) is calcu-
and size of the sample and the control should be as identical
lated by dividing the slope, i.e. the mass increasing amount
as possible.
(mg/day), obtained by the least-squares method, by the
Examine the appearances of the prepared sample and
number of pocket per sheet. Record the moisture transmis-
control with the naked eye or by using a stereomicroscope,
sion rate together with the storage conditions and the name
and use them whose pockets maintain their shapes as
of the desiccant used.
prescribed, and without any forming faults, aberrant wrin-
kles on the sealing material, pinholes or any sealing faults. 7. Information
4.2. Number of samples 7.1. Factors affecting the moisture transmission rate
Five to ten sheets are used for the sample with not less There are as follows:
than 10 pockets per sheet. An appropriate amount of sheets (i) Qualities (molecular structure, density, degree of
equivalent to 20 to 100 pockets (not less than 10 sheets), de- crystallinity, etc.), composition and/or thickness of the
pending on the number of pockets per sheet are used for the molding materials
sample with less than 10 pockets per sheet. The number of (ii) Methods and conditions to form the pocket
2826 General Information Supplement I, JP XVII

(iii) Size and/ or uniformity of wall thickness of the the products release should be made carefully based on the
pocket results of the investigation.
(iv) Storage conditions, water activity inside the pocket Parametric release can be considered a type of real-time
7.2. Measurement of pocket wall thickness release. One example of parametric release is to determine
Measure the wall thickness of at least one position of up- the suitability for release of terminally sterilized drug prod-
per or side face or R part of not less than 10 pockets of the ucts based on the data on sterilizing process instead of the
sampling sheet to a unit of 1 mm, using a micrometer or dial results of sterility testing. In this case, the release of each
gage with an accuracy of better than 1 mm or an equivalent batch is based on satisfactory results from monitoring
measuring instrument, as necessary. Measuring position is specific parameters, e.g., temperature, pressure, and time
selected in consideration of the shape of the pocket or during the terminal sterilization phase(s) of drug product
difficulty of the measurement. It is desirable to identify the manufacturing. Parametric release based on above para-
site that may become thinner in the phase of study for pock- meters is more reliable in predicting sterility assurance than
et forming conditions, and to measure the thickness of the determination of suitability for release based on sterility tes-
site while paying attention to the pressure. ting using limited number of final products. Besides, even if
parametric release is applied, the final product testing need
8. Reference
to be set because the testing is necessary in stability testing
1) Tsuneo Okubo, et al.: PMDRS, 45(2), 155 „ 165, (2014)
and survey after release. The parametric release differs from
RTRT in that the usage of the final product testing as alter-
native should not be applicable in principle in case of
G10 Others failure. If the data of monitoring specific parameters in ter-
minally sterilized process is failed to obtain by a certain rea-
son, the sterilization process is not able to be assured.
Basic Concepts for Quality
Assurance of Drug Substances
Add the following:
and Drug Products
Criteria for Content Uniformity
Change the Basic Concept and section 4. as
follows: in Real Time Release Testing by
Basic Concept Process Analytical Technology
In recent years, the mainstream concept for quality of
1. Introduction
drugs is that their quality is assured by management of
In recent years, the new criteria for Content Uniformity
manufacturing process, including management of raw mate-
Test using a large sample size for Real Time Release Testing
rial and other materials, and quality tests of final products
(RTRT) have become necessary with the rapid development
(drug substances and products) that are conducted mutually
of Process Analytical Technology (PAT). PAT using a non-
complementary.
destructive method such as Near Infrared (NIR) spectromet-
4. Real time release testing (RTRT) and parametric release ry enable to measure a large number of samples in real time,
Determination of the suitability for release can be based resulting in the generation of large amounts of data in a
on the result of RTRT instead of final product testing when short time, and PAT can improve process control and proc-
approved by the regulatory authority. RTRT is a type of ess capability. However, the current pharmacopoeial criter-
tests to evaluate the quality of in-process and/or final prod- ia for Uniformity of Dosage Unit (the sample size is 10 and
ucts based on process data (including results of in-process 30 for first and second stage respectively) may not be used
testing and data on process parameters). The usage of adequately for large sample sizes over a hundred. For exam-
RTRT does not mean unnecessity of setting final product ple, zero tolerance criteria has been used for outliers (no
tests directly. Even if the decision of release is made by unit showing over the 25z deviation from label claim must
RTRT, the tests for final products need to be set as specifi- be observed in the sample tested). However, the probability
cations. It is because final product testing may be requested of occurrence of outliers cannot be ignored when sample
for some reasons such as failure of data acquisition due to size was well over a hundred. This document display the
troubles of equipments used for RTRT and evaluation of consideration about criteria applicable for the large sample
stability of final products. The final products, of course, size over a hundred in RTRT.
need to meet their specifications, when tested. If RTRT
2. Theoretical basis of the criteria
results fail or trending toward failure, RTRT should not
The Content Uniformity Test of pharmacopoeia is a kind
easily be substituted by final product testing. In this case, it
of sampling tests, using small picked sample(s) from a large
is important to investigate the cause properly and need to
population (batch, lot), used for release of products. There-
take corrective action. Also, if RTRT results fail, the prod-
fore, the quality of estimations (test performance) depends
ucts can not be released unless they were caused by equip-
on the sample size. In general, estimate the better the larger
ment failure. If RTRT results are trending toward failure,
Supplement I, JP XVII General Information 2827

A. Constant risk of consumers

Fig. 1 Consumer's risk and producer's risk areas in an OC


curve

B. Constant risk of producers


A. Effects of sample sizes

Fig. 3 OC Curves of Content Uniformity Tests — Risks of


consumers or producers are constant
B. Effects of specification limits

needs to set the appropriate criteria for RTRT using Large-


N.
In setting of a specification limit, the limit value is deter-
mined by the balance between a guaranteed quality limit
(acceptable limit) and the severity of a realistically capable
test. When the specification limit is too strict, the acceptable
quality becomes better, however a stock shortage caused by
a low rate of passing the test of actual products occurs and
the cost may become abnormally high. In order to maintain
an acceptable quality, it is the most reasonable to compare
the consumer's risk (risk of poor quality passing the test)
and the producer's risk (risk of good quality failing the test)
Fig. 2 OC Curves of Content Uniformity Tests — Effects
and to determine the severity of the most suitable test. Fig.
of sample sizes and specification limits
1 shows the OC (operating characteristic) curve describes
the relationship as above.
The consumer's risk level, an acceptable quality corre-
the sample size, and it is considered that a large sample size sponding to pass rate of 5z in release tests, is important to
makes it possible to determine the quality of lots certainly. guarantee the quality of the product to be released. This
On the other hand, usage of a large sample size causes con- means that possibility of releasing low quality products is
sumption of resources. For this reason, compendial tests considered low (<5z). On the other hand, the producer's
like pharmacopoeias tests use a minimum and optimal sam- risk is important for producers. They should consider how
ple size accompanying with strict criteria in order not to good quality is needed to pass (usually 90 „ 95z) the test
release bad products. Now a day, as a large sample size sufficiently. In spite of the sample size, the lot quality corre-
(Large-N) has become popular with development of PAT, it sponding to 50z of pass rate is almost same the quality on
2828 General Information Supplement I, JP XVII

the specification limit. If the sample size is increased with- Table 1 Criteria for Content Uniformity
out change of the specification limit, the OC curves change
Acceptance number*
as in Fig. 2-A, where the quality (x-axis showing variability Sample size (n)
in unit content) of the 50z acceptance level is unchanged in C1** (± 15.0z) C2** (± 25.0z)
all the OC curves while the slope of the OC curves become
Criteria of 6.02 Uniformity
steeper with the larger the sample size. In contrast, if the n < 100
of Dosage Units
limit value is changed to more strict without changing the
sample size, the OC curves shift to the left at a constant 100  n < 150 3 0
slope (Fig. 2-B). To be constant the consumer's risk level
150  n < 200 4 0
regardless of change of sample size, it is necessary to set the
limit value in response to changes in sample size as in Fig. 200  n < 300 6 1
3-A. In general, the large sample size can have the con-
300  n < 500 8 2
sumer's risk level maintain to be constant even if the limit
value becomes loose. 500  n < 1000 13 4
When products is tested by PAT in a large sample size
1000  n < 2000 25 8
and then released, they will be subjected to stability tests
and survey tests using the usual small sample sizes after 2000  n < 5000 47 18
releasing. In this case, though the consumer's risk as in Fig.
5000  n < 10000 112 47
3-A is at a constant, the producer's risk increase. In order
not to increase the producer's risk after releasing, it is neces- 10000  n 217 94
sary to set the test limits so as not to differ very much in
* The requirements are met if the number of outliers is less
producer's risk between the test by PAT and the conven-
than or equal to acceptance number.
tional test. In this case, it is necessary to tighten the test
** Critical acceptance number.
limit in larger sample size, as shown in Fig. 3-B.
Our recommended criteria were determined in considera-
tion of such a point as described above1). It should be noted
that our criteria are simple and non-parametric criteria that 4. References
do not depend on the type of distribution of unit content, 1) Noriko Katori and Haruhiro Okuda, Sakura Bloom
and also has the same attitude with the Alternate 22) of the Tablets P2 Mock by MHLW sponsored QbD Drug
European Pharmacopoeia (EP) being a standard corre- Product Study Group (Mar. 2015).
sponding to the above mentioned Large-N. In the case of 2) European Pharmacopoeia 7.7 DEMONSTRATION
using the Alternate 1 of EP, there could be no problem OF UNIFORMITY OF DOSAGE UNITS USING
from the point of view about quality assurance. LARGE SAMPLE SIZES

3. Criteria for Content Uniformity in sample size equal to


or more than 100
The criteria recommended are consisted of two tests by
attribute (limits are C1 and C2). The sample sizes and ac-
ceptance numbers are shown in Table 1.

Criteria
Select n units representing a lot submitted, and assay the
units individually using an appropriate analytical method
and calculate individual contents expressed by the percen-
tage of label claim. The requirements are met if the number
of dosage units outside 15.0z is less than or equal to C1,
and the number of dosage units outside 25.0z is less than
or equal to C2. The central point of content bias can be
alter to an appropriate value from the label claim if it is
needed by quality control issue.
Supplement I, JP XVII General Information 2829

International Harmonization
Implemented in the Japanese
Pharmacopoeia Seventeenth
Edition
Add the following:

Nov. 2014/July 2015 (Corr. 1)

Harmonized items JP 17 (Supplement I) Remarks

Hydroxypropylcellulose, Low Substit- Hydroxypropylcellulose, Low Substit-


uted uted
Definition limits of content
Description Non-harmonized item
Packing and storage Containers and storage
Identification A Identification (3)
Identification B Identification (1)
Identification C Identification (2)
Purity Heavy metals
pH pH
Loss on drying Loss on drying
Residue on ignition Residue on ignition
Assay for hydroxypropoxy groups Assay

JP's particular description: Purity Heavy metals.

June 2014

Harmonized items JP 17 (Supplement I) Remarks

Glucose Anhydrous Purified Glucose


Definition limits of content
Description Non-harmonized item
Identification A Identification (1)
Identification B Identification (2)
Appearance of solution Purity (1) Clarity and color of solu-
tion
Conductivity Conductivity
Purity (2) Heavy metals Non-harmonized item
Related substances Purity (3) Related substances
Dextrin Purity (4) Dextrin
Soluble starch and sulfite Purity (5) Soluble starch and sulfite
Water Water
Assay Assay
Containers and storage Non-harmonized item

JP's particular description: Identification (1); Purity (3) Related substances Test for required detectability, System
repeatability; Assay System repeatability.
2830 General Information Supplement I, JP XVII

June 2014/Jan. 2015 (Corr. 1)

Harmonized items JP 17 (Supplement I) Remarks

Glucose Monohydrate Glucose Hydrate


Definition limits of content
Description Non-harmonized item
Identification A Identification (1)
Identification B Identification (2)
Appearance of solution Purity (1) Clarity and color of solu-
tion
Conductivity Conductivity
Purity (2) Heavy metals Non-harmonized item
Related substances Purity (3) Related substances
Dextrin Purity (4) Dextrin
Soluble starch and sulfite Purity (5) Soluble starch and sulfite
Water Water
Assay Assay
Containers and storage Non-harmonized item

JP's particular description: Identification (1); Purity (3) Related substances Test for required detectability, System
repeatability; Assay System repeatability.

Nov. 2014

Harmonized items JP 17 (Supplement I) Remarks

Sodium Lauryl Sulfate Sodium Lauryl Sulfate


Definition limits of content
Description Non-harmonized item
Identification A Identification (1)
Identification B Identification (2)
Identification C Identification (3)
Identification D Identification (4)
Alkalinity Purity (1) Alkalinity
Sodium chloride Purity (2) Sodium chloride
Sodium sulfate Purity (3) Sodium sulfate
Unsulfated alcohol Purity (4) Unsulfated alcohols
Water
Total alcohol content
Assay—Content of sodium alkyl sul- Assay
fate
Containers and storage Non-harmonized item

JP's particular description: Water; Total alcohol content.


Supplement I, JP XVII General Information 2831

Change the following as follows:

Nov. 2008

Harmonized items JP 17 (Supplement I) Remarks

Particle-size Analysis by Laser Light 3.06 Laser Diffraction Scattering Meas-


Diffraction urement of Particle size
Introduction Introduction
Principle 3. Measurement
Instrument 1. Instrument
Development of the method 2. Development of the method
Sampling 2.1. Sampling
Evaluation of the dispersion proce- 2.2. Evaluation of the dispersion
dure procedure
Optimisation of the liquid disper- 2.3. Optimization of the liquid disper-
sion sion
Optimisation of the gas dispersion 2.4. Optimization of the gas disper-
sion
Determination of the concentration 2.5. Determination of the concentra-
range tion range
Determination of the measuring time 2.6. Determination of the measuring
time
Selection of an appropriate optical 2.7. Selection of an appropriate opti-
model cal model
Validation 2.8. Validation
Measurement 3. Measurement
Precautions 3.1. Precautions
Measurement of the light scattering 3.2. Measurement of the light scatter-
of dispersed sample(s) ing of dispersed sample(s)
Conversion of scattering pattern into 3.3. Conversion of scattering pattern
particle-size distribution into particle-size distribution
Replicates 3.4. Replicates
Reporting of results 4. Reporting of results
Control of the instrument perfor- 5. Control of the instrument perfor-
mance mance
Calibration 5.1. Calibration
Qualification of the system 5.2. Qualification of the system
Note
Figure 1 Example of a set-up of laser Fig. 3.06-1 Example of a set-up of
light diffraction instrument laser light diffraction instrument
2832 General Information Supplement I, JP XVII

Nov. 2015 (Rev. 2)

Harmonized items JP 17 (Supplement I) Remarks

Uniformity of Dosage Units 6.02 Uniformity of Dosage Units


(Introduction) (Introduction)
Content uniformity 1. Content uniformity
Solid dosage forms (i) Solid dosage forms
Liquid or semi-solid dosage forms (ii) Liquid or semi-solid dosage forms
Calculation of acceptance value 1.1. Calculation of acceptance value
Mass variation 2. Mass variation
Uncoated or film-coated tablets (i) Uncoated or film-coated tablets
Hard capsules (ii) Hard capsules
Soft capsules (iii) Soft capsules
Solid dosage forms other than tablets (iv) Solid dosage forms other than
and capsules tablets and capsules
Liquid dosage forms (v) Liquid dosage forms
Calculation of acceptance value 2.1. Calculation of acceptance value
Criteria 3. Criteria
Solid, semi-solid and liquid dosage (i) Solid, semi-solid and liquid dosage
forms forms
Table 1 Application of content uni- Table 6.02-1 Application of content
formity (CU) and mass variation uniformity (CU) and mass variation
(MV) test for dosage forms (MV) test for dosage forms
Table 2 Table 6.02-2

JP's particular description: (Introduction) Additional explanation on Liquids, Additional explanation for the part not con-
taining drug substance; 2. Mass variation Additional explanation that the test is performed based on the assumption of the
concentration of drug substances being uniform; Table 6.02-1 Additional explanation on ``solids in single-dose packages''
and ``solutions enclosed in unit-dose containers''.

May 2016 (Rev. 2, Corr. 1)

Harmonized items JP 17 (Supplement I) Remarks

Saccharin Sodium Saccharin Sodium Hydrate


Definition limits of content
Identification A Identification (1)
Identification B Identification (2)
Clarity of solution Purity (1) Clarity and color of solu-
tion
Color of solution
Acidity or alkalinity Purity (2) Acidity or alkalinity
Water Water
Readily carbonizable substances Purity (6) Readily carbonizable sub-
stances
Limit of benzoate and salicylate Purity (4) Benzoate and salicylate
Assay Assay
Supplement I, JP XVII General Information 2833

July 2015 (Rev. 1, Corr. 2)

Harmonized items JP 17 (Supplement I) Remarks

Hypromellose Hypromellose
Definition limits of content of methoxy group
and hydroxypropoxy group
Labeling labeling of viscosity
Identification (1) Identification (1)
Identification (2) Identification (2)
Identification (3) Identification (3)
Identification (4) Identification (4)
Identification (5) Identification (5)
Viscosity Viscosity
Method 1 Method I
Method 2 Method II
pH pH
Loss on drying Loss on drying
Residue on ignition Residue on ignition
Assay Assay

July 2016 (Corr. 3)

Harmonized items JP 17 (Supplement I) Remarks

Polysorbate 80 Polysorbate 80
Definition origin
Identification (Composition of fatty Identification
acids)
Acid value Acid value JP's particular description: Applying
Fats and Fatty Oils Test 1.13, using
ethanol (95) as the solvent.
Hydroxyl value Hydroxyl value
Peroxide value Purity (3) Peroxide value
Saponification value Saponification value
Composition of fatty acids Composition of fatty acid
Ethylene oxide and dioxan Purity (2) Ethylene oxide and 1,4-di-
oxane
Water Water
Total ash Residue on ignition
Storage Containers and storage
2834 General Information Supplement I, JP XVII

Oct. 2016 (Rev. 1)

Harmonized items JP 17 (Supplement I) Remarks

General Information
Amino Acid Analysis Amino Acid Analysis
Apparatus Apparatus
General precautions General precautions
Reference standard material Reference standard material
Calibration of instrumentation Calibration of instrumentation
Repeatability Repeatability
Sample preparation Sample preparation
Internal standards Internal standards
Protein hydrolysis Protein hydrolysis
Method 1 Method 1
Hydrolysis solution Hydrolysis solution
Procedure Procedure
Liquid phase hydrolysis Liquid phase hydrolysis
Vapor phase hydrolysis Vapor phase hydrolysis
Method 2 Method 2
Hydrolysis solution Hydrolysis solution
Vapor phase hydrolysis Vapor phase hydrolysis
Method 3 Method 3
Hydrolysis solution Hydrolysis solution
Vapor phase hydrolysis Vapor phase hydrolysis
Method 4 Method 4
Oxidation solution Oxidation solution
Procedure Procedure
Method 5 Method 5
Hydrolysis solution Hydrolysis solution
Liquid phase hydrolysis Liquid phase hydrolysis
Method 6 Method 6
Hydrolysis solution Hydrolysis solution
Vapor phase hydrolysis Vapor phase hydrolysis
Method 7 Method 7
Reducing solution Reducing solution
Procedure Procedure
Method 8 Method 8
Stock solutions Stock solutions
Reducing solution Reducing solution
Procedure Procedure
Method 9 Method 9
Stock solutions Stock solutions
Carboxymethylation solution Carboxymethylation solution
Buffer solution Buffer solution
Supplement I, JP XVII General Information 2835

Procedure Procedure
Method 10 Method 10
Reducing solution Reducing solution
Procedure Procedure
Method 11 Method 11
Reducing solutions Reducing solutions
Procedure Procedure
Methodologies of amino acid analysis Methodologies of amino acid analysis
general principles general principles
Method 1-Postcolumn ninhydrin de- Method 1-Postcolumn ninhydrin de-
tection general principle tection general principle
Method 2-Postcolumn OPA fluoro- Method 2-Postcolumn OPA fluoro-
metric detection general principle metric detection general principle
Method 3-Precolumn PITC derivati- Method 3-Precolumn PITC derivati-
zation general principle zation general principle
Method 4-Precolumn AQC derivati- Method 4-Precolumn AQC derivati-
zation general principle zation general principle
Method 5-Precolumn OPA derivati- Method 5-Precolumn OPA derivati-
zation general principle zation general principle
Method 6-Precolumn DABS-Cl Method 6-Precolumn DABS-Cl
derivatization general principle derivatization general principle
Method 7-Precolumn FMOC-Cl Method 7-Precolumn FMOC-Cl
derivatization general principle derivatization general principle
Method 8-Precolumn NBD-F Method 8-Precolumn NBD-F
derivatization general principle derivatization general principle
Data calculation and analysis Data calculation and analysis
Calculations Calculations
Amino acid mole percent Amino acid mole percent
Unknown protein samples Unknown protein samples
Known protein samples Known protein samples

Add the following: time during which the drug substance is expected to remain
within its specification and, therefore, can be used in the
Stability Testing of Drug manufacture of a given drug product, provided that the
drug substance has been stored under the defined condi-
Substances and Drug Products tions. After this period, a batch of drug substance destined
for use in the manufacture of a drug product should be re-
1. Introduction
tested for compliance with the specification and then used
It is essential that the quality of a drug is maintained dur-
immediately. A batch of drug substance can be re-tested
ing the period from being manufactured to being ad-
multiple times. For certain antibiotics known to be labile, it
ministered in a patient. Stability testing is performed in
is more appropriate to establish a shelf life than a re-test
order to ensure that the quality is maintained during the
period. The shelf life of a drug product is the period in
period. The purpose of stability testing is to provide evi-
which a batch of the product is expected to remain within
dence on how the quality of a drug substance or drug
the approved shelf life specification if stored under defined
product varies with time under the influence of a variety of
conditions.
environmental factors such as temperature, humidity and
This general information mainly illustrates a standard im-
light, and to establish a re-test period for the drug substance
plementation that can be set when we perform stability tests
or a shelf life for the drug product and recommended
of a chemical drug substance and the associated drug
storage conditions.
product, and it is also helpful in stability tests of phar-
The re-test period of a drug substance is the period of
maceuticals other than chemical drugs. Also, this leaves
2836 General Information Supplement I, JP XVII

sufficient flexibility to encompass the variety of different on the drug substance packaged in a container closure sys-
practical situations that may be encountered due to specific tem that is the same as or simulates the packaging proposed
scientific considerations and characteristics of the materials for storage and distribution. The primary batches of the
being evaluated. Alternative approaches can be used when drug product should be of the same formulation and pack-
there are scientifically justifiable reasons. aged in the same container closure system as proposed for
marketing (including, as appropriate, any secondary pack-
2. Conditions of stability testing
aging and container label). The manufacturing process used
Stress testing, long term testing, accelerated testing and if
for primary batches should simulate that to be applied to
necessary intermediate testing are performed as stability tes-
production batches and should provide the product of the
ting for drugs.
same quality and meeting the same specification as that in-
2.1 Stress testing
tended for marketing. Two of the three batches should be at
Stress testing of the drug substance can help identify the
least pilot scale batches and the third one can be smaller, if
likely degradation products, which can in turn help establish
justified. The primary batch may be a production batch.
the degradation pathways and the intrinsic stability of the
Where possible, batches of the drug product should be
molecule and validate the stability indicating power of the
manufactured by using different batches of the drug sub-
analytical procedures used. Stress testing should include the
stance. The pilot scale batch is a batch of a drug substance
effect of temperatures (in 109 C increments (e.g., 509C,
or drug product manufactured by a procedure fully
609C, etc.) above that for accelerated testing), humidity
representative of and simulating that to be applied to a full
(e.g., 75z RH or greater) where appropriate, oxidation,
production scale batch. For solid oral dosage forms, a pilot
and photolysis on the drug substance. The testing should
scale is generally, at a minimum, one-tenth that of a full
also evaluate the susceptibility of the drug substance to hy-
production scale or 100,000 tablets or capsules, whichever is
drolysis across a wide range of pH values when in solution
the larger.
or suspension.
The storage conditions used for stability testing are
Stress testing of the drug product is undertaken to assess
shown as follows.
the effect of severe conditions on the drug product. Such
studies include photostability testing and specific testing on
certain products, (e.g., metered dose inhalers, creams,
emulsions, refrigerated aqueous liquid products).
2.2 Long term testing, accelerated testing and intermediate Table 1 Storage condition
testing Storage condi-
Long term Accelerated Intermediate
Long term testing is undertaken on batches of a drug sub- tion and package
stance or drug product according to a prescribed stability 25 ± 29C/
protocol to establish the re-test period of the drug substance General case
60 ± 5zRH or 40 ± 29C/ 30 ± 29C/
or the shelf life of the drug product. (drug substance
30 ± 29C/ 75 ± 5zRH 65 ± 5zRH2)
and product)
Accelerated testing is a stability study designed to increase 65 ± 5zRH1)
the rate of chemical degradation or physical change of a
Storage in a
drug substance or drug product by using exaggerated refrigerator 25 ± 29C/
storage conditions. Data from these studies, in addition to 5 ± 39C —
(drug substance 60 ± 5zRH
long term stability studies, can be used to assess longer term and product)3)
chemical effects at non-accelerated conditions and to evalu-
Storage in a
ate the effect of short term excursions outside the label
freezer (drug
storage conditions such as might occur during shipping. - 20 ± 59C — —
substance and
Intermediate testing is conducted at 309C/65z RH and product)4)
designed to moderately increase the rate of chemical degra-
Storage below
dation or physical changes for a drug substance or drug
- 209 C
product intended to be stored long term at 259 C. Intermedi- case-by-case basis
(drug substance
ate testing is implemented only when a significant change and product)
occurs in the accelerated testing.
Long term and accelerated testing, also if needed inter- Drug products
packaged in Study can be conducted under any controlled or
mediate testing should be performed on at least three prima-
impermeable ambient humidity condition
ry batches. The primary batches of the drug substance
containers
should be manufactured to a minimum of pilot scale by the
same synthetic route as, and using a method of manufacture Drug products 25 ± 29C/
40 ± 29C/
and procedure that simulates the final process to be used packaged in 40 ± 5zRH or 30 ± 29C/
not more than
semi-permeable 30 ± 29C/ 65 ± 5zRH7)
for, production batches. The overall quality of the batches (NMT) 25zRH
containers5) 35 ± 5zRH6)
of drug substance placed on the stability studies should be
representative of the quality of the material to be made on a 1) It is up to the applicant to decide whether long term stability stu-
production scale. The stability studies should be conducted dies are performed at 25 ± 29C/60 ± 5zRH or 30 ± 29C/65 ±
5zRH.
Supplement I, JP XVII General Information 2837
2) If ``significant change'' occurs at the accelerated storage condi- 3. Testing attributes and testing frequency
tion, additional testing at the intermediate storage condition Stability studies should include testing of those attributes
should be conducted. However, if 30 ± 29C/65 ± 5zRH is the
of the drug substance or the product that are susceptible to
long term condition, there is no intermediate condition. ``Sig-
nificant change'' for a drug substance is defined as failure to change during storage and are likely to influence quality,
meet its specification. In general, ``significant change'' for a safety, and/or efficacy. Validated stability-indicating ana-
drug product is defined as: lytical procedures should be applied. Whether and to what
1. A 5z change in assay from its initial value; or failure to meet extent replication should be performed will depend on the
the acceptance criteria for potency when using biological or im-
results from validation studies.
munological procedures;
2. Any degradation product's exceeding its acceptance criterion; For long term studies, frequency of testing should be
3. Failure to meet the acceptance criteria for appearance, physi- sufficient to establish the stability profile of the drug sub-
cal attributes, and functionality test (e.g., color, phase separation, stance and product. For drug substances or products with a
resuspendibility, caking, hardness, dose delivery per actuation); proposed re-test period or shelf life of at least 12 months,
however, some changes in physical attributes (e.g., softening of
the frequency of testing at the long term storage condition
suppositories, melting of creams) may be expected under accelerat-
ed conditions; should normally be every 3 months over the first year, every
and, as appropriate for the dosage form: 6 months over the second year, and annually thereafter
4. Failure to meet the acceptance criterion for pH; or through the proposed re-test period or shelf life. At the
5. Failure to meet the acceptance criteria for dissolution for 12 accelerated storage condition, a minimum of three time
dosage units.
points, including the initial and final time points (e.g., 0, 3,
6. Physical changes shown in the following may be observed in
accelerated testing, but the changes are not considered as ``sig- and 6 months), from a 6-month study is recommended.
nificant change'' which needs intermediate testing, when there is no When testing at the intermediate storage condition is called
``significant change'' in other attributes. for as a result of significant change at the accelerated
Softening of suppositories designed to melt at 379 C, when its storage condition, a minimum of four time points, includ-
melting point is shown clearly.
ing the initial and final time points (e.g., 0, 6, 9, 12
When it is clear that ``significant change'' is due to crosslink-
ing, the dissolution of gelatin capsules and gel coating tablets do months), from a 12-month study is recommended.
not conform to the acceptance criteria for 12 dosage units. A reduced design, i.e., matrixing or bracketing, where the
When confirming that there is no ``significant change'' in other testing frequency is reduced or certain factor combinations
attributes, consider the possibility that these physical changes affect are not tested at all, can be applied, if justified, for the test-
the other attributes.
3) The drug product is packaged in a semi-permeable container, ap-
ing of combination of drug products having multiple design
propriate information should be provided to assess the extent of factors (e.g., strength, container size and/or fill). A brack-
water loss. In the accelerated testing of drug substances or prod- eting design assumes that the stability of any intermediate
ucts intended for storage in a refrigerator, if significant change levels is represented by the stability of the extremes tested.
occurs within the first 3 months, it is considered unnecessary to This is the design of a stability schedule such that only sam-
continue to test a product through 6 months.
4) Testing on a single batch at an elevated temperature (e.g., 5 ±
ples on the extremes of certain design factors (e.g., strength,
39C or 25 ± 29C) for an appropriate time period should be con- container size and/or fill). Bracketing can be applied to stu-
ducted to address the effect of short term excursions outside the dies with multiple strengths of identical or closely related
label storage condition, e.g., during shipment and handling. formulations. Examples include but are not limited to (1)
5) Aqueous-based products packaged in semi-permeable containers
capsules of different strengths made with different fill plug
should be evaluated for potential water loss under conditions of
sizes from the same powder blend, (2) tablets of different
low relative humidity. Other comparable approaches can be de-
veloped and used for non-aqueous, solvent-based products. strengths manufactured by compressing varying amounts of
6) It is up to the applicant to decide whether long term stability stu- the same granulation, and (3) oral solutions of different
dies are performed at 25 ± 29C/40 ± 5z RH or 30 ± 29C/35 strengths with formulations that differ only in minor ex-
± 5zRH. cipients (e.g., colorants, flavorings). Bracketing can be ap-
7) If ``significant change'' other than water loss occurs during the 6
plied to studies of the same container closure system where
months' testing at the accelerated storage condition. Additional
testing at the intermediate storage condition should be per- either container size or fill varies while the other remains
formed. A significant change in water loss alone at the accelerat- constant. The use of a bracketing design would not be ap-
ed storage condition does not necessitate testing at the intermedi- plicable if it cannot be demonstrated that the strengths or
ate storage condition. However, data should be provided to container sizes and/or fills selected for testing are indeed
demonstrate that the drug product will not have significant water
the extremes. An example of a bracketing design is given in
loss throughout the proposed shelf life if stored at 259C and the
reference relative humidity of 40z RH. If 30 ± 29C/35 ± 5z Table 2. This design is provided for illustrative purpose,
RH is the long term condition, there is no intermediate condi- and should not be considered the only, or the most ap-
tion. A 5z loss in water from its initial value is considered a sig- propriate, design in all cases.
nificant change for a product packaged in a semi-permeable con- A matrixing design assumes that the stability of each sub-
tainer after an equivalent of 3 months' storage at 409C/NMT
set of samples tested represents the stability of all samples at
25z RH. However, for small containers (1 mL or less) or unit-
dose products, a water loss of 5z or more after an equivalent of a given time point. This is the design of a stability schedule
3 months' storage at 409C/NMT 25z RH may be appropriate, if such that a selected subset of the total number of possible
justified. samples for all factor combinations would be tested at a
specified time point. At a subsequent time point, another
subset of samples for all factor combinations would be
2838 General Information Supplement I, JP XVII

Table 2 Example of a Bracketing Design (ii) Option 2 For option 2 the same sample should be
exposed to both the cool white fluorescent and near ultravi-
Strength 50 mg 75 mg 100 mg
olet fluorescent lamp.
Batch 1 2 3 1 2 3 1 2 3 1. A cool white fluorescent lamp designed to produce an
output similar to that specified in ISO10977(1993); and
15 mL bottle T T T T T T
2. A near ultraviolet fluorescent lamp having a spectral
Container
100 mL bottle distribution from 320 nm to 400 nm with a maximum
size
energy emission between 350 nm and 370 nm; a significant
500 mL bottle T T T T T T
proportion of energy emission should be in both bands of
T = Sample tested 320 to 360 nm and 360 to 400 nm.
4.2. Light exposure level and testing condition
For drug substances, photostability testing should consist
Table 3 Example of a Matrixing Design on Time Points of two parts: forced degradation testing and confirmatory
for a Product with Two Strengths testing. The purpose of forced degradation testing studies is
``One-Half Reduction'' to evaluate the overall photosensitivity of the material for
Time point (months) 0 3 6 9 12 18 24 36 method development purposes and/or degradation pathway
elucidation. This testing may involve the drug substance
Batch 1 T T T T T T alone and/or in simple solutions/suspensions to validate the
S1 Batch 2 T T T T T T analytical procedures. In these forced degradation testing
studies, a variety of exposure conditions may be used, de-
Batch 3 T T T T T pending on the photosensitivity of the drug substance in-
Strength
Batch 1 T T T T T volved and the intensity of the light sources used. For devel-
opment and validation purposes it is appropriate to limit
S2 Batch 2 T T T T T T exposure and end the studies if extensive decomposition
Batch 3 T T T T T occurs. For photostable materials, studies may be terminat-
ed after an appropriate exposure level has been used. The
T = Sample tested design of these experiments is left to the applicant's discre-
tion although the exposure levels used should be justified.
Confirmatory studies of drug substance should then be
tested. Matrixing designs can be applied to strengths with undertaken to provide the information necessary for han-
identical or closely related formulations. Examples include dling, packaging, and labeling. For confirmatory studies,
but are not limited to (1) capsules of different strengths samples should be exposed to light providing an overall il-
made with different fill plug sizes from the same powder lumination of not less than 1.2 million lx・h and an inte-
blend, (2) tablets of different strengths manufactured by grated near ultraviolet energy of not less than 200 W・h/m2
compressing varying amounts of the same granulation, and to allow direct comparisons to be made between the drug
(3) oral solutions of different strengths with formulations substance and drug product. Care should be taken to ensure
that differ only in minor excipients (e.g., colorants or that the physical characteristics of the samples under test
flavorings). Other examples of design factors that can be are taken into account and efforts should be made, such as
matrixed include batches made by using the same process cooling and/or placing the samples in sealed containers, to
and equipment, and container sizes and/or fills in the same ensure that the effects of the changes in physical states such
container closure system. as sublimation, evaporation or melting are minimized. All
An example of a matrixing design is given in Table 3. such precautions should be chosen to provide minimal inter-
This design is provided for illustrative purpose, and should ference with the exposure of samples under test. Possible in-
not be considered the only, or the most appropriate, design teractions between the samples and any material used for
in all cases. containers or for general protection of the sample should
also be considered and eliminated wherever not relevant to
4. Photostability testing
the test being carried out. As a direct challenge for samples
Photostability testing is a part of stress testing evaluating
of solid drug substances, an appropriate amount of sample
the photostability characteristics of drug substances and
should be taken and placed in a suitable glass or plastic dish
products.
and protected with a suitable transparent cover if consi-
4.1. Light sources
dered necessary. Solid drug substances should be spread
The light sources described below may be used for
across the container to give a thickness of typically not more
photostability testing.
than 3 mm. Drug substances that are liquids should be ex-
(i) Option 1 Any light source that is designed to pro-
posed in chemically inert and transparent containers. Where
duce an output similar to the D65/ID65 emission standard
practicable when testing samples of the drug product out-
such as an artificial daylight fluorescent lamp combining
side of the primary pack, these should be presented in a way
visible and ultraviolet (UV) outputs, xenon, or metal halide
similar to the conditions mentioned for the drug substance.
lamp.
The samples should be positioned to provide maximum area
Supplement I, JP XVII General Information 2839

of exposure to the light source. For example, tablets, cap-


sules, etc., should be spread in a single layer. If direct ex-
posure is not practical (e.g., due to oxidation of a product),
the sample should be placed in a suitable protective inert
transparent container (e.g., quartz). If testing of the drug
product in the immediate container or as marketed is need-
ed, the samples should be placed horizontally or transverse-
ly with respect to the light source, whichever provides for
the most uniform exposure of the samples. Some
adjustment of testing conditions may have to be made when
testing large volume containers (e.g., dispensing packs).

5. Evaluation of stability data


In the stability data evaluation, data from long term and
accelerated testing, also if needed from intermediate testing
and, as appropriate, supporting data (data of stability test-
ing using drug substances and products in developing stage)
should be evaluated to determine the critical quality attrib-
utes likely to influence the quality and performance of the
drug substance or product. Each attribute should be as-
sessed separately, and an overall assessment should be made
of the findings for the purpose of proposing a re-test period
or shelf life. An approach for analyzing data of a quantita-
tive attribute that is expected to change with time is to deter-
mine the time at which the 95z one-sided confidence limit
for the mean curve intersects the acceptance criterion. The
re-test period or shelf life proposed should not exceed that
predicted for any single attribute.
INDEX
Ajmaline, 381 Injection, 410
A Tablets, 382 Amiodarone Hydrochloride, 411
Akebia Stem, 1793 Tablets, 412
Absorptive Cream, 755 Alacepril, 382 Amitriptyline Hydrochloride, 414
Acacia, 1791 Tablets, 383 Tablets, 414
Powdered, 1791 L-Alanine, 384 Amlexanox, 415
Acebutolol Hydrochloride, 359 Albumin Tannate, 386 Tablets, 416
Aceglutamide Aluminum, 359, 2671 Alcohol, 895 Amlodipine Besilate, 417
Acemetacin, 361 Dehydrated, 896 Orally Disintegrating Tablets, 418
Capsules, 361 for Disinfection, 897 Tablets, 420
Tablets, 362 Aldioxa, 386 Ammonia Water, 420
Acetaminophen, 363 Granules, 387 Amobarbital, 421
Acetazolamide, 364 Tablets, 387 Amomum Seed, 1797
Acetic Acid, 365 Alendronate Powdered, 1797
Glacial, 365 Sodium Hydrate, 388 Amosulalol Hydrochloride, 421
Acetohexamide, 366 Sodium Injection, 389 Tablets, 422
Acetylcholine Chloride for Injection, Sodium Tablets, 390 Amoxapine, 424
367 Alimemazine Tartrate, 391 Amoxicillin
Acetylcysteine, 368 Alisma Tuber, 1793 Capsules, 425
Acetylsalicylic Acid, 449 Powdered, 1794, 2747 Hydrate, 424, 2671
Tablets, 450 Allopurinol, 392 Amphotericin B, 426
Achyranthes Root, 1792 Tablets, 392 for Injection, 427
Aciclovir, 369 Alminoprofen, 393 Syrup, 428
for Injection, 371 Tablets, 394 Tablets, 428
for Syrup, 373 Aloe, 1794 Ampicillin
Granules, 370 Powdered, 1795 Anhydrous, 429
Injection, 371 Alpinia Officinarum Rhizome, 1796 Ethoxycarbonyloxyethyl Hydrochlo-
Ointment, 372 Alprazolam, 395 ride, 468
Ophthalmic Ointment, 372 Alprenolol Hydrochloride, 396 Hydrate, 430, 2671
Syrup, 372 Alprostadil, 396 Sodium, 431
Tablets, 374 Alfadex, 400 Sodium and Sulbactam Sodium for
Aclarubicin Hydrochloride, 375 Injection, 398 Injection, 433
Acrinol Alum, 403 Sodium for Injection, 432
and Zinc Oxide Oil, 377 Solution, 401 Ampicillinphthalidyl Hydrochloride,
and Zinc Oxide Oil, Compound, Powder, Salicylated, 1542 1633
377 Aluminum Ampiroxicam, 434
and Zinc Oxide Ointment, 378 Acetylsalicylate, 450 Capsules, 435
Hydrate, 376 Monostearate, 402 Amyl Nitrite, 436
Actinomycin D, 378 Potassium Sulfate Hydrate, 403 Anemarrhena Rhizome, 1797
Adrenaline, 379 Silicate Hydrate with Silicon Dioxide, Anesthamine, 901
Injection, 379 1796 Anesthetic Ether, 898
Solution, 380 Silicate, Natural, 404 Angelica Dahurica Root, 1798
Adsorbed Silicate, Synthetic, 405 Anhydrous
Diphtheria-Purified Pertussis-Teta- Sucrose Sulfate Ester, 1609 Aminobenzylpenicillin, 429
nus Combined Vaccine, 816 Amantadine Hydrochloride, 405 Ampicillin, 429
Diphtheria-Tetanus Combined Tox- Ambenonium Chloride, 406 Caffeine, 543
oid, 816 Amidotrizoic Acid, 407 Citric Acid, 716
Diphtheria Toxoid for Adult Use, Amikacin Sulfate, 408 Dibasic Calcium Phosphate, 557
816 for Injection, 409 Ethanol, 896, 2693
Habu-venom Toxoid, 1004 Injection, 409 Lactose, 1132, 2714
Hepatitis B Vaccine, 1018 Aminoacetic Acid, 995 Light, Silicic Acid, 1552
Purified Pertussis Vaccine, 1384 Aminobenzylpenicillin, 430 Sodium Sulfate, 1994
Tetanus Toxoid, 1670 Anhydrous, 429 Antipyrine, 437
Afloqualone, 380 Sodium, 431 Apricot Kernel, 1798
Agar, 1792 Aminophylline Water, 1799
Powdered, 1793 Hydrate, 410 Aprindine Hydrochloride, 437

2841
2842 Index Supplement I, JP XVII

Capsules, 438 Bamethan Sulfate, 471 Biphasic Isophane Insulin Human (Ge-
Aralia Rhizome, 1800 Barbital, 472 netical Recombination) Injectable
Arbekacin Sulfate, 439 Barium Sulfate, 473 Aqueous Suspension, 2706
Injection, 440 Bear Bile, 1807 Bisacodyl, 511
Areca, 1800 Bearberry Leaf, 1807 Suppositories, 512
Argatroban Hydrate, 441 Beclometasone Dipropionate, 473 Bismuth
L-Arginine, 442 Beef Tallow, 1808 Subgallate, 512
Hydrochloride, 443 Beeswax Subnitrate, 513
Hydrochloride Injection, 443 White, 1808 Bisoprolol Fumarate, 514
Aromatic Castor Oil, 1826 Yellow, 1809 Tablets, 515
Arotinolol Hydrochloride, 444 Bekanamycin Sulfate, 474 Bitter
Arsenic Trioxide, 445 Belladonna Cardamon, 1812
Arsenical Paste, 445 Extract, 1810 Orange Peel, 1812
Arsenous Acid, 445 Root, 1809 Tincture, 1813
Artemisia Total Alkaloids, 1810 Bleomycin
Capillaris Flower, 1800, 2747 Benidipine Hydrochloride, 475 Hydrochloride, 516
Leaf, 1801 Tablets, 476 Sulfate, 518
Ascorbic Acid, 445 Benincasa Seed, 1811 Bofutsushosan Extract, 1813, 2748
and Calcium Pantothenate Tablets, Benoxinate Hydrochloride, 1345 Boiogito Extract, 1817, 2750
447 Benserazide Hydrochloride, 478 Boric Acid, 520
Injection, 446 Bentonite, 478 Bromazepam, 520
Powder, 446 Benzalkonium Chloride, 479 Bromhexine Hydrochloride, 521
Asiasarum Root, 1801 Solution, 480 Bromocriptine Mesilate, 522
Asparagus Root, 1802 Solution 50, Concentrated, 480 Bromovalerylurea, 523
L-Aspartic Acid, 449 Benzbromarone, 481 Brotizolam, 523
Aspirin, 449 Benzethonium Chloride, 481 Tablets, 524
Aluminum, 450 Solution, 482 Brown Rice, 1820
Tablets, 450 Benzocaine, 901 Bucillamine, 525
Aspoxicillin Hydrate, 451 Benzoic Acid, 482 Tablets, 526
Astragalus Root, 1803 Benzoin, 1812 Bucumolol Hydrochloride, 527
Atenolol, 452 Benzyl Bufetolol Hydrochloride, 528
Atorvastatin Calcium Alcohol, 483 Buformin Hydrochloride, 529
Hydrate, 453 Benzoate, 484 Delayed-release Tablets, 529
Tablets, 454 Benzylpenicillin Tablets, 531
Atractylodes Benzathine Hydrate, 485 Bumetanide, 531
Lancea Rhizome, 1803 Potassium, 486, 2674 Bunazosin Hydrochloride, 532
Lancea Rhizome, Powdered, 1804 Potassium for Injection, 487 Bupivacaine Hydrochloride Hydrate,
Rhizome, 1804 Bepotastine Besilate, 488 533
Rhizome, Powdered, 1805 Tablets, 489 Bupleurum Root, 1820
Atropine Sulfate Beraprost Sodium, 490 Bupranolol Hydrochloride, 534
Hydrate, 456 Tablets, 492 Buprenorphine Hydrochloride, 535
Injection, 456 Berberine Burdock Fruit, 1821
Auranofin, 457 Chloride Hydrate, 493 Burnt Alum, 403
Tablets, 458 Tannate, 494 Busulfan, 536
Azathioprine, 459 Betahistine Mesilate, 495 Butenafine Hydrochloride, 536
Tablets, 460 Tablets, 496 Cream, 537
Azelastine Hydrochloride, 461 Betamethasone, 497 Solution, 538
Granules, 462 Dipropionate, 499 Spray, 538
Azelnidipine, 463 Sodium Phosphate, 500 Butropium Bromide, 539
Tablets, 464 Tablets, 498 Butyl Parahydroxybenzoate, 540
Azithromycin Hydrate, 465 Valerate, 502
Azosemide, 2672 Valerate and Gentamicin Sulfate C
Tablets, 2672 Cream, 502
Aztreonam, 466 Valerate and Gentamicin Sulfate Cacao Butter, 1821
for Injection, 467 Ointment, 504 Cadralazine, 541
Betamipron, 505 Tablets, 542
B Betaxolol Hydrochloride, 506 Caffeine
Bethanechol Chloride, 507 and Sodium Benzoate, 544
Bacampicillin Hydrochloride, 468 Bezafibrate, 507 Anhydrous, 543
Bacitracin, 469, 2674 Extended-release Tablets, 508 Hydrate, 543
Baclofen, 470 Bifonazole, 509 Calciferol, 882
Tablets, 470 Biotin, 510 Calcitonin Salmon, 545
Bakumondoto Extract, 1805, 2747 Biperiden Hydrochloride, 510 Calcium
Supplement I, JP XVII Index 2843

Chloride Hydrate, 550 Ribavirin, 1512 Hydrate, 612


Chloride Injection, 551 Rifampicin, 1519 Tablets, 614
Folinate, 551 Roxatidine Acetate Hydrochloride Cefdinir, 615
Gluconate Hydrate, 552 Extended-release, 1531 Capsules, 617
Hydroxide, 553 Sodium Iodide (123I), 1579 Fine Granules, 617
Lactate Hydrate, 553 Sodium Iodide (131I), 1580 Cefditoren Pivoxil, 618
Leucovorin, 551 Sulpiride, 1621 Fine Granules, 619
Levofolinate Hydrate, 2675 Tacrolimus, 1633 Tablets, 620
Oxide, 554 Teprenone, 1661 Cefepime Dihydrochloride
Pantothenate, 554 Tranexamic Acid, 1709 for Injection, 622
Paraaminosalicylate Granules, 556 Tranilast, 1712 Hydrate, 621
Paraaminosalicylate Hydrate, 555 Trientine Hydrochloride, 1727 Cefixime
Polystyrene Sulfonate, 559 Ubenimex, 1742 Capsules, 624
Sodium Edetate Hydrate, 561 Captopril, 575 Hydrate, 623, 2676
Stearate, 562 Carbamazepine, 575 Cefmenoxime Hydrochloride, 625
Calumba, 1821 Carbazochrome Sodium Sulfonate Hy- Cefmetazole Sodium, 627
Powdered, 1822 drate, 576 for Injection, 628
Camellia Oil, 1822 Carbetapentane Citrate, 1378 Cefminox Sodium Hydrate, 629
Camostat Mesilate, 562 Carbetapentene Citrate, 1378 Cefodizime Sodium, 629
d-Camphor, 563 Carbidopa Hydrate, 577 Cefoperazone Sodium, 631
dl-Camphor, 564 L-Carbocisteine, 578 and Sulbactam Sodium for Injection,
Candesartan Cilexetil, 565 Tablets, 579 632
and Amlodipine Besylate Tablets, Carbolic Acid, 1390 for Injection, 2677
567 Carbon Dioxide, 580 Cefotaxime Sodium, 634
and Hydrochlorothiazide Tablets, Carboplatin, 580 Cefotetan, 635
570 Injection, 582 Cefotiam
Tablets, 566 Carboxymethylcellulose, 583 Hexetil Hydrochloride, 637
Capsicum, 1822 Calcium, 583 Hydrochloride, 639
and Salicylic Acid Spirit, 1824 Sodium, 584 Hydrochloride for Injection, 640
Powdered, 1823 Cardamon, 1825 Cefozopran Hydrochloride, 640
Tincture, 1824 Carmellose, 583 for Injection, 641
Capsules, 574 Calcium, 583 Cefpiramide Sodium, 642
Capsules Sodium, 584 Cefpirome Sulfate, 643
Acemetacin, 361 Carmofur, 586 Cefpodoxime Proxetil, 644
Amoxicillin, 425 Carnauba Wax, 1825 for Syrup, 646
Ampiroxicam, 435 Carteolol Hydrochloride, 587 Tablets, 647
Aprindine Hydrochloride, 438 Carumonam Sodium, 587 Cefroxadine
Cefaclor, 593 Carvedilol, 589 for Syrup, 649
Cefadroxil, 598 Tablets, 590 Hydrate, 648
Cefalexin, 601 Cassia Seed, 1825 Cefsulodin Sodium, 650
Cefdinir, 617 Castor Oil, 1826 Ceftazidime
Cefixime, 624 Aromatic, 1826 for Injection, 653
Cinoxacin, 710 Catalpa Fruit, 1826 Hydrate, 652
Clindamycin Hydrochloride, 722 Cefaclor, 592 Cefteram Pivoxil, 654
Clofibrate, 729 Capsules, 593 Fine Granules, 655
Clorazepate Dipotassium, 740 Combination Granules, 594 Tablets, 656
Diltiazem Hydrochloride Extended- Fine Granules, 596 Ceftibuten Hydrate, 657
release, 807 Cefadroxil, 597 Ceftizoxime Sodium, 658, 2678
Doxifluridine, 835 Capsules, 598 Ceftriaxone Sodium Hydrate, 660
Droxidopa, 842 for Syrup, 599 Cefuroxime Axetil, 662
Emedastine Fumarate Extended- Cefalexin, 599 Cellacefate, 663, 2679
release, 858 Capsules, 601 Cellulose
Ethyl Icosapentate, 904 Combination Granules, 602 , 2-hydroxypropyl ether, 1033
Flopropione, 939 for Syrup, 603 Acetate Phthalate, 663
Fluconazole, 941 Cefalotin Sodium, 604 Microcrystalline, 664
Hypromellose, 574 Cefatrizine Propylene Glycolate, 605 Powdered, 667
Indometacin, 1063 for Syrup, 606 Celmoleukin (Genetical Recombina-
Lansoprazole Delayed-release, 1140 Cefazolin Sodium, 607 tion), 668
Methotrexate, 1225 for Injection, 608 Cetanol, 671
Nifedipine Extended-release, 1310 Hydrate, 609 Cetirizine Hydrochloride, 671
Nizatidine, 1320 Cefbuperazone Sodium, 610 Tablets, 672
Pilsicainide Hydrochloride, 1402 Cefcapene Pivoxil Hydrochloride Cetotiamine Hydrochloride Hydrate,
Pullulan, 574 Fine Granules, 613 673
2844 Index Supplement I, JP XVII

Cetraxate Hydrochloride, 674 Cistanche Herb, 1831 Granules, 751


Chenodeoxycholic Acid, 675 Citicoline, 715 Tablets, 752
Cherry Bark, 1826 Citric Acid Colistin
Chloral Hydrate, 676 Anhydrous, 716 Sodium Methanesulfonate, 752,
Chloramphenicol, 677 Hydrate, 717 2683
and Colistin Sodium Methanesul- Citrus Unshiu Peel, 1832 Sulfate, 753
fonate Ophthalmic Solution, Clarithromycin, 717, 2680 Compound
2679 Tablets, 719 Acrinol and Zinc Oxide Oil, 377
Palmitate, 677 Clebopride Malate, 720 Diastase and Sodium Bicarbonate
Sodium Succinate, 678, 2679 Clemastine Fumarate, 721 Powder, 783
Chlordiazepoxide, 679 Clematis Root, 1833 Hycodenone Injection, 1347
Powder, 680 Clindamycin Iodine Glycerin, 1077
Tablets, 681 Hydrochloride, 721 Methyl Salicylate Spirit, 1238
Chlorhexidine Hydrochloride Capsules, 722 Oxycodone and Atropine Injection,
Gluconate Solution, 682 Phosphate, 724 1348
Hydrochloride, 683 Phosphate Injection, 725 Oxycodone Injection, 1347
Chlorinated Lime, 683 Clinofibrate, 725 Phellodendron Powder for
Chlormadinone Acetate, 684 Clobetasol Propionate, 726 Cataplasm, 1940
Chlorobutanol, 685 Clocapramine Hydrochloride Hydrate, Rhubarb and Senna Powder, 1957
Chlorphenesin Carbamate, 685 727 Salicylic Acid Spirit, 1541
Tablets, 686 Clofedanol Hydrochloride, 728 Scopolia Extract and Diastase Pow-
Chlorpheniramine Maleate, 687 Clofibrate, 728 der, 1976
Injection, 688 Capsules, 729 Thianthol and Salicylic Acid Solu-
Powder, 689 Clomifene Citrate, 730 tion, 1679
Tablets, 690 Tablets, 731 Vitamin B Powder, 1765
d-Chlorpheniramine Maleate, 691 Clomipramine Hydrochloride, 731 Concentrated
Chlorpromazine Hydrochloride, 692 Tablets, 2681 Glycerin, 993
Injection, 692 Clonazepam, 732 Glycerol, 993
Tablets, 693 Fine Granules, 733 Condurango, 1836
Chlorpropamide, 694 Tablets, 733 Fluidextract, 1837
Tablets, 695 Clonidine Hydrochloride, 734 Coptis Rhizome, 1837
Cholecalciferol, 696 Cloperastine Hydrochloride, 735 Powdered, 1838
Cholera Vaccine, 696 Clopidogrel Corn
Cholesterol, 697 Sulfate, 736 Oil, 1839
Chorionic Gonadotrophin, 997 Sulfate Tablets, 738 Starch, 1601
for Injection, 999 Clorazepate Dipotassium, 739 Cornus Fruit, 1839, 2752
Chotosan Extract, 1827, 2751 Capsules, 740 Cortisone Acetate, 754
Chrysanthemum Flower, 1829 Clotiazepam, 741 Corydalis Tuber, 1840
Cibenzoline Succinate, 697 Tablets, 2682 Powdered, 1841
Tablets, 698 Clotrimazole, 742 Crataegus Fruit, 1842
Ciclacillin, 699 Clove, 1833 Creams
Ciclosporin, 700 Oil, 1834 Absorptive, 755
A, 700 Powdered, 1833 Betamethasone Valerate and Gen-
Cilastatin Sodium, 701 Cloxacillin Sodium Hydrate, 743, tamicin Sulfate, 502
Cilazapril 2683 Butenafine Hydrochloride, 537
Hydrate, 702 Cloxazolam, 744 Hydrophilic, 756
Tablets, 703 Cnidium Ibuprofen Piconol, 1044
Cilnidipine, 704 Monnieri Fruit, 1834 Ketoconazole, 1122
Tablets, 705 Rhizome, 1835 Terbinafine Hydrochloride, 1663
Cilostazol, 707 Rhizome, Powdered, 1835 Cresol, 756
Tablets, 708 Cocaine Hydrochloride, 745 Solution, 756
Cimetidine, 709 Coconut Oil, 1835 Solution, Saponated, 757
Cimicifuga Rhizome, 1830 Codeine Phosphate Croconazole Hydrochloride, 757
Cinchocaine Hydrochloride, 786 Hydrate, 746 Croscarmellose Sodium, 585
Cinnamon Powder, 1z, 746 Crospovidone, 758
Bark, 1830 Powder, 10z, 747 Crude Glycyrrhiza Extract, 1864,
Bark, Powdered, 1831 Tablets, 748 2756
Oil, 1831 Cod Liver Oil, 745 Crystal Violet, 1237
Cinoxacin, 709 Codonopsis Root, 1835 Curcuma Rhizome, 2753
Capsules, 710 Coix Seed, 1836 Cyanamide, 759
Ciprofloxacin, 711 Powdered, 1836 Cyanocobalamin, 760
Hydrochloride Hydrate, 712 Colchicine, 749 Injection, 761
Cisplatin, 714 Colestimide, 750 Cyclopentolate Hydrochloride, 761
Supplement I, JP XVII Index 2845

Cyclophosphamide Dibekacin Sulfate, 785 816


Hydrate, 762 Ophthalmic Solution, 785 Dipyridamole, 817
Tablets, 762 Dibucaine Hydrochloride, 786 Disodium Edetate Hydrate, 1573
Cycloserine, 763 Dichlorphenamide, 787 Disopyramide, 818
Cyperus Rhizome, 1842, 2753 Tablets, 789 Distigmine Bromide, 818
Powdered, 1843, 2753 Diclofenac Sodium, 787 Tablets, 819
Cyproheptadine Hydrochloride Hy- Diclofenamide, 787, 2684 Disulfiram, 820
drate, 764 Tablets, 789, 2684 Dobutamine Hydrochloride, 820
L-Cysteine, 765 Dicloxacillin Sodium Hydrate, 789 Docetaxel
Hydrochloride Hydrate, 766 Diethylcarbamazine Citrate, 790 for Injection, 823
L-Cystine, 766 Tablets, 790 Hydrate, 821
Cytarabine, 767 Difenidol Hydrochloride, 791 Injection, 822
Diflorasone Diacetate, 792 Dolichos Seed, 1848
D Diflucortolone Valerate, 793 Domperidone, 824
Digenea, 1846 Donepezil Hydrochloride, 825
Daiokanzoto Extract, 1843, 2754 Digitoxin, 794, 2684 Fine Granules, 826
Daisaikoto Extract, 1844, 2754 Tablets, 795, 2684 Tablets, 827
Danazol, 768 Digoxin, 796, 2685 Dopamine Hydrochloride, 829
Dantrolene Sodium Hydrate, 768 Injection, 797 Injection, 829
Daunorubicin Hydrochloride, 769 Tablets, 798 Dorzolamide Hydrochloride, 830
Deferoxamine Mesilate, 771 Dihydrocodeine Phosphate, 800 Ophthalmic Solution, 831
Dehydrated Alcohol, 896 Powder, 1z, 800 Doxapram Hydrochloride Hydrate,
Dehydrocholate Sodium Injection, Powder, 10z, 801 832
773 Dihydroergotamine Mesilate, 802 Doxazosin Mesilate, 823
Dehydrocholic Acid, 772 Dihydroergotoxine Mesilate, 803 Tablets, 834
Injection, 773 Dilazep Hydrochloride Hydrate, 805 Doxifluridine, 835
Purified, 772 Diltiazem Hydrochloride, 806 Capsules, 835
Demethylchlortetracycline Hydrochlo- Extended-release Capsules, 807 Doxorubicin Hydrochloride, 836,
ride, 774, 2684 Dilute 2685
Dental Hydrochloric Acid, 1024 for Injection, 837
Antiformin, 436 Iodine Tincture, 1076 Doxycycline Hydrochloride
Iodine Glycerin, 1078 Diluted Opium Powder, 1924 Hydrate, 838, 2686
Paraformaldehyde Paste, 1367 Dimemorfan Phosphate, 808 Tablets, 840
Phenol with Camphor, 1392 Dimenhydrinate, 809 Dried
Sodium Hypochlorite Solution, 436 Tablets, 809 Aluminum Hydroxide Gel, 401
Triozinc Paste, 1734 Dimercaprol, 810 Aluminum Hydroxide Gel Fine Gran-
Dermatol, 512 Injection, 811 ules, 402
Deslanoside, 775 Dimorpholamine, 811 Aluminum Potassium Sulfate, 403
Injection, 776 Injection, 812 Sodium Carbonate, 1568
Dexamethasone, 776 Dinoprost, 812 Sodium Sulfite, 1592
Dextran Dionin, 905 Thyroid, 1684
40, 777, 2684 Dioscorea Rhizome, 1847 Yeast, 1779
40 Injection, 778 Powdered, 1847 Droperidol, 841
70, 779 Diphenhydramine, 813 Droxidopa, 842
Sulfate Sodium Sulfur 5, 780 and Bromovalerylurea Powder, 814 Capsules, 842
Sulfate Sodium Sulfur 18, 780 Hydrochloride, 814 Fine Granules, 843
Dextrin, 781 , Phenol and Zinc Oxide Liniment, Dydrogesterone, 844
Dextromethorphan Hydrobromide Hy- 815 Tablets, 845
drate, 782 Tannate, 815
Diagnostic Sodium Citrate Solution, Diphenylhydantoin, 1396 E
1572 Powder, 1396
Diastase, 782 Sodium for Injection, 1398 Ebastine, 845
and Sodium Bicarbonate Powder, Tablets, 1397 Orally Disintegrating Tablets, 846
783 Diphtheria Tablets, 847
and Sodium Bicarbonate Powder, Antitoxin, Equine, Freeze-dried, Ecabet Sodium
Compound, 783 816 Granules, 849
Diazepam, 783 -Purified Pertussis-Tetanus Com- Hydrate, 849
Tablets, 784 bined Vaccine, Absorbed, 816 Ecarazine Hydrochloride, 1700
Dibasic -Tetanus Combined Toxoid, 816 Ecothiopate Iodide, 851
Calcium Phosphate, Anhydrous, -Tetanus Combined Toxoid, Ab- Edaravone, 851
557 sorbed, 816 Injection, 852, 2687
Calcium Phosphate Hydrate, 558 Toxoid, 816 Edrophonium Chloride, 853
Sodium Phosphate Hydrate, 1585 Toxoid for Adult Use, Absorbed, Injection, 854
2846 Index Supplement I, JP XVII

EDTA Sodium Hydrate, 1573 Ether, 898 Saibokuto, 1963, 2783


Elcatonin, 854 Anesthetic, 898 Saikokeishito, 1965, 2785
Eleutherococcus Senticosus Rhizome, Guaiacol Glyceryl, 1002 Saireito, 1968, 2787
1848 Polyoxyethylene Lauryl Alcohol, Scopolia, 1975
Emedastine Fumarate, 857 1144 Shakuyakukanzoto, 1984, 2790
Extended-release Capsules, 858 Ethinylestradiol, 899 Shimbuto, 1985, 2791
Emorfazone, 858 Tablets, 899 Shosaikoto, 1988, 2791
Tablets, 859 Ethionamide, 900 Shoseiryuto, 1990, 2793
Enalapril Maleate, 860 Ethosuximide, 901 Tokakujokito, 1999, 2796
Tablets, 861 Ethoxybenzamide, 897 Tokishakuyakusan, 2002, 2797
Enflurane, 863 Ethyl Yokukansan, 2010, 2798
Enoxacin Hydrate, 863 Aminobenzoate, 901
Entacapone, 2689 Cysteine Hydrochloride, 902 F
Tablets, 2691 L-Cysteine Hydrochloride, 902
Enviomycin Sulfate, 864 Icosapentate, 903 Famotidine, 915
Epalrestat, 865, 2692 Icosapentate Capsules, 904 for Injection, 917
Tablets, 866 Parahydroxybenzoate, 906 Injection, 915
Eperisone Hydrochloride, 867 Ethylenediamine, 903 Powder, 918
Ephedra Herb, 1849 Ethylmorphine Hydrochloride Hy- Tablets, 919
Ephedrine Hydrochloride, 868 drate, 905 Faropenem Sodium
Injection, 869 Etidronate Disodium, 907 for Syrup, 920
Powder, 870 Tablets, 908 Hydrate, 919
Powder, 10z, 870 Etilefrine Hydrochloride, 909 Tablets, 921
Tablets, 870 Tablets, 909 Felbinac, 923
Epimedium Herb, 1850 Etizolam, 910 Cataplasm, 923
Epinephrine, 379 Fine Granules, 911 Tape, 924
Injection, 379 Tablets, 912 Fenbufen, 924
Solution, 380 Etodolac, 913 Fennel, 1851
Epirizole, 871 Etoposide, 914 Oil, 1852
Epirubicin Hydrochloride, 872 Eucalyptus Oil, 1850 Powdered, 1852
Eplerenone, 874 Eucommia Bark, 1851 Fentanyl Citrate, 925
Tablets, 875 Euodia Fruit, 1851, 2755 Ferrous Sulfate Hydrate, 926
Epoetin Exsiccated Gypsum, 1868 Fexofenadine Hydrochloride, 926
Alfa (Genetical Recombination), Extracts Tablets, 927
876 Bakumondoto, 1805, 2747 Filgrastim (Genetical Recombination),
Beta (Genetical Recombination), Belladonna, 1810 929
879 Bofutsushosan, 1813, 2748 Injection, 931
Ergocalciferol, 882 Boiogito, 1817, 2750 Fine Granules
Ergometrine Maleate, 883 Chotosan, 1827, 2751 Cefaclor, 596
Injection, 883 Crude Glycyrrhiza, 1864, 2756 Cefcapene Pivoxil Hydrochloride,
Tablets, 884 Daiokanzoto, 1843, 2754 613
Ergotamine Tartrate, 884 Daisaikoto, 1844, 2754 Cefdinir, 617
Erythromycin, 885, 2692 Glycyrrhiza, 1863, 2755 Cefditoren Pivoxil, 619
Delayed-release Tablets, 886 Goreisan, 2756 Cefteram Pivoxil, 655
Ethylsuccinate, 887 Goshajinkigan, 1865, 2758 Clonazepam, 733
Lactobionate, 887 Hachimijiogan, 1868, 2759 Donepezil Hydrochloride, 826
Stearate, 888 Hangekobokuto, 1871, 2761 Dried Aluminum Hydroxide Gel,
Estazolam, 889 Hangeshashinto, 1873, 2761 402
Estradiol Benzoate, 889 Hochuekkito, 1876, 2763 Droxidopa, 843
Injection (Aqueous Suspension), Juzentaihoto, 1887, 2766 Etizolam, 911
890 Kakkonto, 1889, 2768 Haloperidol, 1005
Estriol, 891 Kakkontokasenkyushin'i, 1892, Ifenprodil Tartrate, 1050
Injection (Aqueous Suspension), 2771 Irsogladine Maleate, 1090
892 Kamikihito, 1895, 2772 Levofloxacin, 1155
Tablets, 892 Kamishoyosan, 1898, 2773 Nifedipine, 1311
Etacrynic Acid, 893 Keishibukuryogan, 1901, 2775 Nifedipine Delayed-release, 1309
Tablets, 893 Maoto, 1912, 2776 Pravastatin Sodium, 1446
Ethacridine Lactate, 376 Mukoi-Daikenchuto, 1917, 2777 Precipitated Calcium Carbonate,
Ethambutol Hydrochloride, 894 Nux Vomica, 1921 548
Ethanol, 895, 2693 Orengedokuto, 1926, 2778 Probucol, 1461
Anhydrous, 896, 2693 Otsujito, 1929, 2778 Quetiapine Fumarate, 1493
for Disinfection, 897 Rikkunshito, 1957, 2780 Risperidone, 1522
Ethenzamide, 897 Ryokeijutsukanto, 1961, 2781 Sarpogrelate Hydrochloride, 1544
Supplement I, JP XVII Index 2847

Tranilast, 1713 Live Attenuated Mumps Vaccine, Concentrated, 993


Troxipide, 1735 1278 Glycerol, 992
Flavin Adenine Dinucleotide Sodium, Live Attenuated Rubella Vaccine, Concentrated, 993
932 1535 Glyceryl Monostearate, 995
Flavoxate Hydrochloride, 934 Mamushi Antivenom, Equine, 1191 Glycine, 995
Flecainide Acetate, 934 Smallpox Vaccine, 1563 Glycyrrhiza, 1862
Tablets, 935 Smallpox Vaccine Prepared in Cell Extract, 1863, 2755
Flomoxef Sodium, 936 Culture, 1563 Extract, Crude, 1864, 2756
for Injection, 938 Tetanus Antitoxin, Equine, 1670 Powdered, 1863
Flopropione, 939 Fritillaria Bulb, 1853 Prepared, 1947
Capsules, 939 Fructose, 967 Gonadorelin Acetate, 996
Fluconazole, 940 Injection, 968 Goreisan Extract, 2756
Capsules, 941 Fudosteine, 968 Goshajinkigan Extract, 1865, 2758
Injection, 942 Tablets, 969 Gramicidin, 1001, 2698
Flucytosine, 943 Furosemide, 970 Granules
Fludiazepam, 944 Injection, 971 Aciclovir, 370
Fludrocortisone Acetate, 944 Tablets, 972 Aldioxa, 387
Fluidextracts Fursultiamine Hydrochloride, 973 Azelastine Hydrochloride, 462
Condurango, 1837 Calcium Paraaminosalicylate, 556
Platycodon, 1943 G Cefaclor Combination, 594
Uva Ursi, 2008 Cefalexin Combination, 602
Flunitrazepam, 945 Gabexate Mesilate, 974 Colestimide, 751
Fluocinolone Acetonide, 946 b-Galactosidase Ecabet Sodium, 849
Fluocinonide, 947 (Aspergillus), 975 L-Isoleucine, L-Leucine and L-Valine,
Fluorescein Sodium, 948 (Penicillium), 975 1096
Fluorometholone, 949 Gallium (67Ga) Citrate Injection, 976 Montelukast Sodium, 2724
Fluorouracil, 950 Gambir, 1853 Pas-calcium, 556
Fluoxymesterone, 950, 2694 Powdered, 1853 Ursodeoxycholic Acid, 1749
Fluphenazine Enanthate, 951 Gardenia Fruit, 1854, 2755 Guaiacol Glyceryl Ether, 1002
Flurazepam Hydrochloride, 952 Powdered, 1854 Guaifenesin, 1002
Flurbiprofen, 953 Gas Gangrene Antitoxin, Equine, 977 Guanabenz Acetate, 1003
Flutamide, 954 Gastrodia Tuber, 1855 Guanethidine Sulfate, 1003
Flutoprazepam, 955 Gefarnate, 977 Gypsum, 1868
Tablets, 955 Gelatin, 978
Fluvoxamine Maleate, 956 Purified, 980 H
Tablets, 958 Gentamicin Sulfate, 982, 2695
Foeniculated Ammonia Spirit, 1852 Ophthalmic Solution, 983 Hachimijiogan Extract, 1868, 2759
Folic Acid, 959, 2694 Gentian, 1856 Haloperidol, 1004
Injection, 959 and Sodium Bicarbonate Powder, Fine Granules, 1005
Tablets, 960 1857 Injection, 1006
Formalin, 961 Powdered, 1856 Tablets, 1006
Water, 961 Geranium Herb, 1857 Halothane, 1007
Formoterol Fumarate Hydrate, 962 Powdered, 1857 Haloxazolam, 1008
Forsythia Fruit, 1852 Ginger, 1857 Hangekobokuto Extract, 1871, 2761
Fosfomycin Powdered, 1858 Hangeshashinto Extract, 1873, 2761
Calcium Processed, 1951 Hedysarum Root, 1875
Calcium for Syrup, 963 Ginseng, 1859 Hemp Fruit, 1876
Calcium Hydrate, 962, 2694 Powdered, 1860 Heparin
Sodium, 964, 2694 Glacial Acetic Acid, 365 Calcium, 1009, 2698
Sodium for Injection, 965 Glehnia Root and Rhizome, 1861 Sodium, 1013, 2699
Fradiomycin Sulfate, 966 Glibenclamide, 984 Sodium Injection, 1017
Freeze-dried Gliclazide, 984 L-Histidine, 1018
BCG Vaccine (for Percutaneous Glimepiride, 985 Hydrochloride Hydrate, 1019
Use), 473 Tablets, 986 Hochuekkito Extract, 1876, 2763
Botulism Antitoxin, Equine, 520 Glucose, 988 Homatropine Hydrobromide, 1020
Diphtheria Antitoxin, Equine, 816 Hydrate, 2695 Homochlorcyclizine Hydrochloride,
Habu Antivenom, Equine, 1004 Injection, 989, 2698 1020
Inactivated Tissue Culture Rabies Purified, 2696 Honey, 1879
Vaccine, 1503 L-Glutamic Acid, 989 Houttuynia Herb, 1880
Japanese Encephalitis Vaccine, L-Glutamine, 990 Human
1111 Glutathione, 991 Chorionic Gonadotrophin, 997
Live Attenuated Measles Vaccine, Glycerin, 992 Chorionic Gonadotrophin for Injec-
1196 and Potash Solution, 994 tion, 999
2848 Index Supplement I, JP XVII

Menopausal Gonadotrophin, 999 Imperata Rhizome, 1880 Docetaxel for, 823


Normal Immunoglobulin, 1021 Indapamide, 1058 Dopamine Hydrochloride, 829
Hycoato Injection, 1348 Tablets, 1059 Doxorubicin Hydrochloride for,
Hydralazine Hydrochloride, 1021 Indenolol Hydrochloride, 1060 837
for Injection, 1022 Indigocarmine, 1061 Edaravone, 852, 2687
Powder, 1022 Injection, 1062 Edrophonium Chloride, 854
Tablets, 1022 Indium (111In) Chloride Injection, Ephedrine Hydrochloride, 869
Hydrochloric Acid, 1023 1062 Epinephrine , 379
Dilute, 1024 Indometacin, 1062 Ergometrine Maleate, 883
Lemonade, 1024 Capsules, 1063 Estradiol Benzoate (Aqueous Suspen-
Hydrochlorothiazide, 1024 Suppositories, 1064 sion), 890
Hydrocortisone, 1025 Influenza HA Vaccine, 1065 Estriol (Aqueous Suspension), 892
Acetate, 1026, 2701 Injection Famotidine, 915
and Diphenhydramine Ointment, Acetylcholine Chloride for, 367 Famotidine for, 917
1027 Aciclovir, 371 Filgrastim (Genetical Recombina-
Butyrate, 1028, 2701 Aciclovir for, 371 tion), 931
Sodium Phosphate, 1028 Adrenaline, 379 Flomoxef Sodium for, 938
Sodium Succinate, 1030 Alendronate Sodium, 389 Fluconazole, 942
Succinate, 1031 Alprostadil, 398 Folic Acid, 959
Hydrocotarnine Hydrochloride Hy- Amikacin Sulfate, 409 Fosfomycin Sodium for, 965
drate, 1031 Amikacin Sulfate for, 409 Fructose, 968
Hydrogenated Oil, 1032 Aminophylline, 410 Furosemide, 971
Hydrophilic Amphotericin B for, 427 Gallium (67Ga) Citrate, 976
Cream, 756 Ampicillin Sodium and Sulbactam Glucose, 989, 2698
Petrolatum, 1387 Sodium for, 433 Haloperidol, 1006
Hydrous Lanolin, 1903 Ampicillin Sodium for, 432 Heparin Sodium, 1017
Hydroxocobalamin Acetate, 1033, Arbekacin Sulfate, 440 Human Chorionic Gonadotrophin
2701 L-Arginine Hydrochloride, 443 for, 999
Hydroxypropylcellulose, 1033, 2702 Ascorbic Acid, 446 Hycoato, 1348
Hydroxypropylmethylcellulose, 1038 Atropine Sulfate, 456 Hydralazine Hydrochloride for,
Hydroxyzine Aztreonam for, 467 1022
Hydrochloride, 1036 Benzylpenicillin Potassium for, 487 Idarubicin Hydrochloride for, 1047
Pamoate, 1036 Calcium Chloride, 551 Imipenem and Cilastatin Sodium for,
Hymecromone, 1037 Carboplatin, 582 1056
Hypromellose, 1038, 2704 Cefazolin Sodium for, 608 Indigocarmine, 1062
Acetate Succinate, 1040 Cefepime Dihydrochloride for, 622 Indium (111In) Chloride, 1062
Capsules, 574 Cefmetazole Sodium for, 628 Insulin Glargine (Genetical Recombi-
Phthalate, 1041 Cefoperazone Sodium and Sulbac- nation), 1067
tam Sodium for, 632 Insulin Human (Genetical Recombi-
I Cefoperazone Sodium for, 2677 nation), 1070
Cefotiam Hydrochloride for, 640 Interferon Alfa (NAMALWA),
Ibudilast, 1042 Cefozopran Hydrochloride for, 641 1074
Ibuprofen, 1043 Ceftazidime for, 653 Iodinated (131I) Human Serum Albu-
Piconol, 1044 Chlorpheniramine Maleate, 688 min, 1075
Piconol Cream, 1044 Chlorpromazine Hydrochloride, Iohexol, 1082, 2710
Piconol Ointment, 1045 692 Iopamidol, 1084
Ichthammol, 1046 Chorionic Gonadotrophin for, 999 Isepamicin Sulfate, 1094
Idarubicin Hydrochloride, 1046 Clindamycin Phosphate, 725 Isoniazid, 1100
for Injection, 1047 Compound Hycodenone, 1347 Isotonic Sodium Chloride, 1571
Idoxuridine, 1048 Compound Oxycodone, 1347 Levallorphan Tartrate, 1152
Ophthalmic Solution, 1049 Compound Oxycodone and Atro- Levofloxacin, 1156
Ifenprodil Tartrate, 1050 pine, 1348 Lidocaine, 1162
Fine Granules, 1050 Cyanocobalamin, 761 Lidocaine Hydrochloride, 1162
Tablets, 1051 Dehydrocholate Sodium, 773 Lincomycin Hydrochloride, 1164
Imidapril Hydrochloride, 1052 Dehydrocholic Acid, 773 Magnesium Sulfate, 1190
Tablets, 1053 Deslanoside, 776 D-Mannite, 1195
Imipenem Dextran 40, 778 D-Mannitol, 1195
and Cilastatin Sodium for Injection, Digoxin, 797 Meglumine Iotalamate, 1206
1056 Dimercaprol, 811 Meglumine Sodium Amidotrizoate,
Hydrate, 1054 Dimorpholamine, 812 1207
Imipramine Hydrochloride, 1057 Diphenylhydantoin Sodium for, Mepivacaine Hydrochloride, 1213
Tablets, 1057 1398 Meropenem for, 1219
Immature Orange, 1880 Docetaxel, 822 Metenolone Enanthate, 1222
Supplement I, JP XVII Index 2849

Minocycline Hydrochloride for, Streptomycin Sulfate for, 1609 Ipecac, 1881


1256 Sulfobromophthalein Sodium, Powdered, 1881
Mitomycin C for, 1262 1619 Syrup, 1882
Morphine and Atropine, 1273 Sulpyrine, 1623 Ipratropium Bromide Hydrate, 1086
Morphine Hydrochloride, 1271 Suxamethonium Chloride, 1628, Ipriflavone, 1087
Nartograstim for (Genetical Recom- 2737 Tablets, 1088
bination), 1293 Suxamethonium Chloride for, 1628 Iproveratril Hydrochloride, 1761
Neostigmine Methylsulfate, 1297 Tazobactam and Piperacillin for, Irbesartan, 1089
Nicardipine Hydrochloride, 1299 1645 and Amlodipine Besilate Tablets,
Nicotinic Acid, 1307 Teceleukin for (Genetical Recombi- 2711
Noradrenaline, 1322 nation), 1652 Tablets, 2710
Noradrenaline Hydrochloride, 1322 Testosterone Enanthate, 1668 Irsogladine Maleate, 1090
Norepinephrine, 1322 Testosterone Propionate, 1669 Fine Granules, 1090
Norepinephrine Hydrochloride, Thallium (201Tl) Chloride, 1672 Tablets, 1091
1322 Thiamine Chloride Hydrochloride, Isepamicin Sulfate, 1093
Operidine, 1385 1675 Injection, 1094
Opium Alkaloids and Atropine, Thiamylal Sodium for, 1678 Isoflurane, 1094
1338 Thiopental Sodium for, 1681 L-Isoleucine, 1096
Opium Alkaloids and Scopolamine, Tobramycin, 1696 , L-Leucine and L-Valine Granules,
1339 Tranexamic Acid, 1710 1096
Opium Alkaloids Hydrochlorides, Vancomycin Hydrochloride for, Isomalt, 1098
1337 1758 Hydrate, 1098
Oxytocin, 1355 Vasopressin, 1759, 2740 Isoniazid, 1099
Ozagrel Sodium, 1357 Vinblastine Sulfate for, 1763 Injection, 1100
Ozagrel Sodium for, 1357 Vitamin B1 Hydrochloride, 1675 Tablets, 1101
Panipenem and Betamipron for, Vitamin B2 Phosphate Ester, 1516 Isophane Insulin Human (Genetical
1361 Vitamin B6, 1490 Recombination) Injectable Aque-
Papaverine Hydrochloride, 1364 Vitamin B12, 761 ous Suspension, 2705
Pazufloxacin Mesilate, 2728 Vitamin C, 446 l-Isoprenaline Hydrochloride, 1102
Peplomycin Sulfate for, 1380 Voriconazole for, 2743 Isopropanol, 1102
Pethidine Hydrochloride, 1385 Water for, 1774 Isopropyl Alcohol, 1102
Phenolsulfonphthalein, 1393 Weak Opium Alkaloids and Scopola- Isopropylantipyrine, 1103
Phenytoin Sodium for, 1398 mine, 1340 Isosorbide, 1103
Piperacillin Sodium for, 1418 Xylitol, 1778 Dinitrate, 1104
Prednisolone Sodium Succinate for, Insulin Dinitrate Tablets, 1105
1456 Aspart (Genetical Recombination), Mononitrate 70z/Lactose 30z,
Procainamide Hydrochloride, 1463 2708 1105, 2714
Procaine Hydrochloride, 1465 Glargine (Genetical Recombination), Mononitrate Tablets, 1107
Progesterone, 1470 1065 Isotonic
Protamine Sulfate, 1484 Glargine (Genetical Recombination) Salt Solution, 1571
Purified Sodium Hyaluronate, 1576 Injection, 1067 Sodium Chloride Injection, 1571
Pyridoxine Hydrochloride, 1490 Human (Genetical Recombination), Sodium Chloride Solution, 1571
Reserpine, 1508 1068 Isoxsuprine Hydrochloride, 1108
Riboflavin Phosphate, 1516 Human (Genetical Recombination) Tablets, 1109
Riboflavin Sodium Phosphate, Injection, 1070 Itraconazole, 1110
1516 Interferon Alfa (NAMALWA), 1071
Roxatidine Acetate Hydrochloride Injection, 1074 J
for, 1533 Iodinated (131I) Human Serum Albumin
Sivelestat Sodium for, 1562 Injection, 1075 Japanese
Sodium Bicarbonate, 1566 Iodine, 1075 Angelica Root, 1883
Sodium Chloride, 0.9z, 1571 Glycerin, Compound, 1077 Angelica Root, Powdered, 1883
Sodium Chloride, 10z, 1570 Glycerin, Dental, 1078 Encephalitis Vaccine, 1111
Sodium Chromate (51Cr), 1571 , Salicylic Acid and Phenol Spirit, Gentian, 1884
Sodium Citrate for Transfusion, 1079 Gentian, Powdered, 1884
1572 Tincture, 1076 Valerian, 1884
Sodium Iodohippurate (131I), 1580 Tincture, Dilute, 1076 Valerian, Powdered, 1885
Sodium Iotalamate, 1580 Iodoform, 1080 Zanthoxylum Peel, 1885
Sodium Pertechnetate (99mTc), 1585 Iohexol, 1080 Zanthoxylum Peel, Powdered, 1886
Sodium Thiosulfate, 1593 Injection, 1082, 2710 Josamycin, 1112
Spectinomycin Hydrochloride for, Iopamidol, 1083 Propionate, 1113
1598 Injection, 1084 Tablets, 1113
Sterile Water for, in Containers, Iotalamic Acid, 1085 Jujube, 1886, 2765
1775 Iotroxic Acid, 1086 Seed, 1886, 2765
2850 Index Supplement I, JP XVII

Juzentaihoto Extract, 1887, 2766 Acetate, 1127 Bark, 1909


Tartrate, 1128 Fruit, 1909
K Leuprorelin Acetate, 1150 Lysine Hydrochloride, 1180
Levallorphan Tartrate, 1152 L-Lysine
Kainic Acid Injection, 1152 Acetate, 1179
and Santonin Powder, 1115 Levodopa, 1153 Hydrochloride, 1180
Hydrate, 1114 Levofloxacin Lysozyme Hydrochloride, 1181
Kakkonto Extract, 1889, 2768 Fine Granules, 1155
Kakkontokasenkyushin'i Extract, Hydrate, 1154 M
1892, 2771 Injection, 1156
Kallidinogenase, 1116 Ophthalmic Solution, 1156 Macrogol
Kamikihito Extract, 1895, 2772 Tablets, 1157 400, 1182
Kamishoyosan Extract, 1898, 2773 Levomepromazine Maleate, 1159 1500, 1182
Kanamycin Levothyroxine Sodium 4000, 1183
Monosulfate, 1118 Hydrate, 1159 6000, 1183
Sulfate, 1119 Tablets, 1160 20000, 1184
Kaolin, 1120 Lidocaine, 1161 Ointment, 1184
Keishibukuryogan Extract, 1901, Hydrochloride Injection, 1162 Magnesium
2775 Injection, 1162 Aluminosilicate, 2715
Ketamine Hydrochloride, 1120 Light Aluminometasilicate, 2716
Ketoconazole, 1121 Anhydrous Silicic Acid, 1552 Carbonate, 1185
Cream, 1122 Liquid Paraffin, 1365 Oxide, 1186
Lotion, 1123 Lilium Bulb, 1906 Silicate, 1187
Solution, 1123 Limaprost Alfadex, 1162 Stearate, 1188
Ketoprofen, 1124 Lincomycin Hydrochloride Sulfate Hydrate, 1189
Ketotifen Fumarate, 1125 Hydrate, 1164 Sulfate Injection, 1190
Kitasamycin, 1126 Injection, 1164 Sulfate Mixture, 1190
Acetate, 1127 Lindera Root, 1906 Magnolia
Tartrate, 1128 Liniments Bark, 1910
Koi, 1903 Diphenhydramine, Phenol and Zinc Bark, Powdered, 1910
Oxide, 815 Flower, 1911
L Phenol and Zinc Oxide, 1392 Mallotus Bark, 1911
Liothyronine Sodium, 1165 Malt, 1912
Labetalol Hydrochloride, 1129 Tablets, 1166 Maltose Hydrate, 1190
Tablets, 1130 Liquefied Phenol, 1391 Manidipine Hydrochloride, 1192
Lactic Acid, 1131 Liquid Paraffin, 1365 Tablets, 1193
L-Lactic Acid, 1131 Lisinopril D-Mannite Injection, 1195
Lactose, 1133 Hydrate, 1167 D-Mannitol, 1194, 2718
Hydrate, 1133, 2715 Tablets, 1168 Injection, 1195
Anhydrous, 1132, 2714 Lithium Carbonate, 1169 Maoto Extract, 1912, 2776
Lactulose, 1134 Lithospermum Root, 1907 Maprotiline Hydrochloride, 1196
Lafutidine, 1135 Live Oral Poliomyelitis Vaccine, 1428 Meclofenoxate Hydrochloride, 1197
Tablets, 1136 Lobenzarit Sodium, 1171 Mecobalamin, 1197
Lanatoside C, 1137, 2715 Longan Aril, 1907 Tablets, 1198
Tablets, 1138, 2715 Longgu, 1907 Medazepam, 1200
Lanolin Powdered, 1908 Medicinal
Hydrous, 1903 Lonicera Leaf and Stem, 1908 Carbon, 1200
Purified, 1904 Loquat Leaf, 1909 Soap, 1201
Lansoprazole, 1139 Lorazepam, 1171 Medroxyprogesterone Acetate, 1202
Delayed-release Capsules, 1140 Losartan Potassium, 1172 Mefenamic Acid, 1203
Delayed-release Orally Disintegrating and Hydrochlorothiazide Tablets, Mefloquine Hydrochloride, 1203
Tablets, 1141 1174 Mefruside, 1204
Lard, 1905 Tablets, 1173 Tablets, 1205
Latamoxef Sodium, 1143 Lotions Meglumine, 1206
Lauromacrogol, 1144 Ketoconazole, 1123 Iotalamate Injection, 1206
Lemonades Sulfur and Camphor, 1619 Sodium Amidotrizoate Injection,
Hydrochloric Acid, 1024 Tacalcitol, 1630 1207
Lenampicillin Hydrochloride, 1144 Low Substituted Hydroxypropylcellu- Melphalan, 1208
Lenograstim (Genetical Recombina- lose, 1035, 2703 Menatetrenone, 1209
tion), 1146 Loxoprofen Sodium Mentha
Leonurus Herb, 1905 Hydrate, 1177 Herb, 1914
L-Leucine, 1149 Tablets, 1178 Oil, 1915
Leucomycin, 1126 Lycium Water, 1915
Supplement I, JP XVII Index 2851

dl-Menthol, 1210 Miconazole, 1249 Narcotine, 1326


l-Menthol, 1211 Nitrate, 1249 Hydrochloride, 1327
Mepenzolate Bromide, 1211 Microcrystalline Cellulose, 664 Nartograstim (Genetical Recombina-
Mepirizole, 871 Micronomicin Sulfate, 1250 tion), 1291
Mepitiostane, 1212 Midecamycin, 1251 for Injection, 1293
Mepivacaine Hydrochloride, 1213 Acetate, 1252 Natamycin, 1404
Injection, 1213 Miglitol, 1253 Nateglinide, 1294
Mequitazine, 1214 Migrenin, 1254 Tablets, 1295
Tablets, 1215 Minocycline Hydrochloride, 1255 Natural Aluminum Silicate, 404
Merbromin, 1216 for Injection, 1256 Nelumbo Seed, 1919
Solution, 1217 Tablets, 1256 Neomycin Sulfate, 966
Mercaptopurine Hydrate, 1215 Mitiglinide Calcium Neostigmine Methylsulfate, 1297
Mercurochrome, 1216, 2718 Hydrate, 1258 Injection, 1297
Solution, 1217, 2718 Tablets, 1259 Nicardipine Hydrochloride, 1298
Meropenem Mitomycin C, 1261 Injection, 1299
for Injection, 1219 for Injection, 1262 Nicergoline, 1300
Hydrate, 1217 Mizoribine, 1262 Powder, 1301
Mesalazine, 2718 Tablets, 1263 Tablets, 1302
Extended-release Tablets, 2720 Monobasic Calcium Phosphate Hy- Niceritrol, 1303
Mestranol, 1220 drate, 558 Nicomol, 1304
Metenolone Monosodium Trichloroethyl Phos- Tablets, 1304
Acetate, 1220 phate, 1725 Nicorandil, 1305
Enanthate, 1221 Syrup, 1726 Nicotinamide, 1306
Enanthate Injection, 1222 Montelukast Sodium, 1264 Nicotinic Acid, 1307
Metformin Hydrochloride, 1222 Chewable Tablets, 1267 Injection, 1307
Tablets, 1223 Granules, 2724 Nifedipine, 1308
Methamphetamine Hydrochloride, Tablets, 1269 Delayed-release Fine Granules,
1223 Morphine 1309
L-Methionine, 1224 and Atropine Injection, 1273 Extended-release Capsules, 1310
Methotrexate, 1225 Hydrochloride Hydrate, 1270 Fine Granules, 1311
Capsules, 1225 Hydrochloride Injection, 1271 Nilvadipine, 1312
Tablets, 2722 Hydrochloride Tablets, 1272 Tablets, 1313
Methoxsalen, 1227 Sulfate Hydrate, 1274 Nitrazepam, 1314
Methyl Mosapride Citrate Nitrendipine, 1314
Parahydroxybenzoate, 1235 Hydrate, 1275 Tablets, 1315
Salicylate, 1238 Powder, 1276 Nitrogen, 1316
Salicylate Spirit, Compound, 1238 Tablets, 1277 Nitroglycerin Tablets, 1317
Methylbenactyzium Bromide, 1227 Moutan Bark, 1915 Nitrous Oxide, 1318
Methylcellulose, 1228, 2723 Powdered, 1916 Nizatidine, 1319
Methyldopa Mukoi-Daikenchuto Extract, 1917, Capsules, 1320
Hydrate, 1229 2777 Noradrenaline, 1321, 2726
Tablets, 1230 Mulberry Bark, 1918 Hydrochloride Injection, 1322
dl-Methylephedrine Hydrochloride, Mupirocin Calcium Injection, 1322
1231 Hydrate, 1279 Norepinephrine, 1321
Powder, 1232 Ointment, 1280 Hydrochloride Injection, 1322
Powder, 10z, 1232 Injection, 1322
Methylergometrine Maleate, 1233 N Norethisterone, 1322
Tablets, 1233 Norfloxacin, 1323
Methylprednisolone, 1236 Nabumetone, 1281 Norgestrel, 1324
Succinate, 1236 Tablets, 1282 and Ethinylestradiol Tablets, 1324
Methylrosanilinium Chloride, 1237 Nadolol, 1283 Nortriptyline Hydrochloride, 1326
Methyltestosterone, 1239 Nafamostat Mesilate, 1283 Noscapine, 1326
Tablets, 1240 Naftopidil, 1284 Hydrochloride Hydrate, 1327
Meticrane, 1241 Orally Disintegrating Tablets, 1285 Notopterygium, 1919
Metildigoxin, 1242 Tablets, 1286 Nuphar Rhizome, 1919
Metoclopramide, 1243 Nalidixic Acid, 1287 Nutmeg, 1920
Tablets, 1243 Naloxone Hydrochloride, 1288 Nux Vomica, 1920
Metoprolol Tartrate, 1244 Naphazoline Extract, 1921
Tablets, 1245 and Chlorpheniramine Solution, Extract Powder, 1922
Metronidazole, 1246 1290 Tincture, 1922
Tablets, 1246 Hydrochloride, 1289 Nystatin, 1328
Metyrapone, 1247 Nitrate, 1289
Mexiletine Hydrochloride, 1248 Naproxen, 1291
2852 Index Supplement I, JP XVII

Oriental Bezoar, 1928, 2778 Pentazocine, 1376


O Otsujito Extract, 1929, 2779 Pentobarbital Calcium, 1377, 2728
Oxapium Iodide, 1342 Tablets, 2729
Ofloxacin, 1328 Oxaprozin, 1343 Pentoxyverine Citrate, 1378
Ointments Oxazolam, 1343 Peony Root, 1935
Aciclovir, 372 Oxetacaine, 1344 Powdered, 1936
Aciclovir Ophthalmic, 372 Oxethazaine, 1344 Peplomycin Sulfate, 1379
Acrinol and Zinc Oxide, 378 Oxprenolol Hydrochloride, 1345 for Injection, 1380
Betamethasone Valerate and Gen- Oxybuprocaine Hydrochloride, 1345 Perilla Herb, 1937
tamicin Sulfate, 504 Oxycodone Hydrochloride Hydrate, Perphenazine, 1381
Hydrocortisone and Diphenhydra- 1346 Maleate, 1383
mine, 1027 Oxydol, 1349 Maleate Tablets, 1383
Ibuprofen Piconol, 1045 Oxygen, 1350 Tablets, 1382
Macrogol, 1184 Oxymetholone, 1351 Pethidine Hydrochloride, 1385
Mupirocin Calcium, 1280 Oxytetracycline Hydrochloride, 1351, Injection, 1385
Polyethylene Glycol, 1184 2726 Petroleum Benzin, 1387
Simple, 1993 Oxytocin, 1353 Peucedanum Root, 1937
Sulfur, Salicylic Acid and Thianthol, Injection, 1355 Pharbitis Seed, 1938
1620 Oyster Shell, 1932 Phellodendron
Tacalcitol, 1631 Powdered, 1932 , Albumin Tannate and Bismuth Sub-
White, 1775 Ozagrel Sodium, 1356 nitrate Powder, 1940
Zinc Oxide, 1784 for Injection, 1357 Bark, 1938
Olive Oil, 1923 Injection, 1357 Bark, Powdered, 1939
Olmesartan Medoxomil, 1329 Powder for Cataplasm, Compound,
Tablets, 1330 P 1940
Olopatadine Hydrochloride, 1332 Phenazone, 437
Tablets, 1333 Panax Japonicus Rhizome, 1932 Phenethicillin Potassium, 1388
Omeprazole, 1334 Powdered, 1933 Phenobarbital, 1389
Delayed-release Tablets, 1335 Pancreatin, 1358 Powder, 1390
Operidine, 1385 Pancuronium Bromide, 1358 Powder, 10z, 1390
Injection, 1385 Panipenem, 1359 Phenol, 1390
Ophiopogon Root, 1923 and Betamipron for Injection, 1361 and Zinc Oxide Liniment, 1392
Ophthalmic Solution Pantethine, 1362 for Disinfection, 1391
Chloramphenicol and Colistin Sodi- Papaverine Hydrochloride, 1363 Liquefied, 1391
um Methanesulfonate, 2679 Injection, 1364 with Camphor, Dental, 1392
Dibekacin Sulfate, 785 Paracetamol, 363 Phenolated Water, 1392
Dorzolamide Hydrochloride, 831 Paraffin, 1364 for Disinfection, 1392
Gentamicin Sulfate, 983 Light Liquid, 1365 Phenolsulfonphthalein, 1393
Idoxuridine, 1049 Liquid, 1365 Injection, 1393
Levofloxacin, 1156 Paraformaldehyde, 1366 L-Phenylalanine, 1394
Pemirolast Potassium, 1373 Paste, Dental, 1367 Phenylbutazone, 1395
Purified Sodium Hyaluronate, 1577 Parnaparin Sodium, 1367 Phenylephrine Hydrochloride, 1395
Silver Nitrate, 1557 Paroxetine Hydrochloride Phenytoin, 1396
Tranilast, 1714 Hydrate, 1369 Powder, 1396
Zinc Sulfate, 1785 Tablets, 1371 Sodium for Injection, 1398
Ophthalmic Ointment Pas-calcium Tablets, 1397
Aciclovir, 372 Granules, 556 Phytomenadione, 1398
Opium Hydrate, 555 Phytonadione, 1398
Ipecac Powder, 1925 Paste Picrasma Wood, 1941
Powder, Diluted, 1924 Arsenical, 445 Powdered, 1941
Powdered, 1924 Paraformaldehyde, Dental, 1367 Pilocarpine Hydrochloride, 1399
Tincture, 1925 Triozinc, Dental, 1734 Tablets, 1400
Opium Alkaloids Pazufloxacin Mesilate, 2726 Pilsicainide Hydrochloride
and Atropine Injection, 1338 Injection, 2728 Capsules, 1402
and Scopolamine Injection, 1339 Peach Kernel, 1933 Hydrate, 1402
Hydrochlorides, 1336 Powdered, 1934 Pimaricin, 1404
Hydrochlorides Injection, 1337 Peanut Oil, 1935 Pimozide, 1405
Orange Pemirolast Potassium, 1372 Pindolol, 1406
Oil, 1925 for Syrup, 1374 Pinellia Tuber, 1941
Peel Syrup, 1926 Ophthalmic Solution, 1373 Pioglitazone Hydrochloride, 1406
Peel Tincture, 1926 Tablets, 1375 and Glimepiride Tablets, 1408
Orciprenaline Sulfate, 1341 Penbutolol Sulfate, 1376 and Metformin Hydrochloride
Orengedokuto Extract, 1926, 2778 Penicillin G Potassium, 486 Tablets, 1411
Supplement I, JP XVII Index 2853

Tablets, 1407 Povidone, 1439, 2731 Vitamin C, 446


Pipemidic Acid Hydrate, 1414 -Iodine, 1441 Zinc Oxide Starch, 1785
Piperacillin Powder Powdered
Hydrate, 1414 Ascorbic Acid, 446 Acacia, 1791
Sodium, 1416 Chlordiazepoxide, 680 Agar, 1793
Sodium for Injection, 1418 Chlorpheniramine Maleate, 689 Alisma Tuber, 1794, 2747
Piperazine Codeine Phosphate, 1z, 746 Aloe, 1795
Adipate, 1418 Codeine Phosphate, 10z, 747 Amomum Seed, 1797
Phosphate Hydrate, 1419 Compound Diastase and Sodium Bi- Atractylodes Lancea Rhizome,
Phosphate Tablets, 1419 carbonate, 783 1804
Pirarubicin, 1420 Compound Phellodendron, for Atractylodes Rhizome, 1805
Pirenoxine, 1421 Cataplasm, 1940 Calumba, 1822
Pirenzepine Hydrochloride Hydrate, Compound Rhubarb and Senna, Capsicum, 1823
1422 1957 Cellulose, 667
Piroxicam, 1423 Compound Scopolia Extract and Di- Cinnamon Bark, 1831
Pitavastatin Calcium astase, 1976 Clove, 1833
Hydrate, 1424 Compound Vitamin B, 1765 Cnidium Rhizome, 1835
Tablets, 1425 Diastase and Sodium Bicarbonate, Coix Seed, 1836
Pivmecillinam Hydrochloride, 1427 783 Coptis Rhizome, 1838
Tablets, 1428 Dihydrocodeine Phosphate, 1z, Corydalis Tuber, 1841
Plantago 800 Cyperus Rhizome, 1843, 2753
Herb, 1942 Dihydrocodeine Phosphate, 10z, Dioscorea Rhizome, 1847
Seed, 1942 801 Fennel, 1852
Platycodon Diluted Opium, 1924 Gambir, 1853
Fluidextract, 1943 Diphenhydramine and Gardenia Fruit, 1854
Root, 1942, 2779 Bromovalerylurea, 814 Gentian, 1856
Root, Powdered, 1943 Diphenylhydantoin, 1396 Geranium Herb, 1857
Pogostemon Herb, 1944 Ephedrine Hydrochloride, 870 Ginger, 1858
Polyethylene Glycol Ephedrine Hydrochloride, 10z, Ginseng, 1860
400, 1182 870 Glycyrrhiza, 1863
1500, 1182 Famotidine, 918 Ipecac, 1881
4000, 1183 Gentian and Sodium Bicarbonate, Japanese Angelica Root, 1883
6000, 1183 1857 Japanese Gentian, 1884
20000, 1184 Hydralazine Hydrochloride, 1022 Japanese Valerian, 1885
Ointment, 1184 Kainic Acid and Santonin, 1115 Japanese Zanthoxylum Peel, 1886
Polygala Root, 1944 dl-Methylephedrine Hydrochloride, Longgu, 1908
Powdered, 1944 1232 Magnolia Bark, 1910
Polygonatum Rhizome, 1945 dl-Methylephedrine Hydrochloride, Moutan Bark, 1916
Polygonum Root, 1945 10z, 1232 Opium, 1924
Polymixin B Sulfate, 1429, 2730 Mosapride Citrate, 1276 Oyster Shell, 1932
Polyoxyethylene Lauryl Alcohol Ether, Nicergoline, 1301 Panax Japonicus Rhizome, 1933
1144 Opium Ipecac, 1925 Peach Kernel, 1934
Polyoxyl 40 Stearate, 1430 Phellodendron, Albumin Tannate Peony Root, 1936
Polyporus Sclerotium, 1946 and Bismuth Subnitrate, 1940 Phellodendron Bark, 1939
Powdered, 1946 Phenobarbital, 1390 Picrasma Wood, 1941
Polysorbate 80, 1430, 2730 Phenobarbital, 10z, 1390 Platycodon Root, 1943
Polyvidone, 1439 Phenytoin, 1396 Polygala Root, 1944
Polyvinylpyrrolidone, 1439 Reserpine, 1508 Polyporus Sclerotium, 1946
Poria Sclerotium, 1946 Reserpine, 0.1z, 1508 Poria Sclerotium, 1947
Powdered, 1947 Riboflavin, 1514 Processed Aconite Root, 1949
Potash Soap, 1432 Scopolia Extract, 1975 Rhubarb, 1956
Potassium Scopolia Extract and Carbon, 1976 Rose Fruit, 1960
Bromide, 1432 Scopolia Extract and Ethyl Scutellaria Root, 1979
Canrenoate, 1433 Aminobenzoate, 1976 Senega, 1981
Carbonate, 1433 Scopolia Extract, Papaverine and Senna Leaf, 1982
Chloride, 1434 Ethyl Aminobenzoate, 1977, Smilax Rhizome, 1993
Clavulanate, 1434 2790 Sophora Root, 1995
Guaiacolsulfonate, 1436 Swertia and Sodium Bicarbonate, Sweet Hydrangea Leaf, 1996, 2796
Hydroxide, 1436 1998 Swertia Herb, 1997
Iodide, 1437 Thiamine Chloride Hydrochloride, Tragacanth, 2004
Permanganate, 1438 1675 Turmeric, 2006, 2798
Sulfate, 1438 Vitamin B1 Hydrochloride, 1675 Pranlukast Hydrate, 1442
Potato Starch, 1602 Vitamin B2, 1514 Pranoprofen, 1443
2854 Index Supplement I, JP XVII

Prasterone Sodium Sulfate Hydrate, Injection, 1484 and Senna Powder, Compound,
1444 Prothionamide, 1484 1957
Pravastatin Sodium, 1444 Protirelin, 1485 Powdered, 1956
Fine Granules, 1446 Tartrate Hydrate, 1486 Ribavirin, 1511
Solution, 1447 Prunella Spike, 1951 Capsules, 1512
Tablets, 1448 Pueraria Root, 1952 Riboflavin, 1513
Prazepam, 1450 Pullulan, 1487 Butyrate, 1514
Tablets, 1450 Capsules, 574 Phosphate, 1515
Prazosin Hydrochloride, 1451 Purified Phosphate Injection, 1516
Precipitated Calcium Carbonate, 548 Dehydrocholic Acid, 772 Powder, 1514
Fine Granules, 548 Gelatin, 980 Sodium Phosphate, 1515
Tablets, 549 Glucose, 2696 Sodium Phosphate Injection, 1516
Prednisolone, 1452 Lanolin, 1904 Ribostamycin Sulfate, 1517
Acetate, 1454 Shellac, 1551 Rice Starch, 1603
Sodium Phosphate, 1455 Sodium Hyaluronate, 1575 Rifampicin, 1518
Sodium Succinate for Injection, Sodium Hyaluronate Injection, Capsules, 1519
1456 1576 Rikkunshito Extract, 1957, 2780
Succinate, 1456 Sodium Hyaluronate Ophthalmic So- Ringer's Solution, 1520
Tablets, 1453 lution, 1577 Risperidone, 1521
Prepared Glycyrrhiza, 1947 Water, 1773 Fine Granules, 1522
Primidone, 1458 Water in Containers, 1774 Oral Solution, 1523
Probenecid, 1458 Pyrantel Pamoate, 1487 Tablets, 1524
Tablets, 1459 Pyrazinamide, 1488 Ritodrine Hydrochloride, 1525
Probucol, 1460 Pyridostigmine Bromide, 1489 Tablets, 1526
Fine Granules, 1461 Pyridoxal Phosphate Hydrate, 2731 Rokitamycin, 1528, 2733
Tablets, 1462 Pyridoxine Hydrochloride, 1489 Tablets, 1529, 2733
Procainamide Hydrochloride, 1462 Injection, 1490 Rose Fruit, 1959
Injection, 1463 Pyroxylin, 1491 Powdered, 1960
Tablets, 1463 Pyrrolnitrin, 1491 Rosin, 1960
Procaine Hydrochloride, 1464 Roxatidine Acetate Hydrochloride,
Injection, 1465 Q 1530
Procarbazine Hydrochloride, 1466 Extended-release Capsules, 1531
Procaterol Hydrochloride Hydrate, Quercus Bark, 1952 Extended-release Tablets, 1532
1466 Quetiapine Fumarate, 1492 for Injection, 1533
Processed Fine Granules, 1493 Roxithromycin, 1534
Aconite Root, 1948 Tablets, 1494 Tablets, 2733
Aconite Root, Powdered, 1949 Quick Lime, 554 Royal Jelly, 1960
Ginger, 1951 Quinapril Hydrochloride, 1496 Ryokeijutsukanto Extract, 1961, 2781
Prochlorperazine Maleate, 1467 Tablets, 1497
Tablets, 1468 Quinidine Sulfate Hydrate, 1498 S
Progesterone, 1469 Quinine
Injection, 1470 Ethyl Carbonate, 1499 Saccharated Pepsin, 1535
Proglumide, 1470 Hydrochloride Hydrate, 1500 Saccharin, 1535
L-Proline, 1471 Sulfate Hydrate, 1501 Sodium, 1536
Promethazine Hydrochloride, 1472 Sodium Hydrate, 1536, 2734
Propafenone Hydrochloride, 1473 R Safflower, 1962
Tablets, 1474 Saffron, 1963
Propantheline Bromide, 1475 Rabeprazole Sodium, 1502 Saibokuto Extract, 1963, 2783
Propiverine Hydrochloride, 1476 Ranitidine Hydrochloride, 1503 Saikokeishito Extract, 1965, 2785
Tablets, 1477 Rape Seed Oil, 1953, 2780 Saireito Extract, 1968, 2787
Propranolol Hydrochloride, 1478 Rebamipide, 1504 Salazosulfapyridine, 1537
Tablets, 1479 Tablets, 1506 Salbutamol Sulfate, 1538
Propylene Glycol, 1480 Red Ginseng, 1953 Salicylated Alum Powder, 1542
Propyl Parahydroxybenzoate, 1481 Rehmannia Root, 1954 Salicylic Acid, 1539
Propylthiouracil, 1482 Reserpine, 1507 Adhesive Plaster, 1540
Tablets, 1482 Injection, 1508 Spirit, 1540
Propyphenazone, 1103 Powder, 1508 Spirit, Compound, 1541
Prostaglandin Powder, 0.1z, 1508 Salvia Miltiorrhiza Root, 1971
E1, 396 Tablets, 1509 Santonin, 1542
E1 a-Cyclodextrin Clathrate Com- Retinol Saponated Cresol Solution, 757
pound, 400 Acetate, 1510 Saposhnikovia Root and Rhizome,
F2a, 812 Palmitate, 1510 1971, 2789
Protamine Sulfate, 1483 Rhubarb, 1955 Sappan Wood, 1972
Supplement I, JP XVII Index 2855

Sarpogrelate Hydrochloride, 1543 Benzoate, 1565 Epinephrine, 380


Fine Granules, 1544 Bicarbonate, 1566 Glycerin and Potash, 994
Tablets, 1545 Bicarbonate and Bitter Tincture Mix- Isotonic Salt, 1571
Saussurea Root, 1972 ture, 1994 Isotonic Sodium Chloride, 1571
Schisandra Fruit, 1973 Bicarbonate Injection, 1566 Ketoconazole, 1123
Schizonepeta Spike, 1973 Bisulfite, 1567 Merbromin, 1217
Scopolamine Borate, 1567 Mercurochrome, 1217, 2718
Butylbromide, 1547 Bromide, 1568 Naphazoline and Chlorpheniramine,
Hydrobromide Hydrate, 1548 Carbonate Hydrate, 1569 1290
Scopolia Chloride, 1569 Pravastatin Sodium, 1447
Extract, 1974 Chloride Injection, 0.9z, 1571 Ringer's, 1520
Extract and Carbon Powder, 1976 Chloride Injection, 10z, 1570 Risperidone Oral, 1523
Extract and Ethyl Aminobenzoate Chromate (51Cr) Injection, 1571 Saponated Cresol, 757
Powder, 1976 Citrate Hydrate, 1571 Silver Protein, 1558
Extract and Tannic Acid Supposito- Citrate Injection for Transfusion, Sodium Iodide (131I), 1580
ries, 1978 1572 Sodium L-Lactate, 1581
Extract, Papaverine and Ethyl Cromoglicate, 1573 Sodium L-Lactate Ringer's, 1582
Aminobenzoate Powder, 1977, Fusidate, 1574 D-Sorbitol, 1597
2790 Hyaluronate, Purified, 1575 Terbinafine Hydrochloride, 1664
Extract Powder, 1975 Hyaluronate Injection, Purified, Tolnaftate, 1705
Rhizome, 1973 1576 Sophora Root, 1995
Scutellaria Root, 1978 Hyaluronate Ophthalmic Solution, Powdered, 1995
Powdered, 1979 Purified, 1577 Sorbitan Sesquioleate, 1595
Senega, 1980 Hydrogen Carbonate, 1566 D-Sorbitol, 1596
Powdered, 1981 Hydrogen Sulfite, 1567 Solution, 1597
Syrup, 1981 Hydroxide, 1578 Soybean Oil, 1995
Senna Leaf, 1981 Iodide, 1579 Spectinomycin Hydrochloride
Powdered, 1982 Iodide (123I) Capsules, 1579 for Injection, 1598
L-Serine, 1548 Iodide (131I) Capsules, 1580 Hydrate, 1597
Serrapeptase, 1549, 2734 Iodide (131I) Solution, 1580 Spiramycin Acetate, 1599, 2735
Sesame, 1983 Iodohippurate (131I) Injection, 1580 Spirit
Oil, 1984 Iotalamate Injection, 1580 Capsicum and Salicylic Acid, 1824
Sevoflurane, 1550 L-Lactate Ringer's Solution, 1582 Compound Methyl Salicylate, 1238
Shakuyakukanzoto Extract, 1984, L-Lactate Solution, 1581 Compound Salicylic Acid, 1541
2790 Lauryl Sulfate, 1584, 2734 Foeniculated Ammonia, 1852
Shellac Metabisulfite, 1587 Iodine, Salicylic Acid and Phenol,
Purified, 1551 Pertechnetate (99mTc) Injection, 1079
White, 1552 1585 Salicylic Acid, 1540
Shimbuto Extract, 1985, 2791 Picosulfate Hydrate, 1585 Spironolactone, 1600
Shosaikoto Extract, 1988, 2791 Polystyrene Sulfonate, 1586 Tablets, 1600
Shoseiryuto Extract, 1990, 2793 Pyrosulfite, 1587 Spray
Silodosin, 1553 Risedronate Hydrate, 1588 Butenafine Hydrochloride, 538
Tablets, 1555 Risedronate Tablets, 1589 Terbinafine Hydrochloride, 1665
Silver Salicylate, 1590 Starch
Nitrate, 1557 Starch Glycolate, 1591 Corn, 1601
Nitrate Ophthalmic Solution, 1557 Sulfate Hydrate, 1994 Potato, 1602
Protein, 1557 Thiosulfate Hydrate, 1592 Rice, 1603
Protein Solution, 1558 Thiosulfate Injection, 1593 Wheat, 1604
Simple Valproate, 1593 Stearic Acid, 1606
Ointment, 1993 Valproate Syrup, 1594 Stearyl Alcohol, 1607
Syrup, 1558 Valproate Tablets, 1594 Sterile
Simvastatin, 1559 Solution Purified Water in Containers, 1774
Tablets, 1560 Adrenaline, 380 Water for Injection in Containers,
Sinomenium Stem and Rhizome, 1993 Alum, 401 1775
Sivelestat Sodium Benzalkonium Chloride, 480 Streptomycin Sulfate, 1608
for Injection, 1562 Benzethonium Chloride, 482 for Injection, 1609
Hydrate, 1561 Butenafine Hydrochloride, 538 Sucralfate Hydrate, 1609
Slaked Lime, 553 Chlorhexidine Gluconate, 682 Sucrose, 1611
Smilax Rhizome, 1993 Compound Thianthol and Salicylic Sulbactam Sodium, 1612, 2735
Powdered, 1993 Acid, 1679 Sulbenicillin Sodium, 1613
Sodium Cresol, 756 Sulfadiazine Silver, 1614
Acetate Hydrate, 1563 Dental Sodium Hypochlorite, 436 Sulfafurazole, 1617
Aurothiomalate, 1564 Diagnostic Sodium Citrate, 1572 Sulfamethizole, 1615
2856 Index Supplement I, JP XVII

Sulfamethoxazole, 1616 Cilazapril, 703


Sulfamonomethoxine Hydrate, 1616 T Cilnidipine, 705
Sulfasalazine, 1537 Cilostazol, 708
Sulfisomezole, 1616 Tablets Clarithromycin, 719
Sulfisoxazole, 1617 Acemetacin, 362 Clomifene Citrate, 731
Sulfobromophthalein Sodium, 1618 Acetylsalicylic Acid, 450 Clomipramine Hydrochloride, 2681
Injection, 1619 Aciclovir, 374 Clonazepam, 733
Sulfur, 1619 Ajmaline, 382 Clopidogrel Sulfate, 738
and Camphor Lotion, 1619 Alacepril, 383 Clotiazepam, 2682
, Salicylic Acid and Thianthol Oint- Aldioxa, 387 Codeine Phosphate, 748
ment, 1620 Alendronate Sodium, 390 Colestimide, 752
Sulindac, 1620 Allopurinol, 392 Cyclophosphamide, 762
Sulpiride, 1621 Alminoprofen, 394 Diazepam, 784
Capsules, 1621 Amiodarone Hydrochloride, 412 Dichlorphenamide, 789
Tablets, 1622 Amitriptyline Hydrochloride, 414 Diclofenamide, 789, 2784
Sulpyrine Amlexanox, 416 Diethylcarbamazine Citrate, 790
Hydrate, 1623 Amlodipine Besilate, 420 Digitoxin, 795, 2684
Injection, 1623 Amlodipine Besilate Orally Disin- Digoxin, 798
Sultamicillin Tosilate tegrating, 418 Dimenhydrinate, 809
Hydrate, 1624, 2736 Amosulalol Hydrochloride, 422 Diphenylhydantoin, 1397
Tablets, 1625 Amphotericin B, 428 Distigmine Bromide, 819
Sultiame, 1626 Ascorbic Acid and Calcium Pan- Donepezil Hydrochloride, 827
Suppositories tothenate, 447 Doxazosin Mesilate, 834
Bisacodyl, 512 Aspirin, 450 Doxycycline Hydrochloride, 840
Indometacin, 1064 Atorvastatin Calcium, 454 Dydrogesterone, 845
Scopolia Extract and Tannic Acid, Auranofin, 458 Ebastine, 847
1978 Azathioprine, 460 Ebastine Orally Disintegrating, 846
Suxamethonium Chloride Azelnidipine, 464 Emorfazone, 859
for Injection, 1628 Azosemide, 2672 Enalapril Maleate, 861
Hydrate, 1627 Baclofen, 470 Entacapone, 2691
Injection, 1628, 2737 Benidipine Hydrochloride, 476 Epalrestat, 866
Sweet Hydrangea Leaf, 1996 Bepotastine Besilate, 489 Ephedrine Hydrochloride, 870
Powdered, 1996, 2796 Beraprost Sodium, 492 Eplerenone, 875
Swertia Betahistine Mesilate, 496 Ergometrine Maleate, 884
and Sodium Bicarbonate Powder, Betamethasone, 498 Erythromycin Delayed-release, 886
1998 Bezafibrate Extended-release, 508 Estriol, 892
Herb, 1996 Bisoprolol Fumarate, 515 Etacrynic Acid, 893
Herb, Powdered, 1997 Brotizolam, 524 Ethinylestradiol, 899
Synthetic Aluminum Silicate, 405 Bucillamine, 526 Etidronate Disodium, 908
Syrup Buformin Hydrochloride, 531 Etilefrine Hydrochloride, 909
Aciclovir, 372 Buformin Hydrochloride Delayed- Etizolam, 912
Aciclovir for, 373 release, 529 Famotidine, 919
Amphotericin B, 428 Cadralazine, 542 Faropenem Sodium, 921
Cefadroxil for, 599 Candesartan Cilexetil, 566 Fexofenadine Hydrochloride, 927
Cefalexin for, 603 Candesartan Cilexetil and Amlodi- Flecainide Acetate, 935
Cefatrizine Propylene Glycolate for, pine Besylate, 567 Flutoprazepam, 955
606 Candesartan Cilexetil and Hydro- Fluvoxamine Maleate, 958
Cefpodoxime Proxetil for, 646 chlorothiazide, 570 Folic Acid, 960
Cefroxadine for, 649 L-Carbocisteine, 579 Fudosteine, 969
Faropenem Sodium for, 920 Carvedilol, 590 Furosemide, 972
Fosfomycin Calcium for, 963 Cefcapene Pivoxil Hydrochloride, Glimepiride, 986
Ipecac, 1882 614 Haloperidol, 1006
Monosodium Trichloroethyl Phos- Cefditoren Pivoxil, 620 Hydralazine Hydrochloride, 1022
phate, 1726 Cefpodoxime Proxetil, 647 Ifenprodil Tartrate, 1051
Orange Peel, 1926 Cefteram Pivoxil, 656 Imidapril Hydrochloride, 1053
Pemirolast Potassium for, 1374 Cetirizine Hydrochloride, 672 Imipramine Hydrochloride, 1057
Senega, 1981 Chlordiazepoxide, 681 Indapamide, 1059
Simple, 1558 Chlorphenesin Carbamate, 686 Ipriflavone, 1088
Sodium Valproate, 1594 Chlorpheniramine Maleate, 690 Irbesartan, 2710
Tranilast for, 1715 Chlorpromazine Hydrochloride, Irbesartan and Amlodipine Besilate,
Triclofos Sodium, 1726 693 2711
Chlorpropamide, 695 Irsogladine Maleate, 1091
Cibenzoline Succinate, 698 Isoniazid, 1101
Supplement I, JP XVII Index 2857

Isosorbide Dinitrate, 1105 Pioglitazone Hydrochloride and Met- Voriconazole, 1770


Isosorbide Mononitrate, 1107 formin Hydrochloride, 1411 Warfarin Potassium, 1772
Isoxsuprine Hydrochloride, 1109 Piperazine Phosphate, 1419 Zaltoprofen, 1780
Josamycin, 1113 Pitavastatin Calcium, 1425 Zolpidem Tartrate, 1789
Labetalol Hydrochloride, 1130 Pivmecillinam Hydrochloride, 1428 Zonisamide, 2745
Lafutidine, 1136 Pravastatin Sodium, 1448 Tacalcitol
Lanatoside C, 1138, 2715 Prazepam, 1450 Hydrate, 1629
Lansoprazole Delayed-release Orally Precipitated Calcium Carbonate, Lotion, 1630
Disintegrating, 1141 549 Ointment, 1631
Levofloxacin, 1157 Prednisolone, 1453 Tacrolimus
Levothyroxine Sodium, 1160 Probenecid, 1459 Capsules, 1633
Liothyronine Sodium, 1166 Probucol, 1462 Hydrate, 1632
Lisinopril, 1168 Procainamide Hydrochloride, 1463 Talampicillin Hydrochloride, 1633
Losartan Potassium, 1173 Prochlorperazine Maleate, 1468 Talc, 1634
Losartan Potassium and Hydrochlo- Propafenone Hydrochloride, 1474 Taltirelin
rothiazide, 1174 Propiverine Hydrochloride, 1477 Hydrate, 1636
Loxoprofen Sodium, 1178 Propranolol Hydrochloride, 1479 Orally Disintegrating Tablets, 1637
Manidipine Hydrochloride, 1193 Propylthiouracil, 1482 Tablets, 1638
Mecobalamin, 1198 Quetiapine Fumarate, 1494 Tamoxifen Citrate, 1640
Mefruside, 1205 Quinapril Hydrochloride, 1497 Tamsulosin Hydrochloride, 1640
Mequitazine, 1215 Rebamipide, 1506 Extended-release Tablets, 1642
Mesalazine Extended-release, 2720 Reserpine, 1509 Tannic Acid, 1643
Metformin Hydrochloride, 1223 Risperidone, 1524 Tartaric Acid, 1643
Methotrexate, 2722 Ritodrine Hydrochloride, 1526 Taurine, 1643
Methyldopa, 1230 Rokitamycin, 1529, 2733 Tazobactam, 1644
Methylergometrine Maleate, 1233 Roxatidine Acetate Hydrochloride and Piperacillin for Injection, 1645
Methyltestosterone, 1240 Extended-release, 1532 Teceleukin
Metoclopramide, 1243 Roxithromycin, 2733 for Injection (Genetical Recombina-
Metoprolol Tartrate, 1245 Sarpogrelate Hydrochloride, 1545 tion), 1652
Metronidazole, 1246 Silodosin, 1555 (Genetical Recombination), 1647
Minocycline Hydrochloride, 1256 Simvastatin, 1560 Tegafur, 1653
Mitiglinide Calcium, 1259 Sodium Risedronate, 1589 Teicoplanin, 1654, 2737
Mizoribine, 1263 Sodium Valproate, 1594 Telmisartan, 1656
Montelukast Sodium, 1269 Spironolactone, 1600 Tablets, 1657
Montelukast Sodium Chewable, Sulpiride, 1622 Temocapril Hydrochloride, 1658
1267 Sultamicillin Tosilate, 1625 Tablets, 1659
Morphine Hydrochloride, 1272 Taltirelin, 1638 Teprenone, 1660
Mosapride Citrate, 1277 Taltirelin Orally Disintegrating, Capsules, 1661
Nabumetone, 1282 1637 Terbinafine Hydrochloride, 1662
Naftopidil, 1286 Tamsulosin Hydrochloride Exten- Cream, 1663
Naftopidil Orally Disintegrating, ded-release, 1642 Solution, 1664
1285 Telmisartan, 1657 Spray, 1665
Nateglinide, 1295 Temocapril Hydrochloride, 1659 Tablets, 1665
Nicergoline, 1302 Terbinafine Hydrochloride, 1665 Terbutaline Sulfate, 1666
Nicomol, 1304 Thiamazole, 1673 Testosterone
Nilvadipine, 1313 Tiapride Hydrochloride, 1685 Enanthate, 1667
Nitrendipine, 1315 Tiaramide Hydrochloride, 1686 Enanthate Injection, 1668
Nitroglycerin, 1317 Ticlopidine Hydrochloride, 1688 Propionate, 1668
Norgestrel and Ethinylestradiol, Tipepidine Hibenzate, 1692 Propionate Injection, 1669
1324 Tolbutamide, 1703 Tetracaine Hydrochloride, 1670
Olmesartan Medoxomil, 1330 Tosufloxacin Tosilate, 1707 Tetracycline Hydrochloride, 1670,
Olopatadine Hydrochloride, 1333 Tranexamic Acid, 1711 2738
Omeprazole Delayed-release, 1335 Trichlormethiazide, 1722 Thallium (201Tl) Chloride Injection,
Paroxetine Hydrochloride, 1371 Trihexyphenidyl Hydrochloride, 1672
Pemirolast Potassium, 1375 1728 Theophylline, 1672
Pentobarbital Calcium, 2729 Trimetazidine Hydrochloride, 1731 Thiamazole, 1672
Perphenazine, 1382 Troxipide, 1736 Tablets, 1673
Perphenazine Maleate, 1383 Ursodeoxycholic Acid, 1750 Thiamine Chloride Hydrochloride,
Phenytoin, 1397 Valaciclovir Hydrochloride, 1753 1674
Pilocarpine Hydrochloride, 1400 Valsartan, 1756 Injection, 1675
Pioglitazone Hydrochloride, 1407 Verapamil Hydrochloride, 1761, Powder, 1675
Pioglitazone Hydrochloride and 2741 Thiamine Nitrate, 1676
Glimepiride, 1408 Voglibose, 1767 Thiamylal Sodium, 1677
2858 Index Supplement I, JP XVII

for Injection, 1678 Tranilast, 1711 Cholera, 696


Thianthol, 1678 Capsules, 1712 Diphtheria-Purified Pertussis-Teta-
and Salicylic Acid Solution, Com- Fine Granules, 1713 nus Combined, Adsorbed, 816
pound, 1679 for Syrup, 1715 Hepatitis B, Adsorbed, 1018
Thiopental Sodium, 1680 Ophthalmic Solution, 1714 Influenza HA, 1065
for Injection, 1681 Trapidil, 1716 Inactivated Tissue Culture Rabies,
Thioridazine Hydrochloride, 1681 Trehalose Hydrate, 1717 Freeze-dried, 1503
L-Threonine, 1682 Trepibutone, 1718 Japanese Encephalitis, 1111
Thrombin, 1682, 2738 Tretoquinol Hydrochloride, 1733 Japanese Encephalitis, Freeze-dried,
Thymol, 1683 Triamcinolone, 1718 1111
Tiapride Hydrochloride, 1684 Acetonide, 1719 Live Attenuated Measles, Freeze-d-
Tablets, 1685 Triamterene, 1720 ried, 1196
Tiaramide Hydrochloride, 1685 Tribulus Fruit, 2005 Live Attenuated Mumps, Freeze-d-
Tablets, 1686 Trichlormethiazide, 1721 ried, 1278
Ticlopidine Hydrochloride, 1687 Tablets, 1722 Live Attenuated Rubella, Freeze-d-
Tablets, 1688 Trichomycin, 1724 ried, 1535
Timepidium Bromide Hydrate, 1689 Trichosanthes Root, 2005, 2798 Live Oral Poliomyelitis, 1428
Timolol Maleate, 1690 Triclofos Sodium, 1725 Purified Pertussis, Adsorbed, 1384
Tincture Syrup, 1726 Smallpox, Freeze-dried, 1563
Bitter, 1813 Trientine Hydrochloride, 1726 Smallpox, Freeze-dried, Prepared in
Capsicum, 1824 Capsules, 1727 Cell Culture, 1563
Iodine, 1076 Trihexyphenidyl Hydrochloride, 1727 Weil's Disease and Akiyami Com-
Iodine, Dilute, 1076 Tablets, 1728 bined, 1775
Nux Vomica, 1922 Trimebutine Maleate, 1729 Valaciclovir Hydrochloride, 1751
Opium, 1925 Trimetazidine Hydrochloride, 1730 Tablets, 1753
Orange Peel, 1926 Tablets, 1731 L-Valine, 1754
Tinidazole, 1691 Trimethadione, 1732 Valsartan, 1755
Tipepidine Hibenzate, 1691 Trimetoquinol Hydrochloride Hydrate, Tablets, 1756
Tablets, 1692 1733 Vancomycin Hydrochloride, 1757
Titanium Oxide, 1693 Tropicamide, 1734 for Injection, 1758
Tizanidine Hydrochloride, 1694 Troxipide, 1735 Vasopressin Injection, 1759, 2740
Toad Cake, 1998 Fine Granules, 1735 Verapamil Hydrochloride, 1761, 2741
Tobramycin, 1695, 2738 Tablets, 1736 Tablets, 1761, 2741
Injection, 1696 L-Tryptophan, 1737 Vinblastine Sulfate, 1762, 2742
Tocopherol, 1697 Tulobuterol, 1738 for Injection, 1763
Acetate, 1697 Hydrochloride, 1740 Vincristine Sulfate, 1764, 2742
Calcium Succinate, 1698 Transdermal Tape, 1739 Vitamin A
Nicotinate, 1699 Turmeric, 2005, 2798 Acetate, 1510
dl-a-Tocopherol, 1697 Powdered, 2006, 2798 Oil, 1765
Acetate, 1697 Turpentine Oil, 2007 Palmitate, 1510
Nicotinate, 1699 L-Tyrosine, 1740 Vitamin B1
Todralazine Hydrochloride Hydrate, Hydrochloride, 1674
1700 U Hydrochloride Injection, 1675
Tofisopam, 1701 Hydrochloride Powder, 1675
Tokakujokito Extract, 1999, 2796 Ubenimex, 1741 Nitrate, 1676
Tokishakuyakusan Extract, 2002, Capsules, 1742 Vitamin B2, 1513
2797 Ubidecarenone, 1743 Phosphate Ester, 1515
Tolazamide, 1702, 2738 Ulinastatin, 1744 Phosphate Ester Injection, 1516
Tolbutamide, 1703 Uncaria Hook, 2007 Powder, 1514
Tablets, 1703 Urapidil, 1746 Vitamin B6, 1489
Tolnaftate, 1704 Urea, 1747 Injection, 1490
Solution, 1705 Urokinase, 1747 Vitamin B12, 760
Tolperisone Hydrochloride, 1705 Ursodeoxycholic Acid, 1748 Injection, 761
Tosufloxacin Tosilate Granules, 1749 Vitamin C, 445
Hydrate, 1706 Tablets, 1750 Injection, 446
Tablets, 1707 Ursodesoxycholic Acid, 1748 Powder, 446
Tragacanth, 2004 Uva Ursi Fluidextract, 2008 Vitamin D2, 882
Powdered, 2004 Vitamin D3, 696
Tramadol Hydrochloride, 2739 V Vitamin E, 1697
Tranexamic Acid, 1708 Acetate, 1697
Capsules, 1709 Vaccine Calcium Succinate, 1698
Injection, 1710 BCG, Freeze-dried, (for Percutane- Nicotinate, 1699
Tablets, 1711 ous Use), 473 Vitamin K1, 1398
Supplement I, JP XVII Index 2859

Voglibose, 1766 Wheat Starch, 1604


Tablets, 1767 White Z
Voriconazole, 1769 Beeswax, 1808
for Injection, 2743 Ointment, 1775 Zaltoprofen, 1780
Tablets, 1770 Petrolatum, 1386 Tablets, 1780
Shellac, 1552 Zedoary, 2012, 2753
W Soft Sugar, 1611 Zidovudine, 1781
Whole Human Blood, 1775 Zinc
Warfarin Potassium, 1771 Wine, 1776 Chloride, 1783
Tablets, 1772 Wood Creosote, 2008 Oxide, 1783
Water, 1773 Oxide Oil, 1784
for Injection, 1774 X Oxide Ointment, 1784
for Injection in Containers, Sterile, Oxide Starch Powder, 1785
1775 Xylitol, 1778 Sulfate Hydrate, 1785
in Containers, Purified, 1774 Injection, 1778 Sulfate Ophthalmic Solution, 1785
in Containers, Sterile Purified, Zinostatin Stimalamer, 1786, 2744
1774 Y Zolpidem Tartrate, 1788
Purified, 1773 Tablets, 1789
Weak Opium Alkaloids and Scopola- Yellow Zonisamide, 2744
mine Injection, 1340 Beeswax, 1809 Tablets, 2745
Weil's Disease and Akiyami Combined Petrolatum, 1387
Vaccine, 1775 Yokukansan Extract, 2010, 2798
INDEX IN LATIN NAME
Fructus Pulveratus, 1823
A Cardamomi Fructus, 1825 F
Carthami Flos, 1962
Achyranthis Radix, 1792 Caryophylli Flos, 1833 Fel Ursi, 1807
Aconiti Radix Processa, 1948 Flos Pulveratus, 1833 Foeniculi Fructus, 1851
Radix Processa et Pulverata, 1949 Cassiae Semen, 1825 Fructus Pulveratus, 1852
Adeps Lanae Purificatus, 1904 Catalpae Fructus, 1826 Forsythiae Fructus, 1852
Suillus, 1905 Cera Alba, 1808 Fossilia Ossis Mastodi, 1907
Agar, 1792 Carnauba, 1825 Ossis Mastodi Pulveratum, 1908
Pulveratum, 1793 Flava, 1809 Fritillariae Bulbus, 1853
Akebiae Caulis, 1793 Chrysanthemi Flos, 1829 Fructus Hordei Germinatus, 1912
Alismatis Tuber, 1793 Cimicifugae Rhizoma, 1830
Tuber Pulveratum, 1794 Cinnamomi Cortex, 1830 G
Aloe, 1794 Cortex Pulveratus, 1831
Pulverata, 1795 Cistanchis Herba, 1831 Gambir, 1853
Alpiniae Fructus, 1812 Citri Unshiu Pericarpium, 1832 Pulveratum, 1853
Officinari Rhizoma, 1796 Clematidis Radix, 1833 Gardeniae Fructus, 1854
Amomi Semen, 1797 Cnidii Monnieris Fructus, 1834 Fructus Pulveratus, 1854
Semen Pulveratum, 1797 Rhizoma, 1835 Gastrodiae Tuber, 1855
Anemarrhenae Rhizoma, 1797 Rhizoma Pulveratum, 1835 Gentianae Radix, 1856
Angelicae Acutilobae Radix, 1883 Codonopsis Radix, 1835 Radix Pulverata, 1856
Acutilobae Radix Pulverata, 1883 Coicis Semen, 1836 Scabrae Radix, 1884
Dahuricae Radix, 1798 Semen Pulveratum, 1836 Scabrae Radix Pulverata, 1884
Apilac, 1960 Condurango Cortex, 1836 Geranii Herba, 1857
Araliae Cordatae Rhizoma, 1800 Coptidis Rhizoma, 1837 Herba Pulverata, 1857
Arctii Fructus, 1821 Rhizoma Pulveratum, 1838 Ginseng Radix, 1859
Arecae Semen, 1800 Corni Fructus, 1839 Radix Pulverata, 1860
Armeniacae Semen, 1798 Corydalis Tuber, 1840 Radix Rubra, 1953
Artemisiae Capillaris Flos, 1800 Tuber Pulveratum, 1841 Glehniae Radix cum Rhizoma, 1861
Folium, 1801 Crataegi Fructus, 1842 Glycyrrhizae Radix, 1862
Asiasari Radix, 1801 Creosotum Ligni, 2008 Radix Praeparata, 1947
Asparagi Radix, 1802 Crocus, 1963 Radix Pulverata, 1863
Astragali Radix, 1803 Curcumae Longae Rhizoma, 2798 Gummi Arabicum, 1791
Atractylodis Lanceae Rhizoma, 1803 Rhizoma Purveratum, 2798 Arabicum Pulveratum, 1791
Lanceae Rhizoma Pulveratum, Curcumae Rhizoma, 2005, 2753 Gypsum Exsiccatum, 1868
1804 Rhizoma Purveratum, 2006 Fibrosum, 1868
Rhizoma, 1804 Cyperi Rhizoma, 1842
Rhizoma Pulveratum, 1805 Rhizoma Pulveratum, 1843 H
Aurantii Fructus Immaturus, 1880
Pericarpium, 1812 D Hedysari Radix, 1875
Houttuyniae Herba, 1880
B Digenea, 1846 Hydrangeae Dulcis Folium, 1996
Dioscoreae Rhizoma, 1847 Dulcis Folium Pulveratum, 1996
Belladonnae Radix, 1809 Rhizoma Pulveratum, 1847
Benincasae Semen, 1811 Dolichi Semen, 1848 I
Benzoinum, 1812
Bezoar Bovis, 1928 E Imperatae Rhizoma, 1880
Bufonis Crustum, 1998 Ipecacuanhae Radix, 1881
Bupleuri Radix, 1820 Eleutherococci senticosi Rhizoma, Radix Pulverata, 1881
1848
C Ephedrae Herba, 1849 K
Epimedii Herba, 1850
Calumbae Radix, 1821 Eriobotryae Folium, 1909 Kasseki, 1796
Radix Pulverata, 1822 Eucommiae Cortex, 1851 Koi, 1903
Cannabis Fructus, 1876 Euodiae Fructus, 1851
Capsici Fructus, 1822

2861
2862 Index in Latin name Supplement I, JP XVII

Saussureae Radix, 1972


L P Schisandrae Fructus, 1973
Schizonepetae Spica, 1973
Leonuri Herba, 1905 Paeoniae Radix, 1935 Scopoliae Rhizoma, 1973
Lilii Bulbus, 1906 Radix Pulverata, 1936 Scutellariae Radix, 1978
Linderae Radix, 1906 Panacis Japonici Rhizoma, 1932 Radix Pulverata, 1979
Lithospermi Radix, 1907 Japonici Rhizoma Pulveratum, Senegae Radix, 1980
Longan Arillus, 1907 1933 Radix Pulverata, 1981
Lonicerae Folium Cum Caulis, 1908 Perillae Herba, 1937 Sennae Folium, 1981
Lycii Cortex, 1909 Persicae Semen, 1933 Folium Pulveratum, 1982
Fructus, 1909 Semen Pulveratum, 1934 Sesami Semen, 1983
Peucedani Radix, 1937 Sevum Bovinum, 1808
M Pharbitidis Semen, 1938 Sinomeni Caulis et Rhizoma, 1993
Phellodendri Cortex, 1938 Smilacis Rhizoma, 1993
Magnoliae Cortex, 1910 Cortex Pulveratus, 1939 Rhizoma Pulveratum, 1993
Cortex Pulveratus, 1910 Picrasmae Lignum, 1941 Sophorae Radix, 1995
Flos, 1911 Lignum Pulveratum, 1941 Radix Pulverata, 1995
Malloti Cortex, 1911 Pinelliae Tuber, 1941 Strychni Semen, 1920
Mel, 1879 Plantaginis Herba, 1942 Swertiae Herba, 1996
Menthae Herba, 1914 Semen, 1942 Herba Pulverata, 1997
Mori Cortex, 1918 Platycodi Radix, 1942
Moutan Cortex, 1915 Radix Pulverata, 1943 T
Cortex Pulveratus, 1916 Pogostemoni Herba, 1944
Myristicae Semen, 1920 Polygalae Radix, 1944 Tinctura Amara, 1813
Radix Pulverata, 1944 Tragacantha, 2004
N Polygonati Rhizoma, 1945 Pulverata, 2004
Polygoni Multiflori Radix, 1945 Tribuli Fructus, 2005
Nelumbis Semen, 1919 Polyporus, 1946 Trichosanthis Radix, 2005
Notopterygii Rhizoma, 1919 Pulveratus, 1946
Nupharis Rhizoma, 1919 Poria, 1946 U
Pulveratum, 1947
O Prunellae Spica, 1951 Uncariae Uncis Cum Ramulus, 2007
Pruni Cortex, 1826 Uvae Ursi Folium, 1807
Oleum Arachidis, 1935 Puerariae Radix, 1952
Aurantii, 1925 V
Cacao, 1821 Q
Camelliae, 1822 Valerianae Fauriei Radix, 1884
Caryophylli, 1834 Quercus Cortex, 1952 Fauriei Radix Pulverata, 1885
Cinnamomi, 1831
Cocois, 1835 R Z
Eucalypti, 1850
Foeniculi, 1852 Rehmanniae Radix, 1954 Zanthoxyli Piperiti Pericarpium, 1885
Maydis, 1839 Resina Pini, 1960 Piperiti Pericarpium Pulveratum,
Menthae Japonicae, 1915 Rhei Rhizoma, 1955 1886
Olivae, 1923 Rhizoma Pulveratum, 1956 Zedoariae Rhizoma, 2012
Rapae, 1953 Rosae Fructus, 1959 Zingiberis Rhizoma, 1857
Ricini, 1826 Fructus Pulveratus, 1960 Rhizoma Processum, 1951
Sesami, 1984 Rhizoma Pulveratum, 1858
Sojae, 1995 S Ziziphi Fructus, 2765
Terebinthinae, 2007 Semen, 2765
Ophiopogonis Radix, 1923 Sal Mirabilis, 1994 Zizyphi Fructus, 1886
Opium Pulveratum, 1924 Mirabilis Anhydricus, 1994 Semen, 1886
Oryzae Fructus, 1820 Salviae Miltiorrhizae Radix, 1971
Ostreae Testa, 1932 Saposhnikoviae Radix, 1971
Testa Pulverata, 1932 Sappan Lignum, 1972
INDEX IN JAPANESE
アトルバスタチンカルシウム水和物 亜硫酸水素ナトリウム 1567
ア 453 アルガトロバン水和物 441
アドレナリン 379 L-アルギニン 442
亜鉛華デンプン 1785 アドレナリン液 380 L-アルギニン塩酸塩 443
亜鉛華軟膏 1784 アドレナリン注射液 379 L-アルギニン塩酸塩注射液 443
アカメガシワ 1911 アトロピン硫酸塩水和物 456 アルジオキサ 386
アクチノマイシン D 378 アトロピン硫酸塩注射液 456 アルジオキサ顆粒 387
アクラルビシン塩酸塩 375 亜ヒ酸パスタ 445 アルジオキサ錠 387
アクリノール・亜鉛華軟膏 378 アプリンジン塩酸塩 437 アルプラゾラム 395
アクリノール水和物 376 アプリンジン塩酸塩カプセル 438 アルプレノロール塩酸塩 396
アクリノール・チンク油 377 アフロクアロン 380 アルプロスタジル 396
アザチオプリン 459 アヘン・トコン散 1925 アルプロスタジル アルファデクス
アザチオプリン錠 460 アヘンアルカロイド・アトロピン注射 400
亜酸化窒素 1318 液 1338 アルプロスタジル注射液 398
アシクロビル 369 アヘンアルカロイド・スコポラミン注 アルベカシン硫酸塩 439
アシクロビル顆粒 370 射液 1339 アルベカシン硫酸塩注射液 440
アシクロビル眼軟膏 372 アヘンアルカロイド塩酸塩 1336 アルミノプロフェン 393
アシクロビル錠 374 アヘンアルカロイド塩酸塩注射液 アルミノプロフェン錠 394
アシクロビルシロップ 372 1337 アレンドロン酸ナトリウム錠 390
アシクロビル注射液 371 アヘン散 1924 アレンドロン酸ナトリウム水和物
アシクロビル軟膏 372 アヘンチンキ 1925 388
アジスロマイシン水和物 465 アヘン末 1924 アレンドロン酸ナトリウム注射液
アジマリン 381 アマチャ 1996 389
アジマリン錠 382 アマチャ末 1996, 2796 アロエ 1794
亜硝酸アミル 436 アマンタジン塩酸塩 405 アロエ末 1795
アスコルビン酸 445 アミオダロン塩酸塩 411 アロチノロール塩酸塩 444
アスコルビン酸・パントテン酸カルシ アミオダロン塩酸塩錠 412 アロプリノール 392
ウム錠 447 アミカシン硫酸塩 408 アロプリノール錠 392
アスコルビン酸散 446 アミカシン硫酸塩注射液 409 安息香酸 482
アスコルビン酸注射液 446 アミドトリゾ酸 407 安息香酸ナトリウム 1565
アズトレオナム 466 アミドトリゾ酸ナトリウムメグルミン 安息香酸ナトリウムカフェイン 544
L-アスパラギン酸 449 注射液 1207 安息香酸ベンジル 484
アスピリン 449 アミトリプチリン塩酸塩 414 アンソッコウ 1812
アスピリンアルミニウム 450 アミトリプチリン塩酸塩錠 414 アンチピリン 437
アスピリン錠 450 アミノ安息香酸エチル 901 アンピシリン水和物 430, 2671
アスポキシシリン水和物 451 アミノフィリン水和物 410 アンピシリンナトリウム 431
アセグルタミドアルミニウム 359, アミノフィリン注射液 410 アンピロキシカム 434
2671 アムホテリシン B 426 アンピロキシカムカプセル 435
アセタゾラミド 364 アムホテリシン B 錠 428 アンベノニウム塩化物 406
アセチルシステイン 368 アムホテリシン B シロップ 428 アンモニア・ウイキョウ精 1852
アセトアミノフェン 363 アムロジピンベシル酸塩 417 アンモニア水 420
アセトヘキサミド 366 アムロジピンベシル酸塩口腔内崩壊錠 アンレキサノクス 415
アセブトロール塩酸塩 359 418 アンレキサノクス錠 416
アセメタシン 361 アムロジピンベシル酸塩錠 420
アセメタシンカプセル 361 アモキサピン 424 イ
アセメタシン錠 362 アモキシシリンカプセル 425
アゼラスチン塩酸塩 461 アモキシシリン水和物 424, 2671 イオウ 1619
アゼラスチン塩酸塩顆粒 462 アモスラロール塩酸塩 421 イオウ・カンフルローション 1619
アゼルニジピン 463 アモスラロール塩酸塩錠 422 イオウ・サリチル酸・チアントール軟膏
アゼルニジピン錠 464 アモバルビタール 421 1620
アセンヤク 1853 アラセプリル 382 イオタラム酸 1085
アセンヤク末 1853 アラセプリル錠 383 イオタラム酸ナトリウム注射液 1580
アゾセミド 2672 L-アラニン 384 イオタラム酸メグルミン注射液 1206
アゾセミド錠 2672 アラビアゴム 1791 イオトロクス酸 1086
アテノロール 452 アラビアゴム末 1791 イオパミドール 1083
アトルバスタチンカルシウム錠 454 アリメマジン酒石酸塩 391 イオパミドール注射液 1084

2863
2864 Index in Japanese Supplement I, JP XVII

イオヘキソール 1080 え)注射液 1067 エチゾラム錠 912


イオヘキソール注射液 1082, 2710 インスリン ヒト(遺伝子組換え) エチドロン酸二ナトリウム 907
イクタモール 1046 1068 エチドロン酸二ナトリウム錠 908
イコサペント酸エチル 903 インスリン ヒト(遺伝子組換え)注射 エチニルエストラジオール 899
イコサペント酸エチルカプセル 904 液 1070 エチニルエストラジオール錠 899
イセパマイシン硫酸塩 1093 インダパミド 1058 L-エチルシステイン塩酸塩 902
イセパマイシン硫酸塩注射液 1094 インダパミド錠 1059 エチルモルヒネ塩酸塩水和物 905
イソクスプリン塩酸塩 1108 インターフェロン アルファ エチレフリン塩酸塩 909
イソクスプリン塩酸塩錠 1109 (NAMALWA) 1071 エチレフリン塩酸塩錠 909
イソソルビド 1103 インターフェロン アルファ エチレンジアミン 903
イソニアジド 1099 (NAMALWA)注射液 1074 エデト酸カルシウムナトリウム水和物
イソニアジド錠 1101 インチンコウ 1800, 2747 561
イソニアジド注射液 1100 インデノロール塩酸塩 1060 エデト酸ナトリウム水和物 1573
イソフェンインスリン ヒト(遺伝子 インドメタシン 1062 エーテル 898
組換え)水性懸濁注射液 2705 インドメタシンカプセル 1063 エテンザミド 897
イソフルラン 1094 インドメタシン坐剤 1064 エトスクシミド 901
l-イソプレナリン塩酸塩 1102 インフルエンザ HA ワクチン 1065 エトドラク 913
イソプロパノール 1102 インヨウカク 1850 エトポシド 914
イソプロピルアンチピリン 1103 エドロホニウム塩化物 853
イソマル水和物 1098 ウ エドロホニウム塩化物注射液 854
L-イソロイシン 1096 エナラプリルマレイン酸塩 860
イソロイシン・ロイシン・バリン顆粒 ウイキョウ 1851 エナラプリルマレイン酸塩錠 861
1096 ウイキョウ末 1852 エノキサシン水和物 863
イダルビシン塩酸塩 1046 ウイキョウ油 1852 エバスチン 845
一硝酸イソソルビド錠 1107 ウコン 2005, 2798 エバスチン口腔内崩壊錠 846
70z一硝酸イソソルビド乳糖末 ウコン末 2006, 2798 エバスチン錠 847
1105, 2714 ウベニメクス 1741 エパルレスタット 865, 2692
イドクスウリジン 1048 ウベニメクスカプセル 1742 エパルレスタット錠 866
イドクスウリジン点眼液 1049 ウヤク 1906 エピリゾール 871
イトラコナゾール 1110 ウラピジル 1746 エピルビシン塩酸塩 872
イフェンプロジル酒石酸塩 1050 ウリナスタチン 1744 エフェドリン塩酸塩 868
イフェンプロジル酒石酸塩細粒 1050 ウルソデオキシコール酸 1748 エフェドリン塩酸塩散10z 870
イフェンプロジル酒石酸塩錠 1051 ウルソデオキシコール酸顆粒 1749 エフェドリン塩酸塩錠 870
イブジラスト 1042 ウルソデオキシコール酸錠 1750 エフェドリン塩酸塩注射液 869
イブプロフェン 1043 ウロキナーゼ 1747 エプレレノン 874
イブプロフェンピコノール 1044 ウワウルシ 1807 エプレレノン錠 875
イブプロフェンピコノールクリーム ウワウルシ流エキス 2008 エペリゾン塩酸塩 867
1044 エポエチン アルファ(遺伝子組換え)
イブプロフェンピコノール軟膏 1045 エ 876
イプラトロピウム臭化物水和物 1086 エポエチン ベータ(遺伝子組換え)
イプリフラボン 1087 エイジツ 1959 879
イプリフラボン錠 1088 エイジツ末 1960 エメダスチンフマル酸塩 857
イミダプリル塩酸塩 1052 エカベトナトリウム顆粒 849 エメダスチンフマル酸塩徐放カプセル
イミダプリル塩酸塩錠 1053 エカベトナトリウム水和物 849 858
イミプラミン塩酸塩 1057 液状フェノール 1391 エモルファゾン 858
イミプラミン塩酸塩錠 1057 エコチオパートヨウ化物 851 エモルファゾン錠 859
イミペネム水和物 1054 エスタゾラム 889 エリスロマイシン 885, 2692
イルソグラジンマレイン酸塩 1090 エストラジオール安息香酸エステル エリスロマイシンエチルコハク酸エス
イルソグラジンマレイン酸塩細粒 889 テル 887
1090 エストラジオール安息香酸エステル水 エリスロマイシンステアリン酸塩
イルソグラジンマレイン酸塩錠 1091 性懸濁注射液 890 888
イルベサルタン 1089 エストリオール 891 エリスロマイシン腸溶錠 886
イルベサルタン・アムロジピンベシル エストリオール錠 892 エリスロマイシンラクトビオン酸塩
酸塩錠 2711 エストリオール水性懸濁注射液 892 887
イルベサルタン錠 2710 エタクリン酸 893 エルカトニン 854
イレイセン 1833 エタクリン酸錠 893 エルゴカルシフェロール 882
インジゴカルミン 1061 エタノール 895, 2693 エルゴタミン酒石酸塩 884
インジゴカルミン注射液 1062 エダラボン 851 エルゴメトリンマレイン酸塩 883
インスリン アスパルト(遺伝子組換 エダラボン注射液 852, 2687 エルゴメトリンマレイン酸塩錠 884
え) 2708 エタンブトール塩酸塩 894 エルゴメトリンマレイン酸塩注射液
インスリン グラルギン(遺伝子組換 エチオナミド 900 883
え) 1065 エチゾラム 910 塩化亜鉛 1783
インスリン グラルギン(遺伝子組換 エチゾラム細粒 911 塩化インジウム(111In)注射液 1062
Supplement I, JP XVII Index in Japanese 2865

塩化カリウム 1434 カンゾウ 1862


塩化カルシウム水和物 550 カ 乾燥亜硫酸ナトリウム 1592
塩化カルシウム注射液 551 カンゾウエキス 1863, 2755
塩化タリウム(201Tl)注射液 1672 カイニン酸・サントニン散 1115 乾燥甲状腺 1684
塩化ナトリウム 1569 カイニン酸水和物 1114 乾燥酵母 1779
10z塩化ナトリウム注射液 1570 ガイヨウ 1801 乾燥細胞培養痘そうワクチン 1563
エンゴサク 1840 カオリン 1120 乾燥ジフテリアウマ抗毒素 816
エンゴサク末 1841 カカオ脂 1821 乾燥弱毒生おたふくかぜワクチン
塩酸 1023 加香ヒマシ油 1826 1278
塩酸リモナーデ 1024 カゴソウ 1951 乾燥弱毒生風しんワクチン 1535
エンタカポン 2689 カシュウ 1945 乾燥弱毒生麻しんワクチン 1196
エンタカポン錠 2691 ガジュツ 2012, 2753 乾燥水酸化アルミニウムゲル 401
エンビオマイシン硫酸塩 864 加水ラノリン 1903 乾燥水酸化アルミニウムゲル細粒
エンフルラン 863 ガスえそウマ抗毒素 977 402
カッコウ 1944 カンゾウ粗エキス 1864, 2756
オ カッコン 1952 乾燥組織培養不活化狂犬病ワクチン
根湯エキス 1889, 2768 1503
オウギ 1803 根湯加川辛夷エキス 1892, 2771 乾燥炭酸ナトリウム 1568
オウゴン 1978 カッセキ 1796 乾燥痘そうワクチン 1563
オウゴン末 1979 過テクネチウム酸ナトリウム(99mTc) 乾燥日本脳炎ワクチン 1111
黄色ワセリン 1387 注射液 1585 乾燥破傷風ウマ抗毒素 1670
オウセイ 1945 果糖 967 乾燥はぶウマ抗毒素 1004
オウバク 1938 果糖注射液 968 乾燥 BCG ワクチン 473
オウバク・タンナルビン・ビスマス散 カドララジン 541 乾燥ボツリヌスウマ抗毒素 520
1940 カドララジン錠 542 カンゾウ末 1863
オウバク末 1939 カナマイシン一硫酸塩 1118 乾燥まむしウマ抗毒素 1191
オウヒ 1826 カナマイシン硫酸塩 1119 乾燥硫酸アルミニウムカリウム 403
オウレン 1837 カノコソウ 1884 カンデサルタン シレキセチル 565
黄連解毒湯エキス 1926, 2778 カノコソウ末 1885 カンデサルタン シレキセチル・アム
オウレン末 1838 カフェイン水和物 543 ロジピンベシル酸塩錠 567
オキサゾラム 1343 カプセル 574 カンデサルタン シレキセチル・ヒド
オキサピウムヨウ化物 1342 カプトプリル 575 ロクロロチアジド錠 570
オキサプロジン 1343 ガベキサートメシル酸塩 974 カンデサルタン シレキセチル錠
オキシコドン塩酸塩水和物 1346 過マンガン酸カリウム 1438 566
オキシテトラサイクリン塩酸塩 加味帰脾湯エキス 1895, 2772 カンテン 1792
1351, 2726 加味逍遙散エキス 1898, 2773 カンテン末 1793
オキシトシン 1353 カモスタットメシル酸塩 562 含糖ペプシン 1535
オキシトシン注射液 1355 b-ガラクトシダーゼ(アスペルギルス) d-カンフル 563
オキシドール 1349 975 dl-カンフル 564
オキシブプロカイン塩酸塩 1345 b-ガラクトシダーゼ(ペニシリウム) 肝油 745
オキシメトロン 1351 975 カンレノ酸カリウム 1433
オキセサゼイン 1344 カリジノゲナーゼ 1116
オクスプレノロール塩酸塩 1345 カリ石ケン 1432 キ
オザグレルナトリウム 1356 カルシトニン サケ 545
オザグレルナトリウム注射液 1357 カルテオロール塩酸塩 587 希塩酸 1024
乙字湯エキス 1929, 2779 カルナウバロウ 1825 キキョウ 1942, 2779
オフロキサシン 1328 カルバゾクロムスルホン酸ナトリウム キキョウ末 1943
オメプラゾール 1334 水和物 576 キキョウ流エキス 1943
オメプラゾール腸溶錠 1335 カルバマゼピン 575 キクカ 1829
オーラノフィン 457 カルビドパ水和物 577 キササゲ 1826
オーラノフィン錠 458 カルベジロール 589 キジツ 1880
オリブ油 1923 カルベジロール錠 590 キシリトール 1778
オルシプレナリン硫酸塩 1341 L-カルボシステイン 578 キシリトール注射液 1778
オルメサルタン メドキソミル 1329 L-カルボシステイン錠 579 キタサマイシン 1126
オルメサルタン メドキソミル錠 カルボプラチン 580 キタサマイシン酢酸エステル 1127
1330 カルボプラチン注射液 582 キタサマイシン酒石酸塩 1128
オレンジ油 1925 カルメロース 583 キナプリル塩酸塩 1496
オロパタジン塩酸塩 1332 カルメロースカルシウム 583 キナプリル塩酸塩錠 1497
オロパタジン塩酸塩錠 1333 カルメロースナトリウム 584 キニジン硫酸塩水和物 1498
オンジ 1944 カルモナムナトリウム 587 キニーネエチル炭酸エステル 1499
オンジ末 1944 カルモフール 586 キニーネ塩酸塩水和物 1500
カロコン 2005, 2798 キニーネ硫酸塩水和物 1501
カンキョウ 1951 牛脂 1808
2866 Index in Japanese Supplement I, JP XVII

吸水クリーム 755 クロナゼパム 732 ケイ酸マグネシウム 1187


キョウカツ 1919 クロナゼパム細粒 733 軽質無水ケイ酸 1552
キョウニン 1798 クロナゼパム錠 733 軽質流動パラフィン 1365
キョウニン水 1799 クロニジン塩酸塩 734 桂枝茯苓丸エキス 1901, 2775
希ヨードチンキ 1076 クロピドグレル硫酸塩 736 ケイヒ 1830
金チオリンゴ酸ナトリウム 1564 クロピドグレル硫酸塩錠 738 ケイヒ末 1831
クロフィブラート 728 ケイヒ油 1831
ク クロフィブラートカプセル 729 ケタミン塩酸塩 1120
クロフェダノール塩酸塩 728 結晶セルロース 664
グアイフェネシン 1002 クロベタゾールプロピオン酸エステル ケツメイシ 1825
グアナベンズ酢酸塩 1003 726 ケトコナゾール 1121
グアネチジン硫酸塩 1003 クロペラスチン塩酸塩 735 ケトコナゾール液 1123
グアヤコールスルホン酸カリウム クロミフェンクエン酸塩 730 ケトコナゾールクリーム 1122
1436 クロミフェンクエン酸塩錠 731 ケトコナゾールローション 1123
クエチアピンフマル酸塩 1492 クロミプラミン塩酸塩 731 ケトチフェンフマル酸塩 1125
クエチアピンフマル酸塩細粒 1493 クロミプラミン塩酸塩錠 2681 ケトプロフェン 1124
クエチアピンフマル酸塩錠 1494 クロム酸ナトリウム(51Cr)注射液 ケノデオキシコール酸 675
クエン酸ガリウム(67Ga)注射液 976 1571 ゲファルナート 977
クエン酸水和物 717 クロモグリク酸ナトリウム 1573 ケンゴシ 1938
クエン酸ナトリウム水和物 1571 クロラゼプ酸二カリウム 739 ゲンタマイシン硫酸塩 982, 2695
クコシ 1909 クロラゼプ酸二カリウムカプセル ゲンタマイシン硫酸塩点眼液 983
クジン 1995 740 ゲンチアナ 1856
クジン末 1995 クロラムフェニコール 677 ゲンチアナ・重曹散 1857
苦味重曹水 1994 クロラムフェニコールコハク酸エステ ゲンチアナ末 1856
苦味チンキ 1813 ルナトリウム 678, 2679 ゲンノショウコ 1857
クラブラン酸カリウム 1434 クロラムフェニコール・コリスチンメ ゲンノショウコ末 1857
グラミシジン 1001, 2698 タンスルホン酸ナトリウム点眼液
クラリスロマイシン 717, 2680 2679 コ
クラリスロマイシン錠 719 クロラムフェニコールパルミチン酸エ
グリクラジド 984 ステル 677 コウイ 1903
グリシン 995 クロルジアゼポキシド 679 コウカ 1962
グリセリン 992 クロルジアゼポキシド散 680 硬化油 1032
グリセリンカリ液 994 クロルジアゼポキシド錠 681 コウジン 1953
クリノフィブラート 725 クロルフェニラミンマレイン酸塩 合成ケイ酸アルミニウム 405
グリベンクラミド 984 687 コウブシ 1842, 2753
グリメピリド 985 d-クロルフェニラミンマレイン酸塩 コウブシ末 1843, 2753
グリメピリド錠 986 691 コウベイ 1820
クリンダマイシン塩酸塩 721 クロルフェニラミンマレイン酸塩散 コウボク 1910
クリンダマイシン塩酸塩カプセル 689 コウボク末 1910
722 クロルフェニラミンマレイン酸塩錠 ゴオウ 1928, 2778
クリンダマイシンリン酸エステル 690 コカイン塩酸塩 745
724 クロルフェニラミンマレイン酸塩注射 ゴシツ 1792
クリンダマイシンリン酸エステル注射 液 688 牛車腎気丸エキス 1865, 2758
液 725 クロルフェネシンカルバミン酸エステ ゴシュユ 1851, 2755
グルコン酸カルシウム水和物 552 ル 685 コデインリン酸塩散1z 746
グルタチオン 991 クロルフェネシンカルバミン酸エステ コデインリン酸塩散10z 747
L-グルタミン 990 ル錠 686 コデインリン酸塩錠 748
L-グルタミン酸 989 クロルプロパミド 694 コデインリン酸塩水和物 746
クレゾール 756 クロルプロパミド錠 695 ゴナドレリン酢酸塩 996
クレゾール水 756 クロルプロマジン塩酸塩 692 ゴボウシ 1821
クレゾール石ケン液 757 クロルプロマジン塩酸塩錠 693 ゴマ 1983
クレボプリドリンゴ酸塩 720 クロルプロマジン塩酸塩注射液 692 ゴマ油 1984
クレマスチンフマル酸塩 721 クロルヘキシジン塩酸塩 683 ゴミシ 1973
クロカプラミン塩酸塩水和物 727 クロルヘキシジングルコン酸塩液 コムギデンプン 1604
クロキサシリンナトリウム水和物 682 コメデンプン 1603
743, 2683 クロルマジノン酢酸エステル 684 コリスチンメタンスルホン酸ナトリウ
クロキサゾラム 744 クロロブタノール 685 ム 752, 2683
クロコナゾール塩酸塩 757 コリスチン硫酸塩 753
クロスカルメロースナトリウム 585 ケ コルチゾン酢酸エステル 754
クロスポビドン 758 コルヒチン 749
クロチアゼパム 741 ケイガイ 1973 五苓散エキス 2756
クロチアゼパム錠 2682 経口生ポリオワクチン 1428 コレカルシフェロール 696
クロトリマゾール 742 ケイ酸アルミン酸マグネシウム 2715 コレスチミド 750
Supplement I, JP XVII Index in Japanese 2867

コレスチミド顆粒 751 シアナミド 759 814


コレスチミド錠 752 シアノコバラミン 760 ジフェンヒドラミン・フェノール・亜鉛
コレステロール 697 シアノコバラミン注射液 761 華リニメント 815
コレラワクチン 696 ジエチルカルバマジンクエン酸塩 ジフェンヒドラミン塩酸塩 814
コロンボ 1821 790 ジブカイン塩酸塩 786
コロンボ末 1822 ジエチルカルバマジンクエン酸塩錠 ジフテリアトキソイド 816
コンズランゴ 1836 790 ジフテリア破傷風混合トキソイド
コンズランゴ流エキス 1837 ジオウ 1954 816
歯科用アンチホルミン 436 ジフルコルトロン吉草酸エステル
サ 歯科用トリオジンクパスタ 1734 793
歯科用パラホルムパスタ 1367 シプロフロキサシン 711
サイクロセリン 763 歯科用フェノール・カンフル 1392 シプロフロキサシン塩酸塩水和物
サイコ 1820 歯科用ヨード・グリセリン 1078 712
柴胡桂枝湯エキス 1965, 2785 ジギトキシン 794, 2684 シプロヘプタジン塩酸塩水和物 764
サイシン 1801 ジギトキシン錠 795, 2684 ジフロラゾン酢酸エステル 792
柴朴湯エキス 1963, 2783 シクラシリン 699 ジベカシン硫酸塩 785
柴苓湯エキス 1968, 2787 ジクロキサシリンナトリウム水和物 ジベカシン硫酸塩点眼液 785
酢酸 365 789 シベレスタットナトリウム水和物
酢酸ナトリウム水和物 1563 シクロスポリン 700 1561
サッカリン 1535 ジクロフェナクナトリウム 787 シベンゾリンコハク酸塩 697
サッカリンナトリウム水和物 1536, ジクロフェナミド 787, 2684 シベンゾリンコハク酸塩錠 698
2734 ジクロフェナミド錠 789, 2684 シメチジン 709
サフラン 1963 シクロペントラート塩酸塩 761 ジメモルファンリン酸塩 808
サラシ粉 683 シクロホスファミド錠 762 ジメルカプロール 810
サラシミツロウ 1808 シクロホスファミド水和物 762 ジメルカプロール注射液 811
サラゾスルファピリジン 1537 シゴカ 1848 ジメンヒドリナート 809
サリチル・ミョウバン散 1542 ジゴキシン 796, 2685 ジメンヒドリナート錠 809
サリチル酸 1539 ジゴキシン錠 798 次没食子酸ビスマス 512
サリチル酸精 1540 ジゴキシン注射液 797 ジモルホラミン 811
サリチル酸ナトリウム 1590 ジコッピ 1909 ジモルホラミン注射液 812
サリチル酸絆創膏 1540 シコン 1907 シャカンゾウ 1947
サリチル酸メチル 1238 次硝酸ビスマス 513 弱アヘンアルカロイド・スコポラミン
ザルトプロフェン 1780 ジスチグミン臭化物 818 注射液 1340
ザルトプロフェン錠 1780 ジスチグミン臭化物錠 819 シャクヤク 1935
サルブタモール硫酸塩 1538 L-シスチン 766 芍薬甘草湯エキス 1984, 2790
サルポグレラート塩酸塩 1543 L-システイン 765 シャクヤク末 1936
サルポグレラート塩酸塩細粒 1544 L-システイン塩酸塩水和物 766 ジャショウシ 1834
サルポグレラート塩酸塩錠 1545 シスプラチン 714 シャゼンシ 1942
酸化亜鉛 1783 ジスルフィラム 820 シャゼンソウ 1942
酸化カルシウム 554 ジソピラミド 818 臭化カリウム 1432
酸化チタン 1693 シタラビン 767 臭化ナトリウム 1568
酸化マグネシウム 1186 シチコリン 715 十全大補湯エキス 1887, 2766
サンキライ 1993 シツリシ 2005 ジュウヤク 1880
サンキライ末 1993 ジドブジン 1781 シュクシャ 1797
サンザシ 1842 ジドロゲステロン 844 シュクシャ末 1797
三酸化二ヒ素 445 ジドロゲステロン錠 845 酒石酸 1643
サンシシ 1854, 2755 シノキサシン 709 ショウキョウ 1857
サンシシ末 1854 シノキサシンカプセル 710 ショウキョウ末 1858
サンシュユ 1839, 2752 ジノスタチン スチマラマー 1786, 小柴胡湯エキス 1988, 2791
サンショウ 1885 2744 硝酸イソソルビド 1104
サンショウ末 1886 ジノプロスト 812 硝酸イソソルビド錠 1105
酸素 1350 ジヒドロエルゴタミンメシル酸塩 硝酸銀 1557
サンソウニン 1886, 2765 802 硝酸銀点眼液 1557
サントニン 1542 ジヒドロエルゴトキシンメシル酸塩 常水 1773
サンヤク 1847 803 ショウズク 1825
サンヤク末 1847 ジヒドロコデインリン酸塩 800 小青竜湯エキス 1990, 2793
ジヒドロコデインリン酸塩散1z 800 焼セッコウ 1868
シ ジヒドロコデインリン酸塩散10z 消毒用エタノール 897
801 消毒用フェノール 1391
ジアスターゼ 782 ジピリダモール 817 消毒用フェノール水 1392
ジアスターゼ・重曹散 783 ジフェニドール塩酸塩 791 ショウマ 1830
ジアゼパム 783 ジフェンヒドラミン 813 ジョサマイシン 1112
ジアゼパム錠 784 ジフェンヒドラミン・バレリル尿素散 ジョサマイシン錠 1113
2868 Index in Japanese Supplement I, JP XVII

ジョサマイシンプロピオン酸エステル スルタミシリントシル酸塩錠 1625 セフィキシムカプセル 624


1113 スルタミシリントシル酸塩水和物 セフィキシム水和物 623, 2676
シラザプリル錠 703 1624, 2736 セフェピム塩酸塩水和物 621
シラザプリル水和物 702 スルチアム 1626 セフォジジムナトリウム 629
シラスタチンナトリウム 701 スルバクタムナトリウム 1612, 2735 セフォゾプラン塩酸塩 640
ジラゼプ塩酸塩水和物 805 スルピリド 1621 セフォタキシムナトリウム 634
ジルチアゼム塩酸塩 806 スルピリドカプセル 1621 セフォチアム塩酸塩 639
ジルチアゼム塩酸塩徐放カプセル スルピリド錠 1622 セフォチアム ヘキセチル塩酸塩
807 スルピリン水和物 1623 637
シルニジピン 704 スルピリン注射液 1623 セフォテタン 635
シルニジピン錠 705 スルファジアジン銀 1614 セフォペラゾンナトリウム 631
シロスタゾール 707 スルファメチゾール 1615 セフカペン ピボキシル塩酸塩細粒
シロスタゾール錠 708 スルファメトキサゾール 1616 613
シロップ用アシクロビル 373 スルファモノメトキシン水和物 1616 セフカペン ピボキシル塩酸塩錠
シロップ用セファトリジンプロピレン スルフイソキサゾール 1617 614
グリコール 606 スルベニシリンナトリウム 1613 セフカペン ピボキシル塩酸塩水和物
シロップ用セファドロキシル 599 スルホブロモフタレインナトリウム 612
シロップ用セファレキシン 603 1618 セフジトレン ピボキシル 618
シロップ用セフポドキシム プロキセ スルホブロモフタレインナトリウム注 セフジトレン ピボキシル細粒 619
チル 646 射液 1619 セフジトレン ピボキシル錠 620
シロップ用セフロキサジン 649 セフジニル 615
シロップ用トラニラスト 1715 セ セフジニルカプセル 617
シロップ用ファロペネムナトリウム セフジニル細粒 617
920 成人用沈降ジフテリアトキソイド セフスロジンナトリウム 650
シロップ用ペミロラストカリウム 816 セフタジジム水和物 652
1374 精製水 1773 セフチゾキシムナトリウム 658, 2678
シロップ用ホスホマイシンカルシウム 精製水(容器入り) 1774 セフチブテン水和物 657
963 精製ゼラチン 980 セフテラム ピボキシル 654
シロドシン 1553 精製セラック 1551 セフテラム ピボキシル細粒 655
シロドシン錠 1555 精製デヒドロコール酸 772 セフテラム ピボキシル錠 656
シンイ 1911 精製白糖 1611 セフトリアキソンナトリウム水和物
シンギ 1875 精製ヒアルロン酸ナトリウム 1575 660
親水クリーム 756 精製ヒアルロン酸ナトリウム注射液 セフピラミドナトリウム 642
親水ワセリン 1387 1576 セフピロム硫酸塩 643
診断用クエン酸ナトリウム液 1572 精製ヒアルロン酸ナトリウム点眼液 セフブペラゾンナトリウム 610
シンバスタチン 1559 1577 セフポドキシム プロキセチル 644
シンバスタチン錠 1560 精製ブドウ糖 2696 セフポドキシム プロキセチル錠
真武湯エキス 1985, 2791 精製ラノリン 1904 647
生理食塩液 1571 セフミノクスナトリウム水和物 629
ス 石油ベンジン 1387 セフメタゾールナトリウム 627
セタノール 671 セフメノキシム塩酸塩 625
水酸化カリウム 1436 セチリジン塩酸塩 671 セフロキサジン水和物 648
水酸化カルシウム 553 セチリジン塩酸塩錠 672 セフロキシム アキセチル 662
水酸化ナトリウム 1578 セッコウ 1868 セボフルラン 1550
スキサメトニウム塩化物水和物 1627 セトチアミン塩酸塩水和物 673 セラセフェート 663, 2679
スキサメトニウム塩化物注射液 セトラキサート塩酸塩 674 ゼラチン 978
1628, 2737 セネガ 1980 セラペプターゼ 1549, 2734
スクラルファート水和物 1609 セネガシロップ 1981 L-セリン 1548
スコポラミン臭化水素酸塩水和物 セネガ末 1981 セルモロイキン(遺伝子組換え) 668
1548 セファクロル 592 センキュウ 1835
ステアリルアルコール 1607 セファクロルカプセル 593 センキュウ末 1835
ステアリン酸 1606 セファクロル細粒 596 ゼンコ 1937
ステアリン酸カルシウム 562 セファクロル複合顆粒 594 センコツ 1919
ステアリン酸ポリオキシル40 1430 セファゾリンナトリウム 607 センソ 1998
ステアリン酸マグネシウム 1188 セファゾリンナトリウム水和物 609 センナ 1981
ストレプトマイシン硫酸塩 1608 セファトリジンプロピレングリコール センナ末 1982
スピラマイシン酢酸エステル 1599, 605 センブリ 1996
2735 セファドロキシル 597 センブリ・重曹散 1998
スピロノラクトン 1600 セファドロキシルカプセル 598 センブリ末 1997
スピロノラクトン錠 1600 セファレキシン 599
スペクチノマイシン塩酸塩水和物 セファレキシンカプセル 601 ソ
1597 セファレキシン複合顆粒 602
スリンダク 1620 セファロチンナトリウム 604 ソウジュツ 1803
Supplement I, JP XVII Index in Japanese 2869

ソウジュツ末 1804 チアミン塩化物塩酸塩散 1675 注射用ドセタキセル 823


ソウハクヒ 1918 チアミン塩化物塩酸塩注射液 1675 注射用ナルトグラスチム(遺伝子組換
ゾニサミド 2744 チアミン硝化物 1676 え) 1293
ゾニサミド錠 2745 チアラミド塩酸塩 1685 注射用パニペネム・ベタミプロン
ソボク 1972 チアラミド塩酸塩錠 1686 1361
ソヨウ 1937 チアントール 1678 注射用バンコマイシン塩酸塩 1758
ソルビタンセスキオレイン酸エステル チオペンタールナトリウム 1680 注射用ヒト絨毛性性腺刺激ホルモン
1595 チオリダジン塩酸塩 1681 999
ゾルピデム酒石酸塩 1788 チオ硫酸ナトリウム水和物 1592 注射用ヒドララジン塩酸塩 1022
ゾルピデム酒石酸塩錠 1789 チオ硫酸ナトリウム注射液 1593 注射用ピペラシリンナトリウム 1418
D-ソルビトール 1596 チクセツニンジン 1932 注射用ビンブラスチン硫酸塩 1763
D-ソルビトール液 1597 チクセツニンジン末 1933 注射用ファモチジン 917
チクロピジン塩酸塩 1687 注射用フェニトインナトリウム 1398
タ チクロピジン塩酸塩錠 1688 注射用プレドニゾロンコハク酸エステ
チザニジン塩酸塩 1694 ルナトリウム 1456
ダイオウ 1955 窒素 1316 注射用フロモキセフナトリウム 938
大黄甘草湯エキス 1843, 2754 チニダゾール 1691 注射用ペプロマイシン硫酸塩 1380
ダイオウ末 1956 チペピジンヒベンズ酸塩 1691 注射用ベンジルペニシリンカリウム
大柴胡湯エキス 1844, 2754 チペピジンヒベンズ酸塩錠 1692 487
ダイズ油 1995 チメピジウム臭化物水和物 1689 注射用ホスホマイシンナトリウム
タイソウ 1886, 2765 チモ 1797 965
ダウノルビシン塩酸塩 769 チモール 1683 注射用ボリコナゾール 2743
タウリン 1643 チモロールマレイン酸塩 1690 注射用マイトマイシン C 1262
タカルシトール水和物 1629 注射用アシクロビル 371 注射用ミノサイクリン塩酸塩 1256
タカルシトール軟膏 1631 注射用アズトレオナム 467 注射用メロペネム 1219
タカルシトールローション 1630 注射用アセチルコリン塩化物 367 注射用ロキサチジン酢酸エステル塩酸
タクシャ 1793 注射用アミカシン硫酸塩 409 塩 1533
タクシャ末 1794, 2747 注射用アムホテリシンB 427 チョウジ 1833
タクロリムスカプセル 1633 注射用アンピシリンナトリウム 432 チョウジ末 1833
タクロリムス水和物 1632 注射用アンピシリンナトリウム・スル チョウジ油 1834
タゾバクタム 1644 バクタムナトリウム 433 チョウトウコウ 2007
ダナゾール 768 注射用イダルビシン塩酸塩 1047 釣藤散エキス 1827, 2751
タムスロシン塩酸塩 1640 注射用イミペネム・シラスタチンナト チョレイ 1946
タムスロシン塩酸塩徐放錠 1642 リウム 1056 チョレイ末 1946
タモキシフェンクエン酸塩 1640 注射用オザグレルナトリウム 1357 L-チロシン 1740
タランピシリン塩酸塩 1633 注射用シベレスタットナトリウム チンク油 1784
タルク 1634 1562 沈降ジフテリア破傷風混合トキソイド
タルチレリン口腔内崩壊錠 1637 注射用水 1774 816
タルチレリン錠 1638 注射用水(容器入り) 1775 沈降精製百日せきジフテリア破傷風混
タルチレリン水和物 1636 注射用スキサメトニウム塩化物 1628 合ワクチン 816
炭酸カリウム 1433 注射用ストレプトマイシン硫酸塩 沈降精製百日せきワクチン 1384
炭酸水素ナトリウム 1566 1609 沈降炭酸カルシウム 548
炭酸水素ナトリウム注射液 1566 注射用スペクチノマイシン塩酸塩 沈降炭酸カルシウム細粒 548
炭酸ナトリウム水和物 1569 1598 沈降炭酸カルシウム錠 549
炭酸マグネシウム 1185 注射用セファゾリンナトリウム 608 沈降破傷風トキソイド 1670
炭酸リチウム 1169 注射用セフェピム塩酸塩 622 沈降はぶトキソイド 1004
単シロップ 1558 注射用セフォゾプラン塩酸塩 641 沈降 B 型肝炎ワクチン 1018
タンジン 1971 注射用セフォチアム塩酸塩 640 チンピ 1832
ダントロレンナトリウム水和物 768 注射用セフォペラゾンナトリウム
単軟膏 1993 2677 ツ
タンニン酸 1643 注射用セフォペラゾンナトリウム・ス
タンニン酸アルブミン 386 ルバクタムナトリウム 632 ツバキ油 1822
タンニン酸ジフェンヒドラミン 815 注射用セフタジジム 653 ツロブテロール 1738
タンニン酸ベルベリン 494 注射用セフメタゾールナトリウム ツロブテロール塩酸塩 1740
628 ツロブテロール経皮吸収型テープ
チ 注射用タゾバクタム・ピペラシリン 1739
1645
チアプリド塩酸塩 1684 注射用チアミラールナトリウム 1678 テ
チアプリド塩酸塩錠 1685 注射用チオペンタールナトリウム
チアマゾール 1672 1681 テイコプラニン 1654, 2737
チアマゾール錠 1673 注射用テセロイキン(遺伝子組換え) 低置換度ヒドロキシプロピルセルロー
チアミラールナトリウム 1677 1652 ス 1035, 2703
チアミン塩化物塩酸塩 1674 注射用ドキソルビシン塩酸塩 837 テオフィリン 1672
2870 Index in Japanese Supplement I, JP XVII

テガフール 1653 トウヒ 1812 トリコマイシン 1724


デキサメタゾン 776 トウヒシロップ 1926 L-トリプトファン 1737
デキストラン40 777, 2684 トウヒチンキ 1926 トリヘキシフェニジル塩酸塩 1727
デキストラン40注射液 778 トウモロコシデンプン 1601 トリヘキシフェニジル塩酸塩錠 1728
デキストラン70 779 トウモロコシ油 1839 トリメタジオン 1732
デキストラン硫酸エステルナトリウム ドキサゾシンメシル酸塩 833 トリメタジジン塩酸塩 1730
イオウ5 780 ドキサゾシンメシル酸塩錠 834 トリメタジジン塩酸塩錠 1731
デキストラン硫酸エステルナトリウム ドキサプラム塩酸塩水和物 832 トリメトキノール塩酸塩水和物 1733
イオウ18 780 ドキシサイクリン塩酸塩錠 840 トリメブチンマレイン酸塩 1729
デキストリン 781 ドキシサイクリン塩酸塩水和物 838, ドルゾラミド塩酸塩 830
デキストロメトルファン臭化水素酸塩 2686 ドルゾラミド塩酸塩点眼液 831
水和物 782 ドキシフルリジン 835 トルナフタート 1704
テストステロンエナント酸エステル ドキシフルリジンカプセル 835 トルナフタート液 1705
1667 ドキソルビシン塩酸塩 836, 2685 トルブタミド 1703
テストステロンエナント酸エステル注 ドクカツ 1800 トルブタミド錠 1703
射液 1668 トコフェロール 1697 トルペリゾン塩酸塩 1705
テストステロンプロピオン酸エステル トコフェロールコハク酸エステルカル L-トレオニン 1682
1668 シウム 1698 トレハロース水和物 1717
テストステロンプロピオン酸エステル トコフェロール酢酸エステル 1697 トレピブトン 1718
注射液 1669 トコフェロールニコチン酸エステル ドロキシドパ 842
デスラノシド 775 1699 ドロキシドパカプセル 842
デスラノシド注射液 776 トコン 1881 ドロキシドパ細粒 843
テセロイキン(遺伝子組換え) 1647 トコンシロップ 1882 トロキシピド 1735
テトラカイン塩酸塩 1670 トコン末 1881 トロキシピド細粒 1735
テトラサイクリン塩酸塩 1670, 2738 トスフロキサシントシル酸塩錠 1707 トロキシピド錠 1736
デヒドロコール酸 772 トスフロキサシントシル酸塩水和物 トロピカミド 1734
デヒドロコール酸注射液 773 1706 ドロペリドール 841
デフェロキサミンメシル酸塩 771 ドセタキセル水和物 821 トロンビン 1682, 2738
テプレノン 1660 ドセタキセル注射液 822 豚脂 1905
テプレノンカプセル 1661 トチュウ 1851 ドンペリドン 824
デメチルクロルテトラサイクリン塩酸 トドララジン塩酸塩水和物 1700
塩 774, 2684 ドネペジル塩酸塩 825 ナ
テモカプリル塩酸塩 1658 ドネペジル塩酸塩細粒 826
テモカプリル塩酸塩錠 1659 ドネペジル塩酸塩錠 827 ナイスタチン 1328
テルビナフィン塩酸塩 1662 ドパミン塩酸塩 829 ナタネ油 1953, 2780
テルビナフィン塩酸塩液 1664 ドパミン塩酸塩注射液 829 ナテグリニド 1294
テルビナフィン塩酸塩クリーム 1663 トフィソパム 1701 ナテグリニド錠 1295
テルビナフィン塩酸塩錠 1665 ドブタミン塩酸塩 820 ナドロール 1283
テルビナフィン塩酸塩スプレー 1665 トブラマイシン 1695, 2738 ナファゾリン・クロルフェニラミン液
テルブタリン硫酸塩 1666 トブラマイシン注射液 1696 1290
テルミサルタン 1656 トラガント 2004 ナファゾリン塩酸塩 1289
テルミサルタン錠 1657 トラガント末 2004 ナファゾリン硝酸塩 1289
テレビン油 2007 トラザミド 1702, 2738 ナファモスタットメシル酸塩 1283
天然ケイ酸アルミニウム 404 トラニラスト 1711 ナフトピジル 1284
デンプングリコール酸ナトリウム トラニラストカプセル 1712 ナフトピジル口腔内崩壊錠 1285
1591 トラニラスト細粒 1713 ナフトピジル錠 1286
テンマ 1855 トラニラスト点眼液 1714 ナブメトン 1281
テンモンドウ 1802 トラネキサム酸 1708 ナブメトン錠 1282
トラネキサム酸カプセル 1709 ナプロキセン 1291
ト トラネキサム酸錠 1711 ナリジクス酸 1287
トラネキサム酸注射液 1710 ナルトグラスチム(遺伝子組換え)
桃核承気湯エキス 1999, 2796 トラピジル 1716 1291
トウガシ 1811 トラマドール塩酸塩 2739 ナロキソン塩酸塩 1288
トウガラシ 1822 トリアムシノロン 1718
トウガラシ・サリチル酸精 1824 トリアムシノロンアセトニド 1719 ニ
トウガラシチンキ 1824 トリアムテレン 1720
トウガラシ末 1823 トリエンチン塩酸塩 1726 ニガキ 1941
トウキ 1883 トリエンチン塩酸塩カプセル 1727 ニガキ末 1941
当帰芍薬散エキス 2002, 2797 トリクロホスナトリウム 1725 ニカルジピン塩酸塩 1298
トウキ末 1883 トリクロホスナトリウムシロップ ニカルジピン塩酸塩注射液 1299
トウジン 1835 1726 ニクジュヨウ 1831
トウニン 1933 トリクロルメチアジド 1721 ニクズク 1920
トウニン末 1934 トリクロルメチアジド錠 1722 ニコチン酸 1307
Supplement I, JP XVII Index in Japanese 2871

ニコチン酸アミド 1306 白色セラック 1552


ニコチン酸注射液 1307 白色軟膏 1775 ヒ
ニコモール 1304 白色ワセリン 1386
ニコモール錠 1304 白糖 1611 ピオグリタゾン塩酸塩 1406
ニコランジル 1305 バクモンドウ 1923 ピオグリタゾン塩酸塩・グリメピリド
ニザチジン 1319 麦門冬湯エキス 1805, 2747 錠 1408
ニザチジンカプセル 1320 バクロフェン 470 ピオグリタゾン塩酸塩・メトホルミン
二酸化炭素 580 バクロフェン錠 470 塩酸塩錠 1411
ニセリトロール 1303 バシトラシン 469, 2674 ピオグリタゾン塩酸塩錠 1407
ニセルゴリン 1300 パズフロキサシンメシル酸塩 2726 ビオチン 510
ニセルゴリン散 1301 パズフロキサシンメシル酸塩注射液 ピコスルファートナトリウム水和物
ニセルゴリン錠 1302 2728 1585
二相性イソフェンインスリン ヒト バソプレシン注射液 1759, 2740 ビサコジル 511
(遺伝子組換え)水性懸濁注射液 八味地黄丸エキス 1868, 2759 ビサコジル坐剤 512
2706 ハチミツ 1879 L-ヒスチジン 1018
ニトラゼパム 1314 ハッカ 1914 L-ヒスチジン塩酸塩水和物 1019
ニトレンジピン 1314 ハッカ水 1915 ビソプロロールフマル酸塩 514
ニトレンジピン錠 1315 ハッカ油 1915 ビソプロロールフマル酸塩錠 515
ニトログリセリン錠 1317 パップ用複方オウバク散 1940 ピタバスタチンカルシウム錠 1425
ニフェジピン 1308 パニペネム 1359 ピタバスタチンカルシウム水和物
ニフェジピン細粒 1311 パパベリン塩酸塩 1363 1424
ニフェジピン徐放カプセル 1310 パパベリン塩酸塩注射液 1364 ビタミン A 油 1765
ニフェジピン腸溶細粒 1309 ハマボウフウ 1861 ヒト下垂体性性腺刺激ホルモン 999
日本脳炎ワクチン 1111 バメタン硫酸塩 471 ヒト絨毛性性腺刺激ホルモン 997
乳酸 1131 パラアミノサリチル酸カルシウム顆粒 人全血液 1775
L-乳酸 1131 556 人免疫グロブリン 1021
乳酸カルシウム水和物 553 パラアミノサリチル酸カルシウム水和 ヒドララジン塩酸塩 1021
L-乳酸ナトリウム液 1581 物 555 ヒドララジン塩酸塩散 1022
L-乳酸ナトリウムリンゲル液 1582 パラオキシ安息香酸エチル 906 ヒドララジン塩酸塩錠 1022
乳糖水和物 1133, 2715 パラオキシ安息香酸ブチル 540 ヒドロキシジン塩酸塩 1036
尿素 1747 パラオキシ安息香酸プロピル 1481 ヒドロキシジンパモ酸塩 1036
ニルバジピン 1312 パラオキシ安息香酸メチル 1235 ヒドロキシプロピルセルロース
ニルバジピン錠 1313 バラシクロビル塩酸塩 1751 1033, 2702
ニンジン 1859 バラシクロビル塩酸塩錠 1753 ヒドロキソコバラミン酢酸塩 1033,
ニンジン末 1860 パラフィン 1364 2701
ニンドウ 1908 パラホルムアルデヒド 1366 ヒドロクロロチアジド 1024
L-バリン 1754 ヒドロコタルニン塩酸塩水和物 1031
ネ バルサルタン 1755 ヒドロコルチゾン 1025
バルサルタン錠 1756 ヒドロコルチゾン・ジフェンヒドラミ
ネオスチグミンメチル硫酸塩 1297 パルナパリンナトリウム 1367 ン軟膏 1027
ネオスチグミンメチル硫酸塩注射液 バルビタール 472 ヒドロコルチゾンコハク酸エステル
1297 バルプロ酸ナトリウム 1593 1031
バルプロ酸ナトリウム錠 1594 ヒドロコルチゾンコハク酸エステルナ
ノ バルプロ酸ナトリウムシロップ 1594 トリウム 1030
バレイショデンプン 1602 ヒドロコルチゾン酢酸エステル
濃グリセリン 993 ハロキサゾラム 1008 1026, 2701
濃ベンザルコニウム塩化物液50 480 パロキセチン塩酸塩錠 1371 ヒドロコルチゾン酪酸エステル
ノスカピン 1326 パロキセチン塩酸塩水和物 1369 1028, 2701
ノスカピン塩酸塩水和物 1327 ハロタン 1007 ヒドロコルチゾンリン酸エステルナト
ノルアドレナリン 1321, 2726 ハロペリドール 1004 リウム 1028
ノルアドレナリン注射液 1322 ハロペリドール細粒 1005 ピブメシリナム塩酸塩 1427
ノルエチステロン 1322 ハロペリドール錠 1006 ピブメシリナム塩酸塩錠 1428
ノルゲストレル 1324 ハロペリドール注射液 1006 ヒプロメロース 1038, 2704
ノルゲストレル・エチニルエストラジ パンクレアチン 1358 ヒプロメロースカプセル 574
オール錠 1324 パンクロニウム臭化物 1358 ヒプロメロース酢酸エステルコハク酸
ノルトリプチリン塩酸塩 1326 ハンゲ 1941 エステル 1040
ノルフロキサシン 1323 半夏厚朴湯エキス 1871, 2761 ヒプロメロースフタル酸エステル
半夏瀉心湯エキス 1873, 2761 1041
ハ バンコマイシン塩酸塩 1757 ピペミド酸水和物 1414
パンテチン 1362 ピペラシリン水和物 1414
バイモ 1853 パントテン酸カルシウム 554 ピペラシリンナトリウム 1416
バカンピシリン塩酸塩 468 ピペラジンアジピン酸塩 1418
バクガ 1912 ピペラジンリン酸塩錠 1419
2872 Index in Japanese Supplement I, JP XVII

ピペラジンリン酸塩水和物 1419 フェノールスルホンフタレイン 1393 フラビンアデニンジヌクレオチドナト


ビペリデン塩酸塩 510 フェノールスルホンフタレイン注射液 リウム 932
ビホナゾール 509 1393 フラボキサート塩酸塩 934
ヒマシ油 1826 フェルビナク 923 プランルカスト水和物 1442
ピマリシン 1404 フェルビナクテープ 924 プリミドン 1458
ヒメクロモン 1037 フェルビナクパップ 923 フルオキシメステロン 950, 2694
ピモジド 1405 フェンタニルクエン酸塩 925 フルオシノニド 947
ビャクゴウ 1906 フェンブフェン 924 フルオシノロンアセトニド 946
ビャクシ 1798 複方アクリノール・チンク油 377 フルオレセインナトリウム 948
ビャクジュツ 1804 複方オキシコドン・アトロピン注射液 フルオロウラシル 950
ビャクジュツ末 1805 1348 フルオロメトロン 949
氷酢酸 365 複方オキシコドン注射液 1347 フルコナゾール 940
ピラジナミド 1488 複方サリチル酸精 1541 フルコナゾールカプセル 941
ピラルビシン 1420 複方サリチル酸メチル精 1238 フルコナゾール注射液 942
ピランテルパモ酸塩 1487 複方ジアスターゼ・重曹散 783 フルジアゼパム 944
ピリドキサールリン酸エステル水和物 複方ダイオウ・センナ散 1957 フルシトシン 943
2731 複方チアントール・サリチル酸液 フルスルチアミン塩酸塩 973
ピリドキシン塩酸塩 1489 1679 フルタミド 954
ピリドキシン塩酸塩注射液 1490 複方ビタミン B 散 1765 フルトプラゼパム 955
ピリドスチグミン臭化物 1489 複方ヨード・グリセリン 1077 フルトプラゼパム錠 955
ピルシカイニド塩酸塩カプセル 1402 複方ロートエキス・ジアスターゼ散 フルドロコルチゾン酢酸エステル
ピルシカイニド塩酸塩水和物 1402 1976 944
ピレノキシン 1421 ブクモロール塩酸塩 527 フルニトラゼパム 945
ピレンゼピン塩酸塩水和物 1422 ブクリョウ 1946 フルフェナジンエナント酸エステル
ピロ亜硫酸ナトリウム 1587 ブクリョウ末 1947 951
ピロカルピン塩酸塩 1399 ブシ 1948 フルボキサミンマレイン酸塩 956
ピロカルピン塩酸塩錠 1400 フシジン酸ナトリウム 1574 フルボキサミンマレイン酸塩錠 958
ピロキシカム 1423 ブシ末 1949 フルラゼパム塩酸塩 952
ピロキシリン 1491 ブシラミン 525 プルラン 1487
ピロールニトリン 1491 ブシラミン錠 526 プルランカプセル 574
ビワヨウ 1909 ブスルファン 536 フルルビプロフェン 953
ビンクリスチン硫酸塩 1764, 2742 ブチルスコポラミン臭化物 1547 ブレオマイシン塩酸塩 516
ピンドロール 1406 ブテナフィン塩酸塩 536 ブレオマイシン硫酸塩 518
ビンブラスチン硫酸塩 1762, 2742 ブテナフィン塩酸塩液 538 フレカイニド酢酸塩 934
ビンロウジ 1800 ブテナフィン塩酸塩クリーム 537 フレカイニド酢酸塩錠 935
ブテナフィン塩酸塩スプレー 538 プレドニゾロン 1452
フ ブドウ酒 1776 プレドニゾロンコハク酸エステル
ブドウ糖 988 1456
ファモチジン 915 ブドウ糖水和物 2695 プレドニゾロン酢酸エステル 1454
ファモチジン散 918 ブドウ糖注射液 989, 2698 プレドニゾロン錠 1453
ファモチジン錠 919 フドステイン 968 プレドニゾロンリン酸エステルナトリ
ファモチジン注射液 915 フドステイン錠 969 ウム 1455
ファロペネムナトリウム錠 921 ブトロピウム臭化物 539 プロカインアミド塩酸塩 1462
ファロペネムナトリウム水和物 919 ブナゾシン塩酸塩 532 プロカインアミド塩酸塩錠 1463
フィトナジオン 1398 ブピバカイン塩酸塩水和物 533 プロカインアミド塩酸塩注射液 1463
フィルグラスチム(遺伝子組換え) ブフェトロール塩酸塩 528 プロカイン塩酸塩 1464
929 ブプラノロール塩酸塩 534 プロカイン塩酸塩注射液 1465
フィルグラスチム(遺伝子組換え)注射 ブプレノルフィン塩酸塩 535 プロカテロール塩酸塩水和物 1466
液 931 ブホルミン塩酸塩 529 プロカルバジン塩酸塩 1466
フェキソフェナジン塩酸塩 926 ブホルミン塩酸塩錠 531 プログルミド 1470
フェキソフェナジン塩酸塩錠 927 ブホルミン塩酸塩腸溶錠 529 プロクロルペラジンマレイン酸塩
フェニトイン 1396 ブメタニド 531 1467
フェニトイン散 1396 フラジオマイシン硫酸塩 966 プロクロルペラジンマレイン酸塩錠
フェニトイン錠 1397 プラステロン硫酸エステルナトリウム 1468
L-フェニルアラニン 1394 水和物 1444 プロゲステロン 1469
フェニルブタゾン 1395 プラゼパム 1450 プロゲステロン注射液 1470
フェニレフリン塩酸塩 1395 プラゼパム錠 1450 フロセミド 970
フェネチシリンカリウム 1388 プラゾシン塩酸塩 1451 フロセミド錠 972
フェノバルビタール 1389 プラノプロフェン 1443 フロセミド注射液 971
フェノバルビタール散10z 1390 プラバスタチンナトリウム 1444 プロタミン硫酸塩 1483
フェノール 1390 プラバスタチンナトリウム液 1447 プロタミン硫酸塩注射液 1484
フェノール・亜鉛華リニメント 1392 プラバスタチンナトリウム細粒 1446 プロチオナミド 1484
フェノール水 1392 プラバスタチンナトリウム錠 1448 ブロチゾラム 523
Supplement I, JP XVII Index in Japanese 2873

ブロチゾラム錠 524 ベポタスチンベシル酸塩錠 489 ホミカチンキ 1922


プロチレリン 1485 ペミロラストカリウム 1372 ホモクロルシクリジン塩酸塩 1020
プロチレリン酒石酸塩水和物 1486 ペミロラストカリウム錠 1375 ボリコナゾール 1769
プロテイン銀 1557 ペミロラストカリウム点眼液 1373 ボリコナゾール錠 1770
プロテイン銀液 1558 ベラドンナエキス 1810 ポリスチレンスルホン酸カルシウム
プロパフェノン塩酸塩 1473 ベラドンナコン 1809 559
プロパフェノン塩酸塩錠 1474 ベラドンナ総アルカロイド 1810 ポリスチレンスルホン酸ナトリウム
プロパンテリン臭化物 1475 ベラパミル塩酸塩 1761, 2741 1586
プロピベリン塩酸塩 1476 ベラパミル塩酸塩錠 1761, 2741 ポリソルベート80 1430, 2730
プロピベリン塩酸塩錠 1477 ベラプロストナトリウム 490 ホリナートカルシウム 551
プロピルチオウラシル 1482 ベラプロストナトリウム錠 492 ポリミキシン B 硫酸塩 1429, 2730
プロピルチオウラシル錠 1482 ペルフェナジン 1381 ホルマリン 961
プロピレングリコール 1480 ペルフェナジン錠 1382 ホルマリン水 961
プロブコール 1460 ペルフェナジンマレイン酸塩 1383 ホルモテロールフマル酸塩水和物
プロブコール細粒 1461 ペルフェナジンマレイン酸塩錠 1383 962
プロブコール錠 1462 ベルベリン塩化物水和物 493 ボレイ 1932
プロプラノロール塩酸塩 1478 ベンザルコニウム塩化物 479 ボレイ末 1932
プロプラノロール塩酸塩錠 1479 ベンザルコニウム塩化物液 480
フロプロピオン 939 ベンジルアルコール 483 マ
フロプロピオンカプセル 939 ベンジルペニシリンカリウム 486,
プロベネシド 1458 2674 マイトマイシン C 1261
プロベネシド錠 1459 ベンジルペニシリンベンザチン水和物 マオウ 1849
ブロマゼパム 520 485 麻黄湯エキス 1912, 2776
ブロムヘキシン塩酸塩 521 ヘンズ 1848 マーキュロクロム 1216, 2718
プロメタジン塩酸塩 1472 ベンズブロマロン 481 マーキュロクロム液 1217, 2718
フロモキセフナトリウム 936 ベンゼトニウム塩化物 481 マクリ 1846
ブロモクリプチンメシル酸塩 522 ベンゼトニウム塩化物液 482 マクロゴール400 1182
ブロモバレリル尿素 523 ベンセラジド塩酸塩 478 マクロゴール1500 1182
L-プロリン 1471 ペンタゾシン 1376 マクロゴール4000 1183
粉末セルロース 667 ペントキシベリンクエン酸塩 1378 マクロゴール6000 1183
ベントナイト 478 マクロゴール20000 1184
ヘ ペントバルビタールカルシウム マクロゴール軟膏 1184
1377, 2728 マシニン 1876
ベカナマイシン硫酸塩 474 ペントバルビタールカルシウム錠 麻酔用エーテル 898
ベクロメタゾンプロピオン酸エステル 2729 マニジピン塩酸塩 1192
473 ペンブトロール硫酸塩 1376 マニジピン塩酸塩錠 1193
ベザフィブラート 507 マプロチリン塩酸塩 1196
ベザフィブラート徐放錠 508 ホ マルトース水和物 1190
ベタキソロール塩酸塩 506 D-マンニトール 1194, 2718
ベタネコール塩化物 507 ボウイ 1993 D-マンニトール注射液 1195
ベタヒスチンメシル酸塩 495 防已黄耆湯エキス 1817, 2750
ベタヒスチンメシル酸塩錠 496 ボウコン 1880 ミ
ベタミプロン 505 ホウ酸 520
ベタメタゾン 497 ホウ砂 1567 ミグリトール 1253
ベタメタゾン吉草酸エステル 502 ボウショウ 1994 ミグレニン 1254
ベタメタゾン吉草酸エステル・ゲンタ 抱水クロラール 676 ミクロノマイシン硫酸塩 1250
マイシン硫酸塩クリーム 502 ボウフウ 1971, 2789 ミコナゾール 1249
ベタメタゾン吉草酸エステル・ゲンタ 防風通聖散エキス 1813, 2748 ミコナゾール硝酸塩 1249
マイシン硫酸塩軟膏 504 ボクソク 1952 ミゾリビン 1262
ベタメタゾンジプロピオン酸エステル ボグリボース 1766 ミゾリビン錠 1263
499 ボグリボース錠 1767 ミチグリニドカルシウム錠 1259
ベタメタゾン錠 498 ホスホマイシンカルシウム水和物 ミチグリニドカルシウム水和物 1258
ベタメタゾンリン酸エステルナトリウ 962, 2694 ミツロウ 1809
ム 500 ホスホマイシンナトリウム 964, 2694 ミデカマイシン 1251
ペチジン塩酸塩 1385 ボタンピ 1915 ミデカマイシン酢酸エステル 1252
ペチジン塩酸塩注射液 1385 ボタンピ末 1916 ミノサイクリン塩酸塩 1255
ベニジピン塩酸塩 475 補中益気湯エキス 1876, 2763 ミノサイクリン塩酸塩錠 1256
ベニジピン塩酸塩錠 476 ポビドン 1439, 2731 ミョウバン水 401
ヘパリンカルシウム 1009, 2698 ポビドンヨード 1441
ヘパリンナトリウム 1013, 2699 ホマトロピン臭化水素酸塩 1020 ム
ヘパリンナトリウム注射液 1017 ホミカ 1920
ペプロマイシン硫酸塩 1379 ホミカエキス 1921 無コウイ大建中湯エキス 1917, 2777
ベポタスチンベシル酸塩 488 ホミカエキス散 1922 無水アンピシリン 429
2874 Index in Japanese Supplement I, JP XVII

無水エタノール 896, 2693 メトロニダゾール 1246 ヨウ化人血清アルブミン(131I)注射液


無水カフェイン 543 メトロニダゾール錠 1246 1075
無水クエン酸 716 メナテトレノン 1209 ヨウ化ヒプル酸ナトリウム(131I)注射
無水乳糖 1132, 2714 メピチオスタン 1212 液 1580
無水ボウショウ 1994 メピバカイン塩酸塩 1213 葉酸 959, 2694
無水リン酸水素カルシウム 557 メピバカイン塩酸塩注射液 1213 葉酸錠 960
ムピロシンカルシウム水和物 1279 メフェナム酸 1203 葉酸注射液 959
ムピロシンカルシウム軟膏 1280 メフルシド 1204 ヨウ素 1075
メフルシド錠 1205 ヨクイニン 1836
メ メフロキン塩酸塩 1203 ヨクイニン末 1836
メペンゾラート臭化物 1211 抑肝散エキス 2010, 2798
メキシレチン塩酸塩 1248 メルカプトプリン水和物 1215 ヨード・サリチル酸・フェノール精
メキタジン 1214 メルファラン 1208 1079
メキタジン錠 1215 メロペネム水和物 1217 ヨードチンキ 1076
メグルミン 1206 dl-メントール 1210 ヨードホルム 1080
メクロフェノキサート塩酸塩 1197 l-メントール 1211
メコバラミン 1197 ラ
メコバラミン錠 1198 モ
メサラジン 2718 ラウリル硫酸ナトリウム 1584, 2734
メサラジン徐放錠 2720 木クレオソート 2008 ラウロマクロゴール 1144
メストラノール 1220 モクツウ 1793 ラクツロース 1134
メタケイ酸アルミン酸マグネシウム モサプリドクエン酸塩散 1276 ラタモキセフナトリウム 1143
2716 モサプリドクエン酸塩錠 1277 ラッカセイ油 1935
メダゼパム 1200 モサプリドクエン酸塩水和物 1275 ラナトシド C 1137, 2715
メタンフェタミン塩酸塩 1223 モッコウ 1972 ラナトシド C 錠 1138, 2715
L-メチオニン 1224 モノステアリン酸アルミニウム 402 ラニチジン塩酸塩 1503
メチクラン 1241 モノステアリン酸グリセリン 995 ラフチジン 1135
メチラポン 1247 モルヒネ・アトロピン注射液 1273 ラフチジン錠 1136
dl-メチルエフェドリン塩酸塩 1231 モルヒネ塩酸塩錠 1272 ラベタロール塩酸塩 1129
dl-メチルエフェドリン塩酸塩散10z モルヒネ塩酸塩水和物 1270 ラベタロール塩酸塩錠 1130
1232 モルヒネ塩酸塩注射液 1271 ラベプラゾールナトリウム 1502
メチルエルゴメトリンマレイン酸塩 モルヒネ硫酸塩水和物 1274 ランソプラゾール 1139
1233 モンテルカストナトリウム 1264 ランソプラゾール腸溶カプセル 1140
メチルエルゴメトリンマレイン酸塩錠 モンテルカストナトリウム顆粒 2724 ランソプラゾール腸溶性口腔内崩壊錠
1233 モンテルカストナトリウム錠 1269 1141
メチルジゴキシン 1242 モンテルカストナトリウムチュアブル
メチルセルロース 1228, 2723 錠 1267 リ
メチルテストステロン 1239
メチルテストステロン錠 1240 ヤ リオチロニンナトリウム 1165
メチルドパ錠 1230 リオチロニンナトリウム錠 1166
メチルドパ水和物 1229 ヤクチ 1812 リシノプリル錠 1168
メチルプレドニゾロン 1236 ヤクモソウ 1905 リシノプリル水和物 1167
メチルプレドニゾロンコハク酸エステ 薬用石ケン 1201 L-リシン塩酸塩 1180
ル 1236 薬用炭 1200 L-リシン酢酸塩 1179
メチルベナクチジウム臭化物 1227 ヤシ油 1835 リスペリドン 1521
メチルロザニリン塩化物 1237 リスペリドン細粒 1522
滅菌精製水(容器入り) 1774 ユ リスペリドン錠 1524
メテノロンエナント酸エステル 1221 リスペリドン内服液 1523
メテノロンエナント酸エステル注射液 ユウタン 1807 リセドロン酸ナトリウム錠 1589
1222 ユーカリ油 1850 リセドロン酸ナトリウム水和物 1588
メテノロン酢酸エステル 1220 輸血用クエン酸ナトリウム注射液 リゾチーム塩酸塩 1181
メトキサレン 1227 1572 六君子湯エキス 1957, 2780
メトクロプラミド 1243 ユビデカレノン 1743 リドカイン 1161
メトクロプラミド錠 1243 リドカイン注射液 1162
メトトレキサート 1225 ヨ リトドリン塩酸塩 1525
メトトレキサート錠 2722 リトドリン塩酸塩錠 1526
メトトレキサートカプセル 1225 ヨウ化カリウム 1437 リバビリン 1511
メトプロロール酒石酸塩 1244 ヨウ化ナトリウム 1579 リバビリンカプセル 1512
メトプロロール酒石酸塩錠 1245 ヨウ化ナトリウム(123I)カプセル リファンピシン 1518
メトホルミン塩酸塩 1222 1579 リファンピシンカプセル 1519
メトホルミン塩酸塩錠 1223 ヨウ化ナトリウム(131I)液 1580 リボスタマイシン硫酸塩 1517
メドロキシプロゲステロン酢酸エステ ヨウ化ナトリウム(131I)カプセル リボフラビン 1513
ル 1202 1580 リボフラビン散 1514
Supplement I, JP XVII Index in Japanese 2875

リボフラビン酪酸エステル 1514 1530


リボフラビンリン酸エステルナトリウ レ ロキサチジン酢酸エステル塩酸塩徐放
ム 1515 カプセル 1531
リボフラビンリン酸エステルナトリウ レセルピン 1507 ロキサチジン酢酸エステル塩酸塩徐放
ム注射液 1516 レセルピン散0.1z 1508 錠 1532
リマプロスト アルファデクス 1162 レセルピン錠 1509 ロキシスロマイシン 1534
リュウガンニク 1907 レセルピン注射液 1508 ロキシスロマイシン錠 2733
リュウコツ 1907 レチノール酢酸エステル 1510 ロキソプロフェンナトリウム錠 1178
リュウコツ末 1908 レチノールパルミチン酸エステル ロキソプロフェンナトリウム水和物
硫酸亜鉛水和物 1785 1510 1177
硫酸亜鉛点眼液 1785 レナンピシリン塩酸塩 1144 ロキタマイシン 1528, 2733
硫酸アルミニウムカリウム水和物 レノグラスチム(遺伝子組換え) 1146 ロキタマイシン錠 1529, 2733
403 レバミピド 1504 ロサルタンカリウム 1172
硫酸カリウム 1438 レバミピド錠 1506 ロサルタンカリウム・ヒドロクロロチ
硫酸鉄水和物 926 レバロルファン酒石酸塩 1152 アジド錠 1174
硫酸バリウム 473 レバロルファン酒石酸塩注射液 1152 ロサルタンカリウム錠 1173
硫酸マグネシウム水 1190 レボチロキシンナトリウム錠 1160 ロジン 1960
硫酸マグネシウム水和物 1189 レボチロキシンナトリウム水和物 ロートエキス 1974
硫酸マグネシウム注射液 1190 1159 ロートエキス・アネスタミン散 1976
リュウタン 1884 レボドパ 1153 ロートエキス・カーボン散 1976
リュウタン末 1884 レボフロキサシン細粒 1155 ロートエキス・タンニン坐剤 1978
流動パラフィン 1365 レボフロキサシン錠 1157 ロートエキス・パパベリン・アネスタミ
リュープロレリン酢酸塩 1150 レボフロキサシン水和物 1154 ン散 1977, 2790
リョウキョウ 1796 レボフロキサシン注射液 1156 ロートエキス散 1975
苓桂朮甘湯エキス 1961, 2781 レボフロキサシン点眼液 1156 ロートコン 1973
リンゲル液 1520 レボホリナートカルシウム水和物 ロベンザリットナトリウム 1171
リンコマイシン塩酸塩水和物 1164 2675 ローヤルゼリー 1960
リンコマイシン塩酸塩注射液 1164 レボメプロマジンマレイン酸塩 1159 ロラゼパム 1171
リン酸水素カルシウム水和物 558 レンギョウ 1852
リン酸水素ナトリウム水和物 1585 レンニク 1919 ワ
リン酸二水素カルシウム水和物 558
ロ ワイル病秋やみ混合ワクチン 1775
ワルファリンカリウム 1771
L-ロイシン1149 ワルファリンカリウム錠 1772
ロキサチジン酢酸エステル塩酸塩

You might also like