文档库 最新最全的文档下载
当前位置:文档库 › FDA对生物类似物的质量要求指南201202

FDA对生物类似物的质量要求指南201202

FDA对生物类似物的质量要求指南201202
FDA对生物类似物的质量要求指南201202

Quality Considerations in

Demonstrating Biosimilarity to a Reference

Protein Product

DRAFT GUIDANCE

This guidance document is being distributed for comment purposes only. Comments and suggestions regarding this draft document should be submitted within 60 days of publication in the Federal Register of the notice announcing the availability of the draft guidance. Submit comments to the Division of Dockets Management (HFA-305), Food and

Drug Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD 20852. All comments

should be identified with the docket number listed in the notice of availability that publishes in

the Federal Register.

For questions regarding this draft document contact (CDER) Sandra Benton at 301-796-2500.

U.S. Department of Health and Human Services

Food and Drug Administration

Center for Drug Evaluation and Research (CDER)

Center for Biologics Evaluation and Research (CBER)

February 2012

Biosimilarity

Quality Considerations in

Demonstrating Biosimilarity to a Reference

Protein Product

Additional copies are available from:

Office of Communications

Division of Drug Information, WO51, Room 2201

Center for Drug Evaluation and Research

Food and Drug Administration

10903 New Hampshire Ave., Silver Spring, MD 20993

Phone: 301-796-3400; Fax: 301-847-8714

druginfo@https://www.wendangku.net/doc/f39543063.html,

https://www.wendangku.net/doc/f39543063.html,/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm

and/or

Office of Communication, Outreach, and

Development, HFM-40

Center for Biologics Evaluation and Research

Food and Drug Administration

1401 Rockville Pike, Rockville, MD 20852-1448

(Tel) 800-835-4709 or 301-827-1800

https://www.wendangku.net/doc/f39543063.html,/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htm

U.S. Department of Health and Human Services

Food and Drug Administration

Center for Drug Evaluation and Research (CDER)

Center for Biologics Evaluation and Research (CBER)

February 2012

Biosimilarity

TABLE OF CONTENTS

I. INTRODUCTION (1)

II. BACKGROUND (2)

III. SCOPE (4)

IV. DEFINITIONS (5)

V. GENERAL PRINCIPLES (6)

VI. FACTORS FOR CONSIDERATION IN ASSESSING WHETHER PRODUCTS ARE HIGHLY SIMILAR (9)

A. Expression System (9)

B. Manufacturing Process (10)

C. Assessment of Physicochemical Properties (10)

D. Functional Activities (11)

E. Receptor Binding and Immunochemical Properties (12)

F. Impurities (12)

G. Reference Product and Reference Standards (13)

H. Finished Drug Product (14)

I. Stability (15)

VII. CONCLUSION (15)

VIII. RELEVANT GUIDANCES (16)

5 10 15 20 25 30 35 40 1 Guidance for Industry 1 2 Quality Considerations in Demonstrating Biosimilarity to a

3

Reference Protein Product

4 6 7 This draft guidance, when finalized, will represent the Food and Drug Administration's (FDA's) current 8 thinking on this topic. It does not create or confer any rights for or on any person and does not operate to 9 bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA 11 staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call 12 the appropriate number listed on the title page of this guidance. 13 14 16 I. INTRODUCTION 17 18 This guidance describes the Agency’s current thinking on factors to consider when

19 demonstrating that a proposed protein product is highly similar to a reference product licensed under section 351(a) of the Public Health Service Act (PHS Act) for purposes of submitting a 21 marketing application under section 351(k) of the PHS Act. Specifically, the guidance is

22 intended to provide recommendations to applicants on the scientific and technical information of 23 the chemistry, manufacturing, and controls (CMC) section of a marketing application for a 24 proposed biosimilar product submitted under section 351(k) of the PHS Act. 26 The Biologics Price Competition and Innovation Act of 2009 (BPCI Act) amends the PHS Act 27 and other statutes to create an abbreviated licensure pathway in section 351(k) of the PHS Act 28 for biological products shown to be biosimilar to, or interchangeable with, an FDA-licensed 29 biological reference product (see sections 7001 through 7003 of the Patient Protection and

Affordable Care Act (Pub. L. 111-148) (Affordable Care Act)). The BPCI Act also amended the 31 definition of biological products to include “protein (except any chemically synthesized 32 polypeptide)” (see section 351(i)(1) of the PHS Act). A 351(k) application for a proposed

33 biosimilar product must include information demonstrating biosimilarity, based on data derived 34 from, among other things, “analytical studies that demonstrate that the biological product is highly similar to the reference product notwithstanding minor differences in clinically inactive 36 components.”2 37 38 Although the 351(k) pathway applies generally to biological products, this guidance focuses on 39 therapeutic protein products and provides an overview of analytical factors to consider in demonstrating biosimilarity between a proposed protein product and the reference product. 41

1

This guidance has been prepared by the Center for Drug Evaluation and Research (CDER) and the Center for Biologics Evaluation and Research (CBER) at the Food and Drug Administration (FDA). 2

See section 351(k)(2)(A)(i)(I)(aa) of the PHS Act.

42 This guidance is one in a series of guidances that FDA is developing to implement the BPCI Act.

43 The guidances will address a broad range of issues, including:3

44

45 ?Quality Considerations in Demonstrating Biosimilarity to a Reference Protein

46 Product

47 ?Scientific Considerations in Demonstrating Biosimilarity to a Reference Product

48 ?Biosimilars: Questions and Answers Regarding Implementation of the Biologics

49 Price Competition and Innovation Act of 2009

50

51 When applicable, references to information in these guidances are included in this guidance.

52

53 FDA’s guidance documents, including this guidance, do not establish legally enforceable

54 responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should

55 be viewed only as recommendations, unless specific regulatory or statutory requirements are

56 cited. The use of the word should in Agency guidances means that something is suggested or

57 recommended, but not required.

58

59

60 II. BACKGROUND

61

62 In the 1980s, FDA began to receive marketing applications for biotechnology-derived protein

63 products, mostly for recombinant DNA-derived versions of a naturally sourced product. In light

64 of these applications, FDA established a regulatory approach for the approval of recombinant

65 DNA-derived protein products, which it announced in a policy document published on June 26,

66 1986 (51 FR 23309), in conjunction with a 1985 document titled Points to Consider in the

67 Production and Testing of New Drugs and Biologicals Produced by Recombinant DNA

68 Technology. The policy requires the submission of an investigational new drug application

69 (IND) to FDA for evaluation before initiation of clinical investigations in human subjects and

70 submission and approval of a new drug application (NDA) or biologics license application

71 (BLA) “before marketing products made with recombinant DNA technology, even if the active

72 ingredient in the product is thought to be identical to a naturally occurring substance or a

73 previously approved product” (51 FR 23309). The policy set forth in those documents was

74 developed in part because of the challenges in evaluating protein products solely by

75 physicochemical and functional testing and because the biological system in which a protein

76 product is produced can have a significant effect on the structure and function of the product

77 itself. Due to the complexities of protein products, FDA has, as a matter of policy, generally

78 required submission of an NDA in accordance with section 505(b)(1) of the FD&C Act or a BLA

79 in accordance with section 351(a) of the PHS Act containing product-specific full safety and

80 efficacy data for recombinant DNA-derived protein drugs. FDA has recognized, however, that

81 “[i]n some instances complete new applications may not be required.” (51 FR 23309).

82

83 Improvements in manufacturing processes, process controls, materials and product testing, as

84 well as characterization tests and studies, have led to a gradual evolution in the regulation of

3 We update guidances periodically. To make sure you have the most recent version of a guidance, check the CDER

guidance page at https://www.wendangku.net/doc/f39543063.html,/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm.

85 protein products. For example, in 1996, FDA provided recommendations in its FDA Guidance

86 Concerning Demonstration of Comparability of Human Biological Products, Including

87 Therapeutic Biotechnology Products, which explains how an applicant may demonstrate,

88 through a combination of analytical testing, functional assays (in vitro and/or in vivo),

89 assessment of pharmacokinetics (PK) and/or pharmacodynamics (PD) and toxicity in animals,

90 and clinical testing (clinical pharmacology, safety, and/or efficacy) that a manufacturing change

91 does not adversely affect identity, purity, or potency of its FDA-approved product.

92

93 Since 1996, FDA has approved many manufacturing process changes for licensed biological

94 products, based on a demonstration of product comparability before and after the process change,

95 as supported by quality criteria and analytical testing and without the need for additional

96 nonclinical data and clinical safety and/or efficacy studies. In some cases, uncertainty about the

97 effect of the change and/or the results of the biochemical/functional comparability studies has

98 necessitated assessment of additional data, including nonclinical and/or clinical testing, to

99 demonstrate product comparability.

100

101 These concepts were further developed in the International Conference on Harmonisation of 102 Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) and resulted 103 in the Q5E guidance on Comparability of Biotechnological/Biological Products Subject to

104 Changes in their Manufacturing Process. Although the scope of ICH Q5E is limited to an

105 assessment of the comparability of a biological product before and after a manufacturing process 106 change made by the same manufacturer, certain general scientific principles described in ICH 107 Q5E are applicable to an assessment of biosimilarity between a proposed biosimilar protein

108 product and its reference product. However, demonstrating that a proposed protein product is 109 biosimilar to an FDA-licensed reference product manufactured by a different manufacturer may 110 require more extensive and comprehensive data than assessing the comparability of a product 111 before and after a manufacturing process change made by the product’s sponsor. Unlike a

112 manufacturer who modifies its own manufacturing process with extensive knowledge and

113 information about the product and the existing process, including established controls and

114 acceptance parameters, the manufacturer of a proposed biosimilar product will likely have a

115 different manufacturing process (e.g., different cell line, raw materials, equipment, processes, 116 process controls, acceptance criteria) from that of the reference product and no direct knowledge 117 of the manufacturing process for the reference product.

118

119 In October 1999, FDA issued a draft guidance for industry on Applications Covered by Section 120 505(b)(2), which, among other things, stated that FDA may accept an application submitted

121 through the approval pathway described by section 505(b)(2) of the FD&C Act for a drug

122 product containing an active ingredient(s) derived from natural sources or recombinant DNA 123 technology. For example, FDA approved a 505(b)(2) application for a follow-on recombinant 124 DNA-derived human growth hormone product in May 2006. Greater knowledge due to

125 advances in science and technology, and improvements in manufacturing processes, process 126 controls, materials and product testing, as well as characterization tests and studies, facilitate the 127 use of an abbreviated pathway for the approval of a protein product.

128

129 The BPCI Act was enacted as part of the Affordable Care Act on March 23, 2010.4 The BPCI 130 Act creates an abbreviated licensure pathway for biological products demonstrated to be

131 biosimilar to, or interchangeable with, a reference product. Section 351(k) of the PHS Act (42 132 U.S.C. 262(k)), added by the BPCI Act, sets forth the requirements for an application for a

133 proposed biosimilar product and an application or a supplement for a proposed interchangeable 134 product.

135

136 Section 351(i) of the PHS Act defines biosimilarity to mean that the biological product is highly 137 similar to the reference product notwithstanding minor differences in clinically inactive

138 components and that there are no clinically meaningful differences between the biological

139 product and the reference product in terms of the safety, purity, and potency of the product (see 140 section 351(i)(2) of the PHS Act).

141

142 To meet the higher standard of “interchangeability,” an applicant must provide sufficient

143 information to demonstrate biosimilarity, and also to demonstrate that the biological product can 144 be expected to produce the same clinical result as the reference product in any given patient and, 145 if the biological product is administered more than once to an individual, the risk in terms of 146 safety or diminished efficacy of alternating or switching between the use of the biological

147 product and the reference product is not greater than the risk of using the reference product

148 without such alternation or switch (see section 351(k)(4) of the PHS Act).

149

150 Analytical studies provide the foundation for an assessment of the proposed protein product 151 intended for submission in a 351(k) application under the PHS Act and whether it is highly

152 similar to the reference product.

153

154

155 III. SCOPE

156

157 This document provides guidance on analytical studies that may be relevant to assessing whether 158 the proposed biosimilar protein product and a reference product are highly similar, which is part 159 of the biosimilarity assessment. This document is not intended to provide an overview of FDA’s 160 approach to determining interchangeability because FDA is continuing to consider the type of 161 information sufficient to enable FDA to determine that a biological product is interchangeable 162 with the reference product. Although this guidance applies specifically to therapeutic protein 163 products, the general scientific principles may be informative for the development of other

164 proteins, such as in vivo protein diagnostic products. If the reference product and the proposed 165 protein product cannot be adequately characterized with state of the art technology as

166 recommended by this guidance, FDA recommends that the sponsor consult FDA for guidance on 167 whether an application for the proposed protein product is appropriate for submission under 168 section 351(k) of the PHS Act.

169

170 All product applications should contain a complete and thorough chemistry, manufacturing and 171 controls (CMC) section that provides the necessary and appropriate information (e.g.,

172 characterization, adventitious agent safety, process controls, and specifications) for the product

4 The BPCI Act appears in title VII, subtitle A of the Affordable Care Act.

173 to be adequately reviewed. This guidance describes considerations for additional CMC 174 information that may be relevant to the assessment of biosimilarity between two protein

175 176 products. This guidance should be used as a companion to other guidances available from FDA that describe the CMC information appropriate for evaluation of protein products.5 We

177 encourage early interaction with FDA to discuss specific CMC issues that may arise for an

178 applicant’s proposed biosimilar product.

179

180 In addition to comparative analytical studies, an assessment of whether a proposed product is 181 biosimilar to a reference product generally will include animal studies (including the assessment

182 183 of toxicity) and a clinical study or studies (including the assessment of immunogenicity and pharmacokinetics and/or pharmacodynamics).6

184

185 This guidance applies to applications submitted under section 351(k) of the PHS Act. However,

186 187 some scientific principles described in this guidance may be informative for the development of certain biological products under section 505(b)(2) of the FD&C Act.7 Section 505(b)(2) of the

188 FD&C Act and section 351(k) of the PHS Act are two separate statutory schemes. This guidance 189 is not intended to describe any relationship between the standards for approval under these

190 schemes.

191

192

193 IV. DEFINITIONS

194

195 For the purpose of this document, the following definitions are applicable:

196

197 Protein means any alpha amino acid polymer with a specific defined sequence that is 198 greater than 40 amino acids in size.

199

200 Chemically synthesized polypeptide means any alpha amino acid polymer that is (a) made 201 entirely by chemical synthesis, and (b) is less than 100 amino acids in size.

202

5 For CMC requirements for submission of a marketing application, applicants should consult current regulations,

the Guidance for Industry for the Submission on Chemistry, Manufacturing, and Controls Information for a

Therapeutic Recombinant DNA-Derived Product or a Monoclonal Antibody Product for In-vivo Use (issued jointly by CBER and CDER, August 1996) and other applicable FDA guidance documents.

6 For a discussion of the Agency’s current thinking on animal and clinical studies relevant to demonstrating

biosimilarity, see Draft Guidance for Industry on Scientific Considerations in Demonstrating Biosimilarity to a

Reference Product (issued jointly by CDER and CBER, February 2012).

7 A 505(b)(2) application is an NDA that contains full reports of investigations of safety and effectiveness, where at

least some of the information required for approval comes from studies not conducted by or for the applicant and for which the applicant has not obtained a right of reference or use (e.g., the Agency’s finding of safety and/or

effectiveness for a listed drug or published literature). A 505(b)(2) application that seeks to rely on a listed drug

(i.e., the reference product) must contain adequate data and information to demonstrate that the proposed product is

sufficiently similar to the listed drug to justify reliance, in part, on FDA’s finding of safety and/or effectiveness for the listed drug. Any aspects of the proposed product that differ from the listed drug must be supported by adequate data and information to show that the differences do not affect the safety and effectiveness of the proposed product.

203 Biosimilar or biosimilarity means that “the biological product is highly similar to the 204 reference product notwithstanding minor differences in clinically inactive components,”

205 206 and “there are no clinically meaningful differences between the biological product and the reference product in terms of the safety, purity, and potency of the product.”8

207

208 Product, when used without modifiers, is intended to refer to intermediates, drug

209 substance, and/or drug product, as appropriate. The use of the term “product” is

210 consistent with the use of the term in ICH Q5E.

211

212 Reference product means the single biological product licensed under section 351(a) of 213 the PHS Act against which a biological product is evaluated in a 351(k) application. 214

215

216 V. GENERAL PRINCIPLES

217

218 Advances in analytical sciences (both physicochemical and biological) enable some protein 219 products to be characterized extensively in terms of their physicochemical and biological

220 properties. These analytical procedures have improved the ability to identify and characterize

221 222 not only the desired product but also product-related substances and product- and process-related impurities.9 Advances in manufacturing science and production methods may enhance the

223 likelihood that a product will be highly similar to another product by better targeting the original 224 product’s physiochemical and functional properties.

225

226 In addition to a complete CMC data submission as required under section 351(a) of the PHS Act, 227 the applicant should assess the analytical similarity to the reference product. The rationale for 228 the analytical similarity assessment should be clearly described with consideration for the known 229 quality attributes and performance characteristics of the specific reference product. Extensive, 230 robust comparative physicochemical and functional studies (these may include bioassays,

231 biological assays, binding assays, and enzyme kinetics) should be performed to evaluate whether 232 the proposed biosimilar product and the reference product are highly similar. A meaningful

233 assessment as to whether the proposed biosimilar product is highly similar to the reference

234 product depends on, among other things, the capabilities of available state-of-the-art analytical 235 assays to assess, for example, the molecular weight of the protein, complexity of the protein

236 (higher order structure and post-translational modifications), degree of heterogeneity, functional 237 properties, impurity profiles, and degradation profiles denoting stability. The capability of the 238 methods used in the analytical assessment, as well as their limitations should be described by the 239 applicant. Physicochemical and functional characterization studies should be sufficient to

240 establish relevant quality attributes including those that define a product’s identity, quantity, 241 purity, potency, and consistency. The product-related impurities, product-related substances, and 242 process-related impurities should be identified, characterized as appropriate, quantified, and

243 compared to those of the reference product to the extent feasible and relevant, as part of an

244 assessment of the potential impact on the safety, purity, and potency of the product.

8 Section 7002(b)(3) of the Affordable Care Act, adding section 351(i)(2) of the PHS Act.

9 The use of the terms “product-related substances” and “product- and process-related impurities” is consistent with

their use and meaning in ICH Q6B.

245

246 Primary structure of some protein products can be highly heterogeneous and could affect the 247 expected clinical performance of a protein product. In addition to the typically low level of

248 replication errors in the DNA encoding the protein sequence and amino acid misincorporation 249 that occurs during translation, most protein products undergo some post-translational

250 modification that can alter the functions of the protein: by attaching it to other biochemical

251 groups such as a phosphate, various lipids and carbohydrates; by proteolytic cleavage following 252 translation; by changing the chemical nature of an amino acid (e.g., formylation); or by many 253 other mechanisms. Such modifications can result from intracellular activities during cell culture 254 or by deliberate modification of the protein, for example, PEGylation. Other post-translational 255 modifications can be a consequence of manufacturing process operations — for example,

256 glycation may occur with exposure of the product to reducing sugars. In other cases, storage 257 conditions may be permissive for certain degradation pathways such as oxidation, deamidation, 258 or aggregation. As all of these product-related variants may alter the biological properties of the 259 expressed recombinant protein, identification and determination of the relative levels of these 260 protein variants should be included in the comparative analytical characterization studies.

261

262 The three dimensional conformation of a protein is an important factor in its biological function. 263 Proteins generally exhibit complex three-dimensional conformations (tertiary structure and, in 264 some cases, quaternary structure) due to their large size and the rotational characteristics of

265 protein alpha carbons. The resulting flexibility enables dynamic, but subtle, changes in protein 266 conformation over time, some of which may be absolutely required for functional activity.

267 These rotations are often dependent on low-energy interactions, such as hydrogen bonds and van 268 der Waals forces, which may be very sensitive to environmental conditions. Current analytical 269 technology is capable of evaluating the three-dimensional structure of many proteins. Methods 270 such as X-ray crystallography and multi-dimensional nuclear magnetic resonance (NMR)

271 spectroscopy can help define tertiary protein structure and, to varying extents, quaternary

272 structure, and can add to the body of information supporting biosimilarity. At the same time, a 273 protein’s three-dimensional conformation can often be difficult to define precisely using current 274 physicochemical analytical technology. Any differences in higher order structure between a 275 proposed biosimilar and a reference product should be evaluated in terms of a potential effect on 276 protein function. Thus, functional assays are also critical tools for evaluating the integrity of the 277 higher order structures.

278

279 A scientifically sound characterization that provides a comprehensive understanding of the

280 chemical, physical, and biological characteristics of the proposed biosimilar product is essential 281 to the design of the manufacturing process and to the conduct of development studies. The body 282 of knowledge that emerges will serve to support product quality during development, at

283 approval, and over the postapproval life of the product. Manufacturers should perform in-depth 284 chemical, physical, and bioactivity comparisons with side-by-side analyses of an appropriate 285 number of lots of the proposed biosimilar product and the reference product and, where available 286 and appropriate, a comparison with the reference standard for specific suitable attributes (e.g., 287 potency). For a discussion of reference standards, see section VI.G of this guidance. The

288 evaluation of multiple lots of reference product and biosimilar product enables determination of 289 product variability across lots and/or range of heterogeneity within a lot of drug product.

290 Identification of the specific lots of the reference product used in the biosimilar studies together

291 with expiration dates and timeframes of actual use would also be of value. This information will 292 be useful in justifying acceptance criteria used for specifications to ensure product consistency, 293 in addition to assessing similarity. However, acceptance criteria should be based on the totality 294 of the analytical data and not simply the observed range of product attributes of the reference 295 product. For example, some product attributes act in combination to define a product’s safety, 296 purity, and potency profile and therefore their potential interaction should be considered when 297 evaluating similarity and setting specifications. Thus, for some glycoproteins, the content and 298 distribution of tetraantennary and N-acetyl lactosamine repeats can affect in vivo potency and 299 should not be evaluated totally independently of each other. Additionally, data obtained for lots 300 used in nonclinical and clinical studies and relevant information on the relationship between an 301 attribute and the performance of the drug product (see ICH Q8) can also be used to help establish 302 acceptance criteria.

303

304 An extensive analytical characterization may also reveal differences between the reference

305 product and the proposed biosimilar product, especially when using analytical techniques

306 capable of discriminating qualitative or quantitative differences in product attributes. Emphasis 307 should be placed on developing orthogonal, quantitative methods to more definitively distinguish 308 any differences in product attributes. If the results show highly similar functional and

309 physicochemical characteristics, including, for example, higher order structure, post-translational 310 modifications, and impurity and degradation profiles, the sponsor may have an appropriate

311 scientific basis for a selective and targeted approach to subsequent animal and/or clinical studies 312 to support a demonstration of biosimilarity. It may be useful to compare differences in the

313 quality attributes of the proposed protein product with those of the reference product using a 314 meaningful fingerprint-like analysis algorithm that covers a large number of additional product 315 attributes and their combinations with high sensitivity using orthogonal methods. Advances in

316 317 manufacturing science and Quality-by-Design approaches may facilitate production processes that can better match a reference product’s fingerprint.10 Such a strategy could further quantify

318 the overall similarity between two molecules and may lead to additional bases for a more

319 selective and targeted approach to subsequent animal and/or clinical studies.

320

321 The type, nature, and extent of any differences between the proposed biosimilar product and the 322 reference product, introduced by design or observed from comprehensive analytical

323 characterization of multiple manufacturing lots, should be clearly described and discussed. The 324 discussion should include identification and comparison of relevant quality attributes from

325 product characterization, as this is an important factor in assessing whether the proposed

326 biosimilar product is highly similar to the reference product. The potential effect of the

327 differences on safety, purity, and potency should be addressed and supported by appropriate data. 328

329 The type and extent of nonclinical or clinical studies that are needed to demonstrate biosimilarity 330 of the proposed biosimilar product can be influenced by several factors, especially the ability to 331 discern differences and their potential effect on safety, purity, and potency. For example, factors 332 such as the ability to robustly characterize the proposed biosimilar product or the reference

333 product (e.g., lack of suitable or sufficiently discriminative analytical techniques) or availability 334 of a relevant drug substance derived from the reference product could impact the nature of the 335 subsequent nonclinical or clinical studies. In addition, if the proposed biosimilar product or

10 See ICH Q8(R2) for guidance.

336 reference product cannot be adequately characterized, the sponsor should consult FDA for

337 guidance on whether an application for such a protein product is appropriate for submission 338 under section 351(k) of the PHS Act.

339

340 In general, a sponsor needs to provide information to demonstrate biosimilarity based on data 341 directly comparing the proposed protein product with the reference product. Analytical studies 342 intended to support a demonstration of biosimilarity for purposes of section 351(k) of the PHS 343 Act must as a scientific matter include an adequate comparison to the reference product licensed 344 under section 351(a). However, under certain circumstances, a sponsor may seek to use data 345 derived from animal or clinical studies comparing a proposed protein product with a non-U.S.-346 licensed product to address, in part, the requirements under section 351(k)(2)(A) of the PHS Act. 347 In such a case, the sponsor should provide adequate data or information to scientifically justify

348 349 the relevance of this comparative data to an assessment of biosimilarity and to establish an acceptable bridge to the U.S.-licensed reference product.11 The scientific bridge between the

350 non-U.S.-licensed product and the U.S.-licensed reference product is likely to include

351 comparative physico-chemical characterization, bioassays/functional assays, and comparative 352 clinical and/or nonclinical PK and/or PD data, as appropriate, and data to address any differences 353 in formulation or primary packaging. Sponsors are encouraged to discuss with FDA during the 354 development program the adequacy of the scientific justification and bridge to the U.S.-licensed 355 reference product; a final determination of the adequacy of the information will be made by FDA 356 during review of the 351(k) application.

357

358 VI. FACTORS FOR CONSIDERATION IN ASSESSING WHETHER PRODUCTS 359 ARE HIGHLY SIMILAR

360

361 When assessing whether products are highly similar, manufacturers should consider a number of 362 factors, including the following.

363

364 A. Expression

System

365

366 Therapeutic protein products can be produced by microbial cells (prokaryotic, eukaryotic), cell 367 lines of human or animal origin (e.g., mammalian, avian, insect), or tissues derived from animals 368 or plants. It is expected that the expression construct for a proposed biosimilar product will

369 encode the same primary amino acid sequence as its reference product. However, minor

370 modifications, such as N or C terminal truncations that will not have an effect on safety, purity, 371 or potency, may be justified by the applicant. Differences between the chosen expression system 372 of the proposed biosimilar product and that of the reference product should be carefully

373 considered because the type of expression system and host cell will significantly affect the types 374 of process- and product-related substances and impurities (including potential adventitious

375 agents) that may be present in the protein product. For example, the expression system can have 376 a significant effect on the types and extent of translational and post-translational modifications

11 Please refer to the Draft Guidance for Industry on Scientific Considerations in Demonstrating Biosimilarity to a

Reference Product (issued jointly by CDER and CBER, February 2012).

377 that are imparted to the proposed protein product, something that may complicate an effort to 378 demonstrate that the proposed biosimilar product is highly similar to the reference product (and 379 thus, for example, affecting the type and extent of nonclinical and clinical data that are needed 380 for demonstrating biosimilarity). Minimizing differences between the proposed and reference 381 expression systems to the extent possible can enhance the likelihood of producing a highly

382 similar protein product. The characterization of the expression construct, including its genetic 383 stability, should be demonstrated in accordance with principles recommended in ICH Q5B.

384

Process

385 B. Manufacturing

386

387 A comprehensive understanding of all steps in the manufacturing process for the proposed

388 biosimilar product should be established during product development. Characterization tests, 389 process controls, and specifications that will emerge from information gained during process 390 development must be specific for the proposed biosimilar product and manufacturing process. 391 The use of Quality-by-Design approaches to pharmaceutical development, along with quality 392 risk management and effective quality systems, will facilitate the consistent manufacturing of a 393 high-quality product. A type II Drug Master File (DMF) would not be acceptable for a 351(k) 394 application because, as with 351(a) BLAs, the license holder needs to have knowledge of and 395 control over the manufacturing process for the biological product.12 Other types of contract

396 manufacturing arrangements can be considered if the applicant does not intend to manufacture 397 the product for licensure.13

398

399 C. Assessment of Physicochemical Properties

400

401 Physicochemical assessment of the proposed biosimilar product and the reference product should 402 consider all relevant characteristics of the protein product (e.g., the primary, secondary, tertiary, 403 and quaternary structure, post-translational modifications, and functional activity(ies)). The 404 objective of this assessment is to maximize the potential for detecting differences in quality

405 attributes between the proposed biosimilar product and the reference product.

406

407 The applicant should address the concept of the desired product (and its variants) as defined in 408 ICH Q6B when designing and conducting the characterization studies. Thus, it will be important 409 to understand the heterogeneity of the proposed biosimilar product and the reference product 410 (e.g., the nature, location, and levels of glycosylation) and the ranges of variability of different 411 isoforms, including those that result from post-translational modifications.

412

12 A type II DMF may, however, be used to support an Investigational New Drug Application (IND) for a biosimilar

product. Assurance of product quality should be provided on each lot of material produced by the DMF holder.

Procedures should also be in place to ensure that the IND sponsor is notified by the DMF holder of significant

changes to the DMF potentially affecting product quality. The sponsor is expected to provide notification to the

Agency of any relevant change in the IND in order to initiate a reevaluation of the DMF.

13 See FDA’s guidance on Cooperative Manufacturing Arrangements for Licensed Biologics (2008).

413 414 Particular analytical methodologies can be used to assess specific physicochemical characteristics of proteins. 14 These methodologies are described in published documents,

415 including scientific literature, regulatory guidelines, and pharmacopeial compendia. Some

416 techniques provide information on multiple characteristics. It is expected that appropriate

417 analytical test methods will be selected based on the nature of the protein being characterized 418 and knowledge regarding the structure and heterogeneity of the reference and the proposed

419 biosimilar product, as well as those characteristics that are critical to product performance. To 420 address the full range of physicochemical properties or biological activities adequately, it is often 421 necessary to apply more than one analytical procedure to evaluate the same quality attribute. 422 Methods that use different physicochemical or biological principles to assess the same attribute 423 are especially valuable because they provide independent data to support the quality of that

424 attribute (e.g., Size Exclusion Chromatography and Analytical Ultracentrifugation or Field Flow 425 Fractionation for the determination of aggregates). In addition, the use of complementary

426 analytical techniques in series, such as peptide mapping or capillary electrophoresis combined 427 with mass spectrometry of the separated molecules, should provide a meaningful and sensitive 428 method for comparing products.

429

430 Tests used to characterize the product do not necessarily need to be validated for routine quality 431 control purposes, but should be scientifically sound, fit for their intended use, and provide results 432 that are reproducible and reliable. In selecting these tests, it is important to consider the

433 characteristics of the protein product, including known and potential impurities. Information 434 regarding the ability of a method to discern relevant differences between a proposed biosimilar 435 product and a reference product should be submitted as part of the comparison.

436

437 Tests chosen to detect and characterize these post-translational protein modifications should be 438 demonstrated to be of appropriate sensitivity and specificity to provide meaningful information 439 as to whether the proposed biosimilar product and the reference product are highly similar.

440

441 D. Functional

Activities

442

443 Functional assays serve multiple purposes in the characterization of protein products. These tests 444 act to complement physicochemical analyses and are a quality measure of the function of the 445 protein product.

446

447 Depending on the structural complexity of the protein and available analytical technology, the 448 physicochemical analysis may be unable to confirm the integrity of the higher order structures. 449 Instead, the integrity of such structures can be inferred from the product’s biological activity. If 450 the clinically relevant mechanism(s) of action are known for the reference product or can

451 reasonably be determined, one or more of the functional assays should reflect these mechanisms 452 of action to the extent possible. The assessment of functional activity is also useful in providing 453 an estimate of the specific activity of a product, as an indicator of manufacturing process

454 consistency, as well as product purity and stability.

455

14 In some cases, in vivo immunogenicity studies may be able to detect subtle differences in structure or impurities

not detected by other methods.

456 If a reference product exhibits multiple functional activities, manufacturers should perform a set 457 of relevant assays designed to evaluate the range of activities. For example, with proteins that 458 possess multiple functional domains that express enzymatic and receptor-mediated activities, 459 manufacturers should evaluate both activities. For products where a single functional activity 460 can be measured by more than one, but related, parameter (e.g., enzyme kinetics or interactions 461 with blood clotting factors), comparative characterization of each parameter between products 462 should be used to provide additional valuable information.

463

464 The manufacturer should recognize the potential limitations of some types of functional assays, 465 such as high variability, that might preclude detection of small but significant differences

466 between the proposed biosimilar product and the reference product. As a highly variable assay 467 may not provide a meaningful assessment as to whether the proposed biosimilar product is

468 highly similar to the reference product, applicants are encouraged to develop assays that are

469 sensitive to changes in the functional activities of the product. In addition, in vitro bioactivity 470 assays may not fully reflect the clinical activity of the protein. For example, these assays

471 generally do not predict the bioavailability (PK and biodistribution) of the product. These

472 factors can impact PD and clinical performance. Also, bioavailability can be dramatically

473 altered by subtle differences in glycoform distribution or other post-translation modifications. 474 Thus, these limitations should be taken into account when assessing the robustness of the quality 475 of data supporting biosimilarity and the need for additional information. Finally, functional

476 assays are critical in assessing the occurrence of neutralizing antibodies in nonclinical and

477 clinical studies.

478

479 E. Receptor Binding and Immunochemical Properties

480

481 When binding or immunochemical properties are part of the activity attributed to the protein 482 product, analytical tests should be performed to characterize the product in terms of these

483 specific properties (e.g., if binding to a receptor is inherent in protein function, this property

484 should be measured and used in comparative studies, see ICH Q6B for additional details).

485 Various methods such as surface plasmon resonance, microcalorimetry, or classical Scatchard 486 analysis can provide information on the kinetics and thermodynamics of binding. Such

487 information can be related to the functional activity and characterization of the proposed

488 biosimilar product’s higher order structure.

489

490 F. Impurities

491

492 The applicant should characterize, identify, and quantify impurities (product- and process-related 493 as defined in ICH Q6B) in the proposed biosimilar product and the reference product. If

494 comparative physicochemical analysis reveals comparable product-related impurities at similar 495 levels between the two products, pharmacological/toxicological studies to characterize potential 496 biological effects of specific impurities may not be necessary. However, if the manufacturing 497 process used to produce the proposed biosimilar product introduces different impurities or higher 498 levels of impurities than those present in the reference product, additional

499 pharmacological/toxicological or other studies may be necessary. As discussed in ICH S6, “[i]t

500 501 is preferable to rely on purification processes to remove impurities . . . rather than to establish a preclinical testing program for their qualification.”15

502

503 Process-related impurities arising from cell substrates (e.g., host cell DNA, host cell proteins), 504 cell culture components (e.g., antibiotics, media components), and downstream processing steps 505 (e.g., reagents, residual solvents, leachables, endotoxin, bioburden) should be evaluated. The 506 potential impact of differences in the impurity profile upon safety should be addressed and

507 supported by appropriate data. In all cases, the chosen analytical procedures should be adequate 508 to detect, identify, and accurately quantify biologically significant levels of impurities (see ICH 509 Q2B). In particular, the results of the immunological methods used to detect host cell proteins 510 depend on the assay reagents and the cell substrate used. Such assays should be validated using

511 512 the product cell substrate and orthogonal methodologies to ensure accuracy and sensitivity. This should be done across both products to the extent relevant and feasible.16

513

514 The safety of the proposed biosimilar product, as with any biological product, with regard to 515 adventitious agents or endogenous viral contamination should be ensured by screening critical 516 raw materials and confirmation of robust virus removal and inactivation achieved by the

517 manufacturing process (see ICH Q5A for guidance).

518

519 G. Reference Product and Reference Standards

520

521 A thorough physicochemical and biological assessment of the reference product should provide a 522 base of information from which to develop the proposed biosimilar product and justify reliance 523 on certain existing scientific knowledge about the reference product. Sufficient evidence that the 524 proposed biosimilar product is highly similar to the reference product must be demonstrated in

525 526 an appropriate time frame to support a selective and targeted approach in early product development (e.g., reduced nonclinical studies, and/or dose-finding clinical studies).17 To justify

527 a selective and targeted approach to a clinical program, a comprehensive physicochemical and 528 functional comparison to the reference product should be performed during early product

529 development and discussed with the appropriate FDA staff. An analytical similarity assessment 530 should support the use of lots that demonstrate the biosimilarity of the proposed biosimilar

531 product used in the principal clinical trial to the reference product and the proposed commercial 532 product. The biosimilar application should include a thorough analytical comparison between 533 the proposed biosimilar product and the reference product. In addition, even when multiple 534 approved products are on the market, a sponsor must demonstrate that the proposed product is 535 biosimilar to a single reference product that previously has been licensed by FDA.

536

537 If the drug substance has been extracted from the reference product in order to assess analytical 538 similarity, the applicant should describe the extraction procedure and provide support that the 539 procedure itself does not alter product quality. This undertaking would include consideration for

15 See ICH S6, page 2.

16 This may be limited by the availability of high levels of reference product host cell proteins or differences in

product and reference substrate.

17 See 21 CFR 312.23 for Investigational New Drug (IND) application content and format.

540 alteration or loss of the desired products and impurities and relevant product-related substances, 541 and it should include appropriate controls that ensure the relevant product characteristics of the 542 reference product are not significantly altered by the extraction procedure.

543

544 If there is a suitable, publicly available and well-established reference standard for the protein, 545 then a physicochemical and/or functional comparison of the proposed biosimilar product with 546 this standard should also be performed. For example, if an international standard for calibration 547 of potency is available, a comparison of the relative potency of the proposed biosimilar product 548 with this potency standard should be performed. As is recommended in ICH Q6B, an in-house 549 reference standard(s) should always be qualified and used for control of the manufacturing

550 process and product.

551

552 In summary, analytical studies carried out to support the approval of a proposed biosimilar

553 product should not focus solely on the characterization of the proposed biosimilar product in 554 isolation. Rather, these studies should be part of a broad comparison that includes, but is not 555 limited to, the proposed biosimilar product, the reference product, applicable reference standards, 556 and consideration of relevant publicly available information.

557

558 H. Finished Drug Product

559

560 Product characterization studies should be performed on the most downstream intermediate best 561 suited for the analytical procedures used. The attributes evaluated should be stable through any

562 563 further processing steps. For these reasons, characterization studies are often performed on bulk drug substance.18 However if bulk drug substance is reformulated and/or exposed to new

564 materials in the finished dosage form, the impact of these changes should be considered.

565

566 If the finished drug product is best suited for a particular analysis, the characterization should 567 compare the proposed finished biosimilar product and the finished reference product. If an

568 analytical method more sensitively detects specific attributes in the drug substance, but the

569 attributes it measures are critical and/or may change during manufacture of the finished drug 570 product, comparative characterization may be called for on both the isolated drug substance and 571 the finished drug product.

572

573 The acceptability of the type, nature, and extent of any differences between the proposed finished 574 biosimilar product and the finished reference product should be evaluated and supported by

575 appropriate data and rationale. Additionally, different excipients in the proposed product should 576 be supported by existing toxicology data for the excipient or by additional toxicity studies with 577 the formulation of the proposed biosimilar product. Excipient interactions as well as direct

578 toxicities should be considered. Proteins are very sensitive to their environment. Therefore, 579 differences in excipients or primary packaging may affect product degradation and/or clinical 580 performance. Differences in formulation between the proposed biosimilar product and the

581 reference product are among the factors that may affect whether subsequent clinical studies may 582 take a selective and targeted approach.

583

18 See 21 CFR 207.3.

584 I. Stability

585

586 An appropriate physicochemical and functional comparison of the stability of the proposed

587 biosimilar product with that of the reference product should be initiated. Accelerated and stress 588 stability studies, or forced degradation studies, should be used to establish degradation profiles 589 and provide direct comparison of the proposed biosimilar product with the reference product. 590 These comparative studies should be conducted under multiple stress conditions (e.g., high

591 temperature, freeze thaw, light exposure, and agitation) that can cause incremental product

592 degradation over a defined time period. Results of these studies may reveal product differences 593 that warrant additional evaluation and also identify conditions under which additional controls 594 should be employed in manufacturing and storage (see ICH Q5C and Q1A(R) for guidance). 595 Sufficient real time, real condition stability data should be provided to support the proposed 596 dating period.

597

598

599 VII. CONCLUSION

600

601 The foundation for an assessment of biosimilarity between a proposed biosimilar product and its 602 reference product involves the robust characterization of the proposed biosimilar product,

603 including comparative physicochemical and functional studies. The information gained from 604 these studies is critical to the overall product assessment that as a scientific matter is necessary 605 for the development of a proposed biosimilar product. In addition, a 351(k) application for a 606 proposed biosimilar product must contain, among other things, information demonstrating

607 biosimilarity based upon data derived from animal studies (including the assessment of toxicity) 608 and a clinical study or studies (including the assessment of immunogenicity and

609 pharmacokinetics or pharmacodynamics), unless the Agency determines that an element is

610 unnecessary in a particular 351(k) application. The ability to discern relevant differences

611 between the proposed product and its reference product will depend on the available analytical 612 technology and complexity of the product. Any information regarding differences between the 613 proposed product and the reference product should be considered to determine whether the

614 statutory standard for biosimilarity can be met.

615

616

617 VIII. RELEVANT GUIDANCES

618

619 The following guidance documents may be relevant to sponsors developing or considering

620 development of a biosimilar product candidate. All Agency guidance documents are available 621 on FDA’s Web page (https://www.wendangku.net/doc/f39543063.html,/RegulatoryInformation/Guidances/default.htm).

622

623

624 1. Draft Guidance for Industry on Scientific Considerations in Demonstrating Biosimilarity 625 to a Reference Product (issued jointly by CDER and CBER, February 2012)

626

627 2. Draft Guidance for Industry, Biosimilars: Questions and Answers Regarding

628 Implementation of the Biologics Price Competition and Innovation Act of 2009 (issued 629 jointly by CDER and CBER, February 2012)

630

631 3. FDA Guidance Concerning Demonstration of Comparability of Human Biological

632 Products, Including Therapeutic Biotechnology-Derived Products (issued jointly by 633 CDER and CBER, April 1996)

634

635 4. Points to Consider in the Manufacture and Testing of Monoclonal Antibody Products for 636 Human Use (issued by CBER, February 1997)

637

638 5. Guidance for Industry for the Submission of Chemistry, Manufacturing, and Controls 639 Information for a Therapeutic Recombinant DNA-Derived Product or a Monoclonal 640 Antibody Product for In Vivo Use (issued jointly by CDER and CBER, August 1996) 641

642 6. FDA Guidance on Cooperative Manufacturing Arrangements for Licensed Biologics 643 (issued jointly by CDER and CBER, November 2008).

644

645 7. ICH M4Q The Common Technical Document

646

647 8. ICH Q2 Text on Validation of Analytical Procedures

648

649 9. ICH Q2B Validation of Analytical Procedures: Methodology

650

651 10. ICH Q3A Impurities in New Drug Substances

652

653 11. ICH Q5A Viral Safety Evaluation of Biotechnology Products Derived from Cell Lines of 654 Human or Animal Origi n

655

656 12. ICH Q5B Quality of Biotechnological Products: Analysis of the Expression Construct in 657 Cells Used for Production of r-DNA Derived Protein Products

658

659 13. ICH Q5C Stability Testing of Biotechnological/Biological Products

660

661 14. ICH Q5D Quality of Biotechnological/Biological Products: Derivation and

662 Characterization of Cell Substrates Used for Production of Biotechnological/Biological 663 Products

664

665 15. ICH Q5E Comparability of Biotechnological/Biological Products Subject to Changes in 666 Their Manufacturing Process

667

668 16. ICH Q6B Specifications: Test Procedures and Acceptance Criteria for

669 Biotechnological/Biological Products

670

671 17. ICH Q7 Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients 672

673 18. ICH Q8 Pharmaceutical Development

674

675 19. ICH Q9 Quality Risk Management

676

677 20. ICH Q10 Pharmaceutical Quality System

678

679 21. ICH S6 Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals

药品经营的质量风险管理规程

编号:SY-AQ-04294 ( 安全管理) 单位:_____________________ 审批:_____________________ 日期:_____________________ WORD文档/ A4打印/ 可编辑 药品经营的质量风险管理规程Regulations on quality risk management of pharmaceutical trade

药品经营的质量风险管理规程 导语:进行安全管理的目的是预防、消灭事故,防止或消除事故伤害,保护劳动者的安全与健康。在安全管 理的四项主要内容中,虽然都是为了达到安全管理的目的,但是对生产因素状态的控制,与安全管理目的关 系更直接,显得更为突出。 一、目的:为规范各经营环节质量风险的评估与控制管理。 二、依据:《药品经营质量管理规范》。 三、范围:适用于质量管理体系中所有经营环节质量风险的管理。 四、职责:质量负责人对本规程的实施负责。 五、内容: 1、成立风险管理小组 公司质量领导小组全面负责公司药品经营质量风险管理工作,由质量负责人主持质量风险管理的日常工作,成员包括各部门经理。质量管理部负责组织各部门对各经营环节质量风险的识别、评估与控制。 2、风险识别 各部门负责人依据GSP相关要求,结合部门经营环节的有关风

险,对可能存在的质量风险因素进行收集整理,填写《质量风险排查表》,报风险管理小组。 3、风险评估 风险管理小组召集与风险相关的部门、人员,依据《质量风险排查表》,对已经被识别的风险进行分析、评价,确定风险等级,确认出现问题的可能性、可识别性以及后果的严重性等,由质量管理部汇总形成《质量风险点列表》。 4、风险控制 4.1、各部门依据风险评估结果制定相应的预防、控制措施,对已经确定的风险点,使其降低或控制到可以接受的水平; 4.2、各部门负责人和质量管理部共同对风险控制措施进行审核、验证,确认能否将风险降低到可接受的水平上,如若不能则应该重新评估、确立防控措施。 5、风险沟通 各部门在实施风险控制措施的过程中,积极与相关部门人员进行沟通与信息交流,促进风险管理的有效实施,及时处理在实施过

风险管理试题及答案

《风险管理》(一~三章) 一、单选 1.由于形成的原因更加复杂和广泛,通常被视为一种综合风险的是(C)。 A.市场风险B.信用风险C.流动性风险D.操作风险 2.巴塞尔委员会以(D)方式把商业银行面临的风险分为八大类。 A.损失结果 B.风险事故 C.风险发生的范围 D.诱发风险的原因 3.( D )是管理利率风险、汇率风险、股票风险和商品风险非常有效的方法。 A.风险分散 B.风险转移 C.风险规避 D.风险对冲 4.我国要求资本充足率不得低于(C) % % % % 5.经风险调整的资本收益率(RAROC)的计算公式是(A) =(税后净利润-预期损失)∕非预期损失 =(税后净利润-非预期损失)∕预期损失 =(非预期损失-预期损失)∕税后净利润 =(预期损失-非预期损失)∕税后净利润 6.( C )是商业银行的最高风险管理/决策机构,承担商业银行风险管理的最终责任。 A.监事会 B.高级管理层 C.董事会 D.风险管理部门 7.商业银行风险识别最基本、最常用的方法是( A )。 A.制作风险清单 B.情景分析法 C.专家预测法 D.录制清单法 8.下列属于商业银行风险管理战略的内容的是( B )。 A.战略目标和战略方式 B.战略目标和实现路径 C.战略目标和实现方式 D.战略目标和战略管理 9.(C)是全面风险管理、资本监管和经济资本配置得以有效实施的重要基础。 A.风险识别 B.风险控制 C.风险计量 D.风险监测与报告 10. ( C )是对经过识别和计量的风险采取分散、对冲、转移、规避和补偿等措施,进行有效 管理和控制的过程。 A.风险监测 B.风险计量 C.风险控制 D.风险识别 11. 客户信用评级中,违约概率的估计包括(A)两个层面。 A.单一借款人的违约概率和某一信用等级所有借款人的违约概率 B.单一借款的违约概率和该借款人所有债项的违约概率 C.某一信用等级所有借款人的违约概率和这些借款人所有债项的违约概率 D.单一借款人的违约频率和某一信用等级所借款人的违约频率 12.下列关于客户信用评级的说法,不正确的是(D)。 A.评价主体是商业银行 B.评从目标是客户违约风险. C.评价结果是信用等级和违约概率 D.评价内容是客户违约后特定债项损失的大小 13.根据对商业银行内部评级法依赖程度的不同,内部评级法分为初级法和高级法两种。 对于非零售暴露,两种评级法都必须估计的风险因素是(A)。 A.违约概率B.违约损失率C.违约风险暴露D.期限

质量风险评估管理规程

1.目的: 为保证公司能适当的应对质量风险,提高质量风险应对的效率和效果,增强行动 的合理性,有效的配置资源——利用有限的资源,最大化的减小风险,特制定本 规程。本规程规定了药品质量风险管理的原理、过程、工具及其应用范围。 2.范围: 本标准适用于公司所有药品生命周期内的质量风险管理的全过程。 3.责任: 质管科长、质量受权人、公司各相关职能部门对本规程的实施负责。

4.内容: 4.1 质量风险管理流程图 不 可 接 受沟通

4.1术语: ◆质量风险管理:是在整个产品生命周期中采用前瞻或回顾的方式,对质量风险进行评估、控制、沟通、审核的系统程序。 ◆产品生命周期:产品从最初的研发、上市直至退市的所有阶段。 4.2 风险管理的组织机构和职责 ◆风险管理通常需要组建一个多学科的团队开展,鉴于风险管理的这一特殊要求,为了更好的开展风险管理活动,应成立风险管理的相应评估小组。 ◆风险管理项目组长由总经理指定,负责组建风险管理项目小组,领导开展具体的风险管理项目。 ◆风险管理项目成员由风险管理项目组长挑选相应人员组成5-7人的团队,团队成员必须包括QA人员,必要时还应包括岗位操作人员,负责开展具体的风险管理项目活动。 ◆公司管理层确保质量风险管理程序的正常运行;协调跨职能跨部门的质量风险管理程序;支持团队合作。 ◆质量受权人任风险管理项目组长,根据风险评估小组的总评意见,批准风险评估报告。 4.3 风险管理实施的内容 ◆有关术语 ●风险:不确定因素对目标的影响,通常表现为危害发生的可能性及其危害程度的综合体。 ●风险管理:即系统的应用管理方针、程序实现对目标任务的风险分析、评价和控制。 ●药品质量风险管理(QRM):是指企业实现确定目标的过程中(进行产品研发、生产、销售和使用等生命周期环节),系统科学地将各种不确

新版GMP之高质量风险管理系统规程

质量风险管理规程 1 目的 建立质量风险管理规程,规范产品生命周期中质量风险的评估、控制、沟通、审核的操作行为,降低产品的质量风险。 2 范围 适用于整个产品生命周期中所有存在风险、需要风险管理的情形。 3 责任 生产、质量管理人员及所有相关人员。 4.标准 4-1质量风险管理(QRM)是整个产品生命周期中就药品的质量风险进行评估、控制、沟通和审核的系统过程。QRM的运用领域如下: (1)文件记录:文件到期修订或药政法规更新,需要较大规模的修订文件记录时,确定修订的范围和深度; (2)质量缺陷:确定可疑的质量缺陷、投诉趋势、不合格品、退回、偏差、OOS,以及产品、洁净室(区)环境、工艺用水等定期回顾中不良趋势对质量的潜在影响; (3)审计/自检:发现潜在的风险范围,制定外部审计/内部自检的范围和深度; (4)变更:分析变更产生的风险; (5)厂房和设备:合理化厂房和设备的设计、安装和使用,设立适当的校正和维护保养计划; (6)确认和验证:确定验证的范围和程度,以及验证后取样、监测、再验证、关键工艺参数等; (7)产品研发:评价是否需要进行与扩大生产和技术转移的附加研究;(8)其他存在风险需要风险管理的情形。 4-2质量风险管理流程

4-2-1启动质量风险管理程序 出现4-1所列的需要进行风险管理的事件后,事件责任部门即报告质量部,经确认后,质量部指定风险管理小组组长及小组成员,即启动本程序。小组成员至少应包括风险事件责任部门负责人及QA 人员,并根据需要也可邀请其他相关部门的专业人员参加。由QA 对风险事件进行编号,编号方式为:QRA YY- XX ,YY 为两位年号,XX 为两位年度流水号,并发放编号后的《质量风险评估表》(见附页)到风险管理小组,同时在《质量风险项目台账》(见附页)上进行登记。 4-2-2风险评估:风险管理程序启动后,即对潜在的危害源进行识别,对接触这些危害源造成的风险进行分析与评价。 4-2-2-1风险识别

美国FDA分析方法验证指南中英文对照

I. INTRODUCTION This guidance provides recommendations to applicants on submitting analytical procedures, validation data, and samples to support the documentation of the identity, strength, quality, purity, and potency of drug substances and drug products. 1. 绪论 本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用于支持原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。 This guidance is intended to assist applicants in assembling information, submitting samples, and presenting data to support analytical methodologies. The recommendations apply to drug substances and drug products covered in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), product license applications (PLAs), and supplements to these applications. 本指南旨在帮助申请者收集资料,递交样品并资料以支持分析方法。这些建议适用于NDA,ANDA,BLA,PLA及其它们的补充中所涉及的原料药和制剂。 The principles also apply to drug substances and drug products covered in Type II drug master files (DMFs). If a different approach is chosen, the applicant is encouraged to discuss the matter in advance with the center with product jurisdiction to prevent the expenditure of resources on preparing a submission that may later be determined to be unacceptable. 这些原则同样适用于二类DMF所涉及的原料药和制剂。如果使用了其它方法,鼓励申请者事先和FDA药品评审中心的官员进行讨论,以免出现这种情况,那就是花了人力物力所准备起来的递交资料后来发现是不可用的。 The principles of methods validation described in this guidance apply to all types of analytical procedures. However, the specific recommendations in this guidance may not be applicable to certain unique analytical procedures for products such as biological, biotechnological, botanical, or radiopharmaceutical drugs. 本指南中所述的分析方法验证的原则适用于各种类型的分析方法。但是,本指南中特定的建议可能不适用于有些产品所用的特殊分析方法,如生物药,生物技术药,植物药或放射性药物等。 For example, many bioassays are based on animal challenge models, 39 immunogenicity assessments, or other immunoassays that have unique features that should be considered when submitting analytical procedure and methods validation information. 比如说,许多生物分析是建立在动物挑战模式,免疫原性评估或其它有着独特特性的免疫分析基础上的,在递交分析方法和分析方法验证资料时需考虑这些独特的性质。Furthermore, specific recommendations for biological and immunochemical tests that may be necessary for characterization and quality control of many drug substances and drug products are beyond the scope of this guidance document. 而且,许多原料药和制剂的界定和质量控制所需的生物和免疫化学检测并不在本指南的范围之内。 Although this guidance does not specifically address the submission of analytical procedures and validation data for raw materials, intermediates, excipients, container closure components, and other materials used in the production of drug

药品经营的质量风险管理规程正式版

Guide operators to deal with the process of things, and require them to be familiar with the details of safety technology and be able to complete things after special training.药品经营的质量风险管理 规程正式版

药品经营的质量风险管理规程正式版 下载提示:此操作规程资料适用于指导操作人员处理某件事情的流程和主要的行动方向,并要求参加施工的人员,熟知本工种的安全技术细节和经过专门训练,合格的情况下完成列表中的每个操作事项。文档可以直接使用,也可根据实际需要修订后使用。 一、目的:为规范各经营环节质量风险的评估与控制管理。 二、依据:《药品经营质量管理规范》。 三、范围:适用于质量管理体系中所有经营环节质量风险的管理。 四、职责:质量负责人对本规程的实施负责。 五、内容: 1、成立风险管理小组 公司质量领导小组全面负责公司药品经营质量风险管理工作,由质量负责人主

持质量风险管理的日常工作,成员包括各部门经理。质量管理部负责组织各部门对各经营环节质量风险的识别、评估与控制。 2、风险识别 各部门负责人依据GSP相关要求,结合部门经营环节的有关风险,对可能存在的质量风险因素进行收集整理,填写《质量风险排查表》,报风险管理小组。 3、风险评估 风险管理小组召集与风险相关的部门、人员,依据《质量风险排查表》,对已经被识别的风险进行分析、评价,确定风险等级,确认出现问题的可能性、可识别性以及后果的严重性等,由质量管理部汇总

SOP01质量风险管理操作规程

质量风险管理操作规程 1.目的:采取有效可行的分析控制方法,最大限度地降低产品质量风险。 2.范围:原辅材料采购、生产、销售的全过程。 3.责任:采购、生产、销售全体高层管理人员,特别是质量管理部门、生产管理部门对执行和实施本规程负有组织检查责任。 4.内容: 4.1风险评估使用的工具:风险排列和过滤 (1)矩阵图:

(2)RRF列表: 4.2风险分析 4.2.1供应商风险评估 4.2.1.1供应商质量风险评估 物料供应商具备不具备供应资质是关键,没有合法的生产经营条件,很难保证其所供物料质量,同时其可追溯性更差,其人员素质、质量保证系统的完善性、售后服务等存在一定风险,现利用风险排列和过滤(RRF列表)的风险评估工具对供应商存在的风险进行评估。 供应商质量风险评估表: 通过对供应商的风险评估,其最大风险为质量保证系统的完善性,为中等风险,能通过供应商现场考察、走访等方法督促其改善或更换体系完善的供应商,进而解决,可以使风险控制在可接受的范围。 4.2.1.2供应商风险等级评估

针对原药材、辅料、直接接触药品的内包材供应商进行等级评估,确定存在风险最大的供应商,采取必要的管理办法。 供应商风险等级评估表: 通过对供应商风险等级评估,显示中药材风险最大,对中药材严格按质量标准采购,采购药材按规定全检,不合格的严禁使用。 4.2.2物料储存风险评估 储存条件的变化可以引起物料成分的变化;虫害、鼠害防范措施不到位,容易污染引起物料质量的变化;温湿度的变化超过范围容易引起霉变、虫蛀等现象;特殊物料的储存,管理不完善容易引起混淆,现利用风险排列和过滤(RRF列表)的风险评估工具对供应商存在的风险进行评估。 供应商风险等级评估表: 通过对物料储存的风险评估,显示仓储色标管理、毒性中药材储存风险最大,色标管理是避免不符合规定物料下转的有效措施,对毒性中药材严格按专库储存、双人双锁管理、色标标示管理、特有标示管理,发放使用双人称量、双人复核管理,使其风险降至可以接受程度。

质量风险标准管理规程

的质量问题并采取处理措施或当产品出现质量问题时改善处理问题的决策过程,确保产品整个生命周期中的质量,以保证产品质量和增强公司处理潜在风险的能力。 应用范围:适用于药品质量各方面,包括药品生命周期中的研发、注册/评审、生产、检验、放行、销售等过程。 责任人:质量受权人负责本规程的批准;质量保证部负责本规程的起草、修订、审核、培训、实施和监督;生产技术部、设备动力部、营销部负责本规程的执行。 内容 1 定义 1.1 产品生命周期:产品从最初的研发到销售,直至最终停产的所有阶段。 1.2 危害源:产生危害的潜在来源。 1.3 风险:危害发生的可能性及其严重程度。 1.4 决策者:有能力和职权在质量风险管理中做出适当和及时决策的人。 1.5 风险评估:规定风险评估过程和程序,概述风险识别、风险分析的过程,以确定风险等级和需制定应对措施的风险。 1.6 风险鉴定:根据风险提问或问题的描述,系统地使用信息来鉴定潜在危害源。 1.7 风险分析:和被确定的危害源有关的风险的分析。 1.8 风险评价:用定性或定量的方法,将被评估的风险与既定的风险标准进行比较,以确定风险的显著性。 1.9 风险控制:实施风险管理决策的行为。 1.10 风险降低:采取措施减少危害发生的可能性和严重程度。 1.11 风险认可:接受风险的决策。 1.12 质量风险管理:贯穿产品生命周期的药品质量风险的评估、控制、交流及回顾的系统化过程。 2 程序 2.1 质量风险管理的原则: 质量风险评估的最终目的在于保护患者的利益。质量风险管理是一种以科学为基础,并且切合实际的决策过程,其严密和正规程度与涉及问题的复杂性和关键性相适应。 2.1.1 组织及人员: 质量风险管理工作小组的成员包括风险管理涉及的相关部门的负责人和专业人员以及公司领导。此外,还可以包括外请相关领域的专家(例如:研发、工程、生产、销售等)。由启动质量风险评估事件的部门负责人决定小组成员,决策者为质量受权人,风险评估涉及重大的财产投资时,决策者为公司总经理。 2.1.2 基本过程:风险管理流程分为五个部分:风险评估、风险控制、风险交流、风险评审和风险回顾。 2.2 风险管理流程 2.2.1 风险评估:

201507fda行业指南:分析方法验证(中英文)(下).doc

201507 FDA行业指南:分析方法验证(中英文)(下) VII. STATISTICAL ANALYSIS AND MODELS 统计学分析和模型 A. Statistics 统计学 Statistical analysis of validation data can be used to evaluate validation characteristics against predetermined acceptance criteria. All statistical procedures and parameters used in the analysis of the data should be based on sound principles and appropriate for the intended evaluation. Several statistical methods are useful for assessing validation characteristics, for example, an analysis of variance (ANOVA) to assess regression analysis R (correlation coefficient) and R squared (coefficient of determination) or linear regression to measure linearity. Many statistical methods used for assessing validation characteristics rely on population normality, and it is important to determine whether or not to reject this assumption. There are many techniques, such as histograms, normality tests, and probability plots that can be used to evaluate the observed distribution. It may be appropriate to transform the data to better fit the normal distribution or apply distribution-free (nonparametric) approaches when the observed data are

药品质量风险管理制度(1)

1目的 建立质量风险管理制度,规范药品生命周期中质量风险的评估、控制与审核操作行为,降低产品的质量风险。 2适用范围 适用于公司所生产药品其质量风险的评估、控制与审核的管理 3术语或定义 质量风险:是一个系统化的过程,是对产品在整个生命周期过程中,对风险的识别、衡量、控制以及评价的过程。质量风险管理:是对药品整个生命周期进行质量风险的识别、评估、控制、沟通、回顾的系统过程,运用时可采用前瞻或回顾的方式。 4职责 质量部:负责组织进行质量风险评估、控制与审核协调、管理等相关事宜。 职能部门:对本规程的实施负责。 5安全注意事项 不适用于本文 6规程 6.1风险管理的内容 6.1.1风险管理包括的内容有风险评估、风险控制、风险沟通和审核等程序,持续地贯穿 于整个产品生命周期。 6.1.2风险评估是风险管理过程的第一步,它包括风险识别,风险分析和风险评价三个部分 即解决三个基本问题:(1)将会出现的问题是什么(2)可能性有多大(3)问题发生的后果是什么 6.1.3风险控制的目的就是将风险降低到可接受的水平。重点归纳为:(1)风险是否在可以 被接受的水平上(2)可以采取什么样的措施来降低、控制或消除风险(3)在控制已经识别的风险时是否会产生新的风险 6.1.4风险沟通:通过风险沟通,能够促进风险管理的实施,使各方掌握更全面的信息从 而调整或改进措施及其效果。 6.1.5风险审核:在风险管理流程的最后阶段,应该对风险管理程序的结果进行审核,尤 其是对那些可能会影响到原先质量管理决策的事件进行审核。 6.2风险管理程序 6.2.1风险管理的启动 6.2.1.1确定问题和/或有关风险的疑问,包括确认风险可能性的相关假设; 6.2.1.2 风险管理小组负责召集与风险相关的部门或专家,收集与所评估的风险相关的可 能性危险、危害或对人体健康的影响的有关背景资料和数据。 6.2.1.3 根据存在的主要风险的性质确定风险管理的组长和必要的资源。 6.2.1.4 确定如何使用这些信息,评估和结论; 6.2.1.5 根据具体的问题,由风险管理的组长负责组织建立风险管理流程,包括详细的时 间计划。 6.2.2风险评估:首先系统地利用各种信息和经验来确认工艺、设备、系统、操作等过程 中存在的风险,指出将会出现的问题在哪里。包括识别可能的后果,为进一步质量风险管理进程的其它步骤提供基础;其次对已经被识别的风险及其问题进行分析,这需要相当有经验的技术人员以及QA相关人员共同完成,通过分析确认将会出现问题的可能性有多大,

质量风险管理操作规程

质量风险管理操作规程 ****药材公司 1.目的:制定公司质量风险操作规程,在公司经营过程中按风险管理预防降低各种质量风险,减少质量风险带来的损失,确保公司经营的持续、稳定、安全运行,保障公司各项业务的正常开展 2.依据: 根据《药品管理法》、新版GSP及其实施细则等相关法律法规规章制定。 3.范围:适用于药品经营质量风险的管理。 4.职责: 4.1、公司全体员工执行风险管理措施。 4.2、质量管理部负责风险管理的日常监督。 4.3、质量风险管理小组负责制定质量风险管理标准、应对计划。 5.内容 5.1、质管部对公司全体员工培训质量风险管理制度、风险操作规程、年度质量风险管理报告、风险清单(质量风险评估标准和应对计划)。公司每一位员工应具有药品质量风险意识。 5.2、每年11月各部门经理组织部门员工采用回顾式和前瞻式的方法,在部门内开展质量风险管理讨论,对每个工作环节的质量风险进行识别、分析、评估。各部门经理在11月30日前将年度质量风险控制情况、下一年度必须继续加强管理的风险控制点,不必控制的风险点、新的可能发生的风险点及防范措施清单上报公司质管部。 5.3、每年12月上旬质管部检查各部门执行风险应对计划情况、收集各部门工作环节的风险项目清单,在此基础上整理提出质量评估报告草案、质量风险预案草案,提交质量风险管理小组。 5.4、风险审核、回顾:质量风险管理小组每年12月召开质量风险评估会议,采用回顾式和前瞻式的方法,对年度质量风险管理工作做出总结--风险评估报告,对下一年度质量风险预案清单进行讨论、分析、对质量风险控制效果进行评价和改进,制定出下一年度质量风险应对计划清单。 5.5、风险控制、风险沟通:各部门经理将公司质量风险小组审议通过的质量

药品质量风险控制管理规程

目的:建立系统的质量风险分析控制管理制度。 范围:适用于公司内进行的药品生产整个生命周期的GMP管理 责任: 内容: 1 定义: 风险:药品风险是与药品有关的、危及人体健康和生命安全的危险。一般是指危害出现的可能性和危害严重性的结合。 质量风险管理:在整个药品生命周期中采用前瞻或回顾的方式,对质量风险进行评估、控制、沟通、审核的系统过程。 2 质量风险管理方针、目的和范围: 2.1 质量风险管理方针:药品质量源于设计、源于生产。 质量风险评估以科学知识为基础,最终目的在于保护患者的利益。 质量风险管理程序实施的力度、形式和文件的要求科学合理,并与风险的程度相匹配 2.2 质量风险管理的目的:按照一个完整有效的风险管理流程,使风险发生的可能性和危害降低到可接受的程度或者提高风险发生的可预测性。 2.3质量风险管理方针适用范围:适用于药品生产整个生命周期的各个环节,主要包括可能直接影响到产品质量的产品研发、物料管理、设备设施管理、生产管理、质量管理等各个方面。 3 人员职责: 3.1风险管理总负责人:为质量部经理,负责为风险管理提供足够的资源支持,并审核批准风险管理计划。 3.2 风险管理小组成员: 3.2.1小组组长,负责组织相关人员对相关类别的风险进行风险管理,并及时与相关的其他部门人员做好风险沟通并制定风险管理计划。 3.2.2 其他小组成员为风险管理提供足够的协助,并参与对风险的评估过程。必要时聘请外部专家进行评估分析。 3.2.3 除参加对风险的评估外,质量保证科还负责对制定的风险管理计划的实施进行监督管理以及对风险管理的回顾。 3 风险管理流程(风险管理流程图见附件一): 3.1 确定事件并启动风险管理流程:需要考虑的风险点包括对患者的风险;产品不符合标准的风险;法规不符合性风险等。 3.2 风险评估:包括风险识别、风险分析和风险评估。由各风险小组组长组织成立风险评估小组,对涉及到的各方面风险进行分析评估,确定优先顺序。(风险管理组织机构图见附件二)在进行风险评估时,需要提问如下三个基本问题: a、将会出现的问题是什么?——风险识别 b、 c、问题发生的后果是什么?——风险分析

风险管理制度及操作规程

大地纺织纺织有限公司 安全会议管理制度 1.为贯彻“安全第一,预防为主”的方针,及时传达上级安全指示精神,计划、布置、总结、评比我公司安全工作,制定本制度。 2.本制度适用于寿光大地纺织有限公司、潍坊公司。 3.公司安委会的会议具体分为年度会议、半年会议和月会议,会议召开时间根据实际情况而定。 4.安委会举行会议时,在家公司级领导都要参加会议,听取安全工作汇报,研究布署安全生产工作。各车间、各部门的第一负责人均要参加,如无特殊情况,不得缺席。 5. 安委会会议召开内容分为: 5.1 传达、贯彻中央及地方上级部门的指示精神、文件及最新的劳动安全卫生法 令、法规。 5.2 审定、分解考核企业年度安全生产经营目标管理责任书、安全技术措施计划, 听取安全部门的工作汇报、情况反映和工作总结。 5.3 研究解决重大隐患,且在会上提出对策,拟定解决的办法及防范措施,协调 各部门在隐患处理过程中的分工和协作,指定隐患整改的具体负责人及隐患整改的要求和期限。 5.4 通报他人事故教训;审议企业的重大事故,审定并通过对重大 事故的处理决定和通报。 5.5 公司级领导布置全公司性安全生产活动或作安全工作报告。 6. 公司安委会会议必须有会议时间、会议地点、会议内容、参加人员等方面的

完整的原始记录。 安全管理制度 安全生产检查制度 1. 为保障企业生产安全,减少事故隐患,促进各车间、部门的安全管理工作, 制定本制度。 2. 本制度适用于寿光大地纺织有限公司、潍坊公司。 3. 企业实行车间(部门)或班组自检、“青安团岗”互检、综合办人员专检的三级安全生产检查制度。 4. 每月28日为我公司安全活动日,各车间、部门要对各自责任区内的安全工作进行认真检查。公司部由综合办组织有关人员对各车间、部门进行检查考核,着重检查企业的重点部位,安全规章制度落实情况;制止“三违”行为,发现和查明事故隐患,并将所查隐患以隐患整改指令书形式及时反馈给有关部门、车间,督促其按时按质整改。 5.综合办专职人员不定期地对各车间、部门进行安全专检,发现问题及时下达隐患整改指令书,限期整改,并进行跟踪考核和验收;责任部门必须按时按质将隐患整改情况以书面形式反馈至综合办。 6. 班组互检工作由公司生产办自行组织安排,每月互检应不少于一次,并按规定填好“互检表”。对所查出的隐患,公司团委应逐条进行跟踪考核,直至整改完毕并将“互检表”反馈给综合办。 7. 各车间、部门每周对本单位安全生产的自检不少于一次,并对所查隐患自行整改;应按规定填好“自检表”,综合办派人不定期到各车间、部门抽查,检

药品质量风险管理规程

药品质量风险管理规程 药品质量风险管理规程目的: 建立质量风险管理制度,规范药品生命周期中质量风险的评估、控 制与审核操作行为,降低产品的质量风险。 适用范围: 适用于公司生产管理和质量管理。 职责: 质量受权人和质量部;负责质量风险管理计划的起草、监督实施和 考核; 生产部;负责确保产品质量风险最小化措施的有效实施; 内容: 1、定义 1.1、风险:指不确定性因素对目标的影响,通常表现为出现危害的可能性和严重性的综合结果。 1.2、质量风险:导致药品达不到预期的质量、安全性和有效性以及生产过程出现不符合法规要求的各种因素的组合。其三要素分别是:可能性、可检测性、严重性。 1.3、质量风险管理:是药品整个生命周期中采用前瞻或回顾的方式,对质量风险进行评估、控制、沟通、审核的系统过程。 1.4、药品整个生命周期:包括从药品研发到技术转移、工业生产直至产品终止的全过程。 1.5、可测定性:发现或测定危险源存在的能力。 1.6、危害:对健康的伤害,包括产品质量缺陷或可获得性造成的伤害。 1.7、危险源:潜在的危害来源。 1.8、可能性:有害事件发生的频率或可能性。 1.9、严重性:对危险源可能造成的后果的衡量。

2、风险评估实施的范围、方式 2.1、目前本公司将开展风险管理的领域及风险管理方法、工具见下表: 开展领域风险管理方法、工具厂房、新设备的URS 失效模式效果分析、矩阵供应商审计调查表(见供应商管理标准) 关键物料失效模式效果分析 质量标准、工艺关键点危害分析和关键控制点(HCAAP)、流程图发运、收回和退回重新发失效模式效果分析 货 返工失效模式效果分析 根据偏差和变更具体内容选择此表中的风险管理方偏差、变更法、工具 投诉失效模式效果分析 质量回顾趋势图、控制图 验证失效模式效果分析、流程图、矩阵、鱼骨图检验异常(OOS) 调查表(见检验异常处理管理标准) 2.2、实施方式: 2.2.1、采用分散式风险评估管理方法,即各类风险所在领域,在处理流程中设置,对于风险处理流程成熟的领域,风险评估可不按本标准流程建立风险管理组织、进行风险管理。可按各自管理标准中规定的流程分析、管理执行。如供应商审计,可按供应商管理标准中规定执行。检验异常可按检验异常处理管理标准中规定执行。 2.2.2、对于一些风险领域已有明确分级的,如偏差、变更、投诉等,对于分级级别高的(一级),存在质量风险较大,应按本管理标准流程,建立风险管理组织、使用风险管理工具,进行系统性的分析,以规避其存在的风险,避免出现产品质量。

药品质量风险管理规程

药品质量风险管理规程 1、目的 建立药品质量风险管理程序,对可能影响到药品质量的因素进行评估和控制,保证产品质量,规避质量事故或药害事件的发生,保护患者利益。 1?范围 适用于在公司质量体系内的药品质疑风险管理,它适用于药品的整个生命周期。 3.责任 与风险管理相关部门人员负责实施本规程;风险管理小组负责质量风险管理过程的评估、控制和追踪检査;风险管理总负责人负责本程序的启动与关闭,对风险管理过程和结果进行最终审批。 4.引用标准及文件 《药品生产质量管理规范》(2010年版)、EU GMP指南Volume 4和ICH Q9。 5.内容 5.1定义 5. 1.1药品风险:与药品有关的、危及人体健康和生命安全的危险。一般是指危害出现的可能性和严重性的结合。 5. 1.2质量风险管理:在整个产品生命周期中采用前瞻或回顾的方式,对质量风险进行评估、控制、沟通、审核的系统过程。质量风险管理过程所采用的方法、措施、形式及形成的文件应当与存在风险的级别相适应。 5.2质量风险管理的组织机构 5. 2. 1公司成立质量风险管理小组,专门负责质量风险管理工作。 5. 2.2公司质量风险管理小组总负责人为质量受权人QP,负责为风险管理提供足够的资源,审批风险管理活动的启动与关闭,评估风险管理过程,回顾风险管理情况。 5.2.3备类风险小组组长一般为风险所在部门或风险控制部门的负责人。组长负责组织本组成员对相关类别的风险进行管理,及时与相关部门做好风险沟通,向本组成员讲解整个风险项目,制左风险管理il?划,组织实施风险评估和风险控制。

页脚内冷5

5?2?4并类风险小组成员至少包括风险发起人、QA人员以及其它相关部门的专业人员.必要时聘请外部专家进行风险评估。组员负责搜集潜在的与质量风险相关的资料和数据,参与风险评估和风险控制过程,并对风险控制措施的实施过程进行追踪检查。 5. 3质量风险管理流程 建立质址风险 倉理流程的结果/输 风P 事件回顾审核K 启动质量风险管理 风也 控借 风险降低 风险接受

质量风险管理操作规程

【最新资料,WORD文档,可编辑修改】 质量风险管理操作规程 文件题目质量风险管理操作规程文件编码SOP01(03)-0001 制定(变更)原 实施《药品生产质量管理规范(2010年修订)》因及目的 起草(修订)人日期年月日版本号0审核人日期年月日页数共9页 批准人日期年月日文件类型操作标准生效日期年月日颁发部门质量管理部分发部门质量管理部、生产技术部、业务部、生产车间、化验室 1.目的:采取有效可行的分析控制方法,最大限度地降低产品质量风险。 2.范围:原辅材料采购、生产、销售的全过程。 3.责任:采购、生产、销售全体高层管理人员,特别是质量管理部门、生产管理部门 对执行和实施本规程负有组织检查责任。 4.内容:

风险评估使用的工具:风险排列和过滤 (1)矩阵图: 风险评估的矩阵图 风险评估评分表 (2)RRF 列表: 严重性 可能性

风险分析 供应商风险评估 供应商质量风险评估 物料供应商具备不具备供应资质是关键,没有合法的生产经营条件,很难保证其所供物料质量,同时其可追溯性更差,其人员素质、质量保证系统的完善性、售后服务等存在一定风险,现利用风险排列和过滤(RRF列表)的风险评估工具对供应商存在的风险进行评估。 供应商质量风险评估表: 通过对供应商的风险评估,其最大风险为质量保证系统的完善性,为中等风险,能通过供应商现场考察、走访等方法督促其改善或更换体系完善的供应商,进而解决,可以使风险控制在可接受的范围。

供应商风险等级评估 针对原药材、辅料、直接接触药品的内包材供应商进行等级评估,确定存在风险最大的供应商,采取必要的管理办法。 供应商风险等级评估表: 通过对供应商风险等级评估,显示中药材风险最大,对中药材严格按质量标准采购,采购药材按规定全检,不合格的严禁使用。 物料储存风险评估 储存条件的变化可以引起物料成分的变化;虫害、鼠害防范措施不到位,容易污染引起物料质量的变化;温湿度的变化超过范围容易引起霉变、虫蛀等现象;特殊物料的储存,管理不完善容易引起混淆,现利用风险排列和过滤(RRF列表)的风险评估工具对供应商存在的风险进行评估。 供应商风险等级评估表:

美国FDA分析方法验证指南中英文对照

美国FDA分析方法验证指南中英文对照 美国FDA分析方法验证指南中英文对 照 I. INTRODUCTION This guidance provides recommendations to applicants on submitting analytical procedures, validation data, and samples to support the documentation of the identity, strength, quality, purity, and potency of drug substances and drug products. 1. 绪论 本指南旨在为申请者提供建议,以帮助其提交分析方法,方法验证资料和样品用 于支持原料药和制剂的认定,剂量,质量,纯度和效力方面的文件。 This guidance is intended to assist applicants in assembling information, submitting samples, and presenting data to support analytical methodologies. The recommendations apply to drug substances and drug products covered in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), product license applications (PLAs), and supplements to these applications. 本指南旨在帮助申请者收集资料,递交样品并资料以支持分析方法。这些建议适 用于NDA,ANDA,BLA,PLA及其它们的补充中所涉及的原料药和制剂。 The principles also apply to drug substances and drug products covered in Type II drug master files (DMFs). If a different approach is chosen, the applicant is encouraged to discuss the matter in advance with

风险辨识管控作业指导书

安全风险辨识管控作业指导书 1.目的 为加强安全管理,消除或减少危害,有效控制重大危害因素,降低安全风险,特制定本制度。 2.适用范围 适用于公司的所有部门、分厂的所有活动,包括生产活动、装臵、设备设施、原料产品、安全防护、正常和异常活动、人为因素、违反规程、规章等。 3.定义: 危害:可能造成人员伤亡、疾病、财产损失、工作环境破坏的根源或状态。 危害识别:认知危害的存在并确定其特征的过程。 风险:风险是发生特定危害事件的可能性及后果的结合。 风险评价: 依据已确定的评价方法、评价准则,定期进行评价风险程度并确定风险是否可容忍的全过程。 事故:造成死亡、职业病、财产损失、环境破坏的事件。 事件:导致或可能导致事故的事情。 4.职责: 风险管理领导小组 公司成立风险管理领导小组,负责制定公司安全生产风险管理作业指导书,全面推行开展安全生产危害辨识工作并指导、审查、批准;负责审查重大风险改进措施方案 安全环保部 安全环保部负责组织危害识别和风险评价工作,指导各单位进行危害识别和风险评价工作,组织重大风险改进措施方案的审查。 各单位确保本单位危害识别和风险评价的人员有足够的培训,组织制定本单位的安全生产风险管理作业指导书,组织本单位工作范围的危害识别和风险评价工作,在日常生产工作中不间断进行危害识别和风险评价工作,编制本单位的危害/风险记录,开展隐患治理工作。 5.危害识别及风险评价程序

成立评价组 公司成立安全生产风险评价组,以公司分管安全生产领导为组长、生产、安全、设备、环境、消防部门的管理人员参加,编制本公司的安全生产风险管理作业指导书和表格。全面开展安全生产危害辨识、风险评价工作,指导、审查、批准。各部门应成立以厂长(部长)为组长、工艺、设备、安全管理人员和操作人员参加的安全生产风险评价小组,所有评价人员需经过专门培训并有能力、资格开展职业安全健康环境的危害识别和风险评价,同时各部门也应鼓励其他员工参与危害和影响的确定。 评价依据《公司安全生产管理制度》、相关安全生产法律法规和技术标准、各单位安全管理制度、操作规程等。 选择和确定评价范围及对象 评估小组应首先识别出本单位风险评价的范围,应包括以下内容: ——规划、设计和建设、投产、运行等阶段; ——常规和异常活动; ——事故及潜在的紧急情况; ——所有进入作业场所的人员的活动; ——原材料、产品的运输和使用过程; ——作业场所的设施、设备、车辆、安全防护用品; ——人为因素,包括违反操作规程和安全生产规章制度; ——丢弃、废弃、拆除与处臵; ——气候、地震及其他自然灾害。 在确定评价范围后,评价小组可按下列方法,确定评价对象: ——按生产流程的各阶段; ——按地理区域或部门; ——按装臵、设备、设施; ——按作业任务。 对所确定的评价对象,可按作业活动进一步细分,以便对危害和环境影响进行全面识别和评价。同时应对工作活动的相关信息有所了解,包括: ——所执行任务的期限、人员及实施任务的频率;

相关文档
相关文档 最新文档