文档库 最新最全的文档下载
当前位置:文档库 › FDA最新指导原则:药物分析程序及方法验证指导原则

FDA最新指导原则:药物分析程序及方法验证指导原则

FDA最新指导原则:药物分析程序及方法验证指导原则
FDA最新指导原则:药物分析程序及方法验证指导原则

Analytical Procedures and Methods Validation for Drugs

and Biologics

DRAFT GUIDANCE

This guidance document is being distributed for comment purposes only. Comments and suggestions regarding this draft document should be submitted within 90 days of publication in the Federal Register of the notice announcing the availability of the draft guidance. Submit electronic comments to https://www.wendangku.net/doc/7a13924693.html,. Submit written 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 Registe r.

For questions regarding this draft document contact (CDER) Lucinda Buhse 314-539-2134, or (CBER) Office of Communication, Outreach and Development at 800-835-4709 or 301-827-1800.

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 2014

CMC

Analytical Procedures and Methods Validation for Drugs

and Biologics

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/7a13924693.html,

https://www.wendangku.net/doc/7a13924693.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

ocod@https://www.wendangku.net/doc/7a13924693.html,

https://www.wendangku.net/doc/7a13924693.html,/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htm

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

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)

Febr uary 2014

CMC

TABLE OF CONTENTS

I.INTRODUCTION (1)

II.BACKGROUND (2)

III.ANALYTICAL METHODS DEVELOPMENT (3)

IV.CONTENT OF ANALYTICAL PROCEDURES (3)

A.Principle/Scope (4)

B.Apparatus/Equipment (4)

C.Operating Parameters (4)

D.Reagents/Standards (4)

E.Sample Preparation (4)

F.Standards Control Solution Preparation (5)

G.Procedure (5)

H.System Suitability (5)

I.Calculations (5)

J.Data Reporting (5)

V.REFERENCE STANDARDS AND MATERIALS (6)

VI.ANALYTICAL METHOD VALIDATION FOR NDA, ANDAs, BLAs, AND DMFs (6)

A.Noncompendial Analytical Procedures (6)

B.Validation Characteristics (7)

https://www.wendangku.net/doc/7a13924693.html,pendial Analytical Procedures (8)

VII.STATISTICAL ANALYSIS AND MODELS (8)

A.Statistics (8)

B.Models (8)

VIII.LIFE CYCLE MANAGEMENT OF ANALYTICAL PROCEDURES (9)

A.Revalidation (9)

B.Analytical Method Comparability Studies (10)

1.Alternative Analytical Procedures (10)

2.Analytical Methods Transfer Studies (11)

C.Reporting Postmarketing Changes to an Approved NDA, ANDA, or BLA (11)

IX.FDA METHODS VERIFICATION (12)

X.REFERENCES (12)

Guidance for Industry1

1

Analytical Procedures and Methods Validation for Drugs and

2

Biologics

3

4

5

This draft guidance, when finalized, will represent the Food and Drug Administration’s (FDA’s) current 6

thinking on this topic. It does not create or confer any rights for or on any person and does not operate to 7

bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of 8

the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA

9

staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call 10

the appropriate number listed on the title page of this guidance.

11

12

13

14

I. INTRODUCTION

15

16

This revised draft guidance supersedes the 2000 draft guidance for industry on Analytical

17

Procedures and Methods Validation2,3 and, when finalized, will also replace the 1987 FDA

18

guidance for industry on Submitting Samples and Analytical Data for Methods Validation. It

19

provides recommendations on how you, the applicant, can submit analytical procedures4 and

20

methods validation data to support the documentation of the identity, strength, quality, purity,

21

and potency of drug substances and drug products.5It will help you assemble information and 22

present data to support your analytical methodologies. The recommendations apply to drug

23

substances and drug products covered in new drug applications (NDAs), abbreviated new drug 24

applications (ANDAs), biologics license applications (BLAs), and supplements to these

25

applications. The principles in this revised draft guidance also apply to drug substances and drug 26

products covered in Type II drug master files (DMFs).

27

28

This revised draft guidance complements the International Conference on Harmonisation (ICH) 29

guidance Q2(R1)Validation of Analytical Procedures: Text and Methodology(Q2(R1)) for

30

developing and validating analytical methods.

31

32

This revised draft guidance does not address investigational new drug application (IND) methods 33

validation, but sponsors preparing INDs should consider the recommendations in this guidance.

34

For INDs, sufficient information is required at each phase of an investigation to ensure proper

35

identity, quality, purity, strength, and/or potency. The amount of information on analytical

36

procedures and methods validation will vary with the phase of the investigation.6 For general

37

1 This guidance has been prepared by the Office of Pharmaceutical Science, in the Center for Drug Evaluation and

Research (CDER) and the Center for Biologics Evaluation and Research (CBER) at the Food and Drug

Administration.

2 Sample submission is described in section IX, FDA Methods Verification.

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

Drugs guidance Web page at

https://www.wendangku.net/doc/7a13924693.html,/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm.

4Analytical procedure is interchangeable with a method or test procedure.

5The terms drug substance and drug product, as used in this guidance, refer to human drugs and biologics.

6 See 21 CFR 312.23(a)(7).

guidance on analytical procedures and methods validation information to be submitted for phase 38

one studies, sponsors should refer to the FDA guidance for industry on Content and Format of

39

Investigational New Drug Applications (INDs) for Phase 1 Studies of Drugs, Including

40

Well-Characterized, Therapeutic, Biotechnology-Derived Products. General considerations for 41

analytical procedures and method validation (e.g., bioassay) before conduct of phase three

42

studies are discussed in the FDA guidance for industry on IND Meetings for Human Drugs and 43

Biologics, Chemistry, Manufacturing, and Controls Information.

44

45

This revised draft guidance does not address specific method validation recommendations for

46

biological and immunochemical assays for characterization and quality control of many drug

47

substances and drug products. For example, some bioassays are based on animal challenge

48

models, and immunogenicity assessments or other immunoassays have unique features that

49

should be considered during development and validation.

50

51

In addition, the need for revalidation of existing analytical methods may need to be considered 52

when the manufacturing process changes during the product’s life cycle. For questions on

53

appropriate validation approaches for analytical procedures or submission of information not

54

addressed in this guidance, you should consult with the appropriate FDA product quality review 55

staff.

56

57

If you choose a different approach than those recommended in this revised draft guidance, we

58

encourage you to discuss the matter with the appropriate FDA product quality review staff before 59

you submit your application.

60

61

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

62

responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and should 63

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

64

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

65

recommended, but not required.

66

67

68

II.BACKGROUND

69

70

Each NDA and ANDA must include the analytical procedures necessary to ensure the identity, 71

strength, quality, purity, and potency of the drug substance and drug product.7 Each BLA must 72

include a full description of the manufacturing methods, including analytical procedures that

73

demonstrate the manufactured product meets prescribed standards of identity, quality, safety,

74

purity, and potency.8 Data must be available to establish that the analytical procedures used in 75

testing meet proper standards of accuracy and reliability and are suitable for their intended

76

purpose.9 For BLAs and their supplements, the analytical procedures and their validation are

77

submitted as part of license applications or supplements and are evaluated by FDA quality

78

review groups.

79

80

7 See 21 CFR 314.50(d)(1) and 314.94(a)(9)(i).

8 See 21 CFR 601.2(a) and 601.2(c).

9 See 21 CFR 211.165(e) and 211.194(a)(2).

Analytical procedures and validation data should be submitted in the corresponding sections of 81

the application in the ICH M2 eCTD: Electronic Common Technical Document Specification.10

82

83

When an analytical procedure is approved/licensed as part of the NDA, ANDA, or BLA, it

84

becomes the FDA approved analytical procedure for the approved product. This analytical

85

procedure may originate from FDA recognized sources (e.g., a compendial procedure from the 86

United States Pharmacopeia/National Formulary (USP/NF)) or a validated procedure you

87

submitted that was determined to be acceptable by FDA. To apply an analytical method to a

88

different product, appropriate validation studies with the matrix of the new product should be

89

considered.

90

91

92

III.ANALYTICAL METHODS DEVELOPMENT

93

94

An analytical procedure is developed to test a defined characteristic of the drug substance or

95

drug product against established acceptance criteria for that characteristic. Early in the

96

development of a new analytical procedure, the choice of analytical instrumentation and

97

methodology should be selected based on the intended purpose and scope of the analytical

98

method. Parameters that may be evaluated during method development are specificity, linearity, 99

limits of detection (LOD) and quantitation limits (LOQ), range, accuracy, and precision.

100

101

During early stages of method development, the robustness of methods should be evaluated

102

because this characteristic can help you decide which method you will submit for approval.

103

Analytical procedures in the early stages of development are initially developed based on a

104

combination of mechanistic understanding of the basic methodology and prior experience.

105

Experimental data from early procedures can be used to guide further development. You should 106

submit development data within the method validation section if they support the validation of 107

the method.

108

109

To fully understand the effect of changes in method parameters on an analytical procedure, you 110

should adopt a systematic approach for method robustness study (e.g., a design of experiments 111

with method parameters). You should begin with an initial risk assessment and follow with

112

multivariate experiments. Such approaches allow you to understand factorial parameter effects 113

on method performance. Evaluation of a method’s performance may include analyses of

114

samples obtained from in-process manufacturing stages to the finished product. Knowledge

115

gained during these studies on the sources of method variation can help you assess the method 116

performance.

117

118

119

IV.CONTENT OF ANALYTICAL PROCEDURES

120

121

You should describe analytical procedures in sufficient detail to allow a competent analyst to 122

reproduce the necessary conditions and obtain results within the proposed acceptance criteria. 123

You should also describe aspects of the analytical procedures that require special attention. An 124

analytical procedure may be referenced from FDA recognized sources (e.g., USP/NF,

125

10 See sections 3.2.S.4 Control of Drug Substance, 3.2.P.4 Control of Excipients, and 3.2.P.5 Control of Drug

Product.

Association of Analytical Communities (AOAC) International)11 if the referenced analytical

126

procedure is not modified beyond what is allowed in the published method. You should provide 127

in detail the procedures from other published sources. The following is a list of essential

128

information you should include for an analytical procedure:

129

130

A.Principle/Scope

131

132

A description of the basic principles of the analytical test/technology (separation, detection, etc.); 133

target analyte(s) and sample(s) type (e.g., drug substance, drug product, impurities or compounds 134

in biological fluids, etc.).

135

136

B.Apparatus/Equipment

137

138

All required qualified equipment and components (e.g., instrument type, detector, column type, 139

dimensions, and alternative column, filter type, etc.).

140

141

C.Operating Parameters

142

143

Qualified optimal settings and ranges (allowed adjustments) critical to the analysis (e.g., flow

144

rate, components temperatures, run time, detector settings, gradient, head space sampler). A

145

drawing with experimental configuration and integration parameters may be used, as applicable. 146

147

D.Reagents/Standards

148

149

The following should be listed:

150

151

?Grade of chemical (e.g., USP/NF, American Chemical Society, High

152

Performance or Pressure Liquid Chromatography, or Gas

153

Chromatography and preservative free).

154

?Source (e.g., USP reference standard or qualified in-house reference material). 155

?State (e.g., dried, undried, etc.) and concentration.

156

?Standard potencies (purity correction factors).

157

?Storage controls.

158

?Directions for safe use (as per current Safety Data Sheet).

159

?Validated or useable shelf life.

160

161

New batches of biological reagents, such as monoclonal antibodies, polyclonal antisera, or cells, 162

may need extensive qualification procedures included as part of the analytical procedure.

163

164

E.Sample Preparation

165

166

Procedures (e.g., extraction method, dilution or concentration, desalting procedures and mixing 167

by sonication, shaking or sonication time, etc.) for the preparations for individual sample tests. 168

A single preparation for qualitative and replicate preparations for quantitative tests with

169

11 See 21 CFR 211.194(a)(2).

appropriate units of concentrations for working solutions (e.g., μg/ml or mg/ml) and information 170

on stability of solutions and storage conditions.

171

172

F.Standards Control Solution Preparation

173

174

Procedures for the preparation and use of all standard and control solutions with appropriate

175

units of concentration and information on stability of standards and storage conditions,

176

including calibration standards, internal standards, system suitability standards, etc.

177

178

G.Procedure

179

180

A step-by-step description of the method (e.g., equilibration times, and scan/injection sequence 181

with blanks, placeboes, samples, controls, sensitivity solution (for impurity method) and

182

standards to maintain validity of the system suitability during the span of analysis) and allowable 183

operating ranges and adjustments if applicable.

184

185

H.System Suitability

186

187

Confirmatory test(s) procedures and parameters to ensure that the system (equipment,

188

electronics, and analytical operations and controls to be analyzed) will function correctly as an 189

integrated system at the time of use. The system suitability acceptance criteria applied to

190

standards and controls, such as peak tailing, precision and resolution acceptance criteria, may be 191

required as applicable. For system suitability of chromatographic systems, refer to CDER

192

reviewer guidance on Validation of Chromatographic Methods and USP General Chapter <621> 193

Chromatography.

194

195

I.Calculations

196

197

The integration method and representative calculation formulas for data analysis (standards,

198

controls, samples) for tests based on label claim and specification (e.g., assay, specified and

199

unspecified impurities and relative response factors). This includes a description of any

200

mathematical transformations or formulas used in data analysis, along with a scientific

201

justification for any correction factors used.

202

203

J.Data Reporting

204

205

A presentation of numeric data that is consistent with instrumental capabilities and acceptance 206

criteria. The method should indicate what format to use to report results (e.g., percentage label 207

claim, weight/weight, and weight/volume etc.) with the specific number of significant figures 208

needed. The American Society for Testing and Materials (ASTM) E29 describes a standard

209

practice for using significant digits in test data to determine conformance with specifications. For 210

chromatographic methods, you should include retention times (RTs) for identification with

211

reference standard comparison basis, relative retention times (RRTs) (known and unknown

212

impurities) acceptable ranges and sample results reporting criteria.

213

214

215

V.REFERENCE STANDARDS AND MATERIALS

216

217

Primary and secondary reference standards and materials are defined and discussed in the

218

following ICH guidances: Q6A Specifications: Test Procedures and Acceptance Criteria for 219

New Drug Substances and New Drug Products: Chemical Substances (ICH Q6A), Q6B

220

Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological

221

Products, and Q7 Good Manufacturing Practice Guidance for Active Pharmaceutical

222

Ingredients. For all standards, you should ensure the suitability for use. Reference standards for 223

drug substances are particularly critical in validating specificity for an identity test. You should 224

strictly follow storage, usage conditions, and handling instructions for reference standards to

225

avoid added impurities and inaccurate analysis. For biological products, you should include

226

information supporting any reference standards and materials that you intend to use in the BLA 227

and in subsequent annual reports for subsequent reference standard qualifications. Information 228

supporting reference standards and materials include qualification test protocols, reports, and 229

certificates of analysis (including stability protocols and relevant known impurity profile

230

information, as applicable).

231

232

Reference standards can often be obtained from USP and may also be available through the

233

European Pharmacopoeia, Japanese Pharmacopoeia, World Health Organization, or National 234

Institute of Standards and Technology. Reference standards for a number of biological products 235

are also available from CBER. For certain biological products marketed in the U.S., reference 236

standards authorized by CBER must be used before the product can be released to the market.12 237

Reference materials from other sources should be characterized by procedures including routine 238

and beyond routine release testing as described in ICH Q6A. You should consider orthogonal 239

methods. Additional testing could include attributes to determine the suitability of the reference 240

material not necessarily captured by the drug substance or product release tests (e.g., more

241

extensive structural identity and orthogonal techniques for purity and impurities, biological

242

activity).

243

244

For biological reference standards and materials, we recommend that you follow a two-tiered 245

approach when qualifying new reference standards to help prevent drift in the quality attributes 246

and provide a long-term link to clinical trial material. A two-tiered approach involves a

247

comparison of each new working reference standard with a primary reference standard so that it 248

is linked to clinical trial material and the current manufacturing process.

249

250

251

VI.ANALYTICAL METHOD VALIDATION FOR NDA, ANDAs, BLAs, AND 252

DMFs

253

254

A.Noncompendial Analytical Procedures

255

256

Analytical method validation is the process of demonstrating that an analytical procedure is

257

suitable for its intended purpose. The methodology and objective of the analytical procedures 258

should be clearly defined and understood before initiating validation studies. This understanding 259

12 See 21 CFR 610.20.

is obtained from scientifically-based method development and optimization studies. Validation 260

data must be generated under an protocol approved by the sponsor following current good

261

manufacturing practices with the description of methodology of each characteristic test and

262

predetermined and justified acceptance criteria, using qualified instrumentation operated under 263

current good manufacturing practices conditions.13 Protocols for both drug substance and

264

product analytes or mixture of analytes in respective matrices should be developed and executed. 265

266

ICH Q2(R1) is considered the primary reference for recommendations and definitions on

267

validation characteristics for analytical procedures. The FDA Reviewer Guidance: Validation of 268

Chromatographic Methods is available as well.

269

270

B.Validation Characteristics

271

272

Although not all of the validation characteristics are applicable for all types of tests, typical

273

validation characteristics are:

274

275

?Specificity

276

?Linearity

277

?Accuracy

278

?Precision (repeatability, intermediate precision, and reproducibility)

279

?Range

280

?Quantitation limit

281

?Detection limit

282

283

If a procedure is a validated quantitative analytical procedure that can detect changes in a quality 284

attribute(s) of the drug substance and drug product during storage, it is considered a stability

285

indicating assay. To demonstrate specificity of a stability-indicating assay, a combination of

286

challenges should be performed. Some challenges include the use of samples spiked with target 287

analytes and all known interferences; samples that have undergone various laboratory stress

288

conditions; and actual product samples (produced by the final manufacturing process) that are

289

either aged or have been stored under accelerated temperature and humidity conditions.

290

291

As the holder of the NDA, ANDA, or BLA, you must:14 (1) submit the data used to establish

292

that the analytical procedures used in testing meet proper standards of accuracy and reliability, 293

and (2) notify the FDA about each change in each condition established in an approved

294

application beyond the variations already provided for in the application, including changes to 295

analytical procedures and other established controls.

296

297

The submitted data should include the results from the robustness evaluation of the method,

298

which is typically conducted during method development or as part of a planned validation

299

study.15

300

13 See 21 CFR 211.165(e); 21 CFR 314.50 (d), and for biologics see 21 CFR 601.2(a), 601.2(c), and 601.12(a).

14 For drugs see 21 CFR 314.50 (d), 314.70(d), and for biologics see 21 CFR 601.2(a), 601.2(c), and 601.12(a). For a

BLA, as discussed below, you must obtain prior approval from FDA before implementing a change in analytical

methods if those methods are specified in FDA regulations

15 See section III and ICH Q2(R1).

https://www.wendangku.net/doc/7a13924693.html,pendial Analytical Procedures

302

303

The suitability of an analytical procedure (e.g., USP/NF, the AOAC International Book of

304

Methods, or other recognized standard references) should be verified under actual conditions of 305

use.16 Compendial general chapters, which are complex and mention multiple steps and/or

306

address multiple techniques, should be rationalized for the intended use and verified. Information 307

to demonstrate that USP/NF analytical procedures are suitable for the drug product or drug

308

substance should be included in the submission and generated under a verification protocol.

309

310

The verification protocol should include, but is not limited to: (1) compendial methodology to 311

be verified with predetermined acceptance criteria, and (2) details of the methodology (e.g.,

312

suitability of reagent(s), equipment, component(s), chromatographic conditions, column, detector 313

type(s), sensitivity of detector signal response, system suitability, sample preparation and

314

stability). The procedure and extent of verification should dictate which validation characteristic 315

tests should be included in the protocol (e.g., specificity, LOD, LOQ, precision, accuracy, etc.). 316

Considerations that may influence what characteristic tests should be in the protocol may depend 317

on situations such as whether specification limits are set tighter than compendial acceptance

318

criteria, or RT or RRT profiles are changing in chromatographic methods because of the

319

synthetic route of drug substance or differences in manufacturing process or matrix of drug

320

product. Robustness studies of compendial assays do not need to be included, if methods are 321

followed without deviations.

322

323

324

VII.STATISTICAL ANALYSIS AND MODELS

325

326

A.Statistics

327

328

Statistical analysis of validation data can be used to evaluate validation characteristics against 329

predetermined acceptance criteria. All statistical procedures and parameters used in the analysis 330

of the data should be based on sound principles and appropriate for the intended evaluation.

331

Reportable statistics of linear regression analysis R (correlation coefficient), R square

332

(coefficient of determination), slope, least square, analysis of variance (ANOVA), confidence 333

intervals, etc., should be provided with justification.For information on statistical techniques 334

used in making comparisons, as well as other general information on the interpretation and

335

treatment of analytical data, appropriate literature or texts should be consulted.17

336

337

B.Models

338

339

Some analytical methods might use chemometric and/or multivariate models. When developing 340

these models, you should include a statistically adequate number and range of samples for model 341

development and comparable samples for model validation. Suitable software should be used for 342

data analysis. Model parameters should be deliberately varied to test model robustness.

343

344

16 See 21 CFR 211.194(a)(2) and USP General Chapter <1226> Verification of Compendial Procedures.

17 See References section for examples including USP <1010> Analytical Data – Interpretation and Treatment.

VIII.LIFE CYCLE MANAGEMENT OF ANALYTICAL PROCEDURES

346

347

Once an analytical procedure (including compendial methods) is successfully validated and

348

implemented, the procedure will be followed during the life cycle of the product. Trend analysis 349

on method performance should be performed at regular intervals to evaluate the need to optimize 350

the analytical procedure or to revalidate all or a part of the analytical procedure. If an analytical 351

procedure can only meet the established system suitability requirements with repeated

352

adjustments to the operating conditions stated in the analytical procedure, the analytical

353

procedure should be reevaluated, revalidated, or amended, as appropriate.

354

355

Over the life cycle of a product, new information (e.g., a better understanding of product CQAs 356

or awareness of a new impurity) may warrant the development and validation of a new or

357

alternative analytical method. New technologies may allow for greater understanding and/or 358

confidence when ensuring product quality. Applicants should periodically evaluate the

359

appropriateness of a product’s analytical methods and consider new or alternative methods.

360

361

In anticipation of life cycle changes in analytics, an appropriate number of samples should be 362

archived to allow for comparative studies. The number should be based on scientific principles 363

and an assessment of risk. For complex products that are sensitive to manufacturing changes, 364

archived samples can be an important tool to make these comparisons. The archived samples 365

used in comparative studies should include samples that represent pivotal clinical trial material 366

and marketed product.

367

368

If a risk-based evaluation or other drivers lead to changes in an analytical procedure or

369

replacement with a new method or if the procedure is transferred to a new testing site;

370

revalidation, a new validation exercise, an analytical method comparability study, or a

371

combination of these exercises should be considered. In some cases, changes to the drug

372

substance or drug product manufacturing process may also warrant analytical procedure

373

revalidation. These additional studies are discussed below.

374

375

A.Revalidation

376

377

Principles described in the validation section (section VI) apply to revalidation. When a change 378

is made to an analytical procedure (e.g., a change in a piece of equipment or reagent or because 379

of a change in manufacturing process or formulation), revalidation of all or part of the analytical 380

procedure should be considered. Analytical method revalidation may also be warranted because 381

of manufacturing process changes, such as an alteration in the drug substance manufacturing

382

process that could impact method performance (e.g., route of synthesis, fermentation) or

383

introduction of a new drug product formulation.

384

385

You should revalidate to ensure that the analytical procedure maintains its critical performance 386

characteristics (e.g., specificity, precision, accuracy, etc). The degree of revalidation depends on 387

the nature of the change.

388

389

B.Analytical Method Comparability Studies

390

391

Analytical method comparability study requests are typically generated when you propose to

392

substitute an FDA approved analytical procedure with an alternative analytical procedure or

393

when an analytical method is transferred from one laboratory to the other. These scenarios are 394

discussed below.

395

396

1.Alternative Analytical Procedures

397

398

An alternative analytical procedure is an analytical procedure that you use in place of the FDA 399

approved analytical procedure. For an NDA or ANDA, you should include any proposed

400

alternate analytical procedures in the application. You must include a description of the

401

procedure.18 After approval, for an NDA or ANDA, or for a procedure approved in a BLA but 402

not included in an FDA regulation, the addition, revision, or deletion of an alternative analytical 403

procedure that provides the same or increased assurance of the identity, strength, quality, purity, 404

or potency of the material being tested as the analytical procedure described in the approved

405

application, must be documented in the next annual report.19 Additions or revisions of analytical 406

procedures in BLAs may require submission as a supplement. FDA recommends discussion with 407

the appropriate review group to determine the appropriate reporting category

408

409

For biological products, rarely an analytical procedure may be included in an FDA regulation. 410

When that occurs, alternative analytical procedures are submitted following 21 CFR 610.9(a). It 411

states that the applicant will present evidence “…demonstrating that the modification will

412

provide assurances of the safety, purity, potency, and effectiveness of the biological product

413

equal to or greater than the assurances provided by the method or process specified in the general 414

standards or additional standards for the biological product.” Modification of such procedures 415

requires FDA approval during application review or in a postapproval supplement.20

416

417

You should identify the use of the alternative analytical procedure (e.g., release, stability testing) 418

and provide a rationale for its inclusion, validation data, and comparative data to the FDA

419

approved analytical procedure. You should perform a comparability study that demonstrates at a 420

minimum that:

421

422

?The new method coupled with any additional control measures is equal or

423

superior for the original method for the intended purpose.

424

425

?The new analytical procedure is not more susceptible to matrix effects than the 426

original procedure.

427

428

If new process or product related variants or any new impurities are discovered with the new

429

procedure, testing on archived samples from historical batches should be performed to

430

demonstrate that the variants/impurities detected by the new method are a result of an increase in 431

18 See 21 CFR 314.50.

19 See 21 CFR 314.70(d)(1), (d)(2)(vii). 314.81(b)(2), and 601.12(d)(vii).

20 See 21 CFR 610.9(b).

the sensitivity or selectivity of the new procedure and not a result of a change to process related 432

impurities.

433

434

If the procedure has stability indicating properties:

435

436

?Appropriate samples should be included that allow a comparison of the ability of 437

the new and original method to detect relevant product variants and degradation 438

species.

439

?The number of batches analyzed for comparison should be statistically relevant 440

and justified for a pre-established confidence interval.

441

?Equivalence, non-inferiority, or superiority studies should be performed with

442

appropriate statistical methods to demonstrate that the new or revised method

443

performance is comparable or better than the original method.

444

?The statistical analyses performed to compare product testing should be

445

identified.

446

?All bias seen with comparative results should be discussed with an explanation, as 447

appropriate.

448

449

2.Analytical Methods Transfer Studies

450

451

Analytical method transfer is typically managed under an internal transfer protocol that details 452

the parameters to be evaluated in addition to the predetermined acceptance criteria that will be 453

applied to the results. Transfer studies usually involve two or more laboratories or sites

454

(originating lab and receiving labs) executing the preapproved transfer protocol. A sufficient 455

number of representative test articles (e.g., same lot(s) of drug substance or drug product) are 456

used by the originating and receiving laboratories. The comparative s tudies are performed to 457

evaluate accuracy and precision, especially with regard to assessment of interlaboratory

458

variability. In cases where the transferred analytical procedure is also a stability indicating

459

method, forced degradation samples or samples containing pertinent product-related impurities 460

should be analyzed at both sites. The USP General Chapter <1224> Transfer of Analytical

461

Procedures provides additional guidance on this topic.

462

463

C.Reporting Postmarketing Changes to an Approved NDA, ANDA, or BLA 464

465

Postmarketing changes to analytical procedures must be reported to the FDA in compliance with 466

21 CFR 314.70 or 21 CFR 601.12.21 Additional information on the appropriate reporting

467

category for various kinds of postapproval changes for NDAs and ANDAs is provided in the

468

FDA guidance for industry on Changes to an Approved NDA or ANDA and Changes to an

469

Approved NDA or ANDA; Specifications – Use of Enforcement Discretion for Compendial

470

Changes. Similar information on postapproval changes to BLAs regulated by CDER and CBER 471

is provided in the FDA guidance Changes to an Approved Application for Specified

472

Biotechnology and Specified Synthetic Biological Products.

473

474

475

21 As noted, for a product licensed under a BLA, if the change is to a procedure prescribed in FDA regulations that

change must be approved by FDA pursuant to 21 CFR 610.9(b).

IX.FDA METHODS VERIFICATION

476

477

Part of the approval process for NDAs and ANDAs may include FDA laboratory assessment to 478

determine whether the analytical procedures are acceptable for quality control and suitable for 479

regulatory purposes.22 If a laboratory assessment will be conducted, the FDA laboratory will 480

send you a request that will detail what samples and supplies to send to the FDA laboratory.

481

These could include product samples, standards, critical reagents, material safety data sheets, and 482

supplies. Laboratory results and comments will be forwarded from the FDA laboratory to the 483

product quality reviewer.

484

485

For certain biological products, samples representative of the product for licensure along with 486

summaries of results of tests performed on the lots represented by these samples should be

487

submitted with the BLA.23 The FDA laboratory verifies the performance of the methods and the 488

results you submit. During the pre-BLA meeting or after submission of the BLA, the FDA

489

laboratory can send you a request to provide standards, controls, reagents, material safety data 490

sheets, and supplies.

491

492

493

X.REFERENCES

494

495

Guidance for Industry24

496

497

ANDAs: Impurities in Drug Products (November 2010)

498

499

ANDAs: Impurities in Drug Substances (July 2009)

500

501

Changes to an Approved NDA or ANDA (April 2004)

502

503

Changes to an Approved Application for Specified Biotechnology and Specified Synthetic

504

Biological Products (July 1997)

505

506

Changes to an Approved NDA or ANDA; Specifications – Use of Enforcement Discretion for 507

Compendial Changes (November 2004)

508

509

Content and Format of Investigational New Drug Applications (INDs) for Phase 1 Studies of 510

Drugs, Including Well-Characterized, Therapeutic, Biotechnology-derived Products (November 511

1995)

512

513

INDs for Phase 2 and 3 Studies of Drugs, Including Specified Therapeutic Biotechnology-

514

Derived Products (February 1999)

515

516

22 See 21 CFR 314.50(e).

23 See 21 CFR 601.2(a).

24 Draft guidances have been included for completeness only. As draft documents, they are not intended to be

implemented until published in final form.

Investigating Out of Specification (OOS) Test Results for Pharmaceutical Production (October 517

2006)

518

519

Submission of Chemistry, Manufacturing, and Controls Information for Synthetic Peptide

520

Substances (November 1994)

521

522

Guidance for Industry: International Conference on Harmonization

523

524

Q1A(R2) Stability Testing of New Drug Substances and Products (November 2003)

525

526

Q1B Stability Testing: Photostability Testing of New Drug Substances and Products (May 527

1997)

528

529

Q1C Stability Testing for New Dosage Forms (May 1997)

530

531

Q2(R1) Validation of Analytical Procedures: Text and Methodology (March 1995, May 1997) 532

533

Q3A(R2) Impurities in New Drug Substances (June 2008)

534

535

Q3B(R2) Impurities in New Drug Products (August 2006)

536

537

Q3C Impurities: Residual Solvents (December1997)

538

539

Q3C Tables and List (February 2012)

540

541

Q5C Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological 542

Products (July 1996)

543

544

Q6A Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and 545

New Drug Products: Chemical Substances (December 2000)

546

547

Q6B Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological 548

Products (August 1999)

549

550

Q7 Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients

551

(August 2001)

552

553

Reviewer Guidance

554

555

Validation of Chromatographic Methods (November 1994)

556

557

United States Pharmacopeia/National Formulary

558

559

General Chapter <621> Chromatography

560

561

General Chapter <1010> Analytical Data – Interpretation and Treatment

562

563

General Chapter <1224> Transfer of Analytical Procedures

564

565

General Chapter <1225> Validation of Compendial Procedures

566

567

General Chapter <1226> Verification of Compendial Procedures

568

569

General Notices and Requirements, Applying to Standards, Tests, Assays, and Other

570

Specifications of the United States Pharmacopeia: 7. Test Results

571

572

Interpretation and Treatment of Analytical Data; USP Pharmacopeial Forum, United States

573

Pharmacopeial Convention, Inc., Rockville MD: 1994, Volume 24, Number 5, pp. 7051 - 7056 574

575

Other

576

577

ASTM Standard, E29 - 2008 Standard Practice for Using Significant Digits in Test Data to

578

Determine Conformance with Specifications, ASTM International, West Conshohocken, PA, 579

(https://www.wendangku.net/doc/7a13924693.html,).

580

581

Miller, J.C., J.N. Miller, and E. Horwood, Statistics for Analytical Chemistry, 3rd edition,

582

Prentice

583

Hall, 1993.

584

585

Saunders, B.D., and R.G. Trapp,Basic and Clinical Biostatistics, 2nd edition, Appleton and 586

Lange,

587

1994.

588

589

590

591

20.化学药物质量控制分析方法验证技术指导原则 2005年颁布

指导原则编号: 【H】G P H 5-1 化学药物质量控制分析方法验证 技术指导原则 二○○五年三月

目 录 一、概述 (1) 二、方法验证的一般原则 (2) 三、方法验证涉及到的三个主要方面 (2) (一)需要验证的检测项目 (2) (二)分析方法 (3) (三)验证内容 (3) 四、方法验证的具体内容 (3) (一)专属性 (3) 1、鉴别反应 (4) 2、杂质检查 (4) 3、含量测定 (4) (二)线性 (5) (三)范围 (5) 1、含量测定 (6) 2、制剂含量均匀度 (6) 3、溶出度或释放度 (6) 4、杂质 (6) (四)准确度 (6) 1、含量测定 (7) 2、杂质定量试验 (7) (五)精密度 (7) 1、重复性 (8) 2、中间精密度 (8) 3、重现性 (8)

(六)检测限 (8) 1、直观法 (8) 2、信噪比法 (9) (七)定量限 (9) 1、直观法 (9) 2、信噪比法 (9) (八)耐用性 (10) (九)系统适用性试验 (10) 五、方法再验证 (11) 六、方法验证的评价 (12) (一)有关方法验证评价的一般考虑 (12) (二)方法验证的整体性和系统性 (12) 七、参考文献 (13) 八、著者 (13)

化学药物质量控制分析方法验证技术指导原则 一、概述 保证药品安全、有效、质量可控是药品研发和评价应遵循的基本原则,其中,对药品进行质量控制是保证药品安全有效的基础和前提。为达到控制质量的目的,需要多角度、多层面来控制药品质量,也就是说要对药物进行多个项目测试,来全面考察药品质量。一般地,每一测试项目可选用不同的分析方法,为使测试结果准确、可靠,必须对所采用的分析方法的科学性、准确性和可行性进行验证,以充分表明分析方法符合测试项目的目的和要求,这就是通常所说的对方法进行验证。 方法验证的目的是判断采用的分析方法是否科学、合理,是否能有效控制药品的内在质量。从本质上讲,方法验证就是根据检测项目的要求,预先设置一定的验证内容,并通过设计合理的试验来验证所采用的分析方法能否符合检测项目的要求。 方法验证在分析方法建立过程中具有重要的作用,并成为质量研究和质量控制的组成部分。只有经过验证的分析方法才能用于控制药品质量,因此方法验证是制订质量标准的基础。方法验证是药物研究过程中的重要内容。 本指导原则重点探讨方法验证的本质,将分析方法验证的要求与所要达到的目的结合起来进行系统和规律性的阐述,重点阐述如何科学合理地进行论证方案的设计。 本指导原则主要包括方法验证的一般原则、方法验证涉及的三个主要方

药品微生物检验替代方法验证指导原则

药品微生物检验替代方法验证指导原则 本指导原则是为所采用的试验方法能否替代药典规定的方法用于药品微生物的检验提供指导。 随着微生物学的迅速发展,制药领域不断引入了一些新的微生物检验技术,大体可分为三类:(1)基于微生物生长信息的检验技术,如生物发光技术、电化学技术、比浊法等;(2)直接测定被测介质中活微生物的检验技术,如固相细胞技术法、流式细胞计数法等;(3)基于微生物细胞所含有特定组成成分的分析技术,如脂肪酸测定技术、核酸扩增技术、基因指纹分析技术等。这些方法与传统检查方法比较,或简便快速,或具有实时或近实时监控的潜力,使生产早期采取纠正措施及监控和指导优良生产成为可能,同时新技术的使用也促进了生产成本降低及检验水平的提高。 在控制药品微生物质量中,微生物实验室出于各种原因如成本、生产量、快速简便及提高药品质量等需要而采用非药典规定的检验方法(即替代方法)时,应进行替代方法的验证,确认其应用效果优于或等同于药典的方法。 微生物检验的类型及验证参数 药品微生物检验方法主要分两种类型:定性试验和定量试验。定性试验就是测定样品中是否存在活的微生物,如无菌检查及控制菌检査。定量试验就是测定样品中存在的微生物数量,如菌落计数试验。 由于生物试验的特殊性,如微生物检验方法中的抽样误差、稀释误差、操作误差、培养误差和计数误差都会对检验结果造成影响,因此,药品质量标准分析方法验证指导原则(附录XIX A)不完全适宜于微生物替代方法的验证。药品微生物检验替代方法的验证参数见表1。 表1 不同微生物检验类型验证参数 注: 尽管替代方法的验证参数与药品质量标准分析方法验证参数有相似之处,但是其具体的内容是依据微生物检验特点而设立的。替代方法验证的实验结果需进行统计分析,当替代方法属于定性检验时,一般采用非参数的统计技术;当替代方法属于定量检验时,需要采用参数统计技术。 进行微生物替代方法的验证时,若替代方法只是针对药典方法中的某一环节进行技术修改,此时,需要验证的对象仅是该项替代技术而不是整个检验方法。如无菌试验若改为使用含培养基的过滤器,然后通过适宜的技术确认活的微生物存在,那么,验证时仅需验证所用的微生物回收系统而不是整个无菌试验方法。 替代方法验证的一般要求 在开展替代方法对样品检验的适用性验证前,有必要对替代方法有一个全面的了解。首先,所选用的替代方法应具备必要的方法适用性证据,表明在不含样品的情况下,替代方法

生物样品定量分析方法验证指导原则

9012 生物样品定量分析方法验证指导原则
1. 范围
准确测定生物基质(如全血、血清、血浆、尿)中的药物浓度,对于药物和 制剂研发非常重要。这些数据可被用于支持药品的安全性和有效性,或根据毒动 学、药动学和生物等效性试验的结果做出关键性决定。因此,必须完整地验证和 记录应用的生物分析方法,以获得可靠的结果。
本指导原则提供生物分析方法验证的要求,也涉及非临床或临床试验样品实 际分析的基本要求,以及何时可以使用部分验证或交叉验证,来替代完整验证。
生物样品定量分析方法验证和试验样品分析应符合本指导原则的技术要求。 应该在相应的生物样品分析中遵守 GLP 原则或 GCP 原则。
2. 生物分析方法验证
2.1 分析方法的完整验证
分析方法验证的主要目的是,证明特定方法对于测定在某种生物基质中分析 物浓度的可靠性。此外,方法验证应采用与试验样品相同的抗凝剂。一般应对每 个物种和每种基质进行完整验证。当难于获得相同的基质时,可以采用适当基质 替代,但要说明理由。
一个生物分析方法的主要特征包括:选择性、定量下限、响应函数和校正范 围(标准曲线性能)、准确度、精密度、基质效应、分析物在生物基质以及溶液 中储存和处理全过程中的稳定性。
有时可能需要测定多个分析物。这可能涉及两种不同的药物,也可能涉及一 个母体药物及其代谢物,或一个药物的对映体或异构体。在这些情况下,验证和 分析的原则适用于所有涉及的分析物。
对照标准物质 在方法验证中,含有分析物对照标准物质的溶液将被加入到空白生物基质 中。此外,色谱方法通常使用适当的内标。 应该从可追溯的来源获得对照标准物质。应该科学论证对照标准物质的适用 性。分析证书应该确认对照标准物质的纯度,并提供储存条件、失效日期和批号。 对于内标,只要能证明其适用性即可,例如显示该物质本身或其相关的任何杂质 不产生干扰。 当在生物分析方法中使用质谱检测时,推荐尽可能使用稳定同位素标记的内 标。它们必须具有足够高的同位素纯度,并且不发生同位素交换反应,以避免结 果的偏差。
1

药物非临床药代动力学研究技术指导原则

附件5 药物非临床药代动力学研究技术指导原则 一、概述 非临床药代动力学研究是通过体外和动物体内的研究方法,揭示药物在体内的动态变化规律,获得药物的基本药代动力学参数,阐明药物的吸收、分布、代谢和排泄(Absorption, Distribution, Metabolism, Excretion, 简称ADME)的过程和特征。 非临床药代动力学研究在新药研究开发的评价过程中起着重要 作用。在药物制剂学研究中,非临床药代动力学研究结果是评价药物制剂特性和质量的重要依据。在药效学和毒理学评价中,药代动力学特征可进一步深入阐明药物作用机制,同时也是药效和毒理研究动物选择的依据之一;药物或活性代谢产物浓度数据及其相关药代动力学参数是产生、决定或阐明药效或毒性大小的基础,可提供药物对靶器官效应(药效或毒性)的依据。在临床试验中,非临床药代动力学研究结果能为设计和优化临床试验给药方案提供有关参考信息。 本指导原则是供中药、天然药物和化学药物新药的非临床药代动力学研究的参考。研究者可根据不同药物的特点,参考本指导原则,科学合理地进行试验设计,并对试验结果进行综合评价。 本指导原则的主要内容包括进行药物非临床药代动力学研究的 基本原则、试验设计的总体要求、生物样品的测定方法、研究项目(血

药浓度-时间曲线、吸收、分布、排泄、血浆蛋白结合、生物转化、对药物代谢酶活性及转运体的影响)、数据处理与分析、结果与评价等,并对研究中其他一些需要关注的问题进行了分析。附录中描述了生物样品分析和放射性同位素标记技术的相关方法和要求,供研究者参考。 二、基本原则 进行非临床药代动力学研究,要遵循以下基本原则: (一)试验目的明确; (二)试验设计合理; (三)分析方法可靠; (四)所得参数全面,满足评价要求; (五)对试验结果进行综合分析与评价; (六)具体问题具体分析。 三、试验设计 (一)总体要求 1. 受试物 中药、天然药物:受试物应采用能充分代表临床试验拟用样品和/或上市样品质量和安全性的样品。应采用工艺路线及关键工艺参数确定后的工艺制备,一般应为中试或中试以上规模的样品,否则应有充分的理由。应注明受试物的名称、来源、批号、含量(或规格)、保存条件、有效期及配制方法等,并提供质量检验报告。由于中药的特殊性,建议现用现配,否则应提供数据支持配制后受试物的质量稳定性及均匀性。当给药时间较

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

美国FDA药物分析程序及方法验证指导原则(中文版)

药品及生物制品的分析方法和方法验证指导原则 目录 1.介绍...................... (1) 2.背景..................... .. (2) 3.分析方法开发. ..................... . (3) 4.分析程序内容.............................................. ......... ..................................... .. 3 A.原则/范围 (4) B.仪器/设备............................................. . (4) C.操作参数.............................................. .. (4) D.试剂/标准............................................. . (4) E.样品制备.............................................. .. (4) F.标准对照品溶液的制备............................................ .. (5) G.步骤......... ....................................... (5) H.系统适应性..... (5) I.计算 (5) J.数据报告 (5) 5.参考标准和教材............................................ (6) 6分析方法验证用于新药,仿制药,生物制品和DMF (6) A.非药典分析方法............................................. (6) B.验证特征 (7) C.药典分析方法............................................. .. (8) 7.统计分析和模型 (8) A.统计 (8) B.模型 (8) 8.生命周期管理分析程序 (9) A.重新验证 (9) B.分析方法的可比性研究............................................ . (10) 1.另一种分析方法............................................... .. (10) 2.分析方法转移的研究 (11) C.报告上市后变更已批准的新药,仿制药,或生物制品 (11) 9.美国FDA方法验证............................................... . (12) 10.参考文献

9012生物样品定量分析方法验证指导原则

中国药典2015年版 9012生物样品定置分析方法验证 指导原则 一、范围 准确测定生物基质(如全血、血清、血浆、尿)中的药物浓度,对于药物和制剂研发非常重要。这些数据可被用于支持药品的安全性和有效性,或根据毒动学、药动学和生物等效性试验的结果做出关键性决定。因此,必须完整地验证和记录应用的生物分析方法,以获得可靠的结果。 本指导原则提供生物分析方法验证的要求,也涉及非临床或临床试验样品实际分析的基本要求,以及何时可以使用部分验证或交叉验证,来替代完整验证。本指导原则二和三主要针对色谱分析方法,四针对配体结合分析方法。 生物样品定量分析方法验证和试验样品分析应符合本指导原则的技术要求。应该在相应的生物样品分析中遵守 G L P原则或GC P原则。 二、生物分析方法验证 (一)分析方法的完整验证 分析方法验证的主要目的是,证明特定方法对于测定在某种生物基质中分析物浓度的可靠性。此外,方法验证应采用与试验样品相同的抗凝剂。一般应对每个新分析方法和新分析物进行完整验证。当难于获得相同的基质时,可以采用适当基质替代,但要说明理由。 一个生物分析方法的主要特征包括:选择性、定量下限、响应函数和校正范围(标准曲线性能)、准确度、精密度、基质效应、分析物在生物基质以及溶液中储存和处理全过程中的稳定性。 有时可能需要测定多个分析物。这可能涉及两种不同的药物,也可能涉及一个母体药物及其代谢物,或一个药物的对映体或异构体。在这些情况下,验证和分析的原则适用于所有涉及的分析物。 对照标准物质 在方法验证中,含有分析物对照标准物质的溶液将被加人到空白生物基质中。此外,色谱方法通常使用适当的内标。 应该从可追溯的来源获得对照标准物质。应该科学论证对照标准物质的适用性。分析证书应该确认对照标准物质的纯度,并提供储存条件、失效日期和批号。对于内标,只要能证明其适用性即可,例如显示该物质本身或其相关的任何杂质不产生干扰。 当在生物分析方法中使用质谱检测时,推荐尽可能使用稳定同位素标记的内标。它们必须具有足够高的同位素纯度,并且不发生同位素交换反应,以避免结果的偏差。 1.选择性 该分析方法应该能够区分目标分析物和内标与基质的内源性组分或样品中其他组分。应该使用至少6个受试者的适宜的空白基质来证明选择性(动物空白基质可以不同批次混 9012生物样品定量分析方法验证指导原则 合),它们被分别分析并评价干扰。当干扰组分的响应低于分析物定量下限响应的20%,并低于内标响应的5%时,通常即可以接受0 应该考察药物代谢物、经样品预处理生成的分解产物以及可能的同服药物引起干扰的程度。在适当情况下,也应该评价代谢物在分析过程中回复转化为母体分析物的可能性。 2.残留 应该在方法建立中考察残留并使之最小。残留可能不影响准确度和精密度。应通过在注射高浓度样品或校正标样后,注射空白样品来估计残留。高浓度样品之后在空白样品中的残留应不超过定量下限的20%,并且不超过内标的5%。如果残留不可避免,应考虑特殊措施,在方法验证时检验并在试验样品分析时应用这些措施,以确保不影响准确度和精密度。这可能包括在高浓度样品后注射空白样品,然后分析下一个试验样品。 3.定量下限 定量下限是能够被可靠定量的样品中分析物的最低浓度,具有可接受的准确度和精密度。定量下限是标准曲线的最低点,应适用于预期的浓度和试验目的。 4.标准曲线 应该在指定的浓度范围内评价仪器对分析物的响应,获得标准曲线。通过加人已知浓度的分析物(和内标)到空白基质中,制备各浓度的校正标样,其基质应该与目标试验样品基质相同。方法验证中研究的每种分析物和每一分析批,都应该有一条标准曲线。 在进行分析方法验证之前,最好应该了解预期的浓度范围。标准曲线范围应该尽量覆盖预期浓度范围,由定量下限和定量上限(校正标样的最髙浓度)来决定。该范围应该足够描述分析物的药动学。 应该使用至少6个校正浓度水平,不包括空白样品(不含分析物和内标的处理过的基质样品)和零浓度样品(含内标的处理过的基质〉。每个校正标样可以被多次处理和分析。 应该使用简单且足够描述仪器对分析物浓度响应的关系式。空白和零浓度样品结果不应参与计算标准曲线参数。 应该提交标准曲线参数,测定校正标样后回算得出的浓度应一并提交。在方法验证中,至少应该评价3条标准曲线。 校正标样回算的浓度一般应该在标示值的:t l5%以内,定量下限处应该在±20%内。至少75%校正标样,含最少6个有效浓度,应满足上述标准。如果某个校正标样结果不符合这些标准,应该拒绝这一标样,不含这一标样的标准曲线应被重新评价,包括回归分析^ 最好使用新鲜配制的样品建立标准曲线,但如果有稳定性数据支持,也可以使用预先配制并储存的校正标样。 5.准确度 分析方法的准确度描述该方法测得值与分析物标示浓度的接近程度,表示为:(测得值/真实值)x l00?^应采用加人已知 ? 363

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

抗药抗体免疫原性分析方法学验证指导原则(中文版)

抗药抗体免疫原性分析方法学验证指导原则 摘要: 几乎所有的生物制药产品都会引起一定的抗药抗体(anti-drug antibody,ADA)反应,抗药抗体反应可能会降低药物疗效或导致严重的不良反应。在人体内,抗药抗体通常不会引起明显的临床反应。但是对于某些治疗性蛋白质,抗药抗体反应能引起各种临床的不良反应,包括温和事件及严重不良事件。临床前研究表明,抗药抗体能对药物暴露、药物毒性作用、药物代谢动力学、药物效应动力学等造成影响。因此治疗性蛋白质的免疫原性引起了临床医生、药企及监管机构的注意。为了评估生物药物分子的免疫原性,以及将实验结果与临床事件联系起来,在临床前研究和临床研究中,很有必要开发可靠的能够有效评估抗药抗体反应的实验方法。这里方法学验证显得尤为重要,并且方法学验证是药物上市申请必不可少的。现行的监管文件对于免疫分析方法的验证的指导相当有限,特别是缺乏有关免疫原性分析方法的验证的指导。因此,本文对抗药抗体免疫分析方法的验证提供科学的建议。在现有的关于生物分析的规范性文件的基础上加入独特的性能验证。笔者建议采用实验和统计学的方法进行免疫分析的方法学验证。这些建议被视为最佳的例子,旨在促进整个医药行业形成一个更加统一的抗体检测方法。 1.简介: 生物制药产品包括氨基酸聚合物、碳水化合物或核酸,一般通过人细胞系、哺乳动物细胞或细菌进行表达,比常规的小分子药物更大(一般大于1~3KD)。由于以上特性,生物制药产品引起免疫反应的潜力更大。生物制药的免疫原性与产品的内在因素(种属特异性表位、外源性、糖基化程度、聚合或变性程度、杂质和制剂)、外在因素(给药途径、慢性或急性给药、药代动力学及内源性当量)、患者因素(自身免疫性疾病、免疫抑制、和替代疗法)相关。 抗药抗体反应可能会导致严重的临床症状,包括过敏、自身免疫和不同的药代动力学特征(例如,药物中和、生物分布异常和药物清除率增强等均可能会使

方法学验证指导原则

一、准确度 准确度系指采用该方法测定的结果与真实值或参考值接近的程度,一般用回收率(%)表示。准确度应在规定的范围内测定。 1.化学药含量测定方法的准确度 原料药采用对照品进行测定,或用本法所得结果与已知准确度的另一个方法测定的结果进行比较。制剂可在处方量空白辅料中,加入已知量被测物对照品进行测定。如不能得到制剂辅料的全部组分,可向待测制剂中加人已知量的被测物对照品进行测定,或用所建立方法的测定结果与已知准确度的另一种方法测定结果进行比较。准确度也可由所测定的精密度、线性和专属性推算出来。 2.化学药杂质定量测定的准确度 可向原料药或制剂处方量空白辅料中加人已知量杂质进行测定。如不能得到杂质或降解产物对照品,可用所建立方法测定的结果与另一成熟的方法进行比较,如药典标准方法或经过验证的方法。在不能测得杂质或降解产物的校正因子或不能测得对主成分的相对校正因子的情况下,可用不加校正因子的主成分自身对照法计算杂质含量。应明确表明单个杂质和杂质总量相当于主成分的重量比(%) 或面积比(% )。 3.中药化学成分测定方法的准确度 可用对照品进行加样回收率测定,即向已知被测成分含量的供试品中再精密加人一定量的被测成分对照品,依法测定。用实测值与供试品中含有量之差,除以加入对照品量计算回收率。在加样回收试验中须注意对照品的加人量与供试品中被测成分含有量之和必须在标准曲线线性范围之内;加入对照品的量要适当,过小则引起较大的相对误差,过大则干扰成分相对减少,真实性差。 回收率:%= (C - A ) /S X 100% 式中:A为供试品所含被测成分量;B 为加入对照品量; C 为实测值。 4.校正因子的准确度 对色谱方法而言,绝对(或定量)校正因子是指单位面积的色谱峰代表的待测物质的量。待测定物质与所选定的参照物质的绝对校正因子之比,即为相对校正因子。相对校正因子计算法常应用于化学药有关物质的测定、中药材及其复方制剂中多指标成分的测定。校正因子的表示方法很多,本指导原则中的校正因

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

化学药物质量控制分析方法验证技术指导原则

化学药物质量控制分析方法验证技术指导原则【】HGPH 5-1指导原则编号: 化学药物质量控制分析方法验证 技术指导原则 二??四年十一月 目录 一、概 述 ..................................................................... ............................................ 1 二、方法验证的一般原 则 ..................................................................... ................ 2 三、方法验证涉及到的三个主要方 面 (2) ,一,需要验证的检测项 目 ..................................................................... . (2) ,二,分析方 法 ..................................................................... .. (3) ,三,验证内 容 ..................................................................... .......................... 3 四、方法验证的具体内 容 ..................................................................... . (3)

,一,专属 性 ..................................................................... (3) 1、鉴别反 应 ..................................................................... (3) 2、杂质检 查 ..................................................................... (4) 3、含量测 定 ..................................................................... (4) ,二,线 性 ..................................................................... . (5) ,三,范 围 ..................................................................... . (5) 1、含量测 定 ..................................................................... (5) 2、制剂含量均匀度...................................................................... (5)

美国FDA分析方法验证指南中文译稿[1]

1 II. 背景 (2) III. 分析方法的类型 (3) A. 法定分析方法 (3) B. 可选择分析方法 (3) 3 C. 稳定性指示分析 (3) IV. 对照品…………………………………………………………………………… 4 A. 对照品的类型 (4) B. 分析报告单 (4) C. 对照品的界定 (4) V. IND 中的分析方法验证 (6) VI. NDA, ANDA, BLA 和PLA 中分析方法验证的内容和格式 (6) A. 原则 (6) B. 取样 (7) C. 仪器和仪器参数 (7) D. 试剂 (7) E. 系统适应性实验 (7) F. 对照品的制备 (7) G. 样品的制备 (8) H. 分析方法 (8) L. 计算 (8) J. 结果报告 (8) VII. NDA,ANDA,BLA 和PLA 中的分析方法验证 (9) A.非法定分析方法 (9) 1.验证项目 (9) 2. 其它分析方法验证信息 (10) a. 耐用性 (11) b. 强降解实验 (11) c. 仪器输出/原始资料 (11) 3.各类检测的建议验证项目 (13) B.法定分析方法 (15) VIII. 统计分析………………………………………………………………………… 15 A. 总则 (15)

C. 统计 (16) IX. 再验证 (16) X. 分析方法验证技术包:内容和过程…………………………………………… 17 A. 分析方法验证技术包 (17) B. 样品的选择和运输 (18) C. 各方责任 (19) XI. 方法……………………………………………………………………………… 20 A. 高效液相色谱(HPLC) (20) B. 气相色谱(GC) (22) C. 分光光度法,光谱学,光谱法和相关的物理方法 (23) D. 毛细管电泳 (23) E. 旋光度 (24) F. 粒径相关的分析方法 (25) G. 溶出度 (26) H. 其它仪器分析方法 (27) 附件A:NDA,ANDA,BLA 和PLA 申请的内容 (28) 附件B:分析方法验证的问题和延误 (29) 参考文献…………………………………………………………………………………… 30 术语表……………………………………………………………………………………… 32 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.

有关物质分析方法验证技术指导原则

摘要:本文介绍了在对有关物质检查所用的分析方法进行方法学验证时,各项指标的可接受标准,以利于判断该分析方法的可行性。 关键词:有关物质检查分析方法验证可接收标准 药品中的有关物质泛指在药品的生产与储存过程中产生的工艺杂质或降解产物。由于这些有关物质的存在会影响到药品的纯度,进而可能会产生毒副作用,所以有关物质的控制是药品研发的一个重要方面,也是我们在药品审评中一直重点关注的要点之一。而要对有关物质进行严格的控制,就离不开专属性强、灵敏度高的分析方法,这就涉及到分析方法的筛选与验证。从现有的申报资料看,药品研发单位已基本上意识到分析方法验证的重要性,但是对验证时各具体指标是否可行尚没有一个明确的可接受标准,从而难以对验证结果进行评判。为解决这一问题,本文结合国外一些大型药品研发企业在此方面的要求,提出了在对有关物质检查方法进行验证时的可接受标准,供国内的药品研发单位在进行研究时参考。 1.准确度 该指标主要是通过回收率来反映。验证时一般要求根据有关物质的定量限与质量标准中该杂质的限度分别配制三个浓度的供试品溶液各三份(例如某杂质的限度为0.2%,则可分别配制该杂质浓度为0.1%、0.2%和0.3%的杂质溶液),分别测定其含量,将实测值与理论值比较,计算回收率,并计算9个回收率数据的相对标准差

(RSD)。 该项目的可接受的标准为:各浓度下的平均回收率均应在80%-120%之间,如杂质的浓度为定量限,则该浓度下的平均回收率可放宽至70%-130%,相对标准差应不大于10%。 2.线性 线性一般通过线性回归方程的形式来表示。具体的验证方法为:在定量限至一定的浓度范围内配制6份浓度不同的供试液,分别测定该杂质峰的面积,计算相应的含量。以含量为横坐标(X),峰面积为纵坐标(Y),进行线性回归分析。 可接受的标准为:回归线的相关系数(R)不得小于0.990,Y轴截距应在100%响应值的25%以内,响应因子的相对标准差应不大于10%。 3.精密度 1)重复性 配制6份杂质浓度(一般为0.1%)相同的供试品溶液,由一个分析人员在尽可能相同的条件下进行测试,所得6份供试液含量的相对标准差应不大于15%。 2)中间精密度 配制6份杂质浓度(一般为0.1%)相同的供试品溶液,分别由两个分析人员使用不同的仪器与试剂进行测试,所得12个含量数据的相对标准差应不大于20%。 4.专属性 可接受的标准为:空白对照应无干扰,该杂质峰与其它峰应能完

化学药物质量控制分析方法验证技术指导原则

指导原则编号: 【H】G P H 5 -1 化学药物质量控制分析方法验证技术指导原则 (第二稿) 二OO四年三月十九日

目录 一、概述 (4) 二、方法验证的一般原则 (5) 三、方法验证涉及到的三个要素 (5) 1、需要验证的检测项目 (5) 2、分析方法 (6) 3、验证内容 (7) 四、方法验证的具体内容 (7) (一)专属性 (7) 1、鉴别反应 (7) 2、杂质检查 (7) 3、含量测定 (8) (二)线性 (8) (三)范围 (9) 1、含量测定 (9) 2、制剂含量均匀度 (9) 3、溶出度或释放度 (9) 4、杂质 (10) (四)准确度 (10) 1、含量测定 (10) 2、杂质定量试验 (10)

(五)精密度 (11) 1、重复性 (11) 2、中间精密度 (12) 3、重现性 (12) (六)检测限 (12) 1、直观法 (12) 2、信噪比法 (12) (七)定量限 (13) 1、直观法 (13) 2、信噪比法 (13) (八)耐用性 (14) (九)系统适用性试验 (14) 五、方法再验证 (15) 六、方法验证的评价 (16) 1、有关方法验证评价的一般考虑 (16) 2、方法验证的整体性和系统性 (16) 七、参考文献 (17) 八、起草说明 (18) 九、著者 (20)

质量控制分析方法验证的技术指导原则 一、概述 保证药品安全、有效、质量可控是药品研发和评价应遵循的基本原则,其中,对药品进行质量控制是保证药品安全有效的基础和前提。为达到控制质量的目的,需要多角度、多层面来控制产品质量,也就是说要对药物进行多个项目测试,来全面考察产品质量。一般地,每一测试项目可选用不同的分析方法,为使测试结果准确、可靠,必须对所采用的分析方法的科学性、准确性和可行性进行验证,以充分表明分析方法符合测试项目的目的和要求,这就是通常所说的对方法进行验证。 方法验证的目的是判断采用的分析方法是否科学、合理,是否能有效控制产品的内在质量。从本质上讲,方法验证就是根据检测项目的要求,预先设置一定的验证内容,并通过设计合理的试验来验证所采用的分析方法是否满足检测项目的要求。 方法验证在分析方法建立过程中具有重要的作用,并成为质量研究和质量控制的组成部分。只有经过验证的分析方法才能用于控制产品质量,因此方法验证是制订质量标准的基础。方法验证是药物研究过程中的重要内容。 本指导原则重点探讨方法验证的本质,将分析方法验证的要求与所要达到的目的结合起来进行系统和规律性的阐述,重点阐述如何科学合理地进行论证方案的设计。

相关文档
相关文档 最新文档