WHO good practices for research and development facilities ...

32
Working document QAS/20.865/.Rev1 July 2021 1 2 DRAFT WORKING DOCUMENT FOR COMMENTS : 3 4 WHO good practices for 5 research and development facilities 6 of pharmaceutical products 7 8 9 Please send your comments to Dr Steve Estevão Cordeiro, Technical Officer, Norms and Standards for Pharmaceuticals, Technical Standards and Specifications ([email protected]), with a copy to Ms Sinéad Jones ([email protected]) before 31 August 2021. Please use the “Table of Comments” document for this purpose. Our working documents are sent out electronically and they will also be placed on the WHO Medicines website (https://www.who.int/teams/health-product-and-policy-standards/standards-and- specifications/pharmaceuticals/current-projects) for comments under the “Working documents in public consultation” link. If you wish to receive our draft guidelines, please send your email address to [email protected] and your name will be added to our electronic mailing list. 10 11 © World Health Organization 2021 12 13 All rights reserved. 14 15 This is a draft. The content of this document is not final, and the text may be subject to revisions before publication. The 16 document may not be reviewed, abstracted, quoted, reproduced, transmitted, distributed, translated or adapted, in part or 17 in whole, in any form or by any means without the permission of the World Health Organization. 18 19 Please send any request for permission to: Sinéad Jones, Norms and Standards for Pharmaceuticals, Technical Standards and 20 Specifications, Department of Health Products Policy and Standards, World Health Organization, CH-1211 Geneva 27, 21 Switzerland, email: [email protected]. 22 23 The designations employed and the presentation of the material in this draft do not imply the expression of any opinion 24 whatsoever on the part of the World Health Organization concerning the legal status of any country, territory, city or area or 25 of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps represent approximate 26 border lines for which there may not yet be full agreement. 27 28 The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or 29 recommended by the World Health Organization in preference to others of a similar nature that are not mentioned. Errors 30 and omissions excepted, the names of proprietary products are distinguished by initial capital letters. 31 32 All reasonable precautions have been taken by the World Health Organization to verify the information contained in this draft. 33 34 However, the printed material is being distributed without warranty of any kind, either expressed or implied. The responsibility 35 for the interpretation and use of the material lies with the reader. In no event shall the World Health Organization be liable 36 for damages arising from its use. 37 38 This draft does not necessarily represent the decisions or the stated policy of the World Health Organization. 39 40

Transcript of WHO good practices for research and development facilities ...

Page 1: WHO good practices for research and development facilities ...

Working document QAS/20.865/.Rev1 July 2021

1

2

DRAFT WORKING DOCUMENT FOR COMMENTS: 3

4

WHO good practices for 5

research and development facilities 6

of pharmaceutical products 7

8

9

Please send your comments to Dr Steve Estevão Cordeiro, Technical Officer, Norms and Standards for Pharmaceuticals, Technical Standards and Specifications ([email protected]), with a copy to Ms Sinéad Jones ([email protected]) before 31 August 2021. Please use the “Table of Comments” document for this purpose.

Our working documents are sent out electronically and they will also be placed on the WHO Medicines website (https://www.who.int/teams/health-product-and-policy-standards/standards-and-specifications/pharmaceuticals/current-projects) for comments under the “Working documents in public consultation” link. If you wish to receive our draft guidelines, please send your email address to [email protected] and your name will be added to our electronic mailing list.

10 11 © World Health Organization 2021 12 13 All rights reserved. 14 15 This is a draft. The content of this document is not final, and the text may be subject to revisions before publication. The 16 document may not be reviewed, abstracted, quoted, reproduced, transmitted, distributed, translated or adapted, in part or 17 in whole, in any form or by any means without the permission of the World Health Organization. 18 19 Please send any request for permission to: Sinéad Jones, Norms and Standards for Pharmaceuticals, Technical Standards and 20 Specifications, Department of Health Products Policy and Standards, World Health Organization, CH-1211 Geneva 27, 21 Switzerland, email: [email protected]. 22 23 The designations employed and the presentation of the material in this draft do not imply the expression of any opinion 24 whatsoever on the part of the World Health Organization concerning the legal status of any country, territory, city or area or 25 of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps represent approximate 26 border lines for which there may not yet be full agreement. 27 28 The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or 29 recommended by the World Health Organization in preference to others of a similar nature that are not mentioned. Errors 30 and omissions excepted, the names of proprietary products are distinguished by initial capital letters. 31 32 All reasonable precautions have been taken by the World Health Organization to verify the information contained in this draft. 33 34 However, the printed material is being distributed without warranty of any kind, either expressed or implied. The responsibility 35 for the interpretation and use of the material lies with the reader. In no event shall the World Health Organization be liable 36 for damages arising from its use. 37 38 This draft does not necessarily represent the decisions or the stated policy of the World Health Organization. 39

40

Page 2: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 2

SCHEDULE FOR DRAFT WORKING DOCUMENT QAS/20.865: 41

WHO good practices for research and development 42

facilities of pharmaceutical products 43

Description of Activity Date

Following a recommendation by WHO Prepublication Inspection Team, the Fifty-fifth Expert Committee on Specifications for Pharmaceutical Preparations (ECSPP) recommended that the WHO Secretariat should develop a new guidance on Good practices in research and development.

October 2020

Preparation of first draft working document. October 2020

Mailing of working document to the Expert Advisory Panel on the International Pharmacopoeia and Pharmaceutical Preparations (EAP) inviting comments and posting of the working document on the WHO website for public consultation

November 2020

Consolidation of comments received and review of feedback. Preparation of working document for discussion.

January 2021

Discussion of the feedback received on the working document in a virtual meeting with an expert working group

February-March 2021

Preparation of working document for next round of public consultation.

March 2021

Mailing of revised working document inviting comments, including to the EAP, and posting the working document on the WHO website for a second round of public consultation.

April 2021

Consolidation of comments received and review of feedback. Preparation of working document for discussion.

June 2021

Discussion of comments in the virtual meeting on Good practices for health product manufacture and inspection

28 June- 2 July 2021

Preparation of working document for next round of public consultation.

July 2021

Mailing of revised working document inviting comments, including to the EAP, and posting the working document on the WHO website for a second round of public consultation.

July – August 2021

Consolidation of comments received and review of feedback. Preparation of working document for discussion in the ECSPP.

September – October 2021

Page 3: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 3

44

45

Presentation to the Fifty-sixth meeting of the ECSPP. TBD

Any other follow-up action as required.

Page 4: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 4

WHO good practices for research and 46

development facilities of 47

pharmaceutical products 48

49

50

Background 51

52

In view of the need for the development of health products, including the research and development 53

for the treatment of COVID-19 therapies, the World Health Organization (WHO) Prequalification 54

Inspection Services Team (PQT INS) raised the urgency for the development of life cycle appropriate 55

good practices text to address the manufacturing of developmental batches, pilot batches and the 56

sequential stability data that are submitted in product applications (dossiers) for marketing 57

authorization and the prequalification of medical products. 58

59

There is currently no other specific WHO guideline which addresses this matter. The data collected from 60

these batches influence the following aspects of the product: 61

• stability; 62

• process validation; and 63

• analytical method development and validation. 64

65

66

1. Introduction 67

2. Scope 68

3. Glossary 69

4. Quality management 70

5. Quality risk management 71

6. Sanitation and hygiene 72

7. Qualification and validation 73

8. Outsourced activities 74

9. Self-inspection and quality audits 75

10. Personnel 76

11. Training 77

12. Premises 78

13. Equipment and instruments 79

14. Materials 80

Page 5: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 5

15. Documentation 81

16. Processing and process design 82

17. Quality control 83

18. Stability studies 84

19. Analytical procedure development 85

20. Technology transfer 86

21. Life cycle approach 87

22. Cleaning procedure development, cleaning verification and cleaning validation 88

89

Abbreviations 90

References 91

Further reading 92

93

94

Page 6: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 6

1. Introduction 95

96

1.1. With an ever increasing awareness of the risks in pharmaceutical production and control and 97

the life cycle approaches being followed, greater emphasis is being placed on ensuring that the 98

research and development of products are appropriately controlled and documented. 99

100

1.2. Consequently, it is necessary that manufacturers of pharmaceutical products submit all relevant 101

data and information related to the their development, including the facilities used, the 102

experimental designs employed in the validations of manufacturing processes and quality 103

control procedures, to the regulators for review to ensure that the facilities, quality systems, 104

data and information meet the appropriate standards and good practices (GxP). 105

106

1.3. This document intends to provide guidance on good manufacturing practices (GMP) to research 107

and development facilities. It further aims to ensure that the correct systems are followed, 108

ensuring appropriateness, reliability and the quality of products, processes, procedures and 109

data. This further helps to help ensure that products meet the requirements for safety, efficacy 110

and quality that they purport to possess. 111

112

1.4 In addition to product development, other activities, including the production of pilot scale 113

batches; process validation; cleaning procedure development; cleaning validation studies; as 114

well as stability studies, are often undertaken in such facilities. 115

116

1.5 The World Health Organization (WHO) document entitled Good manufacturing practices for 117

investigational pharmaceutical products for clinical trials in humans (1) specifically addresses 118

the requirements and recommendations for products used in clinical trials. Other WHO 119

guidelines address specific requirements and recommendations, including but not limited to, 120

data integrity, stability testing, analytical method validation, cleaning validation and the 121

technology transfer (TOT) (see References and Further reading sections). 122

123

1.6 This document should be read in conjunction with other WHO GMP guidelines, as referenced in 124

the document (2-9). Other documents of interest are also listed under the section “Further 125

reading”. 126

127

Page 7: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 7

2. Scope 128

129

2.1. This guideline is specifically applicable to research and development facilities of 130

pharmaceutical products procedures, processes and data that are intended for transfer and 131

submission for approval in marketing authorization applications, process validation, TOT (10)-132

related activities, validation (7), quality control laboratory activities (11) such as stability testing 133

and development, and validation of cleaning procedures (see Figure 1 and section 4 below). 134

135

2.2. The main focus of this document is to provide for GxP in the production and control of pre-136

clinical and not for human use batches, manufactured in pharmaceutical formulation and 137

development facilities, where these are directly supporting; for example, shelf life claims, 138

animal studies or validation activities. The principles described in this document may be 139

applied in facilities where other products, such biopharmaceutical products, vaccines and 140

medical devices, are manufactured. 141

142

2.3. This guide excludes whole cells, whole blood and plasma, blood and plasma derivatives (plasma 143

fractionation), medicinal gases, radiopharmaceuticals and gene therapy products. 144

145

2.4. The GxP outlined below are to be considered general guides and they may be adapted to meet 146

individual needs. The equivalence of alternative approaches, however, should be 147

demonstrated. 148

149

2.5. In this guide, the term “should” indicates recommendations that are expected to apply unless 150

shown to be inapplicable or replaced by an alternative demonstrated to provide an acceptable 151

level of control. 152

153

2.6. This guide, as a whole, does not cover safety aspects for the personnel engaged in the research 154

and development nor the aspects of protection of the environment. These controls are 155

inherent responsibilities of the manufacturer and are governed by national laws. 156

157

2.7. This guide is not intended to define registration requirements or modify pharmacopoeial 158

requirements or other guideline recommendations. For details on process development, it is 159

recommended that other guidelines, such as those published by The International Council for 160

Page 8: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 8

Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), be read 161

in conjunction with this document. 162

163

2.8. This guide does not affect the ability of the responsible regulatory agency to establish specific 164

registration or filing requirements. All commitments in registration and filing documents must 165

be met. This document provides information to consider for a risk- and science-based 166

approach in the research and development of pharmaceutical products. 167

168

2.9. Due to the nature of development work, and an increasing expectation for compliance with 169

standards in manufacture, the guidance in this document would normally be applied based on 170

risk assessment, in an increasing manner, from development to commercial batch 171

manufacturing. The stringency of GMP in research and development should increase as the 172

process proceeds from early development work to the final steps of development and 173

formulation, stability testing, process validation and cleaning validation. 174

175

Figure 1. Application of this guide 176

177

Early research – Research – Development/formulation – Registration batches 178

179

180

181

*The principles described in this guideline are applied, based on risk management principles, in an 182

increased manner from early research to development to registration batches 183

184

3. Glossary 185

186

The definitions given below apply to the terms used in this guideline. They may have different 187

meanings in other contexts. 188

189

batch (or lot). A defined quantity of starting material, packaging material or product processed in a 190

single process or series of processes so that it is expected to be homogeneous. It may sometimes be 191

necessary to divide a batch into a number of sub-batches which are later brought together to form a 192

final homogeneous batch. In the case of terminal sterilization, the batch size is determined by the 193

Increased compliance with Good Manufacturing Practices*

Page 9: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 9

capacity of the autoclave. In continuous manufacture, the batch must correspond to a defined fraction 194

of the production, characterized by its intended homogeneity. The batch size can be defined either as 195

a fixed quantity or as the amount produced in a fixed time interval. 196

197

batch records. All documents associated with the manufacture of a batch of bulk product or finished 198

product. They provide a history of each batch of product and of all circumstances pertinent to the 199

quality of the final product. 200

201

bulk product. Any product that has completed all processing stages up to, but not including, final 202

packaging. 203

204

calibration. The set of operations that establish, under specified conditions, the relationship between 205

values indicated by an instrument or system for measuring (especially weighing), recording and 206

controlling, or the values represented by a material measure, and the corresponding known values of 207

a reference standard. Limits for acceptance of the results of measuring should be established. 208

209

cleaning verification. The act of demonstrating that cleaning was done to an acceptable level; for 210

example, between two batches. 211

212

contamination. The undesired introduction of impurities of a chemical or microbiological nature, or of 213

foreign matter, into or on to a starting material or intermediate during production, sampling, packaging 214

or repackaging, storage or transport. 215

216

cross-contamination. Contamination of a starting material, intermediate product or finished product 217

with another starting material or product during production. 218

219

finished product. A finished dosage form that has undergone all stages of manufacture, including 220

packaging in its final container and labelling. 221

222

in-process control. Checks performed during production in order to monitor and, if necessary, to adjust 223

the process to ensure that the product conforms to its specifications. The control of the environment 224

or equipment may also be regarded as a part of in-process control. 225

226

Page 10: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 10

intermediate product. A partly processed product that must undergo further manufacturing steps 227

before it becomes a bulk product. 228

229

manufacture/manufacturing. Includes all operations of receipt of materials, production, packaging, 230

repackaging, labelling, relabelling, quality control, release, storage, distribution and related controls. 231

232

manufacturer. A company that carries out operations such as production, packaging, repackaging, 233

labelling and relabelling of pharmaceuticals. 234

235

marketing authorization (product licence, registration certificate). A legal document issued by the 236

competent medicines regulatory authority that establishes the detailed composition and formulation 237

of the product and the pharmacopoeial or other recognized specifications of its ingredients and of the 238

final product itself, and includes details of packaging, labelling and shelf life. 239

240

master formula. A document or set of documents specifying the starting materials with their quantities 241

and the packaging materials, together with a description of the procedures and precautions required 242

to produce a specified quantity of a finished product as well as the processing instructions, including 243

the in-process controls. 244

245

master record. A document or set of documents that serve as a basis for the batch documentation 246

(blank batch record). 247

248

packaging. All operations, including filling and labelling, that a bulk product has to undergo in order to 249

become a finished product. The filling of a sterile product under aseptic conditions, or a product 250

intended to be terminally sterilized, would not normally be regarded as part of packaging. 251

252

packaging material. Any material, including printed material, employed in the packaging of a 253

pharmaceutical, but excluding any outer packaging used for transportation or shipment. Packaging 254

materials are referred to as primary or secondary according to whether or not they are intended to be 255

in direct contact with the product. 256

257

Page 11: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 11

pharmaceutical product. Any material or product intended for human or veterinary use presented in 258

its finished dosage form or as a starting material for use in such a dosage form that is subject to control 259

by pharmaceutical legislation in the exporting state and/or the importing state. 260

261

production. All operations involved in the preparation of a pharmaceutical product, from receipt of 262

materials through processing, packaging and repackaging, labelling and relabelling, to completion of 263

the finished product. 264

265

quality audit. An examination and assessment of all or part of a quality system with the specific purpose 266

of improving it. A quality audit is usually conducted by outside or independent specialists or a team 267

designated by the management for this purpose. Such audits may also be extended to suppliers and 268

contractors. 269

270

quality risk management. A systematic process for the assessment, control, communication and review 271

of risks. 272

273

specification. A list of detailed requirements with which the products or materials used or obtained 274

during manufacture have to conform. They serve as a basis for quality evaluation. 275

276

standard operating procedure (SOP). An authorized written procedure giving instructions for 277

performing operations not necessarily specific to a given product or material (e.g. equipment 278

operation, maintenance and cleaning; validation; cleaning of premises and environmental control; 279

sampling and inspection). Certain SOPs may be used to supplement product-specific master and batch 280

production documentation. 281

282

starting material. Any substance of a defined quality used in the production of a pharmaceutical 283

product, but excluding packaging materials. 284

285

validation. The action of proving, in accordance with the principles of GMP, that any procedure, 286

process, equipment, material, activity or system actually leads to the expected results. 287

288

289

290

Page 12: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 12

4. Quality management 291

292

4.1 There should be a quality management system encompassing adequate resources, a written 293

organizational structure and procedures to follow. 294

295

4.2 All parts of the quality system should be adequately resourced and maintained, including with 296

sufficient competent personnel, suitable premises, equipment and facilities. The necessary 297

resources should include, for example: 298

a) a sufficient number of appropriately qualified, trained personnel; 299

b) adequate premises and space; 300

c) suitable equipment and services; 301

d) appropriate materials, containers and labels; and 302

e) suitable storage and transport. 303

304

4.3 Roles, responsibilities and authorities should be defined, communicated and implemented. 305

306

4.4 The quality system should facilitate innovation and continual improvement and strengthen the 307

link between pharmaceutical development and manufacturing activities. 308

309

4.5 Initial research, as well as development activities, should be defined and documented. 310

Development activities, including initial research, should be adequately documented. Controls 311

should be commensurate with the stage of product development (i.e. for testing options or at 312

a final stage for further use where the guideline on Good manufacturing practices for 313

investigational pharmaceutical products for clinical trials in humans applies). 314

315

4.6 The quality system should ensure, as applicable and according to the stage of research and 316

development, that: 317

a) managerial responsibilities are clearly specified in job descriptions; 318

b) personnel are trained; 319

c) instructions and procedures are written in clear and unambiguous language, and 320

followed; 321

d) procedures are correctly carried out; 322

Page 13: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 13

e) records are made (manually and/or by recording instruments) during production and 323

testing; 324

f) records are maintained; 325

g) there is a system for quality risk management (QRM) which is applied, as appropriate; 326

h) arrangements are made for the manufacture, supply and use of the correct starting 327

and packaging materials; 328

i) all necessary controls on starting materials, intermediate products, bulk products and 329

other in-process controls are carried out; 330

j) calibrations and validations are carried out where appropriate; 331

k) the product and process knowledge is managed; 332

l) products are designed and developed in accordance with applicable GxP; 333

m) development procedures should be documented; 334

n) cleaning procedures are developed, verified and validated, where appropriate; 335

o) stability testing is done following written procedures and protocols; and 336

p) data meet ALCOA+ requirements, where applicable. 337

338

4.7 There should be periodic management review with the involvement of senior management. 339

340

5. Quality risk management 341

342

5.1 A system of quality risk management (QRM) should be implemented. The system should 343

ensure that risks are identified based on scientific knowledge and experience. The appropriate 344

controls should be identified and implemented to mitigate risks. 345

346

5.2 The level of effort, formality and documentation of the QRM process is commensurate with 347

the level of risk and the stage from research to development, to commercial batch 348

manufacturing and control (see Figure 1). 349

350

5.3 Systems should be in place to manage and minimize the risks inherent in research and 351

development in order to ensure the ultimate quality, safety and efficacy of products and the 352

reliability of data. 353

354

Page 14: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 14

6. Sanitation and hygiene 355

356

6.1 Procedures should be implemented to maintain sanitation and hygiene. The scope of 357

sanitation and hygiene covers personnel, premises, equipment and apparatus, production 358

materials and containers, and products for cleaning and disinfection. 359

360

6.2 Potential sources of contamination should be identified and controlled. 361

362

7. Qualification and validation 363

364

7.1 Where qualification and validation are performed, the scope and extent should be appropriate 365

using a risk-based approach. 366

367

7.2 The qualification and validation policy and approach should be defined and documented, for 368

example, in a validation master plan. 369

370

7.3 Where qualification and validation is carried out, the responsibility of performing validation 371

should be clearly defined. 372

373

7.4 Where process validation, cleaning validation and analytical procedure validation is done as a 374

part of development, procedures and protocols should be followed. Reports should be 375

available and retained. 376

377

8. Outsourced activities 378

379

8.1 Outsourced activities should be correctly defined, agreed and controlled through a written 380

agreement. 381

382

8.2 All responsibilities and arrangements for activities, such as quality control (QC) testing and 383

technology transfer, should be clearly described. 384

385

Page 15: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 15

The contract giver 386

387

8.3 The contract giver is responsible for assessing the suitability and competence of the contract 388

acceptor to successfully carry out the work or tests required and for approval of the contract 389

activities. 390

391

8.4 The contract giver should provide the contract acceptor with all the information necessary to 392

carry out the contracted operations correctly. 393

394

8.5 The contract giver should ensure that the contract acceptor is fully aware of any hazards 395

associated with the product, work or tests. 396

397

8.6 The contract giver should review and assess the records and results related to the outsourced 398

activities. 399

400

8.7 The contract giver is responsible for ensuring that the contract accepter understands that its 401

activities may be subject to inspection by the competent authorities. 402

403

The contract accepter 404

405

8.8 The contract accepter must have adequate premises, equipment, knowledge, experience and 406

competent, trained personnel to satisfactorily carry out the work ordered by the contract giver. 407

408

8.9 The contract accepter should not pass to a third party any of the work entrusted under the 409

contract without the contract giver’s prior evaluation and approval of the arrangements. 410

411

8.10 The contract accepter should agree to a period of time for retention of documents and data 412

prior to archival or returning to the contract giver. 413

414

The agreement 415

416

8.11 The technical aspects of the agreement should be drawn up by competent persons suitably 417

knowledgeable in the field of law, research, development and GMP. 418

Page 16: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 16

8.12 The agreement should define the roles and responsibilities of all parties. 419

420

8.13 The agreement should permit the contract giver to audit the facilities and activities of the 421

contract accepter. 422

423

9. Self-inspection and quality audits 424

425

9.1 There should be a written self-inspection programme. 426

427

9.2 Self-inspections should be performed routinely and may be, in addition, performed on special 428

occasions. 429

430

9.3 The team responsible for self-inspection should consist of personnel with the appropriate 431

knowledge and experience, free from bias. 432

433

9.4 Self-inspections should cover at least the following items: 434

a) personnel; 435

b) premises including personnel facilities; 436

c) maintenance of buildings and equipment; 437

d) storage of starting materials and finished products; 438

e) equipment; 439

f) production and in-process controls; 440

g) QC; 441

h) documentation; 442

i) data and data integrity; 443

j) sanitation and hygiene; 444

k) qualification and validation; 445

l) calibration of instruments or measurement systems; 446

m) control of labels; and 447

n) results of previous self-inspections and any corrective steps taken. 448

449

Page 17: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 17

9.5 The outcome of the self-inspection should be documented. Corrective actions and preventive 450

actions should be identified and implemented within a defined timeline. There should be an 451

effective follow-up programme. 452

453

9.6 Self-inspections may be supplemented by quality audits. 454

455

10. Personnel 456

457

10.1 Individual responsibilities should be clearly defined and understood by the persons concerned 458

and recorded as written descriptions. 459

460

10.2 All personnel should be aware of the principles of this guideline and other applicable GxP. 461

462

10.3 Steps should be taken to prevent unauthorized people from entering storage, production and 463

QC areas. 464

465

10.4 Smoking, eating, drinking, chewing and keeping plants, food, drink, smoking material and 466

personal medicines should not be permitted in any area where they might adversely influence 467

product quality. 468

10.5 The appropriate protective garments should be worn, based on operation performed and risk. 469

470

10.6 Personnel who are ill should not engage in the manufacture of pharmaceutical products. 471

472

11. Training 473

474

11.1 Training should be provided in accordance with a written programme that covers topics such 475

as the theory and practice of GMP and the duties assigned to them. The appropriate task-476

related training should be further provided based on technical requirements and activities 477

undertaken. 478

479

11.2 The effectiveness of training should be assessed. 480

481

Page 18: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 18

11.3 Training and assessment records should be kept. 482

483

11.4 Where appropriate, specific training should be given on the handling and segregation of highly 484

active, toxic, infectious or sensitizing materials and the need for separate, dedicated facilities 485

where these are required. 486

487

12. Premises 488

489

12.1 Premises should be located, designed, constructed, adapted and maintained to suit the 490

operations to be carried out. 491

492

12.2 The layout and design should aim to minimize the risk of errors and permit effective cleaning 493

and maintenance in order to avoid cross-contamination, build-up of dust or dirt and, in general, 494

any adverse effect on the products and activities. 495

496

12.3 Measures should be taken to avoid cross-contamination and to facilitate cleaning. 497

498

12.4 The premises should be cleaned according to detailed procedures. Records should be 499

maintained. 500

501

12.5 The electrical supply, lighting, temperature, humidity and ventilation should be appropriate. 502

503

12.6 Toilets, rest and refreshment rooms should be separate from production and control areas. 504

505

12.7 Storage areas should be of sufficient capacity with proper separation and segregation between 506

materials. 507

508

12.8 Storage areas should be clean and dry, designed or adapted to ensure the required storage 509

conditions are maintained. Conditions should be controlled, monitored and recorded, where 510

appropriate. 511

512

12.9 Certain materials, such as highly active, radioactive materials and narcotics, should be stored 513

in safe and secure areas. 514

Page 19: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 19

12.10 Materials identified for testing should be sampled and analysed. 515

516

12.11 The stages in production, including weighing, compounding, and packaging, should be done in 517

a manner to prevent contamination, cross-contamination and mix-ups. 518

519

12.12 QC areas should be separated from production areas. They should be designed to suit the 520

operations to be carried out in them. There should be sufficient space, instruments, 521

equipment and the appropriate reference materials, solvents and reagents 522

523

12.13 Poisons or pesticides should not be stored or used in product manufacturing areas. 524

525

13. Equipment and instruments 526

527

13.1 The equipment and instruments should be located, designed, constructed, adapted and 528

maintained to suit the operations to be carried out. They should allow for effective cleaning 529

and maintenance in order to avoid cross-contamination and a build-up of dust or dirt. 530

531

13.2 Pipework, instruments and devices should be adequately marked. 532

533

13.3 Measuring equipment should be available for production and control operations and, where 534

necessary, should be calibrated, verified and serviced on a scheduled basis. Records should be 535

maintained. 536

537

13.4 The equipment and instruments should be thoroughly cleaned on a scheduled basis. 538

539

13.5 Defective equipment and instruments should be removed from operational areas or be clearly 540

labelled as defective in order to prevent use. 541

542

14. Materials 543

544

14.1 Materials should be purchased from approved suppliers. 545

546

Page 20: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 20

14.2 Where so identified, materials should be quarantined immediately after receipt, sampled and 547

tested. 548

549

14.3 Materials within their shelf life should be used. 550

551

14.4 Materials should be stored under the appropriate conditions as specified on their labels and in 552

an orderly fashion to permit segregation. 553

554

14.5 The dispensing of materials for the production of a batch should be recorded. Materials should 555

be accurately weighed or measured into clean and properly labelled containers. 556

557

14.6 No materials used for operations, such as cleaning, the lubrication of equipment and pest 558

control, should come into direct contact with the product. Where possible, such materials 559

should be of a suitable grade (e.g. food grade) to minimize health risks. 560

561

14.7 All materials, including water, should be suitable for its intended use. 562

563

14.8 Packaging and printed materials should be stored in secure conditions so as to exclude the 564

possibility of unauthorized access. 565

566

14.9 Intermediate and bulk products should be kept under appropriate conditions. 567

568

14.10 Finished products should be stored under suitable conditions and appropriately segregated. 569

570

14.11 Rejected materials and products should be clearly marked as such. They should be handled in 571

an appropriate and timely manner. Whatever action is taken should be approved by 572

authorized personnel and recorded. 573

574

14.12 Toxic substances and flammable materials should be stored in suitably designed, separate, 575

enclosed containers and, as required, by national legislation. 576

577

14.13 All waste materials should be stored in a safe manner and disposed of at regular intervals to 578

avoid accumulation. 579

Page 21: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 21

15. Documentation 580

581

15.1 Documentation includes procedures for materials and methods of production and control. The 582

design and use of documents depend upon the research and development facility. 583

584

15.2 Documents should be designed, prepared, reviewed and authorized for use. 585

586

15.3 Standard operating procedures (SOP) should be reviewed periodically and kept up-to-date. 587

Superseded documents should be retained for a defined period of time. 588

589

15.4 Entries of data and information should be clear and legible and meet ALCOA+ principles, as 590

described above. 591

592

15.5 GxP data (including records for storage) may be recorded by electronic data-processing 593

systems or by photographic or other reliable means. Batch production and control records 594

should be protected throughout the defined period of retention. 595

596

15.6 Labels should be clear, unambiguous and in the company’s agreed format. 597

598

15.7 There should be appropriately authorized and dated specifications, including tests on identity, 599

purity and quality, for starting materials and for finished products, as appropriate. 600

601

15.8 Pharmacopoeias, reference standards, reference spectra and other reference materials should 602

be available, where applicable. 603

604

15.9 Specifications should contain appropriate information such as the designated name; internal 605

code reference; and qualitative and quantitative requirements with acceptance criteria. Other 606

data may be added to the specification. 607

608

15.10 The packaging material should be examined for compliance with the specification, as 609

appropriate. 610

611

Page 22: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 22

15.11 Specifications for intermediate and bulk products should be available where the need has been 612

identified, as appropriate. 613

614

15.12 Specifications for finished products should be available and include the required information, 615

where available. 616

617

15.13 A master formula or batch recipe, containing the relevant information, should be available for 618

the product and batch size. 619

620

15.14 Packaging instructions should exist for the products to be packed. 621

622

15.15 A batch processing record should be kept for each batch processed. 623

624

15.16 During processing, detailed information should be recorded at the time each action is taken. 625

Upon completion, the record should be dated and signed by the person responsible in 626

accordance with data integrity expectations. 627

628

15.17 A batch packaging record should be kept for each batch packed. 629

630

15.18 SOP and corresponding records, where required, should be available. These include, but are 631

not limited to, for example: 632

a) equipment assembly and cleaning; 633

b) personnel training, clothing and hygiene; 634

c) maintenance; 635

d) sampling; 636

e) analytical apparatus and instrument calibration; 637

f) testing; 638

g) rejection; and 639

h) pest control. 640

641

15.19 Before any processing operation is started, steps should be taken to ensure that the work area 642

and equipment are clean and free from any starting materials, products, product residues and 643

labels or documents not required for the current operation. 644

Page 23: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 23

16. Processing and process design 645

646

Processing 647

648

Note: For more details on specific aspects relating to process development, see ICH Q 8 (12) and ICH 649

Q11 (13). 650

651

16.1 The selection of the starting materials and manufacturing process should be carefully 652

considered in order to ensure that the intended product will meet the intended standards of 653

safety, efficacy and quality in a consistent manner. 654

655

16.2 Knowledge management and risk assessment principles should be applied. Quality attributes, 656

critical quality attributes, process parameters and critical process parameters should be 657

defined and documented once sufficient data are available. 658

659

16.3 The design of experiments should cover identified variables. 660

661

Process design 662

663

Note: For details on process validation, see WHO Technical Report Series, No. 1019, Annex 3, Appendix 664

7, 2019 (14) as well as EU (15) and FDA Guidelines (16). 665

666

16.4 Process design is usually initiated by research and development facilities. This stage of process 667

validation is also referred to as “process design”. (In a traditional or historical approach, this 668

was often referred to as “prospective validation”.) 669

670

16.5 Product development activities provide key inputs to the process design stage. Laboratory or 671

pilot-scale models designed to be representative of the commercial process can be used to 672

estimate variability. 673

674

16.6 Process design should normally cover the design of experiments, process development, the 675

manufacture of products for use in clinical trials, pilot-scale batches and technology transfer. 676

677

Page 24: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 24

16.7 Process design should be verified during product development. Process design should cover 678

aspects for the selection of materials; expected production variation; selection of production 679

technology/process and qualification of the unitary processes that form the manufacturing 680

process as a whole; selection of in-process controls; tests; inspection; and its suitability for the 681

control strategy. 682

683

16.8 Where the validation data are intended to be used in applications for marketing authorizations, 684

all batch data, results and related information should be clear, detailed and in compliance with 685

ALCOA+. 686

687

17. Quality control 688

689

17.1 There should be adequate resources available to ensure that all the quality control (QC) 690

arrangements are effectively and reliably carried out. 691

692

17.2 Activities and responsibilities of the QC unit include: 693

a) sampling and testing (e.g. starting materials, packaging materials, intermediate 694

products, bulk products and finished products); 695

b) performing the necessary qualification and validation; 696

c) evaluating, maintaining and storing reference materials; 697

d) ensuring that stability programme and testing is done; and 698

e) conducting environmental monitoring. 699

700

17.3 The appropriate records should be kept, demonstrating that all the required activities were 701

performed. 702

703

17.4 Sufficient samples of materials and products should be retained for a defined period of time. 704

705

17.5 The appropriate reference standards should be used. Standards should be stored in an 706

appropriate way. 707

708

17.6 Whenever official reference standards exist, these should preferably be used. 709

710

Page 25: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 25

17.7 Where secondary and working standards are established and used, these should be tested at 711

regular intervals to ensure that they are fit for their intended use. 712

713

17.8 Reference standards should be appropriately labelled with at least the following information: 714

a) name of the material; 715

b) batch or lot number and control number; 716

c) date of preparation; 717

d) shelf life; 718

e) potency; and 719

f) storage conditions. 720

721

18. Stability studies 722

723

Note: See guideline on stability testing of active pharmaceutical ingredients and finished 724

pharmaceutical products , WHO Technical Report Series, No. 1010, Annex 10, 2018 (17). 725

726

18.1 Where stability determination is initiated by research and development organizations, a 727

written programme should be developed and implemented to include elements such as: 728

a) a complete description of the medicine involved in the study; 729

b) the complete set of testing procedure, parameters and limits; 730

c) attributes such as potency or assay, degradation products and physical characteristics; 731

d) evidence that these tests indicate stability; 732

e) the testing schedule for each medicine; 733

f) provision for special storage conditions; and 734

g) provision for adequate sample retention. 735

736

18.2 Sampling should be done in accordance with written procedures. 737

738

18.3 Sample preparation and testing procedures should be detailed and followed. Any deviations 739

from the procedures should be clearly documented. 740

741

18.4 The results and data generated should be documented and include the evaluation and the 742

conclusions of the study. 743

Page 26: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 26

744

18.5 Where stability data are intended to be used in applications for marketing authorizations, all 745

batch data, results and related information should be clear, detailed and in compliance with 746

ALCOA+. 747

748

18.6 Records should be maintained for a defined period of time. 749

750

19. Analytical procedure development 751

752

19.1 Analytical procedures developed by research and development organizations should be 753

appropriately recorded. 754

755

19.2 Analytical procedures developed by research and development facilities should be 756

documented in sufficient detail to facilitate their successful transfer, when required. 757

758

19.3 Analytical procedures should be appropriately validated as fit for purpose. 759

760

Note: For details on analytical procedure validation, see WHO Technical Report Series, No. 1019, Annex 761

3, Appendix 4, 2019 (18). 762

763

20. Technology transfer 764

765

Note: For details on technology transfer, see WHO Technical Report Series, No. 961, Annex 7, 2011 766

(update in progress) (10). 767

768

20.1 Development work, including programmes, procedures, protocols, specifications, process 769

design and validation from research and development facilities, may be transferred to 770

production and QC sites. 771

772

20.2 Data and information relating to equipment, instruments, manufacturing and testing should 773

be in an appropriate level of detail, traceable and available. 774

775

Page 27: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 27

20.3 Authorized procedures should be followed when transferring technology from research and 776

development organizations to production and QC facilities. 777

778

21. Life cycle approach 779

780

21.1 Industry should implement policies and procedures that will encourage science-based and risk-781

based approaches in product research and development. 782

783

21.2 Continual improvement should be encouraged across the entire product life cycle. 784

785

21.3 Knowledge gained from the commercial manufacturing of a product, as well as knowledge 786

gained from other products, can be used to further improve process understanding and 787

process performance. 788

789

21.4 New technologies and the review and interpretation of statistical evaluation of results from 790

process design, validation and other processes, as well as other applicable data and 791

information, should be considered in order to encourage continual improvement during the 792

process development stage of the life cycle of the product. 793

794

21.5 Where appropriate, these should be shared and transferred to commercial manufacturing 795

facilities. 796

797

22. Cleaning procedure development, cleaning 798

verification and cleaning validation 799

800

Note: For details on cleaning validation, see WHO Technical Report Series, No. 1019, Annex 3, Appendix 801

3 (19), 2019 and the WHO Points to consider when including HBELs in cleaning validation, TRS 1033, 802

Annex 2, 2021 (20). 803

804

Page 28: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 28

22.1 Research and development facilities are often involved in the development and validation of 805

cleaning procedures. QRM principles should be applied in cleaning procedure development 806

and cleaning validation. 807

808

22.2 The development of cleaning procedures should include cleanability. 809

810

22.3 Where preparatory work for cleaning validation is done in research and development facilities 811

with a view of technology transfer, the commercial manufacturing sites consideration should 812

be given for inclusion of Health Based Exposure Limits (HBELs) in the approach. 813

814

22.4 The sampling of procedures should include swab and rinse samples. Maximum Safe Residue, 815

Maximum Safe Surface Residue and Visible Residue Limits should be considered in the new 816

cleaning validation approach. 817

818

22.5 The development of the analytical procedures to be used in the testing for residues should be 819

appropriately documented. The procedures should be validated. 820

821

22.6 The procedures for sampling and testing, and the results obtained, should meet ALCOA+ 822

principles. The data and information should be retained over the life cycle of the product. 823

22.7 Procedures and protocols should be followed for the TOT to commercial manufacturing sites. 824

825

22.8 Records should be maintained. 826

827

Abbreviations 828

829

ALCOA+ attributable, legible, contemporaneous, original and accurate, complete, consistent, 830

enduring, and available 831

GMP Good manufacturing practices 832

GxP Good practices 833

ICH International Council for Harmonisation of Technical Requirements for 834

Pharmaceuticals for Human Use 835

QC Quality control 836

QRM Quality risk management 837

Page 29: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 29

TOT Transfer of technology 838

839

References 840

841

1. WHO good manufacturing practices for investigational pharmaceutical products for clinical 842

trials in humans. In: WHO Expert Committee on Specifications for Pharmaceutical 843

Preparations: thirty-fourth report. Geneva: World Health Organization; 1996: Annex 7 (WHO 844

Technical Report Series, No. 863 {under revision: working document QAS/20.863}). 845

2. WHO good manufacturing practices for active pharmaceutical ingredients. In: WHO Expert 846

Committee on Specifications for Pharmaceutical Preparations: forty-fourth report. Geneva: 847

World Health Organization; 2010: Annex 2 (WHO Technical Report Series, No. 957). 848

3. WHO good manufacturing practices for pharmaceutical products: main principles. In: WHO 849

Expert Committee on Specifications for Pharmaceutical Preparations: forty-eight report. 850

Geneva: World Health Organization; 2014: Annex 2 (WHO Technical Report Series, No. 986). 851

4. WHO good manufacturing practices: water for pharmaceutical use. In: WHO Expert 852

Committee on Specifications for Pharmaceutical Preparations: forty--six report. Geneva: 853

World Health Organization; 2012: Annex 2 (WHO Technical Report Series, No. 970). 854

5. WHO guidelines for sampling of pharmaceutical products and related materials. In: WHO 855

Expert Committee on Specifications for Pharmaceutical Preparations: thirty-ninth report. 856

Geneva: World Health Organization; 2005: Annex 4 (WHO Technical Report Series, No. 929). 857

6. WHO guidelines on heating, ventilation and air-conditioning systems for non-sterile 858

pharmaceutical products. In: WHO Expert Committee on Specifications for Pharmaceutical 859

Preparations:. fifty-second report. Geneva: World Health Organization; 2018: Annex 8 (WHO 860

Technical Report Series, No. 1010). 861

7. WHO supplementary guidelines on good manufacturing practices: validation. In: WHO Expert 862

Committee on Specifications for Pharmaceutical Preparations: fortieth report. Geneva: World 863

Health Organization; 2006: Annex 4 (WHO Technical Report Series, No. 937). 864

8. WHO good practices for pharmaceutical products containing hazardous substances. In: WHO 865

Expert Committee on Specifications for Pharmaceutical Preparations: forty-fourth report. 866

Geneva: World Health Organization; 2010: Annex 2 (WHO Technical Report Series, No. 957). 867

9. WHO good manufacturing practices for sterile pharmaceutical products. In: WHO Expert 868

Committee on Specifications for Pharmaceutical Preparations: forty-fifth report. Geneva: 869

Page 30: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 30

World Health Organization; 2011: Annex 6 (WHO Technical Report Series, No. 961 {under 870

revision: consultation closed}). 871

10. WHO guidelines on transfer of technology in pharmaceutical manufacturing. In: WHO Expert 872

Committee on Specifications for Pharmaceutical Preparations: forty-fifth report. Geneva: 873

World Health Organization; 2011: Annex 7 (WHO Technical Report Series, No. 961 {under 874

revision: working document QAS/QAS20.869}). 875

11. WHO good practices for pharmaceutical quality control laboratories. In: WHO Expert 876

Committee on Specifications for Pharmaceutical Preparations: forty-fourth report. Geneva: 877

World Health Organization; 2010: Annex 1 (WHO Technical Report Series, No. 957). 878

12. ICH harmonised tripartite guideline. Pharmaceutical Development Q8. Geneva: International 879

Conference on Harmonisation of Technical Requirements for Registration of Pharmaceutical 880

for Human Use; 2009). 881

13. ICH harmonised tripartite guideline. Quality risk management Q9. Geneva: International 882

Conference on Harmonisation of Technical Requirements for Registration of Pharmaceutical 883

for Human Use; 2005. 884

14. WHO guidelines on good manufacturing practices: validation, Appendix 7: non-sterile process 885

validation. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations: 886

forty-ninth report. Geneva: World Health Organization: 2015: Annex 3 (WHO Technical Report 887

Series, No. 992). 888

15. EMA Guideline on process validation for finished products - information and data to be 889

provided in regulatory submissions. (EMA/CHMP/CVMP/QWP/BWP/70278/2012-Rev1,Corr.1) 890

Committee for Medicinal Products for Human Use (CHMP) and Committee for Medicinal 891

Products for Veterinary Use (CVMP), 2016. 892

16. FDA Guidance for Industry Process Validation: General Principles and Practices, January 2011. 893

17. WHO stability testing of active pharmaceutical ingredients and finished pharmaceutical 894

products. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations: fifty-895

second report. Geneva: World Health Organization; 2018: Annex 10 (WHO Technical Report 896

Series, No. 1010). 897

18. Good manufacturing practices: guidelines on validation. Appendix 4. Analytical procedure 898

validation. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations: fifty-899

third report. Geneva: World Health Organization; 2019: Annex 3 (WHO Technical Report Series, 900

No. 1019). 901

Page 31: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 31

19. Good manufacturing practices: guidelines on validation. Appendix 3. Cleaning validation. In: 902

WHO Expert Committee on Specifications for Pharmaceutical Preparations: fifty-third report. 903

Geneva: World Health Organization; 2019: Annex 3 (WHO Technical Report Series, No. 1019). 904

20. Points to consider when including Health-Based Exposure Limits (HBELs) in cleaning validation. 905

In: WHO Expert Committee on Specifications for Pharmaceutical Preparations: fifty-fifth 906

report. Geneva: World Health Organization; 2021: Annex 2 (WHO Technical Report Series, No. 907

1025). 908

909

Further reading 910

911

• WHO model guidance for the storage and transport of time- and temperature-sensitive 912

pharmaceutical products. In: WHO Expert Committee on Specifications for Pharmaceutical 913

Preparations: forty-fifth report. Geneva: World Health Organization; 2011: Annex 9 (WHO 914

Technical Report Series, No. 961). 915

• WHO general guidelines for the establishment, maintenance and distribution of chemical 916

reference substances. In: WHO Expert Committee on Specifications for Pharmaceutical 917

Preparations: forty-first report. Geneva: World Health Organization; 2007: Annex 3 (WHO 918

Technical Report Series, No. 943). 919

• WHO good practices for pharmaceutical microbiology laboratories. In: WHO Expert 920

Committee on Specifications for Pharmaceutical Preparations: forty-fifth report. Geneva: 921

World Health Organization; 2011: Annex 2 (WHO Technical Report Series, No. 961). 922

• WHO guidelines on quality risk management. In: WHO Expert Committee on Specifications for 923

Pharmaceutical Preparations: forty-seventh report. Geneva: World Health Organization; 2013: 924

Annex 2 (WHO Technical Report Series, No. 981). 925

• WHO guidelines on variation to a prequalified product. In: WHO Expert Committee on 926

Specifications for Pharmaceutical Preparations: forty-seventh report. Geneva: World Health 927

Organization; 2013: Annex 3 (WHO Technical Report Series, No. 981). 928

• WHO guidelines for drafting a site master file. In: WHO Expert Committee on Specifications 929

for Pharmaceutical Preparations: forty-fifth report. Geneva: World Health Organization; 2011: 930

Annex 14 (WHO Technical Report Series, No. 961). 931

Page 32: WHO good practices for research and development facilities ...

Working document QAS/20.865/Rev1 Page 32

• WHO general guidance on hold-time studies. In: WHO Expert Committee on Specifications for 932

Pharmaceutical Preparations: forty-ninth report. Geneva: World Health Organization; 2015: 933

Annex 4 (WHO Technical Report Series, No. 992). 934

• WHO technical supplements to model guidance for storage and transport of time- and 935

temperature-sensitive pharmaceutical products. In: WHO Expert Committee on Specifications 936

for Pharmaceutical Preparations: forty-ninth report. Geneva: World Health Organization; 937

2015: Annex 5 (WHO Technical Report Series, No. 992). 938

• WHO recommendations for quality requirements when plant–derived artemisinin is used as a 939

starting material in the production of antimalarial active pharmaceutical ingredients. In: WHO 940

Expert Committee on Specifications for Pharmaceutical Preparations: forty-ninth report. 941

Geneva: World Health Organization: 2015: Annex 6 (WHO Technical Report Series, No. 992). 942

• Guidance on good data and record management practices. In: WHO Expert Committee on 943

Specifications for Pharmaceutical Preparations: Fiftieth report. Geneva: World Health 944

Organization; 2016: Annex 5 (WHO Technical Report Series, No. 996). 945

• WHO general guidance on variations to multisource pharmaceutical products. In: WHO Expert 946

Committee on Specifications for Pharmaceutical Preparations: Fiftieth report. Geneva: World 947

Health Organization; 2016: (WHO Technical Report Series, No. 996). 948

• International Council for Harmonisation of Technical Requirements for Pharmaceuticals for 949

Human Use (ICH) ICH Q10, Pharmaceutical Quality System, 2008. 950

• International Council for Harmonisation of Technical Requirements for Pharmaceuticals for 951

Human Use (ICH) ICH Q11, Development and Manufacture of Drug Substances, 2018. 952

• ISPE. Good Practice Guide. Technology Transfer. Third edition, 2018. 953

• Pharmaceutical industry, the discovery, development, and manufacture of drugs and 954

medications (pharmaceuticals) by public and private organizations. J. W. Dailey. 2003. 955

• Good manufacturing practices: guidelines on validation. Appendix 5. Validation of 956

computerized systems. In: WHO Expert Committee on Specifications for Pharmaceutical 957

Preparations: fifty-third report. Geneva: World Health Organization; 2019: Annex 3 (WHO 958

Technical Report Series, No. 1019). 959

960

961

*** 962