PROPOSAL FOR REVISION OF THE SUPPLEMENTARY GUIDELINES ON GOOD MANUFACTURING PRACTICES: VALIDATION, APPENDIX 7: NON-STERILE PROCESS VALIDATION



Similar documents
PROPOSED UPDATED TEXT FOR WHO GOOD MANUFACTURING PRACTICES FOR PHARMACEUTICAL PRODUCTS: MAIN PRINCIPLES (JANUARY 2013)

Guideline on Process Validation

GENERAL GUIDANCE FOR INSPECTORS ON HOLD-TIME STUDIES

ICH guideline Q8, Q9 and Q10 - questions and answers volume 4

GUIDE TO GOOD MANUFACTURING PRACTICE FOR MEDICINAL PRODUCTS ANNEX 15 *

Guidance for Industry

Workshop on process validation

DRAFT MONOGRAPH FOR THE INTERNATIONAL PHARMACOPOEIA PARACETAMOL ORAL SUSPENSION (September 2010)

Guidance for Industry

ZINC ACETATE Draft proposal for The International Pharmacopoeia (August 2012)

ICH guideline Q10 on pharmaceutical quality system

Process Validation: Practical Aspects of the New FDA Guidance

Working Party on Control of Medicines and Inspections. Final Version of Annex 15 to the EU Guide to Good Manufacturing Practice

Guidance for Industry. Q10 Pharmaceutical Quality System

White paper: FDA Guidance for Industry Update Process Validation

Annex 2. WHO good manufacturing practices for pharmaceutical products: main principles 1

Workshop B Control Strategy

PHARMACEUTICAL QUALITY SYSTEM Q10

SUPPLEMENTARY GUIDELINES ON GOOD MANUFACTURING PRACTICES (GMP): VALIDATION

FDA Guidance for Industry Update - Process Validation

What is Process Validation?

Annex 2. WHO guidelines on quality risk management. 1. Introduction Glossary Quality risk management process 70

WHO GUIDELINE ON QUALITY RISK MANAGEMENT

LUMEFANTRINE Draft proposal for The International Pharmacopoeia (October 2006)

ICH Q10 Pharmaceutical Quality System (PQS)

Annex 6. Guidance on variations to a prequalified product dossier. Preface

Impact Assessment in a Science & Risk Based Environment. R. Legland 11/04/11

ICH Q10 - Pharmaceutical Quality System

PHARMACEUTICAL DEVELOPMENT

Library Guide: Pharmaceutical GMPs

State of Control Over the Lifecycle and Process Validation (New and Legacy Products)

Implementing New USP Chapters for Analytical Method Validation

Quality by Design Approaches to Analytical Methods -- FDA Perspective. Yubing Tang, Ph.D. FDA/CDER/ONDQA AAPS, Washington DC October 25, 2011

ICH Public Meeting. Joseph C. Famulare. October 2, Acting Deputy Director Office of Compliance CDER / FDA Office of Compliance

Combination Products. Presented by: Karen S. Ginsbury For: IFF March PCI Pharma

Guidance on Qualification of existing facilities, systems, equipment and utilities

EXPERT COMMITTEE ON BIOLOGICAL STANDARDIZATION EXPERT COMMITTEE ON SPECIFICATIONS FOR PHARMACEUTICAL PREPARATIONS Geneva, 18 to 22 October 2010

Preparing for the Pre-Approval Inspection What to do Before the FDA Arrives. Barry A. Friedman, Ph.D. Consultant

ASSESSMENT OF QUALITY RISK MANAGEMENT IMPLEMENTATION

Quality by Design (QbD) Overview

Industry Implications of Pharmaceutical Quality ICH Guidelines

Pharmaceutical Process Validation: An Overview

Cleaning Validation in Active pharmaceutical Ingredient manufacturing plants

ICH guideline Q11 on development and manufacture of drug substances (chemical entities and biotechnological/ biological entities)

QUALITY RISK MANAGEMENT (QRM): A REVIEW

Annex 7 Guidelines on pre-approval inspections

[ABOUT THE AUTHOR. FDA Lifecycle Approach to Process Validation What, Why, and How? PQ Forum. Paul L. Pluta]

GUIDELINES ON VALIDATION APPENDIX 5 VALIDATION OF COMPUTERIZED SYSTEMS

Commercial Manufacturing - Qualification & Validation-related GMP Deficiencies and Other Lifecycle Considerations

WHO GUIDELINE ON QUALITY RISK MANAGEMENT

In 2001, ISPE issued Baseline Guide Volume

IMPURITIES IN NEW DRUG PRODUCTS

Questions and answers on post approval change management protocols

GOOD DOCUMENTATION AND QUALITY MANAGEMENT PRINCIPLES. Vimal Sachdeva Technical Officer (Inspector), WHO Prequalification of Medicines Programme

Workshop A Design Space (DS)

Pharmaceutical Quality Systems: US Perspective

Annex 4 Supplementary guidelines on good manufacturing practices: validation

AN OVERVIEW OF PHARMACEUTICAL VALIDATION: QUALITY ASSURANCE VIEW POINT

International Health Regulations

SPECIFICATIONS AND CONTROL TESTS ON THE FINISHED PRODUCT

A Stability Program for the Distribution of Drug Products

Quality Assurance. Disclosure for Lilli Møller Andersen. No relevant financial relationships exist for any issue mentioned in this presentation

Questions and answers on post approval change management protocols

Workshop on process validation

Annex 4 Good Manufacturing Practices for pharmaceutical products: main principles

COMMISSION DIRECTIVE 2003/94/EC

EUROPEAN COMMISSION HEALTH AND CONSUMERS DIRECTORATE-GENERAL. EudraLex. The Rules Governing Medicinal Products in the European Union

Regulatory Expectations for GMP: What s Happening. Patricia Weideman, PhD Director, Product Quality & Occupational Toxicology Genentech, Inc.

Comparison of EU GMP guidelines with WHO guidelines Identification of the cost-intensive requirements

The purpose of this Supplier Quality Standard is to communicate the expectations and requirements of Baxter Healthcare Corporation to its suppliers.

Recent Updates on European Requirements and what QPs are expected to do

Comparative analysis between the possible regulatory approaches to GMP compliance TITOLO PRESENTAZIONE

Annex 7 Application of Hazard Analysis and Critical Control Point (HACCP) methodology to pharmaceuticals

EU GMP Requirements - Quality Systems - Bernd Boedecker GMP Inspectorate of Hannover / Germany at Turkish Ministry of Health Ankara, Oct 2009

ISO 9001:2015 Internal Audit Checklist

Diogo Filipe Lopes Ferreira

Guide to Master Formulae WHO/FWC/IVB/QSS/VQR

Guidance for Industry PAT A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance

GOOD DISTRIBUTION PRACTICES (GDP) FOR PHARMACEUTICAL PRODUCTS

DNV GL Assessment Checklist ISO 9001:2015

WHO GUIDELINE ON TRANSFER OF TECHNOLOGY

Guidance for Industry

Journal of Chemical and Pharmaceutical Research

Guidance for Industry Quality Systems Approach to Pharmaceutical CGMP Regulations

Guidance for Industry

JANUARY 2013 PREPARATION OF A SITE MASTER FILE FOR A MANUFACTURER OF COSMETIC PRODUCTS

GUIDELINE ON ACTIVE PHARMACEUTICAL INGREDIENT MASTER FILE (APIMF) PROCEDURE 1 (The APIMF procedure guideline does not apply to biological APIs.

How to implement a Quality Management System

STABILITY TESTING OF ACTIVE SUBSTANCES AND PHARMACEUTICAL PRODUCTS

WHO Expert Committee on Specifications for Pharmaceutical Preparations. How does it work?

EUROPEAN COMMISSION HEALTH AND CONSUMERS DIRECTORATE-GENERAL. EudraLex The Rules Governing Medicinal Products in the European Union

EudraLex. The Rules Governing Medicinal Products in the European Union Volume 4

Validation and Calibration. Definitions and Terminology

Conducting a Gap Analysis on your Change Control System. Presented By Miguel Montalvo, President, Expert Validation Consulting, Inc.

Process Performance Qualification. Demonstrating a High Degree of Assurance in Stage 2 of the Process Validation Lifecycle

Quality assurance of pharmaceuticals

Harmonizing Change Control Processes Globally

Pharmaceutical Quality Management System: Current Concept

COMPLIANCE BY DESIGN FOR PHARMACEUTICAL QUALITY CONTROL LABORATORIES INSIGHT FROM FDA WARNING LETTERS

Transcription:

April 2013 RESTRICTED 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 PROPOSAL FOR REVISION OF THE SUPPLEMENTARY GUIDELINES ON GOOD MANUFACTURING PRACTICES: VALIDATION, APPENDIX 7: NON-STERILE PROCESS VALIDATION World Health Organization 2013 All rights reserved. (APRIL 2013) DRAFT FOR COMMENT Please address any comments on this proposal by 24 May 2013 to Dr S. Kopp, Medicines Quality Assurance Programme, World Health Organization, 1211 Geneva 27, Switzerland, fax: (+41 22) 791 4730 or e-mail: kopps@who.int with a copy to gaspardm@who.int. We are sending out our working documents electronically only and they are also placed on the Medicines web site for comment. If you do not already receive our documents please let us have your e-mail address (to bonnyw@who.int) and we will add it to our electronic mailing list. This draft is intended for a restricted audience only, i.e. the individuals and organizations having received this draft. The draft may not be reviewed, abstracted, quoted, reproduced, transmitted, distributed, translated or adapted, in part or in whole, in any form or by any means outside these individuals and organizations (including the organizations' concerned staff and member organizations) without the permission of the World Health Organization. The draft should not be displayed on any web site. Please send any request for permission to: Dr Sabine Kopp, Medicines Quality Assurance Programme, Quality Assurance and Safety: Medicines, Department of Essential Medicines and Health Products, World Health Organization, CH-1211 Geneva 27, Switzerland. Fax: (41-22) 791 4730; e-mail: kopps@who.int. The designations employed and the presentation of the material in this draft do not imply the expression of any opinion whatsoever on the part of the World Health Organization concerning the legal status of any country, territory, city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps represent approximate border lines for which there may not yet be full agreement. The mention of specific companies or of certain manufacturers products does not imply that they are endorsed or recommended by the World Health Organization in preference to others of a similar nature that are not mentioned. Errors and omissions excepted, the names of proprietary products are distinguished by initial capital letters. All reasonable precautions have been taken by the World Health Organization to verify the information contained in this draft. However, the printed material is being distributed without warranty of any kind, either expressed or implied. The responsibility for the interpretation and use of the material lies with the reader. In no event shall the World Health Organization be liable for damages arising from its use. This draft does not necessarily represent the decisions or the stated policy of the World Health Organization.

page 2 38 39 40 41 42 43 SCHEDULE FOR THE PROPOSED ADOPTION PROCESS OF DOCUMENT QAS/13.527: PROPOSAL FOR REVISION OF THE SUPPLEMENTARY GUIDELINE ON GOOD MANUFACTURING PRACTICES: VALIDATION, APPENDIX 7: NON-STERILE PROCESS VALIDATION Need for revision of published good manufacturing practices: validation identified by Prequalification of Medicines Programme March 2013 Wide circulation of draft document for comment April 2013 Compilation of feedback received June 2013 Discussion of feedback during informal consultation on quality assurance guidelines July 2013 Mailing of revision for comment August 2013 Presentation to forty-eighth meeting of the WHO Expert Committee on Specifications for Pharmaceutical Preparations Any further action as necessary October 2013 44

page 3 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 PROPOSAL FOR REVISION OF THE SUPPLEMENTARY GUIDELINE ON GOOD MANUFACTURING PRACTICES: VALIDATION, APPENDIX 7: NON-STERILE PROCESS VALIDATION Note from Secretariat: The current text of the Supplementary Guideline on Good Manufacturing Practices: Validation (Ref: World Health Organization, WHO Technical Report Series, No. 937, 2006, Annex 4) is available on the following web site: http://www.who.int/medicines/areas/quality_safety/quality_assurance/production/en/index.html Moreover, comments are being sought at the same time, if the Appendix 3 on Cleaning validation be revised in line with the current developments on setting health based exposure limits for use in risk identification in the manufacture of different medicinal products in shared facilities; if yes concrete proposals for revision would be appreciated. The Appendixes of the Supplementary Guideline on Good Manufacturing Practices: Validation are currently as follows: Appendix 1- Validation of heating, ventilation and air-conditioning systems Appendix 2 -Validation of water systems for pharmaceutical use Appendix 3 - Cleaning validation Appendix 4 - Analytical method validation need for revision? Appendix 5 - Validation of computerized systems Appendix 6 - Qualification of systems and equipment Appendix 7 - Non-sterile process validation proposed to be revised

page 4 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 Proposal for revision of the Supplementary Guideline on Good Manufacturing Practices: Validation, Appendix 7: Non-sterile process validation Contents page 1. Background and scope 2. Glossary 3. Introduction 4. Phase I. Process design 5. Phase II. Qualification and process verification 6. Phase III. Continued process verification 7. Change control References 1. BACKGROUND AND SCOPE Further to the Supplementary Guideline on good manufacturing practices: validation, as published in the World Health Organization (WHO) Technical Report, No. 937, 1 additional guidelines to support current approaches in GMP are published herewith to further support the scope of process validation (also referred to as process qualification) linked to quality risk management and quality by design principles as described by WHO and the International Conference on Harmonisation (ICH). This guideline allows for different approaches in process validation. The principles described in this guideline are applicable to non-sterile finished pharmaceutical dosage forms. Thorough knowledge of product and process development studies; previous manufacturing experience; and quality risk management (QRM) principles are essential in the all approaches to process validation as the focus is now on the life-cycle approach. The life-cycle approach 1 Supplementary guidelines on good manufacturing practices: validation. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations. Fortieth report. Geneva, World Health Organization. WHO Technical Report Series, No. 937 (Annex 5), 2006.

page 5 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 links product and process development, validation of the commercial manufacturing process and maintenance of the process during routine commercial production. A risk based approach to validation is recommended, linked to in-line or on-line controls and monitoring to ensure that a process is in a state of control during routine manufacture. 2. GLOSSARY control strategy A planned set of controls, derived from current product and process understanding that assures process performance and product quality. The controls can include parameters and attributes related to drug substance and pharmaceutical product materials and components, facility and equipment operating conditions, in-process controls, finished product specifications, and the associated methods and frequency of monitoring and control. continued process verification (CPV) Continued process verification (CPV) can be defined as continuous monitoring of manufacturing performance by using extensive in-line, on-line or at-line monitoring and/or controls to evaluate process performance. It is a science and risk-based real-time approach to verify and demonstrate that a process that operates within the predefined specified parameters consistently produces material which meets all its critical quality attributes (CQAs) and control strategy requirements. continuous process verification An alternative approach to process validation in which manufacturing process performance is continuously monitored and evaluated. critical process parameter (CPP) A process parameter whose variability has an impact on a critical quality attribute and therefore should be monitored or controlled to ensure the process produces the desired quality.

page 6 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 critical quality attribute (CQA) A physical, chemical, biological or microbiological property or characteristic that should be within an appropriate limit, range or distribution to ensure the desired product quality. life-cycle All phases in the life of a product from the initial development through marketing until the product s discontinuation (ICH Q8). pharmaceutical quality system (PQS) Management system to direct and control a pharmaceutical company with regard to quality. process validation Documented evidence which provides a high degree of assurance that a specific process will consistently result in a product that meets its predetermined specifications and quality characteristics. 3. INTRODUCTION Process validation data should be generated for all products to demonstrate the adequacy of the manufacturing process at each site of manufacture. The validation should be carried out in accordance with good manufacturing practices (GMP) and data should be held at the manufacturing location and made available for inspection. Manufacturers should confirm that a manufacturing process is under control before a product is placed on the market. Process validation is associated with the collection and evaluation of data, from the process design stage through commercial production, which provides scientific evidence that a process is capable of consistently delivering a quality product. Process validation provides documented evidence that a process is capable of reliably and repeatedly rendering a product of the required quality. A risk assessment approach should be used to determine the scope and extent to which process(es) and starting material variability may affect product quality. The critical steps and

page 7 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 parameters (e.g. those that may have an impact on the quality of the product) in the process of manufacturing a pharmaceutical product and other relevant studies demonstrating that the process is capable of delivering the desired product, quality should be identified and documented and be based on knowledge of the product or processes concerned, according to the stage of the product life-cycle. Process validation should cover at least these critical steps and parameters. Where necessary, a flow diagram may be helpful, covering all operations and controls in the process to be validated. When applying QRM to a given operation, the steps preceding and following that operation should also be considered. Amendments to the flow diagram may be made where appropriate and should be documented as part of the validation documentation. Different approaches can be followed when validating a process. Manufacturers should ensure that the principles of process validation described in this guideline are implemented. These include and cover phases of validation during process design, scale up, qualification of premises, utilities and equipment, process performance verification and ongoing monitoring of batches manufactured for commercial supply to ensure that the process remains in a state of control. The objectives of process validation include ensuring that: the process design is evaluated to show that it is reproducible, the commercial manufacturing process is defined and controlled, ongoing assurance is gained to show that the process remains in a state of control. The validation should cover all manufactured strengths and all manufacturing sites used for production of the marketed product. A matrix approach may be acceptable based on appropriate risk assessment. There are different approaches to process validation which include traditional process validation with prospective and concurrent validation, continuous process performance verification (where an enhanced approach to development has been employed or where a

page 8 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 substantial amount of product and process knowledge and understanding has been gained through historical data and manufacturing experience) and a combination of traditional process validation and continuous process verification. Continued process verification is considered as a third phase in process validation. Manufacturers should plan towards implementing the new approach in process validation that should consist of three phases in the product life-cycle. Phase I. Process design Phase II. Qualification and process verification Phase IIA. Qualification Phase IIB. Continuous process performance verification Phase III. Continued process verification 4. PHASE I. PROCESS DESIGN As part of the process validation life-cycle some process validation studies may be conducted on pilot-scale batches (corresponding to at least 10% or 100 000 units whichever is the greater) of the production scale. In certain cases, however, it may be necessary to provide production-scale validation data. The number of batches (minimum of three) should be based on the variability of the process, the complexity of the process/product and the experience of the manufacturer. The number of batches produced in this validation exercise should be sufficient to allow the normal extent of variation and trends to be established and to provide sufficient data for evaluation. Extensive testing should be performed on the product at various stages during the manufacturing process of the batches. Manufacturers should define the stage at which the product is considered to be validated and the basis on which that decision was made. It should include a justification for the number of batches used based on the complexity and expected variability of the process.

page 9 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 Validation should be done in accordance with process validation protocols. Data should be collected and reviewed against predetermined acceptance criteria, and reflected in process validation reports. The protocol should include: a description of the process; a description of the experiment; details of the equipment and/or facilities to be used (including measuring or recording equipment) together with its calibration status; the variables to be monitored; the samples to be taken where, when, how, how many and how much (sample size); the product performance characteristics/attributes to be monitored, together with the test methods; the acceptable limits; time schedules; personnel responsibilities; details of methods for recording and evaluating results, including statistical analysis. The results should be documented in the validation report which reflects the validation protocol. A conclusion and recommendation should be made on the extent of monitoring and the inprocess controls necessary for routine production, on the basis of the results obtained. The planned commercial production and control records, which contain the operational limits and overall strategy for process control, should be carried forward to the next phase for confirmation.

page 10 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 5. PHASE II. QUALIFICATION AND PROCESS VERIFICATION Process verification is divided into two phases. These phases include qualification of premises, utilities and equipment where commercial scale batches will be manufactured and continuous process performance verification. Phase IIA. Qualification Premises, utilities, support systems and equipment should be appropriately qualified before process performance verification (as part of validation) is started. In a traditional approach, stages of qualification may include design, installation, operational and performance qualification. In some cases process validation may be conducted concurrently with performance qualification. Phase IIB. Continuous process performance verification After completion of qualification, commercial batches should be subjected to continuous process performance verification as part of process validation. It should confirm that scale up in batch size did not adversely affect the characteristics of a product. CPV demonstrates that a process that operates within the predefined specified parameters consistently produces a product which meets all its critical quality attributes (CQAs) and control strategy requirements. The process should be verified on commercial-scale batches prior to marketing of the product. Extensive in-line or at-line controls should be used to monitor process performance and product quality in a timely manner. Relevant process quality attributes of incoming materials or components, in-process material and finished products should be collected. This should include the verification of attributes, parameters and end points, and assessment of CQA and

page 11 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 critical process parameter (CPP) trends. Process analytical technology applications and multivariate statistical process control (MSPC) can be used. The scope and extent of continuous process verification will be influenced by a number of factors including: prior development and manufacturing knowledge from similar products and/or processes; the extent of process understanding gained from development studies and commercial manufacturing experience; the complexity of the product and/or manufacturing process; the level of process automation and analytical technologies used; process robustness and manufacturing history since point of commercialization as appropriate. Manufacturers should describe the appropriateness and feasibility of the CPV strategy including the process parameters and material attributes that will be monitored as well as the analytical methods that will be employed. Continuous process verification can be introduced at any time of the life-cycle of the product: it can be used to design process validation protocols for the initial commercial production; to revalidate commercialized products as part of process changes; to support continual improvement throughout the remainder of the life-cycle. It is expected that additional monitoring for the first commercial batches should be done where continuous process verification is implemented. Manufacturers should define: the number of batches for which additional monitoring is proposed; the type of testing/monitoring to be performed; the acceptance criteria to be applied; how the data will be evaluated. Statistical models or tools used should be described.

page 12 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 6. PHASE III. CONTINUED PROCESS VERIFICATION Manufacturers should monitor product quality of commercial batches after completion of Phase I and Phase II of process validation. This will provide evidence that a state of control is maintained throughout the product life-cycle. Periods of enhanced sampling and monitoring may help to increase process understanding as part of continuous improvement. Process trends such as the quality of incoming materials or components, in-process and finished product results and non-conformances should be collected and assessed in order to verify the validity of the original process validation or to identify required changes to the control strategy. The extent and frequency of ongoing process validation should be reviewed periodically and modified if appropriate throughout the product life-cycle. Continued process validation (CdPV) should not be confused with product quality review. Table 2 provides a summary of the new approach to process validation. Table 2. New approach to process validation Product life-cycle Process validation Phase I Phase II Phase III Process design Qualification Continuous process verification Continued process verification - Pilot scale (and scale-up batches where appropriate) - Risk assessment to identify critical quality attributes and process control parameters - Premises - Utilities - Equipment - Commercial-scale batches - In-line or on-line monitoring (e.g. PAT) - Defined number of batches - Periodic review of trends - May include sampling and testing

page 13 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 - Protocols and reports - Validate process - Define CQA and CPPs to be monitored in Phase II 7. CHANGE CONTROL Change control GMP Changes during the life-cycle of a product should be managed through a change control procedure. Sufficient data should be generated to demonstrate that the revised process will result in a product of the desired quality, consistent with the approved specification. The change control procedure and records should ensure that all aspects are thoroughly documented and approved including regulatory approval where appropriate (variation). Manufacturers should follow change control procedures when changes are planned to existing systems or processes. Validation should be considered when changes are planned to production and/or control procedures. Changes that are likely to require revalidation may include: changes in the manufacturing process (e.g. mixing times, drying temperatures); changes in the equipment (e.g. addition of automatic detection systems); production area and support system changes (e.g. rearrangement of areas or a new water treatment method); transfer of processes to another site; unexpected changes (e.g. those observed during self-inspection or during routine analysis of process trend data). Based on risk assessment, revalidation should be considered in the case of: changes in the master formula, methods, starting material manufacturer, equipment and/or instruments;

page 14 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 equipment calibrations and preventive maintenance carried out; changes to standard operating procedures (SOPs); changes to cleaning and hygiene programmes. REFERENCES 1. Guideline on Process Validation, 29 March 2012 EMA/CHMP/CVMP/QWP/70278/2012-Rev1, Committee for Medicinal Products for Human Use (CHMP), Committee for Medicinal Products for Veterinary Use (CVMP) www.ema.europa.eu/ uideline/2012/04/wc500125399.pdf. 2. Guidance for Industry, Process Validation: General Principles and Practices, 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), Center for Veterinary Medicine (CVM), January 2011, Current Good Manufacturing Practices (CGMP) Revision 1. 3. ICH Harmonised Tripartite Guideline, Pharmaceutical Development Q8(R2), Current Step 4 version, dated August 2009. 4. ICH Harmonised Tripartite Guideline, Quality Risk Management, Q9, Current Step 4 version, dated 9 November 2005. 5. ICH Harmonised Tripartite Guideline, Pharmaceutical Quality System, Q10, Current Step 4 version, dated 4 June 2008 http://www.ich.org/products/guidelines/quality/article/quality-guidelines.html 6. Quality Assurance of pharmaceuticals. WHO guidelines, related guidance and GXP training materials. Geneva, World Health Organization, 2013 (CD-ROM).

page 15 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 7. WHO good manufacturing practices: main principles for pharmaceutical products. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations. Fortyfifth report. Geneva, World Health Organization. WHO Technical Report Series, No. 961 (Annex 3), 2011 (WHO good manufacturing practices: main principles for pharmaceutical products). 8. WHO guidelines on quality risk management. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations. Forty-seventh report. Geneva, World Health Organization. WHO Technical Report Series, No. 981 (Annex 2), 2013 (in press). ***