Sei sulla pagina 1di 24

Working document QAS/14.

575
February 2014
Draft for comment

1
2

WHO GENERAL GUIDANCE ON


VARIATIONS TO MULTISOURCE
PHARMACEUTICAL PRODUCTS

3
4
5
6
7

(February 2014)

8
9

DRAFT FOR COMMENT

10
11
12
13
14
15

Should you have any comments on the attached revision, please send these to Dr S. Kopp, Group
Lead, Medicines Quality Assurance, Technologies, Standards and Norms (kopps@who.int) with a
copy to Ms Marie Gaspard (gaspardm@who.int) by 7 April 2014.
Our working documents will be sent out electronically only and will also be placed on the
Medicines website for comment under Current projects. If you do not already receive our
draft working documents please let us have your email address (to bonnyw@who.int) and we
will add it to our electronic mailing list.

16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42

World Health Organization 2014


All rights reserved.
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
website.
Please send any request for permission to:
Dr Sabine Kopp, Group Lead, Medicines Quality Assurance, Technologies, Standards and Norms, Department of Essential
Medicines and Health Products, World Health Organization, CH-1211 Geneva 27, Switzerland. Fax: (41-22) 791 4730;
email: 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.

Working document QAS/14.575


page 2
43
44
45
46

SCHEDULE FOR THE PROPOSED ADOPTION PROCESS OF DOCUMENT QAS/14.575:


WHO GENERAL GUIDANCE ON VARIATIONS TO MULTISOURCE
PHARMACEUTICAL PRODUCTS
Development of draft based on WHO Expert Committee
on Specifications for Pharmaceutical Preparations
prepared by Ms G.N. Mahlangu, Director-General,
Medicines Control Authority of Zimbabwe, Harare,
Zimbabwe

47
48
49

October 2013February 2014

Circulation for comments

MarchApril 2014

Compilation of feedback

May 2014

Review of feedback and comments with assessors and


experts

June 2014

Recirculation for feedback

July 2014

Compilation of feedback

September 2014

Discussion at forty-ninth meeting of the WHO Expert


Committee on Specifications for Pharmaceutical
Preparations

1317 October 2014

Any further action, as necessary

Working document QAS/14.575


page 3
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
83

Contents
page
1.

2.
3.

4.
5.

Introduction ...
1.1
Objectives .
1.2
Scope and application of the guideline .
Glossary .
General considerations ..
3.1
Reporting types .
3.1.1 Notification
3.1.2 Minor variation ..
3.1.3 Major variation ..
3.2
New applications and extension applications
3.3
Labelling information
3.4
Conditions to be fulfilled ..
3.5
Documentation required
General stability considerations
Comparative studies ..
5.1
Comparative in vivo studies ..
5.2
Comparative in vitro studies ..
5.3
Variations (changes) to pharmaceutical aspects of registered products which
may be made without prior approval
5.3.1 Annual notification .
5.3.2 Immediate notification Do and tell ..
5.4
Variations (changes) to pharmaceutical aspects of registered products which
require prior approval before implementation
5.4.1 Minor variation ...
5.4.2 Major variation

Working document QAS/14.575


page 4
84

1.

INTRODUCTION

85
86

This guidance document is intended to provide supportive information on how to present an

87

application to implement a change to a registered multisource medicinal product. A marketing

88

authorization holder or applicant is responsible for the safety, efficacy and quality of a

89

medicinal product that is placed on the market throughout its life-cycle. As such, changes are

90

required or necessary for an approved or registered product to account for technical and

91

scientific progress, to improve the quality of the medicinal product, to meet market

92

requirements such as scale-up or additional manufacturing sites, or updates to product

93

information (e.g. updates to information on adverse reactions). The common areas for change

94

are pharmaceutical aspects (quality control, source of raw materials, manufacturing, shelf-life

95

etc.) and product information. Such changes, regardless of the nature of the change, are

96

referred to as variations and may require the approval of national medicines regulatory

97

authority (NMRA) prior to implementation.

98
99

Technical requirements for the different types of variations are set out in these guidelines in

100

order to facilitate the submission of appropriate documentation by applicants and their

101

assessment by NMRAs.

102
103

1.1

Objectives

104
105

This guidance document is intended to assist applicants with the classification of changes

106

made to a registered or approved finished pharmaceutical product (FPP) and provide

107

guidance on the technical and other general data requirements to support changes to the

108

quality attributes of the active pharmaceutical ingredient (API) or FPP. Variation applications

109

are categorized into major variation, minor variation (prior approval) and minor variation

110

(notification).

111
112

1.2

Scope and application of the guideline

113
114

These guidelines can be used by both NMRAs and applicants with respect to changes to the

115

quality sections of product dossiers for an API or an FPP. This guidance should be read in

116

conjunction with the Guidelines on submission of documentation for a multisource finished product

Working document QAS/14.575


page 5
117

(1) and Guidance on variations (2) as well as other related WHO guidelines or applicable

118

national guidelines.

119
120

The NMRAs reserve the rights to request for additional information where necessary, in line

121

with national requirements.

122
123

This guidance document is applicable only to APIs and excipients manufactured by chemical

124

synthesis or semisynthetic processes and FPPs containing such APIs and excipients. APIs

125

produced by fermentation and APIs of biological, biotechnological or herbal origin are treated

126

as special cases.

127
128

It is recommended that the principles established in this guidance document be applied to

129

similar quality changes that occur during the development of the product and that the

130

recommended supporting data be included with the initial application for registration of a

131

medicinal product.

132
133

When a variation leads to a revision of the package insert, the patient information leaflet

134

(PIL) and labelling, 1 the updated product information should be submitted as part of the

135

application.

136
137

For variations that require generation of stability data on the API or FPP, the stability studies

138

required, including commitment batches, should always be continued to cover the currently

139

accepted retest or shelf-life period. The NMRAs should be informed immediately if any

140

problems with the stability of APIs or FPPs occur during storage, e.g. if found to be outside

141

specifications or potentially outside specifications.

142
143

Applicants should be aware that some variations might require the submission of additional

144

consequential variations. Therefore for any given change the applicant should consider

145

whether one or more variations might be required to be submitted.

146
147

If changes to the dossier only concern editorial changes, such changes need not be submitted

148

as a separate variation but can be included as a notification together with a subsequent


1

Different regions/countries use different terminology for product information. In this document package insert,
PIL and label are used to refer to product information.

Working document QAS/14.575


page 6
149

variation concerning that part of the dossier. In such a case a declaration should be provided

150

that the contents of the associated sections of the dossier have not been changed by the

151

editorial changes beyond the substance of the variation submitted.

152
153

2.

GLOSSARY

154
155

The definitions provided below apply to the terms used in this guidance. They may have

156

different meanings in other contexts and documents.

157
158

active pharmaceutical ingredient (API)

159

A substance used in the FPP, intended to furnish pharmacological activity or to otherwise

160

have direct effect in the diagnosis, cure, mitigation, treatment or prevention of disease, or to

161

have direct effect in restoring, correcting or modifying physiological functions in human

162

beings.

163
164

active pharmaceutical ingredient (API) starting material

165

A raw material, intermediate, or an API that is used in the production of an API and that is

166

incorporated as a significant structural fragment into the structure of the API. An API starting

167

material can be an article of commerce, a material purchased from one or more suppliers

168

under contract or commercial agreement, or produced in-house.

169
170

applicant

171

For the purposes of this document, the term applicant refers to any person or entity that holds

172

the legal responsibility for the product on the market by submission of the required

173

documentation on a product that has been listed after evaluation as registered or approved.

174
175

biobatch

176

The batch used to establish bioequivalence or similarity to the comparator product as

177

determined in bioequivalence or biowaiver studies, respectively.

178
179

final intermediate

180

Product formed in the last reaction in the synthetic pathway that undergoes synthetic

181

transformation to the API or the crude API. Purification is not considered to be a synthetic

182

transformation.

Working document QAS/14.575


page 7
183
184

finished pharmaceutical product (FPP)

185

A finished dosage form of a pharmaceutical product, which has undergone all stages of

186

manufacture including packaging in its final container and labelling.

187
188

in-process control

189

Check performed during manufacture to monitor or to adjust the process in order to ensure

190

that the final product conforms to its specifications.

191
192

manufacturer

193

A company that carries out operations such as production, packaging, repackaging, labelling

194

and relabelling of pharmaceuticals.

195
196

multisource (generic) pharmaceutical product

197

Pharmaceutically equivalent products that may or may not be therapeutically equivalent.

198
199

officially recognized pharmacopoeia (or compendium) Those pharmacopoeias recognized by

200

the national regulatory agencies (e.g. national pharmacopoeia (if applicable), The

201

International Pharmacopoeia (Ph.Int.), the European Pharmacopoeia (PhEur.), the British

202

Pharmacopoeia (BP), the Japanese Pharmacopoeia (JP) and the United States Pharmacopeia

203

(USP)).

204
205

pilot-scale batch

206

A batch of an API or FPP manufactured by a procedure fully representative of and simulating

207

that to be applied to a full production-scale batch. For example, for solid oral dosage forms, a

208

pilot scale is generally, at a minimum, one-tenth that of a full production scale or 100 000

209

tablets or capsules, whichever is the larger, unless otherwise adequately justified.

210
211

production batch

212

A batch of an API or FPP manufactured at production scale by using production equipment in

213

a production facility as specified in the application.

214
215

register

216

A list of all the pharmaceutical products authorized for marketing in a particular country. The

Working document QAS/14.575


page 8
217

medicines regulatory authority of the country in question maintains the register.

218
219

registered medicines products

220

Pharmaceutical products that have a marketing authorization.

221
222

stringent regulatory authority (SRA)

223

A stringent regulatory authority is:

224

the medicines regulatory authority in a country which is: (a) a member of the

225

International Conference on Harmonisation (ICH) (European Union (EU), Japan and the

226

United States of America); or (b) an ICH Observer, being the European Free Trade

227

Association (EFTA) as represented by Swiss Medic and Health Canada (as may be

228

updated from time to time); or (c) a regulatory authority associated with an ICH member

229

through a legally-binding, mutual recognition agreement including Australia, Iceland,

230

Liechtenstein and Norway (as may be updated from time to time);

231

only in relation to good manufacturing practices (GMP) inspections: a medicines

232

regulatory authority that is a member of the Pharmaceutical Inspection Co-operation

233

Scheme (PIC/S) as specified at http://www.picscheme.org.

234
235

validation

236

The demonstration, with documentary evidence, that any procedure, process, equipment,

237

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

238
239

variation

240

A change to any aspect of a pharmaceutical product, including but not limited to a change to

241

formulation, method and site of manufacture, specifications for the finished product and

242

ingredients, container and container labelling and product information.

243
244

3. GENERAL CONSIDERATIONS

245
246

3.1

Reporting types

247
248

The definitions outlined in the following reporting types are intended to provide guidance

249

with respect to the classification of quality-related changes. Specific examples of changes are

250

provided in these guidelines. However, it should be noted that a change not covered by these

Working document QAS/14.575


page 9
251

guidelines, should be considered as a major change by default. Whenever the applicant is

252

unclear about the classification of a particular change, the respective NMRA should be

253

contacted. It remains the responsibility of the applicant to submit relevant documentation to

254

justify that the change will not have a negative impact on the quality of the product.

255
256

Individual changes normally require the submission of separate variations. Grouping of

257

variations is acceptable only under the following circumstances:

258
259

260
261

specification that requires a new analytical procedure;

262
263

when variations are consequential to each other, e.g. introduction of a new impurity

when the same change affects multiple FPPs, e.g. addition of a new API
manufacturing site for multiple FPPs;

when all the changes fall under annual notification.

264
265

Applicants are also advised to exercise caution whenever several changes to the same FPP are

266

envisaged. Although each of the individual changes may be classified as a particular reporting

267

type, classification within a higher-risk category may be warranted as a result of the

268

composite effect of these changes. In all such cases, applicants are advised to contact the

269

NMRA prior to submission of the variation application to obtain guidance on classifying such

270

changes.

271
272

3.1.1 Notifications

273
274

Notifications are changes that could have minimal or no adverse effects on the overall safety,

275

efficacy and quality of the FPP. Such notifications may not require prior acceptance, but must

276

be notified to the NMRA immediately after implementation, i.e. immediate notification (IN),

277

or within 12 months following implementation, i.e. annual notification (AN).

278
279

3.1.2 Minor variation

280
281

Minor variations are changes that may have minor effects on the overall safety, efficacy and

282

quality of the FPP. Applicants must satisfy themselves that they meet all of the prescribed

283

conditions for the change and submit all required documentation with the variation

284

application.

Working document QAS/14.575


page 10
285
286

Prior approval by the NMRA may be required, before the changes can be implemented. The

287

timeline and implementation of the variation are subject to the NMRAs specific

288

proposals and should be made publicly available.

289
290

3.1.3 Major variation

291
292

Major variations are changes that could have major effects on the overall safety, efficacy and

293

quality of the FPP. In general, a change that is supported by extensive documentation and/or

294

requires extensive assessment of the supporting documentation would be considered as major

295

variation, e.g. a change supported by in vivo studies. Prior approval by the NMRA is required

296

before the changes can be implemented. The timeline and implementation of the

297

variation are subject to the NMRAs specific proposals.

298
299

The NMRA reserves the right to recategorize the application type, where deemed appropriate.

300

Subject to country specific procedure, recategorization may require the applicant to resubmit

301

a new application or additional data according to the correct category.

302
303

3.2

New applications and extension applications

304
305

Certain changes are so fundamental that they alter the terms of the accepted dossier and

306

consequently cannot be considered as changes. In these cases a new dossier must be

307

submitted in line with applicable national requirements for applications for registration of

308

medicines.

309
310

Examples of such changes are listed below:

311
312

1.

Change of the API to a different API.

313

2.

Inclusion of an additional API in a multicomponent product.

314

3.

Removal of one API from a multicomponent product.

315

4.

Change in the dose and/or strength of one or more APIs.

316

5.

Change from an immediate-release product to an extended or delayed-release dosage form or

317

vice versa.

318

6. Change from a liquid to a powder for reconstitution or vice versa.

319

7. Changes in the route of administration.

Working document QAS/14.575


page 11
320
321

Labelling information

3.3

322
323

For any change to labelling information (package insert, PIL, labels), the NMRA must be

324

notified and submission of the revised labelling information is expected as per country

325

specific proposal or requirements.

326
327

3.4

Conditions to be fulfilled

328
329

For each variation, attempts have been made to identify particular circumstances where lower

330

reporting requirements, e.g. notifications, are possible. A change that does not meet all of the

331

conditions stipulated under general and specific circumstances for the notifications are

332

automatically considered at the next higher level of change, i.e. require prior approval before

333

implementation.

334
335

3.5

Documentation required

336
337

All data recommended to support a change should be provided with the submission.

338
339

When recommended supporting data cannot be submitted, a detailed rationale should be

340

provided. Regardless of the documents specified, applicants should ensure that they have

341

provided all relevant information to support the variation. Additional documentation may be

342

required. For all changes it remains the responsibility of the applicant to provide all necessary

343

documents to demonstrate that the change does not have a negative impact on the safety,

344

efficacy or quality of the FPP.

345
346

Where applicable, the following should be included in the application for variations requiring

347

prior approval:

348
349

350

a covering letter (including a list of changes describing each in sufficient detail to allow for a
quick assessment as to whether the appropriate reporting category has been used);

351

section of the original dossier affected by the change(s);

352

current and proposed condition(s);

353

reason for the change(s);

Working document QAS/14.575


page 12
354

355
356

where relevant, a side-by-side comparison of the previously approved and the proposed
information;

357

replacement of the relevant sections of the dossier as per acceptable dossier format for the
respective NMRAs with the proposed changes clearly annotated;

358

copies of package insert, PIL and labels, if relevant;

359

registration status and date of the proposed change(s) in other countries/agencies that had

360

approved the variation(s), especially the country of origin and the reference agencies.

361
362

It should be noted that the NMRA reserves the right to request further information not

363

explicitly described in these guidelines.

364
365

Alternative approaches to the principles and practices described in this document may be

366

acceptable provided they are supported by adequate scientific justification. It is also important

367

to note that the NMRA may request information or material, or defines conditions not

368

specifically described in this guidance, in order to adequately assess the safety, efficacy and

369

quality of an FPP.

370
371

4. GENERAL STABILITY CONSIDERATIONS

372
373

The effect of the changes on an approved medicinal product on the stability of the medicinal

374

product should be evaluated. For general guidance on conducting stability studies, applicants

375

are referred to the WHO Guideline on Stability Studies [3]. For variation submissions, the

376

following points also should be considered:

377

378
379

In most cases (except those involving scale up), stability data from pilot scale batches will be
acceptable to support the proposed change.

Where stability data show a trend toward potency loss or degradant increase under accelerated

380

conditions, it is recommended that historical accelerated stability data from a representative

381

pre-change batch be submitted for comparison. It is also recommended that under these

382

circumstances, all available long-term data on test batches from ongoing studies be provided

383

in the supplement. Submission of historical accelerated and available long-term data would

384

facilitate review and approval of the supplement.

385

A commitment should be included to conduct long-term stability studies through the

386

expiration-dating period, according to the approved protocol, on the first or first three

387

production batches and to report the results in the annual reports.

388

Working document QAS/14.575


page 13
389

5.

390
391

5.1

COMPARATIVE STUDIES
Comparative in vivo studies

392
393

A number of changes outlined include recommendations for supporting comparative in vivo

394

studies (e.g. comparative bioavailability studies).

395
396

Applicants should consult the ICH Q5E guideline and applicable WHO guidance documents

397

when conducting comparative in vivo studies.

398
399

5.2

Comparative in vitro studies

400
401

A number of changes outlined include recommendations for supporting comparative in vitro

402

studies (e.g. comparative dissolution studies). Where an in vitro comparison is recommended

403

to support a variation, the comparison should be made to the product manufactured according

404

to the same formulation and manufacturing process used in the pivotal clinical and/or

405

comparative bioavailability studies approved for the original drug submission (e.g. including

406

batch formula, manufacturing process). This is referred to as the "approved product" in the

407

appendices.

408
409

Alternative approaches to this recommendation may be acceptable if scientifically justified.

410

For example, a comparison to an applicant's marketed product (rather than the product used in

411

the pivotal clinical and/or comparative bioavailability studies) could be justified if a

412

significant body of information has been established for the marketed drug product. For the

413

purposes of this document, a significant body of information for the marketed drug product is

414

likely to exist after a reasonable number of batches of the drug product will be marketed

415

during the specified period of time (e.g. a minimum of 10 batches).

416
417

Applicants should refer to the General Chapters available in the current Schedule B

418

pharmacopoeia for general dissolution and drug release specifications (e.g. United States

419

Pharmacopeia (USP) <711>, USP <724>, European Pharmacopoeia (Ph.Eur.) 2.9.3).

420
421
422

5.3

Variations (changes) to pharmaceutical aspects of registered products which may be


made without prior approval

Working document QAS/14.575


page 14
423
424

5.3.1 Annual notification

425
426

Changes that may be applied without prior approval but included in annual notifications (AN)

427

are listed below:

428
429

1.

430

currently accepted batch size on condition that (a) the product continues to meet specifications, (b) the

431

dissolution profile is not significantly altered, (c) a stability study has been commenced on at least one

432

full-scale production batch and (d) the change does not concern a sterile API.

Changes in batch size of the API or intermediate involving up to 10-fold compared to the

433
434

2.

435

do not reflect a change in processing, e.g. from a fine to microfine particle size.

Additional tests and limits for starting materials or finished products on condition that these

436
437

3.

A change in the content of an excipient of up to 5%.

439

4.

Alteration of the quantitative composition of a tablet or capsule coating amounting to less than

440

2% of the total weight of the tablet or capsule. On condition that (a) the coating has no modified-

441

release properties, (b) there is no API in the coating, (c) any new colours are permitted by the

442

European Commissions List of Permitted Food Colours (4), or the United States Food and Drug

443

Agencys (US-FDA) Summary of Color Additives for Use in the United States in Foods, Drugs,

444

Cosmetics, and Medical Devices (5) and (d) the change is notified.

438

445
446

5.

447

continues to meet specifications and (b) the dissolution profile is not significantly altered.

Changes to the volume of granulating fluid of up to 15%, provided that (a) the product

448
449

6.

450

viscous. On condition that (a) it has been demonstrated that any solid material present is at least

451

equally well suspended and (b) a stability study has been commenced on at least two batches of the

452

altered product.

Changes to the quantitative content of agents whose only function is to make the product

453
454

7.

455

On condition that (a) multipoint in vitro dissolution profiles of the proposed version of the product

456

(determined in the routine release medium on at least two batches of pilot- or production-scale) are

457

similar to the dissolution profiles of the biobatch, (b) coating is not a critical factor for the release

458

mechanism and (c) specifications for the FPP are updated only with respect to weight and dimensions,

Change in weight of tablet coatings or capsule shells involving immediate-release oral FPPs.

Working document QAS/14.575


page 15
459

if applicable.

460
461

8.

462

product is not a slow- or otherwise modified-release product, (b) a new stability study has been

463

commenced on at least two batches of the altered product, (c) no change in the specifications of the

464

intermediates or the FPP, (d) the dissolution profiles are similar to those of the biobatch and (e) the

465

manufacturing processes for the currently accepted and proposed products use the same principles

466

(e.g. a change from wet to dry granulation, from direct compression to wet or dry granulation, or vice

467

versa, would be considered a change in manufacturing principle), the same processing intermediates

468

and there are no changes to any manufacturing solvent used in the process.

Alteration of methods of manufacture and manufacturing equipment on condition that (a) the

469
470

9.

471

does not affect the reproducibility and/or consistency of the product, (b) the change pertains only to

472

immediate-release oral pharmaceutical forms and to non-sterile liquid forms and (c) changes to the

473

manufacturing method and/or to the in-process controls are only those necessitated by the change in

474

batch size, e.g. use of different-sized equipment.

Change in the batch size of the FPP involving downscaling. On condition that (a) the change

475
476

10.

477

involving (a) tightening of in-process limits, (b) deletion of a test and (c) addition of new tests and

478

limits.

Change to in-process tests or limits applied during the manufacture of the FPP or intermediate

479
480

11.

481

do not reflect a change in processing, e.g. from a fine to microfine particle size.

Additional tests and limits for starting materials or finished products on condition that these

482
483

12.

484

pharmacopoeia provided that there is no change to the specifications other than those required to

485

comply with the pharmacopoeia (e.g. no change in particle size distribution).

Change in specifications of an excipient to comply with an officially-recognized

486
487

13.

488

monograph as a result of an update to this monograph to which the FPP is controlled

Update to the specifications to comply with an officially-recognized pharmacopoeial

489
490

14.

491

with an officially-recognized pharmacopoeial monograph as a result of an update to that monograph.

Change in the analytical procedures for the FPP involving updating the analytical procedure

492
493
494
495

5.3.2

Immediate notification do and tell

Working document QAS/14.575


page 16
496
497
498

Applicants must satisfy themselves that they meet all of the prescribed conditions for the

499

change and submit all required documentation with the notification application. Such changes

500

can be implemented immediately at the time of submission and they can be considered

501

accepted if an objection is not issued by the NMRA within a reasonable period subject to

502

country specific proposal after the date of acknowledgement of receipt of the application.

503
504

Examples of such changes are listed below:

505
5061.
507

Change to the marketing authorization holder (name, address and/or legal entity). On
condition that there is no change to the product, including sites of manufacture.

508
5092.
510

Change in the name and/or corporate address of the supplier of the FPP on condition
that the supplier of the product remains the same legal entity.

511
5123.

Deletion of a manufacturing site or manufacturer involving production or testing of

513

the API/FPP intermediate or API/FPP provided that at least one other site continues to

514

perform the same function(s) as the site(s) intended to be deleted and that the deletion of the

515

site is not a result of critical deficiencies in manufacturing.

516
517

4.

Change in the manufacturing process of the API. On condition (a) that there is no

518

change in the physical state (e.g. crystalline, amorphous) of the API, (b) for low solubility

519

APIs, there is no change in the polymorphic form and whenever particle size is critical

520

(including low solubility APIs) there is no significant change in the particle size distribution

521

compared to that of the API lot used in the preparation of the biobatch, (c) where materials of

522

human or animal origin are used in the process, the manufacturer does not use any new

523

process for which assessment of viral safety data or transmissible spongiform encephalopathy

524

(TSE) risk assessment is required, (d) no change in qualitative and quantitative impurity

525

profile or in physicochemical properties of the API, (e) the change does not affect the

526

sterilization procedures of a sterile API, (f) the change involves only steps before the final

527

intermediate and (g) the change does not require revision of the starting material, intermediate

528

or API specifications.

Working document QAS/14.575


page 17
529
5305.

Change in batch size of the API or intermediate involving downscaling on condition

531

that (a) no changes to the manufacturing process other than those necessitated by changes in

532

scale (e.g. use of a different size of equipment) and (b) the change does not affect the

533

reproducibility of the process.

534
5356.
536

Changes to the test parameters, acceptance criteria or analytical procedures of the API
manufacturer that do not require a change to the FPP manufacturers API specifications.

537
5387.

Change to the test parameters or acceptance criteria of the API specifications of the

539

FPP manufacturer involving (a) replacement of a test parameter, (b) relaxation of an

540

acceptance criterion and (c) addition of test parameter. On condition that (a) for insoluble

541

APIs there is no change in the polymorphic form and whenever particle size is critical

542

(including low-solubility APIs), there is no change in particle size distribution acceptance

543

criteria, (b) no additional impurity found over the International Conference on Harmonisation

544

of Technical Requirements for Registration of Human Medicines (ICH) identification

545

threshold and (c) the change does not concern sterility testing.

546
5478.

Change to the analytical procedures used to control the API by the FPP manufacturer

548

involving change from a currently accepted in-house analytical procedure to an analytical

549

procedure in an officially-recognized pharmacopoeia or from the analytical procedure in one

550

officially-recognized pharmacopoeia to an analytical procedure in another officially-

551

recognized pharmacopoeia.

552
5539.

Changes to the container-closure system in immediate contact with the product or

554

additional types of container-closure on condition that (a) the product is not a sterile product,

555

(b) the new system offers equal or better protection to the product, (c) stability data are

556

available on two batches of the product in the new container for at least three months under

557

accelerated conditions (as defined in relevant guidelines) or one year under non-accelerated

558

conditions, (d) a stability study has been commenced on at least two batches of the altered

559

product for the full duration of the shelf-life and (e) the change is notified. Changes may not

560

be made to labelling without prior approval.

561
56210.

Reduction in the retest period or shelf-life of the API provided the change is not

Working document QAS/14.575


page 18
563

necessitated by unexpected events arising during manufacture or because of stability

564

concerns.

565
56611.

Change in the composition of a solution dosage form. On condition that (a) the

567

affected excipient(s) does/do not function to affect the solubility and/or the absorption of the

568

API, (b) the affected excipient(s) does/do not function as a preservative or preservative

569

enhancer, (c) no change in the specifications of the affected excipient(s) or the FPP, (d) no

570

change in the physical characteristics of the FPP (e.g. viscosity, osmolality, pH), (e) the

571

change does not concern a sterile FPP and (f) the excipients are qualitatively the same. The

572

change in the amount (or concentration) of each excipient is within 10% of the amount (or

573

concentration) of each excipient in the originally approved product.

574
57512.

Changes to flavours, perfumes or colours on condition that (a) any new colours are

576

permitted by the European Commissions List of Permitted Food Colours (4), or the United States

577

Food and Drug Agencys (US-FDA) Summary of Color Additives for Use in the United States in

578

Foods, Drugs, Cosmetics, and Medical Devices (5), (b) the change is notified, (c) stability data are

579

available on two batches of the altered product for at least three months under accelerated

580

conditions (as defined in relevant guidelines) or one year under non-accelerated conditions

581

and (d) a new stability study has been commenced on at least two batches of the altered

582

product for the full duration of the shelf-life.

583
58413.

A change or addition of imprints, embossing or other markings on solid dosage forms,

585

including replacement or addition of inks used for product markings and change in scoring

586

configuration. On condition that (a) these do not imply an unapproved indication or patient

587

population, (b) no unapproved colour (as defined above) is introduced, (c) any changes to

588

scoring are consistent with the dose schedules in the approved product information and (d)

589

the change is notified.

590
59114.

Change in dimensions without change in qualitative or quantitative composition and

592

mean mass of tablets, capsules, suppositories and pessaries. On condition that (a)

593

specifications for the FPP are updated only with respect to dimensions of the FPP and (b)

594

multipoint in vitro dissolution profiles of the current and proposed versions of the product

595

(determined in the routine release medium, on at least one batch of pilot- or production-scale)

596

are comparable.

Working document QAS/14.575


page 19
597
59815.

Addition or replacement of a manufacturing site for part or all of the manufacturing

599

process for an FPP involving (a) secondary packaging of all types of FPPs, (b) primary

600

packaging site of solid FPPs (e.g. tablets, capsules), semi-solid FPPs (e.g. ointments, creams)

601

and solution liquid FPPs and (c) primary packaging site of other liquid FPPs (suspensions,

602

emulsions). On condition that (a) satisfactory good manufacturing practices (GMP) inspection

603

in the last three years and (b) site appropriately authorized by an NMRA (to manufacture the

604

pharmaceutical form and the product concerned).

605
60616.

Change in the batch size of the FPP involving up to and including a factor of 10

607

compared to the biobatch. On condition that (a) the change does not affect the reproducibility

608

and/or consistency of the product, (b) the change pertains only to immediate-release oral

609

pharmaceutical forms and to non-sterile liquid forms, (c) changes to the manufacturing

610

method and/or to the in-process controls are only those necessitated by the change in batch

611

size, e.g. use of different-sized equipment, (d) a validation protocol is available or validation

612

of the manufacture of three production-scale batches has been successfully undertaken in

613

accordance with the current validation protocol and (f) the biobatch size was at least 100 000

614

units in the case of solid oral dosage forms.

615
61617.
617

Change to in-process tests or limits applied during the manufacture of the FPP or
intermediate involving revision or replacement of a test.

618
61918.

Change in the specifications of the FPP involving test parameters and acceptance

620

criteria: (a) relaxation of an acceptance criterion; or (b) replacement of a test parameter

621

provided that the change to the specifications does not affect the stability and the performance

622

of the product and the change does not concern sterility testing.

623
62419.

Analytical methodology for the finished product on condition that (a) validation shows

625

that the new method is equivalent to or better than the existing method and (b) major changes

626

(e.g. ultra violet assay to high-pressure liquid chromatography (HPLC)) are notified.

627
62820.

Change in the package size involving: (a) change in the number of units (e.g. tablets,

629

ampoules, etc.) in a package; and (b) change in the fill weight or fill volume of non-parenteral

630

multidose products. On condition that (a) the change is consistent with the posology and

Working document QAS/14.575


page 20
631

treatment duration accepted in the package insert and (b) no change in the primary packaging

632

material.

633
63421.

Changes to the container-closure system in immediate contact with the product or

635

additional types of container-closure. On condition that (a) the product is not a sterile product,

636

(b) the new system offers equal or better protection to the product, (c) stability data are

637

available on two batches of the product in the new container for at least three months under

638

accelerated conditions (as defined in relevant guidelines) or one year under non-accelerated

639

conditions, (d) a stability study has been commenced on at least two batches of the altered

640

product for the full duration of the shelf-life and (e) the change is notified. Changes may not

641

be made to labelling without prior approval.

642
64322.

Change in any part of the (primary) packaging material not in contact with the FPP

644

formulation (e.g. colour of flip-off caps, colour code rings on ampoules or change of needle

645

shield), provided the change does not concern a fundamental part of the packaging material,

646

which affects the delivery, use, safety or stability of the FPP.

647
64823.
649

Reduction in the shelf-life of the FPP (as packaged for sale) or in the in-use period of
the FPP (after first opening or after reconstitution or dilution).

650
651

New sites of manufacture require prior approval because the NMRA should see evidence of

652

compliance with GMP, e.g. a WHO-type certificate of pharmaceutical product (6). Changes

653

or additions to pack size also require prior approval because the new size must be consistent

654

with the approved uses of the product.

655
656

Changes may not be made to labelling without prior approval, except for changes to layout

657

without alteration of text or meaning. Pictures or diagrams may not be added without prior

658

approval because they may imply an unapproved indication.

659
660

Notifications of variations, and applications to vary, must be accompanied by this statement:

661
662

No variations have been made other than (1) those notified herewith and (2) changes

663

which are permitted without notification or prior approval according to the guidelines of

664

the medicines regulatory authority of ...........

Working document QAS/14.575


page 21
665

5.4

Variations (changes) to pharmaceutical aspects of registered products which require

666

prior approval before implementation

667
668

5.4.1 Minor variation

669
670

Below is a list of variations considered minor. In addition to the general documents stated in

671

section 3.5. Documentation required, specific documentation in support of each type of

672

variation should be provided.

673
674

1.

Replacement or addition of a new manufacturing site or manufacturer of an API involving

675

API testing only provided the transfer of analytical methods has been successfully undertaken and

676

no change in the FPP manufacturers API specifications.

677
678

2.

Change of drug product name.

3.

Change of the specification of drug substance a) specification limits are tightened and b)

679
680
681

addition of new test parameter and limits.

682
683

4.

684

Change in product labelling should be in accordance to country specific labelling requirement


and this includes:

685
686
687

a) change of the layout/artwork without altering meaning;

688

b) addition/deletion/replacement of pictures, diagrams, bar code, logos and/or texts that do not

689

imply an unapproved indication;

690

c) addition/strengthening of warnings, precautions, contraindications and/or adverse

691

events/effects to the approved product labelling;

692

d) tightening of products target population;

693

e) deletion of indication.

694
695

5.

696

Change in batch size of the API or intermediate involving more than 10-fold increase
compared to the currently accepted batch size.

697
698

6.

Change to the specifications or analytical procedures applied to materials used in the

699

manufacture of the API (e.g. raw materials, starting materials, reaction intermediates, solvents,

700

reagents, catalysts) involving addition or replacement of a specification parameter as a result of a

Working document QAS/14.575


page 22
701

safety or quality issue.

702
703

7.

704

Change of the specification of drug substance a) specification limits are tightened and b)
addition of new test parameter and limits.

705
706

8.

Change to the test parameters or acceptance criteria of the API specifications of the FPP

707

manufacturer a) addition of a test parameter, b) replacement of test parameter and c) relaxation of

708

an acceptance criterion.

709
710

9.

711

Change to the analytical procedures used to control the API by the FPP manufacturer
involving modification or replacement of an analytical procedure.

712
713

10.

714

Change in the immediate packaging (primary and functional secondary components) for the
storage and shipment of the API provided the change is not due to instability issues.

715
716

11.

Change in the retest period or shelf-life of the API involving extension provided (a) no change

717

to the primary packaging in direct contact with the API or to the recommended condition of

718

storage and (b) stability data were generated in accordance with the currently accepted stability

719

protocol.

720
721

12.

722

Any change in the labelled storage conditions of the API provided (a) the stability studies
must show compliance with specification and (b) no change in shelf-life/retest period.

723
724

13.

Change or addition of imprints, embossing or other markings, including replacement or

725

addition of inks used for product markings and change in scoring configuration involving addition

726

of a score line. On condition that (a) the change does not affect the stability or performance

727

characteristics (e.g. release rate) of the FPP, (b) changes to the FPP specifications are those

728

necessitated only by the change to the appearance or to the scoring and (c) addition or deletion of

729

a score line from a generic product is consistent with a similar change in the comparator product

730

or was requested by NMRA.

731
732

14.

Change in dimensions without change in qualitative or quantitative composition and mean

733

mass of gastroresistant, modified or prolonged-release FPPs and scored tablets. On condition that

734

specifications for the FPP are updated only with respect to dimensions of the FPP and multipoint

735

in vitro dissolution profiles of the current and proposed versions of the product (determined in the

736

routine release medium, on at least one batch of pilot- or production-scale) are comparable.

737

Working document QAS/14.575


page 23
738

15.

Addition or replacement of a manufacturing site for part or all of the manufacturing process

739

for an FPP involving all other manufacturing operations except batch control and/or release

740

testing. On condition that (a) no change in the batch formula, description of manufacturing

741

process and process controls, equipment class and process controls, controls of critical steps and

742

intermediates or FPP specifications, (b) satisfactory inspection in the last three years, (c) site

743

appropriately authorized by an NMRA (to manufacture the pharmaceutical form and the product

744

concerned) and (d) validation protocol is available or validation of the manufacturing process at

745

the new site has been successfully carried out on at least three production-scale batches in

746

accordance with the current protocol.

747
748

16.

Change in the manufacturing process of the FPP.

17.

Change in the specifications or analytical procedures for an excipient involving change or

749
750
751

replacement of an analytical procedure.

752
753

18.

754

Replacement or addition of a primary packaging type provided the change does not concern a
sterile FPP.

755
756

19.

Change in qualitative and/or quantitative composition of the immediate packaging material for

757

semisolid and liquid FPPs. On condition that (a) the change does not concern a sterile FPP, (b) no

758

change in the packaging type and material (an example of an allowable change is blister to blister)

759

and (c) the relevant properties of the proposed packaging are at least equivalent to those of the

760

currently accepted material.

761
762

20.

Extension in the shelf-life of the FPP (as packaged for sale) provided there is no change to the

763

primary packaging type in direct contact with the FPP and to the recommended conditions of

764

storage and stability data were generated in accordance with the currently accepted stability

765

protocol.

766
767

21.

768

Extension in the in-use period of the FPP (after first opening or after reconstitution or
dilution).

769
770
771

22.

Change in the labelled storage conditions of the FPP (as packaged for sale), the product
during the in-use period or the product after reconstitution or dilution.

772
773
774

5.4.2

Major variation

Working document QAS/14.575


page 24
775
776
777

Variations, which fail to fulfil the conditions for notifications or minor variations

778

automatically, become major variations. In addition, variations, which are not covered by

779

these guidelines, should be considered as a major change by default.

780
781

References

782
783

1.

Guidelines on submission of documentation for a multisource (generic) finished

784

pharmaceutical product for the WHO Prequalification of Medicines Programme: quality part.

785

In: WHO Expert Committee on Specifications for Pharmaceutical Preparations. Forty-sixth

786

report. Geneva, World Health Organization. Technical Report Series, No. 970, 2012, Annex 4.

787
788

2.

Guidance on variations to a prequalified dossier. In: WHO Expert Committee on Specifications

789

for Pharmaceutical Preparations. Forty-first report. Geneva, World Health Organization.

790

Technical Report Series, No. 943, 2007, Annex 6.

791
792

3.

Guidelines for stability testing of active pharmaceutical ingredients and finished pharmaceutical

793

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

794

Forty-third report. Geneva, World Health Organization. Technical Report Series, No. 953,

795

2009, Annex 2.

796
797

4.

798

European Commission. List of permitted food colours: EC Directive 94/36/EC. Official Journal
of the European Communities 1994; L237.

799
800

5.

801

FDA. Summary of Color Additives for Use in the United States in Foods, Drugs, Cosmetics,
and Medical Devices. 2010.

802
803

6.

Guidelines for implementation of the WHO Certification Scheme on the Quality of

804

Pharmaceutical Products Moving in International. In: WHO Expert Committee on Specifications

805

for Pharmaceutical Preparations. Thirty-fourth report. Geneva, World Health Organization.

806

Technical Report Series, No. 863, 1996, Annex 10.

807
808
809

***

Potrebbero piacerti anche