Sei sulla pagina 1di 31

Working document QAS/14.

582
April 2014
Document for comment
1

2
3

FIP-WHO TECHNICAL GUIDELINES:

POINTS TO CONSIDER IN THE

PROVISION BY HEALTH-CARE PROFESSIONALS OF

CHILDREN-SPECIFIC PREPARATIONS THAT ARE NOT

AVAILABLE AS AUTHORIZED PRODUCTS

8
9
10
11

(APRIL 2014)
DRAFT FOR COMMENT
Should you have any comments on the attached text, please send these to:
Dr Sabine Kopp, Group Lead, Medicines Quality Assurance, Technologies, Standards and Norms, World
Health Organization, 1211 Geneva 27, Switzerland; email: kopps@who.int; fax: (+41 22) 791 4730
(kopps@who.int) and to Ms Marie Gaspard (gaspardm@who.int), by 1 May 2014.
Working documents are sent out electronically and they will also be placed on the Medicines website
for comment. If you do not already receive directly our draft guidelines please let us have your email
address (to bonnyw@who.int) and we will add it to our electronic mailing list.

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

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/14.582


page 2
37
38
39
40
41

SCHEDULE FOR THE PROPOSED ADOPTION PROCESS OF DOCUMENT QAS/14.582:


FIP-WHO TECHNICAL GUIDELINES:
POINTS TO CONSIDER IN THE
PROVISION BY HEALTH-CARE PROFESSIONALS OF CHILDREN-SPECIFIC
PREPARATIONS THAT ARE NOT AVAILABLE AS AUTHORIZED PRODUCTS
First draft prepared by Professor A. Nunn, UK,
commissioned by Dr S. Hill, Medicines Access and
Rational Use, Department of Essential Medicines and
Pharmaceutical Policies, World Health Organization

March 2011

Discussion at informal consultation on paediatrics and


generics guidelines development

4-6 May 2011

Revision prepared incorporating amendments by WHO


committee

August 2011

Revision sent out for comments

August 2011

Consolidation of comments received

September 2011

Discussion at forty-sixth meeting of the WHO Expert


Committee on Specifications for Pharmaceutical
Preparations

10-14 October 2011

Revision prepared and sent out for comments

August 2012

Consolidation of comments received

September 2012

Discussion at forty-seventh meeting of the WHO Expert


Committee on Specifications for Pharmaceutical
Preparations

October 2012

Review of comments received and recommendations of


WHO Expert Committee on Specifications for
Pharmaceutical Preparations, and in-depth discussion
between the Secretaries of the FIP Expert Group and
WHO Expert Committee on Specifications for
Pharmaceutical Preparations

February 2013

Preparation of new working document for circulation

March 2013

Circulation of new working document for comments.

March 2013

Discussion during Expert Committee on the Selection


and Use of Essential Medicines

8 April 2013

Compilation of feedback received

June 2013

Any further action by WHO and FIP

July-September 2013

Working document QAS/14.582


page 3

42
43

Presentation to the forty-eighth meeting of the WHO


Expert Committee on Specifications for Pharmaceutical
Preparations

14-18 October 2013

New document received by WHO from FIP

March 2014

Circulation of new working document for comments.

March 2014

Compilation of feedback received

June 2014

Presentation to the forty-eighth meeting of the WHO


Expert Committee on Specifications for Pharmaceutical
Preparations

13-17 October 2014

Any further action as necessary

Working document QAS/14.582


page 4
FIP-WHO TECHNICAL GUIDELINES:
POINTS TO CONSIDER IN THE
PROVISION BY HEALTH-CARE PROFESSIONALS OF CHILDREN-SPECIFIC
PREPARATIONS THAT ARE NOT AVAILABLE AS AUTHORIZED PRODUCTS

44
45
46
47
48
49

The present draft is based on working document QAS/13.525 discussed at the October 2013

50

meeting of the WHO Expert Committee on Specifications for Pharmaceutical Preparations.

51

The working document text has been reorganized and brought into balance with the contents

52

of the WHO document: Development of paediatric medicines: points to consider in

53

formulation. Parts of the original draft (QAS/11.399) have been reintroduced, e.g. Appendix

54

4 on potential problems in compounding. Comments received on the working document are

55

largely taken into account. At the October 2013 meeting experts expressed concern on good

56

manufacturing practices (GMP) aspects of compounded medicines for stock. A new section

57

on GMP aspects is therefore proposed. As the document is intended for a wide audience of

58

practitioners it is advisable to include a glossary.

59
60

1.

Introduction and scope ..

61

1.1

Background ..

62

1.2

Purpose

63

1.3

Target audience and health-care settings

64

2.

Glossary....

65

3.

Alternatives to compounding ....

66

3.1

Sourcing of a commercially available or manufactured product if available

67

3.2

Dose rounding

68

3.3

Therapeutic alternatives .

69

3.4

Manipulation of dosage forms

70

3.4.1 Tablet splitting ..

71

3.4.2 Tablet/capsule dispersion for oral administration .

72

3.4.3 Crushing tablets/opening capsules and mixing powder with food


and drink

73
74

3.4.4 Giving the injectable form by the oral route

75

3.4.5 Splitting suppositories ..

76

3.4.6 Other possibilities .

77

4.

Compounding .

Working document QAS/14.582


page 5
4.1

Good manufacturing practices aspects ...

80

4.2

Potential problems ..

81

4.3

Basic considerations ...

78
79

82

5.

Information, availability and access ....

83

5.1

Standards of practice and guidelines ..

84

5.2

Formularies and compendia ...

85

5.3

Source and supply ...

86

5.4

Networks and information services ....

87

6.

References ...

88

Annex 1. Examples of therapeutic alternatives .....

89

Annex 2. Compounded preparations consideration of potential problems ....

90
91
92

1.

INTRODUCTION AND SCOPE

1.1

Background

93
94
95
96

Paediatric patients should have access to authorized, age-appropriate preparations of

97

medicines that can be administered safely and effectively. Nothing in this document should

98

detract from this objective. However, it is recognized that such preparations are not always

99

available and a safe and effective alternative must be sought.

100
101

In the context of paediatric pharmacy practice, and for the purpose of this document,

102

compounding is the technique applied by pharmacists to produce medicines from active

103

pharmaceutical ingredients (APIs) or using authorized medicines when no commercially

104

available, authorized, age-appropriate or adequate dosage form exists. Unless stated explicitly

105

in this document, the compounded medicine is assumed to be dispensed immediately after

106

preparation and not kept in stock. Compounding does not apply to reconstitution of

107

authorized medicines prior to dispensing.

108
109

Compared to the use of authorized medicines there are significant risks associated with

110

compounding; quality, safety and efficacy can rarely all be assured, and there have been

Working document QAS/14.582


page 6
111

many errors reported in the preparation of such medicines. In some situations compounding

112

of a medicine for a child may be the only option, which may be supported by industry-

113

verified compounding. There may be alternatives to compounding which also should be

114

considered, for example, use of a therapeutic alternative or manipulation of authorized dosage

115

forms.

116
117

This points-to-consider document is supported by a literature review of the evidence available:

118

Report for WHO on findings of a review of existing guidance/advisory documents on how

119

medicines should be administered to children, including general instructions on

120

extemporaneous preparations and manipulation of adult dosage forms (working document

121

QAS/11.400 available on demand).

122
123

The document is to consider as a time-limited document that addresses current needs for

124

advice in the search for an alternative to an authorized, age-appropriate dosage form.

125

Wherever possible the guidance is informed by the relevant evidence. However, the evidence

126

base is weak or non-existent in most situations. Consequently, the guidance is predominantly

127

informed by best practice, based on sound scientific and therapeutic principles and expert

128

consensus. Whilst the guidance is a working practical document it is important to invite

129

comment and input from interested practitioners so that the guidance can be developed

130

further in response to feedback.

131
132

1.2

Purpose

133
134

The purpose of the document is to:

135
136

medicines for paediatric patients;

137
138

provide evidence-based or best practice advice about alternatives to compounding of

describe and educate practitioners in the potential problems of compounding and how
to avoid them;

139
140

provide brief advice on compounding;

141

provide a bibliography of relevant literature, supporting evidence and existing

142
143

guidance.

Working document QAS/14.582


page 7
144

The document will not reproduce areas where existing guidance and standards exist (e.g.

145

GMP standards for facilities and documentation). Where appropriate, reference is made to the

146

relevant resources and publications.

147
148

1.3

Target audience and health-care settings

149
150

The document is intended for a wide audience of health-care stakeholders including:

151
152

pharmacists, physicians, paediatricians and nursing staff;

153
154

all practitioners involved in health care of the paediatric population but mainly

national drug regulatory authorities and professional bodies, e.g. national paediatric
organizations, national pharmacy associations;

155
156

general hospitals and health clinics;

157

specialized paediatric hospitals and primary care clinics;

158

pharmaceutical industry given its role in providing information.

159
160

Pharmaceutical manufacturers can often provide useful information on validated

161

compounded formulae and other information relating to the manipulations and specific

162

characteristics of formulations.

163
164

The document may be particularly useful in resource-poor situations where access to age-

165

appropriate dosage forms is limited and where it may be difficult to obtain relevant

166

information and/or achieve the highest standards of quality assurance when dispensing

167

compounded preparations.

168
169

2. GLOSSARY

170
171

active pharmaceutical ingredient (API)

172

Any substance or mixture of substances intended to be used in the manufacture of a

173

pharmaceutical dosage form and that, when so used, becomes an active ingredient of that

174

pharmaceutical dosage form. Such substances are intended to furnish pharmacological

Working document QAS/14.582


page 8
175

activity or other direct effect in the diagnosis, cure, mitigation, treatment or prevention of

176

disease, or to affect the structure and function of the body.

177
178

authorized dosage form

179

A pharmaceutical dosage form that has been authorized by the competent authority to be

180

marketed for the treatment of specific indications.

181
182

child-resistant container

183

A form of packaging difficult for young children to open but not unduly difficult for adults to

184

open properly.

185
186

compounding

187

The technique applied by pharmacists to produce non-authorized medicines from active

188

pharmaceutical ingredients or using authorized medicines.

189
190

dispensing pharmacy

191

The pharmacy receiving the prescription for a patient and providing the pharmaceutical

192

preparation to the patient. For compounded medicines, the dispensing pharmacy is not

193

necessarily the preparing pharmacy.

194
195

dose rounding

196

197
198

excipient

199

Any ingredient other than the active pharmaceutical ingredient, which has been evaluated for

200

safety and included in a pharmaceutical dosage form to (i) aid the processing of the dosage

201

form during its manufacture, (ii) protect, support or enhance stability, bioavailability or

202

patient acceptability, (iii) assist in product identification, or (iv) enhance any other attribute

203

of the overall safety and effectiveness of the dosage form during storage or use.

204
205

good manufacturing practices (GMP)

206

A philosophy of practice and processes to assure the quality and safety of manufactured

207

pharmaceutical products; specified in for example WHO guidelines.

Working document QAS/14.582


page 9
208
209

labelling information

210

Information to the user provided on the package label or in the patient information leaflet.

211
212

manipulation of a dosage form

213

Operation performed by the pharmacist or parent/caregiver to facilitate the administration of

214

a dosage form. It can be simple, e.g. breaking a tablet, or complex, for example, using tablets

215

as a source for an active pharmaceutical ingredient to prepare a suspension.

216
217

pharmaceutical dosage form

218

The physical form in which a medicine is presented; the name of a dosage forms combines its

219

physical form and the intended route of administration, e.g. a tablet (to be swallowed), oral

220

suspension (liquid suspension of solid particles intended for oral intake and swallowing).

221
222

route of administration

223

The way in which a medicine is given to a patient, e.g. oral administration (administration via

224

the oral route), rectal administration (administration to the rectum).

225
226

SmPC

227

Summary of product characteristics approved by the competent authority

228
229

3.

ALTERNATIVES TO COMPOUNDING

230
231

Before deciding to compound consider possible alternatives that will give the greatest

232

assurance of clinical effectiveness and safety.

233
234

In some situations, for example, if the stability and method of preparation of an oral liquid are

235

well documented, e.g. if compounding has been industry-verified and all facilities and

236

ingredients are available, it may be less compelling to seek an alternative. On the other hand,

237

if there are no stability data and, for example, the API forms a caking suspension in the only

238

available excipients (e.g. a syrup), an alternative must be considered to ensure safe and

239

effective treatment.

240

Working document QAS/14.582


page 10
241

In any case, the decision on how to prepare and/or provide a non-authorized preparation

242

should be based on an assessment of risks and benefits of the dosing strategy. On a case-by-

243

case basis, potential benefits from use should be weighed against all possible risks arising

244

from preparation and administration of such medicines. Even in cases where the compounded

245

preparation can be considered a validated formulation, the impact of compounding on

246

bioavailability may not be known.

247
248

Main alternatives to compounding are:

249
250

3.1

Sourcing of a commercially-available or manufactured product1 if available

251
252

The logistics of supply and access are obvious factors that might work against this but

253

practitioners should liaise with suppliers, importers and regulatory authorities to access these

254

products if possible.

255
256

Importation of products may be expensive and reputable suppliers should be used to avoid

257

spurious/falsely-labelled/falsified/counterfeit (SFFC) medicines. Quality assurance systems

258

should be in place, for example, to ensure that recall systems are available and information

259

provided in the local language.

260
261

Large-scale use of compounded oral liquids for children should not be justified on the

262

grounds that they are cheaper than commercial products. Other options, including local

263

manufacture using GMP standards, should be investigated.

264
265

3.2

Dose rounding

266
267

If the dose prescribed does not correspond to a dosage form which is commercially available,

268

consider whether the dose can be suitably amended whilst maintaining safety and efficacy.

269

The therapeutic index of the medicine and patient characteristics are to be considered before

270

making a decision.

271

This includes products prepared to GMP standards, for example at an accredited hospital manufacturing unit.

Working document QAS/14.582


page 11
272

Some drug doses are calculated accurately on the basis of body weight yet the therapeutic

273

index is such that one dose can be used for a broad weight or age band. Consult the WHO

274

Model Formulary for Children. Available from:

275

http://apps.who.int/medicinedocs/en/m/abstract/Js17151e/

276
277

3.3

Therapeutic alternatives

278
279

If a medicine is prescribed in a formulation which is not available, e.g. in an age-appropriate

280

form, consider the possibility of using a medicine with a similar therapeutic action which is

281

available in a more suitable form. Examples are presented in Appendix 1.

282
283

3.4

Manipulation of dosage forms

284
285

In situations where the prescribed dose is less than what is commercially available, if there

286

are swallowing difficulties, or the stability of a compounded oral liquid cannot be assured,

287

consider the possibilities for manipulation of a dosage form as outlined below.

288
289

The practitioner should have in mind that manipulation, such as tablet splitting, tablet/capsule

290

dispersion or tablet crushing and mixing with food or drink, may increase the potential for

291

inaccurate dosing and it may affect the stability and bioavailability of the dosage form, in

292

particular when mixed with food or drink. Excipients that are safe for adults may not

293

necessarily be so for children.

294
295

When medicines are mixed with food or drink, an unpleasant taste of the mix may cause

296

aversion by the child. Mixing with breast milk could potentially be used in very young

297

children (< 6 months) but may cause aversion in breastfed children; the mother should

298

therefore observe milk intake and signs for breast-milk refusal.

299
300

3.4.1 Tablet splitting

301

Many tablets are designed to allow splitting, either by breaking if scored or by using a

302

purposely designed tablet cutter. If the child is able to take solid dose forms safely (age will

303

vary but usually from age 68 and above), a tablet fraction can be given, otherwise it can be

304

dispersed or mixed with food or juice as below.

Working document QAS/14.582


page 12
305
306

There is evidence that splitting of marketed tablet formulations may result in segments that

307

are far from meeting pharmacopoeia requirements for dose uniformity. Unless the tablet is

308

provided with a score line to facilitate breaking and swallowing the full dose, it is important

309

that regulatory authorities and manufacturers assure that such tablet formulations that may be

310

splitted also meet the relevant uniformity requirements for the tablet segments, and that

311

information about possible splitting occurs in the summary of product characteristics (SmPC).

312
313

In the lack of appropriate information and especially when the API is potent or has a narrow

314

therapeutic index, compounding may be preferable when an accurate dose cannot be assured.

315

Thus, consider on a case-by-case basis whether splitting of tablets might lead to toxicity or

316

reduced effect.

317
318

Not all tablets can be split. In general, those with a sustained release or enteric coating cannot

319

be split but it may be possible to split those with a sustained-release matrix. Formularies or

320

manufacturers information and the SmPC should be consulted.

321
322

Consideration should be given to splitting tablets with an appropriate commercial tablet

323

splitter in the pharmacy. If possible tablets with score lines and uniform distribution of the

324

active drug should be sourced and information sought on the stability of segments. If carers

325

are cutting segments, they should be given a commercial tablet splitter with adequate

326

instruction on the method of preparing and storing tablet segments.

327
328

3.4.2 Tablet/capsule dispersion for oral administration

329

It may be possible to disperse tablets or the contents of capsules in water or other liquid. If

330

the tablet disperses, the tablet or a fraction of the tablet can be dispersed in a small volume

331

appropriate for the concerned child, and the whole dose given when a suspension is formed,

332

mixed with a flavored vehicle if required. Assure that the whole dose is administered by

333

flushing the container. Consider the impact of dispersion and the risk for interactions with the

334

contents of the flavoured vehicle on bioavailability.

335
336

Conventional tablets do not disperse readily but some form a suspension within a short time.

337

Soluble tablets and dispersible tablets disintegrate and dissolve or disperse within a short time

Working document QAS/14.582


page 13
338

in water at room temperature. When the API is known to be soluble, dissolving or dispersing

339

the tablet in a known volume of water can allow a fractional dose to be appropriately

340

measured with a syringe as in the case of captopril. As extraction of soluble API from the

341

tablet excipients may be incomplete the suspension should be shaken or stirred prior to

342

measuring the dose and not filtered unless it has been established that the API is not removed.

343

In the case of a poorly soluble API the measurement of a fractional dose by taking an aliquot

344

from a suspension formed in this way is generally not recommended due to probable rapid

345

sedimentation of insoluble API and resultant dosage inaccuracy.

346
347

The remainder of a tablet or capsule dispersion shall not be stored and reused because of

348

stability reasons (microbiological, chemical, physicochemical).

349
350

Tablet dispersion may not always be practical for infants when the doses required are the

351

equivalent of small tablet fractions that are unable to be reliably prepared, e.g. a fifth of a

352

tablet or if the tablet is not scored.

353
354

When dispersion is intended for tube feeding, parameters such as particle size, viscosity,

355

dosing volume and compatibility of the oral preparation with the tube material should be

356

considered. Dispersions may be too viscous or contain large particles that can make the

357

administration by feeding tube not feasible. Adsorption of API to the tube material results in

358

inappropriate dosing; this concern is most relevant for lipophilic and potent APIs.

359
360

WHO is promoting the use of flexible solid oral dosage forms such as dispersible tablets (see

361

Development of paediatric medicines: points to consider in formulation, WHO Technical

362

Report Series, No. 970, Annex 5). The use of custom-made dispersible tablets for paediatric

363

dosing should be used wherever possible but there is a need to ensure that carers understand

364

how they are to be administered.

365
366

3.4.3 Crushing tablets/opening capsules and mixing powder with food or drink

367

The practice of crushing tablets or opening capsules and adding the powder to a palatable

368

drink or sprinkling onto solid food are common alternatives. However, there may be little

369

evidence to support the efficacy and safety since stability and bioavailability may be altered.

370

In case of potent APIs, consider the risks associated with handling of powdered material for

Working document QAS/14.582


page 14
371

parents/carers. Modified-release tablets and capsules cannot be crushed or opened without

372

affecting bioavailability and/or stability, and this should therefore not be done.

373
374

In general, the possibility to crush tablets should be based on bioavailability studies.

375

Information should be sought from manufacturers (e.g. the SmPC and website) and

376

formularies whenever possible. The process is acceptable only if bioavailability is not

377

affected by food or drink, and when the product is used immediately.

378
379

It is difficult to ensure that a complete dose has been taken and the practice of nurses and

380

carers handling powdered medicines may present health concerns. Tablet dispersion may be a

381

simpler, more reliable and potentially safer method.

382
383

Aliquots should usually not be taken from liquid-filled capsules since it is difficult to remove

384

and measure the total contents.

385
386

3.4.4 Giving the injectable form by the oral route

387

Oral administration is possible, although an expensive option, for some injections. If the

388

injectable form of the API is the same as the oral form (for example, labetalol hydrochloride,

389

ondansetron hydrochloride) it can be assumed that the API will be absorbed enterally from

390

the injectable formulation. However, as the API is in solution more rapid absorption and

391

higher peak levels may occur compared to the slower absorption from a solid oral dose form.

392

When evaluating whether an injection is suitable for oral use specialist advice, e.g.

393

consultation with a medicines information centre, should be sought because there are

394

important factors which must be considered, e.g. first-pass effect, oral bioavailability, gastric

395

acidity (e.g. effect on stability), pH effects (e.g. precipitation of soluble salts of weak acids)

396

and palatability.

397
398

Injections may contain excipients that are undesirable in some patients, e.g. propylene glycol

399

and ethanol. The pH of some injections may mean that they should not be given orally or be

400

diluted before administration to avoid irritation, e.g. furosemide injection (pH 9),

401

pantoprazole injection (pH 910.5), phenytoin sodium injection (pH 1012).

402
403

Working document QAS/14.582


page 15
404

3.4.5 Splitting suppositories

405

There is little information on the accuracy with which suppositories can be split. Splitting is

406

usually associated with major problems with regard to accurate dosing. The majority of

407

commercially available suppositories are formulated as suspensions, which means that

408

sedimentation of the solid API particles may occur during solidification of the suppository;

409

therefore, if suppositories need to be split, this should be done lengthwise.

410
411

Suppositories have been melted and recast into smaller moulds. This option is associated with

412

a risk of recrystallization and for affecting the distribution of the API resulting in over- or

413

under-dosing. It is therefore generally discouraged.

414
415

Therapeutic index and the consequences of over- or under-dosing should be taken into

416

account when determining whether it is safe to split suppositories. If possible, this should be

417

done in the pharmacy and segments should be weighed to ensure the desired weight.

418
419

3.4.6 Other possibilities

420

In any case where a change of the route of administration as intended for an authorized

421

medicine is considered advice should be sought from formularies and the literature and even

422

specialist advice. In general, use of an altered route of administration results in a different

423

pharmacokinetic profile introducing a high risk of dosing errors and may compromise safety

424

and efficacy.

425
426

4.

COMPOUNDING

4.1

Good manufacturing practices aspects

427
428
429
430

The dispensing pharmacy receives the prescription for a patient and provides the

431

compounded preparation to the patient. Preparation may take place in another pharmacy or

432

hospital manufacturing unit.

433
434

When a non-authorized medicine is prepared for stock the preparing pharmacy or hospital

435

unit should meet GMP requirements to personnel, premises and equipment, quality assurance

436

system, documentation and product dossier. Further, an authorization by the competent

Working document QAS/14.582


page 16
437

authority to carry out operations should be considered. Reference in this aspect is made to the

438

PIC/S Guide to Good Practices for the preparation of Medicinal Products in Healthcare

439

Establishments (www.picscheme.org) and to other guidelines, including WHO guidelines on

440

GMP and corresponding national guidelines.

441
442

In case of compounding as a single event and the preparation is dispensed immediately to the

443

patient, requirements may be less strict. Nevertheless, there are quality assurance

444

requirements to take into account:

445
446

the preparing pharmacy should have appropriate premises and equipment;

447

the pharmacist and staff must have sufficient training and background for
compounding;

448
449

access to relevant literature (pharmacopoeias, handbooks, scientific journals, etc.) and


the Internet;

450
451

general instructions for every type of preparation should be available;

452

a record on each preparation should be retained showing the key processing and
packaging steps including the name of the person responsible for each step.

453
454
455

4.2

Potential problems

456
457

Formulation of a compounded medicine is associated with a number of potential problems

458

that may impact on the safety and effectiveness of the formulation. Awareness of the relative

459

complexity of the formulation and the things that can go wrong will help to avoid such

460

problems. Guidance on compounding can be found in, for example, Marc Jackson and

461

Andrew Lowey: Handbook of Extemporaneous Preparation, Pharmaceutical Press, 2010.

462
463

Consideration must be given to the properties of the API (e.g. aqueous solubility, pH effect

464

on solubility, particle size when poorly soluble) and stability of both API and the

465

compounded formulation, i.e. chemical, physical and microbiological instability.

466
467

Further consideration must be given to the selection of excipients and excipients present in

468

manipulated dose forms. It is important to consider any possible adverse effects of the

469

inactive components of the preparation. The use of preservatives, ethanol and sugars must

Working document QAS/14.582


page 17
470

be carefully considered. Some guidance and literature references on the formulation can be

471

found in Development of paediatric medicines: points to consider in formulation (WHO

472

Technical Report Series, No. 970, Annex 5).

473
474

Annex 2 presents examples of potential problems in compounding.

475
476

4.3

Basic considerations

477
478

Quality of API and excipients

479

It is important to ensure that the API and excipients meet pharmacopoeia standards

480

with regard to both identity and purity. The choice of excipients should be restricted

481

to those that have been used in authorized medicines intended for the same route of

482

administration.

483
484

Consider use of an authorized dosage form as a starting point

485

It may be safer and more effective to crush tablets or use the contents of hard capsules

486

with an appropriate suspending vehicle rather than preparing from API and excipients.

487

There are many formulations available with validated shelf-life but sourcing of

488

suspending agents may be difficult and/or expensive.

489
490

There might be instances where the pharmacist crushes a number of tablets or opens a

491

number of capsules, dilutes the powder with a suitable excipient and doses the powder

492

in ready-to-use single dose sachets. Before doing so, consider the stability of the

493

preparation including in-use stability against humidity and exposure to air.

494
495

Consult literature and guidelines if available

496

If possible, always use a validated formulation (i.e. based on literature, stability

497

studies and guidelines). Consult product information and the latest national and

498

international guidelines and/or a specialist information centre if possible.

499
500

Employ the principles of GMP

501

This involves the processes put in place to give the highest level of assurance possible

502

that the product will be safe and effective. It is accepted that few units will be able to

Working document QAS/14.582


page 18
503

conform to the requirements of GMP. However, as stated in Section 3.1, the

504

principles of quality assurance are possible and essential under any conditions.

505
506

Medication error potential

507

There have been many examples of medication error when preparing compounded

508

medicines and some have resulted in death or serious harm to patients. The potential

509

for medication error must be recognized and steps taken to minimize the risk. This

510

will include as a minimum the use of a worksheet listing the formulation ingredients

511

and the identity of ingredients; quantities, calculations and measurements should be

512

double-checked by trained personnel and signatures provided. The pharmacist

513

responsible should check the final product and label against the signed worksheet,

514

ingredients and prescription.

515
516

Caution in extrapolating from other formulations

517

Caution is required if extrapolating the formulation from a published study or

518

formulary. Formulations made from APIs may be more stable than formulations made

519

from solid dose forms and vice versa. Tablet and capsule excipients can increase or

520

decrease the stability of the API in an oral liquid preparation. The salt form of the

521

drug used in a published study could be different to the form locally available and this

522

may affect the drugs solubility and stability. Consult pharmacopoeias and seek

523

specialist advice if possible.

524
525

Similarly, the results of a published study using a drug mixed with a commercial

526

suspending base (e.g. Ora-Plus) cannot generally be extrapolated to a situation

527

where the same drug is mixed with a simple base of syrup or glycerol.

528
529

Formulations for compounded medicines based on APIs and crushed tablets are not

530

interchangeable.

531
532

Dose uniformity may be a problem explain importance of shaking prior to use

533

If the API is poorly soluble in water, it will generally be more chemically stable in an

534

aqueous suspension, but uniformity of dosing the suspension may be a problem. A

535

suspending agent will be required. Always check that the finished preparation

Working document QAS/14.582


page 19
536

resuspends under in-use conditions and explain the importance of resuspension by

537

shaking to patients/carers.

538
539

If the API is soluble, it will generally be less stable in a liquid preparation. As the

540

API is soluble, the inclusion of a suspending agent is less important in a preparation

541

based on crushed tablets.

542
543

As excipients and other formulation components can affect solubility, all compounded

544

liquid formulations should be shaken prior to administration. The entire API may not

545

be in solution even if it is highly soluble. The only exception would be if the

546

preparation is made from pure API and it can be assured that the entire API is in

547

solution.

548
549

Exceptionally, when no published formulation is available

550

The pharmacist must assess the risks for different options and use knowledge and

551

experience to formulate a product.

552
553

o Obtain physicochemical properties of the API if available

554

API solubility and the pH stability profile are useful when considering the

555

approach to formulation or dose administration. If possible, obtain basic

556

physicochemical information about the API, especially the aqueous solubility

557

of the API at the expected pH of the final preparation. This allows a

558

judgement on whether an API solution or suspension is formed.

559
560

o Test the physical characteristics prior to patient use

561

Tablet/capsule formulations vary worldwide, especially with respect to

562

excipients content. The differences can influence the effectiveness and

563

acceptability of the preparation. Basic performance tests should be done

564

before patient use, particularly on formulations prepared for the first time.

565

This includes ease of resuspension and pouring, degree of caking on storage,

566

observation of physical behaviour and characteristics.

567

Working document QAS/14.582


page 20
o Consider risk of microbial growth

568
569

All compounded liquid formulations are highly susceptible to microbial

570

growth. An antimicrobial preservative must be included unless the final

571

product will be used completely within 23 days and stored under

572

refrigeration. Oral liquids that are not adequately preserved will support rapid

573

growth of bacteria and fungi especially at warm to hot temperatures and can

574

pose hazards to patients especially if immunosuppressed.

575
576

Preparation of compounded liquids should be carried out under conditions to

577

minimize the introduction of microbial contamination.

578
579

Use appropriate final containers

580

Final containers and closures should be clean and free from dust and other residues. It

581

is recommended to use new containers. Containers that are reused should be

582

thoroughly washed, rinsed with sterile or freshly boiled water and dried. Light-

583

protective (e.g. dark plastic or amber glass) containers should generally be used.

584

Consider the use of a light protective wrapping such as foil if a light-protective

585

container is not available.

586
587

Selection of the final container should consider potential drug adsorption to plastic

588

containers.

589
590

Dosing device

591

In view of the dosing needs of liquid preparations the feasibility of appropriate dosing

592

should be confirmed as not all dosing devices may allow delivering the required

593

volume. As most compounded liquids should be shaken prior to administration, this

594

may introduce entrapped air in the liquid, which may cause problems with accurate

595

measurement of small volumes.

596
597

Consider in-use storage

598

In-use storage conditions may vary considerably from those in a published study or

599

formulary recommendation. Always consider if it will be possible to store and use the

600

preparation under the optimal conditions described in the study, which usually are

Working document QAS/14.582


page 21
601

refrigeration, protection from light and with minimal possibility of in-use

602

contamination. If these conditions are not possible locally it can be assumed that the

603

preparation will be less stable and more susceptible to microbial growth. Reduce the

604

shelf-life (e.g. from one month to one week) according to professional judgement. If

605

possible, consult expert advice.

606
607

Expiry date

608

Chemical stability and potential for microbial growth under patient-use conditions are

609

seldom tested in published studies.

610
611

It is recommended that each compounded preparation be given an expiry date

612

assigned in a conservative way taking into account API-specific and general stability

613

documentation and literature when available. This will encourage regular fresh

614

preparations and help to assure effectiveness and safety. It also allows the practitioner

615

to regularly review the patients use of the preparation.

616
617

The following items should be considered when determining such expiry date: nature

618

of the API and its degradation mechanisms; dosage form and its components;

619

potential for microbial proliferation in the preparation; container in which the

620

preparation is packaged; expected storage conditions; and the intended duration of

621

therapy.

622
623

When an authorized medicine is used as the source of the API, stability information

624

could be obtained from the manufacturer. Otherwise, applicable information on

625

stability, compatibility and degradation of ingredients and use has to be sought in the

626

literature.

627
628

Give clear instruction to caregivers/patients

629

This may include instructions on storage, resuspension, changes in taste, smell,

630

appearance, adverse effects and other pharmaceutical advices.

631
632

It is common practise that parents/carers add powdered dosage form to a liquid

633

(water, juice, etc.) or sprinkle it onto food to improve palatability and mask

Working document QAS/14.582


page 22
634

unpleasant taste. This should be done only when bioavailability is not affected and

635

intake of the full dose can be ensured. Provide parents/carers with appropriate

636

information.

637
638

If an oral syringe or other measuring device is used it is important to check the

639

technique to ensure the correct dose is administered. Advise the use of clean

640

measuring devices and ways to avoid contaminating the preparation when preparing

641

the dose.

642
643

Label information
In addition to dosage instructions, include at least the following information:

644
645
646

if applicable, the name of the pharmaceutical preparation;

647

the route of administration;

648

the name(s) of the API(s) and excipients of known pharmacological action,


e.g. antimicrobial agents, antioxydants;

649
650

if liquid, concentration(s) of the API(s), e.g. in mg/ml, and the amount or


volume of the preparation in the container;

651
652

if solid, amount(s) of the API(s) in each dose and the number of doses in the
container;

653
654

reference or batch number (or date of preparation);

655

expiry date (do not use after...);

656

any special storage conditions and handling precautions that may be


necessary, e.g. to be shaken before use;

657
658

the pharmacy name and contact information;

659

name of the patient.

660
661

Document concerns and share information

662

Practitioners are encouraged to maintain dialogue with regulatory bodies and

663

international agencies and networks about problems and concerns associated with the

664

preparation and availability of age-appropriate medicines for children. The sharing of

665

solutions to problems is also important.

666

Working document QAS/14.582


page 23
667

5.

INFORMATION, AVAILABILITY AND ACCESS

668
669

A number of networks, websites and other resources are available which provide information

670

on standards of practice, formulas for compounded preparations, manufacturers, suppliers of

671

oral liquid formulations, and networks and responsive information services. These should be

672

consulted by practitioners and regulators to provide the safest and most effective treatment

673

options for children who require an age- appropriate formulation.

674
675

5.1

Standards of practice and guidelines

676
677

Some national, regional and international guidelines for extemporaneous formulations and

678

medicines administration to children have been published. Consulting these documents may

679

assist in forming local policies of practice and educational activities for practitioners.

680
681

5.2

Formularies and compendia

682
683

These may be helpful in providing formulation advice and general advice on dosage

684

manipulations. The information in these formularies may be difficult to transfer to a local

685

situation where the base ingredients (e.g. commercial suspending bases, antimicrobial

686

preservatives, pure drug powder) are not readily available.

687
688

The eMixt database (www.pharminfotech.co.nz) is being developed to provide

689

comprehensive information for all settings and environments.

690
691

Mark Jackson and Andrew Lowey: Handbook of Extemporaneous Preparations,

692

Pharmaceutical Press 2010, contains formulations and associated stability summaries for oral

693

liquid preparations.

694
695

5.3

Source and supply

696
697

A database of sources and prices of medicines for children has been compiled by Unicef. This

698

will be available electronically by mid-2011

699

(http://www.unicef.org/supply/index_47129.html)

Working document QAS/14.582


page 24
700
701

The database can be searched to find worldwide suppliers of oral liquids and other age-

702

appropriate formulations for paediatric use.

703
704

5.4

Networks and information services

705
706

Local, national and international medicines information centres may respond to

707

questions about formulation such as the WHO Paediatric medicines Regulatory

708

Network (PmRN). Partnerships and twinning arrangements between hospitals in

709

poorly-resourced countries and developed countries can be explored and are often

710

beneficial.

711

Questions can also be posted via the eMIxt website www.pharminfotech.co.nz.

712
713
714

Sharing of information and advice on paediatric formulations should be explored


whenever possible.

715
716
717

International discussion lists can be useful for posting questions on formulations and

718

the archives can be searched for previous questions and answers. Examples include

719

eDrug and INDICES accessed via http://www.essentialdrugs.org/.

720
721

6.

REFERENCES

722
723

1.

Pharmaceutical development for multisource (generic) pharmaceutical products. In: WHO

724

Expert Committee on Specifications for Pharmaceutical Preparations. Forty-sixth report.

725

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

726

2012 (http://www.who.int/medicines/areas/quality_safety/quality_assurance/en/).

727
728

2.

Development of paediatric medicines: points to consider in pharmaceutical

729

development. In: WHO Expert Committee on Specifications for Pharmaceutical

730

Preparations. Forty-sixth report. Geneva, World Health Organization. WHO Technical

731

Report Series, No. 970, Annex 5, 2012

732

(http://www.who.int/medicines/areas/quality_safety/quality_assurance/en/).

Working document QAS/14.582


page 25
733
734

3.

WHO good manufacturing practices: main principles for pharmaceutical products

735

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

736

eighth report. Geneva, World Health Organization. WHO Technical Report Series, No.

737

986, Annex 2, 2014 (in print).

738
739

4.

Good manufacturing practices for pharmaceutical products. In: Quality assurance of

740

pharmaceuticals. WHO guidelines, related guidance and GXP training materials,

741

CD-ROM 2013.

742
743

5.

The International Pharmacopoeia, Fourth Edition, including First, Second and Third

744

Supplements (http://who.int/medicines/publications/pharmacopoeia/en/index.html).

745

Available online and CD-ROM version (2013).

746
747

6.

Report for WHO on findings of a review of existing guidance/advisory documents on

748

how medicines should be administered to children, including general instructions on

749

extemporaneous preparations and manipulation of adult dosage forms (working

750

document QAS/11.400).

751
752

7.

The WHO Model Formulary for Children (2010)


(http://www.who.int/selection_medicines/list/WMFc_2010.pdf).

753
754
755

8.

Report of the Informal Expert Meeting on Dosage Forms of Medicines for Children.

756

Geneva, World Health Organization. December 2008

757

[http://www.who.int/selection_medicines/committees/expert/17/application/paediatric/

758

Dosage_form_re-port DEC2008.pdf.

759
760

9.

Handbook of Extemporaneous Preparation: A guide to pharmaceutical compounding.

761

Part A. Standards: In Handbook of Extemporaneous Preparation, pages 165. Mark

762

Jackson and Andrew Lowey. Pharmaceutical Press, 2010.

763
764
765

10.

Pharmaceutical Inspection Co-operation Scheme (http://www.picscheme.org/)


In particular the following documents which can be downloaded free of charge: PE

Working document QAS/14.582


page 26
766

009-9 (Part I). PIC/S GMP guide (Part I: Basic requirements for medicinal products);

767

PE 010-3 Guide to good practices for the preparation of medicinal products in

768

healthcare establishments.

769
770

11.

Kastango ES, Trissel LA, Bradshaw BD. An Ounce of Prevention: Controlling Hazards

771

in Extemporaneous Compounding Practices. International Journal of Pharmacy

772

Compounding. 2003; 7(5): 401-16.

773
774
775
776
777

Working document QAS/14.582


page 27
ANNEX 1

778
779

Examples of therapeutic alternatives to extemporaneous formulations

780
781

Required

Possible alternative

Notes

Enalapril liquid

Captopril liquid

Available commercially in
some countries.

Dispersed captopril tablets

Captopril tablets can be


easily dispersed prior to
giving the dose

Naproxen oral liquid

Ibuprofen liquid if available

NSAID may not be clinically


justified.
Paracetamol (acetaminophen)
may be a suitable and safer
alternative

Felodipine oral liquid

Dispersed amlodipine tablets

Salts of amlodipine are very


soluble and fractional doses
can be prepared.

Tinidazole oral liquid

Metronidazole oral liquid

Very few reasons why


tinidazole should be
preferred over metronidazole

782
783

In some cases the therapeutic alternative may not be available as an oral liquid but as a more

784

easily manipulated form (see felodipine example above).

785
786
787
788
789
790
791
792

Working document QAS/14.582


page 28
ANNEX 2

793
794
795

Compounded preparations consideration of potential problems

796
797

The most frequently used method is to grind the required number of tablets to a fine

798

powder in a mortar and form a slurry by adding a small volume of water. At this stage

799

there is the potential for the operator to be exposed to hazardous powdered drug and

800

microbial contamination of the product if clean equipment is not used.

801
802

Excipients such as antimicrobial preservatives, suspending agents and flavouring agents

803

are added to make the final product. A frequently used base is a mixture of glycerol or

804

syrup, a suspending agent such as methylcellulose and para-hydroxybenzoates (parabens)

805

as a preservative. Other agents sometimes added include alternative solvents such as

806

ethanol, particularly when the drug is poorly soluble in water, and buffer systems to

807

provide the optimum pH for drug stability or activity of the antimicrobial preservative.

808

Whilst ostensibly simple, such formulations can be complex, comprising a mixture of the

809

base and a suspension or solution (usually a combination of both) of tablet excipients and

810

active drug. If the drug is water soluble there is a temptation to filter out the insoluble

811

tablet excipients to leave a clear solution but filtration can remove significant amounts of

812

drug if extraction from tablets is incomplete. Insoluble tablet excipients are in suspension

813

and may compromise product appearance, whereas soluble excipients may alter drug

814

stability, for example, by changing the pH of the preparation. Added ingredients or

815

excipients may not be appropriate for babies and infants, e.g. ethanol, propylene glycol.

816

Whilst there may be advantages in using pure drug powder instead of tablets it may not be

817

easily obtainable.

818
819

The expiry date of a compounded oral liquid is assigned empirically or based on

820

published information on a particular formulation. A conservative approach must be

821

adopted when assigning an expiry date because of lack of information on API stability or

822

limitations in either the design or the conclusions of a published report. Also, it may be

823

impractical to entirely reproduce the conditions of a study which was performed in

824

another institution or country under the controlled conditions of an experiment rather than

825

clinical use. Most studies base their expiry date recommendation on chemical stability but

Working document QAS/14.582


page 29
826

do not address possible physical or microbiological spoilage which may be significant

827

during actual use of the product. For these reasons extemporaneously prepared oral

828

liquids should only be used for a maximum of one month from the date of preparation to

829

minimize any unrecognized product deterioration. Longer expiry dates may be applied if

830

more extensive testing is performed.

831
832

Finally, when deciding on a formulation, it is important to consider any possible adverse

833

effects of the "inactive" components of the preparation. Sucrose (in syrup) can promote

834

the formation of dental caries, ethanol can cause hypoglycaemia and para-

835

hydroxybenzoates can cause hypersensitivity reactions and exacerbate the symptoms of

836

asthma. It has also been suggested that benzoates and para-hydroxybenzoates can

837

aggravate neonatal hyperbilirubinaemia by displacing bilirubin which is bound to plasma,

838

proteins but this effect has not been demonstrated in vivo and the amounts present in oral

839

formulations are unlikely to pose any risk. Limits for the inclusion of ethanol in

840

paediatric formulations have been proposed by the American Academy of Pediatrics but

841

there is little supporting evidence for the recommendation.

842
843

Deterioration of an oral liquid may be due to chemical, physical or microbiological

844

instability which can lead to a subtherapeutic dose of drug, exposure to toxic degradation

845

products or ingestion of unacceptable numbers of microorganisms. It is important for

846

pharmacists, clinicians and nursing staff to be aware of potential problems caused by

847

instability to ensure that drug therapy is effective and safe.

848
849

Chemical instability

850
851

APIs in compounded liquids may be susceptible to chemical reactions leading to

852

degradation. The most common reactions are hydrolysis, oxidation and reduction. Usually

853

the reaction rate or type is influenced by pH, for example, azathioprine is rapidly

854

hydrolysed to 6-mercaptopurine at alkaline pH but is relatively stable in acidic or neutral

855

conditions. Other factors which may increase the rate of reaction include the presence of

856

trace metals which catalyse the oxidation of captopril, methyldopa or exposure to light,

857

which catalyses the oxidative degradation of 6-mercaptopurine. The rate of chemical

858

degradation usually increases with temperature, a factor which is the basis for accelerated

Working document QAS/14.582


page 30
859

stability trials of pharmaceutical formulations. Preparations made from tablets contain

860

excipients such as binders and disintegrating agents in addition to the API. These

861

excipients may reduce chemical stability by changing the pH to a value at which more

862

rapid degradation occurs. This probably explains why amiloride solution prepared from

863

pure API is more stable than an oral liquid prepared from tablets.

864
865

The API in the preparation may be totally or partially in solution or predominantly in the

866

solid state as a suspension. APIs in solution are more susceptible to chemical degradation

867

than APIs in the solid state (i.e. suspensions), thus suspensions of acetazolamide and

868

chlorothiazide are more stable than solutions. However it cannot be assumed in all cases

869

that a compounded suspension is more stable than a solution. In a suspension, an

870

equilibrium exists between the API in the solid state and an API in solution, and even

871

though the amount of drug dissolved may be minimal the conditions could be optimal for

872

degradation. Furosemide is a notable example which undergoes hydrolysis in acidic

873

conditions where the solid state is predominant, but is much more stable at alkaline pH

874

where it is totally in solution.

875
876

Microbiological instability

877
878

Microbial growth in an oral liquid may cause foul odour and turbidity and adversely

879

effect palatability and appearance. High titres of microorganisms may be hazardous to

880

health especially in very young or immunocompromised patients. By-products of

881

microbial metabolism may cause a change in the pH of the preparation and reduce the

882

chemical stability or solubility of the drug. Microbial contamination during preparation

883

must be minimized by using clean equipment, sterile water (Water for Irrigation (BP))

884

and avoiding contaminated raw materials and containers. If sodium benzoate or benzoic

885

acid are used as antimicrobial preservatives the final pH must be less than 5 so that the

886

active unionized form is predominant. Consequently the API must also be stable at this

887

pH.

888
889

Effective preservative systems require rigorous evaluation which is seldom performed on

890

compounded formulations. Many factors can reduce the effectiveness of the preservative

891

including use of contaminated materials, chemical degradation, binding of preservative to

Working document QAS/14.582


page 31
892

suspending agents or tablet excipients, incorrect storage or unhygienic use of the final

893

product.

894
895

Physical instability

896
897

Compounded oral suspensions may be susceptible to sedimentation of insoluble API

898

causing caking. Difficulty in resuspending the API or rapid sedimentation following

899

shaking can lead to erratic dosage measurement as demonstrated with chlorothiazide

900

suspension and this inherent problem with compounded formulations is of considerable

901

concern. Some spironolactone suspensions have been reported to be excessively thick and

902

almost unpourable. Refrigeration, whilst usually desirable to maximize chemical stability

903

and reduce microbial growth, can also increase the viscosity of a suspension making

904

resuspension more difficult or cause the precipitation of API or preservatives. It is

905

important to consider the effect on pH of all components of the formulation and the

906

possible impact on stability. Syrup, for example, is relatively acidic and if used in

907

phenobarbitone sodium oral solution it will cause the precipitation of unionized

908

phenobarbitone.

909
910

***

Potrebbero piacerti anche