Annex 2: WHO Good Manufacturing Practices For Biological Products
Annex 2: WHO Good Manufacturing Practices For Biological Products
Annex 2: WHO Good Manufacturing Practices For Biological Products
1. Introduction 96
2. Scope 96
3. Terminology 100
4. Principles and general considerations 104
5. Pharmaceutical quality system and quality risk management 106
6. Personnel 106
7. Starting materials 107
8. Seed lots and cell banks 109
9. Premises and equipment 111
10. Containment 113
11. Clean rooms 115
12. Production 116
13. Campaign production 118
14. Labelling 119
15. Validation 119
16. Quality control 121
17. Documentation (batch processing records) 122
18. Use of animals 123
19. Authors and acknowledgements 125
20. References 127
93
WHO Expert Committee on Biological Standardization Sixty-sixth report
94
Annex 2
Abbreviations
AEFI adverse event following immunization
ATMP advanced therapy medicinal product
BCG bacille Calmette–Guérin
GMP good manufacturing practice(s)
HEPA high-efficiency particulate air
HVAC heating, ventilation and air conditioning
IgE immunoglobulin E
mAb monoclonal antibody
MCB master cell bank
MSL master seed lot
MVS master virus seed
NRA national regulatory authority
PDL population doubling level
PQR product quality review
PQS pharmaceutical quality system
QRM quality risk management
rDNA recombinant DNA
SPF specific pathogen free
TSE transmissible spongiform encephalopathy
WCB working cell bank
WSL working seed lot
WVS working virus seed
95
WHO Expert Committee on Biological Standardization Sixty-sixth report
1. Introduction
Biological products can be defined according to their source material and
method of manufacture. The source materials and methods employed in the
manufacture of biological products for human use therefore represent critical
factors in shaping their appropriate regulatory control. Biological products are
derived from cells, tissues or microorganisms and reflect the inherent variability
characteristic of living materials. The active substances in biological products
are often too complex to be fully characterized by utilizing physicochemical
testing methods alone and may show a marked heterogeneity from one
preparation and/or batch to the next. Consequently, special considerations
are needed when manufacturing biological products in order to maintain
consistency in product quality.
Good manufacturing practices (GMP) for biological products were
first published by WHO in 1992 (1). This current revision reflects subsequent
developments that have taken place in science and technology, and in the
application of risk-based approaches to GMP (2–14). The content of this
document should be considered complementary to the general recommendations
set out in the current WHO good manufacturing practices for pharmaceutical
products: main principles (2) and in other WHO documents related specifically
to the production and control of biological products.
This document is intended to serve as a basis for establishing national
guidelines for GMP for biological products. If a national regulatory authority
(NRA) so desires, the guidance provided may be adopted as definitive national
requirements, or modifications may be justified and made by the NRA in light
of the risk–benefit balance and legal considerations in each authority. In such
cases, it is recommended that any modification to the principles and technical
specifications set out below should be made only on the condition that the
modifications ensure product quality, safety and efficacy that are at least
equivalent to that recommended in this document.
WHO Technical Report Series No. 999, 2016
2. Scope
The guidance provided in this document applies to the manufacture, control
and testing of biological products for human use – from starting materials
and preparations (including seed lots, cell banks and intermediates) to the
finished product.
Manufacturing procedures within the scope of this document include:
■■ growth of strains of microorganisms and eukaryotic cells;
■■ extraction of substances from biological tissues, including human,
animal and plant tissues, and fungi;
96
Annex 2
97
98
WHO Technical Report Series No. 999, 2016
Table 1
Scope of the current document (illustrative)
Type and source Example products Application of this document to steps in manufacture
of material
1. Animal or plant Heparins, insulin, Collection of plant, Cutting, mixing Isolation and Formulation
sources: non- enzymes, proteins, organ, tissue or and/or initial purification and filling
transgenic allergen extract, ATMPs, fluid processing
animal immune sera
2. Virus or bacteria/ Viral or bacterial Establishment Cell culture Inactivation Formulation
fermentation/cell vaccines, enzymes, and and/or when applicable, and filling
culture proteins maintenance of fermentation isolation and
MCB, WCB, MSL/ purification
MVS, WSL/WVS
3. Biotechnology Recombinant products, Establishment Cell culture Isolation, Formulation
fermentation/cell mAbs, allergens, and and/or purification and and filling
culture vaccines, gene therapy maintenance of fermentation modification
WHO Expert Committee on Biological Standardization Sixty-sixth report
99
WHO Expert Committee on Biological Standardization Sixty-sixth report
3. Terminology
In addition to the terms defined in WHO good manufacturing practices for
pharmaceutical products: main principles (2) and WHO good manufacturing
practices for sterile pharmaceutical products (3), the definitions given below
apply to the terms as used in the current document. These terms may have
different meanings in other contexts.
Active substance: a defined process intermediate containing the active
ingredient, which is subsequently formulated with excipients to produce the drug
product. This may also be referred to as “drug substance” or “active ingredient”
in other documents.
Adventitious agents: contaminating microorganisms of the cell culture
or source materials, including bacteria, fungi, mycoplasmas/spiroplasmas,
mycobacteria, rickettsia, protozoa, parasites, transmissible spongiform
encephalopathy (TSE) agents and viruses that have been unintentionally
introduced into the manufacturing process of a biological product. The source
of these contaminants may be the legacy of the cell line, or the raw materials
used in the culture medium to propagate the cells (in banking, in production or
in their legacy), the environment, personnel, equipment or elsewhere.
Allergen: a molecule capable of inducing an immunoglobulin E (IgE)
response and/or a Type I allergic reaction.
Antibodies: proteins produced naturally by the B-lymphocytes that
bind to specific antigens. Using rDNA technology antibodies are also produced
in other (continuous) cell lines. Antibodies may be divided into two main types
– monoclonal and polyclonal antibodies – based on key differences in their
methods of manufacture. Also called immunoglobulins.
Antigens: substances (for example, toxins, foreign proteins, bacteria,
tissue cells and venoms) capable of inducing specific immune responses.
Axenic: a single organism in culture which is not contaminated with
WHO Technical Report Series No. 999, 2016
bank (WCB).
Monoclonal antibodies (mAbs): homogenous antibody population
obtained from a single clone of lymphocytes or by recombinant technology
and which bind to a single epitope.
Pharmaceutical quality system (PQS): management system used by a
pharmaceutical company to direct and control its activities with regard to quality.
Polyclonal antibodies: antibodies derived from a range of lymphocyte
clones and produced in humans and animals in response to the epitopes on
most “non-self ” molecules.
Primary containment: a system of containment that prevents the
escape of a biological agent into the immediate working environment. It
involves the use of closed containers or biological safety cabinets along with
secure operating procedures.
102
Annex 2
104
Annex 2
6. Personnel
6.1 Personnel responsible for production and control should have an adequate
background in relevant scientific disciplines such as microbiology, biology,
WHO Technical Report Series No. 999, 2016
7. Starting materials
7.1 The source, origin and suitability of active substances, starting materials
(for example, cryo-protectants and feeder cells), buffers and media (for
example, reagents, growth media, serum, enzymes, cytokines, growth
factors and amino acids) and other components of the finished product
should be clearly defined and controlled according to the principles set out
in WHO guidance on GMP for pharmaceutical products (2).
107
WHO Expert Committee on Biological Standardization Sixty-sixth report
7.2 Manufacturers should retain information describing the source and quality
of the biological materials used for at least 1 year after the expiry date
of the finished products and according to local regulations concerning
biological products. It has been found that documents retained for longer
periods may provide useful information related to adverse events following
immunization (AEFIs) and other investigations.
7.3 All starting material suppliers (that is, manufacturers) should be initially
qualified on the basis of documented criteria and a risk-based approach.
Regular assessments of their status should also be carried out. Particular
attention should be given to the identification and monitoring of any
variability that may affect biological processes. When starting materials
are sourced from brokers who could increase the risk of contamination
by performing repackaging operations under GMP (2, 4) they should be
carefully qualified; an audit may form part of such qualification, as needed.
7.4 An identity test, or equivalent, should be performed on each batch of
received starting materials prior to release. The number of containers
sampled should be justified on the basis of QRM principles and in agreement
with all applicable guidelines (2). The identification of all starting materials
should be in compliance with the requirements appropriate to the stage
of manufacture. The level of testing should be commensurate with the
qualification level of the supplier and the nature of the materials used. In the
case of starting material used to manufacture active substances the number
of samples taken should be based on statistically recognized criteria and
QRM principles (2). However, for starting materials and intermediates used
in the formulation of finished product each container should be sampled
for identity testing in accordance with the main principles of GMP for
pharmaceutical products unless reduced testing has been validated.
7.5 The sampling process should not adversely affect the quality of the
WHO Technical Report Series No. 999, 2016
8.8 Each storage container should be adequately sealed, clearly labelled and
kept at an appropriate temperature. A stock inventory should be kept. The
storage temperature should be recorded continuously and, where applicable,
the liquid nitrogen level should be monitored. Any deviation from the set
limits, and any corrective and preventive action taken, should be recorded.
Temperature deviations should be detected as early as possible (for example,
through the use of an alarm system for temperature and nitrogen levels).
8.9 Seed lots and cell banks should be stored and used in such a way as to
minimize the risks of contamination or alteration (for example, stored
in qualified ultra-low temperature freezers or liquid nitrogen storage
containers). Control measures for the storage of different seeds and/or
cells in the same area or equipment should prevent mix-up and should
take into account the infectious nature of the materials in order to prevent
cross-contamination.
8.10 MSLs, MCBs, and preferably also WSLs and WCBs, should be stored in
two or more controlled separate sites in order to minimize the risk of
total loss due to natural disaster, equipment malfunction or human error.
A contingency plan should be in place.
8.11 The storage and handling conditions for the cell or seed banks should
be defined. Access should be controlled and restricted to authorized
personnel, and appropriate access records maintained. Records of location,
identity and inventory of individual containers should also be kept. Once
containers are removed from the seed lot/cell bank management system
they should not be returned to stock.
10. Containment
10.1 Airborne dissemination of live microorganisms and viruses used for the
production process, including those from personnel, should be avoided.
10.2 Adequate precautions should be taken to avoid contamination of the
drainage system with dangerous effluents. Drainage systems should be
designed in such a way that effluents can be effectively neutralized or
decontaminated to minimize the risk of cross-contamination. Specific and
validated decontamination systems should be considered for effluents
when infectious and/or potentially infectious materials are used for
production. Local regulations should be complied with in order to
minimize the risk of contamination of the external environment according
to the risk associated with the biohazardous nature of waste materials.
10.3 Dedicated production areas should be used for the handling of live cells
capable of persistence in the manufacturing environment, for pathogenic
organisms of Biosafety Risk Group 3 or 4 and/or for spore-forming
organisms until the inactivation process is accomplished and verified. For
Bacillus anthracis, Clostridium tetani and Clostridium botulinum strictly
dedicated facilities should be utilized for each individual product.
Up‑to-date information on these and other high-risk or “special” agents
should be sought from major information resources (27). Where campaign
manufacture of spore-forming organisms occurs in a facility or suite of
facilities only one product should be processed at any one time.
Use of any pathogenic organism above Biosafety Risk Group 3
may be permitted by the NRA according to the biohazard classification
of the organism, the risk assessment of the biological product and its
emergency demand.
113
WHO Expert Committee on Biological Standardization Sixty-sixth report
10.9 Areas where Biosafety Risk Group 3 or 4 organisms are handled should
always have a negative air pressure relative to the environment. This will
ensure the containment of the organism in unlikely events such as failure
of the door interlock. Air-lock doors should be interlocked to prevent
them being opened simultaneously. Differential pressure alarms should be
present wherever required, and should be validated and monitored.
10.11 Where the filtration of exhaust air is necessary, the safe changing of filters
should be ensured or bag-in-bag-out housings should be employed. Once
removed, filters should be decontaminated and properly destroyed. In
addition to HEPA filtration other inactivation technologies such as heat
inactivation and steam scavenging may be considered for exhaust air to
ensure effective inactivation of pathogenic organisms of Biosafety Risk
Group 3 and/or 4.
12. Production
12.1 Since cultivation conditions, media and reagents are designed to promote
the growth of cells or microbial organisms, typically in an axenic state,
particular attention should be paid to the control strategy for ensuring that
effective steps are in place for preventing or minimizing the occurrence of
unwanted bioburden, endotoxins, viruses of animal and human origin, and
associated metabolites.
12.2 The QRM process should be the basis for implementing the technical and
organizational measures required to control the risks of contamination
and cross-contamination. These could include, though are not limited to:
■■ carrying out processing and filling in segregated areas;
■■ containing material transfer by means of an airlock and appropriate
type of pass box with validated transfer procedures, clothing change
and effective washing and decontamination of equipment;
■■ recirculation of only treated (HEPA-filtered) air;
■■ acquiring knowledge of the key characteristics (for example,
pathogenicity, detectability, persistence and susceptibility to
WHO Technical Report Series No. 999, 2016
12.9 Antibiotics may be used during the early stages of production to help
prevent inadvertent microbial contamination or to reduce the bioburden
of living tissues and cells. In this case, the use of antibiotics should be well
justified, and they should be cleared from the manufacturing process at
the stage specified in the marketing authorization. Acceptable residual
levels should be defined and validated. Penicillin and other beta-lactam
antibiotics should not be used at any stage of the process.
12.10 A procedure should be in place to address equipment and/or accessories
failure (such as air vent filter failure) which should include a product impact
review. If such failures are discovered following batch release the NRA
should be notified and the need for a batch recall should be considered.
14. Labelling
14.1 The information provided on the inner label (also called the container
label) and on the outer label (on the packaging) should be readable and
legible, and the content approved by the NRA.
14.2 Minimal key information should be printed on the inner label, and
additional information should be provided on the outer label (for example,
carton) and/or product leaflet.
14.3 The suitability of labels for low and ultra-low storage temperatures should
be verified, if applicable. The label should remain properly attached to the
container under different storage conditions during the shelf-life of the
product. The label and its adhesive should have no adverse effect on the
quality of the product caused by leaching, migration and/or other means.
15. Validation
15.1 Biological processes, handling of live materials and using campaign-based
production, if applicable, are the major aspects of biological product
manufacturing which require process and cleaning validation. The
validation of such processes – given the typical variability of biological
products, the possible use of harmful and toxic materials and the need
for inactivation processes – plays an important role in demonstrating
production consistency and in proving that the critical process parameters
119
WHO Expert Committee on Biological Standardization Sixty-sixth report
a defined time period – for example, 1 year, based on the regular product
quality review (PQR) – may indicate a need for process revalidation.
15.8 The integrity and specified hold times of containers used to store
intermediate products should be validated unless such intermediate
products are freshly prepared and used immediately.
Based upon the principles defined in the above working group and
drafting group meetings the first draft of these Guidelines was prepared by Mr R.
Acs, Central Drugs Standard Control Organisation, India; Dr B. Yáñez Chamizo,
Centro para el Control Estatal de Medicamentos, Equipos y Dispositivos Médicos,
Cuba; Dr S. Fakhrzadeh, Ministry of Health and Medical Education, the Islamic
Republic of Iran; Mrs K. Porkaew, Ministry of Public Health, Thailand; Dr S.O.
Rumiano, Consultant, Buenos Aires, Argentina; Dr Y. Wang, National Institutes
for Food and Drug Control, China; Mr B. Wibisono, National Agency of Drug
and Food Control, Indonesia; Mr M. Eisenhawer, WHO Regional Office for
South-East Asia, India; Dr A. Chawla, Consultant, Greater Noida, India; Dr A.R.
Khadem, World Health Organization, Switzerland; V.G. Maqueda, Biologist,
Buenos Aires, Argentina; and Dr D. Lei, World Health Organization, Switzerland.
A second draft was then prepared by V.G. Maqueda, Biologist, Buenos
Aires, Argentina; Dr B. Yáñez Chamizo, Centro para el Control Estatal de
Medicamentos, Equipos y Dispositivos Médicos, Cuba; Dr S. Fakhrzadeh,
Ministry of Health and Medical Education, the Islamic Republic of Iran;
Dr S.O. Rumiano, Consultant, Buenos Aires, Argentina; Dr Y. Wang, National
Institutes for Food and Drug Control, China; Mr B. Wibisono, National Agency
of Drug and Food Control, Indonesia; Mr M. Eisenhawer, WHO Regional
Office for South-East Asia, India; Dr A. Chawla, Consultant, Greater Noida,
India; and Dr A.R. Khadem and Dr D. Lei, World Health Organization,
Switzerland following a consultation held in Tunis, Tunisia, 22–24 July 2014
and attended by: Dr H. Baiao, National Authority for Medicines and Health
Products, Portugal; Mrs R. Bose, Ministry of Health and Family Welfare, India;
Mr C. Cabral, Butantan Institute, Brazil; Dr R. Chaplinsky, GSK Vaccines,
Belgium; Dr A. Chawla, Consultant, Greater Noida, India; Mr M. Diagne,
Direction de la Pharmacie et des Laboratoires, Senegal; Mr M. Eisenhawer,
WHO Regional Office for South-East Asia, India; Dr S. Fakhrzadeh, Ministry
WHO Technical Report Series No. 999, 2016
of Health and Medical Education, the Islamic Republic of Iran; Mrs R. Frikha,
Directorate of Pharmacy Inspection, Tunisia; Dr M. Gershman, Pfizer, the USA;
Ms A.R. Cornelio Geyer, Agência Nacional de Vigilância Sanitária, Brazil; Dr E.
Griffiths, Consultant, Kingston-upon-Thames, the United Kingdom; Dr N.
Harjee, Consultant, Ontario, Canada; Ms D.T.M. Hang, Ministry of Health,
Viet Nam; Dr H. Langar, WHO Regional Office for the Eastern Mediterranean,
Egypt; Dr P. Lauer, Sanofi Pasteur, France; Dr C.K. Lee, Korea Food and Drug
Administration, Republic of Korea; Dr H. Leng, Medicines Regulatory Authority,
South Africa; Dr M.G. Lopez Santos, Comisión Federal para la Protección contra
Riesgos Sanitarios, Mexico; V.G. Maqueda, Biologist, Buenos Aires, Argentina;
Dr A. Mihaylova, Bulgarian Drug Agency, Bulgaria; Dr J. Miteva, Bulgarian
Drug Agency, Bulgaria; Dr S. Pagliusi, DCVMN International, Switzerland;
126
Annex 2
20. References
1. Good manufacturing practices for biological products. In: WHO Expert Committee on Biological
Standardization: forty-second report. Geneva: World Health Organization; 1992: Annex 1 (WHO
Technical Report Series, No. 822; http://www.who.int/biologicals/publications/trs/areas/
vaccines/gmp/WHO_TRS_822_A1.pdf?ua=1, accessed 8 November 2015).
2. WHO good manufacturing practices for pharmaceutical products: main principles. In: WHO Expert
Committee on Specifications for Pharmaceutical Preparations: forty-eighth report. Geneva:
World Health Organization; 2013: Annex 2 (WHO Technical Report Series, No. 986; http://www.
who.int/medicines/areas/quality_safety/quality_assurance/TRS986annex2.pdf?ua=1, accessed 8
November 2015).
3. WHO good manufacturing practices for sterile pharmaceutical products. In: WHO Expert
Committee on Specifications for Pharmaceutical Preparations: forty-fifth report. Geneva: World
Health Organization; 2011: Annex 6 (WHO Technical Report Series, No. 961;
http://www.who.int/medicines/areas/quality_safety/quality_assurance/GMPSterilePharmaceutical
ProductsTRS961Annex6.pdf?ua=1, accessed 8 November 2015).
127
WHO Expert Committee on Biological Standardization Sixty-sixth report
4. WHO good manufacturing practices for active pharmaceutical ingredients. In: WHO Expert
Committee on Specifications for Pharmaceutical Preparations: forty-fourth report. Geneva: World
Health Organization; 2010: Annex 2 (WHO Technical Report Series, No. 957;
http://www.who.int/medicines/areas/quality_safety/quality_assurance/GMPActivePharmaceutical
IngredientsTRS957Annex2.pdf?ua=1, accessed 8 November 2015).
5. Guide to good manufacturing practice for medicinal products. Part I, Annex 2. Manufacture
of biological medicinal substances and products for human use. Pharmaceutical Inspection
Convention and Pharmaceutical Inspection Co-operation Scheme (PIC/S); 1 March 2014 (http://
www.fda.gov.ph/attachments/article/224762/pe-009-11-gmp-guide-xannexes.pdf, accessed 8
November 2015).
6. Guide to good manufacturing practice for medicinal products, Part II. Pharmaceutical Inspection
Convention and Pharmaceutical Inspection Co-operation Scheme (PIC/S); 1 March 2014
(http://www.medsafe.govt.nz/regulatory/Guideline/PE_009-8_GMP_Guide%20_Part_II_Basic_
Requirements_for_API.pdf, accessed 8 November 2015).
7. EU Guidelines for good manufacturing practice for medicinal products for human and
veterinary use. Annex 2: Manufacture of biological active substances and medicinal products
for human use. Brussels: European Commission; 2013.
8. EU Guidelines for good manufacturing practice for medicinal products for human and veterinary
use. Part I. Chapter 6: Quality control. Brussels: European Commission; 2005.
9. Current good manufacturing practice for finished pharmaceuticals. Code of Federal
Regulations Title 21, Vol. 4, revised 1 April 2014. Silver Spring, MD: United States Food and
Drug Administration; 2014 (http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.
cfm?CFRPart=211&showFR=1, accessed 4 July 2015).
10. WHO good practices for pharmaceutical quality control laboratories. In: WHO Expert Committee
on Specifications for Pharmaceutical Preparations: forty-fourth report. Geneva: World Health
Organization; 2010: Annex 1 (WHO Technical Report Series, No. 957;
http://www.who.int/medicines/areas/quality_safety/quality_assurance/Goodpractices
PharmaceuticalQualityControlLaboratoriesTRS957Annex1.pdf?ua=1, accessed 2 February 2016).
11. Good manufacturing practice for drugs (2010 revision). Beijing: China Food and Drug
Administration; 2011 (http://eng.sfda.gov.cn/WS03/CL0768/65113.html, accessed 8 November
2015).
12. WHO guidelines on good manufacturing practices for blood establishments. In: WHO Expert
Committee on Specifications for Pharmaceutical Preparations: forty-fifth report. Geneva: World
WHO Technical Report Series No. 999, 2016
Health Organization; 2011: Annex 4 (WHO Technical Report Series, No. 961; http://www.who.int/
bloodproducts/publications/GMP_Bloodestablishments.pdf?ua=1, accessed 2 February 2016).
13. WHO good distribution practices for pharmaceutical products. In: WHO Expert Committee
on Specifications for Pharmaceutical Preparations: forty-fourth report. Geneva: World Health
Organization; 2010: Annex 5 (WHO Technical Report Series, No. 957; http://www.who.int/
medicines/areas/quality_safety/quality_assurance/GoodDistributionPracticesTRS957Annex5.
pdf?ua=1, accessed 2 February 2016).
14. WHO guidelines on quality risk management. In: WHO Expert Committee on Specifications for
Pharmaceutical Preparations: forty-seventh report. Geneva: World Health Organization; 2013:
Annex 2 (WHO Technical Report Series, No. 981; http://www.who.int/medicines/areas/quality_
safety/quality_assurance/Annex2TRS-981.pdf?ua=1, accessed 2 February 2016).
15. Recommendations for the production, control and regulation of human plasma for fractionation.
In: WHO Expert Committee on Biological Standardization: fifty-sixth report. Geneva: World
Health Organization; 2007: Annex 4 (WHO Technical Report Series, No. 941; http://www.who.int/
bloodproducts/publications/TRS941Annex4blood.pdf?ua=1, accessed 2 February 2016).
128
Annex 2
16. Good manufacturing practices: supplementary guidelines for the manufacture of investigational
pharmaceutical products for clinical trials in humans. In WHO Expert Committee on Specifications
for Pharmaceutical Preparations: thirty-fourth report. Geneva: World Health Organization; 1996:
Annex 7 (WHO Technical Report Series, No. 863;
http://www.who.int/medicines/areas/quality_safety/quality_assurance/Investigational
PharmaceuticalProductsClinicalTrialsHumansTRS863Annex7.pdf?ua=1, accessed 2 February 2016).
17. WHO guidelines on nonclinical evaluation of vaccines. In: WHO Expert Committee on Biological
Standardization: fifty-fourth report. Geneva: World Health Organization; 2005: Annex 1 (WHO
Technical Report Series, No. 927; http://www.who.int/biologicals/publications/trs/areas/vaccines/
nonclinical_evaluation/ANNEX%201Nonclinical.P31-63.pdf?ua=1, accessed 8 November 2015).
18. Guidelines on clinical evaluation of vaccines: regulatory expectations. In: WHO Expert Committee
on Biological Standardization: fifty-second report. Geneva: World Health Organization; 2004:
Annex 1 (WHO Technical Report Series, No. 924; http://www.who.int/biologicals/publications/
trs/areas/vaccines/clinical_evaluation/035-101.pdf?ua=1, accessed 2 February 2016).
19. Guidelines on the nonclinical evaluation of vaccine adjuvants and adjuvanted vaccines. In:
WHO Expert Committee on Biological Standardization: sixty-fourth report. Geneva: World
Health Organization; 2014: Annex 2 (WHO Technical Report Series, No. 987; http://www.who.int/
biologicals/areas/vaccines/TRS_987_Annex2.pdf?ua=1, accessed 2 February 2016).
20. Laboratory biosafety manual, third edition. Geneva: World Health Organization; 2004 (http://
www.who.int/csr/resources/publications/biosafety/Biosafety7.pdf, accessed 8 November 2015).
21. Guidelines on viral inactivation and removal procedures intended to assure the viral safety
of human blood plasma products. In: WHO Expert Committee on Biological Standardization:
fifty-second report. Geneva: World Health Organization; 2004: Annex 4 (WHO Technical Report
Series, No. 924; http://www.who.int/bloodproducts/publications/WHO_TRS_924_A4.pdf?ua=1,
accessed 21 January 2016).
22. ICH Harmonised Tripartite Guideline Q10. Pharmaceutical quality system. Geneva: International
Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals
for Human Use; June 2008 (http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/
Guidelines/Quality/Q10/Step4/Q10_Guideline.pdf, accessed 8 November 2015).
23. Recommendations to assure the quality, safety and efficacy of BCG vaccines. In: WHO Expert
Committee on Biological Standardization: sixty-second report. Geneva: World Health Organization;
2013: Annex 3 (WHO Technical Report Series, No. 979; http://www.who.int/biologicals/areas/
vaccines/TRS_979_Annex_3.pdf?ua=1, accessed 21 January 2016).
24. WHO Guidelines on transmissible spongiform encephalopathies in relation to biological and
pharmaceutical products. Geneva: World Health Organization; 2003 (http://www.who.int/
biologicals/publications/en/whotse2003.pdf?ua=1, accessed 2 February 2016).
25. Environmental monitoring of clean rooms in vaccine manufacturing facilities: points to consider
for manufacturers of human vaccines. Geneva: World Health Organization; 2012 (http://www.who.
int/immunization_standards/vaccine_quality/env_monitoring_cleanrooms_final.pdf?ua=1,
accessed 2 February 2016).
26. Recommendations for the evaluation of animal cell cultures as substrates for the manufacturer
of biological medicinal products and for the characterization of cell banks. In: WHO Expert
Committee on Biological Standardization: sixty-first report. Geneva: World Health Organization;
2013: Annex 3 (WHO Technical Report Series, No. 978; http://www.who.int/biologicals/vaccines/
TRS_978_Annex_3.pdf?ua=1, accessed 2 February 2016).
27. Biosafety [website]. Atlanta, GA: Centers for Disease Control and Prevention (http://www.cdc.gov/
biosafety/, accessed 8 November 2015).
129
WHO Expert Committee on Biological Standardization Sixty-sixth report
28. WHO Global Action Plan to minimize poliovirus facility-associated risk after type-specific
eradication of wild polioviruses and sequential cessation of OPV use. Geneva: World Health
Organization; 2015 (http://www.polioeradication.org/Portals/0/Document/Resources/
PostEradication/GAPIII_2014.pdf, accessed 8 November 2015).
29. Guidelines for the safe production and quality control of inactivated poliomyelitis vaccine
manufactured from wild polioviruses (Addendum, 2003, to the Recommendations for the
production and quality control of poliomyelitis vaccine (inactivated)). In: WHO Expert Committee
on Biological Standardization: fifty-third report. Geneva: World Health Organization; 2004:
Annex 2 (WHO Technical Report Series, No. 926; http://www.who.int/biologicals/publications/
trs/areas/vaccines/polio/Annex%202%20(65-89)TRS926Polio2003.pdf?ua=1, accessed 2 February
2016).
30. Recommendations for the preparation, characterization and establishment of international and
other biological reference standards (revised 2004). In: WHO Expert Committee on Biological
Standardization: fifty-fifth report. Geneva: World Health Organization; 2006: Annex 2 (WHO
Technical Report Series, No. 932; http://www.who.int/immunization_standards/vaccine_
reference_preparations/TRS932Annex%202_Inter%20_biol%20ef%20standards%20rev2004.
pdf?ua=1, accessed 2 February 2016).
31. Guidelines on stability evaluation of vaccines. In: WHO Expert Committee on Biological
Standardization: fifty-seventh report. Geneva: World Health Organization; 2011: Annex 3 (WHO
Technical Report Series, No. 962; http://www.who.int/biologicals/vaccines/Annex_3_WHO_
TRS_962-3.pdf?ua=1, accessed 21 January 2016).
32. Supplementary guidelines on good manufacturing practices: validation. In: WHO Expert
Committee on Specifications for Pharmaceutical Preparations: fortieth report. Geneva: World
Health Organization; 2006: Annex 4 (WHO Technical Report Series, No. 937;
http://www.who.int/medicines/areas/quality_safety/quality_assurance/SupplementaryGMP
ValidationTRS937Annex4.pdf?ua=1, accessed 2 February 2016).
33. A WHO guide to good manufacturing practice (GMP) requirements. Part 2: Validation. Geneva:
World Health Organization; 1997 (WHO/VSQ/97.02; http://apps.who.int/iris/bitstream/10665/
64465/2/WHO_VSQ_97.02.pdf?ua=1, accessed 2 February 2016).
34. Guideline on bioanalytical method validation. Committee for Medicinal Products for Human Use.
London: European Medicines Agency; 2009 (EMEA/CHMP/EWP/192217/2009 Rev.1; http://www.
ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2011/08/WC500109686.
pdf, accessed 8 November 2015).
WHO Technical Report Series No. 999, 2016
35. Guidance for industry. Bioanalytical method validation. Rockville, MD: Center for Veterinary
Medicine; 2013
(http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/
ucm368107.pdf, accessed 8 November 2015).
36. Guidelines for independent lot release of vaccines by regulatory authorities. In: WHO Expert
Committee on Biological Standardization: sixty-first report. Geneva: World Health Organization;
2013: Annex 2 (WHO Technical Report Series, No. 978; http://www.who.int/biologicals/TRS_978_
Annex_2.pdf?ua=1, accessed 2 February 2016).
130