WHO TRS 999 Corrigenda Web PDF
WHO TRS 999 Corrigenda Web PDF
WHO TRS 999 Corrigenda Web PDF
W H O Te c h n i c a l R e p o r t S e r i e s
999
on Biological
guidance documents. Following these discussions, a WHO
guidance document on Regulatory assessment of approved
rDNA-derived biotherapeutics was adopted along with WHO
Standardization
Guidelines on the stability evaluation of vaccines for use under
extended controlled temperature conditions and on WHO good
manufacturing practices for biological products. In addition,
revised WHO Recommendations to assure the quality, safety
and efficacy of recombinant human papillomavirus virus-like
particle vaccines were also adopted by the Committee.
Subsequent sections of the report provide information on the
current status and proposed development of international Sixty-sixth report
reference materials in the areas of antibiotics; biotherapeutics
other than blood products; blood products and related
substances; in vitro diagnostic device reagents; and vaccines
Further information on these and other WHO publications can be obtained from
WHO Press, World Health Organization, 1211 Geneva 27, Switzerland
(tel.: +41 22 791 3264; fax: + 41 22 791 4857; email: [email protected];
order online: www.who.int/bookorders)
W H O Te c h n i c a l R e p o r t S e r i e s
9 9 9
This report contains the collective views of an international group of experts and
does not necessarily represent the decisions or the stated policy of the World Health Organization
WHO Library Cataloguing-in-Publication Data:
WHO Expert Committee on Biological Standardization, sixty-sixth report.
(WHO technical report series ; no. 999)
1.Biological Products - standards. 2.Vaccines - standards. 3.Blood - standards.
4.Anti-Bacterial Agents - standards. 5.Reference Standards. 6.Diagnostic Test Approval.
I. World Health Organization. II. WHO Expert Committee on Biological Standardization
(2015: Geneva, Switzerland). III. Series.
ISBN 978 92 4 120999 1 (NLM classification: QW 800)
ISBN (PDF) 978 92 4 069563 4
ISSN 0512-3054
Committee members1
Professor K. Cichutek, Paul-Ehrlich-Institut, Langen, Germany
Dr J. Epstein, Center for Biologics Evaluation and Research, Food and Drug Administration,
Silver Spring, MD, United States of America (USA) (also Blood Regulators Network (BRN)
representative)
Dr E. Griffiths, Kingston-upon-Thames, the United Kingdom (Chair)
Dr S. Hindawi, Blood Transfusion Services, Jeddah, Saudi Arabia
Mrs T. Jivapaisarnpong, Institute of Biological Products, Ministry of Public Health,
Nonthaburi, Thailand
Dr H. Klein, National Institutes of Health, Bethesda, MD, the USA (Vice-Chair)
Dr P. Minor, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr J. Petricciani, Palm Springs, CA, the USA (Rapporteur)
Mr V.R. Reddy, South African National Blood Service, Weltevreden Park, South Africa
Dr L.S. Slamet, Technical Adviser and Consultant to the National Agency of Drug and
Food Control, Jakarta Selatan, Indonesia
Dr P. Strengers, Sanquin Plasma Products, Amsterdam, Netherlands
Dr Y. Sohn, Ministry of Food and Drug Safety, Chungcheongbuk-do, Republic of Korea
Dr J. Wang, National Institutes for Food and Drug Control, Beijing, China
Dr K. Zoon, National Institutes of Health, Bethesda, MD, the USA
The decisions of the Committee were taken in closed session with only members of the Committee
1
present. Each Committee member had completed a Declaration of Interests form prior to the meeting.
These were assessed by the WHO Secretariat and no declared interests were considered to be in conflict
with full meeting participation.
vii
WHO Expert Committee on Biological Standardization Sixty-sixth report
2
A maximum of two representatives of the DCVMN and two representatives of the IFPMA were present in
the meeting room during discussion of any one agenda item.
3
Participated via teleconference.
viii
WHO Expert Committee on Biological Standardization
Temporary Advisers
Dr A. Chawla, Consultant, Greater Noida, India
Dr C. Conrad, Paul-Ehrlich-Institut, Langen, Germany
Dr P. Krause,4 Center for Biologics Evaluation and Research, Food and Drug Administration,
Bethesda, MD, the USA
Dr M. Lennon, Consultant, Horning, the United Kingdom
Dr V. Maqueda, Biologist, Buenos Aires, Argentina
Dr C. Morris, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom (Rapporteur for the blood products and in vitro diagnostics track)
Dr M. Powell, Medicines and Healthcare Products Regulatory Agency, London, the United
Kingdom
Dr J. Reinhardt, Paul-Ehrlich-Institut, Langen, Germany (Rapporteur for the blood products
and in vitro diagnostics track)
Dr D. Smith, Bacterial and Combination Vaccines Division, Health Canada, Ottawa, Canada
Dr J. Southern, Adviser to the Medicines Control Council of South Africa, Cape Town,
South Africa
Dr Y. Sun, Paul-Ehrlich-Institut, Langen, Germany
Dr E.R. Unger, Centers for Disease Control and Prevention, Atlanta, GA, the USA
Dr A.L. Waddell, Freelance, Stanley, the United Kingdom
Dr T. Wu, Bacterial and Combination Vaccines Division, Health Canada, Ottawa, Canada
Participants
Dr F. Agbanyo, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa,
Canada (also BRN representative)
ix
WHO Expert Committee on Biological Standardization Sixty-sixth report
Dr N. Almond, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr P. Aprea, National Administration of Drugs, Food and Medical Technology, Buenos
Aires, Argentina
Dr S. Baylis,5 Paul-Ehrlich-Institut, Langen, Germany
Dr A. Bristow, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr P. Bowyer,5 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr J. Boyle,5 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr D. Calam, Pewsey, the United Kingdom (International Nonproprietary Names (INN)
expert)
Dr F. Cano, Agence Nationale de Sécurité du Médicament et des Produits du Santé,
Lyons, France
Mr F. Carazzai Reisdorfer, Agência Nacional de Vigilância Sanitaria, Brasilia, Brazil
Dr H. Chan,5 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr K. Chumakov,5 Center for Biologics Evaluation and Research, Food and Drug
Administration, Silver Spring, MD, the USA
Dr H. Chung, Ministry of Food and Drug Safety, Chungcheongbuk-do, Republic of Korea
Dr L. Coombes,5 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr R. Dominguez Morales, Ministerio de Salud Pública, Havana, Cuba
Professor S. Efstahiou, National Institute for Biological Standards and Control, Potters Bar,
WHO Technical Report Series, No. 999, 2016
x
WHO Expert Committee on Biological Standardization
Dr J. Ferguson,6 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr D.A. Garcia Londono, Assesor del Despacho del Viceministro de Salud, Publica y
Prestación de Servicios, Bogota, Colombia
Professor A. Genazzani, Università del Piemonte Orientale, Novara, Italy (INN expert)
Dr K. Grant, Therapeutic Goods Administration, Woden, ACT, Australia (INN expert)
Dr S. Govind,6 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr E. Gray,6 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr M. Gruber,6 Center for Biologics Evaluation and Research, Food and Drug
Administration, Silver Spring, MD, the USA
Dr I. Hamaguchi, National Institute of Infectious Diseases, Tokyo, Japan (also BRN
representative)
Dr K. Haslov, Statens Serum Institute, Copenhagen, Denmark
Dr C.C. Ilonze, National Agency for Food and Drug Administration and Control, Lagos,
Nigeria
Dr S. Inglis, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Mrs W. Jariyapan, Ministry of Public Health, Nonthaburi, Thailand
Dr H. Jia,6 National Institute for Biological Standards and Control, Potters Bar, the United
Kingdom
Ms Juliati, National Agency of Drug and Food Control, Jakarta Pusat, Indonesia
Dr A. Kato, National Institute of Infectious Diseases, Tokyo, Japan
Dr D. Khokal, Health Sciences Authority, Helios, Singapore
Dr S. Kumar,6 Center for Biologics Evaluation and Research, Food and Drug Administration,
Bethesda, MD, the USA
Dr J. Luo, Center for Drug Evaluation, Beijing, China
Dr J. Martin,6 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr F. Mawas,6 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
xi
WHO Expert Committee on Biological Standardization Sixty-sixth report
Dr E. Mee,7 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr M. Moore,7 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Ms N.A.M. Nur, National Pharmaceutical Control Bureau, Selangor, Malaysia
Dr M. Ochiai, National Institute of Infectious Diseases, Tokyo, Japan
Dr H. Oh, Ministry of Food and Drug Safety, Chungcheongbuk-do, Republic of Korea
Dr M. Ovanesov,7 Center for Biologics Evaluation and Research, Food and Drug
Administration, Bethesda, MD, the USA
Ms O. Panova, Department of Medicines Supply and Regulation of Medical Products,
Ministry of Health, Moscow, Russian Federation
Dr E. Parra, Federal Commission for the Protection against Sanitary Risks, Ministry of
Health, Mexico City, Mexico
Dr M. Pfleiderer, Paul-Ehrlich-Institut, Langen, Germany
Dr S. Rijpkema,7 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr C. Schaerer, Swiss Agency for Therapeutic Products, Bern, Switzerland (also BRN
representative)
Professor R. Seitz, Paul-Ehrlich-Institut, Langen, Germany (also BRN representative)
Dr J.S. Shin, Ministry of Food and Drug Safety, Chungcheongbuk-do, Republic of Korea
Dr G. Smith, Therapeutic Goods Administration, Woden, ACT, Australia (also BRN
representative)
Dr K. Sohn, Ministry of Food and Drug Safety, Chungcheongbuk-do, Republic of Korea
Dr E. Spindler-Raffel,7 Paul-Ehrlich-Institut, Langen, Germany
WHO Technical Report Series, No. 999, 2016
xii
WHO Expert Committee on Biological Standardization
Dr A.M.H.P. Van Den Besselaar,8 Leiden University Medical Centre, Leiden, Netherlands
Dr A. Vasheghani Farahani, Food and Drug Organization, Tehran, the Islamic Republic
of Iran
Dr C. Vipond,8 National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr M. Wadhwa, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr J. Weir, Center for Biologics Evaluation and Research, Food and Drug Administration,
Silver Spring, MD, the USA
Dr D. Wilkinson,8 National Institute for Biological Standards and Control, Potters Bar,
the United Kingdom
Dr M. Xu, National Institutes for Food and Drug Control, Beijing, China
WHO Secretariat
Regional Offices
AMRO – Ms M.L. Pombo-Castro
EMRO – Dr H. Langar
SEARO – Mr M. Eisenhawer
WPRO – Dr J. Shin; Dr U. Kim
Headquarters
Mr K. de Joncheere, Director, EMP
Dr J-M Okwo-Bele, Director, IVB and Acting Head, IVR/IVB
Dr L. Rägo, Head, RHT/EMP
Dr D.J. Wood, Coordinator, TSN/EMP (Committee Secretary)
Mr L. Belgharbi, RSS/EMP
Dr J. Fournier-Caruana, PQT/EMP
Dr K. Gao, TSN/EMP
Dr H-N Kang TSN/EMP
Dr I. Knezevic TSN/EMP (Lead for the vaccines and biotherapeutics track)
Dr D. Lei TSN/EMP
xiii
WHO Expert Committee on Biological Standardization Sixty-sixth report
Dr M. Nuebling, TSN/EMP (Lead for the blood products and in vitro diagnostics track)
Ms I. Prat, PQT/EMP
Dr M. Refaat, RSS/EMP
Dr C.A. Rodriguez-Hernandez, PQT/EMP
Dr H. Schmidt, TSN/EMP
Dr W. Zhang, PED/GIP
Dr T. Zhou, TSN/EMP
WHO Technical Report Series, No. 999, 2016
xiv
Abbreviations
AEFI adverse event following immunization
Ag/Ab antigen/antibody
ALIFAR Asociación Latinoamericana de Industrias Farmacéuticas
ATMP advanced therapy medicinal product
BCG bacille Calmette–Guérin
BGTD Biologics and Genetic Therapies Directorate
BQ Biological Qualifier (scheme)
BRN WHO Blood Regulators Network
BSP EDQM biological standardization programme
CBER Center for Biologics Evaluation and Research
CIN cervical intraepithelial neoplasia
CIN2–3 cervical intraepithelial neoplasia grades 2 or 3
CIN2+ cervical intraepithelial neoplasia grade 2 or worse
cLIA competitive Luminex immunoassay
CMI cell-mediated immunity
CTC controlled temperature chain
DCVMN Developing Countries Vaccine Manufacturers Network
DCVRN Developing Country Vaccine Regulators’ Network
DNA deoxyribonucleic acid
ECTC extended controlled temperature conditions
EDQM European Directorate for the Quality of Medicines & HealthCare
EGA European Generic Medicines Association
EIA enzyme immunoassay
ELISA enzyme-linked immunosorbent assay
EMA European Medicines Agency
EMP WHO Department of Essential Medicines and Health Products
EPO erythropoietin
EU ELISA Unit(s)
xv
WHO Expert Committee on Biological Standardization Sixty-sixth report
FIX factor IX
GAPIII WHO Global Action Plan to minimize poliovirus facility-
associated risk after type-specific eradication of wild
polioviruses and sequential cessation of oral polio vaccine use
GCP good clinical practice
GCV geometric coefficient of variation
GMC geometric mean concentration
GMP good manufacturing practice(s)
GMT geometric mean titre
GP glycoprotein
HAV hepatitis B virus
HBsAg hepatitis B surface antigen
HBV hepatitis B virus
HCV hepatitis C virus
HEPA high-efficiency particulate air
HEV hepatitis E virus
HIV human immunodeficiency virus
holoTC holotranscobalamin
HPV human papillomavirus
HVAC heating, ventilation and air conditioning
IARC WHO International Agency for Research on Cancer
WHO Technical Report Series, No. 999, 2016
xix
1. Introduction
The WHO Expert Committee on Biological Standardization met in Geneva
from 12 to 16 October 2015. The meeting was opened by Mr Kees De Joncheere,
Director of the Department of Essential Medicines and Health Products (EMP).
Mr de Joncheere welcomed the Committee, meeting participants
and observers, and reminded the Committee that it had a mandate to review
developments in the field of biological substances used in human medicine that
included antibiotics, biotherapeutics, blood products, in vitro diagnostic device
reagents and vaccines. During its previous 65 meetings the Committee had
established approximately 70 written standards and around 300 international
biological reference preparations essential for the quality control, regulation and
clinical dosing of biological products.
The development and establishment of such standards, and their
subsequent promotion, are crucially important activities. The inclusive standards
development process implemented by WHO facilitates global consensus on
technical matters and is a very significant factor in promoting convergent
regulatory decision-making between countries. After standards have been
established, proactive technical support from WHO was crucial in obtaining
maximum understanding and impact, and facilitating the consistent application
of standards.
Equitable access to safe, quality, affordable and effective medical products
is one of the cornerstones of universal health care. The development and
adoption of norms and standards to regulate the quality, safety, efficacy and cost-
effective use of medical products is a vital foundation upon which this aspiration
is built. The coordinated efforts of WHO Expert Committees, together with
WHO collaborating centres (WHOCCs) and partner organizations, allow for a
global approach to this core normative work.
Increased levels of international collaboration are now required in the
field of regulatory science. As demands grow for reduced regulatory and policy
burdens, expectations of greater transparency will increase. New norms and
standards will need to reflect these aspirations but not to the point where quality
suffers. As the standards-setting expertise and resources needed to address all
needs are likely to be insufficient in any one Member State, the convergence of
international norms and standards was increasingly being recognized as a key
driver of success. Where appropriate, the use of WHO norms and standards as
global benchmarks represents an opportunity to achieve the desired regulatory
and policy convergence.
The standardization of biological products remains high on the agenda
of Member States and there was now broad recognition of the record of success of
biotherapeutic products in treating many life-threatening and chronic diseases.
However, some of these products were currently very expensive and access
1
WHO Expert Committee on Biological Standardization Sixty-sixth report
(consulting and investments); Dr K. Zoon (intellectual property and research support); Dr A. Chawla
(travel support); Professor S. Efstahiou (research support); Dr K. Haslov (consulting); Dr S. Inglis (public
statements); and Dr S. Thorpe (consulting).
3
2. General
2.1 Current directions
2.1.1 Strategic directions in biological standardization: WHO priorities
Dr Wood informed the meeting that in order to facilitate coordination of
overlapping topics and harness synergies, the Expert Committee on Biological
Standardization, the Expert Committee on Specifications for Pharmaceutical
Preparations and the International Nonproprietary Names (INN) Expert
Group were meeting simultaneously. Dr Wood then outlined the three major
categories of outcomes in the area of biological standardization, namely global
written standards, global measurement standards and the provision of support
to regulatory science including through the development of new assays, further
development and refinement of quality control tests, and reviewing of the
scientific basis for the establishment of specifications. In addition, standards-
implementation workshops continue to be conducted and provide extremely
valuable support to participants.
The three global public health priorities of responding to public health
emergencies, promoting access to biotherapeutic products and strengthening
regulatory systems remain key drivers of strategic approaches in this area. In
relation to public health emergencies, an overview from the WHO perspective
was presented on the research and development response to the Ebola outbreak.
The initial challenge had been to compress the usual timeline for unproven
interventions from years to months by working in parallel on product
development, testing, licensure and use. From August 2014, WHO held a series
of consultations with key international experts and stakeholders to identify
potential therapeutic or preventive solutions for Ebola. Based on expert advice,
WHO prioritized a number of products for further investigation through human
testing, namely two candidate vaccines, a shortlist of biotherapeutic products
WHO Technical Report Series, No. 999, 2016
made and high level of commitment in this area further activities were now
planned. A brief overview was also presented of blood regulation in Kenya
and of the collaborative efforts now under way between WHO and the Kenyan
National Blood Transfusion Service to improve the safety of the national blood
supply. Following up on recent efforts in the WHO African Region, a workshop
on the development of a regional strategy for blood safety and the establishment
of national regulatory systems for blood and blood products had been held
in Benin in September 2015 involving regulators and blood-establishment
representatives from 13 countries.
Dr Nuebling then drew attention to the pressing need for the assessment
and evaluation of snake antivenoms. With more than 100 000 fatalities
occurring as a result of snake bites each year, and the largely unregulated
nature of potentially therapeutic products of unknown quality, this continued
to be a highly neglected public health issue. As part of its efforts to address
this situation WHO was extending its prequalification programme to cover
antivenoms. Work was now under way to establish a prioritized prequalification
scheme that would be guided by the deliberations of an Expert Review Panel.
The prequalification of IVDs has been an important WHO activity
for many years as part of facilitating access to safe, appropriate and affordable
IVDs of good quality. Through its standards-setting and guidance-development
activities the Committee was requested to further support WHO prequalification
efforts in this area.
In light of resolution WHA67.20 on regulatory system strengthening and
the paramount requirement for all medical products to be safe and effective, the
development of a model regulatory framework for medical devices (including
IVDs) was also highlighted as a WHO EMP priority. Against a backdrop of a
lack of such regulation in many WHO Member States there was a need for an
established process for the approval of medical devices. The development of a
first draft of the model regulatory framework was scheduled for completion in
WHO Technical Report Series, No. 999, 2016
early 2016. This would then be followed by a round of public consultation prior
to submission of the outcome document for consideration for adoption by the
Expert Committees on Biological Standardization and on Specifications for
Pharmaceutical Preparations in October 2016.
Dr Nuebling then briefly highlighted the main outcomes areas of the
fifth meeting of WHOCCs on the development of WHO biological reference
preparations for in vitro diagnostic devices, which are reported upon more fully
in section 2.2.2 below. Dr Nuebling concluded by summarizing the programme
of measurement standards in the area of blood products and related in vitro
diagnostics proposed for establishment by the Committee in 2016, along with
the new projects to be endorsed.
There was some discussion around the issue of available resources and
expertise to support all the new work areas, particularly the prequalification of
6
General
IVDs. In response, it was noted that although the projected activities in relation
to the prequalification of IVDs were indeed considerable these were longer term
goals with the level of demands likely to build gradually over the coming years,
thus allowing for strengthened collaboration and integration of activities with
other processes.
2
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; www.who.int/biologicals/publications/trs/areas/vaccines/clinical_
evaluation/035-101.pdf?ua=1, accessed 9 February 2016).
3
Guidelines on evaluation of similar biotherapeutic products (SBPs). In: WHO Expert Committee
on Biological Standardization: sixtieth report. Geneva: World Health Organization; 2013: Annex 2
(WHO Technical Report Series, No. 977; http://www.who.int/biologicals/publications/trs/areas/biological_
therapeutics/TRS_977_Annex_2.pdf?ua=1, accessed 9 February 2016).
7
WHO Expert Committee on Biological Standardization Sixty-sixth report
Guidelines on the quality, safety and efficacy of biotherapeutic protein products prepared by
4
recombinant DNA technology. In: WHO Expert Committee on Biological Standardization: sixty-fourth
report. Geneva: World Health Organization; 2014: Annex 4 (WHO Technical Report Series, No. 987; http://
www.who.int/biologicals/biotherapeutics/TRS_987_Annex4.pdf?ua=1, accessed 9 February 2016).
8
General
achieving the goals now required. It was suggested that in view of the recognized
capabilities of other organizations that the WHO implementation workload
might be reduced through improved collaboration and streamlining of efforts.
2.2 Reports
2.2.1 Report from the WHO Blood Regulators Network
Dr Christian Schaerer reminded the meeting that the objectives of the BRN
were: (a) to identify issues and share expertise and information; (b) to promote
the science-based convergence of regulatory policy, including by fostering the
development of international consensus on regulatory approaches; and (c) to
propose solutions to specific issues, especially emerging public health challenges
such as the vulnerability of countries to communicable disease threats. As
of October 2015 the BRN membership comprised representatives of seven
regulatory agencies from different countries and WHO.
Following an update on membership application decisions and other
organizational issues Dr Schaerer summarized the range of recent BRN
activities. Three teleconferences had taken place in relation to the Ebola
outbreak, with further BRN participation in regular discussions on the situation
and on activities to strengthen blood systems in affected countries. An expert
presentation was given at the Alliance of Blood Operators, and discussions held
on their project on risk-based decision-making for blood safety. Discussions
were also held on national decision-making in relation to donor deferral for men
who have sex with men, and on the potential hazards of transfusion in terms
of the increased risks associated with blood transfusions and the postulated late
sequelae of transfusion.
In late 2014 the BRN proposed that an amendment be made to its
position paper on Collection and use of convalescent plasma or serum as an
element in filovirus outbreak response following its posting on the WHO website
to include a statement on the use of non-immune plasma as a control in studies
with convalescent plasma. In addition, a BRN Letter of support was produced
for a request by the World Federation of Hemophilia to add desmopressin to
the WHO Model List of Essential Medicines. This request was subsequently
accepted at the 2015 meeting of the WHO Expert Committee on the Selection
and Use of Essential Medicines.
Following discussions and recommendations made to WHO at the
16th International Conference of Drug Regulatory Authorities (ICDRA) a draft
guidance document was developed on the implementation of national blood
regulatory systems. BRN representatives also facilitated both the WHO workshop
on the development of a regional strategy for blood safety and the WHO
workshop on the assessment of GMP in a blood establishment in Surabaya,
Indonesia as a part of the WHO Achilles project (see section 2.1.2 above).
9
WHO Expert Committee on Biological Standardization Sixty-sixth report
the two organizations which had strengthened the position and activities of
NIBSC. After detailing a number of completed projects and new proposals for
consideration by the Committee in 2015, Dr Inglis briefly outlined the response
of NIBSC to the Ebola outbreak, and acknowledged the contribution of all
those who had been involved in developing materials for establishment at this
meeting in a very short time frame.
Among a number of current broader key issues Dr Inglis pointed out
that the expanding field of standardization activities inevitably raised questions
concerning the suitability of the current process in flexibly responding to and
meeting an increasingly complex range of demands. This raised the issue of
whether the current format and associated processes would be sufficiently
responsive and sustainable, particularly in the face of resourcing challenges.
Dr Inglis also drew attention to the increasing difficulty experienced in publishing
research findings in peer-reviewed journal in cases where data had already
been presented in the annual report of the Committee as part of the WHO
Technical Report Series. This was proving to be restrictive in both conveying
standardization messages to the relevant fields and in staff career progression
through publication in recognized journals. Dr Inglis then drew attention to a
number of upcoming events, which included a symposium on regulatory science
in 2016: shaping the future of biological medicines which was being held to mark
the 40th anniversary of NIBSC.
Dr Inglis concluded his presentation by announcing that he would be
retiring from the position of Director at NIBSC in April 2016 and introduced
his successor Dr Christian Schneider.
The Committee joined in thanking Dr Inglis for all his work over the
past 14 years and discussed a number of the key issues raised in his presentation.
In relation to the specific issue of publication there was some discussion around
the use of the journal Biologicals as this peer-reviewed journal was willing to
WHO Technical Report Series, No. 999, 2016
another proposed for establishment by the Committee this year. As with other
international standards, ISAs require distribution, replacement as needed
and international collaborative studies. Of the current 23 ISAs eight relate to
antibiotics that are on the WHO Model List of Essential Medicines. Dr Buchheit
reaffirmed the willingness of EDQM to continue in its role as the custodian
centre for ISAs.
BSP goals included the establishment of European Pharmacopoeia
biological reference preparations, the standardization of test methods for
the quality control of biological substances, the elaboration of alternative
methods in support of the 3Rs concept (Replacement, Reduction, Refinement)
to minimize the use of animals in research and the provision of support to
international harmonization efforts, including through collaboration with WHO
and non-European partners. BSP achievements to date included the initiation
or conclusion of 102 projects on reference standards and 41 projects on method
development (including 21 projects on 3R methods).
Dr Buchheit reiterated that the development of alternatives to animal
experiments remained a major commitment of EDQM in line with European
Union directives, and WHO was once again strongly urged to consider
the incorporation of the 3R initiative into its written standards and other
guidance, where appropriate. The inclusion of 3R methods in WHO guidance
was viewed by EDQM as being of paramount importance in promoting their
global acceptance. Dr Buchheit also requested that the Committee evaluate
the possibility of its more proactive and earlier involvement in the validation
of 3R alternatives, such as a replacement for the histamine sensitization test
for pertussis vaccines. EDQM projects of potential interest to the Committee
included the development and evaluation of alternative in vitro tests for both
pertussis toxin and pertussis vaccine.
Dr Buchheit also informed the Committee that one of the main outcomes
of a recent International Alliance for Biological Standardization conference on
3R alternatives was the decision to formally request WHO to initiate steps to
delete the abnormal toxicity test from all WHO Recommendations, Guidelines
and other guidance documents. Dr Buchheit went on to suggest that the
Committee might begin to consider the implications for IUs for vaccines when
in vivo tests are replaced with in vitro tests, and the technical ability to perform
the in vivo test has been lost.
Dr Buchheit concluded by highlighting a number of key harmonization
and other implementation issues for regional standard-setting bodies when no
international standard or other WHO guidance was available. The potential
use of international standards and reagents in the veterinary field was also
highlighted as a specific issue requiring clarification of the most appropriate
lead organization.
17
WHO Expert Committee on Biological Standardization Sixty-sixth report
5 Assessment criteria for national blood regulatory systems. In: WHO Expert Committee on Biological
Standardization: sixty-second report. Geneva: World Health Organization; 2013: Annex 7 (WHO Technical
Report Series, No. 979; http://www.who.int/biologicals/expert_committee/TRS_979_62nd_report.pdf?ua=1,
accessed 12 February 2016).
18
General
Center for Biologics Evaluation and Research (CBER), Silver Spring, MD, the USA
Dr Jay Epstein informed the Committee of a wide range of ongoing and
proposed activities in the further development of potency standards, reference
preparations, international standards, reference panels and reagents.
In response to the Ebola epidemic CBER had expedited the review of
investigational new drugs for Ebola vaccine studies and of an investigational
device exemption for pathogen-reduced plasma for patient care. Information
sharing was facilitated by a Confidentiality Arrangement with WHO, with
frequent information exchange and sharing of updates on Ebola-focused
activities taking place with WHO, EMA, Health Canada and others. CBER also
19
WHO Expert Committee on Biological Standardization Sixty-sixth report
NGS was another important new area in which CBER had been cooperatively
engaged, and materials should be available by early 2016. Dr Epstein then
outlined CBER participation in a number of international studies to develop and
evaluate vaccine-related standards for respiratory syncytial virus neutralization
assays, human cytomegalovirus immunoglobulin G (IgG), and Ebola antibody.
A number of completed and ongoing standards activities for plasma-derived
coagulation factors were also described, and current and proposed work on the
development of reference preparations for viruses and other pathogens outlined.
Dr Epstein concluded by highlighting a number of recent workshops
sponsored by CBER and noting the value of such workshops in the successful
implementation of regulatory policy, and the importance of regulatory science
research.
20
General
report plus initial testing results would be shared with interested NRAs to
facilitate national regulatory decision-making in relation to product registration,
variation or withdrawal. The utility of such a collaborative procedure had already
been demonstrated, including during the facilitated licensing of inactivated
poliomyelitis vaccine (IPV) in several countries.
Discussion was held on a broad range of issues, challenges and
opportunities in this area, including in the areas of national and regional
regulatory capacity-building, the role of regional implementation workshops
and associated initiatives, and the need for manufacturer engagement. The
Committee agreed that the revised document would be useful and looked
forward to being updated on its implementation at a future meeting.
22
General
23
WHO Expert Committee on Biological Standardization Sixty-sixth report
Table 1
Recommendations relating to the use of microbiological assays for antibiotics
5. kanamycin monosulfate
4 Monographs for which a concept paper should be developed on the possible
transition from microbiological to physicochemical methods
1. amphotericin B 6. kanamycin acid 11. gentamicin
2. amphotericin B for sulfate sulfate
injection 7. kanamycin for 12. streptomycin
3. bleomycin sulfate injection sulfate
4. erythromycin 8. kanamycin 13. streptomycin for
ethylsuccinate tablets monosulfate injection
5. erythromycin stearate 9. nystatin 14. paromomycin
tablets 10. nystatin tablets sulfate
24
General
Table 1 continued
5 Monographs that should be suppressed
1. bacitracin 4. chlortetracycline 7. oxytetracycline
2. bacitracin zinc hydrochloride dehydrate
3. bleomycin hydrochloride 5. erythromycin (base) 8. oxytetracycline
6. neomycin sulfate hydrochloride
Potential areas for improvement included: (a) reviewing the current WHO
biosafety requirements 6 for manufacturing facilities; (b) reviewing candidate
vaccine virus safety-testing standards and release specifications; (c) improving
the availability of reagents and assays; (d) ensuring clear and effective regulatory
pathways for pandemic vaccines (including for new vaccine platforms and
technologies); and (e) improved understanding of the roles and responsibilities
of stakeholders in terms of communication and interaction.
During discussions, further clarification was given of the vital
importance of promoting and expanding seasonal influenza vaccine use as part
of ensuring that sufficient production capacity would exist for pandemic vaccine
production. The Committee was also informed of the high degree of interest
in moving towards a new paradigm of influenza vaccine virus selection and
development that would allow for the more rapid production of well-matched
seasonal and pandemic influenza vaccines. The Committee expressed its thanks
for the update provided in this important area and asked to be kept informed of
further developments.
WHO biosafety risk assessment and guidelines for the production and quality control of human influenza
6
3.1 General
3.1.1 WHO good manufacturing practices for biological products
The Committee had been informed in 2014 of the history of WHO good
manufacturing practices (GMP) documents for pharmaceuticals, biological
products and blood establishments. The most recent WHO guidance on GMP
for biological products had been published in 1992 (Annex 2, WHO Technical
Report Series, No. 822). Following the initiation of a review process in 2007, a
consultation had been held in 2014 with the resulting draft revised Guidelines
then undergoing two rounds of public consultation in 2015. The text was
further amended by the guidelines drafting group taking into account the
comments received.
The outcome revised Guidelines document (WHO/BS/2015.2253) was
viewed as complementary to the general recommendations set out in the
current WHO good manufacturing practices for pharmaceutical products: main
principles 7 and in other WHO documents related specifically to the production
and control of biological products. As the revised document does not provide
detailed recommendations for specific classes of biological products, attention
WHO Technical Report Series, No. 999, 2016
was drawn in the text to the need to consult other relevant WHO documents,
in particular WHO recommendations to assure the quality, safety and efficacy of
specific products.
The proposed revision reflects recent scientific and technological
developments in the manufacture and control of biological products, and in
the application of risk-based approaches to GMP, while recognizing the wide
variability inherent in this evolving class of medicinal products.
WHO good manufacturing practices for pharmaceutical products: main principles. In: WHO Expert
7
Committee on Specifications for Pharmaceutical Preparations: forty-eighth report. Geneva: World Health
Organization; 2014: Annex 2 (WHO Technical Report Series, No. 986; http://www.who.int/medicines/
areas/quality_safety/quality_assurance/TRS986annex2.pdf?ua=1, accessed 14 February 2016).
28
International Recommendations, Guidelines and other matters
The Committee was then provided with a detailed overview of the precise
scope and principal themes covered by the revised Guidelines. The document does
not cover human whole blood, blood components and plasma-derived products
for therapeutic use as separate comprehensive WHO guidance is available and
should be followed.8,9 The characteristically wide variability typical of biological
products and corresponding manufacturing processes, are emphasized in the
revised document, along with specific aspects of the analytical methods and
approaches required. The importance of in-process controls are emphasized and
the quality risk management (QRM) approach recommended as an effective tool
for managing product variability, preventing quality deviations and strengthening
product and process knowledge.
The Committee reviewed the major comments received during the
second round of public consultation and requested a number of clarifications.
After making further changes, the Committee recommended that the revised
WHO Guidelines be adopted and annexed to its report (Annex 2).
8
WHO guidelines on good manufacturing practices for blood establishments. In: WHO Expert Committee
on Specifications for Pharmaceutical Preparations: forty-fifth report. Geneva: World 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).
9
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).
29
WHO Expert Committee on Biological Standardization Sixty-sixth report
Addendum to: Guidelines on the quality, safety and efficacy of biotherapeutic protein products prepared
10
by recombinant DNA technology. In: WHO Expert Committee on Biological Standardization: sixty-fourth
report. Geneva: World Health Organization; 2014: Annex 4 (WHO Technical Report Series, No. 987; http://
www.who.int/biologicals/biotherapeutics/TRS_987_Annex4.pdf?ua=1, accessed 15 February 2016).
30
International Recommendations, Guidelines and other matters
much more rapid response, and that lessons had been learned that would assist
in defining rapid-response requirements for potential future pathogen outbreaks.
The Committee observed that there had been some degree of criticism
following a perceived slow response from WHO. Nevertheless, it was also
apparent that the scale of work and progress made in a very short space of time
had been considerable.
in 2015. The Committee was provided with a brief overview of the status of
blood regulation in Kenya, the significant challenges still being faced, and the
initiation of collaborative efforts between WHO and the Kenyan National Blood
Transfusion Service to improve the safety of the national blood supply. Following
up on recent efforts in the WHO African Region, a workshop on the development
of a regional strategy for blood safety and the establishment of national regulatory
systems for blood and blood products had been held in Benin in 2015 involving
regulators and blood-establishment representatives from 13 countries, as well as
representatives from WHO, WHO BRN and PEI. This workshop had provided an
opportunity for the sharing of national experiences in blood regulatory activities,
and for the drafting of a regional blood safety strategy for the upcoming 10 years
along with guidelines on the establishing of a regulatory system for blood and
blood products.
32
International Recommendations, Guidelines and other matters
tool may be very useful, and that amendment of the WHO assessment criteria for
national blood regulatory systems would potentially be beneficial in principle, it
would also be important to preserve the details of the different product streams
and/or technologies addressed by individual assessments. It was also reiterated
that the assessments resulting from already existing tools would need to be
accepted after validation to avoid duplication of efforts. It was further pointed
out that the terminology used for the different categories of product needed to
Assessment criteria for national blood regulatory systems. In: WHO Expert Committee on Biological
11
Standardization: sixty-second report. Geneva: World Health Organization; 2013: Annex 7 (WHO
Technical Report Series, No. 979; http://www.who.int/biologicals/expert_committee/TRS_979_62nd_
report.pdf?ua=1, accessed 12 February 2016).
34
International Recommendations, Guidelines and other matters
After further consideration it was decided that that a final version of the
document should be presented to the Committee in 2016 prior to a decision
being made on its placement in the context of current guidance.
Guidelines to assure the quality, safety and efficacy of recombinant human papillomavirus virus-like
12
particle vaccines. In: WHO Expert Committee on Biological Standardization: fifty-seventh report. Geneva:
World Health Organization; 2011: Annex 1 (WHO Technical Report Series, No. 962; http://apps.who.int/
iris/bitstream/10665/44678/1/WHO_TRS_962_eng.pdf, accessed 22 December 2015).
37
WHO Expert Committee on Biological Standardization Sixty-sixth report
Guidelines on stability evaluation of vaccines. In: WHO Expert Committee on Biological Standardization:
13
fifty-seventh report. Geneva: World Health Organization; 2011: Annex 3 (WHO Technical Report
Series, No. 962; http://who.int/biologicals/vaccines/Annex_3_WHO_TRS_962-3.pdf?ua=1, accessed 11
December 2015).
39
WHO Expert Committee on Biological Standardization Sixty-sixth report
and – following careful risk–benefit assessment – to allow the use of vaccines for
which a full regulatory package may not yet be available.
Following an informal consultation in 2015 and subsequent survey
feedback received from 10 NRAs, WHO had now developed a review document
which aimed to provide an overview of various existing regulatory pathways
in selected countries and to encourage countries to review their state of
regulatory preparedness for public health emergencies. Experience of public
health emergencies since 2000 included the severe acute respiratory syndrome
outbreak in several countries, the H1N1 influenza pandemic (and ongoing risk
of an avian influenza pandemic) and the Ebola epidemic in West Africa. Each of
those events served to reinforce the crucial need for a rapid global response.
It was determined by WHO that a certain level of flexibility already
exists in some countries in relation to the evaluating of vaccines during public
health emergencies. However, there were also countries in which vaccine
licensing could only be done on the basis of a full data package with no
alternative pathway. In such cases, it would be important to initiate discussion at
national and regional (or intercountry) level to explore options for establishing
alternative regulatory pathways as part of overall emergency response efforts.
The most important considerations in evaluating vaccines during public
health emergencies included risk–benefit assessments, the need for a well-
defined and transparent decision-making process, risk-management planning
and pharmacovigilance. It had been further concluded that actual case studies
and lessons learnt from previous public health emergencies could serve as
training materials.
The Committee noted the draft review document and sought
clarifications in a number of areas. While it was agreed that a WHO guidance
document might be of particular value to some NRAs and public health
organizations, there was also recognition of the complexity of emergency
situations. The decision to approve the use of an investigational product in the
WHO Technical Report Series, No. 999, 2016
which had included related guidance regarding the quality, safety and efficacy
of Ebola vaccines.
Following expert review, the need for a specific Ebola vaccine document
was highlighted. In March 2015, WHO convened an informal consultation
attended by experts, regulatory professionals and other stakeholders involved
in Ebola vaccine development, production, and regulatory evaluation to review
the resulting draft WHO Guidelines and reach consensus on key technical and
regulatory issues.
The draft Guidelines were intended to provide guidance to NRAs and
vaccine manufacturers on the quality, nonclinical and clinical aspects of Ebola
vaccines, particularly those based on viral vectors as these are currently at the
most advanced stage of development.
As there are currently significant knowledge gaps in the scientific
understanding of Ebola disease and Ebola vaccines, and as no Ebola vaccine
has yet been licensed, the document had been developed as WHO Guidelines
rather than Recommendations. It was recognized that subsequent updating of
the Guidelines may be required in the light of recent and future developments.
The Committee noted the comments and suggestions that had been
received by WHO in relation to the draft Guidelines and indicated its agreement
with the proposed content, scope and proposed approach for its further revision.
The Committee also provided a number of additional recommendations,
including the addition of further guidance on post-marketing surveillance and
the need for fluid approaches to updating and posting the current text in this
rapidly evolving area. The Committee expressed its appreciation for the efforts
that had been made in developing such an excellent and comprehensive draft in
such a short period, and looked forward to considering the revised Guidelines
at its next meeting.
help facilities and oversight bodies better understand and implement GAPIII
requirements as they relate to the certification of poliovirus containment in
essential facilities.
The Committee was informed that current WHO Guidelines for the safe
production and quality control of IPV manufactured from wild polioviruses had
been published as an addendum to the previous WHO Recommendations for
the production and control of poliomyelitis vaccine (inactivated). In light of a
range of associated developments and updates, including in the area of GMP for
biological products, the revision of the current Guidelines had been proposed.
In order to inform the revision process and identify key issues to
be addressed a thorough expert technical review was conducted involving
WHOCCs working in this area. The need for revision of the current guidance
was broadly recognized in order to provide practical and detailed guidance to
poliomyelitis vaccine manufacturers and regulators not fully addressed by any
currently available WHO documents. Ideally the revision would extend the scope
of the current document to cover all poliovirus strains, and provide updated,
detailed, specific and practical guidance on all aspects of poliovirus containment
in vaccine-manufacturing facilities, with an emphasis on issues not addressed
elsewhere. The revision would also take into account the current and anticipated
future global polio eradication situation and be aligned with current resources
and global strategies such as PEESP and GAPIII. Further details and specific
issues will be identified and discussed during consultations with experts from
vaccine manufacturers and NRAs.
The Committee noted the information presented, sought clarifications
in a number of areas and made several specific suggestions. The Committee
agreed with the conclusions and proposals presented, expressed its support
for the development of the revised WHO Guidelines and looked forward to
reviewing progress in 2016.
WHO Technical Report Series, No. 999, 2016
43
4. International Reference Materials – antibiotics
All reference materials established at the meeting are listed in Annex 6.
44
5. International reference materials –
biotherapeutics other than blood products
All reference materials established at the meeting are listed in Annex 6.
was analysed by 18 laboratories from eight countries using their own in-house
assay. Results showed that antibody detection varied between laboratories and
was dependent on antibody characteristics and the method used. Within- and
between-assay repeatability was generally very good with excellent agreement
for most laboratories. Accelerated thermal degradation studies demonstrated
that panel samples stored at 37 °C for 9 months showed no significant loss of
activity. The Committee was informed that 700 vials would be available for use
as an international standard.
The Committee considered the report of the study (WHO/BS/2015.2265)
and recommended that the proposed panel be established as the First WHO
Reference Panel for antibodies to erythropoietin (human).
which allow the relationships between structure and function for various assays
to be explored.
The proposed panel would span a range of properties, principally pI
(a function of the degree of sialylation), to allow users to define the response of
in vitro and in vivo bioassay systems to variations in sialylation. Globally, there
are numerous EPO manufacturers, pharmacopoeias and control laboratories
who would seek access to such materials.
The Committee considered the document (WHO/BS/2015.2273)
outlining the establishment of a panel of bioassay reference reagents for EPO
but was not convinced of the utility of such a panel in the in vitro bioassay of
EPO and did not endorse the proposal.
not been shown to correlate with current in vivo test results. The Committee
recommended that an exploratory study be undertaken to compare unmodified
IFN-α with peg-IFN-α products, and that the results be reported to the
Committee in due course.
50
6. International reference materials – blood
products and related substances
All reference materials established at the meeting are listed in Annex 6.
defined and the decision was made not to proceed with further discussion at
that point. At the same time, a reference reagent for FXIa was established,
followed in 2013 by the establishment of an international standard for FXIa
to support its measurement in immunoglobulin products. In addition, global
stakeholders continued to work on the development and refinement of assays
for procoagulant activity.
The Committee was informed that the international standard for FXIa
is now helping to harmonize FXIa assay methods, and is also used by regulators
and manufacturers to develop other assay methods such as the Non-Activated
Partial Thromboplastin Time and Thrombin Generation Assay. Because of the
matrix heterogeneity of immunoglobulin products, it was clear that in addition
to the purified FXIa standard, matrix-related reference preparations will also
be required. Such matrix-related preparations may be superior to purified FXIa in
54
International reference materials – biotherapeutics other than blood products
56
7. International reference materials –
In vitro diagnostic device reagents
All reference materials established at the meeting are listed in Annex 6.
the candidate material at temperatures used for storage (−20 °C) and laboratory
manipulation (4–20 °C), as well as at the 37–45 °C ambient temperatures
typically encountered during global shipment. Further real-time stability studies
were now in progress to assess the long-term stability of the candidate material.
The Committee was informed that 4103 vials would be available for use as an
international standard.
Discussion was the held on the potential issues relating to the use of
a single subtype, the possibility of matrix effects and the associated issue of
commutability. The Committee was informed that although the commutability
aspects of the study had been limited, there was nothing to indicate that
proposed material would not be suitable, and the possibility of a wider
commutability study was being discussed with study participants and other
stakeholders. Discussion also centred on the assignment of a unit based on
quantitative data or on combined qualitative and quantitative data. Despite
an observed discrepancy between qualitative and quantitative findings, the
Committee concluded that clinical decision-making for JCV relies primarily on
determining the presence or absence of virus rather than its quantitation and
the use of a combined value would be appropriate.
The Committee considered the report of the study (WHO/BS/2015.2259)
and recommended that the candidate material 14/114 be established as the First
WHO International Standard for JC virus DNA for NAT-based assays with an
assigned potency of 7.0 log 10 IU/ml.
of ≥ 4 log 10 /ml for > 3 weeks is presumed predictive for BKVAN, upon which
a reduction in immunosuppression is recommended. A standardized reference
preparation is therefore needed for accurate and comparable viral load
determination. It is envisaged that such a reference preparation would be used
by clinical diagnostic laboratories and IVD manufacturers for the calibration of
secondary reference reagents and working standards.
An international collaborative study was conducted to evaluate
candidate materials for use in the standardization of BKV NAT-based assays.
Two candidate samples of freeze-dried whole BKV preparations (NIBSC
codes 14/202 and 14/212) formulated in a universal buffer were assessed by
33 laboratories from 15 countries, each using their routine NAT-based assays
for BKV detection. The freeze-dried candidates were tested alongside their
liquid equivalents, two patient samples and a BKV plasmid construct. For both
candidate materials a good degree of assay harmonization was already evident
amongst laboratories, and expression of the data as a relative potency for both
candidate samples suggested that either could serve as a suitable candidate for
an international standard. Of the two candidate samples, 14/212 was considered
to be the most suitable based on its geographical representation of the most
relevant circulating strain in Europe and the United States. Accelerated thermal
degradation stability studies performed at 3 months demonstrated that the
candidate material was stable, and exhibited no loss of titre at temperatures
used for storage (−20 °C), laboratory manipulation (4–20 °C), as well as at the
37–45 °C ambient temperature typically encountered during global shipment.
Further real-time stability studies were now in progress to assess the long-term
stability of the candidate material. The Committee was informed that 4092 vials
would be available for use as an international standard.
Discussion was held on the assignment of the unit with agreement
reached that in light of the minority representation of qualitative assays, the
under quantification typically seen in such assays and the primary requirement
to quantify BKV viral load for the management of renal transplant patients a
unit should be derived solely from the quantitative data obtained. Although
the choice of subtype for the candidate material was also approved, there was
also discussion of the potential value of creating a subtype panel that included
subtype 4 as this was also known to be clinically relevant. It was agreed that
NIBSC would investigate this need with stakeholders.
The Committee considered the report of the study (WHO/BS/2015.2270),
indicated that the Instructions for Use should clearly specify that assigned unitage
was based on quantitative NAT assays only, and recommended that the candidate
material 14/212 be established as the First WHO International Standard for BK
virus DNA for NAT-based assays with an assigned potency of 7.2 log 10 IU/ml.
59
WHO Expert Committee on Biological Standardization Sixty-sixth report
was also given of the genotype used to prepare the candidate material, and
confirmation provided that this had been shown, via sequencing, to be the
same genotype used in the previous international standards.
The Committee considered the report of the study (WHO/BS/2015.2262)
and recommended that the candidate material 14/150 be established as the
Fifth WHO International Standard for hepatitis C virus RNA for NAT-based
assays with an assigned value of 5.0 log 10 IU/vial when reconstituted in 1.1 ml
of nuclease-free water.
information only, along with information on the stabilizers used for the stool-
derived viruses.
not be harmonized with other laboratories and methods, thus impairing the
reproducibility of results between laboratories.
Recommendations emerging from a Technical Workshop on the
Standardization of Serological and PCR assays for the detection of Ebola virus
held at NIBSC in 2015 included the urgent development of interim Ebola
standards and, in the longer-term, of international standards in accordance with
published WHO guidelines for formal establishment the Committee.
In an international collaborative study involving 17 laboratories in
five countries nine Ebola virus antibody preparations were evaluated for their
suitability as an interim WHO standard for Ebola antibody assays. These nine
blinded-study samples included three preparations of purified anti-Ebola IgG
derived from transchromosomic bovines that had been pre-bled or vaccinated
with experimental vaccines, three solvent-detergent-treated plasma samples
obtained from three patients who had recovered from Ebola virus disease, one
solvent-detergent-treated Ebola-negative plasma and two non-treated plasma
pools obtained from volunteers vaccinated with the monovalent formulation
of the chimpanzee adenovirus 3-vectored vaccine. Results were obtained using
27 assays covering the three general methodological categories of neutralization
of live Ebola virus, neutralization of Ebola pseudotypes or virus-like particles
and enzyme immunoassay.
The Committee recognized the urgent need for such a material and
congratulated the groups involved on their greatly expedited efforts. Discussions
were held on the reasons for a lack of detectable antibodies in vaccinee material
and on the sufficiency of the available data. As the studies were not sufficiently
complete to establish any of the candidate materials as an international standard,
it was decided that the material should be described as a reference reagent with
this designation reflected in the unit terminology used.
The Committee considered the original proposal for the study (WHO/
WHO Technical Report Series, No. 999, 2016
7.1.10 First WHO reference reagents for Ebola virus RNA for NAT-based assays
The calibration and performance assessment of Ebola virus RNA NAT-based
assays requires reference materials that can be used in parallel with clinical
samples to standardize or control for every step of the assay, from extraction
through to detection and quantification.
An international collaborative study was conducted involving 13
laboratories from seven countries using a range of in-house and commercially
66
International reference materials – In vitro diagnostic device reagents
reagent and the expected remit of the Committee in its establishment it was
clarified that although the relevant data would be available to the Committee
there would be no requirement for its formal establishment.
Following discussion and further consideration, the Committee endorsed
the proposal (WHO/BS/2015.2275) to develop a Fourth WHO International
Standard for hepatitis B virus DNA for NAT-based assays.
of different subtypes it was felt that its outcome may ultimately determine the
correct subtype to be established. In the absence of the immediate resolution
of this issue the Committee requested that full clarification of this point be
provided at its next meeting. Following discussion and further consideration,
the Committee endorsed the proposal (WHO/BS/2015.2275) to develop a First
WHO International Standard for Trypanosoma cruzi DNA for NAT-based assays.
some regions is the most common cause of malaria. Even in low concentrations
P. vivax is capable of causing severe infection and even death, mostly due to
splenomegaly. Addressing P. vivax infection is a recognized priority of the WHO
Malaria Programme, and it is envisaged that the development of a standard in
this area will benefit clinicians, commercial kit manufacturers and researchers.
It was proposed that an international collaborative study be conducted
to evaluate candidate material for use as a First WHO International Standard
for Plasmodium vivax DNA for NAT-based assays. In addition to the candidate
material, the study would also assess clinical samples for commutability purposes.
The Committee recognized the need for this standard while noting that
the study proposal contained only limited information on a number of key
aspects. The Committee suggested that the study should include laboratories
in countries where P. vivax was endemic, and requested that additional study
details be provided at its next meeting. Following discussion and further
consideration, the Committee endorsed the proposal (WHO/BS/2015.2275) to
develop a First WHO International Standard for Plasmodium vivax DNA for
NAT-based assays.
7.2.9 Proposed First WHO Reference Panel for hepatitis E virus antibodies
Diagnosis of infection with hepatitis E virus (HEV) requires a variety of tests
including the detection of IgM and (rising) IgG antibodies. The analysis of
IgM antibodies in particular is useful for the confirmation of acute infection.
Anti‑HEV IgG may also be detectable during acute infection. Anti-HEV IgG is
also a marker of past HEV infection and is thus important in seroprevalence
studies of previous HEV exposure in different populations.
73
WHO Expert Committee on Biological Standardization Sixty-sixth report
7.2.10 Proposed First WHO Reference Panel for HIV-1 p24 antigen
The early detection and monitoring of infection with HIV is a key element in
treatment and prevention activities. Although the use of molecular methods
allows for early viral detection their cost is often prohibitive in low-income
countries. In addition, the genetic diversity of HIV presents a challenge to all
assay development with the accurate detection of HIV across all subtypes known
to be problematic.
An HIV-1 p24 antigen reference reagent (NIBSC code 90/636) was first
established by WHO in 1992 using subtype B patient-derived material that had
been inactivated using a solvent detergent treatment. Further development in
WHO Technical Report Series, No. 999, 2016
7.2.11 Proposed First WHO Reference Panel for Babesia microti antibodies
Babesiosis is a tick-borne zoonosis caused by infection with intra-erythrocytic
protozoans of several species of the genus Babesia. Although several Babesia
species are transmitted in the United States, B. microti is the most prevalent.
Endemic B. microti transmission has also been reported in Canada, parts of
Europe and Japan (B. microti-like). The prevalence of such infections in the
United States and other countries is poorly understood with the full extent of
global transmission believed to be underestimated.
In 2014, a total of 1759 babesiosis cases in 21 states were reported to the
national surveillance programme in the United States highlighting the risk of
infection outside the nine known endemic states. In addition, areas of endemic
transmission are reported to be expanding, particularly in states adjoining the
endemic states. Although there is no FDA-licensed test for diagnostic or donor-
screening purposes, some donor testing using investigational tests has been
conducted in the United States.
It was proposed that CBER develop a First WHO Reference Reagent
panel for Babesia microti antibodies. Panel members would be formulated from
defibrinated plasma samples taken from humans who had a current or recent
B. microti infection. In addition, RBCs from DBA/2 mice infected with B. microti
would be used as a source of antigen in immunofluorescence assays. The
reference panel would consist of four serial dilutions of the pooled B. microti
antibody positive samples and one pooled B. microti antibody negative sample.
75
WHO Expert Committee on Biological Standardization Sixty-sixth report
The need for this panel was acknowledged by the Committee with
discussions held in relation to the degree of assay cross-reactivity between
B. microti and other species of Babesia. Although not specifically tested for
in the material proposed for this panel, reports in the literature suggested
that some degree of cross-reactivity existed between species. Following
discussion and further consideration, the Committee endorsed the proposal
(WHO/BS/2015.2275) to develop a First WHO Reference Panel for Babesia
microti antibodies.
76
8. International reference materials –
vaccines and related substances
All reference materials established at the meeting are listed in Annex 6.
material 13/238 was established as the First WHO Reference Reagent for EV71
neutralization assays with an assigned value of 300 IU/ampoule.
for measuring Lf. Comparable results to the original Ramon flocculation test
were obtained when using a laser light-scattering platelet aggregometer as
the detection system for flocculation. Similarly, Lf results returned by ELISA
methods were not significantly different to flocculation, suggesting that ELISA
may be used as a suitable alternative to the flocculation test for measuring the
Lf of diphtheria toxoid samples, subject to validation.
Real-time stability studies were ongoing and had so far demonstrated
that the candidate material (NIBSC code 13/212) was stable at the normal storage
temperature (−20 °C) and suitable for use up to 3 months after reconstitution
when stored at 4 °C. Accelerated thermal degradation study results suggested
that the material would also exhibit suitable long-term stability with a predicted
degradation rate of 0.018% per year when stored at −20 °C.
The Committee considered the report of the study (WHO/BS/2015.2254)
and recommended that Preparation A (NIBSC code 13/212; 4800 ampoules) be
established as the Third WHO International Standard for diphtheria toxoid for
use in flocculation test with an assigned value of 1870 Lf/ampoule.
and multidrug resistance hampers effective treatment. Field studies have shown
that increased serum levels of anti-O IgG in young children reduce the chances
of reinfection with S. enterica serovar Typhimurium. Anti-O IgGs are also
bactericidal in vitro.
Vaccine manufacturers in developing countries have started to produce
bivalent vaccines containing O4,5 and O9 antigens representing these serovars.
As several vaccines targeting the O-antigen PS will be entering clinical
development, a common global standard is now required for the quantification
and testing of quality parameters of O4,5 and O9 PS used in bioassays to measure
the potency of these vaccines and to compare the immunogenicity of various
vaccines. At present, three types of bivalent vaccines (conjugate, attenuated oral
and outer membrane) are in development, and it was therefore proposed that
an international collaborative study be undertaken to establish a First WHO
82
International reference materials – In vitro diagnostic device reagents
86
Annex 1
WHO Recommendations, Guidelines and other documents
related to the manufacture and quality control of
biological substances used in medicine
WHO Recommendations, Guidelines and other documents are intended to
provide guidance to those responsible for the production of biological substances
as well as to others who may have to decide upon appropriate methods of
assay and control to ensure that products are safe, reliable and potent. WHO
Recommendations (previously called Requirements) and Guidelines are scientific
and advisory in nature but may be adopted by an NRA as national requirements
or used as the basis of such requirements.
Recommendations concerned with biological substances used in
medicine are formulated by international groups of experts and are published
in the WHO Technical Report Series 1 as listed below. A historical list of
Requirements and other sets of Recommendations is available on request from
the World Health Organization, 20 avenue Appia, 1211 Geneva 27, Switzerland.
Reports of the WHO Expert Committee on Biological Standardization
published in the WHO Technical Report Series can be purchased from:
WHO Press
World Health Organization
20 avenue Appia
1211 Geneva 27
Switzerland
Telephone: + 41 22 791 3246
Fax: +41 22 791 4857
Email: [email protected]
Website: www.who.int/bookorders
Individual Recommendations and Guidelines may be obtained free of
charge as offprints by writing to:
Technologies Standards and Norms
Department of Essential Medicines and Health Products
World Health Organization
20 avenue Appia
1211 Geneva 27
Switzerland
87
WHO Expert Committee on Biological Standardization Sixty-sixth report
91
Annex 2
WHO good manufacturing practices for biological products
Replacement of Annex 1 of WHO Technical Report Series, No. 822
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
Annex 3
Regulatory assessment of approved rDNA-derived
biotherapeutics
Addendum to Annex 4 of WHO Technical Report Series, No. 987
1. Introduction 134
2. Regulatory expectations for rDNA-derived biotherapeutics,
including similar biotherapeutic products 134
3. Review of products on the market 135
4. Points to consider in a stepwise regulatory assessment 137
5. Regulatory actions 139
6. Authors and acknowledgements 140
7. References 144
131
WHO Expert Committee on Biological Standardization Sixty-sixth report
132
Annex 3
Abbreviations
ALIFAR Asociación Latinoamericana de Industrias Farmacéuticas
DCVMN Developing Countries Vaccine Manufacturers Network
DNA deoxyribonucleic acid
EGA European Generic Medicines Association
ICDRA International Conference of Drug Regulatory Authorities
IFPMA International Federation of Pharmaceutical Manufacturers
& Associations
IGPA International Generic Pharmaceutical Alliance
NRA national regulatory authority
rDNA recombinant DNA
RBP reference biotherapeutic product
SBP similar biotherapeutic product
133
WHO Expert Committee on Biological Standardization Sixty-sixth report
1. Introduction
This WHO guidance document considers the regulatory assessment needed to
address situations where, for various reasons, biotherapeutic protein products
prepared by recombinant deoxyribonucleic acid (DNA) technology (rDNA-
derived biotherapeutics) were licensed with data packages that do not follow
current international regulatory standards for these biologicals. This includes,
for example, biotherapeutic products licensed via a generic pathway or with
limited analytical, nonclinical and/or clinical data (1, 2). At its 2010 meeting
in Singapore (3) the International Conference of Drug Regulatory Authorities
(ICDRA) discussed such situations and requested WHO assistance in developing
approaches for evaluating these already-licensed products in accordance with
current WHO guidelines. In May 2014 the Sixty-seventh World Health Assembly
adopted two relevant resolutions: one on promoting access to biotherapeutic
products and ensuring their quality, safety and efficacy (4) and the other on
regulatory systems strengthening (5) in which WHO was requested to provide
guidance, especially on dealing with increasingly complex biological products.
Although primarily addressing rDNA-derived biotherapeutic protein
products, some aspects of this document may also be relevant to other
biotherapeutics.
The timeline for completing the overall review exercise will depend
upon the time needed to generate and provide the missing information, taking
into consideration the product-specific points outlined below in section 4.
For example, in 2009 one NRA clarified the “appropriate regulatory pathway”
for dealing with changes in the regulatory oversight of low molecular weight
heparins 1 to reflect the fact that in future they would be regulated in that country
as biologicals and not as small-molecule pharmaceuticals (16). In addition,
it was announced that any biosimilar heparin submissions should follow the
regulatory framework for biosimilars and not the generic pathway. A transition
period of 12 months was set to allow manufacturers to update their files to reflect
the data required for biologicals. Manufacturers were also required to report on
WHO Technical Report Series No. 999, 2016
how much of their licensed product was sold in the country per year following
the official start date of the revised regulatory approach.
Similar transitional provisions have been made by other NRAs when
updating the regulations for biotherapeutics, including biosimilars (15, 17).
Low molecular weight heparin is not an rDNA-derived biotherapeutic product but is highlighted here
1
5. Regulatory actions
On the basis of the outcomes of the regulatory assessment, the NRA should
decide upon the appropriate actions to be taken. The decisions and actions of
NRAs may differ depending upon the assessments made according the points
listed above in section 4, which will be jurisdiction specific. In a stepwise
approach, product supply would not be compromised and authorization might
be regularized after the defined time period during which the product would
have undergone further regulatory evaluation, and on condition that it was
shown to have an acceptable risk–benefit profile.
Capacity-building will be needed where resources and expertise are
considered inadequate. Where the number and level of experience of staff
available to undertake an overall review are limited, consideration could be
given to mentoring or to work-sharing arrangement amongst NRAs. In the case
of mentoring, support could be provided through WHO from an experienced
authority that uses well-established processes that accord with relevant WHO
guidelines. In addition, the sharing of information between NRAs regarding
the basis for regulatory decisions on biotherapeutic products (including SBPs)
and the availability of publically available evaluation reports are considered
important sources of support for regulatory authorities that are less experienced
in dealing with these highly complex products, and may accelerate product
assessment. Communicating the details of what information was reviewed
and how it was incorporated into decision-making is also important for
prescribers, patients and other stakeholders, and can help promote confidence
in biotherapeutic products. The summary basis of decision documents produced
by some regulatory agencies, such as Health Canada, the European Medicines
Agency and the United States Food and Drug Administration, are examples of
informative documents.
139
WHO Expert Committee on Biological Standardization Sixty-sixth report
in Xiamen, China, 28–30 May 2012 and attended by: Mrs A. Abas, Ministry
of Health Malaysia, Malaysia; Dr W.S. Alhaqaish, Jordan Food and Drug
Administration, Jordan; Ms J. Archer (International Generic Pharmaceutical
Alliance (IGPA) representative), Hospira, Australia; Dr B. Boonyapiwat, Ministry
of Public Health, Thailand; Dr L. Gomes Castanheira, Agência Nacional de
Vigilância Sanitária, Brazil; Dr R. Chakrabarti (United States Pharmacopeial
Convention representative), United States Pharmacopeial Convention–India,
India; Dr W. Chang, State Food and Drug Administration, China; Mr D. Cheng,
Beijing Four Rings Bio-Pharmaceutical Co., Ltd, China; Dr Y. Choi, Korea Food
and Drug Administration, Republic of Korea; Ms J. Dahlan, National Agency of
Drug and Food Control, Indonesia; Mr G. Eich (IFPMA representative), Amgen
Inc. Corporate Services/Global Regulatory Affairs & Safety, the USA; Dr K. Gao,
National Institutes for Food and Drug Control, China; Mr T. Go, Health Sciences
140
Annex 3
7. References
1. Praditpornsilpa K, Tiranathanagul K, Kupatawintu P, Jootar S, Intragumtornchai T, Tungsanga K
et al. Biosimilar recombinant human erythropoietin induces the production of neutralizing
antibodies. Kidney Int. 2011;80:88–92 (abstract; http://www.nature.com/ki/journal/v80/n1/pdf/
ki201168a.pdf, accessed 9 November 2015).
2. Pombo ML, Di Fabio JL, Cortes M de los A. Review of regulation of biological and biotechnological
products in Latin American and Caribbean countries. Biologicals. 2009;37(5):271–6 (abstract;
http://www.sciencedirect.com/science/article/pii/S1045105609000906, accessed 9 November
2015).
144
Annex 3
16. Policy Statement: Clarifying the appropriate regulatory pathway for subsequent entry low
molecular weight heparins. Ottawa: Health Canada; Draft date 12 August 2013 (http://www.hc-sc.
gc.ca/dhp-mps/brgtherap/applic-demande/guides/lmwh-pol-hfmm-eng.php, accessed 18 June
2015).
17. Ministerial Resolution, No. 181-2015. Proyecto de reglamento que regula la presentacion
y contenido de los documentos requeridos en la inscripcion y reinscripcion de productos
biologicos similares. Articulo 8–9. Lima, Ministry of Health; 2015 (ftp://ftp2.minsa.gob.pe/
normaslegales/2015/RM-181-2015-MINSA-CONSULTA.PDF, accessed 9 November 2015).
18. The safety of medicines in public health programs: Pharmacovigilance an essential tool. Geneva:
World Health Organization; 2006 (http://www.who.int/medicines/areas/quality_safety/safety_
efficacy/Pharmacovigilance_B.pdf?ua=1, accessed 9 November 2015).
19. Pombo ML. Biotechnological products in Pan American Health Organization (PAHO): Regional
efforts towards harmonization of regulation. Biologicals. 2011;39(5):348 (http://www.sciencedirect.
com/science/article/pii/S1045105611000753#, accessed 9 November 2015).
20. Kohler M. Regulatory pathways in the European Union. MAbs. 2011;3(3):241–2 (http://www.ncbi.
nlm.nih.gov/pmc/articles/PMC3149704/pdf/mabs0303_0241.pdf, accessed 9 November 2015).
WHO Technical Report Series No. 999, 2016
146
Annex 4
Recommendations to assure the quality, safety and
efficacy of recombinant human papillomavirus virus-like
particle vaccines
Replacement of Annex 1 of WHO Technical Report Series, No. 962
Introduction 151
Scope 152
General considerations 152
Terminology 157
Part A. Manufacturing recommendations 160
A.1 Definitions and international reference materials 160
A.2 General manufacturing recommendations 161
A.3 Control of source materials 162
A.4 Control of HPV VLP production 168
A.5 Control of purified monovalent antigen bulk 173
A.6 Control of adsorbed monovalent antigen bulk 176
A.7 Control of final bulk 177
A.8 Filling and containers 179
A.9 Control tests on the final lot 179
A.10 Records 182
A.11 Retained samples 182
A.12 Labelling 182
A.13 Distribution and transport 182
A.14 Stability testing, storage and expiry date 183
Part B. Nonclinical evaluation of recombinant HPV VLP vaccines 184
B.1 Product characterization and process development 184
B.2 Pharmacodynamic studies 185
B.3 Toxicology studies 186
Part C. Clinical evaluation of recombinant HPV VLP vaccines 186
C.1 Introduction 186
C.2 Immunological data 187
C.3 Virological data 195
C.4 Histological data 198
C.5 Evaluation of vaccine efficacy in different settings 198
C.6 Cross-protection 201
147
WHO Expert Committee on Biological Standardization Sixty-sixth report
148
Annex 4
Abbreviations
CIN cervical intraepithelial neoplasia
CIN2–3 CIN grades 2 or 3
CIN2+ cervical intraepithelial neoplasia grade 2 or worse
cLIA competitive Luminex immunoassay
CMI cell-mediated immunity
DCVMN Developing Countries Vaccine Manufacturers Network
DNA deoxyribonucleic acid
EIA enzyme immunoassay
GCP good clinical practice
GMC geometric mean concentration
GMT geometric mean titre
HPV human papillomavirus
IARC WHO International Agency for Research on Cancer
ICP immune correlate of protection
IFPMA International Federation of Pharmaceutical Manufacturers
& Associations
IU International Unit(s)
LAL limulus amebocyte lysate
LLOD lower limit of detection
LLOQ lower limit of quantification
MCB master cell bank
MOI multiplicity of infection
MPL monophosphoryl lipid A
NAT nucleic acid amplification technique
NCL national control laboratory
NRA national regulatory authority
PAGE polyacrylamide gel electrophoresis
PCR polymerase chain reaction
149
WHO Expert Committee on Biological Standardization Sixty-sixth report
150
Annex 4
Introduction
WHO Guidelines to assure the quality, safety and efficacy of recombinant human
papillomavirus (HPV) virus-like particle (VLP) vaccines were first adopted by
the WHO Expert Committee on Biological Standardization in 2006 (1) and were
based largely on experience gained from clinical trials undertaken on the first
two licensed HPV vaccines.
The factors that have prompted this revision include the substantial
amount of data accumulated during vaccine implementation, the development
of prophylactic vaccines with extended valency and the use of other production
methods. In addition, the increasing availability and routine use of HPV VLP
vaccines composed of L1 capsid protein and containing at least types 16 and
18 have important implications for trial designs and end-points for clinical
evaluation of new prophylactic HPV vaccines.
A series of meetings was convened by WHO to review the scientific
evidence needed to initiate and inform the revision process. These meetings were
attended by experts from around the world involved in the research, manufacture,
licensing/authorization, control-testing and release of HPV vaccines. Participants
were drawn from academia, national regulatory authorities (NRAs), national
control laboratories (NCLs) and industry, and included representatives of the
WHO Global HPV LabNet – an initiative that worked towards the international
standardization of HPV testing during 2006–2011. These experts reviewed new
HPV vaccines under development, and the scientific basis and evidence for
accepting alternative end-points for evaluating the clinical efficacy of candidate
HPV vaccines. The first meeting held was in February 2013 and considered issues
relating to the development and evaluation of clinical end-points for trials of new
HPV vaccines and other issues to be addressed in the proposed revision. At a
meeting held at the WHO International Agency for Research on Cancer (IARC),
Lyon, France in September 2013 a Working Group discussed whether it might be
appropriate to consider using a virological end-point – rather than a disease end-
point such as cervical intraepithelial neoplasia (CIN) grade 2 or worse (CIN2+) –
as the primary end-point for future clinical efficacy trials, and the circumstances
under which immunobridging trials might be sufficient for licensure (2, 3).
A third meeting held at WHO headquarters in November 2013 reviewed and
discussed the outcomes of the IARC scientific meeting on appropriate clinical
end-points, reviewed vaccines currently in the development pipeline, and
assessed regulatory and laboratory needs for licensing the vaccines (4).
Major issues addressed in these resulting WHO Recommendations
include updates of:
■■ terminology;
■■ general considerations and other sections to reflect the up-to-date
development of HPV vaccines;
151
WHO Expert Committee on Biological Standardization Sixty-sixth report
Scope
These WHO Recommendations provide guidance to NRAs and manufacturers
on the manufacturing process, and on nonclinical and clinical aspects, of
recombinant HPV VLP vaccines to assure their quality, safety and efficacy.
The scope of the present document encompasses recombinant HPV
VLP vaccines for prophylactic use which contain the L1 capsid protein of one
or more HPV types.
The document does not cover vaccines targeted to L2 capsid proteins
as antigens, as appropriate serological assays have not yet been standardized
and clinical vaccine trials have not started. Non-VLP vaccines (for example,
other forms of subunit vaccines, vectored vaccines and L1 capsomers) and
investigational therapeutic HPV vaccines, which are at an early stage of
development, are also not included. However, some aspects discussed below may
WHO Technical Report Series No. 999, 2016
General considerations
HPV is not a single virus; rather, it includes a group of closely related small,
non-enveloped deoxyribonucleic acid (DNA) viruses in the Papillomaviridae
152
Annex 4
The terms oncogenic/oncogenicity and carcinogenic/carcinogenicity are both used in the scientific
1
humans (Group 2B) suggest that some, but not all, could be upgraded (18). The
distribution and prevalence of the above HPV types in patients with cancer are
generally consistent around the world. Two of the high-risk HPV types (16 and
18) account for approximately 70% of all cervical cancers globally (14). Most
anal cancers are also associated with persistent HPV infection, with HPV type 16
representing an even higher fraction (90%) of HPV-positive cancers of the anus
than is the case for cervical cancer (approximately 50%). In addition, these high-
risk HPV types are associated with a significant fraction of cancers of the vagina,
vulva, penis and oropharynx. The incidence of cervical cancer is substantially
higher than that of all other HPV-related cancers; cervical cancer is the second
most common cancer among women aged 15–44 years.
Low-risk HPV types cause genital warts, recurrent respiratory
papillomatosis (RRP) and low-grade cervical dysplasia. Genital warts affect
both males and females. Data on the worldwide burden of genital warts are
not available, but in developed countries the epidemiology is similar to other
sexually transmitted infections, peaking in young ages (15–24 years) (19). While
not malignant, these lesions are associated with physical and psychological
morbidity and may be difficult to treat. RRP is a devastating, although rare,
disease that manifests as recurrent, rapidly growing benign laryngeal tumours
that require frequent excision to prevent airway obstruction. HPV types 6 and
11 are responsible for over 90% of genital warts and RRP cases, and for 9–12%
of low-grade cervical dysplastic lesions.
Identification of a viral agent such as HPV as a major cause of diseases
implies that prophylactic vaccines or interventions against the viral agent should
prevent the disease(s) it causes. Initial studies in animal models showed that
inoculation with species-specific papillomaviruses induced an immune response
that conferred protection against homologous virus challenge. However, native
papillomaviruses are not good substrates for vaccine development as they
cannot be grown in standard cell culture. Subsequent studies showed that L1
WHO Technical Report Series No. 999, 2016
recommended that the primary target population should be girls within the age
range of 9 or 10 years through to 13 years – that is, before the age of initiation
of sexual activity and exposure to HPV (11).
The initial product licences were for 3-dose schedules (0, 1 or 2 months
and 6 months). Subsequently the European Medicines Agency approved:
(a) a 2-dose schedule for the bivalent vaccine for females aged 9–14 years; and
(b) a 2-dose schedule for the quadrivalent vaccine for females aged 9–13 years.
For both the bivalent and quadrivalent HPV vaccines, SAGE recommended a
2-dose schedule with a 6-month interval between doses for females younger
than 15 years. Those who are ≥ 15 years at the time of the second dose are also
adequately covered by 2 doses (11).
The extended version of the quadrivalent vaccine includes five additional
HPV types (31, 33, 45, 52 and 58). All nine HPV VLPs are prepared from L1
156
Annex 4
Terminology
The definitions given below apply to the terms as used in these WHO
Recommendations. They may have different meanings in other contexts.
Adjuvant: a substance or a combination of substances used in
conjunction with a vaccine antigen to enhance (for example, increase, accelerate,
prolong and/or possibly target) the specific immune response to the vaccine
antigen and the clinical effectiveness of the vaccine. This may also be called a
mineral vehicle or immunostimulant.
Adsorbed monovalent antigen bulk: a batch of purified monovalent
antigen bulk adsorbed on adjuvant. Different batches of adsorbed monovalent
antigen bulks may be pooled before collection into a single vessel.
If a novel adjuvant is used that does not involve adsorption of the VLP
to the adjuvant, the term “adjuvanted monovalent antigen bulk” may
be used.
name of the recombinant protein (for example, types 16 and 18 L1 proteins). The
proper name should be equivalent to the international name in the language of
the country of origin.
The use of the international name should be limited to vaccines that
meet the specifications elaborated below.
160
Annex 4
161
WHO Expert Committee on Biological Standardization Sixty-sixth report
This should include genetic markers of the host cell, the construction, genetics
and structure of the expression vector, and the origin and identification of the
gene that is being cloned. Some techniques (for example, deep sequencing) allow
for the entire construct to be examined, while others (for example, restriction
enzyme analysis) allow for assessment of segments (30, 38). The molecular and
physiological measures used to promote and control the expression of the cloned
gene in the host cell should be described in detail (38).
The nucleotide sequence of the gene insert and the adjacent segments
of the vector and restriction-enzyme mapping of the vector containing the gene
insert should be provided as required by the NRA.
Cells must be maintained in a frozen state that allows for recovery of
viable cells without alteration of genotype. The cells should be recovered from
the frozen state, if necessary in selective media, such that the genotype and
162
Annex 4
phenotype consistent with the recombinant (modified) host and vector are
maintained and clearly identifiable. Cell banks should be identified and fully
characterized by means of appropriate tests.
Data – for example on plasmid restriction enzyme mapping, nutritional
requirements or antibiotic resistance (if applicable) – that demonstrate the
genetic stability of the expression system during passage of the recombinant
WCB up to or beyond the passage level used for production should be provided
to and approved by the NRA. Any instability of the expression system occurring
in the seed culture or after a production-scale run should be documented.
Stability studies should also be performed to confirm cell viability after retrieval
from storage, maintenance of the expression system, etc. These studies may be
performed as part of their routine use in production or may include samples
taken specifically for this purpose.
cell banks (7). It is important to show that the cell banks are free from bacteria,
fungi, mycoplasmas, mycobacterium species (spp.), and adventitious agents
relevant to the species that may be present in raw materials used in its derivation.
For insect cell lines, special emphasis is put on potential insect-borne human
pathogens (for example, arboviruses).
Insect viruses have not been well characterized compared with other
potential adventitious agents, and there is therefore less information about them
– and specifically about their infectivity, replicative life-cycles and pathogenicity,
if any. It should be borne in mind that infection of insect cells with some insect
viruses may occur without showing cytopathic effect. Tests may include specific
nucleic acid amplification technique (NAT) tests such as polymerase chain
reaction (PCR) and other nonspecific tests such as electron microscopy and
co‑cultivation. The specificity and sensitivity of assays should be determined by
the manufacturer and approved by the NRA.
Full characterization may be performed on either the MCB or on the
WCB, with more-limited testing on the other, depending on the strategy chosen
for testing (7). Scientific advice on the testing strategy should be sought from
the NRA.
in sufficient quantities to last the lifetime of the vaccine product and should
be stored in a secure environment, preferably in two geographically separate
locations. The master seed lot is used as the source material for making the
manufacturer’s recombinant baculovirus working seed lots. Either the virus
master seed lots or the virus working seed lots should be fully characterized
and tested extensively for adventitious agents, while the other may be subjected
to more-limited testing. The testing strategy and seed lots should be approved
by the NRA.
The manufacturer’s recombinant baculovirus working seed lot is used
in the production of inoculum intermediates and single antigen harvests and is
prepared from the master recombinant baculovirus seed lot. It is recommended
that a large lot of recombinant baculovirus working seed should be set aside
as the basic material that the manufacturer should use for the preparation of
each batch of vaccine. The recombinant baculovirus working seed lot should be
prepared by a defined number of passages from the recombinant baculovirus
master seed lot using a method and a passage level from the original virus seed
approved by the NRA. Once the acceptable passage level of the working seed
lot is established, it may not be changed in making future lots of working
seed without approval from the NRA.
A.3.2.1.1 Identity
Each baculovirus master and working seed lot should be identified by the HPV
type of the inserted gene using an appropriate method such as PCR. The tests
should be approved by the NRA.
pigs and/or mice. For details of these tests, see the WHO Recommendations
for the evaluation of animal cell cultures as substrates for the manufacture of
biological medicinal products and for the characterization of cell banks (7).
New molecular methods with broad detection capabilities are being
developed for the detection of adventitious agents. These methods
include: (a) degenerate NAT for whole virus families, with analysis of the
amplicons by hybridization, sequencing or mass spectrometry; (b) NAT
with random primers followed by analysis of the amplicons on large
oligonucleotide microarrays of conserved viral sequencing or digital
subtraction of expressed sequences; and (c) high-throughput or deep
sequencing. These methods may be used to supplement existing methods
or as alternative methods to in vivo and/or in vitro tests after appropriate
validation and approval by the NRA (7).
166
Annex 4
as appropriate. The methods used to ensure this should be approved by the
NRA. The source(s) of trypsin of bovine origin, if used, should be approved by
the NRA and should comply with the current WHO guidelines on transmissible
spongiform encephalopathies in relation to biological and pharmaceutical
products (41).
In some countries, irradiation is used to inactivate potential contaminant
viruses. If irradiation is used it is important to ensure that a reproducible
dose is delivered to all batches and to the component units of each batch.
The irradiation dose must be low enough to allow the biological properties
of the reagents to be retained but high enough to reduce virological risk.
Therefore, irradiation cannot be considered a sterilizing process (7). The
irradiation method should be validated and approved by the NRA.
Human serum should not be used. If human serum albumin is used at any
stage of product manufacture, the NRA should be consulted regarding the
requirements, as these may differ from country to country. As a minimum,
the use of human serum albumin should meet the WHO Requirements for
the collection, processing and quality control of blood, blood components and
plasma derivatives (42). In addition, human albumin and materials of animal
origin should comply with the current WHO guidelines on transmissible
spongiform encephalopathies in relation to biological and pharmaceutical
products (41).
Penicillin and other beta-lactams should not be used at any stage of the
manufacture because they are highly sensitizing substances.
Other antibiotics may be used in the manufacture provided that the
WHO Technical Report Series No. 999, 2016
per ml of pooled fluid. At least one culture vessel of each kind of cell culture
should remain un-inoculated as a control.
The inoculated cultures should be incubated at the appropriate growth
temperature and should be observed for cytopathic effects for a period of at
least 14 days.
For the tests to be valid not more than 20% of the culture cells should
have been discarded by the end of the test period and any losses should be due
to nonspecific or accidental reasons.
Some NRAs require that these cells should be tested for the presence of
haemadsorbing viruses at the end of the observation period.
A.4.4.2.1 Sampling
Samples required for the testing of single antigen harvests or single harvest
pools should be taken immediately on harvesting and before further processing.
If tests for sterility and adventitious agents, as described below in sections
171
WHO Expert Committee on Biological Standardization Sixty-sixth report
A.4.4.2.2 and A.4.4.2.4, are not performed immediately then the samples taken
for these tests should be kept at a temperature of −60 °C or below and subjected
to no more than one freeze–thaw cycle.
In addition to sterility tests for bacteria and fungi, each single antigen harvest
or single harvest pool should also be shown to be free from mycoplasmal
contamination by appropriate tests as specified in Part A, section 5.3 of the
WHO General requirements for the sterility of biological substances (40) if
insect or mammalian cells are used in production, or by a method approved by
the NRA.
NAT alone or in combination with cell culture, with an appropriate
detection method, may be used as an alternative to one or both of the
compendial mycoplasma-detection methods following suitable validation
and agreement from the NRA (7).
the NRA. Alternatively, the identity can be confirmed as part of testing of the
purified antigen.
A.4.4.2.4 Tests for adventitious agents if insect or mammalian cells are used in production
Each single antigen harvest or single harvest pool should be tested for adventitious
viruses in cell cultures selected for their appropriateness to the origin and
passage history of the insect cell substrate and recombinant baculovirus or the
mammalian cell substrate. These cell cultures should include, as a minimum, a
monkey kidney cell line and a human cell line. Antisera used for the purpose
of neutralizing the recombinant baculovirus should be free from antibodies
that may neutralize adventitious viruses, and should preferably be generated
by the immunization of specific-pathogen-free animals with an antigen made
172
Annex 4
from a source (other than the production cell line) which has itself been tested
for freedom from adventitious agents. The inoculated indicator cells should be
examined microscopically for cytopathic changes. At the end of the examination
period, the cells should also be tested for haemadsorbing viruses (see section
A.4.2.1.1 above).
Additional testing for specific adventitious viruses may be performed,
for example by using PCR amplification techniques.
A.5.1.1 Identity
Each purified monovalent antigen bulk should be identified as the appropriate
HPV antigen type by a method suitable for distinguishing between HPV types
(for example, an immunological assay). The test for antigen content may also
serve as the identity test.
A.5.1.2 Purity
The degree of purity of each purified monovalent antigen bulk, and levels of
residual host cell protein, should be assessed by suitable methods. One suitable
method for analysing the proportion of potential contaminating proteins in
173
WHO Expert Committee on Biological Standardization Sixty-sixth report
A.5.1.8 Tests for reagents used during purification or other phases of manufacture
A test should be carried out to detect the presence of any potentially hazardous
reagents used during manufacture, using a method(s) approved by the NRA. This
test may be omitted for routine lot release upon demonstration that the process
consistently eliminates the reagent from the purified monovalent antigen bulks.
A.5.1.9 Tests for residual DNA derived from the expression system
The amount of residual host cell DNA derived from the expression system should
be determined in each purified monovalent antigen bulk by suitably sensitive
methods. The level of host cell DNA should not exceed the maximum level
agreed with the NRA, taking into consideration issues such as those discussed
in the WHO Recommendations for the evaluation of animal cell cultures as
substrates for the manufacture of biological medicinal products and for the
characterization of cell banks (7).
These tests may be omitted for routine lot release upon demonstration
that the process consistently inactivates the biological activity of the residual
DNA or reduces the amount and size of the contaminating residual DNA present
in the purified monovalent antigen bulks, subject to the agreement of the NRA.
A.6.2 Storage
Until the adsorbed monovalent antigen bulk is formulated into the final bulk,
the suspension should be stored under conditions shown by the manufacturer
to allow it to retain the desired biological activity. Hold times should be
approved by the NRA.
Each adsorbed monovalent antigen bulk should be tested for bacterial and fungal
sterility, as specified in Part A, section 5.2 of the WHO General requirements for
the sterility of biological substances (39), or by an alternative method approved
by the NRA.
A.6.3.3 Identity
Each adsorbed monovalent antigen bulk should be identified as the appropriate
HPV antigen type by a method suitable for distinguishing between HPV types
(for example, an immunological assay). The test for antigen content may also
serve as the identity test.
A.6.3.6 pH
The pH value of the adsorbed monovalent antigen bulk may be monitored until
production consistency is demonstrated, subject to the agreement of the NRA.
or adjuvants that are added to the product should have been shown to the
satisfaction of the NRA not to impair the safety and efficacy of the vaccine at
the concentration used. The final bulk should be stored under conditions shown
by the manufacturer to allow it to retain the desired biological activity until it is
filled into containers.
A.7.1.5 Potency
The potency of each final bulk should be assessed with an appropriate in vivo or
in vitro method. If an in vivo potency test is used to test final lots, this test may be
omitted on the final bulk. The methods for detection of antibodies to HPV VLPs
178
Annex 4
and the analysis of data should be approved by the NRA. The vaccine potency
should be compared with that of a reference preparation; the NRA should
determine the limits of potency and approve the reference preparation used.
For ethical reasons, it is desirable to apply the 3R principles (Replacement,
Reduction, Refinement) to the use of animals, where scientifically appropriate (44).
A.7.1.6 Osmolality
The osmolality of the final bulk may be tested. The osmolality test may be
omitted if performed on the final lot.
Alternative tests (for example, freezing point) may be used as surrogate
measures for ionic strength/osmolality.
A.9.2 Appearance
The appearance of the vaccine should be described with respect to its form
and colour.
179
WHO Expert Committee on Biological Standardization Sixty-sixth report
A.9.3 Identity
All antigens present in the final lot should be identified by appropriate methods.
The potency test may serve as the identity test.
A.9.6 Preservatives
Each final lot should be tested for the presence of preservative, if added.
180
Annex 4
A.9.11 Potency
An appropriate quantitative test for potency by an in vivo or in vitro method
should be performed on samples that are representative of each final vaccine
lot. The method and the analysis of data from potency tests should be approved
by the NRA. The vaccine potency should be compared with that of a reference
preparation, and the limits of potency should be agreed with the NRA. The NRA
should approve the reference preparation used. If an in vivo potency test is used,
this test may be omitted on the final bulk. The method of testing for antigen
potency in an in vitro test could be quantitative with respect to the antigen
content or relative to a reference preparation.
Because of the diversity in the reactivity of vaccines containing HPV
VLPs produced by different manufacturing techniques and differences in the
adjuvants used for the vaccine formulation, it is unlikely that International
Standards will be suitable for the standardization of assays of vaccines from all
manufacturers. Consequently, International Standards will not be developed for
the potency of each HPV type. Manufacturers should establish a product-specific
reference preparation that is traceable to a lot of vaccine, or bulks used in the
production of such a lot, which has been shown to be efficacious in clinical trials.
The performance of this reference vaccine should be monitored by trend analysis
using relevant test parameters and the reference vaccine should be replaced when
necessary. An acceptable procedure for replacing reference vaccines should be
in place (45, 46).
181
WHO Expert Committee on Biological Standardization Sixty-sixth report
A.10 Records
The requirements given in WHO good manufacturing practices for biological
products (36) should apply.
A.12 Labelling
The requirements given in WHO good manufacturing practices for biological
products (36) should apply, with the addition of the following information.
The label on the carton, the container or the leaflet accompanying the
container should state:
182
Annex 4
183
WHO Expert Committee on Biological Standardization Sixty-sixth report
C.2.1 Assays
Throughout any one clinical development programme it is preferable that the
same assays for determination of anti-HPV antibody are used and that testing is
conducted at a designated central laboratory. Ideally, the same approach should
apply to post-licensing investigations of antibody persistence. This uniformity
becomes essential within any one study and when attempting to compare
immune responses between studies. The assay (or assays, if a change in assay
during clinical development was unavoidable) used to generate the immune-
response data included in the application dossier should be fully validated. The
details and results of the validation exercise(s) should be provided.
In vitro neutralizing antibody assays involve measurement of the
inhibition of HPV pseudovirus infection of cultured cells and usually employ
type-specific pseudovirions carrying a marker plasmid to allow infected cells to
be scored easily. These neutralizing assays require expression plasmid constructs
for L1 and L2 for each viral type, and assay standardization relies on use of the
same source for these constructs. The WHO Human papillomavirus laboratory
manual (24) includes a method for HPV neutralizing assays that has shown good
inter-laboratory performance.
However, neutralization assays are labour intensive, technically
complex and not currently amenable to high throughput. Therefore, following
characterization of the neutralizing antibody response to a candidate HPV
vaccine, the use of alternative assay methods that are less technically demanding
(for example, type-specific competitive Luminex immunoassay (cLIA) or EIA)
WHO Technical Report Series No. 999, 2016
the correlation with neutralizing assays has been generally good because the
strongest host response to vaccines developed to date is to neutralizing epitopes.
Laboratories performing HPV serology testing have to prepare and conduct
quality-control approaches for their own VLPs because no commercial assays
are available. The inability of laboratories to access common key source reagents
for serology assays presents significant challenges to the standardizing of HPV
serology results.
International Standards for serum antibodies to HPV type 16 and HPV
type 18 are available to help improve the comparability of results. The use of the
parallel-line method with standards calibrated to the International Standard is
described in the WHO Human papillomavirus laboratory manual (24) and has
been shown to improve inter-laboratory comparisons. Antibody levels should be
reported in International Units (IU) for HPV types for which an International
Standard is available. It should be kept in mind that the comparison of titres
between HPV types is not appropriate. For each assay the lower limit of detection
(LLOD) and lower limit of quantification (LLOQ) should be clearly established,
along with a justification of the cut-off applied to differentiate samples that are
reported to be seropositive and seronegative.
190
Annex 4
■■ another HPV vaccine, in which case there may be some HPV types
in both vaccines (shared types) and some that are in one vaccine
only (unshared types);
■■ the same candidate vaccine but administered at a different dose,
different schedule or in a different population;
■■ another formulation of the candidate vaccine (for example, with and
without an adjuvant or with variable numbers of HPV types);
■■ data obtained from a group that does not receive HPV vaccine (that
is, a group that receives either a placebo or a non-HPV vaccine).
In each case, it is recommended that:
■■ Due to their clinical importance, comparisons of immune responses
should strongly support a conclusion that vaccine efficacy against
HPV types 16 and 18 is very likely to be comparable to that observed
for the two initially developed HPV vaccines.
■■ The primary analysis population for immune responses to each HPV
type is confined to those who are seronegative for the particular
HPV type at baseline. Therefore, the sample-size calculations should
also take into account the anticipated HPV type-specific baseline
seropositivity in the population under study.
■■ Primary comparisons should be based on antibody titres in sera
obtained at 1 month after the final dose of the intended regimen(s)
unless antibody-kinetic data suggest otherwise. If a test regimen
consists of a different number of doses from the control regimen
(for example, 2 versus 3 doses) or if the last dose is given at a
different time point (for example, at 4 months versus 6 months after
the first dose) then the primary comparison should still be based
on sera obtained at 1 month (or other time point based on kinetics)
after the final dose, whenever that occurs. Secondary analyses
should compare antibody titres measured at predefined time points
from the first dose, including a comparison once antibody levels
have reached a plateau.
C.2.3.1 Comparison with a group that does not receive HPV vaccine
Given the widespread licensing of HPV vaccines and their incorporation into
routine vaccination programmes in many countries, studies of sexually active
men or women that include a group that does not receive HPV vaccine will
be unacceptable in many settings. Comparisons with a group that does not
191
WHO Expert Committee on Biological Standardization Sixty-sixth report
(that is, shared types) are usually used for the primary comparisons between
vaccinated groups. HPV type-specific seroconversion rates should be included
among the secondary end-points.
As a general rule, for the purposes of establishing non-inferiority
between vaccine groups based on GMT ratios for antibody to individual HPV
types, it is suggested that the lower bound of the 95% confidence interval
around the GMT ratio (test versus reference vaccine) should not fall below 0.67.
Under certain circumstances, NRAs may consider allowing a lower bound of
0.5. In future, especially if an ICP can be identified or if a sponsor is able to
offer a sound rationale, it may be appropriate to reconsider these acceptance
criteria. In addition, any marked separations between the reverse cumulative
distributions should be discussed in terms of the potential clinical implications,
even if these occur only at the lower or upper ends of the curves.
192
Annex 4
immune responses to some HPV types have been lower in men than in women
of comparable ages, and lower in women aged 26–45 years compared to women
aged 15–26 years (71, 72). Therefore, whenever an immunobridging approach
is used, it is relevant to consider possible issues of interpretation based on age
range and gender (see section C.5 below).
An immunobridging approach to support use in immunosuppressed
populations is complicated by the likelihood of observing lower immune
responses compared to those in healthy subjects (73–75). The implications of
lower immune responses for vaccine efficacy are uncertain, as an ICP has not
been established. NRAs will have to consider whether the anticipation of some
degree of benefit in immunosuppressed populations, even if potentially lower
than in immunocompetent subjects, is sufficient to support a favourable risk–
benefit conclusion.
C.3.1 Sampling
Because HPV is cell associated, samples must contain cellular material and
separate samples must be obtained from each specific anatomical site of interest.
Methods of sample collection that have been validated in large-scale
epidemiological studies are recommended. The specific method used (for
example, in terms of number of turns and depth of insertion of the device)
should be standardized and adhered to for each study. Ideally, no changes to
the method should be made during each study or during the entire clinical
development programme. If changes are unavoidable, there should be adequate
cross-validation to support the pooling of results obtained with different
methods. The collection medium will influence the volume of sample to be
extracted as well as the method of extraction. Water or collection-medium
blanks should be processed and tested along with samples to ensure that no
cross-contamination occurs during processing (24).
The standard approach for monitoring HPV in the cervix is for samples
to be collected from the ecto-cervix and endo-cervix by clinicians after
visualization of the cervix using speculum examination (24). Although a range
of collection devices may be used, they should target the cervical transformation
zone and each device should be compatible with the selected collection medium.
Alternative methods (such as sampling only from either the ecto-cervix or
the endo-cervix, or self-sampling by study participants) may be considered if
appropriate validation is provided.
195
WHO Expert Committee on Biological Standardization Sixty-sixth report
negative at study baseline for the types in question are sufficiently high to allow
for studies of reasonable size. This is not expected to be a feasible end-point for
studies in populations in which HPV vaccines have been introduced into routine
immunization programmes and in which there has been a very high uptake.
It is also not likely to be a feasible end-point for HPV types that are rarely
encountered, regardless of any vaccine usage.
The demonstration of viral persistence should be based on consecutive
type-specific HPV DNA positive samples obtained from the same anatomical
site over at least 6 months from the time of the first positive result. Because the
timing of incident infections after completion of the vaccination series cannot be
predicted, an event-driven analysis is often employed. Thus, the primary analysis
is conducted when a protocol-defined number of total cases of viral persistence
196
Annex 4
(which may be based on HPV types 16 and 18 or otherwise defined by types) has
been accumulated. This total number should be estimated to provide sufficient
statistical power to detect meaningful differences between treatment groups.
In the primary analysis, the cases of viral persistence should be counted from
a predefined period (for example, at least 2 weeks) after the final dose of the
vaccination series. A secondary analysis could be based on counting all cases
from the time of the first dose. Protocols should address how to handle sequential
results when at least one of a series of samples is negative for HPV DNA but is
followed by one or more positive samples.
C.3.3.1 Comparison with a group that does not receive HPV vaccine
For the same reasons discussed above in section C.2.3.1, studies of viral
persistence that include a group that does not receive HPV vaccine will be
unacceptable in many settings. If such a design is still considered acceptable and
if incidence rates are sufficiently high to make the study feasible (that is, due to
lack of widespread implementation of HPV vaccination in the regions where
the study will be conducted) then a superiority design could be used.
Depending on what is known about incidence rates, there could be co-
primary end-points of viral persistence for each of HPV types 16 and 18 or a
single composite primary end-point based on viral persistence for both types. For
candidate vaccines containing additional HPV types, a primary analysis could be
based on pooled data for HPV types 16 and 18 with a co-primary or secondary
analysis based on viral-persistence data pooled for all other HPV types and
supportive analyses of viral persistence for each HPV type. Separate or combined
studies addressing cervical and anal sites and/or by gender could be considered.
■■ For any additional HPV types shared between the candidate and
comparator vaccines, supportive analyses should compare viral-
persistence data pooled across all shared types as well as for each
individual type.
■■ For any HPV types in the candidate vaccine only, viral persistence
may be pooled across the additional types, but supportive analyses
should be conducted for individual types.
■■ This section does not cover the assessment of efficacy against genital
warts. For a candidate HPV vaccine containing types 6 and 11, the
considerations regarding whether genital warts is a feasible end-
point are the same as those outlined in section C.3.3.1 for viral
persistence as an end-point. In all other settings, efficacy against
genital warts will have to be based on demonstrating similar immune
responses to these two HPV types between the candidate vaccine
and a suitable control vaccine, as outlined in section C.2.4.
C.6 Cross-protection
The sponsor may choose to assess the ability of a vaccine to elicit cross-reacting
neutralizing antibody against non-vaccine HPV types that are closely related to
the types included in a vaccine. However, experience indicates that these data
cannot be used to establish the ability of a vaccine to confer cross-protection (84).
Thus far, claims for cross-protection against related HPV types not
included in a vaccine have been based on relatively short-term histological
and viral-persistence data. Since the assessment of specific epitopes that
elicit cross-protection is not part of the HPV L1 VLP vaccine specifications,
the degree of cross-protection may be very vaccine specific. Currently, or in
the future, it may not be feasible to demonstrate cross-protection based on
virological persistence. Reliance on immunobridging to a licensed vaccine is not
straightforward because:
201
WHO Expert Committee on Biological Standardization Sixty-sixth report
C.7 Safety
The general considerations for the pre-licensure assessment of safety during
clinical studies are those outlined in the WHO Guidelines on clinical evaluation
of vaccines: regulatory expectations (6).
In the specific case of HPV vaccines, it is important that the safety
database adequately covers the entire target age range and both sexes, as
applicable to the authorized indications. However, it is not necessary to generate
sufficient safety data to estimate frequencies of uncommon adverse events
in each subset. The numbers vaccinated within each sex and age subgroup
should be supported by discussion of any anticipated differences that could
preclude assumptions of similar safety profiles according to the characteristics
of vaccinees (for example, if the reactogenicity profile seems to be very different
between men and women or between adults and younger subjects).
Regardless of protocol recommendations for studies conducted among
sexually active women, numerous pregnancies have been documented in
vaccinees. Every effort should be made to estimate the stage of gestation in
relation to vaccine doses and to document the outcome of the pregnancy.
Specific studies in pregnant women are not recommended at this time (85).
Assessment of safety in the post-licensure period is discussed below in
WHO Technical Report Series No. 999, 2016
section C.8.3.
should provide sufficient information on the vaccine lot. The purpose of this
official national release certificate is to facilitate the exchange of vaccines between
countries, and should be provided to importers of the vaccines. A model NRA
Lot Release Certificate is provided in Appendix 2.
References
1. Guidelines to assure the quality, safety and efficacy of recombinant human papillomavirus virus-
like particle vaccines. In: WHO Expert Committee on Biological Standardization: fifty-seventh
report. Geneva: World Health Organization; 2011: Annex 1 (WHO Technical Report Series,
No. 962; http://apps.who.int/iris/bitstream/10665/44678/1/WHO_TRS_962_eng.pdf, accessed
22 December 2015).
2. Primary end-points for prophylactic HPV vaccine trials. Lyons: International Agency for Research
on Cancer; 2014 (IARC Working Group Report, Volume 7; http://www.iarc.fr/en/publications/
pdfs-online/wrk/wrk7/Prophylactic_HPV_VaccineTrials.pdf, accessed 21 December 2015).
3. Lowy DR, Herrero R, Hildesheim A. Primary endpoints for future prophylactic human papillomavirus
vaccine trials: towards infection and immunobridging. Lancet Oncol. 2015;16(5):e226–33
(abstract: http://www.thelancet.com/journals/lanonc/article/PIIS1470-2045%2815%2970075-6/
abstract, accessed 21 December 2015).
4. Meeting on appropriate clinical endpoints for second generation HPV vaccine trials, 19–20
November 2013, Geneva, Switzerland [website]. Geneva: World Health Organization (http://who.
int/immunization/research/meetings_workshops/hpv_vaccine_endpoint_nov13/en/, accessed
21 December 2015).
5. 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 22 December 2015).
6. 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, accessed 22 December 2015).
7. Recommendations for the evaluation of animal cell cultures as substrates for the manufacture
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 22 December 2015).
8. Guidelines on the nonclinical evaluation of vaccine adjuvants and adjuvanted vaccines. In:
WHO Technical Report Series No. 999, 2016
24. Human papillomavirus laboratory manual. First edition, 2009. Geneva: World Health
Organization; 2010 (WHO/IVB/10.12; http://whqlibdoc.who.int/hq/2010/WHO_IVB_10.12_eng.
pdf?ua=1, accessed 22 December 2015).
25. Zhou J, Sun XY, Frazer IH. Glycosylation of human papillomavirus type 16 L1 protein. Virology.
1993;194(1):210–8 (abstract: http://www.ncbi.nlm.nih.gov/pubmed/8386874, accessed 21
December 2015).
26. Kirnbauer R, Taub J, Greenstone H, Roden R, Dürst M, Gissmann L et al. Efficient self-assembly of
human papillomavirus type 16 L1 and L1-L2 into virus-like particles. J Virol. 1993;67(12):6929–36
(http://www.ncbi.nlm.nih.gov/pmc/articles/PMC238150/pdf/jvirol00033-0027.pdf, accessed 21
December 2015).
27. WHO policy statement: Multi-dose vial policy (MDVP). Handling of multi-dose vaccine vials after
opening (Revision 2014). Geneva: World Health Organization; 2014 (WHO/IVB/14.07; http://apps.
who.int/iris/bitstream/10665/135972/1/WHO_IVB_14.07_eng.pdf, accessed 21 December 2015).
28. Inglis S, Shaw A, Koenig S. Chapter 11: HPV vaccines: commercial research & development.
Vaccine. 2006;24(Suppl 3):S99–105 (abstract: http://www.sciencedirect.com/science/article/pii/
S0264410X06007249, accessed 21 December 2015).
29. Endpoints used for relative effectiveness assessment of pharmaceuticals. Composite endpoints.
February 2013. European Network for Health Technology Assessment (http://eunethta.
fedimbo.belgium.be/sites/5026.fedimbo.belgium.be/files/Composite%20endpoints.pdf,
accessed 21 December 2015).
30. Quality of biotechnological products: analysis of the expression construct in cell lines used for
production of r-DNA derived protein products. ICH Topic Q5B (Step 5). ICH Harmonised Tripartite
Guideline. Geneva: International Conference on Harmonisation of Technical Requirements for
Registration of Pharmaceuticals for Human Use; 1995 (CPMP/ICH/139/95; http://www.ema.
europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500002802.pdf,
accessed 21 December 2015).
31. Ferguson M, Wilkinson DE, Heath A, Matejtschuk P. The first international standard for antibodies
to HPV 16. Vaccine. 2011;29(38):6520–6 (abstract: http://www.sciencedirect.com/science/article/
pii/S0264410X11010280, accessed 21 December 2015).
32. Wilkinson DE, Heath AB, Faust H, Panicker G, Unger ER, Dillner J et al. Collaborative Study to
evaluate the proposed 1st WHO International Standard for antibodies to human papillomavirus
WHO Technical Report Series No. 999, 2016
36. WHO good manufacturing practices for biological products. In: WHO Expert Committee on
Biological Standardization: Sixty-sixth report. Geneva: World Health Organization; 2016: Annex 2
(WHO Technical Report Series, No. 999).
37. Derivation and characterisation of cell substrates used for production of biotechnological/
biological products Q5D (Current Step 4 version). ICH Harmonised Tripartite Guideline. Geneva:
International Conference on Harmonisation of Technical Requirements for Registration of
Pharmaceuticals for Human Use; 1997 (http://www.ich.org/fileadmin/Public_Web_Site/ICH_
Products/Guidelines/Quality/Q5D/Step4/Q5D_Guideline.pdf, accessed 22 December 2015).
38. Guidelines on the quality, safety and efficacy of biotherapeutic protein products prepared
by recombinant DNA technology. In: WHO Expert Committee on Biological Standardization:
sixty‑fourth report. Geneva: World Health Organization; 2014: Annex 4 (WHO Technical Report
Series, No. 987; http://apps.who.int/iris/bitstream/10665/129494/1/TRS_987_eng.pdf?ua=1&ua=1,
accessed 22 December 2015).
39. General requirements for the sterility of biological substances (Requirements for Biological
Substances, No. 6) (Revised 1973). In: WHO Expert Committee on Biological Standardization:
twenty-fifth report. Geneva: World Health Organization; 1973: Annex 4 (WHO Technical Report
Series, No. 530; http://www.who.int/biologicals/publications/trs/areas/vaccines/sterility/WHO_
TRS_530_A4.pdf?ua=1, accessed 22 December 2015).
40. General requirements for the sterility of biological substances (Requirements for Biological
Substances, No. 6, revised 1973, amendment 1995). In: WHO Expert Committee on Biological
Standardization: forty-sixth report. Geneva: World Health Organization; 1998: Annex 3 (WHO
Technical Report Series, No. 872; http://whqlibdoc.who.int/trs/WHO_TRS_872.pdf?ua=1, accessed
22 December 2014).
41. 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 22 December 2015).
42. Requirements for the collection, processing and quality control of blood, blood components and
plasma derivatives (Requirements for Biological Substances, No. 27, revised 1992). In: WHO Expert
Committee on Biological Standardization: forty-third report. Geneva: World Health Organization;
1994: Annex 2 (WHO Technical Report Series, No. 840; http://whqlibdoc.who.int/trs/WHO_
TRS_840.pdf?ua=1, accessed 22 December 2015).
43. Quality of biotechnological products: Viral safety evaluation of biotechnology products derived
from cell lines of human or animal origin. ICH Topic Q5A (R1). (Step 5). ICH Harmonised Tripartite
Guideline. Geneva: International Conference on Harmonisation of Technical Requirements for
Registration of Pharmaceuticals for Human Use; 1995 (CPMP/ICH/295/95; http://www.ema.
europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500002801.pdf,
accessed 22 December 2015).
44. Shin J, Lei D, Conrad C, Knezevic I, Wood D. International regulatory requirements for vaccine
safety and potency testing: a WHO perspective. Procedia in Vaccinology. 2011;5:164–70 (http://
www.sciencedirect.com/science/article/pii/S1877282X11000336, accessed 22 December 2015).
45. Recommendations to assure the quality, safety and efficacy of recombinant hepatitis B vaccines.
In: WHO Expert Committee on Biological Standardization: sixty-first report. Geneva: World
Health Organization; 2013: Annex 4 (WHO Technical Report Series, No. 978; http://www.who.int/
biologicals/expert_committee/TRS_978_61st_report.pdf?ua=1, accessed 22 December 2015).
46. WHO manual for the establishment of national and other secondary standards for vaccines.
Geneva: World Health Organization; 2011 (WHO/IVB/11.03; http://whqlibdoc.who.int/hq/2011/
WHO_IVB_11.03_eng.pdf, accessed 22 December 2015).
211
WHO Expert Committee on Biological Standardization Sixty-sixth report
47. Revocation of general safety test regulations that are duplicative of requirements in biologics
license applications. Federal Register. 2015;80(No. 127): Final Rule; Parts 601, 610, and 680
(https://www.federalregister.gov/articles/2015/07/02/2015-16366/revocation-of-general-safety-
test-regulations-that-are-duplicative-of-requirements-in-biologics, accessed 22 December 2015).
48. Model guidance for the storage and transport of time- and temperature-sensitive pharmaceutical
products. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations: forty-fifth
report. Geneva: World Health Organization; 2011: Annex 9 (WHO Technical Report Series, No. 961;
http://whqlibdoc.who.int/trs/WHO_TRS_961_eng.pdf?ua=1, accessed 22 December 2015).
49. 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://whqlibdoc.who.int/trs/WHO_TRS_962_eng.pdf?ua=1,
accessed 22 December 2015).
50. Guidelines for good clinical practice (GCP) for trials on pharmaceutical products. In: WHO Expert
Committee on the Use of Essential Drugs: sixth report. Geneva: World Health Organization; 1995:
Annex 3 (WHO Technical Report Series, No. 850; http://whqlibdoc.who.int/trs/WHO_TRS_850.
pdf?ua=1, accessed 3 December 2014).
51. Castellsagué X, Naud P, Chow S-N, Wheeler CM, Germar MJV, Lehtinen M et al. Risk of newly
detected infections and cervical abnormalities in women seropositive for naturally acquired
human papillomavirus type 16/18 antibodies: analysis of the control arm of PATRICIA. J Infect
Dis. 2014;210(4):517–34 (http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4111909/, accessed 22
December 2015).
52. Nardelli-Haefliger D, Wirthner D, Schiller JT, Lowy DR, Hildesheim A, Ponci F et al. Specific antibody
levels at the cervix during the menstrual cycle of women vaccinated with human papillomavirus
16 virus-like particles. J Natl Cancer Inst. 2003;95(15):1128–37 (http://jnci.oxfordjournals.org/
content/95/15/1128.long, accessed 22 December 2015).
53. Kemp TJ, García-Piñeres A, Falk RT, Poncelet S, Dessy F, Giannini SL et al. Evaluation of systemic
and mucosal anti-HPV16 and anti-HPV18 antibody responses from vaccinated women.
Vaccine. 2008;26(29–30):3608–16 (abstract: http://www.sciencedirect.com/science/article/pii/
S0264410X08005483, accessed 22 December 2015).
54. Schwarz TF, Kocken M, Petäjä T, Einstein MH, Spaczynski M, Louwers JA et al. Correlation between
levels of human papillomavirus (HPV)-16 and 18 antibodies in serum and cervicovaginal
WHO Technical Report Series No. 999, 2016
secretions in girls and women vaccinated with the HPV-16/18 AS04-adjuvanted vaccine. Hum
Vaccin. 2010;6(12):1054–61 (abstract: http://www.ncbi.nlm.nih.gov/pubmed/21157180, accessed
23 December 2015).
55. Petäjä T, Pedersen C, Poder A, Strauss G, Catteau G, Thomas F et al. Long-term persistence of
systemic and mucosal immune response to HPV-16/18 AS04-adjuvanted vaccine in preteen/
adolescent girls and young women. Int J Cancer. 2011;129(9):2147–57 (http://onlinelibrary.wiley.
com/doi/10.1002/ijc.25887/full, accessed 23 December 2015).
56. Scherpenisse M, Mollers M, Schepp RM, Meijer CJLM, de Melker HE, Berbers GAM et al. Detection
of systemic and mucosal HPV-specific IgG and IgA antibodies in adolescent girls one and two
years after HPV vaccination. Hum Vaccin Immunother. 2013;9(2):314–21 (http://www.ncbi.nlm.
nih.gov/pmc/articles/PMC3859753/, accessed 23 December 2015).
57. Schiller JT, Lowy DR. Immunogenicity testing in human papillomavirus virus-like-particle
vaccine trials. J Infect Dis. 2009;200(2):166–71 (http://jid.oxfordjournals.org/content/200/2/166.
long, accessed 23 December 2015).
212
Annex 4
58. Moscicki AB, Wheeler CM, Romanowski B, Hedrick J, Gall S, Ferris D et al. Immune responses
elicited by a fourth dose of the HPV-16/18 AS04-adjuvanted vaccine in previously
vaccinated adult women. Vaccine. 2012;31(1):234–41 (abstract: http://www.ncbi.nlm.nih.gov/
pubmed/23063422, accessed 23 December 2015).
59. Villa LL, Costa RLR, Petta CA, Andrade RP, Ault KA, Giuliano AR et al. Prophylactic quadrivalent
human papillomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in young women:
a randomized double-blind placebo-controlled multicentre phase II efficacy trial. Lancet
Oncol. 2005;6(5):271–78 (abstract: http://www.thelancet.com/journals/lanonc/article/PIIS1470-
2045%2805%2970101-7/abstract, accessed 23 December 2015).
60. Harper DM, Franco EL, Wheeler C, Ferris DG, Jenkins D, Schuind A et al. Efficacy of a bivalent L1
virus-like particle vaccine in prevention of infection with human papillomavirus types 16 and
18 in young women: a randomised controlled trial. Lancet. 2004;364(9447):1757–65 (abstract:
http://www.thelancet.com/journals/lancet/article/PIIS0140-6736%2804%2917398-4/abstract,
accessed 23 December 2015).
61. Pedersen C, Petaja T, Strauss G, Rumke HC, Poder A, Richardus JH et al. Immunization of early
adolescent females with human papillomavirus type 16 and 18 L1 virus-like particle vaccine
containing AS04 adjuvant. J Adolesc Health. 2007;40(6):564–71 (abstract: http://www.ncbi.nlm.
nih.gov/pubmed/17531764, accessed 23 December 2015).
62. Perez G, Lazcano-Ponce E, Hernandez-Avila M, García PJ, Muñoz N, Villa LL et al. Safety,
immunogenicity, and efficacy of quadrivalent human papillomavirus (types 6, 11, 16, 18) L1
virus-like-particle vaccine in Latin American women. Int J Cancer. 2008;122(6):1311–8 (http://
onlinelibrary.wiley.com/doi/10.1002/ijc.23260/full, accessed 23 December 2015).
63. Wheeler CM, Bautista OM, Tomassini JE, Nelson M, Sattler CA, Barr E et al. Safety and
immunogenicity of co-administered quadrivalent human papillomavirus (HPV)-6/11/16/18 L1
virus-like particle (VLP) and hepatitis B (HBV) vaccines. Vaccine. 2008;26(5):686–96 (abstract:
http://www.ncbi.nlm.nih.gov/pubmed/18164106, accessed 23 December 2015).
64. Petäjä T, Keränen H, Karppa T, Kawa A, Lantela S, Siitari-Mattila M et al. Immunogenicity and
safety of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine in healthy boys aged
10–18 years. J Adolesc Health. 2009;44(1):33–41 (abstract: http://www.ncbi.nlm.nih.gov/
pubmed/19101456, accessed 23 December 2015).
65. Krajden M, Cook D, Yu A, Chow R, Mei W, McNeil S et al. Human papillomavirus 16 (HPV 16) and
HPV 18 antibody responses measured by pseudovirus neutralization and competitive Luminex
assays in a two- versus three-dose HPV vaccine trial. Clin Vaccine Immunol. 2011;18(3):418–23
(http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3067374/, accessed 23 December 2015).
66. Romanowski B, Schwarz TF, Ferguson LM, Peters K, Dionne M, Schulze K et al. Immunogenicity and
safety of the HPV-16/18 AS04-adjuvanted vaccine administered as a 2-dose schedule compared
with the licensed 3-dose schedule: results from a randomized study. Hum Vaccin. 2011;7(12):1374–
86 (http://www.tandfonline.com/doi/full/10.4161/hv.7.12.18322, accessed 23 December 2015).
67. Zimmerman RK, Nowalk MP, Lin CJ, Fox DE, Ko FS, Wettick E et al. Randomized trial of an alternate
human papillomavirus vaccine administration schedule in college-aged women. J Womens
Health. 2010;19(8):1441–7 (abstract: http://www.ncbi.nlm.nih.gov/pubmed/20629576, accessed
23 December 2015).
68. Esposito S, Birlutiu V, Jarcuska P, Perino A, Man SC, Vladareanu R et al. Immunogenicity and
safety of human papillomavirus-16/18 AS04-adjuvanted vaccine administered according to an
alternative dosing schedule compared with the standard dosing schedule in healthy women
aged 15 to 25 years: results from a randomized study. Pediatr Infect Dis J. 2011;30(3):e49–55
(abstract: http://www.ncbi.nlm.nih.gov/pubmed/21273939, accessed 23 December 2015).
213
WHO Expert Committee on Biological Standardization Sixty-sixth report
69. Romanowski B, Schwarz TF, Ferguson LM, Ferguson M, Peters K, Dionne M et al. Immune response
to the HPV-16/18 AS04-adjuvanted vaccine administered as a 2-dose or 3-dose schedule
up to 4 years after vaccination: results from a randomized study. Hum Vaccin Immunother.
2014;10(5):1155–65 (http://www.tandfonline.com/doi/abs/10.4161/hv.28022, accessed 23
December 2015).
70. Reisinger KS, Block SL, Lazcano-Ponce E, Samakoses R, Esser MT, Erick J et al. Safety and persistent
immunogenicity of a quadrivalent human papillomavirus types 6, 11, 16, 18 L1 virus-like particle
vaccine in preadolescents and adolescents: a randomized controlled trial. Pediatr Infect Dis
J. 2007;26(3):201–9 (abstract: http://www.ncbi.nlm.nih.gov/pubmed/17484215, accessed 23
December 2015).
71. Hillman RJ, Giuliano AR, Palefsky JM, Goldstone S, Moreira Jr ED, Vardas E et al. Immunogenicity
of the quadrivalent human papillomavirus (type 6/11/16/18) vaccine in males 16 to 26 years
old. Clin Vaccine Immunol. 2012;19(2):261–7 (http://www.ncbi.nlm.nih.gov/pmc/articles/
PMC3272915/pdf/zcd261.pdf, accessed 23 December 2015).
72. Einstein MH, Baron M, Levin MJ, Chatterjee A, Fox B, Scholar S et al. Comparative immunogenicity
and safety of human papillomavirus (HPV)-16/18 vaccine and HPV-6/11/16/18 vaccine: follow-
up from months 12–24 in a Phase III randomized study of healthy women aged 18–45 years.
Hum Vaccin. 2011;7(12):1343–58 (http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3338932/,
accessed 23 December 2015).
73. Levin MJ, Moscicki A-B, Song L-Y, Fenton T, Meyer III WA, Read JS et al. Safety and immunogenicity
of quadrivalent human papillomavirus (types 6, 11, 16, and 18) vaccine in HIV-infected children
7 to 12 years old. J Acquir Immune Defic Syndr. 2010;55(2):197–204 (http://www.ncbi.nlm.nih.
gov/pmc/articles/PMC3033215/, accessed 23 December 2015).
74. Wilkin T, Lee JY, Lensing SY, Stier EA, Goldstone SE, Berry JM et al. Safety and immunogenicity
of the quadrivalent human papillomavirus vaccine in HIV-1-infected men. J Infect Dis.
2010;202(8):1246–53 (http://jid.oxfordjournals.org/content/202/8/1246.long, accessed 23
December 2015).
75. Denny L, Hendricks B, Gordon C, Thomas F, Hezareh M, Dobbelaere K et al. Safety and
immunogenicity of the HPV-16/18 AS04-adjuvanted vaccine in HIV-positive women in South
Africa: a partially-blind randomised placebo-controlled study. Vaccine. 2013;31(48):5745–53
(http://www.sciencedirect.com/science/article/pii/S0264410X13012735, accessed 23 December
2015).
WHO Technical Report Series No. 999, 2016
76. Ferguson M, Wilkinson DE, Zhou T. WHO meeting on the standardization of HPV assays and
the role of the WHO HPV Laboratory Network in supporting vaccine introduction held on
24–25 January 2008, Geneva, Switzerland. Vaccine. 2009;27(3):337–47 (abstract: http://www.
sciencedirect.com/science/article/pii/S0264410X08014588, accessed 23 December 2015).
77. Eklund C, Forslund O, Wallin K-L, Dillner J. Global improvement in genotyping of human
papillomavirus DNA: the 2011 HPV LabNet International Proficiency Study. J Clin Microbiol.
2014;52(2):449–59 (http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3911320/, accessed 23
December 2015).
78. Paavonen J, Naud P, Salmerón J, Wheeler CM, Chow SN, Apter D et al. Efficacy of human
papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer
caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study
in young women. Lancet. 2009;374(9686):301–14 (abstract: http://www.ncbi.nlm.nih.gov/
pubmed/19586656/, accessed 23 December 2015).
214
Annex 4
79. Villa LL, Costa RLR, Petta CA, Andrade RP, Paavonen J, Iversen O-E et al. High sustained efficacy
of a prophylactic quadrivalent human papillomavirus types 6/11/16/18 L1 virus-like particle
vaccine through 5 years of follow-up. Br J Cancer. 2006;95(11):1459–66 (http://www.ncbi.nlm.
nih.gov/pmc/articles/PMC2360730/, accessed 23 December 2015).
80. Lehtinen M, Paavonen J, Wheeler CM, Jaisamrarn U, Garland SM, Castellsagué X et al. on behalf
of the HPV PATRICIA Study Group. Overall efficacy of HPV-16/18 AS04-adjuvanted vaccine
against grade 3 or greater cervical intraepithelial neoplasia: 4-year end-of-study analysis of the
randomised, double-blind PATRICIA trial. Lancet Oncol. 2012;13(1):89–99 (abstract: http://www.
thelancet.com/journals/lanonc/article/PIIS1470-2045%2811%2970286-8/abstract, accessed 23
December 2015).
81. Wheeler CM, Castellsagué X, Garland SM, Szarewski A, Paavonen J, Naud P et al. on behalf of
the HPV PATRICIA Study Group. Cross-protective efficacy of HPV-16/18 AS04-adjuvanted
vaccine against cervical infection and precancer caused by non-vaccine oncogenic HPV types:
4-year end-of-study analysis of the randomised, double-blind PATRICIA trial. Lancet Oncol.
2012;13(1):100–10
(abstract: http://www.thelancet.com/journals/lanonc/article/PIIS1470-2045%2811%2970287-X/
fulltext, accessed 23 December 2015).
82. Radley D, Saah A, Stanley M. Persistent infection with human papillomavirus 16 or 18 is strongly
linked with high-grade cervical disease. Hum Vaccin Immunother. 2015; in press (abstract: http://
www.tandfonline.com/doi/full/10.1080/21645515.2015.1088616, accessed 24 January 2016).
83. Palefsky JM, Giuliano AR, Goldstone S, Moreira Jr ED, Aranda C, Jessen H et al. HPV vaccine
against anal HPV infection and anal intraepithelial neoplasia. N Engl J Med. 2011;365:1576–85
(http://www.nejm.org/doi/pdf/10.1056/NEJMoa1010971, accessed 23 December 2015).
84. Einstein MH, Baron M, Levin MJ, Chatterjee A, Fox B, Scholar S et al. Comparison of the
immunogenicity of the human papillomavirus (HPV)-16/18 vaccine and the HPV-6/11/16/18
vaccine for oncogenic non-vaccine types HPV-31 and HPV-45 in healthy women aged 18–45
years. Hum Vaccin. 2011;7(12):1359–73 (http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3338933/,
accessed 23 December 2015).
85. Vichnin M, Bonanni P, Klein NP, Garland SM, Block SL, Kjaer SK et al. An overview of quadrivalent
human papillomavirus vaccine safety: 2006 to 2015. Pediatr Infect Dis J. 2015;34(9):983–91
(abstract: http://www.ncbi.nlm.nih.gov/pubmed/26107345, accessed 23 December 2015).
86. Guidelines for national authorities on quality assurance for biological products. In: WHO
Expert Committee on Biological Standardization: forty-second report. Geneva: World Health
Organization; 1992: Annex 2 (WHO Technical Report Series, No. 822; http://www.who.int/
biologicals/publications/trs/areas/biological_products/WHO_TRS_822_A2.pdf?ua=1, accessed 23
December 2015).
87. 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/expert_
committee/TRS_978_61st_report.pdf?ua=1, accessed 23 December 2015).
215
WHO Expert Committee on Biological Standardization Sixty-sixth report
Appendix 1
Model protocol for the manufacturing and control of
recombinant human papillomavirus virus-like particle
vaccines
The following protocol is intended for guidance. It indicates the information that
should be provided as a minimum by the manufacturer to the NRA. Information
and tests may be added or omitted as necessary with the approval of the NRA.
It is possible that a protocol for a specific product may differ in
detail from the model provided here. The essential point is that all relevant
details demonstrating compliance with the licence and with the relevant
WHO Recommendations for a particular product should be provided in the
protocol submitted.
The section concerning the final lot must be accompanied by a sample of
the label and a copy of the leaflet (package insert) that accompanies the vaccine
container. If the protocol is being submitted in support of a request to permit
importation, it must also be accompanied by a lot release certificate from the
NRA or from the NCL in the country in which the vaccine was produced or
released, stating that the product meets national requirements as well as the
recommendations in Part A of this document.
Country:
Name and address of manufacturer:
Name and address of licence holder, if different:
Final lot
Batch number(s)
Final lot:
Final bulk:
Type of container:
Total number of filled containers in this final lot:
216
Annex 4
Production information
Batch number of each monovalent bulk
(purified and/or adsorbed):
Site of manufacture of each monovalent bulk:
Date of manufacture of each monovalent bulk:
Site of manufacture of adjuvant(s):
Date of manufacture of adjuvant(s):
Site of manufacture of final bulk:
Date of manufacture of final bulk:
Site of manufacture of final lot:
Date of manufacture of final lot:
Date on which last determination of potency was started or
date of start of period of validity:
Shelf-life approved (months):
Expiry date:
Storage conditions:
Release date:
A genealogy of the lot numbers of all vaccine components used in the formulation of
the final lot will be informative.
The following sections are intended for the reporting of the results of the tests
performed during the production of the vaccine, so that the complete document
will provide evidence of consistency of production. Therefore, if any test has to
be repeated this must be indicated. Any abnormal result must be recorded on a
separate sheet.
Starting materials
The information requested below is to be presented on each submission. Full details
on master and working seed lots, and cell banks are requested upon first submission
only and whenever a change has been introduced.
Adventitious agents
Method:
Specification:
Date:
Result:
Control cell cultures if mammalian or insect cells are used for production and
recombinant viral vector cannot be neutralized, thus interfering with testing.
Karyotype
Method:
Probe:
Reference cells:
Date of start of test:
Date of end of test:
Result:
Method:
Probe:
Reference cells:
Restriction enzymes:
Date of start of test:
Date of end of test:
Result:
Haemadsorbing viruses
Type(s) of red blood cell (RBC):
Storage time and temperature of RBC:
Incubation time and temperature of RBC:
220
Annex 4
221
WHO Expert Committee on Biological Standardization Sixty-sixth report
Mycoplasmas
Method:
Volume inoculated:
Date of start of test:
Date of end of test:
Result:
Specification:
Date:
Result:
222
Annex 4
Mycoplasmas
Method:
Volume inoculated:
Date of start of test:
Date of end of test:
Result:
223
WHO Expert Committee on Biological Standardization Sixty-sixth report
Protein content
Method:
Specification:
Date:
Result:
Date:
Result:
Bovine serum albumin content (if mammalian or insect cells and animal serum are
used for production).
Method:
Specification:
Date:
Result:
Viral clearance
This is performed during vaccine manufacturing development and/or process
validation and is not intended for batch release (see section A.5.1.11).
Method:
Specification:
Date:
Bacterial endotoxins
Method:
Specification:
Date:
Result:
Identity
Method:
Specification:
Date:
Result:
pH
Method:
Specification:
Date:
Result:
WHO Technical Report Series No. 999, 2016
Adjuvants
Method:
Specification:
Date:
Result:
Potency
If an in vitro assay of each type is used
Method:
Batch number of reference vaccine and
assigned potency:
Specification:
Date:
Result:
227
WHO Expert Committee on Biological Standardization Sixty-sixth report
Appearance
Method:
Specification:
228
Annex 4
Date:
Result:
pH
Method:
Specification:
Date:
Result:
229
WHO Expert Committee on Biological Standardization Sixty-sixth report
Pyrogenic substances
Method:
Specification:
Date:
Result:
Adjuvant content
Method:
Specification:
Date:
Result:
Potency
If an in vitro assay of each type is used
Method:
Batch number of reference vaccine and
WHO Technical Report Series No. 999, 2016
assigned potency:
Specification:
Date:
Result:
1
With the exception of provisions on distribution and shipping, which the NRA may not be in a position
to assess.
2
WHO Technical Report Series, No. 999, Annex 4.
231
WHO Expert Committee on Biological Standardization Sixty-sixth report
Appendix 2
Model NRA Lot Release Certificate for recombinant
human papillomavirus virus-like particle vaccines
Certificate No.
1
Name of manufacturer.
2
Country of origin.
3
If any national requirements have not been met, specify which one(s) and indicate why the release of
the lot(s) has nevertheless been authorized by the NRA.
4
With the exception of provisions on distribution and shipping, which the NRA may not be in a position
to assess.
5
WHO Technical Report Series, No. 999, Annex 4.
6
WHO Technical Report Series, No. 986, Annex 2.
7
WHO Technical Report Series, No. 999, Annex 2.
8
WHO Technical Report Series, No. 978, Annex 2.
9
Evaluation of the summary protocol, independent laboratory testing and/or procedures specified in a
defined document etc., as appropriate.
232
Annex 4
233
Annex 5
Guidelines on the stability evaluation of vaccines for use
under extended controlled temperature conditions
1. Introduction 238
2. Scope 239
3. Terminology 239
4. General considerations for the evaluation of vaccines for use
under ECTC 241
5. Stability evaluation of vaccines for use under ECTC 245
6. Monitoring ECTC 250
7. Suggested product labelling information for use under ECTC 251
8. Authors and acknowledgments 252
9. References 255
Appendix Product-specific ECTC evaluation of a model monovalent
polysaccharide conjugate vaccine 257
235
WHO Expert Committee on Biological Standardization Sixty-sixth report
236
Annex 5
Abbreviations
CTC controlled temperature chain
ECTC extended controlled temperature conditions
EU ELISA Unit(s)
IFPMA International Federation of Pharmaceutical Manufacturers
& Associations
LB lower bound
LL lower limit
MRP minimum release potency
NLT not less than
NMR nuclear magnetic resonance
NMT not more than
NRA national regulatory authority
PS polysaccharide
237
WHO Expert Committee on Biological Standardization Sixty-sixth report
1. Introduction
Vaccines are complex biological products and may undergo degradation during
long-term storage under cold chain conditions (for example, 2–8 °C) and this
is typically enhanced at higher temperatures. Consequently, establishing the
stability characteristics of products is a critical element of the overall evaluation
by a national regulatory authority (NRA) to ensure that licensed vaccines
remain efficacious at the end of their shelf-life when stored under the approved
conditions. In response to the stability assessment needs identified by NRAs,
WHO developed guidelines on the stability evaluation of vaccines to assist its
Member States (1). While it is well understood that vaccine quality depends
on cold chain storage, it is also recognized that immunization programmes in
certain regions face substantial challenges in maintaining cold chains in the
field, especially during the final stage of distribution in remote areas (2, 3). To
address these distribution challenges and expand immunization programmes
into specific regions WHO developed a “controlled temperature chain” (CTC)
programme. This programme currently requires that a vaccine exhibits a stability
profile suitable for a single exposure to at least 40 °C for a minimum of 3 days
just prior to administration, while remaining compliant with the approved
vaccine specifications. Additionally, the programme requires that the CTC
provision should be included in the licensure by the relevant NRA and by WHO
prequalification (4).
During the development of these WHO Guidelines, the term “extended
controlled temperature conditions” (ECTC) was proposed to distinguish
regulatory requirements from WHO CTC programme aspects. This terminology
convention is used throughout the following guidance. An ECTC assessment
should assure the performance of a vaccine following short-term exposure
to temperatures above those of a typical cold chain and could consider any
temperature above the traditional 2–8 °C cold chain that might support
WHO Technical Report Series No. 999, 2016
2. Scope
This document provides guidance to NRAs and manufacturers on the scientific
and regulatory issues to be considered in evaluating the stability of vaccines for
use under ECTC. Evaluation criteria are provided for the approval of short-term
temperature conditions, in addition to those defined for long-term storage of
a given vaccine, in situations where the vaccine is exposed to these short-term
conditions immediately prior to administration.
This document does not provide guidance on the stability evaluation of
vaccines that are inadvertently or repeatedly exposed to temperatures for which
they were not licensed.
3. Terminology
The definitions given below apply to the terms as used in these WHO Guidelines.
They may have different meanings in other contexts.
239
WHO Expert Committee on Biological Standardization Sixty-sixth report
obtained are used to recommend storage conditions and to establish the shelf-
life and/or release specifications.
Shelf-life: the period of time during which a vaccine, when stored
under approved conditions, is expected to comply with the specifications. The
shelf-life is determined by stability studies on a number of product batches and
is used to establish the expiry date of each batch of a final product.
Stability of vaccine: the ability of a vaccine to retain its physical,
chemical, biological, biopharmaceutical and microbiological properties within
specified limits to assure clinical performance throughout its shelf-life.
Stability-indicating parameters: quality parameters (direct or indirect
indicators of vaccine efficacy or safety) that are sensitive to storage conditions.
These parameters are used in stability studies to assure product quality throughout
240
Annex 5
subsequent papers (10, 11) and is critical for ECTC applications, to which the
same principles apply. A product-release model (Fig. 1) should be developed
on the basis of studies using the manufacturer’s assays, along with quality
data and other essential information. Labelling a vaccine for ECTC use will
require the support of the manufacturer and the approval of the appropriate
regulatory authorities.
Fig. 1 illustrates the relationship between the MRP specification of 50
ELISA Units (EU) and the shelf-life (24 months) given the rate of decay (slope)
of the potency over both the long-term storage temperature (2–8 °C) and the
maximum ECTC temperature (40 °C), and indicates that the vaccine is above
the approved LL for the potency (30 EU; supported by clinical lots) at the end
of shelf-life. The potency decay assessment should be based on appropriate
242
Annex 5
statistical analysis of multiple lots with a given degree of confidence (for example,
95%) and should include assay variability (not shown in the figure). As noted
below in section 5, logarithmic transformation (log-transformation) of potency
data typically permits analysis of stability data by linear regression.
Fig. 1
Graphic representation of a product-release model for an ECTC application
80
70
Potency (EU = ELISA Unit)
Potency range
60 at release
MRP Potency
range of
50
2–8 °C clinical lots
Available
40
potency
40 °C
30
LL
0
0 6 12 18 24 30 36
Shelf-life (months)
have binary outputs (for example, pass/fail). In such cases, supplemental potency
assays that are capable of showing the decay of the product’s active ingredient (or
that can provide a worst-case estimate of that decay) may be considered for use
in ECTC evaluation, recognizing the need for conservatism in interpreting the
analysis and its results. In the absence of adequate stability-indicating assays for
a vaccine, approval of an ECTC label would not be possible solely on the basis
of a quality data assessment.
While ECTC approval is not a recommendation for shipping or storage,
and unplanned excursions are not within the scope of this document, an
approved ECTC label could potentially be taken into account when making
decisions on product use in cases of temporary temperature excursions.
However, given the finite nature of available potency over the shelf-life of any
given vaccine, potency lost through unplanned excursions with specific final
containers earlier in the shelf-life would not be available to support the use of
those same containers of vaccines during a planned ECTC exposure later in the
shelf-life. This highlights the importance of maintaining the cold chain prior
to the extreme temperature conditions that the vaccine would be subjected to
during a planned ECTC exposure.
For multivalent vaccines, ECTC evaluations must consider all antigens
in the product. If one antigen is known to be less stable than other antigens
within a specific vaccine then the suitability of the product for ECTC should
be based on the least-stable antigen. Potential interference between vaccine
components, including adjuvants, stabilizers and preservatives, may also
need to be considered, as applicable. To simplify the discussion here, with the
exception of comments related to free PS, the focus of this and subsequent
sections will be on how to evaluate and manage the available potency of a
monovalent vaccine.
At release, a product must possess sufficient potency to ensure clinical
effectiveness throughout its shelf-life and to account for assay variability as well
WHO Technical Report Series No. 999, 2016
assay variability, thus reducing the amount of potency required to account for
potential errors in initial potency assignment. Finally, with a more accurate
characterization of vaccine stability, a manufacturer could reduce the amount of
potency required to account for errors in the estimation of shelf-life.
In general, additional clinical assessment for a previously approved
product being considered in an ECTC application should be required only
where a planned ECTC exposure results in a change of product specifications.
For example, a lower end-of-shelf-life potency or a higher release specification
that is not supported by clinical experience would potentially require
further clinical evaluation. If additional clinical studies could demonstrate
that lower potencies were still effective, or that higher-than-approved target
release potencies would not result in new or more-frequent adverse events,
a manufacturer could submit a regulatory amendment and use the broader
potency ranges gained through these approaches to extend the ECTC potential
of a product. Field studies in which the clinical evaluation of a vaccine has
been performed on a product that has been exposed to temperatures higher
than the approved conditions, but without potency and quality testing using
the manufacturer’s assays, are not considered acceptable from a regulatory
perspective. Clinical studies that are intended to support ECTC applications
should be performed using a vaccine with known (or modelled) potency at the
time of the ECTC use, as determined by the manufacturer’s assays.
Finally, when a lyophilized vaccine is being considered for ECTC
applications, a stability analysis should be performed for the reconstituted
product using the rigorous statistical evaluation principles described in this
document. In situations where exposure to an ECTC storage condition could
result in changes to the visual appearance of the lyophilized product that do not
impact on its clinical performance, the manufacturer should provide relevant
supportive data to the NRA considering the ECTC label change. If approved, a
description of the potential change(s) in visual appearance should be included in
the product leaflet and/or package insert. For liquid multi-dose vials, additional
data would be required to demonstrate the antimicrobial effectiveness of the
preservative under ECTC.
statistical evaluation is needed to be able to state that, with a given (usually 95%)
degree of confidence, the potency after ECTC exposure at expiry will still be
above the minimum threshold needed for product efficacy. It is only through
the use of statistical analysis that it is possible to obtain an indication of the
level of confidence in results reported at release or in potencies delivered to the
vaccine recipient. Therefore, statistical analysis is required to assure the quality
of vaccines intended for delivery in the context of ECTC. Because the major
ECTC-related concern is usually that the temperature exposure will reduce
potency to unacceptable levels, the following guidance focuses on ensuring that
the minimum required potency is maintained.
Even when products are sufficiently stable to tolerate an ECTC exposure,
poorly designed studies or inappropriate statistical analyses can reduce the
likelihood that an ECTC exposure is justified. This section describes study-
design and statistical approaches that will improve the likelihood that ECTC
exposure can be justified using sound scientific principles.
Although additional clinical studies will not be required for an ECTC
approval for most licensed products, it is essential to establish the minimum
potency specification for a specific vaccine, through initial licensing studies, for
all stability assessments. Changes in specifications (including lowering of end-
expiry potency specifications) may require supporting clinical data. Thus, the
data package for ECTC applications should include summaries of the initial
clinical studies, including the quality data for the clinical lots, to support the
end-of-shelf-life potency specification.
The data package should also include stability studies that formally
demonstrate that the minimum potency is achieved throughout the time to
expiry, including the ECTC exposure. Estimates of the rate (or slope) at which
the potency decays (hereafter, referred to as “stability estimates”) at the normal
and ECTC temperatures – and an understanding of potential errors in those
estimates – are the most important outcomes of the stability studies. The
WHO Technical Report Series No. 999, 2016
reliability of these stability estimates, and the extent to which the release potency
of any lot can be reliably determined, depend in turn on the potency assay.
As mentioned above in section 4, stability studies to support vaccine
use in an ECTC context should use the manufacturer’s potency assay in order
to preserve a connection between the released product proposed for ECTC use
and the original clinical material used to support product efficacy. It is likely
that key parameters of the potency assay will already be known from assay
validation, including assay accuracy and precision. More reliable estimates of
in-use assay precision may sometimes be obtained by other means (for example,
by comparing actual with modelled results in the stability analyses). Other data
may also be relevant to the estimation of in-use assay precision. Because there
are several possible estimates of assay precision that could be used for ECTC-
related calculations, the choice of estimate should be scientifically justified; if
246
Annex 5
a clear justification cannot be made, the more conservative estimate (from the
perspective of the ECTC label) should be used.
Statistical analysis of vaccine stability is normally based on a
mathematical model and supported by data that describe the kinetics of potency
changes at different temperatures for different periods of time. Statistical release
models must support the conclusion that the mean potency of final containers
in a given lot will, given all stability losses, meet specifications throughout the
shelf-life with a given level of confidence (usually 95%), including permitted
storage periods outside the long-term storage conditions. Typically, the rate
of change (generally loss) of vaccine potency is not a simple linear function of
time. Log-transformation of potency data usually leads to a more predictive
model that permits analysis of stability data by linear regression. Thus, in most
cases, potency data should be log-transformed before analysis. When log-
transformation is not used, scientific justification for the use of a more relevant
model should be provided. Log-transformation usually provides a preferred
model of the biological process (since for most substances the rate of decay at
any point in time depends on the quantity of substance present at that time) as
compared with direct analysis of non-log-transformed potency data.
Moreover, empirical observation supports the conclusion that potency
decay for many vaccines follows first-order kinetics, which are linear following
log-transformation – though low precision of the potency assay may make it
more difficult to determine whether stability results follow the decay model.
In addition, potency measurements are often log-normally distributed; when
this is the case, log-transformation may be required to satisfy the statistical
assumptions of the modelling, and can further improve the precision of the
stability estimate. In all cases, the decay model used should correspond to actual
product decay kinetics as observed in stability studies, and this may support the
use of non-log-transformed decay models (including linear models) if these
models can also be justified as biologically relevant. It should be noted that log-
transformation is not always the best approach for stability-indicating assays.
For example, increases in degradation products over time usually cannot be
modelled on a log scale. Visual examination of the plot of transformed and
untransformed stability data can provide an indication of whether mathematical
transformations can linearize the decay curve. Sometimes no biologically
meaningful model can be identified that fits the data, as may occur when there
are multiple phases in the decay kinetics. In this case, decay estimates during
ECTC exposures can be estimated by using only the beginning and end results
of the stability testing. If the most appropriate choice of model is unclear,
selection of the most conservative option is appropriate.
Stability studies should properly evaluate the kinetics of decay, and
should indicate that decay rates (after any transformation) are not higher at
the end of the observation period than at the beginning. These studies should
247
WHO Expert Committee on Biological Standardization Sixty-sixth report
As noted, the equations listed are for the general case that could include modelling storage, shipping,
1
post-reconstitution and so on. However, when only considering the normal storage condition and a
single ECTC exposure, an example of a simplified form of the equations would be: LB 1-α = MRP + t 1 ∙ b 1 +
t ECTC ∙ b ECTC - U.
249
WHO Expert Committee on Biological Standardization Sixty-sixth report
should also be noted that the equations here are not the only ways to represent
these calculations and that other approaches that encompass similar statistical
principles could potentially be acceptable where justified.
6. Monitoring ECTC
All vaccines should be kept under the recommended long-term storage
conditions with appropriate oversight prior to ECTC exposure. Use of vaccines
under ECTC requires specific monitoring of temperature exposure (for example,
peak threshold indicator) and time, as well as formal procedures to ensure
that the approved maximum temperature and time are not exceeded. Unused
vaccines that exceed the maximum approved temperature or time should be
250
Annex 5
Krause, United States Food and Drug Administration Center for Biologics
Evaluation and Research, the USA; Dr M. (Ferguson) Lennon, Consultant,
Horning, the United Kingdom; Dr H. Meyer, Paul-Ehrlich-Institut, Germany;
Dr V. Oeppling, Paul-Ehrlich-Institut, Germany; Dr M. Pfleiderer, Paul-
Ehrlich-Institut, Germany; Dr J. Shin, World Health Organization, Switzerland;
Dr D. Smith, Health Canada, Canada; Dr R. Wagner, Paul-Ehrlich-Institut,
Germany; Dr T. Wu, Health Canada, Canada; Ms S. Zipursky, World Health
Organization, Switzerland.
Acknowledgments are extended to the following participants in the
consultations held in Ottawa, Canada, 4–6 December 2012 and Langen,
Germany, 4–6 June 2013: Dr M-C. Annequin, Agence nationale de sécurité
du médicament et des produits de santé, France; Dr M. Baca-Estrada, Health
252
Annex 5
9. References
1. 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://who.int/biologicals/vaccines/Annex_3_WHO_TRS_962-3.
pdf?ua=1, accessed 11 December 2015).
2. Meeting of the Strategic Advisory Group of Experts on Immunization (SAGE), Geneva, 10–
12 April 2012 [website]. Geneva: World Health Organization; 2012 (http://www.who.int/
immunization/sage/meetings/2012/april/presentations_background_docs/en/index.html,
accessed 11 December 2015).
3. WHO Immunization Practices Advisory Committee (IPAC), Geneva, 17–18 April 2012. Final meeting
report and recommendations. Geneva: World Health Organization; 2012 (http://www.who.int/
immunization/policy/committees/IPAC_2012_April_report.pdf?ua=1, accessed 7 February 2016).
4. Vaccine management and logistics: Controlled Temperature Chain (CTC) [website]. Geneva: World
Health Organization (http://www.who.int/immunization/programmes_systems/supply_chain/
resources/tools/en/index6.html, accessed 14 December 2015).
5. Butler D. Vaccines endure African temperatures without damage: anti-meningitis campaign
in Benin delivers more than 150,000 doses with no losses from excess heat. Nature. 19
February 2014 (http://www.nature.com/news/vaccines-endure-african-temperatures-without-
damage-1.14744#/ref-link-1, accessed 14 December 2015).
255
WHO Expert Committee on Biological Standardization Sixty-sixth report
6. Zipursky S, Djingarey MH, Lodjo J-C, Olodo L, Tiendrebeogo S, Ronveaux O. Benefits of using
vaccines out of the cold chain: delivering meningitis A vaccine in a controlled temperature chain
during the mass immunization campaign in Benin 2014. Vaccines. 2014;32:1431–5 (https://
www.researchgate.net/publication/260373472_Benefits_of_using_vaccines_out_of_the_cold_
chain_Delivering_Meningitis_A_vaccine_in_a_controlled_temperature_chain_during_the_
mass_immunization_campaign_in_Benin, accessed 14 December 2015).
7. Use of MenAfriVac™ (meningitis A vaccine) in a controlled temperature chain (CTC) during
campaigns. Geneva: World Health Organization; 2013 (WHO/IVB/13.04; http://apps.who.int/iris/
bitstream/10665/86018/1/WHO_IVB_13.04_eng.pdf?ua=1, accessed 7 February 2016).
8. WHO/Health Canada drafting group meeting on scientific and regulatory considerations on
the stability evaluation of vaccines under controlled temperature chain. Ottawa, Canada, 4–6
December 2012. Meeting report. Geneva: World Health Organization; 2012 (http://who.int/
biologicals/areas/vaccines/CTC_FINAL_OTTAWA_Web_Meeting_report_25.11.2013.pdf?ua=1,
accessed 14 December 2015).
9. WHO/Paul-Ehrlich-Institut informal consultation on scientific and regulatory considerations
on the stability evaluation of vaccines under controlled temperature chain (CTC). Paul-Ehrlich-
Institut, Langen, Germany, 4–6 June 2013. Meeting report. Geneva: World Health Organization;
2013 (http://who.int/biologicals/vaccines/CTC_Final_Mtg_Report_Langen.pdf?ua=1, accessed 7
February 2016).
10. Krause PR. Goals of stability evaluation throughout the vaccine life cycle. Biologicals.
2009;37(6):369–78 (abstract available at: http://www.sciencedirect.com/science/article/pii/
S1045105609001201, accessed 14 December 2014).
11. Schofield TL. Vaccine stability study design and analysis to support product licensure. Biologicals.
2009;37(6):387–96 (abstract available at: http://www.ncbi.nlm.nih.gov/pubmed/19717312,
accessed 14 December 2014).
WHO Technical Report Series No. 999, 2016
256
Annex 5
Appendix
Product-specific ECTC evaluation of a model monovalent
polysaccharide conjugate vaccine
The model vaccine and the stability data presented in this appendix were
developed on the basis of Health Canada’s overall experience with conjugate
vaccines and do not represent characteristics or data from any specific product.
The analysis presented is also applicable to other stability-indicating parameters,
such as vaccine potency. The vaccine example under evaluation is a monovalent
conjugate vaccine composed of purified capsular polysaccharide (PS) covalently
attached to diphtheria toxoid protein. The final vaccine product is a non-
adjuvanted liquid formulation presented in single-dose vials. The normal storage
temperature for this model conjugate vaccine is 2–8 °C, with a time to expiry of
three years; the temperature under consideration for the ECTC application is
40 °C. The quality attributes monitored in routine stability studies intended for
licensure included total PS, free PS, molecular size distribution, free protein,
pH and sterility. Free PS is considered a key stability-indicating attribute for
polysaccharide conjugate vaccines since in general this parameter is linked to
the clinical performance of this type of vaccine. The specification for free PS for
this model conjugate vaccine was set as “not more than (NMT) 15%” at release
and “NMT 25%” at the end of the shelf-life. A review of manufacturing data
indicated that, at release, 90% of the commercial lots contained less than 10%
of free PS and 10% of lots contained free PS in the range 10–13%. In addition,
vaccine lots containing 5–25% free PS were shown to be safe and immunogenic
in clinical studies.
Stability data
Real-time and real-condition stability studies were conducted to establish the
shelf-life under normal storage conditions (2–8 °C) and to support the ECTC
application. Although a minimum of three lots is required for statistical
modelling, analysis of a larger data set (more lots) leads to more-precise
estimates. In this example, routine stability-monitoring tests were performed
for four commercial vaccine lots stored at 2–8 °C and for an additional four
commercial lots stored at 40 °C. In addition, O-acetyl content, nuclear magnetic
resonance (NMR) spectrum and immunogenicity (rabbit complement source
serum bactericidal assay and immunoglobulin G) in a mouse model were also
evaluated to characterize vaccine lots exposed to the 40 °C condition. Analysis
257
WHO Expert Committee on Biological Standardization Sixty-sixth report
of routine monitoring data revealed that total PS, molecular size distribution,
free protein and pH were stable for all lots stored under the 2–8 °C and 40 °C
conditions. However, an increase of free PS was observed for all lots, as
summarized in Tables 1 and 2.
Table 1
Summary of free PS content at 2–8 °C
Table 2
Summary of free PS content at 40 °C
Statistical analysis
An initial analysis (1) demonstrated that the free PS data did not fit a linear
regression, with or without log-transformation (plots not provided). Because
the increase in the stability-indicating free PS is due to the hydrolysis of bound
PS, the rate of increase of free PS is the same as the rate of decrease of bound PS.
Therefore, the bound PS at each test point can be calculated from the free and
total PS on the basis of mass balance. The hydrolysis of bound PS can be analysed
as a first-order reaction at a decay rate that is proportional to the concentration
258
Annex 5
of bound PS. Consequently, the rate of increase of free PS was analysed indirectly
through the modelling of bound PS, and log-transformation of the bound PS
content at different test points yielded data that were more amenable to linear
regression analysis (1). Thus, free PS data obtained in stability studies were
converted to percentage bound PS (Tables 3 and 4) and then subjected to log-
transformation. A release model was developed to characterize the relationship
between bound PS at release and end-expiry, thus permitting evaluation of
potential ECTC use.
Table 3
Summary of bound PS content at 2–8 °C
Table 4
Summary of bound PS content at 40 °C
1. For each stability lot, the percentage of bound PS at each test point
was log-transformed and the slope was calculated using a linear
regression model. Plots of the linear regression fit for all stability
lots are presented in Fig. A1.
2. Lots 1, 2, 3 and 4, monitored at 2–8 °C, were assessed with respect
to slope variability, which was considered acceptable for use of the
linear regression model with a pooled (mean) slope for all four lots.
The same analysis was also applied to the data set (lots 5, 6, 7 and 8)
at 40 °C, which supported the use of a pooled slope.
3. The assay precision (sassay) was estimated by the residual error
from the regression analysis using the pooled slope for the
corresponding data set.
4. The uncertainty was calculated using formulae described above in
section 5. Two examples are:
■■ the uncertainty (U) at 2–8 °C for 36 months = z 0.95 ∙ sqrt [(s assay)2
+ (t 2–8 ∙ s(b 2–8))]2
= 1.644854 ∙ sqrt [0.012728112 + (36 ∙ 0.0001845235)2] = 0.02361.
■■ U at 2–8 °C for 36 months followed by 3 days at 40 °C
= z 0.95 ∙ sqrt [(s assay)2 + (t 2–8 ∙ s(b 2–8))2 + (tECTC ∙ s(b ECTC))2]
= 1.644854 ∙ sqrt [0.012728112 + (36 ∙ 0.0001845235)2 +
(0.1 ∙ 0.008625)2]
= 0.02366.
260
Annex 5
Table 5
Summary of statistical analysis of bound PS data at 2–8 °C
Table 6
Summary of statistical analysis of bound PS data at 2–8 °C and 40 °C
261
WHO Expert Committee on Biological Standardization Sixty-sixth report
Fig. A1
Plot of bound PS data for four conjugate vaccine lots
Owing to limited data points at 40 °C, the rate of bound PS decay used
for ECTC application was based on the modelling of a 4-week data set, and a
conservative approach was taken to limit the total decay of bound PS to slightly
below 10%. The statistical analysis summarized in Table 6 revealed an estimated
9.5% loss of bound PS (equal to the increase of free PS) after 36 months storage
at 2–8 °C followed by 3 days at 40 °C, or 24 months storage at 2–8 °C followed
by 7 days at 40 °C.
262
Annex 5
Conclusion
The application of the “product release model” to the analysis of free PS can be
summarized as:
Release specification (15%) = End of shelf-life specification (25%)
minus estimated combined increase at 2–8 °C and 40 °C (upper
bound with 95% confidence level)
On the basis of statistical modelling and product-related information, the
following can be concluded:
■■ Different specifications for release and end of shelf-life should
be established for free PS for this model conjugate vaccine.
A specification of “NMT 25%” at the end of shelf-life is considered
acceptable on the basis of clinical lots shown to be safe and
immunogenic in clinical studies. A release specification of “NMT
15%” was considered appropriate on the basis of manufacturing
capability, which ensures a high compliance rate for commercial lots
at release.
■■ A 36-month shelf-life at 2–8 °C was determined to be appropriate
for this model conjugate vaccine. This conclusion ensures that
worst-case lots, which contain the highest level of free PS permitted
by the release specification (NMT 15%), plus the accumulation of
free PS during the storage period (approximately 8.94%), comply
with the end of the shelf-life specification (NMT 25%).
■■ A single storage period of 3 days at 40 °C, prior to immunization,
was considered acceptable because the worst-case lots, which
contain 15% free PS at release and are stored for almost 36 months
at 2–8 °C followed by an exposure of 3 days at 40 °C, were expected
to contain approximately 24.51% free PS.
■■ If a period longer than 3 days at 40 °C is needed, shortening the
shelf-life at 2–8 °C from 36 months to 24 months would allow for
a single storage period of 7 days at 40 °C. Alternatively, the release
specification of “NMT 15% free PS” might be tightened (for example
to “NMT 13% free PS”) on the basis of additional manufacturing
experience to allow for longer than 3 days of ECTC exposure while
maintaining a 3-year shelf-life at 2–8 °C.
As explained above in section 4, clinical testing of a vaccine stored under
ECTC would not be necessary as long as a battery of stability-monitoring tests
provided sufficient assurance that the critical quality attributes of the vaccine
(such as potency) met the specifications supported by clinical experience.
263
WHO Expert Committee on Biological Standardization Sixty-sixth report
References
1. Wickham H. ggplot2: Elegant Graphics for Data Analysis. New York: Springer-Verlag New York,
2009.
2. R Core Team. R: a language and environment for statistical computing. Vienna: R Foundation for
Statistical Computing; 2014 (available at: http://www.R-project.org/, accessed 22 July 2015).
WHO Technical Report Series No. 999, 2016
264
Annex 6
Biological substances: WHO International Standards,
Reference Reagents and Reference Panels
A list of WHO International Standards, Reference Reagents and Reference Panels
for biological substances is available at: http://www.who.int/biologicals.
At its meeting in October 2015, the WHO Expert Committee on Biological
Standardization made the changes shown below to the previous list.
Additions1
Preparation Activity Status
Antibiotics
Streptomycin* 76 000 IU/vial Fourth WHO International
Standard
Biotherapeutics other than blood products
Tumour necrosis factor 10 000 IU/ampoule First WHO International
receptor Fc fusion protein Standard
(etanercept)
Antibodies to Nine panel members; no First WHO Reference Panel
erythropoietin (human) unitage assigned
Blood products and related substances
Anti-A and anti-B in For DRT: 128 (nominal Second WHO Reference
serum and plasma for use anti-A and anti-B titre) Reagent
in haemagglutination For IAT: 256 (nominal anti-A
assays and anti-B titre)
Blood coagulation factor 10.5 IU/ampoule Fifth WHO International
IX (concentrate) Standard
Unless otherwise indicated, all materials are held and distributed by the National Institute for Biological
1
Standards and Control, Potters Bar, Herts, EN6 3QG, the United Kingdom. Antibiotic reference preparations
identified by an * in the above list are held and distributed by the European Directorate for the Quality
of Medicines & HealthCare, Council of Europe, 7 allée Kastner, CS 30026 F-67081, Strasbourg, France.
Materials identified by an ** in the above list are held and distributed by the Paul-Ehrlich-Institut, 63225
Langen, Germany.
265
WHO Expert Committee on Biological Standardization Sixty-sixth report
266
Annex 6
267
SELECTED WHO PUBLICATIONS OF RELATED INTEREST
Further information on these and other WHO publications can be obtained from
WHO Press, World Health Organization, 1211 Geneva 27, Switzerland
(tel.: +41 22 791 3264; fax: + 41 22 791 4857; email: [email protected];
order online: www.who.int/bookorders)
999
W H O Te c h n i c a l R e p o r t S e r i e s
999
on Biological
guidance documents. Following these discussions, a WHO
guidance document on Regulatory assessment of approved
rDNA-derived biotherapeutics was adopted along with WHO
Standardization
Guidelines on the stability evaluation of vaccines for use under
extended controlled temperature conditions and on WHO good
manufacturing practices for biological products. In addition,
revised WHO Recommendations to assure the quality, safety
and efficacy of recombinant human papillomavirus virus-like
particle vaccines were also adopted by the Committee.
Subsequent sections of the report provide information on the
current status and proposed development of international Sixty-sixth report
reference materials in the areas of antibiotics; biotherapeutics
other than blood products; blood products and related
substances; in vitro diagnostic device reagents; and vaccines