TRS 1039 - 9789240046870 Eng
TRS 1039 - 9789240046870 Eng
TRS 1039 - 9789240046870 Eng
1039
The Organization seeks through its publications to support national health strategies
and address the most pressing public health concerns of populations around the world.
To respond to the needs of Member States at all levels of development, WHO publishes
practical manuals, handbooks and training material for specific categories of health
workers; internationally applicable guidelines and standards; reviews and analyses of
health policies, programmes and research; and state-of-the-art consensus reports that
offer technical advice and recommendations for decision-makers. These books are
closely tied to the Organization’s priority activities, encompassing disease prevention
and control, the development of equitable health systems based on primary health
care, and health promotion for individuals and communities. Progress towards better
health for all also demands the global dissemination and exchange of information
that draws on the knowledge and experience of all WHO’s Member countries and the
collaboration of world leaders in public health and the biomedical sciences.
The WHO Technical Report Series makes available the findings of various international
groups of experts that provide WHO with the latest scientific and technical advice on
a broad range of medical and public health subjects. Members of such expert groups
serve without remuneration in their personal capacities rather than as representatives
of governments or other bodies; their views do not necessarily reflect the decisions or
the stated policy of WHO.
To purchase WHO publications, please contact: WHO Press, World Health Organization,
20 Avenue Appia, 1211 Geneva 27, Switzerland; email: [email protected]; order
online: http://apps.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
1 0 3 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 Expert Committee on Biological Standardization: seventy-fourth report
(WHO Technical Report Series, No. 1039)
ISBN 978-92-4-004687-0 (electronic version)
ISBN 978-92-4-004688-7 (print version)
ISSN 0512-3054
iv
9. International reference materials – vaccines and related substances 65
9.1 WHO international reference standards for vaccines and related substances 65
9.1.1 Second WHO International Standard for diphtheria antitoxin (equine) 65
9.2 Proposed new projects and updates – vaccines and related substances 66
9.2.1 Proposed Fifth WHO International Standard for pertussis vaccine (whole cell) 66
9.2.2 Proposed WHO International Reference Reagent for diphtheria CRM197 antigen 67
Annex 1
WHO Recommendations, Guidelines and other documents related to the manufacture,
quality control and evaluation of biological products 69
Annex 2
Recommendations to assure the quality, safety and efficacy of live attenuated yellow
fever vaccines
Amendment to Annex 5 of WHO Technical Report Series, No. 978 75
Annex 3
Evaluation of the quality, safety and efficacy of messenger RNA vaccines for the
prevention of infectious diseases: regulatory considerations 85
Annex 4
New and replacement WHO international reference standards for biological products 155
v
vi
WHO Expert Committee on Biological Standardization
Seventy-fourth meeting held via video conferencing on 18 to 22 October 2021
Committee members 1
Dr K.M. Boukef, University of Monastir, Monastir, Tunisia
Dr C. Burns, National Institute for Biological Standards and Control, Potters Bar, the United
Kingdom
Dr N. Choudhury, Health City Hospital, Guwahati, India
Professor K. Cichutek, Paul-Ehrlich-Institut, Langen, Germany (Vice-chair; and Chair for the
vaccines and biotherapeutics track)
Dr M. Darko,2 Food and Drugs Authority, Accra, Ghana
Dr A.E. del Pozo, Hospital de Pediatria Garrahan, Buenos Aires, Argentina
Dr I. Feavers, Consultant, Nacton, the United Kingdom (Rapporteur for the plenary sessions
and for the vaccines and biotherapeutics track)
Professor S. Hindawi, King Abdulaziz University, Jeddah, Saudi Arabia (Chair for the blood
products and in vitro diagnostics track)
Mrs T. Jivapaisarnpong, Advisor, King Mongkut’s University of Technology Thonburi,
Bangkok, Thailand (Chair)
Dr G. Kang, Christian Medical College, Vellore, India
Ms C. Morris, National Institute for Biological Standards and Control, Potters Bar, the United
Kingdom
Mr V.R. Reddy,2 South African National Blood Service, Weltevreden Park, South Africa
Dr Y. Sohn, Seoul National University, Seoul, Republic of Korea
Dr D. Teo, Visiting Consultant, Blood Services Group, Health Sciences Authority, Singapore,
Singapore (Rapporteur for the blood products and in vitro diagnostics track)
Dr J. Wang, National Institutes for Food and Drug Control, Beijing, China
Dr Y. Wang, National Institutes for Food and Drug Control, Beijing, China
Dr C. Witten, Center for Biologics Evaluation and Research, Food and Drug Administration,
Silver Spring, MD, United States of America (the USA)
1
The decisions of the Committee were taken in closed session with only members of the Committee
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.
2
Unable to attend.
vii
WHO Expert Committee on Biological Standardization Seventy-fourth report
Temporary advisors
Dr P. Aprea, National Administration of Drugs, Food and Medical Technology, Buenos Aires,
Argentina
Dr A.D. Barrett, University of Texas Medical Branch, Galveston, TX, the USA
Dr C.A. Bravery, Consultant, London, the United Kingdom
Dr M. Buda, European Directorate for the Quality of Medicines & Healthcare, Strasbourg,
France
Dr E. Griffiths, Consultant, Kingston upon Thames, the United Kingdom
Dr A. Hilger, Paul-Ehrlich-Institut, Langen, Germany
Dr A. Kitchen, Consultant, Billericay, the United Kingdom
Dr H. Klein, National Institutes of Health, Bethesda, MD, the USA
Dr M.A. Liu, Consultant, Lafayette, CA, the USA
Dr L. Mallet, European Directorate for the Quality of Medicines & Healthcare, Strasbourg,
France
Dr P. Marks, Center for Biologics Evaluation and Research, Food and Drug Administration,
Silver Spring, MD, the USA
Dr J. Martin, National Institute for Biological Standards and Control, Potters Bar, the UK
Dr H. Meyer, Paul-Ehrlich-Institut, Langen, Germany
Dr P. Minor, St Albans, the United Kingdom
Professor D.H. Muljono, Eijkman Institute for Molecular Biology, Jakarta, Indonesia
Dr M. Page, National Institute for Biological Standards and Control, Potters Bar, the United
Kingdom
Dr K. Peden, Center for Biologics Evaluation and Research, Food and Drug Administration,
Silver Spring, MD, the USA
WHO Technical Report Series, No. 1039, 2022
State actors
Dr N. Almond, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr M. Bailey, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr K. Chumakov, Center for Biologics Evaluation and Research, Food and Drug
Administration, Bethesda, MD, the USA
Dr A. Douglas-Bardsley, National Institute for Biological Standards and Control, Potters
Bar, the United Kingdom
Dr B. Fox, National Institute for Biological Standards and Control, Potters Bar, the United
Kingdom
Dr J, Fryer, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr S. Govind, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr L. Hassall, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr M.M. Ho, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr K. Ishii, National Institute of Infectious Diseases, Tokyo, Japan
Dr A. Ishii-Watabe, National Institute of Health Sciences, Kawasaki, Japan
Dr S. Kempster, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr K.M. Lee, Ministry of Food and Drug Safety, Chungcheongbuk-do, Republic of Korea
Dr G. Mattiuzzo, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr M. Moore, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
ix
WHO Expert Committee on Biological Standardization Seventy-fourth report
United Kingdom
Dr S. Williams, National Institute for Biological Standards and Control, Potters Bar, the
United Kingdom
Dr R. Isbrucker
Dr H-N. Kang
Dr D. Lei
Dr S.H. Yoo
Dr J. Yu
Dr T. Zhou
Medical Devices and Diagnostics (MHP/HPS/ATM/MDD)
Dr F.G. Moussy
In Vitro Diagnostics Assessment (MHP/RPQ/PQT/IVD)
Dr U. Stroher
Vaccines and Immunization Devices Assessment (MHP/RPQ/PQT/VAX)
Dr C. Rodriguez-Hernandez
Dr D. Williams, Consultant on venom and antivenom
Agenda, Policy and Strategy (UHL/IVB/APS)
Dr J. Hombach
Product and Delivery Research (UHL/IVB/PDR)
Dr E. Sparrow
Dr Y. Abdella
WHO Regional Office for the Western Pacific
Dr J. Shin
3
Unable to attend: WHO Regional Office for South-East Asia and WHO Regional Office for Europe.
xii
Abbreviations
ACT WHO Access to COVID-19 Tools (Accelerator)
AFP alpha-fetoprotein
AG-BRA Advisory Group for Blood Regulation, Availability and Safety
ATMP advanced therapy medicinal product
BAU binding antigen unit
BCG bacillus Calmette–Guérin
CBER Center for Biologics Evaluation and Research
CCP COVID-19 convalescent plasma
CEPI Coalition for Epidemic Preparedness Innovations
CGTP cell and gene therapy product
COVID-19 coronavirus disease 2019
CRM197 cross-reacting material 197
Ct cycle threshold
cVDPV2 circulating vaccine-derived poliovirus serotype 2
DAT diphtheria antitoxin
DNA deoxyribonucleic acid
EDQM European Directorate for the Quality of Medicines &
HealthCare
ELISA enzyme-linked immunosorbent assay
EUL emergency use listing
FDA US Food and Drug Administration
FSH follicle-stimulating hormone
FIX blood coagulation factor IX
GBT Plus Blood WHO Global Benchmarking Tool Plus Blood
GP glycoprotein
HBV hepatitis B virus
HIV human immunodeficiency virus
ICDRA International Conference of Drug Regulatory Authorities
xiii
WHO Expert Committee on Biological Standardization Seventy-fourth report
PEI Paul-Ehrlich-Institut
Ph. Eur. European Pharmacopoeia
Ph. Eur. BRP European Pharmacopoeia Biological Reference Preparation
RDT rapid diagnostic test
RNA ribonucleic acid
RSV respiratory syncytial virus
SAGE Strategic Advisory Group of Experts (on Immunization)
SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
sIPV Sabin IPV
xiv
Abbreviations
TB tuberculosis
TgAb thyroglobulin antibody
TPO thyroid peroxidase
TPP target product profile
VOC variant of concern
VWF von Willebrand factor
VZV varicella zoster virus
xv
1. Introduction
The seventy-fourth meeting of the WHO Expert Committee on Biological
Standardization was held from 18 to 22 October 2021 via video conference due to
the travel restrictions imposed during the coronavirus disease 2019 (COVID 19)
pandemic. The meeting was opened on behalf of the Director-General of WHO
and the Assistant Director-General, Access to Medicines and Health Products, by
Dr Clive Ondari, Director, Health Products Policy and Standards. Dr Ondari began
by welcoming Committee members, meeting participants and observers, noting
that this was one of the most long-standing WHO expert committees. Dr Ondari
also noted the particularly large number of new members of the Committee for
the current meeting and expressed his gratitude for the commitment and support
of all members in these challenging times. Dr Ondari singled out Dr Harvey
Klein for particular thanks following his recent retirement from the Committee,
and expressed appreciation for his considerable contribution to the WHO Blood
Regulators Network and to the blood-related activities of the Committee over the
past 15 years.
Dr Ondari noted that the work of WHO had inevitably been impacted
by the additional demands arising from the COVID-19 pandemic. Nevertheless,
there remained a pressing need for WHO to maintain its wide range of global
public health activities. Reflecting on the broad agenda of the current meeting, Dr
Ondari looked forward to receiving the expert advice of the Committee in these
areas, and thanked the secretariat for their efforts in preparing for the meeting.
Dr Ondari acknowledged the hard work of all of the WHO expert committees in
addressing both the COVID-19 and non-COVID-19 activities of WHO.
The WHO response to COVID-19 was continuing at all levels of
the organization, and was geared towards accelerating the development and
availability of diagnostics, therapeutics and vaccines. Dr Ondari noted the crucial
ongoing contribution of the Committee to all three of these pillars of the WHO
Access to COVID-19 Tools (ACT) Accelerator, and expressed the view that the
speed of the Committee in facilitating the development of WHO written and
measurement standards for COVID-19 had in part being made possible by its
previous experience of other emerging pathogens. In this regard, the Executive
Board of the World Health Assembly had placed on record its appreciation for
the rapid development of the WHO regulatory considerations document on the
evaluation of messenger RNA (mRNA) vaccines. Dr Ondari also highlighted
the pivotal role of the National Institute for Biological Standards and Control
(NIBSC) in providing measurement standards to support COVID-19 vaccine
clinical trials with unprecedented speed. In addition, upcoming WHO guidance
documents on the evaluation and use of monoclonal antibodies (mAbs) for the
treatment of infectious diseases, including COVID-19, will make an important
contribution to the therapeutics pillar of the ACT Accelerator. The advice of
1
WHO Expert Committee on Biological Standardization Seventy-fourth report
3
2. General
2.1 Strategic directions in biological standardization
2.1.1 WHO overview
Dr Knezevic began her overview by noting the link between the work of
the Committee and that of other WHO expert committees and groups,
and by outlining the WHO and other resources relevant to WHO biological
standardization activities. Within WHO, the Norms and Standards for Biologicals
(NSB) and the Blood and other Products of Human Origin (BTT) teams were
part of the Technical Standards and Specifications Unit which itself was part
of the Health Policy and Standards Department. Other resources included
WHO collaborating centres, stakeholders, external experts and the Expert
Advisory Panel on Biological Standardization. Among recent improvements,
BTT had been strengthened and the Expert Advisory Panel expanded to ensure
the availability of expertise in new areas of work and to improve geographical
representation. Dr Knezevic then outlined the four strategic priorities of the
WHO five-year plan to help build effective and efficient regulatory systems,
namely: (a) strengthening national and regional regulatory systems; (b)
improving regulatory preparedness for public health emergencies; (c) reinforcing
and expanding WHO prequalification and product risk assessment; and (d)
increasing the impact of WHO regulatory support activities.
Dr Knezevic then highlighted several recent cross-cutting WHO
COVID-19 activities in the context of the three pillars of the ACT Accelerator.
In the case of the vaccines pillar (COVAX), such activities had included the
development of WHO written and measurement standards, and collaboration
with a range of working groups set up by WHO and partner organizations.
WHO standards were now cited in the criteria for EUL of COVID-19 vaccines
and therefore play an important role in WHO prequalification of such vaccines.
WHO Technical Report Series, No. 1039, 2022
given to the revision of the current WHO interim guidance on maintaining a safe
and adequate blood supply during the pandemic, and on the safe collection of CCP.
Reflecting on the importance of WHO collaborating centres during
the pandemic, Dr Knezevic specifically highlighted the rapid development and
evaluation of WHO measurement standards undertaken by NIBSC with the
support of other collaborating centres and laboratories worldwide, as well as
the key contributions made by WHO collaborating centres to the production of
WHO written standards. Dr Knezevic noted that NIBSC in the United Kingdom,
the National Institutes for Food and Drug Control in China and the Paul-Ehrlich-
Institut in Germany would complete their re-designations as WHO collaborating
centres in 2021.
Dr Knezevic continued by briefly reviewing the current status of
WHO EUL of COVID-19 vaccines (see section 2.1.3 below), reiterating that
this clearly emphasized the importance of biological standards in the WHO
prequalification process. In the context of the therapeutics pillar of the ACT
Accelerator, the WHO Therapeutics and COVID-19 living guideline provides
conditional recommendations on the use of the neutralizing mAbs casirivimab
and imdevimab, as well as recommendations for or against the use of a range
of other proposed therapeutic products. WHO had also facilitated a number
of interactions among COVID-19 stakeholders, including product developers,
regulatory groups and networks, and national regulators. In several cases, such
activities had provided direct opportunities to promote the appropriate use of
WHO biological standards.
Dr Knezevic then briefly summarized the current status of regulatory
systems in different jurisdictions, noting that 28% of countries had now met
the World Health Assembly resolution WHA67.20 goal of having a stable, well-
functioning and integrated regulatory system as assessed by the WHO Global
Benchmarking Tool. Dr Knezevic concluded by reflecting on the evolving
scientific and regulatory challenges now being faced, and on the importance of
international cooperation in ensuring the safety, quality and efficacy of locally
used medical products, making best use of available resources and expertise,
avoiding duplication, and concentrating regulatory efforts and resources where
they were most needed.
which was now scheduled for submission to the Committee in April 2022. In
addition, in order to ensure consistency across the range of related documents,
WHO guidance on biosimilar mAbs and the WHO Questions and Answers
document on biosimilars would also be revised in 2022 for submission to the
Committee in 2022–2023. Other new or revised WHO written standards likely
to be submitted to the Committee from 2022 onwards included: (a) revised
WHO Recommendations for oral poliomyelitis vaccines (OPVs) and other
revised WHO guidance on poliomyelitis vaccine production and evaluation; (b)
revised WHO Guidelines on dengue vaccines and on rotavirus vaccines; and
(c) revised WHO Recommendations for the preparation, characterization and
establishment of international and other biological reference standards. Requests
had also been received from external stakeholders for the revision of the current
WHO written standards on post-approval changes to vaccines and on pandemic
6
General
influenza preparedness, and of the WHO global model regulatory framework for
medical devices including in vitro diagnostics (IVDs). Work had also started on
developing WHO guidance on the standardization of enteric vaccines and would
continue during 2022.
Dr Knezevic then turned to the WHO measurement standards
developed since 2013 – including the three standards established on the advice
of the Committee in 2020 to support the development of COVID-19 vaccines,
therapeutics and diagnostics. It was anticipated that use of the WHO COVID-19
antibody standards would facilitate the harmonized interpretation of clinical
study data by allowing antibody titres to be expressed in International Units (IU)
thereby supporting efforts to identify an immune correlate of protection defined
in IU/mL. The announcing of the establishment of the First WHO International
Standard for anti-SARS-CoV-2 immunoglobulin in the scientific press and its
subsequent rapid worldwide distribution highlighted its importance as a key tool
for vaccine developers, national reference laboratories and academic groups.
Among the other main outcomes of recent Committee meetings had been the
endorsement of a proposal to develop a WHO standard for SARS-CoV-2 antigen
to support the development, assessment and comparability of antigen-based
RDTs and an update on progress would be provided at the current meeting (see
section 8.1.3 below).
Going forward, WHO would continue to provide technical support
to users of its COVID-19 standards, for example through dissemination of its
Considerations for evaluation of COVID-19 vaccines document which provides
advice to manufacturers on both the process and criteria that will be used by
WHO to evaluate COVID-19 vaccines submitted either for prequalification or
EUL. Technical assistance will also be provided to users of WHO COVID-19
standards through COVAX, the vaccines pillar of the ACT Accelerator, and
through WHO participation in the recently established Technical Advisory
Group on COVID-19 Vaccine Composition.
Dr Knezevic concluded by outlining recent WHO efforts in the
standardization of CGTPs. In February 2020, WHO had initiated the
development of a white paper on the fundamental principles and issues related to
the regulatory oversight of CGTPs and an update on progress would be provided
at the current meeting (see sections 3.3.1 and 3.3.2 below). A survey had also
been conducted of CGTP regulation in jurisdictions with relevant marketing
authorization experience to better understand the current global regulatory
landscape and inform the development of future WHO guidance on these
advanced products. The survey results will be analyzed and published in 2022.
In addition to these preparatory steps, the Committee was reminded that in 2019
it had recommended the establishment of two WHO measurement standards
relating to the integration of lentiviral vectors, and had subsequently endorsed
7
WHO Expert Committee on Biological Standardization Seventy-fourth report
for example, that guiding principles for the evaluation of viral vectored vaccines
were already provided in the existing WHO Guidelines on the quality, safety and
efficacy of Ebola vaccines.
trials in 2023.
PDVAC has also continued to monitor the development of RSV
immunization products, with several candidate vaccines now in Phase III
trials, including maternal vaccines and mAbs intended to provide passive
immunization to young infants. WHO Preferred Product Characteristics for RSV
vaccines and mAbs were published in 2017 and 2021 respectively. Regulatory
submissions for the most advanced candidate mAbs are expected towards the
end of 2022, and an RSV-specific supplement to the upcoming WHO regulatory
considerations document on the nonclinical and clinical evaluation of mAbs for
infectious diseases (see section 2.1.1 above) is planned. To accelerate access to
these products in LMIC, WHO is raising awareness of the burden of RSV and of
prospective products for disease prevention, and is developing tools to facilitate
in-country decision-making.
10
General
toxicology data still anticipated and there was no WHO recommendation to use
this vaccine in pregnant women. No policy recommendations would be issued
until the vaccine had been granted WHO EUL.
In a joint session, SAGE and the Malaria Policy Advisory Group had
recommended a four-dose schedule of the RTS,S/AS01 malaria vaccine for
children from the age of 5 months in regions with moderate to high malaria
transmission, with the possibility of a five-dose schedule in areas with highly
seasonal malaria. These recommendations were based on the results of a pilot
implementation programme conducted in Ghana, Kenya and Malawi involving
more than 800 000 children. Malaria vaccine delivery through routine childhood
immunization programmes has the ability to reach vulnerable children not
protected by other interventions and in the pilot programme led to statistically
significant reductions in both severe hospitalized malaria cases and hospitalization
12
General
with malaria infection. There was also no evidence that safety signals noted
during Phase III trials had been caused by the vaccine.
SAGE also reinforced the importance of seasonal influenza vaccination,
particularly among the most vulnerable groups, with vaccination in both prior
and current seasons producing higher levels of protection than no or partial
vaccination. In addition, co-administration of inactivated seasonal influenza
vaccine with any WHO EUL COVID-19 vaccine would be acceptable and
expected to maximize the uptake of both vaccines. SAGE also stressed the need
to improve access to hepatitis E vaccine. Although a subunit vaccine has been
available since 2015, it is not widely used and manufacturers were urged to apply
for WHO prequalification. To help address the current lack of data to support
its use, WHO had been urged to work with Gavi, the Vaccine Alliance, on the
inclusion of hepatitis E vaccination for outbreak response, and on hepatitis E
diagnosis and surveillance in Gavi-eligible countries.
During discussion, it was clarified that the data on the impact of prior
vaccination with seasonal influenza vaccine on subsequent levels of protection
had been derived from a systematic review and were based on vaccinations in
consecutive years. Reflecting on the low incidence of poliovirus infections, the
Committee also asked whether bivalent vaccination campaigns were continuing
and was informed that the majority of countries still used bivalent OPV in their
routine schedule, with IPV only recommended for very low risk countries.
The Committee then enquired about the issues to be discussed at
upcoming SAGE sessions and was informed that a decision-making framework
for the implementation of booster doses of COVID-19 vaccines would shortly be
proposed. This was a complex issue as decisions are ultimately made in-country
and are vaccine specific. As it would be premature to issue vaccine-by-vaccine
recommendations at present, the decision-making framework was being put
forward. Discussion then moved on to the possibility of SAGE recommendations
being issued on heterologous boosting and childhood immunization in the
context of COVID-19 vaccine supply issues. The Committee was informed that
SAGE was undertaking a systematic review of the available data and it now looked
likely that it would recommend a more flexible approach to the use of different
booster vaccines in due course. Although children were at low risk of severe
disease, SAGE was considering a recommendation to immunize adolescents
based on the burden of disease and on the potential to reduce transmission as
vaccine supplies increased.
in July 2021. This advisory group had been established to provide technical
advice and recommendations to WHO in the fields of blood regulation and
transfusion medicine. As highlighted by the adoption of several World Health
Assembly resolutions, there is a global need to ensure the safety and availability of
blood products. Furthermore, the development of the WHO Action framework
to advance universal access to safe, effective and quality-assured blood products
2020–2023 now provides a global strategic direction. In January 2020, the merging
of the WHO blood products and blood safety and transplantation teams resulted
in the combined and strengthened WHO Blood and other Products of Human
Origin (BTT) team, and an associated need for wide-ranging expert guidance from
experts in the field. The new advisory group would reflect the need for a suitable
diversity of expertise and experience, while also ensuring representation from all
six WHO regions. Currently comprising 25 members serving in their individual
capacities, the functions of AG-BRAS were: (a) to advise on the development
of WHO norms, standards and guidelines for ensuring the safety, quality and
availability of blood products; (b) to advise on scaling up the implementation
of WHO policies and strategies, and strengthening national systems for blood
supply and regulation; and (c) to provide scientific assessments of current and
emerging threats to the safety and availability of blood and blood products. To
date, two virtual meetings of the advisory group had been conducted, with a third
scheduled in late 2021 to develop its workplan.
Dr Maryuningsih then summarized the outcomes of a recent workshop
on blood and blood products held as part of a virtual meeting of the International
Conference of Drug Regulatory Authorities (ICDRA). Meeting recommendations
to countries included a call for ministries of health to provide effective
leadership and governance in the development of national blood regulation
systems. Countries were also urged to use the WHO Global Benchmarking
Tool Plus Blood (GBT Plus Blood) to identify gaps and needs in national blood
WHO Technical Report Series, No. 1039, 2022
regulation, with training on the use of this tool having been conducted in nine
countries. ICDRA also recommended that national governance mechanisms are
established and implemented to support collaboration among regulators, blood
establishments and hospitals in developing and improving well-functioning
blood systems. ICDRA recommendations for WHO included: (a) ensuring the
provision of support to countries in developing and strengthening their national
blood regulatory systems as a WHO Action framework priority; (b) providing
periodical reports on the progress achieved in implementing the framework;
and (c) supporting the implementation of WHO guidance and blood policies at
country level, particularly in LMIC.
Dr Maryuningsih then provided an update on the current status of several
WHO documents developed in 2021 that were intended to facilitate achievement
of the high-level strategic objectives set out in the WHO Action framework. A
14
General
policy brief on the urgent need to implement patient blood management (see
section 3.2.2 below) and educational modules on clinical use of blood (see section
3.2.1 below) had both been completed with publication expected by the end of
2021. In addition, planning clearance had been obtained for a WHO guidance
document on screening donated blood for transfusion-transmissible infectious
agents, and for tools for the stepwise implementation of a haemovigilance
system, with both resources now under development. WHO had also organized
or contributed to a series of webinars to promote the use of recently published
WHO guidance in this area, and had held a training workshop on the use of the
WHO GBT Plus Blood tool. Dr Maryuningsih noted that consideration would
also need to be given to updating the WHO interim guidance on maintaining
a safe and adequate blood supply and collecting convalescent plasma in the
context of the COVID-19 pandemic. Dr Maryuningsih concluded by reporting
on the eighth meeting of the WHO network of collaborating centres for blood
products and in vitro diagnostics held in September 2021 at which the relevant
WHO measurement standards to be considered by the Committee at the current
meeting had been discussed.
The Committee sought clarification of the envisaged role of AG-BRAS,
particularly with regard to the existing WHO collaborating centre process for
proposing and developing WHO measurement standards for blood products for
review by the Committee. Clarification was provided that AG-BRAS would focus
on WHO written standards rather than measurement standards, and that WHO
would continue to rely on its network of collaborating centres for the latter.
Further clarification was given that in addition to the need for technical advice and
recommendations on both blood regulation and transfusion medicine following
the formation of the BTT team, WHO norms and regulations governing WHO-
managed networks and engagement with expert bodies also required balanced
geographical and other representation in its advisory mechanisms. It was noted
that several members of the long-established WHO Blood Regulators Network
were also now members of AG-BRAS, and it was expected that the establishment
of AG-BRAS would help rationalize all of the various advisory mechanisms in
this field that WHO had relied on in the past.
The Committee acknowledged the many past contributions made by
the WHO Blood Regulators Network in strengthening harmonization and
standardization efforts in blood product safety and quality, expressed its support
for the new advisory group going forward and looked forward to its contributions
in furthering progress in this area.
immunoglobulin G (hyper IgG) had been used early in the COVID-19 pandemic
based on the reported efficacy of convalescent plasma used for diseases such
as severe acute respiratory syndrome in 2003, A(H1N1) influenza in 2009, and
Middle East respiratory syndrome in 2012. However, definitive evidence of the
effectiveness of this approach for COVID-19 was not available. In addition, drugs
shown to be effective in initiating recovery from COVID-19 (such as tocilizumab
and dexamethasone) were not specifically directed towards clearing the virus.
Dr So-Osman outlined the three product types currently available – namely
CCP, hyper IgG and mAbs. CCP can be prepared very quickly once convalescent
donors are available, while hyper IgG and mAbs both take significantly longer
to be produced. In addition, the relatively high costs of producing hyper IgG
and mAbs would likely constrain their availability. The Committee was informed
that Cochrane living systematic reviews were currently in progress for all three
products with the aim of searching the literature for definitive evidence of their
effectiveness.
The first Cochrane living systematic review on the use of CCP or hyper
IgG to treat COVID-19 had been published in May 2020, with a subsequent
review published in July 2020. Both reviews found no difference in primary
outcome (effectiveness) among patients who received or did not receive CCP.
A more recent review had been published in May 2021 based on 13 studies
involving a total of 48 509 participants (including the large RECOVERY study
with more than 11 000 patients). These studies had mostly been carried out in
hospitals, primarily among patients with moderate to severe COVID-19. It was
firmly concluded that CCP conferred no benefit in treating such patients. It was
however unclear whether CCP was of any benefit in treating those with mild or
asymptomatic disease. With 130 studies still ongoing, unpublished or recently
published, these living systematic reviews would continue.
Dr So-Osman reflected on why CCP appeared not to work in these
WHO Technical Report Series, No. 1039, 2022
studies and highlighted the Dutch CONCOVID study which had shown no
difference in viral clearance between hospitalized COVID-19 patients provided
with standard care and those treated with CCP. However, many of the patients
had developed antibodies by day 7 of infection and it was hypothesized that
CCP might be effective if transfused earlier and at a high titre. The results of
studies involving the use of CCP within 7 days of disease onset, and its use in
immunocompromised seronegative patients, had now been published or were
being finalized and would be included in the next Cochrane living systematic
review update. In February 2021, the US Food and Drug Administration (FDA)
issued a Letter of Authorization which stipulated that only early and high-titre
CCP treatments could be used.
Future Cochrane initiatives would evaluate the use of CCP and hyper
IgG to prevent SARS-CoV-2 infection and the use of hyper IgG in treating
16
General
COVID-19. The first review of mAb use in COVID-19 patients had been
published in September 2021 and found insufficient evidence to support the use
of this approach in the overall patient population – though it may have a role to
play in specific patient subgroups.
The Committee observed that there were some indications that the very
early use of high-titre CCP in people at high risk of severe disease may lead to a
less severe disease course. Dr So-Osman noted that of four reported studies on
the use of CCP during the pre-hospitalization stage and during early symptomatic
infection, only one had shown any benefit, with the remaining studies reporting
no difference in outcome. Dr So-Osman further clarified that the Cochrane
reviews had primarily involved randomized control trials which allowed for
specific patient subgroups (for example, immunocompromised patients) to
be evaluated and the results aggregated to improve the available data. A Phase
III randomized control study of CCP had recently been concluded that could
potentially also contribute to the subgroup data. The Committee queried whether
it was possible to identify the specific level of antibody required for treatment
efficacy and was referred to the CONCOR-1 Study in which patients had been
divided into groups receiving either high, medium or low antibody titre CCP,
and which had shown no evidence of benefit. The Committee concluded that
the use of CCP was a developing field and with many studies still ongoing, firm
conclusions on its efficacy in different patient subgroups were yet to be reached.
regional standards would have to take into account differences in snake species,
but agreed that the approach taken should focus on the venom rather than the
snake itself. The use of a well-characterized pool of venom as the immunogen
would help to address the inherent variations.
Center for Biologics Evaluation and Research (CBER), Silver Springs, MD USA
Dr Celia Witten reviewed the standardization activities of CBER during the
last year. CBER COVID-19-related activities had included participation in the
collaborative studies to establish WHO international standards both for nucleic
acid amplification technique (NAT)-based assays and serological assays. In the
context of COVID-19 vaccine development, CBER had participated in WHO
technical working groups on viruses, reagents and immunoassays, and animal
models, with support also provided to WHO workshops on the standardization
of clinical trials and their methodologies. Furthermore, CBER would also
be participating in the forthcoming collaborative studies to establish the First
WHO International Reference Panel for antibodies to SARS-CoV-2 variants of
concern and the Second WHO International Standard for anti-SARS-CoV-2
immunoglobulin (see sections 8.1.1 and 8.1.2 below).
In other areas, CBER had seen an increase in requests for the reference
viruses used to validate high-throughput sequencing as an alternative to in vivo
adventitious virus detection. Replacements stocks were now being developed and
two more viruses (coronavirus and parvovirus) will be included in an expanded
panel, with the results of a collaborative bridging study to be presented to the
Committee for its consideration in due course. Dr Witten also updated the
Committee on a project it had endorsed in 2018 to develop universal reagents
for the potency testing of IPV based on human and mouse mAbs specific for the
D-antigen. These mAbs and other reagents had now been assessed in preliminary
studies and sent to NIBSC for filling so that a collaborative study could start in
late 2021.
With regard to the current meeting, CBER had participated in the
collaborative studies on the First WHO International Standard for anti-Lassa virus
immunoglobulin G (see section 6.1.3 below) and the Third WHO International
Standard for von Willebrand factor (concentrate) (see section 5.1.1 below)
and had been involved in the drafting of the WHO regulatory considerations
document on mRNA vaccines (see section 3.4.2 below). In addition, CBER
was participating in the ongoing revision of the WHO Recommendations for
19
WHO Expert Committee on Biological Standardization Seventy-fourth report
five virus strains, along with a standard operating procedure and reagents, had
been distributed to 31 participants worldwide to evaluate the transferability and
robustness of the method. The results of the study were expected to be reported
to the Committee in 2022. A second project with direct implications for the 3Rs
principles was an initiative to eliminate animal-based pyrogen testing from the
European Pharmacopoeia (Ph. Eur.). Despite changes to relevant texts aimed
at encouraging users to perform in vitro tests (such as the monocyte activation
test) the rabbit pyrogen test continued to be widely used. Dr Mallet set out a
timeline for the complete removal of the rabbit pyrogen test from the Ph. Eur. by
2026 as an essential step towards the exclusive use of in vitro tests for the control
of pyrogens. The current animal-based tests would be replaced by risk-based
analysis of the potential presence of non-endotoxin pyrogens in the product or
intermediate being tested. If the presence of non-endotoxin pyrogens could be
20
General
ruled out in this way, then the revised Ph. Eur. will recommend the use of the
bacterial endotoxin test alone; otherwise, the monocyte activation test will be
recommended, either on its own or in addition to the bacterial endotoxin test.
The Committee acknowledged the progress made towards the removal
of the rabbit pyrogen test from the Ph. Eur. and strongly supported moves to
establish the monocyte activation test as an alternative to animal testing for
pyrogens worldwide. Noting that the use of fresh human blood in the test
remained a challenge, it highlighted the need for research into the development of
alternatives based on the use of suitable cell lines and the application of reporter
gene technology. Regarding the development of a quantitative sandwich ELISA
as an alternative to the in vivo potency test for rabies vaccines, the Committee
noted that essential reagents for the assay were available commercially and their
supply was assured. The Committee further noted that ensuring the ability of the
replacement assay to detect subpotent vaccine batches would be critical.
23
3. International Recommendations, Guidelines and
other matters related to the manufacture, quality
control and evaluation of biological products
3.1 General
3.1.1 Assessment of stability in international collaborative studies
The Committee was presented with an overview of the key issues in assessing
the stability of WHO international reference standards during their development
and evaluation in international collaborative studies. The biological activity of
most such standards was designated in arbitrary “International Units” (IU) and
their degradation cannot therefore be measured directly as by definition they are
the primary standard for the respective biological activity. Although biological
standards were often highly stable lyophilized preparations, it was nevertheless
important to evaluate their predicted degradation, typically using data from
accelerated thermal degradation studies. In this approach, the potency of test
samples stored at various temperatures between 4–56 °C is assayed at several time
points, usually over a period of between 6 months and 2 years. The potency of
these samples relative to that of baseline samples stored at −20 °C is then used
to predict the loss in activity expected at −20 °C using the Arrhenius equation.
To date, a predicted potency loss of less than 0.1% per year has been considered
acceptable.
There are, however, a number of known shortcomings in the use of the
Arrhenius equation to predict the rate of loss of biological activity. Although
many reference materials appear to exhibit Arrhenius-like behaviour over a
limited range of temperatures, the relationship is approximate and may not be
sustained over a wider temperature range. Estimates obtained by this method can
also have poor precision, while highly stable materials may not lose potency even
WHO Technical Report Series, No. 1039, 2022
or there was no loss of activity at elevated temperatures. In other cases there may
be no appropriate model for stability prediction or insufficient time to complete
an accelerated thermal degradation study, especially when a standard is urgently
required. For real-time stability monitoring, options include monitoring the
potency of the standard over time relative to another appropriate preparation
and/or monitoring other parameters of the standard (such as physicochemical
characterization data). As the precise situation will vary for different standards, a
clear vision is required of the stability monitoring goals, potential outcomes, and
the assay and statistical tools to be used.
Noting that the vast majority of reference materials were stable and
relatively few turned out to be unsatisfactory, the Committee discussed the
key issues raised and the stability data expected in collaborative study reports.
Although monitoring the stability of reference materials in real time was
considered ideal, the comprehensive post-establishment stability monitoring
of WHO international standards was considered too resource intensive to be
achieved in practice. Furthermore, as the acceptable annual loss of potency
varied among standard preparations this should be considered on a case-by-
case basis rather than regarding 0.1% per year as a requirement for all reference
materials. Observing that the acceptable level of stability typically relates to the
intended use of the standard and to the precision of the assays involved, it was
agreed that a decision tree to inform the design of stability studies rather than
simply following the Arrhenius model should be included when the current
WHO Recommendations were revised. This would help project leaders to know
what the Committee expected in cases where accelerated thermal degradation
studies failed to predict a degradation rate or where the predicted degradation
rate was too high to assure stability. The Committee commented on the wealth of
experience now available on how the characteristics and formulation of different
reference materials affect their stability, suggesting the possibility of more-flexible
approaches to stability studies conducted as part of collaborative studies. It was
noted in this regard that the stability of reconstituted reference materials was
increasingly being assessed in collaborative studies. The Committee concluded
by highlighting the importance of feedback from users of reference materials in
identifying any emerging stability issues
global audience to help guide decisions and improve training and knowledge in
the field of transfusion medicine. It is intended that the modules will thus support
implementation of the WHO Action framework to advance universal access to
safe, effective and quality-assured blood products 2020–2023, and complement
the WHO Aide-mémoire for national health programmes on clinical use of blood
and other resources.
The Committee was informed that the module development process
had been a collaborative effort between WHO and the International Society of
Blood Transfusion (ISBT). Materials were developed by an international panel
of authors with broad expertise in transfusion medicine, with feedback provided
by a panel of critical readers drawn from countries worldwide and by the WHO/
ISBT steering committee. This process had ensured that critical inputs could
be invited on the modules to ensure their relevance across the diverse range of
settings in which they might be used.
A brief outline of the nine modules comprising the first tranche of
materials was provided and clarification given that these educational materials
were intended to provide simple and clear information that could be readily
grasped by a general audience, including students. As the aim was to provide an
introduction to the topic rather than an encyclopaedic text, the materials focus
on important principles, key points and practical application, and were illustrated
with clear diagrams and examples of clinical scenarios.
Organized as a series of modules that could be individually updated
as needed, the materials would be made available in both electronic and hard
copy formats, and would be accessible online. Publication of the first tranche of
modules was planned for the end of 2021 and would be followed by a series of
webinars hosted by ISBT in collaboration with WHO. The modules would be
promoted and disseminated by WHO and through ISBT and other policy and
professional networks. Based on the extensive use of the 2001 WHO document
WHO Technical Report Series, No. 1039, 2022
and the positive feedback received, a similarly wide uptake was anticipated for
the modules and their translation into multiple languages was anticipated. In
addition, a second tranche of modules was now in preparation and would likely
be finalized shortly. Topics in the second tranche would complement those in the
first tranche and would include the principles of appropriate use of blood and
blood products and patient blood management, major haemorrhage and massive
transfusion, trauma resuscitation, platelet transfusion, plasma transfusion,
adverse events and transfusion in under-resourced countries.
The Committee recognized the timeliness and importance of the
Educational modules as an update of the 2001 document. Given the global reach
of the earlier document, the Committee welcomed the steps taken to ensure
adequate representation of all regions in the authorship and review processes.
This included Latin America where the previous document, translated into
26
International Recommendations, Guidelines and other matters
Portuguese and Spanish, had been widely used. The Committee was assured that
representation from all regions would be ensured in both the finalization of the
first tranche of modules and the development and review of the second tranche.
A number of additional topics for inclusion were then suggested by the
Committee, including a separate module on the blood donor. In response, it was
clarified that references to blood donors were generally restricted to stewardship
and responsibility for the blood as the focus of the materials was on clinical
decision-making given the target audience. However, it was recognized that
the topic of walking donors overlaps both the donor and clinical settings and
could be considered in future updates. Other topics of current interest were also
highlighted including the use of convalescent plasma and advanced therapies, but
in such fast-moving fields requiring regular updating only the broader principles
and aspects could realistically be included in what was intended to become a
reference resource of established information and practices.
in the field. The updated document, scheduled for publication by the end of
2021, is intended to assist countries that require support in developing a national
screening programme to maximize the microbial safety of blood donations.
Guidance will be provided on the key infectious agents for which all donations
should be screened, the most appropriate methodology and screening targets,
and the systems required to support effective and reliable screening. The updated
guidance is also intended to assist national blood systems in advocating for the
political support and resources needed.
The Committee was provided with an overview of the key elements and
general guidance provided in the updated document. Key elements include critical
and screening-specific elements of infectious disease screening programmes,
while the general guidance covers important broader aspects of blood services
and their activities which help to support effective screening programmes. The
28
International Recommendations, Guidelines and other matters
Committee was informed that the overall aims of the update process had been
to ensure that the core guidance remained appropriate while also bringing up
to date the supporting evidence, background information and messages. In
addition, a number of core messages had been strengthened, and the terminology
made clearer and more precise. Developments in understanding and technology
had also been incorporated, along with guidance on the setting of acceptable
risk levels. Specific guidance was now provided on molecular screening and on
a number of “additional” infectious agents for which screening may be required.
During discussion, the Committee enquired whether mention had been
made of the work of the Committee and WHO collaborating centres in developing
assay calibrants and reference materials, and whether these were of relevance.
Confirmation was given that these were indeed relevant to the updated guidance
now provided on quality assurance and quality control. Although the work of the
Committee was not specifically mentioned in the text, reference had been made
to WHO programmes on ensuring quality assurance and quality control, and to
the provision of reference materials. A suggestion to add information, including
website links, on the relevant WHO reference materials now available to support
evaluation of assay performance and assay validation was accepted and would be
included in the final version of the document.
In light of the concerns raised about RDTs and their promotion and use
in many countries, the Committee enquired whether mention of these had been
made in the updated document. Clarification was given that although RDTs were
not directly advocated in the document, they can have a role to play in some
situations. The issue was not the RDTs per se but ensuring their proper evaluation
and validation for their intended use. The Committee emphasized the vital
importance of this issue and the generally poor understanding of the variability
in quality of such tests, citing the SARS-CoV-2 and HIV RDTs as examples. It
was acknowledged that the issues around RDT evaluation and validation were
considerable, and that a process of global evaluation and rapid dissemination of
the outcomes would significantly benefit blood services worldwide.
The Committee noted that the updated document focused primarily on
the screening of donations, and asked if pathogen inactivation might not be a
better approach, considering the threat of future emerging viruses. Clarification
was given that although pathogen inactivation had a role and was mentioned as
part of the overall structure in countries that use the approach, it would not be
of value to many countries until it could be applied effectively across all products
and the associated issues of reduced yield and high cost addressed. The review
group had felt that this evolving situation could not properly be addressed in
the updated document. In addition, advocating for its use might be unhelpful
and could detract from the key messages to be conveyed to the envisaged target
audience of the document.
29
WHO Expert Committee on Biological Standardization Seventy-fourth report
In 2020, a survey had been conducted to identify the priority needs in this
area, with responses received from 87 countries across all six WHO regions. The
provision of educational and training tools and modules in the context of WHO
technical support was identified as the top priority. As this involves a number
of technical and other challenges, including the need to avoid tools becoming
outdated or inaccessible, it had been decided that the ISBT Haemovigilance
website would act as the primary repository, with a smaller set of resources placed
in the WHO Notify Library. The Committee was presented with an overview of
the document structure and contents, which included a directory of the tools and
resources available on the ISBT website. A number of the templates and example
materials were available in English and French, with plans in place to include other
languages. A brief demonstration was then given of how users could navigate the
directory when looking for specific resources. In 2021, the document had been
30
International Recommendations, Guidelines and other matters
widely shared, with positive feedback and suggestions received. Any identified
gaps in the tools would now be addressed and a final draft document produced
and submitted for WHO clearance by the end of 2021.
The Committee commended the contribution the document would make
to the establishment and strengthening of haemovigilance systems, especially
in developing countries. One important area where guidance was needed was
the analysis of haemovigilance data and follow-up actions to be taken. It was a
common misperception in some countries that the simple provision of data to a
medical information system was equivalent to haemovigilance. The Committee
was informed that this issue had been observed among some survey respondents
at all levels of the haemovigilance system. As a result, this issue had been addressed
in the document which emphasizes that comprehensive haemovigilance was not
just about collecting data but also included data analysis and feedback to hospitals
and blood centres so that improvements could be made. With regard to the need
for ongoing maintenance of the online tools and resources, the Committee was
informed that discussions were taking place to identify the best approach.
The Committee enquired as to the reasons for such a broad target
audience, noting for example that patient groups are very different from blood
establishments and ministries of health. The Committee wondered if it might be
more effective to develop specific tools and resources for each group. In response,
it was highlighted that many countries lack strong national health authorities
and that even though the document was primarily intended for national systems
staff, stakeholders at all system levels had been included. In addition, it was
important to recognize that small initial steps could be taken in some countries
by individuals, some of whom may be involved in efforts to improve patient and
donor safety. The Committee noted that in some cases the Ministry of Health will
even delegate responsibility for the implementing of a haemovigilance system,
and expressed its agreement with the overall approach that had been taken.
for the target antigen, challenges in manufacturing and potential toxicity. Similarly,
the number of approved ex vivo retrovirus and lentivirus gene therapy products
was also increasing – with critical challenges here including the risk of integrational
mutagenesis or oncogenesis, and the need to determine optimal treatment timing. In
addition, adeno-associated viruses were now being widely used for the treatment of
monogenic diseases, with ongoing issues including the need to ensure persistence of
the desired effect while avoiding immune responses directed against the virus vector.
By 2020, more than 1200 ATMP clinical trials were ongoing, with rapid
progress being made through the clinical trial phases. As a result, there would
likely be numerous such products submitted for regulatory approval in the near
future. The Committee was reminded of the inherent and unique risks associated
with ATMPs due to their biological complexity and the current lack of long-term
safety data for the majority of approaches. Early risk identification and mitigation
approaches would therefore be key requirements going forward. In addition,
numerous manufacturing, quality control, testing and legislative issues continue
to impact on the clinical development of ATMPs. Among these, the generation of
clinical evidence to support licensure is particularly challenging with regard to the
choice of comparators, dose and end-points. In the case of severe conditions, many
therapies are potentially curative or ameliorating but no relevant standard of care
exists for comparison, while studies often involve “last-line” patients for whom
best supportive care is not attractive. In the case of orphan treatments, the patient
populations eligible for clinical trials were frequently small and heterogeneous.
After being provided with an overview of existing regulatory frameworks
and guidance documents for CGTPs in different jurisdictions, the Committee
discussed a number of specific regulatory aspects in this area. Among these, the
challenge of developing suitable animal-based and in vitro approaches to ensure
the safety of ATMPs prior to their use in humans was highlighted. In addition,
with regard to the evaluation of point-of-care products, the Committee noted
that it was difficult to control products in hospitals and this issue had yet to be
WHO Technical Report Series, No. 1039, 2022
adequately resolved by the major regulators. It was suggested that this and other
issues raised in this presentation, including the need for early risk identification
and mitigation, should be addressed in the WHO white paper now under
development (see section 3.3.2 below).
might simply rely on histological evaluation. The Committee was informed that
this possibility had been discussed by the working group but manufacturers still
felt that clinical signs provided useful information and there was insufficient data
to support the discarding of clinical scoring at present. Having encouraged the
development of high-throughput sequencing technologies to assess the safety of
OPV at its recent meetings, the Committee was also interested in the potential
application of this approach to yellow fever vaccine testing. Clarification was given
that although the implementation of this technology was considered highly likely
in future, there was currently insufficient data on the critical mutations or lot-to-
lot consistency of viral sequences to introduce this as a release test at present.
The Committee was then provided with an overview of a number of
issues addressed by the drafting group following feedback received during public
consultation. These included: (a) the need to be clear on whether testing was for
“neurotropism” or “neurovirulence”; (b) the feasibility of conducting randomized,
double-blind controlled tests in non-human primates; and (c) the potential benefit of
telemetry to make testing more objective. Acknowledging that “neurotropism” was
the term used throughout the current main text of the Recommendations, and that
only the appendix was to be amended, the Committee accepted the proposal to add
a definition of neurotropism to the beginning of the amendment. The Committee
did not feel that randomized, double-blind controlled tests were feasible with non-
human primates and this requirement should therefore not be added. Accepting a
manufacturer’s argument that the benefits of telemetry remain to be demonstrated,
the Committee agreed that the text referring to telemetry should be deleted but a
sentence should be inserted in small print encouraging manufacturers to explore
the possible use of telemetry to render assessment more objective.
The Committee noted that new reference preparations of yellow fever
vaccine virus strains were also now needed for use in non-human primate testing.
However, the development of such standards would be challenging and should
first be discussed with manufacturers and other stakeholders. The Committee
further noted that other yellow fever vaccine platforms were being developed and
that although no attempt had been made at the current time to review the 2010
WHO Recommendations in their entirety, an open stakeholder meeting might
usefully be organized to determine the need for such a revision.
The Committee then recommended that the document WHO/
BS/2021.2401 be adopted and annexed to its report (Annex 2).
3.4.2 Evaluation of the quality, safety and efficacy of messenger RNA vaccines
for the prevention of infectious diseases: regulatory considerations
Recent advances in the manufacturing and stabilization of mRNA have established
the approach as an important vaccine technology and the speed with which
candidate mRNA vaccines can be developed makes them eminently suitable for
35
WHO Expert Committee on Biological Standardization Seventy-fourth report
37
4. International reference materials –
biotherapeutics other than blood products
4.1 WHO international reference standards for
biotherapeutics other than blood products
4.1.1 Third WHO International Standard for follicle-stimulating
hormone (human, recombinant) for bioassay
Follicle-stimulating hormone (FSH) is a glycoprotein produced in the anterior
pituitary gland. It is involved in the regulation of follicular growth, pubertal
maturation and reproductive processes, working in synergy with luteinizing
hormone to regulate ovulation. FSH is used in fertility treatments such as in
vitro fertilization. Early FSH products, which were extracted from urine, were of
variable purity and have largely been replaced by recombinant products. Today,
a number of such human recombinant products are on the market including a
number of biosimilars. The potency of therapeutic FSH is typically determined
using an in vivo bioassay with the dose expressed in IU traceable to the WHO
international standard. Stocks of the Second WHO International Standard
for follicle-stimulating hormone (human, recombinant) for bioassay (NIBSC
code 08/282) were now critically low, prompting the production of a candidate
replacement material to ensure the continuity of the IU for FSH products.
A quantity of manufacturer-donated purified human recombinant FSH
was formulated and filled in 0.5mL aliquots into glass ampoules for evaluation of
its suitability as a replacement standard material. The candidate material (NIBSC
code 20/218) was evaluated in an international collaborative study involving
six laboratories in six different countries. Using a method provided in the study
protocol, each laboratory carried out independent in vivo bioassays to estimate
the content of the candidate material 20/218 relative to the current WHO
WHO Technical Report Series, No. 1039, 2022
performed the Steelman-Pohley assay as described in Ph. Eur., with the exception
of one laboratory which made a minor adjustment to the sample diluent. The
Committee considered the report of the study (WHO/BS/2021.2410) and
recommended that the candidate material 20/218 be established as the Third
WHO International Standard for follicle-stimulating hormone (human,
recombinant) for bioassay, with an assigned unitage of 137 IU/ampoule. The
Committee requested that the report of the study be revised to reflect the minor
assay adjustment made by one of the laboratories and this was duly done (WHO/
BS/2021.2410 rev.1).
40
5. International reference materials – blood
products and related substances
5.1 WHO international reference standards for
blood products and related substances
5.1.1 Third WHO International Standard for von
Willebrand factor (concentrate)
Von Willebrand disease is the most frequent inherited bleeding disorder and
is caused by a deficiency and/or abnormality of von Willebrand factor (VWF).
VWF is essential for platelet subendothelial adhesion and platelet-to-platelet
interactions, and is a specific carrier of blood coagulation factor VIII in plasma.
Where transfusion therapy is necessary, disease treatment relies upon the use of
purified VWF concentrates. The WHO international standard for VWF is used for
the potency estimation and labelling of these therapeutic products. The current
Second WHO International Standard for von Willebrand factor (concentrate)
(NIBSC code 09/182) was in high demand and it was estimated that stocks would
be exhausted by mid-2022.
Candidate replacement materials had therefore been prepared using
clinical grade VWF purified from two plasma-derived VWF concentrates
(NIBSC codes 18/248 and 08/296) and one recombinant VWF product (NIBSC
code 20/156). An international collaborative study involving 48 laboratories
in 22 countries had been conducted to assign potency values for VWF:antigen
(VWF:Ag), VWF:ristocetin cofactor (VWF:RCo) and VWF:collagen binding
(VWF:CB) to the three candidate materials. Values were calculated relative to
the current WHO international standard and to the Sixth WHO International
Standard for factor VIII/VWF (plasma) to ensure continuity of the IU and confirm
the equivalence of the IU values applied to each type of standard (concentrate
and plasma). Study participants also investigated the possibility of assigning
values to the replacement standard for VWF:glycoprotein IbR (VWF:GPIbR)
and VWF:glycoprotein IbM (VWF:GPIbM) to reflect the development of newer
immunological assays based on VWF binding to immobilized recombinant GPIb
receptors. Due to the rapid uptake of these newer assays and the resulting urgent
need for international standardization, the Sixth WHO International Standard
for factor VIII/VWF (plasma) had also been established in 2018 as the WHO
International Reference Reagent for GPIbR and GPIbM methods.
The study results showed no significant variability in overall mean potency
estimates for VWF:Ag and VWF:RCo for all three candidate materials when
measured relative to either the current WHO international standard for concentrate
or the current WHO international standard for plasma. Greater variability was
observed for VWF:CB and was associated with the different methods and different
41
WHO Expert Committee on Biological Standardization Seventy-fourth report
collagen types used. Only candidate material 18/248 showed no significant method
differences for estimates relative to the current international standard, highlighting
the importance of the “like versus like” principle of biological standardization.
Estimates calculated relative to the WHO international standard for concentrate
were observed to be less variable than estimates calculated relative to the WHO
international standard for plasma – supporting the use of a separate international
standard for each material. Candidate material 18/248 was also associated with
the lowest inter-laboratory variability for all three analytes. Mean estimates for
GPIbR and GPIbM were also calculated for the candidate materials relative to
the current WHO international standards. Candidate material 18/248 was
again associated with low inter-laboratory variability and provided the greatest
equivalence between the various estimates made.
An accelerated thermal degradation study carried out to evaluate the
stability of the candidate material 198/248 showed little loss of potency after 3
months. Real-time stability studies of the current WHO international standard
for VWF, concentrate has demonstrated no measurable loss of activity even after
12 years of storage at 20 °C. No stability issues were therefore anticipated and
ongoing monitoring would be conducted. Stability testing of the reconstituted
material indicated that it remained stable when stored on ice for 4 hours.
The Committee queried the short duration of the stability studies and the
resulting limited data and was informed that this had been due to an oversight
in placing the candidate materials into storage for studies after filling. However,
early indications from stability testing at 56 °C storage for 3 months were that
this would not be an issue – a conclusion supported by the extreme stability of
the current international standard during its lifetime. Nonetheless, the stability of
the materials would be checked again in 6 and 12 months.
The Committee questioned the inclusion of a recombinant product in the
study and were informed that licensed recombinant products were not available
WHO Technical Report Series, No. 1039, 2022
normal and low levels of ferritin were also obtained from the Welsh External
Quality Assurance Scheme for use in commutability studies. The sample potencies
obtained were reported by the laboratories according to the assay platform
calibrant, with these potencies then being expressed relative to the current WHO
international standard by NIBSC. It was observed that some laboratories still
claimed traceability of their results to the first and second WHO international
standards despite these not been available for almost 25 years.
Results indicated that inter-laboratory variability was reduced when the
current WHO international standard was used as a reference material. Variability
was also reduced when the results from two laboratories were excluded due to
invalid assays resulting from non-linearity and non-parallelism. All laboratories
correctly identified the commutability samples, though wider variation was
observed for results obtained for the sample with the low ferritin level as this
was below the limit of quantitation for most of the laboratory systems. Inter-
laboratory variability was lower when using either candidate reference material
compared to in-house standards.
Combined study results indicated overall mean potencies of 10.5 µg/
ampoule for candidate material 19/118 and 8.0 µg/ampoule for candidate
material 19/162 relative to the current international standard. Better overall
agreement was observed between all laboratories and between assay methods for
the potency of candidate material 19/118 compared to candidate material 19/162.
Accelerated degradation studies indicated no significant loss of activity in either
candidate material after a period of 2 years and it was anticipated that they would
remain stable under long-term storage at −20 °C.
The Committee considered the report of the study (WHO/BS/2021.2409)
and recommended that candidate material 19/118 be established as the Fourth
WHO International Standard for ferritin (human, recombinant) with an assigned
content of 10.5 µg/ampoule and expanded uncertainty limits of 10.2–10.8 µg/
WHO Technical Report Series, No. 1039, 2022
46
6. International reference materials – in vitro diagnostics
6.1 WHO international reference standards for in vitro diagnostics
6.1.1 First WHO International Standard for Mycobacterium
tuberculosis (H37Rv) DNA for NAT-based assays
Tuberculosis (TB) remains a major cause of death worldwide and is a particular
public health concern in LMIC with inadequate diagnostic facilities. TB is a
respiratory disease caused by the bacterium Mycobacterium tuberculosis and
timely and accurate diagnosis is crucial for effective treatment and the prevention
of transmission. The field testing of TB diagnostics is an important element in
achieving the goal set by the international health community to end global TB
epidemics by 2030. Sputum-smear microscopy and culture techniques are effective
in diagnosing highly infectious TB but less so for the early diagnosis of infection in
people with less-pronounced symptoms, with culture methods in particular having
long turnaround times. More-sensitive RDTs based on nucleic acid amplification
techniques (NATs) are now available with other new tests in development.
The Committee was reminded that experts at a 2018 WHO workshop
had discussed advances in TB diagnostics and had identified the need for a WHO
international standard for M. tuberculosis DNA to serve as the primary calibrator
for NAT-based assays. The use of the international standard would improve
the harmonization of such assays and allow different laboratories to compare
analytical data using different assay formats. At its meeting in October 2018, the
Committee had endorsed a proposal to develop a WHO international standard
based on the commonly used laboratory strain H37Rv.
A single batch of 2992 vials of lyophilized, heat-inactivated H37Rv with
a concentration of approximately 106 genome copies per mL had therefore been
prepared. Preliminary studies confirmed the consistency of filling and effectiveness
of the heat inactivation process. This candidate material (NIBSC code 20/152)
was then evaluated for its suitability to serve as a WHO international standard for
both quantitative PCR assays and RDTs in an international collaborative study
involving eight laboratories in seven countries. It was concluded that the use of
candidate material 20/152 in quantitative PCR assays reduced inter-laboratory
variability. In addition, serial dilution of the material allowed for estimation of the
end-point titres/limit of detection of various RDT kits. There was no observable
loss of genome copies when the vials were stored at elevated temperatures up to
56 °C for 12 months, while the reconstituted material was stable for up to 4 weeks
at −20 °C and up to 1 week at 4 °C.
The Committee commended the study of this necessary standard and
reflected on the difficulty of recruiting diagnostic laboratories that were currently
heavily committed to the COVID-19 response – an issue for recent collaborative
studies in other areas. The Committee did not support a suggestion that “DNA” be
47
WHO Expert Committee on Biological Standardization Seventy-fourth report
omitted from the name of the standard as this would be inconsistent with similar
diagnostic standards. Instead, any potential clarification required of the precise
nature of the reference material should be addressed in the instructions for use
(IFU). However, it did agree that the strain designation should be included in the
name of the standard. Clarification was then given that one of the main intended
uses of the standard would be to evaluate and monitor the fitness for purpose of
RDTs and the Committee noted the importance of such standards in the WHO
prequalification of IVDs.
The Committee considered the report of the study (WHO/BS/2021.2403)
and recommended that the candidate material 20/152 be established as the First
WHO International Standard for Mycobacterium tuberculosis (H37Rv) DNA for
NAT-based assays with an assigned unitage of 6.3 log10 IU/vial.
v-Oka strains), as well as four clinical samples and two spiked samples to permit
a preliminary assessment of commutability. Agreement on the mean potency
estimates reported across laboratories for both qualitative and quantitative assays
significantly improved when results were expressed relative to the candidate
material. In addition, accelerated thermal degradation study data obtained at 12
months and 18 months post-production indicated that the candidate material was
stable and suitable for long-term storage. Nucleotide sequencing data revealed
a pattern of single nucleotide polymorphisms consistent with Clade 3, with no
major genome rearrangements.
Noting the reported higher variability of results relative to the v-Oka
comparator strain, the Committee enquired whether the other samples in
the collaborative study panel had been sequenced. It was informed that work
was ongoing and a wider commutability study would allow the effect of clade
differences to be assessed. However, the higher variability observed might also
have been due to the small number of study participants. The Committee noted
in particular the small number of laboratories that had contributed qualitative
assay data and, despite assurance that the results indicated that the candidate
material had harmonized results, it was suggested that further studies be carried
out post-establishment.
The Committee considered the report of the study (WHO/BS/2021.2405)
and recommended that the candidate material 19/164 be established as the First
WHO International Standard for varicella zoster virus DNA for NAT-based
assays with an assigned unitage of 7.0 log10 IU/vial.
neutralizing and binding assays, though slightly less effectively than the candidate
material 20/202. The stability of the lyophilized plasma pool 20/202 was assessed
in an accelerated thermal degradation study. Relative to a baseline sample stored
at −20 °C, there was minimal loss of potency for up to 1 month at 37 °C and up
to 6 months at ambient temperature (20 °C). Using the Arrhenius equation, the
predicted loss of potency for candidate material 20/202 was estimated at 0.17%
per year when stored at −20 °C.
The Committee queried whether the serum panel samples obtained from
Nigeria and Sierra Leone were sufficiently representative of the broad range of
LASV lineages. It was agreed that other samples from different sources might
usefully be added to the panel in future to improve its representativeness. Reflecting
on the challenge of producing reference materials for emerging pathogens, it was
suggested that further studies into the generation of suitable reference materials
50
International reference materials – in vitro diagnostics
using mAbs might be worthwhile. A key issue to be addressed would the current
focus placed on mAb development in the context of mechanisms of protection
against infectious disease, which would not necessarily reflect the requirements
of diagnostic assays.
The Committee considered the report of the study (WHO/BS/2021.2406)
and recommended that the candidate material 20/202 be established as the First
WHO International Standard for anti-Lassa virus immunoglobulin G with a
unitage of 25 IU/ampoule for neutralizing antibody, 250 IU/ampoule for anti-GP
binding IgG and 250 IU/ampoule for anti-NP binding IgG. The Committee also
recommended that the proposed panel consisting of candidate materials 20/204,
20/222, 20/226, 20/228, 20/244, 20/246 and 20/248 be established as the First
WHO International Reference Panel for anti-Lassa virus immunoglobulin G
without an assigned unitage.
value of 533 IU/ml and a robust mean value of 555 IU/ml. Using a difference-
in-bias approach, the commutability of both materials was demonstrated for all
laboratory methods used in the study. The stability of candidate material 19/260
was assessed in a thermal degradation study over a period of 7 months. Based
on the Arrhenius equation, the annual loss of activity when stored at −20 °C was
predicted to be approximately 0.013%.
It was noted that as autoantibodies were inevitably donor specific, any
replacement standard obtained from different donors would differ from the
previous reference reagent. Accepting that this was an insurmountable issue, the
Committee was content that candidate material 19/260 behaved similarly to the
current reference reagent 66/387 and would harmonize the results of immunoassays.
The Committee considered the report of the study (WHO/BS/2021.2404) and
recommended that the candidate material 19/260 be established as the First WHO
International Standard for anti-thyroid peroxidase antibodies with an assigned
unitage of 555 IU/ampoule. The Committee further recommended that the IFU
should make clear that the replacement material represented a different population
of autoantibodies than the previous reference reagent 66/387.
be made available; (b) the IFU should clearly highlight to users the potential
lack of commutability; (c) stakeholders should reconsider the appropriateness
of quantitative anti-rubella measurement and the use of the 10 IU/mL value as
a cut-off when assessing immune protection; and (d) the use of high-specificity
qualitative assays should be considered as an alternative to antibody quantitation.
The Committee was reminded that stocks of the current WHO
international standard were now limited and likely to be exhausted within
2 years. As the use of this measurement standard had changed over time, the
Committee was asked for its advice on the best approach to its replacement
and whether a single material could realistically harmonize both functional
and binding assays, as well as meet the needs of both vaccine manufacturers
and diagnostic kit manufacturers. A proposal was made to use a panel of anti-
rubella immunoglobulin samples covering a range of antibody concentrations
56
International reference materials – in vitro diagnostics
and calibrated against the current WHO international standard, and a potential
collaborative study design was outlined that would assess usage in different
assays and possibly allow for differentiation between vaccine development and
diagnostic applications.
The Committee agreed that the failure of the current international
standard to harmonize modern diagnostic tests resulted from the lack of a
clearly defined measurand or measuring system. Consistent with its previous
discussions on this issue, the Committee discouraged the quantitative use of a
standard to assign units as this fails to harmonize assays and therefore serves no
purpose. It concluded that only qualitative assays should be used for the diagnosis
of previous infection or vaccination, and that a panel without assigned unitage
would facilitate assay development. Commenting that the rubella field was very
active, the Committee encouraged the involvement of stakeholders in decisions
regarding the development and use of the proposed reference panel.
57
7. International reference materials – standards for
use in high-throughput sequencing technologies
7.1 Proposed new projects and updates – standards for
use in high-throughput sequencing technologies
7.1.1 Proposed test protocol and WHO international
reference reagents for whole-genome sequencing in
the routine lot release of OPV and control of sIPV
Polio eradication remains a top priority for WHO and will depend upon a
continuous supply of safe and effective vaccines. A number of live attenuated
OPV and Sabin IPV (sIPV) products have now been approved for use in many
countries. In addition, following polio eradication, a significant number of
additional manufacturers are expected to begin sIPV production to meet an
increased demand for such vaccines in the near future, with demand for novel
OPVs based on genetically stabilized viruses also anticipated.
Genetic stability is a crucial consideration in ensuring the safety of
vaccines. The Sabin strains currently used to manufacture OPV accumulate
reversions during replication in humans and cell cultures that potentially increase
their virulence. Traditionally, the genetic stability of OPV was assessed using
in vivo neurovirulence tests in monkeys or in transgenic mice expressing the
human poliovirus receptor. Since 2002, the genotypic stability of such vaccines
has also been assessed using the in vitro mutant analysis by polymerase chain
reaction and restriction enzyme cleavage (MAPREC) test, which measures the
proportion of revertant mutations present in the 5′-UTR of the viral genome.
However, MAPREC is technically challenging, requires radioisotopes and only
measures one mutation in the entire genome with implications for the evaluation
of other mutations affecting viral properties.
WHO Technical Report Series, No. 1039, 2022
procedures and reference reagents for the lot release of OPV or sIPV. A series
of consistency lots of monovalent OPV will be analyzed by whole-genome
high-throughput sequencing and the results compared to those obtained in
neurovirulence testing using non-human primates or transgenic mice. The
study, sponsored by PATH, will again be jointly coordinated by NIBSC and FDA.
Several OPV manufacturers had agreed to provide vaccine lots for use in the
study and as prospective reference materials, and a statistical method was being
developed for data analysis.
The Committee discussed the relative sensitivities of the MAPREC and
high-throughput sequencing methods, noting that both were sufficiently sensitive
to detect < 1% mutations in the 5′-UTR – a level of mutation considered to be safe.
Reflecting on the limited sequence variation observed between vaccine lots, and
noting that many mutations arising in vaccine strains are well characterized, the
Committee envisaged that neurovirulence testing may eventually not be required.
Enquiring about the affordability of high-throughput sequencing for vaccine
developers and control laboratories in LMIC, the Committee was assured that
today the approach was technically straightforward, inexpensive and accessible.
Part of this proposed project would be to support manufacturers adopting this
approach through workshops and training sessions. The Committee commended
the progress that had been made and, acknowledging the importance of
implementing modern molecular methods for lot release and the considerable
benefits that would result from the elimination of in vivo neurovirulence testing,
it endorsed the proposal (WHO/BS/2021.2411) to develop a test protocol and
WHO international reference reagents for whole-genome sequencing in the
routine lot release of OPV and control of sIPV.
59
8. International reference materials – standards
for use in public health emergencies
8.1 Proposed new projects and updates – standards
for use in public health emergencies
8.1.1 Proposed First WHO International Reference Panel for
antibodies to SARS-CoV-2 variants of concern
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological
agent of the ongoing COVID-19 pandemic. At its meeting in December
2020, the Committee recommended the establishment of the First WHO
International Standard for anti-SARS-CoV-2 immunoglobulin and the First
WHO International Reference Panel for anti-SARS-CoV-2 immunoglobulin.
These reference materials are intended to facilitate COVID-19 vaccines and
therapeutics development through the harmonization of serological assay data
worldwide. However, in late 2020, SARS-CoV-2 “variants of concern” (VOCs)
began to emerge with mutations that rendered them more transmissible. The
impact of such variants on the effectiveness of vaccines and therapeutics now
requires continual evaluation.
It was proposed that a WHO international reference panel consisting of
CCP or serum from patients infected with VOCs be developed to facilitate the
development of serological assays for VOCs in order to support the continuing
development of vaccines and therapeutic strategies. Following collaboration
between WHO, CEPI and NIBSC, three donations of CCP or serum from
individuals each infected with a VOC had now been offered by a number of
sources. The sourcing and receipt of all required donations was now ongoing.
All candidate materials would be treated to inactivate any contaminating viruses,
sequenced and filled to produce the proposed five-member panel, though
WHO Technical Report Series, No. 1039, 2022
in diagnostic testing. There was also no plan to assign unitages to the panel as this
was a known complex issue and would probably cause considerable confusion
among end users. Instead, data from the collaborative study would be included
in the IFU to guide the interpretation of results.
Noting the importance of the reference panel, the Committee
acknowledged the challenge of adding sera corresponding to newly emerging
VOCs, especially as more people became vaccinated. However, the Committee
also recognized the need for the timely availability of the panel, which should
not be delayed due to difficulties sourcing sera for any specific VOC. It agreed
with the proposal to develop a flexible panel with a clearly defined process
for adding further sera as new VOCs emerged. Reflecting on the challenge of
obtaining serum samples for the current panel, the Committee made a number
of suggestions regarding the sourcing of Gamma variant serum. It would also
be important to make clear how the WHO reference panel differed from similar
national reference panels now in development. It was noted that the First WHO
Repository of red blood cell transfusion relevant bacterial reference strains might
provide a useful precedent for such WHO reference materials.
The Committee endorsed the proposal (WHO/BS/2021.2411) to develop
a First WHO International Reference Panel for antibodies to SARS-CoV-2
variants of concern.
would exhibit high antibody titres against all current VOCs, and be sufficient
to fill at least 5000 ampoules. It had been observed that serum from recovered
and subsequently vaccinated individuals exhibited higher antibody titres and a
broader response to the different VOCs than convalescent sera.
Another significant challenge was the need to avoid the inappropriate
use of the reference material in antibody-binding assays. The current WHO
international standard had been assigned an IU based on neutralizing antibodies
and concern had been raised by the Committee during its establishment that
assigning the same unitage for use in antibody-binding assays based on different
antigens could result in the incorrect use of the standard. To allow for comparison
of relative antibody titres to different antigens, the material would be assigned
an arbitrary binding antigen unit (BAU). Comparison of the current WHO
international standard with clinical samples across a range of antibody-binding
assay platforms had demonstrated its commutability with clinical material but
had also highlighted the potential unsuitability of IU assignment for such assays.
As many commercial assays based on different target antigens were calibrated in
BAU and several publications had reported results accordingly, the continued use
of the unit would be useful.
Data from the collaborative study would be used to assign an IU unitage
relative to the current WHO international standard based on neutralization
assays. A value would be calculated for each VOC with arbitrary values to be
assigned should new VOCs emerge. Regarding the timeline for development of
the replacement standard, it was clarified that if the Committee recommended
the use of high-titre sera from recovered and subsequently vaccinated
individuals then such material would need to be sourced, potentially shifting the
establishment of the replacement standard from early to late 2022. Mindful that
any such change in the source material might also affect its assigned unitage, the
Committee advised that the replacement material should be as similar as possible
WHO Technical Report Series, No. 1039, 2022
to the current WHO international standard and should be assessed using the
same neutralization measurement system. It was noted that the development of
future standards using suitable convalescent serum would be challenging because
of changes in the predominant VOC, the rapidly changing COVID-19 vaccination
status of donors and the declining interest in collecting CCP. Given the interest in
using high-titre sera from infected and subsequently vaccinated individuals, the
Committee suggested that such material could, if sourced in time, be included in
the collaborative study and the data subsequently reviewed.
Acknowledging the ongoing high level of demand for the current WHO
international standard and recognizing its importance in the harmonization
of serological assays globally, the Committee endorsed the proposal (WHO/
BS/2021.2411) to develop a Second WHO International Standard for anti-SARS-
CoV-2 immunoglobulin.
62
International reference materials – standards for use in public health emergencies
tests relied on correct folding of the antigen. Noting that the ED50 data from
the pilot study could be used to rank the sensitivity of the different tests and
that numerous publications were now available on test sensitivity using clinical
materials, it was suggested that it might be worthwhile to compare the results of
the full study with the published scientific literature to allow for a more detailed
comparative analysis of test sensitivity.
WHO Technical Report Series, No. 1039, 2022
64
9. International reference materials –
vaccines and related substances
9.1 WHO international reference standards for
vaccines and related substances
9.1.1 Second WHO International Standard for diphtheria antitoxin (equine)
Diphtheria antitoxin (DAT) products produced from equine serum are essential
medicines used for diphtheria therapy and outbreak management, and for
prophylaxis against suspected cases of diphtheria in countries where the disease
is endemic. In countries with good vaccination coverage, DAT is stockpiled for
emergency use. The use of an equine DAT standard calibrated in IU is essential
for ensuring that products meet minimum potency requirements. The First
WHO International Standard for diphtheria antitoxin (equine) was prepared in
Copenhagen in 1934 and consists of a preparation of dried hyperimmune horse
serum in ampoules. To conserve stocks, a batch of liquid standard has been produced
at NIBSC approximately every 2 years, with a diphtheria antitoxin concentration of
10 IU/mL. In 2016, the Committee had been informed that the stock of dried serum
was running low and had endorsed the preparation of a lyophilized replacement
standard that would provide a single homogenous batch sufficient for 15–20 years.
The procured candidate material (NIBSC code 18/180) consisted of refined
diphtheria immunoglobulin prepared from horse serum and was calibrated using
both in vivo and in vitro (Vero cell) toxin neutralization assays, with potency
expressed relative to the current WHO international standard. Several formulations
were evaluated in a trial fill and were freeze-dried successfully with no loss of
biological activity. An international collaborative study was conducted involving
14 laboratories in nine countries, with 10 participants providing data from in vivo
assays and eight from in vitro methods. Although sample protocols were provided,
participants were encouraged to use their existing in-house assays. Potency was
determined by comparing the dose of DAT necessary to protect against the effects
of diphtheria toxin with the quantity of a reference preparation necessary to give
the same protection. Potency estimates obtained using either the in vivo or in
vitro toxin neutralization tests were comparable, with low inter-assay variability.
Although use of the Vero cell assay is increasing, in vivo assays continue to be
used in many countries and so the potency value for candidate material 18/180
was estimated by combining all data. This gave a value of 57 IU/ampoule, which
was similar to the geometric mean potency obtained using the in vivo assays (54
IU/ampoule). The results of accelerated thermal degradation studies predicted no
significant loss in activity when stored at −20 °C indicating long-term stability of
the material. Real-time stability data indicated that the candidate material would
be suitable for use for up to 1 year after reconstitution when stored at 2–8 °C.
65
WHO Expert Committee on Biological Standardization Seventy-fourth report
The Committee commented on the slow progress towards the use of the
Vero cell assay and was informed that many producers of diphtheria antitoxin still
opted to use the in vivo assay. Reflecting on whether the small difference (3 IU
or 5%) between the potency estimated from all available data and that using only
the in vivo assays would be problematic for borderline potency determinations,
the Committee was satisfied that this difference would be negligible in the overall
context of assay variability. The Committee considered the report of the study
(WHO/BS/2021.2407) and recommended that the candidate material 18/180 be
established as the Second WHO International Standard for diphtheria antitoxin
(equine) with an assigned unitage of 57 IU/ampoule based on calibration by in
vivo and in vitro toxin neutralization tests.
expressed its support for the ongoing development of alternative in vitro methods
and suggested that the use of orthogonal methods be explored further. In order to
reduce the use of the Kendrick test, and concluding that accelerated degradation
studies of the proposed standard would be part of the collaborative study, the
Committee agreed that the stability of reconstituted material only needed to be
determined by the custodian laboratory. The Committee endorsed the proposal
(WHO/BS/2021.2411) to develop a Fifth WHO International Standard for
pertussis vaccine (whole cell).
subtle, with the main differences likely to be in yield, purity and possibly post-
translational modification. The Committee recognized the considerable level of
interest in manufacturing polysaccharide-conjugate vaccines in LMIC, especially
in the WHO African Region. Any reduction in the level of containment required
during vaccine production and the increased yield associated with the novel
CRM197 expression systems would potentially reduce the cost of this important
carrier protein.
Noting the importance of demonstrating that the proposed material
could be used as a control for both native CRM197 and CRM197 produced in
other expression systems, the Committee endorsed the proposal (WHO/
BS/2021.2411) to develop a WHO International Reference Reagent for
diphtheria CRM197 antigen.
WHO Technical Report Series, No. 1039, 2022
68
Annex 1
WHO Recommendations, Guidelines and other documents
related to the manufacture, quality control and evaluation
of biological products
WHO Recommendations, Guidelines and other documents are intended to
provide guidance to those responsible for the development and manufacture of
biological products as well as to others who may have to decide upon appropriate
methods of assay and control to ensure that such 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 and guidance on biological products are formulated
by international groups of experts and published in the WHO Technical Report
Series4 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
Email: [email protected]
Website: http://apps.who.int/bookorders
Individual Recommendations and Guidelines and other documents may
be obtained free of charge as offprints by writing to:
Technical Standards and Specifications unit
Department of Health Product Policy and Standards
Access to Medicines and Health Products
World Health Organization
20 avenue Appia
1211 Geneva 27
Switzerland
4
Abbreviated in the following pages to “TRS”.
69
WHO Expert Committee on Biological Standardization Seventy-fourth report
Blood components and plasma: estimation of Adopted 2016, TRS 1004 (2017)
residual risk of HIV, HBV or HCV infections
Blood establishments: good manufacturing Adopted 2010, TRS 961 (2011)
practices
Blood plasma (human) for fractionation Adopted 2005, TRS 941 (2007)
Blood plasma products (human): viral Adopted 2001, TRS 924 (2004)
inactivation and removal procedures
Blood regulatory systems, assessment criteria Adopted 2011, TRS 979 (2013)
for national
5
Available online at: https://www.who.int/biologicals/expert_committee/QA_for_SBPs_ECBS_2018.pdf?ua=1
70
Annex 1
Dengue tetravalent vaccines (live, attenuated) Revised 2011, TRS 979 (2013)
Diphtheria, tetanus, pertussis (whole cell), and Revised 2012, TRS 980 (2014)
combined (DTwP) vaccines
Haemorrhagic fever with renal syndrome (HFRS) Adopted 1993, TRS 848 (1994)
vaccines (inactivated)
Hepatitis B vaccines prepared from plasma Revised 1994, TRS 858 (1996)
Human immunodeficiency virus rapid diagnostic Adopted 2017, TRS 1011 (2018)
tests for professional use and/or self-testing
Technical Specifications Series for WHO
Prequalification – Diagnostic Assessment
71
WHO Expert Committee on Biological Standardization Seventy-fourth report
72
Annex 1
73
WHO Expert Committee on Biological Standardization Seventy-fourth report
6
Available online at: http://www.who.int/biologicals/publications/en/whotse2003.pdf
74
Annex 2
Recommendations to assure the quality, safety and
efficacy of live attenuated yellow fever vaccines
Amendment to Annex 5 of WHO Technical Report Series, No. 978
Introduction 76
Amendment 77
Authors and acknowledgements 82
References 83
75
WHO Expert Committee on Biological Standardization Seventy-fourth report
Introduction
The WHO Recommendations to assure the quality, safety and efficacy of live
attenuated yellow fever vaccines were adopted in 2010 (1). Appendix 2 of these
Recommendations addresses the testing of new virus master and working
seed lots in non-human primates. Specifically, the appendix sets out the ways
in which such lots should be tested for viscerotropism, immunogenicity and
neurotropism, both in terms of clinical evidence and histological lesions, based
on comparison against a reference virus approved by the NRA. Following
reported discrepancies in the clinical scoring of monkeys during the assessment
of working seed lots, WHO received a request from one yellow fever vaccine
manufacturer to align the neurotropism assessment outlined in the 2010
Recommendations with that used for the neurovirulence testing of oral
poliomyelitis vaccine seed lots in which clinical signs are recorded but do not
form part of the assessment or pass/fail criteria (2).
At its seventy-first meeting in August 2020, the WHO Expert Committee
on Biological Standardization recommended that a drafting group be established
to consult with as many yellow fever vaccine manufacturers and other stakeholders
as possible on a proposed revision of Appendix 2 of the 2010 Recommendations
(3). At its seventy-third meeting in December 2020, the Committee was updated
on the progress that had been made (4). The currently specified approach had
now been associated with several technical challenges including: (a) a paucity of
data on the performance of the test; (b) the difficulties inherent in conducting
a collaborative study involving non-human primates; (c) the lack of an
international reference standard for vaccines of the 17D-204 and 17DD lineages
and consequent use of different reference materials; (d) reported discrepancies
between clinical and histopathological assessments; (e) inconsistencies between
staff in the scoring of clinical and histopathological observations; and (f) the
WHO Technical Report Series, No. 1039, 2022
Amendment
Replace Appendix 2 with the following text:
Appendix 2
Tests in non-human primates of new virus master and working seeds
Neurotropism is defined as the tendency or capacity of a microorganism to cause
disease of the nervous system and the yellow fever test in monkeys is a tropism
test by this definition. However, it also involves examination of the tendency
of the virus to cause viraemia after intracranial inoculation, which could be
interpreted as a surrogate of both viscerotropism and the ability to induce an
immune response in the same way. The test can differentiate between strains of
yellow fever vaccine viruses using all three of these criteria.
Each virus master and working seed lot should be tested for viscerotropism,
immunogenicity and neurotropism in a group of 10 test monkeys compared
against a second similar group of 10 monkeys injected with a reference virus.
The same test and reference groups will be used for all of the viscerotropism,
immunogenicity and neurotropism tests. The allocation of animals to the two
groups should be blinded to the operators throughout the experiment. For the
neurotropism test, the test monkeys inoculated intracranially with the virus seed
lot should be compared against the 10 monkeys injected with the reference virus.
Existing manufacturers should use a homologous reference – for example, where
their working seed is to be replaced by another derived from the same master
seed, the existing seed can be used as the reference material, provided it has been
shown to produce a vaccine with satisfactory properties. It is recommended
that sufficient stocks of such a reference are kept for all future anticipated
replacements of the working seed. New manufacturers using a new seed should
use a homologous preparation known to produce a satisfactory product as a
reference material. The reference virus should be approved by the NRA.
A WHO reference virus, 168-73, is available from the National Institute
for Biological Standards and Control, Potters Bar, England. This virus is of the
same lineage as the WHO primary seed 213-77 (see Appendix 1, Figure 1), but
available published data show that it behaves differently to vaccines of at least one
other lineage in the monkey test, being much less neurovirulent and producing
a higher viraemia. It is likely, though unproven, that 168-73 will be a satisfactory
reference for seeds of the 213-77 lineage. While 168-73 is not suitable as a
77
WHO Expert Committee on Biological Standardization Seventy-fourth report
comparator for vaccines of other lineages, its inclusion in the neurotropism test
as a common material would make it possible to compare different tests, and one
lineage with another, for information.
The monkeys should be Macaca mulatta (rhesus monkeys) or Macaca
fascicularis (cynomolgus monkeys) and should have been demonstrated to be
non-immune to yellow fever virus and other flaviviruses using a relevant test
(such as the haemagglutination inhibition test, ELISA or seroneutralization assay)
immediately prior to injection of the seed virus. Tests should be performed using
healthy macaques of both sexes (weighing at least 2 kg and at least 18 months old).
The monkeys should not have been previously subjected to any experimentation.
The test dose should be injected into one frontal lobe of each monkey, under
anaesthetic, and the monkeys should be observed for a minimum of 30 days.
The test dose should consist of 0.25 mL containing not less than 5000
(3.7 log10) IU and not more than 50 000 (4.7 log10) IU, as shown by titration in
cell culture. In addition, the virus titres of the test virus seed lot and the reference
virus should be as close as possible.
Historically, the test dose has consisted of 0.25 mL containing the
equivalent of not less than 5000 and not more than 50 000 mouse LD50, as
shown by titration in cell culture.
1. Viscerotropism test
The criterion of viscerotropism (indicated by the amount of circulating virus)
should be fulfilled as follows: sera obtained from each of the test monkeys on the
second, fourth and sixth days after injection of the test dose should be inoculated at
dilutions of 1:10, 1:100 and 1:1000 into at least four cell culture vessels per dilution.
In no case should 0.03 mL of serum contain more than 500 (2.7 log10) IU and in no
more than one case should 0.03 mL of serum contain more than 100 (2.0 log10) IU.
WHO Technical Report Series, No. 1039, 2022
2. Immunogenicity test
The criterion of sufficient virus-neutralizing antibody in the sera (immunogenicity)
should be fulfilled as follows: at least 90% of the test monkeys should be shown
to have become immune within 30 days following injection of the test dose, as
determined by examining their sera in the yellow fever virus neutralization test
described below. In some countries it has been shown that, at low dilutions, some
sera contain nonspecific inhibitors that interfere with this test. The NRA may
therefore require sera to be treated to remove such substances.
Dilutions of 1:10, 1:40 and 1:160 of serum from each test monkey should
be mixed with an equal volume of strain 17D vaccine virus at a dilution that has
been shown to yield an optimum number of plaques when assayed according to
one of the cell culture methods given in Appendix 4. These serum–virus mixtures
should be incubated in a water bath at 37 °C for 1 hour and then chilled in an
78
Annex 2
ice-water bath before inoculation of 0.2 mL aliquots of each mixture into each
of four separate cell culture vessels. The vessels should be handled in accordance
with one of the cell culture techniques described in Appendix 4. In addition, 10
vessels should be similarly inoculated with a pre-incubated mixture of the same
virus with an equal volume of a 1:10 dilution of monkey serum known to contain
no neutralizing antibodies to yellow fever virus. At the end of the observation
period, the mean number of plaques in the vessels containing virus and non-
immune serum should be compared with the mean number of plaques in the
vessels containing virus and serum from test monkeys. For the immunogenicity
test to be satisfied, serum at the 1:10 dilution from no more than 10% of the test
monkeys should fail to reduce the mean number of plaques by 50% as compared
with the vessels containing non-immune serum.
3. Neurotropism test
The monkeys in the test group should be compared with 10 monkeys injected
with the reference virus with respect to both clinical evidence of encephalitis and
the severity of histological lesions of the nervous system (5, 6).
The onset and duration of the febrile reaction should not differ between
monkeys injected with the test virus or with the reference virus.
Block IV: the pons and cerebellum at the level of the superior olives;
Block V: the medulla oblongata at the midlevel of the inferior olives.
These blocks should be dehydrated and embedded in paraffin wax and
15 µm sections cut and stained with gallocyanin. Alternatively, 5 µm sections
will be suitable for H&E staining or Nissl staining (gallocyanin, cresyl violet), as
well as for immunohistochemistry techniques. A single section, consisting of two
hemisections, should be cut from each block.
Sections should be examined microscopically and numerical scores
assigned to each hemisection of the cervical and lumbar enlargements, and to
each anatomical structure (see Appendix 3) within each hemisection of the brain
blocks, according to the following grading system:
80
Annex 2
Texas Medical Branch Sealy Center for Vaccine Development, the USA; Dr R.M.
Bretas, Agência Nacional de Vigilância Sanitária, Brazil; Dr G. Cirefice, European
Directorate for the Quality of Medicines & HealthCare , France; Dr M. Diagne,
Direction des Laboratoires, Senegal; Dr A. Dieng, Direction de la Pharmacie et
du Médicament, Senegal; Dr E. Grabski, Paul-Ehrlich-Institut, Germany; Mrs F.
Garnier, Agence nationale de sécurité du médicament et des produits de santé,
France; Dr Y. Li, National Institutes for Food and Drug Control, China; Dr J. Martin,
National Institute for Biological Standards and Control, the United Kingdom;
Dr P. Minor, St Albans, the United Kingdom; Mrs V. Pithon, Agence nationale
de sécurité du médicament et des produits de santé, France; Dr M.F. Reis e Silva
Thees, Agência Nacional de Vigilância Sanitária, Brazil; Dr J. Wang, National
Institutes for Food and Drug Control, China; Dr Y. Wang, National Institutes for
Food and Drug Control, China; Dr M. Xu, National Institutes for Food and Drug
82
Annex 2
References
1. Recommendations to assure the quality, safety and efficacy of live attenuated yellow fever
vaccines. In: WHO Expert Committee on Biological Standardization: sixty-first report. Geneva:
World Health Organization; 2013: Annex 5 (WHO Technical Report Series, No. 978; https://www.
who.int/publications/i/item/9789241209786, accessed 21 June 2021).
2. Recommendations to assure the quality, safety and efficacy of poliomyelitis vaccines (oral, live,
attenuated). In: WHO Expert Committee on Biological Standardization: sixty-third report. Geneva:
World Health Organization; 2014: Annex 2 (WHO Technical Report Series, No. 980; https://www.
who.int/publications/i/item/9789241209802, accessed 21 June 2021).
3. Revision of the WHO Recommendations to assure the quality, safety and efficacy of live attenuated
yellow fever vaccines. In: WHO Expert Committee on Biological Standardization: seventy-first
report. Geneva: World Health Organization; 2021 (WHO Technical Report Series, No. 1028, section
3.2.3, pp. 20–1; https://www.who.int/publications/i/item/9789240020146, accessed 21 June 2021).
4. Amendment to the WHO Recommendations to assure the quality, safety and efficacy of live
attenuated yellow fever vaccines. In: WHO Expert Committee on Biological Standardization:
report of the seventy-second and seventy-third meetings. Geneva: World Health Organization;
2021 (WHO Technical Report Series, No. 1030, section 3.3.4, pp. 36–7; https://www.who.int/
publications/i/item/9789240024373, accessed 21 June 2021).
83
WHO Expert Committee on Biological Standardization Seventy-fourth report
5. Fox JP, Penna HA. Behavior of 17D yellow fever virus in rhesus monkeys: relation to substrain, dose,
and neural or extraneural inoculation. American Journal of Hygiene. 1943;38(2):152–72 (abstract:
https://www.cabdirect.org/cabdirect/abstract/19442900204, accessed 9 June 2021).
6. Levenbook IS, Pelleu LJ, Elisberg BL. The monkey safety test for neurovirulence of yellow fever
vaccines: the utility of quantitative clinical evaluation and histological examination. J Biol
Stand. 1987;15(4):305–13 (abstract: https://www.sciencedirect.com/science/article/abs/pii/
S0092115787800033, accessed 9 June 2021).
WHO Technical Report Series, No. 1039, 2022
84
Annex 3
Evaluation of the quality, safety and efficacy of messenger
RNA vaccines for the prevention of infectious diseases:
regulatory considerations
1. Introduction 88
2. Purpose and scope 89
3. Terminology 91
4. General considerations 95
5. Special considerations 98
6. Manufacture and control of mRNA vaccines 101
6.1 General manufacturing overview 104
6.2 General information and description of vaccine construct and composition 109
6.3 Control of starting and raw materials and excipients 111
6.4 Process development and in-process controls 114
6.5 Product characterization 114
6.6 Consistency of manufacture 116
6.7 Manufacture and control of bulk purified mRNA (drug substance) 116
6.8 Manufacture and control of final formulated vaccine (drug product) 121
6.9 Records 130
6.10 Retained samples 130
6.11 Labelling 130
6.12 Distribution and transport 131
7. Nonclinical evaluation of mRNA vaccines 131
7.1 Pharmacology/immunology/proof-of-concept 132
7.2 Safety/toxicity in animal models 132
7.3 Accelerating nonclinical evaluation in the context of rapid vaccine development
against a priority pathogen during a public health emergency 135
8. Clinical evaluation of mRNA vaccines 137
8.1 Safety and immunogenicity evaluation 137
8.2 Efficacy evaluation 140
8.3 Efficacy evaluation in the context of a public health emergency in which
immune-escape and other variants arise 141
Authors and acknowledgements 141
References 145
85
WHO Expert Committee on Biological Standardization Seventy-fourth report
86
Annex 3
Abbreviations
AESI adverse events of special interest
COVID-19 coronavirus disease 2019
DNA deoxyribonucleic acid
DNase deoxyribonuclease
dsRNA double-stranded RNA
GMP good manufacturing practice(s)
HPLC high-performance liquid chromatography
ICH International Council for Harmonisation of Technical
Requirements for Pharmaceuticals for Human Use
IU International Unit(s)
IVT in vitro transcription
LNP lipid nanoparticle
mRNA messenger RNA
NRA national regulatory authority
ORF open reading frame
PCR polymerase chain reaction
PEG polyethylene glycol
PEGylation polyethylene-glycol-ylation
RNA ribonucleic acid
RT-PCR reverse transcription polymerase chain reaction
sa-mRNA self-amplifying mRNA
SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
tRNA transfer RNA
UTR untranslated region
WHO World Health Organization
87
WHO Expert Committee on Biological Standardization Seventy-fourth report
1. Introduction
Although the immunostimulatory effects of RNA have been known since the
early 1960s (1), the possibility of using direct in vivo administration of in vitro
transcribed messenger RNA (mRNA) to temporarily introduce genes expressing
proteins (including antigens) was demonstrated in 1990 following the direct
injection of “naked” nucleic acids (2). Subsequent improvements in stabilizing
mRNA, increasing the feasibility of manufacturing RNA-based products and
decreasing RNA-associated inflammatory responses have led to significant
advances in the development of mRNA vaccines and therapeutics (3–6). There
are several reasons why the mRNA platform has emerged at the forefront of
vaccine technology. Among these are the rapid speed at which mRNA candidate
vaccines can be constructed and manufactured, and the need to rapidly develop
vaccines against emerging pathogens, such as zoonotic influenza virus strains,
Zika virus and most recently severe acute respiratory syndrome coronavirus 2
(SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19).
A number of publications have now discussed some of the safety,
production and regulatory issues associated with this new technology (7–14). In
addition, the rapidity with which clinical trials have progressed for COVID-19
candidate vaccines, their approval or authorization by NRAs and subsequent
widespread use have created a pressing need for WHO guidance on evaluating
the quality, safety and efficacy of mRNA products used for the prevention
of infectious diseases in humans. Such evaluations must take into account:
(a) the inherent immunological, physiochemical and structural properties
of mRNA; (b) the need for special formulations such as lipid nanoparticles
(LNPs) to ensure in vivo stability and efficient delivery; and (c) the novel cell-
free enzymatic manufacturing process. Because detailed information is not yet
available on the methods used for production, controls are not yet standardized
WHO Technical Report Series, No. 1039, 2022
for safe and efficacious mRNA vaccines, and certain details remain proprietary
and thus not publicly available, it is not feasible to develop specific international
guidelines or recommendations at this time. Consequently, flexibility in the
scientific approach to regulating mRNA vaccines is currently needed. The
detailed production and control procedures, as well as any significant changes
in them that may affect the quality, safety and efficacy of mRNA vaccines, should
be discussed with and approved by the NRA on an individual case-by-case basis.
Nevertheless, the key principles described in this document are applicable to the
class of preventive mRNA vaccines against infectious diseases for human use in
general and are intended to provide guidance until more detailed information
becomes available. For mRNA vaccines that target diseases for which there are
existing vaccines and corresponding WHO guidance, it may be appropriate to
consider the relevant sections of this document for issues specific to mRNA
88
Annex 3
useful. Due to the novelty of mRNA vaccines and their manufacturing process,
a comprehensive approach has been taken to ensure that all relevant aspects can
be considered by manufacturers when developing this type of product, and by
regulators when evaluating such products.
The scope of the current document is limited to mRNA and self-
amplifying mRNA (sa-mRNA) packaged in LNPs for in vivo delivery of the
coding sequences of a target antigen relevant to active immunization for the
prevention of an infectious disease. It is acknowledged that mRNA and sa-mRNA
products in formulations other than LNPs are also in development, and parts of
this document may be applicable to those products as well.
Replicating agents, viral vectors and RNA replicons packaged in viral
proteins or encoded by plasmid DNA are outside the scope of this document.
In addition, mRNA and sa-mRNA products intended for therapeutic purposes
(that is, products for the treatment, mitigation or cure of diseases, including
infectious diseases, as opposed to active immunization for their prevention) are
also outside the scope of this document. In addition, mRNA products expressing
monoclonal antibodies (whether serving as passive immunization for disease
prevention or therapy) are also outside the scope of this document. It may be
the case that some aspects discussed in section 6 and its subsections do apply
to mRNA-based therapeutic products (including those expressing monoclonal
antibodies), as the manufacturing steps of such products may be similar to those
described for vaccines. However, because the nonclinical and clinical evaluations
of such therapeutic products would need to be based on their therapeutic
indication, it is not feasible to include regulatory considerations for them within
this document.
As there may be a need to develop multivalent mRNA vaccines or to
change the existing vaccine strain for some pathogens (for example, influenza
viruses or SARS-CoV-2), specific considerations are provided in this document
WHO Technical Report Series, No. 1039, 2022
3. Terminology
The definitions given below apply to the terms as used in this document. These
terms may have different meaning in other contexts.
Adjuvant: a substance intended to enhance the relevant immune response
and subsequent clinical efficacy of a vaccine.
Biological (or biological product): a medicine produced by a biological
system, as opposed to strictly chemical reactions. These include traditional
biologicals (such as live vaccines) and biotechnologically produced medicines
(such as monoclonal antibodies or subunit vaccines such as human papillomavirus
vaccines). In other documents, these may be referred to as biologics or biological
medicines.
Candidate vaccine: an investigational vaccine that is in the research and
clinical development stages and has not been granted marketing authorization or
licensure by a regulatory agency in the country in which such authorization or
licensure will be sought.
Design of experiments: a structured, organized method for determining
the relationship between factors affecting a process and the output of that process.
Drug product: see final vaccine.
Drug substance: the purified mRNA before final formulation. It is
prepared as a single homogeneous production batch, kept in one or more
containers designated as such and used in the preparation of the final dosage
form (final vaccine or drug product).
Double-stranded RNA (dsRNA): some viruses have genomes
comprising fully double-stranded RNA along their entire length rather than in
distinct segments (such as the secondary structure of mRNA). If present, such
dsRNA is sensed by intracellular receptors and can activate innate immune
responses. Depending on the manufacturing method, dsRNA can be generated
as a by-product during the in vitro transcription (IVT) manufacturing process
for some mRNA vaccines, though some segments may be single stranded. This
type of dsRNA is an impurity that should be removed from the mRNA during
the manufacturing process, or its amount in the product at least determined and
controlled. If the manufacturing method does not produce dsRNA, then the
control of this as an impurity is unnecessary.
Engineering run: a manufacturing campaign conducted to engineer
manufacturing methods in order to improve or confirm those methods for use in
good manufacturing practice (GMP)-compliant production. The materials made
in such a campaign are not intended for use in humans.
Excipient: a constituent of a medicine other than the active drug
substance, added in the formulation for a specific purpose. While most excipients
are considered inactive, some can have a known action or effect in certain
91
WHO Expert Committee on Biological Standardization Seventy-fourth report
circumstances. The excipients must be declared in the labelling and package leaflet
of the medicine to ensure its safe use. In the context of the current document, the
lipids that form the LNPs are excipients but the LNPs if formed separately from
the mRNA are defined as intermediates in the production of the drug product.
Final formulated bulk: an intermediate in the manufacturing process
of the final vaccine, consisting of a homogeneous preparation of the final
formulation of drug substance(s) and excipients at the concentration to be filled
into final containers. Alternatively, the final formulated bulk may be stored at a
higher concentration and diluted immediately prior to filling. In the context of this
document, the term refers to mRNA formulated with LNPs and other excipients
as needed. Note that if more than one drug substance is to be combined (as in
a multivalent or combination vaccine), their mixing would occur as part of the
preparation of this final formulated bulk.
Final lot: a collection of sealed final containers that is homogeneous with
respect to the composition of the product and the avoidance of contamination
during filling. A final lot must therefore have been filled from a final formulated
bulk in one continuous working session. A final formulated bulk might be filled
into more than one final lot.
Final vaccine (or drug product): a final dosage form (for example,
a vialled frozen or liquid suspension or lyophilized cake) that contains one or
more drug substances (active ingredient) typically formulated with excipients
and packaged for use. In the context of this document, the term refers to a
preparation of mRNA formulated with LNPs and other excipients that is filled
into final containers. If filled in concentrated form or lyophilized, a diluent is
needed. Otherwise, the final containers should be filled at the concentration for
the clinical dose (though each container might contain multiple doses). Also
referred to as “finished product” in other documents.
Good manufacturing practice (GMP): a system that ensures that
WHO Technical Report Series, No. 1039, 2022
That is, protection against disease (or infection) caused by a pathogen(s) may
be conferred by the resulting immune response against the target antigen(s)
following vaccination.
Therapeutic: a treatment given after a disease or condition (or signs or
symptoms thereof) is evidenced, in contrast to the prevention of disease before
exposure (or in rare cases following exposure but before the onset of signs or
symptoms) to the infectious pathogenic organism has occurred. Although
preventive vaccines are not considered to be therapeutic in this document, it is
acknowledged that the definition of therapeutic in some regulatory jurisdictions
may differ. Therapeutics as defined here are outside the scope of the current
document.
Transfer RNA (tRNA): an RNA molecule used by ribosomes and that
acts as an adaptor involved in translating the codons of the mRNA into a protein.
4. General considerations
As with all vaccines, the intended clinical use of the mRNA vaccine should
be described, including the pathogen targeted, the target antigen(s) chosen,
disease to be prevented and the target population(s). Given the novel structure
and manufacturing of mRNA candidate vaccines (in contrast to other already
licensed vaccine types with which regulators are familiar), consideration should
be given to the following when evaluating mRNA vaccines for their quality, safety
and efficacy:
■ In particular, the relevant biological characteristics of the specific
mRNA technology used should be described – including for
example the capability of the given mRNA to trigger innate immune
responses as well as target-antigen-specific responses; the quality,
quantity and bias of the immune responses (for example, type 1
T-helper (Th1) or Th2 cell phenotype); and in vivo stability. To
justify the vaccine design, all available information on the type of
immunity (protective and immunopathogenic) considered relevant
to the specific pathogen and disease should also be described.
■ The rationale for the selection of the target antigen(s) or parts
thereof and of any proteins (for example, cytokines) that are
encoded, as well as their contribution to the proposed mode- or
mechanism-of-action (proposed protective process) of the vaccine,
should be clearly described. Likewise, the rationale for the selection
of any coding sequences added to or any modification of the target
antigen, such as those to ensure the folding of the target antigen
into a particular conformation, should be provided. The complete
95
WHO Expert Committee on Biological Standardization Seventy-fourth report
provided.
■ The intended dosing, the route of administration, and a description
and justification of any novel administration device as well as any
required diluent should be provided. Relevant compatibility studies
should be performed where necessary.
■ Although any given manufacturer’s mRNA vaccine product may
be considered to be produced by a platform technology if only the
target antigen sequence is changed, the control, nonclinical testing
and clinical development of each vaccine should be considered
individually, and any special features of that candidate vaccine taken
into account. Early consultation with the NRA(s) will be key to
ensuring the efficient development of any given candidate vaccine.
96
Annex 3
5. Special considerations
The mRNA of vaccines that are currently the most advanced in terms of clinical
development or that are currently in widespread use against COVID-19 is
produced enzymatically rather than biologically within a cell. This approach thus
differs from the production of most other biologicals with which manufacturers
and regulators are familiar (1, 33). Manufacturing either starts with linearized
DNA plasmids that have been produced in bacteria (similar to the way in which
biologicals such as plasmid DNA vaccines are produced) or with a linear DNA
molecule produced enzymatically using the polymerase chain reaction (PCR)
or other synthetic methods. Regardless of whether the manufacture of the RNA
starts with a linearized molecule generated from a plasmid DNA or from an
already linear DNA sequence, mRNA production occurs enzymatically in vitro
by means of a DNA-dependent RNA polymerase that transcribes the linear DNA
template into an mRNA molecule. The mRNA sequence generally consists of the
usual elements of cellular mRNA, such as the coding region, 5′ and 3′ untranslated
regions (UTRs) that regulate mRNA translation, a 5′ cap and a 3′ poly(A) tail.
The nucleotides used in manufacture may contain naturally occurring
nucleosides or modified or synthetic nucleosides (3, 8). Examples of alterations
that might be made to the naturally occurring nucleoside include the use of
pseudouridine or N1-methylpseudouridine in place of uridine (3, 4, 34). In
WHO Technical Report Series, No. 1039, 2022
and generally have a 5′ cap and a 3′ poly(A) tail. The cap can be added to the
mRNA enzymatically or during in vitro transcription (IVT) using appropriate
cap analogues. Likewise, the 3′ poly(A) tail can be encoded in the DNA template
or added enzymatically after IVT. These design features can impact the critical
quality attributes, the manufacturing methods and the control testing of the
mRNA drug substance(s) and/or the final vaccine drug product.
Of relevance to considerations of the safety and efficacy of mRNA vaccines
are the structures adopted by the RNA in the vaccine product. Unlike DNA, which
is normally double stranded, RNA is often represented diagrammatically as being
single stranded. However, depending on its sequence, RNA can form a complex
structure consisting of short double-stranded segments with various single-
stranded loops in between. The reason this is relevant is that double-stranded
RNA (dsRNA) is a form taken by the genome of some RNA viruses and can
induce cells to trigger immune reactivity as an innate response to viral infection.
However, endogenous cellular mRNA does not induce such an effect despite
containing partial double-stranded segments. The in vivo effects, including
potential triggering of innate immunity, of an mRNA candidate vaccine should
therefore be characterized and addressed in the vaccine design, nonclinical
studies and clinical trials.
RNA-based products can take different forms. The most advanced
candidate vaccines and the widely used COVID-19 vaccines take the form
of mRNA encoding the target antigen (35, 36). Because mRNA (and RNA in
general) is subject to degradation by nucleases, the most advanced mRNA
candidate vaccines and widely used COVID-19 vaccines at the time of writing
are formulated in LNPs, which aids in vivo stability and delivery (33, 37–43).
There are different types of LNPs depending on their composition, the types of
lipids employed and the manufacturing process used (44). Some may not yet have
been employed for the delivery of mRNA (45–48). Other stabilizing and delivery
systems using polymer and polypeptide, as well as other lipid-based systems or
combination of polymer and lipid-based systems, may be developed for mRNA
delivery in the future. These drug delivery systems could also be surface modified
for tailored cellular interactions, where necessary.
It is important to note that the drug substance is the mRNA(s). The
lipids which form the LNPs are excipients of the final vaccine or drug product.
The manufacture of LNPs from the different lipids is part of the drug product
manufacturing process. It is acknowledged that some LNPs, depending on their
composition, may also have immunomodulatory effects (47–50) and some lipids
may act as adjuvants without being formulated as LNPs. Nonetheless, vaccine
adjuvants, which are immunomodulating to the vaccine, are also considered to be
excipients. Similarly, as discussed above, RNA itself can be immunomodulating.
Consequently, both components (the mRNA and the lipids in the LNPs) may
99
WHO Expert Committee on Biological Standardization Seventy-fourth report
with GMP. Appropriate attention needs to be given to ensuring the quality and
correct identity of all materials used in production and control. Particular
attention should be given to the sourcing of components of animal (including
human) derivation. Attention should also be given to ensuring freedom from, or
control of, potential adventitious agents supported by relevant evidence and risk
assessment. Many of the general requirements for the quality control of biological
products, such as tests for endotoxin, stability and sterility, should also be applied
to mRNA vaccines. The commercial specifications should be defined on the basis
of the results of tests on lots that have been shown to have acceptable performance
in clinical studies. Additional controls specific to mRNA or sa-mRNA vaccines
formulated in LNPs should be described, including controls for raw materials and
excipients and in-process controls for manufacturing intermediates.
It should be recognized that the level of detail required by a regulatory
authority increases as product development proceeds. During the initial
phases of clinical development, the information contained in a clinical trial
application should be adequate to allow for an assessment of the risks derived
from the drug product and the manufacturing process. This would include, for
example, identification of and specifications for all materials used in the process,
assessment of risks from biologically sourced materials, certification or phase-
appropriate GMP compliance of the manufacturing facility, a brief description of
the processes and tests, results of testing of vaccine lot(s) (and if applicable, for a
clinical trial application, placebo or other comparator) to be used in the proposed
clinical trial and results of preliminary stability testing. As with all vaccines, for
pivotal clinical trials the level of detail provided on the quality (manufacturing
and controls) of an mRNA vaccine would be expected to increase substantially.
While not every mRNA vaccine can be viewed as being made based on
a platform technology, a given manufacturer’s technology might to some extent
be viewed this way. In other words, if essentially no changes are made to the
manufacturing processes (other than process optimization for each candidate
vaccine), tests (except for identity or potency) or specifications, then a new
candidate mRNA vaccine might be supported by data from an earlier candidate
mRNA vaccine or licensed product. For example, this could be the case when
the only changes made are to the sequence and these changes do not change the
size or secondary structure of the resultant new mRNA or its interaction with the
LNP. Supportive data might include data gained on the manufacturing processes,
tests, specifications, stability and nonclinical and clinical safety.
Details of any changes made to product composition (for example, change
in the mRNA sequence, enhanced valency, change in excipients or addition of
preservatives) or manufacture (for example, change in process, site or scale) during
the development of clinical lots should be provided. Depending on the way in
which the final product composition was changed (for example, addition of novel
103
WHO Expert Committee on Biological Standardization Seventy-fourth report
excipients) new nonclinical studies might be warranted (see section 7 below). For
changes to the manufacturing process (such as scale-up or change to the purification
process) the comparability of the resulting drug substance and drug product with
those produced using the previous process should be evaluated. Such comparability
studies might be based on immunogenicity data from animal models, results from
physicochemical analyses, studies of process and product-related impurities, and/
or stability data. The WHO Guidelines on procedures and data requirements for
changes to approved vaccines (31) should be consulted in this regard. All changes
made to the product post-approval should follow the requirements listed in these
same Guidelines (31); other relevant guidance may also be considered such as the
ICH Harmonised Guideline on pharmaceutical product lifecycle management (52).
Defined recombinant nucleic acids used as active drug substances
in vaccines, whether of biological or synthetic origin, could be assigned an
international nonproprietary name (INN) upon request (53, 54).
but prior to purification (8, 34, 55, 56). In addition to removing the DNA
template, the unattached caps, unincorporated nucleotides and the enzymes
(such as RNA polymerase) used in production, all process-related and product-
related impurities (for example, dsRNA and incorrectly sized mRNA molecules)
should also be removed to the extent feasible. Attention should also be paid to
the removal of enzymes possibly involved in DNA template generation, such
as DNA polymerase and restriction enzymes (if not controlled at the level of
the DNA template). The methods of purification and their purposes should be
described and justified. Any purification processes – such as protein digestion
with proteinase(s) as an impurity-reduction step – should be validated at the
appropriate phase of development (see section 6.4 below).
In most cases, the purified mRNA would be considered to correspond
to what is termed for other vaccines “the purified bulk antigen” – even though
104
Annex 3
the mRNA is not the actual antigen but instead mimics the transcript encoding
the antigen. This could also be thought of as the bulk biological substance or
bulk active substance and is referred to in this document as the drug substance
in order to use terminology familiar to most manufacturers and regulators to
describe the active biological element of the vaccine.
As would be expected for any vaccine, a flowchart of production should
be provided that indicates each process step, the samples taken at that process
step and the in-process control tests for which the samples are taken. The process
flowchart should also clarify the steps in the process at which manufacturing
reaches the stages of drug substance, final formulated bulk and final filled vaccine
(drug product), and at which steps in the flowchart samples are being taken for
in-process control and release testing. The tests carried out at each of these steps
should also be indicated. The duration of storage of the concentrated purified
mRNA (drug substance) or any intermediates (such as the final formulated bulk)
that are held or stored should be supported by hold-time/stability studies. As
with any vaccine, an agreed-upon number of lots of the drug product should be
placed on a stability programme.
The mRNA (drug substance) is not suitable for clinical use unless it is
protected and delivered by a given formulation (the preparation of which is part
of drug product manufacture). The formulations chosen for the most advanced
mRNA vaccines so far are based on LNPs. Although there are other approaches to
encapsulating mRNA-based products, the current document only covers systems
that use LNPs. The formulation both stabilizes the mRNA and facilitates its entry
into cells and release into the cytosol, which could be achieved by either active
or passive uptake. The LNPs may also provide an adjuvant activity (47, 49, 50).
In order to protect the mRNA from degradation by nucleases, it is incorporated
into the LNPs to make it inaccessible to such nucleases – however, the LNPs
must also release the mRNA once inside the target cell. The LNPs must also be
of a suitable size range with desirable surface properties for optimal uptake by
target cells. Hence, product development data concerning the optimization of
both the formulation and the manufacturing process should be provided. For
example, consideration should be given to the concentrations of the different
lipids, the mRNA–lipid ratio, pH of buffers/solvents, mRNA encapsulation
efficiency, and the flow rate and mixing rate of the lipids and mRNA, as well as
the thawing temperature of the different components, as these will all have an
impact on the quality of the final vaccine (drug product). In this way, the process
of encapsulation into the LNPs can be carefully controlled and the production
methods and control measures adequately described and suitably validated.
Although sa-mRNA contains the coding sequences (viral nonstructural
genes) for additional proteins that permit its in vivo amplification (but not its
packaging, which requires viral structural genes), the method of manufacture in
105
WHO Expert Committee on Biological Standardization Seventy-fourth report
expected for clinical trial material used in pivotal trials and for commercial
manufacture (18, 19).
Any manufacturing changes made during clinical development,
particularly if made following completion of pivotal safety and efficacy trials
but prior to seeking licensure, need to be described and justified. A comparative
analysis with the clinical efficacy lots should be made. For post-approval changes,
compliance with the WHO Guidelines on procedures and data requirements for
changes to approved vaccines (31) would be expected, though other relevant
guidance might also be considered such as the ICH Harmonised Guideline on
pharmaceutical product lifecycle management (52).
Table 1
Examples of possible methods for characterization or control at various key quality
control points, by potential use(s)
108
Annex 3
Table 1 continued
Examples of possible method Potential use(s)
Electron microscopy; atomic force Physicochemical characterization
microscopy; X-ray diffraction; differential (including surface and morphological
scanning calorimetry analysis properties)
Tests for sterility, endotoxin content Safety attributes
pH determination; gravimetric, azeotropic Quality attributes
or titrimetric method to test residual
moisture
lipids and critical quality attributes of the drug product should also
be included. Sufficient characterization of the mRNA-LNP and of
its uptake into target cells should be provided. This may include an
understanding of the surface chemistry, size, polydispersity, shape,
charge and protein-binding properties of the resultant mRNA-LNP
in order to ensure that adequate protection of the mRNA and the
required stability of the vaccine are achieved. Where the LNPs are
shown to have inherent immunomodulatory effects, relevant data
on the potential benefits and drawbacks should be presented. Thus
any characteristics of the formulation that might impact the safety,
immunogenicity and efficacy of the vaccine should be described
and their effects (positive or negative) should be considered during
formulation development.
c. Additional immunomodulators or adjuvants: the mRNA might
also encode specific immunomodulatory molecules such as
cytokines. Furthermore, a separate adjuvant or immunomodulatory
(stimulatory or suppressive) compound not encoded in the mRNA
might be added to the formulation or as part of the LNP. As a
general principle regarding vaccines formulated with adjuvants, a
demonstration of the contribution of such an addition to vaccine
immunogenicity should be provided (19). Quality aspects of
the separate adjuvant, if included, should also be addressed and
described.
d. Additional peptides/proteins: if additional peptides/proteins are
included to target the mRNA to antigen-presenting cells or other
specific cell types or to increase the release of the mRNA from the
endosome, the sequence and function of these additions need to be
described and evidence provided of their function to support their
proposed mechanism-of-action.
e. Additional excipients (such as preservatives): the composition,
necessity for and (in the case of preservatives) the preservative
efficacy of such additional excipients should be described and
shown not to adversely affect the properties of the LNP.
raw materials (for example, enzymes, buffers and solvents), intermediates and
final excipients (for example, lipids and salts) should be provided. Consideration
should also be given to the use and control of solvents, and to the potential
for contamination with elemental impurities (60–62). Where the recycling
of materials/solvents is proposed, this should be justified and appropriately
controlled. The level of impurities associated with the excipients should also be
suitably controlled and justified. Any purification and isolation steps should be
detailed. To assure the quality of the proposed novel excipients, their manufacturer
should also have available relevant information on the analytical methods used
for the characterization, stability monitoring and batch analyses of the materials.
Since inclusion of a PEGylated lipid plays a critical role in providing in vivo
stability and enhancing the cellular interaction of LNPs (42), adequate controls
(for example, of molecular weight, polydispersity and mole percent) should be in
112
Annex 3
place for the PEGylated lipid. For the manufacturing of excipients, compliance is
expected with WHO Good manufacturing practices: supplementary guidelines
for the manufacture of pharmaceutical excipients (22).
responses, then this may require a redesign of the mRNA sequence. The degree of
consistency of encapsulation of the mRNA in the LNPs should also be addressed
during characterization. Particle-uptake studies could assist in characterizing
potential potency measures through identification of cell types that take up the
particles, mode or mechanism of uptake, and efficiency of uptake, and thus guide
selection of the type of cell-free or in vitro method that best allows for assessment
of these activities. During characterization, it should be determined whether any
of these characteristics should be controlled as critical quality attributes and/or
stability-indicating attributes.
Certain aspects of the LNPs should be very carefully characterized. These
include particle size as determined by different analytical techniques to explore
the morphological and dimensional characteristics of the LNPs containing the
mRNA. Information on the density and distribution of polyethylene glycol
(PEG) within the LNPs would also be useful to help understand the surface
properties of the mRNA-LNP. Measurement of surface charge (for example, zeta
potential) should also be considered as a method for characterizing the LNPs.
These, and other properties, will affect the in vivo stability, cellular interaction
and immunological response properties of the product; such information would
also help to confirm the consistency of the manufactured vaccine.
The immunogenicity elicited by the mRNA-encoded target antigen is a
critical characteristic of the product that should be characterized in nonclinical
studies as a means to understand the product. Additionally, if the LNPs have
inherent immunomodulatory effects these should also be characterized.
Whenever other immunomodulatory elements or genes are included in the
mRNA, their contribution to the mode-of-action (for example, immunogenicity)
of the mRNA-encoded target antigen should also be determined in nonclinical
studies in order to justify their inclusion in the characterized product design
(see section 7 below). Consideration of these aspects is important in gaining
understanding and knowledge of the product in order to optimize its design and
develop appropriate control methods.
Potential impurities that might be introduced by the starting materials,
and potential product- or process-related impurities in the purified mRNA, should
be described and investigated. Such impurities may include residual bacterial
host-cell proteins (if used to manufacture the DNA template), endotoxins,
residual bacterial host-cell RNA and chromosomal DNA (if bacteria were used to
manufacture the DNA template), enzymes (such as DNA and RNA polymerases
and restriction enzymes), unincorporated nucleotides, dsRNA, incomplete or
differently sized RNA, and other materials used in the manufacturing process.
Data should be provided on the impurities present in the purified mRNA in
order to justify the specifications set for their maximum acceptable or lowest
achievable levels. For impurities and residuals with known or potential toxic
115
WHO Expert Committee on Biological Standardization Seventy-fourth report
gene, other gene(s) contained in the mRNA sequence, UTRs, 5′ cap, 3′ poly(A)
tail and regulatory elements used. Any gene expression or other optimization
modifications should be described. Annotated sequences of the complete DNA
template and mRNA should be provided. Both an illustrative and annotated
flowchart and a narrative description of the manufacture, in-process controls
and release tests should be provided. The detailed production and control
procedures along with any significant changes in them that may affect the
quality, safety and efficacy of the mRNA vaccine should be discussed with and
approved by the NRA.
In the case of sa-mRNA, if the replicon and target antigen are expressed
on separate mRNA molecules, this should be described and clearly illustrated in
the provided flowchart, which should also include any additional manufacturing
processes and/or quality control tests. For example, consideration should be
given to controls such as the ratio of replicon-encoding mRNA molecules to
target-antigen-encoding mRNA molecules, or to methods to ensure (or controls
to determine whether or not) both molecules are encapsulated into the same
LNP, if applicable. For sa-mRNA in which the mRNA encoding the replicon and
the mRNA expressing the target antigen are encoded on different molecules, it
will be important that these two RNAs are co-encapsulated in order for them to
be taken up by the same cell in vivo. Therefore, if the two RNAs are encapsulated
separately and then mixed, a justification for this approach will be required.
6.7.1.1 Identity
Each batch of bulk purified mRNA should be tested to confirm its identity.
Confirmation of identity could include determination of the mRNA sequence by
direct RNA sequencing, sequencing (or determining the presence or absence) of
a reverse transcription PCR (RT-PCR) product or high-throughput sequencing.
If identity is based on an RT-PCR amplicon that represents only a portion of the
complete mRNA sequence (including accessory and regulatory regions), then the
sequence chosen should be unique to that mRNA product and not be common
to any others that might be manufactured in the same facility or using the same
equipment. However, it might be more appropriate to sequence the entire mRNA
as this approach could serve to address both identity and potentially purity,
depending on the sequencing method used.
engineering run batch or a batch from which the lot of mRNA vaccine evaluated
in the pivotal nonclinical studies was made may serve as a reference until a suitable
clinical trial batch has been identified and characterized for use as a reference
in advanced development (for example, pivotal clinical trials) and commercial
manufacture. Whatever approach is taken should be clearly described.
6.7.1.7 Stability
A stability assessment should be conducted in accordance with the WHO
Guidelines on stability evaluation of vaccines (27). The types of studies conducted,
the protocols followed, and the study results should all be summarized in an
appropriate format such as tables or graphs along with a narrative document.
The summary should include results as well as conclusions with respect to
120
Annex 3
6.8.1 Composition
The final composition of the vaccine, including the active drug substance
(mRNA) and all excipients (for example, lipids), should be described along with
the quantity of the components in each presentation – particularly if marketing
authorization is being sought for more than one dosage or dosage form. The
function of each of the components should also be described.
candidate vaccines, the impact of the new mRNA drug substance on the critical
quality attributes of the final vaccine product should be determined.
with acceptance criteria that are justified by relevant development data). Details of
the sterilization process and microbial control should also be included.
The general guidance concerning filling and containers provided in WHO
good manufacturing practices for biological products (23) should be applied to
vaccine filled in the final form. The aseptic fill process of the mRNA-LNP should
be adequately validated to ensure all critical quality attributes are maintained
and meet the required specifications. Care should be taken to ensure that the
materials of which the containers and closures (and, if applicable, the transfer
devices) are made do not adversely affect the quality of the vaccine. To this end, a
container-closure integrity test and assessment of extractables and/or leachables
for the final container-closure system are generally required for the qualification
of containers and may be needed as part of stability assessments.
If multi-dose vaccine vials are used and the vaccine does not contain
a preservative, then their use should be time restricted, as is the case for
reconstituted vaccines such as bacillus Calmette–Guérin (BCG) and measles-
containing vaccines (32). In addition, the multi-dose container should prevent
microbial contamination of the contents after opening. Relevant simulation
studies (for example, multi-puncture tests) of the container-closure system
may be required to demonstrate the suitability of the proposed system. Multi-
dose vials should be designed to meet the label claim, with acceptable overfill
to allow for correct dosing. Multi-dose vaccine vials should be evaluated for the
maximum anticipated vial septum punctures to assess the risk of compromising
vial integrity and the potential for vial contamination. The extractable volume
of multi-dose vials should be validated. If multi-dose vaccine vials are supplied
as concentrate, an additional compatibility study should be conducted using the
proposed reconstitution solutions and an appropriate post-dilution hold-time
should be established. The pre-dilution and post-dilution specifications should
be set out and justified. Manufacturers should provide the NRA with adequate
data demonstrating the stability of the product under appropriate conditions of
storage, distribution and during use.
When a final vaccine contains more than one mRNA species (for
example, in a combination or multivalent vaccine, or an sa-mRNA consisting of
separate mRNAs) there may be additional considerations in the manufacture of
that final vaccine. One such consideration will be ensuring the appropriate ratio
of the different mRNAs in the formulation to optimize the expression of each
and to minimize immune interference (in the case of combination or multivalent
vaccines). Another consideration will be whether the mRNAs will be mixed
prior to encapsulation in the LNPs or whether each mRNA will be separately
encapsulated into LNPs and then a mixture of the two or more mRNA-LNPs
prepared. In either case, the approach selected should be described and justified
with relevant data.
123
WHO Expert Committee on Biological Standardization Seventy-fourth report
another should be noted and investigated. The data obtained from such studies,
as well as clinical trial outcomes with various lots, alongside product and process
knowledge and evaluation of the criticality of variations in specific attributes,
should be used as the basis for defining the vaccine specifications and acceptance
criteria to be used for routine lot release. Thus, a comprehensive analysis of the
initial commercial production lots should be undertaken to establish consistency
with regard to the identity, purity, strength/content/quantity, safety, additional
quality parameters, potency and stability of the mRNA vaccine but thereafter a
more limited series of tests may be appropriate, if agreed with the NRA.
When a final vaccine contains more than one mRNA species (for example,
in a combination or multivalent vaccine, or an sa-mRNA consisting of separate
mRNAs) there may be additional considerations in the control of that final vaccine.
Some of these considerations will be based on the approach taken in manufacture
124
Annex 3
– for example, whether the mRNAs were encapsulated together at the same time
as a mixture or were encapsulated separately and then the different mRNA-LNPs
mixed. This may then affect the size, charge and polydispersity of the LNPs. In
addition, validating the consistency of mixing is crucial to ensuring that each dose
contains the appropriate ratio of each of the mRNAs. Ensuring the proper ratios
in the total mRNA content of the final vaccine will be critical as the total mRNA
content is the basis for dosing. Identity testing should address the inclusion of each
mRNA, while still differentiating the vaccine from other products made in the
facility. If one drug substance or component (for example, the mRNA encoding
the replicon) is used in more than one vaccine or product made in the facility, then
such identity testing will also be crucial in preventing mix-ups.
As experience is gained in manufacturing consistency, post-approval
changes might permit reducing the testing and amount of supporting information
required through the use of process validation, product characterization and/or
a comparability protocol (31).
6.8.4.1 Identity
Each lot of vaccine should be subjected to an appropriate test to confirm the
identity of the final product and distinguish it from other products made in the
same facility or using the same equipment. If the vaccine contains more than one
mRNA species (for example, in a combination or multivalent vaccine, or an sa-
mRNA consisting of separate mRNAs), then the identity of each mRNA should
be confirmed. Confirmation of mRNA identity by sequence analysis should be
considered (see section 6.7.1.1 above).
vaccine contains more than one mRNA species (for example, in a combination
or multivalent vaccine, or an sa-mRNA consisting of separate mRNAs), then the
quantity of each mRNA should be measured and confirmation made that the
ratio of each mRNA to the other is as intended and the total mRNA dose has not
been exceeded.
These can include appearance (including presence of both visible and sub-visible
particulate matter), extractable volume and pH. Depending on the product
characteristics, the control of other attributes such as osmolality or viscosity may
also be important. For the final vaccine (drug product), additional attributes
should include lipid/polymer identification and content, nanoparticle size,
mRNA–lipid ratio and polydispersity index.
With respect to nanoparticle size, multiple point control should be
adopted similar to the control of nanoparticle-based therapeutic products, and the
test used for measurement of particle size should be specified, as the results will
be dependent upon the analytical method employed. The degree of encapsulation
of the mRNA in the LNP should also be regarded as a critical quality attribute as
non-encapsulated mRNA is considered to be unstable. Confirmation should be
provided that the structure of the final product does not change due to freeze-
126
Annex 3
6.8.4.6 Potency
The potency of each lot of the final vaccine should be determined using a suitably
quantitative and validated functional method(s). Different tests may be required
to control various aspects of potency (including functionality), which will likely
be disease specific. Immunogenicity in the vaccine recipient is a complex function
of the final vaccine properties, including delivery to target cells by its formulation
as well as expression of the mRNA-encoded protein(s) (which may include a self-
amplifying replicon component). Thus, potential in vitro potency assays may
include cell-based transfection systems or cell-free assays. Such methods would
demonstrate that the correctly sized protein of correct identity is expressed from
the mRNA. However, because potency should be analyzed on the basis of not only
the product type (in this case mRNA vaccines) but also the clinical indication of
the disease to be prevented, it is not possible to indicate a particular assay method
that should be used to measure potency. Scientific justification for the potency
test(s) selected to control the product should be provided and correlated with
clinical performance, as with all quality control tests.
When a new candidate vaccine against a new strain(s) is developed,
consideration should be given to ensuring that the potency assay(s) used is valid
for the strain change.
The potency specifications for mRNA vaccines should be set based on
the minimum dose used to demonstrate efficacy in clinical trials plus human
immunogenicity data. An upper limit should also be defined based on available
human safety data.
Animal-based assays tend to be highly variable and difficult to validate.
Consideration should therefore be given to the use of appropriate in vitro
alternative methods for potency evaluation. It is envisaged that, as with plasmid
DNA vaccines, a combination of biochemical or biophysical measures (such
as nucleic acid quantity and mRNA integrity) might be used to establish and
monitor the potency of mRNA vaccines. Manufacturers are encouraged to work
towards the goal of employing in vitro assays that are suitably quantitative and
assess function. However, it needs to be acknowledged that these measures
only account for the mRNA and not the impact of any formulation, adjuvant,
immunomodulators and so on, and the potency assessment of mRNA vaccines
127
WHO Expert Committee on Biological Standardization Seventy-fourth report
6.8.4.8.1 Stability
Adequate stability studies form an essential part of vaccine development.
To support commercial use, the stability of the final product in the container
proposed for use should therefore be determined and the results used to set a shelf-
life under appropriate storage conditions. Attributes that are stability-indicating
should be measured and these may include appearance (including visible and sub-
visible particulate matter), mRNA quantity, vaccine potency, mRNA integrity,
128
Annex 3
a storage site. The maximum duration of storage should be fixed with the
approval of the NRA based on the results of stability studies and should be such
as to ensure that all quality specifications for the final product, including the
minimum potency specified on the container or package, are maintained until
the end of shelf-life. During clinical trials, this period should ideally be at least
equal to the expected duration of the vaccine administration stage in the fully
enrolled clinical trial.
6.9 Records
The relevant guidance provided in WHO good manufacturing practices for
pharmaceutical products: main principles (21) should apply, as appropriate to
the level of development of the candidate vaccine.
6.11 Labelling
The guidance on labelling provided in WHO good manufacturing practices for
biological products (23) should be followed as appropriate. The label of the carton
enclosing one or more final containers, or the leaflet accompanying the container,
should include, at a minimum and as agreed with the NRA:
■ the common and trade names of the vaccine;
■ INN, if applicable;
■ the names and addresses of the manufacturer and distributer;
■ lot number;
130
Annex 3
7.1 Pharmacology/immunology/proof-of-concept
In addition to the types of studies discussed in the WHO guidelines above (18,
19), additional issues that the NRA might expect nonclinical studies to address
may include:
lipid ratio, and where the total amount of mRNAs and LNPs per
dose remain equal to or below that clinically tested) and where
an approved manufacturing process is used, then, depending on
NRA requirements, the nonclinical programme might be limited
to an immunogenicity study (or studies) or a challenge-protection
study (or studies) in a relevant animal model, if available. As much
safety information as feasible should be collected during these
immunogenicity or challenge-protection studies on the understanding
that such nonclinical proof-of-concept studies are generally
performed without full compliance to good laboratory practices
(GLP). If safety information on veterinary vaccines expressing related
antigens is available, this might also be useful and should be provided.
Any other information concerning the safety of the platform
technology used should also be provided for NRA consideration, for
example, prior toxicology and biodistribution study data.
■ Where the LNPs have been tested clinically with an unrelated
mRNA such that the target antigen is novel (that is, not related to
another antigen that has been clinically tested), then the approach
of limiting nonclinical studies to an immunogenicity or challenge-
protection study might not be sufficient. The decision regarding
what type of nonclinical safety/toxicology information should be
required might be guided by consideration of what and how much
is known about the natural disease in terms of its pathology. If
the natural disease is associated with immunopathology due to
cross-reactivity, molecular mimicry, autoimmunity, allergenicity or
immunity-associated disease enhancement, then toxicology studies
would likely be needed to ensure that the novel target antigen was
not associated with these effects. It should be noted that it may not
WHO Technical Report Series, No. 1039, 2022
where separate mRNAs are used) and to the total dose of LNPs with regard to
the maximally tolerable dose determined during the development of an mRNA
vaccine. For platform technologies, a maximally tolerable dose for a given
population may be suggested by the dose previously determined for vaccines (or
candidate vaccines) produced using that platform.
If boosting following a primary dose or series is being considered due to
waning effectiveness, careful evaluation of any increased frequency or severity
of local or systemic reactions should be performed. As with all vaccines, it
is recommended that a careful exploration of dose, timing and number of
immunizations (primary series and boosters if needed), and kinetics and
durability of immune responses be performed in preliminary clinical trials to
guide the design of the efficacy trial(s). Discussion of these issues can be found in
section 5.5 of the WHO Guidelines on clinical evaluation of vaccines: regulatory
expectations (20). In certain situations, a determination that booster doses are
needed might not be made until post-marketing data have been collected (for
example, indicating waning immunity or protection). The above Guidelines (20)
also discuss the boosting of licensed vaccines in section 5.6.1.2.3, which addresses
the situation in which an alternative posology to the licensed product may need
to be developed for the booster. Waning protection and the necessity for booster
doses is then discussed in section 6.3.8. (20). Differences between the vaccine
and circulating strains, including the potential need to add or replace strains,
are briefly discussed in section 5.3.3 and other sections that discuss influenza
vaccines, for which strain changes are frequently made.
It should be noted that during clinical trials or widespread use of
COVID-19 mRNA vaccines, immunologically relevant adverse events of particular
note (such as anaphylaxis or anaphylactoid reactions) have been observed (87,
88). Anaphylaxis is known to occur very rarely with all vaccines and is not
unique to mRNA vaccines. It is not yet known what aspect of the formulation
is associated with immunological adverse events and it is advised, as with other
vaccines, that individuals with known allergies to specific vaccine components
should not be vaccinated with vaccines containing such components (89–92).
Myocarditis and pericarditis have also been observed during COVID-19 mRNA
vaccine pharmacovigilance and appear to be associated – though the biological
mechanism and associated vaccine component have not yet been identified (93,
94). It should further be noted that recent publications by several regulatory
authorities provide useful relevant information, including publications by the
European Medicines Agency (71, 95), the Medicines and Healthcare products
Regulatory Agency (89, 96) and the US Food and Drug Administration (92, 97).
In line with the WHO Guidelines on clinical evaluation of vaccines:
regulatory expectations (20), the establishment and implementation of active
pharmacovigilance plans are recommended. In the specific case of COVID-19
139
WHO Expert Committee on Biological Standardization Seventy-fourth report
WHO has now published more than 70 Guidelines and Recommendations for
vaccines against specific diseases, any one or several of which may provide relevant
guidance on the evaluation of any given mRNA vaccine (15).
D. Boyce, Pfizer, Ambler, PA, the USA; Dr S. Chiyukula and Dr S. Simons, Sanofi,
Cambridge, MA, the USA; Dr R. Forrat, Sanofi, Lyons, France; Dr S. Gregory
and Dr G. Maruggi, GSK, Rockville, MD, the USA; Dr S. Lockhart, Pfizer,
New York, NY, the USA; and Dr F. Takeshita, Daiichi Sankyo, Japan. Individual
manufacturers/other industries: Dr T. Class, Translate Bio, the USA; Dr S. Gould,
Charles River Laboratories, Lyons, France; Dr A. Kuhn (20 and 21 April), Dr R.
Rizzi (20 and 21 April), Dr C. Lindemann (22 April) and Dr E. Lagkadinou (22
April), BioNTech SE, Germany; Dr K. Lindert, Arcturus Therapeutics, the USA;
Dr J. Maslow, GeneOne Life Science Inc., the USA; Dr J.M. Miller and Dr D.
Parsons, Moderna, Inc., the USA ; Dr F. Neske and Dr L. Oostvogels, CureVac
AG, Germany; Mr Y. Park, GeneOne Life Science Inc., Republic of Korea; and
Dr B. Ying, Suzhou Abogen Biosciences Co., Ltd. P.R., China. WHO staff: Dr
C. Ondari, Dr I. Knezevic, Dr T.Q. Zhou, Dr M. Friede, Dr B. Giersing, Ms
144
Annex 3
References
1. Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics – developing a new class of
drugs. Nat Rev Drug Discov. 2014;13(10):759–80 (https://www.nature.com/articles/
nrd4278?fbclid=IwAR0B8wG6qI3QCvTOPYSOAdlU6GiiM7FQFjD25u1V69BjcdmzhwWbufKOgGs,
accessed 22 September 2021).
2. Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A et al. Direct gene transfer into
mouse muscle in vivo. Science. 1990;247(4949 Pt 1):1465–8 (https://www.researchgate.net/
publication/21464693_Direct_Gene_Transfer_into_Mouse_Muscle_in_Vivo, accessed 22
September 2021).
145
WHO Expert Committee on Biological Standardization Seventy-fourth report
3. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines – a new era in vaccinology. Nat Rev
Drug Discov. 2018;17(4):261–79 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5906799/,
accessed 22 September 2021).
4. Karikó K, Muramatsu H, Welsh FA, Ludwig J, Kato H, Akira S et al. Incorporation of pseudouridine
into mRNA yields superior nonimmunogenic vector with increased translational capacity and
biological stability. Mol Ther. 2008;16(11):1833–40 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC2775451/, accessed 22 September 2021).
5. Thess A, Grund S, Mui BL, Hope MJ, Baumhof P, Fotin-Mleczek M et al. Sequence-engineered
mRNA without chemical nucleoside modifications enables an effective protein therapy in large
animals. Mol Ther. 2015;23(9):1456–64 (https://www.sciencedirect.com/science/article/pii/
S1525001616302738, accessed 22 September 2021).
6. Rauch S, Lutz J, Kowalczyk A, Schlake T, Heidenreich R. RNActive® technology: generation and
testing of stable and immunogenic mRNA vaccines. Methods Mol Biol. 2017;1499:89–107
(abstract: https://pubmed.ncbi.nlm.nih.gov/27987144/, accessed 22 September 2021).
7. Hinz T, Kallen K, Britten CM, Flamion B, Granzer U, Hoos A et al. The European regulatory
environment of RNA-based vaccines. Methods Mol Biol. 2017;1499:203–22 (abstract: https://
pubmed.ncbi.nlm.nih.gov/27987152/3, accessed 22 September 2021).
8. Jackson NAC, Kester KE, Casimiro D, Gurunathan S, DeRosa F. The promise of mRNA vaccines: a
biotech and industrial perspective. NPJ Vaccines. 2020;5(1):11 (https://www.nature.com/articles/
s41541-020-0159-8, accessed 22 September 2021).
9. Naik R, Peden K. Regulatory considerations on the development of mRNA vaccines. Curr Top
Microbiol Immunol. 2020 (abstract: https://link.springer.com/chapter/10.1007%2F82_2020_220,
accessed 22 September 2021).
10. Schmid A. Considerations for producing mRNA vaccines for clinical trials. Methods Mol Biol.
2017;1499:237–51 (abstract: https://pubmed.ncbi.nlm.nih.gov/27987154/, accessed 22
September 2021).
11. Hager G. Nonclinical safety testing of RNA vaccines. Methods Mol Biol. 2017;1499:253–72
(abstract: https://pubmed.ncbi.nlm.nih.gov/27987155/, accessed 22 September 2021).
12. Guideline on the chemistry, manufacture and control (CMC) of prophylactic COVID 19 mRNA
vaccines (current version). Center for Drug Evaluation. Beijing: National Medical Products
Administration; 2020 (https://www.ccfdie.org/en/gzdt/webinfo/2021/09/1632893004599121.
WHO Technical Report Series, No. 1039, 2022
17. Main outcomes of the meeting of the WHO Expert Committee on Biological Standardization held
from 9 to 10 December 2020. Geneva: World Health Organization; 2020 (https://www.who.int/
publications/m/item/ECBS-Executive-Summary.IF.IK.TW-15_Dec_2020, accessed 22 September
2021).
18. 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 September 2021).
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; https://www.who.int/
biologicals/areas/vaccines/TRS_987_Annex2.pdf?ua=1, accessed 22 September 2021).
20. Guidelines on clinical evaluation of vaccines: regulatory expectations. In: WHO Expert Committee
on Biological Standardization: sixty-seventh report. Geneva: World Health Organization; 2017:
Annex 9 (WHO Technical Report Series, No. 1004; http://www.who.int/biologicals/expert_
committee/WHO_TRS_1004_web_Annex_9.pdf?ua=1, accessed 22 September 2021).
21. 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; 2014: Annex 2 (WHO Technical Report Series, No. 986; http://www.who.
int/medicines/areas/quality_safety/quality_assurance/TRS986annex2.pdf?ua=1, accessed 22
September 2021).
22. Good manufacturing practices: supplementary guidelines for the manufacture of pharmaceutical
excipients. In: WHO Expert Committee on Specifications for Pharmaceutical Preparations:
thirty-fifth report. Geneva: World Health Organization; 1999: Annex 5 (WHO Technical Report
Series, No. 885; https://www.who.int/medicines/areas/quality_safety/quality_assurance/
SupplementaryGMPPharmaceuticalExcipientsTRS885Annex5.pdf?ua=1, accessed 22 September
2021).
23. 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; http://www.who.int/biologicals/areas/vaccines/
Annex_2_WHO_Good_manufacturing_practices_for_biological_products.pdf?ua=1, accessed
22 September 2021).
24. 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 (https://www.who.
int/docs/default-source/medicines/norms-and-standards/guidelines/production/trs961-annex6-
gmp-sterile-pharmaceutical-products.pdf?sfvrsn=61682f0c_0, accessed 22 September 2021).
25. 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; https://apps.who.int/iris/bitstream/
handle/10665/37340/WHO_TRS_850.pdf?sequence=1, accessed 9 November 2021).
26. WHO guidelines on transmissible spongiform encephalopathies in relation to biological and
pharmaceutical products. Geneva: World Health Organization; 2003 (https://apps.who.int/iris/
handle/10665/68932, accessed 9 November 2021).
147
WHO Expert Committee on Biological Standardization Seventy-fourth report
27. 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 22 September 2021).
28. 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://www.who.int/medicines/areas/quality_safety/quality_assurance/
ModelGuidanceForStorageTransportTRS961Annex9.pdf, accessed 22 September 2021).
29. Guidelines on the stability evaluation of vaccines for use under extended controlled temperature
conditions. In: WHO Expert Committee on Biological Standardization: sixty-sixth report. Geneva:
World Health Organization; 2016: Annex 5 (WHO Technical Report Series, No. 999; http://www.
who.int/biologicals/areas/vaccines/Annex_5_Guidelines_on_Stability_evaluation_vaccines_
ECTC.pdf?ua=1, accessed 22 September 2021).
30. 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 22 September 2021).
31. Guidelines on procedures and data requirements for changes to approved vaccines. In: WHO Expert
Committee on Biological Standardization: sixty-fifth report. Geneva: World Health Organization;
2015: Annex 4 (WHO Technical Report Series, 993; http://www.who.int/biologicals/vaccines/
Annex4_Guidelines_changes_to_approved_vaccines_eng.pdf?ua=1, accessed 22 September
2021).
32. WHO policy statement: multi-dose vial policy (MDVP). Handling of multi-dose vaccine vials after
opening. Revision 2014. Geneva: World Health Organization; 2014 (Document WHO/IVB/14.07;
http://apps.who.int/iris/bitstream/10665/135972/1/WHO_IVB_14.07_eng.pdf, accessed 22
September 2021).
33. Krammer F. SARS-CoV-2 vaccines in development. Nature. 2020;586(7830):516–27 (https://www.
nature.com/articles/s41586-020-2798-3, accessed 23 September 2021).
34. Zhong Z, Mc Cafferty S, Combes F, Huysmans H, De Temmerman J, Gitsels A et al. mRNA therapeutics
deliver a hopeful message. Nano Today. 2018;23:16–39 (abstract: https://www.sciencedirect.com/
science/article/pii/S1748013218301099, accessed 23 September 2021).
WHO Technical Report Series, No. 1039, 2022
35. Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S et al. Safety and efficacy of the
BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med. 2020;383(27):2603–15 (https://doi.org/10.1056/
NEJMoa2034577, accessed 23 September 2021).
36. Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R et al. Efficacy and safety of the mRNA-
1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384(5):403–16 (https://www.nejm.org/doi/
full/10.1056/NEJMoa2035389, accessed 23 September 2021).
37. Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M et al. COVID-19 vaccine
BNT162b1 elicits human antibody and TH1 T cell responses. Nature. 2020;586(7830):594–9
(https://www.nature.com/articles/s41586-020-2814-7.pdf, accessed 23 September 2021).
38. Lundstrom K. Nanoparticle-based delivery of self-amplifying RNA. Gene Ther. 2020;27(5):183–5
(https://www.nature.com/articles/s41434-020-0132-1, accessed 23 September 2021).
39. Bertrand N, Leroux J-C. The journey of a drug-carrier in the body: an anatomo-physiological
perspective. J Control Release. 2012;161(2):152–63 (https://www.researchgate.net/
publication/51723559_The_journey_of_a_drug-carrier_in_the_body_An_anatomo-
148
Annex 3
150
Annex 3
64. Getting started with vaccine vial monitors. Geneva: World Health Organization; 2002 (Document
WHO/V&B/02.35; https://apps.who.int/iris/bitstream/handle/10665/67806/WHO_V-B_02.35_
eng.pdf;sequence=1, accessed 2 October 2021).
65. Bar-codes, QR codes and Vaccine Vial Monitors in the context of COVID-19 vaccines. Geneva: World
Health Organization; 2020 (https://www.who.int/publications/m/item/bar-codes-qr-codes-and-
vaccine-vial-monitors-in-the-context-of-covid-19-vaccines, accessed 3 October 2021).
66. Stokes A, Pion J, Binazon O, Laffont B, Bigras M, Dubois G et al. Nonclinical safety assessment
of repeated administration and biodistribution of a novel rabies self-amplifying mRNA vaccine
in rats. Regul Toxicol Pharmacol. 2020;113:104648 (abstract: https://pubmed.ncbi.nlm.nih.
gov/32240713/, accessed 3 October 2021).
67. Lou G, Anderluzzi G, Schmidt ST, Woods S, Gallorini S, Brazzoli M et al. Delivery of self-amplifying
mRNA vaccines by cationic lipid nanoparticles: the impact of cationic lipid selection. J Control
Release. 2020;325:370–9 (abstract: https://pubmed.ncbi.nlm.nih.gov/32619745/, accessed 3
October 2021).
68. Pardi N, Tuyishime S, Muramatsu H, Kariko K, Mui BL, Tam YK et al. Expression kinetics of
nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J Control
Release. 2015;217:345–51 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4624045/, accessed
3 October 2021).
69. Bahl K, Senn JJ, Yuzhakov O, Bulychev A, Brito LA, Hassett KJ et al. Preclinical and clinical
demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza
viruses. Mol Ther. 2017;25(6):1316–27 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC5475249/#!po=0.505051, accessed 3 October 2021).
70. Liang F, Lindgren G, Lin A, Thompson EA, Ols S, Röhss J et al. Efficient targeting and activation
of antigen-presenting cells in vivo after modified mRNA vaccine administration in rhesus
macaques. Mol Ther. 2017;25(12):2635–47 (https://www.sciencedirect.com/science/article/pii/
S1525001617303659, accessed 3 October 2021).
71. Assessment report. Comirnaty. Common name: COVID-19 mRNA vaccine (nucleoside-modified).
Procedure no. EMEA/H/C/005735/0000. Amsterdam: European Medicines Agency; 2021
(Document EMA/707383/2020 Corr.1; pp. 46–47; https://www.ema.europa.eu/documents/
assessment-report/comirnaty-epar-public-assessment-report_en.pdf, accessed 3 October 2021).
72. Obeid MA, Tate RJ, Mullen AB, Ferro VA. Lipid-based nanoparticles for cancer treatment. In:
Grumezescu AM, editor. Lipid nanocarriers for drug targeting. Norwich (NY): William Andrew
Publishing; 2018;313–59 (abstract: https://www.sciencedirect.com/science/article/pii/
B9780128136874000086, accessed 3 October 2021).
73. Marlowe JL, Akopian V, Karmali P, Kornbrust D, Lockridge J, Semple S. Recommendations of the
Oligonucleotide Safety Working Group’s Formulated Oligonucleotide Subcommittee for the safety
assessment of formulated oligonucleotide-based therapeutics. Nucleic Acid Ther. 2017;27(4):183–
96 (abstract: https://pubmed.ncbi.nlm.nih.gov/28609186/, accessed 3 October 2021).
74. CHMP Safety Working Party’s response to the Paediatric Committee regarding the use of
PEGylated drug products in the paediatric population. London: European Medicines Agency;
2012 (Document EMA/CHMP/SWP/647258/2012; https://www.ema.europa.eu/en/chmp-safety-
working-partys-response-paediatric-committee-regarding-use-pegylated-drug-products,
accessed 2 October 2021).
75. Turecek PL, Bossard MJ, Schoetens F, Ivens IA. PEGylation of biopharmaceuticals: a review of
chemistry and nonclinical safety information of approved drugs. J Pharm Sci. 2016;105(2):460–75
(abstract: https://www.sciencedirect.com/science/article/abs/pii/S0022354915001306, accessed
151
WHO Expert Committee on Biological Standardization Seventy-fourth report
3 October 2021).
76. Liu G, Li Y, Yang L, Wei Y, Wang X, Wang Z et al. Cytotoxicity study of polyethylene glycol
derivatives. RSC Adv. 2017;7(30):18252–9 (https://pubs.rsc.org/en/content/articlelanding/2017/
ra/c7ra00861a, accessed 9 November 2021).
77. Turecek PL, Siekmann J. 4 - PEG–protein conjugates: nonclinical and clinical toxicity considerations.
In: Pasut G, Zalipsky S, editors. Polymer-protein conjugates. Elsevier; 2020;61–101 (abstract:
https://www.sciencedirect.com/science/article/pii/B9780444640819000048, accessed 3 October
2021).
78. Johnson AA, Ray AS, Hanes J, Suo Z, Colacino JM, Anderson KS et al. Toxicity of antiviral nucleoside
analogs and the human mitochondrial DNA polymerase. J Biol Chem. 2001;276(44):40847–57
(https://www.sciencedirect.com/science/article/pii/S0021925820779317, accessed 3 October
2021).
79. Moyle G. Toxicity of antiretroviral nucleoside and nucleotide analogues: is mitochondrial toxicity
the only mechanism? Drug Saf. 2000;23(6):467–81 (abstract: https://pubmed.ncbi.nlm.nih.
gov/11144657/, accessed 3 October 2021).
80. Nucleoside analogues. In: LiverTox: clinical and research information on drug-induced liver injury.
Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases; 2012 (Overview:
http://www.ncbi.nlm.nih.gov/books/NBK548938/, accessed 3 October 2021).
81. Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart S et al. Phase I/II study of COVID-19
RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–93 (https://www.nature.com/articles/
s41586-020-2639-4, accessed 9 November 2021).
82. Bovay A, Speiser DE, Marraco SAF. Early drop of circulating T cells negatively correlates with
the protective immune response to Yellow Fever vaccination. Hum Vaccines Immunother.
2020;16(12):3103–10 (https://www.tandfonline.com/doi/full/10.1080/21645515.2020.1750249,
accessed 2 October 2021).
83. Faguet GB. The effect of killed influenza virus vaccine on the kinetics of normal human lymphocytes.
J Infect Dis. 1981;143(2):252–8 (abstract: https://pubmed.ncbi.nlm.nih.gov/7217721/, accessed 3
October 2021).
84. Alberer M, Gnad-Vogt U, Hong HS, Mehr KT, Backert L, Finak G et al. Safety and immunogenicity
of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-
human phase 1 clinical trial. Lancet. 2017;390(10101):1511–20 (Summary: https://www.thelancet.
com/journals/lancet/article/PIIS0140-6736(17)31665-3/fulltext, accessed 3 October 2021).
WHO Technical Report Series, No. 1039, 2022
85. Richner JM, Jagger BW, Shan C, Fontes CR, Dowd KA, Cao B et al. Vaccine mediated protection
against Zika virus-induced congenital disease. Cell. 2017;170(2):273–283.e12 (https://www.
sciencedirect.com/science/article/pii/S0092867417307596, accessed 3 October 2021).
86. Heidenreich R, Jasny E, Kowalczyk A, Lutz J, Probst J, Baumhof P et al. A novel RNA-based adjuvant
combines strong immunostimulatory capacities with a favorable safety profile. Int J Cancer.
2015;137(2):372–84 (https://onlinelibrary.wiley.com/doi/full/10.1002/ijc.29402, accessed 3
October 2021).
87. Shimabukuro TT, Cole M, Su JR. Reports of anaphylaxis after receipt of mRNA COVID-19 vaccines in
the US – December 14, 2020-January 18, 2021. JAMA. 2021;325(11):1101–2 (https://jamanetwork.
com/journals/jama/fullarticle/2776557, accessed 3 October 2021).
88. Klein NP, Lewis N, Goddard K, Fireman B, Zerbo O, Hanson KE et al. Surveillance for adverse events
after COVID-19 mRNA vaccination. JAMA. 2021;326(14):1390–9 (https://jamanetwork.com/
journals/jama/fullarticle/2784015, accessed 9 November 2021).
152
Annex 3
89. Regulatory approval of Pfizer/BioNTech vaccine for COVID-19 [website]. London: Medicines
and Healthcare products Regulatory Agency (https://www.gov.uk/government/publications/
regulatory-approval-of-pfizer-biontech-vaccine-for-covid-19, accessed 9 November 2021).
90. Castells MC, Phillips EJ. Maintaining safety with SARS-CoV-2 vaccines. N Engl J Med. 2021;384(7):643–
9 (https://www.nejm.org/doi/10.1056/NEJMra2035343, accessed 3 October 2021).
91. Sellaturay P, Nasser S, Ewan P. Polyethylene glycol-induced systemic allergic reactions (anaphylaxis).
J Allergy Clin Immunol Pract. 2021;9(2):670–5 (abstract: https://www.sciencedirect.com/science/
article/abs/pii/S2213219820310072, accessed 3 October 2021).
92. Moderna COVID-19 vaccine Emergency Use Authorization. Review Memorandum. US Food
and Drug Administration; 2020 (https://www.fda.gov/media/144673/download, accessed 9
November 2021).
93. Gargano JW, Wallace M, Hadler SC, Langley G, Su JR, Oster ME et al. Use of mRNA COVID-19
vaccine after reports of myocarditis among vaccine recipients: update from the Advisory
Committee on Immunization Practices – United States, June 2021. MMWR Morb Mortal Wkly
Rep. 2021;70(27):977–82 (http://www.cdc.gov/mmwr/volumes/70/wr/mm7027e2.htm?s_
cid=mm7027e2_w, accessed 10 November 2021).
94. Høeg TB, Krug A, Stevenson J, Mandrola J. SARS-CoV-2 mRNA vaccination-associated myocarditis
in children ages 12–17: a stratified national database analysis (online preprint: https://www.
medrxiv.org/content/10.1101/2021.08.30.21262866v1, accessed 10 November 2021).
95. Spikevax (previously COVID-19 Vaccine Moderna). European Medicines Agency; 2021 (https://
www.ema.europa.eu/en/medicines/human/EPAR/spikevax, accessed 10 November 2021).
96. Regulatory approval of Spikevax (formerly COVID-19 Vaccine Moderna) [website]. London:
Medicines and Healthcare products Regulatory Agency; 2021 (https://www.gov.uk/government/
publications/regulatory-approval-of-covid-19-vaccine-moderna, accessed 10 November 2021).
97. Comirnaty [website]. US Food and Drug Administration; 2021 (https://www.fda.gov/vaccines-
blood-biologics/comirnaty, accessed 10 November 2021).
98. Goodman S. Considerations for placebo-controlled trial design if an unlicensed vaccine becomes
available. Presentation to the U.S. Food and Drug Administration Vaccines and Related Biological
Products Advisory Committee, 10 December 2020 (https://www.fda.gov/media/144354/
download, accessed 4 October 2021).
99. Goodman S. Considerations for placebo-controlled trial design if an unlicensed vaccine becomes
available. Presentation to the U.S. Food and Drug Administration Vaccines and Related Biological
Products Advisory Committee, 17 December 2020 (https://www.fda.gov/media/144582/
download, accessed 4 October 2021).
100. Expert consultation on the use of placebos in vaccine trials. Meeting report. Geneva: World Health
Organization; 2013 (http://apps.who.int/iris/bitstream/10665/94056/1/9789241506250_eng.pdf,
accessed 4 October 2021).
101. Guideline on influenza vaccines – non-clinical and clinical module. London: European Medicines
Agency; 2016 (Document EMA/CHMP/VWP/457259/2014; https://www.ema.europa.eu/en/
documents/scientific-guideline/influenza-vaccines-non-clinical-clinical-module_en.pdf,
accessed 4 October 2021).
102. Weir JP, Gruber MF. An overview of the regulation of influenza vaccines in the United States.
Influenza Other Respir Viruses. 2016;10(5):354–60 (https://www.ncbi.nlm.nih.gov/pmc/articles/
PMC4947948/?_escaped_fragment_=po=0.925926, accessed 4 October 2021).
153
WHO Expert Committee on Biological Standardization Seventy-fourth report
103. Coronavirus disease (COVID-19) [website]. Geneva: World Health Organization (https://www.
who.int/teams/health-product-and-policy-standards/standards-and-specifications/vaccine-
standardization/coronavirus-disease-(covid-19), accessed 4 October 2021).
104. COVID-19 vaccines [website]. Geneva: World Health Organization (https://www.who.int/
emergencies/diseases/novel-coronavirus-2019/covid-19-vaccines, accessed 10 November 2021).
105. Reflection paper on the regulatory requirements for vaccines intended to provide protection
against variant strain(s) of SARS-CoV-2. Amsterdam: European Medicines Agency; 2021 (Document
EMA/117973/2021; https://www.ema.europa.eu/en/documents/scientific-guideline/reflection-
paper-regulatory-requirements-vaccines-intended-provide-protection-against-variant_en.pdf,
accessed 4 October 2021).
106. Access Consortium’s guidance on strain changes in authorised COVID-19 vaccines [website].
Singapore: Health Sciences Authority (https://www.hsa.gov.sg/therapeutic-products/
international-collaboration/access/guidance-strain-changes-in-authorised-covid-19-vaccines,
accessed 4 October 2021).
107. ICMRA COVID-19 Virus Variants Workshop [website]. International Coalition of Medicines Regulatory
Authorities, 10 February 2021 (http://www.icmra.info/drupal/en/covid-19/10february2021,
accessed 4 October 2021).
108. Recommendations to assure the quality, safety and efficacy of influenza vaccines (human, live
attenuated) for intranasal administration. In: WHO Expert Committee on Biological Standardization:
sixtieth report. Geneva: World Health Organization; 2013: Annex 4 (WHO Technical Report
Series, No. 977; https://www.who.int/publications/m/item/influenza-attenuated-intranasal-
administration-annex-4-trs-no-977, accessed 4 October 2021).
109. Recommendations for the production and control of influenza vaccine (inactivated). In: WHO
Expert Committee on Biological Standardization: fifty-fourth report. Geneva: World Health
Organization; 2005: Annex 3 (WHO Technical Report Series, No. 927; https://www.who.int/
biologicals/publications/trs/areas/vaccines/influenza/ANNEX%203%20InfluenzaP99-134.
pdf?ua=1, accessed 4 October 2021).
WHO Technical Report Series, No. 1039, 2022
154
Annex 4
New and replacement WHO international reference
standards for biological products
The provision of global measurement standards is a core normative WHO activity.
WHO international reference standards are widely used by manufacturers,
regulatory authorities and academic researchers in the development and
evaluation of biological products. The timely development of new reference
standards is crucial in harnessing the benefits of scientific advances in new
biologicals and in vitro diagnosis. At the same time, management of the existing
inventory of WHO international reference standards requires an active and
carefully planned programme of work to replace established materials before
existing stocks are exhausted.
The considerations and guiding principles used to assign priorities
and develop the programme of work in this area have previously been set out
as WHO Recommendations.7 In order to facilitate and improve transparency
in the priority-setting process, a simple tool was developed as Appendix 1 of
these WHO Recommendations. This tool describes the key considerations taken
into account when assigning priorities, and allows stakeholders to review and
comment on any new proposals being considered for endorsement by the WHO
Expert Committee on Biological Standardization.
A list of current WHO international reference standards for biological
products is available at: https://www.who.int/health-topics/Biologicals#tab=tab_1.
At its meetings held via video conference on 18–22 October 2021, the
WHO Expert Committee on Biological Standardization made the changes shown
below to the previous list. Each of the WHO international reference standards
shown in this table should be used in accordance with their instructions for use
(IFU).
7
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 26 October 2021).
155
WHO Expert Committee on Biological Standardization Seventy-fourth report
Additions 8
Material Unitage Status
Biotherapeutics other than blood products
Follicle-stimulating 137 IU/ampoule Third WHO International
hormone (human, Standard
recombinant) for bioassay
Blood products and related substances
von Willebrand factor VWF:Ag 12.0 IU/ampoule Third WHO
(concentrate) VWF:RCo 8.7 IU/ampoule International Standard
VWF:CB 9.8 IU/ampoule
VWF:GPIbR 8.6 IU/ampoule
VWF:GPIbM 7.3 IU/ampoule
Ferritin (human, 10.5 μg/ampoule Fourth WHO
recombinant) Expanded uncertainty International Standard
limits = 10.2–10.8 μg/
ampoule (95% confidence;
k = 2.23)
In vitro diagnostics
Mycobacterium tuberculosis 6.3 log10 IU/vial First WHO International
(H37Rv) DNA for NAT-based Standard
assays
Varicella zoster virus DNA 7.0 log10 IU/vial First WHO International
for NAT-based assays Standard
Anti-Lassa virus 25 IU/ampoule for First WHO International
immunoglobulin G neutralizing antibody Standard
WHO Technical Report Series, No. 1039, 2022
8
Unless otherwise indicated, all materials are held and distributed by the National Institute for Biological
Standards and Control, Potters Bar, Herts, EN6 3QG, the United Kingdom.
156
Annex 4
157
SELECTED WHO PUBLICATIONS OF RELATED INTEREST
Website: https://www.who.int/health-topics/Biologicals#tab=tab_1
ISBN 9789240046870