Nipah Virus Assays and Animal Models for Vaccine Development - Landscape Analysis, January 2021

Page created by Floyd Scott
 
CONTINUE READING
Nipah Virus Assays and Animal Models for Vaccine Development - Landscape Analysis, January 2021
Nipah Virus Assays and
Animal Models for Vaccine
Development
Landscape Analysis, January 2021

Author:
Albert Price, IAVI

Secondary authors/reviewers:
Donata Sizemore, IAVI
Thomas Hassell, IAVI
Raúl Gómez Román, CEPI
Johan Holst, CEPI
Paul Kristiansen, CEPI
Nipah Virus Assays and Animal Models for Vaccine Development - Landscape Analysis, January 2021
TABLE OF CONTENTS
I. Introduction                                                                4

II. Background                                                                  5

      1. Epidemiology                                                           5

      2. NiV Clinical Features and Pathogenesis in Humans                       9

      3. Diagnosis and Treatment                                              10

      4. NiV Molecular Biology and Structure                                    11

      5. Vaccine Development                                                   13

III. Standardization of Assays and Animal Models                               18

IV. NiV Serological Assays                                                     21

      1. Detection of antigen – specific serum IgG                             21

      2. Detection of serum neutralizing antibodies                           22

V. NiV Animal Models                                                          27

      1. Syrian Golden Hamster                                                29

      2. Ferret                                                               32

      3. African Green Monkey (AGM)                                           34

VI. Conclusions                                                               38

VII. References                                                               40

VIII. Statement of Support                                                    46

      1. Article H.20. Publication And Publicity                              46

2                 Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Nipah Virus Assays and Animal Models for Vaccine Development - Landscape Analysis, January 2021
LIST OF FIGURES
Figure 1. Geographic distribution of Pteropid fruit bats
           and Henipavirus outbreaks (Enchery and Horvat, 2017)                5

Figure 2. NiV structure and organization of the 18.2 kB ssRNA
           (-) genome (Sun et al., 2018).                                      11

Figure 3. The Henipavirus infection and replication cycle
           (Aguilar and Lee, 2011).                                           12

LIST OF TABLES
Table 1. NiV Outbreaks by Year and Location*                                   7

Table 2. Differences in Clinical and Epidemiological Characteristics
         Between NiV Malaysia and Bangladesh Outbreaks                        8

Table 3.  NiV Viral-vector Vaccine Candidates Tested in Animals               15

Table 4. NiV Submit Vaccine Candidates Tested in Animals                      16

Table 5. NiV vaccine candidates supported by CEPI                             17

Table 6. Benefits and Potential Challenges of Implementing
          Biological Standards for NiV Vaccine Development                   20

Table 7. Pros and Cons of NiV Serological Assays                             24

Table 8. Serological Assays Used in NiV Pre-Clinical Vaccine Studies         25

Table 9. Serological Assays Used in Other NiV Research Studies               26

Table 10. S
           ummary of clinical signs and pathology in the NiV hamster,
          ferret and AGM challenge models                                    28

Table 11. NiV Hamster Model Challenge Studies                                30

Table 12. NiV Hamster Model Challenge Studies, Continued                      31

Table 13. NiV Ferret Model Challenge Studies                                 33

Table 14. NiV African Green Monkey Model Challenge Studies                   35

Table 15. NiV African Green Monkey Model Challenge Studies, Continued        36

Table 16. Pros and Cons of the Major NiV Animal Challenge Models             37

3                Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
I. INTRODUCTION
Nipah is an emerging, zoonotic viral disease that causes severe neurologic
and respiratory symptoms and has an overall case fatality rate of 59%.

Although relatively rare and                           currently targeted for development   To this end, CEPI is focusing on
currently confined to sporadic                         of prophylactic vaccines as an       developing biological standards,
outbreaks in southern and                              urgent priority2 and is actively     validating assays and supporting
southeast Asia (including Malaysia,                    supporting efforts toward a          the development and refinement of
Singapore, Bangladesh and India),                      protective NiV vaccine.              animal models for three emerging
the extreme virulence, lack of a                                                            diseases in its vaccine development
vaccine or effective therapeutic                       Development of new vaccines          portfolio: Nipah, MERS-CoV
options, broad species tropism                         against any disease is most          and Lassa. The purpose of this
and wide geographical distribution                     efficient when there is              Landscape Analysis, supported by
of the Nipah virus’ (NiV) primary                      standardization of key R&D tools,    NIH/NIAID/DMID and prepared for
animal reservoir (Pteropid fruit                       particularly analytical methods,     CEPI, is to analyze the current state
bats) led the World Health                             reagents and animal models,          of NiV assays and animal models
Organization (WHO) to label NiV                        so that experimental results         currently in use within the context
a “Priority Pathogen” for the                          from different investigators and     of NiV biology, epidemiology,
development of effective medical                       developers can be directly and       and vaccine development. This
countermeasures (MCMs), and in                         confidently compared. CEPI has       document will serve both as an
2017 developed a Target Product                        identified a set of research and     internal resource at CEPI to guide
Profile (TPP) for a NiV vaccine.1                      development activities needed to     scientific discussions and as an
The Coalition for Epidemic                             accelerate vaccine development       external resource to inform the
Preparedness Innovations (CEPI)                        by promoting standardization,        Nipah scientific community.
has selected NiV as one of seven                       transparency and comparability
emerging infectious diseases                           between vaccine candidates.

1
    https://www.who.int/blueprint/priority-diseases/key-action/Nipah_virus_vaccineTPP.pdf
2
    https://cepi.net/research_dev/priority-diseases/

4                        Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
II. BACKGROUND
1. Epidemiology
Fruit bats (‘Flying Foxes’) of the                  concern over the potential spread     Caledonia, and Papua New Guinea
family Pteropodidae, particularly                   of NiV, is illustrated in Figure 1.   (Sun et al., 2018). More recently,
those of the genus Pteropus, are the                                                      a six-year study of Pteropus medius
primary animal reservoir for NiV                    Pteropid bats of the genus Eidolon    bats in Bangladesh indicates
and are asymptomatically infected                   range over most of the African        that Nipah virus may be more
by the virus. Even experimental                     continent and have been found         widespread than previously
infections with very high doses                     to be seropositive for NiV, and       thought: bats throughout the
of NiV cause only sub-clinical                      thus are potentially an additional    country, and not just those in what
infection in fruit bats and viremia                 reservoir (Enchery and Horvat,        is referred to as the “Nipah belt”,
has not been reported, although                     2017). A field survey found           had similar patterns of Nipah virus
the animals do seroconvert against                  that 9% to 25% of fruit bats in       infection throughout the year.3
the virus and virus shedding has                    Malaysia, Cambodia, Thailand
been observed, albeit rarely and                    and Bangladesh were seropositive
only in urine (Geisbert et al., 2012;               for NiV (Sharma et al., 2019) and
Middleton et al., 2007). The broad                  NiV-seropositive bats have also
geographical distribution of these                  been found in China, Vietnam,
animals, one of the factors driving                 Indonesia, Madagascar, New

Figure 1. Geographic distribution of Pteropid fruit bats and
Henipavirus outbreaks (Enchery and Horvat, 2017)

3
    Epstein et al. PNAS 2020: https://doi.org/10.1073/pnas.2000429117

5                        Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
A number of domesticated animals        and Tan, 2014). In the 2014
– pigs, dogs, cats and horses – can     NiV outbreak in the Philippines
be infected by NiV (Geisbert et al.,    (Malaysia strain), the majority
2012), but the virulence and rate of    of cases were acquired by eating
infection is variable. Mortality in     contaminated horse meat or
suckling pigs is high (40%) and 1-6     participating in slaughtering
month-old pigs show respiratory         horses, and the rest were likely due
and neurological symptoms, but          to human to human transmission.
mortality is less than 5%. Adult        65% of the cases presented with
pigs show less serious respiratory      an acute encephalitic syndrome
signs and mortality is rare (McLean     and the overall case fatality rate
and Graham, 2019). During NiV           was 53% (Ching et al., 2015).
outbreaks in Malaysia many dogs
on pig farms were found to be           The NiV outbreaks in Bangladesh
NiV-seropositive, but only 2 had        and India beginning in 2001
active disease (Hooper et al., 2001).   showed a distinct pattern of
Horses can be infected by NiV           transmission and symptomology.
(Hooper et al., 2001), most likely      Humans were infected by drinking
from eating fruit contaminated by       raw date palm sap contaminated
bats, and were intermediate hosts       with bat saliva or urine, and there
in an outbreak in the Philippines       was no intermediate animal host.
in 2014. During that outbreak           There was a higher incidence
investigations found disease among      of respiratory illness (69% and
horses including neurological           a higher fatality rate (75%; see
signs and 10 deaths. Four (4) cats      Table 1). Patients infected with the
and a dog were also likely infected     Bangladesh strain (NiV-B) had
by eating horse meat and died           higher NiV RNA levels in the blood
(Ching et al., 2015). However, no       and more virus in oral secretions
published reports of cats or dogs       (Hossain et al., 2008). Finally,
serving as intermediate hosts for       there was evidence of human-to-
transmission of NiV have been           human transmission, primarily
found.                                  to healthcare workers or family
                                        caregivers, in the Indian and
The locations and human fatality        Bangladeshi outbreaks (Chadha et
rates of all NiV outbreaks to date      al., 2006). The mechanism(s) of
are summarized in Table 1.              human to human transmission has
The first reported NiV outbreaks        not been conclusively established,
occurred in Malaysia and Singapore      but exposure to bodily fluids
in 1998-99. In those outbreaks          (saliva, cough, vomit, blood)
the majority of human cases were        elevates the risk of transmission
due to contact with infected pigs       compared to physical contact
that had acquired NiV by eating         alone or being near the patient
fruit contaminated by bat saliva,       (Kumar et al., 2019). Experiments
urine, or feces. Humans (mostly         in non-human primates have
pig farmers and slaughterhouse          demonstrated infection via small
workers) acquired NiV from              and medium particle size aerosols
infected pig urine or respiratory       (Cong et al., 2017; Hammoud
secretions. A majority of cases         et al., 2018). Comparisons in
were characterized by an acute          transmission rates between the
encephalitic syndrome. The case         Malaysia and Bangladesh outbreaks
fatality rate was 40% (see Table        have not been found in the
1) and, at the time, there was          published literature.
inconclusive evidence of human-
to-human transmission (Ahmad

6                 Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Table 1. NiV Outbreaks by Year and Location*

                                                                                                     Number of   Number of    Case
      Month/Year                Country                              Location
                                                                                                       Cases      Deaths   Fatality (%)

    Sep 1998 – Apr 1999          Malaysia                   Perak, Selangor, Negeri Sebilan             265         105         40

         Mar 1999               Singapore                             Singapore                         11           1           9

      Jan – Feb 2001              India                                Siliguri                         66          45          68

      Apr – May 2001                                                  Meherpur                          13           9          69

         Jan 2003                                                      Naogaon                          12           8          67

         Jan 2004                                                      Rajbari                          31           23         74

         Apr 2004                                                      Faridpur                         36           27         75
                                Bangladesh
      Jan – Mar 2005                                                   Tangail                          12           11         92

      Jan – Feb 2007                                                 Thakurgaon                          7           3          43

         Mar 2007                                               Kushtia, Pabna, Tatore                   8           5          63

         Apr 2007                                                      Naogaon                           3           1          33

         Apr 2007                 India                                 Nadia                            5           5          100

         Feb 2008                                                     Manikgon                           4           4          100

         Apr 2008                                                  Rajbari, Faridpur                     7           5           71

         Jan 2009                                           Gaibandha, Rangpur, Nilphamari               3           0           0

         Jan 2009                                                      Rajbari                           1           1          100

      Feb – Mar 2010                                   Faridpur, Rajbari, Gopalganj, Madaripur          16           14         87.5

                                Bangladesh          Lalmohirhat, Dinajpur, Comilla, Nilphamari ,
      Jan – Feb 2011                                                                                    44          40           91
                                                                     Rangpur

         Feb 2012                                  Joypurhat, Rajshahi, Tatore, Rajbari, Gopalganj      12           10         83

                                                   Gaibandha, Natore, Rajshahi,Naogaon, Rajbari,
      Jan – Feb 2013                                                                                    24           21         87.5
                                                          Pabna, Jhenaidah, Mymensingh

                                                       Manikganj, Magura, Faridpur, Rangpur,
         Feb 2014                                      Shaariatpur, Kushtia, Rajshahi, Tatore,          18           9          50
                                                       Dinajpur, Chapai Nawabganj, Naogaon

     Mar – May 2014             Philippines                      Tinalon, Midtungok                     17           9          53

                                                     Nilphamari, Pnchoghor, Faridpur, Magura,
         Feb 2015               Bangladesh                                                               9           6          67
                                                                 Naugaon, Rajbari

         May 2018                 India                        Kozhikode, Malappuram                    19           17         89

                                            Overall Total                                               643         379         58.9

                              Malaysia/Singapore/Philippines Total                                      293         115         39.2

                                     Bangladesh/India Total                                             350         264         75.4

*Adapted from (Thakur and Bailey, 2019) and (Sharma et al., 2019)

7                         Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
A comparison of key clinical and                the Bangladesh outbreaks.                    and convulsions) were seen in
epidemiological characteristics                 This, coupled with the higher level          the Bangladesh outbreaks at rates
of NiV outbreaks in Malaysia and                of NiV-B RNA in oral secretions              comparable to the Malaysia cases
Bangladesh is compiled in Table                 could be linked to the higher level          (Ang et al., 2018; Hossain et al.,
2. Three clinical characteristics               of human to human transmission,              2008). This phenomenon is largely
between the outbreaks stand out.                which is likely via oral/respiratory         unexplained but could reflect
The first is the shorter time from              secretions or bodily fluids (Ahmad           involvement of different areas of
disease onset to death (7 days vs.              and Tan, 2014). Finally, in the              the central nervous system (CNS)
16 days) and higher case fatality               Malaysia outbreak there was a                due to differences in virus tropism,
rate (74% vs. 38%; Table 1) in                  high incidence of segmented                  differences in the route of infection
the Bangladesh vs. the Malaysia                 myoclonus (muscle jerking),                  or slower disease progression
outbreaks (Ahmad and Tan, 2014;                 which was not reported in the                allowing infection of different
Ang et al., 2018; Hossain et al.,               Bangladesh outbreaks, although               neural tissues.
2008; Lo and Rota, 2008). The                   other indications of encephalitis or
second is the higher incidence                  neurological involvement (such as
of respiratory involvement in                   altered mental status, hyporeflexia

Table 2. Differences in Clinical and Epidemiological Characteristics Between NiV Malaysia
and Bangladesh Outbreaks

    Characteristic                              Malaysia-Singapore                          Bangladesh-India

                                                                                            • Bat to human via consumption of
                                                • Bat to pig → pig to human
    Transmission                                                                               contaminated date palm juice and fruits.
                                                • Rare human to human
                                                                                            • Human to human

    Fever                                       95%                                         100%

    Headache                                    75%                                         73%

    Vomiting                                    32%                                         58%

    Diarrhea                                    18%                                         29%

    Respiratory involvement                     14-29%                                      62-69%

                                                • Segmental myoclonus 32-54%                • Segmental myoclonus not reported

                                                • Hyporeflexia 60.5%                        • Hyporeflexia 65%
    Encephalitis/neurological involvement
                                                • Convulsion 23%                            • Convulsion 23%

                                                • Altered mental status 72%                 • Altered mental status 100%

                                                Disseminated small, high-signal-intensity   Confluent high-signal brain lesions (limited
    MRI
                                                lesions                                     MRIs were performed)

    Relapsed and late-onset encephalitis        ~5-10%                                      4 out of 22 patients (18%) in a follow-up study

    Persistent neurological deficits            ~20%                                        ~30%

    Incubation Period                           Mean = 10 days                              6-11 days

    Average (mean) time from disease onset to
                                                16 days                                     7 days
    death

Sources: (Ang et al., 2018; Hossain et al., 2008); (Ahmad and Tan, 2014; Chong et al., 2002; Lo and Rota, 2008)

8                         Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Further clinical commonalities              • Ferrets infected with NiV-B              AGMs appear to reproduce the NiV
and differences in Nipah                       showed comparable disease                strain differences in virulence and
infections across countries were               progression, and histopathology          pathology seen in humans more
discussed during the NIAID co-                 of the lungs and CNS compared            faithfully than hamster and ferrets,
sponsored Nipah@20 Conference                  to ferrets infected with NiV-M.          and the results suggest that the
in Singapore, December 20204.                  However, NiV-B infected ferrets          distinct clinical characteristics and
The cause of the differences in                shed more virus in oral secretions       epidemiology seen in the Malaysia
disease between the Malaysia/                  than NiV-M infected animals              and Bangladesh outbreaks is at
Singapore and Bangladesh/India                 (Clayton et al 2012). Increased          least partly genetic. However,
outbreaks remains uncertain and                human shedding of NiV-B was              geographic differences in virus
is complicated by the possibility of           seen in the Bangladesh outbreaks         transmission (including route of
differing diagnostic methods and               (Hossain et al., 2008).                  infection and dose), population
case definitions. Experiments in                                                        health and the quality of
animal models have demonstrated             • African Green Monkeys (AGM)              subsequent health care may also
some differences in disease course             Lethality of NiV-B was 100%,             play a role. More information on
and symptomology between the                   compared to 50% for monkeys              the major animal challenge models
Malaysia and Bangladesh strains,               infected with an equivalent dose         for NiV is presented in Section V.
suggesting a genetic component:                of NiV-M. NiV-B also causes more
                                               severe lung histopathology than
•H
  amsters infected with NiV-M                 NiV-M and has a shorter window
 showed accelerated virus                      for therapy with the monoclonal
 replication, pathology and death              antibody m102.4 (Mire et al.,
 compared to hamsters infected                 2016).
 with equivalent doses of NiV-B
 (DeBuysscher et al., 2013). This
 finding is opposite to the human
 fatality rates in the Malaysia and
 Bangladesh outbreaks.

2. NiV Clinical Features and Pathogenesis
    in Humans
The initial clinical presentation of        rapidly to an encephalitic syndrome         encephalitis survivors suffer
NiV infection (by either strain) is         in approximately 60 percent                 long-term neurologic dysfunction
non-specific and characterized by           of patients. The time course of             characterized by persistent
flu-like symptoms including fever,          disease progression from initial            seizures, disabling fatigue and
headache, dizziness, myalgia,               symptoms to the encephalitic                behavioral abnormalities (Mazzola
and loose stools (Banerjee et al.,          syndrome has not been reported              and Kelly-Cirino, 2019; Sejvar
2019). Mild or asymptomatic                 in detail. Neurological symptoms            et al., 2007). In addition, some
infections have also been reported          include meningismus (central                patients with initially mild, non-
in various outbreaks, but the               nervous system inflammation) and            encephalitic disease develop a late-
overall incidence is relatively low         seizures in approximately one-              onset or recurrent neurological
and appears to be strain dependent,         third of patients. A deterioration          disease (Ramphul et al., 2018).
with the Malaysia strain causing            in consciousness, coma and death            Some patients also present with
less severe illness, a lower case           typically occur within an average of        severe respiratory symptoms,
fatality rate and higher prevalence         7 days of disease onset (Bangladesh         and respiratory involvement
of asymptomatic infections than             outbreaks) or an average of 16              has been more common in the
the Bangladesh strain (Kumar et             days (Malaysia outbreak) (Ang               Bangladesh outbreaks compared
al., 2019). The incubation period           et al., 2018; Hossain et al., 2008;         to the Malaysia outbreaks (see
ranges from 7 to 40 days and                Rahman and Chakraborty, 2012).              Epidemiology).
from onset the disease progresses           Approximately 20-30% of NiV

4
    Conference Proceedings:
https://cepi.net/wp-content/uploads/2020/06/2019-CEPI-Duke-WHO-NIAID-Nipah-Conference_FINAL.pdf.

9                       Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Although the route of infection        throughout the body, and the virus     by severe vasculitis and syncytia
in humans has not been                 may then enter the blood stream        formation, resulting in endothelial
conclusively determined, work with     and disseminate throughout the         damage due to vasculitis-induced
experimental animal challenge          host, infecting the brain, spleen      thrombosis and the presence of
models has shown that inhalation       and kidneys (Escaffre et al., 2013).   viral inclusion bodies. Necrotic
of NiV virus particles is sufficient   Experiments in hamsters suggest        plaques are found in both grey and
to initiate infection (Cong et al.,    that entry to the central nervous      white matter of the CNS (Escaffre
2017; Hammoud et al., 2018). In        system (CNS) may occur either          et al., 2013). Lessons learned from
humans, early infection appears        through olfactory neurons or via       pathology and disease course in
to occur in lung epithelial cells,     the choroid plexus and cerebral        humans were discussed in the
and in later stages moves to lung      blood vessels (Baseler et al., 2016;   Transmission/Case Management
endothelial cells. Vasculitis in       Munster et al., 2012). Infection of    Session of the Nipah@20
small blood vessels may be present     the human CNS is characterized         Conference.

3. Diagnosis and Treatment
Laboratory diagnosis of NiV            was not randomized and the             In a lethal challenge study in
infection can be performed using       treated patients may have received     African Green Monkeys (AGMs),
a variety of nucleic acid-based or     better overall care, thus making the   monkeys could be successfully
serological assays. The currently      outcome uncertain (Banerjee et al.,    treated up to 5 days post-infection
preferred methods for detecting        2019). Subsequent animal challenge     with the NiV Malaysia strain
active NiV infection are PCR-          studies in hamsters showed that        (NiV-M). Although half of the
based tests such as conventional       ribavirin delayed, but did not         treated monkeys developed overt
reverse transcriptase (RT) PCR,        prevent, death after NiV infection     clinical signs (fever, respiratory
nested RT-PCR and real-time PCR        (Freiberg et al., 2010; Georges-       and neurological), all the animals
(qPCR). PCR-based tests usually        Courbot et al., 2006). A similar       fully recovered (Geisbert et al.,
target the conserved N, M or P viral   result was obtained after infection    2014). The window for successful
genes. ELISA assays detecting IgM      of African Green Monkeys with the      treatment with m102.4 is only 3
against NiV antigens are typically     closely related Hendra Virus (HeV;     days post-infection when AGMs
the first-line serological tests       (Rockx et al., 2010)).                 are challenged with the NiV
for NiV infection (Mazzola and                                                Bangladesh strain (NiV-B) (Mire
Kelly-Cirino, 2019). Progress and      The broad-spectrum antiviral           et al., 2016). These results suggest
challenges in diagnostics, including   drug Remdesivir (GS-5734) was          that m102.4 may have utility as
a presentation on the WHO Nipah        recently shown to protect African      a post-exposure prophylactic
diagnostics Target Product Profile,    Green Monkeys when administered        or therapeutic in humans. The
were featured in Session 4 of the      24 hours post-inoculation with         m102.4 antibody has also been
Nipah@20 Conference.                   a lethal dose of NiV (Bangladesh       administered on an emergency
                                       strain) (Lo et al., 2019). Treated     basis as post-exposure prophylaxis
Treatment of NiV infection             animals developed mild respiratory     to a handful of humans in cases of
consists primarily of supportive       symptoms, reduced appetite and         high risk of exposure to NiV or HeV
care including maintaining fluids,     showed local virus replication but     (Broder et al., 2013). In all cases the
anticonvulsants, treatment of          no viremia. All the Remdesivir-        patients did not become ill, but it
secondary infection and mechanical     treated animals recovered fully,       is impossible to know if illness was
ventilation. (Ang et al., 2018).       while all the control-treated          prevented by the antibody.
No effective therapeutics for NiV      animals succumbed to the
infection are currently approved       infection.
for use in humans. The antiviral
drug ribavirin was administered to     A human monoclonal antibody,
140 patients during the 1998-99        m102.4, targeting the Ephrin-B2/
NiV Malaysia outbreak, resulting       B3 binding site on the NiV and HeV
in a 36% reduction in mortality        G glycoproteins (see NiV Molecular
compared to 52 untreated control       Biology and Structure), has been
patients (Chong et al., 2001).         tested in NiV animal models for
However, the treatment allocation      prophylactic and therapeutic use.

10               Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
4. NiV Molecular Biology and Structure
NiV is an enveloped, negative-          host cell plasma membrane. Three        glycoprotein mediates target cell
sense, single-stranded RNA virus        non-structural proteins, W, V,          attachment via the cell surface
of the family Paramyxoviridae, a        and C, are produced by alternative      receptors Ephrin-B2 and -B3.
group which also includes measles,      initiation or RNA editing within the    The tissue tropism of Henipavirus
mumps, parainfluenza viruses            P gene open reading frame (Wang         infection is determined by the
and Sendai Virus. NiV shares the        et al., 2001). These gene products      tissue distribution of these
genus Henipavirus with a handful        inhibit host cell antiviral responses   receptors. Ephrin-B2 is expressed
of other recently identified viruses,   such as Type 1 interferon signaling     in neurons, endothelial cells,
including Hendra Virus (HeV) and        and are major determinants of           smooth muscle surrounding
Cedar Virus (Sharma et al., 2019). A    viral pathogenicity (Mathieu et         arteries, placental tissue and
schematic of the NiV viral structure    al., 2012b; Satterfield et al., 2015;   spleen. High levels of Ephrin-B2
and genome organization is shown        Yoneda et al., 2010). The viral         mRNA have also been detected
in Figure 2 below (Sun et al., 2018).   envelope is studded with two            in cardiomyocytes and bronchial
The 18.2 kb NiV genome encodes          transmembrane glycoproteins,            epithelial cells. Ephrin-B3 is
six structural proteins and three       the trimeric F glycoprotein and         expressed in the CNS and in lymph
non-structural proteins. NiV RNA        the tetrameric (dimer of dimers)        nodes (Xu et al., 2012). Ephrin-B2/
is associated with nucleoprotein        G glycoprotein (Aguilar and Lee,        B3 expression levels in target
(N) and phosphoprotein (P) to form      2011). The F and G glycoproteins        tissues also impact the rate of
the virus ribonucleocapsid (RNP).       are the major targets of NiV            virus replication (Sauerhering
The NiV genome encodes its own          neutralizing antibody responses in      et al., 2016), but have not been
RNA-dependent RNA polymerase            animals and humans (Satterfield et      extensively characterized. This
(L) which, together with N and P,       al., 2016b).                            is an important area for future
forms the catalytic subunit of the                                              investigation to understand
replicase complex that enables          The Henipavirus infection and           differences in NiV disease
virus replication. The matrix           replication cycle is depicted           pathogenesis in humans and
(M) protein is required for virion      schematically in Figure 3 (Aguilar      animal challenge models (see
assembly and budding from the           and Lee, 2011). The viral G             Section V.)

Figure 2. NiV structure and organization of the
18.2 kB ssRNA (-) genome (Sun et al., 2018).

11                Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Following attachment, the G             which is then re-exported with G      91.8% similarity at the nucleotide
glycoprotein activates the F            glycoprotein for assembly into the    level (Rockx et al., 2012). The
glycoprotein, which then mediates       budding viral envelope. Assembly      nucleotide changes are not
fusion of the viral envelope            and budding of new viral particles    distributed uniformly; in most
with the host cell membrane.            from the plasma membrane is           cases homologies are higher in the
After cell entry the viral genome       mediated primarily by the M           coding regions than in non-coding
[vRNA(-)] serves as a template for      (matrix) protein. (Aguilar and        regions. Nucleotide homologies
transcription of mRNAs by the           Lee, 2011). The interaction of cell   range from 92.0% to 98.5% in the
viral RNA polymerase (NiV L gene        surface-displayed NiV F and G with    open reading frames. While the 5’
product) which are then translated      Ephrin B2/3 also mediates syncytia    untranslated region of the N gene
into proteins, the vRNA(-) is also      formation by infected cells (Rockx    is 100% conserved between the two
a template for cRNA(+), which is        et al., 2012).                        major strains, homologies in the 5’
then a template for production of                                             and 3’ untranslated regions of all
vRNA(-) genomes for packaging           Two genetically distinct NiV          the other viral genes range from
into new viral particles. Precursor F   strains, Malaysia (NiV-M) and         75.5% to 91.4% (Harcourt et al.,
glycoprotein (Fo) is exported to the    Bangladesh (NiV-B), have been         2005).
plasma membrane, endocytosed            identified. The two strains share
and proteolytically matured to F1/2,    92% amino acid homology and

Figure 3. The Henipavirus infection and replication cycle
(Aguilar and Lee, 2011).

12                Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
5. Vaccine Development
A number of factors suggest that        little attention. One challenge study    Due to safety issues associated
development of a safe, efficacious      conducted in pigs suggested that         with production (i.e., BSL-4
human prophylactic vaccine              cellular immune responses may            containment) and administration
against NiV is scientifically           be important for achieving full          of a live-attenuated or inactivated
feasible. Natural infection by other    protection, but the mechanism was        NiV vaccine, and the need to
paramyxoviruses, such as measles        not defined, and the conclusions         elicit neutralizing antibodies,
and mumps, results in long-term         are complicated by the fact that,        most attempts at NiV vaccine
immunity and vaccines for those         unlike with other animal hosts, NiV      development have focused on
diseases have been successfully         infects a range of porcine immune        recombinant viral vectors and
developed. A vaccine protecting         cells (Pickering et al., 2016). More     adjuvanted protein subunit
horses against the closely related      work is needed to elucidate the role     vaccines. In all cases the target
Hendra Virus (HeV; Equivac®)            of cellular immune responses in          antigen(s) have been the F and/or G
has been approved for use in            protection against NiV infection.        glycoproteins (see Tables 3 and 4).
Australia (Tan et al., 2018). Passive
immunization experiments in a NiV       The WHO developed a Target               The NiV vaccines described below
animal challenge model (hamsters)       Product Profile (TPP) for a human        are all research-stage candidates
using immune sera and monoclonal        NiV vaccine, including preferred         focused on demonstrating
antibodies have demonstrated            as well as critical or minimal           immunogenicity and protection
that neutralizing antibodies            product characteristics.1 Key            against lethal NiV challenge.
confer protection against NiV           vaccine performance attributes           No safety issues associated with
challenge (Guillaume et al., 2004;      recommended in the TPP are:              vaccination or subsequent virus
Guillaume et al., 2006). Finally, as                                             challenge (due to antibody-
discussed below, multiple modes         • Intended use: For reactive use in     dependent disease enhancement;
of active vaccination have resulted        outbreak settings                     ADE) were reported. However,
in protection from lethal NiV                                                    more in-depth safety studies
challenge in animal models.             • Efficacy: ≥ 90% efficacy in           will necessarily be performed
                                           preventing disease (preferred);       on any NiV vaccine candidates
There is broad consensus that              ≥70% (minimal); rapid onset           prior to advancing into human
neutralizing antibodies confer             of protection, less than 2 weeks      clinical testing.
protection against NiV infection           after the first dose (preferred);
and all vaccine development efforts        protection ≤ 2 weeks after the last
to date have focused on their              dose (minimal).
elicitation (Broder et al., 2012;
Prescott et al., 2012; Satterfield et   • Dose Regimen: Single-dose
al., 2016b). However, a correlate of       primary series (preferred); no
protection based on neutralizing           more than 2 doses, with some
antibody titer has not been defined.       protection after the first dose
Neutralizing antibody titers in            (minimal).
animal vaccine challenge studies
where protection was conferred          • Durability of Protection: ≥ 1
are reported in Tables 3 and               year (preferred); ≥ 6 months
4. However, since virtually all            (minimal).
animals were protected in these
studies a threshold of protection       • Product Stability and Storage:
cannot be defined. The role of             Shelf life of 5 years at 2-8oC
cell-mediated immune responses             (preferred); shelf life of at
(CMI) in either natural immunity           least 12 months at -20oC and
or vaccine-induced protection              demonstrated stability of ≥ 1
against NiV has received relatively        month at 2-8oC (minimal).

13                Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Viral Vector Candidates
A number of viral vector platforms     Other recombinant viral vector
expressing the NiV F or G              vaccine platforms expressing NiV
glycoproteins have been tested         F or G have been tested, including:
as vaccine candidates. The most        vaccinia virus (Guillaume et
widely used vector platform to date    al., 2004), canarypox (ALVAC)
has been the Vesicular Stomatitis      (Weingartl et al., 2006), Measles
Virus (VSV). Three types of VSV        virus (Yoneda et al., 2013),
vectors have been employed:            Venezuelan Equine Encephalitis
1) replication-incompetent VSV         Virus (VEEV) (Defang et al.,
pseudotypes expressing NiV             2010), Rabies virus (Keshwara
F or G (Lo et al., 2014); 2) VSV       et al., 2019), Newcastle disease
virions expressing NiV F or G          virus (Kong et al., 2012), Adeno
that can undergo a single round        Associated Virus (AAV) (Ploquin
of replication (Mire et al., 2019);    et al., 2013) and chimpanzee
and 3) replication-competent           adenovirus (ChAd; (van Doremalen
recombinant viruses in which           et al., 2019)). All these candidates
the VSV-G protein is replaced by       conferred full protection against
the Ebola glycoprotein (ZEBOV)         lethal challenge and/or elicited
and also co-expressing NiV F           high titers of neutralizing
or G (DeBuysscher et al., 2014;        antibodies. However, only the
DeBuysscher et al., 2016; Prescott     AAV and chimpanzee adenovirus
et al., 2015). All three VSV-vaccine   (ChAd) vectored vaccines
types, whether expressing NiV F        reported protection after a single
or G antigens and administered         vaccination. A summary of NiV viral
singly, or co-administered, elicited   vector vaccine candidates tested in
neutralizing antibodies and fully      animals is shown in Table 3.
protected immunized animals from
clinical disease in at least one of
the 3 major NiV lethal challenge
models (hamsters, ferrets or
non-human primates; see Section
V). Additionally, all three VSV
vaccine types conferred protection
after a single dose (see Table 3).
Additional details on the animal
challenge models and their use
in vaccine studies are given in
Section V.

14               Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Table 3. NiV Viral-vector Vaccine Candidates Tested in Animals

                                                                                                                            NiV Neutralization
                                                     Animal(s)        Vaccination Route/Regimen/Challenge
     Reference         Vaccine Description                                                                                     Titers (Pre-
                                                    immunized                       (Strain3)
                                                                                                                               challenge)2

    Guillaume et al    Vaccinia virus (VV) vector                       SC/2 vaccinations 1 month apart/ challenge 3
                                                      Hamsters                                                                  ~ 1:10 – 1:25
        (2004)           expressing NiV G or F                               months after last vaccination (M)

    Weingartl et al    Canarypox vector (ALVAC)                       IM/2 vaccinations 14 days apart/ challenge on day
                                                         Pigs                                                                  1:200 – 1:1280
       (2006)            expressing NiV G or F                         28 post 2nd vaccination (NiV strain not specified)

                          Venezuelan Equine
     Defang et al                                                     Footpad inoculation/ 3 vaccinations on week 0, 5
                       Encephalitis Virus (VEEV)         Mice                                                                  ~ 1:215 – 1:217 *
       (2011)                                                                      and 18/ no challenge
                         expressing NiV G or F

                       Newcastle Disease Virus
      Kong et al
                       (NDV) vector expressing           Pigs          IM/2 vaccinations 4 weeks apart/ no challenge            ~ 1:27 – 1:212*
       (2012)
                             NiV F or G

                        Single-cycle replication
     (Mire et al.,                                                     IM/one vaccination/ challenge on day 20 post-
                       VSV-∆G vector expressing         Ferrets                                                                 ~1:40 – 1:160
       2013)                                                                         vaccination (M).
                              NiV G or F

                        Adeno-Associated Virus
    (Ploquin et al.,                                                   IM/one vaccination/ challenge at 5 weeks post-
                        (AAV) vector expressing       Hamsters                                                                  < 1:10 to 1:160
        2013)                                                                        vaccination (M).
                                 NiV G

                                                                         Hamster: IP/ 2 vaccinations 21 days apart/
                         Measles virus vaccine                                                                              Hamster: Not reported
     Yoneda et al                                     Hamsters,       challenge 7 days post 2nd Vaccination AGM. SC/2
                        vector expressing NiV G
       (2013)                                          AGM*            vaccinations 28 days apart/ challenge 2 weeks        AGM: 1:1600 – 1:3200
                             glycoprotein
                                                                       post 2nd vaccination (NiV strain not specified).

                        Replication-competent
    DeBuysscher et                                                     IP/ one vaccination/ challenge on day 28 post-
                       VSV vector expressing NiV      Hamsters                                                                  1:80 - ≥ 1:640
      al (2014)                                                                       vaccination (M).
                                 G or F

                         Replication-defective
                                                                       IM/ one vaccination/ challenge at day 32 post-
    Lo et al (2014)    VSV-∆G vector expressing       Hamsters                                                                ~ 5 x103 – 1 x 104
                                                                                     vaccination (M)
                              NiV G or F.

     (Guillaume-
                       Canarypox vector (ALVAC)
    Vasselin et al.,                                Ponies (horses)     IM/2 vaccinations 21 days apart/ no challenge              ~ 1:2128*
                         expressing HeV G or F
        2016)

     Prescott et al      Live-attenuated VSV                           IM/one vaccination/ challenge on day 29 post-
                                                        AGM1                                                                     1:80 – 1:160
        (2015)          vector expressing NiV G                                      vaccination (M).

    DeBuysscher et        Live attenuated VSV                           IP/one vaccination/ challenge one day post-
                                                      Hamsters                                                                  Not reported
       al 2016          vector expressing NiV G                              vaccination (100% survival) (M).

                        Live-attenuated Rabies                                                                                  ~1:10 to 1:600
    Keshwara et al
                          Virus vaccine vector           Mice          IM/2 vaccinations 28 days apart/ no challenge
       (2019)                                                                                                                  (no challenge)
                       (RABV) expressing NiV G.

                        Single-cycle replication
                                                                       IM/one vaccination/ challenge on day 28 post-
    Mire et al 2019    VSV-∆G vector expressing         AGM1                                                                    1:160 – 1:640
                                                                                     vaccination (B).
                              NiV G or F

         (van           Chimpanzee adenovirus                           IM/ one or two vaccinations (28 days apart)/
    Doremalen et       (ChAd) vector expressing       Hamsters         challenge 70 days post-prime or 42 days post-           ~1:40 - ~1:100
      al., 2019)                NiV G                                                 boost (M and B).

1
 =African Green Monkey 2~ Indicates titer values estimated from data presented graphically 3 Challenge strain M= Malaysia;
B=Bangladesh; *endpoint neutralization titers determined by 2-fold serial dilution and expressed as exponentials of 2.
IM = intramuscular IP = intraperitoneal, SC = sub-cutaneous

15                       Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Subunit Vaccine Candidates

The most widely studied NiV                          does not efficiently generate HeV              comprised of an enveloped virus-
subunit vaccine candidates have                      cross-neutralizing antibodies                  like particle (VLP) created by co-
utilized a purified, recombinant G                   (Ploquin et al., 2013). Various                expression of the NiV M (matrix),
glycoprotein from Hendra Virus                       adjuvant formulations have been                F and G glycoproteins and
(HeV) in which the transmembrane                     tested, including aluminum + CpG               adjuvanted in either aluminum
domain has been removed to allow                     (Bossart et al., 2012; McEachern               hydroxide (Alhydrogel®),
soluble G protein expression (sG).                   et al., 2008), CpG alone (Pallister            monophosphoryl lipid A (MPLA),
The high sequence conservation                       et al., 2013), and Quil A/DEAE-                or CpG has also been tested and
between the NiV and HeV G                            dextran/Montanide (Mungall et al.,             was 100% protective in the hamster
glycoproteins (83% amino acid                        2006). All formulations were 100%              challenge model (Walpita et al.,
homology; (Wang et al., 2001)                        efficacious, eliciting neutralizing            2017). A summary of NiV subunit
allows for the elicitation of                        antibodies and protecting all                  vaccine candidates tested in
potent NiV cross-neutralizing                        vaccinated animals against lethal              animals is shown in Table 4.
antibodies (Sun et al., 2018),                       NiV challenge with no signs of
although vaccination with NiV G                      clinical disease. A vaccine candidate

Table 4. NiV Submit Vaccine Candidates Tested in Animals

                                                                                                                         NiV Neutralization
                                                      Animal(s)     Vaccination Route/Regimen/Challenge
     Reference           Vaccine Description                                                                                Titers (Pre-
                                                     immunized                    (Strain3)
                                                                                                                            challenge)2

                            sGNiV or sGHeV
     Mungall et al          adjuvanted with                           SC/ 3 vaccinations 2 weeks apart/ challenge 15
                                                         Cats                                                              1:2,560 –1:20,480
       (2006)            Montanide/QuilA/DEAE-                             weeks after the first vaccination (M).
                                dextran

                         Recombinant soluble HeV
    McEachern et                                                      IM/ 2 vaccinations 21 days apart/ challenge on
                         G glycoprotein adjuvanted       Cats                                                                  1:32 – 1:512
     al (2008)                                                               day 42 post 1st vaccination (M).
                         with CpG + AlhydrogelTM

                            Virus-like particles
     Walpita et al                                                     SC/ 3 vaccinations on days 0, 15 and 29/ no
                         (VLPs) comprising NiV M,        Mice                                                                  1:5 - >1:80
       (2011)                                                                           challenge
                                  G and F

                         Recombinant soluble HeV
    (Bossart et al.,                                                  IM/ 2 vaccinations 21 days apart/ challenge 21
                         G glycoprotein adjuvanted       AGM1                                                                 1:67 – 1:379
        2012)                                                                 days post 2nd vaccination (M).
                          with CpG + AlhydrogelTM

                         Recombinant soluble HeV
    (Pallister et al.,                                                SC/ 2 vaccinations 20 days apart/ challenge 20
                         G glycoprotein adjuvanted      Ferrets                                                                1:16 – 1:128
         2013)                                                          days or 14 months post 2nd vaccination (B)
                                 with CpG

                           Recombinant soluble
    Pickering et al       HeV G glycoprotein in                       IM/ 2 vaccinations 21 days apart/ challenge 35
                                                         Pigs                                                                ~ 1:25- - 1:450
        (2016)            a proprietary adjuvant                      days post 1st vaccination (Strain not specified)
                               (Zoetis, Inc.)

                                                                     Single dose trial: IM/ one dose/ challenge on day    3-Dose Trial: ~ 1:200
                            Virus-like particles                                 28 post vaccination (M).                      – 1:2500
     Walpita et al
                         (VLPs) containing NiV M,      Hamsters
       (2017)                                                           3 dose trial: 3 doses on days 0, 21 and 42/       1-Dose Trial: ~ 1:10 –
                                 F and G.
                                                                                 challenge on day 58 (M).                        1:200

1
 =African Green Monkey 2~ Indicates titer values estimated from data presented graphically 3 Challenge strain M= Malaysia;
B=Bangladesh; *endpoint neutralization titers determined by 2-fold serial dilution and expressed as exponentials of 2.
IM = intramuscular IP = intraperitoneal, SC = sub-cutaneous

16                         Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
More recently, novel antigen                       challenge with the Malaysian
design options have been evaluated                 strain of Nipah virus. Authors
using a structure-based design.5                   noted immune responses were
A stabilized prefusion F (pre-F),                  suboptimal. It is conceivable
multimeric G constructs, and                       that the protection would have
chimeric proteins containing both                  been superior under a two-dose
pre-F and G were developed as                      regimen.
protein subunit candidate vaccines.
The proteins were evaluated                        In addition, some of the structure-
for antigenicity and structural                    based designs described earlier
integrity using kinetic binding                    for immunogen development5
assays, electron microscopy, and                   are being evaluated in the mRNA
other biophysical properties.                      platform in collaboration with
Immunogenicity of the vaccine                      Moderna, and clinical evaluation
antigens was evaluated in mice                     is planned.
using aluminum hydroxide as
adjuvant.                                          NiV Vaccine Candidates Supported
                                                   by CEPI

mRNA Vaccine Candidates                            As of August 2019, CEPI has four
                                                   NiV vaccine candidates in its
The US CDC has published proof-                    vaccine development portfolio,
of-concept pre-clinical data on a                  three viral-vector platform
Hendra virus glycoprotein mRNA                     candidates and one candidate
vaccine in liquid nanoparticles.6                  comprising an adjuvanted
A single dose of the vaccine                       recombinant protein antigen
protected up to 70% of hamsters                    (Table 5).
against a lethal, intraperitoneal

Table 5. NiV vaccine candidates supported by CEPI

                    Developer                                  Vaccine Platform             Development Stage

                 University of Tokyo                         Recombinant Viral Vector           Pre-clinical

    Profectus Biosciences/Emergent Biosolutions/
                                                               Recombinant Protein             Phase 1 (USA)
                        PATH

       Janssen Vaccines & University of Oxford               Recombinant Viral Vector           Pre-clinical

                                                        Replication-competent rVSV vector
             Public Health Vaccine, LLC                                                         Pre-clinical
                                                                 expressing NiV-G

Source: https://cepi.net/research_dev/our-portfolio/

5
    https://www.frontiersin.org/articles/10.3389/fimmu.2020.00842/full
6
    https://academic.oup.com/jid/article/221/Supplement_4/S493/5637464

17                      Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
III. STANDARDIZATION OF
ASSAYS AND ANIMAL MODELS
Assays and animal models to quantify or characterize immune responses
elicited by vaccination are, by their nature, inherently variable.
The reasons for this include the       Immune Serum Reference                 and commercial manufacturing
molecular complexity of the            Standards                              (https://www.who.int/biologicals/
samples (serum or other biological                                            vaccines/en/). Recent examples
samples), the need to produce          One of the most important tools        include HPV 16 (Ferguson et al.,
reagents in complex biological         for standardization of serological     2011), Typhoid Fever (Rijpkema et
systems such as cell culture or in     assays is immune reference             al., 2018), Respiratory Syncytial
vivo, variability in composition and   serum. Even when similar assay         Virus (McDonald et al., 2018) and
stability of these reagents, and the   formats are used for detection         Zika (Source: WHO/BS/2018.2345).
need to test immune responses in       of antigen binding antibodies or
vivo. Nevertheless, modern vaccine     virus neutralizing antibodies,         Three key factors determine
development requires vaccines          the resulting data can be highly       the fitness of material for use
and samples from vaccinated            variable between laboratories due      as a biological standard. First,
humans and animals be tested           to differences in assay methods        the material must have similar
with the highest possible precision.   and reagents. For example, a           composition and in vitro behavior
The task is further complicated        10-laboratory collaborative study      to the human sera test articles.
by the collaborative and global        assessing the precision of assays      Second, the standard should be
nature of modern vaccine               for detection of serum antibodies      commutable, meaning it should
development. Multiple research         against Human Papillomavirus 16        work for a wide range of serological
laboratories, vaccine developers,      (HPV 16) revealed inter-laboratory     assays and vaccine platforms
non-governmental organizations,        variations in anti-HPV titer of        being tested. Finally, a blinded
and regulatory agencies are often      up to 25-fold for the same test        multi-laboratory collaborative
involved in the development            sample (Ferguson et al., 2006). A      study must demonstrate the
process and vaccine candidates         similar, 15-laboratory collaborative   utility of the standard for reducing
utilizing different platform           study evaluating assays for serum      intra-laboratory assay variability.
technologies are often evaluated for   antibodies against H5N1 influenza      (Source: CEPI 2nd Standards and
the same disease indication. Thus,     showed inter-laboratory variations     Assays Workshop; June 2019).
standardization of methods and         in titer of 10 to 35-fold, depending
reagents is important to facilitate    on the sample and type of assay        Serum reference standards for a
development of new vaccines such       (Stephenson et al., 2009). The         new vaccine are often established
as for NiV. The goal is to enable      purpose of establishing immune         in a staged manner as the
“like versus like” comparisons         reference standards is to provide      development process progresses.
of data generated by different         a common, external control to          This mitigates the risk of producing
laboratories and derived from          improve the comparability of           exhaustively characterized
many assay types. Recognizing          assay data between laboratories.       materials which might not be
the value of phase-appropriate         With the standard in place, test       required if vaccine development
standardization early in the vaccine   results are reported relative          does not progress. For R&D and
development process, CEPI is           to the activity of the reference       early clinical trials a working
promoting assay, reagent, and          standard. In the studies cited         standard or interim standard may
animal model standardization to        above, use of a common reference       be established by a collaborative
accelerate development of vaccines     standard significantly reduced         study involving a relatively limited
for NiV and other priority diseases    intra-laboratory assay variability.    number of laboratories, and
in its portfolio.                      Reference standards have been          relatively low volumes may be
                                       developed for many vaccine             sufficient in the earlier stages.
                                       indications, both in development

18               Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
A number of different sources of        (Source: CEPI 2nd Standards             common reagents (reference
immune sera may be considered.          and Assays Workshop; June               sera and antigens) provided by
For example, a collaborative            2019). A single, large lot of an        regulatory agencies and using a
study for establishment of an           antibody standard is preferred          single, validated assay method
interim standard for antibodies         to avoid potential variability          to test and release new seasonal
to Ebola virus (EBOV) tested            between multiple lots and the           vaccine formulations. In general,
plasma samples from patients who        need for subsequent bridging            standards may improve inter-
recovered from Ebola infection          studies. Once suitable standard         laboratory test performance.
(convalescent sera), anti-EBOV          sera candidates are available for       However, the need, feasibility
IgG preparations from trans-            evaluation, a collaborative study is    and level of standardization is
chromosomal (Tc) cows immunized         performed to evaluate serological       typically considered on a case-
with experimental vaccines and          assays performed by a number of         by-case basis according to the
plasma from vaccinated volunteers       participating laboratories. A broad     stage of vaccine development,
participating in an EBOV vaccine        panel of test samples from different    the types of assays in use and the
trial (Wilkinson et al., 2017). An      sources (e.g., sera from naturally      potential of standards to facilitate
interim standard for NiV will           infected humans, animals infected       development and licensure.
probably be generated from non-         in the laboratory, and vaccinated       Standardized reference sera are
human primates infected with a          humans or animals) is assayed and       relatively easy to implement since
sub-lethal dose of NiV and which        the intra-laboratory variability in     they ideally should be commutable
generate high titers of neutralizing    assay results is assessed. Finally,     across many assay types and are
antibodies (Dhondt and Horvat,          the test sample absolute values         broadly recognized for improving
2013). Obtaining convalescent sera      (for example, geometric mean            both intra- and inter-laboratory
from NiV survivors is also being        titers) are expressed relative to the   assay consistency. In contrast,
considered (Source: CEPI 2nd            activity of the candidate standard      standardized assay formats are
Standards and Assays Workshop;          and the ability of the standard         more challenging to implement,
June 2019). However, given the          to improve intra-laboratory             especially for newer vaccines
sporadic nature of NiV outbreaks        comparability of test results is        and those in development, since
and relatively small number of          assessed. Once the standard has         vaccine antigens may differ
cases (and available survivors),        been chosen a full storage stability    between candidates and there is
obtaining sufficient quantities of      program is conducted to ensure the      less consensus on the ideal assay
convalescent sera for long-term         quality of the material over time.      format. With these considerations
use may be challenging. Therefore,      Trending of assay performance           in mind, the benefits and potential
in the case of emerging infections      over time is also performed. The        challenges of standardizing
such as NiV one of the alternative      International Standard itself is        various assay and animal model
approaches described above may          not intended for routine assay use.     components to accelerate
need to be employed.                    A working reference standard is         NiV vaccine development are
                                        established for routine use and         summarized in Table 6.
Establishment of an interim             a bridging study is conducted to
standard usually precedes               calibrate the working standard to
establishment of an International       the International Standard (Source:
Standard (IS) , often called            CEPI 2nd Standards and Assays
an International Reference              Workshop; June, 2019).
Preparation (IRP) under the
endorsement of the WHO                  Standardization of Other Biological
Expert Committee on Biological          Assay Reagents and Methods
Standardization. This is a more
formal process, taking up to 36         Many aspects of biological
months and involving a larger           assays for vaccine testing may
and more in-depth collaborative         be standardized to improve the
study, often involving more             comparability of intra-laboratory
than 25 laboratories and a wide         data. Common reagents (reference
geographical distribution. This         sera, antigens, virus stocks) may
is the main difference from a           be produced and standardized
working or interim standard.            assay methods established and
Regulatory agencies generally           validated. For example, potency
expect an established International     testing for release of subunit
Standard to be used in pivotal          seasonal influenza vaccines
clinical trials for vaccine approval,   is performed under a high
unless specifically justified           degree of standardization using

19                Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
Table 6. Benefits and Potential Challenges of Implementing Biological Standards for NiV
Vaccine Development

 Standard                                     Benefits                                         Potential challenge(s)

                                                                                               Finding human NiV convalescent donors may
                                              Long track record and high level of acceptance
                                                                                               be challenging (low numbers). Generating
                                              for improving intra-laboratory assay
 Immune Standard Reference Sera                                                                NiV convalescent animal sera by sub-lethal
                                              comparability. Inter-laboratory performance
                                                                                               infection requires BSL-4 containment and
                                              may also be improved.
                                                                                               can be considered an interim mitigation

                                                                                               Choice of genotype/strain and ensuring
                                              Promote standardization of serum antibody        reactivity to diverse serum isolates; storage
 Common stocks of ELISA coating antigen       detection; relatively easy to produce, test,     stability; heterogeneity in post-translational
                                              store and distribute.                            modifications; biochemical differences
                                                                                               between strains.

                                                                                               Current use of pseudovirus assays by
                                                                                               major NiV research groups is rare. Often
 Common pseudovirus(es) for assaying          Promote use of an assay method which can be      overestimate titer compared to wild-type
 neutralizing antibodies                      performed in low-level biocontainment            NiV-based assays, so will require extensive
                                                                                               characterization compared to traditional NiV
                                                                                               neutralization assays to gain acceptance.

                                                                                               Production, testing, storage, stability and
 Common NiV virus stocks for neutralization   Promote standardization of animal challenge      distribution of live NiV and requirement for
 assays and animal challenge models           experiments and NiV neutralization assays        BSL-4; mutations during passaging of ssRNA
                                                                                               virus.

                                                                                               The large number of potential variables in
                                                                                               challenge model performance (ie., challenge
 Standards for performance of animal          Promote standardization of challenge
                                                                                               strain/stock, route, dose, animal species etc.)
 challenge models                             experiments
                                                                                               may complicate agreement on and acceptance
                                                                                               of performance standards.

20                   Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
IV. NIV SEROLOGICAL ASSAYS
Robust serological assays for quantifying and characterizing humoral immune
responses in humans and animals are critical for vaccine development.

A number of methods have been         assays for NiV, as well as newer        experiments is compiled in Table
developed for NiV serology, and the   assays in earlier stages of use and     8 and Table 9 to illustrate the
refinement and standardization        acceptance. An analysis of the pros     prevalence of use, variables in
of such methods will be essential     and cons of different serological       assay performance, how reagents
for facilitating development of       assays for NiV vaccine development      and methods have changed over
safe and effective human vaccines     is presented in Table 7. The usage      time and opportunities for assay
against NiV. This section describes   of assays in a large number of NiV      standardization.
the commonly used serological         vaccine studies and other research

1. Detection of antigen – specific serum IgG
Detection of antigen – specific       The earliest ELISAs for detection       the 17 ELISAs in Tables 8 and 9
serum IgG is essential to the         of NiV antibodies in sera were          use recombinant G or F for target
vaccine development process to        developed by the Centers for            IgG capture. The sFNiV and sGNiV
characterize the specificity and      Disease Control (CDC; USA).             have been produced in a variety of
magnitude of the vaccine-induced      Different ELISAs were developed         recombinant expression systems
humoral immune response. The          for NiV-specific serum IgG and          (E. coli, insect cells, mammalian
most common assay method is           IgM. These ELISAs were used             cells) and are usually epitope
the traditional “indirect” ELISA.     for surveillance and diagnosis of       tagged for ease of purification
In this assay a target antigen is     disease in humans and pigs, and         (Eshaghi et al., 2005; Eshaghi et al.,
plated (adsorbed) onto a 96- well     used detergent and radiation-           2004; Keshwara et al., 2019; Kurup
microtiter plate. After blocking      inactivated, NiV – infected Vero        et al., 2015). The glycosylation and
the plate to suppress non-specific    cell lysates as the target antigen      disulfide bonding in the NiV F and
binding, dilutions of immune          (Daniels et al., 2001). Several of      G glycoproteins make eukaryotic
or control sera are added to          the NiV animal vaccination studies      cells preferable for recombinant
the wells. After washing away         and research experiments detailed       expression of these antigens. The
unbound antibody, bound IgG is        in Table 8 and Table 9 utilized         many successful tests of vaccines
usually detected by the addition      inactivated crude NiV- infected         targeting the G glycoprotein in NiV
of a species-specific anti-IgG        Vero extracts or gradient-purified      animal challenge models make it
secondary antibody conjugated         NiV as the target antigen. However,     likely that this antigen will be used
to a chromogenic enzyme. While        the use of NiV as an assay reagent      in human vaccine candidates.
serum IgG is measured to elucidate    is obviously problematic since
vaccine responses, as well as for     the initial preparation requires
surveillance and epidemiology,        BSL-4 containment. ELISAs using
measurement of NiV-specific           NiV-infected crude extracts also
serum IgM is usually performed        suffered from non-specific binding
for diagnosis of active infection     (Daniels et al., 2001). The discovery
(Mazzola and Kelly-Cirino, 2019).     that NiV F and G glycoproteins are
The advantages of the ELISA assay     the major target of neutralizing
format include its broad use and      antibodies and their use in vaccine
familiarity throughout biomedical     formulations spurred the use
science, relatively low-tech and      of recombinant, soluble NiV F
low-cost application and wide         (sFNiV) and G (sGNiV) as the target
availability of reagents.             antigens in ELISA assays. Six of

21               Nipah Virus Assays and Animal Models for Vaccine Development, Landscape Analysis, January 2021
You can also read