Elsevier

Vaccine

Volume 34, Issue 30, 24 June 2016, Pages 3525-3534
Vaccine

Hendra virus and Nipah virus animal vaccines

https://doi.org/10.1016/j.vaccine.2016.03.075Get rights and content

Abstract

Hendra virus (HeV) and Nipah virus (NiV) are zoonotic viruses that emerged in the mid to late 1990s causing disease outbreaks in livestock and people. HeV appeared in Queensland, Australia in 1994 causing a severe respiratory disease in horses along with a human case fatality. NiV emerged a few years later in Malaysia and Singapore in 1998–1999 causing a large outbreak of encephalitis with high mortality in people and also respiratory disease in pigs which served as amplifying hosts. The key pathological elements of HeV and NiV infection in several species of mammals, and also in people, are a severe systemic and often fatal neurologic and/or respiratory disease. In people, both HeV and NiV are also capable of causing relapsed encephalitis following recovery from an acute infection. The known reservoir hosts of HeV and NiV are several species of pteropid fruit bats. Spillovers of HeV into horses continue to occur in Australia and NiV has caused outbreaks in people in Bangladesh and India nearly annually since 2001, making HeV and NiV important transboundary biological threats. NiV in particular possesses several features that underscore its potential as a pandemic threat, including its ability to infect humans directly from natural reservoirs or indirectly from other susceptible animals, along with a capacity of limited human-to-human transmission. Several HeV and NiV animal challenge models have been developed which have facilitated an understanding of pathogenesis and allowed for the successful development of both active and passive immunization countermeasures.

Introduction

Hendra virus (HeV) and Nipah virus (NiV) are enveloped, single-stranded negative sense RNA viruses and the prototype members of the genus Henipavirus in the family Paramyxoviridae [1]. Recently, a third virus isolate Cedar virus (CedPV) has been added to the Henipavirus genus [2]. Whereas HeV and NiV are bat-borne disease-causing zoonoses, CedPV is not known to be zoonotic nor has it been shown to be pathogenic in any animal, including those susceptible to HeV and NiV, although it does reside in nature in the same bat species as HeV. To date, bats appear to be predominant natural reservoir hosts for henipaviruses [3]. Although nucleic acid based detection studies have identified related Henipavirus species, including complete genomic sequences [4], [5], HeV, NiV, and CedPV are the only virus isolates that have been reported.

In several disease-susceptible animal species, and in people, the major pathological observation of HeV and NiV infection is a severe systemic and often fatal neurologic and/or respiratory disease [6], [7], [8]. However, HeV and particularly NiV can also cause relapsed encephalitis which follows a recovery from an acute infection and appears to result from a recrudescence of virus replication in the central nervous system (CNS) [9], [10]. Outbreaks or spillovers of NiV in Bangladesh and India, since its first emergence in peninsular Malaysia, have continued to occur, as has HeV in Australia, making these henipaviruses important transboundary biological threats [11]. Both HeV and NiV are highly pathogenic in a number of mammalian species and possess several characteristics that distinguish them from all other known paramyxoviruses and are classified as Biosafety Level-4 (BSL-4) agents. Indeed, NiV is considered to have the potential to be a pandemic threat since it can infect humans directly from natural reservoirs (bats) or indirectly following amplification in a susceptible animal species (pig) and has a recognized capacity of, albeit limited, human-to-human transmission [12]. Since their discovery and recognition, a variety of approaches have been taken by multiple research groups to devise countermeasures as a means of addressing the transboundary threat issues brought about by HeV and NiV. Several active and passive immunization approaches have been explored and some have led to successful deployment and human clinical trials. Concurrent with these developments, several animal challenge models of HeV and NiV infection and pathogenesis have been established which have provided insight into the nature of HeV and NiV disease [13], [14] and afforded the possibility of testing vaccine and therapeutic countermeasures [15], [16], [17].

Section snippets

Hendra virus and Nipah virus emergence

In 1994 an outbreak of fatal cases of a severe respiratory disease in horses and humans occurred in the Brisbane suburb of Hendra, Australia. The infectious cause of this event was discovered to be a previously unknown paramyxovirus that was distantly related to certain morbilliviruses [18]. In all, 13 horses and a trainer succumbed to infection together with the non-fatal infection of 7 additional horses and a stable hand. In a separate incident that was retrospectively recognized, this same

The viruses, tropism and entry

HeV and NiV particles are enveloped and pleomorphic with spherical or filamentous forms observed by electron microscopy [70], [71], [72]. The viral envelope carries surface projections composed of the viral transmembrane anchored fusion (F) and attachment (G) glycoproteins which have been the major target of antiviral strategies. HeV and NiV are classified into the Henipavirus genus, Paramyxoviridae family [73] and their genomes are unsegmented, single-stranded, negative-sense RNA [1]. Genomic

Clinical and pathological features of human HeV and NiV infection

Human HeV and NiV infections have an incubation period ranging from a few days to about 3 weeks [101], [102]. All human cases of HeV infection have been the result of exposure and transmission of the virus from infected horses to humans. Following an influenza-like illness (fever, myalgia, headaches, lethargy, vertigo, cough, pharyngitis, and cervical lympadenopathy), the majority of human HeV cases developed severe disease and died; only 3 of 7 patients have survived (∼60% mortality rate) [8],

Clinical and pathological features of HeV and NiV infection in animals

Naturally acquired HeV infections have almost exclusively been observed in horses, and only recently have two dogs been reported HeV antibody positive. Whereas in addition to pigs, naturally acquired NiV infection was noted in dogs, cats and horses in the initial Malaysian outbreak [113]. Since their discovery, however, a large number of different species have been examined under experimental conditions for the purposes of developing infection models and characterizing the viral pathogenic

The pig

Experimental NiV infection of pigs has revealed an incubation period of ∼5–7 days post-infection and the respiratory system serving as a major site of virus replication and pathology, with viral antigen and syncytia formation present in the respiratory epithelium (tracheal, bronchial, bronchiolar, and alveolar) and small blood and lymphatic vessels [113], [121], [123]. Virus was also observed in the kidneys and in endothelial and smooth muscle cells of small blood vessels [121]. CNS involvement

The horse

Naturally acquired HeV infection in horses often results in severe disease, with fever and increased heart rate common, and a rapid deterioration with respiratory and/or neurological clinical signs. The case fatality rate is ∼75%. In some naturally occurring cases HeV infection has presented with more subtle clinical signs including absence of fever. An apparent incubation period in naturally infected horses appears to be 5–16 days based on the events of the two large outbreaks; the first in

Non-human primates

The only nonhuman primate model that has uniformly recapitulated human disease for both NiV and HeV infection was developed using the African green monkey (AGM) [126], [127]. Both NiV and HeV produce a uniformly lethal illness following low dose virus challenge by intratracheal inoculation within 7–10 days post-infection. NiV and HeV spread rapidly to numerous organ systems within the first 3–4 days following challenge, followed by the development of a progressive and severe respiratory disease

Therapeutics and vaccines

There are no approved HeV or NiV antiviral therapeutics or vaccines for human use, however, several countermeasure approaches have been evaluated in animal challenge models and have been recently reviewed in detail elsewhere [16]. Here, the focus will be an up to date summary of the HeV and NiV animal vaccine approaches that have been examined in vivo along with the one therapeutic approach for human use that has reached phase I clinical trial evaluation.

Active immunization strategies

There has been a variety of immunization strategies developed for HeV and NiV infection including live-recombinant virus platforms, protein subunit, virus-like particles and DNA vaccines; many of these approaches have only been examined for their ability to illicit HeV- or NiV-specific neutralizing antibody responses [133], [134], [135], [136]. Some approaches, however, have been examined for both immune response and efficacy using a variety of animal challenge models (Table 1). The first

The passive immunization approach

The first passive immunization studies were conducted in the hamster NiV-challenge model and demonstrated that antibody immunotherapy against NiV infection through the targeting of the viral envelope glycoproteins was possible. Protective passive immunotherapy using either NiV G and F-specific polyclonal antiserums, or mouse monoclonal antibodies (mAbs) specific for the NiV or HeV G or F glycoproteins has been demonstrated [118], [137], [149]. These studies demonstrated the importance of viral

Conclusions and future directions

Because of the potential environmental accessibility of HeV and NiV, their highly pathogenic characteristics and ability to infect a broad range of mammalian hosts including people, the development of effective countermeasures against these biothreats has been a major area of research focus. These efforts have led to the development and testing of potential vaccine candidates and antiviral therapeutics. In 2010, the m102.4 mAb producing cell line was provided to the Queensland Government,

Acknowledgements

C.C.B is supported by NIH grant AI054715-06. The authors thank Dr. Kai Xu for providing Fig. 1.

References (158)

  • K.B. Chua et al.

    Isolation of Nipah virus from Malaysian Island flying-foxes

    Microbes Infect

    (2002)
  • D.J. Middleton et al.

    Experimental Nipah virus infection in pteropid bats (Pteropus poliocephalus)

    J Comp Pathol

    (2007)
  • M.M. Williamson et al.

    Experimental hendra virus infection in pregnant guinea-pigs and fruit bats (Pteropus poliocephalus)

    J Comp Pathol

    (2000)
  • A.D. Hyatt et al.

    Ultrastructure of Hendra virus and Nipah virus within cultured cells and host animals

    Microbes Infect

    (2001)
  • C.S. Goldsmith et al.

    Elucidation of Nipah virus morphogenesis and replication using ultrastructural and molecular approaches

    Virus Res

    (2003)
  • K.B. Chua et al.

    Isolation of Nipah virus from Malaysian Island flying-foxes

    Microbes Infect

    (2002)
  • B.H. Harcourt et al.

    Molecular characterization of Nipah virus, a newly emergent paramyxovirus

    Virology

    (2000)
  • N. Tanimura et al.

    Distribution of viral antigens and development of lesions in chicken embryos inoculated with nipah virus

    J Comp Pathol

    (2006)
  • U. Drescher

    Eph family functions from an evolutionary perspective

    Curr Opin Genet Dev

    (2002)
  • K.N. Bossart et al.

    Functional studies of host-specific ephrin-B ligands as Henipavirus receptors

    Virology

    (2008)
  • N.W. Gale et al.

    Ephrin-B2 selectively marks arterial vessels and neovascularization sites in the adult, with expression in both endothelial and smooth-muscle cells

    Dev Biol

    (2001)
  • A. Poliakov et al.

    Diverse roles of eph receptors and ephrins in the regulation of cell migration and tissue assembly

    Dev Cell

    (2004)
  • E.B. Pasquale

    Eph-ephrin bidirectional signaling in physiology and disease

    Cell

    (2008)
  • B. Lee et al.

    Modes of paramyxovirus fusion: a henipavirus perspective

    Trends Microbiol

    (2011)
  • L.-F. Wang et al.

    Henipaviruses

  • G.A. Marsh et al.

    Cedar virus: a novel Henipavirus isolated from Australian bats

    PLoS Pathog

    (2012)
  • B.A. Clayton et al.

    Henipaviruses: an updated review focusing on the pteropid reservoir and features of transmission

    Zoonoses Public Health

    (2013)
  • Z. Wu et al.

    Novel Henipa-like virus, Mojiang Paramyxovirus, in rats, China, 2012

    Emerg Infect Dis

    (2014)
  • J.F. Drexler et al.

    Bats host major mammalian paramyxoviruses

    Nat Commun

    (2012)
  • K.T. Wong et al.

    Pathology of acute henipavirus infection in humans and animals

    Pathol Res Int

    (2011)
  • E.G. Playford et al.

    Human Hendra virus encephalitis associated with equine outbreak, Australia, 2008

    Emerg Infect Dis

    (2010)
  • K.T. Wong et al.

    Clinical and pathological manifestations of human henipavirus infection

    Curr Top Microbiol Immunol

    (2012)
  • K.T. Wong et al.

    Human Hendra virus infection causes acute and relapsing encephalitis

    Neuropathol Appl Neurobiol

    (2009)
  • T.W. Geisbert et al.

    Animal challenge models of henipavirus infection and pathogenesis

    Curr Top Microbiol Immunol

    (2012)
  • E. de Wit et al.

    Foodborne transmission of nipah virus in Syrian hamsters

    PLoS Pathog

    (2014)
  • D. Middleton et al.

    Hendra virus vaccine, a one health approach to protecting horse, human, and environmental health

    Emerg Infect Dis

    (2014)
  • C.C. Broder et al.

    Immunization strategies against henipaviruses

    Curr Top Microbiol Immunol

    (2012)
  • K. Murray et al.

    A novel morbillivirus pneumonia of horses and its transmission to humans

    Emerg Infect Dis

    (1995)
  • P.T. Hooper et al.

    The retrospective diagnosis of a second outbreak of equine morbillivirus infection

    Aust Vet J

    (1996)
  • R.J. Rogers et al.

    Investigation of a second focus of equine morbillivirus infection in coastal Queensland

    Aust Vet J

    (1996)
  • ProMED-mail

    Hendra virus, equine – Australia (12): (QL) vaccine

    (2012)
  • ProMED-mail

    Hendra virus, equine – Australia (09): New South Wales dog affected

    (2013)
  • ProMED-mail

    Hendra virus, equine – Australia: (08) Queensland, New South Wales

    (2013)
  • ProMED-mail

    Hendra virus, equine – Australia (02): New South Wales

    (2014)
  • ProMED-mail

    Hendra virus, equine – Australia (03): Queensland

    (2014)
  • Hendra virus case confirmed after horse dies in North Queensland

    (2015)
  • ProMED-mail

    Hendra virus, equine – Australia: New South Wales

    (2015)
  • ProMED-mail

    Hendra virus, equine – Australia (04): New South Wales

    (2015)
  • R.K. Plowright et al.

    Urban habituation, ecological connectivity and epidemic dampening: the emergence of Hendra virus from flying foxes (Pteropus spp.)

    Proc R Soc B: Biol Sci

    (2011)
  • R.K. Plowright et al.

    Ecological dynamics of emerging bat virus spillover

    Proc R Soc B: Biol Sci

    (2015)
  • Cited by (61)

    • Update on Viruses in Bats

      2022, Fowler's Zoo and Wild Animal Medicine Current Therapy: Volume 10
    • Safety, tolerability, pharmacokinetics, and immunogenicity of a human monoclonal antibody targeting the G glycoprotein of henipaviruses in healthy adults: a first-in-human, randomised, controlled, phase 1 study

      2020, The Lancet Infectious Diseases
      Citation Excerpt :

      There was a small-to-moderate accumulation of monoclonal antibody in those receiving repeat dose (cohort 5), with elimination kinetics that were similar to those receiving single-dose regimens. A variety of active immunisation strategies have been explored for infection with Hendra virus and Nipah virus involving various recombinant viral vector-based vaccines in animal challenge models.4 These studies showed the possibility of inducing a strong neutralising antibody response that could afford protection in livestock or an emergency-use scenario.4

    View all citing articles on Scopus
    View full text