Elsevier

Vaccine

Volume 27, Supplement 4, 5 November 2009, Pages D80-D85
Vaccine

Vaccines for Venezuelan equine encephalitis

https://doi.org/10.1016/j.vaccine.2009.07.095Get rights and content

Abstract

Arboviruses are capable of causing encephalitis in animals and human population when transmitted by the vector or potentially via infectious aerosol. Recent re-emergence of Venezuelan equine encephalitis virus (VEEV) in South America emphasizes the importance of this pathogen to public health and veterinary medicine. Despite its importance no antivirals or vaccines against VEEV are currently available in the USA. Here we review some of the older and newer approaches aimed at generating a safe and immunogenic vaccine as well as most recent data about the mechanistic of protection in animal models of infection.

Section snippets

Venezuelan equine encephalitis virus and its replication

Venezuelan equine encephalitis virus (VEEV, Alphavirus in the Togaviridae family) is an enveloped virus with a non-segmented, positive-sense RNA genome of approximately 11.4 kb (Fig. 1). The 5′ two-thirds of the genome encodes four nonstructural proteins (nsP1 to nsP4) that form an enzyme complex required for viral replication [1], [2], [3]. The full-length RNA then serves as a template for the synthesis of positive-sense genomic RNA and for transcription of a subgenomic 26S RNA [1]. The

Epidemiology of encephalitic alphaviruses

Most of the encephalitic viruses in the Family Togaviridae, genus Alphavirus are zoonotic pathogens that are transmitted via hematophagous arthropods. These pathogens have a widespread distribution in North, Central and South America (reviewed in [5]). Some of them are highly infectious via the aerosol route, thus have been responsible for numerous laboratory accidents (>150 documented cases without an associated perforating injury) and/or have been developed as a biological weapon in the U.S.

Disease in humans

VEEV infection has an incubation period of 2–10 days, which results typically in non-specific flu-like symptoms. Severe encephalitis is a less common outcome of VEEV infection in comparison to EEEV and WEEV infection, although VEEV-associated encephalitis is a more common outcome in children. Neurological disease, including disorientation, ataxia, mental depression, and convulsions can be detected in up to 14% of infected individuals, especially children, although the human case-fatality rate

Mouse model for VEEV infection

The murine model for VEEV-induced disease is established and typically utilizes subcutaneous inoculation [6], [7], [8], [9]. Previous studies have demonstrated that the murine model is characterized by biphasic disease, which starts with the productive infection of lymphoid tissue and culminates in the destruction of the CNS by viral replication and a “toxic” neuroinflammatory response that is uniformly lethal [10], [11], [12], [13], [14], [15], [16]. By the time the acute encephalitis has

Humoral immunity

Protection from peripheral inoculation or natural alphavirus infection depends mostly on the production of neutralizing antibodies [17], [18]. While virus neutralizing antibody is important for the protection against natural (peripheral) challenge mediated by mosquito-borne transmission, more recent studies demonstrate that even relatively high serum titers of polyclonal neutralizing antibody achieved via passive transfer (not achievable with any vaccination known to authors) do not protect

Alpha Beta (αβ) T cell response

The αβ T cells represent the major proportion of T cells that respond to various pathogens and are subdivided into CD4+ helper and CD8+ cytotoxic cells. These “conventional” T cells have been well characterized functionally. Prior studies in mice vaccinated with TC-83 suggest that Th1-type responses predominate [22]. However, in mice vaccinated parenterally with TC-83, cytotoxic T cell activity could not be detected in the spleen or draining lymph node [23]. It was previously demonstrated that

Gamma delta (γδ) T cell response

Recent studies suggest an important role for another well studied T cell subpopulation, γδ T cells, in disease development and lethal outcomes of VEEV infection [19]. Specifically, qualitative and quantitative changes in the inflammatory cellular infiltrates in vaccinated and challenged mice suggest a regulatory role in the secondary response to virus. However, direct evaluation of their role in pathology vs. protection is limited by the lack of feasible methods for isolating sufficient

Live-attenuated VEE vaccines

Following upon the success of the 17D yellow fever vaccine by Theiler [43], VEEV was attenuated by 83 serial passages in guinea pig heart cells to produce the TC-83 strain [44]. TC-83 was first tested extensively in equids during the 1971 Texas VEE epizootic/epidemic, where it may have contributed to limiting the spread northward. Although the vaccine produces viremia, fever and leucopenia in horses, robust neutralizing antibodies are generated as well as protection from VEEV challenge [45].

Inactivated VEE vaccines

Due to the economic devastation caused by VEE epizootics in regions of Latin American that relied on equids for agriculture and transportation, vaccines were first produced soon after VEEV was isolated in 1938 [59], [60]. Formalin-inactivated preparations were initially made from mouse brain and other animal tissues following infection with wild-type, subtype IAB strains isolated during epizootics [61]. These vaccines were probably efficacious in most animals, but the equid-amplification

Sindbis virus-based chimeric vaccine approach

Recombinant live-attenuated vaccines and, in particular, an alphavirus-based approach, represent a viable approach to the production of safe, immunogenic and efficacious vaccines against the encephalitis alphaviruses [19], [21], [65], [66], [67]. By utilizing as a vector the genome of Sindbis virus (SINV), a relatively nonpathogenic alphavirus in humans, chimeric SIN/VEE virus(es) can be designed to express all of the structural proteins of the virulent alphavirus. These constructs contain the

Alphavirus replicons

Because only the nonstructural proteins and cis-acting RNA sequences are required for alphavirus genome replication, the structural protein genes can be replaced and foreign antigens expressed at high levels [68], [69]. These replicon genomes can be packaged into virus-like particles by capsid and envelope proteins provided in trans from a second genetic construct (Fig. 4). To reduce the probability of recombination between replicon and “helper” RNAs, two separate helpers can be used to encode

References (76)

  • J.F. Rowell et al.

    Contribution of T cells to mortality in neurovirulent Sindbis virus encephalomyelitis

    J Neuroimmunol

    (2002)
  • D. Kagi et al.

    Different roles for cytotoxic T cells in the control of infections with cytopathic versus noncytopathic viruses

    Curr Opin Immunol

    (1996)
  • J.A. Trapani

    Dual mechanisms of apoptosis induction by cytotoxic lymphocytes

    Int Rev Cytol

    (1998)
  • S. Shresta et al.

    How do cytotoxic lymphocytes kill their targets?

    Curr Opin Immunol

    (1998)
  • A.A. Dandekar et al.

    Virus-induced demyelination in nude mice is mediated by gamma delta T cells

    Am J Pathol

    (2002)
  • P.R. Pittman et al.

    Long-term duration of detectable neutralizing antibodies after administration of live-attenuated VEE vaccine and following booster vaccination with inactivated VEE vaccine

    Vaccine

    (1996)
  • W.D. Pratt et al.

    Use of telemetry to assess vaccine-induced protection against parenteral and aerosol infections of Venezuelan equine encephalitis virus in non-human primates

    Vaccine

    (1998)
  • W.D. Pratt et al.

    Genetically engineered, live attenuated vaccines for Venezuelan equine encephalitis: testing in animal models

    Vaccine

    (2003)
  • M.K. Hart et al.

    Improved mucosal protection against Venezuelan equine encephalitis virus is induced by the molecularly defined, live-attenuated V3526 vaccine candidate

    Vaccine

    (2000)
  • V. Rao et al.

    Environmental hazard assessment of Venezuelan equine encephalitis virus vaccine candidate strain V3526

    Vaccine

    (2004)
  • D.S. Reed et al.

    Genetically engineered, live, attenuated vaccines protect nonhuman primates against aerosol challenge with a virulent IE strain of Venezuelan equine encephalitis virus

    Vaccine

    (2005)
  • D.L. Fine et al.

    Neurovirulence evaluation of Venezuelan equine encephalitis (VEE) vaccine candidate V3526 in nonhuman primates

    Vaccine

    (2008)
  • E. Volkova et al.

    IRES-dependent replication of Venezuelan equine encephalitis virus makes it highly attenuated and incapable of replicating in mosquito cells

    Virology

    (2008)
  • E. Wang et al.

    Chimeric Sindbis/eastern equine encephalitis vaccine candidates are highly attenuated and immunogenic in mice

    Vaccine

    (2007)
  • P. Pushko et al.

    Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo

    Virology

    (1997)
  • X. Zhou et al.

    Self-replicating Semliki Forest virus RNA as recombinant vaccine

    Vaccine

    (1994)
  • L.C. Dupuy et al.

    Directed molecular evolution improves the immunogenicity and protective efficacy of a Venezuelan equine encephalitis virus DNA vaccine

    Vaccine

    (2009)
  • E.G. Strauss et al.

    Structure and replication of the alphavirus genome

  • J.H. Strauss et al.

    Togaviridae

  • J.H. Strauss et al.

    The alphaviruses: gene expression, replication, and evolution

    Microbiol Rev

    (1994)
  • C.M. Rice et al.

    Nucleotide sequence of the 26S mRNA of Sindbis virus and deduced sequence of the encoded virus structural proteins

    Proc Natl Acad Sci USA

    (1981)
  • N.L. Davis et al.

    A molecular genetic approach to the study of Venezuelan equine encephalitis virus pathogenesis

    Arch Virol Suppl

    (1994)
  • W.D. Kundin

    Pathogenesis of Venezuelan equine encephalomyelitis virus. II. Infection in young adult mice

    J Immunol

    (1966)
  • A.C. Jackson et al.

    Pathogenesis of Venezuelan equine encephalitis virus infection in mice and hamsters

    Vet Pathol

    (1991)
  • B.A. Schoneboom et al.

    Astrocytes as targets for Venezuelan equine encephalitis virus infection

    J Neurovirol

    (1999)
  • J. Garcia-Tamayo et al.

    Central nervous system alterations as sequelae of Venezuelan equine encephalitis virus infection in rat

    J Pathol

    (1979)
  • L. Gorelkin

    Venezuelan equine encephalomyelitis in an adult animal host. An electron microscopic study

    Am J Pathol

    (1973)
  • A.B. Ryzhikov et al.

    Spread of Venezuelan equine encephalitis virus in mice olfactory tract

    Arch Virol

    (1995)
  • Cited by (0)

    View full text