Elsevier

Vaccine

Volume 30, Issue 7, 8 February 2012, Pages 1276-1282
Vaccine

A vaccine candidate for eastern equine encephalitis virus based on IRES-mediated attenuation

https://doi.org/10.1016/j.vaccine.2011.12.121Get rights and content

Abstract

To develop an effective vaccine against eastern equine encephalitis (EEE), we engineered a recombinant EEE virus (EEEV) that was attenuated and capable of replicating only in vertebrate cells, an important safety feature for live vaccines against mosquito-borne viruses. The subgenomic promoter was inactivated with 13 synonymous mutations and expression of the EEEV structural proteins was placed under the control of an internal ribosomal entry site (IRES) derived from encephalomyocarditis virus (EMCV). We tested this vaccine candidate for virulence, viremia and efficacy in the murine model. A single subcutaneous immunization with 104 infectious units protected 100% of mice against intraperitoneal challenge with a highly virulent North American EEEV strain. None of the mice developed any signs of disease or viremia after immunization or following challenge. Our findings suggest that the IRES-based attenuation approach can be used to develop a safe and effective vaccine against EEE and other alphaviral diseases.

Highlights

► Design of a recombinant EEE virus (EEEV/IRES), as a candidate vaccine for EEEV. ► Subgenomic promoter of wild type EEEV inactivated by IRES-based attenuation approach. ► Virulence, viremia and efficacy of EEEV/IRES tested in murine model. ► EEEV/IRES incapable of infecting mosquitoes.

Introduction

Viruses in the genus Alphavirus, family Togaviridae, have positive strand RNA genomes and cause a spectrum of diseases that includes fever, rash, arthritis, meningitis and encephalitis. Most alphaviruses are mosquito-borne, and transmitted in nature among zoonotic vertebrate reservoir hosts, including birds, primates and rodents. Based on their cross reactivity, several antigenic complexes including the EEE, Venezuelan (VEE) and western equine encephalitis (WEE) complexes comprise the Alphavirus genus [1], [2]. EEEV is considered the most deadly of all the alphaviruses due to the high case fatality rates associated with infections, reaching as high as 90% in horses. In humans, the estimated case fatality rate approaches 80% and many survivors exhibit crippling sequelae such as mental retardation, convulsions, and paralysis that require life-long institutionalized care [3], [4].

Based on antigenic and genetic analyses, EEEV is classified into 4 subtypes: subtype 1 includes strains from North America (NA), whereas the remaining 3 are found in Central and South America (SA) [5], [6]. In general, EEEV strains from SA appear to be less virulent for humans than NA strains [7]. The former can occasionally cause disease and death in horses, but human infections are rarely recognized. In contrast, NA strains are genetically conserved, uniformly virulent and cause severe encephalitis in both humans and equids [8], [9], [10]. However, in mice, both NA and SA strains of EEEV are highly virulent and cause mortality rates as high as 70–90% following subcutaneous, intraperitoneal or intramuscular infection [11], [12]. In outbred laboratory mice, EEEV produces neurologic disease that resembles that following human and equine infections. Virus has been detected in the brain as early as day 1 post-infection (PI), and signs of disease are evident as early as days 3–4. Clinical signs of murine disease include ruffled hair, anorexia, vomiting, lethargy, posterior limb paralysis, convulsions, and coma. Histopathological studies reveal extensive involvement of the brain, including neuronal degeneration, cellular infiltration, and perivascular cuffing, similar to the pathological changes of the central nervous system that are described in naturally infected humans [11]. Immune protection against alphaviruses has been attributed mainly to the humoral response, with titers of neutralizing antibodies directly proportional to the level of protection against disease upon challenge [13], [14], [15], [16].

Despite over 65 years of research there is no licensed human vaccine or effective antiviral treatment available for human EEE, and control depends on mosquito abatement measures and avoidance of exposure to mosquito bites. Several live-attenuated candidate vaccines derived from the wild-type, virulent EEEV were assessed for their safety and efficacy in mice [17], [18] but have not been developed further. A formalin-inactivated vaccine prepared from a wild-type NA strain of EEEV [19] under Investigational New Drug (IND) status is currently available only for researchers and military personnel [20], and a similar inactivated vaccine is sold for equids and other domesticated animals [21], [22], [23], [24], [25], [26]. Recently, the U.S. Department of Agriculture licensed a three-component vaccine composed of inactivated VEE, EEE and WEE viruses, for use in equids [27], [28]. However, all of these inactivated vaccines suffer from poor immunogenicity and from the risk of residual live virus in the vaccine lot [29].

In addition to its importance as a natural pathogen of humans and domesticated animals, EEEV is a potent biological weapon [30], adding further urgency to the need for an effective vaccine. Therefore, we sought to develop an attenuated EEEV strain that would induce rapid, robust and long-lived immunity after a single vaccination. Earlier, Wang et al. developed two chimeric alphavirus strains (SIN/EEEV), both of which replicate efficiently in mammalian and mosquito cell cultures without any adaptation [31]. These vaccines were shown to be highly attenuated, immunogenic and efficacious against EEEV challenge in murine testing. Another chimeric alphavirus, which is derived from a cDNA clone encoding the western equine encephalitis virus (WEEV) nonstructural polyprotein and the EEEV structural polyprotein protects mice against EEEV but not against WEEV challenge [32]. Because the recombinant nature of these chimeras is believed to confer the attenuation phenotype, rather than point mutations that are subject to high rates of reversion, these chimeric viruses appear to be stably attenuated [33]. However, another safety concern for live, genetically engineered vaccines against arboviral diseases is that recombinant viruses might evolve in unforeseen and unexpected ways if they inadvertently underwent vector-borne circulation in the nature. The isolation of the VEEV vaccine strain, TC-83, from naturally infected mosquitoes collected in Louisiana during a 1971 Texas epidemic demonstrated the risk of transmission of attenuated alphaviruses [34], and experimental infections of the chimeric EEE vaccine strains demonstrated some residual mosquito infectivity [35].

To eliminate potential mosquito infectivity of live alphavirus vaccine strains, the findings of Finkelstein et al., that insect cells do not support efficient internal translation initiation from an encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES) [36], were exploited in another vaccine design [37]. The VEEV vaccine strain TC-83 was modified in cDNA clone form to replace the subgenomic promoter with EMCV IRES to drive expression of the structural protein genes from genomic RNA in mammalian cells. The resulting strain is incapable of infecting mosquitoes, and is also further attenuated for mice. Beginning with a wild-type backbone of chikungunya virus, this IRES approach produced a vaccine candidate that appears to have an optimal balance of immunogenicity and attenuation, combined with a lack of mosquito infectivity [38]. In addition to EMCV IRES, attenuation and elimination of mosquito infectivity was also dependent on inactivated subgenomic promoter as previously reported by Volkova et al., and Plante et al. Northern blot analysis performed by both these groups confirmed that insertion of several synonymous point mutations efficiently inactivated the subgenomic promoter [37], [38].

Considering these promising results, we utilized the EMCV IRES approach to generate an attenuated, mosquito-incompetent EEEV vaccine candidate. This recombinant virus was evaluated in mice to assess attenuation, immunogenicity and protection against EEEV challenge, and was also tested for its ability to infect mosquito cells and mosquitoes in vivo. The strong attenuation and mosquito-incompetent phenotypes we measured further support the promise of this approach for alphavirus vaccine development.

Section snippets

Cell lines

Baby hamster kidney (BHK-21) and Vero cells were obtained from the American Type Culture Collection (ATCC, Bethesda, MD) and grown at 37 °C in Eagle's minimal medium (MEM) with 5% fetal bovine serum (FBS) and 0.005% gentamycin sulphate. The mosquito cell line C6/36, also from the ATCC, was maintained in MEM at 32 °C with 5% FBS and 10% tryptose phosphate broth.

Virus

Wild-type (WT) North American EEEV strain FL93-939 was used for the development of the recombinant construct and also for all challenge

Recombinant virus

In the recombinant virus EEEV/IRES, the subgenomic promoter was inactivated by 16 synonymous point mutations, which prevented the reversion to an active SG RNA promoter. To promote synthesis of the EEEV structural proteins, the IRES sequence was cloned to replace the 5′ UTR in the subgenomic RNA. The infectivity of the recombinant viral EEEV/IRES RNA as determined by infectious center assay was 5 × 105 PFU/4 μg, which is comparable to that of other alphavirus cDNA clones [37], [41]. A small and

Discussion

Traditionally, live-attenuated RNA virus vaccines have been generated by serial passage through cell cultures, which results in attenuation that is dependent on a small numbers of point mutations [43], [44], [45], [46]. Because of the instability of the genome during RNA virus replication, these point mutations are likely to revert in vivo, or the virus may acquire a compensatory mutation(s) that can restore virulence. The live-attenuated VEE vaccine, TC-83 is a good example. This vaccine,

Acknowledgements

We thank Rui Mei Yun for excellent technical assistance and Jing Huang for rearing mosquitoes and assistance with mosquito work. This work was supported by a grant from the National Institute of Allergy and Infectious Disease (NIAID) through the Western Regional Center of Excellence for Biodefense and Emerging Infectious Disease Research, National Institutes of Health (NIH) grant U54 AIO57156.

References (54)

  • Y. Finkelstein et al.

    The use of bi-cistronic transfer vectors for the baculovirus expression system

    J Biotechnol

    (1999)
  • 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)
  • P. Minor

    Vaccine-derived poliovirus (VDPV): impact on poliomyelitis eradication

    Vaccine

    (2009)
  • B.L. Jacobs et al.

    Vaccinia virus vaccines: past, present and future

    Antiviral Res

    (2009)
  • A.D. Barrett et al.

    Yellow fever vaccine: how does it work and why do rare cases of serious adverse events take place

    Curr Opin Immunol

    (2009)
  • N.L. Davis et al.

    Attenuating mutations in the E2 glycoprotein gene of Venezuelan equine encephalitis: Construction of single and multiple mutants in a full-length clone

    Virology

    (1991)
  • R.M. Kinney et al.

    The full-length nucleotide sequences of the virulent Trinidad donkey strain of Venezuelan equine encephalitis virus and its attenuated vaccine derivative, strain TC-83

    Virology

    (1989)
  • 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)
  • K.A. Bernard et al.

    Mutations in the E2 glycoprotein of Venezuelan equine encephalitis virus confer heparan sulfate interaction, low morbidity and rapid clearance from blood of mice

    Virology

    (2000)
  • C.H. Calisher et al.

    Arbovirus subgroups: defination and geographical distribution

  • R.F. Feemster et al.

    Easten equine encephalitis

    Neurology

    (1958)
  • R.E. Johnson et al.

    Alphaviruses

  • J. Casals

    Antigenic variants of eastern equine encephalitis virus

    J Exp Med

    (1964)
  • A.C. Brault et al.

    Genetic and antigenic diversity among eastern equine encephalitis viruses from North, Central, and South America

    Am J Trop Med Hyg

    (1999)
  • S.C. Weaver et al.

    Evolution of alphaviruses in the eastern equine encephalomyelitis complex

    J Virol

    (1994)
  • S.C. Weaver

    Eastern equine encephalitis

  • S.C. Weaver et al.

    Alphavirus infections

  • Cited by (23)

    • CpG/Poly (I:C) mixed adjuvant priming enhances the immunogenicity of a DNA vaccine against eastern equine encephalitis virus in mice

      2014, International Immunopharmacology
      Citation Excerpt :

      It is also recommended that researchers be vaccinated with an EEEV vaccine. However, neither a human vaccine nor antiviral drugs are currently available for the prevention and treatment of EEEV [32]. Recently, it has been suggested that a pseudovirus presenting native, neutralizing epitopes on its surface may offer a stable, safe source of alternatives to the live virus [33,34].

    • Novel vaccination approaches against equine alphavirus encephalitides

      2014, Vaccine
      Citation Excerpt :

      The development of equine alphavirus vaccines using a poxvirus-vectored system for equids seems to be a promising approach. Encouraging, but still barely preliminary, are the results obtained with IRES-based technology, which has shown to induce protection against homologous viral challenge, guaranteeing phenotypic stability and inhibiting natural dissemination by limiting viral replication in mosquitoes [6,39]. Considering efficiently proved second generation vaccines in the mouse model, further studies conducted in horses seem to be of high priority.

    • West nile virus and equine encephalitis viruses: New perspectives

      2014, Veterinary Clinics of North America - Equine Practice
      Citation Excerpt :

      Development of new formulations of VEE vaccines is an intense area of research.41–43,46,48,62–67 Several experimental vaccines have been developed and these include subunit vaccines using various constructs and gene deletions, DNA vaccines, adenovirus-vectored vaccines, and encephalomyocarditis virus (internal ribosome entry site) vaccines.41–43,46,48,62–67 Of all of these, the gene-deleted live mutant vaccine, V3526, has been tested in horses and has demonstrated 100% clinical efficacy against challenge with 104 pfu of live virus.68

    • IRES-based Venezuelan equine encephalitis vaccine candidate elicits protective immunity in mice

      2013, Virology
      Citation Excerpt :

      Both murine (Steele et al., 1998) and NHP (Reed et al., 2005) models for VEEV aerosol exposure have been established, allowing for the efficacy of the 68U201/IRES vaccines to be measured. The IRES-based attenuation technology described here has also been successfully applied to other alphaviruses to create safe and efficacious vaccines (Pandya et al., 2012; Plante et al., 2011; Volkova et al., 2008). The technology is easily transferable between different alphaviruses because the mechanism of attenuation is a property of changes the alphavirus gene expression, and not of a particular virus strain.

    View all citing articles on Scopus
    View full text