899
Views
8
CrossRef citations to date
0
Altmetric
Editorial

Can understanding the virulence mechanisms of RNA viruses lead us to a vaccine against eastern equine encephalitis virus and other alphaviruses?

, &

Abstract

Eastern equine encephalitis virus (EEEV) is a highly neurovirulent mosquito-borne alphavirus that causes severe morbidity and mortality upon human infection. Recent emergence of EEEV into nonendemic regions in the USA and Panama demonstrates the need for the development of an effective EEEV vaccine for licensure for human use. The current EEEV vaccine is available to only at-risk laboratory workers but is poorly immunogenic and requires multiple boosters. In this editorial, we summarize recent developments in understanding alphavirus virulence mechanisms that could be utilized to rationally design a live attenuated vaccine against EEEV or other alphaviruses.

In recent years, the spread of mosquito-borne diseases into previously nonendemic areas (e.g., West Nile flavivirus into the USA and chikungunya (CHIKV) alphavirus into Europe and the Caribbean) has raised concerns and increased the urgency for developing effective vaccines to combat these pathogens. Similarly, the known geographical range of eastern equine encephalitis virus (EEEV) and the incidence of disease have increased in the eastern USA and EEEV also has recently emerged in Panama Citation[1,2]. While relatively rare in humans, EEEV causes one of the most severe acute viral infections, resulting in 30–70% mortality among symptomatic cases Citation[3]. Both Venezuelan equine encephalitis alphavirus (VEEV) and EEEV have the capacity to cause severe disease after aerosol delivery, which raises concerns for the possibility of their use as agents of biowarfare/bioterrorism Citation[4].

A formalin-inactivated EEEV vaccine is currently under investigational new drug status and is available to laboratory researchers; however, immunization can be cost-prohibitive, and, like many inactivated vaccines, can require multiple boosters to induce a protective immune response Citation[5]. Two EEEV live attenuated vaccines (LAVs), a chimeric Sindbis virus encoding the structural proteins of EEEV and a similar vaccine encoding an internal ribosome entry site in the subgenomic promoter, have shown protective efficacy in animal models but as yet are unlicensed Citation[6,7]. An LAV against VEEV is similarly available as an investigational new drug, but is not likely to be approved due to poor immunogenicity and adverse reactions in 20% of vaccinees Citation[8]. Finally, a cell culture adapted CHIKV LAV (181/25) has been given to at-risk researchers, but incomplete attenuation has also led to adverse events Citation[9]. The ‘gold standard’ for LAVs is the yellow fever virus 17D vaccine that induces life-long immunity in humans, but, even with this vaccine, adverse events can occur after vaccination Citation[10]. In the following paragraphs, we summarize recent advancements in understanding the pathogenesis of alphaviruses and especially, EEEV, which we feel can be utilized to design next-generation LAVs.

The VEEV LAV, TC-83, encodes two attenuating point mutations that (i) increase replication of the genomic RNA in relation to production of subgenomic RNA while also altering the stem-loop conformation of the 5′ nontranslated region (NTR) of the viral RNA genome rendering TC-83 sensitive to IFIT1-mediated antiviral restriction, or (ii) alter binding to the host glycosaminoglycan heparan sulfate (HS) Citation[11,12]. Importantly, wild-type VEEV strains evade binding of IFIT1 through changes in the stem-loop conformation of the 5′ NTR thus evading one aspect of the host antiviral system Citation[12]. Incorporating mutations to render viral RNA sensitive to IFIT1 antiviral restriction will be an important step in the generation of an alphavirus LAV.

The acquisition of a positively charged amino acid in the E2 glycoprotein of TC-83 renders the virus an efficient binder of HS structures Citation[13]. Passage in culture, natural variation or deliberate mutagenesis can alter HS binding versus wild-type alphavirus strains, typically increasing it with poorly binding viruses (e.g., CHIKV and VEEV), while lowering or altering its qualities with EEEV, thereby attenuating the virus in animal models of disease with attenuation mechanisms different between the viruses Citation[13–16]. Adaptation to HS binding has been proposed as a rational method for the development of alphavirus LAVs previously and should be considered for next-generation LAVs Citation[14,16].

During virus replication, alphavirus proteins arrest host macromolecular synthesis by inhibiting host cell translation and transcription, potentially antagonizing innate immune responses to infection Citation[17]. Mutations in the capsid protein render EEEV or VEEV unable to shut-off host gene transcription, thereby attenuating the virus in vivo Citation[18,19]. Furthermore, mutations in nsP2 have been identified that affect macromolecular synthesis shutoff and interferon (IFN) signaling inhibition Citation[20,21]. However, the nature of the mutations and their effects are different for each alphavirus. The addition of mutations to a LAV for EEEV or other alphaviruses that remove/attenuate their ability to shut-off host macromolecular synthesis or IFN signaling inhibition will further limit the ability of EEEV to replicate and potentially progress to encephalitic disease.

EEEV also encodes a virulence mechanism in the 3′ NTR that contributes to the limited prodrome seen during infection prior to manifestation of encephalitic disease Citation[22,23]. Remarkably, we found that the 3′ NTR of EEEV RNA genome encodes binding sites for the myeloid-specific miRNA, miR-142-3p. Binding of the miRNA to EEEV represses viral translation and replication only in myeloid cells likely limiting both innate and acquired immune responses to EEEV. Removal of the miRNA binding sites rescued myeloid cell replication both in vitro in myeloid cell lines and in vivo in myeloid cells in the popliteal lymph node. Importantly, this myeloid cell replication in vivo increased the production of systemic type I IFN and attenuated the mutant viruses demonstrating a correlation between type I IFN production and the attenuation of EEEV Citation[23]. In combination with removal of the miR-142-3p binding sites, alteration of HS binding of EEEV increased replication in lymphoid tissues, type I IFN production and prodrome in comparison to viral mutants eliminating only miR-142-3p binding or HS binding Citation[23]. Therefore, an effective EEEV LAV would need both HS and miR-142-3p binding altered to efficiently replicate in peripheral lymphoid tissues inducing a protective immune response. Currently, it is unclear whether or not other alphaviruses utilize miRNA inhibition to determine tissue tropism in vivo.

In addition, although humans are considered dead-end hosts for EEEV, an ideal EEEV LAV candidate would not be replication competent in mosquitoes, preventing transmission from vaccinees to mosquitoes as was seen for the VEEV LAV during an equine vaccination campaign during an epizootic outbreak Citation[24]. Interestingly, removal of the miR-142-3p binding sites from EEEV and the EEEV internal ribosome entry site LAV were unable to establish viral replication in mosquitoes, thus eliminating the potential transmission from vaccinees to mosquitoes Citation[6,23].

Both the CHIKV 181/25 and VEEV TC-83 LAV induce adverse events in vaccinees suggesting that the attenuating mutations are not stable and improvements are needed before mass vaccinations Citation[8,9]. To generate a LAV for EEEV, this risk of reverting to virulent phenotypes will need to be considered and mitigated. For example, rather than mutating a single nucleotide, multiple changes in the 5′ NTR nucleotide sequence may be needed to stably change the stem-loop structure or multiple mutations altering HS binding could be combined rather than a single mutation as commonly found in LAVs derived from blind passage Citation[16]. This will be particularly important with viruses such as EEEV that cause extremely severe disease at high frequency.

We propose the formal testing of the individual and combined effects of mutations in the 5′ NTR, nsP2, capsid, E2 glycoprotein and, with EEEV, 3′ NTR upon attenuation and reversion potential as a candidate strategy for an improved alphavirus LAV. The recent discoveries of the mechanisms of action of virulence factors contributing to alphavirus pathogenesis suggest that these can be utilized to rationally design a LAV for EEEV or other arboviruses. However, as noted above, mutations can have different consequences for different viruses or some may only be applicable to a subset (e.g., miRNA restriction of EEEV) such that thorough empirical testing is required for each combination and virus. The current vaccines are either weak, inactivated formulations or can induce adverse events in vaccinees that will not be suitable for mass vaccinations in the case of widespread outbreak or malicious release. We believe that a novel LAV vaccine against EEEV can be developed to induce a protective immune response while limiting virulence and potential reversions to virulence, and that this strategy will be broadly applicable to other arboviruses.

Financial & competing interests disclosure

This work was supported by NIH training grants AI049820 and AI060525 (DW Trobaugh), research grants U54 AI081680 and AI081886 (KD Ryman), and AI083383 and AI095436 (WB Klimstra). The authors have no other relevant affiliations or financial involvement with any organization or entity with an interest in or conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Armstrong PM, Andreadis TG. Eastern equine encephalitis virus – old enemy, new threat. N Engl J Med 2013;368(18):1670-3
  • Carrera J-P, Forrester N, Wang E, et al. Eastern equine encephalitis in Latin America. N Engl J Med 2013;369(8):732-44
  • Silverman MA, Misasi J, Smole S, et al. Eastern equine encephalitis in children, Massachusetts and New Hampshire,USA, 1970–2010. Emerg Infect Dis 2013;19(2):194-201
  • Reed DS, Lackemeyer MG, Garza NL, et al. Severe encephalitis in cynomolgus macaques exposed to aerosolized Eastern equine encephalitis virus. J Infect Dis 2007;196(3):441-50
  • Bartelloni PJ, McKinney RW, Duffy TP, Cole FE. An inactivated eastern equine encephalomyelitis vaccine propagated in chick-embryo cell culture. II. Clinical and serologic responses in man. Am J Trop Med Hyg 1970;19(1):123-6
  • Pandya J, Gorchakov R, Wang E, et al. A vaccine candidate for eastern equine encephalitis virus based on IRES-mediated attenuation. Vaccine 2012;30(7):1276-82
  • Roy CJ, Adams AP, Wang E, et al. A chimeric Sindbis-based vaccine protects cynomolgus macaques against a lethal aerosol challenge of eastern equine encephalitis virus. Vaccine 2013;31(11):1464-70
  • Pittman PR, Makuch RS, Mangiafico JA, 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;14(4):337-43
  • Hoke CH, Pace-Templeton J, Pittman P, et al. US Military contributions to the global response to pandemic chikungunya. Vaccine 2012;30(47):6713-20
  • Barrett ADT, Monath TP, Barban V, et al. 17D yellow fever vaccines: new insights. A report of a workshop held during the World Congress on medicine and health in the tropics, Marseille, France, Monday 12 September 2005. Vaccine 2007;25(15):2758-65
  • Kulasegaran-Shylini R, Thiviyanathan V, Gorenstein DG, Frolov I. The 5′UTR-specific mutation in VEEV TC-83 genome has a strong effect on RNA replication and subgenomic RNA synthesis, but not on translation of the encoded proteins. Virology 2009;387(1):211-21
  • Hyde JL, Gardner CL, Kimura T, et al. A viral RNA structural element alters host recognition of nonself RNA. Science 2014;343(6172):783-7
  • Bernard KA, Klimstra WB, Johnston RE. 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;276(1):93-103
  • Gardner CL, Ebel GD, Ryman KD, Klimstra WB. Heparan sulfate binding by natural eastern equine encephalitis viruses promotes neurovirulence. Proc Natl Acad Sci USA 2011;108(38):16026-31
  • Gardner CL, Choi-Nurvitadhi J, Sun C, et al. Natural variation in the heparan sulfate binding domain of the eastern equine encephalitis virus E2 glycoprotein alters interactions with cell surfaces and virulence in mice. J Virol 2013;87(15):8582-90
  • Gardner CL, Hritz J, Sun C, et al. Deliberate attenuation of chikungunya virus by adaptation to heparan sulfate-dependent infectivity: a model for rational arboviral vaccine design. PLoS Negl Trop Dis 2014;8(2):e2719
  • Yin J, Gardner CL, Burke CW, et al. Similarities and differences in antagonism of neuron alpha/beta interferon responses by Venezuelan equine encephalitis and sindbis alphaviruses. J Virol 2009;83(19):10036-47
  • Aguilar PV, Leung LW, Wang E, et al. A five-amino-acid deletion of the eastern equine encephalitis virus capsid protein attenuates replication in mammalian systems but not in mosquito cells. J Virol 2008;82(14):6972-83
  • Garmashova N, Atasheva S, Kang W, et al. Analysis of Venezuelan equine encephalitis virus capsid protein function in the inhibition of cellular transcription. J Virol 2007;81(24):13552-65
  • Frolov I, Garmashova N, Atasheva S, Frolova EI. Random insertion mutagenesis of sindbis virus nonstructural protein 2 and selection of variants incapable of downregulating cellular transcription. J Virol 2009;83(18):9031-44
  • Fros JJ, Liu WJ, Prow NA, et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferon-stimulated JAK-STAT signaling. J Virol 2010;84(20):10877-87
  • Gardner CL, Burke CW, Tesfay MZ, et al. Eastern and Venezuelan equine encephalitis viruses differ in their ability to infect dendritic cells and macrophages: impact of altered cell tropism on pathogenesis. J Virol 2008;82(21):10634-46
  • Trobaugh DW, Gardner CL, Sun C, et al. RNA viruses can hijack vertebrate microRNAs to suppress innate immunity. Nature 2014;506(7487):245-8
  • Pedersen CE, Robinson DM, Cole FE. Isolation of the vaccine strain of Venezuelan equine encephalomyelitis virus from mosquitoes in Louisiana. Am J Epidemiol 1972;95(5):490-6

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.