6,800
Views
26
CrossRef citations to date
0
Altmetric
Editorial

Development of a Zika vaccine

&
Pages 1083-1085 | Received 19 Mar 2016, Accepted 18 May 2016, Published online: 06 Jun 2016

1. Introduction

Currently, there is no approved vaccine against the Zika virus (ZIKV). However, several organizations are actively developing vaccines using various platforms and technologies. While many of these are in the early stages, several are based upon previously approved platforms and designs against dengue and other infectious disease agents. In March 2016, the World Health Organization (WHO) announced a list of organizations describing for approaches toward Zika vaccine development. This short editorial describes the overview of ZIKV and vaccine development.

2. Zika virus

ZIKV is a mosquito-borne Flavivirus that was first reported in humans in 1952 [Citation1]. ZIKV has recently reemerged in Brazil and tropical/subtropical nations [Citation2,Citation3]. Presently, this noxious virus is being widely transmitted by Aedes (Stegomyia) mosquitoes [Citation4]. Worse yet, the Brazilian Ministry of Health confirmed the relation between ZIKV and microcephaly, and WHO and the US Centers for Diseases Control and Prevention issued an alert about the association of ZIKV infection with congenital malformations and neurological syndromes [Citation5,Citation6]. There is now evidence that ZIKV may also be transmitted sexually [Citation7]. The rapid spread of ZIKV raises increasing concerns for public health, such as the increased frequency of Guillain–Barré syndrome and congenital neurological anomalies [Citation8].

ZIKV is an 11-kb single-stranded, positive-sense RNA virus from the Flaviviridae family, most closely related to the Spondweni virus [Citation9]. Two major lineages, African and Asian, have been identified through phylogenetic analyses [Citation10,Citation11]. Transmission occurs via mosquito vectors from the Aedes genus in a sylvatic cycle involving nonhuman primates [Citation12], but little is known about the seasonal and spatial dynamics. A substantial proportion of ZIKV infections are subclinical, but patients may also present with clinical symptoms similar to other arboviral infections, such as dengue virus (DENV) and chikungunya virus [Citation2]. Commonly reported symptoms include rash, fever, arthralgia, and conjunctivitis [Citation13]. Symptoms tend to be mild; however, there is a concern that ZIKV infection may be related to more severe disease pathogenesis.

Each day, more and more information is available regarding the whole genome of ZIKV that may shed light on viral differences between strains. One major reason is diagnosis of ZIKV infection is hampered by serological cross-reactivity with other Flaviviruses, and relies on the detection of ZIKV RNA in blood through ZIKV-specific reverse transcription polymerase chain reaction (RT-PCR) or pan-flavivirus PCR amplification followed by sequencing or viral isolation [Citation14]. For example, DENV is the most prevalent arbovirus in tropical and subtropical countries. However, most cases are acquired from urban transmission by Aedes aegypti mosquitoes. Every year, DENV infections cause more than 50 million cases, 500,000 hospitalizations, and 12,500 deaths worldwide [Citation15]. DENV like ZIKV belongs to the genus Flavivirus and there are four distinct serotypes (DENV-1 to DENV-4), and infection with 1 serotype does not provide long-term, cross-protective immunity [Citation16].

Before 2007, very few human cases of ZIKV infection were reported, but now there is a full-blown epidemic and the beginnings of a ZIKV pandemic [Citation17]. ZIKV from African and Asia lineages cause infection [Citation11]; however, the ‘African strain’ of ZIKV virus is generally non-symptomatic or in some cases causes acute febrile illness that clinically resembled DENV [Citation18]. In contrast, the emerging Asian strain of ZIKV is known to circulate in the same areas and is associated with a suspected ‘congenital syndrome’, while causing asymptomatic or mild febrile infections that can be DENV-like in infected individuals [Citation13]. For both Africa and Asia lineages, little is known about the genetic relationships. The geographic origin of the strains responsible for the recent epidemics most likely is in the French Polynesian islands. There is a need for vaccine countermeasures to control ZIKV spread.

Recently, a clinical trial in which volunteers were infected with the closely related DENV just 6 months after receiving an experimental DENV vaccine or a placebo showed that tetravalent DENV vaccine induces a robust neutralizing antibody response [Citation19]. The candidate DENV vaccine is made from a mixture of four live, attenuated viruses targeted to each of the four serotypes. All 21 volunteers who received the DENV vaccine, TV003, were protected from infection, while all 20 placebo recipients developed infection. The study is important as it shows the proof-of-concept that a DENV vaccine will provide protection against all serotypes of DENV following administration. DENV–host interaction has challenges including severe disease as a consequence of sequential DENV infection, viral immune evasion, host antibody enhancement, host immune activation, and genetic predisposition [Citation20]. The findings are very encouraging as they clear a significant hurdle for the demonstration of vaccine efficacy and avoidance of disease pathogenesis given the world DENV disease burden. Moreover, these results are informative as they help guide vaccine design for ZIKV showing that neutralizing antibodies are clearly the primary feature for generating a safe and protective vaccine against various strains of ZIKV.

The WHO announced the status of active programs in ZIKV development in March 2016 (Current Zika Product Pipeline, 3 March 2016; http://www.who.int/csr/research-and-development/zika-rd-pipeline.pdf) as shown in . Various companies and academic organizations are using a variety of vaccine approaches, including inactivated virus, virus-like particles, nucleic-acid-based vaccines (DNA and RNA), live vectored vaccines, subunit vaccines, and live recombinant approaches. Most of these are in preclinical development and many are expected to enter Phase I clinical studies in 2017. We anticipate many scientific and clinical research publications outlining the results of these vaccine candidates in the near future. While each of these approaches have merit, live attenuated or viral vectored ZIKV vaccines may have the most likelihood of success based upon previous dengue vaccine trials. Licensed vaccines against Yellow Fever, Japanese Encephalitis, and, most recently, Dengue, all of which are in the same viral family as ZIKV, all use the live vectored approach indicate that this approach may work for ZIKV as well.

Table 1. Current Zika vaccine landscape.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

References

  • Dick GW, Kitchen SF, Haddow AJ. Zika virus. I. Isolations and serological specificity. Trans R Soc Trop Med Hyg. 1952;46:509–520.
  • Keighley CL, Saunderson RB, Kok J, et al. Viral exanthems. Curr Opin Infect Dis. 2015;28:139–150. doi:10.1097/QCO.0000000000000145.
  • Gatherer D, Kohl A. Zika virus: a previously slow pandemic spreads rapidly through the Americas. J Gen Virol. 2016;97:269–273. doi:10.1099/jgv.0.000381.
  • Marcondes CB, Ximenes MF. Zika virus in Brazil and the danger of infestation by Aedes (Stegomyia) mosquitoes. Revista da Sociedade Brasileira de Medicina Tropical. 2015. doi:10.1590/0037-8682-0220-2015.
  • Rubin EJ, Greene MF, Baden LR. Zika virus and microcephaly. N Engl J Med. 2016;374:984–985. doi:10.1056/NEJMe1601862.
  • Petersen EE, Staples JE, Meaney-Delman D, et al. Interim guidelines for pregnant women during a Zika virus outbreak - United States, 2016. MMWR Morb Mortal Wkly Rep. 2016;65:30–33. doi:10.15585/mmwr.mm6502e1.
  • Hills SL, Russell K, Hennessey M, et al. Transmission of Zika virus through sexual contact with travelers to areas of ongoing transmission - continental United States, 2016. MMWR Morb Mortal Wkly Rep. 2016;65:215–216. doi:10.15585/mmwr.mm6508e2.
  • Smith DW, Mackenzie J. Zika virus and Guillain-Barre syndrome: another viral cause to add to the list. Lancet. 2016;387:1486–1488. doi:10.1016/S0140-6736(16)00564-X.
  • Hayes EB. Zika virus outside Africa. Emerg Infect Dis. 2009;15:1347–1350. doi:10.3201/eid1509.090442.
  • Faye O, Freire CCM, Iamarino A, et al. Molecular evolution of Zika virus during its emergence in the 20th century. PLoS Negl Trop Dis. 2014;8:e2636. doi:10.1371/journal.pntd.0002636.
  • Haddow AD, Schuh AJ, Yasuda CY, et al. Genetic characterization of Zika virus strains: geographic expansion of the Asian lineage. PLoS Negl Trop Dis. 2012;6:e1477. doi:10.1371/journal.pntd.0001477.
  • Wolfe ND, Kilbourn AM, Karesh WB, et al. Sylvatic transmission of arboviruses among Bornean orangutans. Am J Trop Med Hyg. 2001;64:310–316.
  • Chan JF, Choi GK, Yip CC, et al. Zika fever and congenital Zika syndrome: an unexpected emerging arboviral disease? J Infect. 2016;72:507–524. doi:10.1016/j.jinf.2016.02.011.
  • Faye O, Faye O, Diallo D, et al. Quantitative real-time PCR detection of Zika virus and evaluation with field-caught mosquitoes. Virol J. 2013;10:311. doi:10.1186/1743-422X-10-311.
  • Guzman MG, Halstead SB, Artsob H, et al. Dengue: a continuing global threat. Nat Rev Microbiol. 2010;8:S7–16. doi:10.1038/nrmicro2460.
  • Rossi SL, Nasar F, Cardosa J, et al. Genetic and phenotypic characterization of sylvatic dengue virus type 4 strains. Virology. 2012;423:58–67. doi:10.1016/j.virol.2011.11.018.
  • Lucey DR, Gostin LO. The emerging Zika pandemic: enhancing preparedness. JAMA. 2016;315:865–866. doi:10.1001/jama.2016.0904.
  • Chang C, Ortiz K, Ansari A, et al. The Zika outbreak of the 21st century. J Autoimmun. 2016;68:1–13. doi:10.1016/j.jaut.2016.02.006.
  • Durbin, A. P, Kirkpatrick, BD, Pierce, KK, et al. A 12-month interval dosing study in adults indicates that a single dose of the NIAID tetravalent dengue vaccine induces a robust neutralizing antibody response. J Infect Dis. 2016. doi:10.1093/infdis/jiw067.
  • Thomas SJ, Endy TP. Critical issues in dengue vaccine development. Curr Opin Infect Dis. 2011;24:442–450. doi:10.1097/QCO.0b013e32834a1b0b.

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.