Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jul 11.
Published in final edited form as: Cell Host Microbe. 2018 Jun 28;24(1):12–17. doi: 10.1016/j.chom.2018.05.021

Zika virus vaccine: Progresses and challenges

Chao Shan 1, Xuping Xie 1, Pei-Yong Shi 1,*
PMCID: PMC6112613  NIHMSID: NIHMS976018  PMID: 30008291

SUMMARY

The explosive emergence of Zika virus has inspired a global effort to develop vaccines. Zika virus, which is a flavivirus primarily transmitted by mosquitoes, can cause devastating congenital syndrome in fetuses of pregnant women, including microcephaly, craniofacial disproportion, spasticity, ocular abnormalities, and miscarriage. In adults, Zika infection has been linked to the autoimmune disorder Guillain-Barré syndrome. Thus, despite the current waning in newly reported Zika infections, an efficacious vaccine is urgently needed to help limit the emergence of another detrimental epidemic. Here we summarize the current status of the Zika vaccine pipeline and highlight the challenges for clinical efficacy trials.

MAIN TEXT

Zika virus (ZIKV) is a mosquito-borne flavivirus that has recently caused global epidemics. Many other flaviviruses, including the four serotypes of dengue virus (DENV), yellow fever virus (YFV), West Nile virus (WNV), Japanese encephalitis virus (JEV), and tick-borne encephalitis virus (TBEV), also cause global outbreaks and epidemics, posing constant threats to public health. ZIKV was first isolated from a sentinel rhesus macaque in the Zika Forest of Uganda in 1947. Outside Africa, ZIKV was isolated for the first time from Aedes aegypti mosquitoes in 1966 in Malaysia. Before 2007, ZIKV had silently circulated between primates and mosquitoes in forests in Africa and Southeast Asia without causing detectable outbreaks and severe human disease. However, ZIKV caused an epidemic on Yap Island, Micronesia in 2007, spawned large epidemics in French Polynesia and other regions of the South Pacific in 2013–14, and arrived in the Americas in 2014, causing >700,000 cases of autochthonous infection (Ikejezie et al., 2017). Importantly, during the recent epidemics, ZIKV infections were shown to cause devastating congenital Zika syndromes (CZS, including microcephaly, congenital malformation, and fetal demise) in about 6–11% of the fetuses from infected pregnant women (Hoen et al., 2018). In adults, Zika infections may cause Guillain-Barré syndrome (GBS), an autoimmune disease caused by the immune system attacking the peripheral nerves, leading to a rapid onset of muscle weakness and even paralysis (Dos Santos et al., 2016). The CZS and GBS potential of ZIKV was unexpected because the virus had only been associated with mild disease before 2013. Due to the explosive epidemics and teratogenic potential, the WHO declared ZIKV to be a Public Health Emergency of International Concern in February 2016. Since then, intensive global efforts have been made to understand ZIKV biology and to develop countermeasures at an unprecedented pace, leading to the establishment of a promising vaccine portfolio.

Licensed flavivirus vaccines

The feasibility to develop a safe and efficacious ZIKV vaccine is supported by the availability of licensed vaccines for other four flaviviruses: YFV (live-attenuated), TBEV (inactivated), JEV (both inactivated and live-attenuated), and DENV (chimeric live-attenuated). Lessons learned from these licensed flavivirus vaccines should be useful to guide the development of a ZIKV vaccine. Due to the four serotypes of DENV (30–35% amino acid variation among serotypes), the development of a tetravalent DENV vaccine has proven challenging. An effective DENV vaccine needs to induce long-lasting immune protection against all four serotypes. Otherwise, a vaccine recipient with incomplete immunization may become sensitized to life-threatening dengue hemorrhagic fever or dengue shock syndrome upon subsequent DENV infections. This challenge was reflected by the results of phase 2b/3 trials of Dengvaxia with an average efficacy of 30–61%. Unfortunately, Dengvaxia seems to increase the risk of hospitalization over time in children <9 years of age who were seronegative at the time of vaccination, possibly through vaccine-induced antibody enhancement. In contrast to DENV, ZIKV exists as two genetic lineages (African and Asian) with <5% amino acid variation among the different strains. Immune serum from ZIKV-infected humans and animals was shown to equivalently neutralize various ZIKV strains from both lineages, demonstrating ZIKV as a single serotype (Dowd et al., 2016a). In support of this idea, serum from non-human primates immunized with an inactivated ZIKV vaccine, a DNA subunit vaccine, or an adenovirus-based subunit vaccine were shown to equally cross-neutralize ZIKV strains from Brazil, Puerto Rico, the Philippines, Thailand, and Uganda (Abbink et al., 2017).

Current Zika vaccine pipeline

Table 1 summarizes the overall pipeline of ZIKV vaccine progress. Vaccine candidates using distinct technologies are being pursued at different stages of development, some of which have already advanced to clinical phase I/II trials. The vaccine candidates belong to three general categories: (i) inactivated vaccine, (ii) subunit vaccine, and (iii) live-attenuated vaccine. Recently, three phase I clinical studies have been published to demonstrate the safety and immunogenicity of inactivated vaccines (Modjarrad et al., 2017) and DNA subunit vaccines (Gaudinski et al., 2017; Tebas et al., 2017). The following sections focus on reviewing the published preclinical and clinical results of different vaccine platforms, particularly on the recent phase I trial data.

Table 1.

Zika Vaccines in Development

Strategy Antigen form Candidate name Sponsor Clinical trial dose regimen Phase I Phase II Reference
Whole inactivated virus Formalin-Inactivated virus ZPIV WRAIR/BIDMC 2-dose (days 1, 29) NCT02963909
NCT02952833
NCT02937233
NCT03008122
- (Abbink et al., 2016)
PIZV/TAV-426 TAKEDA 2-dose (days 1, 29) NCT03343626 -
Zika virus vaccine (strain MR 766) Bharat Biotech International - - - (Sumathy et al., 2017)
Subunit DNA GLS-5700 GeneOne Life Science, Inc./Inovio Pharmaceuticals 3-dose (weeks 0, 4, 12) NCT02809443
NCT02887482
- (Tebas et al., 2017)
VRC-ZKADNA085-00-VP (VRC5288) VRC/NIAID 2~3-dose (weeks 0, 8; 0, 12; 0, 4, 8; or 0, 4, 20) NCT02840487 - (Dowd et al., 2016b)
VRC-ZKADNA090-00-VP (VRC5283) VRC/NIAID 3-dose (weeks 0, 4, 8) NCT02996461 NCT03110770 (Dowd et al., 2016b)
mRNA mRNA-1325 Moderna Therapeutics - NCT03014089 - (Richner et al., 2017b)
mRNA-LNP University of Pennsylvania - - - (Pardi et al., 2017)
Viral vector MV-Zika Themis Bioscience 1~2-dose (day 0 or 0, 28) NCT02996890 -
RhAd52–prM-Env BIDMC - - - (Abbink et al., 2016)
AdC7-M/E BILS - - - (Xu et al., 2018)
E protein Plant-produced ZIKV E (PzE) Arizona State University - - - (Yang et al., 2018)
E protein from S2 insect cell University of Hawaii - - - (To et al., 2018)
Peptide AGS-v NIH 2-dose (days 0 and 21) NCT03055000 -
VLP Zika virus-like particles TechnoVax, Inc - - - (Boigard et al., 2017)
Live attenuated Attenuated Zika virus ZIKV-3′UTR-Δ10 and ZIKV-3′UTR-Δ20 UTMB/PAHO/IEC - - - (Shan et al., 2017b)
Chimeric flavivirus rDEN4Δ30 chimera NIAID/NIH - - -
JEV SA14-14-2-chimera (ChinZIKV) BIME - - - (Li et al., 2018)
YFV 17D chimera Sanofi - - -

Notes: WRAIR, Walter Reed Army Institute of Research; BIDMC, Beth Israel Deaconess Medical Center; NIAID, National Institutes of Allergy and Infectious Diseases; VRC, Vaccine Research Center; NIH, National Institutes of Health; UTMB, University of Texas Medical Branch; PAHO, Pan American Health Organization; IEC, Evandro Chagas Institute, Brazil; BILS, Beijing Institutes of Life Science, Chinese Academy of Sciences; BIME, Beijing Institute of Microbiology and Epidemiology; symbols “-”, data unavailable.

(I) Inactivated vaccine

Purified formalin-inactivated ZIKV vaccine (ZPIV; derived from Puerto Rico strain PRVABC59) was shown to protect against ZIKV infection in mice and non-human primates (Abbink et al., 2016). In clinical phase I trials, human volunteers were given two doses of 5 μg of ZPIV intramuscularly on days 1 and 29. ZPIV caused only mild to moderate adverse events with local pain (60%) or tenderness (47%) at the injection site, fatigue (43%), headache (39%), and malaise (22%). On day 57, 92% and 77% of the vaccine recipients developed neutralizing antibody titers of ≥1:10 and ≥1:100 (measured by a microneutralization assay), respectively. Adoptive transfer of purified IgG from vaccinated people to mice protected against ZIKV challenge when the neutralizing titers reached >1:100 in recipient mice (Modjarrad et al., 2017).

(II) Subunit vaccines

Subunit vaccines express two virion surface glycoproteins of prM (membrane) and E (envelope) using DNA, mRNA, or viral vectors (e.g., adenovirus, measles virus, vesicular stomatitis virus, and modified vaccinia virus). Alternatively, recombinant E protein or purified virus-like particles (formed by prM and E proteins) could be used directly as subunit vaccines. Many of these subunit vaccine candidates have shown efficacy in protecting mice and/or non-human primates from ZIKV infection (Abbink et al., 2016; Dowd et al., 2016b; Larocca et al., 2016; Pardi et al., 2017; Richner et al., 2017b), among which two-dose preconception immunization of female mice with mRNA vaccine protected maternal-to-fetal ZIKV transmission during pregnancy (Richner et al., 2017a).

Two clinical phase I studies have been recently published on DNA subunit vaccines. Gaudinski et al. tested two DNA vaccine candidates: plasmid VRC5288 (ZIKV and JEV chimeric prM-E to increase the secretion level of empty virus-like particles) and plasmid VRC5283 (wild-type ZIKV prM-E). The prM-E sequence of ZIKV was derived from a French Polynesia strain H/HF/2013. Human volunteers were immunized with three 4-mg doses of DNA vaccine via the intramuscular route. Four different administering regimens were compared: single-dose needle injection, split-dose needle injection, single-dose needle-free injection, or split-dose needle-free injection. Both vaccine candidates were safe and well tolerated with local pain and tenderness (46–80%) at the injection site, headache (22–33%), and malaise (27–38%). Among the tested regimens, vaccination at weeks 0, 4, and 8 via split-dose needle-free injection generated the highest immunogenicity: 100% of the recipients developed neutralizing titers of ≥1:100 with geometric mean titer of 304 (measured by a reporter replicon particle assay). The DNA subunit vaccine also elicited a significant T-cell response (Gaudinski et al., 2017). Based on these results, VRC5283 has advanced to phase II efficacy trial of vaccination at 0, 4, and 8 weeks via needle-free delivery (ClinicalTrials.gov identifier NCT03110770).

In a second DNA vaccine phase I study, Yebas et al. tested a DNA subunit vaccine (GLS-5700) containing a consensus sequence of prM-E from pre-2016 human ZIKV strains. Human volunteers received three doses of either 1 mg or 2 mg of DNA via intradermal injection followed by electroporation at weeks 0, 4, and 12. No serious adverse events were found: Approximately 50% of the vaccinated individuals had injection-site pain, redness, swelling, and itching. By week 14, 62% individuals that received the vaccine developed neutralizing titers between 1:18 to 1:317 (measured by a microneutralization assay on Vero cells). Besides humoral response, the DNA vaccine also elicited T cell activation. Adoptive transfer of vaccinated human serum protected 92% of the A129 mice when challenged with a lethal dose of ZIKV. Surprisingly, serum from five vaccinated people with no neutralizing titer (but with positive antibodies in an ELISA assay) protected the A129 mice in an adoptive transfer experiment (Tebas et al., 2017). These data suggest that mouse protection was independent of the neutralizing titer derived from vaccinated humans. The molecular mechanism that contributed to this independency remains to be determined.

Although the inactivated vaccine and DNA subunit vaccines showed promising immunogenicity in the clinical phase I trials, the longevity of protective immunity remains to be followed in humans. In non-human primates, two immunizations with inactivated vaccine or one immunization with the adenovirus-based subunit vaccine afforded protection against ZIKV challenge at one year post-vaccination, whereas two immunizations with a DNA subunit vaccine resulted in reduced protective efficacy and declined neutralizing antibody titers to sub-protective levels at 1 year (Abbink et al., 2017).

(III) Live-attenuated vaccine

Two approaches have been taken to generate live-attenuated ZIKV vaccines: (i) Engineering attenuating mutations in an authentic ZIKV isolate or (ii) generating a chimeric flavivirus using DENV, JEV, or YFV to express ZIKV prM-E genes. For approach (i), Shan et al. developed two live-attenuated vaccine candidates containing a 10-nucleotide (ZIKV-3′UTR-Δ10) or 20-nucleotide (ZIKV-3′UTR-Δ20) deletion within the 3′UTR of the ZIKV genome from a pre-epidemic Cambodian strain FSS13025 (Shan et al., 2017b). The Cambodian strain FSS13025 was selected because, compared with epidemic American isolates, this strain is attenuated in neurovirulence, immune antagonism, and mosquito infectivity (Xia et al., 2018). After a single-dose vaccination, both candidates showed efficacy in mice and non-human primates, among which the 20-nucleotide deletion candidate elicited neutralizing antibody titers of >1:1,000 (measured by an mCherry ZIKV assay) in two weeks and conferred sterilizing immunity in monkeys. The 3′UTR deletion vaccines could also prevent maternal-to-fetal transmission during pregnancy as well as male reproductive tract infection in mice. Importantly, these vaccine candidates exhibited an excellent safety profile, including >1,000-fold less neurovirulence than the licensed live-attenuated vaccines YFV 17D and JEV SA14-14-2 (Shan et al., 2017a; Shan et al., 2017b). For approach (ii), chimeric DENV-2 and JEV SA14-14-2 harboring ZIKV prM-E genes were reported to protect mice from ZIKV infection after a single-dose vaccination (Li et al., 2018; Xie et al., 2017). The JEV SA14-14-2 chimeric ZIKV vaccine was also shown to protect non-human primates from ZIKV infection and maternal-to-fetal transmission in mice (Li et al., 2018). Taking a similar approach, the National Institute of Health is developing a chimeric ZIKV vaccine using a live-attenuated DENV-4 vaccine backbone (rDEN4Δ30; Table 1).

Development of complementary vaccine platforms

Inactivated and subunit vaccines are not infectious, and usually require multiple doses and periodic boosting. In contrast, live-attenuated vaccines usually have the advantage of single dose, quick immunity, durable protection, and low cost. Since ZIKV is mostly endemic in developing countries, a vaccine with single-dose efficacy is of practical importance, particularly when immunizing populations in remote areas where multiple dosing and periodic boosting could be challenging. The unique features of distinct vaccine platforms justify the development of complementary platforms in parallel to provide options for different populations, including pregnant women, women of child-bearing age, children, infants (with immature immune systems), healthy men, and the elderly (immune-senescence with increased risk of GBS). Specifically, non-infectious subunit and inactivated vaccines are desirable for pregnant women, immunocompromised individuals, and senior people; whereas single-dose live-attenuated vaccines may be preferred when immunizing non-pregnant healthy individuals and children (before reaching child-bearing age) living in or travelling to ZIKV-endemic areas, especially in developing countries.

Immune correlates

Neutralizing antibody titers of >1:10 have been established as correlates of protection for most licensed flavivirus vaccines in humans (Hombach et al., 2005). However, the correlates of protection for the DENV vaccine (Dengvaxia) remain elusive. For ZIKV vaccine development, the concept of neutralizing antibody titers as correlates of protection is generally supported by several lines of available evidence. (i) A strong correlation between neutralizing antibody response and protective efficacy has been consistently observed from various ZIKV vaccine platforms in both mice and non-human primates. In non-human primates, the threshold neutralizing titer required for protection was estimated to be log 2.0 to 2.1 (Abbink et al., 2017). (ii) Adoptive transfer of immune serum from vaccinated humans or monkeys to mice have approximated the minimal protective neutralizing titers to be log ≥2 (Modjarrad et al., 2017) and ≥1.77 (Abbink et al., 2016), respectively. However, as mentioned above, an independency was observed between the neutralizing titers in GLS-5700 DNA-vaccinated people and protection in human-sera-transferred mice (Tebas et al., 2017). It should be noted that the quantitative titers required for ZIKV protection may differ among mice, non-human primates, and humans. The minimal neutralizing titer required for protection in humans will be determined in the ongoing phase II efficacy trials. (iii) In DNA subunit vaccine-immunized mice, depletion of CD4+ and/or CD8+ T lymphocytes did not abrogate protective efficacy (Larocca et al., 2016), indicating that neutralizing antibodies represent the primary mechanism of protection, and that a T lymphocyte response may not be essential for protection. However, CD8+ T lymphocytes were recently shown to play a role in reducing ZIKV burden in mice (Elong Ngono et al., 2017). Further, it is known that a cellular immune response could shape the quality and longevity of the humoral response. Therefore, vaccine optimization should also include T cell responses.

Knowledge gaps and challenges

Despite promising progresses, a number of knowledge gaps remain to be addressed for ZIKV vaccine development. First, ZIKV infection causes GBS at an incident rate of about 1 in 4,000 to 5,000 infected adults (Dos Santos et al., 2016). If viral antigen(s) are responsible for the cause of GBS disease, we need to identify the antigen(s) that elicit antibodies that attack the peripheral nerves. Once such viral antigen(s) has been defined, the vaccine candidates should be re-engineered to exclude the cross-reactive epitope(s). Unfortunately, animal models that can recapitulate human GBS are currently not available to study the molecular details of the disease.

Second, whether ZIKV vaccine-induced antibodies enhance the infection of closely related DENV and vice versa. Since ZIKV, DENV, and other flaviviruses co-circulate in many geographic regions, such cross-reactive antibody-mediated enhancement poses a major challenge for ZIKV vaccine development. So far, a complex interplay has been reported between ZIKV and DENV infections. (i) DENV or WNV antibodies can enhance ZIKV replication in cell culture and mice (Bardina et al., 2017; Dejnirattisai et al., 2016), but such enhancement has not been observed in non-human primates (Pantoja et al., 2017). (ii) On the contrary, prior infection of rhesus macaques with ZIKV led to a significant enhancement of DENV-2 viremia that was accompanied by neutropenia, lympocytosis, hyperglycemia, and higher reticulocyte counts, along with the activation of pro-inflammatory monocyte subsets and release of inflammatory mediators (George et al., 2017). This result indicates that prior ZIKV infection could enhance subsequent DENV-2 infections. (iii) Among the four serotypes of DENV, a pediatric cohort study showed that the risk of severe dengue disease was the highest within a narrow range of preexisting anti-DENV antibody titers, whereas all symptomatic dengue diseases were protected at high antibody titers (Katzelnick et al., 2017), supporting cross-serotype enhancement among sequential DENV infections in humans. (iv) For ZIKV itself, sub-neutralizing levels of antibodies against ZIKV did not enhance the replication of subsequent ZIKV challenge in non-human primates, even though these antibodies exhibited enhancement effects in cell culture (Abbink et al., 2017). Besides the antibody-mediated interplays between ZIKV and DENV described above, cross-reactive T cell responses from prior DENV infection could also influence the outcome of ZIKV infection in mice and humans, suggesting a role for prior DENV immunity in shaping the T cell response to subsequent ZIKV infection (Grifoni et al., 2017; Wen et al., 2017). Furthermore, the interplay between T cell and antibody responses is important as the affinity matured, long-lived antibody response is dependent of CD4+ T cells. Given the above knowledge, it is perceivable that the immune response to ZIKV vaccination and its potential impact on subsequent DENV infection will be different between individuals with and without prior flavivirus immunity. An optimal ZIKV vaccine should be designed to elicit virus type-specific, long-lasting protective antibodies with minimal cross reactivity that may enhance other flavivirus infections. For achieving this goal, studies should be pursued to define the epitopes responsible for virus-specific neutralizing antibodies.

Third, given the decline of ZIKV human cases, efficacy testing in both non-pregnant participants and pregnant women is challenging. Development of a controlled human infection model for ZIKV will accelerate many aspects of ZIKV research and countermeasure development, particularly for down-selecting vaccine candidates for clinical development, for demonstrating proof-of-concept efficacy, and for defining correlates of protection in humans. The controlled human infection model will also be invaluable to evaluate antiviral efficacy for therapeutics development. Such controlled human infection model has been successfully developed for DENV-2 and used for DENV vaccine development (Kirkpatrick et al., 2016). Regulatory approval requires careful ethics review about the risks to human volunteers and social benefits of the controlled human infection of ZIKV.

Fourth, since approximately 80% of ZIKV-infected individuals are asymptomatic and even the symptomatic individuals exhibit mild disease, a well-defined correlate of protection and mechanisms of in utero infection are needed for rapid and accurate assessment of vaccine efficacy. As discussed above, correlates of protection derived from the ongoing phase II clinical trials and possibly from controlled human infection model will facilitate the pre- and post-licensure studies.

Fifth, a reliable diagnostic assay for ZIKV infection is needed to support clinical trials (Balmaseda et al., 2017). Viral infection is an important endpoint to measure vaccine efficacy. During viremic period, viral RNAs can be reliably quantified using the well-established RT-PCR assays. After the viremic period, a serologic assay is required for detection of ZIKV infection. Since antibodies against viral prM and E proteins cross-react among different flaviviruses, it is critical to develop a serologic assay that could differentiate among pre-existing flavivirus antibodies, ZIKV vaccine-induced antibodies, and subsequent ZIKV infection-induced antibodies. Such virus-type-specific serologic assay will greatly facilitate vaccine efficacy trials.

Six, vaccines administered before pregnancy have been shown to prevent ZIKV maternal-to-fetal transmission in mice (Richner et al., 2017a; Shan et al., 2017a). It remains to be demonstrated if vaccines administered after pregnancy (i.e., maternal vaccination) can prevent in utero transmission of ZIKV in animal models. It is conceivable that immune responses may differ between non-pregnant and pregnant women. This leads to a number of inter-related questions: Is the correlate of protection for ZIKV infection in non-pregnant individuals different from that for prevention of in utero transmission in pregnant women? Does maternal vaccination require a dose that is different from the dose of preconception vaccination in order to prevent maternal-to-fetal transmission? Does prevention of CZS require both cellular and humoral immunity in pregnant women? Considerable research is required to address these questions through better defining the biology of ZIKV in humans and through designing and analyzing vaccine clinical trials.

Over the past 2.5 years, the collective efforts from academia, industry, and government have established a promising pipeline of ZIKV vaccine development. Although the number of human cases has dropped significantly since 2017, a vaccine that can prevent CZS remains urgently needed. We need to keep the momentum to bring safe and efficacious ZIKV vaccines to licensure.

Acknowledgments

We thank all members of the Shi lab and the collaborators for their hard work and support. P.-Y.S. lab was supported by University of Texas Medical Branch (UTMB) startup award, University of Texas STARs Award, UTMB Technology Commercialization Program Award, CDC grant for the Western Gulf Center of Excellence for Vector-Borne Diseases, Pan American Health Organization grant SCON2016-01353, the Kleberg Foundation Award, UTMB CTSA UL1TR-001439, and NIH grant AI127744.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abbink P, Larocca RA, De La Barrera RA, Bricault CA, Moseley ET, Boyd M, Kirilova M, Li Z, Ng’ang’a D, Nanayakkara O, et al. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science. 2016;353:1129–1132. doi: 10.1126/science.aah6157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Abbink P, Larocca RA, Visitsunthorn K, Boyd M, De La Barrera RA, Gromowski GD, Kirilova M, Peterson R, Li Z, Nanayakkara O, et al. Durability and correlates of vaccine protection against Zika virus in rhesus monkeys. Science translational medicine. 2017;9 doi: 10.1126/scitranslmed.aao4163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Balmaseda A, Stettler K, Medialdea-Carrera R, Collado D, Jin X, Zambrana JV, Jaconi S, Cameroni E, Saborio S, Rovida F, et al. Antibody-based assay discriminates Zika virus infection from other flaviviruses. Proc Natl Acad Sci U S A. 2017;114:8384–8389. doi: 10.1073/pnas.1704984114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bardina SV, Bunduc P, Tripathi S, Duehr J, Frere JJ, Brown JA, Nachbagauer R, Foster GA, Krysztof D, Tortorella D, et al. Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity. Science. 2017 doi: 10.1126/science.aal4365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Boigard H, Alimova A, Martin GR, Katz A, Gottlieb P, Galarza JM. Zika virus-like particle (VLP) based vaccine. PLoS Negl Trop Dis. 2017;11:e0005608. doi: 10.1371/journal.pntd.0005608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Dejnirattisai W, Supasa P, Wongwiwat W, Rouvinski A, Barba-Spaeth G, Duangchinda T, Sakuntabhai A, Cao-Lormeau VM, Malasit P, Rey FA, et al. Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus. Nat Immunol. 2016;17:1102–1108. doi: 10.1038/ni.3515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Dos Santos T, Rodriguez A, Almiron M, Sanhueza A, Ramon P, de Oliveira WK, Coelho GE, Badaro R, Cortez J, Ospina M, et al. Zika Virus and the Guillain-Barre Syndrome - Case Series from Seven Countries. N Engl J Med. 2016;375:1598–1601. doi: 10.1056/NEJMc1609015. [DOI] [PubMed] [Google Scholar]
  8. Dowd KA, DeMaso CR, Pelc RS, Speer SD, Smith ARY, Goo L, Platt DJ, Mascola JR, Graham BS, Mulligan MJ, et al. Broadly Neutralizing Activity of Zika Virus-Immune Sera Identifies a Single Viral Serotype. Cell Rep. 2016a;16:1485–1491. doi: 10.1016/j.celrep.2016.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Dowd KA, Ko SY, Morabito KM, Yang ES, Pelc RS, DeMaso CR, Castilho LR, Abbink P, Boyd M, Nityanandam R, et al. Rapid development of a DNA vaccine for Zika virus. Science. 2016b;354:237–240. doi: 10.1126/science.aai9137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Elong Ngono A, Vizcarra EA, Tang WW, Sheets N, Joo Y, Kim K, Gorman MJ, Diamond MS, Shresta S. Mapping and Role of the CD8(+) T Cell Response During Primary Zika Virus Infection in Mice. Cell Host Microbe. 2017;21:35–46. doi: 10.1016/j.chom.2016.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Gaudinski MR, Houser KV, Morabito KM, Hu Z, Yamshchikov G, Rothwell RS, Berkowitz N, Mendoza F, Saunders JG, Novik L, et al. Safety, tolerability, and immunogenicity of two Zika virus DNA vaccine candidates in healthy adults: randomised, open-label, phase 1 clinical trials. Lancet. 2017 doi: 10.1016/S0140-6736(17)33105-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. George J, Valiant WG, Mattapallil MJ, Walker M, Huang YS, Vanlandingham DL, Misamore J, Greenhouse J, Weiss DE, Verthelyi D, et al. Prior Exposure to Zika Virus Significantly Enhances Peak Dengue-2 Viremia in Rhesus Macaques. Scientific reports. 2017;7:10498. doi: 10.1038/s41598-017-10901-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Grifoni A, Pham J, Sidney J, O’Rourke PH, Paul S, Peters B, Martini SR, de Silva AD, Ricciardi MJ, Magnani DM, et al. Prior Dengue virus exposure shapes T cell immunity to Zika virus in humans. J Virol. 2017 doi: 10.1128/JVI.01469-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Hoen B, Schaub B, Funk AL, Ardillon V, Boullard M, Cabie A, Callier C, Carles G, Cassadou S, Cesaire R, et al. Pregnancy Outcomes after ZIKV Infection in French Territories in the Americas. N Engl J Med. 2018;378:985–994. doi: 10.1056/NEJMoa1709481. [DOI] [PubMed] [Google Scholar]
  15. Hombach J, Solomon T, Kurane I, Jacobson J, Wood D. Report on a WHO consultation on immunological endpoints for evaluation of new Japanese encephalitis vaccines, WHO, Geneva, 2–3 September, 2004. Vaccine. 2005;23:5205–5211. doi: 10.1016/j.vaccine.2005.07.002. [DOI] [PubMed] [Google Scholar]
  16. Ikejezie J, Shapiro CN, Kim J, Chiu M, Almiron M, Ugarte C, Espinal MA, Aldighieri S. Zika Virus Transmission - Region of the Americas, May 15, 2015-December 15, 2016. MMWR Morb Mortal Wkly Rep. 2017;66:329–334. doi: 10.15585/mmwr.mm6612a4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Katzelnick LC, Gresh L, Halloran ME, Mercado JC, Kuan G, Gordon A, Balmaseda A, Harris E. Antibody-dependent enhancement of severe dengue disease in humans. Science. 2017;358:929–932. doi: 10.1126/science.aan6836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Kirkpatrick BD, Whitehead SS, Pierce KK, Tibery CM, Grier PL, Hynes NA, Larsson CJ, Sabundayo BP, Talaat KR, Janiak A, et al. The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a human challenge model. Science translational medicine. 2016;8:330ra336. doi: 10.1126/scitranslmed.aaf1517. [DOI] [PubMed] [Google Scholar]
  19. Larocca RA, Abbink P, Peron JP, Zanotto PM, Iampietro MJ, Badamchi-Zadeh A, Boyd M, Ng’ang’a D, Kirilova M, Nityanandam R, et al. Vaccine protection against Zika virus from Brazil. Nature. 2016;536:474–478. doi: 10.1038/nature18952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Li XF, Dong HL, Wang HJ, Huang XY, Qiu YF, Ji X, Ye Q, Li C, Liu Y, Deng YQ, et al. Development of a chimeric Zika vaccine using a licensed live-attenuated flavivirus vaccine as backbone. Nat Commun. 2018;9:673. doi: 10.1038/s41467-018-02975-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Modjarrad K, Lin L, George SL, Stephenson KE, Eckels KH, De La Barrera RA, Jarman RG, Sondergaard E, Tennant J, Ansel JL, et al. Preliminary aggregate safety and immunogenicity results from three trials of a purified inactivated Zika virus vaccine candidate: phase 1, randomised, double-blind, placebo-controlled clinical trials. Lancet. 2017 doi: 10.1016/S0140-6736(17)33106-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Pantoja P, Perez-Guzman EX, Rodriguez IV, White LJ, Gonzalez O, Serrano C, Giavedoni L, Hodara V, Cruz L, Arana T, et al. Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus. Nat Commun. 2017;8:15674. doi: 10.1038/ncomms15674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, Dowd KA, Sutherland LL, Scearce RM, Parks R, et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature. 2017 doi: 10.1038/nature21428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Richner J, Jagger B, Shan C, Fontes C, Dowd K, Cao B, Himansu S, Caine E, Nunes B, Medeiros D, et al. Vaccine mediated protection against Zika virus induced congenital disease. Cell. 2017a;170:273–283. doi: 10.1016/j.cell.2017.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Richner JM, Himansu S, Dowd KA, Butler SL, Salazar V, Fox JM, Julander JG, Tang WW, Shresta S, Pierson TC, et al. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell. 2017b doi: 10.1016/j.cell.2017.03.016. [DOI] [PubMed] [Google Scholar]
  26. Shan C, Muruato AE, Jagger BW, Richner J, Nunes BTD, Medeiros DBA, Xie X, Nunes JGC, Morabito KM, Kong WP, et al. A single-dose live-attenuated vaccine prevents Zika virus pregnancy transmission and testis damage. Nat Commun. 2017a;8:676. doi: 10.1038/s41467-017-00737-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Shan C, Muruato AE, Nunes BT, Luo H, Xie X, Medeiros DB, Wakamiya M, Tesh RB, Barrett AD, Wang T, et al. A live-attenuated Zika virus vaccine candidate induces sterilizing immunity in mouse models. Nat Med. 2017b doi: 10.1038/nm.4322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Sumathy K, Kulkarni B, Gondu RK, Ponnuru SK, Bonguram N, Eligeti R, Gadiyaram S, Praturi U, Chougule B, Karunakaran L, et al. Protective efficacy of Zika vaccine in AG129 mouse model. Scientific reports. 2017;7:46375. doi: 10.1038/srep46375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Tebas P, Roberts CC, Muthumani K, Reuschel EL, Kudchodkar SB, Zaidi FI, White S, Khan AS, Racine T, Choi H, et al. Safety and Immunogenicity of an Anti-Zika Virus DNA Vaccine - Preliminary Report. N Engl J Med. 2017 doi: 10.1056/NEJMoa1708120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. To A, Medina LO, Mfuh KO, Lieberman MM, Wong TAS, Namekar M, Nakano E, Lai CY, Kumar M, Nerurkar VR, et al. Recombinant Zika Virus Subunits Are Immunogenic and Efficacious in Mice. mSphere. 2018;3 doi: 10.1128/mSphere.00576-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Wen J, Tang WW, Sheets N, Ellison J, Sette A, Kim K, Shresta S. Identification of Zika virus epitopes reveals immunodominant and protective roles for dengue virus cross-reactive CD8(+) T cells. Nat Microbiol. 2017;2:17036. doi: 10.1038/nmicrobiol.2017.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Xia H, Xie X, Shan C, Shi PY. Potential Mechanisms for Enhanced Zika Epidemic and Disease. ACS Infect Dis. 2018 doi: 10.1021/acsinfecdis.8b00004. [DOI] [PubMed] [Google Scholar]
  33. Xie X, Yang Y, Muruato AE, Zou J, Shan C, Nunes BT, Medeiros DB, Vasconcelos PF, Weaver SC, Rossi SL, et al. Understanding Zika Virus Stability and Developing a Chimeric Vaccine through Functional Analysis. MBio. 2017;8 doi: 10.1128/mBio.02134-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Xu K, Song Y, Dai L, Zhang Y, Lu X, Xie Y, Zhang H, Cheng T, Wang Q, Huang Q, et al. Recombinant Chimpanzee Adenovirus Vaccine AdC7-M/E Protects against Zika Virus Infection and Testis Damage. J Virol. 2018;92 doi: 10.1128/JVI.01722-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Yang M, Sun H, Lai H, Hurtado J, Chen Q. Plant-produced Zika virus envelope protein elicits neutralizing immune responses that correlate with protective immunity against Zika virus in mice. Plant Biotechnol J. 2018;16:572–580. doi: 10.1111/pbi.12796. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES