Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ZIKA VIRUS LIKE PARTICLE (VLP) BASED VACCINE AND MICRONEUTRALIZATION ASSAY
Document Type and Number:
WIPO Patent Application WO/2018/237039
Kind Code:
A1
Abstract:
The present invention includes compositions, methods, vectors, vaccines, cell lines and other constructs for making and used Zika virus Reporter Virus Particles (RVPs) and/or Virus Like Particles (VLPs) that are safe for handling and manufacturing and are able to generate an effective immune response against Zika virus and can be readily scaled up for cost-effective production.

Inventors:
GARG HIMANSHU (US)
JOSHI ANJALI (US)
Application Number:
PCT/US2018/038551
Publication Date:
December 27, 2018
Filing Date:
June 20, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV TEXAS TECH SYSTEM (US)
International Classes:
C12N15/86; A61K39/12
Domestic Patent References:
WO2017015463A22017-01-26
WO2016210127A12016-12-29
WO2018152526A12018-08-23
Other References:
BOIGARD, H. ET AL.: "Zika virus-like particle (VLP) based vaccine", PLOS NEGLECTED TROPICAL DISEASES, vol. 11, no. 5, e0005608, 8 May 2017 (2017-05-08), pages 1/20 - 20/20, XP055657701, DOI: 10.1371/journal.pntd.0005608
WHITBECK, C. ET AL.: "Analyzing the immune response to Zika virus: A reporter virus particle (RVP) system for serum and antibody neutralization assays, and a comprehensive Ala-scan mutation library of ZIKV Prm/E to epitope map anti-ZIKV antibodies", THE AMERICAN JOURNAL OF TROPICAL MEDICINE AND HYGIENE, vol. 95, no. 5 suppl., abstract 746, November 2016 (2016-11-01), pages 235
DOWD, K.A. ET AL.: "Evaluating strain specificity of the ZIKA virus neutralizing antibody response", THE AMERICAN JOURNAL OF TROPICAL MEDICINE AND HYGIENE, vol. 95, no. 5 suppl., abstract 1385, November 2016 (2016-11-01), pages 435
GARG, H. ET AL.: "Development of virus-like-particle vaccine and reporter assay for ZIKA virus", JOURNAL OF VIROLOGY, vol. 91, no. 20, e00834-17, 15 October 2017 (2017-10-15), pages 1 - 16, XP055646955, DOI: 10.1128/JVI.00834-17
Attorney, Agent or Firm:
CHALKER, Daniel, J. et al. (US)
Download PDF:
Claims:
What is claimed is:

1. A nucleic acid vector comprising: a recombinant nucleic comprising at least one of: a Zika virus C-prM-E gene, or a recombinant nucleic comprising a portion of a prM-E gene region of Zika virus spanning amino acids 105-795, operably linked to a promoter.

2. The vector of claim 1, wherein the nucleic acid vector is a flavivirus vector.

3. The vector of claim 1, wherein the nucleic acid vector is a plasmid.

4. The vector of claim 1, wherein the C-prM-E, the prM-E gene, or both, are codon optimized. 5. The vector of claim 1, wherein the nucleic acid vector is pcDNA3.1, and the C-prM-E gene, the prM-E gene, or both, are codon optimized for expression in human cells.

6. The vector of claim 5, wherein the nucleic acid vector further comprises an NS2B3 protease gene.

7. The vector of claim 1, wherein the vector is a lentiviral vector pLenti6/5-D-Topo. 8. The vector of claim 7, wherein the vector further comprises an NS2B3 protease gene.

9. The vector of claim 1, further comprising a selectable marker.

10. A method of making a Zika virus Reporter Virus Particles (RVP) comprising: transfecting cells stably expressing a Zika C-prM-E gene; and transfecting the cells stably with a sub-genomic replicon derived from lineage II strain of WNV or ZIKV that also expresses a reporter gene; incubating the cells under conditions in which the proteins are expressed for a period sufficient to form RVPs; and harvesting the RVPs.

11. The method of claim 10, wherein the cells are 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines.

12. The method of claim 10, wherein the reporter gene is selected from at least one of green fluorescent protein; yellow fluorescent protein; blue fluorescent protein; Cerulean fluorescent protein; Cyan fluorescent protein; red fluorescent protein from Zooanthus sp.; red fluorescent protein from Entremacaea quadricolor (RFP), or flavin mononucleotide (FMN)-binding fluorescent proteins (FbFPs).

13. The method of claim 10, wherein the C-prM-E is codon optimized.

14. The method of claim 10, further comprising transfecting an NS2B3 protease gene into the cell.

15. The method of claim 1, wherein the vector further a selectable marker and selecting stable expression with a selectable marker.

16. A vaccine compri sing : a particle that comprises a codon optimized PrM-E protein of Zika virus in a

pharmaceutically acceptable carrier or excipient.

17. The vaccine of claim 10, wherein the particles are manufactured in 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines.

18. The vaccine of claim 10, wherein the particle is a Zika virus like particle.

19. The vaccine of claim 10, wherein the vector further comprises a selectable marker. 20. The vaccine of claim 10, wherein the particle is a Zika virus reporter virus particle capable of a single round infection in cells in a manner identical to native Zika virus.

21. The vaccine of claim 10, wherein the Zika virus reporter virus particle is non-infectious.

22. A vaccine comprising: a Zika virus like particle comprising a matured C-prM-E expressed from a vector that comprises a codon optimized C-prM-E gene and an NS2B3 protease gene.

23. A cell line comprising: a codon optimized C-prM-E gene of Zika virus that is stably expressed.

24. The cell line of claim 23, wherein the cell line comprises 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines. 25. The cell line of claim 23, wherein the cell line is transduced with Lentiviral particles made in cells expressing lentiviral prME and php-dl-NA, and VSVG Env.

26. The cell line of claim 23, wherein the cell line is stably transduced and selected using a selectable marker.

27. The cell line of claim 23, wherein stable expression is by blasticidin selection.

28. The cell line of claim 23, wherein the cell line is further transfected with an NS2B3 protease gene.

29. A cell line comprising: a codon optimized prM-E gene region of Zika virus spanning amino acids 105-795 that is stably expressed. 30. The cell line of claim 29, wherein the cell line is 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines.

31. A method of detecting Zika virus microneutralization using ZIKV reporter virus particles (RVP) or ZIKV Virus Like Particles (VLPs)comprising:

(a) incubating serial dilutions of sera or antibodies with a pre-determined amount of ZIKV RVPs or VLPs comprising a detectable marker at room temperature to make an antibody - RVP mix;

(b) adding the antibody-RVP or VLP mix to cells under conditions in which the detectable marker is expressed;

(c) measuring infection after a pre-determined amount of time by counting the number of detectable marker positive cells; and

(d) comparing the level of the detectable marker in a first set of cells where no sera or neutralizing antibody was used to a second set of cells where the sera or neutralizing antibodies were used, wherein a decrease in the number of marker positive cells in the second set of cells when compared to the first set of cells is indicative of microneutralization that correlates to a level of neutralizing antibodies in the biological sample of the patient.

32. The method of claim 31, wherein the cells are 293T, Raji, or Vero cells.

33. The method of claim 31, further comprising obtaining another biological sample from the patient after a pre-determined time and comparing a titer of neutralizing antibodies between an earlier and a later biological sample to determine antibody titers. 34. The method of claim 31, wherein the method is adapted for use in 4, 6, 8, 12, 24, 48, 96, 384, 1538, 6114, or 9,600 well plates.

35. The method of claim 31, wherein the ZIKV VLPs comprise a PrM-E protein expressed by a codon-optimized construct.

36. The method of claim 31, wherein the detectable marker in the ZIKV RVPs comprises a fluorescent protein expressed in conjunction with a WNV or ZIKV sub-genomic replicon.

37. The method of claim 31, wherein the ZIKV virus like particles are adapted for use as an antigen in a detection assay.

38. A method of making stably transduced cell lines comprising: transfecting cells with lentiviral vector expressing ZIKV prME, CprME, or both prME and CprME, and php-dl-NA and VSVG Env to produce Lentiviral particles; transducing cells with the Lentiviral particles; and selecting stable transduced cells with a selection agent to create the stably transduced cell lines.

39. The method of claim 38, wherein the cell line is prME-F4. 40. The method of claim 38, wherein the cell line is CprME (F6).

41. The method of claim 38, wherein the selection agent is Blasticidin.

42. The method of claim 38, wherein the transfected cells are selected from 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines.

43. The method of claim 38, wherein the ZIKV prME, CprME, or both prME and CprME are codon optimized.

44. A reporter Zika virus Reporter Virus Particles (RVP) made by expressing a matured, codon optimized ZIKV CprME made in a cell line that expressed an NS2B3 protease.

45. A method of making a Zika virus Virus Like Particles (VLPs) comprising: transfecting cells stably expressing a Zika C-prM-E gene; and transfecting the cells stably with a sub-genomic replicon derived from lineage II strain of

WNV or ZIKV; incubating the cells under conditions in which the proteins are expressed for a period sufficient to form VLPs; and harvesting the VLPs. 46. The method of claim 38, wherein the Zika C-prM-E gene is codon optimized.

47. The method of claim 38, wherein the VLP is adapted to be an antigen in an assay.

48. The method of claim 38, wherein the VLP is made in a cell line that expressed an NS2B3 protease.

Description:
ZIKA VIRUS LIKE PARTICLE (VLP) BASED VACCINE AND

MICRONEUTRALIZATION ASSAY

TECHNICAL FIELD OF THE INVENTION

The present invention relates in general to the field of a ZIKA virus like particle (VLP) based vaccine and microneutralization assay, cell lines, and vectors related to the same.

INCORPORATION-BY-REFERENCE OF MATERIALS FILED ON COMPACT DISC

The present application includes a Sequence Listing, which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on June 20, 2018, is named TECH2104WO_SeqList and is 53 kilobytes in size.

BACKGROUND OF THE INVENTION

Without limiting the scope of the invention, its background is described in connection with viruses belonging to the family Flaviviridae.

Since the identification of Zika virus (ZIKV) in 1947 from a Rhesus Monkey in Uganda until 2010, the virus has predominantly circulated between Aedes mosquitoes and non-human primates. Periodic episodes have been identified in the human population, which were characterized by mild self-limiting febrile disease associated with rash, headache, myalgia and conjunctivitis. However, the recent spread of ZIKV infections in the Western continents has caused much concern due to severe clinical outcome in unborn fetuses including cerebral calcifications, microcephaly and other severe congenital malformations. In adults, neurological manifestations are characterized by an autoimmune condition with symptoms of neuropathy and paralysis, also known as the Guillain-Barre syndrome. While Aedes species of mosquitos are the most common source of transmission, the virus has also been shown to transmit sexually both from women to men and men to women and is capable of persisting in semen and vaginal secretions for up to 6 months after infection.

ZIKV is an enveloped RNA virus belonging to the family Flaviviridae. The 11Kb positive sense RNA genome is translated in the cytoplasm to generate three structural and seven nonstructural proteins. The structural proteins C (Capsid), prM/M (Pre-membrane, membrane), and E (Envelope) aid in virus assembly that predominantly occurs in the lumen of the endoplasmic reticulum (ER). Virus maturation occurs during virus egress via the secretory pathway when the acidic environment in the Golgi cleaves the prM followed by release of pr peptide. The E protein is the major target for neutralizing antibodies and monoclonal antibodies against all 3 E protein domain (DI, DII and Dili) target epitopes have been found. The recent outbreaks of ZIKV infection have sparked efforts in the scientific community towards the development of a safe and effective vaccine. These efforts towards a safe and efficacious vaccine encompass the use of established approaches like purified inactivated virus to more advanced approaches like DNA (Pr-M-E), subunit (E) based vaccines, recombinant adenoviral platforms along with recent development of RNA nanoparticle technology, or modified mRNA (prM-E) as vaccine candidates. The studies have demonstrated a neutralizing antibody response capable of protecting against ZIKV infection both in mice and non-human primates leading several clinical trials currently underway (NCT02963909, NCT02840487, NCT02887482, NCT02809443, NCT02952833). One of the advantages of the development of a ZIKV vaccine is that even though the virus exists as two distinct lineages (the African and Asian/ American), the immune response generated against the virus is broadly protective thus obviating the need to incorporate different serotypes in the vaccine.

What is needed is the development of neutralizing antibodies against the infection and availability of a rapid accurate diagnostic assay to quantitate the elicited immune response. A widely used assay for detection of neutralizing antibodies against Flaviviruses is the plaque reduction neutralization test (PRNT) assay, which involves the use of live virus handled under BSL-2 conditions. An ideal assay would be one that could be adapted to a high throughput format with a convenient read out and eliminate the use of live virus making it readily available to laboratories worldwide. With regards to an effective vaccine, the priorities include: safety, efficacy, ease of handling, and economy of production for worldwide dissemination.

Thus, despite many efforts, there is currently no approved vaccine for Zika. A recent report showed a PrME plasmid DNA vaccine to be effective in mouse studies. However, DNA based vaccines includes several risks, e.g., insertional mutagenesis.

SUMMARY OF THE INVENTION

In one embodiment, the present invention includes a nucleic acid vector comprising: a recombinant nucleic comprising at least one of: a Zika virus C-prM-E gene, or a recombinant nucleic comprising a portion of a prM-E gene region of Zika virus spanning amino acids 105- 795, operably linked to a promoter. In one aspect, the nucleic acid vector is a flavivirus vector. In another aspect, the nucleic acid vector is a plasmid. In another aspect, the C-prM-E, the prM- E gene, or both are codon optimized. In another aspect, the nucleic acid vector is pcDNA3.1, and the C-prM-E gene, the prM-E gene, or both, are codon optimized for expression in human cells. In another aspect, the nucleic acid vector further comprises an NS2B3 protease gene. In another aspect, the vector is a lentiviral vector pLenti6/5-D-Topo. In another aspect, the vector further comprises an NS2B3 protease gene. In another aspect, the nucleic acid vector further comprising a selectable marker.

In another embodiment, the present invention includes a method of making a Zika virus Reporter Virus Particles (RVP) comprising: transfecting cells stably expressing a Zika C-prM-E gene; and transfecting the cells stably with a sub-genomic replicon derived from lineage II strain of WNV that also expresses a reporter gene; incubating the cells under conditions in which the proteins are expressed for a period sufficient to form RVPs; and harvesting the RVPs. In one aspect, cells are 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines. In another aspect, the reporter gene is selected from at least one of green fluorescent protein; yellow fluorescent protein; blue fluorescent protein; Cerulean fluorescent protein; Cyan fluorescent protein; red fluorescent protein from Zooanthus sp.; red fluorescent protein from Entremacaea quadricolor (RFP), or flavin mononucleotide (FMN)-binding fluorescent proteins (FbFPs). In another aspect, the C-prM-E is codon optimized. In another aspect, the method further comprises transfecting an NS2B3 protease gene into the cell. In another aspect, the vector further a selectable marker and selecting stable expression with a selectable marker.

In another embodiment, the present invention includes a vaccine comprising: a particle that comprises a codon optimized PrM-E protein of Zika virus in a pharmaceutically acceptable carrier or excipient. In one aspect, the particles are manufactured in 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines. In another aspect, the particle is a Zika virus like particle. In another aspect, the vector further comprises a selectable marker. In another aspect, the particle is a Zika virus reporter virus particle capable of a single round infection in cells in a manner identical to native Zika virus. In another aspect, the Zika virus reporter virus particle is non-infectious.

In yet another embodiment, the present invention includes a vaccine comprising: a Zika virus like particle comprising a matured C-prM-E expressed from a vector that comprises a codon optimized C-prM-E gene and an NS2B3 protease gene.

In another embodiment, the present invention includes a cell line comprising: a codon optimized C-prM-E gene of Zika virus that is stably expressed. In one aspect, the cell line comprises 293T, Raji, or Vero cells. In another aspect, the cell line is transduced with Lentiviral particles made in cells expressing lentiviral prME and php-dl-NA, and VSVG Env. In another aspect, the cell line is stably transduced selected using a selectable marker. In another aspect, the stable expression is by blasticidin selection. In another aspect, the cell line is further transfected with an NS2B3 protease gene. In another embodiment, the present invention includes a cell line comprising: a codon optimized prM-E gene region of Zika virus spanning amino acids 105-795 that is stably expressed. In one aspect, the cell line is 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines. In another embodiment, the present invention includes a method of detecting Zika virus microneutralization using reporter virus particles (RVP) or Virus Like Particles (VLP) comprising: (a) incubating serial dilutions of sera or antibodies with a pre-determined amount of ZIKV RVPs or VLPs comprising a detectable marker at room temperature to make an antibody- RVP mix; (b) adding the antibody-RVP or VLP mix to cells under conditions in which the detectable marker is expressed; (c) measuring infection after a pre-determined amount of time by counting the number of detectable marker positive cells; and (d) comparing the level of the detectable marker in a first set of cells where no sera or neutralizing antibody was used to a second set of cells where the sera or neutralizing antibodies were used, wherein a decrease in the number of marker positive cells in the second set of cells when compared to the first set of cells is indicative of microneutralization that correlates to a level of neutralizing antibodies in the biological sample of the patient. In one aspect, the cells are 293T, Raji, or Vero cells. In another aspect, the method further comprises obtaining another biological sample from the patient after a pre-determined time and comparing a titer of neutralizing antibodies between an earlier and a later biological sample to determine antibody titers. In another aspect, the method is adapted for use in 4, 6, 8, 12, 24, 48, 96, 384, 1538, 6114, or 9,600 well plates. In another aspect, the ZIKV VLPs comprise a PrM-E protein expressed by a codon-optimized construct. In another aspect, the detectable marker in the ZIKV RVPs comprises a fluorescent protein expressed in conjunction with a WNV sub-genomic replicon. In another aspect, the ZIKV VLP is adapted to be an antigen in a diagnostic assay. In another embodiment, the present invention includes a method of making stably transduced cell lines comprising: transfecting cells with lentiviral vector expressing ZIKV prME, CprME, or both prME and CprME, and php-dl-NA and VSVG Env to produce Lentiviral particles; transducing cells with the Lentiviral particles; and selecting stable transduced cells with a selection agent to create the stably transduced cell lines. In one aspect, the cell line is prME-F4. In another aspect, the cell line is CprME (F6). In another aspect, the selection agent is Blasticidin. In another aspect, the transfected cells are selected from 293T cells, HeLa cells, MDCK cells, Vero cells, or vaccine-certified cell lines. In another aspect, the ZIKV prME, CprME, or both prME and CprME are codon optimized. In another embodiment, the present invention includes a reporter Zika virus Reporter Virus Particles (RVP) made by expressing a matured, codon optimized ZIKV CprME made in a cell line that expressed an NS2B3 protease.

BRIEF DESCRIPTION OF THE DRAWINGS

For a more complete understanding of the features and advantages of the present invention, reference is now made to the detailed description of the invention along with the accompanying figures and in which:

FIGS. 1A to 1C show the expression of ZIKV C-prM-E using codon optimized synthetic construct. (FIG. 1A) A codon optimized ZIKV C-prM-E gene was synthesized via gene synthesis technology using the sequence available from the current outbreak in Americas and cloned into the pcDNA3.1 vector. (FIG. IB) 293T cells were transfected with the ZIKV C-prM- E or WNV C-prM-E constructs. Cells were fixed, stained with antibodies MAB10216 and MAB8150 followed by analysis of E protein expression by fluorescence microscopy. (FIG. 1C) 293T cells were transfected as described above. Forty eight hours post transfection, cells were radiolabeled with [ 5 S]Met/Cys. Cell lysates were immunoprecipitated with ProteinA beads coated with MAB 10216 or anti-WNV serum, resolved by SDS-PAGE followed by Phosphorlmager analysis.

FIGS. 2A to 2H show RVP based microneutralization assay for ZIKV using a 96 well plate format and GFP readout. (FIG. 2A) Strategy for generation of ZIKV or WNV reporter virus particles. 293T cells were co-transfected with ZIKV/WNV C-prM-E along with the WNV subgenomic replicon construct Rep/GFP. Culture supernatants were harvested 48h post transfection, used to infect 293T/Vero cells and GFP expression was analyzed as a measure of virus infection. (FIG. 2B) 293T and Vero cells were infected with ZIKV or WNV RVPs and infection analyzed by fluorescence microscopy as GFP positive cells. (FIG. 2C) Vero cells were infected with serial dilutions of ZIKV reporter virus like particles in 96-well plates. Cells were fixed 72h post infection and images of whole wells captured via fluorescence microscopy. Representative fluorescent images of whole wells infected with 6 serial dilutions (dilution 1-6) of ZIKV RVPs is depicted. The top panel shows raw images acquired by fluorescent microscopy. The bottom panel shows the same images analyzed using the automated NIS elements software that marks the GFP positive cells using the cell count function. Numbers below each well represent the number of GFP positive cells in that well. Vero cells were infected with serial dilutions of (FIG. 2D) ZIKV or (FIG. 2E) WNV reporter virus particles in 96-well plates. The experiment was conducted with 6 technical wells infected for each dilution. Number of GFP positive cells was determined 72h post infection as described in part C above. Data shows small variations between the 6 wells for each RVP dilution. (FIG. 2F) Quantitation of GFP positive cells infected with Zika RVPs using the automated software from Nikon versus manual counting. Representative data shows high degree of correlation between the two methods. (FIG. 2G) ZIKV-117 antibody or (FIG. 2H) the indicated antibodies/sera were serially diluted in DMEM and incubated with a predetermined amount of ZIKV RVPs for lh at room temperature. Subsequently, the virus: sera mix was added to Vero cells. The cells were incubated for 72h following which images were acquired and the number of GFP positive cells quantitated as described above. The assay was conducted in technical triplicates for the ZIKV-117 antibody and ZIKV sera and in duplicates for others. One representative of 3 independent experiment is shown.

FIGS. 3A to 3G shows the establishment of a stable cell line expressing ZIKV C-prM-E. (FIG. 3A) The ZIKV C-prM-E was PCR amplified with specific primers using the codon optimized construct as template and cloned into the lentiviral vector pLenti6/5-D-Topo. 293T cells were then transfected with the pLenti-C-prM-E construct along with the helper plasmid and VSV-G envelope and ZIKV-C-prM-E lentiviral particles harvested 48h post transfection. 293T cells were then transduced with the above lentiviral particles and cells selected by culturing in the presence of Blasticidin. Bulk selected cells were confirmed for E protein expression via immunofluorescence. Subsequently, cells were plated in 96 well plates using limiting dilution and clones arising from single viable cells selected. (FIG. 3B) 293T cells transduced with ZIKV C-prM-E lentiviral particles and bulk selected with Blasticidin were stained with antibody MAB10216 and analyzed by fluorescence microscopy. (FIG. 3C) The percentage of C-prM-E positive cells was determined by flow cytometry. The cells only peak represents 293T cells not stained with the antibody and the 293T peak represents 293T cells stained with MAB 10216. (FIG. 3D) 293T cells expressing the pLenti-ZIKV-C-prM-E generated above and bulk selected with blasiticidin were analyzed for E protein expression by western blotting. Non transfected 293T cells (293T) or cells transiently transfected with the ZIKV C-prM-E expression construct (transient) were used as negative and positive controls respectively. (FIG. 3E) Single cells clones of 293T cells expressing the pLenti-ZIKV-C-prM-E were stained using antibody MAB10216 and analyzed by fluorescence microscopy or (FIG. 3F) Flow cytometry. The Mean Fluorescent Intensity (MFI) of E protein expression for each clone is indicated alongside. (FIG. 3G) The pLenti-ZIKV-C-prM-E 293T cells are ideal for producing high titer RVPs. 293T cells were transiently transfected with ZIKV C-prM-E along with the WNV Rep/GFP construct to generate reporter virus particles. Alongside, the pLenti-ZIKV-C-prM-E cell line was transfected with the WNV Rep/GFP construct. RVPs were harvested and serial dilutions used to infect Vero cells. Number of GFP positive cells in each well was quantitated by fluorescent microscopy. Error bars are mean +/- SD. * represents significant difference (p<0.01) in number of GFP+ cells in 293 T versus C-prM-E-F6 RVPs using the unpaired t-test. One representative of 4 independent experiments is shown.

FIGS. 4A to 4H shows the generation of prM-E cell line for VLP production. (FIG. 4A) The ZIKV prM-E was PCR amplified with specific primers using the codon optimized C-prM-E construct as template and cloned into the (FIG. 4B) pCDNA3.1 expression vector. (FIG. 4C) E protein expression was determined by fluorescence microscopy after staining with antibody MAB10216. (FIG. 4D) Culture supernatants were harvested from ZIKV prM-E expressing cells and ultracentrifuged. Cell and virus pellet was lysed and E protein expression determined by western blotting. (FIG. 4E) The ZIKV prM-E was PCR amplified and cloned into the lentiviral vector pLenti6/5-D-Topo. 293T cells were then transfected with the pLenti-prM-E construct along with the helper plasmid and VSV-G envelope and ZIKV-prM-E lentiviral particles harvested 48h post transfection. 293T cells were then transduced with the above lentiviral particles and cells either bulk selected or as single cell clones by culturing in the presence of Blasticidin. (FIG. 4F) Selected cells were confirmed for E protein expression via immunofluorescence and (FIG. 4G) Fluorescence microscopy after staining with antibody MAB10216. The mean Fluorescent Intensity (MFI) of E protein expression for each prM-E clone is indicated alongside. (FIG. 4H) The indicated pLenti-ZIKV-prM-E cell clones were seeded in equal cell numbers and culture supernatants harvested and ultracentrifuged. VLP pellets were lysed, resolved by SDS-PAGE and E protein expression determined by western blotting.

FIGS. 5A to 5E shows the expression of ZIKV prM-E alone releases VLPs in the supernatants while the C-prM-E requires the protease NS2B-3 for efficient VLP release. (FIG. 5A) 293T cells were transfected with the pCDNA3.1 vector expressing the ZIKV prM-E or C-prM-E along with the indicated expression vectors. Cells were radiolabeled with [ 5 S]Met/Cys and culture supernatants harvested and ultracentrifuged. Cell and virion samples were lysed and immunoprecipitated with MAB10216 coated ProteinA beads, resolved by SDS-PAGE followed by Phosphorlmager analysis. (FIG. 5B) Culture supernatants were harvested from cells expressing the ZIKV C-prM-E or prM-E as indicated in the methods. 25-30 ml of supernatant was transferred into ultracentrifuge tubes and carefully underlayed with 5ml of 25% glycerol in TNE buffer. VLPs were pelleted by centrifugation at 110,500 x g for 3h at 4°C. Thereafter, the supernatant was carefully removed and the VLP pellet resuspended in TNE buffer. An aliquot of the concentrated VLPs were lysed using 10X RIPA buffer and E protein in the preps was detected by western blotting. (FIG. 5C) VLPs were concentrated as above and images were acquired after negative staining using the Transmission Electron Microscope JEOL1010 with a Hamamatsu digital camera. Scale Bar~30nm. Immunization studies in mice. (FIG. 5D) Balb/c mice were divided into groups of six mice each. Mice received primary immunization on day 0 followed by 2 boosters at day 14 and 28 and were finally sacrificed at day 63 post primary immunization. (FIG. 5E) Mice were divided into 5 groups and received immunizations with either C-prM-E/prM-E DNA or VLPs. For DNA immunization, a total of 5C^g of DNA in a volume of ΙΟΟμΙ PBS was injected intramuscularly. For VLPs, the first immunization consisted of VLP prep mixed with TiterMax Gold adjuvant in total volume of ΙΟΟμΙ injected intramuscularly. For subsequent boosters, mice received VLPs alone without adjuvant. Control mice were sham injected with PBS.

FIGS. 6A to 6F shows the Anti-ZIKV immune response in mice immunized with prM-E/C-prM- E DNA and VLPs. (FIG. 6A) Serum samples collected from different groups of immunized mice were used in the reporter RVP based microneutralization assay. Sera sample from each mouse was serially diluted in DMEM and incubated with a predetermined amount of ZIKV RVPs for lh at room temperature. All samples were assayed in technical duplicates. Subsequently, the virus: sera mix was added to Vero cells in 96-well plates. The cells were incubated for 72h following which the plates were fixed and images acquired as described in FIGS. 2A-2H. Curves were fit using the GraphPad Prism software and neutralizing antibody (FIG. 6B) EC50 and (FIG. 6C) EC90 values calculated. Statistical analysis was performed using the unpaired t test. Significant difference in EC50 (p=0.0083) and EC90 values (p=0.0006) between the prM-E and C-prM-E VLP immunized mice. The dotted line denotes the limit of detection for the RVP assay (defined as the highest concentration of sera used in the neutralization experiments). Samples with titers <20 are reported at half the limit of confidence (1 : 10). Neutralization data form one of two independent repeats is shown. (FIG. 6D) Neutralization of a clinical ZIKV isolate PRVABC59 with immune sera samples from mice. Pooled sera samples from each immunized group were serially diluted in serum free media as in part (A) and incubated with a predetermined amount of ZIKV for 2h at 37°C. All samples were assayed in technical duplicates. Subsequently, the virus: sera mix was added to Vero cells in 96-well plates. The cells were incubated for 48h following which the plates were stained using MAB10216. Images were acquired as described in Figure 2, antibody positive cells quantitated and curves were fit using the GraphPad Prism software. (FIG. 6E) Protein A beads were coated with 3μ1 or 0.6μ1 of pooled sera samples from each group of immunized mice. The antibody coated beads were then incubated with radiolabeled cell lysates derived from C-prM-E expressing cells. Cell lysates were resolved on an SDS-PAGE gel followed by phosphorlmager analysis. The Photo Stimulated Luminescence (PSL) values for each band are depicted in the graphs underneath. (FIG. 6F) Pooled sera samples from each group of immunized mice were used in technical duplicates to determine inhibition of WNV RVPs as in part (A) above.

DETAILED DESCRIPTION OF THE INVENTION

While the making and using of various embodiments of the present invention are discussed in detail below, it should be appreciated that the present invention provides many applicable inventive concepts that can be embodied in a wide variety of specific contexts. The specific embodiments discussed herein are merely illustrative of specific ways to make and use the invention and do not delimit the scope of the invention.

To facilitate the understanding of this invention, a number of terms are defined below. Terms defined herein have meanings as commonly understood by a person of ordinary skill in the areas relevant to the present invention. Terms such as "a", "an" and "the" are not intended to refer to only a singular entity, but include the general class of which a specific example may be used for illustration. The terminology herein is used to describe specific embodiments of the invention, but their usage does not limit the invention, except as outlined in the claims.

The present invention meets the criteria for an effective vaccine, namely, safety, efficacy, ease of handling, and economy of production for worldwide dissemination. The VLPs-based vaccines of the present invention are safe for manufacturing and handling and generate an effective immune response and can be readily scaled up for cost-effective production. The ZIKV VLPs taught herein can be readily produced in cells expressing the prM-E proteins and the particles although non-infectious resemble the live virus in morphology. Further, addition of the Capsid has been shown to promote virion stability and is effective in inducing a cell- mediated immune response against Flaviviruses. VLP -based vaccines have been successful against viral diseases like Hepatitis B (GlaxoSmithKline's ENGERIX® and Merck and Co., Inc.'s RECOMBIVAX HB®) and human papilloma virus (Merck and Co., Inc.'s GARDISIL®) with others in the pipeline for diseases like influenza, Parvovirus, Norwalk virus etc.

As used herein, the terms "antigen," "antigenic," and "antigenically active," refer to any substance that can be recognized by a specific humoral and/or cell-mediated immune response. As used herein, the terms "immunogen," "immunogenic" and "immunologically active" refer to any substance that is capable of inducing a specific humoral and/or cell-mediated immune response. An antigen or immunogen generally contains at least one epitope. Antigens and immunogens include but are not limited to molecules, including small molecules, peptides, polysaccharides, nucleic acids, and/or lipid, that trigger an immune response. Complexes of peptides with lipids, polysaccharides, or with nucleic acid sequences are also contemplated, including (without limitation) glycopeptide, lipopeptide, glycolipid, etc. These complexes are particularly useful immunogens where smaller molecules with few epitopes do not stimulate a satisfactory immune response by themselves. In certain aspects, the virus like particles of the present invention can be used as an antigen in a diagnostic assay.

As used herein, the term "adjuvant" refers to the addition of an agent that enhances an immune response in an animal. For example, the antigen may be mixed or emulsified in saline, for example, Freund's complete adjuvant ("FCA"), Freund's incomplete adjuvant, alum, CpG, and the mixture is injected parenterally, intraperitoneally, subcutaneously, intramuscularly, orally, etc. The animal is generally boosted 2-6 weeks later with one or more injections of the antigen with or without an adjuvant. Antibodies may also be generated by in vitro immunization, using methods known in the art. When isolated from an animal, polyclonal antisera is then obtained from the immunized animal.

As used herein, the term "cell culture" refers to any in vitro culture of cells, including, e.g., continuous cell lines (immortal), primary cell cultures, transformed cell lines, finite cell lines (e.g., non-transformed cells), and any other cell population capable of being maintained in vitro. Cells may include bacterial, fungal, mammalian, insect, etc. As used herein, the term "codon optimized" refers to a nucleic acid sequence or polynucleotides that is expressed into a polypeptide wherein the codon usage is optimized for a specific host. Codon optimized sequences are engineered to increase the expression of the polypeptide in a given species. To provide optimized polynucleotides coding for the viral and other proteins described herein, the DNA sequence of the gene is modified to 1) include codons preferred by highly expressed genes in a particular species; 2) include an A+T or G+C content in nucleotide base composition to that substantially found in the target species; 3) form an initiation sequence of the target species; or 4) eliminate sequences that cause destabilization, inappropriate polyadenylation, degradation and termination of RNA, or that form secondary structure hairpins or RNA splice sites. Increased expression of the proteins described herein is achieved by using a distribution frequency of codon usage in eukaryotes and prokaryotes, or in a particular species. The term "frequency of codon usage" refers to the usage of nucleotide codons found in a specific host cell to express a given amino acid. There are 20 natural amino acids, most of which are specified by more than one codon. Therefore, all degenerate nucleotide sequences can be codon optimized so as long as the amino acid sequence of the polypeptide encoded by the nucleotide sequence is functionally unchanged.

As used herein, the term "gene" refers to a nucleic acid (e.g., DNA or RNA) sequence that comprises coding sequences necessary for the production of a polypeptide, polypeptide precursors, or RNA (e.g., rRNA, tRNA, RNAi). The polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence so long as the desired activity or functional properties (e.g., immunogenicity) of the full-length or fragment are retained. The term can also encompasses the coding region of a structural gene and the sequences located adjacent to the coding region on both the 5' and 3' ends on either end such that the gene corresponds to the length of the full-length mRNA, e.g., when engineered into a nucleic acid vector. Sequences located 5' of the coding region and present on the mRNA are referred to as 5' non-translated sequences. Sequences located 3' or downstream of the coding region and present on the mRNA are referred to as 3' non-translated sequences. The term "gene" encompasses both cDNA and genomic forms of a gene, e.g., a viral genome. A genomic form or clone of a gene contains the coding region interrupted with non-coding sequences termed "introns" or "intervening regions" or "intervening sequences." Introns are segments of a gene that are transcribed into heterogenous nuclear RNA (hnRNA); introns may contain regulatory elements such as enhancers. Introns are removed or "spliced out" from the nuclear or primary transcript; introns therefore are absent in the messenger RNA (mRNA) transcript. The mRNA functions during translation to specify the sequence or order of amino acids in a nascent polypeptide.

As used herein, the term "heterologous gene" refers to a gene that is not in its natural environment, e.g., a gene from one species introduced into another species. A heterologous gene also includes a gene native to an organism that has been altered in some way (e.g., mutated, added in multiple copies, linked to non-native regulatory sequences, etc). Heterologous genes are distinguished from endogenous genes in that the heterologous gene sequences are typically joined to DNA sequences that are not found naturally associated with the gene sequences in the chromosome or are associated with portions of the chromosome not found in nature (e.g., genes expressed in loci where the gene is not normally expressed).

As used herein, the term "host cell" refers to any cell capable of replicating and/or transcribing and/or translating a heterologous gene. Thus, a "host cell" refers to any eukaryotic or prokaryotic cell (e.g., such as 293T, Raji, or Vero cells, or bacterial cells, E. coli, yeast cells, mammalian cells, avian cells, amphibian cells, plant cells, fish cells, and insect cells), whether located in vitro or in vivo. As used herein, the term "in vitro" refers to events that occur in an artificial environment, e.g., in a plate, well, test tube or reaction vessel, in cell culture, etc., rather than within a multi-cellular organism or tissue.

As used herein, the term "in vivo" refers to events that occur within a multi-cellular organism. For example only and not meant to be limiting, such as a mammal more particularly a human and/or non-human animal.

As used herein, the term "nucleic acid sequence" refers to an oligonucleotide, a nucleotide or a polynucleotide, and fragments or portions thereof, including, DNA or RNA of genomic or synthetic origin, which may be single or double-stranded, and represent the sense or antisense strand. As used herein, the term "amino acid sequence" as used herein refers to peptide or protein sequence.

As used herein, the terms "operably linked", "in operable combination," or "in operable order," refer to the linkage of a nucleic acid sequence in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The terms also refer to the linkage of amino acid sequences in such a manner so that a functional protein is produced.

As used herein, the phrase "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

As used herein, the term "purified" refers to molecules, either polynucleotides or polypeptides that are removed from their natural environment, isolated or separated. An "isolated nucleic acid sequence" is therefore a purified nucleic acid sequence. "Substantially purified" molecules are at least 60% free, preferably at least 75% free, and more preferably at least 90% free from other components with which they are naturally associated.

As used herein, the term "purified" refers to the removal of contaminants from a sample. The removal of contaminating proteins results in an increase in the percent of polypeptide of interest in the sample. In another example, recombinant polypeptides are expressed in plant, bacterial, yeast, or mammalian host cells and the polypeptides are purified by the removal of host cell proteins; the percent of recombinant polypeptides is thereby increased in the sample. As used herein, the term "reporter gene" refers to a gene that, when expressed as a protein, produces a polypeptide that is capable of being identified in an assay. One example of a reporter gene includes that are fluorescent, e.g., luciferase, green fluorescent protein, red fluorescent protein, that can be visually identified (e.g. with marked probes or antibodies) as known to those skilled in the art. Further, while specific examples are given any other means of fluorescent, bioluminescent, luminescent, and related reporter proteins useful for tracking are contemplated by the present invention. Other reporter genes include enzymes that can be used to metabolize or perform a reaction that creates a change in color (β-galactosidase) or the presence of a new metabolite (e.g., acetylation of chloramphenicol). Non-limiting examples of fluorescent proteins include: green fluorescent protein (GFP) PDB designation 1GFL; yellow fluorescent protein (YFP) PDB designation 3DPW; blue fluorescent protein (BFP) PDB designation 1BFP; Cerulean fluorescent protein (CFP) PDB designation 2WSO; Cyan fluorescent protein (CFP) PDB designation 2WSN; red fluorescent protein from Zooanthus sp. (RFP) PDB designation 2ICR; red fluorescent protein from Entremacaea quadricolor (RFP) PDB designation 2PJB, flavin mononucleotide (FMN)-binding fluorescent proteins (FbFPs).

As used herein, the term "subject" or "patient" refers to any organism immunized with the nucleic acid constructs, polypeptides, cells, or Reporter Virus Particles (RVP) invention are administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms; etc.). In vitro systems may also be used (e.g. to express ZIKV proteins or portions thereof for study within the target cell and/or for isolation).

As used herein, the term "vaccine" refers to an immunogenic composition that is administered to a host to provide some degree of protection from an infection and/or disease from a target virus or pathogen, e.g., ZIKV. Immunogenic agents are those that trigger an immune response in an animal. Moreover, some degree of protection includes but is not limited to decreasing, reducing, modifying, and/or ameliorating one or more symptoms of an infection and/or disease.

Vaccines can be administered alone or in combination with various adjuvants/carriers. Pharmaceutical carriers for immunization are known to those skilled in the art. These most typically would be standard carriers for administration of vaccines to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH. Other ingredients can include, e.g., adjuvants, excipients, carriers, thickeners, diluents, buffers, preservatives, and surface-active agents. As used herein, the term "vaccine-certified cells", refers to those cell lines that are routinely used for vaccine manufacturing purposes. A certification process is used, e.g., that taught by Donis, "Performance characteristics of qualified cell lines for isolation and propagation of influenza viruses for vaccine manufacturing", Vaccine, Volume 32, Issue 48, 12 November 2014, Pages 6583-6590, relevant portions incorporated herein by reference.

As used herein, the term "virus-like particle" (VLP) refers to a membrane-surrounded viral core structure having viral envelope proteins. In addition, additional molecules can be expressed in or on the VLP, e.g., adjuvants, antigens, or reporter proteins or enzymes. Typically, viral core proteins are located within the membrane of the VLP and do not contain intact viral nucleic acids, and are non-infectious.

The present invention includes a vaccine against Zika virus as well as a high throughput assay to test anti Zika neutralizing antibodies and anti-viral compounds. The present invention provides for the first time a VLP -based vaccine, due to their protein composition only. Moreover, no VLP-based methodologies have been shown or published for Zika virus. VLP based approaches are both safer and more effective than DNA vaccines. The VLP system of the present invention overcomes the deficiencies in nucleic acid based approaches.

Development of Virus like Particle Vaccine and Reporter Assay for Zika Virus. Recent worldwide outbreaks of Zika virus (ZIKV) infection and the lack of an approved vaccine raise serious concerns regarding preparedness to combat this emerging virus. The present inventors used a Virus Like Particle (VLP) based approach to develop a vaccine and a microneutralization assay against ZIKV. Synthetic C-prM-E gene construct of ZIKV was used to generate Reporter Virus Particles (RVP) that package GFP reporter expressing WNV replicon. The assay was adapted to 96-well format, similar to the Plaque Reduction Neutralization assay (PRNT) and showed high reproducibility with specific detection of ZIKV neutralizing antibodies. Furthermore, C-prM-E and prM-E VLPs were tested as vaccine candidates in mice and compared to DNA vaccination. While flaviviral prM-E constructs alone are sufficient for generating VLPs; it was found that efficient VLP production with the C-prM-E constructs could be achieved in the presence of WNV NS2B-3 protease that cleaves C from prM allowing for VLP release. Immunization studies in mice showed that VLPs generated higher neutralizing antibody titers than DNA vaccine with C-prM-E VLPs slightly better than prM-E VLPs. The superiority of C-prM-E VLPs demonstrates that inclusion of Capsid may have benefits for ZIKV and other flaviviral VLP vaccines. To facilitate the VLP platform, the present inventors generated stable cell line expressing high levels of ZIKV prM-E proteins that constitutively produce VLPs as well as a cell line expressing ZIKV C-prM-E proteins for RVP production. Thus, the present invention provides a scalable platform to tackle the emerging threat of ZIKV by developing a safe, effective and economical ZIKV vaccine.

The present invention addresses two areas highly relevant to Zika virus research-development of an economical, efficacious and safe VLP -based vaccine and a rapid versatile diagnostic assay. The cell lines stably expressing the Zika prM-E were generated to produce high amounts of VLPs in the supernatants and a Zika C-prM-E expressing cell line was developed that produces reporter virus particle upon transfection with GFP replicon plasmid. It was found that the prM-E VLPs induced a strong neutralizing antibody response in mice that was better when the capsid protein was included in the VLPs. The VLP -based vaccines of the present invention showed significantly better neutralizing antibody response when compared to their DNA counterparts. Finally, a RVP -based microneutralization assay was developed that works similar to the plaque reduction neutralization test (PRNT) assay with a rapid GFP readout in a 96 well format. Thus, the VLP based platform provides a source for Zika vaccine, detection, and diagnosis that can be rapidly adapted to current outbreaks.

The present inventors determined the immune response in mice vaccinated with ZIKV VLP generated using a prM-E or the C-prM-E construct and compared it to the DNA based counterparts. To generate PrM-E VLPs, the inventors developed a cell line stably expressing the prM-E proteins while for C-prM-E, VLPs were generated by cotransfection of cells with C-prM- E and NS2B-3 protease of WNV to allow cleavage of Capsid (C) from prM-E. VLPs were purified via ultracentrifugation and used to immunize mice. Concomitantly, the inventors also developed a rapid and quantitative GFP based micro-neutralization assay using Reporter Virus Particles (RVPs) for measuring the neutralizing immune response generated in mice. These data demonstrates that both prM-E and C-prM-E based VLP vaccines were highly effective in generating neutralizing antibodies with the latter being more potent. Interestingly, while the prM-E DNA based vaccine was less effective than both the VLPs, the C-prM-E DNA construct was unable to generate significant neutralizing antibodies, most likely due to the lack of VLP formation in the absence of NS2B-3 protease. The RVP based neutralization assay of the present invention is safe for regular clinical laboratories because it does not require use of live virus and yielded results in <72 hrs compared to the PRNT assay that may require a week for completion. The assay was highly reproducible and effective in measuring the neutralizing antibody response against ZIKV that could be conducted in 96-well format using simple fluorescent microscopy. Thus, this study demonstrates for the first time the use of a VLP platform to tackle the emerging threat of ZIKV by providing a scalable source for a VLP based vaccine and RVP based diagnostic assay.

Cell culture and reagents. 293T and Vero cells were obtained from ATCC and cultured in DMEM supplemented with 10% FBS. All transfections were performed using Turbofect reagent (Thermo Fisher) as per the manufacturer's instructions. The WNV C-prM-E and Rep/GFP plasmids have been described previously (39) and were kindly provided by Dr. Ted Pierson ( AID). Plasmid containing the WNV NS2B-3 accessory fusion protein expressing the active protease has been described previously (52) and was a kind gift from Dr. Frank Scholle (NC State Univ). The ZIKV-117 antibody was kindly provided by Dr. James Crowe (Vanderbilt University Medical Center, Nashville Tennessee) and ZIKV mouse polyclonal sera against the isolate MR766 was kindly provided by the Centers for Disease Control and Prevention (Fort Collins, Colorado). The ZIKV isolate PRVABC59 derived from a human serum specimen from Puerto Rico in December 2015 was obtained from ATCC and propagated in Vero cells strictly following the ATCC recommendations. Generation of vectors expressing ZIKV C-prM-E and prM-E: ZIKV C-prM-E construct was synthesized using the complete ZIKV sequence available from the current outbreak in Americas (accession number KU312312.1). This most current sequence was used to synthesize a codon optimized version of the C-prM-E gene using the Gene Synthesis Technology by GenScript, Piscataway, NJ, USA. The synthesized gene was subcloned into pcDNA3.1 vector (Invitrogen) using BamHl and Xhol restriction sites. The C-prM-E cassette was also cloned into the lentiviral vector pLenti6/V5 vector (Invitrogen) using the above restriction sites to generate plasmid pLenti-C-prM-E. The prM-E construct was generated by PCR amplification of prM-E region spanning amino acids 105-795 using the Phusion high fidelity PCR kit (New England BioLabs) and cloned into the pcDNA3.1+ vector. The prM-E cassette was also subcloned into pLenti6/V5 vector using the BamHl-Xhol sites to generate plasmid pLenti-prM-E. Plasmids were sequenced to confirm sequence identity.

Detection of ZIKV E protein expression. Detection of ZIKV virus E protein was conducted either via immunofluoresscnce or Western blotting. For this, cells were stained using ZIKV E specific antibody MAB10216 (clone 4G2, EMD Millipore) that reacts with Flavivirus group specific antigens followed by secondary antibody Alexa 488 (Invitrogen) and analyzed by fluorescence microscopy. The monoclonal antibody MAB8150 (clone 3.67G, EMD Millipore) was used as control and reacts with E protein of West Nile/Kunjin virus. For Western blotting, lysates were resolved on an SDS-PAGE gel, transferred onto PVDF membranes and probed with ZIKV virus E antibody (GTX133314, GeneTex, 1 :3000) followed by HRP conjugated anti mouse secondary antibody and bands visualized via enhanced chemiluminescence using the Super signal West Femto substrate (Pierce).

Metabolic labeling and immunoprecipitation. The protocol for radiolabeling and immunoprecipitation of cell and virus lysates has been described in detail previously (Garg,

2013. Briefly, transfected cells were washed with RPMI medium lacking Met and Cys.

Thereafter, cells were incubated in RPMI medium supplemented with FBS and [35S]Met/Cys.

Culture supernatants were filtered and subjected to ultracentrifugation at 100,000 x g for 45 min.

Cell and virion samples were lysed with Triton X containing lysis buffer (0.5% Triton X-100, 300 mM NaCl, 50 mM Tris [pH 7.5] containing protease inhibitors [Complete; Roche]).

Thereafter, lysates were immunoprecipitated with anti-WNV serum (kindly provided by Dr.

Robert B. Tesh, University of Texas Medical Branch, Galveston) or MAB 10216 coated Protein

A beads. Immunoprecipitated cell lysates were washed three times with TritonX-100 wash buffer and once with SDS-DOC wash buffer (0.1% sodium dodecyl sulfate, 300 mM NaCl, 50 mM Tris [pH 7.5], 2.5 mM deoxycholic acid), resolved by SDS-PAGE followed by

Phosphorlmager analysis.

Production of RVPs. ZIKV RVPs were generated using protocol described previously (38) with some modifications and originally described by Pierson et al (39). 293T cells were co- transfected with the ZIKV C-prM-E construct along with plasmid containing the sub-genomic GFP expressing replicon derived from lineage II strain of WNV (39). The RVPs were harvested 48h post transfection, aliquoted and stored for future use. RVPs were titrated in Vero cells plated in 96 well clear bottom black plates at 5,000 cells per well. Thereafter, cells were infected with serial two-fold dilutions of the RVPs and incubated for 72h. The plates were fixed using 4% formalin/PBS, images acquired using a Nikon EclipseTi microscope and number of GFP positive cells counted using the NIS elements software (Nikon).

RVP based and clinical ZIKV based microneutralization assay. For neutralization assays, mouse sera or antibodies were serially diluted in DMEM in technical duplicates in a volume of 50μ1. Thereafter, a predetermined concentration of virus producing 200 to 500 GFP positive cells was added to each well in a volume of 50μ1. The sera and RVPs were incubated for lh at room temperature. Subsequently, the virus:sera mix (ΙΟΟμΙ) was added to Vero cells after removing all the media and incubated for ~72h after which the number of GFP positive cells was quantitated. Statistical analysis was performed using the unpaired t test. The limit of detection for the RVP assay was defined as the highest concentration of sera (1 :20 dilution) used in the neutralization experiments.

For neutralization assays using the ZIKV isolate PRVABC59, mouse sera was serially diluted in serum free media in technical duplicates in a volume of 50μ1. Thereafter, diluted ZIKV was added to each sera sample at an MOI of ~5 and incubated for 2h at 37°C. The virus antibody mixtures were then added to Vero cells plated in 96-well plates and incubated for another 2h at 37°C. The virus antibody mix was then removed and cells incubated in DMEM containing 10% FBS for 48h. The cells were then fixed and stained using MAB10216. Images were acquired using fluorescence microscopy and number of antibody stained cells quantitated as described above.

Generation of stable cell lines expressing ZIKV C-prM-E or prM-E: Lentiviral vectors expressing ZIKV C-prM-E and prM-E were packaged in 293T cells by transfecting with pLenti- C- prM-E or pLenti-prM-E along with the helper construct php-dl-NA (NIH AIDS Reagent program) and VSVG Env. The viral supernatants were collected at 48h post transfection, aliquoted and stored. To generate stable cell lines, 293T cells were transduced with the lentiviral particles and the cells were selected using blasticidin at a concentration of 10μg/ml. Bulk selected cells were passaged 8-10 times and stained for ZIKV E protein expression using monoclonal Ab MAB 10216 at regular intervals to confirm selection. Subsequently, single cell clones were generated from the bulk selected cells using limiting dilution cloning in 96 well plates. Up to 6 single cell clones were selected for both C-prM-E and prM-E constructs from wells that showed single colony formation. Each single cell clone was further characterized for ZIKV E protein expression using immunostaining followed by flow cytometry.

Production of DNA and VLPs for immunization: For DNA immunizations in mice, the pcDNA- C-prM-E and pcDNA-prM-E plasmids were purified using the endotoxin free plasmid maxi kit (Qiagen) following the manufacturer's protocol. VLPs for immunization were generated and purified. For C-prM-E VLPs, 293T cells were transfected with pcDNA3.1-CprM-E construct along with WNV NS2B-3 plasmid. The viral supernatants were harvested at 24h and 48h post transfection. For production of prM-E virus particles, the 293T-Lenti-prM-E bulk cell line was cultured in the absence of Blasticidin and supernatants harvested at 72 and 96 hrs. VLPs were concentrated as follows. Harvested supernatants (25-30ml) were transferred into ultracentrifuge tubes and carefully underlayed with 5ml of 25% glycerol in T E buffer. VLPs were pelleted by centrifugation at 110,500 x g for 3 hrs at 4oC. Thereafter, the supernatant was carefully removed and the VLP pellet resuspended in PBS or TNE buffer. The total protein content in the VLP prep was measured using the micro BCA kit (Pierce) and specific E protein in the prep was detected by western blotting using the GTX133314 antibody.

Electron microscopy. VLPs were concentrated as above and imaged by electron microscopy after negative staining. Purified VLPs (3μ1) were applied to glow-discharged carbon-coated 300 mesh grid. After ~1 min, the grid was blotted with filter paper and 3μ1 of 2% uranyl acetate aqueous solution was added for 30 sec. After blotting off excess liquid and drying, images were acquired using the Transmission Electron Microscope JEOL1010 with a Hamamatsu digital camera and AMT Advantage image capture software at 100X magnification.

Mice studies. For immunization studies, 6 to 8 week old Balb/c mice were purchased from Jackson laboratory and housed in pathogen free animal facility at Texas Tech University Health Sciences Center, El Paso. Mice were divided into groups of six mice each and immunized with different preparations. For DNA immunization, a total of 50μg of DNA in a volume of ΙΟΟμΙ PBS was injected intramuscularly in each thigh. Mice received two additional boosts at week 2 and 4 of primary immunization as described above. For VLPs, the first immunization consisted of VLP prep (approximate total protein content 1.7-2.3mg/ml) mixed with TiterMax Gold adjuvant (Sigma) at a 1 : 1 ratio in total volume of ΙΟΟμΙ injected intramuscularly in each thigh. Mice received two additional boosts of VLPs at week 2 and 4 without adjuvant. Control mice were sham injected with PBS. Blood was collected from mice at week 9 post first immunization under terminal isoflurane anesthesia followed by intracardiac puncture. Blood samples were collected in serum separator tubes as per the manufacturer's recommendations. After coagulation, the tubes were centrifuged, sera harvested, aliquoted and stored at -80°C until further use.

Expression of ZIKV C-prM-E using codon optimized synthetic construct: To develop a VLP based platform for ZIKV vaccine and RVP assay, the inventors used the complete ZIKV sequence available from the current outbreak in Americas (KU312312.1) (6) to synthesize a codon optimized C-prM-E gene. The isolate was derived from a 52 year male hospitalized with symptoms of conjunctivitis and exanthema in Paramaribo (Suriname) in 2015. The synthesized gene was cloned into pcDNA3.1 vector that contains a CMV promoter (FIG. 1A). 293T cells transfected with the synthetic ZIKV C-prM-E or WNV C-prM-E construct were tested for E protein expression by immunofluorescence using antibodies MAB10216 (clone 4G2) and MAB8150 (clone 3.67G). As shown in FIG. IB, the MAB 10216 reacted with both ZIKV and WNV E protein while the MAB8150 only reacted specifically with the WNV E protein. This is expected as the MAB10216 reacts with flavivirus group specific antigens (36) and binds to the fusion loop at the extremity of domain II of the E protein while the MAB8150 is specific for the E protein of West Nile/Kunjin virus. The inventors also performed radioimmunoprecipitation analysis of ZIKV C-prM-E expression after transfection in 293T cells and compared it to WNV. As shown in FIG. 1C, both ZIKV and WNV C-prM-E were immunoprecipitated with MAB10216 and also with anti-WNV mouse sera, although with different efficiencies, emphasizing the relatedness between flaviviruses (37). These data demonstrate that the ZIKV C- prM-E synthetic construct expresses the viral proteins at high levels and can be used in downstream assays requiring VLP production.

RVP based microneutralization assay for ZIKV using a 96 well format and GFP readout. To make and test ZIKV RVPs, the inventors used the method described previously by Garg et al (38) and adapted from Pierson et al (39). 293T cells were co-transfected with the ZIKV C-prM-E construct along with the WNV replicon reporter plasmid Rep/GFP (29, 39, 40) that provides the WNV accessory proteins and the GFP reporter gene (FIG. 2A). RVPs generated in a similar manner using WNV C-prM-E alongside were used as positive control. Both WNV and ZIKV RVPs showed robust infection of Vero and 293T cells (FIG. 2B).

Although ZIKV RVPs have recently been used by other labs, the current method of choice is infection of Raji or Vero cells, followed by detection of GFP+ cells via flow cytometry (29, 39, 40). On the other hand, the infectious virus based PRNT utilizes Vero cells in 96 well format and can be tedious and time consuming. The inventors adapted The RVP based assay to a 96 well plate using Vero cells similar to the PRNT method. Cells plated in 96 well clear bottom black tissue culture plates were infected with serial dilutions of the RVPs. The plates were fixed 72h later, analyzed by fluorescent microscopy and the number of GFP positive cells counted using an automated software (NIS Elements, Nikon). Serial dilutions (1-6) of the input virus showed a dose dependent decrease in the number of GFP positive cells (FIG. 2C). The assay was conducted using ZIKV (FIG. 2D) and WNV (FIG. 2E) RVPs and GFP+ cells were quantified using the automated software. The assay showed high level of reproducibility with minimum variation between replicate wells. The linear range of the assay was between 200-800 GFP+ cells. The inventors also compared manual cell counting versus automated software (NIS Elements) and found a high degree of correlation between the two methods (r=0.9650, p<0.0001) (FIG. 2F) suggesting that manual GFP+ counting using a simple fluorescent microscope could also be used for the assay. Hence, this assay, much like the PRNT, can be adapted to give a reasonable number of GFP+ cells that can be counted either manually or using automated software. The inventors next tested whether the assay could detect neutralization of ZIKV RVPs via antibodies or polyclonal mouse sera. Experiments were carried in a manner similar to the standard PRNT, where sera/antibody dilutions were incubated with RVPs for lh prior to addition to Vero cells. As shown in FIG. 2G, a human antibody against the ZIKV E protein (ZIKV- 117) known to prevent infection via cross linking the protein (41) potently inhibited RVP infection in a dose dependent manner. Moreover, WNV pooled sera, polyclonal ZIKV sera and MAB10216 also inhibited ZIKV RVP infection in a dose dependent manner (FIG. 2H). Interestingly, and as expected, the antibody MAB8150 that failed to bind ZIKV E protein in immunofluorescence did not inhibit RVP infection (FIG. 2H). This demonstrates that the assay is specific and can be readily used to test for the presence of ZIKV neutralizing antibodies. Similar to PRNT, this assay can also be used to calculate EC50 and EC90 for serum samples. For example, the EC90 for WNV serum to inhibit 90% ZIKV infection was determined to be 80 and the EC50 to inhibit 50% ZIKV infection was 640 (FIG. 2H). The above data demonstrate that WNV replicon reporter construct along with ZIKV C-prM-E can yield infectious RVPs that can subsequently be used for several applications including a microneutralization assay while obviating the use of infectious virus and high level biosafety containment.

Establishment of a stable cell line expressing ZIKV C-prM-E. The sudden outbreaks of ZIKV infection have caught the research community off guard and the limited number of reagents and resources available for ZIKV studies has hampered research efforts. The inventors hence developed a stable cell line expressing the ZIKV C-prM-E to facilitate generation of RVPs. For this, the codon optimized ZIKV C-prM-E cassette was subcloned into a lentiviral vector (pLenti6/V5) carrying a Blasticidin resistance gene (FIG. 3A). Lentiviral particles generated with this vector were used to transduce 293T cells which were selected in the presence of 10μg/ml Blasticidin for ~2 weeks. Bulk selected cells were tested for expression of ZIKV E protein via fluorescence microscopy (FIG. 3B) and flow cytometry (FIG. 3C) and showed robust expression with more than 90% cells positive for the E antigen. The inventors also confirmed the expression of ZIKV E protein using Western blotting in the selected cell line and compared it to transient protein expression after transfection. As expected, high levels of E protein expression was seen in the 293T-C-prM-E-bulk cell line (FIG. 3D). As lentivirus transduced cells can have different levels of exogenous gene expression (42, 43), the inventors also selected single cell clones from the parent bulk cell line. A total of 6 single clones were selected via limiting dilution cloning and characterized for ZIKV E expression using fluorescent microscopy (FIG. 3E) as well as flow cytometry (FIG. 3F). Based on The flow data, clone F6 (293T-C-prM-E-F6) showed the highest expression and was chosen for subsequent experiments. The inventors generated RVPs using the 293T-C-prM-E-F6 cell line after transfecting with WNV Rep/GFP replicon plasmid and compared it to RVPs generated in 293T cells after transient transfection with pcDNA-C-prM-E and Rep/GFP. RVPs generated using the 293T-C-prM-E-F6 cell line were of significantly higher titers when compared to virus produced form 293 T cells (FIG. 3G). Thus, the 293T-C-prM-E-F6 cell line stably expresses the ZIKV structural proteins and can be used for generation of high titer RVPs.

Generation of prM-E cell line for VLP production. Extensive studies with different flaviviruses including ZIKV have shown that expression of prM-E in the absence of capsid can produce sub viral particles that can be used for immunization and elicit protective antibodies (44-48). These prM-E based constructs are also the basis of DNA and mRNA nanoparticle vaccines currently in development for ZIKV (20-25). The inventors hence developed a stable cell line that would constitutively produce prM-E VLPs in the supernatant and can be used for large-scale production of VLPs for immunization studies. For this, the inventors used the original C-prM-E construct and PCR amplified and cloned the prM-E region spanning amino acids 105-795 (FIG. 4A) into the pcDNA3.1 vector (FIG. 4B). The last 17 amino acids of capsid after the NS2B-3 cleavage site were included in the prM-E construct for proper translocation into the ER lumen. Once the expression of E protein (FIG. 4C) and VLP production in the supernatant (FIG. 4D) was confirmed, the prM-E region was subcloned into the lentivirus vector generating pLenti- prM-E (FIG. 4E). Packaged lentiviral particles were used to transduce 293T cells and generate bulk selected cell line and single clones (FIG. 4E). As above for the C-prM-E cell lines, 6 single cell clones were selected and characterized for ZIKV E protein expression (data not shown). The clones C4 and F4 showed highest expression both by immunofluorescence (FIG. 4F) and flow cytometry (FIG. 4G). Finally, the inventors characterized the single cell clones for production of VLPs in the supernatants. For this, supernatants from the 293T-prM-E cell lines were ultracentrifuged through a glycerol cushion (49), virus pellet was lysed and E protein detected by western blotting (FIG. 4H). Interestingly, the cell line 293T-prM-E-F4 showed significantly higher VLP production than the 293T-prM-E-Bulk and the 293T-prM-E-C4 cell line. Thus, the inventors have generated cell lines that constitutively produce high levels of ZIKV prM-E VLPs in the supernatant that can be used for VLP based vaccine studies. Production of VLPs from The stable cell lines can be readily scaled up for clinical trials.

Immunization studies in mice: Lack of an approved vaccine for ZIKV amidst the recent outbreaks and the association of ZIKV infections with severe congenital birth defects, warrants development of a safe and efficacious vaccine against the virus. For a vaccine to be available worldwide, especially in underdeveloped countries, it should be both easy to prepare and cost effective. In this regard, stable cell lines constitutively producing the ZIKV VLPs would be optimal as they can be readily scaled up with minimal scientific infrastructure and can provide an economical alternative to other forms of vaccination. The inventors hence tested the immunogenicity of the ZIKV VLPs in mice. Although prM-E VLPs are most commonly used in flavivirus vaccine research, the inventors also generated VLPs incorporating the capsid protein using the C-prM-E construct. While prM-E particles can be readily be generated using The stable cell lines (293T-prM-E), for generation of C-prM-E VLPs the WNV NS2B-3 protease was needed for cleavage of C from prM-E in the ER (35, 50). As shown in FIG. 5 A and consistent with published findings (50, 51), expression of the WNV protease NS2B-3 was essential for release of C-prM-E but not prM-E VLPs. Hence, for generation of C-prM-E VLPs, cells were co-transfected with the NS2B-3 expression plasmid (52). Using these two different strategies, the inventors generated both prM-E and C-prM-E VLPs that were purified from culture supernatants by ultracentrifugation through a glycerol cushion (49). The purified VLP pellet was analyzed for E protein expression via western blotting and VLP morphology by electron microscopy. As shown in FIG. 5B, there were high amounts of E protein detected in both the prM-E and C-prM-E VLP preps. The total protein content of the VLP preps ranged from ~1.7-2.3mg/ml. Electron microscopy showed that the VLP particle morphology was consistent with that of ZIKV with the approximate particle diameter ranging from 30-40nm (FIG. 5C).

The purified VLPs were subsequently used to immunize mice. The immunization schedule is shown in FIG. 5D. Mice divided into 5 groups of 6 mice each were immunized with either prM- E or C-prM-E DNA or the corresponding VLPs (FIG. 5E). Mice in the control group were sham injected with PBS. For the first immunization, the VLPs were emulsified with a 1 : 1 mix of TiterMax Gold. All mice received 2 booster injections with the respective DNA or VLPs at day 14 and day 28 and the mice were sacrificed on day 63, blood collected by intra-cardiac puncture and sera was harvested.

Anti-ZIKV virus immune response in mice immunized with prM-E/C-prM-E DNA and VLPs. The inventors next determined the immune response generated in mice upon immunization with the DNA or VLP based vaccines. Two-fold serial dilutions of the individual mice sera were tested in the RVP based microneutralization assay described in FIG. 2. Interestingly, the C-prM- E VLPs elicited the best neutralization titers followed by the prM-E VLPs and prM-E DNA vaccine (FIG. 6A). Interestingly, the C-prM-E DNA worked the poorest in generation of neutralizing antibodies most likely because C-prM-E DNA in the absence of NS2B-3 protease fails to assemble and release VLPs with the proteins being sequestered in the ER. The mean EC50 titers were >1 : 1000 for both the VLP vaccines and 1 : 132 for prM-E DNA while C-prM-E DNA showed <1 :32 titers (FIG. 6B). The EC90 titers followed a similar trend (FIG. 6C). Moreover, there was a significant difference in EC50 (p=0.0083) and EC90 values (p=0.0006) between the prM-E and C-prM-E VLP immunized mice (FIG. 6B and C). The inventors also tested the ability of the immune sera generated in mice to neutralize a clinical ZIKV isolate PRVABC59. As evident in FIG. 6D, the mice sera samples were able to neutralize the clinical ZIKV isolate following a trend similar to that seen with neutralization of ZIKV RVPs in FIG. 6A. To further confirm the presence of ZIKV E specific antibodies, the inventors conducted an immunoprecipitation assay using pooled sera from different groups of immunized mice. For this, C-prM-E expressing cells were radiolabeled with [ 5 S]Met/Cys protein labeling mix and cell lysates were immunoprecipitated with Protein A beads coated with either 3μ1 or 0.6μ1 of pooled sera from respective immunized groups. As shown in FIG. 6E and consistent with microneutralization data, pooled sera form the C-PrM-E VLP immunized group showed highest E protein band intensity followed by the prM-E VLP group. Sera from prM-E DNA group showed low levels of anti-E antibodies followed by the C-prM-E DNA, again consistent with neutralization data presented in FIG. 6A. These results confirm that high levels of neutralizing antibodies against ZIKV virus E protein were generated using either prM-E or C-prM-E VLPs and this approach can be used for generation of safe and effective vaccine.

The inventors have previously seen that immune sera from WNV infected mice is capable of binding to ZIKV E protein and cross neutralize ZIKV RVPs (Figures 1 and 2). The inventors hence determined if pooled sera form The ZIKV immunized mice groups was able to cross neutralize WNV infection using the RVP assay. For this, the inventors prepared WNV reporter VLPs using WNV C-PrM-E and the Rep/GFP construct as described previously (38). Interestingly, sera obtained from ZIKV VLP/DNA immunized mice was able to cross neutralize WNV RVP infection in a trend that was similar to ZIKV RVP inhibition, although with a lower efficacy (FIG. 6F). Here again, sera from ZIKV C-prM-E VLP immunized mice was most effective followed by prM-E VLPs and PrM-E DNA with C-prM-E DNA being the least effective (FIG. 6F). These data demonstrate the efficacy of VLP based vaccine compared to DNA vaccination in generating a robust neutralizing antibody response against ZIKV. Furthermore, incorporation of the capsid in VLPs shows enhanced immune response against ZIKV although prM-E VLPs are sufficient for inducing high level of neutralizing antibodies. The initial outbreaks of ZIKV virus infection in Africa and Southeast Asia and subsequent transfer of infection to western countries including the US has raised concerns regarding lack of preparedness to combat the virus (5-7). Currently there is no approved vaccine or specific treatment for ZIKV infections. Moreover, there is limited information regarding ZIKV spread, its enhanced specificity for neural fetal tissue and the mechanism via which it causes microcephaly. Thus, there is an urgent need to not only understand the basic virus biology but also develop safe and efficacious vaccines to target the virus. Moreover, there is a need for development of rapid, reliable and accurate assays that can be used to test anti-ZIKV immune responses and antiviral agents.

The ZIKV genome consists of a single stranded positive sense RNA and an open reading frame encoding a polyprotein 5'-C-prM-E-NSl-NS2A-NS2B-NS3-NS4A-NS4B-NS5-3' (53, 54). This polyprotein is subsequently cleaved into Capsid (C), precursor of membrane (prM), Envelope (E) and seven non- structural proteins (53, 55). The E protein (~53Kd) is the major virion surface protein involved in virus binding to the cell surface and membrane fusion (53, 55). Co- expression of the flaviviral proteins prM and E leads to secretion of VLPs that do not contain viral RNA and resemble empty particles produced during viral infection (48, 50, 56-58). These VLPs induce neutralizing antibody responses that are more potent than purified proteins (59, 60). One of the major obstacles in flavivirus research is the requirement of high level biosafety containment to undertake infectious virus studies with BSL-2 or higher needed for ZIKV studies. The persistence of ZIKV in body for prolonged periods (12) and the reports of sexual transmission (10, 11) highlight the risks associated with working with infectious virus. The use of VLPs has recently emerged as a powerful technology not only to study basic virus biology but also for vaccine and diagnostic assay development (59, 60). VLPs resemble viruses and are largely composed of viral structural proteins (53, 55). VLPs contain minimal or no genetic material, are non-replicating, and may also contain a reporter gene, thus allowing for easy detection (39, 61). Moreover, the requirement of high level biosafety containment to undertake infectious flavivirus studies (61, 62) can be overcome by using recombinant VLP based assays. This eliminates the use of infectious virus while still allowing testing of various aspects of the pathogen including: (1) mechanism of viral entry; (2) neutralizing antibody sensitivity; (3) vaccine efficacy; and (4) screening compounds that inhibit viral entry (63).

In this study, a construct expressing the C-prM-E polyprotein of ZIKV was used to package GFP reporter containing WNV replicon to generate RVPs similar to those generated with WNV (38, 39). WNV based RVPs have been used by the present inventors and others previously to study WNV E biology (29, 38, 39). ZIKV RVPs were generated using a codon optimized synthetic ZIKV C-prM-E construct that was co-transfected with WNV replicon construct (Rep/GFP). These RVPs were infectious in multiple cell lines and could be neutralized using anti-ZIKV antibodies or cross-neutralizing WNV immune sera. Although similar ZIKV RVPs have recently been used for anti-ZIKV response, the current approach involves detection of infected cells by flow cytometry (29, 40). While flow cytometry has its benefits, especially in minimizing human error and bias and can be readily used for large sample sizes, the plate-based assay may be more helpful in resource limiting conditions where flow cytometry is not available.

The inventors adapted the RVP assay to a 96-well plate format with a GFP readout that could be quantitated using a simple fluorescent microscope. The objective was to develop an assay that can replace PRNT, requires less time to complete, does not involve use of live ZIKV, is highly reproducible and can be used in resource limiting situations. The inventors tested the assay extensively in 96 well format and found that both the number of GFP+ cells and the neutralization sensitivity was highly reproducible. Both automated counting using imaging software or manual counting yielded similar results suggesting that the assay needs minimum infrastructure. For further ease and to facilitate the use of the assay, the inventors generated a stable cell line expressing ZIKV structural proteins (293T-C-prM-E-F6) that can be used for RVP production by transfecting with a GFP replicon construct, in the case WNV replicon. However, the same cell line could also be utilized to package ZIKV reporter replicons when those become available. Besides the elimination of the use of infectious virus, the assay also provides several key advantages that make it attractive for resource limiting areas. The assay requires simple to grow and maintain cell lines (293T-C-prM-E-F6 and Vero), standard transfection protocols, 96 well plates and a basic fluorescent microscope with a 4X objective. As in the case of PRNT, the plates can be fixed using formalin and saved for extended periods for quantitation of GFP positive cells at a later time. In The hands, the inventors have saved the fixed plates for up to 6 months without loss of GFP signal and quantified at different time points with comparable accuracy. With rapid spread of ZIKV in countries with limited scientific infrastructure, this assay would be a valuable tool to assess ZIKV neutralizing antibodies in response to either vaccination drives and/or natural infection with ZIKV or related flaviviruses. Recently, a number of studies have investigated vaccine candidates for ZIKV. These include conventional approaches like use of purified inactivated virus (PIV) (20, 21), DNA (20-22), adenoviral-based subunit vaccines incorporating prM-E or M-E regions of ZIKV (20, 23) as well as lipid nanoparticle (LNP) encapsulated RNA or modified mRNA as vaccine candidates (24- 26). These studies have demonstrated effective neutralizing antibody responses capable of protecting against ZIKV infection in various animal models. Recruitment of human subjects for testing some of these vaccine candidates is ongoing (NCT02963909, NCT02840487, NCT02887482, NCT02809443, NCT02952833). The geographical distribution of ZIKV is largely in developing and underdeveloped areas of the world (64). Hence, besides safety and efficacy being the top priorities for a successful vaccine, an important practical aspect for ZIKV vaccine is cost effectiveness and ease of production. In this regard, VLP based vaccines especially from cells lines stably expressing and releasing ZIKV proteins are highly attractive. The 293T-prM-E cell lines provide a much needed resource to take ZIKV VLP vaccines from the bench to the bedside. Not only do these cell lines release copious amount of E protein in the supernatant, but VLPs produced from these cells generate a robust neutralizing antibody response in mice making it ideal for further vaccine development.

The inventors also found that VLP vaccines were more efficacious than their DNA counterparts in inducing a neutralizing antibody response. In contrast, other studies have reported higher antibody titers upon immunization with a single dose of prM-E DNA (22) or prM-E modified RNA (25). This could be due to use of heterologous signal sequence from JEV to improve expression, use of stem/transmembrane region of the E protein from JEV to improve particle secretion (22) or use of signal peptide from MHC class II (25). Moreover, while C-prM-E VLPs worked better than prM-E VLPs, the C-prM-E DNA based vaccine was relatively non- efficacious. This is largely because the flavivirus NS3 enzyme along with the cofactor NS2B forms an active protease that cleaves the flavivirus C protein that spans the ER membrane producing its mature form (35, 50). Processing of the C protein by the viral protease in the ER is important for subsequent cleavage of E protein from prM by ER resident signal peptidases (50, 65). Interestingly, this study finds that inclusion of capsid in VLPs generates a better neutralizing immune response. However, it has been proposed that in the absence of an RNA genome, the nucleocapsid does not form and hence no capsid would be released into the supernatant (66) debating the benefit of a C-prM-E based vaccine.

Studies have suggested that T cells play an important role in generating a functional immune response in the presence of the viral capsid for Hepatitis B and C viruses (67, 68). Similarly, for DENV-4, epitopes in the capsid were shown to be recognized by CTLs that were cross reactive with other dengue serotypes (33). In fact, immunization by capsid alone was shown to generate a protective immune response independent of neutralizing antibodies and largely dependent on cell-mediated immunity (34). Moreover, CD4 T cells may also be involved in protection as specialized subsets have been implicated in lysing flavivirus infected cells (33, 69). Interestingly, The study shows that inclusion of capsid in VLPs requires a functional flavivivral protease, in the case WNV NS2B-3 fusion protein. The NS2B-3 fusion protein sequence itself is about 2Kb and can be easily included in VLP platforms, DNA vaccines as well as modified mRNA vaccines. In summary, the inventors describe the development, testing and efficacy of a VLP based vaccine against ZIKV and the generation of stable cell lines to facilitate this platform. The inventors also describe the optimization of a RVP based microneutralization assay using ZIKV C-PrM-E cell line and WNV replicon/GFP. This assay recapitulates the standard PRNT routinely used by virologists with several advantages including ease of use, reproducibility and eliminating infectious virus use. Thus, the study addresses two most relevant aspects of ZIKV infection, a safe, effective and economical vaccine and a neutralization assay that could be employed in the fight against the current ZIKV outbreaks.

SEQ ID NO: l, Zika virus isolate Zl 106033 nucleic acid sequence, GenBank Accession No. KU312312.1. Enfissi, A., Codrington, J., Roosblad, J., Kazanji, M. and Rousset, D., "Zika virus genome from the Americas", Lancet 387 (10015), 227-228 (2016).

1 acaggtttta ttttggattt ggaaacgaga gtttctggtc atgaaaaacc caaaaaagaa 61 atccggagga ttccggattg tcaatatgct aaaacgcgga gtagcccgtg tgagcccctt 121 tgggggcttg aagaggctgc cagccggact tctgctgggt catgggccca tcaggatggt 181 cttggcgatt ctagcctttt tgagattcac ggcaatcaag ccatcactgg gtctcatcaa 241 tagatggggt tcagtgggga aaaaagaggc tatggaaata ataaagaagt tcaagaaaga 301 tctggctgcc atgctgagaa taatcaatgc taggaaggag aagaagagac gaggcgcaga 361 tactagtgtc ggaattgttg gcctcctgct gaccacagct atggcagcgg aggtcactag 421 acgtgggagt gcatactata tgtacttgga cagaaacgat gctggggagg ccatatcttt 481 tccaaccaca ttggggatga ataagtgtta tatacagatc atggatcttg gacacacgtg 541 tgatgccacc atgagctatg aatgccctat gctggatgag ggggtggaac cagatgacgt 601 cgattgttgg tgcaacacga cgtcaacttg ggttgtgtac ggaacctgcc atcacaaaaa 661 aggtgaagca cggagatcta gaagagctgt gacgctcccc tcccattcca ctaggaagct 721 gcaaacgcgg tcgcaaacct ggttggaatc aagagaatac acaaagcact tgattagagt 781 cgaaaattgg atattcagga accctggctt cgcgttagca gcagctgcca tcgcttggct 841 tttgggaagc tcaacgagcc aaaaagtcat atacttggtc atgatactgc tgattgcccc 901 ggcatacagc atcaggtgca taggagtcag caatagggac tttgtggaag gtatgtcagg 961 tgggacttgg gttgatgttg tcttggaaca tggaggttgt gtcactgtaa tggcacagga 1021 caaaccgact gtcgacatag agctggttac aacaacagtc agcaacatgg cggaggtaag 1081 atcctactgc tatgaggcat caatatcaga catggcttcg gacagccgct gcccaacaca 1141 aggtgaagcc taccttgaca agcaatcaga cactcaatat gtctgcaaaa gaacgttagt 1201 ggacagaggc tggggaaatg gatgtggact ttttggcaaa gggagcctgg tgacatgcgc 1261 taagtttgca tgctccaaga aaatgaccgg gaagagcatc cagccagaga atctggagta 1321 ccggataatg ctgtcagttc atggctccca gcacagtggg atgatcgtta atgacacagg 1381 acatgaaact gatgagaata gagcgaaagt tgagataacg cccaattcac caagagccga 1441 agccaccctg ggggggtttg gaagcctagg acttgattgt gaaccgagga caggccttga 1501 cttttcagat ttgtattact tgactatgaa taacaagcac tggctggttc acaaggagtg 1561 gttccacgac attccattac cttggcacgc tggggcagac accggaactc cacactggaa 1621 caacaaagaa gcactggtag agttcaagga cgcacatgcc aaaaggcaaa ctgtcgtggt 1681 tctagggagt caagaaggag cagttcacac ggcccttgct ggagctctgg aggctgagat 1741 ggatggtgca aagggaaggc tgtcctctgg ccacttgaaa tgtcgcctga aaatggataa 1801 acttagattg aagggcgtgt catactcctt gtgtactgca gcgttcacat tcaccaagat 1861 cccggctgaa acactgcacg ggacagtcac agtggaggta cagtacgcag ggacagatgg 1921 accttgcaag gttccagctc agatggcggt ggacatgcaa actctgaccc cagttgggag 1981 gttgataacc gctaaccccg taatcactga aagcactgag aactctaaga tgatgctgga

2041 acttgatcca ccatttgggg actcttacat tgtcatagga gtcggggaga agaagatcac 2101 ccaccactgg cacaggagtg gcagcaccat tggaaaagca tttgaagcca ctgtgagagg 2161 tgccaagaga atggcagtct tgggagacac agcctgggac tttggatcag ttggaggcgc 2221 tctcaactca ttgggcaagg gcatccatca aatctttgga gcagctttca aatcattgtt 2281 tggaggaatg tcctggttct cacaaattct cattggaacg ttgctgatgt ggttgggtct 2341 gaacgcaaag aatggatcta tttcccttat gtgcttggcc ttagggggag tgttgatctt 2401 cttatccaca gccgtctctg ctgatgtggg gtgctcggtg gacttctcaa agaaggagac 2461 gagatgcggt acaggggtgt tcgtctataa cgacgttgaa gcctggaggg acaggtacaa 2521 gtaccatcct gactcccccc gtagattggc agcagcagtc aagcaagcct gggaagatgg 2581 tatctgcggg atctcctctg tttcaagaat ggaaaacatc atgtggagat cagtagaagg 2641 ggagctcaac gcaatcctgg aagagaatgg agttcaactg acggtcgttg tgggatctgt 2701 aaaaaacccc atgtggagag gtccacagag attgcccgtg cctgtgaacg agctgcccca 2761 cggctggaag gcttggggga aatcgtactt cgtcagagca gcaaagacaa ataacagctt 2821 tgtcgtggat ggtgacacac tgaaggaatg cccactcaaa catagagcat ggaacagctt 2881 tcttgtggag gatcatgggt tcggggtatt tcacactagt gtctggctca aggttagaga

2941 agattattca ttagagtgtg atccagccgt tattggaaca gctgttaagg gaaaggaggc

3001 tgtacacagt gatctaggct actggattga gagtgagaag aatgacacat ggaggctgaa 3061 gagggcccat ctgatcgaga tgaaaacatg tgaatggcca aagtcccaca cattgtggac 3121 agatggaata gaagagagtg atctgatcat acccaagtct ttagctgggc cactcagcca 3181 tcacaatacc agagagggct acaggaccca aatgaaaggg ccatggcaca gtgaagagct 3241 tgaaattcgg tttgaggaat gcccaggcac taaggtccac gtggaggaaa catgtggaac 3301 gagaggacca tctctgagat caaccactgc aagcggaagg gtgatcgagg aatggtgctg 3361 cagggagtgc acaatgcccc cactgtcgtt ccgggctaaa gatggctgtt ggtatggaat 3421 ggagataagg cccaggaaag aaccagaaag caacttagta aggtcaatgg tgactgcagg 3481 atcaactgat cacatggacc acttctccct tggagtgctt gtgattctgc tcatggtgca 3541 ggaagggttg aagaagagaa tgaccacaaa gatcatcata agcacatcaa tggcagtgct 3601 ggtagctatg atcctgggag gattttcaat gagtgacctg gctaagcttg caattttgat 3661 gggtgccacc ttcgcggaaa tgaacactgg aggagatgta gctcatctgg cgctgatagc 3721 ggcattcaaa gtcagaccag cgttgctggt atctttcatc ttcagagcta attggacacc 3781 ccgtgaaagc atgctgctgg ccttggcctc gtgtcttttg caaactgcga tctccgcctt

3841 ggaaggcgac ctgatggttc tcatcaatgg ttttgctttg gcctggttgg caatacgagc 3901 gatggttgtt ccacgcactg ataacatcac cttggcaatc ctggctgctc tgacaccact 3961 ggcccggggc acactgcttg tggcgtggag agcaggcctt gctacttgcg gggggtttat

4021 gctcctctct ctgaagggaa aaggcagtgt gaagaagaac ttaccatttg tcatggccct 4081 gggactaacc gctgtgaggc tggtcgaccc catcaacgtg gtgggactgc tgttgctcac

4141 aaggagtggg aagcggagct ggccccctag cgaagtactc acagctgttg gcctgatatg 4201 cgcattggct ggagggttcg ccaaggcaga tatagagatg gctgggccca tggccgcggt 4261 cggtctgcta attgtcagtt acgtggtctc aggaaagagt gtggacatgt acattgaaag 4321 agcaggtgac atcacatggg aaaaagatgc ggaagtcact ggaaacagtc cccggctcga 4381 tgtggcgcta gatgagagtg gtgatttctc cctggtggag gatgacggtc cccccatgag 4441 agagatcata ctcaaggtgg tcctgatgac catctgtggc atgaacccaa tagccatacc 4501 ctttgcagct ggagcgtggt acgtatacgt gaagactgga aaaaggagtg gtgctctatg 4561 ggatgtgcct gctcccaagg aagtaaaaaa gggggagacc acagatggag tgtacagagt 4621 aatgactcgt agactgctag gttcaacaca agttggagtg ggagttatgc aagagggggt 4681 ctttcacact atgtggcacg tcacaaaagg atccgcgctg agaagcggtg aagggagact 4741 tgatccatac tggggagatg tcaagcagga tctggtgtca tactgtggtc catggaagct 4801 agatgccgcc tgggacgggc acagcgaggt gcagctcttg gccgtgcccc ccggagagag 4861 agcgaggaac atccagactc tgcccggaat atttaagaca aaggatgggg acattggagc 4921 ggttgcgctg gattacccag caggaacttc aggatctcct atcctagaca agtgtgggag 4981 agtgatagga ctttatggca atggggtcgt gatcaaaaat gggagttatg ttagtgccat 5041 cacccaaggg aggagggagg aagagactcc tgttgagtgc ttcgagcctt cgatgctgaa 5101 gaagaagcag ctaactgtct tagacttgca tcctggagct gggaaaacca ggagagttct 5161 tcctgaaata gtccgtgaag ccataaaaac aagactccgt actgtgatct tagctccaac 5221 cagggttgtc gctgctgaaa tggaggaggc ccttagaggg cttccagtgc gttatatgac 5281 aacagcagtc aatgtcaccc actctggaac agaaatcgtc gacttaatgt gccatgccac 5341 cttcacttcg cgtctactac agccaatcag agtccccaac tataatctgt atattatgga 5401 tgaggcccac ttcacagatc cctcaagtat agcagcaaga ggatacattt caacaagggt 5461 tgagatgggc gaggcggccg ccatcttcat gaccgccacg ccaccaggaa cccgtgacgc 5521 atttccggac tccaactcac caattatgga caccgaagtg gaagtcccag agagagcctg 5581 gagctcaggc tttgattggg tgacggatca ttctggaaaa acagtttggt ttgttccaag 5641 cgtgaggaac ggcaatgaga tcgcagcttg tctgacaaag gctggaaaac gggtcataca 5701 gctcagcaga aagacttttg agacagagtt ccagaaaaca aaacatcaag agtgggactt 5761 tgtcgtgaca actgacattt cagagatggg cgccaacttt aaagctgacc gtgtcataga 5821 ttccaggaga tgcctaaagc cggtcatact tgatggcgag agagtcattc tggctggacc 5881 catgcctgtc acacatgcca gcgctgccca gaggaggggg cgcataggca ggaatcccaa 5941 caaacctgga gatgagtatc tgtatggagg tgggtgcgca gagactgacg aagaccatgc 6001 acactggctt gaagcaagaa tgctccttga caatatttac ctccaagatg gcctcatagc 6061 ctcgctctat cgacctgagg ccgacaaagt agcagccatt gagggagagt tcaagcttag 6121 gacggagcaa aggaagacct ttgtggaact catgaaaaga ggagatcttc ctgtttggct 6181 ggcctatcag gttgcatctg ccggaataac ctacacagat agaagatggt gctttgatgg 6241 cacgaccaac aacaccataa tggaagacag tgtgccggca gaagtgtgga ccagacacgg 6301 agagaaaaga gtgctcaaac cgaggtggat ggacgccaga gtttgttcag atcatgcggc 6361 cctgaagtca ttcaaggagt ttgccgctgg gaaaagagga gcggcttttg gagtgatgga 6421 agccctggga acactgccag gacacatgac agagagattc caggaagcca ttgacaacct 6481 cgctgtgctc atgcgggcag agactggaag caggccttac aaagccgcgg cggcccaatt 6541 gccggagacc ctagagacca ttatgctttt ggggttgctg ggaacagtct cgctgggaat 6601 cttcttcgtc ttgatgagga acaagggcat agggaagatg ggctttggaa tggtgactct 6661 tggggccagc gcatggctca tgtggctctc ggaaattgag ccagccagaa ttgcatgtgt 6721 cctcattgtt gtgttcctat tgctggtggt gctcatacct gagccagaaa agcaaagatc 6781 tccccaggac aaccaaatgg caatcatcat catggtagca gtaggtcttc tgggcttgat 6841 taccgccaat gaactcggat ggttggagag aacaaagagt gacctaagcc atctaatggg 6901 aaggagagag gagggggcaa ccataggatt ctcaatggac attgacctgc ggccagcctc 6961 agcttgggcc atctatgctg ccttgacaac tttcattacc ccagccgtcc aacatgcagt 7021 gaccacctca tacaacaact actccttaat ggcgatggcc acgcaagctg gagtgttgtt 7081 tggtatgggc aaagggatgc cattctacgc atgggacttt ggagtcccgc tgctaatgat 7141 aggttgctac tcacaattaa cacccctgac cctaatagtg gccatcattt tgctcgtggc 7201 gcactacatg tacttgatcc cagggctgca ggcagcagct gcgcgtgctg cccagaagag 7261 aacggcagct ggcatcatga agaaccctgt tgtggatgga atagtggtga ctgacattga 7321 cacaatgaca attgaccccc aagtggagaa aaagatggga caggtgctac tcatagcagt 7381 agccgtctcc agcgccatac tgtcgcggac cgcctggggg tggggggagg ctggggccct 7441 gatcacagcc gcaacttcca ctttgtggga aggctctccg aacaagtact ggaactcctc 7501 tacagccact tcactgtgta acatttttag gggaagttac ttggctggag cttctctaat 7561 ctacacagta acaagaaacg ctggcttggt caagagacgt gggggtggaa caggagagac 7621 cctgggagag aaatggaagg cccgcttgaa ccagatgtcg gccctggagt tctactccta 7681 caaaaagtca ggcatcaccg aggtgtgcag agaagaggcc cgccgcgccc tcaaggacgg 7741 tgtggcaacg ggaggccatg ctgtgtcccg aggaagtgca aagctgagat ggttggtgga 7801 gcggggatac ctgcagccct atggaaaggt cattgatctt ggatgtggca gagggggctg 7861 gagttactac gccgccacca tccgcaaagt tcaagaagtg aaaggataca caaaaggagg 7921 ccctggtcat gaagaacccg tgttggtgca aagctatggg tggaacatag tccgtcttaa 7981 gagtggggtg gacgtctttc atatggcggc tgagccgtgt gacacgttgc tgtgtgacat 8041 aggtgagtca tcatctagtc ctgaagtgga agaagcacgg acgctcagag tcctctccat 8101 ggtgggggat tggcttgaaa aaagaccagg agccttttgt ataaaagtgt tgtgcccata 8161 caccagcact atgatggaaa ccctggagcg actgcagcgt aggtatgggg gaggactggt 8221 cagagtgcca ctctcccgca actctacaca tgagatgtac tgggtctctg gagcgaaaag 8281 caacaccata aaaagtgtgt ccaccacgag ccagctcctc ttggggcgca tggacgggcc 8341 taggaggcca gtgaaatatg aggaggatgt gaatctcggc tctggcacgc gggctgtggt 8401 aagctgcgct gaagctccca acatgaagat cattggtaac cgcattgaaa ggatccgcag 8461 tgagcacgcg gaaacgtggt tctttgacga gaaccaccca tataggacat gggcttacca 8521 tggaagctat gaggccccca cacaagggtc agcgtcctct ctaataaacg gggttgtcag 8581 gctcctgtca aaaccctggg atgtggtgac tggagtcaca ggaatagcca tgaccgacac 8641 cacaccgtat ggtcagcaaa gagttttcaa ggaaaaagtg gacactaggg tgccagaccc 8701 ccaagaaggc actcgtcagg ttatgagcat ggtctcttcc tggttgtgga aagagctagg 8761 caaacacaaa cggccacgag tctgtaccaa agaagagttc atcaacaagg ttcgtagcaa 8821 tgcagcatta ggggcaatat ttgaagagga aaaagagtgg aagactgcag tggaagctgt 8881 gaacgatcca aggttctggg ctctagtgga caaggaaaga gagcaccacc tgagaggaga 8941 gtgccagagt tgtgtgtaca acatgatggg aaaaagagaa aagaaacaag gggaatttgg 9001 aaaggccaag ggcagccgcg ccatctggta tatgtggcta ggggctagat ttctagagtt 9061 cgaagccctt ggattcttga acgaggatca ctggatgggg agagagaact caggaggtgg 9121 tgttgaaggg ctgggattac aaagactcgg atatgtccta gaagagatga gtcgtatacc 9181 aggaggaagg atgtatgcag atgacactgc tggctgggac acccgcatta gcaggtttga 9241 tctggagaat gaagctctaa tcaccaacca aatggagaaa gggcacaggg ccttggcatt 9301 ggccataatc aagtacacat accaaaacaa agtggtaaag gtccttagac cagctgaaaa 9361 agggaaaaca gttatggaca ttatttcgag acaagaccaa agggggagcg gacaagttgt 9421 cacttacgct cttaacacat ttaccaacct agtggtgcaa ctcattcgga atatggaggc 9481 tgaggaagtt ctagagatgc aagacttgtg gctgctgcgg aggtcagaga aagtgactaa 9541 ctggttgcag agcaacggat gggataggct caaacgaatg gcagtcagtg gagatgattg 9601 cgttgtgaag ccaattgatg ataggtttgc acatgccctc aggttcttga atgatatggg 9661 aaaagttagg aaggacacac aagagtggaa accctcaact ggatgggaca actgggaaga 9721 agttccgttt tgctcccacc acttcaacaa gctccatctc aaggacggga ggtccattgt 9781 ggttccctgc cgccaccaag atgaactgat tggccgggcc cgcgtctctc caggggcggg 9841 atggagcatc cgggagactg cttgcctagc aaaatcatat gcgcaaatgt ggcagctcct 9901 ttatttccac agaagggacc tccgactgat ggccaatgcc atttgttcat ctgtgccagt 9961 tgactgggtt ccaactggga gaactacctg gtcaatccat ggaaagggag aatggatgac 10021 cactgaagac atgcttgtgg tgtggaacag agtgtggatt gaggagaacg

accacatgga

10081 agacaagacc ccagttacga aatggacaga cattccctat ttgggaaaaa

gggaagactt

10141 gtggtgtgga tctctcatag ggcacagacc gcgcaccacc tgggctgaga

acattaaaaa

10201 cacagtcaac atggtgcgca ggatcatagg tgatgaagaa aagtacatgg

actacctatc

10261 cacccaagtt cgctacttgg gtgaagaagg gtctacacct ggagtgctgt

aagcaccaat

10321 cttaatgttg tcaggcctgc tagtcagcca cagcttgggg aaagctgtgc agcc

SEQ ID NO:2. Zika vims isolate Zl 106033 polyprotein GenBank Accession No. KU312312.1. MKNPKKKSGGFRIVNMLKRGVARVSPFGGLKRLPAGLLLGHGPIRMVLAILAFLRFTAI KPSLGLINRWGSVGKKEAMEIIKKFKKDLAAMLRIINARKEKKRRGADTSVGIVGLLLT TAMAAEVTRRGSAYYMYLDRNDAGEAISFPTTLGMNKCYIQF DLGHTCDATMSYEC PMLDEGVEPDDVDCWCNTTSTWVVYGTCHHKKGEARRSRRAVTLPSHSTRKLQTRSQ TWLESREYTKHLIRVENWIFRNPGFALAAAAIAWLLGSSTSQKVIYLVMILLIAPAYSIR C IGVSNRDF VEGMSGGTWVD VVLEHGGC VTVMAQDKPTVDIELVTTT VSNMAEVRS YC YEASISDMASDSRCPTQGEAYLDKQSDTQYVCKRTLVDRGWGNGCGLFGKGSLVTCA KFACSKKMTGKSIQPENLEYRIMLSVHGSQHSGMIVNDTGHETDENRAKVEITPNSPRA EATLGGFGSLGLDCEPRTGLDFSDLYYLTMNNKHWLVHKEWFHDIPLPWHAGADTGT PHWNNKEALVEFKDAHAKRQTVVVLGSQEGAVHTALAGALEAEMDGAKGRLSSGHL KCRLKMDKLRLKGVS YSLCT AAFTFTKIPAETLHGT VTVEVQ YAGTDGPCKVP AQMAV DMQTLTPVGRLITANPVITESTENSKMMLELDPPFGDSYIVIGVGEKKITHHWHRSGSTI GK AFE AT VRGAKRM A VLGDT A WDF GS VGGALN SLGKGIHQIF GA AFK SLF GGM SWF S QILIGTLLMWLGLNAKNGSISLMCLALGGVLIFLSTAVSADVGCSVDFSKKETRCGTGV FVYNDVEAWRDRYKYHPDSPRRLAAAVKQAWEDGICGISSVSRMENF WRSVEGELN AILEENGVQLTVVVGSVKNPMWRGPQRLPWVNELPHGWKAWGKSYFVRAAKTNNSF VVDGDTLKECPLKHRAWNSFLVEDHGFGVFHTSVWLKVREDYSLECDPAVIGTAVKG

KEAVHSDLGYWIESEKNDTWRLKRAHLIEMKTCEWPKSHTLWTDGIEESDLIIPKSL AG

PLSHHNTREGYRTQMKGPWHSEELEIRFEECPGTKVHVEETCGTRGPSLRSTTASGR VIE EWCCRECTMPPLSFRAKDGCWYGMEIRPRKEPES LVRSMVTAGSTDHMDHFSLGVL VILLMVQEGLKKRMTTKIIISTSMAVLVAMILGGFSMSDLAKLAILMGATFAEMNTGGD VAHLALIAAFKVRPALLVSFIFRANWTPRESMLLALASCLLQTAISALEGDLMVLINGFA LAWLAIRAMVVPRTDNITLAILAALTPLARGTLLVAWRAGLATCGGFMLLSLKGKGSV KK LPFVMALGLTAVRLVDPINVVGLLLLTRSGKRSWPPSEVLTAVGLICALAGGFAKA DIEMAGPMAAVGLLIVSYVVSGKSVDMYIERAGDITWEKDAEVTGNSPRLDVALDESG DFSLVEDDGPPMREIILKVVLMTICGMNPIAIPFAAGAWYVYVKTGKRSGALWDVPAP KEVKKGETTDGVYRVMTRRLLGSTQVGVGVMQEGVFHTMWHVTKGSALRSGEGRLD PYWGDVKQDLVSYCGPWKLDAAWDGHSEVQLLAVPPGERARNIQTLPGIFKTKDGDI GAVALDYPAGTSGSPILDKCGRVIGLYGNGVVIKNGS YVSAITQGRREEETPVECFEPSM LKKKQLTVLDLHPGAGKTRRVLPEIVREAIKTRLRTVILAPTRVVAAEMEEALRGLPVR YMTTAVNVTHSGTEIVDLMCHATFTSRLLQPIRVPNYNLYIMDEAHFTDPSSIAARGYIS TRVEMGEAAAIFMTATPPGTRD AFPD SNSPIMDTEVEVPERAWS SGFDWVTDHSGKTV WFVPSVRNG EIAACLTKAGKRVIQLSRKTFETEFQKTKHQEWDFVVTTDISEMGA F K ADRVID SRRCLKP VILDGERVIL AGPMP VTH AS A AQRRGRIGR P KPGDE YL YGGGC AETDEDHAHWLEARMLLDNIYLQDGLIASLYRPEADKVAAIEGEFKLRTEQRKTFVEL MKRGDLPVWLAYQVASAGITYTDRRWCFDGTTNNTIMEDSVPAEVWTRHGEKRVLKP RWMDARVCSDHAALKSFKEFAAGKRGAAFGVMEALGTLPGHMTERFQEAID LAVL MRAETGSRPYKAAAAQLPETLETF LLGLLGTVSLGIFFVLMRNKGIGKMGFGMVTLG ASAWLMWLSEIEPARIACVLIVVFLLLVVLIPEPEKQRSPQDNQMAIIIMVAVGLLGLIT A ELGWLERTK SDL SHLMGRREEGATIGF SMDIDLRP AS AW AI YAALTTFITP AVQHAVT TSYNNYSLMAMATQAGVLFGMGKGMPFYAWDFGVPLLMIGCYSQLTPLTLIVAIILLV AHYMYLIPGLQAAAARAAQKRTAAGIMK PVVDGIVVTDIDTMTIDPQVEKKMGQVL LIAVAVSSAILSRTAWGWGEAGALITAATSTLWEGSP KYWNSSTATSLCNIFRGSYLA GASLIYTVTRNAGLVKRRGGGTGETLGEKWKARLNQMSALEFYSYKKSGITEVCREEA RRALKDGVATGGHAVSRGSAKLRWLVERGYLQPYGKVIDLGCGRGGWSYYAATIRKV QEVKGYTKGGPGHEEPVLVQSYGWNIVRLKSGVDVFHMAAEPCDTLLCDIGESSSSPE VEE ARTLRVL SM VGD WLEKRPGAF CIK VLCP YT S TMMETLERLQRRYGGGL VRVPL SR NSTHEMYWVSGAKSNTIKSVSTTSQLLLGRMDGPRRPVKYEEDVNLGSGTRAVVSCAE AP MKIIG RIERIRSEHAETWFFDE HP YRTWAYHGS YEAPTQGS AS SLING VVRLL SK PWDVVTGVTGIAMTDTTPYGQQRVFKEKVDTRVPDPQEGTRQVMSMVSSWLWKELG KHKRPRVCTKEEFINKVRSNAALGAIFEEEKEWKTAVEAVNDPRFWALVDKEREHHLR GECQSCVYNMMGKREKKQGEFGKAKGSRAIWYMWLGARFLEFEALGFLNEDFIWMG RENS GGGVEGLGLQRLGYVLEEMSRIPGGRM Y ADD T AGWD TRI SRFDLENE ALITNQM EKGHRALALAIIKYTYQ KVVKVLRPAEKGKTVMDIISRQDQRGSGQVVTYALNTFTN LVVQLIRNMEAEEVLEMQDLWLLRRSEKVTNWLQSNGWDRLKRMAVSGDDCVVKPI DDRFAHALRFL DMGKVRKDTQEWKPSTGWDNWEEVPFCSHHF KLHLKDGRSIVVP CRHQDELIGRARVSPGAGWSIRETACLAKSYAQMWQLLYFHRRDLRLMANAICSSVPV DWWTGRTTWSIHGKGEWMTTEDMLVVW RVWIEE DHMEDKTPVTKWTDIPYLGK REDLWCGSLIGHRPRTTWAENIKNTVNMVRRIIGDEEKYMDYLSTQVRYLGEEGSTPG VL

SEQ ID NO:3, codon optimized Zika C-prM-E synthesized.

ATGAAGAATCCCAAGAAGAAATCTGGCGGGTTCCGAATCG TC AAT ATGCTGAAGAGAGGAGTGGC AAGAGTGTC ACCTTTTGGCG GGCTGAAGAGGCTGCCTGCAGGACTGCTGCTGGGGCACGGACCAA TCAGGATGGTGCTGGCAATTCTGGCCTTCCTGCGCTTTACCGCTA TCAAACCCAGCCTGGGCCTGATTAATCGCTGGGGGTCCGTGGGAA AGAAAGAGGCTATGGAGATCATCAAGAAGTTCAAGAAAGACCTGG CCGCTATGCTGCGGATCATTAACGCTAGAAAGGAGAAGAAACGGA GAGGGGCAGATACCTCTGTGGGCATCGTCGGGCTGCTGCTGACCA CAGCAATGGCAGCCGAGGTGACAAGGCGCGGATCAGCCTACTATA TGTACCTGGACCGGAATGATGCTGGCGAAGCAATCAGCTTCCCAA CTACCCTGGGGATGAACAAGTGCTACATCCAGATTATGGACCTGG GCCACACATGCGATGCCACCATGAGCTATGAGTGTCCAATGCTGG ACGAGGGGGTGGAACCCGACGATGTCGATTGCTGGTGTAATACAA CTTCCACTTGGGTGGTCTACGGCACCTGTCACCATAAGAAAGGAG AAGCTCGGCGGAGCCGGAGGGCAGTGACACTGCCATCACACAGCA CTAGGAAGCTGCAGACACGCAGCCAGACTTGGCTGGAGTCCAGAG AATATACAAAACATCTGATCAGAGTGGAGAACTGGATCTTCCGGA ATCCAGGATTCGCACTGGCTGCAGCCGCTATCGCATGGCTGCTGG GCAGCTCCACCTCTCAGAAAGTGATCTACCTGGTCATGATCCTGC TGATTGCCCCCGCTTATTCTATCCGCTGCATTGGGGTGAGTAATC GAGACTTCGTCGAGGGAATGAGCGGCGGGACATGGGTGGATGTGG TCCTGGAAC ACGGAGGCTGCGTGACTGTGATGGCTC AGGAC AAGC CTACCGTGGATATCGAGCTGGTGACCACAACTGTCTCAAACATGG CCGAGGTGAGGAGCTACTGCTATGAAGCCTCCATTTCTGACATGG CTAGTGATTCACGCTGTCCAACCCAGGGCGAGGCCTACCTGGACA AGCAGAGTGATACCCAGTACGTGTGCAAACGAACACTGGTCGACC GGGGCTGGGGGAATGGATGTGGCCTGTTTGGGAAGGGAAGCCTGG TGACATGCGCCAAATTCGCTTGTTCCAAGAAAATGACTGGCAAGT CTATCCAGCCTGAGAACCTGGAATACAGGATTATGCTGAGCGTGC ACGGATCACAGCATAGCGGCATGATCGTCAACGACACCGGCCACG AGACAGATGAAAATCGAGCCAAAGTGGAGATTACCCCTAACTCTC CAAGAGCAGAAGCCACACTGGGGGGATTTGGAAGTCTGGGCCTGG ACTGCGAGCCACGAACCGGCCTGGACTTCTCCGATCTGTACTATC TGACAATGAACAATAAGCACTGGCTGGTGCATAAAGAATGGTTTC ACGAC ATCCC ACTGCCCTGGC ATGCTGGAGC AGATACCGGC AC AC CTCACTGGAACAATAAGGAGGCCCTGGTGGAGTTCAAGGATGCCC ATGCTAAACGGCAGACAGTGGTCGTGCTGGGGAGCCAGGAGGGAG CAGTGCACACTGCACTGGCCGGCGCTCTGGAGGCAGAAATGGACG GGGCCAAGGGAAGACTGTCTAGTGGGCATCTGAAATGCCGGCTGA AGATGGATAAACTGAGACTGAAGGGAGTGAGCTACTCCCTGTGC A CTGCAGCCTTCACTTTTACCAAAATCCCAGCTGAGACACTGCACG GCACAGTCACTGTGGAAGTCCAGTATGCCGGCACTGACGGCCCTT GTAAGGTGCCTGCACAGATGGCCGTCGATATGCAGACCCTGACAC CAGTGGGCCGGCTGATCACCGCCAATCCTGTCATTACTGAGAGTA CCGAAAACTCAAAAATGATGCTGGAGCTGGACCCCCCTTTTGGGG ATTCCTATATCGTGATTGGCGTCGGGGAAAAGAAAATCACACACC ATTGGCACCGGAGCGGCAGTACAATTGGGAAGGCTTTTGAGGCAA CTGTGCGCGGCGCCAAACGAATGGCTGTCCTGGGAGACACCGCAT GGGATTTCGGCAGTGTGGGAGGGGCTCTGAACTCACTGGGAAAGG GCATCCATCAGATTTTCGGAGCTGCCTTCAAGAGCCTGTTCGGAG GCATGTCCTGGTTCTCTCAGATCCTGATTGGCACTCTGCTGATGT GGCTGGGGCTGAACGCCAAGAATGGCAGCATCAGTCTGATGTGCC TGGCCCTGGGGGGGGTCCTGATTTTCCTGTCAACCGCAGTCTCCG CTGACTGATGA SEQ ID NO:4, codon optimized amino acid sequence of Zika C-prM-E

MKNPKKKSGGFRIVNMLKRGVARVSPFGGLKRLPAGLLLGHGPIRMVLAI LAFLRFTAIKPSLGLINRWGSVGKKEAME11KKFKKDLAAMLRI INARKE KKRRGADTSVGIVGLLLTTAMAAEVTRRGSAYYMYLDRNDAGEAISFPTT LGMNKCYIQIMDLGHTCDATMSYECPMLDEGVEPDDVDCWCNTTSTWWY GTCHHKKGEARRSRRAVTLPSHSTRKLQTRSQTWLESREYTKHLIRVENW

IFRNPGFALAAAAIAWLLGSSTSQKVIYLVMILLIAPAYSIRCIGVSNRD FVEGMSGGTWVDWLEHGGCVTVMAQDKPTVDIELVTTTVSNMAEVRSYC YEASISDMASDSRCPTQGEAYLDKQSDTQYVCKRTLVDRGWGNGCGLFGK GSLVTCAKFACSKKMTGKSIQPENLEYRIMLSVHGSQHSGMIVNDTGHET DENRAKVEITPNSPRAEATLGGFGSLGLDCEPRTGLDFSDLYYLTMNNKH WLVHKEWFHDIPLPWHAGADTGTPHWNNKEALVEFKDAHAKRQTVWLGS QEGAVH ALAGALEAEMDGAKGRLSSGHLKCRLKMDKLRLKGVSYSLC A AFTFTKIPAETLHGTVTVEVQYAGTDGPCKVPAQMAVDMQTLTPVGRLIT ANPVITESTENSKMMLELDPPFGDSYIVIGVGEKKITHHWHRSGSTIGKA FEA VRGAKRMAVLGD AWDFGSVGGALNSLGKGIHQIFGAAFKSLFGGM

SWFSQILIGTLLMWLGLNAKNGSISLMCLALGGVLIFLSTAVSAD

It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method, kit, reagent, or composition of the invention, and vice versa. Furthermore, compositions of the invention can be used to achieve methods of the invention.

It will be understood that particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims.

All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.

The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.

As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps. In embodiments of any of the compositions and methods provided herein, "comprising" may be replaced with "consisting essentially of or "consisting of. As used herein, the phrase "consisting essentially of requires the specified integer(s) or steps as well as those that do not materially affect the character or function of the claimed invention. As used herein, the term "consisting" is used to indicate the presence of the recited integer (e.g., a feature, an element, a characteristic, a property, a method/process step or a limitation) or group of integers (e.g., feature(s), element(s), characteristic(s), property(ies), method/process steps or limitation(s)) only. The term "or combinations thereof as used herein refers to all permutations and combinations of the listed items preceding the term. For example, "A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.

As used herein, words of approximation such as, without limitation, "about", "substantial" or "substantially" refers to a condition that when so modified is understood to not necessarily be absolute or perfect but would be considered close enough to those of ordinary skill in the art to warrant designating the condition as being present. The extent to which the description may vary will depend on how great a change can be instituted and still have one of ordinary skilled in the art recognize the modified feature as still having the required characteristics and capabilities of the unmodified feature. In general, but subject to the preceding discussion, a numerical value herein that is modified by a word of approximation such as "about" may vary from the stated value by at least ±1, 2, 3, 4, 5, 6, 7, 10, 12 or 15%.

All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

REFERENCES:

1. Dick GW. 1952. Zika virus. II. Pathogenicity and physical properties. Trans R Soc Trop Med Hyg 46:521-34. 2. Dick GW, Kitchen SF, Haddow AJ. 1952. Zika virus. I. Isolations and serological specificity. Trans R Soc Trop Med Hyg 46:509-20.

3. Duffy MR, Chen TH, Hancock WT, Powers AM, Kool JL, Lanciotti RS, Pretrick M, Marfel M, Holzbauer S, Dubray C, Guillaumot L, Griggs A, Bel M, Lambert AJ, Laven J, Kosoy O, Panella A, Biggerstaff BJ, Fischer M, Hayes EB. 2009. Zika virus outbreak on Yap Island, Federated States of Micronesia. N Engl J Med 360:2536-43.

4. Simpson DI. 1964. Zika Virus Infection in Man. Trans R Soc Trop Med Hyg 58:335-8.

5. Brasil P, Pereira JP, Jr., Moreira ME, Ribeiro Nogueira RM, Damasceno L, Wakimoto M, Rabello RS, Valderramos SG, Halai UA, Salles TS, Zin AA, Horovitz D, Daltro P, Boechat M, Raja Gabaglia C, Carvalho de Sequeira P, Pilotto JH, Medialdea-Carrera R, Cotrim da Cunha D, Abreu de Carvalho LM, Pone M, Machado Siqueira A, Calvet GA, Rodrigues Baiao AE, Neves ES, Nassar de Carvalho PR, Hasue RH, Marschik PB, Einspieler C, Janzen C, Cherry JD, Bispo de Filippis AM, Nielsen-Saines K. 2016. Zika Virus Infection in Pregnant Women in Rio de Janeiro. N Engl J Med 375:2321-2334.

6. Enfissi A, Codrington J, Roosblad J, Kazanji M, Rousset D. 2016. Zika virus genome from the Americas. Lancet 387:227-8.

7. Rasmussen SA, Jamieson DJ, Honein MA, Petersen LR. 2016. Zika Virus and Birth Defects—Reviewing the Evidence for Causality. N Engl J Med 374: 1981-7.

8. Cao-Lormeau VM, Blake A, Mons S, Lastere S, Roche C, Vanhomwegen J, Dub T, Baudouin L, Teissier A, Larre P, Vial AL, Decam C, Choumet V, Halstead SK, Willison HJ, Musset L, Manuguerra JC, Despres P, Fournier E, Mallet HP, Musso D, Fontanet A, Neil J, Ghawche F. 2016. Guillain-Barre Syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet 387: 1531-9.

9. Oehler E, Watrin L, Larre P, Leparc-Goffart I, Lastere S, Valour F, Baudouin L, Mallet H, Musso D, Ghawche F. 2014. Zika virus infection complicated by Guillain-Barre syndrome— case report, French Polynesia, December 2013. Euro Surveill 19.

10. Davidson A, Slavinski S, Komoto K, Rakeman J, Weiss D. 2016. Suspected Female-to- Male Sexual Transmission of Zika Virus - New York City, 2016. MMWR Morb Mortal Wkly Rep 65:716-7.

11. Mansuy JM, Suberbielle E, Chapuy-Regaud S, Mengelle C, Bujan L, Marchou B, Delobel P, Gonzalez-Dunia D, Malnou CE, Izopet J, Martin-Blondel G. 2016. Zika virus in semen and spermatozoa. Lancet Infect Dis 16: 1106-7.

12. Murray KO, Gorchakov R, Carlson AR, Berry R, Lai L, Natrajan M, Garcia MN, Correa A, Patel SM, Aagaard K, Mulligan MJ. 2017. Prolonged Detection of Zika Virus in Vaginal Secretions and Whole Blood. Emerg Infect Dis 23 :99-101. 13. Shi Y, Gao GF. 2017. Structural Biology of the Zika Virus. Trends Biochem Sci doi: 10.1016/j .tibs.2017.02.009.

14. Stadler K, Allison SL, Schalich J, Heinz FX. 1997. Proteolytic activation of tick-borne encephalitis virus by furin. J Virol 71 :8475-81.

15. Sapparapu G, Fernandez E, Kose N, Bin C, Fox JM, Bombardi RG, Zhao H, Nelson CA, Bryan AL, Barnes T, Davidson E, Mysorekar IU, Fremont DH, Doranz BJ, Diamond MS,

Crowe JE. 2016. Neutralizing human antibodies prevent Zika virus replication and fetal disease in mice. Nature 540:443-447.

16. Stettler K, Beltramello M, Espinosa DA, Graham V, Cassotta A, Bianchi S, Vanzetta F, Minola A, Jaconi S, Mele F, Foglierini M, Pedotti M, Simonelli L, Dowall S, Atkinson B, Percivalle E, Simmons CP, Varani L, Blum J, Baldanti F, Cameroni E, Hewson R, Harris E, Lanzavecchia A, Sallusto F, Corti D. 2016. Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection. Science 353 :823-6.

17. Swanstrom JA, Plante JA, Plante KS, Young EF, McGowan E, Gallichotte EN, Widman DG, Heise MT, de Silva AM, Baric RS. 2016. Dengue Virus Envelope Dimer Epitope Monoclonal Antibodies Isolated from Dengue Patients Are Protective against Zika Virus. MBio 7. 18. Wang Q, Yang H, Liu X, Dai L, Ma T, Qi J, Wong G, Peng R, Liu S, Li J, Li S, Song J, Liu J, He J, Yuan H, Xiong Y, Liao Y, Li J, Yang J, Tong Z, Griffin BD, Bi Y, Liang M, Xu X, Qin C, Cheng G, Zhang X, Wang P, Qiu X, Kobinger G, Shi Y, Yan J, Gao GF. 2016. Molecular determinants of human neutralizing antibodies isolated from a patient infected with Zika virus. Sci Transl Med 8:369ral79.

19. Zhao H, Fernandez E, Dowd KA, Speer SD, Piatt DJ, Gorman MJ, GoVero J, Nelson CA, Pierson TC, Diamond MS, Fremont DH. 2016. Structural Basis of Zika Virus-Specific Antibody Protection. Cell 166: 1016-27.

20. Abbink P, Larocca RA, De La Barrera RA, Bricault CA, Moseley ET, Boyd M, Kirilova M, Li Z, Ng'ang'a D, Nanayakkara O, Nityanandam R, Mercado NB, Borducchi EN, Agarwal A,

Brinkman AL, Cabral C, Chandrashekar A, Giglio PB, Jetton D, Jimenez J, Lee BC, Mojta S, Molloy K, Shetty M, Neubauer GH, Stephenson KE, Peron JP, Zanotto PM, Misamore J, Finneyfrock B, Lewis MG, Alter G, Modjarrad K, Jarman RG, Eckels KH, Michael NL, Thomas SJ, Barouch DH. 2016. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science 353 : 1129-32.

21. Larocca RA, Abbink P, Peron JP, Zanotto PM, Iampietro MJ, Badamchi-Zadeh A, Boyd M, Ng'ang'a D, Kirilova M, Nityanandam R, Mercado NB, Li Z, Moseley ET, Bricault CA, Borducchi EN, Giglio PB, Jetton D, Neubauer G, Nkolola JP, Maxfield LF, De La Barrera RA, Jarman RG, Eckels KH, Michael NL, Thomas SJ, Barouch DH. 2016. Vaccine protection against Zika virus from Brazil. Nature 536:474-8.

22. Dowd KA, Ko SY, Morabito KM, Yang ES, Pelc RS, DeMaso CR, Castilho LR, Abbink P, Boyd M, Nityanandam R, Gordon DN, Gallagher JR, Chen X, Todd JP, Tsybovsky Y, Harris A, Huang YS, Higgs S, Vanlandingham DL, Andersen H, Lewis MG, De La Barrera R, Eckels KH, Jarman RG, Nason MC, Barouch DH, Roederer M, Kong WP, Mascola JR, Pierson TC, Graham BS. 2016. Rapid development of a DNA vaccine for Zika virus. Science 354:237-240.

23. Kim E, Erdos G, Huang S, Kenniston T, Falo LD, Jr., Gambotto A. 2016. Preventative Vaccines for Zika Virus Outbreak: Preliminary Evaluation. EBioMedicine 13 :315-320.

24. Chahal JS, Fang T, Woodham AW, Khan OF, Ling J, Anderson DG, Ploegh HL. 2017. An RNA nanoparticle vaccine against Zika virus elicits antibody and CD8+ T cell responses in a mouse model. Sci Rep 7:252.

25. Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, Dowd KA, Sutherland LL, Scearce RM, Parks R, Wagner W, Granados A, Greenhouse J, Walker M, Willis E, Yu JS, McGee CE, Sempowski GD, Mui BL, Tarn YK, Huang YJ, Vanlandingham D, Holmes VM, Balachandran H, Sahu S, Lifton M, Higgs S, Hensley SE, Madden TD, Hope MJ, Kariko K, Santra S, Graham BS, Lewis MG, Pierson TC, Haynes BF, Weissman D. 2017. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature 543 :248- 251.

26. Richner JM, Himansu S, Dowd KA, Butler SL, Salazar V, Fox JM, Julander JG, Tang WW, Shresta S, Pierson TC, Ciaramella G, Diamond MS. 2017. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell 169: 176.

27. Boigard H, Alimova A, Martin GR, Katz A, Gottlieb P, Galarza JM. 2017. Zika virus- like particle (VLP) based vaccine. PLoS Negl Trop Dis 11 :e0005608.

28. Haddow AD, Schuh AJ, Yasuda CY, Kasper MR, Heang V, Huy R, Guzman H, Tesh RB, Weaver SC. 2012. Genetic characterization of Zika virus strains: geographic expansion of the Asian lineage. PLoS Negl Trop Dis 6:el477.

29. Dowd KA, DeMaso CR, Pelc RS, Speer SD, Smith AR, Goo L, Piatt DJ, Mascola JR, Graham BS, Mulligan MJ, Diamond MS, Ledgerwood JE, Pierson TC. 2016. Broadly

Neutralizing Activity of Zika Virus-Immune Sera Identifies a Single Viral Serotype. Cell Rep 16: 1485-91.

30. Maeda A, Maeda J. 2013. Review of diagnostic plaque reduction neutralization tests for flavivirus infection. Vet J 195:33-40. 31. Jeong H, Seong BL. 2017. Exploiting virus-like particles as innovative vaccines against emerging viral infections. J Microbiol 55:220-230.

32. Oliveira ER, Mohana-Borges R, de Alencastro RB, Horta BA. 2017. The flavivirus capsid protein: Structure, function and perspectives towards drug design. Virus Res 227: 115-123.

33. Gagnon SJ, Zeng W, Kurane I, Ennis FA. 1996. Identification of two epitopes on the dengue 4 virus capsid protein recognized by a serotype-specific and a panel of serotype-cross- reactive human CD4+ cytotoxic T-lymphocyte clones. J Virol 70: 141-7.

34. Lazo L, Hermida L, Zulueta A, Sanchez J, Lopez C, Silva R, Guillen G, Guzman MG. 2007. A recombinant capsid protein from Dengue-2 induces protection in mice against homologous virus. Vaccine 25: 1064-70. 35. Stocks CE, Lobigs M. 1998. Signal peptidase cleavage at the flavivirus C-prM junction: dependence on the viral NS2B-3 protease for efficient processing requires determinants in C, the signal peptide, and prM. J Virol 72:2141-9.

36. Henchal EA, Gentry MK, McCown JM, Brandt WE. 1982. Dengue virus-specific and flavivirus group determinants identified with monoclonal antibodies by indirect immunofluorescence. Am J Trop Med Hyg 31 :830-6.

37. Lazear HM, Diamond MS. 2016. Zika Virus: New Clinical Syndromes and Its Emergence in the Western Hemisphere. J Virol 90:4864-75.

38. Garg H, Lee RT, Tek NO, Maurer-Stroh S, Joshi A. 2013. Identification of conserved motifs in the West Nile virus envelope essential for particle secretion. BMC Microbiol 13 : 197.

39. Pierson TC, Sanchez MD, Puffer BA, Ahmed AA, Geiss BJ, Valentine LE, Altamura LA, Diamond MS, Doms RW. 2006. A rapid and quantitative assay for measuring antibody -mediated neutralization of West Nile virus infection. Virology 346:53-65.

40. Dowd KA, DeMaso CR, Pierson TC. 2015. Genotypic Differences in Dengue Virus Neutralization Are Explained by a Single Amino Acid Mutation That Modulates Virus

Breathing. MBio 6:e01559-15.

41. Hasan SS, Miller A, Sapparapu G, Fernandez E, Klose T, Long F, Fokine A, Porta JC, Jiang W, Diamond MS, Crowe JE, Jr., Kuhn RJ, Rossmann MG. 2017. A human antibody against Zika virus crosslinks the E protein to prevent infection. Nat Commun 8: 14722. 42. Garg H, Lee RT, Maurer-Stroh S, Joshi A. 2016. HIV-1 adaptation to low levels of CCR5 results in V3 and V2 loop changes that increase envelope pathogenicity, CCR5 affinity and decrease susceptibility to Maraviroc. Virology 493 :86-99.

43. Joshi A, Nyakeriga AM, Ravi R, Garg H. 2011. HIV ENV glycoprotein-mediated bystander apoptosis depends on expression of the CCR5 co-receptor at the cell surface and ENV fusogenic activity. J Biol Chem 286:36404-13.

44. Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, Bowen R, Bunning ML. 2001. West Nile virus recombinant DNA vaccine protects mouse and horse from virus challenge and expresses in vitro a noninfectious recombinant antigen that can be used in enzyme-linked immunosorbent assays. J Virol 75:4040-7. 45. Ferlenghi I, Clarke M, Ruttan T, Allison SL, Schalich J, Heinz FX, Harrison SC, Rey FA, Fuller SD. 2001. Molecular organization of a recombinant subviral particle from tick-borne encephalitis virus. Mol Cell 7:593-602.

46. Hunt AR, Cropp CB, Chang GJ. 2001. A recombinant particulate antigen of Japanese encephalitis virus produced in stably-transformed cells is an effective noninfectious antigen and subunit immunogen. J Virol Methods 97: 133-49.

47. Mason PW, Pincus S, Fournier MJ, Mason TL, Shope RE, Paoletti E. 1991. Japanese encephalitis virus-vaccinia recombinants produce particulate forms of the structural membrane proteins and induce high levels of protection against lethal JEV infection. Virology 180:294-305. 48. Pincus S, Mason PW, Konishi E, Fonseca BA, Shope RE, Rice CM, Paoletti E. 1992. Recombinant vaccinia virus producing the prM and E proteins of yellow fever virus protects mice from lethal yellow fever encephalitis. Virology 187:290-7.

49. Brien JD, Lazear HM, Diamond MS. 2013. Propagation, quantification, detection, and storage of West Nile virus. Curr Protoc Microbiol 31 : 15D 3 1-15D 3 18. 50. Lobigs M. 1993. Flavivirus premembrane protein cleavage and spike heterodimer secretion require the function of the viral proteinase NS3. Proc Natl Acad Sci U S A 90:6218-22.

51. Bera AK, Kuhn RJ, Smith JL. 2007. Functional characterization of cis and trans activity of the Flavivirus NS2B-NS3 protease. J Biol Chem 282: 12883-92.

52. Wilson JR, de Sessions PF, Leon MA, Scholle F. 2008. West Nile virus nonstructural protein 1 inhibits TLR3 signal transduction. J Virol 82:8262-71.

53. Chambers TJ, Hahn CS, Galler R, Rice CM. 1990. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol 44:649-88.

54. Kuno G, Chang GJ. 2007. Full-length sequencing and genomic characterization of Bagaza, Kedougou, and Zika viruses. Arch Virol 152:687-96. 55. Lindenbach BD, Rice CM. 2003. Molecular biology of flaviviruses. Adv Virus Res 59:23-61.

56. Gehrke R, Ecker M, Aberle SW, Allison SL, Heinz FX, Mandl CW. 2003. Incorporation of tick-borne encephalitis virus replicons into virus-like particles by a packaging cell line. J Virol 77:8924-33. 57. Lorenz IC, Kartenbeck J, Mezzacasa A, Allison SL, Heinz FX, Helenius A. 2003. Intracellular assembly and secretion of recombinant subviral particles from tick-borne encephalitis virus. J Virol 77:4370-82.

58. Pugachev KV, Mason PW, Shope RE, Frey TK. 1995. Double-subgenomic Sindbis virus recombinants expressing immunogenic proteins of Japanese encephalitis virus induce significant protection in mice against lethal JEV infection. Virology 212:587-94.

59. Noad R, Roy P. 2003. Virus-like particles as immunogens. Trends Microbiol 11 :438-44.

60. Pattenden LK, Middelberg AP, Niebert M, Lipin DI. 2005. Towards the preparative and large-scale precision manufacture of virus-like particles. Trends Biotechnol 23 :523-9. 61. Lo MK, Tilgner M, Shi PY. 2003. Potential high-throughput assay for screening inhibitors of West Nile virus replication. J Virol 77: 12901-6.

62. Shi PY, Tilgner M, Lo MK. 2002. Construction and characterization of subgenomic replicons of New York strain of West Nile virus. Virology 296:219-33.

63. Pijlman GP. 2015. Enveloped virus-like particles as vaccines against pathogenic arboviruses. Biotechnol J 10:659-70.

64. Hayes EB. 2009. Zika virus outside Africa. Emerg Infect Dis 15: 1347-50.

65. Yamshchikov VF, Compans RW. 1995. Formation of the flavivirus envelope: role of the viral NS2B-NS3 protease. J Virol 69: 1995-2003.

66. Khromykh AA, Varnavski AN, Westaway EG. 1998. Encapsidation of the flavivirus kunjin replicon RNA by using a complementation system providing Kunjin virus structural proteins in trans. J Virol 72:5967-77.

67. Duenas-Carrera S, Alvarez-Lajonchere L, Alvarez-Obregon JC, Herrera A, Lorenzo LJ, Pichardo D, Morales J. 2000. A truncated variant of the hepatitis C virus core induces a slow but potent immune response in mice following DNA immunization. Vaccine 19:992-7. 68. Xing YP, Huang ZH, Wang SX, Cai J, Li J, Chou TH, Lu S. 2005. Novel DNA vaccine based on hepatitis B virus core gene induces specific immune responses in Balb/c mice. World J Gastroenterol 11 :4583-6.

69. Aihara H, Takasaki T, Matsutani T, Suzuki R, Kurane I. 1998. Establishment and characterization of Japanese encephalitis virus-specific, human CD4(+) T-cell clones: flavivirus cross-reactivity, protein recognition, and cytotoxic activity. J Virol 72:8032-6.