Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Human antibodies to the dengue virus E-dimer epitope have therapeutic activity against Zika virus infection

An Author Correction to this article was published on 30 January 2020

Abstract

The Zika virus (ZIKV) epidemic has resulted in congenital abnormalities in fetuses and neonates. Although some cross-reactive dengue virus (DENV)-specific antibodies can enhance ZIKV infection in mice, those recognizing the DENV E-dimer epitope (EDE) can neutralize ZIKV infection in cell culture. We evaluated the therapeutic activity of human monoclonal antibodies to DENV EDE for their ability to control ZIKV infection in the brains, testes, placentas, and fetuses of mice. A single dose of the EDE1-B10 antibody given 3 d after ZIKV infection protected against lethality, reduced ZIKV levels in brains and testes, and preserved sperm counts. In pregnant mice, wild-type or engineered LALA variants of EDE1-B10, which cannot engage Fcg receptors, diminished ZIKV burden in maternal and fetal tissues, and protected against fetal demise. Because neutralizing antibodies to EDE have therapeutic potential against ZIKV, in addition to their established inhibitory effects against DENV, it may be possible to develop therapies that control disease caused by both viruses.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: EDE1-B10 is a human mAb to DENV that cross-neutralizes ZIKV infection.
Figure 2: EDE1-B10 protects against ZIKV-induced lethality and viral burden.
Figure 3: EDE1-B10 protects against testis infection and injury.
Figure 4: EDE1-B10 protects Ifnar1−/− pregnant dams.
Figure 5: Therapeutic effect of EDE1-B10 in WT pregnant dams.
Figure 6: Treatment with EDE1-B10 prevents maternal and fetal ZIKV infection after intravaginal inoculation of pregnant dams.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Weaver, S.C. et al. Zika virus: History, emergence, biology, and prospects for control. Antiviral Res. 130, 69–80 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Brasil, P. et al. Zika virus infection in pregnant women in Rio de Janeiro. N. Engl. J. Med. 375, 2321–2334 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Schaub, B. et al. Analysis of blood from Zika-virus-infected fetuses: a prospective case series. Lancet Infect. Dis. 3099, 26–28 (2017).

    Google Scholar 

  4. Honein, M.A. et al. Birth defects among fetuses and infants of US women with evidence of possible Zika virus infection during pregnancy. J. Am. Med. Assoc. 317, 59–68 (2017).

    Article  Google Scholar 

  5. Cao-Lormeau, V.-M. et al. Guillain–Barré Syndrome outbreak associated with Zika virus infection in French Polynesia: a case–control study. Lancet 387, 1531–1539 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Parra, B. et al. Guillain–Barré Syndrome associated with Zika virus infection in Colombia. N. Engl. J. Med. 375, 1513–1523 (2016).

    Article  PubMed  Google Scholar 

  7. Dai, L. et al. Structures of the Zika virus envelope protein and its complex with a flavivirus broadly protective antibody. Cell Host Microbe 19, 696–704 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Kuhn, R.J. et al. Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell 108, 717–725 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Dowd, K.A. et al. Broadly neutralizing activity of Zika-virus-immune sera identifies a single viral serotype. Cell Rep. 16, 1485–1491.

    Article  CAS  PubMed  Google Scholar 

  10. Barba-Spaeth, G. et al. Structural basis of potent Zika–dengue virus antibody cross-neutralization. Nature 536, 48–53 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Dejnirattisai, W. et al. A new class of highly potent, broadly neutralizing antibodies isolated from viremic patients infected with dengue virus. Nat. Immunol. 16, 170–177 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Fernandez, E. & Diamond, M.S. Vaccination strategies against Zika virus. Curr. Opin. Virol. 23, 59–67 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nelson, S. et al. Maturation of West Nile virus modulates sensitivity to antibody-mediated neutralization. PLoS Pathog. 4, e1000060 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Kam, Y.-W. et al. Cross-reactive dengue human monoclonal antibody prevents severe pathologies and death from Zika virus infections. JCI Insight 2, http://dx.doi.org/10.1172/jci.insight.92428 (2017).

  15. Halstead, S.B., Mahalingam, S., Marovich, M.A., Ubol, S. & Mosser, D.M. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. Lancet Infect. Dis. 10, 712–722 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dejnirattisai, W. et al. Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with Zika virus. Nat. Immunol. 17, 1102–1108 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Stettler, K. et al. Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection. Science 353, 823–826 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Zhao, H. et al. Structural basis of Zika-virus-specific antibody protection. Cell 166, 1016–1027 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sapparapu, G. et al. Neutralizing human antibodies prevent Zika virus replication and fetal disease in mice. Nature 540, 443–447 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Robbiani, D.F. et al. Recurrent potent human neutralizing antibodies to Zika virus in Brazil and Mexico. Cell 169, 597–609 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang, Q. et al. Molecular determinants of human neutralizing antibodies isolated from a patient infected with Zika virus. Sci. Transl. Med. 8, 369ra179 (2016).

    PubMed  Google Scholar 

  22. Swanstrom, J.A. et al. Dengue virus envelope dimer epitope monoclonal antibodies isolated from dengue patients are protective against Zika virus. MBio 7, e01123–e16 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hirsch, A.J. et al. Zika virus infection of rhesus macaques leads to viral persistence in multiple tissues. PLoS Pathog. 13, e1006219 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Miner, J.J. et al. Zika Virus infection in mice causes panuveitis with shedding of virus in tears. Cell Rep. 16, 3208–3218 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bhatnagar, J. et al. Zika virus RNA replication and persistence in brain and placental tissue. Emerg. Infect. Dis. 23, 405–414 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Aid, M. et al. Zika virus persistence in the central nervous system and lymph nodes of rhesus monkeys. Cell 169, 610–620 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Govero, J. et al. Zika virus infection damages the testes in mice. Nature 540, 438–442 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ma, W. et al. Zika virus causes testis damage and leads to male infertility in mice. Cell 167, 1511–1524 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Yockey, L.J. et al. Vaginal exposure to Zika virus during pregnancy leads to fetal brain infection. Cell 166, 1247–1256 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rouvinski, A. et al. Covalently linked dengue virus envelope glycoprotein dimers reduce exposure of the immunodominant fusion loop epitope. Nat. Commun. 8, 15411 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pierson, T.C. & Diamond, M.S. A game of numbers: the stoichiometry of antibody-mediated neutralization of flavivirus infection. Prog. Mol. Biol. Transl. Sci. 129, 141–166 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Hessell, A.J. et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449, 101–104 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Magdelaine-Beuzelin, C., Pinault, C., Paintaud, G. & Watier, H. Therapeutic antibodies in ophthalmology: old is new again. MAbs 2, 176–180 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Miner, J.J. et al. Zika virus infection during pregnancy in mice causes placental damage and fetal demise. Cell 165, 1081–1091 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cugola, F.R. et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 534, 267–271 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Foy, B.D. et al. Probable non-vector-borne transmission of Zika virus, Colorado, USA. Emerg. Infect. Dis. 17, 880–882 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Musso, D. et al. Potential sexual transmission of Zika virus. Emerg. Infect. Dis. 21, 359–361 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Russell, K. et al. Male-to-female sexual transmission of Zika virus—United States, January–April 2016. Clin. Infect. Dis. 64, 211–213 (2017).

    Article  PubMed  Google Scholar 

  39. Davidson, A., Slavinski, S., Komoto, K., Rakeman, J. & Weiss, D. Suspected female-to-male sexual transmission of Zika virus—New York City, 2016. MMWR Morb. Mortal. Wkly. Rep. 65, 716–717 (2016).

    Article  PubMed  Google Scholar 

  40. Deckard, D.T. et al. Male-to-male sexual transmission of Zika virus—Texas, January 2016. MMWR Morb. Mortal. Wkly. Rep. 65, 372–374 (2016).

    Article  PubMed  Google Scholar 

  41. Barzon, L. et al. Infection dynamics in a traveller with persistent shedding of Zika virus RNA in semen for six months after returning from Haiti to Italy, January 2016. Euro Surveill. 21, 30316 (2016).

    PubMed Central  Google Scholar 

  42. Richner, J.M. et al. Vaccine-mediated protection against Zika-virus-induced congenital disease. Cell 170, 273–283 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sukupolvi-Petty, S. et al. Functional analysis of antibodies against dengue virus type 4 reveals strain-dependent epitope exposure that impacts neutralization and protection. J. Virol. 87, 8826–8842 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mansuy, J.M. et al. Zika virus: high infectious viral load in semen, a new sexually transmitted pathogen? Lancet Infect. Dis. 16, 405 (2016).

    Article  PubMed  Google Scholar 

  45. Murray, K.O. et al. Prolonged detection of Zika virus in vaginal secretions and whole blood. Emerg. Infect. Dis. 23, 99–101 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zeitlin, L. et al. Enhanced potency of a fucose-free monoclonal antibody being developed as an Ebola virus immunoprotectant. Proc. Natl. Acad. Sci. USA 108, 20690–20694 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. DiLillo, D.J., Tan, G.S., Palese, P. & Ravetch, J.V. Broadly neutralizing hemagglutinin stalk-specific antibodies require FcγR interactions for protection against influenza virus in vivo. Nat. Med. 20, 143–151 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hiatt, A. et al. Glycan variants of a respiratory syncytial virus antibody with enhanced effector function and in vivo efficacy. Proc. Natl. Acad. Sci. USA 111, 5992–5997 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Carroll, T. et al. Zika virus preferentially replicates in the female reproductive tract after vaginal inoculation of rhesus macaques. PLoS Pathog. 13, e1006537 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Halstead, S.B. Biologic evidence required for Zika disease enhancement by dengue antibodies. Emerg. Infect. Dis. 23, 569–573 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Brien, J.D., Lazear, H.M. & Diamond, M.S. Propagation, quantification, detection, and storage of West Nile virus. Curr. Protoc. Microbiol. 31, 15D.3.1–15D.3.18 (2013).

    Article  Google Scholar 

  52. National Research Council. Guide for the Care and Use of Laboratory Animals 8th edn. (The National Academies Press, 2011).

  53. Lanciotti, R.S. et al. Genetic and serologic properties of Zika virus associated with an epidemic, Yap State, Micronesia, 2007. Emerg. Infect. Dis. 14, 1232–1239 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hansen, D.A., Esakky, P., Drury, A., Lamb, L. & Moley, K.H. The aryl hydrocarbon receptor is important for proper seminiferous tubule architecture and sperm development in mice. Biol. Reprod. 90, 8 (2014).

    Article  PubMed  CAS  Google Scholar 

  55. Goodson, S.G., Zhang, Z., Tsuruta, J.K., Wang, W. & O'Brien, D.A. Classification of mouse sperm motility patterns using an automated multiclass support vector machines model. Biol. Reprod. 84, 1207–1215 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Haina Shin for advice on the intravaginal infection experiments, S. Whitehead (US National Institutes of Health (NIH)) for ZIKV-Brazil (Paraiba, 2015), S. Shresta(La Jolla Institute of Allergy and Immunology) for DENV-2 strain D2S20, P. Malasit and S. Noisakran (Mahidol University) for DENV-1, DENV-3, and DENV-4 isolates from patients, C. Simmons (University of Melbourne) for DENV-2 strain DF-699, A. Kohl, R.F. de Oliveira Freitas, and L.J. Pena (University of Glasgow) for ZIKV-Brazil PE243, and A. Sakuntabhai (Institut Pasteur) for ZIKV-Africa HD78788. Supported by grants and contracts from the NIH (R01 AI073755 (M.S.D.), R01 AI127828 (M.S.D.), R01 HD091218 (I.U.M. and M.S.D.), HHSN272201400018C (M.S.D.), T32 AI007163 (E.F.)), the Wellcome Trust (G.R.S.), MRC-NEWTON UK (J.M.), and the National Institute for Health Research Biomedical Research Centre funding scheme UK (G.R.S.).

Author information

Authors and Affiliations

Authors

Contributions

E.F., J.M., W.D., S.M.S., B.C., G.R.S., and M.S.D. designed the experiments; E.F., W.D., S.M.S., B.C., P.S., and W.W. performed the experiments; E.F., J.M., W.D., S.M.S., B.C., P.E., A.D., I.U.M., K.H.M., G.R.S., and M.S.D. analyzed the data; E.F. and M.S.D. wrote the first draft of the paper; and all authors participated in editing the final version of the manuscript.

Corresponding authors

Correspondence to Gavin R Screaton or Michael S Diamond.

Ethics declarations

Competing interests

M.S.D. is a consultant for Inbios, Visterra, Aviana and Sanofi-Pasteur and is on the Scientific Advisory Boards of Moderna and OvaGene. G.R.S. is on the Vaccine Scientific Advisory Board of GlaxoSmithKline plc.

Integrated supplementary information

Supplementary Figure 1 EDE-specific mAbs protect against ZIKV-induced lethality.

Four to five week-old WT male mice were treated with anti-Ifnar1 mAb followed by subcutaneous infection with 103 FFU of mouse-adapted ZIKV-Dakar. Mice then were treated with isotype-control, EDE1-C8, or EDE2-A11 mAbs at day +1 (100 μg, left) or day +3 (250 μg, right). Data were pooled from two (isotype-control mAb) or three (EDE1-C8 and EDE2-A11) independent experiments (isotype-control mAb, n = 19; EDE1-C8, n = 10; EDE2-A11, n = 10). Statistical significance was analyzed (log-rank test: ****, P < 0.0001).

Supplementary Figure 2 Levels of EDE1-B10 mAb in tissues at day +5 after infection.

Eight to nine week old WT male mice were treated with a single dose of EDE1-B10 mAb at day +1 or +3 as described in Fig 2. a. At D+5, tissues were harvested and EDE1-B10 levels were assessed by ELISA using a standard curve. Bars indicate median values. Data were pooled from two independent experiments, and symbols correspond to individual mice (n = 8 per group). Statistical analysis was determined (Mann-Whitney test: **, P < 0.01; ***, P < 0.001).

Supplementary Figure 3 ISH and histological analysis of epididymis from mice treated with EDE1-B10.

Eight- to nine- week old male WT mice were treated with isotype control or EDE1-B10 mAb at day +1 (n=6 mice), day +3 (n=4 mice), day +5 (n=8 mice) after infection, as described in Figure 2. a. RNA in situ hybridization (ISH) staining of epididymis at day +21 using ZIKV-specific RNA probes. Low power (scale bar = 500 μm) and high power (scale bar = 20 μm) images are presented in sequence. The images in the panels are representative of sections from 4 to 6 mice. b. H & E staining of epididymis. Low power (scale bar = 500 μm) and high power (scale bar = 20 μm) images are shown in sequence. The images are representative of sections from 3 to 5 mice.

Supplementary Figure 4 Protection of pregnant mice with WT and LALA EDE1-C8 mAbs.

WT female mice were mated with WT sires. At E5.5, dams were treated with anti-Ifnar1 mAb. At E6.5, dams were infected subcutaneously with 103 FFU of mouse-adapted ZIKV-Dakar. At E7.5 (day +1), dams were treated with 250 μg of either isotype-control mAb or EDE1-C8 (wild-type or LALA variant). At E13.5, placentas and fetal heads were harvested, and viral RNA was assessed by qRT-PCR. Bars indicate median values. Data were pooled from two independent experiments, and symbols correspond to individual mice (isotype mAb, n = 16; EDE1-C8, n = 20; EDE1-C8 LALA, n = 12). Statistical significance was determined (Kruskal-Wallis test: ***, P < 0.001; ****, P < 0.0001). Dashed line indicates the limit of detection for the assay.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fernandez, E., Dejnirattisai, W., Cao, B. et al. Human antibodies to the dengue virus E-dimer epitope have therapeutic activity against Zika virus infection. Nat Immunol 18, 1261–1269 (2017). https://doi.org/10.1038/ni.3849

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3849

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing