G. Kuno, G. J. Chang, K. R. Tsuchiya, N. Karabatsos, and C. B. Cropp, Phylogeny of the genus Flavivirus, J. Virol, vol.72, pp.73-83, 1998.

G. W. Dick, S. F. Kitchen, and A. J. Haddow, Zika virus. I. Isolations and serological specificity, Trans. R. Soc. Trop. Med. Hyg, vol.46, pp.509-520, 1952.

M. R. Duffy, T. Chen, T. Hancock, A. M. Powers, J. L. Kool et al., Zika virus outbreak on Yap Island, Federated States of Micronesia, N. Engl. J. Med, vol.360, pp.2536-2543, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00734543

R. S. Lanciotti, O. L. Kosoy, J. J. Laven, J. O. Velez, A. J. Lambert et al., Genetic and serologic properties of Zika virus associated with an epidemic, Yap State, Micronesia, Emerg. Infect. Dis, vol.14, pp.1232-1239, 2007.

V. M. Cao-lormeau, C. Roche, A. Teissier, E. Robin, A. Berry et al., Zika virus, French polynesia, South pacific, Emerg. Infect. Dis, vol.20, pp.1085-1086, 2013.

G. S. Campos, A. C. Bandeira, and S. I. Sardi, Zika virus outbreak, Brazil. Emerg. Infect. Dis, vol.21, pp.1885-1886, 2015.

M. ;. Dupont-rouzeyrol, O. O'connor, E. Calvez, M. Daurès, M. John et al., Co-infection with Zika and dengue viruses in 2 patients, Emerg. Infect. Dis, vol.21, pp.381-382, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01114780

C. Zanluca, V. C. De-melo, A. L. Mosimann, G. I. Santos, C. N. Santos et al., First report of autochthonous transmission of Zika virus in Brazil, Mem. Inst. Oswaldo Cruz, vol.110, pp.569-572, 2015.

O. Pacheco, M. Beltrán, C. A. Nelson, D. Valencia, N. Tolosa et al., Zika virus disease in Colombia-Preliminary report, N. Engl. J. Med, 2016.

E. Oehler, L. Watrin, P. Larre, I. Leparc-goffart, S. Lastère et al., Zika virus infection complicated by Guillain-Barre syndrome-Case report, 2013.

V. M. Cao-lormeau, A. Blake, S. Mons, S. Lastère, C. Roche et al., Guillain-Barre Syndrome outbreak associated with Zika virus infection in French Polynesia: A case-control study, Lancet, vol.387, pp.1531-1539, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01453407

S. Cauchemez, M. Besnard, P. Bompard, T. Dub, P. Guillemette-artur et al., Association between Zika virus and microcephaly in French Polynesia, A retrospective study, vol.387, pp.2125-2132, 2016.

J. S. Soares-de-araujo, C. Teixeira-regis, R. G. Silva-gomes, T. Tavares, . Rocha et al., Microcephaly in north-east Brazil: A retrospective study on neonates born between, Bull. World Health Organ, vol.94, pp.835-840, 2012.

A. Enfissi, J. Codrington, J. Roosblad, M. Kazanji, and D. Rousset, Zika virus genome from the Americas, Lancet, vol.387, pp.227-228, 2016.

D. Musso, D. J. Gubler, . Zika, and . Virus, Clin. Microbiol. Rev, vol.29, pp.487-524, 2016.

Y. Liu, J. Liu, S. Du, C. Shan, K. Nie et al., Evolutionary enhancement of Zika virus infectivity in Aedes aegypti mosquitoes, Nature, vol.545, pp.482-486, 2017.

H. C. Metsky, C. B. Matranga, S. Wohl, S. F. Schaffner, C. A. Freije et al., Zika virus evolution and spread in the Americas, Nature, vol.546, pp.411-415, 2017.

L. Yuan, X. Huang, Z. Liu, F. Zhang, X. Zhu et al., A single mutation in the prM protein of Zika virus contributes to fetal microcephaly, vol.358, pp.933-936, 2017.

L. C. Katzelnick, J. Coloma, E. Harris, and . Dengue, Knowledge gaps, unmet needs, and research priorities, Lancet Infect. Dis, vol.17, pp.88-100, 2017.

G. Barba-spaeth, W. Dejnirattisai, A. Rouvniski, M. Vaney, I. Medits et al., Structural basis of potent Zika-dengue virus antibody cross-neutralization, Nature, vol.536, pp.48-53, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01408100

D. Sirohi, Z. Chen, L. Sun, T. Klose, T. C. Pierson et al., The 3.8 A resolution cryo-EM structure of Zika virus, Science, vol.352, pp.467-470, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01439629

X. Xu, H. Song, J. Qi, Y. Liu, H. Wang et al., Contribution of intertwined loop to membrane association revealed by Zika virus full-length NS1 structure, EMBO J, vol.35, pp.2170-2178, 2016.

L. Priyamvada, K. M. Quicke, W. H. Hudson, N. Onlamoon, J. Sewatanon et al., Human antibody responses after dengue virus infection are highly cross-reactive to Zika virus, Proc. Natl. Acad. Sci, vol.113, pp.7852-7857, 2016.

K. Stettler, M. Beltramello, D. A. Espinosa, V. Graham, A. Cassotta et al., cross-reactivity, and function of antibodies elicited by Zika virus infection, Science, vol.353, pp.823-826, 2016.

D. V. Andrade and E. Harris, Recent advances in understanding the adaptive immune response to Zika virus and the effect of previous flavivirus exposure, Virus Res, 2017.

S. V. Bardina, P. Bunduc, S. Tripathi, J. Duehr, J. J. Frere et al., Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity, vol.356, pp.175-180, 2017.

T. F. Rogers, E. C. Goodwin, B. Briney, D. Sok, N. Beutler et al., Zika virus activates de novo and cross-reactive memory B cell responses in dengue-experienced donors

S. B. Halstead, Dengue antibody-dependent enhancement: Knowns and unknowns. Microbiol. Spectr. 2014, 2

L. Priyamvada, W. Hudson, R. Ahmed, and J. Wrammert, Humoral cross-reactivity between Zika and dengue viruses: Implications for protection and pathology, Emerg. Microbes Infect, vol.6, 2017.

X. Xu, K. Vaughan, D. Weiskopf, A. Grifoni, M. S. Diamond et al., Identifying Candidate Targets of Immune Responses in Zika Virus Based on Homology to Epitopes in Other Flavivirus Species, PLoS Curr, vol.8, 2016.

S. J. Thomas, A. Nisalak, K. B. Anderson, D. H. Libraty, S. Kalayanarooj et al., Dengue plaque reduction neutralization test (PRNT) in primary and secondary dengue virus infections: How alterations in assay conditions impact performance, Am. J. Trop. Med. Hyg, vol.81, pp.825-833, 2009.

R. De-alwis and A. M. Silva, Measuring antibody neutralization of dengue virus (DENV) using a flow cytometry-based technique, Methods Mol. Biol, vol.1138, pp.27-39, 2014.

E. P. Calvo, F. Sanchez-quete, S. Duran, I. Sandoval, and J. E. Castellanos, Easy and inexpensive molecular detection of dengue, chikungunya and zika viruses in febrile patients, Acta Trop, vol.163, pp.32-37, 2016.

M. Aubry, A. Teissier, M. Huart, S. Merceron, J. Vanhomwegen et al., Zika virus seroprevalence, Emerg. Inf. Dis, vol.23, pp.669-672, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01521256

R. De-alwis, S. A. Smith, N. P. Olivarez, W. B. Messer, J. P. Huynh et al., Identification of human neutralizing antibodies that bind to complex epitopes on dengue virions, Proc. Natl. Acad. Sci, vol.109, pp.7439-7444, 2012.

S. B. Halstead, Neutralization and antibody-dependent enhancement of dengue viruses, Adv. Virus Res, vol.60, pp.421-467, 2003.

B. Patel, P. Longo, M. J. Miley, M. Montoya, E. Harris et al., Dissecting the human serum antibody response to secondary dengue virus infections

S. Olkowski, B. M. Forshey, A. C. Morrison, C. Rocha, S. Vilcarromero et al., Reduced risk of disease during postsecondary dengue virus infections, J. Infect. Dis, vol.208, pp.1026-1033, 2013.

M. Beltramello, K. L. Williams, C. P. Simmons, A. Macagno, L. Simonelli et al., The human immune response to dengue virus is dominated by highly cross-reactive antibodies endowed with neutralizing and enhancing activity, Cell Host Microbe, vol.8, pp.271-283, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-00530282

W. Dejnirattisai, A. Jumnainsong, N. Onsirisakul, P. Fitton, S. Vasanawathana et al., Cross-reacting antibodies enhance dengue virus infection in humans, Science, vol.328, pp.745-748, 2010.

S. A. Smith, Y. Zhou, N. P. Olivarez, A. H. Broadwater, A. M. Silva et al., Persistence of circulating memory B cell clones with potential for dengue virus disease enhancement for decades following infection, J. Virol, vol.86, pp.2665-2675, 2012.

W. Dejnirattisai, P. Supasa, W. Wongwiwat, A. Rouvinski, G. Barba-spaeth et al., Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus, Nat. Immunol, vol.17, pp.1102-1108, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-02003950

L. Priyamvada, A. Cho, N. Onlamoon, N. Zheng, M. Huang et al., B cell responses during secondary dengue virus infection are dominated by highly cross-reactive, memory-derived plasmablasts, J. Virol, vol.90, pp.5574-5585, 2016.

D. F. Robbiani, L. Bozzacco, J. R. Keeffe, R. Khouri, P. C. Olsen et al., Recurrent potent human neutralizing antibodies to Zika virus in Brazil and Mexico, vol.169, pp.597-609, 2017.

M. H. Collins, E. Mcgowan, R. Jadi, E. Young, C. A. Lopez et al., Lack of durable cross-neutralizing antibodies against Zika virus from dengue virus infection, Emerg. Infect. Dis, vol.23, pp.773-781, 2017.

A. Puschnik, L. Lau, E. A. Cromwell, A. Balmaseda, S. Zompi et al., Correlation between dengue-specific neutralizing antibodies and serum avidity in primary and secondary dengue virus 3 natural infections in humans

K. S. Corbett, L. Katzelnick, H. Tissera, A. Amerasinghe, A. Dharshan-de-silva et al., Preexisting neutralizing antibody responses distinguish clinically inapparent and apparent dengue virus infections in a Sri Lankan pediatric cohort, J. Infect. Dis, vol.211, pp.590-599, 2015.

H. E. Clapham, I. Rodriguez-barraquer, A. S. Azman, B. M. Althouse, H. Salje et al., Dengue virus (DENV) neutralizing antibody kinetics in children after symptomatic primary and postprimary DENV infection, J. Infect. Dis, vol.213, pp.1428-1435, 2016.

L. C. Katzelnick, M. Montoya, L. Gresh, A. Balmaseda, and E. Harris, Neutralizing antibody titers against dengue virus correlate with protection from symptomatic infection in a longitudinal cohort, Proc. Natl. Acad. Sci, vol.113, pp.728-733, 2016.

L. C. Katzelnick, L. Gresh, M. E. Halloran, J. C. Mercado, G. Kuan et al., Antibody-dependent enhancement of severe dengue disease in humans, vol.358, pp.929-932, 2017.

A. B. Sabin, Research on dengue during World War II, Am. J. Trop. Med. Hyg, vol.1, pp.30-50, 1952.

S. B. Halstead, J. Casals, H. Shotwell, and N. Palumbo, Studies on the immunization of monkeys against dengue. I. Protection derived from single and sequential virus infections, Am. J. Trop. Med. Hyg, vol.22, pp.365-374, 1973.

B. M. Forshey, R. C. Reiner, S. Olkowski, A. C. Morrison, A. Espinoza et al., Incomplete protection against dengue virus type 2 re-infection in Peru, PLoS Neglect. Trop. Dis, vol.10, 2016.

J. J. Waggoner, A. Balmaseda, L. Gresh, M. K. Sahoo, M. Montoya et al., Homotypic dengue virus reinfections in Nicaraguan children, J. Infect. Dis, vol.214, pp.986-993, 2016.

A. Culshaw, J. Mongkolsapaya, and G. R. Screaton, The immunopathology of dengue and Zika virus infections, Curr. Opin. Immunol, vol.48, pp.1-6, 2017.

L. Rivino, T cell immunity to dengue virus and implications for vaccine design, Expert Rev. Vaccines, vol.15, pp.443-453, 2016.

L. C. Katzelnick and E. Harris, Participants in the Summit on Dengue Immune Correlates of P Immune correlates of protection for dengue: State of the art and research agenda, vol.35, pp.4659-4669, 2017.

J. Mongkolsapaya, W. Dejnirattisai, X. Xu, S. Vasanawathana, N. Tangthawornchaikul et al., Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever, Nat. Med, vol.9, pp.921-927, 2003.

P. P. Yachi, J. Ampudia, T. Zal, and N. R. Gascoigne, Altered peptide ligands induce delayed CD8-T cell receptor interaction-A role for CD8 in distinguishing antigen quality, Immunity, vol.25, pp.203-211, 2006.

T. Duangchinda, W. Dejnirattisai, S. Vasanawathana, W. Limpitikul, N. Tangthawornchaikul et al., Immunodominant T-cell responses to dengue virus NS3 are associated with DHF, Proc. Natl. Acad. Sci, vol.107, pp.16922-16927, 2010.

H. S. Bashyam, S. Green, and A. L. Rothman, Dengue virus-reactive CD8 + T cells display quantitative and qualitative differences in their response to variant epitopes of heterologous viral serotypes, J. Immunol, vol.176, pp.2817-2824, 2006.

M. M. Mangada and A. L. Rothman, Altered cytokine responses of dengue-specific CD4 + T cells to heterologous serotypes, J. Immunol, vol.175, pp.2676-2683, 2005.

J. Mongkolsapaya, T. Duangchinda, W. Dejnirattisai, S. Vasanawathana, P. Avirutnan et al., T cell responses in dengue hemorrhagic fever: Are cross-reactive T cells suboptimal?, J. Immunol, vol.176, pp.3821-3829, 2006.

J. Zivny, M. Defronzo, W. Jarry, J. Jameson, J. Cruz et al., Partial agonist effect influences the CTL response to a heterologous dengue virus serotype, J. Immunol, vol.163, pp.2754-2760, 1999.

D. Weiskopf, M. A. Angelo, D. J. Bangs, J. Sidney, S. Paul et al., The human CD8 + T cell responses induced by a live attenuated tetravalent dengue vaccine are directed against highly conserved epitopes, J. Virol, vol.89, pp.120-128, 2015.

D. Weiskopf, C. Cerpas, M. A. Angelo, D. J. Bangs, J. Sidney et al., Human CD8 + T-cell responses against the 4 dengue virus serotypes are associated with distinct patterns of protein targets, J. Infect. Dis, vol.212, pp.1743-1751, 2015.

S. Hatch, T. P. Endy, S. Thomas, A. Mathew, J. Potts et al., Intracellular cytokine production by dengue virus-specific T cells correlates with subclinical secondary infection, J. Infect. Dis, vol.203, pp.1282-1291, 2011.

H. Loke, D. B. Bethell, C. X. Phuong, M. Dung, J. Scheinder et al., HLA class I-restricted T cell responses in dengue hemorrhagic fever: A double-edged sword?, J. Infect. Dis, vol.184, pp.1369-1373, 2001.

H. A. Stephens, R. Klaythong, M. Sirikong, D. W. Vaughn, S. Green et al., HLA-A and-B allele associations with secondary dengue virus infections correlate with disease severity and the infecting viral serotype in ethnic Thais, Tissue Antigens, vol.60, pp.309-318, 2002.

B. Sierra, R. Alegre, A. B. Pérez, G. García, K. Sturn-ramirez et al., and-DRB1 allele frequencies in Cuban individuals with antecedents of dengue 2 disease: Advantages of the Cuban population for HLA studies of dengue virus infection, Hum. Immunol, vol.68, pp.531-540, 2007.

T. P. Nguyen, M. Kikuchi, V. T. Huong, D. Q. Ha, T. T. Thuy et al., Protective and enhancing HLA alleles, HLA-DRB1*0901 and HLA-A*24, for severe forms of dengue virus infection, dengue hemorrhagic fever and dengue shock syndrome

L. L. Coffey, E. Mertens, A. Brehin, M. D. Fernandez-garcia, A. Amara et al., Human genetic determinants of dengue virus susceptibility, vol.11, pp.143-156, 2009.

G. N. Malavige, T. Rostron, L. T. Rohanachandra, N. Fernando, A. Dharshan-de-silva et al., HLA class I and class II associations in dengue viral infections in a Sri Lankan population, PLoS ONE, vol.6, 2011.

L. E. Yauch, R. M. Zellweger, M. F. Kotturi, A. Qutubuddin, J. Sidney et al., A protective role for dengue virus-specific CD8 + T cells, J. Immunol, vol.182, pp.4865-4873, 2009.

D. Weiskopf, M. A. Angelo, E. L. De-azeredo, J. Sidney, J. A. Greenbaum et al., Comprehensive analysis of dengue virus-specific responses supports an HLA-linked protective role for CD8 + T cells, Proc. Natl. Acad. Sci, vol.110, 2013.

E. Simon-lorière, V. Duong, A. Tawfik, S. Ung, S. Ly et al., Increased adaptive immune responses and proper feedback regulation protect against clinical dengue, Sci. Transl. Med, vol.9, 2017.

P. G. Livingston, I. Kurane, L. C. Dai, Y. Okamoto, C. J. Lai et al., Dengue virus-specific, HLA-B35-restricted, human CD8 + cytotoxic T lymphocyte (CTL) clones. Recognition of NS3 amino acids 500 to 508 by CTL clones of two different serotype specificities, J. Immunol, vol.154, pp.1287-1295, 1995.

I. Kurane, L. Zeng, M. A. Brinton, and F. A. Ennis, Definition of an epitope on NS3 recognized by human CD4 + cytotoxic T lymphocyte clones cross-reactive for dengue virus types 2, 3, and 4, Virology, vol.240, pp.169-174, 1998.

L. Rivino, E. A. Kumaran, V. Jovanovic, K. Nadua, E. W. Teo et al., Differential targeting of viral components by CD4 + versus CD8 + T lymphocytes in dengue virus infection, J. Virol, vol.87, pp.2693-2706, 2013.

L. Rivino, A. T. Tan, A. Chia, E. A. Kumaran, G. M. Grotenbreg et al., Defining CD8 + T cell determinants during human viral infection in populations of Asian ethnicity, J. Immunol, vol.191, pp.4010-4019, 2013.

D. Weiskopf, D. J. Bangs, J. Sidney, R. V. Kolla, A. D. Silva et al., Dengue virus infection elicits highly polarized CX3CR1 + cytotoxic CD4 + T cells associated with protective immunity, Proc. Natl. Acad. Sci, vol.112, 2015.

L. Zeng, I. Kurane, Y. Okamoto, F. A. Ennis, and M. A. Brinton, Identification of amino acids involved in recognition by dengue virus NS3-specific, HLA-DR15-restricted cytotoxic CD4 + T-cell clones, J. Virol, vol.70, pp.3108-3117, 1996.

Y. Okamoto, I. Kurane, A. M. Leporati, and F. A. Ennis, Definition of the region on NS3 which contains multiple epitopes recognized by dengue virus serotype-cross-reactive and flavivirus-cross-reactive, HLA-DPw2-restricted CD4 + T cell clones, J. Gen. Virol, vol.79, pp.697-704, 1998.

C. P. Simmons, T. Dong, N. V. Chau, N. T. Dung, T. N. Chau et al., Early T-cell responses to dengue virus epitopes in Vietnamese adults with secondary dengue virus infections, J. Virol, vol.79, pp.5665-5675, 2005.

E. J. Nascimento, R. B. Mailliard, A. M. Khan, J. Sidney, A. Sette et al., Identification of conserved and HLA promiscuous DENV3 T-cell epitopes

A. Alam, S. Ali, S. Ahamad, M. Z. Malik, and R. Ishrat, From ZikV genome to vaccine: In silico approach for the epitope-based peptide vaccine against Zika virus envelope glycoprotein, Immunology, vol.149, pp.386-399, 2016.

H. Dar, T. Zaheer, M. Talha-rehman, A. Ali, A. Javed et al., Prediction of promiscuous T-cell epitopes in the Zika virus polyprotein: An in silico approach. Asian Pac, J. Trop. Med, vol.9, pp.844-850, 2016.

U. Mirza, M. Rafique, S. Ali, A. Munir, M. Ikram et al., Towards peptide vaccines against Zika virus: Immunoinformatics combined with molecular dynamics simulations to predict antigenic epitopes of Zika viral proteins, Sci. Rep, vol.6, 2016.

L. Rivino and M. Q. Lim, CD4 + and CD8 + T-cell immunity to Dengue-Lessons for the study of Zika virus, Immunology, vol.150, pp.146-154, 2016.

J. Wen, W. Tang, N. Sheets, J. Ellison, A. Sette et al., Identification of Zika virus epitopes reveals immunodominant and protective roles for dengue virus cross-reactive CD8 + T cells, Nat. Microbiol, 2017.

C. J. Reynolds, O. M. Sulyeman, A. M. Ortega-prieto, J. K. Skelton, P. Bonnesoeur et al., T cell immunity to Zika virus targets immunodominant epitopes that show cross-reactivity with other Flaviviruses

R. Zust, Y. Toh, I. Valdés, D. Cerny, J. Heinrich et al., Type I Interferon signals in macrophages and dendritic cells control dengue virus infection: Implications for a new mouse model to test dengue vaccines, J. Virol, vol.88, pp.7276-7285, 2014.

A. Elong-ngono, E. A. Vizcarra, W. W. Tang, N. Sheets, Y. Joo et al., Mapping and role of the CD8 + T cell response during primary Zika virus infection in mice, Cell Host Microbe, vol.21, pp.35-46, 2017.

J. Wen, A. Elong-ngono, J. A. Regla-nava, K. Kim, M. J. Gorman et al., Dengue virus-reactive CD8 + T cells mediate cross-protection against subsequent Zika virus challenge, Nat. Commun, vol.8, 1459.

R. A. Larocca, P. Abbink, J. P. Peron, P. M. Zanotto, M. J. Iampietro et al., Vaccine protection against Zika virus from Brazil, Nature, vol.536, pp.474-478, 2016.

P. Abbink, R. A. Larocca, R. A. Barrera, C. A. Bricault, E. T. Moseley et al., Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys, Science, vol.353, pp.1129-1132, 2016.

N. S. Lima, M. Rolland, K. Modjarrad, and L. Trautmann, Cell immunity and Zika virus vaccine development, Trends Immunol, vol.38, pp.594-605, 2017.

H. Huang, S. Li, Y. Zhang, X. Han, B. Jia et al., CD8 + T cell immune response in immunocompetent mice during Zika virus infection, J. Virol, p.91, 2017.

R. D. Pardy, M. M. Rajah, S. A. Condotta, N. G. Taylor, S. M. Sagan et al., Analysis of the T cell response to Zika virus and identification of a novel CD8 + T cell epitope in immunocompetent mice, PLoS Pathog, vol.13, 2017.

C. W. Winkler, L. M. Myers, T. A. Woods, R. J. Messer, A. B. Carmody et al., Adaptive immune responses to Zika virus are important for controlling virus infection and preventing infection in brain and testes, J. Immunol, vol.198, pp.3526-3535, 2017.

M. M. Naiyer, S. A. Cassidy, A. Magri, V. Cowton, K. Chen et al.,

A. Grifoni, J. Pham, J. ;. Sidney, P. O'rourke, S. Paul et al., Prior Dengue virus exposure shapes T cell immunity to Zika virus in humans, J. Virol, vol.91, 2017.

S. B. Halstead, Which Dengue Vaccine Approach Is the Most Promising, and Should We Be Concerned about Enhanced Disease after Vaccination? There Is Only One True Winner, C.S.H. Perspect. Biol, 2017.

R. M. Zellweger, W. E. Eddy, W. W. Tang, R. Miller, and S. Shresta, CD8 + T cells prevent antigen-induced antibody-dependent enhancement of dengue disease in mice, J. Immunol, vol.193, pp.4117-4124, 2014.

R. M. Zellweger, W. W. Tang, W. E. Eddy, K. King, M. C. Sanchez et al., CD8 + T cells can mediate short-term protection against heterotypic dengue virus reinfection in mice, J. Virol, vol.89, pp.6494-6505, 2015.