A Chimeric Japanese Encephalitis Vaccine Protects against Lethal Yellow Fever Virus Infection without Inducing Neutralizing Antibodies

Niraj Mishra, Robbert Boudewijns, Michael Alexander Schmid, Rafael Elias Marques, Sapna Sharma, Johan Neyts, Kai Dallmeier, Niraj Mishra, Robbert Boudewijns, Michael Alexander Schmid, Rafael Elias Marques, Sapna Sharma, Johan Neyts, Kai Dallmeier

Abstract

Recent outbreaks of yellow fever virus (YFV) in West Africa and Brazil resulted in rapid depletion of global vaccine emergency stockpiles and raised concerns about being unprepared against future YFV epidemics. Here we report that a live attenuated virus similar to the Japanese encephalitis virus (JEV) vaccine JE-CVax/Imojev that consists of YFV-17D vaccine from which the structural (prM/E) genes have been replaced with those of the JEV SA14-14-2 vaccine strain confers full protection in mice against lethal YFV challenge. In contrast to the YFV-17D-mediated protection against YFV, this protection is not mediated by neutralizing antibodies but correlates with YFV-specific nonneutralizing antibodies and T cell responses against cell-associated YFV NS1 and other YFV nonstructural (NS) proteins. Our findings reveal the potential of YFV NS proteins to mediate protection and demonstrate that chimeric flavivirus vaccines, such as Imojev, could confer protection against two flaviviruses. This dual protection may have implications for the possible off-label use of JE-CVax in case of emergency and vaccine shortage during YFV outbreaks. In addition, populations in Asia that have been vaccinated with Imojev may already be protected against YFV should outbreaks ever occur on that continent, as several countries/regions in the Asia-Pacific are vulnerable to international spread of the YFV.IMPORTANCE Efficient and safe vaccines against yellow fever (e.g., YFV-17D) that provide long-lasting protection by rapidly inducing neutralizing antibody responses exist. However, the vaccine supply cannot cope with an increasing demand posed by urban outbreaks in recent years. Here we report that JE-CVax/Imojev, a YFV-17D-based chimeric Japanese encephalitis vaccine, also efficiently protects against YFV infection in mice. In case of shortage of the YFV vaccine during yellow fever outbreaks, (off-label) use of JE-CVax/Imojev may be considered. Moreover, wider use of JE-CVax/Imojev in Asia may lower the risk of the much-feared YFV spillover to the continent. More generally, chimeric vaccines that combine surface antigens and replication machineries of two distinct flaviviruses may be considered dual vaccines for the latter pathogen without induction of surface-specific antibodies. Following this rationale, novel flavivirus vaccines that do not hold a risk for antibody-dependent enhancement (ADE) of infection (inherent to current dengue vaccines and dengue vaccine candidates) could be designed.

Keywords: antibody-dependent cellular cytotoxicity (ADCC); antibody-dependent enhancement; chimeric YFV-17D vaccine; chimeric flavivirus vaccine; cross-protection; dual protection; flavivirus; nonneutralizing antibodies; off-label use of vaccine; protective T cell responses.

Copyright © 2020 Mishra et al.

Figures

FIG 1
FIG 1
In vivo evaluation of JE-CVax-mediated dual protection against lethal JEV SA14-14-2 and YFV-17D challenge. (A to D) AG129 and C57BL/6 mice were first vaccinated via the i.p. route with either 104 PFU of JE-CVax (blue), 1/6th of a human dose of Ixiaro (green), or assay medium as a negative control (red). Animals vaccinated with Ixiaro were boosted with another 1/6th of a human dose of Ixiaro 14 dpv. In order to facilitate vaccine virus replication (28), wild-type C57BL/6 mice receiving JE-CVax vaccination were treated with MAR1-5A3 antibody. AG129 mice were i.p. challenged with 103 of PFU JEV SA14-14-2 at 28 dpv (A) or with 103 PFU of YFV-17D at 28 dpv (B) or at 0, 4, 7, 14, 21, and 28 dpv (C). C57BL/6 mice were i.c. challenged with 104 PFU of YFV-17D at 28 dpv (D). Animals were observed for 5 weeks after challenge and were euthanized when humane endpoints were reached. The data represent cumulative results of at least two independent experiments. Log rank (Mantel-Cox) survival analysis test was performed for statistical significance. **, P ≤ 0.01; ****, P ≤ 0.001 compared to the nonvaccinated group.
FIG 2
FIG 2
Serological analysis of serum of JE-CVax-vaccinated and YFV-17D- or ZIKV-MR766-challenged animals. (A) Detection of nAbs against JEV and YFV. CPE neutralization tests (CPENT) for JE-CVax (circles) and YFV-17D (squares) were performed on sera day 0 prior to vaccination (preimmune, red), day 28 after vaccination (blue), and after challenge (study endpoint, orange) for samples of JE-CVax-vaccinated AG129 mice, of JE-CVax-vaccinated mice after subsequent YFV-17D-challenge (n = 34), of mice hyperimmunized with JE-CVax (n = 13; first bleed 2 weeks post last booster immunization, blue), and of mice vaccinated with Ixiaro (green). Limit of detection (LOD) for virus neutralization was log10 20 (1.3). Data are presented as log10 CPENT50 (mean ± SD). The data presented are from ≥3 independent experiments. Statistical significance was determined using one-way ANOVA. ****, P ≤ 0.0001 for mean log10 CPENT50 titers against JEV or YFV compared to mean log10 CPENT50 titers before JE-CVax vaccination and before YFV-17D-challenge, respectively. (B) Quantitation of anti-YFV NS1 binding antibodies by direct ELISA. Serum from naive, nonvaccinated mice (red) or mice that had been vaccinated with 103 to 105 of PFU JE-CVax (blue) or that had been infected with 104 PFU of YFV-17D (orange) or with 105 PFU of ZIKV-MR766 (pink) were collected either 28 days postimmunization or when euthanized at the humane endpoint (n ≥ 5). The data are means of two independent analyses. Statistical significance was determined using one-way ANOVA. **, P ≤ 0.01 compared to YFV-17D. (C) Binding of serum antibodies to NS1-expressing cells. HEK 293 cells were transfected with a plasmid expressing YFV-17D NS1 as a transcriptional fusion to GFP (top) or infected with the YFV-17D-mCherry reporter virus (bottom). Either 48 h after transfection or 72 h after infection, cells were stained with the anti-YFV NS1-specific MAb 1A5 (left), with serum from mice that were vaccinated with JE-CVax (center), or with serum from naive, nonvaccinated mice (right). Graphs show flow cytometric analysis of GFP or mCherry fluorescence and visualization of anti-YFV NS1 antibody binding using a PE-Cy7-conjugated goat anti-mouse IgG secondary antibody. The fraction of NS1-positive cells (GFP or mCherry) stained by MAb 1A5 or serum of JE-CVax-immunized mice (anti-mouse IgG) is given as a percentage in the upper right quadrant. Data from one representative experiment out of four independent experiments are shown.
FIG 3
FIG 3
Role of antibody-dependent cell-mediated cytotoxicity (ADCC) conferred by JE-CVax hyperimmune serum in the protection against YFV. JE-CVax hyperimmune serum was tested for its ability to mediate ADCC activity compared to serum of nonvaccinated mice (normal serum) at 3:1 (A) and 10:1 (B) effector (E)-to-target (T) cell ratios. Experiments were conducted twice, each in triplicate, and data are presented as means ± SEMs for fold changes compared to control (CC) (i.e., mean reporter signal plus three SDs from E:T in the absence of hyperimmune serum). Values from noninfected target cells incubated with E in the presence of either hyperimmune serum or normal serum at highest antibody concentrations (dilution 1:9) are indicated as Control-9. Statistical significance was determined using two-way ANOVA. * and ****, P ≤ 0.05 and 0.0001 compared to normal serum.
FIG 4
FIG 4
Detection of protective T cell responses directed against YFV. (A to C) ELISpot assay data showing TNF-α (A) IFN-γ (B and C) and production by splenocytes of AG129 mice (n = 5; A and B) or C57BL/6 (n = 10; C) at 18 and 4 weeks, respectively, after vaccination with 104 PFU of JE-CVax, following 16 h ex vivo restimulation with either an MHC class I-restricted peptide derived from YFV-17D NS3 (32) or the lysate of YFV-17D- or JEV SA14-14-2-infected Vero E6 cells. Stimulation using lysate of noninfected Vero E6 cells served as a negative control. The data are derived from two independent experiments. Spot counts were normalized by subtraction of the number of spots in corresponding wells stimulated with uninfected Vero E6 cell lysate. (D) Cytokine expression profile of YFV-specific T cells. Shown are IFN-γ and TNF-α production profiles of YFV-specific CD4+ and CD8+ T cells from JE-CVax-vaccinated AG129 and C57BL/6 mice 18 and 4 weeks, respectively, postvaccination, as determined by intracellular cytokine staining. Mouse splenocytes were stimulated 16 h ex vivo with either an MHC class I-restricted NS3 peptide, cell lysate of YFV-17D-infected Vero E6 cells, or lysate of uninfected Vero E6 cells. The data are derived from two independent experiments and normalized by subtraction of number of cytokine-secreting T cells in corresponding samples in which uninfected Vero E6 cell lysate was used as recall antigen. (E) T cell-mediated in vivo protection against YFV. Loss of protection resulting from antibody-mediated T cell depletion (30, 52) suggests a direct functional involvement of CD4+ and CD8+ T cells in JE-CVax-mediated immunity against YFV in C57BL/6 mice (n ≥ 7) that had been vaccinated with 104 PFU of JE-CVax and subsequently challenged intracranially with 104 PFU of YFV-17D. Depletions were performed by administration of 0.5 mg of anti-mouse CD4 and/or anti-mouse CD8 antibodies i.p. on day −2 and day 0 each prior to YFV challenge. Log rank (Mantel-Cox) survival analysis test was performed for statistical significance. * and **, P ≤ 0.05 and 0.01 compared to vaccinated group (n = 5); +, P ≤ 0.05 compared to CD4-depleted group (n = 8).

References

    1. Ishikawa T, Yamanaka A, Konishi E. 2014. A review of successful flavivirus vaccines and the problems with those flaviviruses for which vaccines are not yet available. Vaccine 32:1326–1337. doi:10.1016/j.vaccine.2014.01.040.
    1. Petersen LR, Jamieson DJ, Powers AM, Honein MA. 2016. Zika virus. N Engl J Med 374:1552–1563. doi:10.1056/NEJMra1602113.
    1. Wilder-Smith A, Monath TP. 2017. Responding to the threat of urban yellow fever outbreaks. Lancet Infect Dis 17:248–250. doi:10.1016/S1473-3099(16)30588-6.
    1. Simon-Loriere E, Faye O, Prot M, Casademont I, Fall G, Fernandez-Garcia MD, Diagne MM, Kipela JM, Fall IS, Holmes EC, Sakuntabhai A, Sall AA. 2017. Autochthonous Japanese encephalitis with yellow fever coinfection in Africa. N Engl J Med 376:1483–1485. doi:10.1056/NEJMc1701600.
    1. WHO. 2015. Vaccines and vaccination against yellow fever: WHO Position Paper, June 2013—recommendations. Vaccine 33:76–77. doi:10.1016/j.vaccine.2014.05.040.
    1. Barrett AD. 2016. Yellow fever in Angola and beyond—the problem of vaccine supply and demand. N Engl J Med 375:301–303. doi:10.1056/NEJMp1606997.
    1. Gershman MD, Angelo KM, Ritchey J, Greenberg DP, Muhammad RD, Brunette G, Cetron MS, Sotir MJ. 2017. Addressing a yellow fever vaccine shortage—United States, 2016-2017. MMWR Morb Mortal Wkly Rep 66:457–459. doi:10.15585/mmwr.mm6617e2.
    1. Wasserman S, Tambyah PA, Lim PL. 2016. Yellow fever cases in Asia: primed for an epidemic. Int J Infect Dis 48:98–103. doi:10.1016/j.ijid.2016.04.025.
    1. Chen Z, Liu L, Lv Y, Zhang W, Li J, Zhang Y, Di T, Zhang S, Liu J, Li J, Qu J, Hua W, Li C, Wang P, Zhang Q, Xu Y, Jiang R, Wang Q, Chen L, Wang S, Pang X, Liang M, Ma X, Li X, Wang Q, Zhang F, Li D. 2016. A fatal yellow fever virus infection in China: description and lessons. Emerg Microbes Infect 5:e69. doi:10.1038/emi.2016.89.
    1. Li X, Ma SJ, Liu X, Jiang LN, Zhou JH, Xiong YQ, Ding H, Chen Q. 2014. Immunogenicity and safety of currently available Japanese encephalitis vaccines: a systematic review. Hum Vaccin Immunother 10:3579–3593. doi:10.4161/21645515.2014.980197.
    1. Monath TP, Seligman SJ, Robertson JS, Guy B, Hayes EB, Condit RC, Excler JL, Mac LM, Carbery B, Chen RT, Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG). 2015. Live virus vaccines based on a yellow fever vaccine backbone: standardized template with key considerations for a risk/benefit assessment. Vaccine 33:62–72. doi:10.1016/j.vaccine.2014.10.004.
    1. Dans AL, Dans LF, Lansang MAD, Silvestre MAA, Guyatt GH. 2018. Controversy and debate on dengue vaccine series—paper 1: review of a licensed dengue vaccine: inappropriate subgroup analyses and selective reporting may cause harm in mass vaccination programs. J Clin Epidemiol 95:137–139. doi:10.1016/j.jclinepi.2017.11.019.
    1. Aguiar M, Stollenwerk N. 2018. Dengvaxia efficacy dependency on serostatus: a closer look at more recent data. Clin Infect Dis 66:641–642. doi:10.1093/cid/cix882.
    1. Wilder-Smith A. 2018. Four-year safety follow-up of the tetravalent dengue vaccine CYD-TDV. Clin Microbiol Infect 24:680–681. doi:10.1016/j.cmi.2018.03.024.
    1. Pierson TC, Fremont DH, Kuhn RJ, Diamond MS. 2008. Structural insights into the mechanisms of antibody-mediated neutralization of flavivirus infection: implications for vaccine development. Cell Host Microbe 4:229–238. doi:10.1016/j.chom.2008.08.004.
    1. Watson AM, Lam LK, Klimstra WB, Ryman KD. 2016. The 17D-204 vaccine strain-induced protection against virulent yellow fever virus is mediated by humoral immunity and CD4+ but not CD8+ T cells. PLoS Pathog 12:e1005786. doi:10.1371/journal.ppat.1005786.
    1. Putnak JR, Schlesinger JJ. 1990. Protection of mice against yellow fever virus encephalitis by immunization with a vaccinia virus recombinant encoding the yellow fever virus non-structural proteins, NS1, NS2a and NS2b. J Gen Virol 71:1697–1702. doi:10.1099/0022-1317-71-8-1697.
    1. Schlesinger JJ, Foltzer M, Chapman S. 1993. The Fc portion of antibody to yellow fever virus NS1 is a determinant of protection against YF encephalitis in mice. Virology 192:132–141. doi:10.1006/viro.1993.1015.
    1. Schlesinger JJ, Brandriss MW, Cropp CB, Monath TP. 1986. Protection against yellow fever in monkeys by immunization with yellow fever virus nonstructural protein NS1. J Virol 60:1153–1155. doi:10.1128/JVI.60.3.1153-1155.1986.
    1. Rastogi M, Sharma N, Singh SK. 2016. Flavivirus NS1: a multifaceted enigmatic viral protein. Virol J 13:131. doi:10.1186/s12985-016-0590-7.
    1. Chung KM, Thompson BS, Fremont DH, Diamond MS. 2007. Antibody recognition of cell surface-associated NS1 triggers Fc-gamma receptor-mediated phagocytosis and clearance of West Nile virus-infected cells. J Virol 81:9551–9555. doi:10.1128/JVI.00879-07.
    1. Bailey MJ, Broecker F, Duehr J, Arumemi F, Krammer F, Palese P, Tan GS. 2019. Antibodies elicited by an NS1-based vaccine protect mice against Zika virus. mBio 10:e02861-18. doi:10.1128/mBio.02861-18.
    1. Bassi MR, Larsen MA, Kongsgaard M, Rasmussen M, Buus S, Stryhn A, Thomsen AR, Christensen JP. 2016. Vaccination with replication deficient adenovectors encoding YF-17D antigens induces long-lasting protection from severe yellow fever virus infection in mice. PLoS Negl Trop Dis 10:e0004464. doi:10.1371/journal.pntd.0004464.
    1. Desprès P, Dietrich J, Girard M, Bouloy M. 1991. Recombinant baculoviruses expressing yellow fever virus E and NS1 proteins elicit protective immunity in mice. J Gen Virol 72:2811–2816. doi:10.1099/0022-1317-72-11-2811.
    1. Chan KR, Wang X, Saron WAA, Gan ES, Tan HC, Mok DZL, Zhang SL, Lee YH, Liang C, Wijaya L, Ghosh S, Cheung YB, Tannenbaum SR, Abraham SN, St John AL, Low JGH, Ooi EE. 2016. Cross-reactive antibodies enhance live attenuated virus infection for increased immunogenicity. Nat Microbiol 1:16164. doi:10.1038/nmicrobiol.2016.164.
    1. Li J, Gao N, Fan D, Chen H, Sheng Z, Fu S, Liang G, An J. 2016. Cross-protection induced by Japanese encephalitis vaccines against different genotypes of dengue viruses in mice. Sci Rep 6:19953. doi:10.1038/srep19953.
    1. Schlesinger JJ, Brandriss MW, Monath TP. 1983. Monoclonal antibodies distinguish between wild and vaccine strains of yellow fever virus by neutralization, hemagglutination inhibition, and immune precipitation of the virus envelope protein. Virology 125:8–17. doi:10.1016/0042-6822(83)90059-4.
    1. Kum DB, Mishra N, Boudewijns R, Gladwyn-Ng I, Alfano C, Ma J, Schmid MA, Marques RE, Schols D, Kaptein S, Nguyen L, Neyts J, Dallmeier K. 2018. A yellow fever-Zika chimeric virus vaccine candidate protects against Zika infection and congenital malformations in mice. NPJ Vaccines 3:56. doi:10.1038/s41541-018-0092-2.
    1. Cheng ZJ, Garvin D, Paguio A, Moravec R, Engel L, Fan F, Surowy T. 2014. Development of a robust reporter-based ADCC assay with frozen, thaw-and-use cells to measure Fc effector function of therapeutic antibodies. J Immunol Methods 414:69–81. doi:10.1016/j.jim.2014.07.010.
    1. Akondy RS, Monson ND, Miller JD, Edupuganti S, Teuwen D, Wu H, Quyyumi F, Garg S, Altman JD, Del Rio C, Keyserling HL, Ploss A, Rice CM, Orenstein WA, Mulligan MJ, Ahmed R. 2009. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J Immunol 183:7919–7930. doi:10.4049/jimmunol.0803903.
    1. Li XF, Deng YQ, Yang HQ, Zhao H, Jiang T, Yu XD, Li SH, Ye Q, Zhu SY, Wang HJ, Zhang Y, Ma J, Yu YX, Liu ZY, Li YH, Qin ED, Shi PY, Qin CF. 2013. A chimeric dengue virus vaccine using Japanese encephalitis virus vaccine strain SA14-14-2 as backbone is immunogenic and protective against either parental virus in mice and nonhuman primates. J Virol 87:13694–13705. doi:10.1128/JVI.00931-13.
    1. Bassi MR, Kongsgaard M, Steffensen MA, Fenger C, Rasmussen M, Skjødt K, Finsen B, Stryhn A, Buus S, Christensen JP, Thomsen AR. 2015. CD8+ T cells complement antibodies in protecting against yellow fever virus. J Immunol 194:1141–1153. doi:10.4049/jimmunol.1402605.
    1. Calvert AE, Dixon KL, Delorey MJ, Blair CD, Roehrig JT. 2014. Development of a small animal peripheral challenge model of Japanese encephalitis virus using interferon deficient AG129 mice and the SA14-14-2 vaccine virus strain. Vaccine 32:258–264. doi:10.1016/j.vaccine.2013.11.016.
    1. Thibodeaux BA, Garbino NC, Liss NM, Piper J, Blair CD, Roehrig JT. 2012. A small animal peripheral challenge model of yellow fever using interferon-receptor deficient mice and the 17D-204 vaccine strain. Vaccine 30:3180–3187. doi:10.1016/j.vaccine.2012.03.003.
    1. Meier KC, Gardner CL, Khoretonenko MV, Klimstra WB, Ryman KD. 2009. A mouse model for studying viscerotropic disease caused by yellow fever virus infection. PLoS Pathog 5:e1000614. doi:10.1371/journal.ppat.1000614.
    1. Yang J, Yang H, Li Z, Lin H, Zhao Y, Wang W, Tan S, Zeng X, Li Y. 2017. The chimeric Japanese encephalitis/dengue 2 virus protects mice from challenge by both dengue virus and JEV virulent virus. Protein Cell 8:225–229. doi:10.1007/s13238-016-0363-5.
    1. Zellweger RM, Tang WW, Eddy WE, King K, Sanchez MC, Shresta S. 2015. CD8+ T cells can mediate short-term protection against heterotypic dengue virus reinfection in mice. J Virol 89:6494–6505. doi:10.1128/JVI.00036-15.
    1. Xu X, Vaughan K, Weiskopf D, Grifoni A, Diamond MS, Sette A, Peters B. 2016. Identifying candidate targets of immune responses in Zika virus based on homology to epitopes in other flavivirus species. PLoS Curr 8:ecurrents.outbreaks.9aa2e1fb61b0f632f58a098773008c4b. doi:10.1371/currents.outbreaks.9aa2e1fb61b0f632f58a098773008c4b.
    1. Nasveld PE, Marjason J, Bennett S, Aaskov J, Elliott S, McCarthy K, Kanesa-Thasan N, Feroldi E, Reid M. 2010. Concomitant or sequential administration of live attenuated Japanese encephalitis chimeric virus vaccine and yellow fever 17D vaccine: randomized double-blind phase II evaluation of safety and immunogenicity. Hum Vaccin 6:906–914. doi:10.4161/hv.6.11.12854.
    1. Chambers TJ, Nestorowicz A, Mason PW, Rice CM. 1999. Yellow fever/Japanese encephalitis chimeric viruses: construction and biological properties. J Virol 73:3095–3101. doi:10.1128/JVI.73.4.3095-3101.1999.
    1. Monath TP, Soike K, Levenbook I, Zhang ZX, Arroyo J, Delagrave S, Myers G, Barrett AD, Shope RE, Ratterree M, Chambers TJ, Guirakhoo F. 1999. Recombinant, chimaeric live, attenuated vaccine (ChimeriVax) incorporating the envelope genes of Japanese encephalitis (SA14-14-2) virus and the capsid and nonstructural genes of yellow fever (17D) virus is safe, immunogenic and protective in non-human primates. Vaccine 17:1869–1882. doi:10.1016/S0264-410X(98)00487-3.
    1. Stiasny K, Kiermayr S, Holzmann H, Heinz FX. 2006. Cryptic properties of a cluster of dominant flavivirus cross-reactive antigenic sites. J Virol 80:9557–9568. doi:10.1128/JVI.00080-06.
    1. Schlesinger JJ, Brandriss MW, Putnak JR, Walsh EE. 1990. Cell surface expression of yellow fever virus non-structural glycoprotein NS1: consequences of interaction with antibody. J Gen Virol 71:593–599. doi:10.1099/0022-1317-71-3-593.
    1. Gould EA, Buckley A, Barrett AD, Cammack N. 1986. Neutralizing (54K) and non-neutralizing (54K and 48K) monoclonal antibodies against structural and non-structural yellow fever virus proteins confer immunity in mice. J Gen Virol 67:591–595. doi:10.1099/0022-1317-67-3-591.
    1. James EA, LaFond RE, Gates TJ, Mai DT, Malhotra U, Kwok WW. 2013. Yellow fever vaccination elicits broad functional CD4+ T cell responses that recognize structural and nonstructural proteins. J Virol 87:12794–12804. doi:10.1128/JVI.01160-13.
    1. Guy B, Nougarede N, Begue S, Sanchez V, Souag N, Carre M, Chambonneau L, Morrisson DN, Shaw D, Qiao M, Dumas R, Lang J, Forrat R. 2008. Cell-mediated immunity induced by chimeric tetravalent dengue vaccine in naive or flavivirus-primed subjects. Vaccine 26:5712–5721. doi:10.1016/j.vaccine.2008.08.019.
    1. Harenberg A, Begue S, Mamessier A, Gimenez-Fourage S, Ching Seah C, Wei Liang A, Li Ng J, Yun Toh X, Archuleta S, Wilder-Smith A, Shek LP, Wartel-Tram A, Bouckenooghe A, Lang J, Crevat D, Caillet C, Guy B. 2013. Persistence of Th1/Tc1 responses one year after tetravalent dengue vaccination in adults and adolescents in Singapore. Hum Vaccin Immunother 9:2317–2325. doi:10.4161/hv.25562.
    1. Lund O. 2011. Human leukocyte antigen (HLA) class I restricted epitope discovery in yellow fewer and dengue viruses: importance of HLA binding strength. PLoS One 6:e26494. doi:10.1371/journal.pone.0026494.
    1. de Melo AB, Nascimento EJM, Braga-Neto U, Dhalia R, Silva AM, Oelke M, Schneck JP, Sidney J, Sette A, Montenegro SML, Marques ETA. 2013. T-cell memory responses elicited by yellow fever vaccine are targeted to overlapping epitopes containing multiple HLA-I and-II binding motifs. PLoS Negl Trop Dis 7:e1938. doi:10.1371/journal.pntd.0001938.
    1. Blom K, Braun M, Ivarsson MA, Gonzalez VD, Falconer K, Moll M, Ljunggren H-G, Michaëlsson J, Sandberg JK. 2013. Temporal dynamics of the primary human T cell response to yellow fever virus 17D as it matures from an effector- to a memory-type response. J Immunol 190:2150–2158. doi:10.4049/jimmunol.1202234.
    1. Kongsgaard M, Bassi MR, Rasmussen M, Skjødt K, Thybo S, Gabriel M, Hansen MB, Christensen JP, Thomsen AR, Buus S, Stryhn A. 2017. Adaptive immune responses to booster vaccination against yellow fever virus are much reduced compared to those after primary vaccination. Sci Rep 7:1–14. doi:10.1038/s41598-017-00798-1.
    1. Barrett AD, Gould EA. 1986. Comparison of neurovirulence of different strains of yellow fever virus in mice. J Gen Virol 67:631–637. doi:10.1099/0022-1317-67-4-631.
    1. Perelygin AA, Scherbik SV, Zhulin IB, Stockman BM, Li Y, Brinton MA. 2002. Positional cloning of the murine flavivirus resistance gene. Proc Natl Acad Sci U S A 99:9322–9327. doi:10.1073/pnas.142287799.
    1. Theiler M, Smith HH. 1937. The use of yellow fever virus modified by in vitro cultivation for human immunization. J Exp Med 65:787–800. doi:10.1084/jem.65.6.787.
    1. Chokephaibulkit K, Sirivichayakul C, Thisyakorn U, Sabchareon A, Pancharoen C, Bouckenooghe A, Gailhardou S, Boaz M, Feroldi E. 2010. Safety and immunogenicity of a single administration of live-attenuated Japanese encephalitis vaccine in previously primed 2- to 5-year-olds and naive 12- to 24-month-olds: multicenter randomized controlled trial. Pediatr Infect Dis J 29:1111–1117. doi:10.1097/INF.0b013e3181f68e9c.
    1. Monath TP, Guirakhoo F, Nichols R, Yoksan S, Schrader R, Murphy C, Blum P, Woodward S, McCarthy K, Mathis D, Johnson C, Bedford P. 2003. Chimeric live, attenuated vaccine against Japanese encephalitis (ChimeriVax-JE): phase 2 clinical trials for safety and immunogenicity, effect of vaccine dose and schedule, and memory response to challenge with inactivated Japanese encephalitis antigen. J Infect Dis 188:1213–1230. doi:10.1086/378356.
    1. Guirakhoo F, Kitchener S, Morrison D, Forrat R, McCarthy K, Nichols R, Yoksan S, Duan X, Ermak TH, Kanesa-Thasan N, Bedford P, Lang J, Quentin-Millet MJ, Monath TP. 2006. Live attenuated chimeric yellow fever dengue type 2 (ChimeriVax-DEN2) vaccine: phase I clinical trial for safety and immunogenicity: effect of yellow fever pre-immunity in induction of cross neutralizing antibody responses to all 4 dengue serotypes. Hum Vaccin 2:60–67. doi:10.4161/hv.2.2.2555.
    1. Xie X, Yang Y, Muruato AE, Zou J, Shan C, Nunes BT, Medeiros DB, Vasconcelos PF, Weaver SC, Rossi SL, Shi PY. 2017. Understanding Zika virus stability and developing a chimeric vaccine through functional analysis. mBio 8:e02134-16. doi:10.1128/mBio.02134-16.
    1. Arroyo J, Guirakhoo F, Fenner S, Zhang ZX, Monath TP, Chambers TJ. 2001. Molecular basis for attenuation of neurovirulence of a yellow fever Virus/Japanese encephalitis virus chimera vaccine (ChimeriVax-JE). J Virol 75:934–942. doi:10.1128/JVI.75.2.934-942.2001.
    1. Zmurko J, Marques RE, Schols D, Verbeken E, Kaptein SJ, Neyts J. 2016. The viral polymerase inhibitor 7-deaza-2′-c-methyladenosine is a potent inhibitor of in vitro Zika virus replication and delays disease progression in a robust mouse infection model. PLoS Negl Trop Dis 10:e0004695. doi:10.1371/journal.pntd.0004695.
    1. Bredenbeek PJ, Kooi EA, Lindenbach B, Huijkman N, Rice CM, Spaan WJ. 2003. A stable full-length yellow fever virus cDNA clone and the role of conserved RNA elements in flavivirus replication. J Gen Virol 84:1261–1268. doi:10.1099/vir.0.18860-0.
    1. Demidenko AA, Blattman JN, Blattman NN, Greenberg PD, Nibert ML. 2013. Engineering recombinant reoviruses with tandem repeats and a tetravirus 2A-like element for exogenous polypeptide expression. Proc Natl Acad Sci U S A 110:E1867–E1876. doi:10.1073/pnas.1220107110.
    1. Avirutnan P, Fuchs A, Hauhart RE, Somnuke P, Youn S, Diamond MS, Atkinson JP. 2010. Antagonism of the complement component C4 by flavivirus nonstructural protein NS1. J Exp Med 207:793–806. doi:10.1084/jem.20092545.
    1. Barba-Spaeth G, Longman RS, Albert ML, Rice CM. 2005. Live attenuated yellow fever 17D infects human DCs and allows for presentation of endogenous and recombinant T cell epitopes. J Exp Med 202:1179–1184. doi:10.1084/jem.20051352.
    1. Li XF, Li XD, Deng CL, Dong HL, Zhang QY, Ye Q, Ye HQ, Huang XY, Deng YQ, Zhang B, Qin CF. 2017. Visualization of a neurotropic flavivirus infection in mouse reveals unique viscerotropism controlled by host type I interferon signaling. Theranostics 7:912–925. doi:10.7150/thno.16615.
    1. Aguirre S, Maestre AM, Pagni S, Patel JR, Savage T, Gutman D, Maringer K, Bernal-Rubio D, Shabman RS, Simon V, Rodriguez-Madoz JR, Mulder LC, Barber GN, Fernandez-Sesma A. 2012. DENV inhibits type I IFN production in infected cells by cleaving human STING. PLoS Pathog 8:e1002934. doi:10.1371/journal.ppat.1002934.
    1. Ashour J, Morrison J, Laurent-Rolle M, Belicha-Villanueva A, Plumlee CR, Bernal-Rubio D, Williams KL, Harris E, Fernandez-Sesma A, Schindler C, García-Sastre A. 2010. Mouse STAT2 restricts early dengue virus replication. Cell Host Microbe 8:410–421. doi:10.1016/j.chom.2010.10.007.
    1. Grant A, Ponia SS, Tripathi S, Balasubramaniam V, Miorin L, Sourisseau M, Schwarz MC, Sánchez-Seco MP, Evans MJ, Best SM, García-Sastre A. 2016. Zika virus targets human STAT2 to inhibit type i interferon signaling. Cell Host Microbe 19:882–890. doi:10.1016/j.chom.2016.05.009.
    1. Laurent-Rolle M, Boer EF, Lubick KJ, Wolfinbarger JB, Carmody AB, Rockx B, Liu W, Ashour J, Shupert WL, Holbrook MR, Barrett AD, Mason PW, Bloom ME, García-Sastre A, Khromykh AA, Best SM. 2010. The NS5 protein of the virulent West Nile virus NY99 strain is a potent antagonist of type I interferon-mediated JAK-STAT signaling. J Virol 84:3503–3515. doi:10.1128/JVI.01161-09.
    1. Muñoz-Jordán JL, Laurent-Rolle M, Ashour J, Martínez-Sobrido L, Ashok M, Lipkin WI, García-Sastre A. 2005. Inhibition of alpha/beta interferon signaling by the NS4B protein of flaviviruses. J Virol 79:8004–8013. doi:10.1128/JVI.79.13.8004-8013.2005.
    1. Erickson AK, Pfeiffer JK. 2015. Spectrum of disease outcomes in mice infected with YFV-17D. J Gen Virol 96:1328–1339. doi:10.1099/vir.0.000075.
    1. Julander JG. 2016. Animal models of yellow fever and their application in clinical research. Curr Opin Virol 18:64–69. doi:10.1016/j.coviro.2016.03.010.
    1. Lam LKM, Watson AM, Ryman KD, Klimstra WB. 2018. Gamma-interferon exerts a critical early restriction on replication and dissemination of yellow fever virus vaccine strain 17D-204. NPJ Vaccines 3:5. doi:10.1038/s41541-017-0039-z.
    1. Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, Kamijo R, Vilcek J, Zinkernagel RM, Aguet M. 1993. Immune response in mice that lack the interferon-gamma receptor. Science 259:1742–1745. doi:10.1126/science.8456301.
    1. Kruisbeek AM. 2001. In vivo depletion of CD4- and CD8-specific T cells. Curr Protoc Immunol Chapter 4:Unit 4.1. doi:10.1002/0471142735.im0401s01.
    1. Johnson BW, Kosoy O, Hunsperger E, Beltran M, Delorey M, Guirakhoo F, Monath T. 2009. Evaluation of chimeric Japanese encephalitis and dengue viruses for use in diagnostic plaque reduction neutralization tests. Clin Vaccine Immunol 16:1052–1059. doi:10.1128/CVI.00095-09.
    1. Reed LJ, Muench H. 1938. A simple method of estimating fifty per cent endpoints. Am J Epidemiol 27:493–497. doi:10.1093/oxfordjournals.aje.a118408.
    1. Jochmans D, Leyssen P, Neyts J. 2012. A novel method for high-throughput screening to quantify antiviral activity against viruses that induce limited CPE. J Virol Methods 183:176–179. doi:10.1016/j.jviromet.2012.04.011.
    1. Bardi MS, Jarduli LR, Jorge AJ, Camargo RB, Carneiro FP, Gelinski JR, Silva RA, Lavado EL. 2012. HLA-A, B and DRB1 allele and haplotype frequencies in volunteer bone marrow donors from the north of Parana State. Rev Bras Hematol Hemoter 34:25–30. doi:10.5581/1516-8484.20120010.
    1. Chao DY, Galula JU, Shen WF, Davis BS, Chang GJ. 2015. Nonstructural protein 1-specific immunoglobulin M and G antibody capture enzyme-linked immunosorbent assays in diagnosis of flaviviral infections in humans. J Clin Microbiol 53:557–566. doi:10.1128/JCM.02735-14.
    1. Falgout B, Bray M, Schlesinger JJ, Lai CJ. 1990. Immunization of mice with recombinant vaccinia virus expressing authentic dengue virus nonstructural protein NS1 protects against lethal dengue virus encephalitis. J Virol 64:4356–4363. doi:10.1128/JVI.64.9.4356-4363.1990.

Source: PubMed

3
Subscribe