A tetravalent live attenuated dengue virus vaccine stimulates balanced immunity to multiple serotypes in humans

Usha K Nivarthi, Jesica Swanstrom, Matthew J Delacruz, Bhumi Patel, Anna P Durbin, Steve S Whitehead, Beth D Kirkpatrick, Kristen K Pierce, Sean A Diehl, Leah Katzelnick, Ralph S Baric, Aravinda M de Silva, Usha K Nivarthi, Jesica Swanstrom, Matthew J Delacruz, Bhumi Patel, Anna P Durbin, Steve S Whitehead, Beth D Kirkpatrick, Kristen K Pierce, Sean A Diehl, Leah Katzelnick, Ralph S Baric, Aravinda M de Silva

Abstract

The four-dengue virus (DENV) serotypes infect several hundred million people annually. For the greatest safety and efficacy, tetravalent DENV vaccines are designed to stimulate balanced protective immunity to all four serotypes. However, this has been difficult to achieve. Clinical trials with a leading vaccine demonstrated that unbalanced replication and immunodominance of one vaccine component over others can lead to low efficacy and vaccine enhanced severe disease. The Laboratory of Infectious Diseases at the National Institutes of Health has developed a live attenuated tetravalent DENV vaccine (TV003), which is currently being tested in phase 3 clinical trials. Here we report, our study to determine if TV003 stimulate balanced and serotype-specific (TS) neutralizing antibody (nAb) responses to each serotype. Serum samples from twenty-one dengue-naive individuals participated under study protocol CIR287 (ClinicalTrials.gov NCT02021968) are analyzed 6 months after vaccination. Most subjects (76%) develop TS nAbs to 3 or 4 DENV serotypes, indicating immunity is induced by each vaccine component. Vaccine-induced TS nAbs map to epitopes known to be targets of nAbs in people infected with wild type DENVs. Following challenge with a partially attenuated strain of DENV2, all 21 subjects are protected from the efficacy endpoints. However, some vaccinated individuals develop post challenge nAb boost, while others mount post-challenge antibody responses that are consistent with sterilizing immunity. TV003 vaccine induced DENV2 TS nAbs are associated with sterilizing immunity. Our results indicate that nAbs to TS epitopes on each serotype may be a better correlate than total levels of nAbs currently used for guiding DENV vaccine development.

Conflict of interest statement

A.M.D. has consulted on Dengue vaccine for Takeda vaccines, Sanofi Pasteur, GSK, and Merck Pharmaceuticals and also an inventor in patents related to Dengue vaccines. R.S.B. has consulted on Dengue vaccines for Takeda Vaccines and Sanofi Pasteur and is also an inventor in patents related to Dengue vaccines. All other authors report no potential conflicts of interest.

Figures

Fig. 1. DENV antibody depletion assay to…
Fig. 1. DENV antibody depletion assay to estimate proportions of serotype-specific and cross-reactive-neutralizing antibodies in a subject who received TV003.
Subject 6 was tested 6 months after vaccination to measure levels of TS binding and neutralizing antibodies to each DENV serotype. The sample was incubated with immobilized bovine serum albumin (control depleted), DENV2 antigen (DENV2 depleted) and a mix of DENV1, 3 and 4 antigens (DENV1, 3, 4 depleted) to remove specific populations of antibodies. The DENV2 depletion resulted in the removal of DENV cross-reactive and DENV2 TS antibodies, while retaining any DENV1, 3, or 4 TS antibodies in the sample. The DENV1, 3, 4 depletion resulted in the removal of DENV cross-reactive and DENV1, 3, and 4 TS antibodies, while retaining any DENV2 TS antibodies in the sample. The sample depleted of specific antibody populations was tested by a ELISA to detect any DENV1, 2, 3, or 4 type specific binding antibodies. Data was analyzed from subject 6 and represented as bars from a single independent experiment and expressed as mean ODs with corresponding data points shown. b Neutralization test to measure levels of TS-neutralizing antibodies to each DENV serotype. Data was analyzed from subject 6 and represented as dots from a single independent experiment and expressed as 50% neutralization titers.
Fig. 2. Proportion and level of homotypic-neutralizing…
Fig. 2. Proportion and level of homotypic-neutralizing antibodies to each serotype in DENV naive subjects who received TV003.
The antibody depletion assay was used to measure proportions of DENV TS (homotypic)-neutralizing antibodies induced by TV003 in (n = 21) DENV naive subjects. a The percentage of the total-neutralizing antibody response to each serotype contributed by the serotype-specific antibodies for each subject. b The absolute level of homotypic-neutralizing antibody to each serotype for each subject. c Number and % of subjects that developed an absolute TS nab titer >20 is depicted. Data were analyzed from a total number of 21 subjects indicated as dots from a single independent experiment and expressed as mean titers ± 95% confidence intervals (a, b). Nonparametric one-way Anova with Friedman Dunn’s multiple comparisons test was used for determining the statistical differences between the level of TS nAbs across serotypes (b). Each p-value was adjusted to account for multiple comparisons. (****p < 0.0001; ***p < 0.001; **p < 0.01; *p < 0.05). The serotypes with significantly high levels of TS nabs compared to other serotypes is shown (b) (DENV3: **p = 0.0092; CI:148.4–481.5; DENV4: **p = 0.0061; CI: 172.9–469.8). The dotted line indicates the cut off for the absolute type specific titer.
Fig. 3. Characterization of DENV4 126 and…
Fig. 3. Characterization of DENV4 126 and 131 epitope transplant rDENV (rDENV2/4) used for mapping the DENV4 responses.
a Recombinant DENV2 containing EDI/II hinge, EDII residues from DENV4 that includes epitopes of the DENV4-126 and DENV4-131 mAbs. The figure depicts a model of DENV2 E protein dimer, with the mutated residues colored in blue. b Amino acid alignment of DENV2, rDENV2/4, DENV4, protein sequences in the EDI/II hinge; EDII regions with mutated residues in rDENV2/4 colored in blue. c Monoclonal antibodies (2D22, 3F9, 4J23: DENV2 TS mAbs, 5H2, 126,131: DENV4 TS mAbs, EDE1 C8, EDE2 B7 pan DENV specific mAbs) neutralization of rDENV2/4 compared to each parental strain’s is shown. Data shown here was from a single independent experiment and represented as mean neutralization titers. d Characterizing the neutralizing properties of DENV2 (n = 3) and DENV4 (n = 3) polyclonal natural infection sera against rDENV2/4 and the parental viruses. Data were obtained from a single independent experiment and expressed as mean titers.
Fig. 4. TV003 induce varying levels of…
Fig. 4. TV003 induce varying levels of DENV4 serotype-specific responses against known DENV4-neutralizing epitopes.
a DENV2 wt (green), DENV4 wt (blue), rDENV2/4 with DENV4-specific 126, 131 mAb epitopes transplanted into DENV2 (shown in blue) are depicted. b Panel of 19 sera (n = 19) with evidence of DENV4 TS responses were subjected to depletions against DENV2 to map the DENV4 TS nAb epitopes using the rDENV2/4 virus. This is a gain of function virus. Figure shows the 50% neutralization antibody titers for the BSA depleted and the DENV2 depleted serum samples against DENV2 wt, DENV4 wt and the rDENV2/4 viruses. Data were analyzed from a total number of 19 subjects indicated as dots from a single independent experiment and expressed as geometric mean titers ± 95% confidence intervals of the grouped samples (b).
Fig. 5. TV003 induce varying levels of…
Fig. 5. TV003 induce varying levels of DENV1 serotype-specific responses against a known DENV1-neutralizing epitope.
a DENV1wt (yellow), DENV2wt (green), rDENV2/1 with DENV1-specific 1F4 mAb epitope transplanted into DENV2 (shown in yellow) are depicted. b Panel of 8 TV003 sera (n = 8) with evidence of DENV1 TS responses were subjected to depletions against DENV2 to map the DENV1 TS nAb epitopes using the rDENV2/1 virus. Figure shows the 50% nAb titers for the BSA (control) depleted and the DENV2 depleted serum samples against DENV1 wt, DENV2 wt and the rDENV2/1 viruses. Data were analyzed from a total number of eight subjects indicated as dots from a single independent experiment and expressed as geometric mean titers ± 95% confidence intervals of the grouped samples (b).
Fig. 6. TV003 induce varying levels of…
Fig. 6. TV003 induce varying levels of DENV3 serotype-specific responses that do not track with known DENV3-neutralizing epitope.
a DENV3 wt (orange), DENV4 wt (blue), rDENV4/3 with DENV3-specific 5J7 mAb epitope transplanted into DENV4 (shown in orange) are depicted. b Panel of 8 TV003 sera (n = 8) with evidence of DENV3 TS responses were subjected to depletions against DENV4 to map the DENV3 TS nAb epitopes using the rDENV4/3 virus. Figure shows the Neut50 titers for the BSA depleted and the DENV4 depleted serum samples against DENV3 wt, DENV4 wt and the rDENV4/3 viruses. Data were analyzed from a total number of eight subjects indicated as dots from a single independent experiment and expressed as geometric mean titers ± 95% confidence intervals of the grouped samples (b).
Fig. 7. TV003 induce varying levels of…
Fig. 7. TV003 induce varying levels of DENV2 serotype-specific responses against known DENV2-neutralizing epitope domain.
a DENV2 wt (green), DENV4 wt (blue) rDENV4/2 with the entire DENV2 EDIII domain transplanted into DENV4 (shown in green) are depicted. b Panel of 16 TV003 sera (n = 16) with evidence of DENV2 TS responses were subjected to depletions against DENV4 to map the DENV2 TS nAb epitopes using the rDENV4/2 virus. Figure shows the Neut50 titers for the BSA depleted and the DENV4 depleted serum samples against DENV2 wt, DENV4 wt and the rDENV4/2 viruses. Data were analyzed from a total number of 16 subjects indicated as dots from a single independent experiment and expressed as geometric mean titers ± 95% confidence intervals of the grouped samples. Nonparametric one-way Anova with Friedman Dunn’s multiple comparisons test (b) was used for the epitope mapping data to determine the loss or gain in neutralization to the recombinant chimeric viruses compared to the wild type parental backbone viruses. Each p-value was adjusted to account for multiple comparisons. (****p < 0.0001; ***p < 0.001; **p < 0.01; *p < 0.05). The significant gain in neutralization of DENV2 TS nAbs against rDENV4/2 is indicated (***p = 0.0007; CI: 95.0–535.7).

References

    1. Bhatt S, et al. The global distribution and burden of dengue. Nature. 2013;496:504–507. doi: 10.1038/nature12060.
    1. Radke EG, et al. Dengue outbreak in Key West, Florida, USA, 2009. Emerg. Infect. Dis. 2012;18:135–137. doi: 10.3201/eid1801.110130.
    1. Schaffner F, Mathis A. Dengue and dengue vectors in the WHO European region: past, present, and scenarios for the future. Lancet Infect. Dis. 2014;14:1271–1280. doi: 10.1016/S1473-3099(14)70834-5.
    1. Sabin AB. Research on dengue during World War II. Am. J. Trop. Med. Hyg. 1952;1:30–50. doi: 10.4269/ajtmh.1952.1.30.
    1. Montoya M, et al. Symptomatic versus inapparent outcome in repeat dengue virus infections is influenced by the time interval between infections and study year. PLoS Negl. Trop. Dis. 2013;7:e2357. doi: 10.1371/journal.pntd.0002357.
    1. Halstead SB. Etiologies of the experimental dengues of Siler and Simmons. Am. J. Trop. Med. Hyg. 1974;23:974–982. doi: 10.4269/ajtmh.1974.23.974.
    1. Guzman MG, et al. Epidemiologic studies on Dengue in Santiago de Cuba, 1997. Am. J. Epidemiol. 2000;152:793–799. doi: 10.1093/aje/152.9.793.
    1. de Alwis R, et al. In-depth analysis of the antibody response of individuals exposed to primary dengue virus infection. PLoS Negl. Trop. Dis. 2011;5:e1188. doi: 10.1371/journal.pntd.0001188.
    1. de Alwis R, et al. Dengue viruses are enhanced by distinct populations of serotype cross-reactive antibodies in human immune sera. PLoS Pathog. 2014;10:e1004386. doi: 10.1371/journal.ppat.1004386.
    1. Larsen CP, Whitehead SS, Durbin AP. Dengue human infection models to advance dengue vaccine development. Vaccine. 2015;33:7075–7082. doi: 10.1016/j.vaccine.2015.09.052.
    1. Kirkpatrick BD, et al. The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a human challenge model. Sci. Transl. Med. 2016;8:330ra336. doi: 10.1126/scitranslmed.aaf1517.
    1. Capeding MR, et al. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. Lancet. 2014;384:1358–1365. doi: 10.1016/S0140-6736(14)61060-6.
    1. Hadinegoro SR, et al. Efficacy and long-term safety of a dengue vaccine in regions of endemic disease. N. Engl. J. Med. 2015;373:1195–1206. doi: 10.1056/NEJMoa1506223.
    1. Sridhar S, et al. Effect of dengue serostatus on dengue vaccine safety and efficacy. N. Engl. J. Med. 2018;379:327–340. doi: 10.1056/NEJMoa1800820.
    1. Guirakhoo F, et al. Viremia and immunogenicity in nonhuman primates of a tetravalent yellow fever-dengue chimeric vaccine: genetic reconstructions, dose adjustment, and antibody responses against wild-type dengue virus isolates. Virology. 2002;298:146–159. doi: 10.1006/viro.2002.1462.
    1. Guy B, et al. Evaluation of interferences between dengue vaccine serotypes in a monkey model. Am. J. Trop. Med. Hyg. 2009;80:302–311. doi: 10.4269/ajtmh.2009.80.302.
    1. Osorio JE, et al. Efficacy of a tetravalent chimeric dengue vaccine (DENVax) in Cynomolgus macaques. Am. J. Trop. Med. Hyg. 2011;84:978–987. doi: 10.4269/ajtmh.2011.10-0592.
    1. Morrison D, et al. A novel tetravalent dengue vaccine is well tolerated and immunogenic against all 4 serotypes in flavivirus-naive adults. J. Infect. Dis. 2010;201:370–377. doi: 10.1086/649916.
    1. Dayan GH, et al. Assessment of bivalent and tetravalent dengue vaccine formulations in flavivirus-naive adults in Mexico. Hum. Vaccin. Immunother. 2014;10:2853–2863. doi: 10.4161/21645515.2014.972131.
    1. Torresi J, et al. Replication and excretion of the live attenuated tetravalent dengue vaccine CYD-TDV in a flavivirus-naive adult population: assessment of vaccine viremia and virus shedding. J. Infect. Dis. 2017;216:834–841. doi: 10.1093/infdis/jix314.
    1. Henein S, et al. Dissecting antibodies induced by a chimeric yellow fever-dengue, live-attenuated, tetravalent dengue vaccine (CYD-TDV) in naive and dengue-exposed individuals. J. Infect. Dis. 2017;215:351–358.
    1. Dayan GH, et al. Assessment of the long-term efficacy of a dengue vaccine against symptomatic, virologically-confirmed dengue disease by baseline dengue serostatus. Vaccine. 2020;38:3531–3536. doi: 10.1016/j.vaccine.2020.03.029.
    1. Durbin AP, et al. A single dose of any of four different live attenuated tetravalent dengue vaccines is safe and immunogenic in flavivirus-naive adults: a randomized, double-blind clinical trial. J. Infect. Dis. 2013;207:957–965. doi: 10.1093/infdis/jis936.
    1. Durbin AP, et al. rDEN4delta30, a live attenuated dengue virus type 4 vaccine candidate, is safe, immunogenic, and highly infectious in healthy adult volunteers. J. Infect. Dis. 2005;191:710–718. doi: 10.1086/427780.
    1. Durbin AP, et al. rDEN2/4Delta30(ME), a live attenuated chimeric dengue serotype 2 vaccine is safe and highly immunogenic in healthy dengue-naive adults. Hum. Vaccin. 2006;2:255–260. doi: 10.4161/hv.2.6.3494.
    1. Durbin AP, et al. The live attenuated dengue serotype 1 vaccine rDEN1Delta30 is safe and highly immunogenic in healthy adult volunteers. Hum. Vaccin. 2006;2:167–173. doi: 10.4161/hv.2.4.2944.
    1. Durbin AP, Kirkpatrick BD, Pierce KK, Schmidt AC, Whitehead SS. Development and clinical evaluation of multiple investigational monovalent DENV vaccines to identify components for inclusion in a live attenuated tetravalent DENV vaccine. Vaccine. 2011;29:7242–7250. doi: 10.1016/j.vaccine.2011.07.023.
    1. Kirkpatrick BD, et al. Robust and balanced immune responses to all 4 dengue virus serotypes following administration of a single dose of a live attenuated tetravalent dengue vaccine to healthy, flavivirus-naive adults. J. Infect. Dis. 2015;212:702–710. doi: 10.1093/infdis/jiv082.
    1. Gallichotte EN, et al. Genetic variation between dengue virus type 4 strains impacts human antibody binding and neutralization. Cell Rep. 2018;25:1214–1224. doi: 10.1016/j.celrep.2018.10.006.
    1. Fibriansah G, et al. A potent anti-dengue human antibody preferentially recognizes the conformation of E protein monomers assembled on the virus surface. EMBO Mol. Med. 2014;6:358–371. doi: 10.1002/emmm.201303404.
    1. Fibriansah G, et al. DENGUE VIRUS. Cryo-EM structure of an antibody that neutralizes dengue virus type 2 by locking E protein dimers. Science. 2015;349:88–91. doi: 10.1126/science.aaa8651.
    1. Fibriansah G, et al. A highly potent human antibody neutralizes dengue virus serotype 3 by binding across three surface proteins. Nat. Commun. 2015;6:6341. doi: 10.1038/ncomms7341.
    1. Nivarthi, U. K. et al. Mapping the human memory B cell and serum neutralizing antibody responses to dengue virus serotype 4 infection and vaccination. J. Virol. 9110.1128/JVI.02041-16 (2017).
    1. Swanstrom JA, et al. Beyond neutralizing antibody levels: the epitope specificity of antibodies induced by national institutes of health monovalent dengue virus vaccines. J. Infect. Dis. 2019;220:219–227. doi: 10.1093/infdis/jiz109.
    1. Gallichotte EN, et al. A new quaternary structure epitope on dengue virus serotype 2 is the target of durable type-specific neutralizing antibodies. MBio. 2015;6:e01461-01415. doi: 10.1128/mBio.01461-15.
    1. Gallichotte EN, et al. Human dengue virus serotype 2 neutralizing antibodies target two distinct quaternary epitopes. PLoS Pathog. 2018;14:e1006934. doi: 10.1371/journal.ppat.1006934.
    1. Widman DG, et al. Transplantation of a quaternary structure neutralizing antibody epitope from dengue virus serotype 3 into serotype 4. Sci. Rep. 2017;7:17169. doi: 10.1038/s41598-017-17355-5.
    1. Lai CJ, et al. Epitope determinants of a chimpanzee dengue virus type 4 (DENV-4)-neutralizing antibody and protection against DENV-4 challenge in mice and rhesus monkeys by passively transferred humanized antibody. J. Virol. 2007;81:12766–12774. doi: 10.1128/JVI.01420-07.
    1. Dejnirattisai W, et al. A new class of highly potent, broadly neutralizing antibodies isolated from viremic patients infected with dengue virus. Nat. Immunol. 2015;16:170–177. doi: 10.1038/ni.3058.
    1. Smith SA, et al. Isolation of dengue virus-specific memory B cells with live virus antigen from human subjects following natural infection reveals the presence of diverse novel functional groups of antibody clones. J. Virol. 2014;88:12233–12241. doi: 10.1128/JVI.00247-14.
    1. Guy B, Briand O, Lang J, Saville M, Jackson N. Development of the Sanofi Pasteur tetravalent dengue vaccine: one more step forward. Vaccine. 2015;33:7100–7111. doi: 10.1016/j.vaccine.2015.09.108.
    1. Osorio JE, et al. Safety and immunogenicity of a recombinant live attenuated tetravalent dengue vaccine (DENVax) in flavivirus-naive healthy adults in Colombia: a randomised, placebo-controlled, phase 1 study. Lancet Infect. Dis. 2014;14:830–838. doi: 10.1016/S1473-3099(14)70811-4.
    1. Osorio JE, Wallace D, Stinchcomb DT. A recombinant, chimeric tetravalent dengue vaccine candidate based on a dengue virus serotype 2 backbone. Expert Rev. Vaccines. 2016;15:497–508. doi: 10.1586/14760584.2016.1128328.
    1. Moodie Z, et al. Neutralizing antibody correlates analysis of tetravalent dengue vaccine efficacy trials in Asia and Latin America. J. Infect. Dis. 2018;217:742–753. doi: 10.1093/infdis/jix609.
    1. Biswal S, et al. Efficacy of a tetravalent dengue vaccine in healthy children and adolescents. N. Engl. J. Med. 2019;381:2009–2019. doi: 10.1056/NEJMoa1903869.
    1. Swanstrom JA, et al. Beyond neutralizing antibody levels: the epitope specificity of antibodies induced by NIH monovalent dengue virus vaccines. J. Infect. Dis. 2019 doi: 10.1093/infdis/jiz109.
    1. Lindow JC, et al. Vaccination of volunteers with low-dose, live-attenuated, dengue viruses leads to serotype-specific immunologic and virologic profiles. Vaccine. 2013;31:3347–3352. doi: 10.1016/j.vaccine.2013.05.075.
    1. Wahala WM, Kraus AA, Haymore LB, Accavitti-Loper MA, de Silva AM. Dengue virus neutralization by human immune sera: role of envelope protein domain III-reactive antibody. Virology. 2009;392:103–113. doi: 10.1016/j.virol.2009.06.037.
    1. de Alwis R, et al. Identification of human neutralizing antibodies that bind to complex epitopes on dengue virions. Proc. Natl Acad. Sci. USA. 2012;109:7439–7444. doi: 10.1073/pnas.1200566109.
    1. Swanstrom, J. A. et al. Dengue virus envelope dimer epitope monoclonal antibodies isolated from dengue patients are protective against Zika virus. MBio7, 10.1128/mBio.01123-16 (2016).
    1. Collins MH, et al. Lack of durable cross-neutralizing antibodies against Zika virus from dengue virus infection. Emerg. Infect. Dis. 2017;23:773–781. doi: 10.3201/eid2305.161630.
    1. Messer WB, et al. Functional transplant of a dengue virus serotype 3 (DENV3)-specific human monoclonal antibody epitope into DENV1. J. Virol. 2016;90:5090–5097. doi: 10.1128/JVI.00155-16.
    1. Messer WB, et al. Development and characterization of a reverse genetic system for studying dengue virus serotype 3 strain variation and neutralization. PLoS Negl. Trop. Dis. 2012;6:e1486. doi: 10.1371/journal.pntd.0001486.

Source: PubMed

3
Iratkozz fel