Defining levels of dengue virus serotype-specific neutralizing antibodies induced by a live attenuated tetravalent dengue vaccine (TAK-003)

Laura J White, Ellen F Young, Mark J Stoops, Sandra R Henein, Elizabeth C Adams, Ralph S Baric, Aravinda M de Silva, Laura J White, Ellen F Young, Mark J Stoops, Sandra R Henein, Elizabeth C Adams, Ralph S Baric, Aravinda M de Silva

Abstract

The four dengue virus serotypes (DENV1-4) infect several hundred million people each year living in tropical and sub-tropical regions. Clinical development of DENV vaccines is difficult because immunity to a single serotype increases risk of severe disease during a second infection with a new serotype. Leading vaccines are based on tetravalent formulations to induce simultaneous and balanced protective immunity to all 4 serotypes. TAK-003 is a tetravalent live attenuated dengue vaccine candidate developed by Takeda Vaccines Inc, which is currently being evaluated in phase 3 efficacy trials. Here, we use antibody depletion methods and chimeric, epitope transplant DENVs to characterize the specificity of neutralizing antibodies in dengue-naïve adults and non-human primates immunized with TAK-003. Our results demonstrate that TAK-003 induced high levels of DENV2 neutralizing antibodies that recognized unique (type-specific) epitopes on DENV2. In contrast, most vaccinated subjects developed lower levels of DENV1, DENV3 and DENV4 neutralizing antibodies that mainly targeted epitopes that were conserved (cross-reactive) between serotypes. Trial Registration: ClinicalTrials.gov NCT02425098.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: RB and AdS have served as consultants for dengue vaccine developers and they are inventors on patents filed by the University of North Carolina at Chapel Hill related to dengue vaccines.

Figures

Fig 1
Fig 1
50% Neutralization titers (Neut50) in undepleted (A) and Heterologous virus-depleted (B) baseline seronegative NHP vaccinated with TDV (left) or infected with WT DV1, DV2 or DV3 (right). Non-human primates were vaccinated with one dose (filled circles) or two doses (half filled circles) of TDV, or infected once with wild type DV1, DV2 or DV3 (S2 Table). Animals were bled 180 days after infection or last vaccination. Each sample underwent BSA-depletion (undepleted control) (A), depletion of cross-reactive nAbs by incubation with heterologous serotype viruses (HET depleted) (B) and depletion with the homologous serotype, before determining Neut50 titers by mFRNT assay. Each value represents the titer after subtraction of the background homologous-depletion titer. Each dot represents one animal. Line represents the GMT of the Neut50 titers. The lowest serum dilution tested was 20 (horizontal dotted line). Statistical analysis was done using nonparametric Friedman test with Dunn’s multiple comparison among serotypes after TDV vaccine. * P < 0.05; ** P < 0,005; *** P < 0.0005.
Fig 2
Fig 2
50% Neutralization titers (Neut50) in undepleted (A) and Heterologous virus-depleted (B) baseline seronegative subjects vaccinated with one dose of TDV (left) or after exposure to primary DV1 or DV2 infection (right). The TDV vaccine cohort included sera from 30 subjects collected 180 days after TDV vaccination. The primary infection cohort included 6 primary DENV1 and 8 primary DENV2, collected at baseline in study DEN-205. Each sample was BSA-depleted (Undepleted control) (A) or depleted of cross-reactive nAbs using heterologous serotype viruses (HET Depleted) (B). To measure DV1 and DV3 TS neutralization, HET depletion was done with a mix of DV2+DV4 dynabeads. For DV2 and DV4 TS titers, Het depletion was done with a mix of DV1+DV3 dynabeads. Each value represents the titer after subtraction of background homologous depletion titer. Each dot represents one subjects. Line represents the GMT of the Neut50 titers. The # of subjects with evidence of TS nAbs >20 after HET depletion/#total subjects analyzed is shown at the bottom of panel B. % TS is the percent of subjects with TS nAbs = or > 20. The lowest serum dilution tested was 20 (horizontal dotted line). Statistical analysis was done using nonparametric Friedman test for multiple comparison among serotypes after TDV vaccine. ** P < 0,005; **** P < 0.0001.
Fig 3. Analysis of the levels of…
Fig 3. Analysis of the levels of TS nAbs in a subset of DEN-205 clinical samples that met enhanced inclusion criteria for TS analysis.
Samples were included in this analysis only if the efficiency of Ab depletion for the serotype used as heterologous was >90%, or if the HET-depleted titer was

Fig 4

Tracking the molecular specificity of…

Fig 4

Tracking the molecular specificity of DV2 TS nAbs to epitopes on EDIII in…
Fig 4
Tracking the molecular specificity of DV2 TS nAbs to epitopes on EDIII in sera from TDV vaccinated NHPs (A), WT DV2 infected NHPs (B), TDV vaccinated, baseline seronegative adults (C) or adults who had a DV2 natural infection (D). Neut50 titers to parental viruses DV2 (EDIII epitope donor), DV4 (backbone recipient of epitope transplant) and chimeric virus DV4/2EDIII (DV2 EDIII transplanted into DV4 E protein) were determined in sera collected 180 days after infection or vaccination (A, B, C). Sera used in (D) were collected in subjects pre-TDV vaccination, with evidence of past primary DENV2 infection based on Neut50 titers to DV2 > 4-fold higher than those to the other 3 serotypes. (A) (B) and (D) show titers in control-depleted (BSA-D) and DV4-depleted sera. (C) shows titers in undepleted sera. Horizontal lines and error bars represent GMT +/- 95% CI. Dotted line represents lower limit of detection.
Fig 4
Fig 4
Tracking the molecular specificity of DV2 TS nAbs to epitopes on EDIII in sera from TDV vaccinated NHPs (A), WT DV2 infected NHPs (B), TDV vaccinated, baseline seronegative adults (C) or adults who had a DV2 natural infection (D). Neut50 titers to parental viruses DV2 (EDIII epitope donor), DV4 (backbone recipient of epitope transplant) and chimeric virus DV4/2EDIII (DV2 EDIII transplanted into DV4 E protein) were determined in sera collected 180 days after infection or vaccination (A, B, C). Sera used in (D) were collected in subjects pre-TDV vaccination, with evidence of past primary DENV2 infection based on Neut50 titers to DV2 > 4-fold higher than those to the other 3 serotypes. (A) (B) and (D) show titers in control-depleted (BSA-D) and DV4-depleted sera. (C) shows titers in undepleted sera. Horizontal lines and error bars represent GMT +/- 95% CI. Dotted line represents lower limit of detection.

References

    1. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, et al.. The global distribution and burden of dengue. Nature. 2013;496(7446):504–7. 10.1038/nature12060
    1. Radke EG, Gregory CJ, Kintziger KW, Sauber-Schatz EK, Hunsperger EA, Gallagher GR, et al.. Dengue outbreak in Key West, Florida, USA, 2009. Emerg Infect Dis. 2012;18(1):135–7. 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(12):1271–80. 10.1016/S1473-3099(14)70834-5
    1. Montoya M, Gresh L, Mercado JC, Williams KL, Vargas MJ, Gutierrez G, 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(8):e2357. 10.1371/journal.pntd.0002357
    1. Sabin AB. Research on dengue during World War II. Am J Trop Med Hyg. 1952;1(1):30–50. 10.4269/ajtmh.1952.1.30
    1. Halstead SB. Etiologies of the experimental dengues of Siler and Simmons. Am J Trop Med Hyg. 1974;23(5):974–82. 10.4269/ajtmh.1974.23.974
    1. Wahala WM, Silva AM. The human antibody response to dengue virus infection. Viruses. 2011;3(12):2374–95. 10.3390/v3122374
    1. Katzelnick LC, Montoya M, Gresh L, Balmaseda A, Harris E. Neutralizing antibody titers against dengue virus correlate with protection from symptomatic infection in a longitudinal cohort. Proc Natl Acad Sci U S A. 2016;113(3):728–33. 10.1073/pnas.1522136113
    1. de Alwis R, Beltramello M, Messer WB, Sukupolvi-Petty S, Wahala WM, Kraus A, et al.. In-depth analysis of the antibody response of individuals exposed to primary dengue virus infection. PLoS Negl Trop Dis. 2011;5(6):e1188. 10.1371/journal.pntd.0001188
    1. de Alwis R, Williams KL, Schmid MA, Lai CY, Patel B, Smith SA, et al.. Dengue viruses are enhanced by distinct populations of serotype cross-reactive antibodies in human immune sera. PLoS Pathog. 2014;10(10):e1004386. 10.1371/journal.ppat.1004386
    1. Appanna R, Kg S, Xu MH, Toh YX, Velumani S, Carbajo D, et al.. Plasmablasts During Acute Dengue Infection Represent a Small Subset of a Broader Virus-specific Memory B Cell Pool. EBioMedicine. 2016;12:178–88. 10.1016/j.ebiom.2016.09.003
    1. Patel B, Longo P, Miley MJ, Montoya M, Harris E, de Silva AM. Dissecting the human serum antibody response to secondary dengue virus infections. PLoS Negl Trop Dis. 2017;11(5):e0005554. 10.1371/journal.pntd.0005554
    1. Dejnirattisai W, Wongwiwat W, Supasa S, Zhang X, Dai X, Rouvinski A, et al.. A new class of highly potent, broadly neutralizing antibodies isolated from viremic patients infected with dengue virus. Nat Immunol. 2015;16(2):170–7. 10.1038/ni.3058
    1. Tsai WY, Lai CY, Wu YC, Lin HE, Edwards C, Jumnainsong A, et al.. High-avidity and potently neutralizing cross-reactive human monoclonal antibodies derived from secondary dengue virus infection. J Virol. 2013;87(23):12562–75. 10.1128/JVI.00871-13
    1. Weiskopf D, Angelo MA, Sidney J, Peters B, Shresta S, Sette A. Immunodominance changes as a function of the infecting dengue virus serotype and primary versus secondary infection. J Virol. 2014;88(19):11383–94. 10.1128/JVI.01108-14
    1. de Silva AM, Harris E. Which Dengue Vaccine Approach Is the Most Promising, and Should We Be Concerned about Enhanced Disease after Vaccination? The Path to a Dengue Vaccine: Learning from Human Natural Dengue Infection Studies and Vaccine Trials. Cold Spring Harb Perspect Biol. 2018;10(6).
    1. Capeding MR, Tran NH, Hadinegoro SR, Ismail HI, Chotpitayasunondh T, Chua MN, 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(9951):1358–65. 10.1016/S0140-6736(14)61060-6
    1. Hadinegoro SR, Arredondo-Garcia JL, Capeding MR, Deseda C, Chotpitayasunondh T, Dietze R, et al.. Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease. N Engl J Med. 2015;373(13):1195–206. 10.1056/NEJMoa1506223
    1. Sridhar S, Luedtke A, Langevin E, Zhu M, Bonaparte M, Machabert T, et al.. Effect of Dengue Serostatus on Dengue Vaccine Safety and Efficacy. N Engl J Med. 2018;379(4):327–40. 10.1056/NEJMoa1800820
    1. Barban V, Mantel N, De Montfort A, Pagnon A, Pradezynski F, Lang J, et al.. Improvement of the Dengue Virus (DENV) Nonhuman Primate Model via a Reverse Translational Approach Based on Dengue Vaccine Clinical Efficacy Data against DENV-2 and -4. J Virol. 2018;92(12).
    1. Guirakhoo F, Pugachev K, Zhang Z, Myers G, Levenbook I, Draper K, et al.. Safety and efficacy of chimeric yellow Fever-dengue virus tetravalent vaccine formulations in nonhuman primates. J Virol. 2004;78(9):4761–75. 10.1128/jvi.78.9.4761-4775.2004
    1. Guy B. Immunogenicity of sanofi pasteur tetravalent dengue vaccine. J Clin Virol. 2009;46 Suppl 2:S16–9. 10.1016/S1386-6532(09)70290-2
    1. Guy B, Barban V, Mantel N, Aguirre M, Gulia S, Pontvianne J, et al.. Evaluation of interferences between dengue vaccine serotypes in a monkey model. Am J Trop Med Hyg. 2009;80(2):302–11.
    1. Henein S, Swanstrom J, Byers AM, Moser JM, Shaik SF, Bonaparte M, 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(3):351–8. 10.1093/infdis/jiw576
    1. Biswal S, Reynales H, Saez-Llorens X, Lopez P, Borja-Tabora C, Kosalaraksa P, et al.. Efficacy of a Tetravalent Dengue Vaccine in Healthy Children and Adolescents. N Engl J Med. 2019;381(21):2009–19. 10.1056/NEJMoa1903869
    1. Biswal S, Borja-Tabora C, Martinez Vargas L, Velasquez H, Theresa Alera M, Sierra V, et al.. Efficacy of a tetravalent dengue vaccine in healthy children aged 4–16 years: a randomised, placebo-controlled, phase 3 trial. Lancet. 2020. 10.1016/S0140-6736(20)30414-1
    1. Osorio JE, Brewoo JN, Silengo SJ, Arguello J, Moldovan IR, Tary-Lehmann M, et al.. Efficacy of a tetravalent chimeric dengue vaccine (DENVax) in Cynomolgus macaques. Am J Trop Med Hyg. 2011;84(6):978–87. 10.4269/ajtmh.2011.10-0592
    1. Rupp R, Luckasen GJ, Kirstein JL, Osorio JE, Santangelo JD, Raanan M, et al.. Safety and immunogenicity of different doses and schedules of a live attenuated tetravalent dengue vaccine (TDV) in healthy adults: A Phase 1b randomized study. Vaccine. 2015;33(46):6351–9. 10.1016/j.vaccine.2015.09.008
    1. Tricou V, Low JG, Oh HM, Leo YS, Kalimuddin S, Wijaya L, et al.. Safety and immunogenicity of a single dose of a tetravalent dengue vaccine with two different serotype-2 potencies in adults in Singapore: A phase 2, double-blind, randomised, controlled trial. Vaccine. 2020;38(6):1513–9. 10.1016/j.vaccine.2019.11.061
    1. Gallichotte EN, Widman DG, Yount BL, Wahala WM, Durbin A, Whitehead S, et al.. A new quaternary structure epitope on dengue virus serotype 2 is the target of durable type-specific neutralizing antibodies. mBio. 2015;6(5):e01461–15. 10.1128/mBio.01461-15
    1. Swanstrom JA, Nivarthi UK, Patel B, Delacruz MJ, Yount B, Widman DG, 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(2):219–27. 10.1093/infdis/jiz109
    1. Collins MH, McGowan E, Jadi R, Young E, Lopez CA, Baric RS, et al.. Lack of Durable Cross-Neutralizing Antibodies Against Zika Virus from Dengue Virus Infection. Emerg Infect Dis. 2017;23(5):773–81. 10.3201/eid2305.161630
    1. Sariol CA, White LJ. Utility, limitations, and future of non-human primates for dengue research and vaccine development. Front Immunol. 2014;5:452. 10.3389/fimmu.2014.00452
    1. Gallichotte EN, Baric TJ, Yount BL Jr, Widman DG, Durbin A, Whitehead S, et al.. Human dengue virus serotype 2 neutralizing antibodies target two distinct quaternary epitopes. PLoS Pathog. 2018;14(2):e1006934. 10.1371/journal.ppat.1006934
    1. Nivarthy U; Durbin A; Whitehead S; Kirkpatrick B; Pierce K; Diehl S; Katzelnick L; Baric R; deSilva A. A tetravalent live attenuated dengue virus vaccine stimulates balanced immunity to multiple serotypes in humans. Accepted in Nature Communications. 2021.
    1. Swanstrom JA, Henein S, Plante JA, Yount BL, Widman DG, Gallichotte EN, et al.. Analyzing the Human Serum Antibody Responses to a Live Attenuated Tetravalent Dengue Vaccine Candidate. J Infect Dis. 2018;217(12):1932–41. 10.1093/infdis/jiy063
    1. Anderson KB, Gibbons RV, Cummings DA, Nisalak A, Green S, Libraty DH, et al.. A shorter time interval between first and second dengue infections is associated with protection from clinical illness in a school-based cohort in Thailand. J Infect Dis. 2014;209(3):360–8. 10.1093/infdis/jit436

Source: PubMed

3
Iratkozz fel