Antibody effector analysis of prime versus prime-boost immunizations with a recombinant measles-vectored chikungunya virus vaccine

Roland Tschismarov, Raphaël M Zellweger, Min Jie Koh, Yan Shan Leong, Jenny G Low, Eng Eong Ooi, Christian W Mandl, Katrin Ramsauer, Ruklanthi de Alwis, Roland Tschismarov, Raphaël M Zellweger, Min Jie Koh, Yan Shan Leong, Jenny G Low, Eng Eong Ooi, Christian W Mandl, Katrin Ramsauer, Ruklanthi de Alwis

Abstract

Chikungunya is a mosquito-borne disease that causes periodic but explosive epidemics of acute disease throughout the tropical world. Vaccine development against chikungunya virus (CHIKV) has been hampered by an inability to conduct efficacy trials due to the unpredictability of CHIKV outbreaks. Therefore, immune correlates are being explored to gain inference into vaccine-induced protection. This study is an in-depth serological characterization of Fab- and Fc-mediated antibody responses in selected phase II clinical trial participants following immunization with the recombinant measles-vectored CHIKV vaccine, MV-CHIK. Antibody comparisons were conducted between participants who received prime and those who received prime-boost vaccine regimens. MV-CHIK vaccination elicited potent Fab-mediated antibody responses (such as CHIKV-specific IgG, neutralization, and avidity), including dominant IgG3 responses, which translated into strong antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. At 1 month, prime-boost immunization led to significantly greater responses in every measured Fab and Fc antibody parameter. Interestingly, prime-boost-elicited antibodies decreased rapidly over time, until at 6 months both vaccine regimens displayed similar antibody profiles. Nonetheless, antibody avidity and antibody-dependent cellular phagocytosis remained significantly greater following boost immunization. Our observations suggest that a prime-boost administration of MV-CHIK will be more appropriate for CHIKV-endemic regions, while a prime-only regimen may be sufficient for travel purposes or outbreak situations.

Trial registration: ClinicalTrials.gov NCT02861586.

Keywords: Adaptive immunity; Immunology; Vaccines.

Conflict of interest statement

Conflict of interest: This study was partially funded by Themis Bioscience GmbH, a subsidiary of Merck & Co. Inc. RT and KR are employees of Themis Bioscience GmbH.

Figures

Figure 1. Study design and characterization of…
Figure 1. Study design and characterization of the CHIK-specific IgG response elicited by MV-CHIK prime and prime-boost vaccination strategies.
(A) Participants were vaccinated with either MV-CHIK prime at day 0 (P only) or prime (at month –1) and boost (at day 0) (P+B), and serum antibody responses were compared at 1 and 6 months after vaccination. MV-vaccinated groups served as controls. (B–F) Total IgG binding to CHIKV virus was measured using ELISA and compared (B) between groups and (C) at 1 and 6 months for paired MV-CHIK–vaccinated samples. (D) Fold change in IgG (mean ± 95% CI) in response to CHIKV virus was compared between MV-CHIK P only and P+B. IgG binding to 2 CHIK surface glycoproteins, E1 and E2, was measured using Luminex immunoassay and compared between groups at (E) 1 month and (F) 6 months after vaccination. (G) Distribution of the CHIK antigens targeted by the antibody response was estimated by the ratio of E2-specific to E1-specific IgG (mean ± 95% CI) and compared between prime and prime-boost MV-CHIK vaccinations. IgG responses between groups were compared using Mann-Whitney U test, while paired analysis between groups at 1 and 6 months was conducted using paired Wilcoxon signed-rank test (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001).
Figure 2. Avidity and neutralizing capacity of…
Figure 2. Avidity and neutralizing capacity of CHIK virus–specific antibody responses elicited by MV-CHIK prime and prime-boost immunization.
(A) Binding strength of CHIK virus–specific antibodies, as indicated by avidity index estimations following 4, 6, and 8 M urea washes. (B) CHIK virus neutralization (measured by PRNT assay) and (C) neutralization normalized to total IgG (mean ± 95% CI) compared between vaccinated groups. (D and E) Longitudinal assessment of the CHIKV-neutralizing antibody response using (D) paired analysis between 1 and 6 months and (E) fold change in CHIKV neutralization from 1 to 6 months (mean ± 95% CI) in MV-CHIK prime (P) and prime-boost (P+B) immunization. Avidity index and PRNT50 titers were compared between groups using Mann-Whitney U test, and analysis between groups at 1 and 6 months was conducted using paired Wilcoxon signed-rank test (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001).
Figure 3. IgG subclass distribution of CHIK…
Figure 3. IgG subclass distribution of CHIK virus–specific antibody responses following MV-CHIK prime and prime-boost vaccinations.
(A) IgG1, IgG2, IgG3, and IgG4 CHIK virus–binding endpoint titers (measured by ELISA) in MV-CHIK–vaccinated individuals. (B and C) Longitudinal analysis of CHIK virus–specific IgG1 and IgG3. (B) Endpoint titers at 1 and 6 months. (C) Fold change in antibody titers from 1 to 6 months (mean ± 95% CI). (D–G) The ratio of each IgG subclass (IgG1, IgG2, IgG3, and IgG4) to total IgG, and (H) the ratio of IgG1 to IgG2 (mean ± 95% CI). IgG subclass titers were compared between groups using Mann-Whitney U test, and comparisons between groups at 1 and 6 months used paired Wilcoxon signed-rank test (**P < 0.01, ***P < 0.001, ****P < 0.0001).
Figure 4. Antibody-dependent cellular phagocytosis of CHIK…
Figure 4. Antibody-dependent cellular phagocytosis of CHIK virus by a monocytic cell line following vaccination with MV-CHIK.
(A) Phagocytic scores (measured using a whole-virus phagocytic assay) compared between vaccinated groups. (B) Paired longitudinal antibody-dependent cellular phagocytosis (ADCP) responses compared at 1 and 6 months. (C) Normalized phagocytic scores (i.e., phagocytic scores normalized to CHIKV-specific IgG titers) compared between P- and P+B MC-CHIK–vaccinated groups. Phagocytic scores and normalized phagocytic scores were compared between groups using Mann-Whitney U test, and paired analysis between groups at 1 and 6 months was conducted using paired Wilcoxon signed-rank test (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001).
Figure 5. NK-mediated ADCC following MV-CHIK vaccination.
Figure 5. NK-mediated ADCC following MV-CHIK vaccination.
ADCC was assessed through the proxy measures of ADNKA degranulation (CD107α) and cytokine release (IFN-γ). (A) Percentage of CD107α+ cells and (B) IFN-γ+ cells in vaccinated groups. Paired longitudinal analysis of the percentage of (C) CD107α+ cells and (D) IFN-γ+ cells at 1 and 6 months after vaccination. Normalized percentage of (E) CD107α+ cells and (F) IFN-γ+ cells (to CHIKV-specific IgG titers) in P- and P+B MV-CHIK–vaccinated groups. Percentages and normalized percentages of CD17α+ and IFN-γ+ were compared between groups using Mann-Whitney U test, and paired analysis between groups at 1 and 6 months was conducted using paired Wilcoxon signed-rank test (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001).
Figure 6. Multidimensional unsupervised analysis of MV-CHIK–vaccinated…
Figure 6. Multidimensional unsupervised analysis of MV-CHIK–vaccinated groups and measured antibody phenotypes.
(A and B) Principal component analysis (PCA) was conducted on MV-CHIK vaccine–induced antibody responses following prime (P) and prime-boost (P+B) regiments at 1 and 6 months. (A) Individual plot with vaccine groups highlighted in different colors. Ellipses in the PCA individual plot represent the confidence ellipses around the centroid (marked by the larger symbol for each group) of each vaccine group. (B) Circle plot highlighting the variables contributing to the spread of points. (C) Correlation matrix with hierarchical clustering of variables utilized in the PCA analysis. Variables included in the PCA and hierarchical correlation matrix include neutralization (PRNT50 titers); virus-binding total IgG, IgG1, IgG2, IgG3, and IgG4 (endpoint titers); E1-binding IgG (MFI); E2-binding IgG (MFI); avidity at 8 M (AI); ADCP (phagocytic score); and ADCC (ADNKA degranulation as percentage of CD107α+ cells, and ADNKA cytokine release as percentage of IFN-γ+ cells).

References

    1. Schrauf S, et al. Current efforts in the development of vaccines for the prevention of Zika and Chikungunya virus infections. Front Immunol. 2020;11:592. doi: 10.3389/fimmu.2020.00592.
    1. Rezza G, Weaver SC. Chikungunya as a paradigm for emerging viral diseases: evaluating disease impact and hurdles to vaccine development. PLoS Negl Trop Dis. 2019;13(1):e0006919.
    1. Nsoesie EO, et al. Global distribution and environmental suitability for Chikungunya virus, 1952 to 2015. Euro Surveill. 2016;21(20)
    1. Microbe TL. Arboviruses and COVID-19: the need for a holistic view. Lancet Microbe. 2020;1(4)
    1. Erasmus JH, et al. Development of vaccines for Chikungunya fever. J Infect Dis. 2016;214(suppl 5):S488–S496. doi: 10.1093/infdis/jiw271.
    1. Gao S, et al. Recent progress in vaccine development against Chikungunya virus. Front Microbiol. 2019;10:2881. doi: 10.3389/fmicb.2019.02881.
    1. Food and Drug Administration Center for Biologics Evaluation and Research. Summary Minutes 158th Vaccines and Related Biological Products Advisory Committee. Accessed October 6, 2021.
    1. Yoon IK, et al. High rate of subclinical Chikungunya virus infection and association of neutralizing antibody with protection in a prospective cohort in the Philippines. PLoS Negl Trop Dis. 2015;9(5):e0003764. doi: 10.1371/journal.pntd.0003764.
    1. Yoon IK, et al. Pre-existing Chikungunya virus neutralizing antibodies correlate with risk of symptomatic infection and subclinical seroconversion in a Philippine cohort. Int J Infect Dis. 2020;95:167–173. doi: 10.1016/j.ijid.2020.03.073.
    1. Auerswald H, et al. Broad and long-lasting immune protection against various Chikungunya genotypes demonstrated by participants in a cross-sectional study in a Cambodian rural community. Emerg Microbes Infect. 2018;7(1):13.
    1. Galatas B, et al. Long-lasting immune protection and other epidemiological findings after Chikungunya emergence in a Cambodian rural community, April 2012. PLoS Negl Trop Dis. 2016;10(1):e0004281. doi: 10.1371/journal.pntd.0004281.
    1. Akahata W, et al. A virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat Med. 2010;16(3):334–338. doi: 10.1038/nm.2105.
    1. Brandler S, et al. A recombinant measles vaccine expressing Chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with Chikungunya virus. Vaccine. 2013;31(36):3718–3725. doi: 10.1016/j.vaccine.2013.05.086.
    1. Kumar M, et al. Evaluation of recombinant E2 protein-based and whole-virus inactivated candidate vaccines against Chikungunya virus. Vaccine. 2012;30(43):6142–6149. doi: 10.1016/j.vaccine.2012.07.072.
    1. Langsjoen RM, et al. Chikungunya virus strains show lineage-specific variations in virulence and cross-protective ability in murine and nonhuman primate models. mBio. 2018;9(2):e02449–17.
    1. Arnold KB, Chung AW. Prospects from systems serology research. Immunology. 2018;153(3):279–289. doi: 10.1111/imm.12861.
    1. Tay MZ, et al. Antibody-dependent cellular phagocytosis in antiviral immune responses. Front Immunol. 2019;10:332. doi: 10.3389/fimmu.2019.00332.
    1. Chung AW, et al. Dissecting polyclonal vaccine-induced humoral immunity against HIV using systems serology. Cell. 2015;163(4):988–998. doi: 10.1016/j.cell.2015.10.027.
    1. He W, et al. Epitope specificity plays a critical role in regulating antibody-dependent cell-mediated cytotoxicity against influenza A virus. Proc Natl Acad Sci U S A. 2016;113(42):11931–11936. doi: 10.1073/pnas.1609316113.
    1. Jegaskanda S, et al. Influenza-specific antibody-dependent cellular cytotoxicity: toward a universal influenza vaccine. J Immunol. 2014;193(2):469–475. doi: 10.4049/jimmunol.1400432.
    1. Gunn BM, et al. A role for Fc function in therapeutic monoclonal antibody-mediated protection against Ebola virus. Cell Host Microbe. 2018;24(2):221–233. doi: 10.1016/j.chom.2018.07.009.
    1. Fox JM, et al. Optimal therapeutic activity of monoclonal antibodies against Chikungunya virus requires Fc-FcγR interaction on monocytes. Sci Immunol. 2019;4(32):eaav5062. doi: 10.1126/sciimmunol.aav5062.
    1. Jin J, et al. Neutralizing antibodies inhibit Chikungunya virus budding at the plasma membrane. Cell Host Microbe. 2018;24(3):417–428. doi: 10.1016/j.chom.2018.07.018.
    1. Wressnigg N, et al. Single-shot live-attenuated Chikungunya vaccine in healthy adults: a phase 1, randomised controlled trial. Lancet Infect Dis. 2020;20(10):1193–1203. doi: 10.1016/S1473-3099(20)30238-3.
    1. Chang LJ, et al. Safety and tolerability of Chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation trial. Lancet. 2014;384(9959):2046–2052. doi: 10.1016/S0140-6736(14)61185-5.
    1. Ramsauer K, et al. Immunogenicity, safety, and tolerability of a recombinant measles-virus-based Chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. Lancet Infect Dis. 2015;15(5):519–527. doi: 10.1016/S1473-3099(15)70043-5.
    1. Reisinger EC, et al. Immunogenicity, safety, and tolerability of the measles-vectored Chikungunya virus vaccine MV-CHIK: a double-blind, randomised, placebo-controlled and active-controlled phase 2 trial. Lancet. 2019;392(10165):2718–2727.
    1. Kam YW, et al. Longitudinal analysis of the human antibody response to Chikungunya virus infection: implications for serodiagnosis and vaccine development. J Virol. 2012;86(23):13005–13015. doi: 10.1128/JVI.01780-12.
    1. Schiller JT. The potential benefits of HPV vaccination in previously infected women. EBioMedicine. 2016;10:5–6. doi: 10.1016/j.ebiom.2016.08.005.
    1. Eisen HN. Affinity enhancement of antibodies: how low-affinity antibodies produced early in immune responses are followed by high-affinity antibodies later and in memory B-cell responses. Cancer Immunol Res. 2014;2(5):381–392. doi: 10.1158/2326-6066.CIR-14-0029.
    1. Rodrigo WW, et al. Dengue virus neutralization is modulated by IgG antibody subclass and Fcgamma receptor subtype. Virology. 2009;394(2):175–182. doi: 10.1016/j.virol.2009.09.024.
    1. Richardson SI, et al. IgG3 enhances neutralization potency and Fc effector function of an HIV V2-specific broadly neutralizing antibody. PLoS Pathog. 2019;15(12):e1008064. doi: 10.1371/journal.ppat.1008064.
    1. Kam YW, et al. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with Chikungunya virus clearance and long-term clinical protection. J Infect Dis. 2012;205(7):1147–1154. doi: 10.1093/infdis/jis033.
    1. Kam YW, et al. Early neutralizing IgG response to Chikungunya virus in infected patients targets a dominant linear epitope on the E2 glycoprotein. EMBO Mol Med. 2012;4(4):330–343. doi: 10.1002/emmm.201200213.
    1. Rossi SL, et al. Immunogenicity and efficacy of a measles virus-vectored Chikungunya vaccine in nonhuman primates. J Infect Dis. 2019;220(5):735–742. doi: 10.1093/infdis/jiz202.
    1. Akkaya M, et al. B cell memory: building two walls of protection against pathogens. Nat Rev Immunol. 2020;20(4):229–238. doi: 10.1038/s41577-019-0244-2.
    1. Mesin L, et al. Restricted clonality and limited germinal center reentry characterize memory B cell reactivation by boosting. Cell. 2020;180(1):92–106. doi: 10.1016/j.cell.2019.11.032.
    1. Hepburn MJ, et al. Neutralizing antibody response to booster vaccination with the 17d yellow fever vaccine. Vaccine. 2006;24(15):2843–2849. doi: 10.1016/j.vaccine.2005.12.055.
    1. Fong RH, et al. Exposure of epitope residues on the outer face of the Chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol. 2014;88(24):14364–14379. doi: 10.1128/JVI.01943-14.
    1. Goh LY, et al. Neutralizing monoclonal antibodies to the E2 protein of Chikungunya virus protects against disease in a mouse model. Clin Immunol. 2013;149(3):487–497. doi: 10.1016/j.clim.2013.10.004.
    1. Jin J, et al. Neutralizing monoclonal antibodies block Chikungunya virus entry and release by targeting an epitope critical to viral pathogenesis. Cell Rep. 2015;13(11):2553–2564. doi: 10.1016/j.celrep.2015.11.043.
    1. Henss L, et al. Analysis of humoral immune responses in Chikungunya virus (CHIKV)-infected patients and individuals vaccinated with a candidate CHIKV vaccine. J Infect Dis. 2020;221(10):1713–1723. doi: 10.1093/infdis/jiz658.
    1. Vidarsson G, et al. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014;5:520.
    1. Alter G, et al. Antibody glycosylation in inflammation, disease and vaccination. Semin Immunol. 2018;39:102–110. doi: 10.1016/j.smim.2018.05.003.
    1. Quast I, et al. Regulation of antibody effector functions through IgG Fc N-glycosylation. Cell Mol Life Sci. 2017;74(5):837–847. doi: 10.1007/s00018-016-2366-z.
    1. Wang J, et al. Fc-glycosylation of IgG1 is modulated by B-cell stimuli. Mol Cell Proteomics. 2011;10(5):M110.004655.
    1. Palgen JL, et al. Prime and boost vaccination elicit a distinct innate myeloid cell immune response. Sci Rep. 2018;8(1):3087. doi: 10.1038/s41598-018-21222-2.
    1. Guo N, et al. Repeated immunization induces the increase in fucose content on antigen-specific IgG N-linked oligosaccharides. Clin Biochem. 2005;38(2):149–153. doi: 10.1016/j.clinbiochem.2004.10.002.
    1. Tsai WY, et al. Use of urea wash ELISA to distinguish Zika and Dengue virus infections. Emerg Infect Dis. 2018;24(7):1355–1359. doi: 10.3201/eid2407.171170.
    1. Mani S, et al. Serological cross reactivity between Zika and Dengue viruses in experimentally infected monkeys. Virol Sin. 2018;33(4):378–381. doi: 10.1007/s12250-018-0048-8.
    1. Winarski KL, et al. Antibody-dependent enhancement of influenza disease promoted by increase in hemagglutinin stem flexibility and virus fusion kinetics. Proc Natl Acad Sci U S A. 2019;116(30):15194–15199. doi: 10.1073/pnas.1821317116.
    1. Le S, et al. FactoMineR: An R package for multivariate analysis. J Stat Softw. 2008;25(1):1–18.
    1. Kassambara A, Mundt F. factoextra: Extract and Visualize the Results of Multivariate Data Analyses. Version 1.0.7. Cran R package; 2020. Accessed October 6, 2021. .
    1. Wei T, Simko V. 2021. R package ‘corrplot’: Visualization of a Correlation Matrix. (Version 0.90). Accessed October 21, 2021.
    1. R Core Team. Vienna, Austria: R Foundation for Statistical Computing; 2017. Accessed October 6, 2021.
    1. Wickham H, ed. ggplot2: Elegant Graphics for Data Analysis. Springer-Verlag; 2009.

Source: PubMed

3
구독하다