Detectable Vesicular Stomatitis Virus (VSV)-Specific Humoral and Cellular Immune Responses Following VSV-Ebola Virus Vaccination in Humans

Joseph H Poetsch, Christine Dahlke, Madeleine E Zinser, Rahel Kasonta, Sebastian Lunemann, Anne Rechtien, My L Ly, Hans C Stubbe, Verena Krähling, Nadine Biedenkopf, Markus Eickmann, Sarah K Fehling, Flaminia Olearo, Thomas Strecker, Piyush Sharma, Karl S Lang, Ansgar W Lohse, Stefan Schmiedel, Stephan Becker, VSV-Ebola Consortium (VEBCON), Marylyn M Addo, Claire-Anne Siegrist, Angela Huttner, Marylyn M Addo, Stephan Becker, Verena Krähling, Phillip Bejon, Patricia Njuguna, Francis Ndungu, Peter G Kremsner, Jessica S Brosnahan, Selidji Todagbe Agnandji, Sanjeev Krishna, Marie Paule Kieny, Kayvon Modjarrad, Vasee Moorthy, Patricia Fast, Barbara Savarese, Olivier Lapujade, Joseph H Poetsch, Christine Dahlke, Madeleine E Zinser, Rahel Kasonta, Sebastian Lunemann, Anne Rechtien, My L Ly, Hans C Stubbe, Verena Krähling, Nadine Biedenkopf, Markus Eickmann, Sarah K Fehling, Flaminia Olearo, Thomas Strecker, Piyush Sharma, Karl S Lang, Ansgar W Lohse, Stefan Schmiedel, Stephan Becker, VSV-Ebola Consortium (VEBCON), Marylyn M Addo, Claire-Anne Siegrist, Angela Huttner, Marylyn M Addo, Stephan Becker, Verena Krähling, Phillip Bejon, Patricia Njuguna, Francis Ndungu, Peter G Kremsner, Jessica S Brosnahan, Selidji Todagbe Agnandji, Sanjeev Krishna, Marie Paule Kieny, Kayvon Modjarrad, Vasee Moorthy, Patricia Fast, Barbara Savarese, Olivier Lapujade

Abstract

In response to the Ebola virus (EBOV) crisis of 2013-2016, a recombinant vesicular stomatitis virus (VSV)-based EBOV vaccine was clinically tested (NCT02283099). A single-dose regimen of VSV-EBOV revealed a safe and immunogenic profile and demonstrated clinical efficacy. While EBOV-specific immune responses to this candidate vaccine have previously been investigated, limited human data on immunity to the VSV vector are available. Within the scope of a phase 1 study, we performed a comprehensive longitudinal analysis of adaptive immune responses to internal VSV proteins following VSV-EBOV immunization. While no preexisting immunity to the vector was observed, more than one-third of subjects developed VSV-specific cytotoxic T-lymphocyte responses and antibodies.

Figures

Figure 1.
Figure 1.
Humoral responses against vesicular stomatitis virus (VSV). A, Structure and design of VSV–Ebola virus (EBOV) vaccine. VSV glycoprotein G (G) is replaced by EBOV glycoprotein (GP), while nucleoprotein (N), phosphoprotein (P), matrix protein (M), and RNA-dependent RNA polymerase (L) correspond to the VSV backbone vector. B, VSV-M–specific antibodies were generated following VSV-EBOV immunization in humans. VSV-M antibody titers were assessed by enzyme-linked immunosorbent assay at baseline and days 14, 28, 56, 84, and 180 postvaccination. Results are expressed as corrected optical density (OD) values. The dashed line depicts the threshold for a positive antibody response, calculated as the median on day 0 of all subjects ± 3 standard deviations. VSV-M–specific antibodies are detectable in 8 subjects (3 × 105 plaque-forming units [PFU], 5 of 10 subjects; 3 × 106 PFU, 1 of 10 subjects; 2 × 107 PFU, 2 of 9 subjects). C, Positive correlation between OD values of VSV-M and EBOV-GP–specific immunoglobulin G (IgG) at day 56 postvaccination. D, VSV-M–positive subjects were analyzed for generation of neutralizing antibodies against VSV wild-type (VSVwt; (n = 8). Neutralizing antibodies against infectious EBOV isolate Mayinga but not against VSV-M were detected. Statistical analysis was performed with Mann–Whitney–Wilcoxon test.
Figure 2.
Figure 2.
Antigen-specific T cells against vesicular stomatitis virus (VSV). A, VSV wild-type (VSVwt)–specific T-cell responses. Peripheral blood mononuclear cells were stimulated with ultraviolet-inactivated VSVwt. Graph depicts the observed interferon gamma (IFN-γ)/interleukin 2 (IL-2)/tumor necrosis factor alpha (TNF-α) secretion of CD8+ T cells measured by flow cytometry. Each dot represents summarized cytokine responses of CD8+ T cells for 1 subject (3 × 105 plaque-forming units [PFU]: n = 6; 3 × 106 PFU: n = 5; 2 × 107 PFU: n = 4). A significant intergroup difference between the low- and high-dose group was observed on day 56 (Mann–Whitney test, P = .01). B and C, Cytokine responses of CD8+ and CD4+ T cells (IFN-γ/IL-2/TNF-α) following stimulation with VSV nucleoprotein (VSV-N) overlapping peptide pools, respectively. Cytokine secretion was measured by flow cytometry (3 × 105 PFU: n = 8; 3 × 106 PFU: n = 7; 2 × 107 PFU: n = 9). D, Pie charts represent the functionality of specific T cells to VSV-N peptide pools at day 28 following immunization. Shown are the proportions of VSV-N–specific memory CD8+ (left) and CD4+ (right) cells that produce any combinations of the 3 measured cytokines. Pie charts represent the mean value of 9 subjects from the high-dose cohort. E, Cytotoxic T-lymphocyte (CTL) response following stimulation with VSV-N peptides. Flow cytometry analysis of the degranulation marker CD107a in the CD8+ T-cell subset (3 × 105 PFU: n = 10; 3 × 106 PFU: n = 5; 2 × 107 PFU: n = 8). The magnitude of CTL responses revealed significant intergroup differences on days 28 and 56 (Mann–Whitney–Wilcoxon test, P = .0045 and P = .0095, respectively). Comparing T-cell responses following VSV-N peptide stimulation revealed an increased response to VSV-N peptides in 3 vaccinees, showing induced cytokine or CD107a expression in CD8+ or CD4+ T cells. Box and whiskers show minimum to maximum; line shows the median. Statistical analysis was performed with Mann–Whitney–Wilcoxon test (*P < .05). Green: 3 × 105 PFU; blue: 3 × 106 PFU; red: 2 × 107 PFU.

References

    1. Agnandji ST, Huttner A, Zinser ME, et al. . Phase 1 trials of rVSV Ebola vaccine in Africa and Europe. N Engl J Med 2016; 374:1647–60.
    1. Coller BG, Blue J, Das R, et al. . Clinical development of a recombinant Ebola vaccine in the midst of an unprecedented epidemic. Vaccine 2017; 35:4465–9.
    1. Geisbert TW, Feldmann H. Recombinant vesicular stomatitis virus-based vaccines against Ebola and Marburg virus infections. J Infect Dis 2011; 204(Suppl 3):S1075–81.
    1. Marzi A, Robertson SJ, Haddock E, et al. . Ebola vaccine. VSV-EBOV rapidly protects macaques against infection with the 2014/15 Ebola virus outbreak strain. Science 2015; 349:739–42.
    1. Henao-Restrepo AM, Longini IM, Egger M, et al. . Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: interim results from the Guinea ring vaccination cluster-randomised trial. Lancet 2015; 386:857–66.
    1. Zhu FC, Hou LH, Li JX, et al. . Safety and immunogenicity of a novel recombinant adenovirus type-5 vector-based Ebola vaccine in healthy adults in China: preliminary report of a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet 2015; 385:2272–9.
    1. Bukreyev A, Marzi A, Feldmann F, et al. . Chimeric human parainfluenza virus bearing the Ebola virus glycoprotein as the sole surface protein is immunogenic and highly protective against Ebola virus challenge. Virology 2009; 383:348–61.
    1. Dahlke C, Kasonta R, Lunemann S, et al. . VEBCON Consortium Dose-dependent T-cell dynamics and cytokine cascade following rVSV-ZEBOV immunization. EBioMedicine 2017; 19:107–18.
    1. Medaglini D, Santoro F, Siegrist CA. Correlates of vaccine-induced protective immunity against Ebola virus disease. Semin Immunol 2018. doi:10.1016/j.smim.2018.07.003.
    1. Li SS, Kochar NK, Elizaga M, et al. . DNA priming increases frequency of T-cell responses to a vesicular stomatitis virus HIV vaccine with specific enhancement of CD8(+) T-cell responses by interleukin-12 plasmid DNA. Clin Vaccine Immunol 2017; 24. doi:10.1128/CVI.00263-17.
    1. Harrington LE, Most Rv Rv, Whitton JL, Ahmed R. Recombinant vaccinia virus-induced T-cell immunity: quantitation of the response to the virus vector and the foreign epitope. J Virol 2002; 76:3329–37.
    1. Khurana S, Fuentes S, Coyle EM, Ravichandran S, Davey RT Jr, Beigel JH. Human antibody repertoire after VSV-Ebola vaccination identifies novel targets and virus-neutralizing IgM antibodies. Nat Med 2016; 22:1439–47.
    1. Marzi A, Feldmann F, Geisbert TW, Feldmann H, Safronetz D. Vesicular stomatitis virus-based vaccines against Lassa and Ebola viruses. Emerg Infect Dis 2015; 21:305–7.
    1. Monath TP, Gershman M, Staples JE. Yellow fever vaccine. In: Plotkin SA Orenstein WA, Offit PA, eds. Vaccines. 6th ed. London: W.B. Saunders, 2013: 870–968.
    1. Furman D, et al. . Cytomegalovirus infection improves immune responses to influenza. Sci Transl Med 2015; 7:281ra43.

Source: PubMed

3
Abonneren