Increased neutralization and IgG epitope identification after MVA-MERS-S booster vaccination against Middle East respiratory syndrome

Anahita Fathi, Christine Dahlke, Verena Krähling, Alexandra Kupke, Nisreen M A Okba, Matthijs P Raadsen, Jasmin Heidepriem, Marcel A Müller, Grigori Paris, Susan Lassen, Michael Klüver, Asisa Volz, Till Koch, My L Ly, Monika Friedrich, Robert Fux, Alina Tscherne, Georgia Kalodimou, Stefan Schmiedel, Victor M Corman, Thomas Hesterkamp, Christian Drosten, Felix F Loeffler, Bart L Haagmans, Gerd Sutter, Stephan Becker, Marylyn M Addo, Anahita Fathi, Christine Dahlke, Verena Krähling, Alexandra Kupke, Nisreen M A Okba, Matthijs P Raadsen, Jasmin Heidepriem, Marcel A Müller, Grigori Paris, Susan Lassen, Michael Klüver, Asisa Volz, Till Koch, My L Ly, Monika Friedrich, Robert Fux, Alina Tscherne, Georgia Kalodimou, Stefan Schmiedel, Victor M Corman, Thomas Hesterkamp, Christian Drosten, Felix F Loeffler, Bart L Haagmans, Gerd Sutter, Stephan Becker, Marylyn M Addo

Abstract

Vaccine development is essential for pandemic preparedness. We previously conducted a Phase 1 clinical trial of the vector vaccine candidate MVA-MERS-S against the Middle East respiratory syndrome coronavirus (MERS-CoV), expressing its full spike glycoprotein (MERS-CoV-S), as a homologous two-dose regimen (Days 0 and 28). Here, we evaluate the safety (primary objective) and immunogenicity (secondary and exploratory objectives: magnitude and characterization of vaccine-induced humoral responses) of a third vaccination with MVA-MERS-S in a subgroup of trial participants one year after primary immunization. MVA-MERS-S booster vaccination is safe and well-tolerated. Both binding and neutralizing anti-MERS-CoV antibody titers increase substantially in all participants and exceed maximum titers observed after primary immunization more than 10-fold. We identify four immunogenic IgG epitopes, located in the receptor-binding domain (RBD, n = 1) and the S2 subunit (n = 3) of MERS-CoV-S. The level of baseline anti-human coronavirus antibody titers does not impact the generation of anti-MERS-CoV antibody responses. Our data support the rationale of a booster vaccination with MVA-MERS-S and encourage further investigation in larger trials. Trial registration: Clinicaltrials.gov NCT03615911.

Conflict of interest statement

The authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Study design and trial profile.
Fig. 1. Study design and trial profile.
Twenty-three individuals completed a homologous primary immunization with two vaccinations (Day (D) 0 (D0) and D28) of either 1 × 107 plaque-forming units (PFU) (low dose (LD), blue) or 1 × 108 PFU MVA-MERS-S (high dose (HD), red), and were followed-up for 180 days, which concluded this part of the study (End of Study). Participants were invited to return for an additional third vaccination as a booster 1 year ±4 months after prime. 10 participants (3 from the LD, 7 from the HD group) were re-enrolled and received a dose of 1 × 108 PFU MVA-MERS-S. Safety and tolerability were assessed on Boost (B) Days (B:D) 0 (B:D0, baseline), B:D1, B:D3 (not depicted), B:D7, B:D14, and B:D28; humoral immunogenicity was assessed on B:D0, B:D7, B:D14, and B:D28 (End of Study). All 10 participants completed the extension trial and were included in the analyses. Created with BioRender.com.
Fig. 2. Biologic monitoring.
Fig. 2. Biologic monitoring.
Graphs represent changes in a leukocyte, b neutrophil, c thrombocyte, and d lymphocyte counts, as well as e C-reactive protein (CRP) levels, in all booster study participants (n = 10) after booster vaccination. We observed an increase in a leukocyte (p = 0.014) and, by extension, b neutrophil counts (p = 0.002) as well as e CRP levels (p = 0.031) and a decrease in c thrombocyte (p = 0.049) and d lymphocyte (p = 0.002) counts on boost (B) Day 1 (B:D1) compared to B:D0. By B:D3, leukocyte and neutrophil counts had decreased (p = 0.004 for both neutrophil and leukocyte count on B:D1 vs B:D3 and B:D0 vs. BD3). These changes from baseline were transient and not clinically significant, but indicate biologic activity after vaccination. CRP levels of <5 mg/L (below the limit of detection) were set to 5 mg/L for visualization. Boxes indicate 25–75 percentile; whiskers are min. to max.; medians are shown as horizonal lines within the boxes. ULN = upper limit of normal. LLN = lower limit of normal. *p < 0.05, **p < 0.005, differences assessed using a two-sided Wilcoxon matched-pairs signed rank test. Source data are provided as a Source Data file.
Fig. 3. Increase in anti-MERS-CoV-S-specific binding antibodies…
Fig. 3. Increase in anti-MERS-CoV-S-specific binding antibodies after booster vaccination.
Anti-MERS-CoV-S-specific binding antibodies were measured as optical density values (y-axes) by two distinct ELISAs a in-house ELISA, b EUROIMMUN ELISA) at multiple timepoints after first (Day (D) 0 (D0), D35, D42, D84, D180) and booster (B) (B:D0, B:D7, B:D14, B:D28) vaccinations (x-axes). Former low dose (LD, n = 3) and high dose (HD, n = 7) vaccinees are depicted in blue and red, respectively, controls in gray (n = 2 in (a) and n = 4 in (b)). Boxes indicate 25–75 percentile; whiskers are min. to max.; medians are shown as horizonal lines within the boxes. Arrows indicate vaccinations. The horizontal dashed line in (a) indicates the cut-off level for positivity. Source data are provided as a Source Data file.
Fig. 4. Increase in MERS-CoV neutralizing antibodies…
Fig. 4. Increase in MERS-CoV neutralizing antibodies after booster vaccination.
MERS-CoV neutralization was measured by a ≥80% plaque reduction neutralization test (PRNT80) and b, c virus neutralization test (VNT). Neutralizing antibody levels were measured in reciprocal titers (y-axes) at multiple timepoints after first (Day (D) 0 (D0), D35, D42, D84, and D180) and booster (B) (B:D0, B:D7, B:D14, and B:D28) vaccinations (x-axes). Arrows indicate vaccinations. Horizontal dashed lines indicate the cut-off levels for positivity. a+b Boxes indicate 25–75 percentile; whiskers are min. to max.; medians are shown as horizonal lines within the boxes. Former low dose (LD, n = 3) and high dose (HD, n = 7) vaccinees are depicted in blue and red, respectively, controls in gray (n = 4). c VNT titers after prime vaccination measured longitudinally in the five individuals who did not develop nAb after prime vaccinations. Source data are provided as a Source Data file.
Fig. 5. Identification of four immunogenic MERS-CoV-S…
Fig. 5. Identification of four immunogenic MERS-CoV-S epitopes.
a Schematic representation of the MERS-CoV-S protein. The N-terminal domain (NTD), receptor-binding domain (RBD), S1/S2 cleavage site (S1/S2), fusion peptide (FP), S2‘ cleavage site (S2‘), heptad repeat 1 and 2 (HR1, HR2), transmembrane domain (TM) and cytoplasmic domain (CD) are illustrated. b Microarray of 15-mer peptides spanning the complete MERS-CoV-S protein with a 13 amino acid (AA) overlap. Immunogenic B-cell peptides are marked with red lines. cf IgG binding to the respective peptides on MERS-CoV-S was measured in fluorescence intensity (as arbitrary fluorescence units, AFU), depicted as transformed values (areas sinus hyperbolicus (asinh), y axis), in all booster study participants (n = 10). Mean levels of peptide-binding IgG at baseline (Day (D) 0 (D0), white boxes) and 28 days after booster vaccination (Boost Day (B:D) 28 (B:D28), gray boxes) were compared using a two-sided Wilcoxon matched-pairs signed rank test and are depicted for each peptide (x-axis) within the immunogenic epitopes c AA 535-553 (AA 535–549, p = 0.014; AA 537–551, p = 0.002; AA 539–553, p = 0.002), d AA 887–913 (AA 887–901, p = 0.027; AA 889–903, p = 0.006; AA 891–905, p = 0.002; AA 897–911, p = 0.002; AA 899–913, p = 0.002), e AA 1225–1247 (AA 1225–1239, p = 0.002; AA 1227–1241, p = 0.002; AA 1229–1243, p = 0.002; AA 1231–1245, p = 0.002; AA 1233–1247, p = 0.004) and f AA 1333–1353 (AA 1333–1347, p = 0.002; AA 1335–1349, p = 0.002; AA 1337–1351, p = 0.002; AA 1339–1353, p = 0.002). Boxes indicate 25–75 percentile; whiskers are min. to max.; medians are shown as horizonal lines within the boxes. *p < 0.05, **p < 0.005. Source data are provided as a Source Data file.
Fig. 6. Immunity to endemic HCoV compared…
Fig. 6. Immunity to endemic HCoV compared to anti-MERS-CoV immunity.
Reciprocal anti-CoV spike protein antibody titers of a human (H) Coronavirus (CoV) (HCoV)-OC43, b HCoV-229E, c HCoV-HKU1, d HCoV-NL63, e MERS-CoV, and f SARS-CoV (used as control antigen) were measured on Day (D) 0 (D0), pre-vaccination) and D42 via immunofluorescence assay in all participants who completed the primary immunization schedule with MVA-MERS-S on D0 and D28 (n = 23) and unvaccinated controls (n = 6). Low dose (n = 12) and high dose (n = 11) vaccinees are depicted in blue and red, respectively, unvaccinated controls in gray. A titer of <40 (dotted line) was considered negative. While we observed increased anti-MERS-CoV titers on Day 42 compared to baseline, there was no significant increase of anti-HCoV titers. Boxes indicate 25–75 percentile; whiskers are min. to max.; medians are shown as horizonal lines within the boxes. e ***p = 0.001 (LD) and p = 0.001 (HD), assessed via two-sided Wilcoxon matched-pairs signed rank test. Source data are provided as a Source Data file.

References

    1. World Health Organization. Prioritizing diseases for research and development in emergency contexts, (2021).
    1. Fathi A, Dahlke C, Addo MM. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum. Vaccin Immunother. 2019;15:2269–2285. doi: 10.1080/21645515.2019.1649532.
    1. Ewer K, et al. Chimpanzee adenoviral vectors as vaccines for outbreak pathogens. Hum. Vaccin. Immunother. 2017;13:3020–3032. doi: 10.1080/21645515.2017.1383575.
    1. Nagata LP, Irwin CR, Hu WG, Evans DH. Vaccinia-based vaccines to biothreat and emerging viruses. Biotechnol. Genet. Eng. Rev. 2018;34:107–121. doi: 10.1080/02648725.2018.1471643.
    1. Choi MJ, et al. Use of Ebola vaccine: Recommendations of the Advisory Committee on Immunization Practices, United States, 2020. MMWR Recomm. Rep. 2021;70:1–12. doi: 10.15585/mmwr.rr7001a1.
    1. European Medicines Agency. New vaccine for prevention of Ebola virus disease recommended for approval in the European Union, (2020).
    1. European Medicines Agency, Committee for Medicinal Products for Human Use. SmPC COVID-19 Vaccine AstraZeneca, (2021).
    1. European Medicines Agency, Committee for Medicinal Products for Human Use. SmPC JCOVDEN, INN-COVID-19 vaccine (Ad26.COV2-S [recombinant]) (2022).
    1. Volkmann A, et al. The Brighton Collaboration standardized template for collection of key information for risk/benefit assessment of a Modified Vaccinia Ankara (MVA) vaccine platform. Vaccine. 2021;39:3067–3080. doi: 10.1016/j.vaccine.2020.08.050.
    1. Kreijtz JH, Gilbert SC, Sutter G. Poxvirus vectors. Vaccine. 2013;31:4217–4219. doi: 10.1016/j.vaccine.2013.06.073.
    1. Greenough TC, et al. Safety and immunogenicity of recombinant poxvirus HIV-1 vaccines in young adults on highly active antiretroviral therapy. Vaccine. 2008;26:6883–6893. doi: 10.1016/j.vaccine.2008.09.084.
    1. Mayr A, Stickl H, Muller HK, Danner K, Singer H. [The smallpox vaccination strain MVA: Marker, genetic structure, experience gained with the parenteral vaccination and behavior in organisms with a debilitated defence mechanism (author’s transl)] Zentralbl Bakteriol. B. 1978;167:375–390.
    1. Krammer F. SARS-CoV-2 vaccines in development. Nature. 2020;586:516–527. doi: 10.1038/s41586-020-2798-3.
    1. Mou H, et al. The receptor binding domain of the new Middle East respiratory syndrome coronavirus maps to a 231-residue region in the spike protein that efficiently elicits neutralizing antibodies. J. Virol. 2013;87:9379–9383. doi: 10.1128/JVI.01277-13.
    1. Shehata MM, Gomaa MR, Ali MA, Kayali G. Middle East respiratory syndrome coronavirus: A comprehensive review. Front. Med. 2016;10:120–136. doi: 10.1007/s11684-016-0430-6.
    1. Modjarrad K, et al. Safety and immunogenicity of an anti-Middle East respiratory syndrome coronavirus DNA vaccine: A phase 1, open-label, single-arm, dose-escalation trial. Lancet Infect. Dis. 2019;19:1013–1022. doi: 10.1016/S1473-3099(19)30266-X.
    1. Folegatti PM, et al. Safety and immunogenicity of a candidate Middle East respiratory syndrome coronavirus viral-vectored vaccine: A dose-escalation, open-label, non-randomised, uncontrolled, phase 1 trial. Lancet Infect. Dis. 2020;20:816–826. doi: 10.1016/S1473-3099(20)30160-2.
    1. Koch T, et al. Safety and immunogenicity of a modified vaccinia virus Ankara vector vaccine candidate for Middle East respiratory syndrome: an open-label, phase 1 trial. Lancet Infect. Dis. 2020;20:827–838. doi: 10.1016/S1473-3099(20)30248-6.
    1. Drosten C, et al. Transmission of MERS-coronavirus in household contacts. N. Engl. J. Med. 2014;371:828–835. doi: 10.1056/NEJMoa1405858.
    1. Wang C, et al. A conserved immunogenic and vulnerable site on the coronavirus spike protein delineated by cross-reactive monoclonal antibodies. Nat. Commun. 2021;12:1715. doi: 10.1038/s41467-021-21968-w.
    1. Agnandji ST, et al. Phase 1 trials of rVSV Ebola vaccine in Africa and Europe. N. Engl. J. Med. 2016;374:1647–1660. doi: 10.1056/NEJMoa1502924.
    1. European Medicines Agency. SmPC Imvanex, (2018).
    1. National Center for Immunization and Respiratory Diseases. General recommendations on immunization—recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm. Rep. 2011;60:1–64.
    1. Atmar RL, et al. Homologous and heterologous Covid-19 booster vaccinations. N. Engl. J. Med. 2022;386:1046–1057. doi: 10.1056/NEJMoa2116414.
    1. Falsey AR, et al. SARS-CoV-2 neutralization with BNT162b2 vaccine dose 3. N. Engl. J. Med. 2021;385:1627–1629. doi: 10.1056/NEJMc2113468.
    1. Arbel R, et al. BNT162b2 vaccine booster and mortality due to Covid-19. N. Engl. J. Med. 2021;385:2413–2420. doi: 10.1056/NEJMoa2115624.
    1. Hall VG, et al. Randomized trial of a third dose of mRNA-1273 vaccine in transplant recipients. N. Engl. J. Med. 2021;385:1244–1246. doi: 10.1056/NEJMc2111462.
    1. Bar-On YM, et al. Protection against Covid-19 by BNT162b2 booster across age groups. N. Engl. J. Med. 2021;385:2421–2430. doi: 10.1056/NEJMoa2115926.
    1. Robert Koch Institut. Beschluss der STIKO zur 16. Aktualisierung der COVID-19-Impfempfehlung. Epidemiologisches Bull. 2022;2:3–15.
    1. Mbaeyi S, et al. The Advisory Committee on Immunization Practices’ interim recommendations for additional primary and booster doses of COVID-19 vaccines - United States, 2021. MMWR Morb. Mortal. Wkly Rep. 2021;70:1545–1552. doi: 10.15585/mmwr.mm7044e2.
    1. Kreijtz JH, et al. Safety and immunogenicity of a modified-vaccinia-virus-Ankara-based influenza A H5N1 vaccine: A randomised, double-blind phase 1/2a clinical trial. Lancet Infect. Dis. 2014;14:1196–1207. doi: 10.1016/S1473-3099(14)70963-6.
    1. Guardo AC, et al. Safety and vaccine-induced HIV-1 immune responses in healthy volunteers following a late MVA-B boost 4 years after the last immunization. PLoS One. 2017;12:e0186602. doi: 10.1371/journal.pone.0186602.
    1. Nilsson C, et al. Broad and potent cellular and humoral immune responses after a second late HIV-modified vaccinia virus ankara vaccination in HIV-DNA-primed and HIV-modified vaccinia virus Ankara-boosted Swedish vaccinees. AIDS Res Hum. Retroviruses. 2014;30:299–311. doi: 10.1089/aid.2013.0149.
    1. Palgen JL, et al. Innate and secondary humoral responses are improved by increasing the time between MVA vaccine immunizations. NPJ Vaccines. 2020;5:24. doi: 10.1038/s41541-020-0175-8.
    1. Ying T, et al. Exceptionally potent neutralization of Middle East respiratory syndrome coronavirus by human monoclonal antibodies. J. Virol. 2014;88:7796–7805. doi: 10.1128/JVI.00912-14.
    1. Wang L, et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nat. Commun. 2015;6:7712. doi: 10.1038/ncomms8712.
    1. Song F, et al. Middle East respiratory syndrome coronavirus spike protein delivered by modified vaccinia virus Ankara efficiently induces virus-neutralizing antibodies. J. Virol. 2013;87:11950–11954. doi: 10.1128/JVI.01672-13.
    1. Widjaja I, et al. Towards a solution to MERS: protective human monoclonal antibodies targeting different domains and functions of the MERS-coronavirus spike glycoprotein. Emerg. Microbes Infect. 2019;8:516–530. doi: 10.1080/22221751.2019.1597644.
    1. Wang L, et al. Importance of neutralizing monoclonal antibodies targeting multiple antigenic sites on the Middle East respiratory syndrome coronavirus spike glycoprotein to avoid neutralization escape. J. Virol. 2018;92:e02002–02017.
    1. Poh CM, et al. Two linear epitopes on the SARS-CoV-2 spike protein that elicit neutralising antibodies in COVID-19 patients. Nat. Commun. 2020;11:2806. doi: 10.1038/s41467-020-16638-2.
    1. Nguyen-Contant P, et al. S Protein-reactive IgG and memory B cell production after human SARS-CoV-2 infection includes broad reactivity to the S2 subunit. mBio. 2020;11:e01991–01920. doi: 10.1128/mBio.01991-20.
    1. Routhu NK, et al. A modified vaccinia Ankara vector-based vaccine protects macaques from SARS-CoV-2 infection, immune pathology, and dysfunction in the lungs. Immunity. 2021;54:542–556.e549. doi: 10.1016/j.immuni.2021.02.001.
    1. Tscherne A, et al. Immunogenicity and efficacy of the COVID-19 candidate vector vaccine MVA-SARS-2-S in preclinical vaccination. Proc. Natl Acad. Sci. USA. 2021;118:e2026207118. doi: 10.1073/pnas.2026207118.
    1. U.S. Department of health and human services. Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0, (2009).
    1. Food and Drug Administration. Guidance for Industry Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials, (2007).
    1. Okba NMA, et al. Sensitive and specific detection of low-level antibody responses in mild middle east respiratory syndrome coronavirus infections. Emerg. Infect. Dis. 2019;25:1868–1877. doi: 10.3201/eid2510.190051.
    1. Heidepriem J, et al. Longitudinal development of antibody responses in COVID-19 patients of different severity with ELISA, peptide, and glycan arrays: An immunological case series. Pathogens. 2021;10:438. doi: 10.3390/pathogens10040438.
    1. Corman VM, et al. Assays for laboratory confirmation of novel human coronavirus (hCoV-EMC) infections. Eur. Surveill. 2012;17:20334.

Source: PubMed

3
Subscribe