Immunological correlates of protection afforded by PHV02 live, attenuated recombinant vesicular stomatitis virus vector vaccine against Nipah virus disease

Thomas P Monath, Richard Nichols, Friederike Feldmann, Amanda Griffin, Elaine Haddock, Julie Callison, Kimberly Meade-White, Atsushi Okumura, Jamie Lovaglio, Patrick W Hanley, Chad S Clancy, Carl Shaia, Wasima Rida, Joan Fusco, Thomas P Monath, Richard Nichols, Friederike Feldmann, Amanda Griffin, Elaine Haddock, Julie Callison, Kimberly Meade-White, Atsushi Okumura, Jamie Lovaglio, Patrick W Hanley, Chad S Clancy, Carl Shaia, Wasima Rida, Joan Fusco

Abstract

Introduction: Immune correlates of protection afforded by PHV02, a recombinant vesicular stomatitis (rVSV) vector vaccine against Nipah virus (NiV) disease, were investigated in the African green monkey (AGM) model. Neutralizing antibody to NiV has been proposed as the principal mediator of protection against future NiV infection.

Methods: Two approaches were used to determine the correlation between neutralizing antibody levels and outcomes following a severe (1,000 median lethal doses) intranasal/intratracheal (IN/IT) challenge with NiV (Bangladesh): (1) reduction in vaccine dose given 28 days before challenge and (2) challenge during the early phase of the antibody response to the vaccine.

Results: Reduction in vaccine dose to very low levels led to primary vaccine failure rather than a sub-protective level of antibody. All AGMs vaccinated with the nominal clinical dose (2 × 107 pfu) at 21, 14, or 7 days before challenge survived. AGMs vaccinated at 21 days before challenge had neutralizing antibodies (geometric mean titer, 71.3). AGMs vaccinated at 7 or 14 days before challenge had either undetectable or low neutralizing antibody titers pre-challenge but had a rapid rise in titers after challenge that abrogated the NiV infection. A simple logistic regression model of the combined studies was used, in which the sole explanatory variable was pre-challenge neutralizing antibody titers. For a pre-challenge titer of 1:5, the predicted survival probability is 100%. The majority of animals with pre-challenge neutralizing titer of ≥1:20 were protected against pulmonary infiltrates on thoracic radiograms, and a majority of those with titers ≥1:40 were protected against clinical signs of illness and against a ≥fourfold antibody increase following challenge (indicating sterile immunity). Controls receiving rVSV-Ebola vaccine rapidly succumbed to NiV challenge, eliminating the innate immunity stimulated by the rVSV vector as a contributor to survival in monkeys challenged as early as 7 days after vaccination.

Discussion and conclusion: It was concluded that PHV02 vaccine elicited a rapid onset of protection and that any detectable level of neutralizing antibody was a functional immune correlate of survival.

Keywords: Nipah virus; immune correlate; neutralizing antibody; recombinant VSV; vaccine.

Conflict of interest statement

TM is employed by Crozet Biopharma LLC. RN and JF are employed by Public Health Vaccines Inc. WR is an independent consultant. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2023 Monath, Nichols, Feldmann, Griffin, Haddock, Callison, Meade-White, Okumura, Lovaglio, Hanley, Clancy, Shaia, Rida and Fusco.

Figures

Figure 1
Figure 1
Experimental design of two studies in African green monkeys (AGMs) designed to test the protective efficacy of PHV02 and to define the immunological correlates of protection. D, Day.
Figure 2
Figure 2
(A) Survival ratios, African green monkeys (AGMs) (N= 4 per group) vaccinated with graded doses of PHV02 vaccine 28 days before intranasal/intratracheal (IN/IT) challenge with 2 x 105 TCID50 of Nipah (Bangladesh). Controls (n=4) received rVSV-EBOV. (B) Survival ratios, AGMs (N= 6 per group) vaccinated with PHV02 2 x 107 pfu 21, 14 or 7 days before IN/IT challenge with 1 x 105 TCID50 of Nipah (Bangladesh). Controls (n=2) received rVSV-EBOV 14 or 7 days before challenge.
Figure 3
Figure 3
Pre-challenge (Day -1) Nipah virus neutralizing antibody titers, AGMs vaccinated with graded doses of PHV02 or with rVSV-EBOV (Controls) 28, 21, 14, or 7 days before IN/IT challenge (Day +1) with 2 x 105 TCID50 of Nipah (Bangladesh). Animals in all dose groups in Study 1 are combined since there were no statistical differences between groups, (see text and Supplemental Figure 1). Day -1 neutralizing antibody titers are displayed for animals in study 2 that were vaccinated 21, 14 or 7 days before challenge. Individual animal titers and geometric mean (horizontal bar) and geometric mean standard deviation (GSD) are shown. The two animals in study 1 vaccinated with the low dose on day -28 that did not seroconvert and died are shown in open circles (o).
Figure 4
Figure 4
Nipah virus neutralizing antibody titers, AGMs vaccinated with graded doses of PH VO2 or with rVSV-EBOV(Controls) 28 days before IN/IT challenge (Day +1) with 2 x 105 TCID50 of Nipah (Bangladesh), Study 1. Individual animal titers and geometric mean (horizontal bar) are shown by day with respect to challenge. The two monkeys in the low dose group that failed to develop neutralizing antibodies (designated X) died 7 days after challenge.
Figure 5
Figure 5
Logistic regression analysis to assess the relationship between pre-challenge (Day -1) log2 neutralizing antibody titer (NT) and survival in 38 African green monkeys (Studies 1 and 2 combined). (A) Simple logistic regression model with the sole explanatory variable pre-challenge log2 transformed NT. For a pre-challenge titer of 1:5, the predicted survival probability is 100%, but the confidence interval cannot be estimated. (B) Random noise (±1.0 log2) added to assess the impact of measurement error on predicted survival. For a pre-challenge titer of 1:5, the predicted survival probability (95% CI) is 73.2% (47.7, 89.1%).
Figure 6
Figure 6
Linear regression analysis, maximum clinical scores by pre-challenge (Day -2) neutralizing antibody titers (NT) (A). Study 1 Maximum clinical score by neutralizing titer pre-challenge (Day -1), animals vaccinated IM with graded doses of PHV02 28 days before IN/IT challenge with Nipah (Bangladesh). Regression analysis without (left panel) and with random noise [± 1.0log2) added to the pre-challenge NTs to assess the impact of measurement error] (right panel) (B). Study 2 Maximum clinical score by neutralizing titer pre-challenge (Day -1), animals vaccinated IM with PHV02 (2x107 pfu) 21, 14 or 7 days before or with rVSV-EBOV 14 or 7 days before IN/IT challenge with Nipah (Bangladesh) without (left panel) and with random noise (right panel). Individual data points are color coded by days from vaccination at the time of challenge. Note, in these plots in which noise has been added to pre-challenge titers unmask data points which are identical for different AGMs. For example, 3 control AGMs in Study 2 had the same maximum clinical score of 35. The estimated linear regression lines are superimposed on each plot.
Figure 7
Figure 7
Geometric mean (±GSD) Nipah virus neutralizing antibody titers following challenge (Day +1) with 2 x 105 TCID50 of Nipah (Bangladesh). AGMs were vaccinated with graded doses of PHV02 or with rVSV-EBOV (Controls) 21, 14, or 7 days before IN/IT challenge.

References

    1. Siegrist CA. Vaccine immunology. In: Plotkin SA, Offit PA, Orenstein WA, Edwards KM, editors. Plotkin’s vaccines, 7th Edit. Phila. PA: Elsevier; (2018). p. Pp16–34.
    1. Plotkin SA, Gilbert P. Immune correlates. In: Plotkin SA, Offit PA, Orenstein WA, Edwards KM, editors. Plotkin’s vaccines, 7th Edit. Phila. PA: Elsevier; (2018). p. Pp. 35–40.
    1. US Dept. Health and Human Services, Food and Drug Administration . Guidance for industry expedited programs for serious conditions – drugs and biologics. (2020). Available at: .
    1. Roozendaal R, Hendriks J, van Effelterre T, Spiessens B, Dekking L, Solforosi L, et al. . Nonhuman primate to human immunobridging to infer the protective effect of an Ebola virus vaccine candidate. NPJ Vaccines (2020) 5(1):112. doi: 10.1038/s41541-020-00261-9
    1. Khoury DS, Cromer D, Reynaldi A, Schlub TE, Wheatley AK, Juno JA, et al. . Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med (2021) 27(7):1205–11. doi: 10.1038/s41591-021-01377-8
    1. Aditi, Shariff M. Nipah virus infection: A review. Epidemiol Infect (2019) 147:e95. doi: 10.1017/S0950268819000086
    1. Tan CT, Goh KJ, Wong KT, Sarji SA, Chua KB, Chew NK, et al. . Relapsed and late-onset Nipah encephalitis. Ann Neurol (2002) 51(6):703–8. doi: 10.1002/ana.10212
    1. WHO . R&D blueprint (2015). Available at: .
    1. Looi LM, Chua KB. Lessons from the Nipah virus outbreak in Malaysia. Malays J Pathol (2007) 29(2):63–7.
    1. Black S, Bloom DE, Kaslow DC, Pecetta S, Rappuoli R. Transforming vaccine development. Semin Immunol (2020) 50:101413. doi: 10.1016/j.smim.2020.101413
    1. Prescott J, de Wit E, Feldmann H, Munster VJ. The immune response to Nipah virus infection. Arch Virol (2012) 157(9):1635–41. doi: 10.1007/s00705-012-1352-5
    1. Liew YJM, Ibrahim PAS, Ong HM, Chong CN, Tan CT, Schee JP, et al. . The immunobiology of nipah virus. Microorganisms (2022) 10:1162. doi: 10.3390/microorganisms10061162
    1. DeBuysscher BL, Scott D, Marzi A, Prescott J, Feldmann H. Single-dose live-attenuated Nipah virus vaccines confer complete protection by eliciting antibodies directed against surface glycoproteins. Vaccine (2014) 32(22):2637–44. doi: 10.1016/j.vaccine.2014.02.087
    1. Bossart KN, Zhu Z, Middleton D, Klippel J, Crameri G, Bingham J, et al. . A neutralizing human monoclonal antibody protects against lethal disease in a new ferret model of acute nipah virus infection. PLoS Pathog (2009) 5(10):e1000642. doi: 10.1371/journal.ppat.1000642
    1. Dong J, Cross RW, Doyle MP, Kose N, Mousa JJ, Annand EJ, et al. . Potent henipavirus neutralization by antibodies recognizing diverse sites on hendra and nipah virus receptor binding protein. Cell (2020) 183(6):1536–1550.e17. doi: 10.1016/j.cell.2020.11.023
    1. Doyle MP, Kose N, Borisevich V, Binshtein E, Amaya M, Nagel M, et al. . Cooperativity mediated by rationally selected combinations of human monoclonal antibodies targeting the henipavirus receptor binding protein. Cell Rep (2021) 36(9):109628. doi: 10.1016/j.celrep.2021.109628
    1. Dang HV, Cross RW, Borisevich V, Bornholdt ZA, West BR, Chan YP, et al. . Broadly neutralizing antibody cocktails targeting Nipah virus and Hendra virus fusion glycoproteins. Nat Struct Mol Biol (2021) 28(5):426–34. doi: 10.1038/s41594-021-00584-8
    1. Nie J, Liu L, Wang Q, Chen R, Ning T, Liu Q, et al. . Nipah pseudovirus system enables evaluation of vaccines in vitro and in vivo using non-BSL-4 facilities. Emerg Microbes Infect (2019) 8(1):272–81. doi: 10.1080/22221751.2019.1571871
    1. de Wit E, Munster VJ. Animal models of disease shed light on Nipah virus pathogenesis and transmission. J Pathol (2015) 235(2):196–205. doi: 10.1002/path.4444
    1. Prescott J, DeBuysscher BL, Feldmann F, Gardner DJ, Haddock E, Martellaro C, et al. . Single-dose live-attenuated vesicular stomatitis virus-based vaccine protects African green monkeys from Nipah virus disease. Vaccine (2015) 33(24):2823–9. doi: 10.1016/j.vaccine.2015.03.089
    1. Mire CE, Satterfield BA, Geisbert JB, Agans KN, Borisevich V, Yan L, et al. . Pathogenic differences between nipah virus Bangladesh and Malaysia strains in primates: Implications for antibody therapy. Sci Rep (2016) 6:30916. doi: 10.1038/srep30916
    1. Prasad AN, Woolsey C, Geisbert JB, Agans KN, Borisevich V, Deer DJ, et al. . Resistance of cynomolgus monkeys to nipah and hendra virus disease is associated with cell-mediated and humoral immunity. J Infect Dis (2020) 221(Suppl 4):S436–47. doi: 10.1093/infdis/jiz613
    1. Hossain MJ, Gurley ES, Montgomery JM, Bell M, Carroll DS, Hsu VP, et al. . Clinical presentation of nipah virus infection in Bangladesh. Clin Infect Dis (2008) 46(7):977–84. doi: 10.1086/529147
    1. Chong HT, KunJapan SR, Thayaparan T, Tong J, Petharunam V, Jusoh MR, et al. . Nipah encephalitis outbreak in Malaysia, clinical features in patients from Seremban. Can J Neurol Sci (2002) 29(1):83–7. doi: 10.1017/S0317167100001785
    1. Thomas B, Chandran P, Lilabi MP, George B, Sivakumar CP, Jayadev VK, et al. . Nipah virus infection in kozhikode, kerala, south India, in 2018: Epidemiology of an outbreak of an emerging disease. Indian J Community Med (2019) 44(4):383–7.
    1. Gurley ES, Montgomery JM, Hossain MJ, Bell M, Azad AK, Islam MR, et al. . Person-to-person transmission of Nipah virus in a Bangladeshi community. Emerg Infect Dis (2007) 13(7):1031–7. doi: 10.3201/eid1307.061128
    1. Cong Y, Lentz MR, Lara A, Alexander I, Bartos C, Bohannon JK, et al. . Loss in lung volume and changes in the immune response demonstrate disease progression in African green monkeys infected by small-particle aerosol and intratracheal exposure to Nipah virus. PLoS Negl Trop Dis (2017) 11(4):e0005532. doi: 10.1371/journal.pntd.0005532
    1. Rockx B, Brining D, Kramer J, Callison J, Ebihara H, Mansfield K, et al. . Clinical outcome of henipavirus infection in hamsters is determined by the route and dose of infection. J Virol (2011) 85(15):7658–71. doi: 10.1128/JVI.00473-11
    1. Heppner DG, Kemp TL, Martin BK, Ramsey WJ, Nichols R, Dasen EJ, et al. . Safety and immunogenicity of the rVSVΔG-ZEBOV-GP Ebola virus vaccine candidate in healthy adults: a phase 1b randomised, multicentre, double-blind, placebo-controlled, dose-response study. Lancet Infect Dis (2017) 17(8):854–66. doi: 10.1016/S1473-3099(17)30313-4
    1. Henao-Restrepo AM, Camacho A, Longini IM, Watson CH, Edmunds WJ, Egger M, et al. . Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Lancet (2017) 389(10068):505–18. doi: 10.1016/S0140-6736(16)32621-6
    1. Huttner A, Combescure C, Grillet S, Haks MC, Quinten E, Modoux C, et al. . A dose-dependent plasma signature of the safety and immunogenicity of the rVSV-Ebola vaccine in Europe and Africa. Sci Transl Med (2017) 9(385):eaaj1701. doi: 10.1126/scitranslmed.aaj1701
    1. Marzi A, Jankeel A, Menicucci AR, Callison J, O'Donnell KL, Feldmann F, et al. . Single dose of a VSV-based vaccine rapidly protects macaques from marburg virus disease. Front Immunol (2021) 12:774026. doi: 10.3389/fimmu.2021.774026
    1. Marzi A, Hanley PW, Haddock E, Martellaro C, Kobinger G, Feldmann H. Efficacy of vesicular stomatitis virus-ebola virus postexposure treatment in rhesus macaques infected with ebola virus makona. J Infect Dis (2016) 214(suppl 3):S360–6. doi: 10.1093/infdis/jiw218
    1. DeBuysscher BL, Scott D, Thomas T, Feldmann H, Prescott J. Peri-exposure protection against Nipah virus disease using a single-dose recombinant vesicular stomatitis virus-based vaccine. NPJ Vaccines (2016) 1:16002–. doi: 10.1038/npjvaccines.2016.2
    1. Liu J, Coffin KM, Johnston SC, Babka AM, Bell TM, Long SY, et al. . Nipah virus persists in the brains of nonhuman primate survivors. JCI Insight (2019) 4(14):e129629. doi: 10.1172/jci.insight.129629
    1. Monath TP, Nichols R, Tussey L, Scappaticci K, Pullano TG, Whiteman MD, et al. . Recombinant vesicular stomatitis vaccine against Nipah virus has a favorable safety profile: Model for assessment of live vaccines with neurotropic potential. PLoS Pathog (2022) 18(6):e1010658. doi: 10.1371/journal.ppat.1010658
    1. Brining DL, Mattoon JS, Kercher L, LaCasse RA, Safronetz D, Feldmann H, et al. . Thoracic radiography as a refinement methodology for the study of H1N1 influenza in cynomologus macaques (Macaca fascicularis). Comp Med (2010) 60(5):389–95.

Source: PubMed

3
Iratkozz fel