Immunogenicity and Efficacy of a Measles Virus-Vectored Chikungunya Vaccine in Nonhuman Primates

Shannan L Rossi, Jason E Comer, Eryu Wang, Sasha R Azar, William S Lawrence, Jessica A Plante, Katrin Ramsauer, Sabrina Schrauf, Scott C Weaver, Shannan L Rossi, Jason E Comer, Eryu Wang, Sasha R Azar, William S Lawrence, Jessica A Plante, Katrin Ramsauer, Sabrina Schrauf, Scott C Weaver

Abstract

Background: Chikungunya virus (CHIKV) infection can result in chikungunya fever (CHIKF), a self-limited acute febrile illness that can progress to chronic arthralgic sequelae in a large percentage of patients. A new measles virus-vectored vaccine was developed to prevent CHIKF, and we tested it for immunogenicity and efficacy in a nonhuman primate model.

Methods: Nine cynomolgus macaques were immunized and boosted with the measles virus-vectored chikungunya vaccine or sham-vaccinated. Sera were taken at multiple times during the vaccination phase to assess antibody responses against CHIKV. Macaques were challenged with a dose of CHIKV previously shown to cause fever and viremia, and core body temperature, viremia, and blood cell and chemistry panels were monitored.

Results: The vaccine was well tolerated in all macaques, and all seroconverted (high neutralizing antibody [PRNT80 titers, 40-640] and enzyme-linked immunosorbent assay titers) after the boost. Furthermore, the vaccinated primates were protected against viremia, fever, elevated white blood cell counts, and CHIKF-associated cytokine changes after challenge with the virulent La Reunión CHIKV strain.

Conclusions: These results further document the immunogenicity and efficacy of a measles-vectored chikungunya vaccine that shows promise in Phase I-II clinical trials. These findings are critical to human health because no vaccine to combat CHIKF is yet licensed.

Keywords: chikungunya virus; measles virus; nonhuman primate; vaccine.

© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Anti-Chikungunya virus immunoglobulin titers in sera collected on days 0, 28, and 56 of the study as assayed by enzyme-linked immunosorbent assay. Sera yielding absorbance values that exceeded the means of negative control sera by more than 2 standard deviations were considered positive. The mean absorbance of the negative control sera was 0.08 at 450 nm. Error bars denote standard error.
Figure 2.
Figure 2.
Body temperatures generated using a 2-hour moving average obtained from telemetric data. The vaccinated versus sham-vaccinated groups were compared statistically by one-way analysis of variance followed by Tukey’s test at each 2-hour time point beginning from 24 to 48 hours postchallenge. The mean temperature of the sham-vaccinated animals was significantly (P < .05) higher between 34 to 44 hours postchallenge as indicated by the asterisk.
Figure 3.
Figure 3.
Nonhuman primate activity curves were generated using a 2-hour moving average. The vaccinated versus sham-vaccinated groups were compared statistically by one-way analysis of variance followed by Tukey’s test at each 2-hour time point beginning from 42 to 90 hours postchallenge. The mean activity of the sham-vaccinated animals was lower (P < .05) at hour 44 postchallenge as indicated by the asterisk. There are no units for activity.
Figure 4.
Figure 4.
Viremia measured by quantitative real-time polymerase chain reaction assays. Ribonucleic acid (RNA) was isolated from sera and assayed using Chikungunya virus La Reunión (CHIKV-LR)-specific primers and probe. Cycle threshold (Ct) values were converted to plaque-forming units per milliliter (PFU/mL) from a standard curve generated from serially diluted RNA prepared from CHIKV-LR that was titered by plaque assay. Black symbols represent MV-CHIK vaccinated nonhuman primates (NHPs), whereas the red symbols indicate sham-vaccinated control animals.
Figure 5.
Figure 5.
Mean levels of circulating cytokines in nonhuman primate serum assayed using the Monkey Cytokine Magnetic 29-Plex Panel (Invitrogen). Mean levels of interferon (IFN)-γ (A), interleukin (IL)-1RA (B), and monocyte chemoattractant protein-1 (MCP-1) are presented. One-way analysis of variance followed by Tukey’s multiple group comparisons were performed on the different time points for a given group; +, P < .05. Differences between groups were determined using an unpaired t test; *, P < .05 and **, P < .001. Bars indicate standard errors.

References

    1. Weaver SC, Lecuit M. Chikungunya virus and the global spread of a mosquito-borne disease. N Engl J Med 2015; 372:1231–9.
    1. Weaver SC, Charlier C, Vasilakis N, Lecuit M. Zika, Chikungunya, and other emerging vector-borne viral diseases. Annu Rev Med 2018; 69:395–408.
    1. Weaver SC. Prediction and prevention of urban arbovirus epidemics: a challenge for the global virology community. Antiviral Res 2018; 156:80–4.
    1. Halstead SB. Reappearance of chikungunya, formerly called dengue, in the Americas. Emerg Infect Dis 2015; 21:557–61.
    1. Chretien JP, Anyamba A, Bedno SA, et al. . Drought-associated chikungunya emergence along coastal East Africa. Am J Trop Med Hyg 2007; 76:405–7.
    1. Rezza G, Nicoletti L, Angelini R, et al. . Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet 2007; 370:1840–6.
    1. Grandadam M, Caro V, Plumet S, et al. . Chikungunya virus, southeastern France. Emerg Infect Dis 2011; 17:910–3.
    1. Cassadou S, Boucau S, Petit-Sinturel M, Huc P, Leparc-Goffart I, Ledrans M. Emergence of chikungunya fever on the French side of Saint Martin island, October to December 2013. Euro Surveill 2014; 19:pii: 20752.
    1. Nunes MR, Faria NR, de Vasconcelos JM, et al. . Emergence and potential for spread of chikungunya virus in Brazil. BMC Med 2015; 13:102.
    1. Brandler S, Ruffié C, Combredet C, 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; 31:3718–25.
    1. Ramsauer K, Schwameis M, Firbas C, 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; 15:519–27.
    1. Reisinger EC, Tschismarov R, Beubler E, 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; 392:2718–27.
    1. Broeckel R, Haese N, Messaoudi I, Streblow DN. Nonhuman primate models of chikungunya virus infection and disease (CHIKV NHP model). Pathogens 2015; 4:662–81.
    1. Roy CJ, Adams AP, Wang E, et al. . Chikungunya vaccine candidate is highly attenuated and protects nonhuman primates against telemetrically monitored disease following a single dose. J Infect Dis 2014; 209:1891–9.
    1. Labadie K, Larcher T, Joubert C, et al. . Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J Clin Invest 2010; 120:894–906.
    1. Erasmus JH, Auguste AJ, Kaelber JT, et al. . A chikungunya fever vaccine utilizing an insect-specific virus platform. Nat Med 2017; 23:192–9.
    1. Plante K, Wang E, Partidos CD, et al. . Novel chikungunya vaccine candidate with an IRES-based attenuation and host range alteration mechanism. PLoS Pathog 2011; 7:e1002142.
    1. Langsjoen RM, Haller SL, Roy CJ, et al. . Chikungunya virus strains show lineage-specific variations in virulence and cross-protective ability in murine and nonhuman primate models. MBio 2018; 9: doi: 10.1128/mBio.02449-17.
    1. Beaty BJ, Calisher CH, Shope RE. Arboviruses. In: Lennete ET, Lennete DA, eds. Diagnostic procedures for viral, rickettsial and chlamydial infections, 7th edition. Washington, DC: American Public Health Association; 1995: pp. 189–212.
    1. Erasmus JH, Needham J, Raychaudhuri S, et al. . Utilization of an Eilat virus-based chimera for serological detection of chikungunya infection. PLoS Negl Trop Dis 2015; 9:e0004119.
    1. Minor PD. Live attenuated vaccines: historical successes and current challenges. Virology 2015; 479–480:379–92.
    1. Wang E, Volkova E, Adams AP, et al. . Chimeric alphavirus vaccine candidates for chikungunya. Vaccine 2008; 26:5030–9.
    1. Hallengard D, Kakoulidou M, Lulla A, et al. . Novel attenuated chikungunya vaccine candidates elicit protective immunity in C57BL/6 mice. J Virol 2014; 88:2858–66.
    1. Lorin C, Mollet L, Delebecque F, et al. . A single injection of recombinant measles virus vaccines expressing human immunodeficiency virus (HIV) type 1 clade B envelope glycoproteins induces neutralizing antibodies and cellular immune responses to HIV. J Virol 2004; 78:146–57.
    1. Weaver SC, Paessler S. Alphaviral Encephalitides. In: Barrett ADT, Stanberry L, eds. Vaccines against biothreats and emerging infections. London: Elsevier; 2009:339–59.
    1. Chua CL, Sam IC, Merits A, Chan YF. Antigenic variation of East/Central/South African and Asian chikungunya virus genotypes in neutralization by immune sera. PLoS Negl Trop Dis 2016; 10:e0004960.
    1. Yoon IK, Alera MT, Lago CB, 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:e0003764.
    1. Wauquier N, Becquart P, Nkoghe D, Padilla C, Ndjoyi-Mbiguino A, Leroy EM. The acute phase of chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J Infect Dis 2011; 204:115–23.
    1. Chaaitanya IK, Muruganandam N, Sundaram SG, et al. . Role of proinflammatory cytokines and chemokines in chronic arthropathy in CHIKV infection. Viral Immunol 2011; 24:265–71.
    1. Ruiz Silva M, van der Ende-Metselaar H, Mulder HL, Smit JM, Rodenhuis-Zybert IA. Mechanism and role of MCP-1 upregulation upon chikungunya virus infection in human peripheral blood mononuclear cells. Sci Rep 2016; 6:32288.

Source: PubMed

3
Suscribir