Safety and Immunogenicity of a Respiratory Syncytial Virus Prefusion F (RSVPreF3) Candidate Vaccine in Older Adults: Phase 1/2 Randomized Clinical Trial

Isabel Leroux-Roels, Matthew G Davis, Katie Steenackers, Brandon Essink, Corinne Vandermeulen, Charles Fogarty, Charles P Andrews, Edward Kerwin, Marie-Pierre David, Laurence Fissette, Carline Vanden Abeele, Delphine Collete, Magali de Heusch, Bruno Salaun, Nathalie De Schrevel, Juliane Koch, Céline Verheust, Nancy Dezutter, Frank Struyf, Narcisa Mesaros, Jelena Tica, Veronica Hulstrøm, Isabel Leroux-Roels, Matthew G Davis, Katie Steenackers, Brandon Essink, Corinne Vandermeulen, Charles Fogarty, Charles P Andrews, Edward Kerwin, Marie-Pierre David, Laurence Fissette, Carline Vanden Abeele, Delphine Collete, Magali de Heusch, Bruno Salaun, Nathalie De Schrevel, Juliane Koch, Céline Verheust, Nancy Dezutter, Frank Struyf, Narcisa Mesaros, Jelena Tica, Veronica Hulstrøm

Abstract

Background: The aim of this study was to investigate safety and immunogenicity of vaccine formulations against respiratory syncytial virus (RSV) containing the stabilized prefusion conformation of RSV fusion protein (RSVPreF3).

Methods: This phase 1/2, randomized controlled, observer-blind study enrolled 48 young adults (YAs; aged 18-40 years) and 1005 older adults (OAs; aged 60-80 years) between January and August 2019. Participants were randomized into equally sized groups to receive 2 doses of unadjuvanted (YAs and OAs) or AS01-adjuvanted (OAs) vaccine or placebo 2 months apart. Vaccine safety and immunogenicity were assessed until 1 month (YAs) or 12 months (OAs) after second vaccination.

Results: The RSVPreF3 vaccines boosted humoral (RSVPreF3-specific immunoglobulin G [IgG] and RSV-A neutralizing antibody) responses, which increased in an antigen concentration-dependent manner and were highest after dose 1. Compared to prevaccination, the geometric mean frequencies of polyfunctional CD4+ T cells increased after each dose and were significantly higher in adjuvanted than unadjuvanted vaccinees. Postvaccination immune responses persisted until end of follow-up. Solicited adverse events were mostly mild to moderate and transient. Despite a higher observed reactogenicity of AS01-containing vaccines, no safety concerns were identified for any assessed formulation.

Conclusions: Based on safety and immunogenicity profiles, the AS01E-adjuvanted vaccine containing 120 μg of RSVPreF3 was selected for further clinical development.

Clinical trials registration: NCT03814590.

Keywords: AS01 adjuvant; F protein; RSV neutralizing antibodies; cell-mediated immunity; respiratory syncytial virus.

Conflict of interest statement

Potential conflicts of interest. D. C., M.-P. D., M. d. H., N. D. S., N. D., L. F., V. H., J. K., N. M., B. S., F. S., J. T., C. V. A., and C. Ve. are/were employees of the GSK group of companies at the time of the study conduct. C. Va. is currently an employee of the GSK group of companies. D. C., M.-P. D., M. d. H., N. D. S., N. D., B. S., F. S., and C. Ve. hold shares from the GSK group of companies as part of their past/current employee remuneration. F. S. is currently an employee of Janssen Pharmaceutical Companies of Johnson & Johnson and holds restricted shares from Johnson & Johnson as part of his employee remuneration. All current/previous employees of the GSK groups of companies declare financial and nonfinancial relationships and activities. C. P. A., E. K., I. L.-R., K. S., and C. Va. report grant/research support from the GSK group of companies to their institution for study conduct and, except for C. Va., they have no nonfinancial relationships and activities to declare. E. K. has served as consultant, in advisory boards, in speaker’s bureaus, or received travel reimbursement from Amphastar, AstraZeneca, Boehringer Ingelheim, Forest, Cipla, Chiesi, GSK, Mylan, Novartis, Sunovion, Teva, Pearl Pharmaceuticals, and Theravance. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

© The Author(s) 2022. Published by Oxford University Press on behalf of Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Study design. Relevant points in the study timeline are designated as day (D) and month (M). N indicates number of participants in each study part/group. Dose 1 and 2 indicate vaccine and placebo administration timepoints. Blood sample S was the blood sample drawn for analysis of hematological (blood cell counts and hemoglobin levels) and biochemical (alanine and aspartate aminotransferases, creatinine, blood urea nitrogen, and uric acid) laboratory parameters. Blood sample H was the blood sample collected to measure concentrations of RSVPreF3-specific immunoglobulin and titers of respiratory syncytial virus-specific neutralizing antibodies. Blood sample C was the blood sample collected for analysis of polyfunctional T-cell responses. 30 μg, 60 μg, and 120 μg indicate RSVPreF3 antigen concentration. Abbreviations: AS01B and AS01E, adjuvanted vaccine formulations with the corresponding vaccine adjuvant systems; IDMC, independent data monitoring committee; Plain, unadjuvanted vaccine formulations; RSVPreF3, prefusion conformation of the respiratory syncytial virus F protein.
Figure 2.
Figure 2.
Consolidated Standards of Reporting Trials (CONSORT) flow diagram of participant cohorts in study parts A and B. Abbreviations: AS01B and AS01E, adjuvanted vaccine formulations with the corresponding vaccine adjuvant systems; pIMD, potential immune-mediated disease; Plain, unadjuvanted vaccine formulations; RSVPreF3, prefusion conformation of the respiratory syncytial virus F protein; SAE, serious adverse event.
Figure 3.
Figure 3.
Summary of solicited adverse events (AEs) reported within 7 days after any dose (A) and unsolicited AEs (any within 30 days after any dose, serious AE [SAE], or fatality until study end) (B) in part B (older adults [60–80 years of age]) (exposed set). B, Percentages of participants experiencing at least 1 AE from the following categories and periods: any unsolicited AE within 30 days after any vaccination; an SAE until study end; fatal outcome until study end. 30 µg, 60 µg, and 120 µg indicate prefusion conformation of the respiratory syncytial virus F protein antigen concentration. Abbreviations: AE, adverse event; AS01B and AS01E, adjuvanted vaccine formulations with the corresponding vaccine adjuvant systems; CI, confidence interval; EoS, end of study; Plain, unadjuvanted vaccine formulation; SAE, serious adverse event.
Figure 4.
Figure 4.
Solicited administration-site (A) and systemic adverse events (B) reported within 7 days after any vaccination in part B (older adults [60–80 years of age]) (exposed set). 30 µg, 60 µg, and 120 µg indicate prefusion conformation of the respiratory syncytial virus F protein antigen concentration. Abbreviations: AE, adverse event; AS01B and AS01E, adjuvanted vaccine formulations with the corresponding vaccine adjuvant systems; CI, confidence interval; GI, gastrointestinal; Plain, unadjuvanted vaccine formulation.
Figure 5.
Figure 5.
Prefusion conformation of the respiratory syncytial virus F protein (RSVPreF3)-specific immunoglobulin G geometric mean concentration (A), respiratory syncytial virus (RSV) A-specific neutralizing antibody (nAb) geometric mean titer (GMT) (B), and RSV-B-specific nAb GMT values (C) in part B (older adults [60–80 years of age]) (per-protocol set). *The timepoints in months (0, 1, 2, and 3) reflect days 1 (vaccination 1), 31, 61 (vaccination 2), and 91, respectively. Month 14 data derive from the long-term evaluation subset. C, The data at timepoint month 1 (day 31) were only tested for formulations with the selected concentration of the RSVPreF3 antigen (120 μg) and placebo; the participants were also vaccinated twice and at same timepoints, but the blood samples were not analyzed on day 61. Error bars represent 95% confidence intervals. The syringe symbols above the x-axis designate vaccination timepoints. 30 µg, 60 µg, and 120 µg indicate prefusion conformation of the respiratory syncytial virus F protein antigen concentration. Abbreviations: AS01B and AS01E, adjuvanted vaccine formulations with the corresponding vaccine adjuvant systems; ED60, estimated dilution 60; ELU, enzyme-linked immunosorbent assay unit; GMC, geometric mean concentration; GMT, geometric mean titer; IgG, immunoglobulin G; nAb, neutralizing antibody; Plain, unadjuvanted vaccine formulations; RSV, respiratory syncytial virus; RSVPreF3, prefusion conformation of the respiratory syncytial virus F protein.
Figure 6.
Figure 6.
Geometric mean frequency (GMF) (A) and fold increase (calculated as the ratio of T-cell concentration relative to day 1 values for the corresponding vaccine formulation) (B) in prefusion conformation of the respiratory syncytial virus F protein (RSVPreF3)-specific CD4+ T cells expressing at least 2 markersa and geometric mean frequency of RSVPreF3-specific CD4+ T cells expressing at least interferon gamma (IFN-γ)b (C) in part B (older adults [60–80 years of age]) (per-protocol set). aAt least 2 of the following in vitro markers: interleukin (IL) 2, CD40 ligand, tumor necrosis factor-α, IFN-γ. bAt least IFN-γ among IFN-γ, IL-13, and IL-17. *The timepoints in months (0, 1, 2, and 3) reflect days 1 (vaccination 1), 31, 61 (vaccination 2), and 91, respectively. Month 14 data derive from the long-term evaluation subset. A and C, The syringe symbols above the x-axis designate vaccination timepoints; dotted lines represent the assay cutoff of 590. GMF values are plotted as the median; error bars denote the range (min, max). 30 µg, 60 µg, and 120 µg indicate RSVPreF3 antigen concentration. Abbreviations: AS01B and AS01E, adjuvanted vaccine formulations with the corresponding vaccine adjuvant systems; GMF, geometric mean frequency; IFN-γ, interferon gamma; Plain, unadjuvanted vaccine formulations; RSVPreF3, prefusion conformation of the respiratory syncytial virus F protein.

References

    1. Simoes EA. Respiratory syncytial virus infection. Lancet 1999; 354:847–52.
    1. Ascough S, Paterson S, Chiu C. Induction and subversion of human protective immunity: contrasting influenza and respiratory syncytial virus. Front Immunol 2018; 9:323.
    1. Hause AM, Henke DM, Avadhanula V, Shaw CA, Tapia LI, Piedra PA. Sequence variability of the respiratory syncytial virus (RSV) fusion gene among contemporary and historical genotypes of RSV/A and RSV/B. PLoS One 2017; 12:e0175792.
    1. Mas V, Nair H, Campbell H, Melero JA, Williams TC. Antigenic and sequence variability of the human respiratory syncytial virus F glycoprotein compared to related viruses in a comprehensive dataset. Vaccine 2018; 36:6660–73.
    1. Russell CD, Unger SA, Walton M, Schwarze J. The human immune response to respiratory syncytial virus infection. Clin Microbiol Rev 2017; 30:481–502.
    1. Falsey AR, Hennessey PA, Formica MA, Cox C, Walsh EE. Respiratory syncytial virus infection in elderly and high-risk adults. N Engl J Med 2005; 352:1749–59.
    1. Falsey AR, McElhaney JE, Beran J, et al. . Respiratory syncytial virus and other respiratory viral infections in older adults with moderate to severe influenza-like illness. J Infect Dis 2014; 209:1873–81.
    1. Shi T, Denouel A, Tietjen AK, et al. . Global disease burden estimates of respiratory syncytial virus–associated acute respiratory infection in older adults in 2015: a systematic review and meta-analysis. J Infect Dis 2020; 222:S577–83.
    1. Cherukuri A, Patton K, GasserRA, Jret al. . Adults 65 years old and older have reduced numbers of functional memory T cells to respiratory syncytial virus fusion protein. Clin Vaccine Immunol 2013; 20:239–47.
    1. Malloy AM, Falsey AR, Ruckwardt TJ. Consequences of immature and senescent immune responses for infection with respiratory syncytial virus. Curr Top Microbiol Immunol 2013; 372:211–31.
    1. Boyoglu-Barnum S, Chirkova T, Anderson LJ. Biology of infection and disease pathogenesis to guide RSV vaccine development. Front Immunol 2019; 10:1675.
    1. Graham BS, Modjarrad K, McLellan JS. Novel antigens for RSV vaccines. Curr Opin Immunol 2015; 35:30–8.
    1. Didierlaurent AM, Laupeze B, Di Pasquale A, Hergli N, Collignon C, Garcon N. Adjuvant system AS01: helping to overcome the challenges of modern vaccines. Expert Rev Vaccines 2017; 16:55–63.
    1. Garcon N, Van Mechelen M. Recent clinical experience with vaccines using MPL- and QS-21-containing adjuvant systems. Expert Rev Vaccines 2011; 10:471–86.
    1. Leroux-Roels G, Marchant A, Levy J, et al. . Impact of adjuvants on CD4(+) T cell and B cell responses to a protein antigen vaccine: results from a phase II, randomized, multicenter trial. Clin Immunol 2016; 169:16–27.
    1. Chlibek R, Smetana J, Pauksens K, et al. . Safety and immunogenicity of three different formulations of an adjuvanted varicella-zoster virus subunit candidate vaccine in older adults: a phase II, randomized, controlled study. Vaccine 2014; 32:1745–53.
    1. Schwarz TF, Volpe S, Catteau G, et al. . Persistence of immune response to an adjuvanted varicella-zoster virus subunit vaccine for up to year nine in older adults. Hum Vaccin Immunother 2018; 14:1370–7.
    1. Voic H, de Vries RD, Sidney J, et al. . Identification and characterization of CD4(+) T cell epitopes after Shingrix vaccination. J Virol 2020; 94:e01641–20.
    1. Schwarz TF, Johnson C, Grigat C, et al. . Three dose levels of a maternal respiratory syncytial virus vaccine candidate are well tolerated and immunogenic in a randomized trial in non-pregnant women. J Infect Dis 2022; 225:2067–76.
    1. Ngwuta JO, Chen M, Modjarrad K, et al. . Prefusion F-specific antibodies determine the magnitude of RSV neutralizing activity in human sera. Sci Transl Med 2015; 7:309ra162.
    1. Boudreau CM, Yu WH, Suscovich TJ, Talbot HK, Edwards KM, Alter G. Selective induction of antibody effector functional responses using MF59-adjuvanted vaccination. J Clin Invest 2020; 130:662–72.
    1. Suscovich TJ, Fallon JK, Das J, et al. . Mapping functional humoral correlates of protection against malaria challenge following RTS, S/AS01 vaccination. Sci Transl Med 2020; 12:eabb4757.
    1. Ruckwardt TJ, Morabito KM, Phung E, et al. . Safety, tolerability, and immunogenicity of the respiratory syncytial virus prefusion F subunit vaccine DS-Cav1: a phase 1, randomised, open-label, dose-escalation clinical trial. Lancet Resp Med 2021; 9:1111–20.
    1. Beran J, Lickliter JD, Schwarz TF, et al. . Safety and immunogenicity of 3 formulations of an investigational respiratory syncytial virus vaccine in nonpregnant women: results from 2 phase 2 trials. J Infect Dis 2018; 217:1616–25.
    1. Langley JM, Aggarwal N, Toma A, et al. . A randomized, controlled, observer-blinded phase 1 study of the safety and immunogenicity of a respiratory syncytial virus vaccine with or without alum adjuvant. J Infect Dis 2017; 215:24–33.
    1. Walsh EE, Falsey AR, Scott DA, et al. . A randomized phase 1/2 study of a respiratory syncytial virus prefusion F vaccine. J Infect Dis 2022; 225:1357–66.
    1. Schwarz TF, McPhee RA, Launay O, et al. . Immunogenicity and safety of 3 formulations of a respiratory syncytial virus candidate vaccine in nonpregnant women: a phase 2, randomized trial. J Infect Dis 2019; 220:1816–25.
    1. Williams K, Bastian AR, Feldman RA, et al. . Phase 1 safety and immunogenicity study of a respiratory syncytial virus vaccine with an adenovirus 26 vector encoding prefusion F (Ad26.RSV.preF) in adults aged ≥60 years. J Infect Dis 2020; 222:979–88.
    1. Stephens LM, Varga SM. Considerations for a respiratory syncytial virus vaccine targeting an elderly population. Vaccines 2021; 9:624.
    1. van Erp EA, Luytjes W, Ferwerda G, van Kasteren PB. Fc-mediated antibody effector functions during respiratory syncytial virus infection and disease. Front Immunol 2019; 10:548.
    1. Saeland E, van der Fits L, Bolder R, et al. . Immunogenicity and protective efficacy of adenoviral and subunit RSV vaccines based on stabilized prefusion F protein in pre-clinical models. Vaccine 2022; 40:934–44.
    1. Zohar T, Hsiao JC, Mehta N, et al. . Upper and lower respiratory tract correlates of protection against respiratory syncytial virus following vaccination of nonhuman primates. Cell Host Microbe 2022; 30:41–52.e5.
    1. Walsh EE, Falsey AR. Humoral and mucosal immunity in protection from natural respiratory syncytial virus infection in adults. J Infect Dis 2004; 190:373–8.
    1. Habibi MS, Jozwik A, Makris S, et al. . Impaired antibody-mediated protection and defective IgA B-cell memory in experimental infection of adults with respiratory syncytial virus. Am J Respir Crit Care Med 2015; 191:1040–9.
    1. Lambert ND, Ovsyannikova IG, Pankratz VS, Jacobson RM, Poland GA. Understanding the immune response to seasonal influenza vaccination in older adults: a systems biology approach. Expert Rev Vaccines 2012; 11:985–94.

Source: PubMed

Подписаться