Safety and immunogenicity of a parenteral trivalent P2-VP8 subunit rotavirus vaccine: a multisite, randomised, double-blind, placebo-controlled trial

Michelle J Groome, Lee Fairlie, Julie Morrison, Alan Fix, Anthonet Koen, Maysseb Masenya, Lisa Jose, Shabir A Madhi, Nicola Page, Monica McNeal, Len Dally, Iksung Cho, Maureen Power, Jorge Flores, Stanley Cryz, Michelle J Groome, Lee Fairlie, Julie Morrison, Alan Fix, Anthonet Koen, Maysseb Masenya, Lisa Jose, Shabir A Madhi, Nicola Page, Monica McNeal, Len Dally, Iksung Cho, Maureen Power, Jorge Flores, Stanley Cryz

Abstract

Background: A monovalent, parenteral, subunit rotavirus vaccine was well tolerated and immunogenic in adults in the USA and in toddlers and infants in South Africa, but elicited poor responses against heterotypic rotavirus strains. We aimed to evaluate safety and immunogenicity of a trivalent vaccine formulation (P2-VP8-P[4],[6],[8]).

Methods: A double-blind, randomised, placebo-controlled, dose-escalation, phase 1/2 study was done at three South African research sites. Healthy adults (aged 18-45 years), toddlers (aged 2-3 years), and infants (aged 6-8 weeks, ≥37 weeks' gestation, and without previous receipt of rotavirus vaccination), all without HIV infection, were eligible for enrolment. In the dose-escalation phase, adults and toddlers were randomly assigned in blocks (block size of five) to receive 30 μg or 90 μg of vaccine, or placebo, and infants were randomly assigned in blocks (block size of four) to receive 15 μg, 30 μg, or 90 μg of vaccine, or placebo. In the expanded phase, infants were randomly assigned in a 1:1:1:1 ratio to receive 15 μg, 30 μg, or 90 μg of vaccine, or placebo, in block sizes of four. Participants, parents of participants, and clinical, data, and laboratory staff were masked to treatment assignment. Adults received an intramuscular injection of vaccine or placebo in the deltoid muscle on the day of randomisation (day 0), day 28, and day 56; toddlers received a single injection of vaccine or placebo in the anterolateral thigh on day 0. Infants in both phases received an injection of vaccine or placebo in the anterolateral thigh on days 0, 28, and 56, at approximately 6, 10, and 14 weeks of age. Primary safety endpoints were local and systemic reactions (grade 2 or worse) within 7 days and adverse events and serious adverse events within 28 days after each injection in all participants who received at least one injection. Primary immunogenicity endpoints were analysed in infants in either phase who received all planned injections, had blood samples analysed at the relevant timepoints, and presented no major protocol violations considered to have an effect on the immunogenicity results of the study, and included serum anti-P2-VP8 IgA, IgG, and neutralising antibody geometric mean titres and responses measured 4 weeks after the final injection in vaccine compared with placebo groups. This trial is registered with ClinicalTrials.gov, NCT02646891.

Findings: Between Feb 15, 2016, and Dec 22, 2017, 30 adults (12 each in the 30 μg and 90 μg groups and six in the placebo group), 30 toddlers (12 each in the 30 μg and 90 μg groups and six in the placebo group), and 557 infants (139 in the 15 μg group, 140 in the 30 μg group, 139 in the 90 μg group, and 139 in the placebo group) were randomly assigned, received at least one dose, and were assessed for safety. There were no significant differences in local or systemic adverse events, or unsolicited adverse events, between vaccine and placebo groups. There were no serious adverse events within 28 days of injection in adults, whereas one serious adverse event occurred in a toddler (febrile convulsion in the 30 μg group) and 23 serious adverse events (four in placebo, ten in 15 μg, four in 30 μg, and five in 90 μg groups) occurred among 20 infants, most commonly respiratory tract infections. One death occurred in an infant within 28 days of injection due to pneumococcal meningitis. In 528 infants (130 in placebo, 132 in 15 μg, 132 in 30 μg, and 134 in 90 μg groups), adjusted anti-P2-VP8 IgG seroresponses (≥4-fold increase from baseline) to P[4], P[6], and P[8] antigens were significantly higher in the 15 μg, 30 μg, and 90 μg groups (99-100%) than in the placebo group (10-29%; p<0·0001). Although significantly higher than in placebo recipients (9-10%), anti-P2-VP8 IgA seroresponses (≥4-fold increase from baseline) to each individual antigen were modest (20-34%) across the 15 μg, 30 μg, and 90 μg groups. Adjusted neutralising antibody seroresponses in infants (≥2·7-fold increase from baseline) to DS-1 (P[4]), 1076 (P[6]), and Wa (P[8]) were higher in vaccine recipients than in placebo recipients: p<0·0001 for all comparisons.

Interpretation: The trivalent P2-VP8 vaccine was well tolerated, with promising anti-P2-VP8 IgG and neutralising antibody responses across the three vaccine P types. Our findings support advancing the vaccine to efficacy testing.

Funding: Bill & Melinda Gates Foundation.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile for infants in the dose-escalation and expanded cohorts PP=per protocol. *254 at the Respiratory and Meningeal Pathogens Research Unit, 168 at Wits Shandukani Research Centre, and 136 at the Family Clinical Research Unit. †PP population included participants who received two doses and had immunogenicity data at baseline and day 56 (for secondary immunogenicity analyses) or who received all three doses and had immunogenicity data at baseline and day 84 (for the primary immunogenicity analysis). ‡The infant who did not receive the third dose because they were ineligible attended the day 56 follow-up visit, had a blood sample collected, and was included in the day 56 PP population.
Figure 2
Figure 2
Neutralising antibodies to Wa, DS-1, and 1076 rotavirus strains 4 weeks after the second and third injection of trivalent P2-VP8 or placebo in the per-protocol infant population, according to treatment group GMT and 95% CI unadjusted for decrease in maternal antibodies. GMT=geometric mean titre.

References

    1. Burnett E, Jonesteller CL, Tate JE, Yen C, Parashar UD. Global impact of rotavirus vaccination on childhood hospitalizations and mortality from diarrhea. J Infect Dis. 2017;215:1666–1672.
    1. GBD 2016 Diarrhoeal Disease Collaborators Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect Dis. 2018;18:1211–1228.
    1. Jonesteller CL, Burnett E, Yen C, Tate JE, Parashar UD. Effectiveness of rotavirus vaccination: a systematic review of the first decade of global postlicensure data, 2006–2016. Clin Infect Dis. 2017;65:840–850.
    1. Yen C, Tate JE, Patel MM. Rotavirus vaccines: update on global impact and future priorities. Hum Vaccin. 2011;7:1282–1290.
    1. Levine MM. Immunogenicity and efficacy of oral vaccines in developing countries: lessons from a live cholera vaccine. BMC Biol. 2010;8:129.
    1. Patriarca PA, Wright PF, John TJ. Factors affecting the immunogenicity of oral poliovirus vaccine in developing countries: review. Rev Infect Dis. 1991;13:926–939.
    1. Velasquez DE, Parashar U, Jiang B. Decreased performance of live attenuated, oral rotavirus vaccines in low-income settings: causes and contributing factors. Expert Rev Vaccines. 2018;17:145–161.
    1. Desselberger U. Differences of rotavirus vaccine effectiveness by country: likely causes and contributing factors. Pathogens. 2017;6:e65.
    1. Steele AD, Victor JC, Carey ME. Experiences with rotavirus vaccines: can we improve rotavirus vaccine impact in developing countries? Hum Vaccin Immunother. 2019;15:1215–1227.
    1. Yen C, Healy K, Tate JE. Rotavirus vaccination and intussusception—science, surveillance, and safety: a review of evidence and recommendations for future research priorities in low and middle income countries. Hum Vaccin Immunother. 2016;12:2580–2589.
    1. Buttery JP, Standish J, Bines JE. Intussusception and rotavirus vaccines: consensus on benefits outweighing recognized risk. Pediatr Infect Dis J. 2014;33:772–773.
    1. Tate JE, Mwenda JM, Armah G. Evaluation of intussusception after monovalent rotavirus vaccination in Africa. N Engl J Med. 2018;378:1521–1528.
    1. Groome MJ, Tate JE, Arnold M. Evaluation of intussusception after oral monovalent rotavirus vaccination in South Africa. Clin Infect Dis. 2019:ciz431.
    1. Tissera MS, Cowley D, Bogdanovic-Sakran N. Options for improving effectiveness of rotavirus vaccines in developing countries. Hum Vaccin Immunother. 2017;13:921–927.
    1. Glass RI, Jiang B, Parashar U. The future control of rotavirus disease: can live oral vaccines alone solve the rotavirus problem? Vaccine. 2018;36:2233–2236.
    1. Jiang B, Gentsch JR, Glass RI. Inactivated rotavirus vaccines: a priority for accelerated vaccine development. Vaccine. 2008;26:6754–6758.
    1. Wen X, Cao D, Jones RW, Li J, Szu S, Hoshino Y. Construction and characterization of human rotavirus recombinant VP8* subunit parenteral vaccine candidates. Vaccine. 2012;30:6121–6126.
    1. Wen X, Wen K, Cao D. Inclusion of a universal tetanus toxoid CD4(+) T cell epitope P2 significantly enhanced the immunogenicity of recombinant rotavirus ΔVP8* subunit parenteral vaccines. Vaccine. 2014;32:4420–4427.
    1. Fix AD, Harro C, McNeal M. Safety and immunogenicity of a parenterally administered rotavirus VP8 subunit vaccine in healthy adults. Vaccine. 2015;33:3766–3772.
    1. Groome MJ, Koen A, Fix A. Safety and immunogenicity of a parenteral P2-VP8-P[8] subunit rotavirus vaccine in toddlers and infants in South Africa: a randomised, double-blind, placebo-controlled trial. Lancet Infect Dis. 2017;17:843–853.
    1. Seheri M, Nemarude L, Peenze I. Update of rotavirus strains circulating in Africa from 2007 through 2011. Pediatr Infect Dis J. 2014;33(suppl 1):S76–S84.
    1. Santos N, Hoshino Y. Global distribution of rotavirus serotypes/genotypes and its implication for the development and implementation of an effective rotavirus vaccine. Rev Med Virol. 2005;15:29–56.
    1. Ward RL, Kapikian AZ, Goldberg KM, Knowlton DR, Watson MW, Rappaport R. Serum rotavirus neutralizing-antibody titers compared by plaque reduction and enzyme-linked immunosorbent assay-based neutralization assays. J Clin Microbiol. 1996;34:983–985.
    1. Leshem E, Lopman B, Glass R. Distribution of rotavirus strains and strain-specific effectiveness of the rotavirus vaccine after its introduction: a systematic review and meta-analysis. Lancet Infect Dis. 2014;14:847–856.
    1. Westerman LE, McClure HM, Jiang B, Almond JW, Glass RI. Serum IgG mediates mucosal immunity against rotavirus infection. Proc Natl Acad Sci USA. 2005;102:7268–7273.
    1. Jafari H, Deshpande JM, Sutter RW. Polio eradication. Efficacy of inactivated poliovirus vaccine in India. Science. 2014;345:922–925.
    1. Clarke E, Desselberger U. Correlates of protection against human rotavirus disease and the factors influencing protection in low-income settings. Mucosal Immunol. 2015;8:1–17.
    1. Patel M, Glass RI, Jiang B, Santosham M, Lopman B, Parashar U. A systematic review of anti-rotavirus serum IgA antibody titer as a potential correlate of rotavirus vaccine efficacy. J Infect Dis. 2013;208:284–294.
    1. Liu GF, Hille D, Kaplan SS, Goveia MG. Postdose 3 G1 serum neutralizing antibody as correlate of protection for pentavalent rotavirus vaccine. Hum Vaccin Immunother. 2017;13:2357–2363.
    1. Moon SS, Wang Y, Shane AL. Inhibitory effect of breast milk on infectivity of live oral rotavirus vaccines. Pediatr Infect Dis J. 2010;29:919–923.

Source: PubMed

3
Tilaa