Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18-59 years: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial

Yanjun Zhang, Gang Zeng, Hongxing Pan, Changgui Li, Yaling Hu, Kai Chu, Weixiao Han, Zhen Chen, Rong Tang, Weidong Yin, Xin Chen, Yuansheng Hu, Xiaoyong Liu, Congbing Jiang, Jingxin Li, Minnan Yang, Yan Song, Xiangxi Wang, Qiang Gao, Fengcai Zhu, Yanjun Zhang, Gang Zeng, Hongxing Pan, Changgui Li, Yaling Hu, Kai Chu, Weixiao Han, Zhen Chen, Rong Tang, Weidong Yin, Xin Chen, Yuansheng Hu, Xiaoyong Liu, Congbing Jiang, Jingxin Li, Minnan Yang, Yan Song, Xiangxi Wang, Qiang Gao, Fengcai Zhu

Abstract

Background: With the unprecedented morbidity and mortality associated with the COVID-19 pandemic, a vaccine against COVID-19 is urgently needed. We investigated CoronaVac (Sinovac Life Sciences, Beijing, China), an inactivated vaccine candidate against COVID-19, containing inactivated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), for its safety, tolerability and immunogenicity.

Methods: In this randomised, double-blind, placebo-controlled, phase 1/2 clinical trial, healthy adults aged 18-59 years were recruited from the community in Suining County of Jiangsu province, China. Adults with SARS-CoV-2 exposure or infection history, with axillary temperature above 37·0°C, or an allergic reaction to any vaccine component were excluded. The experimental vaccine for the phase 1 trial was manufactured using a cell factory process (CellSTACK Cell Culture Chamber 10, Corning, Wujiang, China), whereas those for the phase 2 trial were produced through a bioreactor process (ReadyToProcess WAVE 25, GE, Umea, Sweden). The phase 1 trial was done in a dose-escalating manner. At screening, participants were initially separated (1:1), with no specific randomisation, into two vaccination schedule cohorts, the days 0 and 14 vaccination cohort and the days 0 and 28 vaccination cohort, and within each cohort the first 36 participants were assigned to block 1 (low dose CoronaVac [3 μg per 0·5 mL of aluminium hydroxide diluent per dose) then another 36 were assigned to block 2 (high-dose Coronavc [6 μg per 0·5 mL of aluminium hydroxide diluent per dse]). Within each block, participants were randomly assigned (2:1), using block randomisation with a block size of six, to either two doses of CoronaVac or two doses of placebo. In the phase 2 trial, at screening, participants were initially separated (1:1), with no specific randomisation, into the days 0 and 14 vaccination cohort and the days 0 and 28 vaccination cohort, and participants were randomly assigned (2:2:1), using block randomisation with a block size of five, to receive two doses of either low-dose CoronaVac, high-dose CoronaVac, or placebo. Participants, investigators, and laboratory staff were masked to treatment allocation. The primary safety endpoint was adverse reactions within 28 days after injection in all participants who were given at least one dose of study drug (safety population). The primary immunogenic outcome was seroconversion rates of neutralising antibodies to live SARS-CoV-2 at day 14 after the last dose in the days 0 and 14 cohort, and at day 28 after the last dose in the days 0 and 28 cohort in participants who completed their allocated two-dose vaccination schedule (per-protocol population). This trial is registered with ClinicalTrials.gov, NCT04352608, and is closed to accrual.

Findings: Between April 16 and April 25, 2020, 144 participants were enrolled in the phase 1 trial, and between May 3 and May 5, 2020, 600 participants were enrolled in the phase 2 trial. 743 participants received at least one dose of investigational product (n=143 for phase 1 and n=600 for phase 2; safety population). In the phase 1 trial, the incidence of adverse reactions for the days 0 and 14 cohort was seven (29%) of 24 participants in the 3 ug group, nine (38%) of 24 in the 6 μg group, and two (8%) of 24 in the placebo group, and for the days 0 and 28 cohort was three (13%) of 24 in the 3 μg group, four (17%) of 24 in the 6 μg group, and three (13%) of 23 in the placebo group. The seroconversion of neutralising antibodies on day 14 after the days 0 and 14 vaccination schedule was seen in 11 (46%) of 24 participants in the 3 μg group, 12 (50%) of 24 in the 6 μg group, and none (0%) of 24 in the placebo group; whereas at day 28 after the days 0 and 28 vaccination schedule, seroconversion was seen in 20 (83%) of 24 in the 3 μg group, 19 (79%) of 24 in the 6 μg group, and one (4%) of 24 in the placebo group. In the phase 2 trial, the incidence of adverse reactions for the days 0 and 14 cohort was 40 (33%) of 120 participants in the 3 μg group, 42 (35%) of 120 in the 6 μg group, and 13 (22%) of 60 in the placebo group, and for the days 0 and 28 cohort was 23 (19%) of 120 in the 3 μg group, 23 (19%) of 120 in the 6 μg group, and 11 (18%) of 60 for the placebo group. Seroconversion of neutralising antibodies was seen for 109 (92%) of 118 participants in the 3 μg group, 117 (98%) of 119 in the 6 μg group, and two (3%) of 60 in the placebo group at day 14 after the days 0 and 14 schedule; whereas at day 28 after the days 0 and 28 schedule, seroconversion was seen in 114 (97%) of 117 in the 3 μg group, 118 (100%) of 118 in the 6 μg group, and none (0%) of 59 in the placebo group.

Interpretation: Taking safety, immunogenicity, and production capacity into account, the 3 μg dose of CoronaVac is the suggested dose for efficacy assessment in future phase 3 trials.

Funding: Chinese National Key Research and Development Program and Beijing Science and Technology Program.

Copyright © 2021 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Study profile *7 days after first dose, safety observation was done, and safety criteria were met, as determined by the data monitoring committee, participants in block 2 were then given their first dose of vaccine. †7 days after first dose of study drug in block 2, if safety criteria were met as determined by the data monitoring committee, participants enrolled in phase 2 were started on study treatment. ‡A participant in the 6 μg group was mistakenly given placebo rather than vaccine at the second dose; therefore, this participant was included in the 6 μg group dataset in the overall safety evaluation but not in the immunogenicity analysis. §Two participants did not have available antibody results, and so were not included in the immunogenicity analysis. ¶One participant did not have available antibody results, and so was not included in the immunogenicity analysis.
Figure 1
Figure 1
Study profile *7 days after first dose, safety observation was done, and safety criteria were met, as determined by the data monitoring committee, participants in block 2 were then given their first dose of vaccine. †7 days after first dose of study drug in block 2, if safety criteria were met as determined by the data monitoring committee, participants enrolled in phase 2 were started on study treatment. ‡A participant in the 6 μg group was mistakenly given placebo rather than vaccine at the second dose; therefore, this participant was included in the 6 μg group dataset in the overall safety evaluation but not in the immunogenicity analysis. §Two participants did not have available antibody results, and so were not included in the immunogenicity analysis. ¶One participant did not have available antibody results, and so was not included in the immunogenicity analysis.
Figure 2
Figure 2
Incidence of adverse reactions reported within 28 days after second dose of study drug, in the days 0 and 14 vaccination cohort in phase 1 (A) and phase 2 (C) and in the days 0 and 28 vaccination cohort in phase 1 (B) and phase 2 (D) Adverse reactions refer to the adverse events related to the vaccination. Rare injection-site symptoms reported only in the days 0 and 14 vaccination cohort are not shown in the figure and are listed in appendix 2 along with all adverse reactions after the first and second dose (pp 4–13). *The p value of comparison among three groups is significant for the incidence of any injection-site symptoms (p=0·02) and injection-site pain (p=0·04).
Figure 3
Figure 3
Antibody titres of neutralising antibodies to live SARS-CoV-2 (A–D) and RBD-specific IgG (E–H) induced after two doses of CoronaVac or placebo given in the days 0 and 14 and days 0 and 28 vaccination cohorts, in the phase 1 and phase 2 trials The error bars indicate the 95% CI of the GMT and the spots indicated the individual antibody titres, with the numbers above the spots showing the GMT estimate. Only p values for significant differences are shown on the figure, all p values for all data are in appendix 2 (p 19). GMT=geometric mean titre. RBD=receptor binding domain. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.

References

    1. Walker PGT, Whittaker C, Watson OJ, et al. The impact of COVID-19 and strategies for mitigation and suppression in low- and middle-income countries. Science. 2020;369:413–422.
    1. WHO . World Health Organization; Geneva: 2020. Coronavirus disease (COVID-19) situation report - 163.
    1. Flaxman S, Mishra S, Gandy A, et al. Estimating the effects of non-pharmaceutical interventions on COVID-19 in Europe. Nature. 2020;584:257–261.
    1. Sanche S, Lin YT, Xu C, Romero-Severson E, Hengartner N, Ke R. High contagiousness and rapid spread of severe acute respiratory syndrome coronavirus 2. Emerg Infect Dis. 2020;26:1470–1477.
    1. WHO . World Health Organization; Geneva: Oct 19, 2020. Draft landscape of COVID-19 candidate vaccines.
    1. Krammer F. SARS-CoV-2 vaccines in development. Nature. 2020;586:516–527.
    1. Callaway E. The race for coronavirus vaccines: a graphical guide. Nature. 2020;580:576–577.
    1. Corey L, Mascola JR, Fauci AS, Collins FS. A strategic approach to COVID-19 vaccine R&D. Science. 2020;368:948–950.
    1. Graham BS. Rapid COVID-19 vaccine development. Science. 2020;368:945–946.
    1. Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2 — preliminary report. N Engl J Med. 2020 doi: 10.1056/NEJMoa2022483. published July 14.
    1. Zhu F-C, Li Y-H, Guan X-H, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet. 2020;395:1845–1854.
    1. Xia S, Zhang Y, Wang Y, et al. Safety and immunogenicity of an inactivated SARS-CoV-2 vaccine, BBIBP-CorV: a randomised, double-blind, placebo-controlled, phase 1/2 trial. Lancet Infect Dis. 2020 doi: 10.1016/S1473-3099(20)30831-8. published online Oct 15.
    1. Folegatti PM, Ewer KJ, Aley PK, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet. 2020;396:467–478.
    1. Xia S, Duan K, Zhang Y, et al. Effect of an inactivated vaccine against SARS-CoV-2 on safety and immunogenicity outcomes: interim analysis of 2 randomized clinical trials. JAMA. 2020;324:951–960.
    1. Gao Q, Bao L, Mao H, Wang L, Qin C. Development of an inactivated vaccine candidate for SARS-CoV-2. Science. 2020;369:77–81.
    1. China National Medical Products Administration Guidelines for grading standards of adverse events in clinical trials of preventive vaccines. 2019.
    1. Li P, Liang Z, Gao Q, et al. Safety and immunogenicity of a novel human enterovirus 71 (EV71) vaccine: a randomized, placebo-controlled, double-blinded, phase I clinical trial. Vaccine. 2012;30:3295–3303.
    1. Nie J, Li Q, Wu J, et al. Establishment and validation of a pseudovirus neutralization assay for SARS-CoV-2. Emerg Microbes Infect. 2020;9:680–686.
    1. Wang X, Guo X, Xin, et al. Neutralizing antibody responses to severe acute respiratory syndrome coronavirus 2 in coronavirus disease 2019 inpatients and convalescent patients. Clin Infect Dis. 2020 doi: 10.1093/cid/ciaa721. published online June 4.
    1. Hao B, Chen Z, Zeng G, et al. Efficacy, safety and immunogenicity of live attenuated varicella vaccine in healthy children in China: double-blind, randomized, placebo-controlled clinical trial. Clin Microbiol Infect. 2019;25:1026–1031.
    1. Jin P, Li J, Zhou Y, et al. Immunological surrogate endpoints to evaluate vaccine efficacy. Chin J Prev Med. 2015;49:1110–1114. (in Chinese).
    1. Chen W, Xu Z, Mu J, et al. Antibody response and viraemia during the course of severe acute respiratory syndrome (SARS)-associated coronavirus infection. J Med Microbiol. 2004;53:435–438.
    1. Prévost J, Gasser R, Beaudoin-Bussières G, et al. Cross-sectional evaluation of humoral responses against SARS-CoV-2 spike. Cell Rep Med. 2020;1
    1. Payne DC, Iblan I, Rha B, et al. Persistence of antibodies against Middle East respiratory syndrome coronavirus. Emerg Infect Dis. 2016;22:1824–1826.
    1. Zhang K, Lau JY-N, Yang L, Ma Z-G. SARS-CoV-2 reinfection in two patients who have recovered from COVID-19. Precis Clin Med. 2020 doi: 10.1093/pcmedi/pbaa031. published online Sept 4.
    1. Chan PKS, Lui G, Hachim A, et al. Serologic responses in healthy adult with SARS-CoV-2 reinfection, Hong Kong, August 2020. Emerg Infect Dis. 2020 doi: 10.3201/eid2612.203833. published online Oct 22.
    1. Huang AT, Garcia-Carreras B, Hitchings MDT, et al. A systematic review of antibody mediated immunity to coronaviruses: kinetics, correlates of protection, and association with severity. Nat Commun. 2020;11

Source: PubMed

3
購読する