Safety and immunogenicity of an inactivated SARS-CoV-2 vaccine, BBV152: a double-blind, randomised, phase 1 trial

Raches Ella, Krishna Mohan Vadrevu, Harsh Jogdand, Sai Prasad, Siddharth Reddy, Vamshi Sarangi, Brunda Ganneru, Gajanan Sapkal, Pragya Yadav, Priya Abraham, Samiran Panda, Nivedita Gupta, Prabhakar Reddy, Savita Verma, Sanjay Kumar Rai, Chandramani Singh, Sagar Vivek Redkar, Chandra Sekhar Gillurkar, Jitendra Singh Kushwaha, Satyajit Mohapatra, Venkat Rao, Randeep Guleria, Krishna Ella, Balram Bhargava, Raches Ella, Krishna Mohan Vadrevu, Harsh Jogdand, Sai Prasad, Siddharth Reddy, Vamshi Sarangi, Brunda Ganneru, Gajanan Sapkal, Pragya Yadav, Priya Abraham, Samiran Panda, Nivedita Gupta, Prabhakar Reddy, Savita Verma, Sanjay Kumar Rai, Chandramani Singh, Sagar Vivek Redkar, Chandra Sekhar Gillurkar, Jitendra Singh Kushwaha, Satyajit Mohapatra, Venkat Rao, Randeep Guleria, Krishna Ella, Balram Bhargava

Abstract

Background: To mitigate the effects of COVID-19, a vaccine is urgently needed. BBV152 is a whole-virion inactivated SARS-CoV-2 vaccine formulated with a toll-like receptor 7/8 agonist molecule adsorbed to alum (Algel-IMDG) or alum (Algel).

Methods: We did a double-blind, multicentre, randomised, controlled phase 1 trial to assess the safety and immunogenicity of BBV152 at 11 hospitals across India. Healthy adults aged 18-55 years who were deemed healthy by the investigator were eligible. Individuals with positive SARS-CoV-2 nucleic acid and/or serology tests were excluded. Participants were randomly assigned to receive either one of three vaccine formulations (3 μg with Algel-IMDG, 6 μg with Algel-IMDG, or 6 μg with Algel) or an Algel only control vaccine group. Block randomisation was done with a web response platform. Participants and investigators were masked to treatment group allocation. Two intramuscular doses of vaccines were administered on day 0 (the day of randomisation) and day 14. Primary outcomes were solicited local and systemic reactogenicity events at 2 h and 7 days after vaccination and throughout the full study duration, including serious adverse events. Secondary outcome was seroconversion (at least four-fold increase from baseline) based on wild-type virus neutralisation. Cell-mediated responses were evaluated by intracellular staining and ELISpot. The trial is registered at ClinicalTrials.gov (NCT04471519).

Findings: Between July 13 and 30, 2020, 827 participants were screened, of whom 375 were enrolled. Among the enrolled participants, 100 each were randomly assigned to the three vaccine groups, and 75 were randomly assigned to the control group (Algel only). After both doses, solicited local and systemic adverse reactions were reported by 17 (17%; 95% CI 10·5-26·1) participants in the 3 μg with Algel-IMDG group, 21 (21%; 13·8-30·5) in the 6 μg with Algel-IMDG group, 14 (14%; 8·1-22·7) in the 6 μg with Algel group, and ten (10%; 6·9-23·6) in the Algel-only group. The most common solicited adverse events were injection site pain (17 [5%] of 375 participants), headache (13 [3%]), fatigue (11 [3%]), fever (nine [2%]), and nausea or vomiting (seven [2%]). All solicited adverse events were mild (43 [69%] of 62) or moderate (19 [31%]) and were more frequent after the first dose. One serious adverse event of viral pneumonitis was reported in the 6 μg with Algel group, unrelated to the vaccine. Seroconversion rates (%) were 87·9, 91·9, and 82·8 in the 3 μg with Algel-IMDG, 6 μg with Algel-IMDG, and 6 μg with Algel groups, respectively. CD4+ and CD8+ T-cell responses were detected in a subset of 16 participants from both Algel-IMDG groups.

Interpretation: BBV152 led to tolerable safety outcomes and enhanced immune responses. Both Algel-IMDG formulations were selected for phase 2 immunogenicity trials. Further efficacy trials are warranted.

Funding: Bharat Biotech International.

Copyright © 2021 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile Due to a sudden increase in interest from potential participants, parallel enrolment at several sites, and an oversight in the trial software, 27 of the 132 people who reached target enrolment after the planned size of 375 people was reached received a first dose and 26 of them a second dose. They were not included in further safety or immunogenicity trial data, but were monitored for safety with 3 people reporting injection site pain and 1 person a tingling sensation after the first dose, and 1 person drowsiness, 2 people headache, and 1 person injection site pain after the second dose. No serious adverse events occurred. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. *Unable to contact the participant for vaccination or withdrawal of consent.
Figure 2
Figure 2
SARS-CoV-2 IgG titres against anti-spike protein (A), receptor-binding domain (B), and nucleocapsid IgG (C) and anti-spike protein IgG1/IgG4 ratio (D) ELISA results at baseline (day 0) and 2 weeks after the second vaccination (day 28). In A–C, error bars show 95% CIs. The cutoff for detectable antibodies was 1/500. Some samples were positive for SARS-CoV-2 in the control group, as evident by the antibody titres on day 28. Endpoint titre dilution for day 28 sera samples was established with baseline (day 0), interpolated from the absorbance of the corresponding day 0 sample. Cutoff (mean ± 3 SD) for day 0 was calculated considering the absorbance of all sera dilutions (1/500 to 1/32000) tested, except the lowest dilution (1/500). ELISA titres (endpoint titres) on day 14 were not analysed. In D, the isotyping ratio was calculated (in a randomly selected subset) as IgG1/IgG4; dots show the individual datapoints and horizontal bars show means with error bars for 95% CIs. Endpoint titre=the highest sera dilution at which the absorbance was above the cutoff. GMT=geometric mean titre. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.
Figure 3
Figure 3
SARS-CoV-2 wild-type MNT50 seroconversion rates (A) and GMT (B) and PRNT50 seroconversion rates (C) and medians (D) Results at baseline (day 0), 2 weeks after the first vaccination (day 14), and 2 weeks after the second vaccination in the immunogenicity cohort. Seroconversion rates were defined by the proportion of titres achieving at least four-fold greater than baseline. In A–C, error bars show 95% CIs. In B, the human convalescent serum panel included specimens from participants with PCR-confirmed symptomatic or asymptomatic COVID-19, obtained at least 30 days after diagnosis (41 samples for MNT50). In D, randomly selected serum samples from day 28 were analysed by PRNT50 at the National Institute of Virology for homologous (NIV-2020-770) and heterologous (nCoV-Q11 and nCoV-Q100) assessments; dots show individual datapoints and horizontal bars show medians with error bars for IQRs. GMT=geometric mean titre. MNT50=microneutralisation assay. PRNT50=plaque-reduction neutralisation test. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.

References

    1. WHO Draft landscape of COVID-19 candidate vaccines. Dec 8, 2020.
    1. Graham BS. Rapid COVID-19 vaccine development. Science. 2020;368:945–946.
    1. Johnson TR, Graham BS. Contribution of respiratory syncytial virus G antigenicity to vaccine-enhanced illness and the implications for severe disease during primary respiratory syncytial virus infection. Pediatr Infect Dis J. 2004;23(suppl):S46–S57.
    1. Delgado MF, Coviello S, Monsalvo AC, et al. Lack of antibody affinity maturation due to poor toll-like receptor stimulation leads to enhanced respiratory syncytial virus disease. Nat Med. 2009;15:34–41.
    1. Philbin VJ, Dowling DJ, Gallington LC, et al. Imidazoquinoline toll-like receptor 8 agonists activate human newborn monocytes and dendritic cells through adenosine-refractory and caspase-1-dependent pathways. J Allergy Clin Immunol. 2012;130:195–204.e9.
    1. Shukla NM, Salunke DB, Balakrishna R, Mutz CA, Malladi SS, David SA. Potent adjuvanticity of a pure TLR7-agonistic imidazoquinoline dendrimer. PLoS One. 2012;7
    1. Ganneru B, Jogdand H, Dharam VK, et al. Evaluation of safety and immunogenicity of an adjuvanted, TH-1 skewed, whole virion inactivated SARS-CoV-2 vaccine—BBV152. bioRxiv. 2020 doi: 10.1101/2020.09.09.285445. published online Sept 9. (preprint)
    1. Yadav P, Ella R, Kumar S, et al. Remarkable immunogenicity and protective efficacy of BBV152, an inactivated SARS-CoV-2 vaccine in rhesus macaques. Res Square. 2020 doi: 10.21203/-65715/v1. published online Sept 10, 2020. (preprint)
    1. Mohandas S, Yadav PD, Shete A, et al. Immunogenicity and protective efficacy of BBV152: a whole virion inactivated SARS CoV-2 vaccine in the Syrian hamster model. Res Square. 2020 doi: 10.21203/-76768/v1. published online Sept 16. (preprint).
    1. Sarkale P, Patil S, Yadav PD, et al. First isolation of SARS-CoV-2 from clinical samples in India. Indian J Med Res. 2020;151:244–250.
    1. WHO Weekly epidemiological record. Jan 24, 2020.
    1. Sampath G, Madhusudana SN, Sudarshan MK, et al. Immunogenicity and safety study of indirab: a vero cell based chromatographically purified human rabies vaccine. Vaccine. 2010;28:4086–4090.
    1. Vadrevu KM, Potula V, Khalatkar V, Mahantshetty NS, Shah A, Ella R. Persistence of immune responses with an inactivated Japanese encephalitis single-dose vaccine, JENVAC and interchangeability with a live-attenuated vaccine. J Infect Dis. 2020;222:1478–1487.
    1. Bhandari N, Rongsen-Chandola T, Bavdekar A, et al. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian children in the second year of life. Vaccine. 2014;32(suppl 1):A110–A116.
    1. Ella R, Babji S, Ciarlet M, Blackwelder WC, Vadrevu KM. A randomized, open-labelled, non-inferiority phase 4 clinical trial to evaluate the immunogenicity and safety of the live, attenuated, oral rotavirus vaccine, ROTAVAC in comparison with a licensed rotavirus vaccine in healthy infants. Vaccine. 2019;37:4407–4413.
    1. Ella R, Bobba R, Muralidhar S, Babji S, Vadrevu KM, Bhan MK. A phase 4, multicentre, randomized, single-blind clinical trial to evaluate the immunogenicity of the live, attenuated, oral rotavirus vaccine (116E), ROTAVAC, administered simultaneously with or without the buffering agent in healthy infants in India. Hum Vaccin Immunother. 2018;14:1791–1799.
    1. Potdar V, Cherian SS, Deshpande GR, et al. Genomic analysis of SARS-CoV-2 strains among Indians returning from Italy, Iran & China, & Italian tourists in India. Indian J Med Res. 2020;151:255–260.
    1. Mulligan MJ, Lyke KE, Kitchin N, et al. Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;586:589–593.
    1. Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2 - preliminary report. N Engl J Med. 2020;383:1920–1931.
    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. 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. Zhu F-C, Guan X-H, Li Y-H, et al. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2020;396:479–488.
    1. Walsh EE, Frenck RW, Jr, Falsey AR, et al. Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N Engl J Med. 2020 doi: 10.1056/NEJMoa2027906. published online Oct 14.
    1. Zhang Y-J, Zeng G, Pan H-X, et al. Immunogenicity and safety of a SARS-CoV-2 inactivated vaccine in healthy adults aged 18–59 years: report of the randomized, double-blind, and placebo-controlled phase 2 clinical trial. medRxiv. 2020 doi: 10.1101/2020.07.31.20161216. published online Aug 10. (preprint)
    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. 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;21:39–51.
    1. Graepel KW, Kochhar S, Clayton EW, Edwards KE. Balancing Expediency and scientific rigor in severe acute respiratory syndrome coronavirus 2 vaccine development. J Infect Dis. 2020;222:180–182.
    1. Hotez PJ, Corry DB, Bottazzi ME. COVID-19 vaccine design: the Janus face of immune enhancement. Nat Rev Immunol. 2020;20:347–348.
    1. Diamond MS, Pierson TC. The challenges of vaccine development against a new virus during a pandemic. Cell Host Microbe. 2020;27:699–703.
    1. Weiskopf D, Schmitz KS, Raadsen MP, et al. Phenotype and kinetics of SARS-CoV-2-specific T cells in COVID-19 patients with acute respiratory distress syndrome. Sci Immunol. 2020;5
    1. Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020;181:1489. 501.e15.
    1. Arvin AM, Fink K, Schmid MA, et al. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. Nature. 2020;584:353–363.
    1. He P, Zou Y, Hu Z. Advances in aluminum hydroxide-based adjuvant research and its mechanism. Hum Vaccin Immunother. 2015;11:477–488.
    1. Schnare M, Barton GM, Holt AC, Takeda K, Akira S, Medzhitov R. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2001;2:947–950.
    1. Geeraedts F, Goutagny N, Hornung V, et al. Superior immunogenicity of inactivated whole virus H5N1 influenza vaccine is primarily controlled by toll-like receptor signalling. PLoS Pathog. 2008;4
    1. Korber B, Fischer WM, Gnanakaran S, et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell. 2020;182:812. 27.e19.
    1. Anderson EJ, Rouphael NG, Widge AT, et al. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. N Engl J Med. 2020 doi: 10.1056/NEJMoa2028436. published online Sept 29.
    1. Weissman D, Alameh M-G, de Silva T, et al. D614G spike mutation increases SARS CoV-2 susceptibility to neutralization. medRxiv. 2020 doi: 10.1101/2020.07.22.20159905. published online Sept 12. (preprint)

Source: PubMed

3
구독하다