Heterologous prime-boost: breaking the protective immune response bottleneck of COVID-19 vaccine candidates

Qian He, Qunying Mao, Chaoqiang An, Jialu Zhang, Fan Gao, Lianlian Bian, Changgui Li, Zhenglun Liang, Miao Xu, Junzhi Wang, Qian He, Qunying Mao, Chaoqiang An, Jialu Zhang, Fan Gao, Lianlian Bian, Changgui Li, Zhenglun Liang, Miao Xu, Junzhi Wang

Abstract

COVID-19 vaccines emerging from different platforms differ in efficacy, duration of protection, and side effects. To maximize the benefits of vaccination, we explored the utility of employing a heterologous prime-boost strategy in which different combinations of the four types of leading COVID-19 vaccine candidates that are undergoing clinical trials in China were tested in a mouse model. Our results showed that sequential immunization with adenovirus vectored vaccine followed by inactivated/recombinant subunit/mRNA vaccine administration specifically increased levels of neutralizing antibodies and promoted the modulation of antibody responses to predominantly neutralizing antibodies. Moreover, a heterologous prime-boost regimen with an adenovirus vector vaccine also improved Th1-biased T cell responses. Our results provide new ideas for the development and application of COVID-19 vaccines to control the SARS-CoV-2 pandemic.

Keywords: COVID-19 vaccine; Heterologous prime-boost; T cell response; neutralizing antibody; th1/th2 balance.

Conflict of interest statement

No potential conflict of interest was reported by the author(s).

Figures

Figure 1.
Figure 1.
Comparison of humeral immune responses induced by COVID-19 vaccines of different technology platforms and heterologous prime-boost regimens. (A). Schematic representation of experimental protocols and immunization groups. Mice in 9 groups were immunized with different COVID-19 vaccines or vaccine combinations: rAd, 2 × INA, 2 × rRBD,, rAd > INA, INA > rAd, rAd > rRBD, rRBD > rAd, INA > rRBD, rRBD > INA. (rAd: recombinant Ad5 vectored vaccine, INA: inactivated vaccine, rRBD: recombinant RBD vaccine), Mice in a blank control group were sham-vaccinated with PBS. For rAd group, mice were immunized with one dose of rAd vaccine and blood samples were collected 14 days post -vaccination; for other groups, blood samples were only collected 14 days after the second vaccine dose (B,C). Serum Nab levels measured by live SARS-CoV-2 virus (B) and pseudovirus (C). NAb titres are expressed as 50% inhibitory dilution (EC50) of serum. D. Spike-specific binding IgG titres were measured by ELISA (n = 8–10 per group, one spot represents one sample). Bars represent means ± SD, **p < 0.01, ****p < 0.0001, ns: p > 0.05.
Figure 1.
Figure 1.
Comparison of humeral immune responses induced by COVID-19 vaccines of different technology platforms and heterologous prime-boost regimens. (A). Schematic representation of experimental protocols and immunization groups. Mice in 9 groups were immunized with different COVID-19 vaccines or vaccine combinations: rAd, 2 × INA, 2 × rRBD,, rAd > INA, INA > rAd, rAd > rRBD, rRBD > rAd, INA > rRBD, rRBD > INA. (rAd: recombinant Ad5 vectored vaccine, INA: inactivated vaccine, rRBD: recombinant RBD vaccine), Mice in a blank control group were sham-vaccinated with PBS. For rAd group, mice were immunized with one dose of rAd vaccine and blood samples were collected 14 days post -vaccination; for other groups, blood samples were only collected 14 days after the second vaccine dose (B,C). Serum Nab levels measured by live SARS-CoV-2 virus (B) and pseudovirus (C). NAb titres are expressed as 50% inhibitory dilution (EC50) of serum. D. Spike-specific binding IgG titres were measured by ELISA (n = 8–10 per group, one spot represents one sample). Bars represent means ± SD, **p < 0.01, ****p < 0.0001, ns: p > 0.05.
Figure 2.
Figure 2.
Humeral immune responses induced by heterologous prime-boost regimen of adenovirus vectored and mRNA-based COVID-19 vaccine. (A). Schematic representation of experimental protocol and immunization groups. Mice in 4 groups were immunized with adenovirus vectored vaccine or mRNA vaccine: rAd, 2 × mRNA, rAd > mRNA, mRNA > rAd. (rAd: recombinant Ad5 vectored vaccine, RNA: mRNAbased vaccine). For the rAd group, mice were immunized with one dose of rAd vaccine and blood samples were collected 14 days post-vaccination; for other groups, bloods were collected 14 days post the second vaccine dose. (B). NAbs of serum measured by live SARS-CoV-2 virus and expressed as 50% inhibitory dilution (EC50) of serum. (C). Spike-specific binding IgG titres were measured by ELISA (n = 8–10 per group, one spot represents one sample). Bars represent means ± SD, **p < 0.01, ****p < 0.0001, ns: p > 0.05.
Figure 2.
Figure 2.
Humeral immune responses induced by heterologous prime-boost regimen of adenovirus vectored and mRNA-based COVID-19 vaccine. (A). Schematic representation of experimental protocol and immunization groups. Mice in 4 groups were immunized with adenovirus vectored vaccine or mRNA vaccine: rAd, 2 × mRNA, rAd > mRNA, mRNA > rAd. (rAd: recombinant Ad5 vectored vaccine, RNA: mRNAbased vaccine). For the rAd group, mice were immunized with one dose of rAd vaccine and blood samples were collected 14 days post-vaccination; for other groups, bloods were collected 14 days post the second vaccine dose. (B). NAbs of serum measured by live SARS-CoV-2 virus and expressed as 50% inhibitory dilution (EC50) of serum. (C). Spike-specific binding IgG titres were measured by ELISA (n = 8–10 per group, one spot represents one sample). Bars represent means ± SD, **p < 0.01, ****p < 0.0001, ns: p > 0.05.
Figure 3.
Figure 3.
T cell responses to SARS-CoV-2 spike peptides measured by IFN-γ ELISPOT. 6 mice in Figure 1 were sacrificed and T cell response were measured. Isolated lymphocytes were stimulated with 4 peptide pools spanning spike respectively, and the IFN-γ secreting cells were quantified by ELISPOT assay. (n = 6 per group, one spot represents one sample). Bars represent means ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Figure 3.
Figure 3.
T cell responses to SARS-CoV-2 spike peptides measured by IFN-γ ELISPOT. 6 mice in Figure 1 were sacrificed and T cell response were measured. Isolated lymphocytes were stimulated with 4 peptide pools spanning spike respectively, and the IFN-γ secreting cells were quantified by ELISPOT assay. (n = 6 per group, one spot represents one sample). Bars represent means ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
Figure 4.
Figure 4.
Multiplex cytokine analysis for different immunization regimens. Isolated lymphocytes in 9 regimens identified in Figure 1 were stimulated with 4 spike peptide pools. Supernatants pooled by different groups (A) or different peptide pools (B) were collected. IL-2, IL-4, IL-10 and TNF-α levels in supernatants were measured by MSD; the concentration of each cytokine(pg/mL) is represented by histogram (A) or heatmap (B) (n = 6 per group). Bars represent means ± SEM, *p < 0.05, **p < 0.01, ****p < 0.0001.
Figure 4.
Figure 4.
Multiplex cytokine analysis for different immunization regimens. Isolated lymphocytes in 9 regimens identified in Figure 1 were stimulated with 4 spike peptide pools. Supernatants pooled by different groups (A) or different peptide pools (B) were collected. IL-2, IL-4, IL-10 and TNF-α levels in supernatants were measured by MSD; the concentration of each cytokine(pg/mL) is represented by histogram (A) or heatmap (B) (n = 6 per group). Bars represent means ± SEM, *p < 0.05, **p < 0.01, ****p < 0.0001.

References

    1. Polack FP, Thomas SJ, Kitchin N, et al. . Safety and efficacy of the BNT162b2 mRNA covid-19 vaccine. N Engl J Med. 2020;383:2603–2615.
    1. Voysey M, Clemens SAC, Madhi SA, et al. . Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. The Lancet. 2020;397:99–111.
    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. Implications of the Emerging SARS-CoV-2 Variant VOC 202012/01.
    1. Davies NG, Barnard RC, Jarvis CI, et al. . Estimated transmissibility and severity of novel SARS-CoV-2 Variant of concern 202012/01 in england. MedRxiv. 2020.
    1. Widge AT, Rouphael NG, Jackson LA, et al. . Durability of responses after SARS-CoV-2 mRNA-1273 vaccination. N Engl J Med. 2020;384:80–82.
    1. Ramasamy MN, Minassian AM, Ewer KJ, et al. . Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): a single-blind, randomised, controlled, phase 2/3 trial. The Lancet. 2020;396:1979–1993.
    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. 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;324:1–10.
    1. Keech C, Albert G, Cho I, et al. . Phase 1-2 trial of a SARS-CoV2 recombinant spike protein nanoparticle vaccine. N Engl J Med. 2020;383:2320–2332.
    1. Zhang Y, Zeng G, Pan H, et al. . Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18-59 years: a randomised, doubleblind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect Dis. 2020;21:181–192.
    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. 2021;21:39–51.
    1. Yang S, Li Y, Dai L, et al. . Safety and immunogenicity of a recombinant tandem-repeat dimeric RBD protein vaccine against COVID-19 in adults: pooled analysis of two randomized, double-blind, placebo-controlled, phase 1 and 2 trials. medRxiv. 2020;396:479–488.
    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. Dagotto G, Yu J, Barouch DH.. Approaches and challenges in SARS-CoV-2 vaccine development. Cell Host Microbe. 2020;28:364–370.
    1. Excler JL, Kim JH.. Novel prime-boost vaccine strategies against HIV-1. Expert Rev Vaccines. 2019;18:765–779.
    1. Lu S. Heterologous prime-boost vaccination. Curr Opin Immunol. 2009;21:34651.
    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. McMahan K, Yu J, Mercado NB, et al. . Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature. 2020;590:630–634.
    1. Wang S, Parker C, Taaffe J, et al. . Heterologous HA DNA vaccine prime–inactivated influenza vaccine boost is more effective than using DNA or inactivated vaccine alone in eliciting antibody responses against H1 or H3 serotype influenza viruses. Vaccine. 2008;26:3626–3633.
    1. Gil A, Shen S, Coley S, et al. . DNA vaccine prime followed by boost with live attenuated virus significantly improves antigen-specific T cell responses against human cytomegalovirus. Hum Vaccin Immunother. 2013;9:2120–2132.
    1. Chen P, Nirula A, Heller B, et al. . SARS-CoV-2 neutralizing antibody LY-CoV555 in outpatients with covid-19. N Engl J Med. 2020;384:229–237.
    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;11:4704.
    1. Baden LR, El Sahly HM, Essink B, et al. . Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2020;384:403–416.
    1. Walsh EE, Frenck RW, Falsey AR, et al. . Safety and immunogenicity of two RNA-based covid-19 vaccine candidates. N Engl J Med. 2020;383:2439–2450.
    1. Diamond MS, Pierson TC.. Molecular insight into dengue virus pathogenesis and Its Implications for disease control. Cell. 2015;162:488–492.
    1. Lee WS, Wheatley AK, Kent SJ, et al. . Antibody-dependent enhancement and SARSCoV-2 vaccines and therapies. Nature Microbiology. 2020;5:1185–1191.
    1. Logunov DY, Dolzhikova IV, Zubkova OV, et al. . Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous primeboost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from russia. Lancet. 2020;396:887–897.
    1. Hollister K, Chen Y, Wang S, et al. . The role of follicular helper T cells and the germinal center in HIV-1 gp120 DNA prime and gp120 protein boost vaccination. Hum Vaccin Immunother. 2014;10:1985–1992.
    1. Qi H, Chen X, Chu C, et al. . Tfh cell differentiation and their function in promoting B-cell responses. Adv Exp Med Biol. 2014;841:15380.
    1. Law H, Venturi V, Kelleher A, et al. . Tfh cells in health and immunity: potential targets for systems biology Approaches to vaccination. Int J Mol Sci. 2020;21:8524.
    1. Wang S, Arthos J, Lawrence JM, et al. . Enhanced immunogenicity of gp120 protein when combined with recombinant DNA priming To generate antibodies that neutralize the JR-FL primary isolate of human immunodeficiency virus type 1. J Virol. 2005;79:7933–7937.

Source: PubMed

3
Abonneren