SARS-CoV-2 immunity: review and applications to phase 3 vaccine candidates

Gregory A Poland, Inna G Ovsyannikova, Richard B Kennedy, Gregory A Poland, Inna G Ovsyannikova, Richard B Kennedy

Abstract

Understanding immune responses to severe acute respiratory syndrome coronavirus 2 is crucial to understanding disease pathogenesis and the usefulness of bridge therapies, such as hyperimmune globulin and convalescent human plasma, and to developing vaccines, antivirals, and monoclonal antibodies. A mere 11 months ago, the canvas we call COVID-19 was blank. Scientists around the world have worked collaboratively to fill in this blank canvas. In this Review, we discuss what is currently known about human humoral and cellular immune responses to severe acute respiratory syndrome coronavirus 2 and relate this knowledge to the COVID-19 vaccines currently in phase 3 clinical trials.

Copyright © 2020 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
The structure of the SARS-CoV-2 virion SARS-CoV-2 is a spherical, enveloped virus, with three structural proteins present in the lipid bilayer: the spike glycoprotein, the membrane protein, and the envelope protein. The nucleocapsid protein is associated with the membrane protein and is complexed with the viral RNA genome. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.
Figure 2
Figure 2
SARS-CoV-2 infection and the development of immunity The illustration depicts the major steps in the viral lifecycle and in the development of immune responses. (1) Attachment of the SARS-CoV-2 virion to the cell surface via interactions with the ACE2 cellular receptor. (2) Entry into the cell. Viral proteins can be recognised by pattern recognition receptors (eg, TLR3, TLR4, and TLR7), leading to the release of danger-associated molecular patterns, the inflammatory response, and the activation of innate anti-viral pathways. (3) Membrane fusion and release of RNA into the cell. (4) RNA translation to produce viral proteins. (5) RNA genome is copied and attached to the nucleocapsid protein. (6) Assembly of daughter SARS-CoV-2 virions. (7) Recognition of the spike glycoprotein and nucleocapsid protein (structural proteins) by the B-cell receptor. (8) B cell produces spike glycoprotein-binding antibodies and neutralising antibodies targeting the RBD region of the spike glycoprotein. (9) Viral uptake by APCs. (10) Presentation of antigens, including epitopes from structural and non-structural proteins, to T cells. (11) Activation of Th cells. (12) Activation of CTLs. (13) Th cells produce cytokines (mainly IFNγ, IL-2, and TNFα). (14) CTL recognition and killing of infected cells. ACE2=angiotensin-converting enzyme 2. APC=antigen-presenting cell. CTL=cytotoxic T lymphocyte. RBD=receptor-binding domain. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2. Th=T-helper. TLR=toll-like receptor. TNF=tumour necrosis factor.
Figure 3
Figure 3
SARS-CoV-2 proteins targeted by adaptive immune responses The four structural proteins are shown in the red boxes. Non-structural proteins and accessory factors are shown in the blue boxes. Arrows link antibodies to the viral proteins they target and identify viral proteins shown to contain epitopes targeted by CD4+ T cells or CD8+ T cells. SARS-CoV-2=severe acute respiratory syndrome coronavirus 2.

References

    1. To KK-W, Hung IF-N, Ip JD. COVID-19 re-infection by a phylogenetically distinct SARS-coronavirus-2 strain confirmed by whole genome sequencing. Clin Infect Dis. 2020 doi: 10.1093/cid/ciaa1275. published online Aug 25.
    1. Cavanaugh D. Coronaviruses and toroviruses. In: Zuckerman AJ, Banatvala JE, Pattinson JR, Griffiths P, Schoub B, editors. Principles and practice of clinical virology. 5th edn. John Wiley & Sons; London, UK: 2004. pp. 379–397.
    1. Wu LP, Wang NC, Chang YH. Duration of antibody responses after severe acute respiratory syndrome. Emerg Infect Dis. 2007;13:1562–1564.
    1. Payne DC, Iblan I, Rha B. Persistence of antibodies against Middle East respiratory syndrome coronavirus. Emerg Infect Dis. 2016;22:1824–1826.
    1. Moderbacher CR, Ramirez SI, Dan JM. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell. 2020 doi: 10.1016/j.cell.2020.09.038. published online Sept 16.
    1. Ou X, Liu Y, Lei X. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun. 2020;11
    1. Wong SK, Li W, Moore MJ, Choe H, Farzan M. A 193-amino acid fragment of the SARS coronavirus S protein efficiently binds angiotensin-converting enzyme 2. J Biol Chem. 2004;279:3197–3201.
    1. Tai W, He L, Zhang X. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell Mol Immunol. 2020;17:613–620.
    1. Jiang H-w, Li Y, Zhang H-n. Global profiling of SARS-CoV-2 specific IgG/IgM responses of convalescents using a proteome microarray. medRxiv. 2020 doi: 10.1101/2020.03.20.20039495. published online March 27. (preprint)
    1. Okba NMA, Müller MA, Li W. Severe acute respiratory syndrome coronavirus 2-specific antibody responses in coronavirus disease patients. Emerg Infect Dis. 2020;26:1478–1488.
    1. Martinez-Fleta P, Alfranca A, González-Álvaro I. SARS-Cov-2 cysteine-like protease (Mpro) is immunogenic and can be detected in serum and saliva of COVID-19-seropositive individuals. medRxiv. 2020 doi: 10.1101/2020.07.16.20155853. published online July 18. (preprint)
    1. Ni L, Ye F, Cheng ML. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. 2020;52:971. 77.e3.
    1. Padoan A, Sciacovelli L, Basso D. IgA-Ab response to spike glycoprotein of SARS-CoV-2 in patients with COVID-19: a longitudinal study. Clin Chim Acta. 2020;507:164–166.
    1. Lou B, Li T-D, Zheng S-F. Serology characteristics of SARS-CoV-2 infection since exposure and post symptom onset. Eur Respir J. 2020 doi: 10.1183/13993003.00763-2020. published online May 19.
    1. Adams ER, Ainsworth M, Anand R. Antibody testing for COVID-19: a report from the National COVID Scientific Advisory Panel. medRxiv. 2020 doi: 10.1101/2020.04.15.20066407. published online July 7. (preprint)
    1. Ibarrondo FJ, Fulcher JA, Goodman-Meza D. Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild COVID-19. N Engl J Med. 2020;383:1085–1087.
    1. Röltgen K, Wirz OF, Stevens BA. SARS-CoV-2 antibody responses correlate with resolution of RNAemia but are short-lived in patients with mild illness. medRxiv. 2020 doi: 10.1101/2020.08.15.20175794. published online Aug 17. (preprint)
    1. Liu T, Wu S, Tao H. Prevalence of IgG antibodies to SARS-CoV-2 in Wuhan—implications for the ability to produce long-lasting protective antibodies against SARS-CoV-2. medRxiv. 2020 doi: 10.1101/2020.06.13.20130252. published online June 16. (preprint)
    1. Tan W, Lu Y, Zhang J. Viral kinetics and antibody responses in patients with COVID-19. medRxiv. 2020 doi: 10.1101/2020.03.24.20042382. published online March 26. (preprint)
    1. Long QX, Tang XJ, Shi QL. Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat Med. 2020;26:1200–1204.
    1. Seow J, Graham C, Merrick B. Longitudinal evaluation and decline of antibody responses in SARS-CoV-2 infection. medRxiv. 2020 doi: 10.1101/2020.07.09.20148429. published online July 11. (preprint)
    1. Gudbjartsson DF, Norddahl GL, Melsted P. Humoral immune response to SARS-CoV-2 in Iceland. N Engl J Med. 2020 doi: 10.1056/NEJMoa2026116. published online Sept 1.
    1. Robbiani DF, Gaebler C, Muecksch F. Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature. 2020;584:437–442.
    1. Takahashi T, Ellingson MK, Wong P. Sex differences in immune responses that underlie COVID-19 disease outcomes. Nature. 2020 doi: 10.1038/s41586-020-2700-3. published online Aug 26.
    1. Jin JM, Bai P, He W. Gender differences in patients with COVID-19: focus on severity and mortality. Front Public Health. 2020;8:152.
    1. Zhang Q, Bastard P, Liu Z. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science. 2020 doi: 10.1126/science.abd4570. published online Sept 24.
    1. Bastard P, Rosen LB, Zhang Q. Auto-antibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020 doi: 10.1126/science.abd4585. published online Sept 24.
    1. Fontanet A, Cauchemez S. COVID-19 herd immunity: where are we? Nat Rev Immunol. 2020;20:583–584.
    1. Deng W, Bao L, Liu J. Primary exposure to SARS-CoV-2 protects against reinfection in rhesus macaques. Science. 2020;369:818–823.
    1. Thevarajan I, Nguyen THO, Koutsakos M. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26:453–455.
    1. Xu Z, Shi L, Wang Y. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420–422.
    1. Kuri-Cervantes L, Pampena MB, Meng W. Immunologic perturbations in severe COVID-19/SARS-CoV-2 infection. bioRxiv. 2020 doi: 10.1101/2020.05.18.101717. published online May 18. (preprint)
    1. Grifoni A, Weiskopf D, Ramirez SI. 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. Le Bert N, Tan AT, Kunasegaran K. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature. 2020;584:457–462.
    1. Braun J, Loyal L, Frentsch M. SARS-CoV-2 reactive T cells in healthy donors and patients with COVID-19. Nature. 2020 doi: 10.1038/s41586-020-2598-9. published online July 29.
    1. Peng Y, Mentzer AJ, Liu G. Broad and strong memory CD4 (+) and CD8 (+) T cells induced by SARS-CoV-2 in UK convalescent COVID-19 patients. bioRxiv. 2020 doi: 10.1101/2020.06.05.134551. published online June 8. (preprint)
    1. Sekine T, Perez-Potti A, Rivera-Ballesteros O. Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID-19. bioRxiv. 2020 doi: 10.1101/2020.06.29.174888. published online June 29. (preprint)
    1. Zhu F-C, Li Y-H, Guan X-H. 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. Crotty S. Follicular helper CD4 T cells (TFH) Annu Rev Immunol. 2011;29:621–663.
    1. Kaneko N, Kuo H-H, Boucau J. The loss of Bcl-6 expressing T follicular helper cells and the absence of Germinal Centers in COVID-19. Cell. 2020 doi: 10.1016/j.cell.2020.08.025. published online Aug 19.
    1. Meckiff BJ, Ramirez-Suastegui C, Fajardo V. Single-cell transcriptomic analysis of SARS-CoV-2 reactive CD4 (+) T cells. SSRN. 2020 doi: 10.2139/ssrn.3641939. published online July 7.
    1. Orlov M, Wander PL, Morrell ED, Mikacenic C, Wurfel MM. A case for targeting Th17 cells and IL-17A in SARS-CoV-2 infections. J Immunol. 2020;205:892–898.
    1. Tang Y, Liu J, Zhang D, Xu Z, Ji J, Wen C. Cytokine storm in COVID-19: the current evidence and treatment strategies. Front Immunol. 2020;11
    1. Hotez PJ, Bottazzi ME, Corry DB. The potential role of Th17 immune responses in coronavirus immunopathology and vaccine-induced immune enhancement. Microbes Infect. 2020;22:165–167.
    1. Liu J, Li S, Liu J. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. 2020;55
    1. Chen G, Wu D, Guo W. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130:2620–2629.
    1. Diao B, Wang C, Tan Y. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19) Front Immunol. 2020;11:827.
    1. De Biasi S, Meschiari M, Gibellini L. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat Commun. 2020;11
    1. Zheng M, Gao Y, Wang G. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell Mol Immunol. 2020;17:533–535.
    1. Laing AG, Lorenc A, Del Molino Del Barrio I. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat Med. 2020 doi: 10.1038/s41591-020-1038-6. published online Aug 17.
    1. Gallais F, Velay A, Wendling M-J. Intrafamilial exposure to SARS-CoV-2 induces cellular immune response without seroconversion. medRxiv. 2020 doi: 10.1101/2020.06.21.20132449. published online June 22. (preprint)
    1. Graham BS. Rapid COVID-19 vaccine development. Science. 2020;368:945–946.
    1. Poland GA, Ovsyannikova IG, Crooke SN, Kennedy RB. SARS-CoV-2 vaccine development: current status. Mayo Clin Proc. 2020 doi: 10.1016/j.mayocp.2020.07.021. published online Aug 30.
    1. Kaur SP, Gupta V. COVID-19 vaccine: a comprehensive status report. Virus Res. 2020;288
    1. Amanat F, Krammer F. SARS-CoV-2 vaccines: status report. Immunity. 2020;52:583–589.
    1. Hassan AO, Kafai NM, Dmitriev IP. A single-dose intranasal ChAd vaccine protects upper and lower respiratory tracts against SARS-CoV-2. Cell. 2020 doi: 10.1016/j.cell.2020.08.026. published online Aug 19.
    1. Bloomberg News China starts testing COVID-19 nasal spray vaccine. Sept 11, 2020.
    1. MediciNova MediciNova announces that its intranasal COVID-19 vaccine successfully induced systemic IgG and mucosal IgA neutralizing antibodies against SARS-CoV-2 in mice using BC-PIV vector technology. Sept 23, 2020.
    1. US Food and Drug Administration Coronavirus (COVID-19) update: FDA takes action to help facilitate timely development of safe, effective COVID-19 vaccines. June 30, 2020.
    1. Long SW, Olsen RJ, Christensen PA. Molecular architecture of early dissemination and massive second wave of the SARS-CoV-2 virus in a major metropolitan area. medRxiv. 2020 doi: 10.1101/2020.09.22.20199125. published online Sept 23. (preprint)
    1. Poland GA. Tortoises, hares, and vaccines: a cautionary note for SARS-CoV-2 vaccine development. Vaccine. 2020;38:4219–4220.
    1. van Doremalen N, Lambe T, Spencer A. ChAdOx1 nCoV-19 vaccination prevents SARS-CoV-2 pneumonia in rhesus macaques. bioRxiv. 2020 doi: 10.1101/2020.05.13.093195. published online May 13. (preprint)
    1. Folegatti PM, Ewer KJ, Aley PK. 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. Corbett KS, Flynn B, Foulds KE. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N Engl J Med. 2020 doi: 10.1056/NEJMoa2024671. published online July 28.
    1. Jackson LA, Anderson EJ, Rouphael NG. An mRNA vaccine against SARS-CoV-2—preliminary report. N Engl J Med. 2020 doi: 10.1056/NEJMoa2022483. published online July 28.
    1. Mulligan MJ, Lyke KE, Kitchin N. Phase 1/2 study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020 doi: 10.1038/s41586-020-2639-4. published online Aug 12.
    1. Loftus P. For COVID-19 vaccine, J&J plans 60,000-subject pivotal trial. Aug 20, 2020.
    1. Mercado NB, Zahn R, Wegmann F. Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques. Nature. 2020 doi: 10.1038/s41586-020-2607-z. published online July 30.
    1. Logunov DY, Dolzhikova IV, Zubkova OV. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet. 2020;396:887–897.
    1. Bucci E. Note of concern. Sept 17, 2020.
    1. Wee S-L, Simões M. In coronavirus vaccine race, China strays from the official paths. July 16, 2020.
    1. Deng C. China injects hundreds of thousands with experimental COVID-19 vaccines. Sept 12, 2020.
    1. Zhang Y-J, Zeng G, Pan H-X. 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 August 10. (preprint)
    1. Xia S, Duan K, Zhang Y. 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. Arvin AM, Fink K, Schmid MA. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. Nature. 2020;584:353–363.
    1. Tetro JA. Is COVID-19 receiving ADE from other coronaviruses? Microbes Infect. 2020;22:72–73.
    1. Taylor A, Foo SS, Bruzzone R, Dinh LV, King NJ, Mahalingam S. Fc receptors in antibody-dependent enhancement of viral infections. Immunol Rev. 2015;268:340–364.
    1. Ruckwardt TJ, Morabito KM, Graham BS. Immunological lessons from respiratory syncytial virus vaccine development. Immunity. 2019;51:429–442.
    1. Iankov ID, Pandey M, Harvey M, Griesmann GE, Federspiel MJ, Russell SJ. Immunoglobulin g antibody-mediated enhancement of measles virus infection can bypass the protective antiviral immune response. J Virol. 2006;80:8530–8540.
    1. Katzelnick LC, Gresh L, Halloran ME. Antibody-dependent enhancement of severe dengue disease in humans. Science. 2017;358:929–932.
    1. Lambert PH, Ambrosino DM, Andersen SR. Consensus summary report for CEPI/BC March 12-13, 2020 meeting: assessment of risk of disease enhancement with COVID-19 vaccines. Vaccine. 2020;38:4783–4791.
    1. Yang ZY, Werner HC, Kong WP. Evasion of antibody neutralization in emerging severe acute respiratory syndrome coronaviruses. Proc Natl Acad Sci USA. 2005;102:797–801.
    1. Yip MS, Leung NH, Cheung CY. Antibody-dependent infection of human macrophages by severe acute respiratory syndrome coronavirus. Virol J. 2014;11:82.
    1. Jaume M, Yip MS, Cheung CY. Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR pathway. J Virol. 2011;85:10582–10597.
    1. Liu L, Wei Q, Lin Q. Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight. 2019;4
    1. Chandrashekar A, Liu J, Martinot AJ. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020;369:812–817.
    1. Smatti MK, Al Thani AA, Yassine HM. Viral-induced enhanced disease illness. Front Microbiol. 2018;9
    1. Peignier A, Parker D. Trained immunity and host-pathogen interactions. Cell Microbiol. 2020 doi: 10.1111/cmi.13261. published online Sept 9.
    1. Xing Z, Afkhami S, Bavananthasivam J. Innate immune memory of tissue-resident macrophages and trained innate immunity: re-vamping vaccine concept and strategies. J Leukoc Biol. 2020;108:825–834.
    1. WHO Key criteria for the ethical acceptability of COVID-19 human challenge studies. May 6, 2020.

Source: PubMed

3
購読する