Register      Login
Microbiology Australia Microbiology Australia Society
Microbiology Australia, bringing Microbiologists together
RESEARCH ARTICLE (Open Access)

Long-term and short-term immunity to SARS-CoV-2: why it matters

John Zaunders A C and Chansavath Phetsouphanh B
+ Author Affiliations
- Author Affiliations

A Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia

B Kirby Institute, UNSW Sydney, Australia

C Email: j.zaunders@amr.org.au

Microbiology Australia 42(1) 34-38 https://doi.org/10.1071/MA21010
Submitted: 29 January 2021  Accepted: 10 March 2021   Published: 13 April 2021

Journal Compilation © The Authors 2021 Open Access CC BY, published (by CSIRO Publishing) on behalf of the ASM

Abstract

The adaptive immune system, regulated by CD4 T cells, is essential for control of many viral infections. Endemic coronavirus infections generally occur as short-term upper respiratory tract infections which in many cases appear to be cleared before adaptive immunity is fully involved, since adaptive immunity takes approximately 1.5–2 weeks to ramp up the response to a primary infection, or approximately 1 week for a recurrent infection. However, the adaptive immune response to SARS-CoV-2 infection will be critical to full recovery with minimal long-lasting effects, and to either prevention of recurrence of infection or at least reduced severity of symptoms. The detailed kinetics of this infection versus the dynamics of the immune response, including in vaccinated individuals, will largely determine these outcomes.

Introduction

SARS-CoV-2 is the third highly pathogenic coronavirus zoonosis from bats, which have evolved interferon and adaptive immune responses that allow many such viruses to co-exist without disease, but still permitting transmission1. In humans, transmission of SARS-CoV-2 to other individuals will typically occur at the peak of viral load in the first 5 days of symptoms2.


Innate immunity

The earliest mechanism combating an acute, short-lived viral infection of an epithelial barrier, such as with SARS-CoV-2, involves type-1 and type-III interferons (IFN-I and IFN-λ)3, prior to the adaptive immune response. Therefore, the first phase of the current pandemic is likely due to sufficient evasion of the early IFN response. Amongst patients with severe life-threatening pneumonia, 3% have inborn genetic errors in the IFN-I response pathway4, and 13% have autoantibodies that prevent IFN-I signalling5, much higher rates than in the general population. Also, it has been reported that COVID-19 patients with severe disease have lower6, or delayed IFN production7,8, consistent with evidence that exogenous IFN can limit coronavirus replication in vitro, and that these viruses have non-structural proteins that help evade and antagonise innate immunity9,10.

Highly specialised circulating plasmacytoid dendritic cells (PDCs) also sense dsRNA and produce large amounts of exogenous IFN-Is11. Importantly, numerically, these PDC’s decrease with age12, which may add to the reasons that older patients do worse after SARS-CoV-2 infection than paediatric and adolescent cases13, who can be PCR negative in the same household as PCR+ adults14. Overall, later adaptive immune responses could become more critical in older patients.


Primary human immune responses to viral infection

Neutralising and opsonising antibodies will likely be extremely important to finally clear all virus in older adults. Neutralising antibody (nAb) responses to viral infections are regarded as the main protective mechanism of effective vaccines15, and generally require a concomitant CD4 T-cell response to initiate germinal centres where they help B cells increase the avidity of their antibodies by somatic hypermutation and switch immunoglobulin to important sub-classes of IgG16. CD4 T cells also help expanded specific B cell clones to generate long-term memory B cells, and are believed to be important also for CD8 T-cell responses17.

During a primary response, naïve B cells are multipotent with different fate pathways, including: (1) short-term proliferating IgM and IgG antibody-secreting plasmablasts in the circulation and the spleen that are independent of CD4 T-cell help; (2) longer lived plasma cells, generated via germinal centre reactions with CD4 T-cell help, that can mature into bone marrow-resident, non-proliferating cells that produce antibody in large amounts; and (3) memory B cells from germinal centres that can rapidly become antibody-secreting upon re-encounter with antigen in secondary lymphoid tissues (reviewed in Akkaya et al.18). Important variables include: B cell receptor affinity; antigen structural pattern, especially valency; signalling via TLR’s, especially TLR7 and TLR9 for viral antigens; activation of CD4 T cells with expression of CD40, IL-4 and IL-21; and generation of germinal centres18. Different B cell fates are mediated by differential expression of transcription factors, Bcl6 in germinal centre B cells, Blimp in plasma cells, and the level of IRF4 expression in response to the affinity and strength of the B cell receptor signalling18,19. Empirically, all these different fate decisions can be effective, such as the examples of immunity resulting from: T-cell-independent responses to multivalent pneumococcal vaccine without generating memory B cells20; nAb responses, with minimal somatic hypermutation, to highly multivalent virus like particles of the HPV vaccine21; and T-cell-associated lifelong nAb responses to measles infection and vaccinia inoculation15.

Most studies of T cells during human acute viral infections have begun after diagnosis, and exposure date is often unclear, but we closely studied the immune response before and after inoculation of healthy volunteers with vaccinia virus22. Typically, the draining axillary lymph node was tender by days 7–8, the inoculation site was edematous at days 9–11, antigen-specific CD4 T cells appeared in the circulation at days 11–14 and serum nAb responses appeared between days 14–2122, as later confirmed for antigen-specific CD8 T cells23, and consistent with murine primary responses to influenza infection24.

Overall, primary adaptive immune responses take longer than the time course of a typical mildly symptomatic SARS-CoV-2 infection, with 65% of such individuals reporting a return to usual health within a median of 7 days from onset of symptoms25.


Secondary immune responses to viral re-infection

By comparison, annual vaccination to influenza leads to enrichment of circulating antigen-specific CD4 T cells at day 726, which is 4–5 days later than the peak and start of the decline of influenza viral titres in human challenge studies27.

In murine influenza challenge models, large populations of antigen-specific CD8 T cells present in the lungs are associated with shortening the duration of peak viral load in lungs by about 2–3 days, compared to the viral load during the primary response28.


Antibody responses to SARS-CoV-2 infection

The kinetics of antibody responses to SARS-CoV-2 show that most patients seroconvert with the appearance of specific IgG between day 14 and day 21, peaking around day 3029, but IgG levels thereafter decrease in serum by about half in the ensuing month30. Numerous studies have found nAb in the vast majority of COVID-19 patients, but there is a very wide range of titres, with highest titres associated with severe symptoms29,31,32.

Acute SARS-CoV-2 infection is associated with a variable increase in plasmablasts in the circulation3335, which is resolved during convalescence35. These relatively immature B cells spontaneously secrete immunoglobulins independent of CD4 help, but may be short-lived cells, lost soon after viral clearance18,35. Overall, the early drop in antibody levels30 may be mainly due to resolution of the acute plasmablast response.

However, around the time of antibody appearance, COVID-19 patients who go on to develop life-threatening pneumonia begin to exhibit worsening symptoms2. This raises the question of whether the immune response could also be unfavourable. It is possible that high levels of opsonising antibodies enhance viral entry into cells with Fc receptors, such as mucosal epithelial cells that transcytose immunoglobulins to and from the lumen at intestinal sites, possibly making the infection worse36. Gastrointestinal symptoms are very common37, and antibody dependent enhancement could be involved, in addition to direct infection of ACE2+ enterocytes.

Therefore, it is imperative to better understand the role of B cells and antibodies in SARS-CoV-2 infection, as well as vaccination, including: the affinity of naïve B cells for epitopes in spike protein, particularly the receptor binding domain (RBD); does SARS-CoV-2, as an RNA virus, trigger TLR7 and TLR9 signalling in B cells; what are the CD4 epitopes in spike and RBD to help B cells; numbers of naïve CD4 T cells available to respond and form germinal centres; and how much somatic hypermutation is actually required for the most effective nAb. It has been reported that nAb to SARS-CoV-2 may not require a large number of mutations relative to germ-line immunoglobulin gene sequences38. While two reports from autopsy studies suggest a lack of germinal centres in fatal COVID-1939,40, another study did not find abnormal lymph node architecture if tissues with autolysis were stringently excluded41, so that the role of germinal centres is unresolved.


What do we know from other coronavirus infections?

A study from the Netherlands found that most children were seropositive for antibodies to the nucleocapsid of NL63 by ages 3–642. However, detailed measurements of anti-NL63, or anti-229E or anti-OC43 antibodies, using longitudinal serum samples from adult males over 30 years, shows a pattern of boosted antibodies typically every 1–3 years, presumed to be due to intermittent re-infections, with waning of antibody levels between re-infections43.

It is also possible that, in these recurrent infections, viral mutation in circulating strains might be as important as waning antibodies, but this is unknown. In the case of HIV-1 infection, chronic viral replication is partly enabled by escape mutants from nAb and chronic germinal centre responses44. Therefore, newly arising mutant SARS-CoV-2 strains that can escape nAb32 will be globally important.

There have been few published studies of memory T cells specific for endemic coronaviruses, and these have shown quite low levels of IFN-γ ELISPOT responses in PBMC, to spike proteins from NL63, 229E or OC4345, or to nucleocapsid from SARS-CoV-1 in survivors, 17 years after recovery46. Notably, these T-cell responses were undetectable in approximately half of the patients studied, which contrasts with the higher prevalence of specific antibodies.


T-cell responses in SARS-CoV-2 infection

Many studies have addressed the early dynamics of the T-cell immune response to SARS-CoV-2 infection47,48. During the acute infection, activated, proliferating T cells are readily detected by either flow cytometry3335 or single cell RNA sequencing49, as previously well described for antigen-specific lymphocytes during various other human acute viral infections22,23,50,51. Most experimental studies of murine antiviral T cells have concentrated on the acute cytotoxic CD8 T-cell response28, but eventually it was found that a later germinal centre and nAb response was critical to full viral clearance52.

Importantly, then, CD4 T-cells responses to SARS-CoV-2 antigens were generally more prevalent in COVID-19 patients than CD8 responses47,48, although both CD4 and CD8 activated cells are found in some but not all acute patients at day 734. Most studies have used PBMC ELISPOT assays to quantify SARS-CoV-2 specific T cells, but others have used the intracellular cytokine assay or the AIM assay of upregulation of activation markers47,48, similar to our original OX40 assay53, to ascribe responses to CD4 or CD8 T-cell subsets. We have confirmed the presence of SARS-CoV-2 specific CD4 T cells in recovered patients in the ADAPT Study at St Vincent’s Hospital, Sydney, using the OX40 assay, but like other studies, have found that about half of healthy non-COVD-19 controls had responses to pools of peptides from the full SARS-CoV-2 spike protein sequence, presumably cross-reacting with endemic coronaviruses54.

SARS-CoV-2 specific memory CD4 and CD8 T cells were still present in recovered patients at 9 months after symptom onset in one study55, and at 6-8 months in another study, with an apparent half-life of only 3–5 months56.

We have also concentrated on proliferative CD4 T-cell responses to the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, which we only found in COVID-19 patients and which were highly correlated with the patients’ serum anti-spike IgG and IgM antibodies and nAb. Proliferative CD4 T-cell responses will likely allow rapid expansion of RBD-specific CD4 T cells in vivo, on re-infection, or vaccination, which will help rapid expansion of RBD-specific memory B cells and boost nAb levels. It has not yet been reported whether the new vaccines induce long-term proliferative memory CD4 T cells.

However, the T-cell response could also possibly be detrimental, since many of the cytokines associated with acute respiratory distress syndrome (ARDS) may involve T cells, particularly those recruiting neutrophils to the lungs, including IL-17 produced by Th17 CD4 T cells57. Infected alveolar macrophages may also amplify damaging T-cell pro-inflammatory responses58, and it has been speculated that development of highly activated cytotoxic T cells may also cause damage by widespread killing of infected epithelial cells in the lung59.

Recently, two early studies of real-world post-vaccine protection in Israel60 and in healthcare workers (HCW) in the UK61 have reported that vaccine effectiveness was 51% and 72%, respectively. The latter study concluded that HCW will still require PPE and physical distancing, as well as regular testing for asymptomatic infection.


Conclusions

There are many important questions that so far remain unanswered including: (1) whether pre-existing cross-reactive antibodies or T cells influence the outcome of infection; (2) whether, overall, antibodies, T cells and inflammatory cytokines are beneficial or even detrimental; (3) the longevity of natural and vaccinated immune responses; (4) whether individuals with weaker responses are still protected; and (5) whether SARS-CoV-2 mutant strains can evade neutralising antibodies, and could T-cell immunity compensate?

Both short-term innate immunity and the later-to-develop adaptive immunity dictate outcomes to infection, and longer-term adaptive immunity in conjunction with vaccination, will determine whether COVID-19 becomes a relatively benign seasonal illness.


Conflicts of interest

The authors declare no conflicts of interest.



Acknowledgements

This research did not receive any specific funding.


References

[1]  Clayton, E. and Munir, M. (2020) Fundamental characteristics of bat interferon systems. Front. Cell. Infect. Microbiol. 10, 527921.
Fundamental characteristics of bat interferon systems.Crossref | GoogleScholarGoogle Scholar | 33363045PubMed |

[2]  Cevik, M. et al. (2020) Virology, transmission, and pathogenesis of SARS-CoV-2. BMJ 371, m3862.
Virology, transmission, and pathogenesis of SARS-CoV-2.Crossref | GoogleScholarGoogle Scholar | 33097561PubMed |

[3]  Andreakos, E. et al. (2019) Lambda interferons come to light: dual function cytokines mediating antiviral immunity and damage control. Curr. Opin. Immunol. 56, 67–75.
Lambda interferons come to light: dual function cytokines mediating antiviral immunity and damage control.Crossref | GoogleScholarGoogle Scholar | 30399529PubMed |

[4]  Zhang, Q. et al. (2020) Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 370, eabd4570.
Inborn errors of type I IFN immunity in patients with life-threatening COVID-19.Crossref | GoogleScholarGoogle Scholar | 33243877PubMed |

[5]  Bastard, P. et al. (2020) Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 370, eabd4585.
Autoantibodies against type I IFNs in patients with life-threatening COVID-19.Crossref | GoogleScholarGoogle Scholar | 32972996PubMed |

[6]  Hadjadj, J. et al. (2020) Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724.
Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients.Crossref | GoogleScholarGoogle Scholar | 32661059PubMed |

[7]  Blanco-Melo, D. et al. (2020) Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181, 1036–1045.e9.
Imbalanced host response to SARS-CoV-2 drives development of COVID-19.Crossref | GoogleScholarGoogle Scholar | 32416070PubMed |

[8]  Galani, I.E. et al. (2021) Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 22, 32–40.
Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison.Crossref | GoogleScholarGoogle Scholar | 33277638PubMed |

[9]  Lei, X. et al. (2020) Activation and evasion of type I interferon responses by SARS-CoV-2. Nat. Commun. 11, 3810.
Activation and evasion of type I interferon responses by SARS-CoV-2.Crossref | GoogleScholarGoogle Scholar | 32733001PubMed |

[10]  Kikkert, M. (2020) Innate immune evasion by human respiratory RNA viruses. J. Innate Immun. 12, 4–20.
Innate immune evasion by human respiratory RNA viruses.Crossref | GoogleScholarGoogle Scholar | 31610541PubMed |

[11]  Liu, Y.J. (2005) IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu. Rev. Immunol. 23, 275–306.
IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors.Crossref | GoogleScholarGoogle Scholar | 15771572PubMed |

[12]  Splunter, M.V. et al. (2019) Plasmacytoid dendritic cell and myeloid dendritic cell function in ageing: a comparison between elderly and young adult women. PLoS One 14, e0225825.
Plasmacytoid dendritic cell and myeloid dendritic cell function in ageing: a comparison between elderly and young adult women.Crossref | GoogleScholarGoogle Scholar | 31830086PubMed |

[13]  Götzinger, F. et al. (2020) COVID-19 in children and adolescents in Europe: a multinational, multicentre cohort study. Lancet Child Adolesc. Health 4, 653–661.
COVID-19 in children and adolescents in Europe: a multinational, multicentre cohort study.Crossref | GoogleScholarGoogle Scholar | 32593339PubMed |

[14]  Tosif, S. et al. (2020) Immune responses to SARS-CoV-2 in three children of parents with symptomatic COVID-19. Nat. Commun. 11, 5703.
Immune responses to SARS-CoV-2 in three children of parents with symptomatic COVID-19.Crossref | GoogleScholarGoogle Scholar | 33177504PubMed |

[15]  Pantaleo, G. and Koup, R.A. (2004) Correlates of immune protection in HIV-1 infection: what we know, what we don’t know, what we should know. Nat. Med. 10, 806–810.
Correlates of immune protection in HIV-1 infection: what we know, what we don’t know, what we should know.Crossref | GoogleScholarGoogle Scholar | 15286782PubMed |

[16]  Crotty, S. (2011) Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663.
Follicular helper CD4 T cells (TFH).Crossref | GoogleScholarGoogle Scholar | 21314428PubMed |

[17]  Walker, B. and McMichael, A. (2012) The T-cell response to HIV. Cold Spring Harb. Perspect. Med. 2, a007054.
The T-cell response to HIV.Crossref | GoogleScholarGoogle Scholar | 23002014PubMed |

[18]  Akkaya, M. et al. (2020) B cell memory: building two walls of protection against pathogens. Nat. Rev. Immunol. 20, 229–238.
B cell memory: building two walls of protection against pathogens.Crossref | GoogleScholarGoogle Scholar | 31836872PubMed |

[19]  Tellier, J. et al. (2016) Blimp-1 controls plasma cell function through the regulation of immunoglobulin secretion and the unfolded protein response. Nat. Immunol. 17, 323–330.
Blimp-1 controls plasma cell function through the regulation of immunoglobulin secretion and the unfolded protein response.Crossref | GoogleScholarGoogle Scholar | 26779600PubMed |

[20]  Papadatou, I. et al. (2019) The role of serotype-specific immunological memory in pneumococcal vaccination: current knowledge and future prospects. Vaccines (Basel) 7, 13.
The role of serotype-specific immunological memory in pneumococcal vaccination: current knowledge and future prospects.Crossref | GoogleScholarGoogle Scholar |

[21]  Harper, D.M. and DeMars, L.R. (2017) HPV vaccines – a review of the first decade. Gynecol. Oncol. 146, 196–204.
HPV vaccines – a review of the first decade.Crossref | GoogleScholarGoogle Scholar | 28442134PubMed |

[22]  Zaunders, J.J. et al. (2006) CD127+CCR5+CD38+++ CD4+ Th1 effector cells are an early component of the primary immune response to vaccinia virus and precede development of interleukin-2+ memory CD4+ T cells. J. Virol. 80, 10151–10161.
CD127+CCR5+CD38+++ CD4+ Th1 effector cells are an early component of the primary immune response to vaccinia virus and precede development of interleukin-2+ memory CD4+ T cells.Crossref | GoogleScholarGoogle Scholar | 17005692PubMed |

[23]  Miller, J.D. et al. (2008) Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity 28, 710–722.
Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines.Crossref | GoogleScholarGoogle Scholar | 18468462PubMed |

[24]  Hornick, E.E. et al. (2019) Kinetics and phenotype of the CD4 T cell response to influenza virus infections. Front. Immunol. 10, 2351.
Kinetics and phenotype of the CD4 T cell response to influenza virus infections.Crossref | GoogleScholarGoogle Scholar | 31632414PubMed |

[25]  Tenforde, M.W. et al. (2020) Symptom duration and risk factors for delayed return to usual health among outpatients with COVID-19 in a multistate health care systems network – United States, March-June 2020. MMWR Morb. Mortal. Wkly Rep. 69, 993–998.
Symptom duration and risk factors for delayed return to usual health among outpatients with COVID-19 in a multistate health care systems network – United States, March-June 2020.Crossref | GoogleScholarGoogle Scholar | 32730238PubMed |

[26]  Herati, R.S. et al. (2017) Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells. Sci. Immunol. 2, eaag2152.
Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells.Crossref | GoogleScholarGoogle Scholar | 28620653PubMed |

[27]  Baccam, P. et al. (2006) Kinetics of influenza A virus infection in humans. J. Virol. 80, 7590–7599.
Kinetics of influenza A virus infection in humans.Crossref | GoogleScholarGoogle Scholar | 16840338PubMed |

[28]  Doherty, P.C. and Christensen, J.P. (2000) Accessing complexity: the dynamics of virus-specific T cell responses. Annu. Rev. Immunol. 18, 561–592.
Accessing complexity: the dynamics of virus-specific T cell responses.Crossref | GoogleScholarGoogle Scholar | 10837069PubMed |

[29]  Seow, J. et al. (2020) Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans. Nat. Microbiol. 5, 1598–1607.
Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans.Crossref | GoogleScholarGoogle Scholar | 33106674PubMed |

[30]  Ibarrondo, F.J. et al. (2020) Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild Covid-19. N. Engl. J. Med. 383, 1085–1087.
Rapid decay of anti-SARS-CoV-2 antibodies in persons with mild Covid-19.Crossref | GoogleScholarGoogle Scholar | 32706954PubMed |

[31]  Wajnberg, A. et al. (2020) Robust neutralizing antibodies to SARS-CoV-2 infection persist for months. Science 370, 1227–1230.
Robust neutralizing antibodies to SARS-CoV-2 infection persist for months.Crossref | GoogleScholarGoogle Scholar | 33115920PubMed |

[32]  Tea  F.et al2020 SARS-CoV-2 neutralizing antibodies; longevity, breadth, and evasion by emerging viral variants.medRxiv[Preprint].

[33]  Thevarajan, I. et al. (2020) Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat. Med. 26, 453–455.
Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19.Crossref | GoogleScholarGoogle Scholar | 32284614PubMed |

[34]  Mathew, D. et al. (2020) Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369, eabc8511.
Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications.Crossref | GoogleScholarGoogle Scholar | 32669297PubMed |

[35]  Kuri-Cervantes, L. et al. (2020) Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 5, eabd7114.
Comprehensive mapping of immune perturbations associated with severe COVID-19.Crossref | GoogleScholarGoogle Scholar | 32669287PubMed |

[36]  Lee, W.S. et al. (2020) Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies. Nat. Microbiol. 5, 1185–1191.
Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies.Crossref | GoogleScholarGoogle Scholar | 32908214PubMed |

[37]  Parasa, S. et al. (2020) Prevalence of gastrointestinal symptoms and fecal viral shedding in patients with coronavirus disease 2019: a systematic review and meta-analysis. JAMA Netw. Open 3, e2011335.
Prevalence of gastrointestinal symptoms and fecal viral shedding in patients with coronavirus disease 2019: a systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar | 32525549PubMed |

[38]  Seydoux, E. et al. (2020) Analysis of a SARS-CoV-2-infected individual reveals development of potent neutralizing antibodies with limited somatic mutation. Immunity 53, 98–105.e5.
Analysis of a SARS-CoV-2-infected individual reveals development of potent neutralizing antibodies with limited somatic mutation.Crossref | GoogleScholarGoogle Scholar | 32561270PubMed |

[39]  Kaneko, N. et al. (2020) Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19. Cell 183, 143–157.e13.
Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19.Crossref | GoogleScholarGoogle Scholar | 32877699PubMed |

[40]  Lax, S.F. et al. (2020) Pulmonary arterial thrombosis in COVID-19 with fatal outcome: results from a prospective, single-center, clinicopathologic case series. Ann. Intern. Med. 173, 350–361.
Pulmonary arterial thrombosis in COVID-19 with fatal outcome: results from a prospective, single-center, clinicopathologic case series.Crossref | GoogleScholarGoogle Scholar | 32422076PubMed |

[41]  Elsoukkary, S.S. et al. (2021) Autopsy findings in 32 patients with COVID-19: a single-institution experience. Pathobiology 88, 56–68.
Autopsy findings in 32 patients with COVID-19: a single-institution experience.Crossref | GoogleScholarGoogle Scholar | 32942274PubMed |

[42]  Dijkman, R. et al. (2008) Human coronavirus NL63 and 229E seroconversion in children. J. Clin. Microbiol. 46, 2368–2373.
Human coronavirus NL63 and 229E seroconversion in children.Crossref | GoogleScholarGoogle Scholar | 18495857PubMed |

[43]  Edridge, A.W.D. et al. (2020) Seasonal coronavirus protective immunity is short-lasting. Nat. Med. 26, 1691–1693.
Seasonal coronavirus protective immunity is short-lasting.Crossref | GoogleScholarGoogle Scholar |

[44]  Zaunders, J. et al. (2017) Divergent expression of CXCR5 and CCR5 on CD4+ T cells and the paradoxical accumulation of T follicular helper cells during HIV infection. Front. Immunol. 8, 495.
Divergent expression of CXCR5 and CCR5 on CD4+ T cells and the paradoxical accumulation of T follicular helper cells during HIV infection.Crossref | GoogleScholarGoogle Scholar | 28553284PubMed |

[45]  Woldemeskel, B.A. et al. (2020) Healthy donor T cell responses to common cold coronaviruses and SARS-CoV-2. J. Clin. Invest. 130, 6631–6638.
Healthy donor T cell responses to common cold coronaviruses and SARS-CoV-2.Crossref | GoogleScholarGoogle Scholar | 32966269PubMed |

[46]  Le Bert, N. et al. (2020) SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 584, 457–462.
SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls.Crossref | GoogleScholarGoogle Scholar | 32668444PubMed |

[47]  Altmann, D.M. and Boyton, R.J. (2020) SARS-CoV-2 T cell immunity: specificity, function, durability, and role in protection. Sci. Immunol. 5, eabd6160.
SARS-CoV-2 T cell immunity: specificity, function, durability, and role in protection.Crossref | GoogleScholarGoogle Scholar | 33361161PubMed |

[48]  Bertoletti, A. et al. (2021) The T cell response to SARS-CoV-2: kinetic and quantitative aspects and the case for their protective role. Oxford Open Immunology 2, iqab006.
The T cell response to SARS-CoV-2: kinetic and quantitative aspects and the case for their protective role.Crossref | GoogleScholarGoogle Scholar |

[49]  Wilk, A.J. et al. (2020) A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 26, 1070–1076.
A single-cell atlas of the peripheral immune response in patients with severe COVID-19.Crossref | GoogleScholarGoogle Scholar | 32514174PubMed |

[50]  Zaunders, J.J. et al. (2005) Early proliferation of CCR5+ CD38+++ antigen-specific CD4+ Th1 effector cells during primary HIV-1 infection. Blood 106, 1660–1667.
Early proliferation of CCR5+ CD38+++ antigen-specific CD4+ Th1 effector cells during primary HIV-1 infection.Crossref | GoogleScholarGoogle Scholar | 15905189PubMed |

[51]  Ellebedy, A.H. et al. (2016) Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination. Nat. Immunol. 17, 1226–1234.
Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination.Crossref | GoogleScholarGoogle Scholar | 27525369PubMed |

[52]  Fahey, L.M. et al. (2011) Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells. J. Exp. Med. 208, 987–999.
Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells.Crossref | GoogleScholarGoogle Scholar | 21536743PubMed |

[53]  Zaunders, J.J. et al. (2009) High levels of human antigen-specific CD4+ T cells in peripheral blood revealed by stimulated coexpression of CD25 and CD134 (OX40). J. Immunol. 183, 2827–2836.
High levels of human antigen-specific CD4+ T cells in peripheral blood revealed by stimulated coexpression of CD25 and CD134 (OX40).Crossref | GoogleScholarGoogle Scholar | 19635903PubMed |

[54]  Mateus, J. et al. (2020) Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 370, 89–94.
Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans.Crossref | GoogleScholarGoogle Scholar | 32753554PubMed |

[55]  Li  Z.et al2020 SARS-CoV-2-specific T cell memory is long-lasting in the majority of convalsecent COVID-19 individuals.bioRxiv [Preprint]. 10.1101/2020.11.15.383463

[56]  Dan, J.M. et al. (2021) Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science 371, eabf4063.
Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection.Crossref | GoogleScholarGoogle Scholar | 33408181PubMed |

[57]  Orlov, M. et al. (2020) A case for targeting Th17 cells and IL-17A in SARS-CoV-2 infections. J. Immunol. 205, 892–898.
A case for targeting Th17 cells and IL-17A in SARS-CoV-2 infections.Crossref | GoogleScholarGoogle Scholar | 32651218PubMed |

[58]  Grant, R.A. et al. (2021) Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature 590, 635–641.
Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia.Crossref | GoogleScholarGoogle Scholar | 33429418PubMed |

[59]  Chen, Z. and John Wherry, E. (2020) T cell responses in patients with COVID-19. Nat. Rev. Immunol. 20, 529–536.
T cell responses in patients with COVID-19.Crossref | GoogleScholarGoogle Scholar | 32728222PubMed |

[60]  Chodick  G.et al2021 The effectiveness of the first dose of vaccine in reducing SARS-CoV-2 infection 13-24 days after immunization: real world evidence.medRxiv[Preprint].

[61]  Hall  V.et al2021 Effectiveness of BNT162b2 mRNA vaccine against infection and COVID-19 vaccine coverage in healthcare workers in England, multicentre prospective cohort study (the SIREN study).medRxiv[Preprint].


Biographies

Dr John Zaunders is a Senior Scientist at the Centre for Applied Medical Research at St Vincent’s Hospital, Sydney. He has studied CD4 T cells in HIV infection for over 25 years, particularly antigen-specific CD4 and CD8 T cells during acute HIV infection, and other acute infections, using high parameter flow cytometry.

Dr Chan Phetsouphanh recently returned to Sydney from his postdoctoral position at the University of Oxford (UK). He is currently working at the Kirby Institute (UNSW) as a senior research associate focusing on T-cell immunology. His projects involve the investigation of T-cell responses during primary SARS-CoV-2 infection and evaluating immune signatures of Long-COVID. He has keen interests in infectious diseases, immunotherapy, T-cell biology and immunopeptidomics.