ArticlePDF Available

Cellular immune responses to covid-19

Authors:
Cellular immune responses to covid-19
T cells could be valuable allies in pandemic control
Herb F Sewell, 1Raymond M Agius, 2Marcia Stewart, 3Denise Kendrick1
Protective and enduring immune responses to viral
infections or vaccines usually arise from the
combined actions of lymphocytes: B cells (responsible
for humoral antibody immunity) and T cells
(responsible for cellular immunity and helping B cell
responses).
B cells produce detectable antibodies in classes IgM,
IgG, and IgA along with lesser amounts of IgD and
IgE. For SARS-CoV-2, the causative agent of covid-19,
the focus is mainly on IgM, IgG, and IgA antibodies
that can neutralise the virus by binding to the spike
and other membrane proteins and thus preventing
infection.1Understanding the lesser known roles of
T cells and cellular immunity will deepen our insights
into covid-19 pathogenesis and help inform both
vaccine development and pandemic containment
strategies.
An effective immune response to SARS-CoV-2 involves
four types or subsets of T cells: T helper cells (CD4)
are responsible for cellular immunity and for helping
B cells to produce neutralising antibodies; cytotoxic
or killer T cells (CD8) directly kill infected cellsaided
by helper T cells2; other T cells (including T-17 (Th17)
cells) drive the inflammatory responses that help to
control infections3; and regulatory T cells (T regs)
help to contain the immune response, to prevent
over-reaction and damage to tissues.
CD4 T cells ensure all these components work
together by secreting small short acting cytokines
that bind to receptors on target cells. Importantly, all
B and T cell types have immunological memory after
a first encounter with a pathogen. This enables a
faster effective response after a second encounter
with the same pathogen or one that is closely related
(cross reaction).
Preliminary studies from the US and Europe recently
documented T cells specific to SARS-CoV-2 in people
with acute covid-19 and in those recovering from
infection.4 -6 They report helper and killer T cells
specific to SARS-CoV-2 in people with and without
antibodies.6More unexpectedly, they found specific
T cells in people with no history of exposure to
SARS-CoV-2individuals who had repeatedly
swabbed negative for the virus.4These cells have
even been found in stored blood taken before the
pandemic (2015-18). Finally, the studies identified
strong T cell memory responses in people recovering
from covid-19. Memory cells are critical for protective
and enduring immunity.
What are the implications of these early findings?
Principally, these studies show that a good T cell
immune response and immunological memory
accompany natural exposure to or infection with
SARS-CoV-2, that evidence of these responses is
present in some people who have apparently never
encountered the virus, and that T cell immune
responses can exist in the absence of detectable
antibodies.
Collectively, these features suggest that candidate
vaccines7should aim to stimulate both B cell
(neutralising) antibodies and T cell antiviral
responses.8Early phase clinical trials of candidate
vaccines developed in Oxford, UK, and in China do
show concomitant B cell neutralising antibodies and
antiviral T cells in vaccinated healthy volunteers,9 10
improving prospects for protective immunity. This
combined response is a feature of many successful
vaccines, including vaccines against varicella
(chickenpox), influenza, measles, and hepatitis B.
Immune memory
That some virus naiveparticipants in early studies
had pre-existing memory helper (50% of participants)
and killer T (20%) cells with potential activity against
SARS-CoV-2 is intriguing. These cells might arise from
cross reactions to other circulating coronaviruses,
such as some common cold viruses, and might be a
welcome hint of possible background immunity to
covid-19 in populations at riskeven in the absence
of antibodies.
Any cellular immune memory for SARS-CoV-2 in the
population could enhance responses to vaccines and
might also give a vaccination programme a head start
towards herd immunity. Herd immunity is population
resistance to spread of an infection, achieved when
a high enough proportion of individuals are immune,
usually through vaccination.11 12
Pre-existing memory helper T cells specific to
SARS-CoV-2 could boost the production of
neutralising IgG antibodies in the blood of newly
exposed people and could also enhance antibody
protection at mucosal surfaces through IgA in saliva,
tears, or nasal secretions.13 -15 Such IgA antibodies
act as a protective barrier at common viral entry
points. Research is now required to further
characterise these possible immune pathways
including memory B and T cells in mucosal tissues.
Research should also explore the role of regulatory
T cells in severe covid-19, particularly cytokine storm
syndrome16 and the documented association between
high titres of IgG antibodies and poorer disease
outcomes including death.17 Both might reflect
ineffective control of inflammation by regulatory T
cells.
Recent findings on the role of T cells in covid-19 give
us cause to be cautiously optimistic that cellular
immune responses could be a valuable ally in global
efforts to control this and future pandemics.
1the bmj | BMJ 2020;370:m3018 | doi: 10.1136/bmj.m3018
EDITORIALS
1University of Nottingham,
Nottingham, UK
2University of Manchester,
Manchester, UK
3De Montfort University, Leicester, UK
Correspondence to: H Sewell
herb.sewell@nottingham.ac.uk
Cite this as:
BMJ
2020;370:m3018
http://dx.doi.org/10.1136/bmj.m3018
Published: 31 July 2020
on 26 September 2020 by guest. Protected by copyright.http://www.bmj.com/BMJ: first published as 10.1136/bmj.m3018 on 31 July 2020. Downloaded from
Competing interests: The BMJ has judged that there are no disqualifying financial ties to commercial
companies. The authors declare the following other interests: None
Provenance and peer review: Commissioned, not peer reviewed
1Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity
of the SARSCoV-2 spike glycoprotein.
Cell
2020;181:281-292.e6. .
doi: 10.1016/j.cell.2020.02.058 pmid: 32155444
2Ahrends T, Busselaar J, Severson TM, etal. CD4+T cell help creates memory CD8+T cells with
innate and help-independent recall capacities.
Nat Commun
2019;10:5531.
doi: 10.1038/s41467-019-13438-1 pmid: 31797935
3Bettelli E, Oukka M, Kuchroo VK. T(H)-17 cells in the circle of immunity and autoimmunity.
Nat
Immunol
2007;8:345-50. doi: 10.1038/ni0407-345 pmid: 17375096
4Grifoni A, Weiskopf D, Ramirez SI, etal. Targets of T cell responses to SARS-CoV-2 coronavirus
in humans with COVID-19 disease and unexposed individuals.
Cell
2020;181:1489-1501.e15.
doi: 10.1016/j.cell.2020.05.015 pmid: 32473127
5Le Bert N, Tan AT, Kunasegaran K, et al. Different pattern of pre-existing SARS-COV-2 specific T
cell immunity in SARS-recovered and uninfected individuals. BioRxiv [Preprint].
https://www.biorxiv.org/content/10.1101/2020.05.26.115832v1
6Sekine T, Perez-Potti A, Rivera-Ballesteros O, et al. Robust T cell immunity in convalescent
individuals with asymptomatic or mild COVID-19. BioRxiv [Preprint] https://www.biorxiv.org/con-
tent/10.1101/2020.06.29.174888v1.
7Caddy S. Developing a vaccine for covid-19.
BMJ
2020;369:m1790.
doi: 10.1136/bmj.m1790 pmid: 32366511
8Panagioti E, Klenerman P, Lee LN, van der Burg SH, Arens R. Features of effective T cell-inducing
vaccines against chronic viral infections.
Front Immunol
2018;9:276.
doi: 10.3389/fimmu.2018.00276 pmid: 29503649
9Folegatti PM, Ewer KJ, Aley PK, etalOxford COVID Vaccine Trial Group. 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;.
doi: 10.1016/S0140-6736(20)31604-4. pmid: 32702298
10 Zhu F-C, Guan X-H, Li Y-H, etal. 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;.
doi: 10.1016/S0140-6736(20)31605-6. pmid: 32702299
11 Britton T, Ball F, Trapman P. A mathematical model reveals the influence of population
heterogeneity on herd immunity to SARS-CoV-2.
Science
2020;.
doi: 10.1126/science.abc6810. pmid: 32576668
12 Lourenço J, Pinotti F, Thompson C, Gupta S. The impact of host resistance on cumulative mortality
and the threshold of herd immunity for SARS-CoV-2. MedRxiv [Preprint]. https://www.medrx-
iv.org/content/10.1101/2020.07.15.20154294v1.
13 Strugnell RA, Wijburg OLC. The role of secretory antibodies in infection immunity.
Nat Rev
Microbiol
2010;8:656-67. doi: 10.1038/nrmicro2384 pmid: 20694027
14 Brandtzaeg P. Secretory IgA: designed for anti-microbial defense.
Front Immunol
2013;4:222.
doi: 10.3389/fimmu.2013.00222 pmid: 23964273
15 Zhao J, Zhao J, Mangalam AK, etal. Airway memory CD4(+) T cells mediate protective immunity
against emerging respiratory coronaviruses.
Immunity
2016;44:1379-91.
doi: 10.1016/j.immuni.2016.05.006 pmid: 27287409
16 Blanco-Melo D, Nilsson-Payant BE, Liu WC, etal. Imbalanced host response to SARS-CoV-2
drives development of covid-19.
Cell
2020;181:1036-1045.e9. .
doi: 10.1016/j.cell.2020.04.026 pmid: 32416070
17 Zhao J, Yuan Q, Wang H, etal. Antibody responses to SARS-CoV-2 in patients of novel coronavirus
disease 2019.
Clin Infect Dis
2020: . doi: 10.1093/cid/ciaa344 pmid: 32221519
This article is made freely available for use in accordance with BMJ's website terms and conditions for
the duration of the covid-19 pandemic or until otherwise determined by BMJ. You may use, download
and print the article for any lawful, non-commercial purpose (including text and data mining) provided
that all copyright notices and trade marks are retained.
the bmj | BMJ 2020;370:m3018 | doi: 10.1136/bmj.m30182
EDITORIALS
on 26 September 2020 by guest. Protected by copyright.http://www.bmj.com/BMJ: first published as 10.1136/bmj.m3018 on 31 July 2020. Downloaded from
... Antibodies can also tag the virus for destruction by other immune cells. The spike protein of the virus, which is responsible for binding to the host cell receptor, is the primary target of neutralizing antibodies [15]. ...
... 15 represents the inception V4 results on the CT scan images. ...
... 15: Performance Metrics of Inception V3 on X-ray Images Results of Inception V4 on CT Scan ImagesThe implementation of the Inception V4 model from scratch on CT scan images depicting COVID-19 cases resulted in exceptional outcomes. During training, the model achieved a remarkable accuracy of 100 percent, indicating that it accurately classified all instances in the training set. ...
Thesis
Full-text available
In recent times, the use of chest Computed Tomography (CT) images for detecting coronavirus infections has gained significant attention, owing to their ability to reveal bilateral changes in affected individuals. However, classifying patients from medical images presents a formidable challenge, particularly in identifying such bilateral changes. To tackle this challenge, our study harnesses the power of deep learning models for the precise classification of infected patients. Our research involves a comparative analysis of deep transfer learning-based classification models, including DenseNet201, GoogleNet, and AlexNet, against carefully chosen supervised learning models. Additionally, our work encompasses Covid-19 classification, which involves the identification and differentiation of medical images, such as X-rays and electrocardiograms, that exhibit telltale signs of Covid-19 infection. This comprehensive approach ensures that our models can handle a wide range of medical image types and effectively identify characteristic patterns indicative of Covid-19. By conducting meticulous research and employing advanced deep learning techniques, we have made significant strides in enhancing the accuracy and speed of Covid-19 diagnosis. Our results demonstrate the effectiveness of these models and their potential to make substantial contributions to the global effort to combat COVID-19.
... During SARS-CoV-2 infection, adaptive immune responses are needed to prevent disease progression to its severe stages [52] . The presence of specific immunoglobulin antibodies, such as IgA, IgM, and IgG, contributes to a better outcome for infected individuals [53][54][55][56][57] . These antibodies can neutralize the viral particles by binding to the S glycoprotein and other membrane proteins [58] . ...
... These antibodies can neutralize the viral particles by binding to the S glycoprotein and other membrane proteins [58] . Early and rapid antibody responses can be achieved by either naïve B cells or memory B cells that have been informed by prior human CoVs [56] . The balance between these two types of immune cells may have a positive effect on the clinical consequences of the viral infection [58] . ...
... T cells play a crucial role in combating SARS-CoV-2 infection both during acute illness and in recovered individuals. Specific CD4+ Thelper cells and killer CD8+ T cells have been detected in patients with and without humoral-mediated immunity, including those who repeatedly tested negative for the virus [56] . T-cell memory responses are also important for long-lasting cell immunity and can contribute to the production of virus-neutralizing antibodies in the blood and at mucosal infection sites [84] . ...
Article
Full-text available
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed a significant threat to the world, causing a respiratory illness with a wide range of symptoms and affecting people of all sexes and age groups. Although the innate immune response to SARS-CoV-2 is not entirely understood, recent findings suggest that a high number of innate immune cells and inflammatory cytokines can help in fighting the virus. However, an excessive expression of cytokines can lead to a cytokine storm, resulting in a severe disease state with high morbidity and mortality. The coordinated actions of the innate and adaptive immune responses are responsible for eliminating virus-infected cells and inhibiting reinfection. The actual role of T-cell immunity and antibody responses is unclear, and further research is necessary to gain insights into the nature of immune protection. The SARS-CoV-2 S protein is considered a sensitive and useful protein that can be targeted by antibodies and T cells, providing significant biological targets for finding effective vaccines and treatments. Despite challenges in developing effective vaccines, the small number of reinfections suggests that a primary infection may offer some protection during subsequent exposure to the same virus. A deeper understanding of the vital role of B and T cells in COVID-19 may help in managing, controlling, and halting this new pandemic.
... Based on a series of immune responses, we selected 15 molecules and 33 SNPs categorized as priming associated APC and T cell activation, B cell activation with T-B interaction, and B cell survival, which are key immunological steps in enhancing specific antibody production after antigen exposure (11,57,58). To understand the immune response as a multi-directional interacting network, genetic polymorphisms were analyzed using unbiased statistical methods such as stepwise regression. ...
Article
Full-text available
Background Vaccination against severe acute respiratory syndrome coronavirus type 2 is highly effective in preventing infection and reducing the severity of coronavirus disease (COVID-19). However, acquired humoral immunity wanes within six months. Focusing on the different tempo of acquisition and attenuation of specific antibody titers in individuals, we investigated the impact of genetic polymorphisms on antibody production after COVID-19 vaccination. Methods In total 236 healthcare workers from a Japanese municipal hospital, who received two doses of the vaccine were recruited. We employed a candidate gene approach to identify the target genetic polymorphisms affecting antibody production after vaccination. DNA samples from the study populations were genotyped for 33 polymorphisms in 15 distinct candidate genes encoding proteins involved in antigen-presenting cell activation, T cell activation, T-B interaction, and B cell survival. We measured total anti-SARS-Cov2 spike IgG antibody titers and analyzed the association with genetic polymorphisms at several time points after vaccination using an unbiased statistical method, and stepwise logistic regression following multivariate regression. Results Significant associations were observed between seven SNPs in NLRP3, OAS1, IL12B, CTLA4, and IL4, and antibody titers at 3 weeks after the first vaccination as an initial response. Six SNPs in NLRP3, TNF, OAS1, IL12B, and CTLA4 were associated with high responders with serum antibody titer > 4000 BAU/ml as boosting effect at 3 weeks after the second vaccination. Analysis of long-term maintenance showed the significance of the three SNPs in IL12B, IL7R, and MIF for the maintenance of antibody titers and that in BAFF for attenuation of neutralizing antibodies. Finally, we proposed a predictive model composed of gene profiles to identify the individuals with rapid antibody attenuation by receiver operating characteristic (ROC) analysis (area under the curve (AUC)= 0.76, sensitivity = 82.5%, specificity=67.8%). Conclusions The candidate gene approach successfully showed shifting responsible gene profiles and initial and boosting effect mainly related to the priming phase into antibody maintenance including B cell survival, which traces the phase of immune reactions. These gene profiles provide valuable information for further investigation of humoral immunity against COVID-19 and for building a strategy for personalized vaccine schedules.
... We reported increased GC content, altered size and end motif patterns ( Fig. 2A-D), which extended previous studies on elevated cfDNA concentration and neutrophil extracellular traps (NETs) in COVID-19 (Ng et al. 2020;Thierry and Roch 2020;Zuo et al. 2020;Hammad et al. 2021). COVID-19 patients suffer from active immune response to the viral infection and produces high level of immunoglobulins (Sewell et al. 2020;Wang et al. 2020b), which prefer binding/protecting GC-rich DNA (e.g., DNA molecules originated from the open chromatin regions) (Uccellini et al. 2012), suggesting that immune response may be responsible to the abnormalities in plasma cfDNA characteristics in COVID-19 patients. However, more efforts are needed to elucidate the underline biological mechanisms (e.g., chromatin status, replication timing, etc.). ...
Article
Full-text available
Coronavirus 2019 (COVID-19) is a complex disease that affects billions of people worldwide. Currently, effective etiological treatment of COVID-19 is still lacking; COVID-19 also causes damages to various organs that affects therapeutics and mortality of the patients. Surveillance of the treatment responses and organ injury assessment of COVID-19 patients are of high clinical value. In this study, we investigated the characteristic fragmentation patterns and explored the potential in tissue injury assessment of plasma cell-free DNA in COVID-19 patients. Through recruitment of 37 COVID-19 patients, 32 controls and analysis of 208 blood samples upon diagnosis and during treatment, we report gross abnormalities in cfDNA of COVID-19 patients, including elevated GC content, altered molecule size and end motif patterns. More importantly, such cfDNA fragmentation characteristics reflect patient-specific physiological changes during treatment. Further analysis on cfDNA tissue-of-origin tracing reveals frequent tissue injuries in COVID-19 patients, which is supported by clinical diagnoses. Hence, our work demonstrates and extends the translational merit of cfDNA fragmentation pattern as valuable analyte for effective treatment monitoring, as well as tissue injury assessment in COVID-19.
... Later, the importance of T-cell mediated immunity following COVID-19 also became the focus of investigation. However, similar B and T cell responses are expected following vaccination and now the scientific community is probing as to what role COVID-19 vaccines might play in developing protective and durable immune response to SARS-CoV-2 [2]. ...
Article
Full-text available
To determine the antibody levels at 6 months in SARS-CoV-2 vaccinated individuals in COVID-recovered versus non-infected groups to determine the need to administer booster COVID vaccine in each group. Prospective longitudinal study. Pathology Department, Combined Military Hospital, Lahore for a period of eight months from July 2021 to February 2022. Two hundred and thirty three study participants in both COVID recovered and non-infected groups (105 participants in infected group, 128 participants in non-infected group) were subjected to blood sampling at 6 months post-vaccination. Anti-SARS-CoV-2 IgG antibody test was done using Chemiluminescence method. Comparison of antibody levels between COVID-recovered and non-infected groups was made. Results were compiled and statistically analyzed using SPSS version 21. Out of 233 study participants, males were 183 (78%) while females were 50 (22%), mean age being 35.93 years ± 8.298. Mean Anti-SARS-CoV-2 S IgG levels among COVID-recovered group was 1342 U/ml and among non-infected group was 828 U/ml at 6 months post-vaccination. Mean antibody titers in COVID-19 recovered group are higher than in non-infected group at 6 months post-vaccination in both groups.
... Moreover, in this study, we also noted that the oral and nasal vaccination could induce the cellular immune response through the activation of CD8 + T cells in the intestine and lymph. The cellular immune response is crucial to counter the viral cells, including SARS-CoV2, and therefore the cellular immune response induced by the vaccine will bring many benefits [38,39]. ...
Article
Full-text available
The COVID-19 pandemic began at the end of 2019 in Wuhan, China, and has spread throughout the world. Vaccination is still the most effective method of prevention of pathogenic infections, including viral infections. However, there is little evidence that vaccination can protect against SARS-CoV-2 virus for a long time. Thus, regular re-vaccination is necessary to control COVID-19. Vaccination by injection is invasive, and is one of the reasons people refuse to get re-vaccinated. Therefore, we developed a less invasive vaccine based on oral or nasal administration. The gene encoding the high conserved region (HCR) spike protein was inserted into pNZ8149 and expressed in L.lactis NZ3900. Mice were immunized at 3-week intervals with oral or nasal routes. Anti-SARS-CoV2 spike antibody (IgG and IgA) level were measured using ELISA method before and after treatment. Plasma cells population in lymph were analyzed using flowcytometry and the CD4 + and CD8 + cells in lymph and intestine were analyzed using immunofluorescence method. The results of nasal and oral administration in experimental animals showed that L.lactis carrying the HCR gene could induce a humoral immune response, as indicated by increased levels of IgG and IgA against SARS-CoV-2 (IgG/IgA-SARS-CoV-2). The plasma cell population after nasal and oral vaccination in mice were significantly different with control group (p < 0.05). The CD4 + and CD8 + cells in intestine were significantly higher in orally immunized group mice than control group. The CD8 + cells in lymph were significantly higher in intranasal immunized group mice than control group. Our data demonstrate L.lactis expressing spike protein can be developed into a less invasive alternative to nasal and oral vaccination.
... Currently, more data are needed on the use of homologous boosters (same vaccine as double-dose vaccination) versus heterologous boosters (different vaccine as double-dose vaccination) in patients with cancer. In addition to the production of antiviral antibodies, the cellular immune response-in particular T-cell-mediated immune responsehas proven to be of significant importance in the defense against SARS-CoV-2 (21). Moreover, it has been described that T-cell responses are negatively correlated with COVID-19 severity (22). ...
Article
Full-text available
Purpose: Cancer patients display reduced humoral responses after double-dose COVID-19 vaccination while their cellular response is more comparable to that in healthy individuals. Recent studies demonstrated that a third vaccination dose boosts these immune responses, both in healthy people and cancer patients. Due to the availability of many different COVID-19 vaccines, many people have been boosted with a different vaccine from the one used for double-dose vaccination. Data on such alternative vaccination schedules are scarce. This prospective study compares a third dose of BNT162b2 after double-dose BNT162b2 (homologous) versus ChAdOx1 (heterologous) vaccination in cancer patients. Experimental design: 442 subjects (315 patients and 127 healthy) received a third dose of BNT162b2 (230 homologous vs 212 heterologous). Vaccine-induced adverse events (AE) were captured up to 7 days after vaccination. Humoral immunity was assessed by SARS-CoV-2 anti-S1 IgG antibody levels and SARSCoV-2 50% neutralization titers (NT50) against Wuhan and BA.1 Omicron strains. Cellular immunity was examined by analyzing CD4+ and CD8+ T cell responses against SARS-CoV-2 specific S1 and S2 peptides. Results: Local AEs were more common after heterologous boosting. SARS-CoV-2 anti-S1 IgG antibody levels did not differ significantly between homologous and heterologous boosted subjects (GMT 1755.90 BAU/mL [95% CI 1276.95-2414.48] vs 1495.82 BAU/mL (95% CI 1131.48-1977.46)). However, homologous boosted subjects show significantly higher NT50 values against BA.1 Omicron. Subjects receiving heterologous boosting demonstrated increased spike-specific CD8+ T cells, including higher IFNγ and TNFα levels. Conclusions: In cancer patients who received double-dose ChAdOx1, a third heterologous dose of BNT162b2 was able to close the gap in antibody response.
Article
Full-text available
Understanding the antibody response to SARS-CoV-2, the virus responsible for COVID-19, is crucial to comprehending disease progression and the significance of vaccine and therapeutic development. The emergence of highly contagious variants poses a significant challenge to humoral immunity, underscoring the necessity of grasping the intricacies of specific antibodies. This review emphasizes the pivotal role of antibodies in shaping immune responses and their implications for diagnosing, preventing, and treating SARS-CoV-2 infection. It delves into the kinetics and characteristics of the antibody response to SARS-CoV-2 and explores current antibody-based diagnostics, discussing their strengths, clinical utility, and limitations. Furthermore, we underscore the therapeutic potential of SARS-CoV-2-specific antibodies, discussing various antibody-based therapies such as monoclonal antibodies, polyclonal antibodies, anti-cytokines, convalescent plasma, and hyperimmunoglobulin-based therapies. Moreover, we offer insights into antibody responses to SARS-CoV-2 vaccines, emphasizing the significance of neutralizing antibodies in order to confer immunity to SARS-CoV-2, along with emerging variants of concern (VOCs) and circulating Omicron subvariants. We also highlight challenges in the field, such as the risks of antibody-dependent enhancement (ADE) for SARS-CoV-2 antibodies, and shed light on the challenges associated with the original antigenic sin (OAS) effect and long COVID. Overall, this review intends to provide valuable insights, which are crucial to advancing sensitive diagnostic tools, identifying efficient antibody-based therapeutics, and developing effective vaccines to combat the evolving threat of SARS-CoV-2 variants on a global scale.
Article
Full-text available
SARS-CoV-2-specific memory T cells will likely prove critical for long-term immune protection against COVID-19. We here systematically mapped the functional and phenotypic landscape of SARS-CoV-2-specific T cell responses in unexposed individuals, exposed family members, and individuals with acute or convalescent COVID-19. Acute phase SARS-CoV-2-specific T cells displayed a highly activated cytotoxic phenotype that correlated with various clinical markers of disease severity, whereas convalescent phase SARS-CoV-2-specific T cells were polyfunctional and displayed a stem-like memory phenotype. Importantly, SARS-CoV-2-specific T cells were detectable in antibody-seronegative exposed family members and convalescent individuals with a history of asymptomatic and mild COVID-19. Our collective dataset shows that SARS-CoV-2 elicits robust, broad and highly functional memory T cell responses, suggesting that natural exposure or infection may prevent recurrent episodes of severe COVID-19.
Article
Full-text available
Background The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) might be curtailed by vaccination. We assessed the safety, reactogenicity, and immunogenicity of a viral vectored coronavirus vaccine that expresses the spike protein of SARS-CoV-2. Methods We did a phase 1/2, single-blind, randomised controlled trial in five trial sites in the UK of a chimpanzee adenovirus-vectored vaccine (ChAdOx1 nCoV-19) expressing the SARS-CoV-2 spike protein compared with a meningococcal conjugate vaccine (MenACWY) as control. Healthy adults aged 18–55 years with no history of laboratory confirmed SARS-CoV-2 infection or of COVID-19-like symptoms were randomly assigned (1:1) to receive ChAdOx1 nCoV-19 at a dose of 5 × 10¹⁰ viral particles or MenACWY as a single intramuscular injection. A protocol amendment in two of the five sites allowed prophylactic paracetamol to be administered before vaccination. Ten participants assigned to a non-randomised, unblinded ChAdOx1 nCoV-19 prime-boost group received a two-dose schedule, with the booster vaccine administered 28 days after the first dose. Humoral responses at baseline and following vaccination were assessed using a standardised total IgG ELISA against trimeric SARS-CoV-2 spike protein, a muliplexed immunoassay, three live SARS-CoV-2 neutralisation assays (a 50% plaque reduction neutralisation assay [PRNT50]; a microneutralisation assay [MNA50, MNA80, and MNA90]; and Marburg VN), and a pseudovirus neutralisation assay. Cellular responses were assessed using an ex-vivo interferon-γ enzyme-linked immunospot assay. The co-primary outcomes are to assess efficacy, as measured by cases of symptomatic virologically confirmed COVID-19, and safety, as measured by the occurrence of serious adverse events. Analyses were done by group allocation in participants who received the vaccine. Safety was assessed over 28 days after vaccination. Here, we report the preliminary findings on safety, reactogenicity, and cellular and humoral immune responses. The study is ongoing, and was registered at ISRCTN, 15281137, and ClinicalTrials.gov, NCT04324606. Findings Between April 23 and May 21, 2020, 1077 participants were enrolled and assigned to receive either ChAdOx1 nCoV-19 (n=543) or MenACWY (n=534), ten of whom were enrolled in the non-randomised ChAdOx1 nCoV-19 prime-boost group. Local and systemic reactions were more common in the ChAdOx1 nCoV-19 group and many were reduced by use of prophylactic paracetamol, including pain, feeling feverish, chills, muscle ache, headache, and malaise (all p<0·05). There were no serious adverse events related to ChAdOx1 nCoV-19. In the ChAdOx1 nCoV-19 group, spike-specific T-cell responses peaked on day 14 (median 856 spot-forming cells per million peripheral blood mononuclear cells, IQR 493–1802; n=43). Anti-spike IgG responses rose by day 28 (median 157 ELISA units [EU], 96–317; n=127), and were boosted following a second dose (639 EU, 360–792; n=10). Neutralising antibody responses against SARS-CoV-2 were detected in 32 (91%) of 35 participants after a single dose when measured in MNA80 and in 35 (100%) participants when measured in PRNT50. After a booster dose, all participants had neutralising activity (nine of nine in MNA80 at day 42 and ten of ten in Marburg VN on day 56). Neutralising antibody responses correlated strongly with antibody levels measured by ELISA (R²=0·67 by Marburg VN; p<0·001). Interpretation ChAdOx1 nCoV-19 showed an acceptable safety profile, and homologous boosting increased antibody responses. These results, together with the induction of both humoral and cellular immune responses, support large-scale evaluation of this candidate vaccine in an ongoing phase 3 programme. Funding UK Research and Innovation, Coalition for Epidemic Preparedness Innovations, National Institute for Health Research (NIHR), NIHR Oxford Biomedical Research Centre, Thames Valley and South Midland's NIHR Clinical Research Network, and the German Center for Infection Research (DZIF), Partner site Gießen-Marburg-Langen.
Article
Full-text available
Heterogeneity and herd immunity In response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), some politicians have been keen to exploit the idea of achieving herd immunity. Countering this possibility are estimates derived from work on historical vaccination studies, which suggest that herd immunity may only be achieved at an unacceptable cost of lives. Because human populations are far from homogeneous, Britton et al. show that by introducing age and activity heterogeneities into population models for SARS-CoV-2, herd immunity can be achieved at a population-wide infection rate of ∼40%, considerably lower than previous estimates. This shift is because transmission and immunity are concentrated among the most active members of a population, who are often younger and less vulnerable. If nonpharmaceutical interventions are very strict, no herd immunity is achieved, and infections will then resurge if they are eased too quickly. Science , this issue p. 846
Article
Full-text available
Understanding adaptive immunity to SARS-CoV-2 is important for vaccine development, interpreting coronavirus disease 2019 (COVID-19) pathogenesis, and calibration of pandemic control measures. Using HLA class I and II predicted peptide ‘megapools’, circulating SARS-CoV-2−specific CD8⁺ and CD4⁺ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, respectively. CD4⁺ T cell responses to spike, the main target of most vaccine efforts, were robust and correlated with the magnitude of the anti-SARS-CoV-2 IgG and IgA titers. The M, spike and N proteins each accounted for 11-27% of the total CD4⁺ response, with additional responses commonly targeting nsp3, nsp4, ORF3a and ORF8, among others. For CD8⁺ T cells, spike and M were recognized, with at least eight SARS-CoV-2 ORFs targeted. Importantly, we detected SARS-CoV-2−reactive CD4⁺ T cells in ∼40-60% of unexposed individuals, suggesting cross-reactive T cell recognition between circulating ‘common cold’ coronaviruses and SARS-CoV-2.
Article
Full-text available
Viral pandemics, such as the one caused by SARS-CoV-2, pose an imminent threat to humanity. Because of its recent emergence, there is a paucity of information regarding viral behavior and host response following SARS-CoV-2 infection. Here we offer an in-depth analysis of the transcriptional response to SARS-CoV-2 compared with other respiratory viruses. Cell and animal models of SARS-CoV-2 infection, in addition to transcriptional and serum profiling of COVID-19 patients, consistently revealed a unique and inappropriate inflammatory response. This response is defined by low levels of type I and III interferons juxtaposed to elevated chemokines and high expression of IL-6. We propose that reduced innate antiviral defenses coupled with exuberant inflammatory cytokine production are the defining and driving features of COVID-19.
Article
Full-text available
Background: The novel coronavirus SARS-CoV-2 is a newly emerging virus. The antibody response in infected patient remains largely unknown, and the clinical values of antibody testing have not been fully demonstrated. Methods: A total of 173 patients with SARS-CoV-2 infection were enrolled. Their serial plasma samples (n=535) collected during the hospitalization were tested for total antibodies (Ab), IgM and IgG against SARS-CoV-2. The dynamics of antibodies with the disease progress was analyzed. Results: Among 173 patients, the seroconversion rate for Ab, IgM and IgG was 93.1%, 82.7% and 64.7%, respectively. The reason for the negative antibody findings in 12 patients might due to the lack of blood samples at the later stage of illness. The median seroconversion time for Ab, IgM and then IgG were day-11, day-12 and day-14, separately. The presence of antibodies was <40% among patients within 1-week since onset, and rapidly increased to 100.0% (Ab), 94.3% (IgM) and 79.8% (IgG) since day-15 after onset. In contrast, RNA detectability decreased from 66.7% (58/87) in samples collected before day-7 to 45.5% (25/55) during day 15-39. Combining RNA and antibody detections significantly improved the sensitivity of pathogenic diagnosis for COVID-19 (p<0.001), even in early phase of 1-week since onset (p=0.007). Moreover, a higher titer of Ab was independently associated with a worse clinical classification (p=0.006). Conclusions: The antibody detection offers vital clinical information during the course of SARS-CoV-2 infection. The findings provide strong empirical support for the routine application of serological testing in the diagnosis and management of COVID-19 patients.
Article
Full-text available
CD4⁺ T cell help is required for the generation of CD8⁺ cytotoxic T lymphocyte (CTL) memory. Here, we use genome-wide analyses to show how CD4⁺ T cell help delivered during priming promotes memory differentiation of CTLs. Help signals enhance IL-15-dependent maintenance of central memory T (TCM) cells. More importantly, help signals regulate the size and function of the effector memory T (TEM) cell pool. Helped TEM cells produce Granzyme B and IFNγ upon antigen-independent, innate-like recall by IL-12 and IL-18. In addition, helped memory CTLs express the effector program characteristic of helped primary CTLs upon recall with MHC class I-restricted antigens, likely due to epigenetic imprinting and sustained mRNA expression of effector genes. Our data thus indicate that during priming, CD4⁺ T cell help optimizes CTL memory by creating TEM cells with innate and help-independent antigen-specific recall capacities.
Article
Background This is the first randomised controlled trial for assessment of the immunogenicity and safety of a candidate non-replicating adenovirus type-5 (Ad5)-vectored COVID-19 vaccine, aiming to determine an appropriate dose of the candidate vaccine for an efficacy study. Methods This randomised, double-blind, placebo-controlled, phase 2 trial of the Ad5-vectored COVID-19 vaccine was done in a single centre in Wuhan, China. Healthy adults aged 18 years or older, who were HIV-negative and previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection-free, were eligible to participate and were randomly assigned to receive the vaccine at a dose of 1 × 10¹¹ viral particles per mL or 5 × 10¹⁰ viral particles per mL, or placebo. Investigators allocated participants at a ratio of 2:1:1 to receive a single injection intramuscularly in the arm. The randomisation list (block size 4) was generated by an independent statistician. Participants, investigators, and staff undertaking laboratory analyses were masked to group allocation. The primary endpoints for immunogenicity were the geometric mean titres (GMTs) of specific ELISA antibody responses to the receptor binding domain (RBD) and neutralising antibody responses at day 28. The primary endpoint for safety evaluation was the incidence of adverse reactions within 14 days. All recruited participants who received at least one dose were included in the primary and safety analyses. This study is registered with ClinicalTrials.gov, NCT04341389. Findings 603 volunteers were recruited and screened for eligibility between April 11 and 16, 2020. 508 eligible participants (50% male; mean age 39·7 years, SD 12·5) consented to participate in the trial and were randomly assigned to receive the vaccine (1 × 10¹¹ viral particles n=253; 5 × 10¹⁰ viral particles n=129) or placebo (n=126). In the 1 × 10¹¹ and 5 × 10¹⁰ viral particles dose groups, the RBD-specific ELISA antibodies peaked at 656·5 (95% CI 575·2–749·2) and 571·0 (467·6–697·3), with seroconversion rates at 96% (95% CI 93–98) and 97% (92–99), respectively, at day 28. Both doses of the vaccine induced significant neutralising antibody responses to live SARS-CoV-2, with GMTs of 19·5 (95% CI 16·8–22·7) and 18·3 (14·4–23·3) in participants receiving 1 × 10¹¹ and 5 × 10¹⁰ viral particles, respectively. Specific interferon γ enzyme-linked immunospot assay responses post vaccination were observed in 227 (90%, 95% CI 85–93) of 253 and 113 (88%, 81–92) of 129 participants in the 1 × 10¹¹ and 5 × 10¹⁰ viral particles dose groups, respectively. Solicited adverse reactions were reported by 183 (72%) of 253 and 96 (74%) of 129 participants in the 1 × 10¹¹ and 5 × 10¹⁰ viral particles dose groups, respectively. Severe adverse reactions were reported by 24 (9%) participants in the 1 × 10¹¹ viral particles dose group and one (1%) participant in the 5 × 10¹⁰ viral particles dose group. No serious adverse reactions were documented. Interpretation The Ad5-vectored COVID-19 vaccine at 5 × 10¹⁰ viral particles is safe, and induced significant immune responses in the majority of recipients after a single immunisation. Funding National Key R&D Programme of China, National Science and Technology Major Project, and CanSino Biologics.
Article
The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3,000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We determined cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.